1
|
Schulze K, Weber U, Schuy C, Durante M, Guzmán CA. Influenza Virus Inactivated by Heavy Ion Beam Irradiation Stimulates Antigen-Specific Immune Responses. Pharmaceutics 2024; 16:465. [PMID: 38675126 PMCID: PMC11054185 DOI: 10.3390/pharmaceutics16040465] [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: 02/21/2024] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 04/28/2024] Open
Abstract
The COVID-19 pandemic has made clear the need for effective and rapid vaccine development methods. Conventional inactivated virus vaccines, together with new technologies like vector and mRNA vaccines, were the first to be rolled out. However, the traditional methods used for virus inactivation can affect surface-exposed antigen, thereby reducing vaccine efficacy. Gamma rays have been used in the past to inactivate viruses. We recently proposed that high-energy heavy ions may be more suitable as an inactivation method because they increase the damage ratio between the viral nucleic acid and surface proteins. Here, we demonstrate that irradiation of the influenza virus using heavy ion beams constitutes a suitable method to develop effective vaccines, since immunization of mice by the intranasal route with the inactivated virus resulted in the stimulation of strong antigen-specific humoral and cellular immune responses.
Collapse
Affiliation(s)
- Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Zentrum für Infektionsforschung (HZI), 38124 Braunschweig, Germany;
| | - Ulrich Weber
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany; (U.W.); (C.S.); (M.D.)
- Fachbereich Mathematik, Naturwissenschaften und Informatik, Technische Hochschule Mittelhessen, 35390 Gießen, Germany
| | - Christoph Schuy
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany; (U.W.); (C.S.); (M.D.)
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany; (U.W.); (C.S.); (M.D.)
- Institute of Condensed Matter Physics, Technische Universität Darmstadt, 64289 Darmstadt, Germany
- Department of Physics “Ettore Pancini”, University Federico II, 80138 Naples, Italy
| | - Carlos Alberto Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Zentrum für Infektionsforschung (HZI), 38124 Braunschweig, Germany;
| |
Collapse
|
2
|
Sayadmanesh A, Azadbakht M, Yari K, Abedelahi A, Shafaei H, Shanehbandi D, Baradaran B, Basiri M. Characterization of CAR T Cells Manufactured Using Genetically Engineered Artificial Antigen Presenting Cells. CELL JOURNAL 2023; 25:674-687. [PMID: 37865876 PMCID: PMC10591261 DOI: 10.22074/cellj.2023.2001712.1304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/22/2023] [Accepted: 07/01/2023] [Indexed: 10/23/2023]
Abstract
OBJECTIVE Chimeric antigen receptor (CAR) T cell therapy has recently emerged as a promising approach for the treatment of different types of cancer. Improving CAR T cell manufacturing in terms of costs and product quality is an important concern for expanding the accessibility of this therapy. One proposed strategy for improving T cell expansion is to use genetically engineered artificial antigen presenting cells (aAPC) expressing a membrane-bound anti-CD3 for T cell activation. The aim of this study was to characterize CAR T cells generated using this aAPC-mediated approach in terms of expansion efficiency, immunophenotype, and cytotoxicity. MATERIALS AND METHODS In this experimental study, we generated an aAPC line by engineering K562 cells to express a membrane-bound anti-CD3 (mOKT3). T cell activation was performed by co-culturing PBMCs with either mitomycin C-treated aAPCs or surface-immobilized anti-CD3 and anti-CD28 antibodies. Untransduced and CD19-CARtransduced T cells were characterized in terms of expansion, activation markers, interferon gamma (IFN-γ) secretion, CD4/CD8 ratio, memory phenotype, and exhaustion markers. Cytotoxicity of CD19-CAR T cells generated by aAPCs and antibodies were also investigated using a bioluminescence-based co-culture assay. RESULTS Our findings showed that the engineered aAPC line has the potential to expand CAR T cells similar to that using the antibody-based method. Although activation with aAPCs leads to a higher ratio of CD8+ and effector memory T cells in the final product, we did not observe a significant difference in IFN-γ secretion, cytotoxic activity or exhaustion between CAR T cells generated with aAPC or antibodies. CONCLUSION Our results show that despite the differences in the immunophenotypes of aAPC and antibody-based CAR T cells, both methods can be used to manufacture potent CAR T cells. These findings are instrumental for the improvement of the CAR T cell manufacturing process and future applications of aAPC-mediated expansion of CAR T cells.
Collapse
Affiliation(s)
- Ali Sayadmanesh
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamad Azadbakht
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Kheirollah Yari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Shafaei
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
3
|
Meng J, Lv Y, Bao W, Meng Z, Wang S, Wu Y, Li S, Jiao Z, Tian Z, Ma G, Wei W. Generation of whole tumor cell vaccine for on-demand manipulation of immune responses against cancer under near-infrared laser irradiation. Nat Commun 2023; 14:4505. [PMID: 37495590 PMCID: PMC10372023 DOI: 10.1038/s41467-023-40207-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 07/18/2023] [Indexed: 07/28/2023] Open
Abstract
The therapeutic efficacy of whole tumor cell vaccines (TCVs) is modest, which has delayed their translation into personalized immunotherapies in the clinic. Here, we develop a TCV platform based on photothermal nanoparticle-loaded tumor cells, which can be rationally applied to diverse tumor types to achieve on-demand boost of anti-tumor immune responses for inhibiting tumor growth. During the fabrication process, mild photothermal heating by near-infrared (NIR) laser irradiation induces the nanoparticle-bearing tumor cells to express heat shock proteins as endogenous adjuvants. After a single vaccination at the back of tumor-bearing mice, non-invasive NIR laser irradiation further induces mild hyperthermia at vaccination site, which promotes the recruitment, activation, and antigen presentation by dendritic cells. Using an indicator we term fluctuation of tumor growth rate, we determine appropriate irradiation regimens (including optimized irradiation intervals and times). This TCV platform enables on-demand NIR manipulation of immune responses, and we demonstrate potent therapeutic efficacy against six murine models that mimick a range of clinical scenarios, including a model based on humanized mice and patient-derived tumor xenografts.
Collapse
Affiliation(s)
- Jiaqi Meng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanlin Lv
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Weier Bao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zihui Meng
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, P. R. China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yuanbin Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Shuping Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Zhouguang Jiao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Zhiyuan Tian
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China.
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China.
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China.
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China.
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China.
| |
Collapse
|
4
|
Webster JA, Robinson TM, Blackford AL, Warlick E, Ferguson A, Borrello I, Zahurak M, Jones RJ, Smith BD. A randomized, phase II trial of adjuvant immunotherapy with durable TKI-free survival in patients with chronic phase CML. Leuk Res 2021; 111:106737. [PMID: 34768161 DOI: 10.1016/j.leukres.2021.106737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/14/2021] [Accepted: 10/25/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE Chronic myeloid leukemia (CP-CML) patients can achieve undetectable minimal residual disease (UMRD) and discontinue tyrosine kinase inhibitors (TKIs). Cellular immunity plays an important role in CML disease control. We conducted a randomized, non-blinded phase II trial of adjuvant immunotherapy with TKIs to facilitate TKI discontinuation. METHODS TKI-treated patients with CP-CML were randomized to receive the K562/GM-CSF vaccine (vaccine) OR Interferon-α + Sargramostim (IFN). If UMRD was achieved, then all treatment was stopped. Patients who did not achieve UMRD within one year, had a molecular relapse, or discontinued therapy for toxicity could crossover. RESULTS Thirty-four patients were randomized to IFN (n = 18) or vaccine (n = 16), and 21 patients crossed over (IFN⟶vaccine: n = 9, vaccine⟶IFN, n = 12). TKIs at enrollment included imatinib (n = 31), nilotinib (n = 2), and dasatinib (n = 1). No patients discontinued vaccine due to side effects, while 33 % of IFN-treated patients discontinued treatment. More patients randomized to IFN (47.4 %, 95 % CI: 16.7-66.7 %) versus vaccine (25.0 %, 95 % CI: 0.5-43.5 %) achieved UMRD within one year. Seven patients randomized to IFN discontinued treatment with 28.6 % (95 % CI: 8.9-92.2 %) sustaining treatment-free remission (TFR) at 1 year, while three patients randomized to vaccine discontinued treatment with none sustaining TFR. Including crossover, there was a cumulative discontinuation success rate of 36.4 % (95 % CI: 16.6 %-79.5 %) after adjuvant IFN. Patients who sustained TFR received a median of 29 months of imatinib prior to discontinuation. CONCLUSION Adjuvant IFN led to durable TFRs with limited prior TKI exposure with comparable success to prior discontinuation trials, but many patients stopped IFN early.
Collapse
Affiliation(s)
- Jonathan A Webster
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of MedIcine, Baltimore, MD, United States.
| | | | - Amanda L Blackford
- Division of Biostatistics and Bioinformatics, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Erica Warlick
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Anna Ferguson
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of MedIcine, Baltimore, MD, United States
| | - Ivan Borrello
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of MedIcine, Baltimore, MD, United States
| | - Marianna Zahurak
- Division of Biostatistics and Bioinformatics, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Richard J Jones
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of MedIcine, Baltimore, MD, United States
| | - B Douglas Smith
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of MedIcine, Baltimore, MD, United States
| |
Collapse
|
5
|
Schmidts A, Marsh LC, Srivastava AA, Bouffard AA, Boroughs AC, Scarfò I, Larson RC, Bedoya F, Choi BD, Frigault MJ, Bailey SR, Leick MB, Vatsa S, Kann MC, Prew MS, Kleinstiver BP, Joung JK, Maus MV. Cell-based artificial APC resistant to lentiviral transduction for efficient generation of CAR-T cells from various cell sources. J Immunother Cancer 2020; 8:jitc-2020-000990. [PMID: 32900862 PMCID: PMC7477986 DOI: 10.1136/jitc-2020-000990] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2020] [Indexed: 01/14/2023] Open
Abstract
Background Adoptive cell therapy with chimeric antigen receptor T cells (CAR-T) has become a standard treatment for patients with certain aggressive B cell malignancies and holds promise to improve the care of patients suffering from numerous other cancers in the future. However, the high manufacturing cost of CAR-T cell therapies poses a major barrier to their broader clinical application. Among the key cost drivers of CAR-T production are single-use reagents for T cell activation and clinical-grade viral vector. The presence of variable amounts of contaminating monocytes in the starting material poses an additional challenge to CAR-T manufacturing, since they can impede T cell stimulation and transduction, resulting in manufacturing failure. Methods We created K562-based artificial antigen-presenting cells (aAPC) with genetically encoded T cell stimulation and costimulation that represent an inexhaustible source for T cell activation. We additionally disrupted endogenous expression of the low-density lipoprotein receptor (LDLR) on these aAPC (aAPC-ΔLDLR) using CRISPR-Cas9 gene editing nucleases to prevent inadvertent lentiviral transduction and avoid the sink effect on viral vector during transduction. Using various T cell sources, we produced CD19-directed CAR-T cells via aAPC-ΔLDLR-based activation and tested their in vitro and in vivo antitumor potency against B cell malignancies. Results We found that lack of LDLR expression on our aAPC-ΔLDLR conferred resistance to lentiviral transduction during CAR-T production. Using aAPC-ΔLDLR, we achieved efficient expansion of CAR-T cells even from unpurified starting material like peripheral blood mononuclear cells or unmanipulated leukapheresis product, containing substantial proportions of monocytes. CD19-directed CAR-T cells that we produced via aAPC-ΔLDLR-based expansion demonstrated potent antitumor responses in preclinical models of acute lymphoblastic leukemia and B-cell lymphoma. Conclusions Our aAPC-ΔLDLR represent an attractive approach for manufacturing of lentivirally transduced T cells that may be simpler and more cost efficient than currently available methods.
Collapse
Affiliation(s)
- Andrea Schmidts
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Leah C Marsh
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ambike A Srivastava
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Amanda A Bouffard
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Angela C Boroughs
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Irene Scarfò
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca C Larson
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Felipe Bedoya
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bryan D Choi
- Cellular Immunotherapy Program and Department of Neurosurgery, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew J Frigault
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefanie R Bailey
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark B Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Sonika Vatsa
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael C Kann
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michelle S Prew
- Molecular Pathology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Benjamin P Kleinstiver
- Center for Genomic Medicine and Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - J Keith Joung
- Molecular Pathology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA .,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Özgür Yurttaş N, Eşkazan AE. Novel therapeutic approaches in chronic myeloid leukemia. Leuk Res 2020; 91:106337. [PMID: 32200189 DOI: 10.1016/j.leukres.2020.106337] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022]
Abstract
The tyrosine kinase inhibitors (TKIs) have revolutionized the management of chronic myeloid leukemia (CML) and BCR-ABL1 inhibitors form the mainstay of CML treatment. Although patients with CML generally do well under TKI therapy, there is a subgroup of patients who are resistant and/or intolerant to TKIs. In these group of patients, there is the need of additional treatment strategies. In this review, we provide an update on the current knowledge of these novel treatment approaches that can be used alone and/or in combination with TKIs.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Clinical Trials as Topic
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Everolimus/therapeutic use
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/immunology
- Gene Expression
- Histone Deacetylase Inhibitors/therapeutic use
- Homoharringtonine/therapeutic use
- Humans
- Immunotherapy/methods
- Interferon-alpha/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Molecular Targeted Therapy/methods
- Niacinamide/analogs & derivatives
- Niacinamide/therapeutic use
- Piperidines/therapeutic use
- Polyethylene Glycols/therapeutic use
- Protein Kinase Inhibitors/therapeutic use
- Pyrazoles/therapeutic use
- Pyridines/therapeutic use
- Quinolones/therapeutic use
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- Nurgül Özgür Yurttaş
- Division of Hematology, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ahmet Emre Eşkazan
- Division of Hematology, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| |
Collapse
|
7
|
Music M, Prassas I, Diamandis EP. Optimizing cancer immunotherapy: Is it time for personalized predictive biomarkers? Crit Rev Clin Lab Sci 2018; 55:466-479. [PMID: 30277835 DOI: 10.1080/10408363.2018.1499706] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer immunotherapy, a treatment that selectively augments a patient's anti-tumor immune response, is a breakthrough advancement in personalized medicine. A subset of cancer patients undergoing immunotherapy have displayed robust and long-lasting therapeutic responses. Currently, the spotlight is on the use of blocking antibodies against the T-cell checkpoint molecules, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programed cell death-1 (PD-1)/programed death-ligand 1 (PD-L1), which have been effectively used to combat many cancers types. Despite the overall enthusiasm, immune checkpoint blockade inhibitors suffer from significant limitations such as high cost, serious toxicity in a substantial proportion of patients, and a response rate as low as 10%-40% in some clinical trials. Consequently, there is an urgent and unmet medical need for companion biomarkers that could both predict the response of individual patients to these therapies, and provide the means for precise monitoring of their therapeutic outcome. In this era of precision medicine, predictive biomarkers are a hot commodity because they can effectively separate responders from non-responders, and spare non-responders from serious therapy-related toxicity. Emerging predictive biomarkers for immune checkpoint blockade are: PD-L1 expression, increased amounts of tumor-infiltrating lymphocytes, increased mutational load and mismatch repair deficiency. Other well-studied biomarkers include inflammatory infiltrate, absolute lymphocyte count and lactate dehydrogenase levels. We review recent progress on predictive cancer biomarkers in immunotherapy, with a special emphasis on serum autoantibodies that have the potential to be personalized for optimal clinical outcomes.
Collapse
Affiliation(s)
- Milena Music
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada
| | - Ioannis Prassas
- b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada
| | - Eleftherios P Diamandis
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada.,b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada.,c Department of Clinical Biochemistry , University Health Network , Toronto , Canada.,d Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital , Toronto , Canada
| |
Collapse
|
8
|
Kavanagh S, Nee A, Lipton JH. Emerging alternatives to tyrosine kinase inhibitors for treating chronic myeloid leukemia. Expert Opin Emerg Drugs 2018; 23:51-62. [PMID: 29480034 DOI: 10.1080/14728214.2018.1445717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION BCR-ABL-directed tyrosine kinase inhibitors (TKIs) have revolutionised therapy for chronic myeloid leukemia. However, despite the availability and efficacy of this class of agents, lifelong treatment is still required in a significant proportion of patients Areas covered: We give an overview of the currently available BCR-ABL-directed TKIs and other conventional therapies for CML. We proceed to review the current market and some of the scientific rationale for new drug development before outlining a number of novel therapies, considered broadly as immunotherapies and targeted agents. Published English-language literature was reviewed regarding currently available TKIs; clinical trials repositories were reviewed to identify novel agents recently investigated or under active study. Expert opinion: We recommend discussion with patients and enrolment on an appropriate clinical trial where feasible. In situations where no trials are available, or if patients decline enrolment, we recommend use of an appropriate BCR-ABL directed TKI, selected on the basis of an evaluation of patient risk factors and side effect profile. Allogeneic stem cell transplant continues to have a role though this is generally limited to cases with advanced phases of disease or in cases with resistance-conferring mutations.
Collapse
Affiliation(s)
- Simon Kavanagh
- a Princess Margaret Cancer Centre , University Health Network , Toronto , ON , Canada
| | - Aisling Nee
- a Princess Margaret Cancer Centre , University Health Network , Toronto , ON , Canada
| | - Jeffrey H Lipton
- a Princess Margaret Cancer Centre , University Health Network , Toronto , ON , Canada
| |
Collapse
|
9
|
Vaccination with autologous myeloblasts admixed with GM-K562 cells in patients with advanced MDS or AML after allogeneic HSCT. Blood Adv 2017; 1:2269-2279. [PMID: 29296875 DOI: 10.1182/bloodadvances.2017009084] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/08/2017] [Indexed: 01/05/2023] Open
Abstract
We report a clinical trial testing vaccination of autologous myeloblasts admixed with granulocyte-macrophage colony-stimulating factor secreting K562 cells after allogeneic hematopoietic stem cell transplantation (HSCT). Patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) with ≥5% marrow blasts underwent myeloblast collection before HSCT. At approximately day +30, 6 vaccines composed of irradiated autologous myeloblasts mixed with GM-K562 were administered. Tacrolimus-based graft-versus-host disease (GVHD) prophylaxis was not tapered until vaccine completion (∼day 100). Thirty-three patients with AML (25) and MDS (8) enrolled, 16 (48%) had ≥5% marrow blasts at transplantation. The most common vaccine toxicity was injection site reactions. One patient developed severe eosinophilia and died of eosinophilic myocarditis. With a median follow-up of 67 months, cumulative incidence of grade 2-4 acute and chronic GVHD were 24% and 33%, respectively. Relapse and nonrelapse mortality were 48% and 9%, respectively. Progression-free survival (PFS) and overall survival (OS) at 5 years were 39% and 39%. Vaccinated patients who were transplanted with active disease (≥5% marrow blasts) had similar OS and PFS at 5 years compared with vaccinated patients transplanted with <5% marrow blasts (OS, 44% vs 35%, respectively, P = .81; PFS, 44% vs 35%, respectively, P = .34). Postvaccination antibody responses to angiopoietin-2 was associated with superior OS (hazard ratio [HR], 0.43; P = .031) and PFS (HR, 0.5; P = .036). Patients transplanted with active disease had more frequent angiopoeitin-2 antibody responses (62.5% vs 20%, P = .029) than those transplanted in remission. GM-K562/leukemia cell vaccination induces biologic activity, even in patients transplanted with active MDS/AML. This study is registered at www.clinicaltrials.gov as #NCT 00809250.
Collapse
|
10
|
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: 1.8] [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
|
11
|
Hosen N, Maeda T, Hashii Y, Tsuboi A, Nishida S, Nakata J, Oji Y, Oka Y, Sugiyama H. Wilms tumor 1 peptide vaccination after hematopoietic stem cell transplant in leukemia patients. Stem Cell Investig 2016; 3:90. [PMID: 28078270 DOI: 10.21037/sci.2016.11.08] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/07/2016] [Indexed: 01/03/2023]
Abstract
Although the prognosis of leukemia patients after allogeneic hematopoietic stem cell transplantation (HSCT) has greatly improved, relapse is still a major cause of death after HSCT. Cancer vaccines may have the potential to enhance the graft-versus-leukemia (GVL) effect. The post-allogeneic HSCT period provides a unique platform for vaccination, because (I) tumor burden is minimal, (II) lymphopenia allows for rapid expansion of cytotoxic T cells (CTLs), (III) donor-derived CTLs are not exhausted, (IV) inflammation is caused by alloreactions, and (V) the abundance of regulatory T cells is low due to their late recovery. Tumor cell lysates, dendritic cells (DCs), and peptides derived from leukemia-associated antigens (LAAs) have been used as vaccines. Clinical trials with several types of vaccines for post-HSCT patients revealed that the vaccination induced an immunological response and might benefit patients with minimal residual disease; however, the efficacy of this approach must be examined in randomized studies. In addition, it is important to consider the combination of cancer vaccine with checkpoint antibodies, recently shown to be useful in treating leukemia relapse after HSCT.
Collapse
Affiliation(s)
- Naoki Hosen
- Department of Cancer stem cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Maeda
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sumiyuki Nishida
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Nakata
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Oji
- Department of Cancer stem cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Oka
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
12
|
Šmahelová J, Kaštánková I, Poláková KM, Klamová H, Zemanová K, Tachezy R, Hamšíková E, Šmahel M. Expression of genes encoding centrosomal proteins and the humoral response against these proteins in chronic myeloid leukemia. Oncol Rep 2016; 37:547-554. [DOI: 10.3892/or.2016.5226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/25/2016] [Indexed: 11/06/2022] Open
|
13
|
Zebrafish Models of Human Leukemia: Technological Advances and Mechanistic Insights. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:335-69. [PMID: 27165361 DOI: 10.1007/978-3-319-30654-4_15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insights concerning leukemic pathophysiology have been acquired in various animal models and further efforts to understand the mechanisms underlying leukemic treatment resistance and disease relapse promise to improve therapeutic strategies. The zebrafish (Danio rerio) is a vertebrate organism with a conserved hematopoietic program and unique experimental strengths suiting it for the investigation of human leukemia. Recent technological advances in zebrafish research including efficient transgenesis, precise genome editing, and straightforward transplantation techniques have led to the generation of a number of leukemia models. The transparency of the zebrafish when coupled with improved lineage-tracing and imaging techniques has revealed exquisite details of leukemic initiation, progression, and regression. With these advantages, the zebrafish represents a unique experimental system for leukemic research and additionally, advances in zebrafish-based high-throughput drug screening promise to hasten the discovery of novel leukemia therapeutics. To date, investigators have accumulated knowledge of the genetic underpinnings critical to leukemic transformation and treatment resistance and without doubt, zebrafish are rapidly expanding our understanding of disease mechanisms and helping to shape therapeutic strategies for improved outcomes in leukemic patients.
Collapse
|
14
|
Sanders KL, Fox BA, Bzik DJ. Attenuated Toxoplasma gondii therapy of disseminated pancreatic cancer generates long-lasting immunity to pancreatic cancer. Oncoimmunology 2015; 5:e1104447. [PMID: 27141388 DOI: 10.1080/2162402x.2015.1104447] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/25/2015] [Accepted: 09/29/2015] [Indexed: 12/21/2022] Open
Abstract
We have recently reported that treatment of disseminated pancreatic cancer with an attenuated Toxoplasma gondii uracil auxotroph vaccine promoted antitumor CD8+ T cell responses and long-term survival. Here, we optimized the treatment strategy for disseminated pancreatic cancer and show that attenuated Toxoplasma gondii therapy stimulated effective long-term immunity to pancreatic cancer through mechanisms involving CD4+ T cells and pancreatic tumor-specific IgG. Our results suggest that cell-mediated immunity in conjunction with humoral antibody immunity may offer greater resistance to recurrence of highly aggressive tumors. Cancer immunotherapeutic strategies using attenuated Toxoplasma gondii vaccines merit further investigation as a novel strategy to reawaken immunity to primary pancreatic carcinoma and to generate long-lasting immunity to pancreatic cancer recurrences.
Collapse
Affiliation(s)
- Kiah L Sanders
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth , Lebanon, NH, USA
| | - Barbara A Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth , Lebanon, NH, USA
| | - David J Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth , Lebanon, NH, USA
| |
Collapse
|
15
|
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
|
16
|
Abstract
The introduction of protein tyrosine kinase inhibitors (TKIs) in 1998 transformed the management of chronic myeloid leukemia (CML), leading to significantly reduced mortality and improved 5 year survival rates. However, the CML community is faced with several clinical issues that need to be addressed. Ten to 15% of CML patients are diagnosed in advanced phase, and small numbers of chronic phase (CP) cases experience disease progression each year during treatment. For these patients, TKIs induce only transient responses and alternative treatment strategies are urgently required. Depending on choice of first line TKI, approximately 30% of CML CP cases show suboptimal responses, due to a combination of poor compliance, drug intolerance, and drug resistance, with approximately 50% of TKI-resistance caused by kinase domain mutations and the remainder due to unknown mechanisms. Finally, the chance of successful treatment discontinuation is on the order of only 10-20% related to disease persistence. Disease persistence is a poorly understood phenomenon; all CML patients have functional Philadelphia positive (Ph+) stem and progenitor cells in their bone marrows and continue to express BCR-ABL1 by DNA PCR, even when in very deep remission and following treatment discontinuation. What controls the maintenance of these persisting cells, whether it is necessary to fully eradicate the malignant clone to achieve cure, and how that might be approached therapeutically are open questions.
Collapse
Affiliation(s)
- Tessa L Holyoake
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
17
|
Seo HS. Application of radiation technology in vaccines development. Clin Exp Vaccine Res 2015; 4:145-58. [PMID: 26273573 PMCID: PMC4524899 DOI: 10.7774/cevr.2015.4.2.145] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/10/2015] [Accepted: 06/20/2015] [Indexed: 12/11/2022] Open
Abstract
One of the earliest methods used in the manufacture of stable and safe vaccines is the use of chemical and physical treatments to produce inactivated forms of pathogens. Although these types of vaccines have been successful in eliciting specific humoral immune responses to pathogen-associated immunogens, there is a large demand for the development of fast, safe, and effective vaccine manufacturing strategies. Radiation sterilization has been used to develop a variety of vaccine types, because it can eradicate chemical contaminants and penetrate pathogens to destroy nucleic acids without damaging the pathogen surface antigens. Nevertheless, irradiated vaccines have not widely been used at an industrial level because of difficulties obtaining the necessary equipment. Recent successful clinical trials of irradiated vaccines against pathogens and tumors have led to a reevaluation of radiation technology as an alternative method to produce vaccines. In the present article, we review the challenges associated with creating irradiated vaccines and discuss potential strategies for developing vaccines using radiation technology.
Collapse
Affiliation(s)
- Ho Seong Seo
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea
| |
Collapse
|
18
|
Hosen N, Maeda T, Hashii Y, Tsuboi A, Nishida S, Nakata J, Nakae Y, Takashima S, Oji Y, Oka Y, Kumanogoh A, Sugiyama H. Vaccination strategies to improve outcome of hematopoietic stem cell transplant in leukemia patients: early evidence and future prospects. Expert Rev Hematol 2014; 7:671-81. [DOI: 10.1586/17474086.2014.953925] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
19
|
Tesfatsion DA. Dendritic cell vaccine against leukemia: advances and perspectives. Immunotherapy 2014; 6:485-96. [PMID: 24815786 DOI: 10.2217/imt.14.12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
As with many other types of malignancies, sustainable eradication of leukemia has been a challenge. This is related to the inevitable failure of conventional chemotherapeutic agents and radiation therapy to target the relatively quiescent leukemia stem cells, which are believed to have multidrug resistance, antiapoptotic capacity and enhanced DNA repair mechanisms allowing them to evade the immune system. Considering other therapeutic options that are minimally toxic to normal cells and effectively target not only the majority and more differentiated cancer cells, but also the rare residual leukemia cells, is of paramount importance. A number of immunotherapeutic options have been proposed to counter this challenge. One of the remarkable achievements in the field of immunotherapy has been the successful use of antigen presenting cells as vehicles of tumor/pathogenic antigens to the T-cell compartments. This review will focus on advances and perspectives of this arm of immunotherapy against leukemia.
Collapse
|
20
|
|