1
|
Zhang D, Jiang Y, Wang M, Zhao J, Wan J, Li Z, Huang D, Yu J, Li J, Liu J, Huang F, Hao S. A novel costimulatory molecule gene-modified leukemia cell-derived exosome enhances the anti-leukemia efficacy of DC vaccine in mouse models. Vaccine 2024; 42:126097. [PMID: 38960787 DOI: 10.1016/j.vaccine.2024.06.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVES Leukemia cell-derived exosomes (LEXs), carrying leukemia cell-specific antigens, can serve as a source of antigen for dendritic cell (DC) vaccine loading. However, LEX-targeted DC-based vaccines have demonstrated limited antitumor immune effects in clinical trials, attributed to the low immunogenicity of LEXs and the scant levels of costimulatory molecules on DCs. The costimulatory molecules CD80 and CD86, which are crucial to DC function, play a significant role in enhancing immune efficacy. In this study, we explored the anti-leukemia immune response of costimulatory molecule gene-modified LEX-targeted DCs (LEX-8086) in vitro and in animal models. METHODS DCs were incubated with LEX-8086 to produce LEX-8086-targeted DCs (DCsLEX-8086). ELISA, cytotoxicity assays and flow cytometry utilized to assess the antitumor efficacy of DCsLEX8086 in vitro. Flow cytometry was used to evaluate the immunomodulatory function of DCsLEX8086 in animal models. RESULTS Our findings indicated that LEX-8086 enhanced the maturation and antigen-presenting ability of DCs. Immunization with DCsLEX8086 significantly activated CD8+ T cells and boosted the CTL response in vitro. More importantly, DCsLEX-8086 effectively suppressed tumor growth and exerted anti-leukemia effects in both prophylactic and therapeutic animal models. Furthermore, DCsLEX-8086 promoted the proportion of CD4+ T cells, CD8+ T cells and M1 macrophages in the tumor environments both prophylactically and therapeutically. Treatment with DCsLEX-8086 showed no significant difference in the levels of M2 macrophages but decreased the proportion of Tregs within the tumor bed during therapeutic experiments. CONCLUSION The results suggested that DCsLEX-8086 induces a more effective anti-leukemia immunity compared to DCsLEX-null in vivo and in vitro. DCsLEX-8086 might achieve antitumor effects by elevating the numbers of CD4+ T cells, CD8+ T cells, and M1 macrophages in tumors. Our findings indicate that DCsLEX-8086 could be leveraged to develop a new, highly effective vaccine for anti-leukemia immunity.
Collapse
Affiliation(s)
- Difan Zhang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Jiang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minghui Wang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhao
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiangbo Wan
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhichao Li
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Huang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Yu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Li
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayu Liu
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Huang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Siguo Hao
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
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
|
3
|
Kwaśnik P, Giannopoulos K. Treatment-Free Remission-A New Aim in the Treatment of Chronic Myeloid Leukemia. J Pers Med 2021; 11:697. [PMID: 34442340 PMCID: PMC8399881 DOI: 10.3390/jpm11080697] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 02/08/2023] Open
Abstract
Tyrosine kinases inhibitors (TKIs) revolutionized chronic myeloid leukemia (CML) treatment for many years, prolonging patients' life expectancy to be comparable to age-matched healthy individuals. According to the latest the European LeukemiaNet (ELN) recommendations, CML treatment aims to achieve long-term remission without treatment (TFR), which is feasible in more than 40% of patients. Nearly all molecular relapses occur during the first 6 months after TKI withdrawal and do not progress to clinical relapse. The mechanisms that are responsible for CML relapses remain unexplained. It is suggested that maintaining TFR is not directly related to the total disposing of the gene transcript BCR-ABL1, but it might be a result of the restoration of the immune surveillance in CML. The importance of the involvement of immunocompetent cells in the period of TKI withdrawal is also emphasized by the presence of specific symptoms in some patients with "withdrawal syndrome". The goal of this review is to analyze data from studies regarding TFRs in order to characterize the elements of the immune system of patients that might prevent CML molecular relapse. The role of modern droplet digital polymerase chain reaction (ddPCR) and next-generation sequencing (NGS) in better identification of low levels of BCR-ABL1 transcripts was also taken into consideration for refining the eligibility criteria to stop TKI therapy.
Collapse
Affiliation(s)
- Paulina Kwaśnik
- Department of Experimental Hematooncology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Krzysztof Giannopoulos
- Department of Experimental Hematooncology, Medical University of Lublin, 20-093 Lublin, Poland;
- Department of Hematology, St John’s Cancer Center, 20-090 Lublin, Poland
| |
Collapse
|
4
|
Jo T, Shioya H, Tominaga H, Sakai T, Hayashi S, Matsuzaka K, Kaneko Y, Matsuo M, Taguchi J. Induction of Effector and Memory Cellular Immunity in a Patient with Long-Term Complete Molecular Response to Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia. Case Rep Oncol 2020; 13:990-996. [PMID: 32999661 PMCID: PMC7506382 DOI: 10.1159/000508997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 11/19/2022] Open
Abstract
This case report is about a patient who suffered from Philadelphia chromosome (Ph1)-positive acute lymphoblastic leukemia. The blasts were positive for myeloid-lineage markers including CD13 and CD33, as well as B-cell-lineage markers. Minor bcr-abl1 mRNA was detected by real-time quantitative polymerase chain reaction. Chromosomal abnormality monosomy 7 was also observed, in addition to Ph1. Despite treatment difficulties that were anticipated based on these findings, the patient had long-time complete molecular response (CMR) for approximately 5 years using chemotherapy and two tyrosine kinase inhibitors, imatinib and dasatinib. Lymphocytes were elevated after the patient switched from imatinib to dasatinib, and a T-cell receptor (TCR) V beta gene repertoire analysis revealed oligoclonal expansion of effector and memory cytotoxic T lymphocytes (CTLs), including Wilms tumor 1-specific CTLs. More specifically, the two memory CTLs expressing TCR V beta 3 and V beta 7.1 gradually increased after dasatinib administration. The activation and maintenance of anti-leukemia immunity may have allowed the patient to obtain long-time CMR. These results highlight that obtaining memory CTLs for leukemia cells may lead to safe withdrawal from dasatinib in the patient.
Collapse
Affiliation(s)
- Tatsuro Jo
- Department of Hematology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Haruna Shioya
- Department of Laboratory, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Hiroo Tominaga
- Department of Laboratory, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Takahiro Sakai
- Department of Laboratory, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Shizuka Hayashi
- Department of Laboratory, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Kaori Matsuzaka
- Department of Laboratory, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Yohei Kaneko
- Department of Laboratory, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Masatoshi Matsuo
- Department of Hematology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Jun Taguchi
- Department of Hematology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| |
Collapse
|
5
|
Hu W, Huang F, Ning L, Hao J, Wan J, Hao S. Enhanced immunogenicity of leukemia-derived exosomes via transfection with lentiviral vectors encoding costimulatory molecules. Cell Oncol (Dordr) 2020; 43:889-900. [PMID: 32578140 PMCID: PMC7581614 DOI: 10.1007/s13402-020-00535-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2020] [Indexed: 12/31/2022] Open
Abstract
Background: Tumor cell-derived exosomes (TEXs) have been widely used to induce antitumor immune responses in animal models and clinical trials. Similarly, leukemia cell-derived exosomes (LEXs) can induce antileukemia immune responses in animal models. However, the antileukemia immunity induced by LEXs is less effective, which may be due to an inadequate costimulatory capacity. Methods: In this study, we transduced L1210 leukemia cells with a lentiviral vector encoding two B7 costimulatory molecules (CD80, CD86) and obtained LEXs that highly expressed CD80 and CD86. The antileukemia immune response derived from these LEXs was examined in vitro and in vivo in animal models. Results: We found that B7 gene-modified LEXs, including LEX-CD80, LEX-CD86, and LEX-8086, could significantly boost the expression of CD80 and CD86 in dendritic cells (DCs) and promote the secretion of functional cytokines such as TNF-α and IL-12. Moreover, these B7 gene-modified LEXs, particularly LEX-CD8086, could effectively induce CD4+ T cell proliferation, Th1 cytokine secretion, and an antigen-specific anti-leukemia cytotoxic T lymphocyte (CTL) response. Additional animal studies indicated that immunization with B7 gene-modified LEXs, in particular LEX-CD8086, could significantly retard tumor growth compared to the control LEXnull group. Conclusions: This study sheds light on the feasibility of obtaining LEXs that overexpress costimulatory molecules via genetically modified leukemia cells, thereby enhancing their anti-leukemia immunity and providing a potential therapeutic strategy that contributes to leukemia immunotherapy.
Collapse
Affiliation(s)
- Weiwei Hu
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665# Kongjiang Road, Shanghai, 200090, China
| | - Fang Huang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665# Kongjiang Road, Shanghai, 200090, China
| | - Liuxin Ning
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665# Kongjiang Road, Shanghai, 200090, China
| | - Jun Hao
- Interdisciplinary Oncology Program, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Jiangbo Wan
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665# Kongjiang Road, Shanghai, 200090, China
| | - Siguo Hao
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665# Kongjiang Road, Shanghai, 200090, China.
| |
Collapse
|
6
|
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
|
7
|
Jin Z, Xu L, Li Y. Approaches for generation of anti-leukemia specific T cells. CELL REGENERATION 2019; 7:40-44. [PMID: 30671229 PMCID: PMC6326242 DOI: 10.1016/j.cr.2018.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/13/2018] [Accepted: 09/26/2018] [Indexed: 02/06/2023]
Abstract
As three decades ago, it was reported that adoptive T cell immunotherapy by infusion of autologous tumor infiltrating lymphocytes (TILs) mediated objective cancer regression in patients with metastatic melanoma. A new era of T cell immunotherapy arose since the improvement and clinical use of anti-CD19 chimeric antigen receptor T cells (CAR-T) for the treatment of refractory and relapsed B lymphocyte leukemia. However, several challenges and difficulties remain on the way to reach generic and effective T cell immunotherapy, including lacking a generic method for generating anti-leukemia-specific T cells from every patient. Here, we summarize the current methods of generating anti-leukemia-specific T cells, and the promising approaches in the future.
Collapse
Key Words
- ACT, adoptive cellular immunotherapy
- APL, promyelocytic leukemia
- Anti-leukemia T cell
- B-ALL, cell acute lymphoblastic leukemia
- CAR-T
- CAR-T, chimeric antigen receptor T cells
- CML, chronic myelogenous leukemia
- CR, complete remission
- CTLs, cytotoxic T cells
- DLI, donor lymphocyte infusion
- FLT3-ITD, FLT3 internal tandem duplication
- GVHD, graft-versus-host disease
- GVL, graft-versus-leukemia
- HLA, human leukocyte antigen
- HPCs, hematopoietic progenitor cells
- IL-2, interleukin-2
- Ig, immunoglobulin
- T cell immunotherapy
- T cell reprogramming
- TAA, tumor-associated antigen
- TCR-T
- TCR-T, TCR gene-modified T cell
- TIL, infiltrating lymphocytes
- TKI, tyrosine kinase inhibitor
- WT1, Wilm's tumor antigen 1
- allo-HSCT, allogeneic hematopoietic stem cell transplantation
- hESC, human embryonic stem cell
- iPSCs, induced pluripotent stem cells
- iTs, induced functional T cells
- scFv, single-chain variable fragment
Collapse
Affiliation(s)
- Zhenyi Jin
- Key Laboratory for Regenerative Medicine of Ministry of Education; Institute of Hematology, School of Medicine; Jinan University, Guangzhou, 510632, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Ling Xu
- Key Laboratory for Regenerative Medicine of Ministry of Education; Institute of Hematology, School of Medicine; Jinan University, Guangzhou, 510632, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education; Institute of Hematology, School of Medicine; Jinan University, Guangzhou, 510632, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
8
|
Imamura T, Komatsu S, Ichikawa D, Miyamae M, Okajima W, Ohashi T, Kiuchi J, Nishibeppu K, Kosuga T, Konishi H, Shiozaki A, Fujiwara H, Okamoto K, Tsuda H, Otsuji E. Overexpression of ZRF1 is related to tumor malignant potential and a poor outcome of gastric carcinoma. Carcinogenesis 2018; 39:263-271. [PMID: 29228320 DOI: 10.1093/carcin/bgx139] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022] Open
Abstract
Zuotin-related factor 1 (ZRF1) is a recently characterized epigenetic factor involved in transcriptional regulation and is highly overexpressed in several malignancies, but it is not known whether it plays a role in gastric cancer (GC). In this study, we investigated whether ZRF1 acts as a cancer-promoting gene through its activation/overexpression in GC. We analyzed five GC cell lines and 133 primary tumors, which had been curatively resected in our hospital between 2001 and 2003. Overexpression of ZRF1 was detected in GC cell lines (four out of five lines, 80.0%) and was detected in primary tumor samples of GC (52 out of 133 cases, 39.1%) and significantly correlated with differentiated histological type, venous invasion, lymphatic invasion, advanced stage and a higher recurrence rate. ZRF1-overexpressing tumors had a worse survival rate than those with non-expressing tumors (P < 0.01, log-rank test). ZRF1 positivity was independently associated with a worse outcome in the multivariate analysis (P < 0.01; hazard ratio 4.92; 95% confidence interval: 1.6-21.1). In ZRF1-overexpressing GC cells, knockdown of ZRF1 using specific siRNAs inhibited the cell proliferation, migration and invasion and induced apoptosis in a p53-dependent manner. These findings suggest that ZRF1 plays a crucial role in tumor malignant potential through its overexpression and highlight its usefulness as a prognostic factor and potential therapeutic target in GC.
Collapse
Affiliation(s)
- Taisuke Imamura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Daisuke Ichikawa
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Mahito Miyamae
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Wataru Okajima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Keiji Nishibeppu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hitoshi Tsuda
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan.,Department of Basic Pathology, National Defense Medical College, Tokorozawa, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| |
Collapse
|
9
|
Kaymak A, Sayols S, Papadopoulou T, Richly H. Role for the transcriptional activator ZRF1 in early metastatic events in breast cancer progression and endocrine resistance. Oncotarget 2018; 9:28666-28690. [PMID: 29983888 DOI: 10.18632/oncotarget.25596] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 05/24/2018] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is one of the most common malignancies among women which is often treated with hormone therapy and chemotherapy. Despite the improvements in detection and treatment of breast cancer, the vast majority of breast cancer patients are diagnosed with metastatic disease either at the beginning of the disease or later during treatment. Still, the molecular mechanisms causing a therapy resistant metastatic breast cancer are still elusive. In the present study we addressed the function of the transcriptional activator ZRF1 during breast cancer progression. We provide evidence that ZRF1 plays an essential role for the early metastatic events in vitro and acts like a tumor suppressor protein during the progression of breast invasive ductal carcinoma into a more advanced stage. Hence, depletion of ZRF1 results in the acquisition of metastatic behavior by facilitating the initiation of the metastatic cascade, notably for cell adhesion, migration and invasion. Furthermore absence of ZRF1 provokes endocrine resistance via misregulation of cell death and cell survival related pathways. Taken together, we have identified ZRF1 as an important regulator of breast cancer progression that holds the potential to be explored for new treatment strategies in the future.
Collapse
Affiliation(s)
- Aysegül Kaymak
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology, Mainz, Germany
| | - Sergi Sayols
- Bioinformatics Core Facility, Institute of Molecular Biology, Mainz, Germany
| | - Thaleia Papadopoulou
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology, Mainz, Germany.,Department of Developmental and Stem Cell Biology, Institute Pasteur, Paris, France
| | - Holger Richly
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology, Mainz, Germany
| |
Collapse
|
10
|
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
|
11
|
Abstract
Historically, immune-based therapies have played a leading role in the treatment of hematologic malignancies, with the efficacy of stem cell transplantation largely attributable to donor immunity against malignant cells. As new and more targeted immunotherapies have developed, their role in the treatment of hematologic malignancies is evolving and expanding. Herein, we discuss approaches for antigen discovery and review known and novel tumor antigens in hematologic malignancies. We further explore the role of established and investigational immunotherapies in hematologic malignancies, with a focus on personalization of treatment modalities such as cancer vaccines and adoptive cell therapy. Finally, we identify areas of active investigation and development. Immunotherapy is at an exciting crossroads for the treatment of hematologic malignancies, with further investigation aimed at producing effective, targeted immune therapies that maximize antitumor effects while minimizing toxicity.
Collapse
Affiliation(s)
- David A. Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
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: 0.3] [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
|
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
|
14
|
Aloia L, Demajo S, Di Croce L. ZRF1: a novel epigenetic regulator of stem cell identity and cancer. Cell Cycle 2015; 14:510-5. [PMID: 25665097 DOI: 10.4161/15384101.2014.988022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The Zuotin-related factor 1, ZRF1, has recently been identified as an epigenetic regulator of gene transcription in stem cells and cancer. During differentiation of human teratocarcinoma cells, ZRF1 promotes transcriptional induction of developmental genes that are repressed by Polycomb complexes. Importantly, ZRF1 has recently been shown to be required for both neural differentiation of embryonic stem cells (ESCs) and for maintenance of neural progenitor cell (NPC) identity. Moreover, a dual role has now emerged for ZRF1 in cancer: on the one hand, ZRF1 plays a crucial role in oncogene-induced senescence (OIS) by activating the INK4/ARF locus, thus working as a tumor suppressor; on the other hand, ZRF1 promotes leukemogenesis in acute myeloid leukemia (AML) in a Polycomb-independent fashion. Therefore, increasing evidence points to ZRF1 as a novel target for therapy of neurodegenerative diseases and cancer.
Collapse
Key Words
- AML, acute myeloid leukemia
- ChIP, chromatin immunoprecipitation
- ESC, embryonic stem cells
- H2Aub1, mono-ubiquitinated histone H2A
- HDAC, histone deacetylase
- NPC, neural progenitor cells
- OIS, oncogene-induced senescence
- PRC1, polycomb repressive complex 1
- PRC2, polycomb repressive complex 2
- RA, retinoic acid
- RARa, retinoic acid receptor a
- UBD, ubiquitin binding domain
- ZRF1
- cancer
- cell fate
- development
- differentiation
- epigenetics
- polycomb
- retinoic acid
- senescence
- stem cell
- transcription
Collapse
Affiliation(s)
- Luigi Aloia
- a Centre for Genomic Regulation (CRG) ; Barcelona , Spain
| | | | | |
Collapse
|
15
|
Demajo S, Uribesalgo I, Gutiérrez A, Ballaré C, Capdevila S, Roth M, Zuber J, Martín-Caballero J, Di Croce L. ZRF1 controls the retinoic acid pathway and regulates leukemogenic potential in acute myeloid leukemia. Oncogene 2014; 33:5501-10. [PMID: 24292673 DOI: 10.1038/onc.2013.501] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 10/15/2013] [Accepted: 10/18/2013] [Indexed: 11/09/2022]
Abstract
Acute myeloid leukemia (AML) is frequently linked to epigenetic abnormalities and deregulation of gene transcription, which lead to aberrant cell proliferation and accumulation of undifferentiated precursors. ZRF1, a recently characterized epigenetic factor involved in transcriptional regulation, is highly overexpressed in human AML, but it is not known whether it plays a role in leukemia progression. Here, we demonstrate that ZRF1 depletion decreases cell proliferation, induces apoptosis and enhances cell differentiation in human AML cells. Treatment with retinoic acid (RA), a differentiating agent currently used to treat certain AMLs, leads to a functional switch of ZRF1 from a negative regulator to an activator of differentiation. At the molecular level, ZRF1 controls the RA-regulated gene network through its interaction with the RA receptor α (RARα) and its binding to RA target genes. Our genome-wide expression study reveals that ZRF1 regulates the transcription of nearly half of RA target genes. Consistent with our in vitro observations that ZRF1 regulates proliferation, apoptosis, and differentiation, ZRF1 depletion strongly inhibits leukemia progression in a xenograft mouse model. Finally, ZRF1 knockdown cooperates with RA treatment in leukemia suppression in vivo. Taken together, our data reveal that ZRF1 is a key transcriptional regulator in leukemia progression and suggest that ZRF1 inhibition could be a novel strategy to be explored for AML treatment.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/genetics
- Blotting, Western
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Disease Progression
- Gene Expression Regulation, Neoplastic/physiology
- Humans
- Immunoprecipitation
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, SCID
- Molecular Chaperones
- Oligonucleotide Array Sequence Analysis
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- RNA-Binding Proteins
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Transcription, Genetic
- Transfection
- Tretinoin/metabolism
- Tretinoin/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- S Demajo
- 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain
| | - I Uribesalgo
- 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain
| | - A Gutiérrez
- 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain
| | - C Ballaré
- 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain
| | - S Capdevila
- Unidad de Animal de Laboratorio, PRBB, Barcelona, Spain
| | - M Roth
- Institute of Molecular Pathology (IMP), Vienna, Austria
| | - J Zuber
- Institute of Molecular Pathology (IMP), Vienna, Austria
| | | | - L Di Croce
- 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain [3] Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
16
|
Brayer JB, Pinilla-Ibarz J. Developing strategies in the immunotherapy of leukemias. Cancer Control 2013; 20:49-59. [PMID: 23302907 DOI: 10.1177/107327481302000108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In the current treatment paradigms for leukemias, hematopoietic stem cell transplant (HSCT) is considered the best option with a curative potential although more often than not it simply delays disease progression. Advances are needed, both in current therapies and in the development of new strategies. Partly from studying the nuances of the curative potential of stem cell transplant, we have come to appreciate the relevance of the immune response and the potential of immunotherapy. METHODS This review article summarizes the recent advances in the field of immunology and immunotherapy for leukemia. RESULTS In passive immunotherapy, recent progress in chimeric T-cell antigen receptor technology has been encouraging. In active immunotherapy, a cancer vaccine may potentially enhance HSCT. An overview of various clinical studies of peptide vaccination strategies focusing on molecular targets such as the Wilms' tumor gene 1 (WT1), proteinase 3 (PR3), and receptor for hyaluronan acid-mediated motility (RHAMM) is provided. Cell-based vaccination strategies are also briefly explored. CONCLUSIONS The immune system clearly has the capacity to recognize and react to leukemic cells, and recent evidence directs our attention to the importance of mounting inflammatory and CD4 T-cell responses to complement and support the cytotoxic activity elicited by peptide vaccines.
Collapse
Affiliation(s)
- Jason B Brayer
- Malignant Hematology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | | |
Collapse
|
17
|
Snauwaert S, Vanhee S, Goetgeluk G, Verstichel G, Van Caeneghem Y, Velghe I, Philippé J, Berneman ZN, Plum J, Taghon T, Leclercq G, Thielemans K, Kerre T, Vandekerckhove B. RHAMM/HMMR (CD168) is not an ideal target antigen for immunotherapy of acute myeloid leukemia. Haematologica 2012; 97:1539-47. [PMID: 22532518 DOI: 10.3324/haematol.2012.065581] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Criteria for good candidate antigens for immunotherapy of acute myeloid leukemia are high expression on leukemic stem cells in the majority of patients with acute myeloid leukemia and low or no expression in vital tissues. It was shown in vaccination trials that Receptor for Hyaluronic Acid Mediated Motility (RHAMM/HMMR) generates cellular immune responses in patients with acute myeloid leukemia and that these responses correlate with clinical benefit. It is not clear however whether this response actually targets the leukemic stem cell, especially since it was reported that RHAMM is expressed maximally during the G2/M phase of the cell cycle. In addition, tumor specificity of RHAMM expression remains relatively unexplored. DESIGN AND METHODS Blood, leukapheresis and bone marrow samples were collected from both acute myeloid leukemia patients and healthy controls. RHAMM expression was assessed at protein and mRNA levels on various sorted populations, either fresh or after manipulation. RESULTS High levels of RHAMM were expressed by CD34(+)CD38(+) and CD34(-) acute myeloid leukemia blasts. However, only baseline expression of RHAMM was measured in CD34(+)CD38(-) leukemic stem cells, and was not different from that in CD34(+)CD38(-) hematopoietic stem cells from healthy controls. RHAMM was significantly up-regulated in CD34(+) cells from healthy donors during in vitro expansion and during in vivo engraftment. Finally, we demonstrated an explicit increase in the expression level of RHAMM after in vitro activation of T cells. CONCLUSIONS RHAMM does not fulfill the criteria of an ideal target antigen for immunotherapy of acute myeloid leukemia. RHAMM expression in leukemic stem cells does not differ significantly from the expression in hematopoietic stem cells from healthy controls. RHAMM expression in proliferating CD34+ cells of healthy donors and activated T cells further compromises RHAMM-specific T-cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Sylvia Snauwaert
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wang D, Zhang L, Li Y, Wang H, Xiao Q, Cao W, Feng W. Construction and expression of humanized chimeric T cell receptor specific for chronic myeloid leukemia cells. Biotechnol Lett 2012; 34:1193-201. [PMID: 22447097 DOI: 10.1007/s10529-012-0896-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 02/23/2012] [Indexed: 10/28/2022]
Abstract
Chimeric T cell receptors (chTCRs), composed of the single-chain variable fragments (scFv) of murine antibodies and human signaling molecules, are used to redirect the specificity of autologous or allogeneic T lymphocytes. To develop novel therapeutic agents for treatment of chronic myeloid leukemia (CML), we engineered a scFv from the hybridoma cell line CMA1 which produces monoclonal antibody specific against CML. The genes encoding the heavy and light chain variable regions were amplified from CMA1 cDNA and a humanized chTCR was constructed. Expression of the novel hchTCR was verified in NIH3T3 cells transduced with retroviral vectors. The results demonstrated that hchTCR can be expressed and presented on cell surface normally. These results suggest that retroviral vectors expressing hchTCR specific for CML cells may be used to redirect human T lymphocytes.
Collapse
Affiliation(s)
- Dong Wang
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
19
|
Identification of a novel peptide derived from the M-phase phosphoprotein 11 (MPP11) leukemic antigen recognized by human CD8+ cytotoxic T lymphocytes. Hematol Oncol Stem Cell Ther 2012; 3:24-33. [PMID: 20231810 DOI: 10.1016/s1658-3876(10)50053-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND AND OBJECTIVES There is an urgent need for the development of leukemia-targeted immunotherapeutic approaches using defined leukemia-associated antigens that are preferentially expressed by most leukemia subtypes and absent or minimally expressed in vital tissues. M-phase phosphoprotein 11 protein (MPP11) is extensively overexpressed in leukemic cells and therefore is considered an attractive target for leukemia T cell therapy. We sought to identify potential CD8+ cytotoxic T lymphocytes that specifically recognised peptides derived from the MPP11 antigen. METHODS A computer-based epitope prediction program SYFPEITHI, was used to predict peptides from the MPP11 protein that bind to the most common HLA- A*0201 molecule. Peptide binding capacity to the HLA-A*0201 molecule was measured using the T2 TAP-deficient, HLA-A*0201-positive cell line. Dendritic cells were pulsed with peptides and then used to generate CD8+ cytotoxic T lymphocytes (CTL). The CML leukemic cell line K562-A2.1 naturally expressing the MPP11 antigen and engineered to express the HLA-A*0201 molecule was used as the target cell. RESULTS We have identified a potential HLA-A*0201 binding epitope (STLCQVEPV) named MPP-4 derived from the MPP11 protein which was used to generate a CTL line. Interestingly, this CTL line specifically recognized peptide-loaded target cells in both ELISPOT and cytotoxic assays. Importantly, this CTL line exerted a cytotoxic effect towards the CML leukemic cell line K562-A2.1. CONCLUSION This is the first study to describe a novel epitope derived from the MPP11 antigen that has been recognized by human CD8+ CTL.
Collapse
|
20
|
Smahel M. Antigens in chronic myeloid leukemia: implications for vaccine development. Cancer Immunol Immunother 2011; 60:1655-68. [PMID: 22033582 PMCID: PMC11028763 DOI: 10.1007/s00262-011-1126-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 10/06/2011] [Indexed: 12/16/2022]
Abstract
Treatment with imatinib mesylate and other tyrosine kinase inhibitors (TKI) revolutionized the therapy of chronic myeloid leukemia (CML). However, it alone does not cure this disease. Moreover, some patients develop resistance or adverse effects to this therapy. As successful treatment of a portion of CML patients by hematopoietic stem cell transplantation (HSCT) suggests the importance of immune mechanisms in the elimination of leukemic cells, including leukemia stem cells, TKI administration or HSCT might be combined with vaccination to cure CML patients. However, antigens implicated in the immune responses have not yet been sufficiently identified. Therefore, in this report, we compiled and characterized a list of 165 antigens associated with CML (CML-Ag165) and analyzed the expression of the corresponding genes in CML phases, subpopulations of leukemic cells, and CML-derived cell lines using available datasets from microarray transcriptional-profiling studies. From the CML-Ag165 list, we selected antigens most suitable for vaccine development and evaluated their appropriate characteristics.
Collapse
Affiliation(s)
- Michal Smahel
- Laboratory of Molecular Oncology, Department of Experimental Virology, Institute of Hematology and Blood Transfusion, U Nemocnice 1, Prague 2, Czech Republic.
| |
Collapse
|
21
|
Alatrash G, Molldrem JJ. Vaccines as consolidation therapy for myeloid leukemia. Expert Rev Hematol 2011; 4:37-50. [PMID: 21322777 DOI: 10.1586/ehm.10.80] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immunotherapy for myeloid leukemias remains a cornerstone in the management of this highly aggressive group of malignancies. Allogeneic (allo) stem cell transplantation (SCT), which can be curative in acute and chronic myeloid leukemias, exemplifies the success of immunotherapy for cancer management. However, because of its nonspecific immune response against normal tissue, allo-SCT is associated with high rates of morbidity and mortality, secondary to graft-versus-host disease, which can occur in up to 50% of allo-SCT recipients. Targeted immunotherapy using leukemia vaccines has been heavily investigated, as these vaccines elicit specific immune responses against leukemia cells while sparing normal tissue. Peptide and cellular vaccines have been developed against tumor-specific and leukemia-associated self-antigens. Although not yet considered the standard of care, leukemia vaccines continue to show promising results in the management of the myeloid leukemias.
Collapse
Affiliation(s)
- Gheath Alatrash
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 900, Houston, TX 77030, USA
| | | |
Collapse
|
22
|
Westers TM, van den Ancker W, Bontkes HJ, Janssen JJWM, van de Loosdrecht AA, Ossenkoppele GJ. Chronic myeloid leukemia lysate-loaded dendritic cells induce T-cell responses towards leukemia progenitor cells. Immunotherapy 2011; 3:569-76. [DOI: 10.2217/imt.11.3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Treatment of chronic myeloid leukemia with tyrosine kinase inhibitors, such as imatinib mesylate, dasatinib and nilotinib, results in high rates of cytogenetic and molecular responses. However, in many patients, minimal residual disease is detected by molecular techniques. Since chronic myeloid leukemia cells are particularly good targets for immune surveillance mechanisms, we explored active specific immunotherapy using leukemia lysate-loaded dendritic cells in vitro. Our data show the potency of dendritic cell-based vaccination strategies for the induction of T cell-mediated responses to eradicate minimal residual disease.
Collapse
Affiliation(s)
| | - Willemijn van den Ancker
- Department of Hematology, Cancer Center Amsterdam/VUmc Institute for Cancer & Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Hetty J Bontkes
- Department of Hematology, Cancer Center Amsterdam/VUmc Institute for Cancer & Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jeroen JWM Janssen
- Department of Hematology, Cancer Center Amsterdam/VUmc Institute for Cancer & Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Arjan A van de Loosdrecht
- Department of Hematology, Cancer Center Amsterdam/VUmc Institute for Cancer & Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Gert J Ossenkoppele
- Department of Hematology, Cancer Center Amsterdam/VUmc Institute for Cancer & Immunology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Vonka V. Immunotherapy of chronic myeloid leukemia: present state and future prospects. Immunotherapy 2010; 2:227-41. [PMID: 20635930 DOI: 10.2217/imt.10.2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In spite of the considerable successes that have been achieved in the treatment of chronic myeloid leukemia (CML), cure for the disease can only be obtained by the present means in a rather small minority of patients. During the past decade, considerable progress has been made in the understanding of the immunology of CML, which has raised hopes that this disease may be curable by supplementing the current targeted chemotherapy with immunotherapeutic approaches. More than ten small-scale clinical trials have been carried out with experimental vaccines predominantly based on the p210bcr-abl fusion protein. Their results suggested beneficial effects in some patients. Recent data obtained in human patients as well as in animal models indicate that the p210bcr-abl protein does not carry the immunodominant epitope(s). These observations, combined with the recognition of an ever increasing number of other immunogenic proteins in CML cells, strongly support the concept that gene-modified, cell-based vaccines containing the full spectrum of tumor antigens might be the most effective immunotherapeutic approach. Recently created mathematical models have provided important leads for the timing of the combination of targeted drug therapy with vaccine administration. A strategy of how targeted drug therapy might be combined with vaccination is outlined.
Collapse
Affiliation(s)
- Vladimír Vonka
- Department of Experimental Virology, Institutute of Hematology & Blood Transfusion, Prague, Czech Republic.
| |
Collapse
|
24
|
Hickey MJ, Malone CC, Erickson KL, Jadus MR, Prins RM, Liau LM, Kruse CA. Cellular and vaccine therapeutic approaches for gliomas. J Transl Med 2010; 8:100. [PMID: 20946667 PMCID: PMC2964608 DOI: 10.1186/1479-5876-8-100] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Accepted: 10/14/2010] [Indexed: 12/25/2022] Open
Abstract
Despite new additions to the standard of care therapy for high grade primary malignant brain tumors, the prognosis for patients with this disease is still poor. A small contingent of clinical researchers are focusing their efforts on testing the safety, feasibility and efficacy of experimental active and passive immunotherapy approaches for gliomas and are primarily conducting Phase I and II clinical trials. Few trials have advanced to the Phase III arena. Here we provide an overview of the cellular therapies and vaccine trials currently open for patient accrual obtained from a search of http://www.clinicaltrials.gov. The search was refined with terms that would identify the Phase I, II and III immunotherapy trials open for adult glioma patient accrual in the United States. From the list, those that are currently open for patient accrual are discussed in this review. A variety of adoptive immunotherapy trials using ex vivo activated effector cell preparations, cell-based and non-cell-based vaccines, and several combination passive and active immunotherapy approaches are discussed.
Collapse
Affiliation(s)
- Michelle J Hickey
- The Joan S, Holmes Memorial Biotherapeutics Research Laboratory, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Naugler C, Liwski R. Human leukocyte antigen class I alleles and the risk of chronic myelogenous leukemia: a meta-analysis. Leuk Lymphoma 2010; 51:1288-92. [DOI: 10.3109/10428191003802340] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Kanojia D, Garg M, Saini S, Agarwal S, Kumar R, Suri A. Sperm associated antigen 9 expression and humoral response in chronic myeloid leukemia. Leuk Res 2010; 34:858-63. [PMID: 20138665 DOI: 10.1016/j.leukres.2010.01.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 12/12/2009] [Accepted: 01/15/2010] [Indexed: 02/08/2023]
Abstract
Early diagnosis and cure for patients with chronic myeloid leukemia (CML) exhibit significant clinical challenges because of the disease progression from chronic phase (CP) into a rapidly fatal blast crisis. Our earlier studies suggested an association of sperm associated antigen 9 (SPAG9) with various human malignancies. The present investigation revealed that SPAG9 mRNA and protein are expressed in CML patients (88%), in K562 and KCL-22 cells. Further, SPAG9 protein expression was also detected on cell surface suggesting that this molecule may be a suitable target for immunotherapy. Interestingly, 90% CML-CP patients showed humoral response against SPAG9, suggesting its important role in early diagnostic of CML-CP. Further investigation is warranted in establishing the potential of SPAG9 as a biomarker and immunotherapeutic target for early treatment of CML-CP patients.
Collapse
Affiliation(s)
- Deepika Kanojia
- Cancer Microarray, Genes and Proteins Laboratory, National Institute of Immunology, New Delhi, India
| | | | | | | | | | | |
Collapse
|
27
|
Naugler C, Liwski R. HLA risk markers for chronic myelogenous leukemia in Eastern Canada. Leuk Lymphoma 2009; 50:254-9. [DOI: 10.1080/10428190802668873] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
28
|
Pinilla-Ibarz J, Shah B, Dubovsky JA. The biological basis for immunotherapy in patients with chronic myelogenous leukemia. Cancer Control 2009; 16:141-52. [PMID: 19337200 DOI: 10.1177/107327480901600206] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Chronic myelogenous leukemia (CML) has long been recognized as an entity responsive to immunotherapeutic interventions. Despite the success of the tyrosine kinase inhibitors (TKIs) in this disease, CML remains incurable. Only allogeneic bone marrow transplantation can provide long-term eradication of CML. METHODS This review summarizes the recent advances in the field of immunology in CML, specifically in tumor antigen discovery, that have been incorporated into the design of new clinical trials. RESULTS Multiple vaccine approaches are currently under clinical investigation. Recent laboratory and clinical data also point to a unique interaction of TKIs with the immune system. CONCLUSIONS A better understanding of these interactions combined with advances in the field of immunotherapy will likely lead to incorporation of TKIs in future therapeutic interventions to develop a cure for this disease.
Collapse
Affiliation(s)
- Javier Pinilla-Ibarz
- Department of Malignant Hematology at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, USA.
| | | | | |
Collapse
|
29
|
The candidate immunotherapeutical target, the receptor for hyaluronic acid-mediated motility, is associated with proliferation and shows prognostic value in B-cell chronic lymphocytic leukemia. Leukemia 2008; 23:519-27. [DOI: 10.1038/leu.2008.338] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
30
|
Ex vivo characterization of polyclonal memory CD8+ T-cell responses to PRAME-specific peptides in patients with acute lymphoblastic leukemia and acute and chronic myeloid leukemia. Blood 2008; 113:2245-55. [PMID: 18988867 DOI: 10.1182/blood-2008-03-144071] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Preferentially expressed antigen of melanoma (PRAME) is aberrantly expressed in hematologic malignancies and may be a useful target for immunotherapy in leukemia. To determine whether PRAME is naturally immunogenic, we studied CD8(+) T-cell responses to 4 HLA-A*0201-restricted PRAME-derived epitopes (PRA100, PRA142, PRA300, PRA425) in HLA-A*0201-positive patients with acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), and healthy donors. CD8(+) T cells recognizing PRAME peptides could be detected ex vivo in 4 of 10 ALL, 6 of 10 AML, 3 of 10 CML patients, and 3 of 10 donors by HLA-A2 tetramer analysis and flow cytometry for intracellular interferon-gamma. The frequency of PRAME-specific CD8(+) T cells was greater in patients with AML, CML, and ALL than healthy controls. All peptides were immunogenic in patients, while responses were only detected to PRA300 in donors. High PRAME expression in patient peripheral blood mononuclear cells was associated with responses to greater than or equal to 2 PRAME epitopes compared with low PRAME expression levels (4/7 vs 0/23, P = .001), suggesting a PRAME-driven T-cell response. PRAME-specific T cells were readily expanded in short-term cultures in donors and patients. These results provide evidence for spontaneous T cell reactivity against multiple epitopes of PRAME in ALL, AML, and CML. The potential for developing PRAME as a target for immunotherapy in leukemia deserves further exploration.
Collapse
|
31
|
Dasatinib exerts an immunosuppressive effect on CD8+ T cells specific for viral and leukemia antigens. Exp Hematol 2008; 36:1297-308. [DOI: 10.1016/j.exphem.2008.05.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 05/05/2008] [Accepted: 05/09/2008] [Indexed: 11/18/2022]
|
32
|
Identification of NM23-H2 as a tumour-associated antigen in chronic myeloid leukaemia. Leukemia 2008; 22:1542-50. [DOI: 10.1038/leu.2008.107] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
33
|
Greiner J, Schmitt M. Leukemia-associated antigens as target structures for a specific immunotherapy in chronic myeloid leukemia. Eur J Haematol 2008; 80:461-8. [DOI: 10.1111/j.1600-0609.2008.01053.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
34
|
RHAMM-R3 peptide vaccination in patients with acute myeloid leukemia, myelodysplastic syndrome, and multiple myeloma elicits immunologic and clinical responses. Blood 2007; 111:1357-65. [PMID: 17978170 DOI: 10.1182/blood-2007-07-099366] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The receptor for hyaluronic acid-mediated motility (RHAMM) is an antigen eliciting both humoral and cellular immune responses in patients with acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), and multiple myeloma (MM). We initiated a phase 1 clinical trial vaccinating 10 patients with R3 (ILSLELMKL), a highly immunogenic CD8(+) T-cell epitope peptide derived from RHAMM. In 7 of 10 patients, we detected an increase of CD8(+)/HLA-A2/RHAMM R3 tetramer(+)/CD45RA(+)/CCR7(-)/CD27(-)/CD28(-) effector T cells in accordance with an increase of R3-specific CD8(+) T cells in enzyme linked immunospot (ELISpot) assays. In chromium release assays, a specific lysis of RHAMM-positive leukemic blasts was shown. Three of 6 patients with myeloid disorders (1/3 AML, 2/3 MDS) achieved clinical responses: one patient with AML and one with MDS showed a significant reduction of blasts in the bone marrow after the last vaccination. One patient with MDS no longer needed erythrocyte transfusions after 4 vaccinations. Two of 4 patients with MM showed a reduction of free light chain serum levels. Taken together, RHAMM-R3 peptide vaccination induced both immunologic and clinical responses, and therefore RHAMM constitutes a promising target for further immunotherapeutic approaches. This study is registered at http://ISRCTN.org as ISRCTN32763606 and is registered with EudraCT as 2005-001706-37.
Collapse
|
35
|
Brauer KM, Werth D, von Schwarzenberg K, Bringmann A, Kanz L, Grünebach F, Brossart P. BCR-ABL Activity Is Critical for the Immunogenicity of Chronic Myelogenous Leukemia Cells. Cancer Res 2007; 67:5489-97. [PMID: 17545631 DOI: 10.1158/0008-5472.can-07-0302] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chronic myelogenous leukemia (CML) is a myeloproliferative disorder caused by excessive granulopoiesis due to the formation of the constitutively active tyrosine kinase BCR-ABL. An effective drug against CML is imatinib mesylate, a tyrosine kinase inhibitor acting on Abl kinases, c-KIT, and platelet-derived growth factor receptor. Recently, a study revealed that patients treated with imatinib showed impaired CTL responses compared with patients treated with IFN-alpha, which might be due to a treatment-induced reduction in immunogenicity of CML cells or immunosuppressive effects. In our study, we found that inhibition of BCR-ABL leads to a down-regulation of immunogenic antigens on the CML cells in response to imatinib treatment, which results in the inhibition of CML-directed immune responses. By treating CML cells with imatinib, we could show that the resulting inhibition of BCR-ABL leads to a decreased expression of tumor antigens, including survivin, adipophilin, hTERT, WT-1, Bcl-x(L), and Bcl-2 in correlation to a decreased development of CML-specific CTLs. In contrast, this reduction in immunogenicity was not observed when a CML cell line resistant to the inhibitory effects of imatinib was used, but could be confirmed by transfection with specific small interfering RNA against BCR-ABL or imatinib treatment of primary CML cells.
Collapse
MESH Headings
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/immunology
- Antineoplastic Agents/pharmacology
- Benzamides
- Dendritic Cells/immunology
- Down-Regulation
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/immunology
- Humans
- Imatinib Mesylate
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Lymphocyte Activation/drug effects
- Piperazines/pharmacology
- Pyrimidines/pharmacology
- RNA, Small Interfering/genetics
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Katharina M Brauer
- Department of Hematology, Oncology, Immunology, Rheumatology, and Pulmonology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
36
|
Roman-Gomez J, Jimenez-Velasco A, Agirre X, Castillejo JA, Navarro G, Jose-Eneriz ES, Garate L, Cordeu L, Cervantes F, Prosper F, Heiniger A, Torres A. Epigenetic regulation of PRAME gene in chronic myeloid leukemia. Leuk Res 2007; 31:1521-8. [PMID: 17382387 DOI: 10.1016/j.leukres.2007.02.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2006] [Revised: 02/15/2007] [Accepted: 02/24/2007] [Indexed: 01/14/2023]
Abstract
Tumor associated antigens (TAA) provide attractive targets for cancer-specific immunotherapy. PRAME is a TAA gene up-regulated in advanced phases of chronic myeloid leukemia (CML). To date, molecular mechanisms for the expression of PRAME have never been studied. We found that some Ph'-positive cell lines did not express PRAME. The expression of PRAME was restored in these cell lines by treatment with 5'-aza-2'-deoxycytidine, suggesting that the expression of PRAME is mainly suppressed by hypermethylation. Bisulfite sequencing analysis of the CpG sites of the PRAME exon 2 in these cancer cell lines revealed a close relationship between the methylation status of the PRAME gene and its expression. A methylation-specific PCR analysis demonstrated that hypomethylation of PRAME was significantly more frequent in CML blast crisis (70%) than in chronic phase (36%) (P=0.01) and was correlated with high expression levels of PRAME transcripts (P<0.0001). These results suggest that hypomethylation of PRAME up-regulates its expression in CML and might play a significant role in the progression of the disease.
Collapse
Affiliation(s)
- Jose Roman-Gomez
- Hematology Department, Reina Sofia Hospital, Avda, Menendez Pidal s/n, 14004, Cordoba, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|