1
|
Wan X, Wang J, Fang F, Hu Y, Zhang Z, Tao Y, Zhang Y, Yu J, Wu Y, Zhou B, Yin H, Ma L, Li X, Zhuo R, Cheng W, Zhang S, Pan J, Lu J, Hu S. Super enhancer related gene ANP32B promotes the proliferation of acute myeloid leukemia by enhancing MYC through histone acetylation. Cancer Cell Int 2024; 24:81. [PMID: 38383388 PMCID: PMC10882810 DOI: 10.1186/s12935-024-03271-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/13/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a malignancy of the hematopoietic system, and childhood AML accounts for about 20% of pediatric leukemia. ANP32B, an important nuclear protein associated with proliferation, has been found to regulate hematopoiesis and CML leukemogenesis by inhibiting p53 activity. However, recent study suggests that ANP32B exerts a suppressive effect on B-cell acute lymphoblastic leukemia (ALL) in mice by activating PU.1. Nevertheless, the precise underlying mechanism of ANP32B in AML remains elusive. RESULTS Super enhancer related gene ANP32B was significantly upregulated in AML patients. The expression of ANP32B exhibited a negative correlation with overall survival. Knocking down ANP32B suppressed the proliferation of AML cell lines MV4-11 and Kasumi-1, along with downregulation of C-MYC expression. Additionally, it led to a significant decrease in H3K27ac levels in AML cell lines. In vivo experiments further demonstrated that ANP32B knockdown effectively inhibited tumor growth. CONCLUSIONS ANP32B plays a significant role in promoting tumor proliferation in AML. The downregulation of ANP32B induces cell cycle arrest and promotes apoptosis in AML cell lines. Mechanistic analysis suggests that ANP32B may epigenetically regulate the expression of MYC through histone H3K27 acetylation. ANP32B could serve as a prognostic biomarker and potential therapeutic target for AML patients.
Collapse
Affiliation(s)
- Xiaomei Wan
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatrics, The First Affiliated Hospital of Wannan Medical College, Wuhu, 24100, China
| | - Jianwei Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Yixin Hu
- Department of Hematology, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, Jiangsu, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Yanfang Tao
- Department of Hematology, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, Jiangsu, China
| | - Yongping Zhang
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Juanjuan Yu
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yumeng Wu
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Bi Zhou
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Suzhou Hospital of AnHui Medical University, Suzhou, 234000, China
| | - Hongli Yin
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Li Ma
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Xiaolu Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Ran Zhuo
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Wei Cheng
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatrics, The First Affiliated Hospital of Wannan Medical College, Wuhu, 24100, China
| | - Shuqi Zhang
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatrics, The First Affiliated Hospital of Wannan Medical College, Wuhu, 24100, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China.
| | - Jun Lu
- Department of Hematology, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, Jiangsu, China.
| | - Shaoyan Hu
- Department of Hematology, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, Jiangsu, China.
| |
Collapse
|
2
|
Yedier-Bayram O, Gokbayrak B, Kayabolen A, Aksu AC, Cavga AD, Cingöz A, Kala EY, Karabiyik G, Günsay R, Esin B, Morova T, Uyulur F, Syed H, Philpott M, Cribbs AP, Kung SHY, Lack NA, Onder TT, Bagci-Onder T. EPIKOL, a chromatin-focused CRISPR/Cas9-based screening platform, to identify cancer-specific epigenetic vulnerabilities. Cell Death Dis 2022; 13:710. [PMID: 35973998 PMCID: PMC9381743 DOI: 10.1038/s41419-022-05146-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/24/2022] [Accepted: 07/28/2022] [Indexed: 01/21/2023]
Abstract
Dysregulation of the epigenome due to alterations in chromatin modifier proteins commonly contribute to malignant transformation. To interrogate the roles of epigenetic modifiers in cancer cells, we generated an epigenome-wide CRISPR-Cas9 knockout library (EPIKOL) that targets a wide-range of epigenetic modifiers and their cofactors. We conducted eight screens in two different cancer types and showed that EPIKOL performs with high efficiency in terms of sgRNA distribution and depletion of essential genes. We discovered novel epigenetic modifiers that regulate triple-negative breast cancer (TNBC) and prostate cancer cell fitness. We confirmed the growth-regulatory functions of individual candidates, including SS18L2 and members of the NSL complex (KANSL2, KANSL3, KAT8) in TNBC cells. Overall, we show that EPIKOL, a focused sgRNA library targeting ~800 genes, can reveal epigenetic modifiers that are essential for cancer cell fitness under in vitro and in vivo conditions and enable the identification of novel anti-cancer targets. Due to its comprehensive epigenome-wide targets and relatively high number of sgRNAs per gene, EPIKOL will facilitate studies examining functional roles of epigenetic modifiers in a wide range of contexts, such as screens in primary cells, patient-derived xenografts as well as in vivo models.
Collapse
Affiliation(s)
- Ozlem Yedier-Bayram
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Bengul Gokbayrak
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Alisan Kayabolen
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Ali Cenk Aksu
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Ayse Derya Cavga
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Biostatistics, Bioinformatics and Data Management Core, KUTTAM, Istanbul, Türkiye
| | - Ahmet Cingöz
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Ezgi Yagmur Kala
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Goktug Karabiyik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Rauf Günsay
- Koç University School of Medicine, Istanbul, Türkiye
| | - Beril Esin
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Tunc Morova
- Koç University School of Medicine, Istanbul, Türkiye
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Fırat Uyulur
- Koç University Department of Computational Biology, Istanbul, Türkiye
| | - Hamzah Syed
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Biostatistics, Bioinformatics and Data Management Core, KUTTAM, Istanbul, Türkiye
- Koç University School of Medicine, Istanbul, Türkiye
| | - Martin Philpott
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Adam P Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Sonia H Y Kung
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Nathan A Lack
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Koç University School of Medicine, Istanbul, Türkiye
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Tamer T Onder
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye.
- Koç University School of Medicine, Istanbul, Türkiye.
| | - Tugba Bagci-Onder
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye.
- Koç University School of Medicine, Istanbul, Türkiye.
| |
Collapse
|
3
|
Xu H, Wen Y, Jin R, Chen H. Epigenetic modifications and targeted therapy in pediatric acute myeloid leukemia. Front Pediatr 2022; 10:975819. [PMID: 36147798 PMCID: PMC9485478 DOI: 10.3389/fped.2022.975819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy resulting from the genetic alterations and epigenetic dysregulations of the hematopoietic progenitor cells. One-third of children with AML remain at risk of relapse even though outcomes have improved in recent decades. Epigenetic dysregulations have been identified to play a significant role during myeloid leukemogenesis. In contrast to genetic changes, epigenetic modifications are typically reversible, opening the door to the development of epigenetic targeted therapy. In this review, we provide an overview of the landscape of epigenetic alterations and describe the current progress that has been made in epigenetic targeted therapy, and pay close attention to the potential value of epigenetic abnormalities in the precision and combinational therapy of pediatric AML.
Collapse
Affiliation(s)
- Huan Xu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxi Wen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo Chen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Cheng T, Kiser K, Grasse L, Iles L, Bartholomeusz G, Samaniego F, Orlowski RZ, Chandra J. Expression of histone deacetylase (HDAC) family members in bortezomib-refractory multiple myeloma and modulation by panobinostat. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:888-902. [PMID: 34888496 PMCID: PMC8653980 DOI: 10.20517/cdr.2021.44] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIM Multiple myeloma (MM) is a hematological malignancy of antibody-producing mature B cells or plasma cells. The proteasome inhibitor, bortezomib, was the first-in-class compound to be FDA approved for MM and is frequently utilized in induction therapy. However, bortezomib refractory disease is a major clinical concern, and the efficacy of the pan-histone deacetylase inhibitor (HDACi), panobinostat, in bortezomib refractory disease indicates that HDAC targeting is a viable strategy. Here, we utilized isogenic bortezomib resistant models to profile HDAC expression and define baseline and HDACi-induced expression patterns of individual HDAC family members in sensitive vs. resistant cells to better understanding the potential for targeting these enzymes. METHODS Gene expression of HDAC family members in two sets of isogenic bortezomib sensitive or resistant myeloma cell lines was examined. These cell lines were subsequently treated with HDAC inhibitors: panobinostat or vorinostat, and HDAC expression was evaluated. CRISPR/Cas9 knockdown and pharmacological inhibition of specific HDAC family members were conducted. RESULTS Interestingly, HDAC6 and HDAC7 were significantly upregulated and downregulated, respectively, in bortezomib-resistant cells. Panobinostat was effective at inducing cell death in these lines and modulated HDAC expression in cell lines and patient samples. Knockdown of HDAC7 inhibited cell growth while pharmacologically inhibiting HDAC6 augmented cell death by panobinostat. CONCLUSION Our data revealed heterogeneous expression of individual HDACs in bortezomib sensitive vs. resistant isogenic cell lines and patient samples treated with panobinostat. Cumulatively our findings highlight distinct roles for HDAC6 and HDAC7 in regulating cell death in the context of bortezomib resistance.
Collapse
Affiliation(s)
- Tiewei Cheng
- Department of Pediatrics Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kendall Kiser
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Leslie Grasse
- Department of Pediatrics Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lakesla Iles
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Geoffrey Bartholomeusz
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Felipe Samaniego
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Z Orlowski
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joya Chandra
- Department of Pediatrics Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
5
|
Bruserud Ø, Tsykunova G, Hernandez-Valladares M, Reikvam H, Tvedt THA. Therapeutic Use of Valproic Acid and All-Trans Retinoic Acid in Acute Myeloid Leukemia-Literature Review and Discussion of Possible Use in Relapse after Allogeneic Stem Cell Transplantation. Pharmaceuticals (Basel) 2021; 14:ph14050423. [PMID: 34063204 PMCID: PMC8147490 DOI: 10.3390/ph14050423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Even though allogeneic stem cell transplantation is the most intensive treatment for acute myeloid leukemia (AML), chemo-resistant leukemia relapse is still one of the most common causes of death for these patients, as is transplant-related mortality, i.e., graft versus host disease, infections, and organ damage. These relapse patients are not always candidates for additional intensive therapy or re-transplantation, and many of them have decreased quality of life and shortened expected survival. The efficiency of azacitidine for treatment of posttransplant AML relapse has been documented in several clinical trials. Valproic acid is an antiepileptic fatty acid that exerts antileukemic activity through histone deacetylase inhibition. The combination of valproic acid and all-trans retinoic acid (ATRA) is well tolerated even by unfit or elderly AML patients, and low-toxicity chemotherapy (e.g., azacitidine) can be added to this combination. The triple combination of azacitidine, valproic acid, and ATRA may therefore represent a low-intensity and low-toxicity alternative for these patients. In the present review, we review and discuss the general experience with valproic acid/ATRA in AML therapy and we discuss its possible use in low-intensity/toxicity treatment of post-allotransplant AML relapse. Our discussion is further illustrated by four case reports where combined treatments with sequential azacitidine/hydroxyurea, valproic acid, and ATRA were used.
Collapse
Affiliation(s)
- Øystein Bruserud
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway; (G.T.); (T.H.A.T.)
- Correspondence:
| | - Galina Tsykunova
- Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway; (G.T.); (T.H.A.T.)
| | - Maria Hernandez-Valladares
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, N-5021 Bergen, Norway;
| | - Hakon Reikvam
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway; (G.T.); (T.H.A.T.)
| | | |
Collapse
|
6
|
Assi R, Masri N, Abou Dalle I, El-Cheikh J, Bazarbachi A. Post-Transplant Maintenance Therapy for Patients with Acute Myeloid Leukemia: Current Approaches and the Need for More Trials. J Blood Med 2021; 12:21-32. [PMID: 33531851 PMCID: PMC7847363 DOI: 10.2147/jbm.s270015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/05/2021] [Indexed: 12/18/2022] Open
Abstract
Relapse rates following allogeneic stem cell transplantation for acute myeloid leukemia remain unacceptably high and a major cause of death. Maintenance therapies post-transplant administered either to patients with impending relapse or at high risk of relapse could present a strategy to improve survival and overall outcomes. With the increasing use of molecular and genomic characterization of the disease, more novel therapies became available as maintenance strategies. These options were, however, hindered by excessive toxicities, mostly hematologic, especially with the use of myeloablative conditioning regimens. Several key questions have also emerged including the efficacy of these therapies, the duration of maintenance, as well as the potential modulation of the graft and the immune microenvironment. These issues are further complicated by the paucity of well-designed prospective randomized clinical trials evaluating these agents. Future directions in this field should include better risk stratification and patient selection based on assays of minimal residual disease, as well as the incorporation of novel targets and pathways of leukemogenesis. In this article, we highlight the current evidence behind the use of post-transplant maintenance therapy, the optimal patient and disease selection, as well as the challenges faced by these strategies in an area that remains quite controversial. We will focus on therapies targeting leukemia stem cells that directly or indirectly modulate the allografted immune microenvironment and augment the graft-versus-leukemia impact.
Collapse
Affiliation(s)
- Rita Assi
- Division of Hematology-Oncology, Lebanese American University and Lebanese American University Medical Center-Rizk Hospital, Beirut, Lebanon
| | - Nohad Masri
- Division of Hematology-Oncology, Lebanese American University and Lebanese American University Medical Center-Rizk Hospital, Beirut, Lebanon
| | - Iman Abou Dalle
- Division of Hematology-Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jean El-Cheikh
- Division of Hematology-Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Bazarbachi
- Division of Hematology-Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
7
|
Thomopoulos TP, Bouhla A, Papageorgiou SG, Pappa V. Chronic myelomonocytic leukemia - a review. Expert Rev Hematol 2020; 14:59-77. [PMID: 33275852 DOI: 10.1080/17474086.2021.1860004] [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] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Chronic myelomonocytic leukemia (CMML) is a clonal myeloid neoplasm, denoted by overlapping myelodysplastic and myeloproliferative features, with poor overall survival and high transformation rate to acute myeloid leukemia. AREAS COVERED This review, following a thorough Medline search of pertinent published literature, discusses the diagnostic criteria, the pathogenesis, and the complex genetic landscape of the disease. Prognostication, response criteria, therapeutic management of patients, efficacy of established and novel treatment modalities are thoroughly reviewed. EXPERT OPINION Cytogenetic abnormalities and mutations in genes involved in epigenetic and transcriptional regulation, and cell-signaling are abundant in CMML and implicated in its complex pathogenesis. As presence of these mutations carry a prognostic impact, they are increasingly incorporated in risk-stratification schemes. Novel response criteria have been proposed, considering the unique features of the disease. Although allogeneic hematopoietic stem cell transplantation remains the only treatment with curative intent, it is reserved for a minority of patients; therefore, there is an unmet need for optimizing treatment modalities, such as hypomethylating agents, and introducing novel agents, which could substantially improve survival and quality of life of CMML patients. Clinical trials dedicated specifically to CMML are needed to explore the efficacy and safety of novel treatment modalities.
Collapse
Affiliation(s)
- Thomas P Thomopoulos
- 2 Department of Internal Medicine - Propaedeutic and Research Unit, National and Kapodistrian University of Athens, Medical School, University General Hospital "Attikon" , Athens, Greece
| | - Anthi Bouhla
- 2 Department of Internal Medicine - Propaedeutic and Research Unit, National and Kapodistrian University of Athens, Medical School, University General Hospital "Attikon" , Athens, Greece
| | - Sotirios G Papageorgiou
- 2 Department of Internal Medicine - Propaedeutic and Research Unit, National and Kapodistrian University of Athens, Medical School, University General Hospital "Attikon" , Athens, Greece
| | - Vasiliki Pappa
- 2 Department of Internal Medicine - Propaedeutic and Research Unit, National and Kapodistrian University of Athens, Medical School, University General Hospital "Attikon" , Athens, Greece
| |
Collapse
|
8
|
Jiang Y, Xu Z, Ma N, Yin L, Hao C, Li J. Effects of signaling pathway inhibitors on hematopoietic stem cells. Mol Med Rep 2020; 23:9. [PMID: 33179097 PMCID: PMC7687261 DOI: 10.3892/mmr.2020.11647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/17/2020] [Indexed: 12/23/2022] Open
Abstract
While there are numerous small molecule inhibitory drugs available for a wide range of signalling pathways, at present, they are generally not used in combination in clinical settings. Previous reports have reported that the effects of glycogen synthase kinase (GSK)3β, p38MAPK, mTOR and histone deacetylase signaling combined together to suppress the stem-like nature of hematopoietic stem cells (HSCs), driving these cells to differentiate, cease proliferating and thereby impairing normal hematopoietic functionality. The present study aimed to determine the effect of HDACs, mTOR, GSK-3β and p38MAPK inhibitor combinations on the efficient expansion of HSCs using flow cytometry. Moreover, it specifically aimed to determine how inhibitors of the GSK3β signaling pathway, in combination with inhibitors of P38MAPK and mTOR signaling or histone deacetylase (HDAC) inhibitors, could affect HSC expansion, with the goal of identifying novel combination strategies useful for the expansion of HSCs. The results indicated that p38MAPK and/or GSK3β inhibitors increased Lin− cell and Lin−Sca-1+c-kit+ (LSK) cell numbers in vitro. Taken together, these results suggested that a combination of p38MAPK and GSK3β signaling may regulate HSC differentiation in vitro. These findings further indicated that the suppression of p38MAPK and/or GSK3β signalling may modulate HSC differentiation and self-renewal to enhance HSC expansion.
Collapse
Affiliation(s)
- Yuyu Jiang
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Zhaofeng Xu
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Na Ma
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Lizhi Yin
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Caiqin Hao
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Jing Li
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| |
Collapse
|
9
|
Yan B, Claxton D, Huang S, Qiu Y. AML chemoresistance: The role of mutant TP53 subclonal expansion and therapy strategy. Exp Hematol 2020; 87:13-19. [PMID: 32569759 DOI: 10.1016/j.exphem.2020.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/02/2020] [Accepted: 06/17/2020] [Indexed: 12/24/2022]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous clonal disease characterized by the proliferation and accumulation of myeloid blast cells in the bone marrow, which eventually lead to hematopoietic failure. Chemoresistance presents as a major burden for therapy of AML patients. p53 is the most important tumor suppressor protein that regulates cellular response to various stress. It is also important for hematopoietic stem cell development and hematopoiesis. Mutation or deletion of TP53 has been found to be linked to cancer progression, therapy-related resistance, and poor prognosis. TP53 mutation occurs in less than 10% of AML patients; however, it represents a subset of AML with therapy resistance and poor outcome. In addition, there is a subgroup of patients with low-frequency TP53 mutations. The percentage ranges from 1% to 3% of all AML patients. These patients have outcomes comparable to those of the high-frequency TP53 mutation patients. TP53-mutated clones isolated from the parental cells exhibit a survival advantage under drug treatment compared with cells with wild-type TP53, and have a higher population of leukemia stem cell (LSC) marker-positive cells, a characteristic of chemo-resistant cells. Therefore, low-frequency TP53 mutation, which is currently underappreciated, is an important prognosis factor for AML patients. Epigenetic drugs, such as hypomethylating agent and histone deacetylase inhibitors, have been found effective in targeting TP53-mutated AML. Histone deacetylase inhibitors can preferentially target the TP53-mutated subpopulation by reactivating p53-targeted genes and by eradicating LSC marker-positive cells. Therefore, combined treatment with epigenetic drugs may represent a new therapeutic strategy for treatments of TP53-mutated AML.
Collapse
Affiliation(s)
- Bowen Yan
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine Hershey, PA
| | - David Claxton
- Department of Medicine, Pennsylvania State University College of Medicine Hershey, PA; Penn State Cancer Institute, Pennsylvania State University College of Medicine Hershey, PA
| | - Suming Huang
- Penn State Cancer Institute, Pennsylvania State University College of Medicine Hershey, PA; Department of Pediatrics, Pennsylvania State University College of Medicine Hershey, PA
| | - Yi Qiu
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine Hershey, PA; Penn State Cancer Institute, Pennsylvania State University College of Medicine Hershey, PA.
| |
Collapse
|
10
|
Chandhok NS, Lewis R, Prebet T. Hypomethylating agent based combinations in higher risk myelodysplastic syndrome. Leuk Lymphoma 2020; 61:1012-1027. [PMID: 31814484 DOI: 10.1080/10428194.2019.1697812] [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] [Indexed: 12/11/2022]
Abstract
For over a decade the hypomethylating agents (HMA) azacitidine and decitabine have been the mainstay of therapy for myelodysplastic syndrome (MDS). There is a critical need to improve frontline therapy, given that only up to half of high-risk MDS patients will respond to HMA therapy, and responses are short-lived. Currently, a key strategy has been to combine HMAs with other novel agents to improve patient outcomes. While synergy of agents is the goal of combination therapy, combinations often come at the cost of increased side effects that are often intolerable in this vulnerable population. The purpose of this review is to critically examine clinically relevant HMA combinations and discuss the future of MDS management.
Collapse
Affiliation(s)
- Namrata S Chandhok
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Russell Lewis
- Smilow Cancer Center at Yale New Haven Hospital, New Haven, CT, USA
| | - Thomas Prebet
- Smilow Cancer Center at Yale New Haven Hospital, New Haven, CT, USA
| |
Collapse
|
11
|
Xu QY, Yu L. Epigenetic therapies in acute myeloid leukemia: the role of hypomethylating agents, histone deacetylase inhibitors and the combination of hypomethylating agents with histone deacetylase inhibitors. Chin Med J (Engl) 2020; 133:699-715. [PMID: 32044818 PMCID: PMC7190219 DOI: 10.1097/cm9.0000000000000685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Indexed: 12/24/2022] Open
Abstract
Epigenetic regulation includes changes of DNA methylation and modifications of histone proteins and is essential for normal physiologic functions, especially for controlling gene expression. Epigenetic dysregulation plays a key role in disease pathogenesis and progression of some malignancies, including acute myeloid leukemia (AML). Epigenetic therapies, including hypomethylating agents (HMAs) and histone deacetylase (HDAC) inhibitors, were developed to reprogram the epigenetic abnormalities in AML. However, the molecular mechanisms and therapeutic effects of the two agents alone or their combination remain unknown. An overview of these epigenetic therapies is given here. A literature search was conducted through PubMed database, looking for important biological or clinical studies related to the epigenetic regimens in the treatment of AML until October 15th, 2019. Various types of articles, including original research and reviews, were assessed, identified, and eventually summarized as a collection of data pertaining the mechanisms and clinical effects of HMAs and HDAC inhibitors in AML patients. We provided here an overview of the current understanding of the mechanisms and clinical therapeutic effects involved in the treatment with HMAs and HDAC inhibitors alone, the combination of epigenetic therapies with intensive chemotherapy, and the combination of both types of epigenetic therapies. Relevant clinical trials were also discussed. Generally speaking, the large number of studies and their varied outcomes demonstrate that effects of epigenetic therapies are heterogeneous, and that HMAs combination regimens probably contribute to significant response rates. However, more research is needed to explore therapeutic effects of HDAC inhibitors and various combinations of HMAs and HDAC inhibitors.
Collapse
Affiliation(s)
- Qing-Yu Xu
- Department of Hematology-Oncology, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen, Guangdong 518000, China
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim 68169, Germany
| | - Li Yu
- Department of Hematology-Oncology, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen, Guangdong 518000, China
| |
Collapse
|
12
|
Shallis RM, Boddu PC, Bewersdorf JP, Zeidan AM. The golden age for patients in their golden years: The progressive upheaval of age and the treatment of newly-diagnosed acute myeloid leukemia. Blood Rev 2020; 40:100639. [DOI: 10.1016/j.blre.2019.100639] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/25/2022]
|
13
|
The Development Process: from SAHA to Hydroxamate HDAC Inhibitors with Branched CAP Region and Linear Linker. Chem Biodivers 2019; 17:e1900427. [DOI: 10.1002/cbdv.201900427] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/02/2019] [Indexed: 12/31/2022]
|
14
|
Varadarajan I, Ballen KK. What have we learned from transplanting older patients? Best Pract Res Clin Haematol 2019; 32:101110. [PMID: 31779974 DOI: 10.1016/j.beha.2019.101110] [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] [Indexed: 11/26/2022]
Abstract
Aggressive curative therapies have now been extended to patients older than 65 years, a fast-growing segment of the population. As the number of allogeneic transplants in patients older than age 65 is increasing, attention is now focused on improving outcomes in this group. This paper discusses important aspects of allogeneic transplant in the older patient, focusing on donor and patient selection, choice of conditioning regimen and graft source, and the importance of timely access to a transplant center.
Collapse
Affiliation(s)
- Indumathy Varadarajan
- University of Virginia Cancer Center, 1300 Jefferson Park Avenue, Charlottesville, VA, 22908, USA
| | - Karen K Ballen
- University of Virginia Cancer Center, 1300 Jefferson Park Avenue, Charlottesville, VA, 22908, USA.
| |
Collapse
|
15
|
Targeting epigenetic modifications in cancer therapy: erasing the roadmap to cancer. Nat Med 2019; 25:403-418. [PMID: 30842676 DOI: 10.1038/s41591-019-0376-8] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 01/25/2019] [Indexed: 12/31/2022]
Abstract
Epigenetic dysregulation is a common feature of most cancers, often occurring directly through alteration of epigenetic machinery. Over the last several years, a new generation of drugs directed at epigenetic modulators have entered clinical development, and results from these trials are now being disclosed. Unlike first-generation epigenetic therapies, these new agents are selective, and many are targeted to proteins which are mutated or translocated in cancer. This review will provide a summary of the epigenetic modulatory agents currently in clinical development and discuss the opportunities and challenges in their development. As these drugs advance in the clinic, drug discovery has continued with a focus on both novel and existing epigenetic targets. We will provide an overview of these efforts and the strategies being employed.
Collapse
|
16
|
Zhang Q, Wang S, Chen J, Yu Z. Histone Deacetylases (HDACs) Guided Novel Therapies for T-cell lymphomas. Int J Med Sci 2019; 16:424-442. [PMID: 30911277 PMCID: PMC6428980 DOI: 10.7150/ijms.30154] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022] Open
Abstract
T-cell lymphomas are a heterogeneous group of cancers with different pathogenesis and poor prognosis. Histone deacetylases (HDACs) are epigenetic modifiers that modulate many key biological processes. In recent years, HDACs have been fully investigated for their roles and potential as drug targets in T-cell lymphomas. In this review, we have deciphered the modes of action of HDACs, HDAC inhibitors as single agents, and HDACs guided combination therapies in T-cell lymphomas. The overview of HDACs on the stage of T-cell lymphomas, and HDACs guided therapies both as single agents and combination regimens endow great opportunities for the cure of T-cell lymphomas.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Shaobin Wang
- Health Management Center of Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Junhui Chen
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Zhendong Yu
- China Central Laboratory of Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| |
Collapse
|
17
|
Bewersdorf JP, Stahl M, Zeidan AM. Are we witnessing the start of a therapeutic revolution in acute myeloid leukemia? Leuk Lymphoma 2019; 60:1354-1369. [DOI: 10.1080/10428194.2018.1546854] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jan Philipp Bewersdorf
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Maximilian Stahl
- Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
18
|
Bewersdorf JP, Shallis R, Stahl M, Zeidan AM. Epigenetic therapy combinations in acute myeloid leukemia: what are the options? Ther Adv Hematol 2019; 10:2040620718816698. [PMID: 30719265 PMCID: PMC6348528 DOI: 10.1177/2040620718816698] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/08/2018] [Indexed: 01/01/2023] Open
Abstract
Epigenetics refers to the regulation of gene expression mainly by changes in DNA methylation and modifications of histone proteins without altering the actual DNA sequence. While epigenetic modifications are essential for normal cell differentiation, several driver mutations in leukemic pathogenesis have been identified in genes that affect epigenetic processes, such as DNA methylation and histone acetylation. Several therapeutic options to target epigenetic alterations in acute myeloid leukemia (AML) have been successfully tested in preclinical studies and various drugs have already been approved for use in clinical practice. Among these already approved therapeutics are hypomethylating agents (azacitidine and decitabine) and isocitrate dehydrogenase inhibitors (ivosidenib, enasidenib). Other agents such as bromodomain-containing epigenetic reader proteins and histone methylation (e.g. DOT1L) inhibitors are currently in advanced clinical testing. As several epigenetic therapies have only limited efficacy when used as single agents, combination therapies that target AML pathogenesis at different levels and exploit synergistic mechanisms are also in clinical trials. Combinations of either epigenetic therapies with conventional chemotherapy, different forms of epigenetic therapies, or epigenetic therapies with immunotherapy are showing promising early results. In this review we summarize the underlying pathophysiology and rationale for epigenetically-based combination therapies, review current preclinical and clinical data and discuss the future directions of epigenetic therapy combinations in AML.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Rory Shallis
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Maximilian Stahl
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, 333 Cedar Street, PO Box 208028, New Haven, CT 06520-8055, USA
| |
Collapse
|
19
|
Yang F, Zhao N, Ge D, Chen Y. Next-generation of selective histone deacetylase inhibitors. RSC Adv 2019; 9:19571-19583. [PMID: 35519364 PMCID: PMC9065321 DOI: 10.1039/c9ra02985k] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022] Open
Abstract
Histone deacetylases (HDACs) are clinically validated epigenetic drug targets for cancer treatment. HDACs inhibitors (HDACis) have been successfully applied against a series of cancers. First-generation inhibitors are mainly pan-HDACis that target multiple isoforms which might lead to serious side effects. At present, the next-generation HDACis are mainly focused on being class- or isoform-selective which can provide improved risk–benefit profiles compared to non-selective inhibitors. Because of the rapid development in next-generation HDACis, it is necessary to have an updated and state-of-the-art overview. Here, we summarize the strategies and achievements of the selective HDACis. Histone deacetylases (HDACs) are clinically validated epigenetic drug targets for cancer treatment.![]()
Collapse
Affiliation(s)
- Feifei Yang
- School of Biological Science and Technology
- University of Jinan
- Jinan
- China
- Shanghai Key Laboratory of Regulatory Biology
| | - Na Zhao
- School of Biological Science and Technology
- University of Jinan
- Jinan
- China
| | - Di Ge
- School of Biological Science and Technology
- University of Jinan
- Jinan
- China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology
- The Institute of Biomedical Sciences and School of Life Sciences
- East China Normal University
- Shanghai
- China
| |
Collapse
|
20
|
Yang F, Shan P, Zhao N, Ge D, Zhu K, Jiang CS, Li P, Zhang H. Development of hydroxamate-based histone deacetylase inhibitors containing 1,2,4-oxadiazole moiety core with antitumor activities. Bioorg Med Chem Lett 2018; 29:15-21. [PMID: 30455152 DOI: 10.1016/j.bmcl.2018.11.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/17/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022]
Abstract
Histone deacetylases (HDACs) has proved to be promising target for the development of antitumor drugs. In this study, we reported the design and synthesis of a class of novel hydroxamate-based bis-substituted aromatic amide HDAC inhibitors with 1,2,4-oxadiazole core. Most newly synthesized compounds displayed excellent HDAC1 inhibitory effects and significant anti-proliferative activities. Among them, compounds 11a and 11c increased acetylation of histone H3 and H4 in dose-dependent manner. Furthermore, 11a and 11c remarkably induced apoptosis in HepG2 cancer cells. Finally, the high potency of compound 11a was rationalized by molecular docking studies.
Collapse
Affiliation(s)
- Feifei Yang
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Peipei Shan
- Institute for Translation Medicine, Qingdao University, Qingdao, Shandong Province 266071, China
| | - Na Zhao
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Di Ge
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Kongkai Zhu
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Peifeng Li
- Institute for Translation Medicine, Qingdao University, Qingdao, Shandong Province 266071, China.
| | - Hua Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China.
| |
Collapse
|
21
|
Ungerstedt JS. Epigenetic Modifiers in Myeloid Malignancies: The Role of Histone Deacetylase Inhibitors. Int J Mol Sci 2018; 19:ijms19103091. [PMID: 30304859 PMCID: PMC6212943 DOI: 10.3390/ijms19103091] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/01/2018] [Accepted: 10/05/2018] [Indexed: 01/18/2023] Open
Abstract
Myeloid hematological malignancies are clonal bone marrow neoplasms, comprising of acute myeloid leukemia (AML), the myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia (CMML), the myeloproliferative neoplasms (MPN) and systemic mastocytosis (SM). The field of epigenetic regulation of normal and malignant hematopoiesis is rapidly growing. In recent years, heterozygous somatic mutations in genes encoding epigenetic regulators have been found in all subtypes of myeloid malignancies, supporting the rationale for treatment with epigenetic modifiers. Histone deacetylase inhibitors (HDACi) are epigenetic modifiers that, in vitro, have been shown to induce growth arrest, apoptotic or autophagic cell death, and terminal differentiation of myeloid tumor cells. These effects were observed both at the bulk tumor level and in the most immature CD34+38− cell compartments containing the leukemic stem cells. Thus, there is a strong rationale supporting HDACi therapy in myeloid malignancies. However, despite initial promising results in phase I trials, HDACi in monotherapy as well as in combination with other drugs, have failed to improve responses or survival. This review provides an overview of the rationale for HDACi in myeloid malignancies, clinical results and speculations on why clinical trials have thus far not met the expectations, and how this may be improved in the future.
Collapse
Affiliation(s)
- Johanna S Ungerstedt
- Department of Medicine, Huddinge, Karolinska Institutet, and Hematology Center, and Karolinska University Hospital, S-141 86 Stockholm, Sweden.
| |
Collapse
|
22
|
Post-remission strategies for the prevention of relapse following allogeneic hematopoietic cell transplantation for high-risk acute myeloid leukemia: expert review from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Bone Marrow Transplant 2018; 54:519-530. [PMID: 30104717 DOI: 10.1038/s41409-018-0286-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 01/06/2023]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematopoietic malignancy generally associated with poor prognosis. Allogeneic hematopoietic cell transplantation (alloHCT) continues to be the most potent anti-leukemia treatment for adult patients with intermediate and high-risk AML. However, disease relapse after alloHCT remains unacceptably high and is the primary cause of treatment failure and mortality following alloHCT. It is therefore that post-transplant early cellular or pharmacologic maintenance or preemptive strategies to enhance the graft-versus-leukemia effect or to eradicate persistent minimal residual disease have been of renewed interest, particularly with the availability of more sensitive technologies to measure residual AML. Although preliminary studies have demonstrated improved outcomes with the use of post-alloHCT remission therapies, prospective randomized trials are required to determine their clinical efficacy and role in the treatment of AML. On behalf of the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation, we summarize the available evidence on the use and efficacy of available pharmacologic post-remission therapies, including hypomethylating agents, deacetylase inhibitors, and tyrosine kinase inhibitors, as well as cellular therapies, including preemptive and prophylactic donor lymphocyte infusions for the prevention of relapse of AML.
Collapse
|
23
|
Vega-García N, Malatesta R, Estella C, Pérez-Jaume S, Esperanza-Cebollada E, Torrebadell M, Català A, Gassiot S, Berrueco R, Ruiz-Llobet A, Alonso-Saladrigues A, Mesegué M, Pont-Martí S, Rives S, Camós M. Paediatric patients with acute leukaemia andKMT2A (MLL)rearrangement show a distinctive expression pattern of histone deacetylases. Br J Haematol 2018; 182:542-553. [DOI: 10.1111/bjh.15436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/26/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Nerea Vega-García
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Roberta Malatesta
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Camino Estella
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Sara Pérez-Jaume
- Developmental Tumor Biology Laboratory; Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Elena Esperanza-Cebollada
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Montserrat Torrebadell
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
| | - Albert Català
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Susanna Gassiot
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Rubén Berrueco
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Anna Ruiz-Llobet
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Anna Alonso-Saladrigues
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Montserrat Mesegué
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Sandra Pont-Martí
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Susana Rives
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Mireia Camós
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
| |
Collapse
|
24
|
Kubasch AS, Platzbecker U. Beyond the Edge of Hypomethylating Agents: Novel Combination Strategies for Older Adults with Advanced MDS and AML. Cancers (Basel) 2018; 10:E158. [PMID: 29795051 PMCID: PMC6025349 DOI: 10.3390/cancers10060158] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/19/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022] Open
Abstract
Higher-risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) of the elderly exhibit several commonalities, including first line treatment with hypomethylating agents (HMA) like azacitidine (AZA) or decitabine (DAC). Until today, response to treatment occurs in less than 50 percent of patients, and is often short-lived. Moreover, patients failing HMA have a dismal prognosis. Current developments include combinations of HMA with novel drugs targeting epigenetic or immunomodulatory pathways. Other efforts focus on the prevention of resistance to HMA using checkpoint inhibitors to enhance immune attack. This review focuses on recent advances in the field of HMA-based front-line therapies in elderly patients with myeloid diseases.
Collapse
Affiliation(s)
- Anne Sophie Kubasch
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
| | - Uwe Platzbecker
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), 01307 Dresden, Germany.
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
25
|
Abstract
Acute myeloid leukemia (AML) is one of the best studied malignancies, and significant progress has been made in understanding the clinical implications of its disease biology. Unfortunately, drug development has not kept pace, as the '7+3' induction regimen remains the standard of care for patients fit for intensive therapy 40 years after its first use. Temporal improvements in overall survival were mostly confined to younger patients and driven by improvements in supportive care and use of hematopoietic stem cell transplantation. Multiple forms of novel therapy are currently in clinical trials and are attempting to bring bench discoveries to the bedside to benefit patients. These novel therapies include improved chemotherapeutic agents, targeted molecular inhibitors, cell cycle regulators, pro-apoptotic agents, epigenetic modifiers, and metabolic therapies. Immunotherapies in the form of vaccines; naked, conjugated and bispecific monoclonal antibodies; cell-based therapy; and immune checkpoint inhibitors are also being evaluated in an effort to replicate the success seen in other malignancies. Herein, we review the scientific basis of these novel therapeutic approaches, summarize the currently available evidence, and look into the future of AML therapy by highlighting key clinical studies and the challenges the field continues to face.
Collapse
|
26
|
Newcombe AA, Gibson BES, Keeshan K. Harnessing the potential of epigenetic therapies for childhood acute myeloid leukemia. Exp Hematol 2018; 63:1-11. [PMID: 29608923 DOI: 10.1016/j.exphem.2018.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/19/2018] [Accepted: 03/27/2018] [Indexed: 12/31/2022]
Abstract
There is a desperate need for new and effective therapeutic approaches to acute myeloid leukemia (AML) in both children and adults. Epigenetic aberrations are common in adult AML, and many novel epigenetic compounds that may improve patient outcomes are in clinical development. Mutations in epigenetic regulators occur less frequently in AML in children than in adults. Investigating the potential benefits of epigenetic therapy in pediatric AML is an important issue and is discussed in this review.
Collapse
Affiliation(s)
| | - Brenda E S Gibson
- Department of Paediatric Haematology, Royal Hospital for Children, Glasgow, UK
| | - Karen Keeshan
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
27
|
Lee SI, Celik S, Logsdon BA, Lundberg SM, Martins TJ, Oehler VG, Estey EH, Miller CP, Chien S, Dai J, Saxena A, Blau CA, Becker PS. A machine learning approach to integrate big data for precision medicine in acute myeloid leukemia. Nat Commun 2018; 9:42. [PMID: 29298978 PMCID: PMC5752671 DOI: 10.1038/s41467-017-02465-5] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 11/30/2017] [Indexed: 02/06/2023] Open
Abstract
Cancers that appear pathologically similar often respond differently to the same drug regimens. Methods to better match patients to drugs are in high demand. We demonstrate a promising approach to identify robust molecular markers for targeted treatment of acute myeloid leukemia (AML) by introducing: data from 30 AML patients including genome-wide gene expression profiles and in vitro sensitivity to 160 chemotherapy drugs, a computational method to identify reliable gene expression markers for drug sensitivity by incorporating multi-omic prior information relevant to each gene’s potential to drive cancer. We show that our method outperforms several state-of-the-art approaches in identifying molecular markers replicated in validation data and predicting drug sensitivity accurately. Finally, we identify SMARCA4 as a marker and driver of sensitivity to topoisomerase II inhibitors, mitoxantrone, and etoposide, in AML by showing that cell lines transduced to have high SMARCA4 expression reveal dramatically increased sensitivity to these agents. Identification of markers of drug response is essential for precision therapy. Here the authors introduce an algorithm that uses prior information about each gene’s importance in AML to identify the most predictive gene-drug associations from transcriptome and drug response data from 30 AML samples.
Collapse
Affiliation(s)
- Su-In Lee
- Paul G. Allen School of Computer Science and Engineering, University of Washington, 185 E Stevens Way NE, Seattle, WA, 98195, USA. .,Department of Genome Sciences, University of Washington, 3720 15th Ave NE, Seattle, WA, 98195, USA. .,Center for Cancer Innovation, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA.
| | - Safiye Celik
- Paul G. Allen School of Computer Science and Engineering, University of Washington, 185 E Stevens Way NE, Seattle, WA, 98195, USA
| | | | - Scott M Lundberg
- Paul G. Allen School of Computer Science and Engineering, University of Washington, 185 E Stevens Way NE, Seattle, WA, 98195, USA
| | - Timothy J Martins
- Quellos High Throughput Screening Core, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - Vivian G Oehler
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA.,Division of Hematology, Department of Medicine and Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - Elihu H Estey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA.,Division of Hematology, Department of Medicine and Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - Chris P Miller
- Division of Hematology, Department of Medicine and Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - Sylvia Chien
- Division of Hematology, Department of Medicine and Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - Jin Dai
- Division of Hematology, Department of Medicine and Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - Akanksha Saxena
- Division of Hematology, Department of Medicine and Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - C Anthony Blau
- Center for Cancer Innovation, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA.,Division of Hematology, Department of Medicine and Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - Pamela S Becker
- Center for Cancer Innovation, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA.,Division of Hematology, Department of Medicine and Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| |
Collapse
|
28
|
Hay JF, Lappin K, Liberante F, Kettyle LM, Matchett KB, Thompson A, Mills KI. Integrated analysis of the molecular action of Vorinostat identifies epi-sensitised targets for combination therapy. Oncotarget 2017; 8:67891-67903. [PMID: 28978082 PMCID: PMC5620222 DOI: 10.18632/oncotarget.18910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 05/31/2017] [Indexed: 11/25/2022] Open
Abstract
Several histone deacetylase inhibitors including Vorinostat have received FDA approval for the treatment of haematological malignancies. However, data from these trials indicate that Vorinostat has limited efficacy as a monotherapy, prompting the need for rational design of combination therapies. A number of epi-sensitised pathways, including sonic hedgehog (SHH), were identified in AML cells by integration of global patterns of histone H3 lysine 9 (H3K9) acetylation with transcriptomic analysis following Vorinostat-treatment. Direct targeting of the SHH pathway with SANT-1, following Vorinostat induced epi-sensitisation, resulted in synergistic cell death of AML cells. In addition, xenograft studies demonstrated that combination therapy induced a marked reduction in leukemic burden compared to control or single agents. Together, the data supports epi-sensitisation as a potential component of the strategy for the rational development of combination therapies in AML.
Collapse
Affiliation(s)
- Jodie F Hay
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.,Molecular Oncology Laboratory, MRC - University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Katrina Lappin
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Fabio Liberante
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.,Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Laura M Kettyle
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.,Haematopoietic Stem Cell Biology, MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Kyle B Matchett
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Alexander Thompson
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.,Division of Cancer and Stem Cells, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Ken I Mills
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| |
Collapse
|
29
|
|
30
|
Uy N, Singh A, Gore SD, Prebet T. Hypomethylating agents (HMA) treatment for myelodysplastic syndromes: alternatives in the frontline and relapse settings. Expert Opin Pharmacother 2017; 18:1213-1224. [PMID: 28675065 PMCID: PMC6121132 DOI: 10.1080/14656566.2017.1349100] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/27/2017] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Hypomethylating agents (HMA) have played a pivotal role for treating myelodysplastic syndromes (MDS) over the past decade, inducing sustained hematological responses and delaying progression to leukemia. However, a vast majority of patients will experience treatment failure within 2 years, with poor prognoses and limited options, and management of this growing patient population remains unclear. Areas covered: With the introduction of new agents in the MDS field, a better understanding of the biology of MDS, and updated information on standard of care options (including allogeneic transplantation), we re-evaluate the global treatment strategy in MDS via novel agents, focusing in particular on investigational approaches for patients who fail to respond to HMA when applicable. This review aims to address two questions: what are reasonable alternatives to HMA in MDS, and what strategies can be used for patients experiencing HMA failure. Expert opinion/commentary: HMA therapy remains a mainstay of treatment, even if additional research is still warranted to maximize its benefits for the different groups of patients. The outcome of patients experiencing HMA failure remains grim, without standard of care, but several new approaches seem promising, as there is an increasing focus on studying treatments for patients refractory to HMA treatment.
Collapse
Affiliation(s)
- Natalie Uy
- a Section of Hematology, Department of Internal Medicine , Yale School of Medicine , New Haven , CT , USA
| | - Abhay Singh
- a Section of Hematology, Department of Internal Medicine , Yale School of Medicine , New Haven , CT , USA
| | - Steven D Gore
- a Section of Hematology, Department of Internal Medicine , Yale School of Medicine , New Haven , CT , USA
| | - Thomas Prebet
- a Section of Hematology, Department of Internal Medicine , Yale School of Medicine , New Haven , CT , USA
| |
Collapse
|
31
|
Zeidan AM, Stahl M, Sekeres MA, Steensma DP, Komrokji RS, Gore SD. A call for action: Increasing enrollment of untreated patients with higher-risk myelodysplastic syndromes in first-line clinical trials. Cancer 2017; 123:3662-3672. [DOI: 10.1002/cncr.30903] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/22/2017] [Accepted: 06/12/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Amer M. Zeidan
- Department of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center; Yale University School of Medicine; New Haven Connecticut
| | - Maximilian Stahl
- Department of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center; Yale University School of Medicine; New Haven Connecticut
| | | | - David P. Steensma
- Dana-Farber Cancer Institute; Harvard Medical School; Boston Massachusetts
| | - Rami S. Komrokji
- H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Steven D. Gore
- Department of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center; Yale University School of Medicine; New Haven Connecticut
| |
Collapse
|
32
|
Gbolahan OB, Zeidan AM, Stahl M, Abu Zaid M, Farag S, Paczesny S, Konig H. Immunotherapeutic Concepts to Target Acute Myeloid Leukemia: Focusing on the Role of Monoclonal Antibodies, Hypomethylating Agents and the Leukemic Microenvironment. Int J Mol Sci 2017; 18:E1660. [PMID: 28758974 PMCID: PMC5578050 DOI: 10.3390/ijms18081660] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/22/2017] [Accepted: 07/24/2017] [Indexed: 12/20/2022] Open
Abstract
Intensive chemotherapeutic protocols and allogeneic stem cell transplantation continue to represent the mainstay of acute myeloid leukemia (AML) treatment. Although this approach leads to remissions in the majority of patients, long-term disease control remains unsatisfactory as mirrored by overall survival rates of approximately 30%. The reason for this poor outcome is, in part, due to various toxicities associated with traditional AML therapy and the limited ability of most patients to tolerate such treatment. More effective and less toxic therapies therefore represent an unmet need in the management of AML, a disease for which therapeutic progress has been traditionally slow when compared to other cancers. Several studies have shown that leukemic blasts elicit immune responses that could be exploited for the development of novel treatment concepts. To this end, early phase studies of immune-based therapies in AML have delivered encouraging results and demonstrated safety and feasibility. In this review, we discuss opportunities for immunotherapeutic interventions to enhance the potential to achieve a cure in AML, thereby focusing on the role of monoclonal antibodies, hypomethylating agents and the leukemic microenvironment.
Collapse
Affiliation(s)
- Olumide Babajide Gbolahan
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Amer M Zeidan
- Department of Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Maximilian Stahl
- Department of Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Mohammad Abu Zaid
- Department of Medicine, Bone Marrow and Stem Cell Transplantation, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Sherif Farag
- Department of Medicine, Bone Marrow and Stem Cell Transplantation, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Sophie Paczesny
- Wells Center for Pediatric Research, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Heiko Konig
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
33
|
Santini V. First-line Therapeutic Strategies for Myelodysplastic Syndromes. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17S:S31-S36. [DOI: 10.1016/j.clml.2017.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 01/27/2023]
|
34
|
Abstract
INTRODUCTION Myelofibrosis (MF) is characterized by bone marrow fibrosis with subsequent extramedullary hematopoiesis and abnormal cytokine expression leading to splenomegaly, constitutional symptoms and cytopenias. The discovery of the JAK2 V617F mutation in the majority of MF patients has been followed by significant progress in drug development for MF. Areas covered: In this article, we review advances in the understanding of the underlying disease biology, prognostic assessment and therapeutic modalities for MF. We provide clinical trial evidence behind using the JAK2 inhibitor ruxolitinib, erythropoiesis stimulating agents, androgens, immunomodulatory drugs, interferon, cytoreductive drugs and hypomethylating agents in MF. Finally, we review novel therapeutic options for MF including the new JAK1/2 inhibitors, ruxolitinib based combination approaches as well as novel therapeutic agents. Expert commentary: Despite significant reduction of splenomegaly and improvement of symptom burden and a signal for survival improvement, ruxolitinib does not lead to major reductions in JAK2 V617F allele burden and bone marrow fibrosis. No ruxolitinib-based combination approach has so far demonstrated superiority over ruxolitinib monotherapy. The novel JAK2 inhibitors pacritinib and momelotinib, other JAK inhibitors, telomerase inhibitors, anti-fibrosis agents and hsp90 inhibitors are in different stages of development.
Collapse
Affiliation(s)
- Maximilian Stahl
- a Yale University School of Medicine , Department of Internal Medicine, Section of Hematology and the Yale Cancer Center , New Haven , CT , USA
| | - Amer M Zeidan
- a Yale University School of Medicine , Department of Internal Medicine, Section of Hematology and the Yale Cancer Center , New Haven , CT , USA
| |
Collapse
|
35
|
Stahl M, Zeidan AM. Management of lower-risk myelodysplastic syndromes without del5q: current approach and future trends. Expert Rev Hematol 2017; 10:345-364. [PMID: 28277851 DOI: 10.1080/17474086.2017.1297704] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Myelodysplastic syndromes (MDS) are characterized by progressive bone marrow failure manifesting as blood cytopenia and a variable risk of progression into acute myeloid leukemia. MDS is heterogeneous in biology and clinical behavior. MDS are generally divided into lower-risk (LR) and higher-risk (HR) MDS. Goals of care in HR-MDS focus on changing the natural history of the disease, whereas in LR-MDS symptom control and quality of life are the main goals. Areas covered: We review the epidemiology, tools of risk assessment, and the available therapeutic modalities for LR-MDS. We discuss the use of erythropoiesis stimulating agents (ESAs), immunosuppressive therapy (IST), lenalidomide and the hypomethylating agents (HMAs). We also discuss the predictors of response, combination treatment modalities, and management of iron overload. Lastly, we overview the most promising investigational agents for LR-MDS. Expert commentary: It remains unclear how to best incorporate a wealth of new genetic and epigenetic prognostic markers into risk assessment tools especially for LR-MDS patients. Only a subset of patients respond to current treatment modalities and most responders eventually lose their response. Once standard therapeutic options fail, management becomes more challenging. Combination-based approaches have been largely unsuccessful. Among the most promising investigational are the TPO agonists, TGF- β pathway inhibitors, telomerase inhibitors, and the splicing modifiers.
Collapse
Affiliation(s)
- Maximilian Stahl
- a Section of Hematology, Department of Internal Medicine, Section of Hematology, Yale University and the Yale Comprehensive Cancer Center , Yale University School of Medicine , New Haven , CT , USA
| | - Amer M Zeidan
- a Section of Hematology, Department of Internal Medicine, Section of Hematology, Yale University and the Yale Comprehensive Cancer Center , Yale University School of Medicine , New Haven , CT , USA
| |
Collapse
|
36
|
Stahl M, Zeidan AM. Hypomethylating agents in combination with histone deacetylase inhibitors in higher risk myelodysplastic syndromes: Is there a light at the end of the tunnel? Cancer 2017; 123:911-914. [PMID: 28094843 DOI: 10.1002/cncr.30532] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 11/23/2016] [Accepted: 11/28/2016] [Indexed: 01/29/2023]
Affiliation(s)
- Maximilian Stahl
- Section of Hematology, Department of Internal Medicine, Yale University and Yale Cancer Center, New Haven, Connecticut
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University and Yale Cancer Center, New Haven, Connecticut
| |
Collapse
|
37
|
Vey N, Prebet T, Thalamas C, Charbonnier A, Rey J, Kloos I, Liu E, Luan Y, Vezan R, Graef T, Recher C. Phase 1 dose-escalation study of oral abexinostat for the treatment of patients with relapsed/refractory higher-risk myelodysplastic syndromes, acute myeloid leukemia, or acute lymphoblastic leukemia. Leuk Lymphoma 2016; 58:1880-1886. [PMID: 27911138 DOI: 10.1080/10428194.2016.1263843] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Histone deacetylase (HDAC) inhibitor abexinostat is under investigation for the treatment of various cancers. Epigenetic changes including aberrant HDAC activity are associated with cancers, including myelodysplastic syndromes (MDS), acute myeloid leukemia (AML), and acute lymphoblastic leukemia (ALL). In this phase 1 dose-escalation study, 17 patients with relapsed/refractory higher-risk MDS, AML, or ALL received oral abexinostat (60, 80 [starting dose], 100, or 120 mg) twice daily (bid) on Days 1-14 of 21-day cycles. The most common treatment-related grade ≥3 adverse events were thrombocytopenia (29%) and neutropenia (24%), none of which led to discontinuation. Maximum-tolerated dose was not reached. Of 12 evaluable patients, best response was stable disease in 1 patient. This study was closed due to limited clinical benefit. Future development of oral abexinostat 100 mg bid in patients with MDS, AML, or ALL should focus on combination regimens. ISRCTN registry: 99680465.
Collapse
Affiliation(s)
- Norbert Vey
- a Department of Hematology , Institut Paoli Calmettes, SIRIC-Marseille , Marseille , France.,b Aix-Marseille University , Marseille , France
| | - Thomas Prebet
- c Internal Medicine Department, Section of Hematology , Yale Cancer Center at Yale University , New Haven , CT , USA
| | - Claire Thalamas
- d Department of Medical Pharmacology , CIC 1436, Université Toulouse Hospital, Inserm , Toulouse , France
| | - Aude Charbonnier
- a Department of Hematology , Institut Paoli Calmettes, SIRIC-Marseille , Marseille , France
| | - Jerome Rey
- a Department of Hematology , Institut Paoli Calmettes, SIRIC-Marseille , Marseille , France
| | - Ioana Kloos
- e Department of Oncology Research and Development , Institut de Recherches Internationales Servier , Suresnes , France
| | - Emily Liu
- f Biometrics, Pharmacyclics , LLC, an AbbVie Company , Sunnyvale , CA , USA
| | - Ying Luan
- f Biometrics, Pharmacyclics , LLC, an AbbVie Company , Sunnyvale , CA , USA
| | - Remus Vezan
- g Department of Clinical Science, Pharmacyclics , LLC, an AbbVie Company , Sunnyvale , CA , USA
| | - Thorsten Graef
- g Department of Clinical Science, Pharmacyclics , LLC, an AbbVie Company , Sunnyvale , CA , USA
| | - Christian Recher
- h Service d'Hématologie , Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole , Toulouse , France.,i Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294-CNRS , Toulouse , France
| |
Collapse
|
38
|
Lee CJ, Shiraz P, Muffly L. Pharmacologic maintenance strategies following allogeneic hematopoietic cell transplantation for acute myeloid leukemia. Leuk Lymphoma 2016; 58:516-527. [PMID: 27685315 DOI: 10.1080/10428194.2016.1205744] [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: 10/21/2022]
Abstract
The use of pharmacologic agents to maintain remission following allogeneic hematopoietic cell transplantation (HCT) is a topic of increasing interest and exploration for patients with high-risk acute myeloid leukemia (AML). This review details published and ongoing studies focused on post-transplant pharmacologic maintenance for AML. While early phase studies have demonstrated the safety and tolerability of various maintenance approaches following HCT, the results of several ongoing randomized prospective studies will be required to determine the clinical efficacy needed to expand this approach from experimental to standard of care.
Collapse
Affiliation(s)
- Catherine J Lee
- a Huntsman Cancer Institute, University of Utah, Blood and Marrow Transplantation Program , Salt Lake City , UT , USA
| | - Parveen Shiraz
- b Division of Blood and Marrow Transplantation , Stanford University , Stanford , CA , USA
| | - Lori Muffly
- b Division of Blood and Marrow Transplantation , Stanford University , Stanford , CA , USA
| |
Collapse
|
39
|
Zeidan AM, Stahl M, Komrokji R. Emerging biological therapies for the treatment of myelodysplastic syndromes. Expert Opin Emerg Drugs 2016; 21:283-300. [DOI: 10.1080/14728214.2016.1220534] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
40
|
Morabito F, Voso MT, Hohaus S, Gentile M, Vigna E, Recchia AG, Iovino L, Benedetti E, Lo-Coco F, Galimberti S. Panobinostat for the treatment of acute myelogenous leukemia. Expert Opin Investig Drugs 2016; 25:1117-31. [PMID: 27485472 DOI: 10.1080/13543784.2016.1216971] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Therapeutic strategies in patients with acute myeloid leukemia (AML) have not changed significantly over the last decades. Appropriate strategies are ultimately driven by the assessment of patients' fitness to define suitability for intensive induction chemotherapy, which produces high initial remission rates but, increased likelihood of relapse. Old/unfit AML patients still represent an urgent and unmet therapeutic need. Epigenetic deregulation represents a strategic characteristic of AML pathophysiology whereby aberrant gene transcription provides an advantage to leukemic cell survival. Efforts to re-establish impaired epigenetic regulation include hypomethylating agents and histone deacetylase inhibitors (HDACi). AREAS COVERED The review discusses the underlying mechanisms leading to disruption of lysine acetyltransferases (KAT or HAT)/deacetylase (KDAC or HDAC) balance and the rationale for using the HDACi panobinostat (LBH-589) in AML. EXPERT OPINION Although panobinostat has produced significant results in myeloma, its efficacy remains limited in AML. Panobinostat exerts pleiotropic activity and lack of specificity, which likely contributes to its inadequate safety in elderly AML patients. Phase I-II trials, utilizing panobinostat associated with well-known chemotherapeutic agents are ongoing and combinations with other druggable targets may likely be evaluated in future trials. The clinical use of this HDACi in AML the near future does not appearing promising.
Collapse
Affiliation(s)
- Fortunato Morabito
- a Hematology Unit, Department of Onco-Hematology , A.O. of Cosenza , Cosenza , Italy.,b Biotechnology Research Unit , ASP of Cosenza , Aprigliano (CS) , Italy
| | - Maria Teresa Voso
- c Department of Biomedicine and Prevention , Universita' Tor Vergata , Rome , Italy
| | - Stefan Hohaus
- d Department of Hematology , Universita' Cattolica S. Cuore , Rome , Italy
| | - Massimo Gentile
- a Hematology Unit, Department of Onco-Hematology , A.O. of Cosenza , Cosenza , Italy
| | - Ernesto Vigna
- a Hematology Unit, Department of Onco-Hematology , A.O. of Cosenza , Cosenza , Italy
| | | | - Lorenzo Iovino
- e Department of Clinical and Experimental Medicine, Hematology Division , University of Pisa , Pisa , Italy
| | - Edoardo Benedetti
- e Department of Clinical and Experimental Medicine, Hematology Division , University of Pisa , Pisa , Italy
| | - Francesco Lo-Coco
- c Department of Biomedicine and Prevention , Universita' Tor Vergata , Rome , Italy
| | - Sara Galimberti
- e Department of Clinical and Experimental Medicine, Hematology Division , University of Pisa , Pisa , Italy
| |
Collapse
|