1
|
Kim DY, Shin DY, Oh S, Kim I, Kim EJ. Gene Expression and DNA Methylation Profiling Suggest Potential Biomarkers for Azacitidine Resistance in Myelodysplastic Syndrome. Int J Mol Sci 2024; 25:4723. [PMID: 38731939 PMCID: PMC11083267 DOI: 10.3390/ijms25094723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Myelodysplastic syndrome/neoplasm (MDS) comprises a group of heterogeneous hematopoietic disorders that present with genetic mutations and/or cytogenetic changes and, in the advanced stage, exhibit wide-ranging gene hypermethylation. Patients with higher-risk MDS are typically treated with repeated cycles of hypomethylating agents, such as azacitidine. However, some patients fail to respond to this therapy, and fewer than 50% show hematologic improvement. In this context, we focused on the potential use of epigenetic data in clinical management to aid in diagnostic and therapeutic decision-making. First, we used the F-36P MDS cell line to establish an azacitidine-resistant F-36P cell line. We performed expression profiling of azacitidine-resistant and parental F-36P cells and used biological and bioinformatics approaches to analyze candidate azacitidine-resistance-related genes and pathways. Eighty candidate genes were identified and found to encode proteins previously linked to cancer, chronic myeloid leukemia, and transcriptional misregulation in cancer. Interestingly, 24 of the candidate genes had promoter methylation patterns that were inversely correlated with azacitidine resistance, suggesting that DNA methylation status may contribute to azacitidine resistance. In particular, the DNA methylation status and/or mRNA expression levels of the four genes (AMER1, HSPA2, NCX1, and TNFRSF10C) may contribute to the clinical effects of azacitidine in MDS. Our study provides information on azacitidine resistance diagnostic genes in MDS patients, which can be of great help in monitoring the effectiveness of treatment in progressing azacitidine treatment for newly diagnosed MDS patients.
Collapse
Affiliation(s)
- Da Yeon Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Dong-Yeop Shin
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.-Y.S.); (S.O.)
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Somi Oh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.-Y.S.); (S.O.)
| | - Inho Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.-Y.S.); (S.O.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Eun Ju Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
- Institute for Molecular Bioscience, The University of Queensland, Carmody Rd., St Lucia, Brisbane, QLD 4072, Australia
- Genomics and Machine Learning Lab, QIMR Berghofer Medical Research Institute, Herston Rd., Herston, Brisbane, QLD 4006, Australia
| |
Collapse
|
2
|
Mendoza H, Siddon AJ. Molecular Techniques and Gene Mutations in Myelodysplastic Syndromes. Clin Lab Med 2023; 43:549-563. [PMID: 37865502 DOI: 10.1016/j.cll.2023.06.002] [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] [Indexed: 10/23/2023]
Abstract
Sequencing technology, particularly next-generation sequencing, has highlighted the importance of gene mutations in myelodysplastic syndromes (MDSs). Mutations affecting DNA methylation, chromatin modification, RNA splicing, cohesin complex, and other pathways are present in most MDS cases and often have prognostic and clinical implications. Updated international diagnostic guidelines as well as the new International Prognostic Scoring System-Molecular incorporate molecular data into the diagnosis and prognostication of MDS. With whole-genome sequencing predicted to become the future standard of genetic evaluation, it is likely that MDS diagnosis and management will become increasingly personalized based on an individual's clinical and genomic profile.
Collapse
Affiliation(s)
- Hadrian Mendoza
- Department of Internal Medicine, Yale School of Medicine, PO Box 208030, New Haven, CT 06520, USA
| | - Alexa J Siddon
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
A geno-clinical decision model for the diagnosis of myelodysplastic syndromes. Blood Adv 2021; 5:4361-4369. [PMID: 34592765 PMCID: PMC8579270 DOI: 10.1182/bloodadvances.2021004755] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/28/2021] [Indexed: 11/28/2022] Open
Abstract
We developed a machine learning–based model to assist in the differential diagnosis of myeloid malignancies. Our work also describes genotype-phenotype correlations in different myeloid malignancies.
The differential diagnosis of myeloid malignancies is challenging and subject to interobserver variability. We used clinical and next-generation sequencing (NGS) data to develop a machine learning model for the diagnosis of myeloid malignancies independent of bone marrow biopsy data based on a 3-institution, international cohort of patients. The model achieves high performance, with model interpretations indicating that it relies on factors similar to those used by clinicians. In addition, we describe associations between NGS findings and clinically important phenotypes and introduce the use of machine learning algorithms to elucidate clinicogenomic relationships.
Collapse
|
4
|
Schwind S, Jentzsch M, Kubasch AS, Metzeler KH, Platzbecker U. Myelodysplastic syndromes: Biological and therapeutic consequences of the evolving molecular aberrations landscape. Neoplasia 2021; 23:1101-1109. [PMID: 34601234 PMCID: PMC8495032 DOI: 10.1016/j.neo.2021.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/02/2021] [Indexed: 11/29/2022]
Abstract
Myelodysplastic syndromes (MDS) are clonal hematopoietic disorders with heterogeneous presentation, ranging from indolent disease courses to aggressive diseases similar to acute myeloid leukemia (AML). Approximately 90% of MDS patients harbor recurrent mutations , which – with the exception of mutated SF3B1 –have not (yet) been included into the diagnostic criteria or risk stratification for MDS. Accumulating evidence suggests their utility for diagnostic workup, treatment indication and prognosis. Subsequently, in patients with unexplained cytopenia or dysplasia identification of these mutations may lead to earlier diagnosis. The acquisition and expansion of additional driver mutations usually antecedes further disease progression to higher risk MDS or secondary AML and thus, can be clinically helpful to detect individuals that may benefit from aggressive treatment approaches. Here, we review our current understanding of somatic gene mutations, gene expression patterns and flow cytometry regarding their relevance for disease evolution from pre-neoplastic states to MDS and potentially AML.
Collapse
Affiliation(s)
- Sebastian Schwind
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Madlen Jentzsch
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Anne Sophie Kubasch
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Klaus H Metzeler
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany; German MDS Study Group (G-MDS), Leipzig, Germany; European Myelodysplastic Syndromes Cooperative Group, Leipzig, Germany.
| |
Collapse
|
5
|
Lee P, Yim R, Yung Y, Chu HT, Yip PK, Gill H. Molecular Targeted Therapy and Immunotherapy for Myelodysplastic Syndrome. Int J Mol Sci 2021; 22:10232. [PMID: 34638574 PMCID: PMC8508686 DOI: 10.3390/ijms221910232] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/22/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous, clonal hematological disorder characterized by ineffective hematopoiesis, cytopenia, morphologic dysplasia, and predisposition to acute myeloid leukemia (AML). Stem cell genomic instability, microenvironmental aberrations, and somatic mutations contribute to leukemic transformation. The hypomethylating agents (HMAs), azacitidine and decitabine are the standard of care for patients with higher-risk MDS. Although these agents induce responses in up to 40-60% of patients, primary or secondary drug resistance is relatively common. To improve the treatment outcome, combinational therapies comprising HMA with targeted therapy or immunotherapy are being evaluated and are under continuous development. This review provides a comprehensive update of the molecular pathogenesis and immune-dysregulations involved in MDS, mechanisms of resistance to HMA, and strategies to overcome HMA resistance.
Collapse
Affiliation(s)
| | | | | | | | | | - Harinder Gill
- Division of Haematology, Medical Oncology and Haemopoietic Stem Cell Transplantation, Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (P.L.); (R.Y.); (Y.Y.); (H.-T.C.); (P.-K.Y.)
| |
Collapse
|
6
|
Luo M, Yang X, Chen HN, Nice EC, Huang C. Drug resistance in colorectal cancer: An epigenetic overview. Biochim Biophys Acta Rev Cancer 2021; 1876:188623. [PMID: 34481016 DOI: 10.1016/j.bbcan.2021.188623] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. Despite significant progress that has been made in therapies against CRC over the past decades, drug resistance is still a major limitation in CRC treatment. Numerous investigations have unequivocally shown that epigenetic regulation plays an important role in CRC drug resistance because of the high rate of epigenetic alterations in multiple genes during cancer development or drug treatment. Furthermore, the reversibility of epigenetic alterations provides novel therapeutic strategies to overcome drug resistance using small molecules, which can target non-coding RNAs or reverse histone modification and DNA methylation. In this review, we discuss epigenetic regulation in CRC drug resistance and the possible role of preventing or reversing CRC drug resistance using epigenetic therapy in CRC treatment.
Collapse
Affiliation(s)
- Maochao Luo
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315020, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xingyue Yang
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315020, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Hai-Ning Chen
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.
| | - Canhua Huang
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315020, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
7
|
Palacios-Berraquero ML, Alfonso-Piérola A. Current Therapy of the Patients with MDS: Walking towards Personalized Therapy. J Clin Med 2021; 10:2107. [PMID: 34068316 PMCID: PMC8153316 DOI: 10.3390/jcm10102107] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis, dysplasia and peripheral cytopenias. Nowadays, MDS therapy is selected based on risk. The goals of therapy are different in low-risk and high-risk patients. In low-risk MDS, the goal is to decrease transfusion needs and to increase the quality of life. Currently, available drugs for newly diagnosed low-risk MDS include growth factor support, lenalidomide and immunosuppressive therapy. Additionally, luspatercept has recently been added to treat patients with MDS with ring sideroblasts, who are not candidates or have lost the response to erythropoiesis-stimulating agents. Treatment of high-risk patients is aimed to improve survival. To date, the only currently approved treatments are hypomethylating agents and allogeneic stem cell transplantation. However, the future for MDS patients is promising. In recent years, we are witnessing the emergence of multiple treatment combinations based on hypomethylating agents (pevonedistat, magrolimab, eprenetapopt, venetoclax) that have proven to be effective in MDS, even those with high-risk factors. Furthermore, the approval in the US of an oral hypomethylating agent opens the door to exclusively oral combinations for these patients and their consequent impact on the quality of life of these patients. Relapsed and refractory patients remain an unmet clinical need. We need more drugs and clinical trials for this profile of patients who have a dismal prognosis.
Collapse
Affiliation(s)
| | - Ana Alfonso-Piérola
- Hematology and Hemotherapy Department, Clínica Universidad de Navarra, 31008 Pamplona, Spain;
| |
Collapse
|
8
|
Zhang Z, Jia Y, Xv F, Song LX, Shi L, Guo J, Chang CK. Decitabine Induces Change of Biological Traits in Myelodysplastic Syndromes via FOXO1 Activation. Front Genet 2021; 11:603956. [PMID: 33584800 PMCID: PMC7873873 DOI: 10.3389/fgene.2020.603956] [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: 09/08/2020] [Accepted: 12/04/2020] [Indexed: 11/26/2022] Open
Abstract
Decitabine (DAC) is considered to be a profound global DNA demethylation, which can induce the re-expression of silenced tumor suppressor genes. Little is known about the function of tumor suppressor gene FOXO1 in myelodysplastic syndromes (MDS). To address this issue, the study firstly investigated differentially expressed genes (DEGs) for DAC treatment in MDS cell lines, then explored the role of FOXO1 through silencing its expression before DAC treatment in MDS. The results showed that FOXO1 exists in a hyperphosphorylated, inactive form in MDS-L cells. DAC treatment both induces FOXO1 expression and reactivates the protein in its low phosphorylation level. Additionally, the results also demonstrated that this FOXO1 activation is responsible for the DAC-induced apoptosis, cell cycle arrest, antigen differentiation, and immunoregulation in MDS-L cells. We also demonstrated DAC-induced FOXO1 activation upregulates anti-tumor immune response in higher-risk MDS specimens. Collectively, these results suggest that DAC induces FOXO1 activation, which plays an important role in anti-MDS tumors.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yan Jia
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Feng Xv
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lu-Xi Song
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lei Shi
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Juan Guo
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chun-Kang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
9
|
Ramos Perez J, Montalban-Bravo G. Emerging drugs for the treatment of chronic myelomonocytic leukemia. Expert Opin Emerg Drugs 2020; 25:515-529. [PMID: 33280448 DOI: 10.1080/14728214.2020.1854224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Introduction: Chronic myelomonocytic leukemia (CMML) is a clonal hematologic disorder with heterogenous prognosis, but with no curative therapies with exception of allogeneic transplant. Therapeutic options for patients with CMML are limited, and although hypomethylating agents such as azacitidine and decitabine are the standard of care, only 40% of patients achieve a response, and most responses are transient. Over the last 5 years, significant advances have been made in the understanding of the clonal landscape of CMML, some of the mechanisms associated to resistance to HMA, and other key biological processes involved in disease pathogenesis. Areas covered: The current article reviews the most relevant emerging therapies currently undergoing clinical trials for the treatment of previously untreated or relapsed CMML. Expert opinion: The presence of recurrent somatic mutations in CMML represents therapeutic opportunities to utilize specific small molecule inhibitors such as IDH, FLT3, MEK/ERK, PLK1, or splicing inhibitors and modulators. In addition, other novel agents such as immune therapies, BCL2 or MCL1 inhibitors and other monoclonal antibodies could lead to therapeutic advances. Identifying specific patient populations likely to benefit from some of these interventions, and development of optimal combinations will remain the challenge when determining their role in therapy.
Collapse
Affiliation(s)
- Jorge Ramos Perez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | | |
Collapse
|
10
|
Albany C, Fazal Z, Singh R, Bikorimana E, Adra N, Hanna NH, Einhorn LH, Perkins SM, Sandusky GE, Christensen BC, Keer H, Fang F, Nephew KP, Spinella MJ. A phase 1 study of combined guadecitabine and cisplatin in platinum refractory germ cell cancer. Cancer Med 2020; 10:156-163. [PMID: 33135391 PMCID: PMC7826483 DOI: 10.1002/cam4.3583] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Purpose Germ cell tumors (GCTs) are cured with therapy based on cisplatin, although a clinically significant number of patients are refractory and die of progressive disease. Based on preclinical studies indicating that refractory testicular GCTs are hypersensitive to hypomethylating agents (HMAs), we conducted a phase I trial combining the next‐generation HMA guadecitabine (SGI‐110) with cisplatin in recurrent, cisplatin‐resistant GCT patients. Methods Patients with metastatic GCTs were treated for five consecutive days with guadecitabine followed by cisplatin on day 8, for a 28‐day cycle for up to six cycles. The primary endpoint was safety and toxicity including dose‐limiting toxicity (DLT) and maximum tolerated dose (MTD). Results The number of patients enrolled was 14. The majority of patients were heavily pretreated. MTD was determined to be 30 mg/m2 guadecitabine followed by 100 mg/m2 cisplatin. The major DLTs were neutropenia and thrombocytopenia. Three patients had partial responses by RECIST criteria, two of these patients, including one with primary mediastinal disease, completed the study and qualified as complete responses by serum tumor marker criteria with sustained remissions of 5 and 13 months and survival of 16 and 26 months, respectively. The overall response rate was 23%. Three patients also had stable disease indicating a clinical benefit rate of 46%. Conclusions The combination of guadecitabine and cisplatin was tolerable and demonstrated activity in patients with platinum refractory germ cell cancer.
Collapse
Affiliation(s)
- Costantine Albany
- Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zeeshan Fazal
- Department of Comparative Biosciences and the Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ratnakar Singh
- Department of Comparative Biosciences and the Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Emmanuel Bikorimana
- Department of Comparative Biosciences and the Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Nabil Adra
- Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nasser H Hanna
- Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lawrence H Einhorn
- Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Susan M Perkins
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - George E Sandusky
- Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Harold Keer
- Astex Pharmaceuticals, Inc, Pleasanton, CA, USA
| | - Fang Fang
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, USA
| | - Kenneth P Nephew
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, USA
| | - Michael J Spinella
- Department of Comparative Biosciences and the Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
11
|
Zhao H, Wang C, Yu F, Guo Q. Decitabine combined with CAG regimen in the treatment of elderly patients with acute myeloid leukemia. Pak J Med Sci 2019; 36:141-145. [PMID: 32063948 PMCID: PMC6994887 DOI: 10.12669/pjms.36.2.850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: To analyze the efficacy and safety of decitabine combined with CAG ((cytarabine + aclacinomycin + granulocyte colony stimulating factor)) regimen and CAG regimen alone in the treatment of elderly acute myeloid leukemia. Methods: 96 elderly patients with acute myeloid leukemia who were admitted to our hospital from July 2015 to July 2017 were randomly divided into an observation group and a control group, 48 cases in each group. The patients in the control group were treated with CAG regimen, while the patients in the observation group were treated with decitabine on the basis of the control group. The clinical curative effect, changes of immune indicators, occurrence of adverse reactions and survival rate at different time after treatment were compared between the two groups. Results: The total effective rate of the observation group was significantly higher than that of the control group (P<0.05). After treatment, the indicators of cellular immunity in the two groups were significantly lower than those before treatment, and the indicators of cellular immunity in the observation group were significantly lower than those in the control group (P<0.05). There was no significant difference in the incidence of adverse reactions between the two groups (P>0.05). The 9-month survival rate and 1-year survival rate in the observation group were significantly higher than those in the control group (P<0.05). Conclusion: The combination of decitabine and CAG regimen is effective in the treatment of elderly patients with acute myeloid leukemia. The therapy can fully inhibit cellular immune function and improve long-term survival rate, and its safety has a small difference with that of CAG regimen alone. It is worth clinical promotion.
Collapse
Affiliation(s)
- Haitao Zhao
- Haitao Zhao, Department of Hematology, Binzhou People's Hospital, Shandong 256610, China
| | - Chunyan Wang
- Chunyan Wang, Department of Hematology, Binzhou People's Hospital, Shandong 256610, China
| | - Fengying Yu
- Fengying Yu, Department of Pharmaceutical, Binzhou People's Hospital, Shandong 256610, China
| | - Qingwei Guo
- Qingwei Guo, Department of Hematology, Qilu Children's Hospital of Shandong University, Shandong 250022, China
| |
Collapse
|
12
|
Qin H, Jiang Y, Zhang J, Deng C, Zhong Z. Oncoprotein Inhibitor Rigosertib Loaded in ApoE-Targeted Smart Polymersomes Reveals High Safety and Potency against Human Glioblastoma in Mice. Mol Pharm 2019; 16:3711-3719. [PMID: 31299161 DOI: 10.1021/acs.molpharmaceut.9b00691] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The unbiased cytotoxicity and blood-brain barrier (BBB) impermeability render common chemotherapeutics nonviable for treating glioblastoma (GBM) patients. Although rigosertib (RGS), a RAS effector protein inhibitor, has shown low toxicity to healthy cells and high efficacy toward various cancer cells by inactivating PI3K-Akt, it hardly overcomes the BBB barricade. Here, we report that RGS loaded in apolipoprotein E derived peptide (ApoE)-targeted chimaeric polymersomes (ApoE-CP) is safe and highly potent against human GBM in vivo. ApoE-CP exhibited stable loading of RGS in its lumen, giving RGS nanoformulations (ApoE-CP-RGS) with a size of 60 nm and reduction-triggered drug release behavior. Notably, ApoE-CP-RGS induction markedly enhanced the G2/M cell cycle arrest and inhibitory effect in U-87 MG glioblastoma cells compared with the nontargeted CP-RGS and free RGS. The therapeutic outcomes in orthotopic U-87 MG GBM models demonstrated that ApoE-CP-RGS brought about effective GBM inhibition, greatly prolonged survival time, and depleted adverse effects. Rigosertib formulated in ApoE-targeted chimaeric polymersomes has emerged as a novel, highly specific, efficacious, and nontoxic treatment for glioblastoma.
Collapse
Affiliation(s)
- Huazhen Qin
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| | - Yu Jiang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| | - Jian Zhang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| | - Chao Deng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| |
Collapse
|
13
|
Banerjee T, Calvi LM, Becker MW, Liesveld JL. Flaming and fanning: The Spectrum of inflammatory influences in myelodysplastic syndromes. Blood Rev 2019; 36:57-69. [PMID: 31036385 PMCID: PMC6711159 DOI: 10.1016/j.blre.2019.04.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/11/2019] [Accepted: 04/16/2019] [Indexed: 12/22/2022]
Abstract
The myelodysplastic syndromes (MDS) represent neoplasms derived from the expansion of mutated clonal hematopoietic cells which often demonstrate aberrant differentiation potential with resultant cytopenias and a propensity to evolve into acute myelogenous leukemia. While multiple mutations have been identified which may serve as drivers of the MDS clone, there is accumulating evidence that MDS clones and subclones are subject to modulation by the marrow microenvironment and its inflammatory milieu. There is also a strong link between autoimmune disorders and MDS. In this review, we examine the role of inflammatory cytokines, toll like receptors, pyroptosis, stromal cells, and cellular inflammatory mediators in MDS initiation, propagation, and progression. These contributions in a background of mutational, epigenetic, and aging changes in the marrow are also reviewed. Such inflammatory mediators may be subject to therapeutic agents which will enhance suppression of the MDS clone with potential to improve therapeutic outcomes in this disease which is usually incurable in aged patients not eligible for stem cell transplantation.
Collapse
Affiliation(s)
- Titas Banerjee
- Department of Medicine, University of Rochester, Rochester, NY, USA.
| | - Laura M Calvi
- Division of Endocrinology and Metabolism, Department of Medicine, and the James P Wilmot Cancer Institute, USA.
| | - Michael W Becker
- Division of Hematology/Oncology, Department of Medicine, James P Wilmot Cancer Institute, USA.
| | - Jane L Liesveld
- Division of Hematology/Oncology, Department of Medicine, James P Wilmot Cancer Institute, USA.
| |
Collapse
|
14
|
Gil-Perez A, Montalban-Bravo G. Management of myelodysplastic syndromes after failure of response to hypomethylating agents. Ther Adv Hematol 2019; 10:2040620719847059. [PMID: 31156799 PMCID: PMC6515843 DOI: 10.1177/2040620719847059] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023] Open
Abstract
Hypomethylating agents (HMAs) are the standard of care for patients with myelodysplastic syndrome (MDS). However, only around 50% of patients respond to these agents, and responses tend to be transient, with loss of response frequently happening within 2 years and being associated with very poor prognosis and limited therapeutic options. Identification of patients who will respond to HMAs is challenging. Mechanisms underlying resistance to HMAs are not clear yet. Recently, absence of response has been associated with increased cell-cycle quiescence among the hematopoietic progenitor cells. There are no standard-of-care options for patients after HMA failure. However, the increasing knowledge of MDS pathogenesis has led to the development of new potential therapies, including HMAs with longer half-life and exposure, inhibition of the antiapoptotic BCL2 protein with venetoclax or inhibition of immune-checkpoint regulatory proteins such as PD-1 or CTLA-4, innate immunity and targeting of CD33/CD3 with multiple monoclonal antibodies. In addition, multiple targeted agents are opening opportunities to treat subgroups of patients whose disease harbors mutations in TP53, IDH, FLT3, and genes involved in splicing machinery. Newer formulations of intensive chemotherapy and its different combinations may be considered a valid option in selected patients after HMA failure. Finally, decision making at the time of failure of response to HMAs should be personalized, taking into account that allogenic stem-cell transplantation remains the only therapeutic approach with curative potential in these patients. In the current review, we will focus on all the above aspects.
Collapse
Affiliation(s)
| | - Guillermo Montalban-Bravo
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77015, USA
| |
Collapse
|
15
|
Comont T, Delavigne K, Cougoul P, Bertoli S, Delabesse E, Fenaux P, Beyne-Rauzy O. [Management of myelodysplastic syndromes in 2019: An update]. Rev Med Interne 2019; 40:581-589. [PMID: 31054780 DOI: 10.1016/j.revmed.2019.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/10/2019] [Indexed: 01/04/2023]
Abstract
Myelodysplastic syndromes are a heterogeneous group of clonal myeloid disorders characterized by peripheral cytopenias and an increased risk of progression to acute myeloid leukemia. Inflammatory, auto-immune or syndromic symptoms can make the diagnosis difficult. Diagnosis is currently based on bone marrow cytology but cytogenetics and molecular features are currently overpassing their initial prognostic function (allowing early diagnosis and prediction of therapeutic response). The prognostic classification is based on the Revised International Prognostic Scoring System, which also provides guidance for therapeutic management. The treatment of low-risk myelodysplastic syndromes is based on the correction of cytopenias (erythropoiesis stimulating agents, transfusions, lenalidomide, etc.), whereas in high-risk group, the goal is the control of the leukemic clone (hypomethylating agents, allograft of hematopoietic stem cell transplantation). Other molecules are used to manage complications of cytopenias or transfusion (anti-infectious prophylaxis and treatments, martial chelation). New molecules are being studied with some interesting results (luspatercept, venetoclax). This article aims to provide an update on the knowledge that an internist should know for the practical management of myelodysplastic syndromes in 2019.
Collapse
Affiliation(s)
- T Comont
- Service de médecine interne et immunopathologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France; UFR Purpan, université Toulouse III Paul Sabatier, 31400 Toulouse, France; UMR1037-Inserm, ERL5294 CNRS, centre de recherche en cancérologie de Toulouse, 31100 Toulouse, France.
| | - K Delavigne
- Service de médecine interne et immunopathologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France
| | - P Cougoul
- Service de médecine interne et immunopathologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France
| | - S Bertoli
- UFR Purpan, université Toulouse III Paul Sabatier, 31400 Toulouse, France; UMR1037-Inserm, ERL5294 CNRS, centre de recherche en cancérologie de Toulouse, 31100 Toulouse, France; Service d'hématologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France
| | - E Delabesse
- Laboratoire d'hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopôle, 31100 Toulouse, France
| | - P Fenaux
- Service d'hématologie, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - O Beyne-Rauzy
- Service de médecine interne et immunopathologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France; UFR Purpan, université Toulouse III Paul Sabatier, 31400 Toulouse, France; UMR1037-Inserm, ERL5294 CNRS, centre de recherche en cancérologie de Toulouse, 31100 Toulouse, France
| |
Collapse
|
16
|
Short NJ, Jabbour E, Naqvi K, Patel A, Ning J, Sasaki K, Nogueras-Gonzalez GM, Bose P, Kornblau SM, Takahashi K, Andreeff M, Sanchez-Petitto G, Estrov Z, Dinardo CD, Montalban-Bravo G, Konopleva M, Alvarado Y, Bhalla KN, Fiskus W, Khouri M, Islam R, Kantarjian H, Garcia-Manero G. A phase II study of omacetaxine mepesuccinate for patients with higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia after failure of hypomethylating agents. Am J Hematol 2019; 94:74-79. [PMID: 30328139 DOI: 10.1002/ajh.25318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/04/2018] [Accepted: 10/15/2018] [Indexed: 11/09/2022]
Abstract
The outcome of patients with myelodysplastic syndromes (MDSs) after failure of hypomethylating agents (HMAs) failure is poor with a median overall survival (OS) of only 4-6 months. Omacetaxine mepesuccinate (OM) is safe and effective in myeloid malignancies but has not been studied in MDS with HMA failure. We conducted a phase II study of OM in patients with MDS or chronic myelomonocytic leukemia (CMML) who had previously failed or been intolerant to HMAs. Patients received OM at a dose of 1.25 mg/m2 subcutaneously every 12 hours for 3 consecutive days on a 4- to 7-week schedule. The primary endpoints were the overall response rate (ORR) and OS. A total of 42 patients were enrolled with a median age of 76 years. The ORR was 33%. Patients with diploid cytogenetics were more likely to respond to OM than were those with cytogenetic abnormalities (58% vs 23%, respectively; P = .03). Overall, the median OS was 7.5 months and 1-year OS rate was 25%. Patients with diploid cytogenetics had superior OS to those with cytogenetic abnormalities (median OS 14.8 vs 6.8 months, respectively; P = .01). Two patients had ongoing response to OM of 2 years or longer (both MDS with diploid cytogenetics and RUNX1 mutation). The most common grade ≥ 3 adverse events were infections in 11 patients (26%), febrile neutropenia in 4 (10%), and hemorrhage in 3 (7%). Overall, OM was safe and active in patients with MDS or CMML who experienced HMA failure. These results support the further development of OM in this setting.
Collapse
Affiliation(s)
- Nicholas J. Short
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Elias Jabbour
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Kiran Naqvi
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Ami Patel
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Jing Ning
- Department of Biostatistics; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Koji Sasaki
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | | | - Prithviraj Bose
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Steven M. Kornblau
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Koichi Takahashi
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Michael Andreeff
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Gabriela Sanchez-Petitto
- Department of Internal Medicine; The University of Texas Health Science Center at Houston; Houston Texas
| | - Zeev Estrov
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Courtney D. Dinardo
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | | | - Marina Konopleva
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Yesid Alvarado
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Kapil N. Bhalla
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Warren Fiskus
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Maria Khouri
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Rubiul Islam
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Hagop Kantarjian
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | | |
Collapse
|
17
|
Heudobler D, Klobuch S, Thomas S, Hahn J, Herr W, Reichle A. Cutaneous Leukemic Infiltrates Successfully Treated With Biomodulatory Therapy in a Rare Case of Therapy-Related High Risk MDS/AML. Front Pharmacol 2018; 9:1279. [PMID: 30483125 PMCID: PMC6243099 DOI: 10.3389/fphar.2018.01279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/18/2018] [Indexed: 12/29/2022] Open
Abstract
Cutaneous manifestations in hematologic malignancies, especially in leukemia, are not common and may be very variable. Here we report a very unusual case of a patient (female, 70 years old) who was admitted to the hospital in 2016 because of skin lesions on the face, the trunk of the body and the extremities. She had a history of breast cancer in the year 2004 (pT1b, pN0, cM0, L0, V0, R0) which had been resected and treated with adjuvant radiation and chemotherapy (cyclophosphamide, methotrexate, 5-fluorouracile) as well as psoriasis treated with methotrexate and cyclosporine. Because of mild cytopenia a bone marrow aspirate/biopsy was performed showing myelodysplastic syndrome (MDS) with multilineage dysplasia. Cytogenetic review revealed a complex aberrant karyotype denoting adverse outcome. Simultaneously, a skin biopsy could confirm leukemic skin infiltration. Consequently, a therapy with azacitidine was started. After the first cycle the patient developed severe pancytopenia with a percentage of 13% peripheral blasts (previously 0–2%) as well as fever without evidence for infection which was interpreted as progressive disease. Therefore, the therapeutic regimen was changed to a biomodulatory therapy consisting of low-dose azacitidine 75 mg/day (given sc d1-7 of 28), pioglitazone 45 mg/day per os, and all-trans-retinoic acid (ATRA) 45 mg/m2/day per os. After cycle 1 of this combined biomodulatory therapy the patient showed hematologic recovery; besides a mild anemia (hemoglobin 11.1 g/dl) she developed a normal blood count. Moreover, the cutaneous leukemic infiltrates which had been unaffected by the azacitidine ameliorated tremendously after 2 cycles resulting in a complete remission of the skin lesions after cycle 6. In conclusion, we report a very unusual case with cutaneous infiltrates being the first clinical manifestation of hematologic disease, preceding the development of acute myeloid leukemia. While azacitidine alone was ineffective, a combined biomodulatory approach resulted in a complete remission of the cutaneous manifestation.
Collapse
Affiliation(s)
- Daniel Heudobler
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Sebastian Klobuch
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Simone Thomas
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Joachim Hahn
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Albrecht Reichle
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
18
|
Steensma DP, Brunner AM, DeZern AE, Garcia-Manero G, Komrokji RS, Odenike OS, Roboz GJ, Savona MR, Stone RM, Sekeres MA. Low clinical trial accrual of patients with myelodysplastic syndromes: Causes and potential solutions. Cancer 2018; 124:4601-4609. [PMID: 30289970 DOI: 10.1002/cncr.31769] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/03/2018] [Accepted: 08/21/2018] [Indexed: 11/11/2022]
Abstract
Despite few effective therapies, only a small percentage of patients diagnosed with myelodysplastic syndromes (MDS) in the United States are enrolled in prospective, interventional clinical trials. MDS-specific barriers to trial accrual include a high frequency of elderly patients with comorbid conditions, atypical disease features and uncertainty regarding the diagnosis (because other nonclonal processes also can cause dysplasia and cytopenias), a history of another nonmyeloid neoplasm resulting in therapy-related MDS, rapid disease recurrence after allogeneic stem cell transplantation, and an arbitrary division between MDS and acute myeloid leukemia. In addition, barriers to accrual that are common to other oncology populations, such as difficulty traveling to clinical trial enrollment sites and narrow trial eligibility criteria, also prevent patients with MDS from enrolling in studies. Collectively these barriers must be assessed systematically, and creative solutions are needed to improve outcomes for this needy patient population.
Collapse
Affiliation(s)
- David P Steensma
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Andrew M Brunner
- Division of Hematology and Medical Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Amy E DeZern
- Division of Hematological Malignancies, Sidney Kimmel Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | | | - Rami S Komrokji
- Moffitt Cancer Center, Malignant Hematology Department, Tampa, Florida
| | - Olatoyosi S Odenike
- Division of Hematology and Medical Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Gail J Roboz
- Division of Hematology & Oncology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Michael R Savona
- Division of Hematology & Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Richard M Stone
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mikkael A Sekeres
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| |
Collapse
|
19
|
Abaza Y, Hidalgo-Lopez JE, Verstovsek S, Jabbour E, Ravandi F, Borthakur G, Estrov Z, Alvarado Y, Burger J, Schneider H, Soltysiak KA, Wei Y, Kantarjian HM, Bueso-Ramos CE, Garcia-Manero G. Phase I study of ruxolitinib in previously treated patients with low or intermediate-1 risk myelodysplastic syndrome with evidence of NF-kB activation. Leuk Res 2018; 73:78-85. [PMID: 30245189 DOI: 10.1016/j.leukres.2018.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/28/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Abstract
Therapeutic options for patients with lower-risk myelodysplastic syndrome (MDS) who have failed prior therapies are limited particularly after hypomethylating agent. Several studies have indicated that deregulation of innate immunity signaling is critical in the pathogenesis of MDS. This process involves Toll-like receptor stimulation, cytokine overexpression, and nuclear factor-kB (NF-kB) activation. Since ruxolitinib, a JAK1/JAK2 inhibitor, suppresses NF-kB expression, we conducted a phase 1 dose-escalation study to determine the safety and efficacy of ruxolitinib in previously treated lower-risk MDS patients with evidence of NF-kB activation. Nineteen patients, 8 with chronic myelomonocytic leukemia and 11 with MDS, were enrolled. No dose limiting toxicity was observed and the maximum tolerated dose was 20 mg twice daily. Responses were restricted to MDS patients with an overall response rate of 22% [hematological improvement in platelets (HI-P) = 2, hematological improvement in erythrocytes (HI-E) = 1, partial cytogenetic response (PCyR) = 1]. Of these patients, 2 relapsed (HI-P and PCyR) and 2 continue to be in HI-P and HI-E, respectively, with ongoing therapy. Meaningful improvement in bone marrow dysplasia was only seen in a patient who achieved HI-E. Phosphorylated p65 (pp65) decreased in 6 of 15 patients (40%) including the 2 patients with continued response to treatment and increased in a patient who relapsed after a short-lived HI-P. This suggests potential correlation between reduction in pp65 expression and response duration. In conclusion, ruxolitinib was well-tolerated in previously treated lower-risk MDS patients with evidence of NF-kB activation and resulted in low but significant frequency of responses. (NCT01895842).
Collapse
Affiliation(s)
- Yasmin Abaza
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Juliana E Hidalgo-Lopez
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Zeev Estrov
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Yesid Alvarado
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jan Burger
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Heather Schneider
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kelly A Soltysiak
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Yue Wei
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Carlos E Bueso-Ramos
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
20
|
Somatic mutations precede acute myeloid leukemia years before diagnosis. Nat Med 2018; 24:1015-1023. [PMID: 29988143 DOI: 10.1038/s41591-018-0081-z] [Citation(s) in RCA: 408] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/23/2018] [Indexed: 01/08/2023]
Abstract
The pattern of somatic mutations observed at diagnosis of acute myeloid leukemia (AML) has been well-characterized. However, the premalignant mutational landscape of AML and its impact on risk and time to diagnosis is unknown. Here we identified 212 women from the Women's Health Initiative who were healthy at study baseline, but eventually developed AML during follow-up (median time: 9.6 years). Deep sequencing was performed on peripheral blood DNA of these cases and compared to age-matched controls that did not develop AML. We discovered that mutations in IDH1, IDH2, TP53, DNMT3A, TET2 and spliceosome genes significantly increased the odds of developing AML. All subjects with TP53 mutations (n = 21 out of 21 patients) and IDH1 and IDH2 (n = 15 out of 15 patients) mutations eventually developed AML in our study. The presence of detectable mutations years before diagnosis suggests that there is a period of latency that precedes AML during which early detection, monitoring and interventional studies should be considered.
Collapse
|
21
|
Myelodysplastic syndromes current treatment algorithm 2018. Blood Cancer J 2018; 8:47. [PMID: 29795386 PMCID: PMC5967332 DOI: 10.1038/s41408-018-0085-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/09/2018] [Accepted: 02/15/2018] [Indexed: 01/03/2023] Open
Abstract
Myelodysplastic syndromes (MDS) include a group of clonal myeloid neoplasms characterized by cytopenias due to ineffective hematopoiesis, abnormal blood and marrow cell morphology, and a risk of clonal evolution and progression to acute myeloid leukemia (AML). Because outcomes for patients with MDS are heterogeneous, individual risk stratification using tools such as the revised International Prognostic Scoring System (IPSS-R) is important in managing patients-including selecting candidates for allogeneic hematopoietic stem cell transplantation (ASCT), the only potentially curative therapy for MDS. The IPSS-R can be supplemented by molecular genetic testing, since certain gene mutations such as TP53 influence risk independent of established clinicopathological variables. For lower risk patients with symptomatic anemia, treatment with erythropoiesis-stimulating agents (ESAs) or lenalidomide (especially for those with deletion of chromosome 5q) can ameliorate symptoms. Some lower risk patients may be candidates for immunosuppressive therapy, thrombopoiesis-stimulating agents, or a DNA hypomethylating agent (HMA; azacitidine or decitabine). Among higher risk patients, transplant candidates should undergo ASCT as soon as possible, with HMAs useful as a bridge to transplant. Non-transplant candidates should initiate HMA therapy and continue if tolerated until disease progression. Supportive care with transfusions and antimicrobial drugs as needed remains important in all groups.
Collapse
|