1
|
Tanriver U, Geppert M, Filges I, Menter T, Schmugge M, Güngör T, Gengler C, Strahm B, Erlacher M, Diesch-Furlanetto T, von der Weid N, Schifferli A. Managing High-Risk PTPN11-Mutated Juvenile Myelomonocytic Leukemia in a Patient with SETBP1 Mosaicism. Pediatr Blood Cancer 2025; 72:e31491. [PMID: 39690457 DOI: 10.1002/pbc.31491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/19/2024]
Affiliation(s)
- Ursula Tanriver
- Pediatric Hematology-Oncology Department, University Children's Hospital of Basel, Basel, Switzerland
| | - Mario Geppert
- Pediatric Hematology-Oncology Department, University Children's Hospital of Basel, Basel, Switzerland
| | - Isabel Filges
- Medical Genetics, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Thomas Menter
- Medical Genetics, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Markus Schmugge
- Division of Pediatric Hematology, University Children's Hospital of Zurich, Zurich, Switzerland
| | - Tayfun Güngör
- Division of Stem Cell Transplantation and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Carole Gengler
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Brigitte Strahm
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Miriam Erlacher
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Tamara Diesch-Furlanetto
- Pediatric Hematology-Oncology Department, University Children's Hospital of Basel, Basel, Switzerland
| | - Nicolas von der Weid
- Pediatric Hematology-Oncology Department, University Children's Hospital of Basel, Basel, Switzerland
| | - Alexandra Schifferli
- Pediatric Hematology-Oncology Department, University Children's Hospital of Basel, Basel, Switzerland
| |
Collapse
|
2
|
Peng Z, Gao J, Huang L, He Y, Tang H, Zong S, Pei Y, Pei F, Ge J, Liu X, Yue L, Zhou J, Li X, Yue D, Chen Y, Chen C, Wu X, Feng X, Li C. Decitabine-based treatment strategy improved the outcome of HSCT in JMML: a retrospective cohort study. Front Immunol 2024; 15:1426640. [PMID: 39253078 PMCID: PMC11381242 DOI: 10.3389/fimmu.2024.1426640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/01/2024] [Indexed: 09/11/2024] Open
Abstract
Introduction Pre-HSCT disease control, suboptimal long-term prognosis, and a high recurrence incidence (RI) continue to pose significant challenges for hematopoietic stem cell transplantation (HSCT) in juvenile myelomonocytic leukemia (JMML) patients. Methods This retrospective cohort study assessed the effectiveness of a decitabine (DAC)-based protocol in JMML patients undergoing HSCT. The pre-HSCT treatment includes initial and bridging treatment. The efficacy of DAC monotherapy versus DAC combined with cytotoxic chemotherapy(C-DAC) as initial treatment was compared, followed by DAC plus FLAG (fludarabine, cytarabine, and GCSF) as bridging treatment. The HSCT regimens were based on DAC, fludarabine, and busulfan. Post-HSCT, low-dose DAC was used as maintenance therapy. The study endpoints focused on pretransplantation simplified clinical response and post-HSCT survival. Results There were 109 patients, including 45 receiving DAC monotherapy and 64 undergoing C-DAC treatment. 106 patients completed bridging treatment. All patients were administered planned HSCT regimens and post-HSCT treatment. The initial treatment resulted in 88.1% of patients achieving clinical remission without a significant difference between the DAC and C-DAC groups (p=0.769). Clinical remission rates significantly improved following bridging treatment (p=0.019). The 5-year overall survival, leukemia-free survival, and RI were 92.2%, 88.4%, and 8.0%, respectively. A poor clinical response to pre-HSCT treatment emerged as a risk factor for OS (hazard ratio: 9.8, 95% CI: 2.3-41.1, p=0.002). Conclusion Implementing a DAC-based administration strategy throughout the pre-HSCT period, during HSCT regimens, and in post-HSCT maintenance significantly reduced relapse and improved survival in JMML patients. Both DAC monotherapy and the DAC plus FLAG protocol proved effective as pre-HSCT treatments.
Collapse
Affiliation(s)
- Zhiyong Peng
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Jingyu Gao
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Litao Huang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Yuelin He
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Haoran Tang
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Sa Zong
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Yanru Pei
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Fuyu Pei
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Ge
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Liu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Yue
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Jun Zhou
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Xia Li
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Dan Yue
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Yun Chen
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Chen Chen
- Department of Biostatistics, Gobroad Research Center, Shanghai, China
| | - Xuedong Wu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoqin Feng
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunfu Li
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| |
Collapse
|
3
|
Hayatigolkhatmi K, Valzelli R, El Menna O, Minucci S. Epigenetic alterations in AML: Deregulated functions leading to new therapeutic options. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 387:27-75. [PMID: 39179348 DOI: 10.1016/bs.ircmb.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
Acute myeloid leukemia (AML) results in disruption of the hematopoietic differentiation process. Crucial progress has been made, and new therapeutic strategies for AML have been developed. Induction chemotherapy, however, remains the main option for the majority of AML patients. Epigenetic dysregulation plays a central role in AML pathogenesis, supporting leukemogenesis and maintenance of leukemic stem cells. Here, we provide an overview of the intricate interplay of altered epigenetic mechanisms, including DNA methylation, histone modifications, and chromatin remodeling, in AML development. We explore the role of epigenetic regulators, such as DNMTs, HMTs, KDMs, and HDACs, in mediating gene expression patterns pushing towards leukemic cell transformation. Additionally, we discuss the impact of cytogenetic lesions on epigenomic remodeling and the potential of targeting epigenetic vulnerabilities as a therapeutic strategy. Understanding the epigenetic landscape of AML offers insights into novel therapeutic avenues, including epigenetic modifiers and particularly their use in combination therapies, to improve treatment outcomes and overcome drug resistance.
Collapse
Affiliation(s)
- Kourosh Hayatigolkhatmi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy.
| | - Riccardo Valzelli
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Oualid El Menna
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy; Department of Hemato-Oncology, Università Statale di Milano, Milan, Italy.
| |
Collapse
|
4
|
Liu J, Huang B, Ding F, Li Y. Environment factors, DNA methylation, and cancer. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2023; 45:7543-7568. [PMID: 37715840 DOI: 10.1007/s10653-023-01749-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023]
Abstract
Today, the rapid development of science and technology and the rapid change in economy and society are changing the way of life of human beings and affecting the natural, living, working, and internal environment on which human beings depend. At the same time, the global incidence of cancer has increased significantly yearly, and cancer has become the number one killer that threatens human health. Studies have shown that diet, living habits, residential environment, mental and psychological factors, intestinal flora, genetics, social factors, and viral and non-viral infections are closely related to human cancer. However, the molecular mechanisms of the environment and cancer development remain to be further explored. In recent years, DNA methylation has become a key hub and bridge for environmental and cancer research. Some environmental factors can alter the hyper/hypomethylation of human cancer suppressor gene promoters, proto-oncogene promoters, and the whole genome, causing low/high expression or gene mutation of related genes, thereby exerting oncogenic or anticancer effects. It is expected to develop early warning markers of cancer environment based on DNA methylation, thereby providing new methods for early detection of cancers, diagnosis, and targeted therapy. This review systematically expounds on the internal mechanism of environmental factors affecting cancer by changing DNA methylation, aiming to help establish the concept of cancer prevention and improve people's health.
Collapse
Affiliation(s)
- Jie Liu
- Department of General Surgery, Second Hospital of Lanzhou University, Lan Zhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lan Zhou, China
| | - Binjie Huang
- Department of General Surgery, Second Hospital of Lanzhou University, Lan Zhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lan Zhou, China
| | - Feifei Ding
- Department of General Surgery, Second Hospital of Lanzhou University, Lan Zhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lan Zhou, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lan Zhou, China.
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lan Zhou, China.
| |
Collapse
|
5
|
Goyal A, Bauer J, Hey J, Papageorgiou DN, Stepanova E, Daskalakis M, Scheid J, Dubbelaar M, Klimovich B, Schwarz D, Märklin M, Roerden M, Lin YY, Ma T, Mücke O, Rammensee HG, Lübbert M, Loayza-Puch F, Krijgsveld J, Walz JS, Plass C. DNMT and HDAC inhibition induces immunogenic neoantigens from human endogenous retroviral element-derived transcripts. Nat Commun 2023; 14:6731. [PMID: 37872136 PMCID: PMC10593957 DOI: 10.1038/s41467-023-42417-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/11/2023] [Indexed: 10/25/2023] Open
Abstract
Immunotherapies targeting cancer-specific neoantigens have revolutionized the treatment of cancer patients. Recent evidence suggests that epigenetic therapies synergize with immunotherapies, mediated by the de-repression of endogenous retroviral element (ERV)-encoded promoters, and the initiation of transcription. Here, we use deep RNA sequencing from cancer cell lines treated with DNA methyltransferase inhibitor (DNMTi) and/or Histone deacetylase inhibitor (HDACi), to assemble a de novo transcriptome and identify several thousand ERV-derived, treatment-induced novel polyadenylated transcripts (TINPATs). Using immunopeptidomics, we demonstrate the human leukocyte antigen (HLA) presentation of 45 spectra-validated treatment-induced neopeptides (t-neopeptides) arising from TINPATs. We illustrate the potential of the identified t-neopeptides to elicit a T-cell response to effectively target cancer cells. We further verify the presence of t-neopeptides in AML patient samples after in vivo treatment with the DNMT inhibitor Decitabine. Our findings highlight the potential of ERV-derived neoantigens in epigenetic and immune therapies.
Collapse
Affiliation(s)
- Ashish Goyal
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jens Bauer
- Department of Peptide-based Immunotherapy, University of Tübingen and University Hospital Tübingen, Tübingen, Germany
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Joschka Hey
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German-Israeli Helmholtz Research School in Cancer Biology, Heidelberg, Germany
- German Center for Lung Research, (DZL) partner site Heidelberg, Heidelberg, Germany
| | - Dimitris N Papageorgiou
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg University, Medical Faculty, Heidelberg, Germany
| | - Ekaterina Stepanova
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Daskalakis
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Jonas Scheid
- Department of Peptide-based Immunotherapy, University of Tübingen and University Hospital Tübingen, Tübingen, Germany
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Marissa Dubbelaar
- Department of Peptide-based Immunotherapy, University of Tübingen and University Hospital Tübingen, Tübingen, Germany
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Boris Klimovich
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Dominic Schwarz
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Märklin
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Malte Roerden
- Department of Peptide-based Immunotherapy, University of Tübingen and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Yu-Yu Lin
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tobias Ma
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Mücke
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Georg Rammensee
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Michael Lübbert
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Fabricio Loayza-Puch
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeroen Krijgsveld
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg University, Medical Faculty, Heidelberg, Germany
| | - Juliane S Walz
- Department of Peptide-based Immunotherapy, University of Tübingen and University Hospital Tübingen, Tübingen, Germany.
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.
| | - Christoph Plass
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Center for Lung Research, (DZL) partner site Heidelberg, Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
6
|
He C, Peng Z, Zhang D, Guo Y, Liang T, Zhao Y, Yu L, Zhang Q, Chang Z, Xiao Y, Li N, Xue H, Wu S, Zhao ZJ, Zhang C, Chen Y. Sunitinib selectively targets leukemogenic signaling of mutant SHP2 in juvenile myelomonocytic leukemia. Biochem Pharmacol 2023; 213:115588. [PMID: 37187274 DOI: 10.1016/j.bcp.2023.115588] [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: 02/23/2023] [Revised: 05/08/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Leukemogenic SHP2 mutations occur in 35% of patients with juvenile myelomonocytic leukemia (JMML), a hematopoietic malignancy with poor response to cytotoxic chemotherapy. Novel therapeutic strategies are urgently needed for patients with JMML. Previously, we established a novel cell model of JMML with HCD-57, a murine erythroleukemia cell line that depends on EPO for survival. SHP2-D61Y or -E76K drove the survival and proliferation of HCD-57 in absence of EPO. In this study, we identified sunitinib as a potent compound to inhibit SHP2-mutant cells by screening a kinase inhibitor library with our model. We used cell viability assay, colony formation assay, flow cytometry, immunoblotting, and a xenograft model to evaluate the effect of sunitinib against SHP2-mutant leukemia cells in vitro and in vivo. The treatment of sunitinib selectively induced apoptosis and cell cycle arrest in mutant SHP2-transformed HCD-57, but not parental cells. It also inhibited cell viability and colony formation of primary JMML cells with mutant SHP2, but not bone marrow mononuclear cells from healthy donors. Immunoblotting showed that the treatment of sunitinib blocked the aberrantly activated signals of mutant SHP2 with deceased phosphorylation levels of SHP2, ERK, and AKT. Furthermore, sunitinib effectively reduced tumor burdens of immune-deficient mice engrafted with mutant-SHP2 transformed HCD-57. Our data demonstrated that sunitinib selectively inhibited SHP2-mutant leukemia cells, which could serve as an effective therapeutic strategy for SHP2-mutant JMML in the future.
Collapse
Affiliation(s)
- Chunxiao He
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhiyong Peng
- Nanfang-Chunfu Children's Institute of Hematology, Taixin Hospital, Dongguan, Guangdong, China
| | - Dengyang Zhang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yao Guo
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Tianqi Liang
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yuming Zhao
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Liuting Yu
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Qi Zhang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhiguang Chang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yan Xiao
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Na Li
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Hongman Xue
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Shunjie Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China.
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
7
|
Yi ES, Kim SK, Ju HY, Lee JW, Cho B, Kim BK, Kang HJ, Baek HJ, Kook H, Yang EJ, Lim YT, Ahn WK, Hahn SM, Park SK, Yoo ES, Yoo KH. Allogeneic hematopoietic cell transplantation in patients with juvenile myelomonocytic leukemia in Korea: a report of the Korean Pediatric Hematology-Oncology Group. Bone Marrow Transplant 2023; 58:20-29. [PMID: 36167906 DOI: 10.1038/s41409-022-01826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/08/2022] [Accepted: 09/05/2022] [Indexed: 01/07/2023]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a life-threatening myeloproliferative neoplasm. This multicenter study evaluated the characteristics, outcomes, and prognostic factors of allogeneic hematopoietic cell transplantation (HCT) in recipients with JMML who were diagnosed between 2000 and 2019 in Korea. Sixty-eight patients were retrospectively enrolled-28 patients (41.2%) received HCT during 2000-2010 and 40 patients (58.8%) during 2011-2020. The proportion of familial mismatched donors increased from 3.6 to 37.5%. The most common conditioning therapy was changed from Busulfan/Cyclophosphamide-based to Busulfan/Fludarabine-based therapy. The 5-year probabilities of event-free survival (EFS) and overall survival (OS) were 52.6% and 62.3%, respectively. The 5-year incidence of transplant-related mortality was 30.1%. Multivariate analysis revealed that the proportion of hemoglobin F ≥ 40%, abnormal cytogenetics, and matched sibling donors were independent risk factors for a higher relapse rate. Patients whose donor chimerism was below 99% had a significantly higher relapse rate. Better OS and lower treatment-related mortality were observed in patients with chronic graft-versus-host disease (GVHD), whereas grade III or IV acute GVHD was associated with worse EFS. In conclusion, the number of transplant increased along with the increase in alternative donor transplants, nevertheless, similar results were maintained. Alternative donor transplantation should be encouraged.
Collapse
Affiliation(s)
- Eun Sang Yi
- Department of Pediatrics, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea.,Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seong Koo Kim
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Wook Lee
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bin Cho
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bo Kyung Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Cancer Research Institute, Wide River Institute of Immunology, Seoul National University Children's Hospital, Seoul, Korea
| | - Hyoung Jin Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Cancer Research Institute, Wide River Institute of Immunology, Seoul National University Children's Hospital, Seoul, Korea
| | - Hee Jo Baek
- Department of Pediatrics, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Hoon Kook
- Department of Pediatrics, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Eu Jeen Yang
- Department of Pediatrics, Pusan National University School of Medicine, Pusan National University Children's Hospital, Yangsan, Korea
| | - Young Tak Lim
- Department of Pediatrics, Pusan National University School of Medicine, Pusan National University Children's Hospital, Yangsan, Korea
| | - Won Kee Ahn
- Department of Pediatrics, Yonsei University College of Medicine, Severance Hospital, Seoul, Korea
| | - Seung Min Hahn
- Department of Pediatrics, Yonsei University College of Medicine, Severance Hospital, Seoul, Korea
| | - Sang Kyu Park
- Department of Pediatrics, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Eun Sun Yoo
- Department of Pediatrics, Ewha Womans University College of Medicine, Ewha Womans University Seoul Hospital, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea. .,Department of Health Science and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea. .,Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea.
| |
Collapse
|
8
|
Zhang T, Liu X, Fei H, Zhang L, Ma R, Shen Y, Pang A, Yang D, Chen X, Zhang R, Wei J, He Y, Jiang E, Han M, Feng S. Improved myeloablative conditioning regimen for allogeneic stem cell transplantation in adult patients with chronic myelomonocytic leukaemia. Br J Haematol 2023; 200:256-260. [PMID: 36367209 DOI: 10.1111/bjh.18527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022]
Affiliation(s)
- Tingting Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xin Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Hairong Fei
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lining Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Runzhi Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yuyan Shen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Aiming Pang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Donglin Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xin Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Rongli Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jialin Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yi He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Mingzhe Han
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
9
|
De Vos N, Hofmans M, Lammens T, De Wilde B, Van Roy N, De Moerloose B. Targeted therapy in juvenile myelomonocytic leukemia: Where are we now? Pediatr Blood Cancer 2022; 69:e29930. [PMID: 36094370 DOI: 10.1002/pbc.29930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/07/2022]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare and aggressive clonal neoplasm of early childhood, classified as an overlap myeloproliferative/myelodysplastic neoplasm by the World Health Organization. In 90% of the patients with JMML, typical initiating mutations in the canonical Ras pathway genes NF1, PTPN11, NRAS, KRAS, and CBL can be identified. Hematopoietic stem cell transplantation (HSCT) currently is the established standard of care in most patients, although long-term survival is still only 50-60%. Given the limited therapeutic options and the important morbidity and mortality associated with HSCT, new therapeutic approaches are urgently needed. Hyperactivation of the Ras pathway as disease mechanism in JMML lends itself to the use of targeted therapy. Targeted therapy could play an important role in the future treatment of patients with JMML. This review presents a comprehensive overview of targeted therapies already developed and evaluated in vitro and in vivo in patients with JMML.
Collapse
Affiliation(s)
- Nele De Vos
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University, Ghent, Belgium
| | - Mattias Hofmans
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Tim Lammens
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bram De Wilde
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Nadine Van Roy
- Cancer Research Institute Ghent, Ghent, Belgium.,Center for Medical Genetics Ghent, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Barbara De Moerloose
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
10
|
Meier R, Greve G, Zimmer D, Bresser H, Berberich B, Langova R, Stomper J, Rubarth A, Feuerbach L, Lipka DB, Hey J, Grüning B, Brors B, Duyster J, Plass C, Becker H, Lübbert M. The antileukemic activity of decitabine upon PML/RARA-negative AML blasts is supported by all-trans retinoic acid: in vitro and in vivo evidence for cooperation. Blood Cancer J 2022; 12:122. [PMID: 35995769 PMCID: PMC9395383 DOI: 10.1038/s41408-022-00715-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/03/2022] [Accepted: 07/29/2022] [Indexed: 12/02/2022] Open
Abstract
The prognosis of AML patients with adverse genetics, such as a complex, monosomal karyotype and TP53 lesions, is still dismal even with standard chemotherapy. DNA-hypomethylating agent monotherapy induces an encouraging response rate in these patients. When combined with decitabine (DAC), all-trans retinoic acid (ATRA) resulted in an improved response rate and longer overall survival in a randomized phase II trial (DECIDER; NCT00867672). The molecular mechanisms governing this in vivo synergism are unclear. We now demonstrate cooperative antileukemic effects of DAC and ATRA on AML cell lines U937 and MOLM-13. By RNA-sequencing, derepression of >1200 commonly regulated transcripts following the dual treatment was observed. Overall chromatin accessibility (interrogated by ATAC-seq) and, in particular, at motifs of retinoic acid response elements were affected by both single-agent DAC and ATRA, and enhanced by the dual treatment. Cooperativity regarding transcriptional induction and chromatin remodeling was demonstrated by interrogating the HIC1, CYP26A1, GBP4, and LYZ genes, in vivo gene derepression by expression studies on peripheral blood blasts from AML patients receiving DAC + ATRA. The two drugs also cooperated in derepression of transposable elements, more effectively in U937 (mutated TP53) than MOLM-13 (intact TP53), resulting in a “viral mimicry” response. In conclusion, we demonstrate that in vitro and in vivo, the antileukemic and gene-derepressive epigenetic activity of DAC is enhanced by ATRA.
Collapse
Affiliation(s)
- Ruth Meier
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gabriele Greve
- Institute of Genetic Epidemiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dennis Zimmer
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Helena Bresser
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bettina Berberich
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ralitsa Langova
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Bioscience, University of Heidelberg, Heidelberg, Germany
| | - Julia Stomper
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anne Rubarth
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lars Feuerbach
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel B Lipka
- Section Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center (DKFZ) & National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Faculty of Medicine, Otto-von-Guericke-University, Magdeburg, Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
| | - Joschka Hey
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Björn Grüning
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Freiburg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Justus Duyster
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Freiburg, Freiburg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heiko Becker
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Lübbert
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Freiburg, Freiburg, Germany.
| |
Collapse
|
11
|
Wang Y, Sun Y, Xie J, Hu J, Liu N, Chen J, Li B, Lan S, Niu J, Wang L, Qiao Z, Zhang Y, Ren J, Zhang B, Qian L, Tan Y, Dou L, Li Y, Hu L. Allogeneic haematopoietic stem cell transplantation with decitabine-containing preconditioning regimen in TP53-mutant myelodysplastic syndromes: A case study. Front Oncol 2022; 12:928324. [PMID: 35924157 PMCID: PMC9339648 DOI: 10.3389/fonc.2022.928324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/27/2022] [Indexed: 11/23/2022] Open
Abstract
Myelodysplastic syndrome (MDS) with TP53 mutations has a poor prognosis after transplantation, and novel therapeutic means are urgently needed. Decitabine (Dec) monotherapy has demonstrated improved overall response rates in MDS and acute myeloid leukaemia, although these responses were not durable. This study aimed to preliminary evaluate the efficacy of a Dec-containing allogeneic haematopoietic stem cell transplantation (allo-HSCT) preconditioning regimen in TP53-mutant MDS. Nine patients with TP53-mutant myelodysplastic syndromes received the decitabine-containing preconditioning regimen and subsequent myeloablative allo-HCT between April 2013 and September 2021 in different centres. At a median follow-up of 42 months (range, 5 to 61 months), the overall survival (OS) was 89% (8/9), progression-free survival (PFS) was 89% (8/9), and relapse incidence was 11.1%. The incidence of severe acute (grade III-IV) graft-versus-host disease (GVHD) was 22.2% (2/9) and that of chronic moderate-to-severe GVHD was 11.1% (1/9). The 1-year GVHD-free/relapse-free survival (GRFS) was 56% (5/9). In conclusion, we found real-world clinical data that supports the use of a Dec-containing preconditioning regimen before allo-HSCT for possible improved outcomes in TP53-mutant MDS patients; there is therefore an urgent call for an in-depth exploration of the involved mechanism to confirm these preliminary findings.
Collapse
Affiliation(s)
- Yuxin Wang
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yao Sun
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Jing Xie
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Jiangwei Hu
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Na Liu
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Jianlin Chen
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Botao Li
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Sanchun Lan
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Jingwen Niu
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Lei Wang
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Zhuoqing Qiao
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yu Zhang
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Jing Ren
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Bin Zhang
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Liren Qian
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yehui Tan
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Liping Dou
- Department of Hematology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yuhang Li
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Liangding Hu
- Senior Department of Hematology, The Fifth Medical Centre of Chinese People’s Liberation Army General Hospital, Beijing, China
| |
Collapse
|
12
|
Li J, Li J, Hu T, Cui X, Song Z, Shi X, Liu R. Response to trametinib of two pediatric myeloid malignancies cases harboring RAS mutation and monosomy 7. Leuk Lymphoma 2022; 63:2238-2242. [PMID: 35694895 DOI: 10.1080/10428194.2022.2067998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Juanjuan Li
- Department of Hematology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Junhui Li
- Department of Hematology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Tao Hu
- Department of Hematology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Xiaodai Cui
- Experiment Cencer, Capital Institute of Pediatrics, Beijing, China
| | - Zeliang Song
- Department of Hematology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Xiaodong Shi
- Department of Hematology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Rong Liu
- Department of Hematology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
13
|
Gao J, Hu Y, Gao L, Xiao P, Lu J, Hu S. The effect of decitabine-combined minimally myelosuppressive regimen bridged allo-HSCT on the outcomes of pediatric MDS from 10 years' experience of a single center. BMC Pediatr 2022; 22:312. [PMID: 35624441 PMCID: PMC9137053 DOI: 10.1186/s12887-022-03376-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 05/19/2022] [Indexed: 11/21/2022] Open
Abstract
Background Myelodysplastic syndrome (MDS) is a rare disease in children and the treatment option before the allogeneic hematopoietic stem cell transplantation (allo-HSCT) is rarely reported. Our main objective was to report our single-center experience with the DNA-hypomethylating agent, decitabine-combined minimally myelosuppressive regimen (DAC + MMR) bridged allo-HSCT in children with MDS. Methods Twenty-eight children with de novo MDS who underwent allo-HSCT between 2011 and 2020 were enrolled. Patients were divided into subgroups (refractory cytopenia of childhood [RCC] and advanced MDS [aMDS]) and treated by HSCT alone or pre-transplant combination treatment based on risk stratification. The patients’ clinical characteristics, treatment strategies and outcomes were retrospectively evaluated. Results Twenty patients with aMDS had received pre-transplant treatment (three were treated with decitabine alone, thirteen with DAC + MMR, and four with acute myeloid leukemia type [AML-type] induction therapy). DAC + MMR was well tolerated and the most common adverse events were myelosuppression and gastrointestinal reaction. DAC + MMR had shown an improved marrow complete remission (mCR) compared with AML-type chemotherapy (13/13, 100% versus 2/4, 50%, P = 0.044). The median follow-up for total cohort was 53.0 months (range, 2.3-127.0 months) and the 4-year overall survival (OS) was 71.4 ± 8.5%. In the subgroup of aMDS, pretreatment of DAC + MMR resulted in a much better survival rate than AML-type chemotherapy (84.6 ± 10.0% versus 0.0 ± 0.0%, P < 0.001). Conclusions The DAC + MMR bridged allo-HSCT may be recommended as a novel and effective approach. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-022-03376-1.
Collapse
Affiliation(s)
- Junyan Gao
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Pediatrics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Yixin Hu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Li Gao
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Peifang Xiao
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Lu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shaoyan Hu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
14
|
Pan T, Han S, Zhou M, Qi J, Wang H, Xu X, Li X, Yao Y, Han Y. Efficacy of azacitidine in preventing relapse after hematopoietic stem cell transplantation for advanced myeloid malignancies: a systematic review and meta-analysis. Expert Rev Hematol 2022; 15:457-464. [PMID: 35535679 DOI: 10.1080/17474086.2022.2073214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Relapse is the leading cause of death from myeloid malignancies after allogeneic hematopoietic stem cell transplantation (HSCT). Azacitidine has gained attention in recent years in the prophylaxis of relapsed refractory hematologic malignancies. This study evaluated the efficacy of AZA in preventing relapse after HSCT in patients with myeloid malignancies. METHODS A systematic review and meta-analysis of all available cohort studies were performed regarding the application of AZA for prophylaxis of relapse after HSCT for advanced MDS and AML. Databases were searched for relevant studies. Endpoints included 2-year relapse rate, survival, relapse-related mortality, as well as the incidence of graft-versus-host disease (GVHD). RESULTS A total of 444 patients from 13 studies were included in this analysis. The pooled estimate of the cumulative incidence of relapse after two years in enrolled patients was 25% (95% confidence interval [CI], 18%-33%). The pooled estimates of 2-year survival probabilities were 65% (95% CI, 50%-79%). The pooled cumulative incidence of relapse-related mortality was 28% (95% CI, 22%-34%). The pooled estimated incidence of acute and chronic GVHD, respectively, were 28% (95% CI, 22%-34%) and 38% (95% CI, 27%-49%). CONCLUSION AZA administration is efficacious for relapse prevention after HSCT in myeloid malignancies.
Collapse
Affiliation(s)
- Tingting Pan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, Jiangsu, China
| | - Shiyu Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Meng Zhou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Jiaqian Qi
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Hong Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Xiaoyan Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Xueqian Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Yifang Yao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Yue Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, Jiangsu, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
15
|
Comparative analysis of Decitabine intensified BUCY2 and BUCY2 conditioning regimen for high-risk MDS patients undergoing allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2022; 57:1063-1071. [PMID: 35459877 DOI: 10.1038/s41409-022-01645-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022]
Abstract
The optimal conditioning regimen for high-risk myelodysplastic syndrome (MDS) patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains elusive. This study aimed to explore the anti-leukemic efficacy and toxicity of Decitabine (Dec, 20 mg/m2/day, day -11 to -7) intensified BUCY2 vs. traditional regimen in high-risk MDS population. We retrospectively evaluated 93 consecutive high-risk MDS patients undergoing allo-HSCT in our institution, comparing discrepancies in clinical characteristics and outcomes between cases using Dec-intensified BUCY2 (n = 52) and traditional BUCY2 regimen (n = 41). Three-year cumulative incidence of relapse after Dec-intensified BUCY2 conditioning was remarkably lower than that of patients using BUCY2 regimen (20.2% vs. 39.0%, p = 0.034). Overall survival and disease-free survival at 3 years for Dec-intensified BUCY2 group were 70.2% and 64.9%, respectively, which were significantly improved when compared with BUCY2 group (51.1% and 43.9%, p = 0.031 and p = 0.027). Furthermore, overall survival and disease-free survival for MDS cases receiving cytoreduction therapy were dramatically better than patients in non-cytoreduction group (p = 0.041, p = 0.047). In summary, the Dec-intensified conditioning regimen could be effective and feasible, providing prominent recurrence control with moderate toxicity for high-risk MDS patients. These patients might also benefit from pre-transplant cytoreductive therapeutic schedules. Larger randomized controlled trials are still needed to further confirm these conclusions.
Collapse
|
16
|
Oliveira AF, Tansini A, Toledo T, Balceiro R, Lee MLM, Villela N, Ikeuty P, Metze K, Lopes LF, Lorand-Metze I. Immunophenotypic changes in juvenile myelomonocytic leukaemia after treatment with hypomethylating agent: Do they help to evaluate dept of response? Br J Haematol 2022; 197:339-348. [PMID: 35187646 DOI: 10.1111/bjh.18089] [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: 11/03/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 11/29/2022]
Abstract
5-Azacitidine has been used before stem cell transplantation in juvenile myelomonocytic leukaemia (JMML) patients. Recently, we have described immunophenotypic features in JMML at diagnosis. Here, our aim was to examine the changes in the immunophenotypic features during azacitidine treatment, correlating it with clinical response. Patients treated with 5-azacitidine were evaluated at diagnosis and after three and six cycles of medication. Among 32 patients entering the study, 28 patients were examined after three cycles and 25 patients after six. Patients showed a reduction in CD34/CD117+ cells: median 3.35% at diagnosis, 2.8% after three cycles and 1.63% after six. B-cell progenitors were decreased at diagnosis and decreased after treatment. Monocytes decreased: 11.91% to 6.4% and 4.18% respectively. Complete response was associated with increase in classical monocytes. T lymphocytes, reduced at diagnosis, increased in patients responding to 5-azacitidine. Immunophenotypic aberrancies including expression of CD7 in myeloid progenitors remained after treatment. This feature was associated with a worse response to treatment, as well as presence of NF1. Immunophenotyping was feasible in all patients. Clinical response was associated with a decrease of myeloid progenitors and monocytes and a rise in T lymphocytes although phenotypic aberrancies persisted. The largest effect was observed after three cycles.
Collapse
Affiliation(s)
- Anita Frisanco Oliveira
- Barretos Children´s Cancer Hospital, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Myeloproliferative Diseases Committee, Barretos, Brazil
| | - Aline Tansini
- Barretos Children´s Cancer Hospital, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Morfology and Flow Cytometry Committee, Barretos, Brazil
| | - Thais Toledo
- Barretos Children´s Cancer Hospital, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Morfology and Flow Cytometry Committee, Barretos, Brazil
| | | | - Maria Lucia Martino Lee
- Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Myeloproliferative Diseases Committee, Barretos, Brazil
| | - Neysimelia Villela
- Barretos Children´s Cancer Hospital, Hematopoietic Stem Cell Transplantation, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), HSCT Committee, Barretos, Brazil
| | - Patricia Ikeuty
- Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), HSCT Committee, Barretos, Brazil
| | - Konradin Metze
- Department of Pathology, Faculty Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Luiz Fernando Lopes
- Barretos Children´s Cancer Hospital, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Barretos, Brazil
| | - Irene Lorand-Metze
- Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Morfology and Flow Cytometry Committee, Barretos, Brazil.,Department of Internal Medicine, Faculty Medical Sciences, State University of Campinas, Campinas, Brazil
| |
Collapse
|
17
|
Cheng S, Xiao P, Wang J, Li Z, Gao L, Zheng J, Hu Y, Ding X, Ling J, Lu Q, Pan J, Li B, Lu J, Wang Y, Ribeiro RC, Hu S. Decitabine combined with minimally myelosuppressive therapy for induction of remission in pediatric high-risk acute myeloid leukemia with chromosome 5q deletion: a report of three cases. Int J Hematol 2022; 116:146-151. [PMID: 35181851 DOI: 10.1007/s12185-022-03309-9] [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: 07/14/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022]
Abstract
Cases of pediatric acute myeloid leukemia (AML) with complex karyotypes including chromosome 5 abnormalities are rare and have a very poor prognosis. Management of AML with monosomy 5/del(5q) has been inconsistent. We treated three adolescents with this AML subtype using combined low-dose cytarabine and mitoxantrone, concurrently with decitabine and G-CSF, for remission induction. Decitabine was also included in the conditioning regimen before hematopoietic cell transplantation (HCT). All three patients achieved complete remission after treatment with this combination therapy. The treatment was well tolerated, and the patients are alive and free of disease at 3.6, 3.2, and 3.0 years after HCT, respectively.
Collapse
Affiliation(s)
- Shengqin Cheng
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Peifang Xiao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Juxiang Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China.,Department of Hematology and Oncology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhiheng Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Li Gao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Jiajia Zheng
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Yixin Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Xin Ding
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Jing Ling
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Qin Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Jian Pan
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Bohan Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Jun Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Yi Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Raul C Ribeiro
- Department of Oncology, Division of Leukemia/Lymphoma, and Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Shaoyan Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China.
| |
Collapse
|
18
|
Fang K, Qi J, Zhou M, Zhang Z, Han Y. Clinical Characteristics, Prognosis, and Treatment Strategies of TP53 Mutations in Myelodysplastic Syndromes. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:224-235. [PMID: 34690091 DOI: 10.1016/j.clml.2021.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 11/28/2022]
Abstract
TP53 gene mutations are common in myelodysplastic syndromes (MDS). Previous studies have reported their detrimental effects on patient survival. However, current treatment strategies mainly based on hypomethylating agent therapy (HMA) and hematopoietic stem cell transplantation (HSCT) still leave a lot to be desired. And there is also a lack of studies on large sample with a view to the refinement of specific characteristics and disease progression. So we performed a meta-analysis including 20 studies compromising 5067 patients to assess the prognostic impact and clinical characteristics of TP53 mutations in MDS patients. The overall hazard ratio for overall survival (OS) was 2.14 (95% confidence interval 1.94-2.37, P < .00001) compared with patients with MDS without TP53 mutations. Lower progression-free survival and leukemia-free survival were associated with TP53 mutations. Subgroup analysis revealed that TP53 mutations were significantly associated with high levels of blast cells and karyotypic aberrations. And among Asian population, the adverse impact on OS of TP53 mutations seemed worse than those in Western countries. (HR 2.87 vs. 2.02, P = .01). In addition, TP53 mutations had no effect on response to HMA therapy, and HSCT improved OS in patients carrying TP53 mutations.
Collapse
Affiliation(s)
- Kun Fang
- National clinical research center for hematologic diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jiaqian Qi
- National clinical research center for hematologic diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China; Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Meng Zhou
- National clinical research center for hematologic diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China; Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Ziyan Zhang
- National clinical research center for hematologic diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yue Han
- National clinical research center for hematologic diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China; Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China.
| |
Collapse
|
19
|
Li Y, Cheng L, Xu C, Chen J, Hu J, Liu N, Lan S, Xie J, Sun T, Wang L, Zhang Y, Sun Y, Chen S, Hu L. A Retrospective Observation of Treatment Outcomes Using Decitabine-Combined Standard Conditioning Regimens Before Transplantation in Patients With Relapsed or Refractory Acute Myeloid Leukemia. Front Oncol 2021; 11:702239. [PMID: 34504785 PMCID: PMC8421765 DOI: 10.3389/fonc.2021.702239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/02/2021] [Indexed: 12/11/2022] Open
Abstract
Hypomethylating agents, decitabine (DAC) and azacitidine, can act as prophylactic and pre-emptive approaches after allogeneic hematopoietic stem cell transplantation (allo-HSCT) and a non-intensive bridging approach before allo-HSCT. However, they are rarely used as a part of conditioning regimens in patients with relapsed or refractory acute myeloid leukemia (AML). This retrospectively study included a total of 65 patients (median, 37; range, 13–63) with relapsed or refractory AML who were treated by allo-HSCT after myeloablative conditioning regimens without or with DAC (high-dose DAC schedule, 75 mg/m2 on day −9 and 50 mg/m2 on day −8; low-dose DAC schedule, 25 mg/m2/day on day −10 to −8). DAC exerted no impact on hematopoietic reconstitution. However, patients who were treated with the high-dose DAC schedule had significantly higher incidence of overall survival (OS, 50.0%) and leukemia-free survival (LFS, 35.0%), and lower incidence of relapse (41.1%) and grade II–IV acute graft versus host disease (aGVHD, 10.0%) at 3 years, when compared with those treated with standard conditioning regimens or with the low-dose DAC schedule. In conclusion, high-dose DAC combined with standard conditioning regimens before allo-HSCT is feasible and efficient and might improve outcomes of patients with relapsed or refractory AML, which provides a potential approach to treat these patients.
Collapse
Affiliation(s)
- Yuhang Li
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Longcan Cheng
- Department of Hematology, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Chen Xu
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jianlin Chen
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jiangwei Hu
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Na Liu
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Sanchun Lan
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jing Xie
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ting Sun
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lei Wang
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yu Zhang
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yao Sun
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shuiping Chen
- Department of Laboratory Medicine, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Liangding Hu
- Department of Hematology, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
20
|
Zhang Y, Gao Q, Wu Y, Peng Y, Zhuang J, Yang Y, Jiang W, Liu X, Guan G. Hypermethylation and Downregulation of UTP6 Are Associated With Stemness Properties, Chemoradiotherapy Resistance, and Prognosis in Rectal Cancer: A Co-expression Network Analysis. Front Cell Dev Biol 2021; 9:607782. [PMID: 34485268 PMCID: PMC8416280 DOI: 10.3389/fcell.2021.607782] [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: 09/18/2020] [Accepted: 07/12/2021] [Indexed: 12/28/2022] Open
Abstract
Background To identify the hub genes associated with chemoradiotherapy resistance in rectal cancer and explore the potential mechanism. Methods Weighted gene co-expression network analysis (WGCNA) was performed to identify the gene modules correlated with the chemoradiotherapy resistance of rectal cancer. Results The mRNA expression of 31 rectal cancer patients receiving preoperative chemoradiotherapy was described in our previous study. Through WGCNA, we demonstrated that the chemoradiotherapy resistance modules were enriched for translation, DNA replication, and the androgen receptor signaling pathway. Additionally, we identified and validated UTP6 as a new effective predictor for chemoradiotherapy sensitivity and a prognostic factor for the survival of colorectal cancer patients using our data and the GSE35452 dataset. Low UTP6 expression was correlated with significantly worse disease-free survival (DFS), overall survival (OS), and event- and relapse-free survival both in our data and the R2 Platform. Moreover, we verified the UTP6 expression in 125 locally advanced rectal cancer (LARC) patients samples by immunohistochemical analysis. The results demonstrated that low UTP6 expression was associated with worse DFS and OS by Kaplan-Meier and COX regression model analyses. Gene set enrichment and co-expression analyses showed that the mechanism of the UTP6-mediated chemoradiotherapy resistance may involve the regulation of FOXK2 expression by transcription factor pathways. Conclusion Low expression of the UTP6 was found to be associated with chemoradiotherapy resistance and the prognosis of colorectal cancer possibly via regulating FOXK2 expression by transcription factor pathways.
Collapse
Affiliation(s)
- Yiyi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qiao Gao
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yong Wu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yong Peng
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jinfu Zhuang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yuanfeng Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Weizhong Jiang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xing Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guoxian Guan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
21
|
Zhao F, Wang J, Yao L, Qin YT, Tuerxun N, Wang H, Jiang M, Hao JP. Synergistic inhibitory effect of Smo inhibitor jervine and its combination with decitabine can target Hedgehog signaling pathway to inhibit myelodysplastic syndrome cell line. ACTA ACUST UNITED AC 2021; 26:518-528. [PMID: 34314648 DOI: 10.1080/16078454.2021.1950897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Hypomethylating agents (HMAs) have been reported to target the Sonic Hedgehog (Shh) signaling pathway in myelodysplastic syndrome (MDS). However, the synergistic inhibitory effect of Smo inhibitor jervine and its combination with decitabine in MUTZ-1 cell lines remains lacking. METHODS We used a CCK-8 assay to detect the in-vitro proliferation rate of MUTZ-1 cell lines. Besides, the Annexin V-FITC/PI double staining flow cytometry was utilized to detect the apoptosis rate and cell cycle changes. The expression levels of mRNA were quantified by using qRT-PCR, and the western blot was employed to detect the expression of proteins. RESULTS We found that the single-agent jervine or decitabine can significantly inhibit the proliferation rate of MUTZ-1 cell lines, and this inhibitory effect is time-dependent and concentration-dependent. The combined intervention of the jervine and decitabine can more significantly inhibit cell proliferation, induce cell apoptosis, and block the G1 phase of the cell cycle. The combined intervention of the two drugs significantly reduced Smo and G1i-1 mRNA expression in MUTZ-1 cells. Furthermore, after combining both of the drug treatments, the proteins levels of Smo, G1i-1, PI3K, p-AKT, Bcl2, and Cyclin Dl were significantly downregulated, and Caspase-3 is upregulated, indicating that jervine with its combination of decitabine might be effective for controlling the proliferation, apoptosis, and cell cycle. CONCLUSION The Smo inhibitor jervine and its combination with decitabine have a synergistic effect on the proliferation, cell cycle, and apoptosis of MUTZ-1 cells, and its mechanism may be achieved by interfering with the Shh signaling pathway.
Collapse
Affiliation(s)
- Fang Zhao
- Department of Hematology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Jie Wang
- Department of Pharmacy, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Liu Yao
- The First Clinical Medical College of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Yu-Ting Qin
- Department of Hematology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Niluopaer Tuerxun
- Department of Hematology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Huan Wang
- Department of Hematology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Ming Jiang
- Department of Hematology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Jian-Ping Hao
- Department of Hematology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| |
Collapse
|
22
|
Liu J, Liang L, Li X, Peng YL, Zhang J, Wang XL, Liu J, Nie L. AICAR suppresses cell proliferation and synergizes with decitabine in myelodysplastic syndrome via DNA damage induction. Biotechnol Lett 2021; 43:1131-1142. [PMID: 33788127 DOI: 10.1007/s10529-021-03112-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/28/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the efficacy and safety of the AMPK activator AICAR alone or in combination with decitabine on myelodysplastic syndromes (MDS). RESULTS p-AMPK (Thr172) expression was lower in MDS samples than in healthy donors. AMPK agonist AICAR inhibited the proliferation of MDS cell lines (SKM1 and MDS-L) (P < 0.05). The results from flow cytometry suggested that AICAR induced G0/G1 phase arrest and apoptosis through inducing DNA damage, as confirmed by immunofluorescence analysis in MDS cell lines. AICAR alone or in combination with decitabine was applied to the two MDS cell lines, and the combination index values at all concentrations were significantly < 1. This strong synergistic effect was also corroborated in the primary MDS patient samples and in an MDS cell line xenograft mouse model. Furthermore, immunohistochemical staining showed that there was more DNA damage accumulation in the combination group than that in any other groups. CONCLUSION This is the first report on how the AICAR suppresses MDS cell proliferation and synergizes with decitabine via DNA damage induction. AICAR in combination with decitabine may be a promising therapeutic strategy in MDS.
Collapse
Affiliation(s)
- Jin Liu
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Molecular Biology Research Center, Central South University, Changsha, 410078, Hunan, China
| | - Long Liang
- Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410078, Hunan, China
| | - Xin Li
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Molecular Biology Research Center, Central South University, Changsha, 410078, Hunan, China
| | - Yuan Liang Peng
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Molecular Biology Research Center, Central South University, Changsha, 410078, Hunan, China
| | - Ji Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of South China University, Hengyang, 421000, Hunan, China
| | - Xiao Long Wang
- School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Jing Liu
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Molecular Biology Research Center, Central South University, Changsha, 410078, Hunan, China
| | - Ling Nie
- Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410078, Hunan, China.
| |
Collapse
|
23
|
McCullough KB, Kuhn AK, Patnaik MM. Treatment advances for pediatric and adult onset neoplasms with monocytosis. Curr Hematol Malig Rep 2021; 16:256-266. [PMID: 33728588 DOI: 10.1007/s11899-021-00622-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW For decades, the management of chronic myelomonocytic leukemia (CMML) or juvenile myelomonocytic leukemia (JMML) has been largely inextricable from myelodysplastic syndromes (MDS), myeloproliferative neoplasms, and acute myeloid leukemia. Hallmarks of these diseases have been the emergence of unique genomic signatures and discouraging responses to available therapies. Here, we will critically examine the current options for management and review the rapidly developing opportunities based on advances in CMML and JMML disease biology. RECENT FINDINGS Few clinical trials have exclusively been done in CMML, and in JMML, the rarity of the disease limits wide scale participation. Recent case series in JMML suggest that hypomethylating agents (HMAs) are a viable option for bridging to curative intent with allogeneic hematopoietic stem cell transplant or as posttransplant maintenance. Emerging evidence has demonstrated targeting the RAS-pathway via MEK inhibition may also be considered. In CMML, treatment with HMAs is largely derived from data inclusive of MDS patients, including a small number of patients with dysplastic CMML variants. Based on CMML disease biology, additional therapeutic targets being investigated include inhibitors of splicing, CD123/dendritic cell axis, inherent GM-CSF progenitor cell hypersensitivity, and targeting the JAK/STAT pathway. Current evidence is also expanding for oral HMAs. The management of CMML and JMML is rapidly evolving and clinicians must be aware of the genetic landscape and expanding treatment options to ensure these rare populations are afforded therapeutic interventions best suited to their needs.
Collapse
Affiliation(s)
- Kristen B McCullough
- Department of Pharmacy Services, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
| | - Alexis K Kuhn
- Department of Pharmacy Services, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Mrinal M Patnaik
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
24
|
Greenmyer JR, Kohorst M. Pediatric Neoplasms Presenting with Monocytosis. Curr Hematol Malig Rep 2021; 16:235-246. [PMID: 33630234 DOI: 10.1007/s11899-021-00611-x] [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] [Accepted: 02/01/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW Juvenile myelomonocytic leukemia (JMML) is a rare but severe pediatric neoplasm with hematopoietic stem cell transplant as its only established curative option. The development of targeted therapeutics for JMML is being guided by an understanding of the pathobiology of this condition. Here, we review JMML with an emphasis on genetics in order to (i) demonstrate the relationship between JMML genotype and clinical phenotype and (ii) explore potential genetic targets of novel JMML therapies. RECENT FINDINGS DNA hypermethylation studies have demonstrated consistently that methylation is related to disease severity. Increasing understanding of methylation in JMML may open the door to novel therapies, such as DNA methyltransferase inhibitors. The PI3K/AKT/MTOR, JAK/STAT, and RAF/MEK/ERK pathways are being investigated as therapeutic targets for JMML. Future therapy for JMML will be driven by an increased understanding of pathobiology. Targeted therapeutic approaches hold potential for improving outcomes in patients with JMML.
Collapse
Affiliation(s)
| | - Mira Kohorst
- Pediatric Hematology and Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
25
|
Mancarella D, Plass C. Epigenetic signatures in cancer: proper controls, current challenges and the potential for clinical translation. Genome Med 2021; 13:23. [PMID: 33568205 PMCID: PMC7874645 DOI: 10.1186/s13073-021-00837-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 01/21/2021] [Indexed: 12/26/2022] Open
Abstract
Epigenetic alterations are associated with normal biological processes such as aging or differentiation. Changes in global epigenetic signatures, together with genetic alterations, are driving events in several diseases including cancer. Comparative studies of cancer and healthy tissues found alterations in patterns of DNA methylation, histone posttranslational modifications, and changes in chromatin accessibility. Driven by sophisticated, next-generation sequencing-based technologies, recent studies discovered cancer epigenomes to be dominated by epigenetic patterns already present in the cell-of-origin, which transformed into a neoplastic cell. Tumor-specific epigenetic changes therefore need to be redefined and factors influencing epigenetic patterns need to be studied to unmask truly disease-specific alterations. The underlying mechanisms inducing cancer-associated epigenetic alterations are poorly understood. Studies of mutated epigenetic modifiers, enzymes that write, read, or edit epigenetic patterns, or mutated chromatin components, for example oncohistones, help to provide functional insights on how cancer epigenomes arise. In this review, we highlight the importance and define challenges of proper control tissues and cell populations to exploit cancer epigenomes. We summarize recent advances describing mechanisms leading to epigenetic changes in tumorigenesis and briefly discuss advances in investigating their translational potential.
Collapse
Affiliation(s)
- Daniela Mancarella
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany. .,Faculty of Biosciences, Ruprecht-Karls-University of Heidelberg, 69120, Heidelberg, Germany.
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,German Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| |
Collapse
|
26
|
Cabezón M, Malinverni R, Bargay J, Xicoy B, Marcé S, Garrido A, Tormo M, Arenillas L, Coll R, Borras J, Jiménez MJ, Hoyos M, Valcárcel D, Escoda L, Vall-Llovera F, Garcia A, Font LL, Rámila E, Buschbeck M, Zamora L. Different methylation signatures at diagnosis in patients with high-risk myelodysplastic syndromes and secondary acute myeloid leukemia predict azacitidine response and longer survival. Clin Epigenetics 2021; 13:9. [PMID: 33446256 PMCID: PMC7809812 DOI: 10.1186/s13148-021-01002-y] [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] [Received: 07/15/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Epigenetic therapy, using hypomethylating agents (HMA), is known to be effective in the treatment of high-risk myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) patients who are not suitable for intensive chemotherapy and/or allogeneic stem cell transplantation. However, response rates to HMA are low and there is an unmet need in finding prognostic and predictive biomarkers of treatment response and overall survival. We performed global methylation analysis of 75 patients with high-risk MDS and secondary AML who were included in CETLAM SMD-09 protocol, in which patients received HMA or intensive treatment according to age, comorbidities and cytogenetic. RESULTS Unsupervised analysis of global methylation pattern at diagnosis did not allow patients to be differentiated according to the cytological subtype, cytogenetic groups, treatment response or patient outcome. However, after a supervised analysis we found a methylation signature defined by 200 probes, which allowed differentiating between patients responding and non-responding to azacitidine (AZA) treatment and a different methylation pattern also defined by 200 probes that allowed to differentiate patients according to their survival. On studying follow-up samples, we confirmed that AZA decreases global DNA methylation, but in our cohort the degree of methylation decrease did not correlate with the type of response. The methylation signature detected at diagnosis was not useful in treated samples to distinguish patients who were going to relapse or progress. CONCLUSIONS Our findings suggest that in a subset of specific CpGs, altered DNA methylation patterns at diagnosis may be useful as a biomarker for predicting AZA response and survival.
Collapse
Affiliation(s)
- M Cabezón
- Hematology Laboratory Service, ICO Badalona-Hospital Germans Trias I Pujol, Myeloid Neoplasms Group, Josep Carreras Leukemia Research Institute (IJC), Badalona, Spain.,Departament de Medicina, Universitat Autònoma de Barcelona, Badalona, Spain
| | - R Malinverni
- Cancer and Leukemia Epigenetics and Biology Program, Josep Carreras Leukemia Research Institute (IJC), Campus ICO-GTP-UAB, Badalona, Spain
| | - J Bargay
- Hematology Service, Hospital Son Llàtzer, Palma de Mallorca, Spain
| | - B Xicoy
- Hematology Laboratory Service, ICO Badalona-Hospital Germans Trias I Pujol, Myeloid Neoplasms Group, Josep Carreras Leukemia Research Institute (IJC), Badalona, Spain.,Departament de Medicina, Universitat Autònoma de Barcelona, Badalona, Spain
| | - S Marcé
- Hematology Laboratory Service, ICO Badalona-Hospital Germans Trias I Pujol, Myeloid Neoplasms Group, Josep Carreras Leukemia Research Institute (IJC), Badalona, Spain
| | - A Garrido
- Hematology Service, Hospital de Sant Pau, Barcelona, Spain
| | - M Tormo
- Hematology Service, Hospital Clínico de Valencia, Valencia, Spain
| | - L Arenillas
- Hematology Service, Hospital del Mar, Barcelona, Spain
| | - R Coll
- Hematology Service, ICO Girona - Hospital Josep Trueta, Girona, Spain
| | - J Borras
- Hematology Service, Hospital Son Llàtzer, Palma de Mallorca, Spain
| | - M J Jiménez
- Hematology Laboratory Service, ICO Badalona-Hospital Germans Trias I Pujol, Myeloid Neoplasms Group, Josep Carreras Leukemia Research Institute (IJC), Badalona, Spain
| | - M Hoyos
- Hematology Service, Hospital de Sant Pau, Barcelona, Spain
| | - D Valcárcel
- Hematology Service, Hospital Vall D'Hebron, Barcelona, Spain
| | - L Escoda
- Hematology Service, Hospital Joan XXIII, Tarragona, Spain
| | - F Vall-Llovera
- Hematology Service, Hospital Mútua de Terrassa, Terrassa, Spain
| | - A Garcia
- Hematology Service, Hospital Arnau de Vilanova, Lleida, Spain
| | - L L Font
- Hematology Service, Hospital Verge de La Cinta, Tortosa, Spain
| | - E Rámila
- Hematology Service, Hospital Parc Taulí, Sabadell, Spain
| | - M Buschbeck
- Cancer and Leukemia Epigenetics and Biology Program, Josep Carreras Leukemia Research Institute (IJC), Campus ICO-GTP-UAB, Badalona, Spain.,Program for Predictive and Personalized Medicine of Cancer, Germans Trias I Pujol Research Institute (PMPPC-IGTP), Badalona, Spain
| | - L Zamora
- Hematology Laboratory Service, ICO Badalona-Hospital Germans Trias I Pujol, Myeloid Neoplasms Group, Josep Carreras Leukemia Research Institute (IJC), Badalona, Spain.
| | | |
Collapse
|
27
|
Pei Y, Lou X, Li K, Xu X, Guo Y, Xu D, Yang Z, Xu D, Cui W, Zhang D. Peripheral Blood Leukocyte N6-methyladenosine is a Noninvasive Biomarker for Non-small-cell Lung Carcinoma. Onco Targets Ther 2020; 13:11913-11921. [PMID: 33239892 PMCID: PMC7682600 DOI: 10.2147/ott.s267344] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Background N6-methyladenosine (m6A) triggers a new layer of epi-transcription. However, the potential noninvasive screening and diagnostic value of peripheral blood m6A for cancer are still unknown. Here, we intend to investigate whether leukocyte m6A can be a novel biomarker for non-small-cell lung cancer (NSCLC). Materials and Methods Peripheral blood was collected from 119 NSCLC patients and 74 age-matched healthy controls. Total RNA was isolated from leukocytes for m6A measurement, and clinical information of participants was reviewed. The sensitivity, specificity, and area under the curve (AUC) of m6A for cancer diagnosis were evaluated by the receiver-operating characteristic (ROC) curve analysis. Flow cytometry and the Human Protein Atlas (HPA) database were used to characterize m6A in leukocyte differentials. Pearson's correlation was applied to indicate the relationship between m6A level and hematology variables. qPCR and bioinformatic analysis were used to identity the expression of m6A regulators in leukocyte. Results Leukocyte m6A was significantly elevated in 119 NSCLC patients compared with 74 healthy controls (P<0.001). We did not find significant association between m6A and age or gender. Elevated m6A level in NSCLC was associated with tumor stage (P<0.05) and tumor differentiation (P<0.05), and was significantly reduced after surgery (P<0.01). ROC curve analysis revealed that leukocyte m6A could significantly discriminate patients with lung adenocarcinoma (LUAD) (AUC=0.736, P<0.001) and lung squamous cell carcinoma (LUSC) (AUC=0.963, P<0.001) from healthy individuals. m6A displayed superior sensitivity (100%) and specificity (85.7%) for LUSC than squamous cell carcinoma (SCC) antigen and cytokeratin fragment 211 (Cyfra211). Flow cytometry analysis showed m6A modification was mainly localized on T cells and monocytes among leukocyte differentials. Leukocyte m6A was positively correlated with the number of lymphocytes and negatively correlated with monocytes in NSCLC but not in healthy controls. qPCR and bioinformatic analysis showed that elevated leukocyte m6A in NSCLC was caused by upregulated methyltransferase complex and downregulated FTO and ALKBH5. Conclusion Leukocyte m6A represents a potential noninvasive biomarker for NSCLC screening, monitoring and diagnosis.
Collapse
Affiliation(s)
- Yuqing Pei
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Xiaoying Lou
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Kexin Li
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Xiaotian Xu
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Ye Guo
- Department of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, People's Republic of China
| | - Danfei Xu
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Zhenxi Yang
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Dongsheng Xu
- Hematopathology Program, CBL Path, Inc, Rye Brook, NY 10753, USA
| | - Wei Cui
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Donghong Zhang
- Center for Molecular and Translational Medicine, Georgia State University, Research Science Center, Atlanta, GA 30303, USA
| |
Collapse
|
28
|
Haltom AR, Toll SA, Cheng D, Maegawa S, Gopalakrishnan V, Khatua S. Medulloblastoma epigenetics and the path to clinical innovation. J Neurooncol 2020; 150:35-46. [PMID: 32816225 DOI: 10.1007/s11060-020-03591-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/06/2020] [Indexed: 12/30/2022]
Abstract
INTRODUCTION In the last decade, a number of genomic and pharmacological studies have demonstrated the importance of epigenetic dysregulation in medulloblastoma initiation and progression. High throughput approaches including gene expression array, next-generation sequencing (NGS), and methylation profiling have now clearly identified at least four molecular subgroups within medulloblastoma, each with distinct clinical and prognostic characteristics. These studies have clearly shown that despite the overall paucity of mutations, clinically relevant events do occur within the cellular epigenetic machinery. Thus, this review aims to provide an overview of our current understanding of the spectrum of epi-oncogenetic perturbations in medulloblastoma. METHODS Comprehensive review of epigenetic profiles of different subgroups of medulloblastoma in the context of molecular features. Epigenetic regulation is mediated mainly by DNA methylation, histone modifications and microRNAs (miRNA). Importantly, epigenetic mis-events are reversible and have immense therapeutic potential. CONCLUSION The widespread epigenetic alterations present in these tumors has generated intense interest in their use as therapeutic targets. We provide an assessment of the progress that has been made towards the development of molecular subtypes-targeted therapies and the current status of clinical trials that have leveraged these recent advances.
Collapse
Affiliation(s)
- Amanda R Haltom
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.,Center for Cancer Epigenetics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie A Toll
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, USA
| | - Donghang Cheng
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.,Center for Cancer Epigenetics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Shinji Maegawa
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.,Center for Cancer Epigenetics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Vidya Gopalakrishnan
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA. .,Department of Molecular and Cellular Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA. .,Center for Cancer Epigenetics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA. .,Brain Tumor Center, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Soumen Khatua
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA. .,Brain Tumor Center, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
29
|
Zhou QB, Liu ZT, Wang HZ, Guo XQ, Xu YG, Hu XM. Arsenic Disulfide Promoted Hypomethylation by Increasing DNA Methyltransferases Expression in Myelodysplastic Syndrome. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1641-1650. [PMID: 32431489 PMCID: PMC7201013 DOI: 10.2147/dddt.s239158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/25/2020] [Indexed: 12/25/2022]
Abstract
Background Previous studies have shown that DNA methylation plays a significant role in myelodysplastic syndrome (MDS). In addition to hypermethylation, aberrant hypomethylation can result in the transcriptional activation of oncogenes in cancer, including MDS. Therefore, drugs targeting DNA hypomethylation are needed for the treatment of MDS. This study aimed to investigate whether As2S2 promoted hypomethylation by increasing DNA methyltransferases (DNMTs) expression in MDS. Patients and Methods Ten bone marrow samples from MDS patients and 3 healthy donors were obtained for the examination of the DNA methylation with a Human Methylation 850K BeadChip. The mRNA expressions for the DNMTs in the ten MDS patients and 3 controls were compared by Q-PCR. Then, the MDS cell line SKM-1 was treated with As2S2. After 2 days of treatment, Human Methylation 850K BeadChip was applied to analyze the changes of gene methylation status in the cells. Q-PCR and Western blot were taken to test the changes of mRNA and protein expressions for DNMTs in SKM-1 cells after treatment. Results Five hundred ninety-two abnormally hypomethylated genes were found in MDS patients compared to those in controls by Human Methylation 850K. The mRNA expressions of DNMTs (DNMT1, DNMT3a and DNMT3b) in MDS patients were significantly lower than those in healthy individuals. The IC50 value of As2S2 for SKM-1 cells was 4.97 μmol/L.Treatment with As2S2 at 2 μmoL/L resulted in significant alterations in the methylation levels at 1718 sites in SKM-1 cells compared to those in the controls. Hypermethylation was observed in 1625 sites (94.58%), corresponding to 975 genes, compared to those in the controls. Finally, the expression levels of DNMTs (DNMT1, DNMT3a, and DNMT3b) significantly increased in SKM-1 cells treated with As2S2 at 2 μmoL/L and 4 μmoL/L. Conclusion These data show a potential clinical application of As2S2 as an innovative hypermethylation agent in MDS.
Collapse
Affiliation(s)
- Qing-Bing Zhou
- China Academy of Chinese Medical Sciences, Institute of Geriatric Medicine, Xiyuan Hospital, Beijing, People's Republic of China
| | - Zheng-Tang Liu
- China Academy of Chinese Medical Sciences, Institute of Geriatric Medicine, Xiyuan Hospital, Beijing, People's Republic of China
| | - Hong-Zhi Wang
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Xiao-Qing Guo
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Yong-Gang Xu
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Xiao-Mei Hu
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
30
|
Traditional Chinese Medicine Containing Arsenic Treated MDS Patients Effectively through Regulating Aberrant Hypomethylation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7469809. [PMID: 32215045 PMCID: PMC7085376 DOI: 10.1155/2020/7469809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/20/2019] [Accepted: 10/26/2019] [Indexed: 11/18/2022]
Abstract
Aberrant hypermethylation and hypomethylation both play important roles in myelodysplastic syndrome (MDS). Hypomethylating agents targeting hypermethylation have been employed for the MDS treatment, but the treatment effect is limited. Novel drugs for DNA hypomethylation-targeted therapy may be needed to improve clinic efficacy for the treatment of MDS. Chinese medicine (CM) herbs have been used to treat MDS for many years in our hospital. However, the long-term treatment effect and mechanism remain unclear. In this study, all 135 patients received CM treatment for at least 36 months. The response rates for CM treatment were 81.53% (106/130) for hematological improvement in 130 MDS-RCMD patients and 80% (4/5) for bone marrow CR in 5 MDS-RAEB patients, respectively. The Human Methylation 850K BeadChip showed that 115 genes (50.88%) were aberrantly hypomethylated in 5 MDS patients compared with 3 healthy individuals. GO-analysis showed that these hypomethylated genes participated in many cancer-related biological functions and pathways. Furthermore, 60 genes were hypermethylated and the protein expression level of DNMT1 was significantly increased in the 5 MDS patients after 6 months of CM treatment. Our study suggests that CM can improve aberrant hypomethylation by increasing DNMT1 expression in MDS. The data support the clinical application of CM herbs containing arsenic as an innovative hypermethylation-inducing regimen for the treatment of MDS.
Collapse
|
31
|
After 95 years, it's time to eRASe JMML. Blood Rev 2020; 43:100652. [PMID: 31980238 DOI: 10.1016/j.blre.2020.100652] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 12/07/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022]
Abstract
Juvenile myelomonocytic leukaemia (JMML) is a rare clonal disorder of early childhood. Constitutive activation of the RAS pathway is the initial event in JMML. Around 90% of patients diagnosed with JMML carry a mutation in the PTPN11, NRAS, KRAS, NF1 or CBL genes. It has been demonstrated that after this first genetic event, an additional somatic mutation or epigenetic modification is involved in disease progression. The available genetic and clinical data have enabled researchers to establish relationships between JMML and several clinical conditions, including Noonan syndrome, Ras-associated lymphoproliferative disease, and Moyamoya disease. Despite scientific progress and the development of more effective treatments, JMML is still a deadly disease: the 5-year survival rate is ~50%. Here, we report on recent research having led to a better understanding of the genetic and molecular mechanisms involved in JMML.
Collapse
|
32
|
Yoshida N, Sakaguchi H, Yabe M, Hasegawa D, Hama A, Hasegawa D, Kato M, Noguchi M, Terui K, Takahashi Y, Cho Y, Sato M, Koh K, Kakuda H, Shimada H, Hashii Y, Sato A, Kato K, Atsuta Y, Watanabe K. Clinical Outcomes after Allogeneic Hematopoietic Stem Cell Transplantation in Children with Juvenile Myelomonocytic Leukemia: A Report from the Japan Society for Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2019; 26:902-910. [PMID: 31790827 DOI: 10.1016/j.bbmt.2019.11.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/01/2019] [Accepted: 11/27/2019] [Indexed: 01/16/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) is the only curative treatment for juvenile myelomonocytic leukemia (JMML), but few large studies of HSCT for JMML exist. Using data from the Japan Society for Hematopoietic Cell Transplantation registry, we analyzed the outcomes of 129 children with JMML who underwent HSCT between 2000 and 2011. The 5-year overall survival (OS) rate and cumulative incidence of relapse were 64% and 34%, respectively. A regimen of busulfan/fludarabine/melphalan was the most commonly used (59 patients) and provided the best outcomes; the 5-year OS rate reached 73%, and the cumulative incidences of relapse and transplantation-related mortality were 26% and 9%, respectively. In contrast, the use of the irradiation-based myeloablative regimen was the most significant risk factor for OS (hazard ratio [HR], 2.92; P = .004) in the multivariate model. In addition, chronic graft-versus-host disease (GVHD) was strongly associated with lower relapse (HR, 0.37; P = .029) and favorable survival (HR, 0.22; P = .006). The current study has shown that a significant proportion of children with JMML can be cured with HSCT, especially those receiving the busulfan/fludarabine/melphalan regimen. Based on the lower relapse and better survival observed in patients with chronic GVHD, additional treatment strategies that focus on enhancing graft-versus-leukemia effects may further improve survival.
Collapse
Affiliation(s)
- Nao Yoshida
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan.
| | - Hirotoshi Sakaguchi
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Miharu Yabe
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan
| | - Daiichiro Hasegawa
- Departments of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Asahito Hama
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Motohiro Kato
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Maiko Noguchi
- Department of Pediatrics, National Kyushu Cancer Center, Fukuoka, Japan
| | - Kiminori Terui
- Department of Pediatrics, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuko Cho
- Department of Pediatrics, Hokkaido University Hospital, Sapporo, Japan
| | - Maho Sato
- Department of Hematology/Oncology, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Harumi Kakuda
- Department of Hematology/Oncology, Chiba Children's Hospital, Chiba, Japan
| | - Hiroyuki Shimada
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiko Hashii
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Sato
- Departments of Hematology and Oncology, Miyagi Children's Hospital, Sendai, Japan
| | - Koji Kato
- Central Japan Cord Blood Bank, Seto, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan; Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | | |
Collapse
|
33
|
Zhao XL, Jiang EL, Zhai WH, Ma QL, Pang AM, Wei JL, He Y, Yang DL, Feng SZ, Han MZ. [Decitabine-based conditioning regimen is feasible and effective in the treatment of myelodysplastic syndrome and chronic myelomonocytic leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:467-471. [PMID: 31340618 PMCID: PMC7342392 DOI: 10.3760/cma.j.issn.0253-2727.2019.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Indexed: 01/09/2023]
Abstract
Objective: To assess the efficacy and toxicity of decitabine-based conditioning regimen in patients with myelodysplastic syndrome (MDS) , acute myeloid leukemia secondary to MDS (MDS-AML) or chronic myelomonocytic leukemia (CMML) . Methods: From March 1, 2013 to May 25, 2015, 22 patients who underwent allogenic hematopoietic stem cell transplantation (allo-HSCT) with decitabine-based conditioning regimen were analyzed retrospectively. Results: ①22 patients, 14 males and 8 females with a median age of 42.5 (24-56) years old, were diagnosed as MDS (n=14) , CMML (n=4) , MDS-AML (n=4) . ②15 patients were treated with the conditioning regimen of decitabine combined with busulfan, cyclophosphamide, fludarabine, and cytarabine, the other 7 cases were treated with decitabine, busulfan, fludarabine, and cytarabine. The dose of decitabine was 20 mg·m(-2)·d(-1) for 5 days.Rabbit anti-human anti-thymocyte globulin (2.5 mg·kg(-1)·d(-1) for 4 days) was involved in conditioning regimen in patients with unrelated donor or haploidentical transplantation. ③Except 1 patient died of infection in 2 months after transplantation, the other patients were engrafted successfully. The median time of granulocyte engraftment was 13 (12-18) days, and the median time of platelet engraftment was 16 (13-81) days. ④The incidence of acute graft versus host disease (aGVHD) was (41.3±10.6) %, and severe aGVHD (grade of III-IV) was (18.4±9.7) %. The incidence of chronic graft versus host disease (cGVHD) was (56.4±11.3) %, and extensive cGVHD was (36.4±12.1) %. ⑤8 patients were suffered with cytomegalovirus (CMV) viremia. Among the 18 patients with definitely infection, 6 occurred during myelosuppression and 12 cases occurred after hematopoietic reconstruction. The 2-year and 3-year non-relapse mortality was (13.9±7.4) % and (24.3±9.5) %, respectively. ⑥The 2-year and 3-year overall survival (OS) was (77.3±8.9) % and (67.9±10.0) %, respectively. The 2-year and 3-year relapse-free survival (RFS) was (72.7±9.5) % and (63.6±10.3) %, respectively. Conclusions: allo-HSCT with decitabine-based conditioning regimen is feasible in the treatment of MDS, MDS-AML or CMML.
Collapse
Affiliation(s)
- X L Zhao
- Institute of Hematology and Blood Diseases Hospital, CAMS & PUMC, Tianjin 300020, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Stomper J, Lübbert M. Can we predict responsiveness to hypomethylating agents in AML? Semin Hematol 2019; 56:118-124. [PMID: 30926087 DOI: 10.1053/j.seminhematol.2019.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 02/18/2019] [Accepted: 02/22/2019] [Indexed: 11/11/2022]
Abstract
DNA-hypomethylating agents (HMAs) were developed as nonintensive treatment alternatives to standard chemotherapy in older, unfit patients with acute myeloid leukemia and myelodysplastic syndrome. Given their distinct effects on the methylome and transcriptome of malignant cells compared to cytarabine (Ara-C) and other cytotoxic drugs not inhibiting DNA methyltransferases, it is of great interest to define their specific clinical ``signature.'' Here, we present and discuss clinical, genetic, and epigenetic predictors of responsiveness to HMAs. Indeed, mounting evidence supports the notion that HMAs are not "just another kind of low-dose Ara-C." Not only patient factors (age, performance status, comorbidities, etc.), blast counts, and early platelet response, but also adverse genetics (monosomal karyotype and/or a TP53 mutation) have predictive potential. Given the surprising-and initially counterintuitive-responses observed in patients with the latter features, these are subject to mechanistic studies to elucidate their as yet unresolved interaction with HMAs. Finally, other potential biomarkers for HMA response such as elevated fetal hemoglobin might also contribute to overcome the present challenges in predicting responsiveness to HMAs.
Collapse
Affiliation(s)
- Julia Stomper
- Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Michael Lübbert
- Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany; German Cancer Research Consortium (DKTK), Freiburg, Germany.
| |
Collapse
|
35
|
Azacitidine is effective for targeting leukemia-initiating cells in juvenile myelomonocytic leukemia. Leukemia 2019; 33:1805-1810. [DOI: 10.1038/s41375-018-0343-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/15/2018] [Accepted: 10/24/2018] [Indexed: 02/05/2023]
|
36
|
Cai YL, Zhang JL, Zhu XF. [Advances in the treatment of juvenile myelomonocytic leukemia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:958-963. [PMID: 30477631 PMCID: PMC7389026 DOI: 10.7499/j.issn.1008-8830.2018.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 09/11/2018] [Indexed: 06/09/2023]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare chronic myeloid leukemia in children and has the features of both myelodysplastic syndrome and myeloproliferative neoplasm. It is highly malignant and has a poor treatment outcome. Children with JMML have a poor response to conventional chemotherapy. At present, hematopoietic stem cell transplantation is the only possible cure for this disease. In recent years, significant progress has been made in targeted therapy for mutant genes in the Ras signaling pathway and demethylation treatment of aberrant methylation of polygenic CpG islands. This article reviews the treatment and efficacy evaluation of JMML.
Collapse
Affiliation(s)
- Yu-Li Cai
- Department of Pediatrics, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China.
| | | | | |
Collapse
|
37
|
N6-Methyladenosine in RNA and DNA: An Epitranscriptomic and Epigenetic Player Implicated in Determination of Stem Cell Fate. Stem Cells Int 2018; 2018:3256524. [PMID: 30405719 PMCID: PMC6199872 DOI: 10.1155/2018/3256524] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 07/24/2018] [Indexed: 02/06/2023] Open
Abstract
Vast emerging evidences are linking the base modifications and determination of stem cell fate such as proliferation and differentiation. Among the base modification markers extensively studied, 5-methylcytosine (5-mC) and its oxidative derivatives (5-hydroxymethylcytosine (5-hmC), 5-formylcytosine (5-fC), and 5-carboxylcytosine (5-caC)) dynamically occur in DNA and RNA and have been acknowledged as important epigenetic markers involved in regulation of cellular biological processes. N6-Methyladenosine modification in DNA (m6dA), mRNA (m6A), tRNA, and other noncoding RNAs has been defined as another important epigenetic and epitranscriptomic marker in eukaryotes in recent years. The mRNA m6A modification has been characterized biochemically, molecularly, and phenotypically, including elucidation of its methyltransferase complexes (m6A writer), demethylases (m6A eraser), and direct interaction proteins (readers), while limited information on the DNA m6dA is available. The levels and the landscapes of m6A in the epitranscriptomes and epigenomes are precisely and dynamically regulated by the fine-tuned coordination of the writers and erasers in accordance with stages of the growth, development, and reproduction as naturally programmed during the lifespan. Additionally, progress has been made in appreciation of the link between aberrant m6A modification in stem cells and diseases, like cancers and neurodegenerative disorders. These achievements are inspiring scientists to further uncover the epigenetic mechanisms for stem cell development and to dissect pathogenesis of the multiple diseases conferred by development aberration of the stem cells. This review article will highlight the research advances in the role of m6A methylation modifications of DNA and RNA in the regulation of stem cell and genesis of the closely related disorders. Additionally, this article will also address the research directions in the future.
Collapse
|
38
|
Pan T, Qi J, You T, Yang L, Wu D, Han Y, Zhu L. Addition of histone deacetylase inhibitors does not improve prognosis in patients with myelodysplastic syndrome and acute myeloid leukemia compared with hypomethylating agents alone: A systematic review and meta-analysis of seven prospective cohort studies. Leuk Res 2018; 71:13-24. [DOI: 10.1016/j.leukres.2018.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/06/2018] [Accepted: 06/08/2018] [Indexed: 11/29/2022]
|
39
|
Dvorak CC, Satwani P, Stieglitz E, Cairo MS, Dang H, Pei Q, Gao Y, Wall D, Mazor T, Olshen AB, Parker JS, Kahwash S, Hirsch B, Raimondi S, Patel N, Skeens M, Cooper T, Mehta PA, Grupp SA, Loh ML. Disease burden and conditioning regimens in ASCT1221, a randomized phase II trial in children with juvenile myelomonocytic leukemia: A Children's Oncology Group study. Pediatr Blood Cancer 2018; 65. [PMID: 29528181 PMCID: PMC5980696 DOI: 10.1002/pbc.27034] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Most patients with juvenile myelomonocytic leukemia (JMML) are curable only with allogeneic hematopoietic cell transplantation (HCT). However, the current standard conditioning regimen, busulfan-cyclophosphamide-melphalan (Bu-Cy-Mel), may be associated with higher risks of morbidity and mortality. ASCT1221 was designed to test whether the potentially less-toxic myeloablative conditioning regimen containing busulfan-fludarabine (Bu-Flu) would be associated with equivalent outcomes. PROCEDURE Twenty-seven patients were enrolled on ASCT1221 from 2013 to 2015. Pre- and post-HCT (starting Day +30) mutant allele burden was measured in all and pre-HCT therapy was administered according to physician discretion. RESULTS Fifteen patients were randomized (six to Bu-Cy-Mel and nine to Bu-Flu) after meeting diagnostic criteria for JMML. Pre-HCT low-dose chemotherapy did not appear to reduce pre-HCT disease burden. Two patients, however, received aggressive chemotherapy pre-HCT and achieved low disease-burden state; both are long-term survivors. All four patients with detectable mutant allele burden at Day +30 post-HCT eventually progressed compared to two of nine patients with unmeasurable allele burden (P = 0.04). The 18-month event-free survival of the entire cohort was 47% (95% CI, 21-69%), and was 83% (95% CI, 27-97%) and 22% (95% CI, 03-51%) for Bu-Cy-Mel and Bu-Flu, respectively (P = 0.04). ASCT1221 was terminated early due to concerns that the Bu-Flu arm had inferior outcomes. CONCLUSIONS The regimen of Bu-Flu is inadequate to provide disease control in patients with JMML who present to HCT with large burdens of disease. Advances in molecular testing may allow better characterization of biologic risk, pre-HCT responses to chemotherapy, and post-HCT management.
Collapse
Affiliation(s)
| | | | | | - Mitchell S. Cairo
- Maria Fareri Children’s Hospital, Westchester Medical Center, New York Medical College
| | - Ha Dang
- University of Southern California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
In the last decade, epigenetic drugs (such as inhibitors of DNA methyltransferases and histone deacetylases) have been intensively used for cancer treatment. Their applications have shown high anticancer effectivity and tolerable side effects. However, they are unfortunately not effective in the treatment of some types and phenotypes of cancers. Nevertheless, several studies have demonstrated that problems of drug efficacy can be overcome through the combined application of therapeutic modulates. Therefore, combined applications of epigenetic agents with chemotherapy, radiation therapy, immunotherapy, oncolytic virotherapy and hyperthermia have been presented. This review summarizes and discusses the general principles of this approach, as introduced and supported by numerous examples. In addition, predictions of the future potential applications of this methodology are included.
Collapse
|