1
|
Motallebzadeh Khanmiri J, Alizadeh M, Esmaeili S, Gholami Z, Safarzadeh A, Khani-Eshratabadi M, Baghbanzadeh A, Alizadeh N, Baradaran B. Dendritic cell vaccination strategy for the treatment of acute myeloid leukemia: a systematic review. Cytotherapy 2024; 26:427-435. [PMID: 38483358 DOI: 10.1016/j.jcyt.2024.02.009] [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: 09/03/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND AIMS Acute myeloid leukemia (AML) is classified as a hematologic malignancy characterized by the proliferation of immature blood cells within the bone marrow (BM), resulting in an aberrant and unregulated cellular growth. The primary therapeutic modalities for AML include chemotherapy and hematopoietic stem cell transplantation. However, it is important to note that these treatments are accompanied by important adverse effects and mortality rates. Therefore, the need for more effective treatment options seems necessary, and dendritic cell (DC) vaccine therapy can be one of these options. In this study, we aim to investigate the effectiveness of DC vaccination therapy for the management of AML. METHODS PubMed, Scopus, ProQuest, Web of Science, and Google Scholar databases were searched for this systematic review. The articles were evaluated based on the inclusion criteria of this study and initially compared in terms of titles or abstracts. Finally, the articles related to the topic of this review were obtained in full text. The complete remission and partial remission, survival, correlative immune assays, and health-related metrics were used to evaluate this cellular immunotherapy effectiveness. The quality of the studies was assessed independently using the Cochrane risk-of-bias tools. The compiled data were input into a standard Excel spreadsheet. Each domain was evaluated as having either a "low risk," "high risk," or "unclear risk" of bias. RESULTS Among the 3986 studies that were determined, a total of 11 correlated trials were selected for inclusion in this systematic review. DC vaccine therapy was effective in inducing complete and partial remission, and stabilization of the disease. Additionally, it was discovered that the treatment strengthened the immune system as seen by increased levels of CD4+ and CD8+ T cells, Th1 cytokines, WT1-specific T cells, and activated NK cells. CONCLUSION We conducted a systematic review that supports the use of DC vaccine therapy as an effective treatment for AML. The therapy demonstrated potentials in achieving remission, enhancing the immune system function, and increasing overall survival. However, more studies are required to improve the methods of preparing and delivering the DC vaccine, and to confirm its long-term safety and effectiveness.
Collapse
Affiliation(s)
- Jamal Motallebzadeh Khanmiri
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Esmaeili
- Student Research Committee, Shahed University, Tehran, Iran
| | - Zeinab Gholami
- Faculty of Medicine, University of Medical Sciences, Tabriz, Iran
| | - Ali Safarzadeh
- Department of Biology, University of Padova, Padova, Italy
| | | | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
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
|
Dendritic Cell-Triggered Immune Activation Goes along with Provision of (Leukemia-Specific) Integrin Beta 7-Expressing Immune Cells and Improved Antileukemic Processes. Int J Mol Sci 2022; 24:ijms24010463. [PMID: 36613907 PMCID: PMC9820538 DOI: 10.3390/ijms24010463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Integrin beta 7 (β7), a subunit of the integrin receptor, is expressed on the surface of immune cells and mediates cell-cell adhesions and interactions, e.g., antitumor or autoimmune reactions. Here, we analyzed, whether the stimulation of immune cells by dendritic cells (of leukemic derivation in AML patients or of monocyte derivation in healthy donors) leads to increased/leukemia-specific β7 expression in immune cells after T-cell-enriched mixed lymphocyte culture-finally leading to improved antileukemic cytotoxicity. Healthy, as well as AML and MDS patients' whole blood (WB) was treated with Kit-M (granulocyte-macrophage colony-stimulating factor (GM-CSF) + prostaglandin E1 (PGE1)) or Kit-I (GM-CSF + Picibanil) in order to generate DCs (DCleu or monocyte-derived DC), which were then used as stimulator cells in MLC. To quantify antigen/leukemia-specific/antileukemic functionality, a degranulation assay (DEG), an intracellular cytokine assay (INTCYT) and a cytotoxicity fluorolysis assay (CTX) were used. (Leukemia-specific) cell subtypes were quantified via flow cytometry. The Kit treatment of WB (compared to the control) resulted in the generation of DC/DCleu, which induced increased activation of innate and adaptive cells after MLC. Kit-pretreated WB (vs. the control) led to significantly increased frequencies of β7-expressing T-cells, degranulating and intracellular cytokine-producing β7-expressing immune cells and, in patients' samples, increased blast lysis. Positive correlations were found between the Kit-M-mediated improvement of blast lysis (vs. the control) and frequencies of β7-expressing T-cells. Our findings indicate that DC-based immune therapies might be able to specifically activate the immune system against blasts going along with increased frequencies of (leukemia-specific) β7-expressing immune cells. Furthermore, β7 might qualify as a predictor for the efficiency and the success of AML and/or MDS therapies.
Collapse
|
4
|
Bastin DJ, Quizi J, Kennedy MA, Kekre N, Auer RC. Current challenges in the manufacture of clinical-grade autologous whole cell vaccines for hematological malignancies. Cytotherapy 2022; 24:979-989. [PMID: 35562303 DOI: 10.1016/j.jcyt.2022.03.010] [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: 01/17/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/03/2022]
Abstract
Autologous whole cell vaccines use a patient's own tumor cells as a source of antigen to elicit an anti-tumor immune response in vivo. Recently, the authors conducted a systematic review of clinical trials employing these products in hematological cancers that showed a favorable safety profile and trend toward efficacy. However, it was noted that manufacturing challenges limit both the efficacy and clinical implementation of these vaccine products. In the current literature review, the authors sought to define the issues surrounding the manufacture of autologous whole cell products for hematological cancers. The authors describe key factors, including the acquisition, culture, cryopreservation and transduction of malignant cells, that require optimization for further advancement of the field. Furthermore, the authors provide a summary of pre-clinical work that informs how the identified challenges may be overcome. The authors also highlight areas in which future basic research would be of benefit to the field. The goal of this review is to provide a roadmap for investigators seeking to advance the field of autologous cell vaccines as it applies to hematological malignancies.
Collapse
Affiliation(s)
- Donald J Bastin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Schulich School of Medicine, Western University, London, Canada
| | - Jennifer Quizi
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Michael A Kennedy
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Natasha Kekre
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Rebecca C Auer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada; Department of Surgery, University of Ottawa, Ottawa, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
5
|
Klauer LK, Schutti O, Ugur S, Doraneh-Gard F, Amberger DC, Rogers N, Krämer D, Rank A, Schmid C, Eiz-Vesper B, Schmetzer HM. Interferon Gamma Secretion of Adaptive and Innate Immune Cells as a Parameter to Describe Leukaemia-Derived Dendritic-Cell-Mediated Immune Responses in Acute Myeloid Leukaemia in vitro. Transfus Med Hemother 2022; 49:44-61. [PMID: 35221867 PMCID: PMC8832209 DOI: 10.1159/000516886] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/28/2021] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION Myeloid leukaemic blasts can be converted into leukaemia-derived dendritic cells (DCleu), characterised by the simultaneous expression of dendritic- and leukaemia-associated antigens, which have the competence to prime and enhance (leukaemia-specific) immune responses with the whole leukaemic antigen repertoire. To display and further specify dendritic cell (DC)- and DCleu-mediated immune responses, we analysed the interferon gamma (IFNy) secretion of innate and adaptive immune cells. METHODS DC/DCleu were generated from leukaemic whole blood (WB) with (blast)modulatory Kit-I (granulocyte-macrophage colony-stimulating factor [GM-CSF] + Picibanil [OK-432]) and Kit-M (GM-CSF + prostaglandin E1) and were used to stimulate T cell-enriched immunoreactive cells. Initiated anti-leukaemic cytotoxicity was investigated with a cytotoxicity fluorolysis assay. Initiated IFNy secretion of T, NK, CIK, and iNKT cells was investigated with a cytokine secretion assay (CSA). IFNy positivity was additionally evaluated with an intracellular cytokine assay (ICA). Recent activation of leukaemia-specific cells was verified through addition of leukaemia-associated antigens (LAA; WT-1 and Prame). RESULTS We found Kit-I and Kit-M competent to generate mature DC and DCleu from leukaemic WB without induction of blast proliferation. Stimulation of immunoreactive cells with DC/DCleu regularly resulted in an increased anti-leukaemic cytotoxicity and increased IFNy secretion of T, NK, and CIK cells, pointing to the significant role of DC/DCleu in leukaemia-specific alongside anti-leukaemic reactions. Interestingly, an addition of LAA did not further increase IFNy secretion, suggesting an efficient activation of leukaemia-specific cells. Here, both the CSA and ICA yielded comparable frequencies of IFNy-positive cells. Remarkably, the anti-leukaemic cytotoxicity positively correlated with the IFNy secretion in TCD3+, TCD4+, TCD8+, and NKCD56+ cells. CONCLUSION Ultimately, the IFNy secretion of innate and adaptive immune cells appeared to be a suitable parameter to assess and monitor the efficacy of in vitro and potentially in vivo acute myeloid leukaemia immunotherapy. The CSA in this regard proved to be a convenient and reproducible technique to detect and phenotypically characterise IFNy-secreting cells. In respect to our studies on DC-based immunomodulation, we were able to display the potential of DC/DCleu to induce or improve leukaemia-specific and anti-leukaemic activity.
Collapse
Affiliation(s)
| | - Olga Schutti
- Department of Medicine III, University Hospital of Munich, Munich, Germany
| | - Selda Ugur
- Department of Medicine III, University Hospital of Munich, Munich, Germany
| | | | | | - Nicole Rogers
- Department of Medicine III, University Hospital of Munich, Munich, Germany
| | - Doris Krämer
- Department of Haematology and Oncology, St.-Josefs-Hospital, Hagen, Germany
| | - Andreas Rank
- Department of Haematology and Oncology, University Hospital of Augsburg, Augsburg, Germany
| | - Christoph Schmid
- Department of Haematology and Oncology, University Hospital of Augsburg, Augsburg, Germany
| | - Britta Eiz-Vesper
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
6
|
Yu J, Sun H, Cao W, Song Y, Jiang Z. Research progress on dendritic cell vaccines in cancer immunotherapy. Exp Hematol Oncol 2022; 11:3. [PMID: 35074008 PMCID: PMC8784280 DOI: 10.1186/s40164-022-00257-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/16/2022] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell (DC) vaccines induce specific immune responses that can selectively eliminate target cells. In recent years, many studies have been conducted to explore DC vaccination in the treatment of hematological malignancies, including acute myeloid leukemia and myelodysplastic syndromes, as well as other nonleukemia malignancies. There are at least two different strategies that use DCs to promote antitumor immunity: in situ vaccination and canonical vaccination. Monocyte-derived DCs (mo-DCs) and leukemia-derived DCs (DCleu) are the main types of DCs used in vaccines for AML and MDS thus far. Different cancer-related molecules such as peptides, recombinant proteins, apoptotic leukemic cells, whole tumor cells or lysates and DCs/DCleu containing a vaster antigenic repertoire with RNA electroporation, have been used as antigen sources to load DCs. To enhance DC vaccine efficacy, new strategies, such as combination with conventional chemotherapy, monospecific/bispecific antibodies and immune checkpoint-targeting therapies, have been explored. After a decade of trials and tribulations, much progress has been made and much promise has emerged in the field. In this review we summarize the recent advances in DC vaccine immunotherapy for AML/MDS as well as other nonleukemia malignancies.
Collapse
Affiliation(s)
- Jifeng Yu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan International Joint Laboratory of Nuclear Protein Gene Regulation, Henan University College of Medicine, Kaifeng, 475004, Henan, China
| | - Hao Sun
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Weijie Cao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yongping Song
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, Henan, China.
| | - Zhongxing Jiang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
7
|
Acute myeloid leukemia cell membrane-coated nanoparticles for cancer vaccination immunotherapy. Leukemia 2022; 36:994-1005. [PMID: 34845316 PMCID: PMC8979812 DOI: 10.1038/s41375-021-01432-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 09/08/2021] [Accepted: 09/17/2021] [Indexed: 12/21/2022]
Abstract
Cancer vaccines are promising treatments to prevent relapse after chemotherapy in acute myeloid leukemia (AML) patients, particularly for those who cannot tolerate intensive consolidation therapies. Here, we report the development of an AML cell membrane-coated nanoparticle (AMCNP) vaccine platform, in which immune-stimulatory adjuvant-loaded nanoparticles are coated with leukemic cell membrane material. This AMCNP vaccination strategy stimulates leukemia-specific immune responses by co-delivering membrane-associated antigens along with adjuvants to antigen-presenting cells. To demonstrate that this AMCNP vaccine enhances leukemia-specific antigen presentation and T cell responses, we modified a murine AML cell line to express membrane-bound chicken ovalbumin as a model antigen. AMCNPs were efficiently acquired by antigen-presenting cells in vitro and in vivo and stimulated antigen cross-presentation. Vaccination with AMCNPs significantly enhanced antigen-specific T cell expansion and effector function compared with control vaccines. Prophylactic vaccination with AMCNPs enhanced cellular immunity and protected against AML challenge. Moreover, in an AML post-remission vaccination model, AMCNP vaccination significantly enhanced survival in comparison to vaccination with whole leukemia cell lysates. Collectively, AMCNPs retained AML-specific antigens, elicited enhanced antigen-specific immune responses, and provided therapeutic benefit against AML challenge.
Collapse
|
8
|
Bastin DJ, Khan ST, Montroy J, Kennedy MA, Forbes N, Martel AB, Baker L, Gresham L, Boucher DM, Wong B, Shorr R, Diallo JS, Fergusson DA, Lalu MM, Auer RC, Kekre N. Safety and efficacy of autologous whole cell vaccines in hematologic malignancies: A systematic review and meta-analysis. Hematol Oncol 2021; 39:448-464. [PMID: 33963789 DOI: 10.1002/hon.2875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/26/2021] [Indexed: 01/07/2023]
Abstract
Autologous cell vaccines use a patient's tumor cells to stimulate a broad antitumor response in vivo. This approach shows promise for treating hematologic cancers in early phase clinical trials, but overall safety and efficacy remain poorly described. We conducted a systematic review assessing the use of autologous cell vaccination in treating hematologic cancers. Primary outcomes of interest were safety and clinical response, with secondary outcomes including survival, relapse rate, correlative immune assays and health-quality related metrics. We performed a search of MEDLINE, Embase and the Cochrane Register of Controlled Trials including any interventional trial employing an autologous, whole cell product in any hematologic malignancy. Risk of bias was assessed using a modified Institute of Health Economics tool. Across 20 single arm studies, only 341 of 592 enrolled participants received one or more vaccinations. Primary reasons for not receiving vaccination included rapid disease progression/death and manufacturing challenges. Overall, few high-grade adverse events were observed. One death was reported and attributed to a GM-CSF producing allogeneic cell line co-administered with the autologous vaccine. Of 58 evaluable patients, the complete response rate was 21.0% [95% CI, 10.4%-37.8%)] and overall response rate was 35.8% (95% CI, 24.4%-49.0%). Of 97 evaluable patients for survival, the 5-years overall survival rate was 64.9% (95% CI, 52.6%-77.2%) and disease-free survival was 59.7% (95% CI, 47.7%-71.7%). We conclude that, in hematologic malignancies, based on limited available data, autologous cell vaccines are safe and display a trend towards efficacy but that challenges exist in vaccine manufacture and administration.
Collapse
Affiliation(s)
- Donald J Bastin
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Schulich School of Medicine, Western University, London, ON, Canada
| | - Sarwat T Khan
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Joshua Montroy
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Michael A Kennedy
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Nicole Forbes
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Andre B Martel
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Laura Baker
- Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Louise Gresham
- Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Dominique M Boucher
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Boaz Wong
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Risa Shorr
- Learning Services, The Ottawa Hospital, Ottawa, ON, Canada
| | - Jean-Simon Diallo
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Manoj M Lalu
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada.,Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Rebecca C Auer
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Natasha Kekre
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Medicine and The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
9
|
Amberger DC, Schmetzer HM. Dendritic Cells of Leukemic Origin: Specialized Antigen-Presenting Cells as Potential Treatment Tools for Patients with Myeloid Leukemia. Transfus Med Hemother 2021; 47:432-443. [PMID: 33442338 DOI: 10.1159/000512452] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/12/2020] [Indexed: 11/19/2022] Open
Abstract
The prognosis of elderly patients with acute myeloid leukemia (AML) and high-grade myelodysplastic syndrome (MDS) is limited due to the lack of therapy options and high relapse rates. Dendritic cell (DC)-based immunotherapy seems to be a promising treatment tool. DC are potent antigen-presenting cells and play a pivotal role on the interface of the innate and the adaptive immune system. Myeloid leukemia blasts can be converted to DC of leukemic origin (DCleu), expressing costimulatory molecules along with the whole leukemic antigen repertoire of individual patients. These generated DCleu are potent stimulators of various immune reactive cells and increase antileukemic immunity ex vivo. Here we review the generating process of DC/DCleu from leukemic peripheral blood mononuclear cells as well as directly from leukemic whole blood with "minimized" Kits to simulate physiological conditions ex vivo. The purpose of adoptive cell transfer of DC/DCleu as a vaccination strategy is discussed. A new potential therapy option with Kits for patients with myeloid leukemia, which would render an adoptive DC/DCleu transfer unnecessary, is presented. In summary, DC/DCleu-based therapies seem to be promising treatment tools for patients with AML or MDS but ongoing research including trials in animals and humans have to be performed.
Collapse
Affiliation(s)
| | - Helga Maria Schmetzer
- Department of Medicine III, University Hospital, Hematopoetic Cell Transplantation, Munich, Germany
| |
Collapse
|
10
|
Motais B, Charvátová S, Hrdinka M, Šimíček M, Jelínek T, Ševčíková T, Kořístek Z, Hájek R, Bagó JR. A Bird's-Eye View of Cell Sources for Cell-Based Therapies in Blood Cancers. Cancers (Basel) 2020; 12:E1333. [PMID: 32456165 PMCID: PMC7281611 DOI: 10.3390/cancers12051333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/25/2022] Open
Abstract
: Hematological malignancies comprise over a hundred different types of cancers and account for around 6.5% of all cancers. Despite the significant improvements in diagnosis and treatment, many of those cancers remain incurable. In recent years, cancer cell-based therapy has become a promising approach to treat those incurable hematological malignancies with striking results in different clinical trials. The most investigated, and the one that has advanced the most, is the cell-based therapy with T lymphocytes modified with chimeric antigen receptors. Those promising initial results prepared the ground to explore other cell-based therapies to treat patients with blood cancer. In this review, we want to provide an overview of the different types of cell-based therapies in blood cancer, describing them according to the cell source.
Collapse
Affiliation(s)
- Benjamin Motais
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
| | - Sandra Charvátová
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
| | - Matouš Hrdinka
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Michal Šimíček
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Tomáš Jelínek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Tereza Ševčíková
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Zdeněk Kořístek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Roman Hájek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Juli R. Bagó
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| |
Collapse
|
11
|
Ji YS, Park SK, Ryu S. Whole leukemia cell vaccines: Past progress and future directions. Vaccine 2020; 38:3811-3820. [PMID: 32280046 DOI: 10.1016/j.vaccine.2020.03.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/17/2020] [Accepted: 03/21/2020] [Indexed: 11/28/2022]
Abstract
It has long been recognized that allogeneic hematopoietic stem cell transplantation can reduce the risk of leukemia relapse by inducing the graft-versus-leukemia effect. However, allogeneic stem cell transplantation is also known to be able to cause graft-versus-host disease, which can cause considerable morbidity and even mortality in patients receiving allogeneic hematopoietic stem cell transplantation. Therefore, to elicit leukemia-specific immune responses without alloimmune reaction, the possibilities of active immunotherapy methods such as leukemia vaccines have been studied for decades. Among various types of leukemia vaccines, whole leukemia cell vaccines are known to be able to induce immune responses against multiple unknown antigens without the need for adoptive transfer of dendritic cells. In this review, we will discuss the past progress of whole leukemia cell vaccines, with a focus on strategies to enhance their immunogenicity. We will also present the future directions of whole leukemia cell vaccines along with addressing newly emerging concepts, such as immunogenic cell death and necroptosis. We will not discuss in detail other factors that can reduce the therapeutic efficacy of whole leukemia cell vaccines such as various immunosuppressive mechanisms of leukemia.
Collapse
Affiliation(s)
- Young Sok Ji
- Department of Pathology, School of Medicine, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31151, Republic of Korea.
| | - Seong Kyu Park
- Division of Hemato-Oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14584, Republic of Korea.
| | - Seongho Ryu
- Department of Pathology, School of Medicine, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31151, Republic of Korea; Soonchunhyang Institute of Medi-bio Sciences (SIMS), Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31151, Republic of Korea.
| |
Collapse
|
12
|
O'Brien LJ, Guillerey C, Radford KJ. Can Dendritic Cell Vaccination Prevent Leukemia Relapse? Cancers (Basel) 2019; 11:cancers11060875. [PMID: 31234526 PMCID: PMC6627518 DOI: 10.3390/cancers11060875] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/14/2019] [Accepted: 06/20/2019] [Indexed: 01/02/2023] Open
Abstract
Leukemias are clonal proliferative disorders arising from immature leukocytes in the bone marrow. While the advent of targeted therapies has improved survival in certain subtypes, relapse after initial therapy is a major problem. Dendritic cell (DC) vaccination has the potential to induce tumor-specific T cells providing long-lasting, anti-tumor immunity. This approach has demonstrated safety but limited clinical success until recently, as DC vaccination faces several barriers in both solid and hematological malignancies. Importantly, vaccine-mediated stimulation of protective immune responses is hindered by the aberrant production of immunosuppressive factors by cancer cells which impede both DC and T cell function. Leukemias present the additional challenge of severely disrupted hematopoiesis owing to both cytogenic defects in hematopoietic progenitors and an abnormal hematopoietic stem cell niche in the bone marrow; these factors accentuate systemic immunosuppression and DC malfunction. Despite these obstacles, several recent clinical trials have caused great excitement by extending survival in Acute Myeloid Leukemia (AML) patients through DC vaccination. Here, we review the phenotype and functional capacity of DCs in leukemia and approaches to harness DCs in leukemia patients. We describe the recent clinical successes in AML and detail the multiple new strategies that might enhance prognosis in AML and other leukemias.
Collapse
Affiliation(s)
- Liam J O'Brien
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| | - Camille Guillerey
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| | - Kristen J Radford
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
13
|
Van Acker HH, Versteven M, Lichtenegger FS, Roex G, Campillo-Davo D, Lion E, Subklewe M, Van Tendeloo VF, Berneman ZN, Anguille S. Dendritic Cell-Based Immunotherapy of Acute Myeloid Leukemia. J Clin Med 2019; 8:E579. [PMID: 31035598 PMCID: PMC6572115 DOI: 10.3390/jcm8050579] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/16/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is a type of blood cancer characterized by the uncontrolled clonal proliferation of myeloid hematopoietic progenitor cells in the bone marrow. The outcome of AML is poor, with five-year overall survival rates of less than 10% for the predominant group of patients older than 65 years. One of the main reasons for this poor outcome is that the majority of AML patients will relapse, even after they have attained complete remission by chemotherapy. Chemotherapy, supplemented with allogeneic hematopoietic stem cell transplantation in patients at high risk of relapse, is still the cornerstone of current AML treatment. Both therapies are, however, associated with significant morbidity and mortality. These observations illustrate the need for more effective and less toxic treatment options, especially in elderly AML and have fostered the development of novel immune-based strategies to treat AML. One of these strategies involves the use of a special type of immune cells, the dendritic cells (DCs). As central orchestrators of the immune system, DCs are key to the induction of anti-leukemia immunity. In this review, we provide an update of the clinical experience that has been obtained so far with this form of immunotherapy in patients with AML.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Maarten Versteven
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Felix S Lichtenegger
- Department of Medicine III, LMU Munich, University Hospital, 80799 Munich, Germany.
| | - Gils Roex
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Marion Subklewe
- Department of Medicine III, LMU Munich, University Hospital, 80799 Munich, Germany.
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
- Division of Hematology and Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Antwerp, Belgium.
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
- Division of Hematology and Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Antwerp, Belgium.
| |
Collapse
|
14
|
Cirillo M, Tan P, Sturm M, Cole C. Cellular Immunotherapy for Hematologic Malignancies: Beyond Bone Marrow Transplantation. Biol Blood Marrow Transplant 2017; 24:433-442. [PMID: 29102721 DOI: 10.1016/j.bbmt.2017.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/25/2017] [Indexed: 02/06/2023]
Abstract
Immunotherapy has changed treatment practices for many hematologic malignancies. Even in the current era of targeted therapy, chemotherapy remains the backbone of treatment for many hematologic malignancies, especially in acute leukemias, where relapse remains the major cause of mortality. Application of novel immunotherapies in hematology attempts to harness the killing power of the immune system against leukemia and lymphoma. Cellular immunotherapy is evolving rapidly for high-risk hematologic disorders. Recent advances include chimeric antigen-receptor T cells, mesenchymal stromal/stem cells, dendritic cell tumor vaccines, cytokine-induced killer cells, and virus-specific T cells. The advantages of nontransplantation cellular immunotherapy include suitability for patients for whom transplantation has failed or is contraindicated, and a potentially less-toxic treatment alternative to transplantation for relapsed/refractory patients. This review examines those emerging cellular immunotherapies that are changing treatment paradigms for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Melita Cirillo
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia.
| | - Peter Tan
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Marian Sturm
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Catherine Cole
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
15
|
Galati D, Zanotta S. Hematologic neoplasms: Dendritic cells vaccines in motion. Clin Immunol 2017; 183:181-190. [PMID: 28870867 DOI: 10.1016/j.clim.2017.08.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 07/28/2017] [Accepted: 08/29/2017] [Indexed: 12/17/2022]
Abstract
Dendritic cells (DCs) are bone-marrow-derived immune cells accounted for a key role in cancer vaccination as potent antigen-presenting cells within the immune system. Cancer microenvironment can modulate DCs maturation resulting in their accumulation into functional states associated with a reduced antitumor immune response. In this regard, a successful cancer vaccine needs to mount a potent antitumor immune response able to overcome the immunosuppressive tumor milieu. As a consequence, DCs-based approaches are a safe and promising strategy for improving the therapeutic efficacy in hematological malignancies, particularly in combinations with additional treatments. This review summarizes the most significant evidence about the immunotherapeutic strategies performed to target hematologic neoplasms including the tumoral associated antigens (TAA) pulsed on DCs, whole tumor cell vaccines or leukemia-derived DCs.
Collapse
Affiliation(s)
- Domenico Galati
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Hematology, National Cancer Institute, Fondazione 'G. Pascale', IRCCS, Via Mariano Semmola 49, 80131 Naples, Italy.
| | - Serena Zanotta
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Hematology, National Cancer Institute, Fondazione 'G. Pascale', IRCCS, Via Mariano Semmola 49, 80131 Naples, Italy
| |
Collapse
|
16
|
Hoffmann JM, Schmitt M, Ni M, Schmitt A. Next-generation dendritic cell-based vaccines for leukemia patients. Immunotherapy 2017; 9:173-181. [PMID: 28128712 DOI: 10.2217/imt-2016-0116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Up to today treatment of leukemia patients remains challenging and different therapies have been developed, among them the generation of dendritic cell (DC) vaccines. DCs, highly specific for immunogenic cancer antigens, are generated either ex vivo or in vivo and boost the immune response against leukemic cells. Nevertheless, response rates are still heterogeneous and DC vaccines need improvement. New methods for generating DC vaccines have been summed up under the term 'next-generation DC vaccines'. They range from the analysis of human leukocyte antigen-ligandomes to immunogenic cell death inducers, from the production of viral vectors to mRNA transfection and finally from delivering peptides to DCs in vivo through either antibodies or cell-penetrating peptides. This review gives an overview of the latest developments in this still evolving field.
Collapse
Affiliation(s)
- Jean-Marc Hoffmann
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, University Hospital of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Michael Schmitt
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, University Hospital of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Ming Ni
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, University Hospital of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Anita Schmitt
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, University Hospital of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
17
|
Tao Z, Li S, Ichim TE, Yang J, Riordan N, Yenugonda V, Babic I, Kesari S. Cellular immunotherapy of cancer: an overview and future directions. Immunotherapy 2017; 9:589-606. [DOI: 10.2217/imt-2016-0086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The clinical success of checkpoint inhibitors has led to a renaissance of interest in cancer immunotherapies. In particular, the possibility of ex vivo expanding autologous lymphocytes that specifically recognize tumor cells has attracted much research and clinical trial interest. In this review, we discuss the historical background of tumor immunotherapy using cell-based approaches, and provide some rationale for overcoming current barriers to success of autologous immunotherapy. An overview of adoptive transfer of lymphocytes, tumor infiltrating lymphocytes and dendritic cell therapies is provided. We conclude with discussing the possibility of gene-manipulating immune cells in order to augment therapeutic activity, including silencing of the immune-suppressive zinc finger orphan nuclear receptor, NR2F6, as an attractive means of overcoming tumor-associated immune suppression.
Collapse
Affiliation(s)
- Ziqi Tao
- The Affiliated XuZhou Center Hospital of Nanjing University of Chinese Medicine, The Affiliated XuZhou Hospital of Medical College of Southeast University, Jiangsu, China
| | - Shuang Li
- Department of Endocrinology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | | | - Junbao Yang
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Neil Riordan
- Medistem Panama, Inc., City of Knowledge, Clayton, Republic of Panama
| | - Venkata Yenugonda
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Ivan Babic
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Santosh Kesari
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
- John Wayne Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA 90404, USA
| |
Collapse
|
18
|
Fischbacher D, Merle M, Liepert A, Grabrucker C, Kroell T, Kremser A, Dreyßig J, Freudenreich M, Schuster F, Borkhardt A, Kraemer D, Koehne CH, Kolb HJ, Schmid C, Schmetzer HM. Cytokine Release Patterns in Mixed Lymphocyte Culture (MLC) of T-Cells with Dendritic Cells (DC) Generated from AML Blasts Contribute to Predict anti-Leukaemic T-Cell Reactions and Patients’ Response to Immunotherapy. ACTA ACUST UNITED AC 2016; 22:49-65. [DOI: 10.1080/15419061.2016.1223634] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Dorothea Fischbacher
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
| | - Marion Merle
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
| | - Anja Liepert
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
| | - Christine Grabrucker
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
| | - Tanja Kroell
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
| | - Andreas Kremser
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
| | - Julia Dreyßig
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
| | - Markus Freudenreich
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
| | - Friedhelm Schuster
- Department for pediatric Haematology and Oncology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department for pediatric Haematology and Oncology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | | | | | - Hans-Jochem Kolb
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
- Helmholtz Center, Munich, Clinical cooperative group Human Cell Transplantation (CCG-HCT), Munich, Germany
| | | | - Helga Maria Schmetzer
- Department for Haematopoietic Transplantations, University Hospital of Munich, Munich, Germany
- Helmholtz Center, Munich, Clinical cooperative group Human Cell Transplantation (CCG-HCT), Munich, Germany
| |
Collapse
|
19
|
Ni M, Hoffmann JM, Schmitt M, Schmitt A. Progress of dendritic cell-based cancer vaccines for patients with hematological malignancies. Expert Opin Biol Ther 2016; 16:1113-23. [PMID: 27238400 DOI: 10.1080/14712598.2016.1196181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Dendritic cells (DCs) are the most professional antigen-presenting cells eliciting cellular and humoral immune responses against cancer cells by expressing these antigens on MHC class I/II complexes to T cells. Therefore, they have been employed in many clinical trials as cancer vaccines for patients with cancer. This review focuses on the use of DCs in leukemia patients expressing leukemia-associated antigens (LAAs). AREAS COVERED The contribution of both stimulating vs. tolerogenic DCs as well as of other factors to the milieu of anti-leukemia immune responses are discussed. Several DC vaccination strategies like leukemia lysate, proteins and peptides have been developed. Next generation DC vaccines comprise transduction of DCs with retroviral vectors encoding for LAAs, cytokines and costimulatory molecules as well as transfection of DCs with naked RNA encoding for LAAs. Published as well as ongoing clinical trials are reported and critically reviewed. EXPERT OPINION Future results will demonstrate whether next-generation DCs are really superior to conventional pulsing with peptide, protein or tumor lysate. However, currently available methods based on nucleic acid transfection/transduction are tempting in terms of material production costs and time for clinical application according to good manufacturing practice (GMP).
Collapse
Affiliation(s)
- Ming Ni
- a Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V , Heidelberg University Hospital , Heidelberg , Germany
| | - Jean-Marc Hoffmann
- a Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V , Heidelberg University Hospital , Heidelberg , Germany
| | - Michael Schmitt
- a Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V , Heidelberg University Hospital , Heidelberg , Germany
| | - Anita Schmitt
- a Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V , Heidelberg University Hospital , Heidelberg , Germany
| |
Collapse
|
20
|
Buckley SA, Walter RB. Update on antigen-specific immunotherapy of acute myeloid leukemia. Curr Hematol Malig Rep 2016; 10:65-75. [PMID: 25896530 DOI: 10.1007/s11899-015-0250-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Among the few drugs that have shown a benefit for patients with acute myeloid leukemia (AML) in randomized clinical trials over the last several decades is the CD33 antibody-drug conjugate, gemtuzumab ozogamicin (GO). Undoubtedly, this experience has highlighted the value of antigen-specific immunotherapy in AML. A wide variety of therapeutics directed against several different antigens on AML cells are currently explored in preclinical and early clinical studies. On the one hand, these include passive strategies such as unconjugated antibodies targeting one or more antigens, antibodies armed with drugs, toxic proteins, or radionuclides, or adoptive immunotherapies, in particular utilizing T cells engineered to express chimeric antigen receptors (CARs) or modified T cell receptor (TCR) genes; on the other hand, these include active strategies such as vaccinations. With the documented benefit for GO and the emerging data with several classes of therapeutics in other leukemias, in particular small bispecific antibodies and CAR T cells, the future is bright. Nevertheless, a number of important questions related to the choice of target antigen(s), patient population, exact treatment modality, and supportive care needs remain open. Addressing such questions in upcoming studies will ultimately be required to optimize the clinical use of antigen-specific immunotherapies in AML and ensure that such treatments become an effective, versatile tool for this disease for which the outcomes have remained unsatisfactory in many patients.
Collapse
Affiliation(s)
- Sarah A Buckley
- Hematology/Oncology Fellowship Program, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
21
|
Ru YX, Dong SX, Zhao SX, Liang HY, Wang HJ, Hu X, Mi YC, Wang JX. Histiocytic differentiation in acute monocytic leukemia. Ultrastruct Pathol 2016; 40:18-23. [PMID: 26771450 DOI: 10.3109/01913123.2015.1120838] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Myeloid histocytes of dendritic cells (DCs), Langerhans cells (LCs), and macrophages in varied tissues, as leukemic blasts in acute monoblastic and monocytic leukemia (AML-M5a and M5b), are derived from monocyte progenitors in bone marrow. Based on DC induction from hematopoietic stem cells, myeloid progenitors, and monocytes, and occasional expressions of histocyte-related antigens (HRAs) in M5, we presume some M5 cases share histiocytic phenotypes originally. To clarify the conception, 93 M5 cases were tested with antibodies for HRAs, CD1a, CD163, S100, fascin, and langerin by immunostaining, and their morphologic characteristics were studied by light and transmission electron microscopy. The study revealed that 23 M5 cases were positive for two or more kinds of HRAs and shared a serial of histocytic immunophenotype and morphologic features, which were closely associated with M5b subtype and expression of CD14 in M5.
Collapse
Affiliation(s)
- Yong-xin Ru
- a Institute of Hematology & Blood Diseases Hospital , State Key Laboratory of Experimental Hematology, Peking Union Medical College , Tianjin , China
| | - Shu-xu Dong
- a Institute of Hematology & Blood Diseases Hospital , State Key Laboratory of Experimental Hematology, Peking Union Medical College , Tianjin , China
| | - Shi-xuan Zhao
- a Institute of Hematology & Blood Diseases Hospital , State Key Laboratory of Experimental Hematology, Peking Union Medical College , Tianjin , China
| | - Hao-yue Liang
- a Institute of Hematology & Blood Diseases Hospital , State Key Laboratory of Experimental Hematology, Peking Union Medical College , Tianjin , China
| | - Hui-jun Wang
- a Institute of Hematology & Blood Diseases Hospital , State Key Laboratory of Experimental Hematology, Peking Union Medical College , Tianjin , China
| | - Xiao Hu
- a Institute of Hematology & Blood Diseases Hospital , State Key Laboratory of Experimental Hematology, Peking Union Medical College , Tianjin , China
| | - Ying-chang Mi
- a Institute of Hematology & Blood Diseases Hospital , State Key Laboratory of Experimental Hematology, Peking Union Medical College , Tianjin , China
| | - Jian-xiang Wang
- a Institute of Hematology & Blood Diseases Hospital , State Key Laboratory of Experimental Hematology, Peking Union Medical College , Tianjin , China
| |
Collapse
|
22
|
Le Pogam C, Patel S, Gorombei P, Guerenne L, Krief P, Omidvar N, Tekin N, Bernasconi E, Sicre F, Schlageter MH, Chopin M, Noguera ME, West R, Abu A, Mathews V, Pla M, Fenaux P, Chomienne C, Padua RA. DNA-mediated adjuvant immunotherapy extends survival in two different mouse models of myeloid malignancies. Oncotarget 2015; 6:32494-508. [PMID: 26378812 PMCID: PMC4741708 DOI: 10.18632/oncotarget.5572] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/14/2015] [Indexed: 02/06/2023] Open
Abstract
We have previously shown that a specific promyelocytic leukemia-retinoic acid receptor alpha (PML-RARA) DNA vaccine combined with all-trans retinoic acid (ATRA) increases the number of long term survivors with enhanced immune responses in a mouse model of acute promyelocytic leukemia (APL). This study reports the efficacy of a non-specific DNA vaccine, pVAX14Flipper (pVAX14), in both APL and high risk myelodysplastic syndrome (HR-MDS) models. PVAX14 is comprised of novel immunogenic DNA sequences inserted into the pVAX1 therapeutic plasmid. APL mice treated with pVAX14 combined with ATRA had increased survival comparable to that obtained with a specific PML-RARA vaccine. Moreover, the survival advantage correlated with decreased PML-RARA transcript levels and increase in anti-RARA antibody production. In HR-MDS mice, pVAX14 significantly improved survival and reduced biomarkers of leukemic transformation such as phosphorylated mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) 1. In both preclinical models, pVAX14 vaccine significantly increased interferon gamma (IFNγ) production, memory T-cells (memT), reduced the number of colony forming units (CFU) and increased expression of the adapter molecule signalling to NF-κB, MyD88. These results demonstrate the adjuvant properties of pVAX14 providing thus new approaches to improve clinical outcome in two different models of myeloid malignancies, which may have potential for a broader applicability in other cancers.
Collapse
Affiliation(s)
- Carole Le Pogam
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Satyananda Patel
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Petra Gorombei
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Laura Guerenne
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Patricia Krief
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Nader Omidvar
- Haemotology Department, Cardiff University School of Medicine, Cardiff, UK
| | - Nilgun Tekin
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Elena Bernasconi
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Flore Sicre
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Marie-Helene Schlageter
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Martine Chopin
- Département d'Expérimentation Animale, Institut Universitaire d'Hématologie, University Paris Diderot, Paris, France
| | - Maria-Elena Noguera
- Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Robert West
- Welsh Heart Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - Ansu Abu
- Department of Hematology, Christian Medical College and Hospital, Vellore, India
| | - Vikram Mathews
- Department of Hematology, Christian Medical College and Hospital, Vellore, India
| | - Marika Pla
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Pierre Fenaux
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Christine Chomienne
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Rose Ann Padua
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| |
Collapse
|
23
|
Grosso DA, Hess RC, Weiss MA. Immunotherapy in acute myeloid leukemia. Cancer 2015; 121:2689-704. [PMID: 26095886 DOI: 10.1002/cncr.29378] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 01/23/2015] [Accepted: 02/09/2015] [Indexed: 11/08/2022]
Abstract
Despite the remarkable progress made in some leukemias such as CML and CLL, cytotoxic treatment for AML remains essentially unchanged over the last 4 decades. Several lines of evidence, including the graft versus leukemia effect associated with allogeneic hematopoietic stem cell transplantation (HSCT), suggest that immunotherapy is an active modality in AML. Given the lack of progress for chemotherapy in this disease, many novel immunologic treatment approaches have been explored. The goals of non-transplant-based immune approaches have largely consisted of the stimulation or restoration of endogenous immune responses or the targeting of specific tumor antigens by immune cells. These strategies have been associated with less toxicity than allogeneic HSCT but typically have inferior efficacy. Allogeneic HSCT exploits major and minor histocompatibility differences between the donor and recipient in order to recognize and eradicate malignancy. With the recognition that the immune system itself provides a basis for treating AML, immunotherapy continues to be an attractive modality to exploit in the treatment of this disease.
Collapse
Affiliation(s)
- Dolores A Grosso
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Rosemary C Hess
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Mark A Weiss
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Buckley SA, Walter RB. Antigen-specific immunotherapies for acute myeloid leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2015; 2015:584-595. [PMID: 26637776 DOI: 10.1182/asheducation-2015.1.584] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Antigen-specific immunotherapies have emerged as important components of curative treatment algorithms for many cancers. In acute myeloid leukemia (AML), success has been less obvious. Nonetheless, among the few drugs shown to improve survival in recent randomized trials is the CD33 antibody-drug conjugate gemtuzumab ozogamicin. Significant antileukemic activity is also well documented for radioimmunoconjugates targeting CD33, CD45, or CD66. These therapeutics can intensify conditioning before hematopoietic cell transplantation, but their effect on patient outcomes needs clarification. Emerging data now suggest clinical antileukemic activity of several novel antibodies and perhaps some adoptive T-cell immunotherapies and vaccines. In parallel, numerous other agents targeting a wider variety of antigens are currently being explored. However, the antigenic heterogeneity characteristic of AML is a considerable limitation for all these therapeutics, and many important questions related to the ideal target antigen(s), disease situation in which to use these therapies, most suitable patient populations, exact treatment modalities, and details of supportive care needs remain open. Addressing such questions in upcoming studies will be required to ensure that antigen-directed therapies become an effective tool in AML, a disease for which outcomes with standard "3 + 7"-based chemotherapy have remained unsatisfactory in many patients.
Collapse
Affiliation(s)
| | - Roland B Walter
- Department of Medicine, Division of Hematology, and Department of Epidemiology, University of Washington, Seattle, WA; and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
25
|
van den Ancker W, Wijnands PGJB, Ruben JM, Westers TM, Punt B, Bachas C, Ravenshorst N, van Wetering S, Kruisbeek AM, Bontkes HJ, Ossenkoppele GJ, van de Loosdrecht AA, de Gruijl TD. Procedures for the expansion of CD14(+) precursors from acute myeloid leukemic cells to facilitate dendritic cell-based immunotherapy. Immunotherapy 2014; 5:1183-90. [PMID: 24188673 DOI: 10.2217/imt.13.125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS Vaccination with acute myeloid leukemia (AML)-derived dendritic cells (DCs) is a promising immunotherapeutic approach to prevent relapse of AML. However, in clinical practice AML-derived DC culture is unfeasible in 40% of cases. Here, we demonstrate that AML cells can be expanded in vitro prior to differentiation with cocktails of cytokines with known myeloid growth-promoting effects. RESULTS Nine out of 13 initially CD14(-) samples gain de novo CD14 (>10%) expression (69% increment; p = 0.01) after in vitro expansion. These expanded CD14(+) leukemic cells displayed a high probability (six out of six initially CD14(-) samples tested) to differentiate into DCs upon culture with GM-CSF, TNF-α and IL-4. CONCLUSION Induction of CD14 on initially CD14(-) AML cells potentially increases the number of patients eligible for DC-based immunotherapy.
Collapse
Affiliation(s)
- Willemijn van den Ancker
- Department of Hematology, VU University Medical Center, VU Institute for Cancer & Immunology, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Rosenblatt J, Avigan D. Can leukemia-derived dendritic cells generate antileukemia immunity? Expert Rev Vaccines 2014; 5:467-72. [PMID: 16989627 DOI: 10.1586/14760584.5.4.467] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Tumor vaccines are being explored as a means of generating antitumor immune responses in patients with cancer. Based on the efficacy of allogeneic transplantation, acute myelogenous leukemia appears to be susceptible to cellular immune-based therapy. Dendritic cells (DCs) are the most potent antigen-presenting cells and, as such, are being studied as a platform for the design of cancer vaccines. In acute leukemia, a promising approach involves the generation of DCs from leukemic blasts via cytokine exposure ex vivo. Leukemia-derived DCs potentially retain the tumor-associated antigens of the leukemic clone, which are presented in the context of the immune stimulating machinery of the mature DC. However, the efficacy of this approach may be limited by intrinsic abnormalities in the malignant clone that prevent differentiation towards a normal DC phenotype.
Collapse
Affiliation(s)
- Jacalyn Rosenblatt
- Beth Israel Deaconess Medical Center, Hematologic Malignancy Bone Marrow Transplant Program, 330 Brookline Avenue, KS 121, Boston, MA 02215, USA.
| | | |
Collapse
|
27
|
Pyzer AR, Avigan DE, Rosenblatt J. Clinical trials of dendritic cell-based cancer vaccines in hematologic malignancies. Hum Vaccin Immunother 2014; 10:3125-31. [PMID: 25625926 PMCID: PMC4514037 DOI: 10.4161/21645515.2014.982993] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/17/2014] [Accepted: 10/05/2014] [Indexed: 11/19/2022] Open
Abstract
The potential for the immune system to target hematological malignancies is demonstrated in the allogeneic transplant setting, where durable responses can be achieved. However, allogeneic transplantation is associated with significant morbidity and mortality related to graft versus host disease. Cancer immunotherapy has the capacity to direct a specific cytotoxic immune response against cancer cells, particularly residual cancer cells, in order to reduce the likelihood of disease relapse in a more targeted and tolerated manner. Ex vivo dendritic cells can be primed in various ways to present tumor associated antigen to the immune system, in the context of co-stimulatory molecules, eliciting a tumor specific cytotoxic response in patients. Several approaches to prime dendritic cells and overcome the immunosuppressive microenvironment have been evaluated in pre-clinical and early clinical trials with promising results. In this review, we summarize the clinical data evaluating dendritic cell based vaccines for the treatment of hematological malignancies.
Collapse
Key Words
- AML, Acute Myeloid Leukemia
- ASCT, Autologous Stem Cell Transplant
- Apo-DC, Apoptotic body loaded- dendritic cells
- CML, Chronic Myeloid Leukemia
- CR, Complete response
- CTLA-4, Cytotoxic T-Lymphocyte Antigen 4
- DC/AML, Dendritic cell Acute Myeloid Leukemia fusion vaccine
- DC/MM, Dendritic cell Multiple Myeloma fusion vaccine
- DNA Deoxyribonucleic acid
- FLT-ITD, Fms-like Tyrosine Kinase with Internal Tandem Duplication
- GMCSF, Granulocyte macrophage colony-stimulating factor
- GVHD, Graft vs Host Disease
- HLA-A*2402, Human Leukocyte antigen A*2402
- IFN, Interferon
- IFNg, Interferon gamma
- IL, Interleukin
- Id, Idiotype
- KLH, Keyhole limpet hemocyanin
- MDS, Myelodysplastic syndrome
- MHC, Major histocompatibility complex
- OS, Overall Survival
- PD-1, Programmed death 1
- PD-L1, Programmed death-ligand 1
- PR, Partial response
- PRR, Pathogen recognition receptor
- RNA, Ribonucleic acid
- SCT, Stem cell transplant
- TGFB, Transforming growth factor β
- TNFα, Tumor necrosis factor α
- VEGF, Vascular endothelial growth factor
- VGPR, Very good partial response
- WT-1, Wilm's tumor suppressor gene 1
- cancer
- dendritic cell
- immunotherapy
- leukemia
- mRNA, mRNA
- myeloma
- pDCs, Plasmacytoid Dendritic cell
- trial
- vaccine
Collapse
Affiliation(s)
- Athalia R Pyzer
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| | - David E Avigan
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| | - Jacalyn Rosenblatt
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| |
Collapse
|
28
|
Kitawaki T. DC-based immunotherapy for hematological malignancies. Int J Hematol 2013; 99:117-22. [DOI: 10.1007/s12185-013-1496-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 12/15/2013] [Accepted: 12/16/2013] [Indexed: 12/29/2022]
|
29
|
Avigan D, Hari P, Battiwalla M, Bishop MR, Giralt SA, Hardy NM, Kröger N, Wayne AS, Hsu KC. Proceedings from the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: part II. Autologous Transplantation-novel agents and immunomodulatory strategies. Biol Blood Marrow Transplant 2013; 19:1661-9. [PMID: 24018393 PMCID: PMC3914636 DOI: 10.1016/j.bbmt.2013.08.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 08/30/2013] [Indexed: 12/11/2022]
Abstract
In the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation, the Scientific/Educational Session on Autologous Transplantation addressed the role of novel agents and immunomodulatory strategies in management of relapse after autologous hematopoietic stem cell transplantation (AHSCT). Concepts were illustrated through in-depth discussion of multiple myeloma, with broader discussion of areas relevant for relapse of other malignancies as well as in the setting of allogeneic transplantation. Dr. Hari provided an overview of the epidemiology of relapse after AHSCT in multiple myeloma, addressing clinical patterns, management implications, and treatment options at relapse, highlighting the implications of novel therapeutic agents in initial, maintenance, and relapse treatment. Dr. Avigan discussed current concepts in tumor vaccine design, including whole cell and antigen-specific strategies, use of an AHSCT platform to reverse tumor-associated immunosuppression and tolerance, and combining vaccines with immunomodulatory agents to promote establishment of durable antitumor immunity. Dr. Hsu reviewed the immunogenetics of natural killer (NK) cells and general NK biology, the clinical importance of autologous NK activity (eg, lymphoma and neuroblastoma), the impact of existing therapies on promotion of NK cell activity (eg, immunomodulatory drugs, monoclonal antibodies), and strategies for enhancing autologous and allogeneic NK cell effects through NK cell gene profiling.
Collapse
Affiliation(s)
- David Avigan
- Division of Hematology Oncology, Hematologic Malignancies/Bone Marrow Transplant Program, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lichtenegger FS, Schnorfeil FM, Hiddemann W, Subklewe M. Current strategies in immunotherapy for acute myeloid leukemia. Immunotherapy 2013; 5:63-78. [PMID: 23256799 DOI: 10.2217/imt.12.145] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The prognosis of acute myeloid leukemia, particularly when associated with adverse chromosomal or molecular aberrations, is poor due to a high relapse rate after induction chemotherapy. Postremission therapy for elimination of minimal residual disease remains a major challenge. Allogeneic hematopoietic stem cell transplantation has proven to provide a potent antileukemic effect. Novel strategies are needed for patients ineligible for this treatment. Here current immunotherapeutic concepts in acute myeloid leukemia in a nonallogeneic hematopoietic stem cell transplantation setting are reviewed. Data gathered with different monoclonal antibodies are discussed. Adoptive transfer of NK and T cells is reviewed, including evolving data on T-cell engineering. Results of systemic cytokine administration and of therapeutic vaccinations with peptides, modified leukemic cells and dendritic cells are presented. One particular focus of this review is the integration of currently running clinical trials. Recent immunotherapeutic studies have been encouraging and further interesting results are to be expected.
Collapse
Affiliation(s)
- Felix S Lichtenegger
- Department of Internal Medicine III, Klinikum der Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | | | | | | |
Collapse
|
31
|
Kerkhoff N, Bontkes HJ, Westers TM, de Gruijl TD, Kordasti S, van de Loosdrecht AA. Dendritic cells in myelodysplastic syndromes: from pathogenesis to immunotherapy. Immunotherapy 2013; 5:621-37. [DOI: 10.2217/imt.13.51] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are clonal disorders of the hematopoietic stem cell characterized by ineffective hematopoiesis leading to peripheral cytopenias. Different processes are involved in its pathogenesis, such as (epi)genetic alterations and immunological dysfunctions. The nature of immune dysregulation is markedly different between various MDS risk groups. In low-risk MDS, the immune system is in a proinflammatory state, whereas in high-risk disease, immunosuppressive features facilitate expansion of the dysplastic clone and can eventually lead to disease progression to acute myeloid leukemia. Various cell types contribute to dysregulation of immune responses in MDS. Dendritic cells (DCs) are important regulators of immunity. However, the role of DCs in MDS has yet to be elucidated. It has been suggested that impaired DC function can hamper adequate immune responses. This review focuses on the involvement of DCs in immune dysregulation in low- and high-risk MDS and the implications for DC-targeted therapies.
Collapse
Affiliation(s)
- Nathalie Kerkhoff
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Hetty J Bontkes
- Department of Pathology, Unit Medical Immunology, VU University Medical Center, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Theresia M Westers
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Shahram Kordasti
- Department of Haematological Medicine, King’s College Hospital London, Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU, UK
| | - Arjan A van de Loosdrecht
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
32
|
|
33
|
Upregulation of CD200 is associated with Foxp3+ regulatory T cell expansion and disease progression in acute myeloid leukemia. Tumour Biol 2012. [DOI: 10.1007/s13277-012-0578-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
34
|
Abstract
The long-term outlook for adult patients with acute myeloid leukemia (AML) remains dismal. The main reason for this state of affairs lies in the fact that the majority of AML patients will eventually relapse, even after obtaining complete remission following front-line chemotherapy. Relapses are generally attributed to the persistence of a small number of chemotherapy-resistant leukemic (stem) cells, a condition known as minimal residual disease (MRD). The eradication of MRD, with the eventual aim of reducing the risk of relapse, therefore represents a high-priority goal of modern AML therapy. It is now well established that the immune system plays a crucial role in the defense against AML. This knowledge has fuelled the development of immune-based approaches to control MRD and, ultimately, to prevent relapse. One of the promising strategies that have emerged in this regard involves the use of dendritic cells for therapeutic vaccination. This review article aims to introduce the reader into the conceptual and practical aspects of DC-based vaccination for AML. Next, we will review the first clinical results obtained with this immunotherapeutic approach in AML patients. Finally, we will briefly reflect on the potential place of DC vaccination in the future therapy of AML.
Collapse
|
35
|
Anguille S, Willemen Y, Lion E, Smits EL, Berneman ZN. Dendritic cell vaccination in acute myeloid leukemia. Cytotherapy 2012; 14:647-56. [DOI: 10.3109/14653249.2012.693744] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Yuan C, Song G, Jiang G. The characterization and role of leukemia cell-derived dendritic cells in immunotherapy for leukemic diseases. Intractable Rare Dis Res 2012; 1:53-65. [PMID: 25343074 PMCID: PMC4204560 DOI: 10.5582/irdr.2012.v1.2.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 03/28/2012] [Accepted: 04/13/2012] [Indexed: 11/05/2022] Open
Abstract
Usually, an effective anti-leukemia immune response cannot be initiated effectively in patients with leukemia. This is probably related to immunosuppression due to chemotherapy, down-regulation of major histocompatibility complex (MHC) II molecules, and the lack of co-stimulatory molecules on dendritic cells (DC). In light of this problem, some methods had been used to induce leukemia cells to differentiate into mature DCs, causing them to present leukemia-associated antigens and activating naïve T cells. Furthermore, leukemia-derived DCs could be modified with tumor antigens or tumor-associated antigens to provide a new approach to anti-leukemia therapy. Numerous studies have indicated factors related to the induction and functioning of leukemia-derived DCs and the activation of cytotoxic T-lymphocytes (CTLs). These include the amount of purified DCs, cytokine profiles appropriate for inducing leukemia-derived DCs, effective methods of activating CTLs, reasonable approaches to DC vaccines, and the standardization of their clinical use. Determining these factors could lead to more effective leukemia treatment and benefit both mankind and scientific development. What follows in a review of advances in and practices of inducing leukemia-derived DCs and the feasibility of their clinical use.
Collapse
Affiliation(s)
- Changjin Yuan
- Key Laboratory for Tumor Immunology & Traditional Chinese Medicine Immunology, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Department of Hemato-Oncology, Shandong Academy of Medical Sciences, Key Laboratory for Biotech-Drugs of the Ministry of Health, Key Laboratory for Modern Medicine and Technology of Shandong Province, Ji'nan, Shandong, China
| | - Guanhua Song
- Key Laboratory for Tumor Immunology & Traditional Chinese Medicine Immunology, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Department of Hemato-Oncology, Shandong Academy of Medical Sciences, Key Laboratory for Biotech-Drugs of the Ministry of Health, Key Laboratory for Modern Medicine and Technology of Shandong Province, Ji'nan, Shandong, China
| | - Guosheng Jiang
- Key Laboratory for Tumor Immunology & Traditional Chinese Medicine Immunology, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Department of Hemato-Oncology, Shandong Academy of Medical Sciences, Key Laboratory for Biotech-Drugs of the Ministry of Health, Key Laboratory for Modern Medicine and Technology of Shandong Province, Ji'nan, Shandong, China
- Address correspondence to: Prof. Guosheng Jiang, Department of Hemato-oncology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jingshi Road 18877, Ji'nan 250062, Shandong, China. E-mail:
| |
Collapse
|
37
|
Recent advance in antigen-specific immunotherapy for acute myeloid leukemia. Clin Dev Immunol 2011; 2011:104926. [PMID: 22028726 PMCID: PMC3199067 DOI: 10.1155/2011/104926] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 08/18/2011] [Indexed: 11/18/2022]
Abstract
Relapse after chemotherapy is inevitable in the majority of patients with acute myeloid leukemia (AML). Thus, it is necessary to develop novel therapies that have different antileukemic mechanisms. Recent advances in immunology and identification of promising leukemia-associated antigens open the possibilities for eradicating minimal residual diseases by antigen-specific immunotherapy after chemotherapy. Several methods have been pursued as immunotherapies for AML: peptide vaccines, granulocyte-macrophage colony-stimulating factor-secreting tumor vaccines, dendritic cell vaccines, and adoptive T cell therapy. Whereas immunogenicity and clinical outcomes are improving in these trials, severe adverse events were observed in highly avid engineered T cell therapies, indicating the importance of the balance between effectiveness and side effects in advanced immunotherapy. Such progress in inducing antitumor immune responses, together with strategies to attenuate immunosuppressive factors, will establish immunotherapy as an important armament to combat AML.
Collapse
|
38
|
Dreyssig J, Kremser A, Liepert A, Grabrucker C, Freudenreich M, Schmid C, Kroell T, Scholl N, Tischer J, Kufner S, Salih H, Kolb HJ, Schmetzer HM. Various ‘dendritic cell antigens’ are already expressed on uncultured blasts in acute myeloid leukemia and myelodysplastic syndromes. Immunotherapy 2011; 3:1113-24. [DOI: 10.2217/imt.11.108] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim and methods: Leukemia-derived dendritic cells (DCleu) potentially present the whole leukemic antigen repertoire. We studied antigen-expression profiles of blasts/dendritic cells (DCs) generated from 137 acute myeloid leukemia (AML)/49 myelodysplastic syndromes (MDS) patients with six different DC-generating media by flow-cytometry combining expression of blast/maturation and DC antigens (DCA:CD1a,b,c, CD25, CD40, CD80, CD83, CD86, CD137-L and CD206). Results: First, DCA are regularly and variably expressed on uncultured blasts/mononuclear cells (MNCs). Individual patients’ DCA profiles must be evaluated before DC-culture to find suitable DCA to estimate quality/quantity of DC after culture. Second, after culture in every patient, at least one marker fulfilled these criteria. Third, different DC-generating methods showed varying efficiency to generate DC: not every method was always successful. Fourth, individual FACS-DCA profiles showed a successful DC/DCleu generation with at least one of three previously tested methods in every given AML/MDS case. Fifth, pooling results of all selected best methods in every given case, 28/30% DC were generated from AML/MDS samples: >60% viable DC, on average 49/56% mature DC and on average 36% of blasts were convertible to DCleu resulting in on average 49% DCleu of AML-DC. Conclusions: Individual DCA-expression profiles should be evaluated before culture to evaluate DC counts/subtypes (mature/viableDC, DCleu) in individual patients.
Collapse
Affiliation(s)
- Julia Dreyssig
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Andreas Kremser
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Anja Liepert
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Christine Grabrucker
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Markus Freudenreich
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | | | - Tanja Kroell
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Nina Scholl
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Johanna Tischer
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Stephany Kufner
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Helmut Salih
- Department of Internal Medicine II, University Hospital of the Eberhard Karls University, Tuebingen, Germany
| | - Hans-Jochem Kolb
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
- Helmholtz Center Munich, German Research Center for Environmental Health/Clinical Cooperative Group Haematopoetic Cell Transplantation (CCG-HCT), Marchioninistr. 15, 81377 Munich, Germany
| | | |
Collapse
|
39
|
Abstract
The microenviroment of acute myelogenous leukemia (AML) is suppressive for immune effector cells. Regulatory T cells (Tregs) have been recognized as a contributor factor and may be recruited and exploited by leukemic cells to evade immunesurveillance. Studies have shown that the frequencies of marrow and blood Tregs are greater in patients with AML than in control patients. Although increased Tregs have been associated with a decreased risk of GVHD after allogeneic HCT and hence may impede the graft-versus-tumor effect, recent findings indicate that that this may not be the case. Because there is a need to improve outcomes of standard treatment (chemotherapy with or without allogeneic HCT) in AML, targeting Tregs present an outstanding opportunity in AML because discoveries may apply throughout its treatment. Here, we review data on the roles of Tregs in mediating immune system-AML interactions. We focused on in vitro, animal, and observational human studies of Tregs in AML biology, development, prognosis, and therapy in different settings (eg, vaccination and HCT). Manipulation of Tregs or other types of immunomodulation may become a part of AML treatment in the future.
Collapse
|
40
|
Smits ELJ, Lee C, Hardwick N, Brooks S, Van Tendeloo VFI, Orchard K, Guinn BA. Clinical evaluation of cellular immunotherapy in acute myeloid leukaemia. Cancer Immunol Immunother 2011; 60:757-69. [PMID: 21519825 PMCID: PMC11029703 DOI: 10.1007/s00262-011-1022-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 04/08/2011] [Indexed: 02/07/2023]
Abstract
Immunotherapy is currently under active investigation as an adjuvant therapy to improve the overall survival of patients with acute myeloid leukaemia (AML) by eliminating residual leukaemic cells following standard therapy. The graft-versus-leukaemia effect observed following allogeneic haematopoietic stem cell transplantation has already demonstrated the significant role of immune cells in controlling AML, paving the way to further exploitation of this effect in optimized immunotherapy protocols. In this review, we discuss the current state of cellular immunotherapy as adjuvant therapy for AML, with a particular focus on new strategies and recently published results of preclinical and clinical studies. Therapeutic vaccines that are being tested in AML include whole tumour cells as an autologous source of multiple leukaemia-associated antigens (LAA) and autologous dendritic cells loaded with LAA as effective antigen-presenting cells. Furthermore, adoptive transfer of cytotoxic T cells or natural killer cells is under active investigation. Results from phase I and II trials are promising and support further investigation into the potential of cellular immunotherapeutic strategies to prevent or fight relapse in AML patients.
Collapse
Affiliation(s)
- Evelien L J Smits
- Laboratory of Experimental Haematology, Vaccine and Infectious Disease Institute, Antwerp University Hospital, University of Antwerp, Wilrijkstraat 10, 2650, Antwerp, Belgium.
| | | | | | | | | | | | | |
Collapse
|
41
|
Kitawaki T, Kadowaki N, Fukunaga K, Kasai Y, Maekawa T, Ohmori K, Itoh T, Shimizu A, Kuzushima K, Kondo T, Ishikawa T, Uchiyama T. Cross-priming of CD8+ T cells in vivo by dendritic cells pulsed with autologous apoptotic leukemic cells in immunotherapy for elderly patients with acute myeloid leukemia. Exp Hematol 2011; 39:424-433.e2. [DOI: 10.1016/j.exphem.2011.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 12/28/2010] [Accepted: 01/01/2011] [Indexed: 10/18/2022]
|
42
|
Serrano-López J, Sanchez-Garcia J, Serrano J, Alvarez-Rivas MA, Garcia-Castellano JM, Roman-Gomez J, Rosa ODL, Herrera-Arroyo C, Torres-Gomez A. Nonleukemic myeloid dendritic cells obtained from autologous stem cell products elicit antileukemia responses in patients with acute myeloid leukemia. Transfusion 2011; 51:1546-55. [DOI: 10.1111/j.1537-2995.2010.03042.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
43
|
van den Ancker W, van Luijn MM, Westers TM, Bontkes HJ, Ruben JM, de Gruijl TD, Ossenkoppele GJ, van de Loosdrecht AA. Recent advances in antigen-loaded dendritic cell-based strategies for treatment of minimal residual disease in acute myeloid leukemia. Immunotherapy 2010; 2:69-83. [PMID: 20635890 DOI: 10.2217/imt.09.85] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Therapeutic vaccination with dendritic cells (DCs) is recognized as an important experimental therapy for the treatment of minimal residual disease in acute myeloid leukemia. Many sources of leukemia-associated antigens and different methods for antigen loading of DCs have been used in an attempt to optimize anti-tumor responses. For instance, monocyte-derived DCs have been loaded with apoptotic whole-cell suspensions, necrotic cell lysates, tumor-associated peptides, eluted peptides and cellular DNA or RNA. Furthermore, monocyte-derived DCs can be chemically or electrically fused with leukemic blasts, and DCs have been cultured out of leukemic blasts. However, it remains a challenge in cancer immunotherapy to identify which of these methods is the most optimal for antigen loading and activation of DCs. This review discusses recent advances in DC research and the application of this knowledge towards new strategies for antigen loading of DCs in the treatment of minimal residual disease in acute myeloid leukemia.
Collapse
Affiliation(s)
- Willemijn van den Ancker
- Department of Hematology, VU Institute for Cancer & Immunology, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Van Tendeloo VF, Van de Velde A, Van Driessche A, Cools N, Anguille S, Ladell K, Gostick E, Vermeulen K, Pieters K, Nijs G, Stein B, Smits EL, Schroyens WA, Gadisseur AP, Vrelust I, Jorens PG, Goossens H, de Vries IJ, Price DA, Oji Y, Oka Y, Sugiyama H, Berneman ZN. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms' tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci U S A 2010; 107:13824-9. [PMID: 20631300 PMCID: PMC2922237 DOI: 10.1073/pnas.1008051107] [Citation(s) in RCA: 272] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Active immunization using tumor antigen-loaded dendritic cells holds promise for the adjuvant treatment of cancer to eradicate or control residual disease, but so far, most dendritic cell trials have been performed in end-stage cancer patients with high tumor loads. Here, in a phase I/II trial, we investigated the effect of autologous dendritic cell vaccination in 10 patients with acute myeloid leukemia (AML). The Wilms' tumor 1 protein (WT1), a nearly universal tumor antigen, was chosen as an immunotherapeutic target because of its established role in leukemogenesis and superior immunogenic characteristics. Two patients in partial remission after chemotherapy were brought into complete remission after intradermal administration of full-length WT1 mRNA-electroporated dendritic cells. In these two patients and three other patients who were in complete remission, the AML-associated tumor marker returned to normal after dendritic cell vaccination, compatible with the induction of molecular remission. Clinical responses were correlated with vaccine-associated increases in WT1-specific CD8+ T cell frequencies, as detected by peptide/HLA-A*0201 tetramer staining, and elevated levels of activated natural killer cells postvaccination. Furthermore, vaccinated patients showed increased levels of WT1-specific IFN-gamma-producing CD8+ T cells and features of general immune activation. These data support the further development of vaccination with WT1 mRNA-loaded dendritic cells as a postremission treatment to prevent full relapse in AML patients.
Collapse
Affiliation(s)
- Viggo F Van Tendeloo
- University of Antwerp, Faculty of Medicine, Vaccine and Infectious Disease Institute (Vaxinfectio) and Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp B-2650, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dendritic cells (DCs) can be successfully generated from leukemic blasts in individual patients with AML or MDS: an evaluation of different methods. J Immunother 2010; 33:185-99. [PMID: 20139775 DOI: 10.1097/cji.0b013e3181b8f4ce] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Myeloid-leukemic cells (AML, MDS, CML) can be differentiated to leukemia-derived dendritic cell [DC (DCleu)] potentially presenting the whole leukemic antigen repertoire without knowledge of distinct leukemia antigens and are regarded as promising candidates for a vaccination strategy. We studied the capability of 6 serum-free DC culture methods, chosen according to different mechanisms, to induce DC differentiation in 137 cases of AML and 52 cases of MDS. DC-stimulating substances were cytokines ("standard-medium", "MCM-Mimic", "cytokine-method"), bacterial lysates ("Picibanil"), double-stranded RNA ["Poly (I:C)"] or a cytokine bypass method ("Ca-ionophore"). The quality/quantity of DC generated was estimated by flow cytometry studying (co) expressions of "DC"antigens, costimulatory, maturation, and blast-antigens. Comparing these methods on average 15% to 32% DC, depending on methods used, could be obtained from blast-containing mononuclear cells (MNC) in AML/MDS cases with a DC viability of more than 60%. In all, 39% to 64% of these DC were mature; 31% to 52% of leukemic blasts could be converted to DCleu and DCleu-proportions in the suspension were 2% to 70% (13%). Average results of all culture methods tested were comparable, however not every given case of AML could be differentiated to DC with 1 selected method. However performing a pre-analysis with 3 DC-generating methods (MCM-Mimic, Picibanil, Ca-ionophore) we could generate DC in any given case. Functional analyses provided proof, that DC primed T cells to antileukemia-directed cytotoxic cells, although an anti-leukemic reaction was not achieved in every case. In summary our data show that a successful, quantitative DC/DCleu generation is possible with the best of 3 previously tested methods in any given case. Reasons for different functional behaviors of DC-primed T cells must be evaluated to design a practicable DC-based vaccination strategy.
Collapse
|
46
|
The Quality and Quantity of Leukemia-derived Dendritic Cells From Patients With Acute Myeloid Leukemia and Myelodysplastic Syndrome Are a Predictive Factor for the Lytic Potential of Dendritic Cells-primed Leukemia-Specific T Cells. J Immunother 2010; 33:523-37. [DOI: 10.1097/cji.0b013e3181d87ffd] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
47
|
Abstract
While chemotherapy is successful at inducing remission of acute myeloid leukaemia (AML), the disease has a high probability of relapse. Strategies to prevent relapse involve consolidation chemotherapy, stem cell transplantation and immunotherapy. Evidence for immunosurveillance of AML and susceptibility of leukaemia cells to both T cell and natural killer (NK) cell attack and justifies the application of immune strategies to control residual AML persisting after remission induction. Immune therapy for AML includes allogeneic stem cell transplantation, adoptive transfer of allogeneic or autologous T cells or NK cells, vaccination with leukaemia cells, dendritic cells, cell lysates, peptides and DNA vaccines and treatment with cytokines, antibodies and immunomodulatory agents. Here we describe what is known about the immunological features of AML at presentation and in remission, the current status of immunotherapy and strategies combining treatment approaches with a view to achieving leukaemia cure.
Collapse
Affiliation(s)
- A J Barrett
- Stem Cell Allotransplantation Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1202, USA.
| | | |
Collapse
|
48
|
Massumoto C, Sousa-Canavez JM, Leite KRM, Camara-Lopes LH. Stabilization of acute myeloid leukemia with a dendritic cell vaccine. Hematol Oncol Stem Cell Ther 2010; 1:239-40. [PMID: 20058480 DOI: 10.1016/s1658-3876(08)50011-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
49
|
Bund D, Buhmann R, Gökmen F, Kremser A, Dreyssig J, Kolb HJ, Schmetzer HM. Canine-DCs using different serum-free methods as an approach to provide an animal-model for immunotherapeutic strategies. Cell Immunol 2010; 263:88-98. [DOI: 10.1016/j.cellimm.2010.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 02/24/2010] [Accepted: 03/01/2010] [Indexed: 10/19/2022]
|
50
|
Schmitt M, Casalegno-Garduño R, Xu X, Schmitt A. Peptide vaccines for patients with acute myeloid leukemia. Expert Rev Vaccines 2009; 8:1415-25. [PMID: 19803762 DOI: 10.1586/erv.09.90] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The majority of patients with acute myeloid leukemia (AML) under 60 years of age reach a complete hematological remission after intensive chemotherapy. However, only 20-40% of all patients with AML achieve a disease-free survival of more than 5 years. The graft-versus-leukemia effect observed after allogeneic stem cell transplantation and donor lymphocyte infusions strongly suggests that T lymphocytes play a major role in the rejection of leukemic cells. Vaccination with leukemia-associated antigen (LAA) peptides might constitute a way to augment the graft-versus-leukemia effect. Peptide vaccination causes no major side effects, which is of particular note as most AML patients are people over 60 years of age, often suffering from concomitant disease. This review summarizes approaches to define appropriate LAAs as targets of a T-cell-based vaccine immunotherapy. Current clinical LAA peptide vaccination protocols targeting Wilms' tumor gene, proteinase-3 and the receptor for hyaluronan-mediated motility are reviewed and an outlook to dendritic cells, adjuvants and short oligodenucleotides is given.
Collapse
Affiliation(s)
- Michael Schmitt
- Head of Clinical Stem Cell Transplantation and Cellular Therapy, Department of Internal Medicine III, University of Rostock, Ernst-Heydemann-Str. 6, 18057 Rostock, Germany.
| | | | | | | |
Collapse
|