201
|
Récher C. The beginning of a new therapeutic era in acute myeloid leukemia. EJHAEM 2021; 2:823-833. [PMID: 35845213 PMCID: PMC9175720 DOI: 10.1002/jha2.252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/17/2022]
Abstract
In the field of AML, the early 2000s were shaped by the advent of novel molecular biology technologies including high-throughput sequencing that improved prognostic classification, response evaluation through the quantification of minimal residual disease, and the launch of research on targeted therapies. Our knowledge of leukemogenesis, AML genetic diversity, gene-gene interactions, clonal evolution, and treatment response assessment has also greatly improved. New classifications based on chromosomal abnormalities and gene mutations are now integrated on a routine basis. These considerable efforts contributed to the discovery and development of promising drugs which specifically target gene mutations, apoptotic pathways and cell surface antigens as well as reformulate classical cytotoxic agents. In less than 2 years, nine novels drugs have been approved for the treatment of AML patients, and many others are being intensively investigated, in particular immune therapies. There are now numerous clinical research opportunities offered to clinicians, thanks to these new treatment options. We are only at the start of a new era which should see major disruptions in the way we understand, treat, and monitor patients with AML.
Collapse
Affiliation(s)
- Christian Récher
- Service d'HématologieCentre Hospitalier Universitaire de ToulouseInstitut Universitaire du Cancer de Toulouse OncopoleUniversité Toulouse III Paul SabatierCentre de Recherches en Cancérologie de ToulouseToulouseFrance
| |
Collapse
|
202
|
Fink A, Hung E, Singh I, Ben-Neriah Y. Immunity in acute myeloid leukemia: Where the immune response and targeted therapy meet. Eur J Immunol 2021; 52:34-43. [PMID: 34648664 DOI: 10.1002/eji.202048945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 07/29/2021] [Accepted: 10/05/2021] [Indexed: 12/19/2022]
Abstract
Acute myeloid leukemia (AML) is a highly aggressive disease with high relapse and mortality rates. Recent years have shown a surge in novel therapeutic development for AML, both in clinical and preclinical stages. These developments include targeted therapies based on AML-specific molecular signatures as well as more general immune modulation and vaccination studies. In this review, we will explore the evolving arena of AML therapy and suggest some intriguing connections between immune system modulation and targeted therapy. Improved understanding of the immune system involvement in various stages of the disease and the crosstalk between immune effectors, targeted therapy, and AML cells can provide a better framework for designing the next generation of AML therapies.
Collapse
Affiliation(s)
- Avner Fink
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Eric Hung
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Indranil Singh
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yinon Ben-Neriah
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
203
|
Wang Z, Cai J, Ren J, Chen Y, Wu Y, Cheng J, Jia K, Huang F, Cheng Z, Sheng T, Song S, Heng H, Zhu Y, Tang W, Li H, Lu T, Chen Y, Lu S. Discovery of a Potent FLT3 Inhibitor (LT-850-166) with the Capacity of Overcoming a Variety of FLT3 Mutations. J Med Chem 2021; 64:14664-14701. [PMID: 34550682 DOI: 10.1021/acs.jmedchem.1c01196] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Secondary mutations of FLT3 have become the main mechanism of FLT3 inhibitor resistance that presents a significant clinical challenge. Herein, a series of pyrazole-3-amine derivatives were synthesized and optimized to overcome the common secondary resistance mutations of FLT3. The structure-activity relationship and molecular dynamics simulation studies illustrated that the ribose region of FLT3 could be occupied to help address the obstacle of secondary mutations. Among those derivatives, compound 67 exhibited potent and selective inhibitory activities against FLT3-ITD-positive acute myeloid leukemia (AML) cells and possessed equivalent potency against transformed BaF3 cells with a variety of secondary mutations. Besides, cellular mechanism assays demonstrated that 67 strongly inhibited phosphorylation of FLT3 and its downstream signaling factors, as well as induced cell cycle arrest and apoptosis in MV4-11 cells. In the MV4-11 xenograft models, 67 exhibited potent antitumor potency without obvious toxicity. Taken together, these results demonstrated that 67 might be a drug candidate for the treatment of FLT3-ITD-positive AML.
Collapse
Affiliation(s)
- Zhijie Wang
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jiongheng Cai
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jiwei Ren
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Yingli Wu
- Chemical Biology Division of Shanghai Universities E-Institutes, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Jie Cheng
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Kun Jia
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Fei Huang
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Zitian Cheng
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Tiancheng Sheng
- School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Shiyu Song
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing 210038, P. R. China
| | - Hao Heng
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yifan Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Weifang Tang
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
204
|
Genomic Abnormalities as Biomarkers and Therapeutic Targets in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13205055. [PMID: 34680203 PMCID: PMC8533805 DOI: 10.3390/cancers13205055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary AML is a heterogenous malignancy with a variety of underlying genomic abnormalities. Some of the genetic aberrations in AML have led to the development of specific inhibitors which were approved by the Food and Drug Administration (FDA) and are currently used to treat eligible patients. In this review, we describe five gene mutations for which approved inhibitors have been developed, the response of AML patients to these inhibitors, and the known mechanism(s) of resistance. This review also highlights the significance of developing function-based screens for target discovery in the era of personalized medicine. Abstract Acute myeloid leukemia (AML) is a highly heterogeneous malignancy characterized by the clonal expansion of myeloid stem and progenitor cells in the bone marrow, peripheral blood, and other tissues. AML results from the acquisition of gene mutations or chromosomal abnormalities that induce proliferation or block differentiation of hematopoietic progenitors. A combination of cytogenetic profiling and gene mutation analyses are essential for the proper diagnosis, classification, prognosis, and treatment of AML. In the present review, we provide a summary of genomic abnormalities in AML that have emerged as both markers of disease and therapeutic targets. We discuss the abnormalities of RARA, FLT3, BCL2, IDH1, and IDH2, their significance as therapeutic targets in AML, and how various mechanisms cause resistance to the currently FDA-approved inhibitors. We also discuss the limitations of current genomic approaches for producing a comprehensive picture of the activated signaling pathways at diagnosis or at relapse in AML patients, and how innovative technologies combining genomic and functional methods will improve the discovery of novel therapeutic targets in AML. The ultimate goal is to optimize a personalized medicine approach for AML patients and possibly those with other types of cancers.
Collapse
|
205
|
Kunadt D, Stölzel F. Effective Immunosurveillance After Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemia. Cancer Manag Res 2021; 13:7411-7427. [PMID: 34594134 PMCID: PMC8478160 DOI: 10.2147/cmar.s261721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/15/2021] [Indexed: 12/25/2022] Open
Abstract
The number of patients receiving allogeneic hematopoietic stem cell transplantation (alloHCT) has increased constantly over the last years due to advances in transplant technology development, supportive care, transplant safety, and donor availability. Currently, acute myeloid leukemia (AML) is the most frequent indication for alloHCT. However, disease relapse remains the main cause of therapy failure. Therefore, concepts of maintaining and, if necessary, reinforcing a strong graft-versus-leukemia (GvL) effect is crucial for the prognosis and long-term survival of the patients. Over the last decades, it has become evident that effective immunosurveillance after alloHCT is an entangled complex of donor-specific characteristics, leukemia-associated geno- and phenotypes, and acquired resistance mechanisms. Furthermore, adoption of effector cells such as natural killer (NK) cells, alloreactive and regulatory T-cells with their accompanying receptor repertoire, and cell–cell interactions driven by messenger molecules within the stem cell and the bone marrow niche have important impact. In this review of pre- and posttransplant elements and mechanisms of immunosurveillance, we highlight the most important mechanisms after alloHCT.
Collapse
Affiliation(s)
- Desiree Kunadt
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University of Dresden, Dresden, Germany
| | - Friedrich Stölzel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University of Dresden, Dresden, Germany
| |
Collapse
|
206
|
Machado CB, de Pinho Pessoa FMC, da Silva EL, da Costa Pantoja L, Ribeiro RM, de Moraes Filho MO, de Moraes MEA, Montenegro RC, Burbano RMR, Khayat AS, Moreira-Nunes CA. Kinase Inhibition in Relapsed/Refractory Leukemia and Lymphoma Settings: Recent Prospects into Clinical Investigations. Pharmaceutics 2021; 13:1604. [PMID: 34683897 PMCID: PMC8540545 DOI: 10.3390/pharmaceutics13101604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/21/2021] [Accepted: 09/25/2021] [Indexed: 01/19/2023] Open
Abstract
Cancer is still a major barrier to life expectancy increase worldwide, and hematologic neoplasms represent a relevant percentage of cancer incidence rates. Tumor dependence of continuous proliferative signals mediated through protein kinases overexpression instigated increased strategies of kinase inhibition in the oncologic practice over the last couple decades, and in this review, we focused our discussion on relevant clinical trials of the past five years that investigated kinase inhibitor (KI) usage in patients afflicted with relapsed/refractory (R/R) hematologic malignancies as well as in the pharmacological characteristics of available KIs and the dissertation about traditional chemotherapy treatment approaches and its hindrances. A trend towards investigations on KI usage for the treatment of chronic lymphoid leukemia and acute myeloid leukemia in R/R settings was observed, and it likely reflects the existence of already established treatment protocols for chronic myeloid leukemia and acute lymphoid leukemia patient cohorts. Overall, regimens of KI treatment are clinically manageable, and results are especially effective when allied with tumor genetic profiles, giving rise to encouraging future prospects of an era where chemotherapy-free treatment regimens are a reality for many oncologic patients.
Collapse
Affiliation(s)
- Caio Bezerra Machado
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Medicine, Federal University of Ceará, Fortaleza 60430-275, Brazil; (C.B.M.); (F.M.C.d.P.P.); (E.L.d.S.); (M.O.d.M.F.); (M.E.A.d.M.); (R.C.M.)
| | - Flávia Melo Cunha de Pinho Pessoa
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Medicine, Federal University of Ceará, Fortaleza 60430-275, Brazil; (C.B.M.); (F.M.C.d.P.P.); (E.L.d.S.); (M.O.d.M.F.); (M.E.A.d.M.); (R.C.M.)
| | - Emerson Lucena da Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Medicine, Federal University of Ceará, Fortaleza 60430-275, Brazil; (C.B.M.); (F.M.C.d.P.P.); (E.L.d.S.); (M.O.d.M.F.); (M.E.A.d.M.); (R.C.M.)
| | - Laudreísa da Costa Pantoja
- Department of Pediatrics, Octávio Lobo Children’s Hospital, Belém 60430-275, Brazil;
- Oncology Research Center, Department of Biological Sciences, Federal University of Pará, Belém 66073-005, Brazil; (R.M.R.B.); (A.S.K.)
| | | | - Manoel Odorico de Moraes Filho
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Medicine, Federal University of Ceará, Fortaleza 60430-275, Brazil; (C.B.M.); (F.M.C.d.P.P.); (E.L.d.S.); (M.O.d.M.F.); (M.E.A.d.M.); (R.C.M.)
| | - Maria Elisabete Amaral de Moraes
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Medicine, Federal University of Ceará, Fortaleza 60430-275, Brazil; (C.B.M.); (F.M.C.d.P.P.); (E.L.d.S.); (M.O.d.M.F.); (M.E.A.d.M.); (R.C.M.)
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Medicine, Federal University of Ceará, Fortaleza 60430-275, Brazil; (C.B.M.); (F.M.C.d.P.P.); (E.L.d.S.); (M.O.d.M.F.); (M.E.A.d.M.); (R.C.M.)
| | - Rommel Mário Rodriguez Burbano
- Oncology Research Center, Department of Biological Sciences, Federal University of Pará, Belém 66073-005, Brazil; (R.M.R.B.); (A.S.K.)
| | - André Salim Khayat
- Oncology Research Center, Department of Biological Sciences, Federal University of Pará, Belém 66073-005, Brazil; (R.M.R.B.); (A.S.K.)
| | - Caroline Aquino Moreira-Nunes
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Medicine, Federal University of Ceará, Fortaleza 60430-275, Brazil; (C.B.M.); (F.M.C.d.P.P.); (E.L.d.S.); (M.O.d.M.F.); (M.E.A.d.M.); (R.C.M.)
- Oncology Research Center, Department of Biological Sciences, Federal University of Pará, Belém 66073-005, Brazil; (R.M.R.B.); (A.S.K.)
| |
Collapse
|
207
|
Hultmark S, Baudet A, Schmiderer L, Prabhala P, Palma-Tortosa S, Sandén C, Fioretos T, Sasidharan R, Larsson C, Lehmann S, Juliusson G, Ek F, Magnusson M. Combinatorial molecule screening identified a novel diterpene and the BET inhibitor CPI-203 as differentiation inducers of primary acute myeloid leukemia cells. Haematologica 2021; 106:2566-2577. [PMID: 32855276 PMCID: PMC8485661 DOI: 10.3324/haematol.2020.249177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Indexed: 12/24/2022] Open
Abstract
Combination treatment has proven effective for patients with acute promyelocytic leukemia, exemplifying the importance of therapy targeting multiple components of oncogenic regulation for a successful outcome. However, recent studies have shown that the mutational complexity of acute myeloid leukemia (AML) precludes the translation of molecular targeting into clinical success. Here, as a complement to genetic profiling, we used unbiased, combinatorial in vitro drug screening to identify pathways that drive AML and to develop personalized combinatorial treatments. First, we screened 513 natural compounds on primary AML cells and identified a novel diterpene (H4) that preferentially induced differentiation of FLT3 wild-type AML, while FLT3-ITD/mutations conferred resistance. The samples responding to H4, displayed increased expression of myeloid markers, a clear decrease in the nuclear-cytoplasmic ratio and the potential of re-activation of the monocytic transcriptional program reducing leukemia propagation in vivo. By combinatorial screening using H4 and molecules with defined targets, we demonstrated that H4 induces differentiation by the activation of the protein kinase C (PKC) signaling pathway, and in line with this, activates PKC phosphorylation and translocation of PKC to the cell membrane. Furthermore, the combinatorial screening identified a bromo- and extra-terminal domain (BET) inhibitor that could further improve H4-dependent leukemic differentiation in FLT3 wild-type monocytic AML. These findings illustrate the value of an unbiased, multiplex screening platform for developing combinatorial therapeutic approaches for AML.
Collapse
Affiliation(s)
- Simon Hultmark
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| | - Aurélie Baudet
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| | - Ludwig Schmiderer
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| | - Pavan Prabhala
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sara Palma-Tortosa
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Carl Sandén
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thoas Fioretos
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Christer Larsson
- Division of Translational Cancer Research, Lund University, Lund, Sweden
| | - Sören Lehmann
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Gunnar Juliusson
- Department of Hematology, Skane University Hospital, Lund, Sweden
| | - Fredrik Ek
- Chemical Biology and Therapeutics, Lund University, Lund, Sweden
| | - Mattias Magnusson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| |
Collapse
|
208
|
Stenmarck MS, Engen C, Strand R. Reframing cancer: challenging the discourse on cancer and cancer drugs-a Norwegian perspective : Reframing Cancer. BMC Med Ethics 2021; 22:126. [PMID: 34548091 PMCID: PMC8454291 DOI: 10.1186/s12910-021-00693-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/09/2021] [Indexed: 01/05/2023] Open
Abstract
Background As the range of therapeutic options in the field of oncology increases, so too does the strain on health care budgets. The imbalance between what is medically possible and financially feasible is frequently rendered as an issue of tragic choices, giving rise to public controversies around health care rationing. Main body We analyse the Norwegian media discourse on expensive cancer drugs and identify four underlying premises: (1) Cancer drugs are de facto expensive, and one does not and should not question why. (2) Cancer drugs have an indubitable efficacy. (3) Any lifetime gained for a cancer patient is an absolute good, and (4) cancer patients and doctors own the truth about cancer. Applying a principle-based approach, we argue that these premises should be challenged on moral grounds. Within the Norwegian public discourse, however, the premises largely remain unchallenged due to what we find to be unjustified claims of moral superiority. We therefore explore alternative framings of the issue of expensive cancer drugs and discuss their potential to escape the predicament of tragic choices. Conclusions In a media discourse that has seemingly stagnated, awareness of the framings within it is necessary in order to challenge the current tragic choices predicament the discourse finds itself in. In order to allow for a discourse not solely concerned with the issue of tragic choices, the premises that underlie it must be subjected to critical examination. As the field of oncology advances rapidly, we depend on a discussion of its opportunities and challenges that is meaningful, and that soberly addresses the future of cancer care—both its potential and its limits.
Collapse
Affiliation(s)
- Mille Sofie Stenmarck
- Centre for Cancer Biomarkers, Centre for the Study of the Sciences and the Humanities, University of Bergen, Bergen, Norway
| | - Caroline Engen
- Centre for Cancer Biomarkers, Centre for the Study of the Sciences and the Humanities, University of Bergen, Bergen, Norway
| | - Roger Strand
- Centre for Cancer Biomarkers, Centre for the Study of the Sciences and the Humanities, University of Bergen, Bergen, Norway.
| |
Collapse
|
209
|
Novatcheva ED, Anouty Y, Saunders I, Mangan JK, Goodman AM. FMS-Like Tyrosine Kinase 3 Inhibitors for the Treatment of Acute Myeloid Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:e161-e184. [PMID: 34649791 DOI: 10.1016/j.clml.2021.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022]
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia of adults, with a five-year survival that remains poor (approximately 25%). Knowledge and understanding of AML genomics have expanded tremendously over the past decade and are now included in AML prognostication and treatment decisions. FMS-like tyrosine kinase 3 (FLT3) is a Class III receptor tyrosine kinase (RTK) expressed primarily in the cell membranes of early hematopoietic progenitor cells, found in 28% of all patients with AML. FLT3 is the second most frequent mutation in adult AML following Nuclear-cytoplasmic shuttling phosphoprotein (NPM1), which is found in 50% of cases.1 FLT3 inhibitors are promising new molecular therapeutics increasingly becoming standard of care for both newly diagnosed and relapsed/refractory FLT3 positive AML. This review will focus on the clinical trials/evidence, similarities, differences, clinical toxicities, and drug interactions relevant to treating clinicians as pertains to 5 FLT3-inhibitors: midostaurin, sorafenib, gilteritinib, crenolanib, and quizartinib.
Collapse
Affiliation(s)
| | - Yasmine Anouty
- Department of Pharmacy, University of California San Diego Health, La Jolla, CA
| | - Ila Saunders
- Department of Pharmacy, University of California San Diego Health, La Jolla, CA; UC San Diego Skaggs School of Pharmacy & Pharmaceutical Sciences, La Jolla, CA
| | - James K Mangan
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, CA
| | - Aaron M Goodman
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, CA.
| |
Collapse
|
210
|
Döhner H, Wei AH, Löwenberg B. Towards precision medicine for AML. Nat Rev Clin Oncol 2021; 18:577-590. [PMID: 34006997 DOI: 10.1038/s41571-021-00509-w] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2021] [Indexed: 02/08/2023]
Abstract
With rapid advances in sequencing technologies, tremendous progress has been made in understanding the molecular pathogenesis of acute myeloid leukaemia (AML), thus revealing enormous genetic and clonal heterogeneity, and paving the way for precision medicine approaches. The successful development of precision medicine for patients with AML has been exemplified by the introduction of targeted FLT3, IDH1/IDH2 and BCL-2 inhibitors. When used as single agents, these inhibitors display moderate antileukaemic activity. However, augmented clinical activity has been demonstrated when they are administered in combination with drugs with broader mechanisms of action targeting epigenetic and/or other oncogenic signalling pathways or with conventional cytotoxic agents. The development of immunotherapies has been hampered by the expression of antigens that are expressed by both leukaemic and non-malignant haematopoietic progenitor cells; nonetheless, a diverse range of immunotherapies are now entering clinical development. This myriad of emerging agents also creates challenges, such as how to safely combine agents with different mechanisms of action, the need to circumvent primary and secondary resistance, and new challenges in future clinical trial design. In this Review, we discuss the current state of precision medicine for AML, including both the potential to improve patient outcomes and the related challenges.
Collapse
Affiliation(s)
- Hartmut Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany.
| | - Andrew H Wei
- Department of Clinical Hematology, The Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - Bob Löwenberg
- Department of Hematology, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| |
Collapse
|
211
|
Abstract
The outcomes associated with pediatric acute myeloid leukemia (AML) have improved over the last few decades, with the implementation of intensive chemotherapy, hematopoietic stem cell transplant, and improved supportive care. However, even with intensive therapy and the use of HSCT, both of which carry significant risks of short- and long-term side effects, approximately 30% of children are not able to be cured. The characterization of AML in pediatrics has evolved over time and it currently involves use of a variety of diagnostic tools, including flow cytometry and comprehensive genomic sequencing. Given the adverse effects of chemotherapy and the need for additional therapeutic options to improve outcomes in these patients, the genomic and molecular architecture is being utilized to inform selection of targeted therapies in pediatric AML. This review provides a summary of current, targeted therapy options in pediatric AML.
Collapse
|
212
|
Almatani MF, Ali A, Onyemaechi S, Zhao Y, Gutierrez L, Vaikari VP, Alachkar H. Strategies targeting FLT3 beyond the kinase inhibitors. Pharmacol Ther 2021; 225:107844. [PMID: 33811956 PMCID: PMC11490306 DOI: 10.1016/j.pharmthera.2021.107844] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/20/2022]
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by clonal expansion and differentiation arrest of the myeloid progenitor cells, which leads to the accumulation of immature cells called blasts in the bone marrow and peripheral blood. Mutations in the receptor tyrosine kinase FLT3 occur in 30% of normal karyotype patients with AML and are associated with a higher incidence of relapse and worse survival. Targeted therapies against FLT3 mutations using small-molecule FLT3 tyrosine kinase inhibitors (TKIs) have long been investigated, with some showing favorable clinical outcomes. However, major setbacks such as limited clinical efficacy and the high risk of acquired resistance remain unresolved. FLT3 signaling, mutations, and FLT3 inhibitors are topics that have been extensively reviewed in recent years. Strategies to target FLT3 beyond the small molecule kinase inhibitors are expanding, nevertheless they are not receiving enough attention. These modalities include antibody-based FLT3 targeted therapies, immune cells mediated targeting strategies, and approaches targeting downstream signaling pathways and FLT3 translation. Here, we review the most recent advances and the challenges associated with the development of therapeutic modalities targeting FLT3 beyond the kinase inhibitors.
Collapse
Affiliation(s)
- Mohammed F Almatani
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Atham Ali
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Sandra Onyemaechi
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Yang Zhao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Lucas Gutierrez
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Vijaya Pooja Vaikari
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Houda Alachkar
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States; USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, United States.
| |
Collapse
|
213
|
Heinicke T, Krahl R, Kahl C, Cross M, Scholl S, Wolf HH, Hähling D, Hegenbart U, Peter N, Schulze A, Florschütz A, Schmidt V, Reifenrath K, Zojer N, Junghanss C, Sayer HG, Maschmeyer G, Späth C, Hochhaus A, Fischer T, Al-Ali HK, Niederwieser D. Allogeneic hematopoietic stem cell transplantation improves long-term outcome for relapsed AML patients across all ages: results from two East German Study Group Hematology and Oncology (OSHO) trials. Ann Hematol 2021; 100:2387-2398. [PMID: 34232360 PMCID: PMC8357692 DOI: 10.1007/s00277-021-04565-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 06/04/2021] [Indexed: 12/14/2022]
Abstract
Relapse of acute leukemia is a frequent complication with uncertain outcome and poorly defined risk factors. From 1621 patients entered into two prospective clinical trials (AML02; n = 740 and AML04; n = 881), 74.2% reached complete remission (CR) 1 after induction(s) and 59 patients after additional induction ± hematopoietic cell transplantation (HCT). Of the non-refractory patients, 48.4% with a median age of 63 (range 17-85) years relapsed. Relapses occurred within 6 months after CR in 46.5%, between 7 and 18 months in 38.7%, and after 18 months in 14.8% of patients. Relapse treatment resulted in CR2 in 39% of patients depending upon age (54.5% of ≤ 60 and 28.6% of > 60 years), duration of CR1, and treatment of relapse. Overall survival (OS) was 10.9 (7.4-16.2) %, but OS after HCT ± intensive chemotherapy (ICT) was 39.3% (31.8-48.6) at 5 years and not different in younger and older patients. Donor lymphocyte infusion ± chemotherapy and ICT alone resulted only in OS of 15.4% and of 5%, respectively. Independent favorable factors for OS were long CR1 duration, and HCT, while non-monosomal disease was beneficial for OS in elderly patients. Leukemia-free survival [LFS; 24.9 (19.5-31.7) % at 10 years] was affected by similar risk factors. In a competing risk model, the relapse incidence at 5 years was 53.5 ± 3.5% and the non-relapse mortality rate 21.7 ± 2.9%. Lower relapse incidence was observed in patents with HCT, long CR1 duration, and female gender. Risk factors for non-relapse mortality were HCT in younger and type of AML in elderly patients. In conclusion, allogeneic HCT ± IC improved the results in relapsed AML in younger and elderly patients. Increasing CR2 rates and HCT frequency will be the challenge for the next years. Relapse of the disease remains the major problem.
Collapse
Affiliation(s)
- Thomas Heinicke
- Department of Hematology and Oncology, University of Magdeburg, Magdeburg, Germany
| | - Rainer Krahl
- University Hospital Leipzig, 04106, Leipzig, Germany
| | - Christoph Kahl
- Department of Hematology and Oncology, Hospital Magdeburg, Magdeburg, Germany
| | - Michael Cross
- University Hospital Leipzig, 04106, Leipzig, Germany
| | - Sebastian Scholl
- Klinik Für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Hans-Heinrich Wolf
- Department of Hematology and Oncology, University Hospital, Halle, Germany
| | - Detlev Hähling
- Department of Hematology and Oncology, Klinikum Schwerin, Schwerin, Germany
| | - Ute Hegenbart
- Departement of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Norma Peter
- Medizinische Klinik, Carl-Thieme-Klinikum GmbH, Cottbus, Germany
| | - Antje Schulze
- Department of Hematology and Oncology, Helios Klinikum Erfurt, Erfurt, Germany
| | | | - Volker Schmidt
- Department of Hematology and Oncology, Helios Klinikum Erfurt, Erfurt, Germany
| | | | - Niklas Zojer
- Department of Medicine I, Wilhelminen Cancer Research Institute, Wilhelminenhospital, , Vienna, Austria
| | - Christian Junghanss
- Hematology, Oncology, Palliative Medicine, University of Rostock, Rostock, Germany
| | - Herbert G Sayer
- Department of Hematology and Oncology, Helios Klinikum Erfurt, Erfurt, Germany
| | - Georg Maschmeyer
- Department of Hematology, Oncology and Palliative Care, Ernst Von Bergmann Hospital, Potsdam, Germany
| | - Christian Späth
- Hematology and Oncology, University of Greifswald, Greifswald, Germany
| | - Andreas Hochhaus
- Klinik Für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Thomas Fischer
- Department of Hematology and Oncology, University of Magdeburg, Magdeburg, Germany
| | | | - Dietger Niederwieser
- University Hospital Leipzig, 04106, Leipzig, Germany.
- Lithuanian University of Health Sciences, Kaunas, Lithuania.
- Aichi Medical University, Nagakute, Japan.
| |
Collapse
|
214
|
Larson RA, Mandrekar SJ, Huebner LJ, Sanford BL, Laumann K, Geyer S, Bloomfield CD, Thiede C, Prior TW, Döhner K, Marcucci G, Voso MT, Klisovic RB, Galinsky I, Wei AH, Sierra J, Sanz MA, Brandwein JM, de Witte T, Niederwieser D, Appelbaum FR, Medeiros BC, Tallman MS, Krauter J, Schlenk RF, Ganser A, Serve H, Ehninger G, Amadori S, Gathmann I, Döhner H, Stone RM. Midostaurin reduces relapse in FLT3-mutant acute myeloid leukemia: the Alliance CALGB 10603/RATIFY trial. Leukemia 2021; 35:2539-2551. [PMID: 33654204 PMCID: PMC8591906 DOI: 10.1038/s41375-021-01179-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/13/2021] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
The prospective randomized, placebo-controlled CALGB 10603/RATIFY trial (Alliance) demonstrated a statistically significant overall survival benefit from the addition of midostaurin to standard frontline chemotherapy in a genotypically-defined subgroup of 717 patients with FLT3-mutant acute myeloid leukemia (AML). The risk of death was reduced by 22% on the midostaurin-containing arm. In this post hoc analysis, we analyzed the cumulative incidence of relapse (CIR) on this study and also evaluated the impact of 12 4-week cycles of maintenance therapy. CIR analyses treated relapses and AML deaths as events, deaths from other causes as competing risks, and survivors in remission were censored. CIR was improved on the midostaurin arm (HR = 0.71 (95% CI, 0.54-0.93); p = 0.01), both overall and within European LeukemiaNet 2017 risk classification subsets when post-transplant events were considered in the analysis as events. However, when transplantation was considered as a competing risk, there was overall no significant difference between the risks of relapse on the two randomized arms. Patients still in remission after consolidation with high-dose cytarabine entered the maintenance phase, continuing with either midostaurin or placebo. Analyses were inconclusive in quantifying the impact of the maintenance phase on the overall outcome. In summary, midostaurin reduces the CIR.
Collapse
Affiliation(s)
- Richard A Larson
- Department of Medicine and Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA.
| | - Sumithra J Mandrekar
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Lucas J Huebner
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Ben L Sanford
- Alliance Statistics and Data Center, Duke University, Durham, NC, USA
| | - Kristina Laumann
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Susan Geyer
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Clara D Bloomfield
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Christian Thiede
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus der TU Dresden, Dresden, Germany
| | - Thomas W Prior
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Konstanze Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Guido Marcucci
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy
| | | | - Ilene Galinsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andrew H Wei
- Department of Clinical Haematology, The Alfred Hospital and Monash University, Melbourne, Australia
| | - Jorge Sierra
- Hematology Department, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau and Jose Carreras Leukemia Research Institute, Autonomous University of Barcelona, Barcelona, Spain
| | - Miguel A Sanz
- Department of Hematology, Hospital Universitario y Politécnico La Fe and Department of Medicine, University of Valencia, Valencia, Spain
| | | | - Theo de Witte
- Radboud University Medical Centre, Nijmegen, Netherlands
| | | | - Frederick R Appelbaum
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bruno C Medeiros
- Division of Hematology, Stanford Comprehensive Cancer Center, Stanford University, Stanford, CA, USA
| | - Martin S Tallman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jürgen Krauter
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Richard F Schlenk
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
- NCT Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Hubert Serve
- Department of Medicine II, Hematology/Oncology, Goethe University Hospital Frankfurt, Frankfurt/Main, Germany
| | - Gerhard Ehninger
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus der TU Dresden, Dresden, Germany
| | - Sergio Amadori
- Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy
| | | | - Hartmut Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
215
|
Cerchione C, Peleteiro Raíndo A, Mosquera Orgueira A, Mosquera Torre A, Bao Pérez L, Marconi G, Isidori A, Pérez Encinas MM, Martinelli G. Safety of FLT3 inhibitors in patients with acute myeloid leukemia. Expert Rev Hematol 2021; 14:851-865. [PMID: 34424108 DOI: 10.1080/17474086.2021.1969911] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction: Acute myeloblastic leukemia (AML) is the most frequent type of acute leukemia in adults with an incidence of 4.2 cases per 100,000 inhabitants and poor 5-year survival. Patients with mutations in the FMS-like tyrosine kinase 3 (FLT3) gene have poor survival and higher relapse rates compared with wild-type cases.Areas covered: Several FLT3 inhibitors have been proved in FLT3mut AML patients, with differences in their pharmacokinetics, kinase inhibitory and adverse events profiles. First-generation multi-kinase inhibitors (midostaurin, sorafenib, lestaurtinib) target multiple proteins, whereassecond-generation inhibitors (crenolanib, quizartinib, gilteritinib) are more specific and potent inhibitors of FLT3, so they are associated with less off-target toxic effects. All of these drugs have primary and acquired mechanisms of resistance, and therefore their combinations with other drugs (checkpoint inhibitors, hypomethylating agents, standard chemotherapy) and its application in different clinical settings are under study.Expert opinion: The recent clinical development of various FLT3 inhibitors for the treatment of FLT3mut AML is an effective therapeutic strategy. However, there are unique toxicities and drug-drug interactions that need to be resolved. It is necessary to understand the mechanisms of toxicity in order to recognize and manage them adequately.
Collapse
Affiliation(s)
- Claudio Cerchione
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Andrés Peleteiro Raíndo
- Health Research Institute of Santiago De Compostela (Idis), Santiago De Compostela, Spain.,Division of Hematology, Complejo Hospitalario Universitario De Santiago (Chus), Santiago De Compostela, Spain
| | - Adrián Mosquera Orgueira
- Health Research Institute of Santiago De Compostela (Idis), Santiago De Compostela, Spain.,Division of Hematology, Complejo Hospitalario Universitario De Santiago (Chus), Santiago De Compostela, Spain
| | - Alicia Mosquera Torre
- Health Research Institute of Santiago De Compostela (Idis), Santiago De Compostela, Spain.,Division of Hematology, Complejo Hospitalario Universitario De Santiago (Chus), Santiago De Compostela, Spain
| | - Laura Bao Pérez
- Health Research Institute of Santiago De Compostela (Idis), Santiago De Compostela, Spain.,Division of Hematology, Complejo Hospitalario Universitario De Santiago (Chus), Santiago De Compostela, Spain
| | - Giovanni Marconi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro Isidori
- Hematology and Stem Cell Transplant Center, Aormn Hospital, Pesaro, Italy
| | - Manuel Mateo Pérez Encinas
- Health Research Institute of Santiago De Compostela (Idis), Santiago De Compostela, Spain.,Division of Hematology, Complejo Hospitalario Universitario De Santiago (Chus), Santiago De Compostela, Spain.,University of Santiago De Compostela, Santiago De Compostela, Spain
| | - Giovanni Martinelli
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
216
|
Hosono N, Yokoyama H, Aotsuka N, Ando K, Iida H, Ishikawa T, Usuki K, Onozawa M, Kizaki M, Kubo K, Kuroda J, Kobayashi Y, Shimizu T, Chiba S, Nara M, Hata T, Hidaka M, Fujiwara SI, Maeda Y, Morita Y, Kusano M, Lu Q, Miyawaki S, Berrak E, Hasabou N, Naoe T. Gilteritinib versus chemotherapy in Japanese patients with FLT3-mutated relapsed/refractory acute myeloid leukemia. Int J Clin Oncol 2021; 26:2131-2141. [PMID: 34363558 PMCID: PMC8522999 DOI: 10.1007/s10147-021-02006-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023]
Abstract
Background Until recently, no effective targeted therapies for FLT3-mutated (FLT3mut+) relapsed/refractory (R/R) acute myeloid leukemia (AML) were available in Japan. The FLT3 inhibitor, gilteritinib, was approved in Japan for patients with FLT3mut+ R/R AML based on the phase 3 ADMIRAL trial, which demonstrated the superiority of gilteritinib over salvage chemotherapy (SC) with respect to overall survival (OS; median OS, 9.3 vs 5.6 months, respectively; hazard ratio, 0.64 [95% confidence interval 0.49, 0.83]; P < 0.001). Methods We evaluated the Japanese subgroup (n = 48) of the ADMIRAL trial, which included 33 patients randomized to 120-mg/day gilteritinib and 15 randomized to SC. Results Median OS was 14.3 months in the gilteritinib arm and 9.6 months in the SC arm. The complete remission/complete remission with partial hematologic recovery rate was higher in the gilteritinib arm (48.5%) than in the SC arm (13.3%). After adjustment for drug exposure, fewer adverse events (AEs) occurred in the gilteritinib arm than in the SC arm. Common grade ≥ 3 AEs related to gilteritinib were febrile neutropenia (36%), decreased platelet count (27%), and anemia (24%). Conclusion Findings in Japanese patients are consistent with those of the overall ADMIRAL study population.
Collapse
Affiliation(s)
- Naoko Hosono
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan.
| | - Hisayuki Yokoyama
- Sendai Medical Center, Sendai, Japan.,Tohoku University, Sendai, Japan
| | | | - Kiyoshi Ando
- Tokai University School of Medicine, Isehara, Japan
| | | | | | | | | | - Masahiro Kizaki
- Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Kohmei Kubo
- Aomori Prefectural Central Hospital, Aomori, Japan
| | - Junya Kuroda
- Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yukio Kobayashi
- International University of Health and Welfare (IUHW), Mita Hospital, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | - Qiaoyang Lu
- Astellas Pharma US, Inc., Northbrook, IL, USA
| | | | | | | | | |
Collapse
|
217
|
Swaminathan M, Kantarjian HM, Levis M, Guerra V, Borthakur G, Alvarado Y, DiNardo CD, Kadia T, Garcia-Manero G, Ohanian M, Daver N, Konopleva M, Pemmaraju N, Ferrajoli A, Andreeff M, Jain N, Estrov Z, Jabbour EJ, Wierda WG, Pierce S, Pinsoy MR, Xiao L, Ravandi F, Cortes JE. A phase I/II study of the combination of quizartinib with azacitidine or low-dose cytarabine for the treatment of patients with acute myeloid leukemia and myelodysplastic syndrome. Haematologica 2021; 106:2121-2130. [PMID: 33853292 PMCID: PMC8327731 DOI: 10.3324/haematol.2020.263392] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
The FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) mutation in acute myeloid leukemia (AML) is associated with poor prognosis. We hypothesized that quizartinib, a selective and potent FLT3 inhibitor, with azacitidine (AZA) or low-dose cytarabine (LDAC) might improve the outcomes in patients with FLT3-ITD-mutated AML. In this open-label phase I/II trial, patients of any age receiving first-salvage treatment for FLT3-ITD AML or age >60 years with untreated myelodysplastic syndrome or AML were treated with quizartinib plus AZA or LDAC. Seventy-three patients were treated (34 frontline, 39 first salvage). With regard to previously untreated patients, the composite response (CRc) rate was 87% (n=13/15: 8 complete responses [CR], 4 CR with incomplete hematologic recovery [CRi], 1 CR without platelet recovery [CRp]) among the patients treated with quizartinib/AZA and 74% (n=14/19: 1 CR, 8 CRi, 5 CRp) among those treated with quizartinib/LDAC. The median overall survival was 19.2 months for the cohort treated with quizartinib/AZA cohort and 8.5 months for the patients treated with quizartinib/LDAC; the corresponding relapse-free survival figures were 10.5 and 6.4 months, respectively. With regard to previously treated patients, the CRc rate was 64% (n=16/25 in the quizartinib/AZA cohort and 29% (n=4/14)) in the quizartinib/LDAC cohort. The median overall survival for patients treated with quizartinib/AZA and quizartinib/LDAC was 12.8 versus 4 months, respectively. QTc prolongation grade 3 occurred in only one patient in each cohort. Quizartinib-based combinations, particularly with AZA, appear effective in both frontline and first salvage therapy for patients with FLT3-ITD-mutated AML and are well tolerated. ClinicalTrials.gov identifier: NCT01892371.
Collapse
Affiliation(s)
| | | | - Mark Levis
- Department of Hematological Malignancies, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center
| | - Veronica Guerra
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Gautam Borthakur
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Yesid Alvarado
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | | | - Tapan Kadia
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | | | - Maro Ohanian
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Naval Daver
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Marina Konopleva
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Naveen Pemmaraju
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | | | - Michael Andreeff
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Nitin Jain
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Zeev Estrov
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Elias J Jabbour
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - William G Wierda
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Sherry Pierce
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | | | - Lianchun Xiao
- Department of Biostatistics, University of Texas MD Anderson Cancer Center
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center
| | - Jorge E Cortes
- Department of Leukemia, University of Texas MD Anderson Cancer Center.
| |
Collapse
|
218
|
Ma J, Ge Z. Recent advances of targeted therapy in relapsed/refractory acute myeloid leukemia. Bosn J Basic Med Sci 2021; 21:409-421. [PMID: 33577442 PMCID: PMC8292864 DOI: 10.17305/bjbms.2020.5485] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/26/2021] [Indexed: 12/22/2022] Open
Abstract
Despite advances in the understanding of disease pathobiology, treatment for relapsed or refractory acute myeloid leukemia (R/R AML) remains challenging. The prognosis of R/R AML remains extremely poor despite chemotherapy and bone marrow transplants. Discoveries on recurrent and novel genetic mutations, such as FLT3-ITD and IDH1/IDH2, critical signaling pathways, and unique molecular markers expressed on the surface of leukemic cells have been under investigation for the management of R/R AML. Other than monoclonal antibodies, diabodies, and triabodies are new targeted therapies developed in recent years and will be the new direction of immunotherapy. Targeted agents combined intensive regimens can be viable options for salvage therapy and as bridges to allogeneic transplant. Future directions will focus on novel, efficient and targeted combinations, low-toxicity maintenance, and individualized precision strategies. Here, we review the major recent advances of targeted therapies in the treatment of R/R AML.
Collapse
Affiliation(s)
- Jiale Ma
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China; Department of Hematology, Xuzhou Central Hospital, Xuzhou, China
| | - Zheng Ge
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China
| |
Collapse
|
219
|
Kidoguch K, Shibusawa M, Tanimoto T. A critical appraisal of Japan's new drug approval process: a case study of FLT3-ITD inhibitor quizartinib. Invest New Drugs 2021; 39:1457-1459. [PMID: 34268710 DOI: 10.1007/s10637-021-01151-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
In the last two decades, simultaneous global development of novel drugs become more common by conducting multiregional clinical trials. However, regulatory authorities of different regions often make different decisions on the approvals of the same new drugs. We would like to discuss the appropriateness of Japanese regulatory approach through a case study of quizartinib, a novel anti-leukemia drug developed in Japan. The pivotal clinical trial "QuANTUM-R" conducted in 19 countries showed a modest increase in median overall survival with quizartinib than the conventional chemotherapy. However, because several critical defects in this trial were pointed out by the United States Food and Drug Administration (US FDA) and the European Medicines Agency (EMA), quizartinib has not been approved in the US and Europe to date. On the contrary, the regulatory authority of Japan gave a notice of approval to quizartinib as a "standard of care", and the country becomes the sole country that granted market authorization. In our paper, we provide more detailed discussion about the methodology for scientific evaluation of the new drug.
Collapse
Affiliation(s)
- Keisuke Kidoguch
- Saga-Ken medical centre KOSEIKAN, Department of Hematology, Saga, Japan.
| | - Motoharu Shibusawa
- Department of Hematology, Shinmatsudo Central General Hospital, Chiba, Japan
| | | |
Collapse
|
220
|
Bernasconi P, Borsani O. Eradication of Measurable Residual Disease in AML: A Challenging Clinical Goal. Cancers (Basel) 2021; 13:3170. [PMID: 34202000 PMCID: PMC8268140 DOI: 10.3390/cancers13133170] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 12/18/2022] Open
Abstract
In non-promyelocytic (non-M3) AML measurable residual disease (MRD) detected by multi-parameter flow cytometry and molecular technologies, which are guided by Consensus-based guidelines and discover very low leukemic cell numbers far below the 5% threshold of morphological assessment, has emerged as the most relevant predictor of clinical outcome. Currently, it is well-established that MRD positivity after standard induction and consolidation chemotherapy, as well as during the period preceding an allogeneic hematopoietic stem cell transplant (allo-HSCT), portends to a significantly inferior relapse-free survival (RFS) and overall survival (OS). In addition, it has become absolutely clear that conversion from an MRD-positive to an MRD-negative state provides a favorable clinical outcome similar to that associated with early MRD negativity. Thus, the complete eradication of MRD, i.e., the clearance of the few leukemic stem cells-which, due to their chemo-radiotherapy resistance, might eventually be responsible of disease recurrence-has become an un-met clinical need in AML. Nowadays, this goal might potentially be achieved thanks to the development of novel innovative treatment strategies, including those targeting driver mutations, apoptosis, methylation patterns and leukemic proteins. The aim of this review is to analyze these strategies and to suggest any potential combination able to induce MRD negativity in the pre- and post-HSCT period.
Collapse
Affiliation(s)
- Paolo Bernasconi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
- Hematology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Oscar Borsani
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
221
|
Hellesøy M, Engen C, Grob T, Löwenberg B, Valk PJM, Gjertsen BT. Sex disparity in acute myeloid leukaemia with FLT3 internal tandem duplication mutations: implications for prognosis. Mol Oncol 2021; 15:2285-2299. [PMID: 34101344 PMCID: PMC8410575 DOI: 10.1002/1878-0261.13035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/22/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022] Open
Abstract
Incidence, molecular presentation and outcome of acute myeloid leukaemia (AML) are influenced by sex, but little attention has been directed at untangling sex‐related molecular and phenotypic differences between female and male patients. While increased incidence and poor risk are generally associated with a male phenotype, the poor prognostic FLT3 internal tandem duplication (FLT3‐ITD) mutation and co‐mutations with NPM1 and DNMT3A are overrepresented in female AML. Here, we have investigated the relationship between sex and FLT3‐ITD mutation status by comparing clinical data, mutational profiles, gene expression and ex vivo drug sensitivity in four cohorts: Beat AML, LAML‐TCGA and two independent HOVON/SAKK cohorts, comprising 1755 AML patients in total. We found prevalent sex‐associated molecular differences. Co‐occurrence of FLT3‐ITD, NPM1 and DNMT3A mutations was overrepresented in females, while males with FLT3‐ITDs were characterized by additional mutations in RNA splicing and epigenetic modifier genes. We observed diverging expression of multiple leukaemia‐associated genes as well as discrepant ex vivo drug responses, suggestive of discrete functional properties. Importantly, significant prognostication was observed only in female FLT3‐ITD‐mutated AML. Thus, we suggest optimization of FLT3‐ITD mutation status as a clinical tool in a sex‐adjusted manner and hypothesize that prognostication, prediction and development of therapeutic strategies in AML could be improved by including sex‐specific considerations.
Collapse
Affiliation(s)
- Monica Hellesøy
- Haematology Section, Department of Medicine, Haukeland University Hospital, Helse Bergen HF, Norway
| | - Caroline Engen
- Department of Clinical Science, Center for Cancer Biomarkers CCBIO, University of Bergen, Norway
| | - Tim Grob
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bob Löwenberg
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter J M Valk
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bjørn T Gjertsen
- Haematology Section, Department of Medicine, Haukeland University Hospital, Helse Bergen HF, Norway.,Department of Clinical Science, Center for Cancer Biomarkers CCBIO, University of Bergen, Norway
| |
Collapse
|
222
|
Boluda B, Martínez-Cuadrón D, Algarra L, Cano I, Sayas MJ, Acuña-Cruz E, Blanco A, Marco-Ayala J, DeLapuerta R, Díaz-González Á, Tormo M, Rodríguez-Veiga R, García R, Piñana JL, López-Pavía M, Barragán E, Amigo ML, Sargas C, López A, Solana-Altabella A, Gil C, Megías-Vericat JE, Sanz MA, Montesinos P. Evolving patterns of care and outcomes in relapsed/refractory FLT3 mutated acute myeloid leukemia adult patients. Leuk Lymphoma 2021; 62:2727-2736. [PMID: 34121593 DOI: 10.1080/10428194.2021.1938031] [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
We have analyzed treatment patterns and outcomes of relapsed/refractory(R/R) FLT3mut AML adult patients registered in our institutional data base between 1998 and 2018. Overall, 147 patients were evaluable: 34 from 1998 to 2009, 113 from 2010 to 2018. Salvage treatments were intensive chemotherapy (n = 25, 74%), and supportive care (n = 9, 26%) in the 1998-2009 period, and intensive chemotherapy (n = 63, 56%), hypomethylating agent (n = 7, 6%), low-dose cytarabine-based (n = 8, 7%), clinical trial (n = 16, 14%) and supportive care (n = 19, 17%) in the 2010-2018 period. Complete remission (CR) or with incomplete recovery (CRi) rate was 44%, 49% among patients treated intensively (vs 30% with non-intensive p = 0.005). Median overall survival since first R/R was 5.8 months, and 16.3 months in subjects receiving an allo-HSCT in CR/CRi after first salvage (vs 3.8 in the remaining patients p < 0.0001). Clinical outcomes of R/R FLT3mut AML remain unsatisfactory. Inclusion in clinical trials and expanding options could lead to improved outcomes.
Collapse
Affiliation(s)
- Blanca Boluda
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - David Martínez-Cuadrón
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Lorenzo Algarra
- Hematology Department, Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | - Isabel Cano
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - María J Sayas
- Hematology Department, Hospital Doctor Peset, Valencia, Spain
| | - Evelyn Acuña-Cruz
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Albert Blanco
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Javier Marco-Ayala
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Rosalía DeLapuerta
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Álvaro Díaz-González
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Mar Tormo
- Hematology Department, Hospital Clínico Universitario, Valencia, Spain
| | - Rebeca Rodríguez-Veiga
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Raimundo García
- Hematology Department, Hospital General Castellón, Castellón, Spain
| | - José L Piñana
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - María López-Pavía
- Hematology Department, Hospital General Universitario, Valencia, Spain
| | - Eva Barragán
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - María L Amigo
- Hematology Department, Hospital Morales Meseguer, Murcia, Spain
| | - Claudia Sargas
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Aurelio López
- Hematology Department, Hospital Arnau de Vilanova, Valencia, Spain
| | - Antonio Solana-Altabella
- Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Pharmacy Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Cristina Gil
- Hematology Department, Hospital General Universitario de Alicante, Alicante, Spain
| | - Juan Eduardo Megías-Vericat
- Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Pharmacy Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Miguel A Sanz
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Pau Montesinos
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
223
|
Smith CC, Viny AD, Massi E, Kandoth C, Socci ND, Rapaport F, Najm M, Medina-Martinez JS, Papaemmanuil E, Tarver TC, Hsu HH, Le MH, West B, Bollag G, Taylor BS, Levine RL, Shah NP. Recurrent Mutations in Cyclin D3 Confer Clinical Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia. Clin Cancer Res 2021; 27:4003-4011. [PMID: 34103301 DOI: 10.1158/1078-0432.ccr-20-3458] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/25/2021] [Accepted: 05/14/2021] [Indexed: 12/28/2022]
Abstract
PURPOSE Biomarkers of response and resistance to FLT3 tyrosine kinase inhibitors (TKI) are still emerging, and optimal clinical combinations remain unclear. The purpose of this study is to identify co-occurring mutations that influence clinical response to the novel FLT3 inhibitor pexidartinib (PLX3397). EXPERIMENTAL DESIGN We performed targeted sequencing of pretreatment blasts from 29 patients with FLT3 internal tandem duplication (ITD) mutations treated on the phase I/II trial of pexidartinib in relapsed/refractory FLT3-ITD+ acute myeloid leukemia (AML). We sequenced 37 samples from 29 patients with available material, including 8 responders and 21 non-responders treated at or above the recommended phase II dose of 3,000 mg. RESULTS Consistent with other studies, we identified mutations in NRAS, TP53, IDH2, and a variety of epigenetic and transcriptional regulators only in non-responders. Among the most frequently mutated genes in non-responders was Cyclin D3 (CCND3). A total of 3 individual mutations in CCND3 (Q276*, S264R, and T283A) were identified in 2 of 21 non-responders (one patient had both Q276* and S264R). No CCND3 mutations were found in pexidartinib responders. Expression of the Q276* and T283A mutations in FLT3-ITD MV4;11 cells conferred resistance to apoptosis, decreased cell-cycle arrest, and increased proliferation in the presence of pexidartinib and other FLT3 inhibitors. Inhibition of CDK4/6 activity in CCND3 mutant MV4;11 cells restored pexidartinib-induced cell-cycle arrest but not apoptosis. CONCLUSIONS Mutations in CCND3, a gene not commonly mutated in AML, are a novel cause of clinical primary resistance to FLT3 inhibitors in AML and may have sensitivity to CDK4/6 inhibition.
Collapse
Affiliation(s)
- Catherine C Smith
- Division of Hematology/Oncology, University of California, San Francisco, California. .,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Aaron D Viny
- Human Oncology & Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Evan Massi
- Division of Hematology/Oncology, University of California, San Francisco, California
| | - Cyriac Kandoth
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicholas D Socci
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Franck Rapaport
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthieu Najm
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Juan S Medina-Martinez
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elli Papaemmanuil
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Theodore C Tarver
- Division of Hematology/Oncology, University of California, San Francisco, California
| | | | - Mai H Le
- Plexxikon Inc, Berkeley, California
| | | | | | - Barry S Taylor
- Human Oncology & Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ross L Levine
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Human Oncology & Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neil P Shah
- Division of Hematology/Oncology, University of California, San Francisco, California. .,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| |
Collapse
|
224
|
Xu Q, He S, Yu L. Clinical Benefits and Safety of FMS-Like Tyrosine Kinase 3 Inhibitors in Various Treatment Stages of Acute Myeloid Leukemia: A Systematic Review, Meta-Analysis, and Network Meta-Analysis. Front Oncol 2021; 11:686013. [PMID: 34150652 PMCID: PMC8209493 DOI: 10.3389/fonc.2021.686013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/11/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Given the controversial roles of FMS-like tyrosine kinase 3 inhibitors (FLT3i) in various treatment stages of acute myeloid leukemia (AML), this study was designed to assess this problem and further explored which FLT3i worked more effectively. METHODS A systematic review, meta-analysis and network meta-analysis (NMA) were conducted by filtering PubMed, Embase, Cochrane library, and Chinese databases. We included studies comparing therapeutic effects between FLT3i and non-FLT3i group in AML, particularly FLT3(+) patients, or demonstrating the efficiency of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in FLT3(+) AML. Relative risk (RR) with 95% confidence intervals (CI) was used for estimating complete remission (CR), early death and toxicity. Hazard ratio (HR) was used to assess overall survival (OS), event-free survival (EFS), relapse-free survival (RFS) and cumulative incidence of relapse (CIR). RESULTS After addressing all criteria, 39 studies were eventually analyzed. Better CR was accomplished by FLT3i in untreated AML (RR 0.88, p = 0.04) and refractory and relapsed FLT3(+) AML (rrAML) (RR 0.61, p < 0.01) compared to non-FLT3i arm, followed by improved survival (untreated AML: OS, HR 0.76; EFS, HR 0.67; RFS, HR 0.72; all p < 0.01; FLT3(+) rrAML: OS, HR 0.60, p < 0.01; RFS, HR 0.40, p = 0.01). In addition, allo-HSCT improved survival in FLT3(+) AML (OS, HR 0.53; EFS, HR 0.50; RFS, HR 0.57; CIR, HR 0.26; all p < 0.01), which was further prolonged by FLT3i administrated after allo-HSCT (OS, HR 0.45; RFS, HR 0.34; CIR, HR 0.32; all p < 0.01). Additionally, FLT3i consistently improved OS (p < 0.05) regardless of FLT3-ITD ratio, when compared to non-FLT3i group. Besides, FLT3i showed significantly increased risk of thrombocytopenia, neutropenia, anemia, skin- and cardiac-related adverse effects, increased alanine aminotransferase, and increased risk of cough and dyspnea (p < 0.05). In NMA, gilteritinib showed the highest probability for improved prognosis. CONCLUSIONS FLT3i safely improved prognosis in induction/reinduction stage of FLT3(+) AML and further boosted survival benefits from allo-HSCT as maintenance therapy, suggesting better prognosis if FLT3i is combined before and after allo-HSCT. In NMA, gilteritinib potentially achieved the best prognosis, which should be identified in direct trials.
Collapse
Affiliation(s)
- Qingyu Xu
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, China
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Shujiao He
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, China
| | - Li Yu
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
225
|
Abstract
Patients with relapsed or refractory (r/r) acute myeloid leukemia (AML) have a poor prognosis and treatment remains challenging. For the majority of r/r patients, allogeneic hematopoietic stem cell transplantation (HSCT) is the only curative treatment approach. Salvage therapy is given in order to reduce the leukemia load prior to transplantation. Patients achieving complete remission prior to allogeneic HSCT have a more favorable outcome. Intensive salvage regimens commonly consist of an anthracycline and high-dose cytarabine backbone. Donor lymphocyte infusions have shown efficacy in patients relapsing after allogeneic HSCT. For patients who cannot be intensively treated (eg, elderly AML patients), outcome is generally very poor and combinations with novel agents are currently under investigation. Mutational analysis should be repeated at the time of relapse to identify aberrations that can be targeted with new agents. For r/r AML patients with mutated fms-related tyrosine kinase 3 (FLT3), gilteritinib has shown superior results to intensive salvage regimens. The US Food and Drug Administration (FDA) and European Medicines Agency (EMA) approved gilteritinib for FLT3 mutated r/r AML patients. Ivosidenib and enasidenib, inhibitors for mutated isocitrate dehydrogenase (IDH) 1 and 2, respectively, have received approval for IDH1/IDH2 mutated r/r AML by the FDA (not EMA). APR-246 restores the function of mutated TP53 and early study results are promising. Other agents targeting CD47, menin, neural-precursor-cell-expressed developmentally down-regulated 8, as well as bispecific antibodies or chimeric antigen receptor T cells are under investigation. Further trials are needed to understand how to best combine novel agents with each other or with chemotherapy.
Collapse
|
226
|
Fathi AT, Stein EM, DiNardo CD, Levis MJ, Montesinos P, Botton S. Differentiation syndrome with lower-intensity treatments for acute myeloid leukemia. Am J Hematol 2021; 96:735-746. [PMID: 33625753 DOI: 10.1002/ajh.26142] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/16/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022]
Abstract
Differentiation Syndrome (DS) has been identified in a subset of patients undergoing treatment with novel classes of differentiating therapies for acute myeloid leukemia (AML) such as IDH and FLT3 inhibitors. While DS is a well-known treatment-related complication in acute promyelocytic leukemia (APL), efforts are still ongoing to standardize diagnostic and treatment parameters for DS in AML. Though the rates of incidence vary, many of the signs and symptoms of DS are common between APL and AML. So, DS can lead to fatal complications in AML, but prompt management is usually effective and rarely necessitates interruption or discontinuation of AML therapy.
Collapse
Affiliation(s)
- Amir T. Fathi
- Massachusetts General Hospital Cancer Center Boston Massachusetts USA
- Harvard Medical School Boston Massachusetts USA
| | - Eytan M. Stein
- Memorial Sloan Kettering Cancer Center New York New York USA
- Weill Cornell Medical College New York New York USA
| | | | - Mark J. Levis
- Sidney Kimmel Comprehensive Cancer Center Johns Hopkins University Baltimore Maryland USA
| | | | | |
Collapse
|
227
|
Roydhouse JK, Mishra-Kalyani PS, Bhatnagar V, Gutman R, King-Kallimanis BL, Sridhara R, Kluetz PG. Does Knowledge of Treatment Assignment Affect Patient Report of Symptoms, Function, and Health Status? An Evaluation Using Multiple Myeloma Trials. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2021; 24:822-829. [PMID: 34119080 DOI: 10.1016/j.jval.2020.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 11/22/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVES Unblinded trials are common in oncology, but patient knowledge of treatment assignment may bias response to questionnaires. We sought to ascertain the extent of possible bias arising from patient knowledge of treatment assignment. METHODS This is a retrospective analysis of data from 2 randomized trials in multiple myeloma, 1 double-blind and 1 open label. We compared changes in patient reports of symptoms, function, and health status from prerandomization (screening) to baseline (pretreatment but postrandomization) across control and investigational arms in the 2 trials. Changes from prerandomization scores at ~2 and 6 months on treatment were evaluated only across control arms to avoid comparisons between 2 different experimental drugs. All scores were on 0- to 100-point scales. Inverse probability weighting, entropy balancing, and multiple imputation using propensity score splines were used to compare score changes across similar groups of patients. RESULTS Minimal changes from screening were seen at baseline in all arms. In the control arm, mean changes of <7 points were seen for all domains at 2 and 6 months. The effect of unblinding at 6 months in social function was a decline of less than 6 points (weighting: -3.09; 95% confidence interval -8.41 to 2.23; balancing: -4.55; 95% confidence interval -9.86 to 0.76; imputation: -5.34; 95% confidence interval -10.64 to -0.04). CONCLUSION In this analysis, we did not find evidence to suggest that there was a meaningful differential effect on how patients reported their symptoms, function or health status after knowing their treatment assignment.
Collapse
Affiliation(s)
- Jessica K Roydhouse
- ORISE Fellow, Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA; Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| | | | - Vishal Bhatnagar
- Oncology Center of Excellence, Food and Drug Administration, Silver Spring, MD, USA
| | - Roee Gutman
- Department of Biostatistics, Brown University School of Public Health, Providence, RI, USA
| | | | - Rajeshwari Sridhara
- Office of Biostatistics, Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | - Paul G Kluetz
- Oncology Center of Excellence, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
228
|
FLT3 Tyrosine Kinase Inhibitors for the Treatment of Fit and Unfit Patients with FLT3-Mutated AML: A Systematic Review. Int J Mol Sci 2021; 22:ijms22115873. [PMID: 34070902 PMCID: PMC8198781 DOI: 10.3390/ijms22115873] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 11/17/2022] Open
Abstract
FLT3-mutated acute myeloid leukemia accounts for around 30% of acute myeloid leukemia (AML). The mutation carried a poor prognosis until the rise of tyrosine kinase inhibitors (TKIs). New potent and specific inhibitors have successfully altered the course of the disease, increasing the complete response rate and the survival of patients with FLT3-mutated AML. The aim of this article is to review all the current knowledge on these game-changing drugs as well as the unsolved issues raised by their use for fit and unfit FLT3-mutated AML patients. To this end, we analyzed the results of phase I, II, III clinical trials evaluating FLT3-TKI both in the first-line, relapse monotherapy or in combination referenced in the PubMed, the American Society of Hematology, the European Hematology Association, and the Clinicaltrials.gov databases, as well as basic science reports on TKI resistance from the same databases. The review follows a chronological presentation of the different trials that allowed the development of first- and second-generation TKI and ends with a review of the current lines of evidence on leukemic blasts resistance mechanisms that allow them to escape TKI.
Collapse
|
229
|
Targeted Therapeutic Approach Based on Understanding of Aberrant Molecular Pathways Leading to Leukemic Proliferation in Patients with Acute Myeloid Leukemia. Int J Mol Sci 2021; 22:ijms22115789. [PMID: 34071627 PMCID: PMC8198876 DOI: 10.3390/ijms22115789] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/14/2021] [Accepted: 05/26/2021] [Indexed: 12/21/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous hematopoietic neoplasm with various genetic abnormalities in myeloid stem cells leading to differentiation arrest and accumulation of leukemic cells in bone marrow (BM). The multiple genetic alterations identified in leukemic cells at diagnosis are the mainstay of World Health Organization classification for AML and have important prognostic implications. Recently, understanding of heterogeneous and complicated molecular abnormalities of the disease could lead to the development of novel targeted therapeutic agents. In the past years, gemtuzumab ozogamicin, BCL-2 inhibitors (venetovlax), IDH 1/2 inhibitors (ivosidenib and enasidenib) FLT3 inhibitors (midostaurin, gilteritinib, and enasidenib), and hedgehog signaling pathway inhibitors (gladegib) have received US Food and Drug Administration (FDA) approval for the treatment of AML. Especially, AML patients with elderly age and/or significant comorbidities are not currently suitable for intensive chemotherapy. Thus, novel therapeutic planning including the abovementioned target therapies could lead to improve clinical outcomes in the patients. In the review, we will present various important and frequent molecular abnormalities of AML and introduce the targeted agents of AML that received FDA approval based on the previous studies.
Collapse
|
230
|
Zuanelli Brambilla C, Lobaugh SM, Ruiz JD, Dahi PB, Goldberg AD, Young JW, Gyurkocza B, Shaffer BC, Ponce DM, Tamari R, Sanchez Escamilla M, Castillo Flores N, Politikos I, Scordo M, Shah GL, Cho C, Lin RJ, Maloy MA, Devlin SM, Jakubowski AA, Berman E, Stein EM, Papadopoulos EB, Perales MA, Tallman MS, Giralt SA, Smith M. Relapse after Allogeneic Stem Cell Transplantation of Acute Myelogenous Leukemia and Myelodysplastic Syndrome and the Importance of Second Cellular Therapy. Transplant Cell Ther 2021; 27:771.e1-771.e10. [PMID: 34033977 DOI: 10.1016/j.jtct.2021.05.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/30/2021] [Accepted: 05/13/2021] [Indexed: 10/01/2022]
Abstract
Patients with acute myelogenous leukemia (AML) or myelodysplastic syndrome (MDS) who relapse after allogeneic hematopoietic cell transplantation (allo-HCT) generally have poor overall survival (OS). Interventions that result in improved OS after relapse are not well established. The efficacy of second cellular therapy and specific indications are matters of debate. This study was conducted to evaluate factors associated with postrelapse survival and the efficacy of a second course of cellular therapy. We retrospectively analyzed consecutive patients with AML and MDS who underwent a first allo-HCT between 2010 and 2017 at our center but subsequently relapsed. One hundred and four patients with AML and 44 patients with MDS were included (total n = 148). Bone marrow (BM) and peripheral blood stem cell grafts were either unmodified or T cell-depleted (TCD) by CD34+ selection ex vivo. Forty-five patients (30.4%) received a second cellular therapy after relapse, either a second allo-HCT (n = 28; 18.9%) or donor leukocyte infusion (DLI) (n = 17; 11.5%). The median age at transplantation was 60 years (range, 24 to 78 years). The median time to relapse (TTR) after transplantation was 6.5 months (range, 1 to 60.9 months), and the ensuing median OS was 6 months (95% confidence interval [CI], 4.8 to 8.9 months). In univariable analysis, longer TTR, relapse type (measurable residual disease versus morphologic), relapse occurring in the most recent years, and receipt of cellular therapy after relapse were associated with better outcomes, whereas adverse cytogenetics and/or abnormality of TP53, as well as NPM1 mutation in patients with AML, were associated with adverse outcomes. Relapse type, year of relapse, and a variable resulting from the combination of TTR and receipt of second cellular therapy remained significantly associated with postrelapse survival in multivariable analysis. In a separate multivariable model, adjusted only for TTR, relapse type, and receipt of second cellular therapy, an adverse effect of NPM1 mutation on survival was confirmed. We could not show an effect of post-transplantation maintenance on survival after relapse. In both univariable and multivariable analysis, we found a positive association for second cellular therapy with survival after relapse in patients who relapsed early (<6 months) after allo-HCT and a similar trend in patients who relapsed late (>12 months) after transplantation. Two-year OS after a second cellular therapy was 44.9% (95% CI, 28.5% to 61.4%), and it was significantly better in patients with <5% BM blasts before cell infusion. We could not show different effects on survival after second cellular therapy for DLI versus second allo-HCT in univariable analysis. Survival after relapse is improving over time, but this remains a challenging event, especially for patients who relapse early after transplantation. We found that a second cellular therapy could offer a benefit even in these cases. Nonetheless, more research is needed to clarify the most appropriate treatment choices after relapse. These are probably driven by underlying genetic and immunologic conditions, which should be the focus of future studies.
Collapse
Affiliation(s)
- Corrado Zuanelli Brambilla
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medical Biotechnologies, University of Siena, Siena, Italy; Hematology Unit, Department of Oncology, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Stephanie M Lobaugh
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Josel D Ruiz
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Parastoo B Dahi
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Aaron D Goldberg
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James W Young
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York; The Rockefeller University, New York, New York
| | - Boglarka Gyurkocza
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Brian C Shaffer
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Doris M Ponce
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Roni Tamari
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Miriam Sanchez Escamilla
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Hematological Malignancies and Stem Cell Transplantation, Research Institute Marqués de Valdecilla, Santander, Spain
| | - Nerea Castillo Flores
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ioannis Politikos
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Christina Cho
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Richard J Lin
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Molly A Maloy
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ann A Jakubowski
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ellin Berman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eytan M Stein
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Esperanza B Papadopoulos
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Martin S Tallman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sergio A Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Melody Smith
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York; Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
231
|
Gilteritinib: potent targeting of FLT3 mutations in AML. Blood Adv 2021; 4:1178-1191. [PMID: 32208491 DOI: 10.1182/bloodadvances.2019000174] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/26/2020] [Indexed: 01/13/2023] Open
Abstract
Since the discovery of FMS-like tyrosine kinase-3 (FLT3)-activating mutations as genetic drivers in acute myeloid leukemia (AML), investigators have tried to develop tyrosine kinase inhibitors that could effectively target FLT3 and alter the disease trajectory. Giltertinib (formerly known as ASP2215) is a novel compound that entered the field late, but moved through the developmental process with remarkable speed. In many ways, this drug's rapid development was facilitated by the large body of knowledge gained over the years from efforts to develop other FLT3 inhibitors. Single-agent gilteritinib, a potent and selective oral FLT3 inhibitor, improved the survival of patients with relapsed or refractory FLT3-mutated AML compared with standard chemotherapy. This continues to validate the approach of targeting FLT3 itself and establishes a new backbone for testing combination regimens. This review will frame the preclinical and clinical development of gilteritinib in the context of the lessons learned from its predecessors.
Collapse
|
232
|
Real-life experience with CPX-351 and impact on the outcome of high-risk AML patients: a multicentric French cohort. Blood Adv 2021; 5:176-184. [PMID: 33570629 DOI: 10.1182/bloodadvances.2020003159] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/12/2020] [Indexed: 01/22/2023] Open
Abstract
CPX-351 is a liposomal formulation of cytarabine and daunorubicin approved for the treatment of adults with newly diagnosed, therapy-related acute myeloid leukemia (t-AML) or AML with myelodysplasia-related changes (MRC-AML). We retrospectively analyzed the efficacy and safety of CPX-351 in a real-world setting in 103 patients from 12 French centers, including the evaluation of molecular abnormalities at baseline and minimal residual disease (MRD) in responding patients, compared with a historical data set from Bordeaux-Toulouse DATAML registry. A favorable safety profile was observed, with a low frequency of alopecia (11%) and gastrointestinal toxicity (50%). The overall response rate after induction was 59%, and MRD <10-3 was achieved in 57% of complete response (CR)/CR with incomplete hematological recovery (CRi) patients. Only the presence of mutated TP53 (P = .02) or PTPN11 (P = .004) predicted lower response in multivariate analysis. Interestingly, high-risk molecular prognosis subgroups defined by 2017 European LeukemiaNet risk stratification, including ASXL1 and RUNX1 mutations, were not associated with a significantly lower response rate using CPX-351. With a median follow-up of 8.6 months, median overall survival (OS) was 16.1 months. Thirty-six patients underwent allogeneic stem cell transplantation with a significantly longer median OS compared with nontransplanted patients (P < .001). In multivariate analyses, only spliceosome mutations were associated with better OS (P = .04). In comparison with intensive chemotherapy, there was no difference in OS for patients <60 years. These data confirm the efficacy and safety of CPX-351 in high-risk AML (t-AML and MRC-AML) in a real-life setting. CPX-351 is a treatment of choice for patients aged ≥60 years.
Collapse
|
233
|
Loke J, Buka R, Craddock C. Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia: Who, When, and How? Front Immunol 2021; 12:659595. [PMID: 34012445 PMCID: PMC8126705 DOI: 10.3389/fimmu.2021.659595] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022] Open
Abstract
Although the majority of patients with acute myeloid leukemia (AML) treated with intensive chemotherapy achieve a complete remission (CR), many are destined to relapse if treated with intensive chemotherapy alone. Allogeneic stem cell transplant (allo-SCT) represents a pivotally important treatment strategy in fit adults with AML because of its augmented anti-leukemic activity consequent upon dose intensification and the genesis of a potent graft-versus-leukemia effect. Increased donor availability coupled with the advent of reduced intensity conditioning (RIC) regimens has dramatically increased transplant access and consequently allo-SCT is now a key component of the treatment algorithm in both patients with AML in first CR (CR1) and advanced disease. Although transplant related mortality has fallen steadily over recent decades there has been no real progress in reducing the risk of disease relapse which remains the major cause of transplant failure and represents a major area of unmet need. A number of therapeutic approaches with the potential to reduce disease relapse, including advances in induction chemotherapy, the development of novel conditioning regimens and the emergence of the concept of post-transplant maintenance, are currently under development. Furthermore, the use of genetics and measurable residual disease technology in disease assessment has improved the identification of patients who are likely to benefit from an allo-SCT which now represents an increasingly personalized therapy. Future progress in optimizing transplant outcome will be dependent on the successful delivery by the international transplant community of randomized prospective clinical trials which permit examination of current and future transplant therapies with the same degree of rigor as is routinely adopted for non-transplant therapies.
Collapse
Affiliation(s)
- Justin Loke
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Richard Buka
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
234
|
Engen C, Hellesøy M, Grob T, Al Hinai A, Brendehaug A, Wergeland L, Bedringaas SL, Hovland R, Valk PJM, Gjertsen BT. FLT3-ITD mutations in acute myeloid leukaemia - molecular characteristics, distribution and numerical variation. Mol Oncol 2021; 15:2300-2317. [PMID: 33817952 PMCID: PMC8410560 DOI: 10.1002/1878-0261.12961] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/02/2021] [Accepted: 04/01/2021] [Indexed: 11/07/2022] Open
Abstract
Recurrent somatic internal tandem duplications (ITD) in the FMS-like tyrosine kinase 3 (FLT3) gene characterise approximately one third of patients with acute myeloid leukaemia (AML), and FLT3-ITD mutation status guides risk-adapted treatment strategies. The aim of this work was to characterise FLT3-ITD variant distribution in relation to molecular and clinical features, and overall survival in adult AML patients. We performed two parallel retrospective cohort studies investigating FLT3-ITD length and expression by cDNA fragment analysis, followed by Sanger sequencing in a subset of samples. In the two cohorts, a total of 139 and 172 mutant alleles were identified in 111 and 123 patients, respectively, with 22% and 28% of patients presenting with more than one mutated allele. Further, 15% and 32% of samples had a FLT3-ITD total variant allele frequency (VAF) < 0.3, while 24% and 16% had a total VAF ≥ 0.7. Most of the assessed clinical features did not significantly correlate to FLT3-ITD numerical variation nor VAF. Low VAF was, however, associated with lower white blood cell count, while increasing VAF correlated with inferior overall survival in one of the cohorts. In the other cohort, ITD length above 50 bp was identified to correlate with inferior overall survival. Our report corroborates the poor prognostic association with high FLT3-ITD disease burden, as well as extensive inter- and intrapatient heterogeneity in the molecular features of FLT3-ITD. We suggest that future use of FLT3-targeted therapy could be accompanied with thorough molecular diagnostics and follow-up to better predict optimal therapy responders.
Collapse
Affiliation(s)
- Caroline Engen
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIO, University of Bergen, Norway
| | - Monica Hellesøy
- Haematology Section, Department of Medicine, Haukeland University Hospital, Helse Bergen HF, Norway
| | - Tim Grob
- Department of Haematology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Adil Al Hinai
- Department of Haematology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Atle Brendehaug
- Department of Medical Genetics, Haukeland University Hospital, Helse Bergen HF, Norway
| | - Line Wergeland
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIO, University of Bergen, Norway
| | - Siv Lise Bedringaas
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIO, University of Bergen, Norway
| | - Randi Hovland
- Department of Medical Genetics, Haukeland University Hospital, Helse Bergen HF, Norway.,Department of Biosciences, University of Bergen, Norway
| | - Peter J M Valk
- Department of Haematology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Bjørn T Gjertsen
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIO, University of Bergen, Norway.,Haematology Section, Department of Medicine, Haukeland University Hospital, Helse Bergen HF, Norway
| |
Collapse
|
235
|
Westermann J, Bullinger L. Precision medicine in myeloid malignancies. Semin Cancer Biol 2021; 84:153-169. [PMID: 33895273 DOI: 10.1016/j.semcancer.2021.03.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022]
Abstract
Myeloid malignancies have always been at the forefront of an improved understanding of the molecular pathogenesis of cancer. In accordance, over the last years, basic research focusing on the aberrations underlying malignant transformation of myeloid cells has provided the basis for precision medicine approaches and subsequently has led to the development of powerful therapeutic strategies. In this review article, we will recapitulate what has happened since in the 1980s the use of all-trans retinoic acid (ATRA), as a first targeted cancer therapy, has changed one of the deadliest leukemia subtypes, acute promyelocytic leukemia (APL), into one that can be cured without classical chemotherapy today. Similarly, imatinib, the first molecularly designed cancer therapy, has revolutionized the management of chronic myeloid leukemia (CML). Thus, targeted treatment approaches have become the paradigm for myeloid malignancy, but many questions still remain unanswered, especially how identical mutations can be associated with different phenotypes. This might be linked to the impact of the cell of origin, gene-gene interactions, or the tumor microenvironment including the immune system. Continuous research in the field of myeloid neoplasia has started to unravel the molecular pathways that are not only crucial for initial treatment response, but also resistance of leukemia cells under therapy. Ongoing studies focusing on leukemia cell vulnerabilities do already point to novel (targetable) "Achilles heels" that can further improve myeloid cancer therapy.
Collapse
Affiliation(s)
- Jörg Westermann
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine Berlin, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine Berlin, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
236
|
Loke J, Vyas H, Craddock C. Optimizing Transplant Approaches and Post-Transplant Strategies for Patients With Acute Myeloid Leukemia. Front Oncol 2021; 11:666091. [PMID: 33937080 PMCID: PMC8083129 DOI: 10.3389/fonc.2021.666091] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Acute Myeloid Leukemia (AML) is the commonest indication for allogeneic stem cell transplantation (allo-SCT) worldwide. The increasingly important role of allo-SCT in the management of AML has been underpinned by two important advances. Firstly, improvements in disease risk stratification utilizing genetic and Measurable Residual Disease (MRD) technologies permit ever more accurate identification of allo-mandatory patients who are at high risk of relapse if treated by chemotherapy alone. Secondly, increased donor availability coupled with the advent of reduced intensity conditioning (RIC) regimens has substantially expanded transplant access for patients with high risk AML In patients allografted for AML disease relapse continues to represent the commonest cause of transplant failure and the development of novel strategies with the potential to reduce disease recurrence represents a major unmet need.
Collapse
Affiliation(s)
- Justin Loke
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom.,Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Hrushikesh Vyas
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom.,Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom.,Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
237
|
Piccini M, Pilerci S, Merlini M, Grieco P, Scappini B, Bencini S, Peruzzi B, Caporale R, Signori L, Pancani F, Vannucchi AM, Gianfaldoni G. Venetoclax-Based Regimens for Relapsed/Refractory Acute Myeloid Leukemia in a Real-Life Setting: A Retrospective Single-Center Experience. J Clin Med 2021; 10:jcm10081684. [PMID: 33919958 PMCID: PMC8070927 DOI: 10.3390/jcm10081684] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/31/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022] Open
Abstract
Relapsed/refractory (R/R) acute myeloid leukemia (AML) is a largely unmet medical need, owing to the lack of standardized, effective treatment approaches, resulting in an overall dismal outcome. The only curative option for R/R AML patients is allogeneic hematopoietic stem cell transplantation (HSCT) which is only applicable in a fraction of patients due to the scarce efficacy and high toxicity of salvage regimens. Recently, a number of targeted agents with relatively favorable toxicity profiles have been explored in clinical trials for R/R AML patients. The Bcl-2 inhibitor venetoclax, in combination with hypomethylating agents or low dose cytarabine, has produced impressive results for newly diagnosed AML, while its role in R/R disease is not well defined yet. We retrospectively analyzed the clinical outcomes of 47 R/R AML patients treated with venetoclax-based regimens between March 2018 and December 2020 at our institution. Overall, we report a composite complete response rate of 55% with an overall acceptable toxicity profile. Outcomes were particularly favorable for NPM1 mutated patients, unlike for FLT3-ITD positive patients irrespective of NPM1 status. For patients treated with intention to transplant, the procedure could be finally performed in 54%. These findings suggest a role for venetoclax-based regimens in R/R AML patients and support the design of prospective studies.
Collapse
Affiliation(s)
- Matteo Piccini
- SOD Ematologia, Università degli Studi di Firenze e Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (M.P.); (B.S.); (A.M.V.)
| | - Sofia Pilerci
- Scuola di Specializzazione in Ematologia, Università degli Studi di Firenze, 50139 Firenze, Italy; (S.P.); (M.M.)
| | - Marta Merlini
- Scuola di Specializzazione in Ematologia, Università degli Studi di Firenze, 50139 Firenze, Italy; (S.P.); (M.M.)
| | - Pietro Grieco
- SOD Ematologia, Ospedale San Donato, Azienda Usl Toscana Sud-Est, 20121 Milano, Italy;
| | - Barbara Scappini
- SOD Ematologia, Università degli Studi di Firenze e Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (M.P.); (B.S.); (A.M.V.)
| | - Sara Bencini
- SOD Centro Diagnostico di Citofluorimetria e Immunoterapia, Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (S.B.); (B.P.); (R.C.)
| | - Benedetta Peruzzi
- SOD Centro Diagnostico di Citofluorimetria e Immunoterapia, Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (S.B.); (B.P.); (R.C.)
| | - Roberto Caporale
- SOD Centro Diagnostico di Citofluorimetria e Immunoterapia, Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (S.B.); (B.P.); (R.C.)
| | - Leonardo Signori
- Centro di Ricerca e Innovazione per le Malattie Mieloproliferative (CRIMM), Università degli Studi e Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (L.S.); (F.P.)
| | - Fabiana Pancani
- Centro di Ricerca e Innovazione per le Malattie Mieloproliferative (CRIMM), Università degli Studi e Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (L.S.); (F.P.)
| | - Alessandro Maria Vannucchi
- SOD Ematologia, Università degli Studi di Firenze e Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (M.P.); (B.S.); (A.M.V.)
- Centro di Ricerca e Innovazione per le Malattie Mieloproliferative (CRIMM), Università degli Studi e Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (L.S.); (F.P.)
| | - Giacomo Gianfaldoni
- SOD Ematologia, Università degli Studi di Firenze e Azienda Ospedaliera Universitaria Careggi, 50139 Firenze, Italy; (M.P.); (B.S.); (A.M.V.)
- Correspondence:
| |
Collapse
|
238
|
Marconi G, Giannini MB, Bagnato G, Simonetti G, Cerchione C, Mosquera Orgueira A, Musuraca G, Martinelli G. The safety profile of FLT3 inhibitors in the treatment of newly diagnosed or relapsed/refractory acute myeloid leukemia. Expert Opin Drug Saf 2021; 20:791-799. [PMID: 33853481 DOI: 10.1080/14740338.2021.1913120] [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: 12/19/2022]
Abstract
INTRODUCTION FLT3 inhibitors are important drugs in the therapy of FLT3 positive acute myeloid leukemia (AML). Midostaurin was registered in combination with chemotherapy to treat newly diagnosed AML. Gilteritinib and quizartinib demonstrate effectiveness in a randomized trial in relapsed/refractory AML. Several promising FLT3 inhibitors are being evaluated in clinical research. AREAS COVERED This review will report the safety of FLT3 inhibitors that are registered for acute myeloid leukemia induction and rescue therapy. EXPERT OPINION In the near future, it is possible that all the FLT3 positive non M3-AML patients will receive a FLT3 inhibitor. Therapy adherence and strategies to mitigate adverse events must be pursued. The treatment with FLT3 inhibitors may be optimized in terms of toxicities with a rational evaluation of antifungal prophylaxis and concomitant therapy, cardiology monitoring, and keeping in mind rare adverse events. Future studies on unfit patients, special populations, and maintenance settings are warranted, together with post-market studies and real-life experiences. Whenever new FLT3 inhibitors will come to the clinic, we could face a scenario in which profound knowledge of effectiveness, toxicities, and off-target effects will be relevant to choose the best drug for each patient.
Collapse
Affiliation(s)
- Giovanni Marconi
- Istituto Romagnolo per Lo Studio Dei Tumori "Dino Amadori" - IRST, Meldola (FC), Italy
| | | | - Gianmarco Bagnato
- Istituto Romagnolo per Lo Studio Dei Tumori "Dino Amadori" - IRST, Meldola (FC), Italy
| | - Giorgia Simonetti
- Istituto Romagnolo per Lo Studio Dei Tumori "Dino Amadori" - IRST, Meldola (FC), Italy
| | - Claudio Cerchione
- Istituto Romagnolo per Lo Studio Dei Tumori "Dino Amadori" - IRST, Meldola (FC), Italy
| | - Adrián Mosquera Orgueira
- Health Research Institute of Santiago De Compostela (IDIS), Santiago De Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario De Santiago De Compostela (CHUS - SERGAS), Santiago De Compostela, Spain.,University of Santiago De Compostela, Santiago De Compostela, Spain
| | - Gerardo Musuraca
- Istituto Romagnolo per Lo Studio Dei Tumori "Dino Amadori" - IRST, Meldola (FC), Italy
| | - Giovanni Martinelli
- Istituto Romagnolo per Lo Studio Dei Tumori "Dino Amadori" - IRST, Meldola (FC), Italy
| |
Collapse
|
239
|
Miura K, Iriyama N, Hatta Y, Takei M. Personalized patient care with aggressive hematological malignancies in non-responders to first-line treatment. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1903314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Katsuhiro Miura
- Tumor Center, Nihon University Itabashi Hospital, 173-8610, Itabashi city, Japan
- Department of Hematology and Rheumatology, Nihon University School of Medicine, 173-8610, Itabashi city, Tokyo, Japan
| | - Noriyoshi Iriyama
- Department of Hematology and Rheumatology, Nihon University School of Medicine, 173-8610, Itabashi city, Tokyo, Japan
| | - Yoshihiro Hatta
- Department of Hematology and Rheumatology, Nihon University School of Medicine, 173-8610, Itabashi city, Tokyo, Japan
| | - Masami Takei
- Department of Hematology and Rheumatology, Nihon University School of Medicine, 173-8610, Itabashi city, Tokyo, Japan
| |
Collapse
|
240
|
Zhang H, Wang P, Li Z, He Y, Gan W, Jiang H. Anti-CLL1 Chimeric Antigen Receptor T-Cell Therapy in Children with Relapsed/Refractory Acute Myeloid Leukemia. Clin Cancer Res 2021; 27:3549-3555. [PMID: 33832948 DOI: 10.1158/1078-0432.ccr-20-4543] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/20/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE The survival rate of children with refractory/relapsed acute myeloid leukemia (R/R-AML) by salvage chemotherapy is minimal. Treatment with chimeric antigen receptor T cells (CAR T) has emerged as a novel therapy to improve malignancies treatment. C-type lectin-like molecule 1 (CLL1) is highly expressed on AML stem cells, blast cells, and monocytes, but not on normal hematopoietic stem cells, indicating the therapeutic potential of anti-CLL1 CAR T in AML treatment. This study aimed to test the safety and efficacy of CAR T-cell therapy in R/R-AML. PATIENTS AND METHODS Four pediatric patients with R/R-AML were enrolled in the ongoing phase I/II anti-CLL1 CAR T-cell therapy trial. The CAR design was based on an apoptosis-inducing gene, FKBP-caspase 9, to establish a safer CAR (4SCAR) application. Anti-CLL1 CAR was transduced into peripheral blood mononuclear cells of the patients via lentivector 4SCAR, followed by infusion into the recipients after lymphodepletion chemotherapy. Cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and other adverse events were documented. Treatment response was evaluated by morphology and flow cytometry-based minimal residual disease assays. RESULTS Three patients with R/R-AML achieved complete remission and minimal residual disease negativity, while the other patient remained alive for 5 months. All these patients experienced low-grade and manageable adverse events. CONCLUSIONS On the basis of our single-institution experience, autologous anti-CLL1 CAR T-cell therapy has the potential to be a safe and efficient alternative treatment for children with R/R-AML, and therefore requires further investigation.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, P.R. China.
| | - Pengfei Wang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, P.R. China
| | - Zhuoyan Li
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, P.R. China
| | - Yingyi He
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, P.R. China
| | - Wenting Gan
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, P.R. China.
| | - Hua Jiang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|
241
|
Synergistic targeting of FLT3 mutations in AML via combined menin-MLL and FLT3 inhibition. Blood 2021; 136:2442-2456. [PMID: 32589720 DOI: 10.1182/blood.2020005037] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022] Open
Abstract
The interaction of menin (MEN1) and MLL (MLL1, KMT2A) is a dependency and provides a potential opportunity for treatment of NPM1-mutant (NPM1mut) and MLL-rearranged (MLL-r) leukemias. Concomitant activating driver mutations in the gene encoding the tyrosine kinase FLT3 occur in both leukemias and are particularly common in the NPM1mut subtype. In this study, transcriptional profiling after pharmacological inhibition of the menin-MLL complex revealed specific changes in gene expression, with downregulation of the MEIS1 transcription factor and its transcriptional target gene FLT3 being the most pronounced. Combining menin-MLL inhibition with specific small-molecule kinase inhibitors of FLT3 phosphorylation resulted in a significantly superior reduction of phosphorylated FLT3 and transcriptional suppression of genes downstream of FLT3 signaling. The drug combination induced synergistic inhibition of proliferation, as well as enhanced apoptosis, compared with single-drug treatment in models of human and murine NPM1mut and MLL-r leukemias harboring an FLT3 mutation. Primary acute myeloid leukemia (AML) cells harvested from patients with NPM1mutFLT3mut AML showed significantly better responses to combined menin and FLT3 inhibition than to single-drug or vehicle control treatment, whereas AML cells with wild-type NPM1, MLL, and FLT3 were not affected by either of the 2 drugs. In vivo treatment of leukemic animals with MLL-r FLT3mut leukemia reduced leukemia burden significantly and prolonged survival compared with results in the single-drug and vehicle control groups. Our data suggest that combined menin-MLL and FLT3 inhibition represents a novel and promising therapeutic strategy for patients with NPM1mut or MLL-r leukemia and concurrent FLT3 mutation.
Collapse
|
242
|
Waksal JA, Tallman MS. Incorporation of Novel therapies for the treatment of acute myeloid leukemia: a perspective. Leuk Lymphoma 2021; 62:779-790. [PMID: 33541192 PMCID: PMC8631181 DOI: 10.1080/10428194.2020.1842403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 10/22/2022]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of diseases that poses an array of therapeutic challenges. For decades two chemotherapeutic agents, cytarabine and daunorubicin, remained the backbone of AML therapy protocols. However, since 2017 nine novel therapies have been approved for the management of AML. With the rapid expansion of therapeutic options, hematologists must adapt their practice to optimize the benefits of these novel therapy options and minimize treatment toxicity. Here, we discuss the novel therapies that have changed the standard of care in management of patients with AML. We summarize the pivotal clinical trials that lead to the approval of these agents, and ongoing trials evaluating additional potential indications. We discuss several promising therapy candidates and their corresponding clinical trials. We discuss therapeutic strategies to incorporate these therapies into practice and pose unanswered questions that have arisen along with the expansion of treatment options.
Collapse
|
243
|
Kang D, Ludwig E, Jaworowicz D, Huang H, Fiedler-Kelly J, Cortes J, Ganguly S, Khaled S, Krämer A, Levis M, Martinelli G, Perl A, Russell N, Abutarif M, Choi Y, Yin O. Concentration-QTc analysis of quizartinib in patients with relapsed/refractory acute myeloid leukemia. Cancer Chemother Pharmacol 2021; 87:513-523. [PMID: 33415416 PMCID: PMC7946665 DOI: 10.1007/s00280-020-04204-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/16/2020] [Indexed: 11/29/2022]
Abstract
PURPOSE This analysis evaluated the relationship between concentrations of quizartinib and its active metabolite AC886 and QT interval corrected using Fridericia's formula (QTcF) in patients with relapsed/refractory acute myeloid leukemia (AML) treated in the phase 3 QuANTUM-R study (NCT02039726). METHODS The analysis dataset included 226 patients with AML. Quizartinib dihydrochloride was administered as daily doses of 20, 30, and 60 mg. Nonlinear mixed-effects modeling was performed using observed quizartinib and AC886 concentrations and time-matched mean electrocardiogram measurements. RESULTS Observed QTcF increased with quizartinib and AC886 concentrations; the relationship was best described by a nonlinear maximum effect (Emax) model. The predicted mean increase in QTcF at the maximum concentration of quizartinib and AC886 associated with 60 mg/day was 21.1 ms (90% CI, 18.3-23.6 ms). Age, body weight, sex, race, baseline QTcF, QT-prolonging drug use, hypomagnesemia, and hypocalcemia were not significant predictors of QTcF. Hypokalemia (serum potassium < 3.5 mmol/L) was a statistically significant covariate affecting baseline QTcF, but no differences in ∆QTcF (change in QTcF from baseline) were predicted between patients with versus without hypokalemia at the same quizartinib concentration. The use of concomitant QT-prolonging drugs did not increase QTcF further. CONCLUSION QTcF increase was dependent on quizartinib and AC886 concentrations, but patient factors, including sex and age, did not affect the concentration-QTcF relationship. Because concomitant strong cytochrome P450 3A (CYP3A) inhibitor use significantly increases quizartinib concentration, these results support the clinical recommendation of quizartinib dose reduction in patients concurrently receiving a strong CYP3A inhibitor. CLINICAL TRIAL REGISTRATION NCT02039726 (registered January 20, 2014).
Collapse
Affiliation(s)
| | - Elizabeth Ludwig
- Cognigen Corporation, a Simulations Plus Company, Buffalo, NY, USA
| | - David Jaworowicz
- Cognigen Corporation, a Simulations Plus Company, Buffalo, NY, USA
| | - Hannah Huang
- Cognigen Corporation, a Simulations Plus Company, Buffalo, NY, USA
| | | | - Jorge Cortes
- Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | | | | | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Mark Levis
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Giovanni Martinelli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alexander Perl
- Division of Hematology and Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Nigel Russell
- Centre for Clinical Haematology, Nottingham University Hospital, Nottingham, UK
| | | | | | | |
Collapse
|
244
|
Dillon R, Potter N, Freeman S, Russell N. How we use molecular minimal residual disease (MRD) testing in acute myeloid leukaemia (AML). Br J Haematol 2021; 193:231-244. [PMID: 33058194 DOI: 10.1111/bjh.17185] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In recent years there have been major advances in the use of molecular diagnostic and monitoring techniques for patients with acute myeloid leukaemia (AML). Coupled with the simultaneous explosion of new therapeutic agents, this has sown the seeds for significant improvements to treatment algorithms. Here we show, using a selection of real-life examples, how molecular monitoring can be used to refine clinical decision-making and to personalise treatment in patients with AML with nucleophosmin (NPM1) mutations, core binding factor translocations and other fusion genes. For each case we review the established evidence base and provide practical recommendations where evidence is lacking or conflicting. Finally, we review important technical considerations that clinicians should be aware of in order to safely exploit these technologies as they undergo widespread implementation.
Collapse
Affiliation(s)
- Richard Dillon
- Cancer Genetics Laboratory, Department of Medical and Molecular Genetics, King's College, London, UK
- Department of Haematology, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| | - Nicola Potter
- Cancer Genetics Laboratory, Department of Medical and Molecular Genetics, King's College, London, UK
| | - Sylvie Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Nigel Russell
- Department of Haematology, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| |
Collapse
|
245
|
Cortes JE, Mehta P. Determination of fitness and therapeutic options in older patients with acute myeloid leukemia. Am J Hematol 2021; 96:493-507. [PMID: 33368536 PMCID: PMC7986910 DOI: 10.1002/ajh.26079] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/15/2020] [Accepted: 12/20/2020] [Indexed: 12/11/2022]
Abstract
Treatment of older patients with AML remains challenging. Although age, performance status, and comorbidities are commonly employed to determine fitness for intensive treatment, several studies have demonstrated improved outcomes with treatment in older and classically unfit patients, highlighting the importance of other disease-related and patient-related factors that have prognostic value for treatment outcome in AML. However, consistent and objective assessments for fitness are lacking. Multi-parameter geriatric assessment tools offer more comprehensive evaluation, but are limited by the required resources and lack of standardization and consensus regarding prognostic value. These assessments are particularly important considering the emerging new AML therapies that represent a spectrum of intensities. Patients should therefore be evaluated holistically for fitness to receive a specific treatment, with the aim of providing individualized care, and such definitions of fitness should also consistently be applied to clinical trials. This review will examine evolving criteria for the determination of fitness among AML patients and discuss treatment options for older and/or unfit patients with AML.
Collapse
Affiliation(s)
- Jorge E. Cortes
- Georgia Cancer Center Augusta University Augusta Georgia USA
| | - Priyanka Mehta
- Department of Haematology University Hospitals Bristol, NHS Foundation Trust Bristol UK
| |
Collapse
|
246
|
Perl AE. Which novel agents will have a clinically meaningful impact in AML at diagnosis? Best Pract Res Clin Haematol 2021; 34:101257. [PMID: 33762111 DOI: 10.1016/j.beha.2021.101257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
New drug approvals now afford AML physicians a wider choice of initial treatment options than ever before. Although chemotherapy for AML is by no means ready to be replaced entirely by novel agents, the role of traditional cytotoxics in AML therapy is rapidly changing. In particular, biologically targeted agents such as the BCL2 inhibitor venetoclax and inhibitors of FLT3 and IDH mutations stand out as drugs likely to take AML therapy in important new directions. Maximum response and survival benefits likely require combinations of novel agents and chemotherapy or multiple novel agents together. The recently-published phase 3 VIALE-A study demonstrates a very successful example of a new combination approach, which led to venetoclax plus azacitidine establishing itself as the new standard of care for patients unfit for intensive chemotherapy. One could reasonably expect other subsets of AML to benefit from this regimen or other applications of venetoclax combinations. Building on this experience, venetoclax-based regimens also have the potential to replace standard intensive cytarabine/anthracycline "7&3" induction approach for some if not many patients who are fit for induction. This review will describe novel agents with the greatest potential for impactful frontline applications that will change the AML treatment paradigm.
Collapse
Affiliation(s)
- Alexander E Perl
- Perelman School of Medicine at the University of Pennsylvania, Division of Hematology-Oncology, Abramson Cancer Center, Leukemia Program, Phiadelphia, PA, USA.
| |
Collapse
|
247
|
Abstract
Until recently, acute myeloid leukemia (AML) patients used to have limited treatment options, depending solely on cytarabine + anthracycline (7 + 3) intensive chemotherapy and hypomethylating agents. Allogeneic stem cell transplantation (Allo-SCT) played an important role to improve the survival of eligible AML patients in the past several decades. The exploration of the genomic and molecular landscape of AML, identification of mutations associated with the pathogenesis of AML, and the understanding of the mechanisms of resistance to treatment from excellent translational research helped to expand the treatment options of AML quickly in the past few years, resulting in noteworthy breakthroughs and FDA approvals of new therapeutic treatments in AML patients. Targeted therapies and combinations of different classes of therapeutic agents to overcome treatment resistance further expanded the treatment options and improved survival. Immunotherapy, including antibody-based treatment, inhibition of immune negative regulators, and possible CAR T cells might further expand the therapeutic armamentarium for AML. This review is intended to summarize the recent developments in the treatment of AML.
Collapse
Affiliation(s)
- Hongtao Liu
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago Medical Center, 5841 S. Maryland Ave, MC 2115, Chicago, IL, 60637-1470, USA.
| |
Collapse
|
248
|
Wang Z, Cai J, Cheng J, Yang W, Zhu Y, Li H, Lu T, Chen Y, Lu S. FLT3 Inhibitors in Acute Myeloid Leukemia: Challenges and Recent Developments in Overcoming Resistance. J Med Chem 2021; 64:2878-2900. [PMID: 33719439 DOI: 10.1021/acs.jmedchem.0c01851] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene are often present in newly diagnosed acute myeloid leukemia (AML) patients with an incidence rate of approximately 30%. Recently, many FLT3 inhibitors have been developed and exhibit positive preclinical and clinical effects against AML. However, patients develop resistance soon after undergoing FLT3 inhibitor treatment, resulting in short durable responses and poor clinical effects. This review will discuss the main mechanisms of resistance to clinical FLT3 inhibitors and summarize the emerging strategies that are utilized to overcome drug resistance. Basically, medicinal chemistry efforts to develop new small-molecule FLT3 inhibitors offer a direct solution to this problem. Other potential strategies include the combination of FLT3 inhibitors with other therapies and the development of multitarget inhibitors. It is hoped that this review will provide inspiring insights into the discovery of new AML therapies that can eventually overcome the resistance to current FLT3 inhibitors.
Collapse
Affiliation(s)
- Zhijie Wang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jiongheng Cai
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jie Cheng
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Wenqianzi Yang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Yifan Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P.R. China
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| |
Collapse
|
249
|
Burchert A. Maintenance therapy for FLT3-ITD-mutated acute myeloid leukemia. Haematologica 2021; 106:664-670. [PMID: 33472354 PMCID: PMC7927878 DOI: 10.3324/haematol.2019.240747] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
FLT3-ITD is a constitutively activated variant of the FLT3 tyrosine kinase receptor. Its expression in acute myeloid leukemia (AML) is associated with a poor prognosis. Due to this, the development of tyrosine kinase inhibitors (TKI) blocking FLT3-ITD became a rational therapeutic concept. This review describes key milestones in the clinical development of different FLT3-specific TKI with a particular focus on FLT3-TKI maintenance therapy in remission after allogeneic hematopoietic stem cell transplantation (HCT). Recent evidence from randomized trials using sorafenib in FLT3-ITD mutated AML provided a proof of concept that targeted post-HCT maintenance therapy could become a new treatment paradigm in AML.
Collapse
Affiliation(s)
- Andreas Burchert
- Department of Internal Medicine, Hematology, Oncology and Immunology, Philipps University Marburg and University Hospital Giessen and Marburg, Campus Marburg, Marburg.
| |
Collapse
|
250
|
Abstract
Aberrant FLT3 receptor signaling is common in acute myeloid leukemia (AML) and has important implications for the biology and clinical management of the disease. Patients with FLT3-mutated AML frequently present with critical illness, are more likely to relapse after treatment, and have worse clinical outcomes than their FLT3 wild type counterparts. The clinical management of FLT3-mutated AML has been transformed by the development of FLT3 inhibitors, which are now in use in the frontline and relapsed/refractory settings. However, many questions regarding the optimal approach to the treatment of these patients remain. In this paper, we will review the rationale for targeting the FLT3 receptor in AML, the impact of FLT3 mutation on patient prognosis, the current standard of care approaches to FLT3-mutated AML management, and the diverse array of FLT3 inhibitors in use and under investigation. We will also explore new opportunities and strategies for targeting the FLT3 receptor. These include targeting the receptor in patients with non-canonical FLT3 mutations or wild type FLT3, pairing FLT3 inhibitors with other novel therapies, using minimal residual disease (MRD) testing to guide the targeting of FLT3, and novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alexander J Ambinder
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark Levis
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|