1
|
Lin WT, Chao CM, Lin CY, Hsu YT, Hsiao SY, Weng TS. Efficacy and safety of second‑generation FLT3 inhibitors in acute myeloid leukemia: A systematic review and meta‑analysis of randomized controlled trials. Mol Clin Oncol 2024; 21:93. [PMID: 39478693 PMCID: PMC11523226 DOI: 10.3892/mco.2024.2791] [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: 06/07/2024] [Accepted: 08/15/2024] [Indexed: 11/02/2024] Open
Abstract
Acute myeloid leukemia (AML) is one of the most frequent forms of acute leukemia and the second most common leukemia subtype in adults. In 2020, the incidence of AML in the United States was estimated to be ~4 cases per 100,000 adults. The FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) and tyrosine kinase domain (TKD) mutation are major prognostic indicators of AML. They are more frequently observed in younger AML patients (aged <60 years), likely due to their association with de novo. Additionally, these mutations have a stronger negative impact on survival in younger patients. Therefore, quizartinib and gilteritinib are second-generation FLT3 inhibitors that are frequently applied for treating patients with AML. However, to the best of our knowledge, few studies have compared the efficacy of second-generation FLT3 inhibitors for AML treatment. Therefore, the present study conducted a comprehensive search for studies on the efficacy and safety of FLT3 inhibitors across PubMed, Embase, the Cochrane Library and ClinicalTrials.gov. The search criteria were limited to randomized controlled trials (RCTs). Subsequently, a meta-analysis was performed on a total of five randomized controlled trials, involving 1,543 participants in total, using a random-effects model. In each RCT, compared to the salvage chemotherapy used in the control group, the groups that received second-generation FLT3 inhibitors experienced significant improvements in overall survival (hazard ratio, 0.717; 95% CI, 0.604-0.850; P<0.001). In addition, overall survival was found to be consistent across the different types of second-generation FLT3 inhibitors used and different types of AML. The risks associated with a prolonged heart-rate corrected QT interval (QTc) interval were next evaluated. Compared with the salvage chemotherapy used in the control group, the second-generation FLT3 inhibitor group exhibited a significantly higher risk of having a prolonged QTc interval (odds ratio, 6.311; 95% CI, 3.061-13.013; P<0.001). In conclusion, these findings suggest that second-generation FLT3 inhibitors can improve the overall survival of patients with AML. However, QTc prolongation is a potential adverse effect that should be monitored.
Collapse
Affiliation(s)
- Wei-Ting Lin
- Department of Orthopedics, Chi Mei Medical Center, Tainan 710033, Taiwan, R.O.C
| | - Chien-Ming Chao
- Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan 736402, Taiwan, R.O.C
| | - Cheng-Yao Lin
- Department of Senior Welfare and Services, Southern Taiwan University of Science and Technology, Tainan 710301, Taiwan, R.O.C
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
- Division of Hematology-Oncology, Department of Internal Medicine, Chi Mei Medical Center, Liouying, Tainan 736402, Taiwan, R.O.C
| | - Ya-Ting Hsu
- Division of Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan, R.O.C
| | - Sheng-Yen Hsiao
- Division of Hematology-Oncology, Department of Internal Medicine, Chi Mei Medical Center, Liouying, Tainan 736402, Taiwan, R.O.C
- Department of Nursing, Chung Hwa University of Medical Technology, Tainan 717302, Taiwan, R.O.C
| | - Teng-Song Weng
- Department of Pharmacy, Chi Mei Medical Center, Liouying, Tainan 736402, Taiwan, R.O.C
| |
Collapse
|
2
|
Cortes J. Quizartinib: a potent and selective FLT3 inhibitor for the treatment of patients with FLT3-ITD-positive AML. J Hematol Oncol 2024; 17:111. [PMID: 39538314 PMCID: PMC11558990 DOI: 10.1186/s13045-024-01617-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Mutations in FMS-related receptor tyrosine kinase 3 (FLT3) are among the most common alterations in acute myeloid leukemia (AML), present in ≈30% of newly diagnosed AML cases. Internal tandem duplications (ITD) in FLT3 (FLT3-ITD) occur in ≈25% of newly diagnosed AML cases and are associated with unfavorable outcomes. Quizartinib (formerly AC220) is a novel, second-generation, highly potent, and selective type II FLT3 inhibitor. Quizartinib is approved in Japan as monotherapy for the treatment of adult patients with FLT3-ITD-positive relapsed/refractory (R/R) AML. Quizartinib is also approved in the United States, Japan, Europe, and United Kingdom in combination with chemotherapy during induction and consolidation, and as maintenance monotherapy (but, in the United States, not after allogeneic hematopoietic cell transplantation [allo-HCT]), for the treatment of adult patients with newly diagnosed FLT3-ITD-positive AML. In this review, we summarize preclinical studies that established quizartinib as a potent and selective type II FLT3 inhibitor as well as early and pivotal phase 3 clinical studies (QuANTUM-R and QuANTUM-First) that led to the approvals of quizartinib. We also summarize mechanisms of resistance to quizartinib along with its safety profile. Furthermore, we review the ongoing post hoc analyses of the QuANTUM-First data elucidating the impact of allo-HCT, the presence of measurable residual disease, and number and length of ITD on the clinical outcomes of quizartinib. We also describe the impact of quizartinib on patient-reported outcomes. Finally, we highlight some of the ongoing studies that test quizartinib in patients with FLT3-ITD-positive AML, patients with FLT3-ITD-negative AML, in both the first-line and R/R settings, in patients fit or unfit for intensive chemotherapy, including studies for quizartinib-based combination with other compounds such as decitabine and venetoclax. Future research should aim to further optimize the clinical value of quizartinib and explore its use in additional clinical settings, which could be achieved by testing quizartinib with other drugs, better characterization of the mechanisms of resistance, identification of the role of quizartinib as a maintenance therapy after allo-HCT, and investigating quizartinib in patients with FLT3-ITD-negative AML.
Collapse
Affiliation(s)
- Jorge Cortes
- Georgia Cancer Center at Augusta University, 1410 Laney Walker Blvd., CN2222, Augusta, GA, 30912, USA.
| |
Collapse
|
3
|
Schlaweck S, Radcke A, Kampmann S, Becker BV, Brossart P, Heine A. The Immunomodulatory Effect of Different FLT3 Inhibitors on Dendritic Cells. Cancers (Basel) 2024; 16:3719. [PMID: 39518156 PMCID: PMC11545830 DOI: 10.3390/cancers16213719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND FMS-like tyrosine kinase 3 (FLT3) mutations or internal tandem duplication occur in 30% of acute myeloid leukemia (AML) cases. In these cases, FLT3 inhibitors (FLT3i) are approved for induction treatment and relapse. Allogeneic hematopoietic stem cell transplantation (alloHSCT) remains the recommended post-induction therapy for suitable patients. However, the role of FLT3i therapy after alloHSCT remains unclear. Therefore, we investigated the three currently available FLT3i, gilteritinib, midostaurin, and quizartinib, in terms of their immunosuppressive effect on dendritic cells (DCs). DCs are professional antigen-presenting cells inducing T-cell responses to infectious stimuli. Highly activated DCs can also cause complications after alloHSCT, such as triggering Graft versus Host disease, a serious and potentially life-threatening complication after alloHSCT. METHODS To study the immunomodulatory effects on DCs, we differentiated murine and human DCs in the presence of FLT3i and performed immunophenotyping by flow cytometry and cytokine measurements and investigated gene and protein expression. RESULTS We detected a dose-dependent immunosuppressive effect of midostaurin, which decreased the expression of costimulatory markers like CD86, and found a reduced secretion of pro-inflammatory cytokines such as IL-12, TNFα, and IL-6. Mechanistically, we show that midostaurin inhibits TLR and TNF signaling and NFκB, PI3K, and MAPK pathways. The immunosuppressive effect of gilteritinib was less pronounced, while quizartinib did not show truncation of relevant signaling pathways. CONCLUSIONS Our results suggest different immunosuppressive effects of these three FLT3i and may, therefore, provide an additional rationale for optimal maintenance therapy after alloHSCT of FLT3-positive AML patients to prevent infectious complications and GvHD mediated by DCs.
Collapse
Affiliation(s)
- Sebastian Schlaweck
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
- Faculty of Medicine, Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), University Hospital of Bonn, 53127 Bonn, Germany
| | - Alea Radcke
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
| | - Sascha Kampmann
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
| | - Benjamin V. Becker
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany;
| | - Peter Brossart
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
| | - Annkristin Heine
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
| |
Collapse
|
4
|
Moallem FE, Gholami Chahkand MS, Dadkhah PA, Azarm E, Shahrokhi M, Deyhimi MS, Karimi MA. Quizartinib: a new hope in acute myeloid leukemia, an applied comprehensive review. Future Oncol 2024; 20:2791-2810. [PMID: 39297694 PMCID: PMC11572082 DOI: 10.1080/14796694.2024.2399425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 08/29/2024] [Indexed: 11/16/2024] Open
Abstract
Acute myeloid leukemia (AML) is caused by a defective precursor leading to malignant clonal expansion, often with FMS-like tyrosine kinase-3 receptor (FLT3) mutations, particularly internal tandem duplication (ITD), which has a poor prognosis. Quizartinib, a second-generation FLT3 inhibitor, has FDA approval for relapsed/refractory AML with FLT3/ITD mutation. It has shown promise in clinical studies since 2013 due to its excellent oral absorption and potent activity on FLT3. This review explores Quizartinib's mechanism of action, efficacy in monotherapy or combination with chemotherapy, drug interactions, adverse events, resistance mechanisms and future research directions.
Collapse
Affiliation(s)
| | | | | | - Eftekhar Azarm
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | | - Mohammad Amin Karimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Leifheit ME, Johnson G, Kuzel TM, Schneider JR, Barker E, Yun HD, Ustun C, Goldufsky JW, Gupta K, Marzo AL. Enhancing Therapeutic Efficacy of FLT3 Inhibitors with Combination Therapy for Treatment of Acute Myeloid Leukemia. Int J Mol Sci 2024; 25:9448. [PMID: 39273395 PMCID: PMC11394928 DOI: 10.3390/ijms25179448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) mutations are genetic changes found in approximately thirty percent of patients with acute myeloid leukemia (AML). FLT3 mutations in AML represent a challenging clinical scenario characterized by a high rate of relapse, even after allogeneic hematopoietic stem cell transplantation (allo-HSCT). The advent of FLT3 tyrosine kinase inhibitors (TKIs), such as midostaurin and gilteritinib, has shown promise in achieving complete remission. However, a substantial proportion of patients still experience relapse following TKI treatment, necessitating innovative therapeutic strategies. This review critically addresses the current landscape of TKI treatments for FLT3+ AML, with a particular focus on gilteritinib. Gilteritinib, a highly selective FLT3 inhibitor, has demonstrated efficacy in targeting the mutant FLT3 receptor, thereby inhibiting aberrant signaling pathways that drive leukemic proliferation. However, monotherapy with TKIs may not be sufficient to eradicate AML blasts. Specifically, we provide evidence for integrating gilteritinib with mammalian targets of rapamycin (mTOR) inhibitors and interleukin-15 (IL-15) complexes. The combination of gilteritinib, mTOR inhibitors, and IL-15 complexes presents a compelling strategy to enhance the eradication of AML blasts and enhance NK cell killing, offering a potential for improved patient outcomes.
Collapse
Affiliation(s)
- Malia E Leifheit
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Gunnar Johnson
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Timothy M Kuzel
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jeffrey R Schneider
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Edward Barker
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Hyun D Yun
- Hematology, Oncology, Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA
- Department of Medicine, Division of Hematology, Oncology, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Celalettin Ustun
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Josef W Goldufsky
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kajal Gupta
- Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Amanda L Marzo
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
6
|
Lap CJ, Abrahim MS, Nassereddine S. Perspectives and challenges of small molecule inhibitor therapy for FLT3-mutated acute myeloid leukemia. Ann Hematol 2024; 103:2215-2229. [PMID: 37975931 DOI: 10.1007/s00277-023-05545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous clonal disease characterized overall by an aggressive clinical course. The underlying genetic abnormalities present in leukemic cells contribute significantly to the AML phenotype. Mutations in FMS-like tyrosine kinase 3 (FLT3) are one of the most common genetic abnormalities identified in AML, and the presence of these mutations strongly influences disease presentation and negatively impacts prognosis. Since mutations in FLT3 were identified in AML, they have been recognized as a valid therapeutic target resulting in decades of research to develop effective small molecule inhibitor treatment that could improve outcome for these patients. Despite the approval of several FLT3 inhibitors over the last couple of years, the treatment of patients with FLT3-mutated AML remains challenging and many questions still need to be addressed. This review will provide an up-to-date overview of our current understanding of FLT3-mutated AML and discuss what the current status is of the available FLT3 inhibitors for the day-to-day management of this aggressive disease.
Collapse
Affiliation(s)
- Coen J Lap
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Marwa Sh Abrahim
- The George Washington Cancer Center, George Washington University, Washington, DC, USA
| | - Samah Nassereddine
- The George Washington Cancer Center, George Washington University, Washington, DC, USA.
| |
Collapse
|
7
|
Zhao Y, Zhang X, Ding X, Wang Y, Li Z, Zhao R, Cheng HE, Sun Y. Efficacy and safety of FLT3 inhibitors in monotherapy of hematological and solid malignancies: a systemic analysis of clinical trials. Front Pharmacol 2024; 15:1294668. [PMID: 38828446 PMCID: PMC11140126 DOI: 10.3389/fphar.2024.1294668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
Introduction: FLT3 mutations are closely associated with the occurrence of hematological and solid malignancies, especially with acute myeloid leukemia. Currently, several FLT3 inhibitors are in clinical trials, and some have been applied in clinic. However, the safety, efficacy and pharmacodynamics of these FLT3 inhibitors have not been systemically analyzed before. Methods: We searched and reviewed clinical trial reports on the monotherapy of 13 FLT3 inhibitors, including sorafenib, lestaurtinib, midostaurin, gilteritinib, quizartinib, sunitinib, crenolanib, tandutinib, cabozantinib, pexidartinib, pacritinib, famitinib, and TAK-659 in patients with hematological and solid malignancies before May 31, 2023. Results: Our results showed the most common adverse events (AEs) were gastrointestinal adverse reactions, including diarrhea, hand-foot syndrome and nausea, while the most common hematological AEs were febrile neutropenia, anemia, and thrombocytopenia. Based on the published data, the mean overall survival (OS) and the mean progression-free survival (PFS) were 9.639 and 5.905 months, respectively. The incidence of overall response rate (ORR), complete remission (CR), partial response (PR), and stable disease (SD) for all these FLT3 inhibitors was 29.0%, 8.7%, 16.0%, and 42.3%, respectively. The ORRs of FLT3 inhibitors in hematologic malignancies and solid tumors were 40.8% and 18.8%, respectively, indicating FLT3 inhibitors were more effective for hematologic malignancies than for solid tumors. In addition, time to maximum plasma concentration (Tmax) in these FLT3 inhibitors ranged from 0.7-12.0 hours, but the elimination half-life (T1/2) range was highly variable, from 6.8 to 151.8 h. Discussion: FLT3 inhibitors monotherapy has shown significant anti-tumor effect in clinic, and the effectiveness may be further improved through combination medication.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hai-En Cheng
- School of Medical Laboratory, Shandong Second Medical University, Weifang, China
| | - Yanli Sun
- School of Medical Laboratory, Shandong Second Medical University, Weifang, China
| |
Collapse
|
8
|
Monogiou Belik D, Bernasconi R, Xu L, Della Verde G, Lorenz V, Grüterich V, Balzarolo M, Mochizuki M, Pfister O, Kuster GM. The Flt3-inhibitor quizartinib augments apoptosis and promotes maladaptive remodeling after myocardial infarction in mice. Apoptosis 2024; 29:357-371. [PMID: 37945814 PMCID: PMC10873224 DOI: 10.1007/s10495-023-01911-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) targeting fms-like tyrosine kinase 3 (Flt3) such as quizartinib were specifically designed for acute myeloid leukemia treatment, but also multi-targeting TKIs applied to solid tumor patients inhibit Flt3. Flt3 is expressed in the heart and its activation is cytoprotective in myocardial infarction (MI) in mice. OBJECTIVES We sought to test whether Flt3-targeting TKI treatment aggravates cardiac injury after MI. METHODS AND RESULTS Compared to vehicle, quizartinib (10 mg/kg/day, gavage) did not alter cardiac dimensions or function in healthy mice after four weeks of therapy. Pretreated mice were randomly assigned to MI or sham surgery while receiving quizartinib or vehicle for one more week. Quizartinib did not aggravate the decline in ejection fraction, but significantly enhanced ventricular dilatation one week after infarction. In addition, apoptotic cell death was significantly increased in the myocardium of quizartinib-treated compared to vehicle-treated mice. In vitro, quizartinib dose-dependently decreased cell viability in neonatal rat ventricular myocytes and in H9c2 cells, and increased apoptosis as assessed in the latter. Together with H2O2, quizartinib potentiated the phosphorylation of the pro-apoptotic mitogen activated protein kinase p38 and augmented H2O2-induced cell death and apoptosis beyond additive degree. Pretreatment with a p38 inhibitor abolished apoptosis under quizartinib and H2O2. CONCLUSION Quizartinib potentiates apoptosis and promotes maladaptive remodeling after MI in mice at least in part via a p38-dependent mechanism. These findings are consistent with the multi-hit hypothesis of cardiotoxicity and make cardiac monitoring in patients with ischemic heart disease under Flt3- or multi-targeting TKIs advisable.
Collapse
Affiliation(s)
- Daria Monogiou Belik
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Riccardo Bernasconi
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Lifen Xu
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Giacomo Della Verde
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Vera Lorenz
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Vivienne Grüterich
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Melania Balzarolo
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Michika Mochizuki
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Otmar Pfister
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Basel, Basel, Switzerland
| | - Gabriela M Kuster
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland.
- Department of Cardiology, University Heart Center, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
9
|
Murdock HM, Ho VT, Garcia JS. Innovations in conditioning and post-transplant maintenance in AML: genomically informed revelations on the graft-versus-leukemia effect. Front Immunol 2024; 15:1359113. [PMID: 38571944 PMCID: PMC10987864 DOI: 10.3389/fimmu.2024.1359113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/20/2024] [Indexed: 04/05/2024] Open
Abstract
Acute Myeloid Leukemia (AML) is the prototype of cancer genomics as it was the first published cancer genome. Large-scale next generation/massively parallel sequencing efforts have identified recurrent alterations that inform prognosis and have guided the development of targeted therapies. Despite changes in the frontline and relapsed standard of care stemming from the success of small molecules targeting FLT3, IDH1/2, and apoptotic pathways, allogeneic stem cell transplantation (alloHSCT) and the resulting graft-versus-leukemia (GVL) effect remains the only curative path for most patients. Advances in conditioning regimens, graft-vs-host disease prophylaxis, anti-infective agents, and supportive care have made this modality feasible, reducing transplant related mortality even among patients with advanced age or medical comorbidities. As such, relapse has emerged now as the most common cause of transplant failure. Relapse may occur after alloHSCT because residual disease clones persist after transplant, and develop immune escape from GVL, or such clones may proliferate rapidly early after alloHSCT, and outpace donor immune reconstitution, leading to relapse before any GVL effect could set in. To address this issue, genomically informed therapies are increasingly being incorporated into pre-transplant conditioning, or as post-transplant maintenance or pre-emptive therapy in the setting of mixed/falling donor chimerism or persistent detectable measurable residual disease (MRD). There is an urgent need to better understand how these emerging therapies modulate the two sides of the GVHD vs. GVL coin: 1) how molecularly or immunologically targeted therapies affect engraftment, GVHD potential, and function of the donor graft and 2) how these therapies affect the immunogenicity and sensitivity of leukemic clones to the GVL effect. By maximizing the synergistic action of molecularly targeted agents, immunomodulating agents, conventional chemotherapy, and the GVL effect, there is hope for improving outcomes for patients with this often-devastating disease.
Collapse
Affiliation(s)
- H. Moses Murdock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Vincent T. Ho
- Bone Marrow Transplant Program, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Jacqueline S. Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| |
Collapse
|
10
|
Bhatia K, Sandhu V, Wong MH, Iyer P, Bhatt S. Therapeutic biomarkers in acute myeloid leukemia: functional and genomic approaches. Front Oncol 2024; 14:1275251. [PMID: 38410111 PMCID: PMC10894932 DOI: 10.3389/fonc.2024.1275251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/17/2024] [Indexed: 02/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is clinically and genetically a heterogeneous disease characterized by clonal expansion of abnormal hematopoietic progenitors. Genomic approaches to precision medicine have been implemented to direct targeted therapy for subgroups of AML patients, for instance, IDH inhibitors for IDH1/2 mutated patients, and FLT3 inhibitors with FLT3 mutated patients. While next generation sequencing for genetic mutations has improved treatment outcomes, only a fraction of AML patients benefit due to the low prevalence of actionable targets. In recent years, the adoption of newer functional technologies for quantitative phenotypic analysis and patient-derived avatar models has strengthened the potential for generalized functional precision medicine approach. However, functional approach requires robust standardization for multiple variables such as functional parameters, time of drug exposure and drug concentration for making in vitro predictions. In this review, we first summarize genomic and functional therapeutic biomarkers adopted for AML therapy, followed by challenges associated with these approaches, and finally, the future strategies to enhance the implementation of precision medicine.
Collapse
Affiliation(s)
- Karanpreet Bhatia
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Vedant Sandhu
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Mei Hsuan Wong
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Prasad Iyer
- Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore, Singapore
- Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
Wang X, DeFilippis RA, Leung YK, Shah NP, Li HY. N-(3-Methoxyphenyl)-6-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)pyridin-2-amine is an inhibitor of the FLT3-ITD and BCR-ABL pathways, and potently inhibits FLT3-ITD/D835Y and FLT3-ITD/F691L secondary mutants. Bioorg Chem 2024; 143:106966. [PMID: 37995643 PMCID: PMC11586108 DOI: 10.1016/j.bioorg.2023.106966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023]
Abstract
Activating mutations within FLT3 make up 30 % of all newly diagnosed acute myeloid leukemia (AML) cases, with the most common mutation being an internal tandem duplication (FLT3-ITD) in the juxtamembrane region (25 %). Currently, two generations of FLT3 kinase inhibitors have been developed, with three inhibitors clinically approved. However, treatment of FLT3-ITD mutated AML is limited due to the emergence of secondary clinical resistance, caused by multiple mechanism including on-target FLT3 secondary mutations - FLT3-ITD/D835Y and FLT3-ITD/F691L being the most common, as well as the off-target activation of alternative pathways including the BCR-ABL pathway. Through the screening of imidazo[1,2-a]pyridine derivatives, N-(3-methoxyphenyl)-6-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)pyridin-2-amine (compound 1) was identified as an inhibitor of both the FLT3-ITD and BCR-ABL pathways. Compound 1 potently inhibits clinically related leukemia cell lines driven by FLT3-ITD, FLT3-ITD/D835Y, FLT3-ITD/F691L, or BCR-ABL. Studies indicate that it mediates proapoptotic effects on cells by inhibiting FLT3 and BCR-ABL pathways, and other possible targets. Compound 1 is more potent against FLT3-ITD than BCR-ABL, and it may have other possible targets; however, compound 1 is first step for further optimization for the development of a balanced FLT3-ITD/BCR-ABL dual inhibitor for the treatment of relapsed FLT3-ITD mutated AML with multiple secondary clinical resistant subtypes such as FLT3-ITD/D835Y, FLT3-ITD/F691L, and cells co-expressing FLT3-ITD and BCR-ABL.
Collapse
Affiliation(s)
- Xiuqi Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Rosa Anna DeFilippis
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Yuet-Kin Leung
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Neil P Shah
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
12
|
Wang X, DeFilippis RA, Weldemichael T, Gunaganti N, Tran P, Leung YK, Shah NP, Li HY. An imidazo[1,2-a]pyridine-pyridine derivative potently inhibits FLT3-ITD and FLT3-ITD secondary mutants, including gilteritinib-resistant FLT3-ITD/F691L. Eur J Med Chem 2024; 264:115977. [PMID: 38056299 PMCID: PMC11590664 DOI: 10.1016/j.ejmech.2023.115977] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 11/07/2023] [Accepted: 11/18/2023] [Indexed: 12/08/2023]
Abstract
FLT3 activating mutations are detected in approximately 30 % of newly diagnosed acute myeloid leukemia (AML) cases, most commonly consisting of internal tandem duplication (ITD) mutations in the juxtamembrane region. Recently, several FLT3 inhibitors have demonstrated clinical activity and three are currently approved - midostaurin, quizartinib, and gilteritinib. Midostaurin is a first-generation FLT3 inhibitor with minimal activity as monotherapy. Midostaurin lacks selectivity and is only approved by the USFDA for use in combination with other chemotherapy agents. The second-generation inhibitors quizartinib and gilteritinib display improved specificity and selectivity, and have been approved for use as monotherapy. However, their clinical efficacies are limited in part due to the emergence of drug-resistant FLT3 secondary mutations in the tyrosine kinase domain at positions D835 and F691. Therefore, in order to overcome drug resistance and further improve outcomes, new compounds targeting FLT3-ITD with secondary mutants are urgently needed. In this study, through the structural modification of a reported compound Ling-5e, we identified compound 24 as a FLT3 inhibitor that is equally potent against FLT3-ITD and the clinically relevant mutants FLT3-ITD/D835Y, and FLT3-ITD/F691L. Its inhibitory effects were demonstrated in both cell viability assays and western blots analyses. When tested against cell lines lacking activating mutations in FLT3, no non-specific cytotoxicity effect was observed. Interestingly, molecular docking results showed that compound 24 may adopt different binding conformations with FLT3-F691L compared to FLT3, which may explain its retained activity against FLT3-ITD/F691L. In summary, compound 24 has inhibition potency on FLT3 comparable to gilteritinib, but a more balanced inhibition on FLT3 secondary mutations, especially FLT3-ITD/F691L which is gilteritinib resistant. Compound 24 may serve as a promising lead for the drug development of either primary or relapsed AML with FLT3 secondary mutations.
Collapse
Affiliation(s)
- Xiuqi Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Rosa Anna DeFilippis
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Tsigereda Weldemichael
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Naresh Gunaganti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Phuc Tran
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yuet-Kin Leung
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Neil P Shah
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
13
|
Bergeron J, Capo-Chichi JM, Tsui H, Mahe E, Berardi P, Minden MD, Brandwein JM, Schuh AC. The Clinical Utility of FLT3 Mutation Testing in Acute Leukemia: A Canadian Consensus. Curr Oncol 2023; 30:10410-10436. [PMID: 38132393 PMCID: PMC10742150 DOI: 10.3390/curroncol30120759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) mutations are detected in approximately 20-30% of patients with acute myeloid leukemia (AML), with the presence of a FLT3 internal tandem duplication (FLT3-ITD) mutation being associated with an inferior outcome. Assessment of FLT3 mutational status is now essential to define optimal upfront treatment in both newly diagnosed and relapsed AML, to support post-induction allogeneic hematopoietic stem cell transplantation (alloSCT) decision-making, and to evaluate treatment response via measurable (minimal) residual disease (MRD) evaluation. In view of its importance in AML diagnosis and management, the Canadian Leukemia Study Group/Groupe canadien d'étude sur la leucémie (CLSG/GCEL) undertook the development of a consensus statement on the clinical utility of FLT3 mutation testing, as members reported considerable inter-center variability across Canada with respect to testing availability and timing of use, methodology, and interpretation. The CLSG/GCEL panel identified key clinical and hematopathological questions, including: (1) which patients should be tested for FLT3 mutations, and when?; (2) which is the preferred method for FLT3 mutation testing?; (3) what is the clinical relevance of FLT3-ITD size, insertion site, and number of distinct FLT3-ITDs?; (4) is there a role for FLT3 analysis in MRD assessment?; (5) what is the clinical relevance of the FLT3-ITD allelic burden?; and (6) how should results of FLT3 mutation testing be reported? The panel followed an evidence-based approach, taken together with Canadian clinical and laboratory experience and expertise, to create a consensus document to facilitate a more uniform approach to AML diagnosis and treatment across Canada.
Collapse
Affiliation(s)
- Julie Bergeron
- CEMTL Installation Maisonneuve-Rosemont, Institut Universitaire d’Hématologie-Oncologie et de Thérapie Cellulaire, Université de Montréal, Montréal, QC H1T 2M4, Canada
| | - Jose-Mario Capo-Chichi
- Division of Clinical Laboratory Genetics, Department of Laboratory Medicine and Pathobiology, Laboratory Medicine Program, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada;
| | - Hubert Tsui
- Division of Hematological Pathology, Department of Laboratory Medicine and Molecular Diagnostics, Precision Diagnostics and Therapeutics Program, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Laboratory Medicine and Pathobiology, Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Etienne Mahe
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Division of Hematology and Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Philip Berardi
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital/Eastern Ontario Regional Laboratory Association, Ottawa, ON K1H 8M2, Canada;
- Department of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mark D. Minden
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.D.M.); (A.C.S.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Joseph M. Brandwein
- Division of Hematology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada;
| | - Andre C. Schuh
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.D.M.); (A.C.S.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
14
|
Pan X, Chang Y, Ruan G, Wei F, Jiang H, Jiang Q, Huang X, Zhao X. Prognostic impact of FLT3-ITD mutation on NPM1 + acute myeloid leukaemia patients and related molecular mechanisms. Br J Haematol 2023; 203:212-223. [PMID: 37621257 DOI: 10.1111/bjh.18973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/12/2023] [Accepted: 06/28/2023] [Indexed: 08/26/2023]
Abstract
The prognosis of acute myeloid leukaemia (AML) patients carrying NPM1 mutations is significantly worse when accompanied by FLT3-ITD mutations. However, accurate quantitative detection of FLT3-ITD mutations remains challenging. To identify a novel biomarker in NPM1+ FLT3-ITD+ AML patients for more accurate stratification, we analysed the differential gene expression between the NPM1+ FLT3-ITD+ and NPM1+ FLT3-ITD- groups in five public AML datasets and identified a biomarker by taking the intersection of differentially expressed genes. We validated this biomarker in bone marrow samples from NPM1+ AML patients at the Peking University Institute of Haematology and analysed its prognostic significance. BCAT1 expression was higher in the NPM1+ FLT3-ITD+ group than in the NPM1+ FLT3-ITD- group in all seven cohorts. BCAT1 was able to predict the prognosis of NPM1+ FLT3-ITD+ AML patients, and its predictive ability was superior to that of the FLT3-ITD allelic ratio (AR). FLT3-targeted inhibitor quizartinib reduced BCAT1 expression. BCAT1 knockdown using lentiviral vectors led to the downregulation of MYC expression. Thus, we identified BCAT1 as a novel biomarker for NPM1+ FLT3-ITD+ AML patients. The FLT3-ITD/BCAT1/MYC signalling pathway may play a biological role in promoting the occurrence and development of AML in FLT3-ITD+ cell lines.
Collapse
Affiliation(s)
- Xin'an Pan
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Yingjun Chang
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Guorui Ruan
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Fangfang Wei
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Hao Jiang
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Qian Jiang
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Xiaojun Huang
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaosu Zhao
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Cremer A, Enssle JC, Pfaff S, Kouidri K, Lang F, Brandts C, Zeiher A, Cremer S, Steffen B, Serve H, Bug G. Treatment with midostaurin and other FLT3 targeting inhibitors is associated with an increased risk of cardiovascular adverse events in patients who underwent allogeneic hematopoietic stem cell transplantation with FLT3-mutated AML. Ann Hematol 2023; 102:2903-2908. [PMID: 37552323 PMCID: PMC10492676 DOI: 10.1007/s00277-023-05396-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/29/2023] [Indexed: 08/09/2023]
Abstract
The addition of midostaurin to standard chemotherapy has improved survival in patients with FLT3-mutated AML. However, the impact of midostaurin and other FLT3 inhibitors (FLT3i) on cardiovascular adverse events (CAEs) has not been studied in patients who underwent allogeneic hematopoietic stem cell transplantation in a real-world setting. We reviewed 132 patients with AML who were treated with intensive induction therapy and consecutive allogeneic stem cell transplantation at our institution (42 FLT3-mutated AML and 90 with FLT3 wildtype). We identified treatment with midostaurin and/or FLT3i as an independent risk factor for CAEs not resulting in higher non-relapse mortality (NRM) or impaired overall survival (OS). Hence, close monitoring for CAEs is warranted for these patients.
Collapse
Affiliation(s)
- Anjali Cremer
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany.
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany.
| | - Julius C Enssle
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany
| | - Saskia Pfaff
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Khouloud Kouidri
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Fabian Lang
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Christian Brandts
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University, Frankfurt, Germany
| | - Andreas Zeiher
- Department of Medicine, Cardiology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Sebastian Cremer
- Department of Medicine, Cardiology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Björn Steffen
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany
| | - Hubert Serve
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany
| | - Gesine Bug
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany
| |
Collapse
|
16
|
Jaramillo S, Le Cornet L, Kratzmann M, Krisam J, Görner M, Hänel M, Röllig C, Wass M, Scholl S, Ringhoffer M, Reichart A, Steffen B, Kayser S, Mikesch JH, Schaefer-Eckart K, Schubert J, Geer T, Martin S, Kieser M, Sauer T, Kriegsmann K, Hundemer M, Serve H, Bornhäuser M, Müller-Tidow C, Schlenk RF. Q-HAM: a multicenter upfront randomized phase II trial of quizartinib and high-dose Ara-C plus mitoxantrone in relapsed/refractory AML with FLT3-ITD. Trials 2023; 24:591. [PMID: 37715270 PMCID: PMC10504729 DOI: 10.1186/s13063-023-07421-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/27/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND About 50% of older patients with acute myeloid leukemia (AML) fail to attain complete remission (CR) following cytarabine plus anthracycline-based induction therapy. Salvage chemotherapy regimens are based on high-dose cytarabine (HiDAC), which is frequently combined with mitoxantrone (HAM regimen). However, CR rates remain low, with less than one-third of the patients achieving a CR. FLT3-ITD has consistently been identified as an unfavorable molecular marker in both relapsed and refractory (r/r)-AML. One-quarter of patients who received midostaurin are refractory to induction therapy and relapse rate at 2 years exceeds 40%. The oral second-generation bis-aryl urea tyrosine kinase inhibitor quizartinib is a very selective FLT3 inhibitor, has a high capacity for sustained FLT3 inhibition, and has an acceptable toxicity profile. METHODS In this multicenter, upfront randomized phase II trial, all patients receive quizartinib combined with HAM (cytarabine 3g/m2 bidaily day one to day three, mitoxantrone 10mg/m2 days two and three) during salvage therapy. Efficacy is assessed by comparison to historical controls based on the matched threshold crossing approach with achievement of CR, complete remission with incomplete hematologic recovery (CRi), or complete remission with partial recovery of peripheral blood counts (CRh) as primary endpoint. During consolidation therapy (chemotherapy and allogeneic hematopoietic cell transplantation), patients receive either prophylactic quizartinib therapy or measurable residual disease (MRD)-triggered preemptive continuation therapy with quizartinib according to up-front randomization. The matched threshold crossing approach is a novel study-design to enhance the classic single-arm trial design by including matched historical controls from previous clinical studies. It overcomes common disadvantages of single-armed and small randomized studies, since the expected outcome of the observed study population can be adjusted based on the matched controls with a comparable distribution of known prognostic and predictive factors. Furthermore, balanced treatment groups lead to stable statistical models. However, one of the limitations of our study is the inability to adjust for unobserved or unknown confounders. Addressing the primary endpoint, CR/CRi/CRh after salvage therapy, the maximal sample size of 80 patients is assessed generating a desirable power of the used adaptive design, assuming a logistic regression is performed at a one-sided significance level α=0.05, the aspired power is 0.8, and the number of matching partners per intervention patient is at least 1. After enrolling 20 patients, the trial sample size will be recalculated in an interim analysis based on a conditional power argument. CONCLUSION Currently, there is no commonly accepted standard for salvage chemotherapy treatment. The objective of the salvage therapy is to reduce leukemic burden, achieve the best possible remission, and perform a hemopoietic stem-cell transplantation. Thus, in patients with FLT3-ITD mutation, the comparison of quizartinib with intensive salvage therapy versus chemotherapy alone appears as a logical consequence in terms of efficacy and safety. ETHICS AND DISSEMINATION Ethical approval and approvals from the local and federal competent authorities were granted. Trial results will be reported via peer-reviewed journals and presented at conferences and scientific meetings. TRIAL REGISTRATION ClinicalTrials.gov NCT03989713; EudraCT Number: 2018-002675-17.
Collapse
Affiliation(s)
- Sonia Jaramillo
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.
| | - Lucian Le Cornet
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Markus Kratzmann
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Johannes Krisam
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | - Martin Görner
- Department of Hematology, Oncology and Palliative Medicine, Community Hospital Bielefeld, Bielefeld, Germany
| | - Mathias Hänel
- Department of Medicine III, Hospital Chemnitz gGmbH, Chemnitz, Germany
| | - Christoph Röllig
- Department of Medicine and Polyclinic I, TU Dresden University Hospital, Dresden, Germany
| | - Maxi Wass
- Department of Medicine IV, Halle (Saale) University Hospital, Halle, Germany
| | - Sebastian Scholl
- Department of Medicine II, Jena University Hospital, Jena, Germany
| | - Mark Ringhoffer
- Department of Medicine, III, Hospital Karlsruhe, Karlsruhe, Germany
| | - Alexander Reichart
- Department of Hematology, Oncology and Palliative Medicine, Hospital Winnenden, Winnenden, Germany
| | - Björn Steffen
- Department of Medicine II, Frankfurt University Hospital, Frankfurt, Germany
| | - Sabine Kayser
- Department of Medicine I - Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | | | | | - Jörg Schubert
- Department of Inner Medicine II, Elbland Hospital Riesa, Riesa, Germany
| | - Thomas Geer
- Department of Medicine II, Diaconal Hospital Schwäbisch-Hall, Schwäbisch Hall, Germany
| | - Sonja Martin
- Department of Hematology, Oncology and Palliative Medicine, Robert-Bosch Hospital, Stuttgart, Germany
| | - Meinhard Kieser
- Department of Hematology, Oncology and Palliative Medicine, Robert-Bosch Hospital, Stuttgart, Germany
| | - Tim Sauer
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Katharina Kriegsmann
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Hundemer
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Hubert Serve
- Department of Medicine II, Frankfurt University Hospital, Frankfurt, Germany
| | - Martin Bornhäuser
- Department of Medicine and Polyclinic I, TU Dresden University Hospital, Dresden, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard F Schlenk
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
17
|
Li X, Wang P, Wang C, Jin T, Xu R, Tong L, Hu X, Shen L, Li J, Zhou Y, Liu T. Discovery of 2-Aminopyrimidine Derivatives as Potent Dual FLT3/CHK1 Inhibitors with Significantly Reduced hERG Inhibitory Activities. J Med Chem 2023; 66:11792-11814. [PMID: 37584545 DOI: 10.1021/acs.jmedchem.3c00245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
FLT3 inhibitors as single agents have limited effects because of acquired and adaptive resistance and the cardiotoxicity related to human ether-a-go-go-related gene (hERG) channel blockade further impedes safe drugs to the market. Inhibitors having potential to overcome resistance and reduce hERG affinity are highly demanded. Here, we reported a dual FLT3/CHK1 inhibitor 18, which displayed potencies to overcome varying acquired resistance in BaF3 cells with FLT3-TKD and FLT3-ITD-TKD mutations. Moreover, 18 displayed high selectivity over c-KIT more than 1700-fold and greatly reduced hERG affinity, with an IC50 value of 58.4 μM. Further mechanistic studies demonstrated 18 can upregulate p53 and abolish the outgrowth of adaptive resistant cells. In the in vivo studies, 18 demonstrated favorable PK profiles and good safety, suppressed the tumor growth in the MV-4-11 cell inoculated mouse xenograft model, and prolonged the survival in the Molm-13 transplantation model, supporting its further development.
Collapse
Affiliation(s)
- Xuemei Li
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Peipei Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Chang Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Tingting Jin
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou310006, P.R. China
| | - Ran Xu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Lexian Tong
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, P. R. China
| | - Xiaobei Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District,Guangdong 528400, P. R. China
| | - Liteng Shen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou310058, P.R. China
| | - Jia Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District,Guangdong 528400, P. R. China
| | - Yubo Zhou
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District,Guangdong 528400, P. R. China
| | - Tao Liu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou310058, P.R. China
| |
Collapse
|
18
|
Wu RH, Zhu CY, Yu PH, Ma Y, Hussain L, Naranmandura H, Wang QQ. The landscape of novel strategies for acute myeloid leukemia treatment: Therapeutic trends, challenges, and future directions. Toxicol Appl Pharmacol 2023; 473:116585. [PMID: 37302559 DOI: 10.1016/j.taap.2023.116585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous subtype of hematological malignancies with a wide spectrum of cytogenetic and molecular abnormalities, which makes it difficult to manage and cure. Along with the deeper understanding of the molecular mechanisms underlying AML pathogenesis, a large cohort of novel targeted therapeutic approaches has emerged, which considerably expands the medical options and changes the therapeutic landscape of AML. Despite that, resistant and refractory cases caused by genomic mutations or bypass signalling activation remain a great challenge. Therefore, discovery of novel treatment targets, optimization of combination strategies, and development of efficient therapeutics are urgently required. This review provides a detailed and comprehensive discussion on the advantages and limitations of targeted therapies as a single agent or in combination with others. Furthermore, the innovative therapeutic approaches including hyperthermia, monoclonal antibody-based therapy, and CAR-T cell therapy are also introduced, which may provide safe and viable options for the treatment of patients with AML.
Collapse
Affiliation(s)
- Ri Han Wu
- College of Life Sciences, Changchun Normal University, Changchun 130032, China
| | - Chen Ying Zhu
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Pei Han Yu
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yafang Ma
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liaqat Hussain
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Hua Naranmandura
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Qian Qian Wang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
19
|
Yang J, Friedman R. Combination strategies to overcome drug resistance in FLT + acute myeloid leukaemia. Cancer Cell Int 2023; 23:161. [PMID: 37568211 PMCID: PMC10416533 DOI: 10.1186/s12935-023-03000-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Acute myeloid leukaemia (AML) remains difficult to treat despite the development of novel formulations and targeted therapies. Activating mutations in the FLT3 gene are common among patients and make the tumour susceptible to FLT3 inhibitors, but resistance to such inhibitors develops quickly. METHODS We examined combination therapies aimed at FLT3+-AML, and studied the development of resistance using a newly developed protocol. Combinations of FLT3, CDK4/6 and PI3K inhibitors were tested for synergism. RESULTS We show that AML cells express CDK4 and that the CDK4/6 inhibitors palbociclib and abemaciclib inhibit cellular growth. PI3K inhibitors were also effective in inhibiting the growth of AML cell lines that express FLT3-ITD. Whereas resistance to quizartinib develops quickly, the combinations overcome such resistance. CONCLUSIONS This study suggests that a multi-targeted intervention involving a CDK4/6 inhibitor with a FLT3 inhibitor or a pan-PI3K inhibitor might be a valuable therapeutic strategy for AML to overcome drug resistance. Moreover, many patients cannot tolerate high doses of the drugs that were studied (quizartinib, palbociclib and PI3K inhibitors) for longer periods, and it is therefore of high significance that the drugs act synergistically and lower doses can be used.
Collapse
Affiliation(s)
- Jingmei Yang
- Department of Chemistry and Biomedical Science, Linnaeus University, Kalmar Campus, 391 82, Kalmar, Sweden
| | - Ran Friedman
- Department of Chemistry and Biomedical Science, Linnaeus University, Kalmar Campus, 391 82, Kalmar, Sweden.
| |
Collapse
|
20
|
Darici S, Jørgensen HG, Huang X, Serafin V, Antolini L, Barozzi P, Luppi M, Forghieri F, Marmiroli S, Zavatti M. Improved efficacy of quizartinib in combination therapy with PI3K inhibition in primary FLT3-ITD AML cells. Adv Biol Regul 2023; 89:100974. [PMID: 37245251 DOI: 10.1016/j.jbior.2023.100974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/09/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Acute myeloid leukemia is a heterogeneous hematopoietic malignancy, characterized by uncontrolled clonal proliferation of abnormal myeloid progenitor cells, with poor outcomes. The internal tandem duplication (ITD) mutation of the Fms-like receptor tyrosine kinase 3 (FLT3) (FLT3-ITD) represents the most common genetic alteration in AML, detected in approximately 30% of AML patients, and is associated with high leukemic burden and poor prognosis. Therefore, this kinase has been regarded as an attractive druggable target for the treatment of FLT3-ITD AML, and selective small molecule inhibitors, such as quizartinib, have been identified and trialled. However, clinical outcomes have been disappointing so far due to poor remission rates, also because of acquired resistance. A strategy to overcome resistance is to combine FLT3 inhibitors with other targeted therapies. In this study, we investigated the preclinical efficacy of the combination of quizartinib with the pan PI3K inhibitor BAY-806946 in FLT3-ITD cell lines and primary cells from AML patients. We show here that BAY-806946 enhanced quizartinib cytotoxicity and, most importantly, that this combination increases the ability of quizartinib to kill CD34+ CD38-leukemia stem cells, whilst sparing normal hematopoietic stem cells. Because constitutively active FLT3 receptor tyrosine kinase is known to boost aberrant PI3K signaling, the increased sensitivity of primary cells to the above combination can be the mechanistic results of the disruption of signaling by vertical inhibition.
Collapse
Affiliation(s)
- Salihanur Darici
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, Largo del Pozzo 71, University of Modena and Reggio Emilia, Modena, 41125, Italy; Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Heather G Jørgensen
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Xu Huang
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Valentina Serafin
- Department of Surgery Oncology and Gastroenterology Oncology and Immunology Section University of Padova, Italy
| | - Ludovica Antolini
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, Largo del Pozzo 71, University of Modena and Reggio Emilia, Modena, 41125, Italy
| | - Patrizia Barozzi
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Di Modena, Via del Pozzo 71, 41124, Modena, Italy
| | - Mario Luppi
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Di Modena, Via del Pozzo 71, 41124, Modena, Italy.
| | - Fabio Forghieri
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Di Modena, Via del Pozzo 71, 41124, Modena, Italy.
| | - Sandra Marmiroli
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, Largo del Pozzo 71, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Manuela Zavatti
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, Largo del Pozzo 71, University of Modena and Reggio Emilia, Modena, 41125, Italy
| |
Collapse
|
21
|
Choi JH, Shukla M, Abdul-Hay M. Acute Myeloid Leukemia Treatment in the Elderly: A Comprehensive Review of the Present and Future. Acta Haematol 2023; 146:431-457. [PMID: 37459852 DOI: 10.1159/000531628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 06/17/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a disease of the hematopoietic system that remains a therapeutic challenge despite advances in our understanding of the underlying cancer biology in the past decade. It is also an affliction of the elderly that predominantly affects patients over 60 years of age. Standard therapy involves intensive chemotherapy that is often difficult to tolerate in older populations. Fortunately, recent developments in molecular targeting have shown promising results in treating leukemia, paving the way for novel treatment strategies that are easier to tolerate. SUMMARY Venetoclax, a BCL-2 inhibitor, when combined with a hypomethylating agent, has proven to be a highly effective and well-tolerated drug and established itself as a new standard for treating AML in patients who are unfit for standard intensive therapy. Other targeted therapies include clinically proven and FDA-approved agents, such as IDH1/2 inhibitors, FLT3 inhibitors, and Gemtuzumab, as well as newer and more experimental drugs such as magrolimab, PI-kinase inhibitors, and T-cell engaging therapy. Some of the novel agents such as magrolimab and menin inhibitors are particularly promising, providing therapeutic options to a wider population of patients than ever before. Determining who will benefit from intense or novel low-intense therapy remains a challenge, and it requires careful assessment of individual patient's fitness and disease characteristics. KEY MESSAGES This article reviews past and current treatment strategies that harness various mechanisms of leukemia-targeting agents and introduces novel therapies on the horizon aimed at exploring therapeutic options for the elderly and unfit patient population. It also provides a strategy to select the best available therapy for elderly patients with both newly diagnosed and relapsed/refractory AML.
Collapse
Affiliation(s)
- Jun H Choi
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
- Division of Hematology and Medical Oncology, New York University Perlmutter Cancer Center, New York, New York, USA
| | - Mihir Shukla
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Maher Abdul-Hay
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
- Division of Hematology and Medical Oncology, New York University Perlmutter Cancer Center, New York, New York, USA
| |
Collapse
|
22
|
Bhansali RS, Pratz KW, Lai C. Recent advances in targeted therapies in acute myeloid leukemia. J Hematol Oncol 2023; 16:29. [PMID: 36966300 PMCID: PMC10039574 DOI: 10.1186/s13045-023-01424-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/14/2023] [Indexed: 03/27/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults. While survival for younger patients over the last several decades has improved nearly sixfold with the optimization of intensive induction chemotherapy and allogeneic stem cell transplantation (alloHSCT), this effect has been largely mitigated in older and less fit patients as well as those with adverse-risk disease characteristics. However, the last 10 years has been marked by major advances in the molecular profiling of AML characterized by a deeper understanding of disease pathobiology and therapeutic vulnerabilities. In this regard, the classification of AML subtypes has recently evolved from a morphologic to a molecular and genetic basis, reflected by recent updates from the World Health Organization and the new International Consensus Classification system. After years of stagnation in new drug approvals for AML, there has been a rapid expansion of the armamentarium against this disease since 2017. Low-intensity induction therapy with hypomethylating agents and venetoclax has substantially improved outcomes, including in those previously considered to have a poor prognosis. Furthermore, targeted oral therapies against driver mutations in AML have been added to the repertoire. But with an accelerated increase in treatment options, several questions arise such as how to best sequence therapy, how to combine therapies, and if there is a role for maintenance therapy in those who achieve remission and cannot undergo alloHSCT. Moreover, certain subtypes of AML, such as those with TP53 mutations, still have dismal outcomes despite these recent advances, underscoring an ongoing unmet need and opportunity for translational advances. In this review, we will discuss recent updates in the classification and risk stratification of AML, explore the literature regarding low-intensity and novel oral combination therapies, and briefly highlight investigative agents currently in early clinical development for high-risk disease subtypes.
Collapse
Affiliation(s)
- Rahul S Bhansali
- Division of Hematology/Oncology, Department of Medicine, Hospital of the University of Pennsylvania, South Pavilion, 12th Floor, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Keith W Pratz
- Division of Hematology/Oncology, Department of Medicine, Hospital of the University of Pennsylvania, South Pavilion, 12th Floor, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Catherine Lai
- Division of Hematology/Oncology, Department of Medicine, Hospital of the University of Pennsylvania, South Pavilion, 12th Floor, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
23
|
Solana-Altabella A, Megías-Vericat JE, Ballesta-López O, Martínez-Cuadrón D, Montesinos P. Drug-drug interactions associated with FLT3 inhibitors for acute myeloblastic leukemia: current landscape. Expert Rev Clin Pharmacol 2023; 16:133-148. [PMID: 36708283 DOI: 10.1080/17512433.2023.2174523] [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: 01/29/2023]
Abstract
INTRODUCTION FLT3 inhibitors (FLT3i) are drugs in which there is limited experience and not yet enough information on the mechanisms of absorption, transport, and elimination; but especially on the potential drug-drug interactions (DDIs). There are therefore risks in the management of FLT3i DDIs (i.e. sorafenib, ponatinib, crenolanib, midostaurin, quizartinib, and gilteritinib) and ignoring them can compromise therapeutic success in acute myeloid leukemia (AML) treatment, in complex patients and secondary pathologies. AREAS COVERED This review summarizes the DDIs of FLT3i with P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), organic anion transporting (OAT), organic cationic transporting (OCT), cytochrome P450 (CYP) subunits, and other minor metabolic/transport pathways. EMBASE, PubMed, the Cochrane Central Register and the Web of Science were searched. The last literature search was performed on the 14 February 2022. EXPERT OPINION FLT3i will be combined with other therapeutic agents (supportive care, doublet, or triplet therapy) and in different clinical settings, which means a greater chance of controlling and even eradicating the disease effectively, but also an increased risk to patients due to potential DDIs. Healthcare professionals should be aware of the potential interactions that may occur and be vigilant in monitoring those patients who are receiving any potentially interacting drug.
Collapse
Affiliation(s)
- Antonio Solana-Altabella
- Servicio de Farmacia Área del Medicamento, Hospital Universitari i Politècnic La Fe Av. Valencia, Spain.,Grupo de Investigación en Hematología y Hemoterapia, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain
| | | | - Octavio Ballesta-López
- Servicio de Farmacia Área del Medicamento, Hospital Universitari i Politècnic La Fe Av. Valencia, Spain.,Grupo de Investigación en Hematología y Hemoterapia, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain
| | - David Martínez-Cuadrón
- Grupo de Investigación en Hematología y Hemoterapia, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain.,Servicio de Hematología y Hemoterapia Hospital Universitari i Politècnic La Fe. Valencia Spain
| | - Pau Montesinos
- Grupo de Investigación en Hematología y Hemoterapia, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain.,Servicio de Hematología y Hemoterapia Hospital Universitari i Politècnic La Fe. Valencia Spain
| |
Collapse
|
24
|
Chen Y, Wang J, Zhang F, Liu P. A perspective of immunotherapy for acute myeloid leukemia: Current advances and challenges. Front Pharmacol 2023; 14:1151032. [PMID: 37153761 PMCID: PMC10154606 DOI: 10.3389/fphar.2023.1151032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 05/10/2023] Open
Abstract
During the last decade, the underlying pathogenic mechanisms of acute myeloid leukemia (AML) have been the subject of extensive study which has considerably increased our understanding of the disease. However, both resistance to chemotherapy and disease relapse remain the principal obstacles to successful treatment. Because of acute and chronic undesirable effects frequently associated with conventional cytotoxic chemotherapy, consolidation chemotherapy is not feasible, especially for elderly patients, which has attracted a growing body of research to attempt to tackle this problem. Immunotherapies for acute myeloid leukemia, including immune checkpoint inhibitors, monoclonal antibodies, dendritic cell (DC) vaccines, together with T-cell therapy based on engineered antigen receptor have been developed recently. Our review presents the recent progress in immunotherapy for the treatment of AML and discusses effective therapies that have the most potential and major challenges.
Collapse
Affiliation(s)
- Ying Chen
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Guiyang, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Guiyang, China
- *Correspondence: Jishi Wang,
| | - Fengqi Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Guiyang, China
| | - Ping Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Guiyang, China
| |
Collapse
|
25
|
Tecik M, Adan A. Therapeutic Targeting of FLT3 in Acute Myeloid Leukemia: Current Status and Novel Approaches. Onco Targets Ther 2022; 15:1449-1478. [PMID: 36474506 PMCID: PMC9719701 DOI: 10.2147/ott.s384293] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/19/2022] [Indexed: 08/13/2023] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) is mutated in approximately 30% of acute myeloid leukemia (AML) patients. The presence of FLT3-ITD (internal tandem duplication, 20-25%) mutation and, to a lesser extent, FLT3-TKD (tyrosine kinase domain, 5-10%) mutation is associated with poorer diagnosis and therapy response since the leukemic cells become hyperproliferative and resistant to apoptosis after continuous activation of FLT3 signaling. Targeting FLT3 has been the focus of many pre-clinical and clinical studies. Hence, many small-molecule FLT3 inhibitors (FLT3is) have been developed, some of which are approved such as midostaurin and gilteritinib to be used in different clinical settings, either in combination with chemotherapy or alone. However, many questions regarding the best treatment strategy remain to be answered. On the other hand, various FLT3-dependent and -independent resistance mechanisms could be evolved during FLT3i therapy which limit their clinical impact. Therefore, identifying molecular mechanisms of resistance and developing novel strategies to overcome this obstacle is a current interest in the field. In this review, recent studies of approved FLT3i and knowledge about major resistance mechanisms of clinically approved FLT3i's will be discussed together with novel treatment approaches such as designing novel FLT3i and dual FLT3i and combination strategies including approved FLT3i plus small-molecule agents targeting altered molecules in the resistant cells to abrogate resistance. Moreover, how to choose an appropriate FLT3i for the patients will be summarized based on what is currently known from available clinical data. In addition, strategies beyond FLT3i's including immunotherapeutics, small-molecule FLT3 degraders, and flavonoids will be summarized to highlight potential alternatives in FLT3-mutated AML therapy.
Collapse
Affiliation(s)
- Melisa Tecik
- Bioengineering Program, Graduate School of Engineering and Science, Abdullah Gul University, Kayseri, Turkey
| | - Aysun Adan
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, Turkey
| |
Collapse
|
26
|
Park HJ, Gregory MA, Zaberezhnyy V, Goodspeed A, Jordan CT, Kieft JS, DeGregori J. Therapeutic resistance in acute myeloid leukemia cells is mediated by a novel ATM/mTOR pathway regulating oxidative phosphorylation. eLife 2022; 11:e79940. [PMID: 36259537 PMCID: PMC9645811 DOI: 10.7554/elife.79940] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
While leukemic cells are susceptible to various therapeutic insults, residence in the bone marrow microenvironment typically confers protection from a wide range of drugs. Thus, understanding the unique molecular changes elicited by the marrow is of critical importance toward improving therapeutic outcomes. In this study, we demonstrate that aberrant activation of oxidative phosphorylation serves to induce therapeutic resistance in FLT3 mutant human AML cells challenged with FLT3 inhibitor drugs. Importantly, our findings show that AML cells are protected from apoptosis following FLT3 inhibition due to marrow-mediated activation of ATM, which in turn upregulates oxidative phosphorylation via mTOR signaling. mTOR is required for the bone marrow stroma-dependent maintenance of protein translation, with selective polysome enrichment of oxidative phosphorylation transcripts, despite FLT3 inhibition. To investigate the therapeutic significance of this finding, we tested the mTOR inhibitor everolimus in combination with the FLT3 inhibitor quizartinib in primary human AML xenograft models. While marrow resident AML cells were highly resistant to quizartinib alone, the addition of everolimus induced profound reduction in tumor burden and prevented relapse. Taken together, these data provide a novel mechanistic understanding of marrow-based therapeutic resistance and a promising strategy for improved treatment of FLT3 mutant AML patients.
Collapse
Affiliation(s)
- Hae J Park
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
- Medical Scientist Training Program, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Mark A Gregory
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Vadym Zaberezhnyy
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Andrew Goodspeed
- Department of Pharmacology, University of Colorado Anschutz Medical CampusAuroraUnited States
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Craig T Jordan
- Department of Medicine, Section of Hematology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Jeffrey S Kieft
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Medicine, Section of Hematology, University of Colorado Anschutz Medical CampusAuroraUnited States
| |
Collapse
|
27
|
Acharya B, Saha D, Armstrong D, Lakkaniga NR, Frett B. FLT3 inhibitors for acute myeloid leukemia: successes, defeats, and emerging paradigms. RSC Med Chem 2022; 13:798-816. [PMID: 35923716 PMCID: PMC9298189 DOI: 10.1039/d2md00067a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/21/2022] [Indexed: 09/10/2023] Open
Abstract
FLT3 mutations are one of the most common genetic aberrations found in nearly 30% of acute myeloid leukemias (AML). The mutations are associated with poor prognosis despite advances in the understanding of the biological mechanisms of AML. Numerous small molecule FLT3 inhibitors have been developed in an effort to combat AML. Even with the development of these inhibitors, the five-year overall survival for newly diagnosed AML is less than 30%. In 2017, midostaurin received FDA approval to treat AML, which was the first approved FLT3 inhibitor in the U.S. and Europe. Following, gilteritinib received FDA approval in 2018 and in 2019 quizartinib received approval in Japan. This review parallels these clinical success stories along with other pre-clinical and clinical investigations of FLT3 inhibitors.
Collapse
Affiliation(s)
- Baku Acharya
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Debasmita Saha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Daniel Armstrong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad Jharkhand 826004 India
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| |
Collapse
|
28
|
Knight TE, Edwards H, Meshinchi S, Taub JW, Ge Y. "FLipping" the Story: FLT3-Mutated Acute Myeloid Leukemia and the Evolving Role of FLT3 Inhibitors. Cancers (Basel) 2022; 14:3398. [PMID: 35884458 PMCID: PMC9315611 DOI: 10.3390/cancers14143398] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 12/19/2022] Open
Abstract
The treatment of many types of cancers, including acute myeloid leukemia (AML), has been revolutionized by the development of therapeutics targeted at crucial molecular drivers of oncogenesis. In contrast to broad, relatively indiscriminate conventional chemotherapy, these targeted agents precisely disrupt key pathways within cancer cells. FMS-like tyrosine kinase 3 (FLT3)-encoding a critical regulator of hematopoiesis-is the most frequently mutated gene in patients with AML, and these mutations herald reduced survival and increased relapse in these patients. Approximately 30% of newly diagnosed AML carries an FLT3 mutation; of these, approximately three-quarters are internal tandem duplication (ITD) mutations, and the remainder are tyrosine kinase domain (TKD) mutations. In contrast to its usual, tightly controlled expression, FLT3-ITD mutants allow constitutive, "run-away" activation of a large number of key downstream pathways which promote cellular proliferation and survival. Targeted inhibition of FLT3 is, therefore, a promising therapeutic avenue. In April 2017, midostaurin became both the first FLT3 inhibitor and the first targeted therapy of any kind in AML to be approved by the US FDA. The use of FLT3 inhibitors has continued to grow as clinical trials continue to demonstrate the efficacy of this class of agents, with an expanding number available for use as both experimental standard-of-care usage. This review examines the biology of FLT3 and its downstream pathways, the mechanism of FLT3 inhibition, the development of the FLT3 inhibitors as a class and uses of the agents currently available clinically, and the mechanisms by which resistance to FLT3 inhibition may both develop and be overcome.
Collapse
Affiliation(s)
- Tristan E. Knight
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA 98105, USA;
- Division of Hematology and Oncology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48202, USA; (H.E.); (Y.G.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Soheil Meshinchi
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA 98105, USA;
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jeffrey W. Taub
- Division of Hematology/Oncology, Children’s Hospital of Michigan, Detroit, MI 48201, USA;
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Department of Pediatrics, Central Michigan University, Mt. Pleasant, MI 48859, USA
| | - Yubin Ge
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48202, USA; (H.E.); (Y.G.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
29
|
Ishii H, Yano S. New Therapeutic Strategies for Adult Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:2806. [PMID: 35681786 PMCID: PMC9179253 DOI: 10.3390/cancers14112806] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 12/19/2022] Open
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous hematological malignancy. Chromosomal and genetic analyses are important for the diagnosis and prognosis of AML. Some patients experience relapse or have refractory disease, despite conventional cytotoxic chemotherapies and allogeneic transplantation, and a variety of new agents and treatment strategies have emerged. After over 20 years during which no new drugs became available for the treatment of AML, the CD33-targeting antibody-drug conjugate gemtuzumab ozogamicin was developed. This is currently used in combination with standard chemotherapy or as a single agent. CPX-351, a liposomal formulation containing daunorubicin and cytarabine, has become one of the standard treatments for secondary AML in the elderly. FMS-like tyrosine kinase 3 (FLT3) inhibitors and isocitrate dehydrogenase 1/2 (IDH 1/2) inhibitors are mainly used for AML patients with actionable mutations. In addition to hypomethylating agents and venetoclax, a B-cell lymphoma-2 inhibitor is used in frail patients with newly diagnosed AML. Recently, tumor protein p53 inhibitors, cyclin-dependent kinase inhibitors, and NEDD8 E1-activating enzyme inhibitors have been gaining attention, and a suitable strategy for the use of these drugs is required. Antibody drugs targeting cell-surface markers and immunotherapies, such as antibody-drug conjugates and chimeric antigen receptor T-cell therapy, have also been developed for AML.
Collapse
Affiliation(s)
| | - Shingo Yano
- Division of Clinical Oncology & Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 1058461, Japan;
| |
Collapse
|
30
|
FLT3-targeted treatment for acute myeloid leukemia. Int J Hematol 2022; 116:351-363. [DOI: 10.1007/s12185-022-03374-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 12/17/2022]
|
31
|
Senapati J, Kadia TM. Which FLT3 Inhibitor for Treatment of AML? Curr Treat Options Oncol 2022; 23:359-380. [PMID: 35258791 DOI: 10.1007/s11864-022-00952-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2022] [Indexed: 12/17/2022]
Abstract
OPINION STATEMENT Treatment options in acute myeloid leukemia (AML) have improved significantly over the last decade with better understanding of disease biology and availability of a multitude of targeted therapies. The use of FLT3 inhibitors (FLT3i) in FLT3-mutated (FLT3mut) AML is one such development; however, the clinical decisions that govern their use and dictate the choice of the FLT3i are evolving. Midostaurin and gilteritinib are FDA-approved in specific situations; however, available data from clinical trials also shed light on the utility of sorafenib maintenance post-allogeneic stem cell transplantation (allo-SCT) and quizartinib as part of combination therapy in FLT3mut AML. The knowledge of the patient's concurrent myeloid mutations, type of FLT3 mutation, prior FLT3i use, and eligibility for allo-SCT helps to refine the choice of FLT3i. Data from ongoing studies will further precisely define their use and help in making more informed choices. Despite improvements in FLT3i therapy, the definitive aim is to enable the eligible patient with FLT3mut AML (esp. ITD) to proceed to allo-SCT with regimens containing FLT3i incorporated prior to SCT and as maintenance after SCT.
Collapse
Affiliation(s)
- Jayastu Senapati
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd. - Unit 428, Houston, 77030, USA
| | - Tapan Mahendra Kadia
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd. - Unit 428, Houston, 77030, USA.
| |
Collapse
|
32
|
Suga M, Fukushima K, Ueda T, Arai Y, Nakagawa S, Minami Y, Toda J, Hino A, Fujita J, Yokota T, Hosen N. Clinical implications of combination therapy with quizartinib and craniospinal irradiation for refractory acute myeloid leukemia positive for FMS-like tyrosine kinase 3-internal tandem duplication with central nervous system involvement. Clin Case Rep 2022; 10:e05384. [PMID: 35140970 PMCID: PMC8815089 DOI: 10.1002/ccr3.5384] [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: 10/21/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/10/2022] Open
Abstract
FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) mutation-positive acute myeloid leukemia (AML) has a poor prognosis. We report the first case of successful bridge therapy of novel FLT3 inhibitor, quizartinib, to umbilical cord blood stem cell transplantation for FLT3-ITD-positive AML-primary induction failure patients with central nervous system involvement.
Collapse
Affiliation(s)
- Makiko Suga
- Department of Hematology and OncologyOsaka University Graduate School of MedicineSuitaJapan
| | - Kentaro Fukushima
- Department of Hematology and OncologyOsaka University Graduate School of MedicineSuitaJapan
| | - Tomoaki Ueda
- Department of Hematology and OncologyOsaka University Graduate School of MedicineSuitaJapan
| | - Yasuyuki Arai
- Department of Hematology and OncologyKyoto University HospitalKyotoJapan
| | - Shunsaku Nakagawa
- Department of Clinical Pharmacology and TherapeuticsKyoto University HospitalKyotoJapan
| | - Yosuke Minami
- Department of HematologyNational Cancer Center Hospital EastKashiwaJapan
| | - Jun Toda
- Department of Hematology and OncologyOsaka University Graduate School of MedicineSuitaJapan
| | - Akihisa Hino
- Department of Hematology and OncologyOsaka University Graduate School of MedicineSuitaJapan
| | - Jiro Fujita
- Department of Hematology and OncologyOsaka University Graduate School of MedicineSuitaJapan
| | - Takafumi Yokota
- Department of Hematology and OncologyOsaka University Graduate School of MedicineSuitaJapan
| | - Naoki Hosen
- Department of Hematology and OncologyOsaka University Graduate School of MedicineSuitaJapan
- Laboratory of Cellular Immunotherapy, World Premier Interenational Immunology Frontier Research CenterOsaka UniversitySuitaJapan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI)Osaka UniversitySuitaJapan
| |
Collapse
|
33
|
Solana-Altabella A, Ballesta-López O, Megías-Vericat JE, Martínez-Cuadrón D, Montesinos P. Emerging FLT3 inhibitors for the treatment of acute myeloid leukemia. Expert Opin Emerg Drugs 2022; 27:1-18. [DOI: 10.1080/14728214.2021.2009800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Antonio Solana-Altabella
- Servicio de Farmacia, Área del Medicamento. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia– Spain
- Instituto de Investigación Sanitaria La Fe (IISLAFE). Av. Fernando Abril Martorell, Valencia–Spain
| | - Octavio Ballesta-López
- Servicio de Farmacia, Área del Medicamento. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia– Spain
- Instituto de Investigación Sanitaria La Fe (IISLAFE). Av. Fernando Abril Martorell, Valencia–Spain
| | - Juan Eduardo Megías-Vericat
- Servicio de Farmacia, Área del Medicamento. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia– Spain
| | - David Martínez-Cuadrón
- Servicio de Farmacia, Área del Medicamento. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia– Spain
- Instituto de Investigación Sanitaria La Fe (IISLAFE). Av. Fernando Abril Martorell, Valencia–Spain
| | - Pau Montesinos
- Instituto de Investigación Sanitaria La Fe (IISLAFE). Av. Fernando Abril Martorell, Valencia–Spain
- Servicio de Hematología y Hemoterapia. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia-Spain
| |
Collapse
|
34
|
Dupont M, Huart M, Lauvinerie C, Bidet A, Guitart AV, Villacreces A, Vigon I, Desplat V, El Habhab A, Pigneux A, Ivanovic Z, Brunet De la Grange P, Dumas PY, Pasquet JM. Autophagy Targeting and Hematological Mobilization in FLT3-ITD Acute Myeloid Leukemia Decrease Repopulating Capacity and Relapse by Inducing Apoptosis of Committed Leukemic Cells. Cancers (Basel) 2022; 14:cancers14020453. [PMID: 35053612 PMCID: PMC8796021 DOI: 10.3390/cancers14020453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/19/2022] Open
Abstract
Targeting FLT3-ITD in AML using TKI against FLT3 cannot prevent relapse even in the presence of complete remission, suggesting the resistance and/or the persistence of leukemic-initiating cells in the hematopoietic niche. By mimicking the hematopoietic niche condition with cultures at low oxygen concentrations, we demonstrate in vitro that FLT3-ITD AML cells decrease their repopulating capacity when Vps34 is inhibited. Ex vivo, AML FLT3-ITD blasts treated with Vps34 inhibitors recovered proliferation more slowly due to an increase an apoptosis. In vivo, mice engrafted with FLT3-ITD AML MV4-11 cells have the invasion of the bone marrow and blood in 2 weeks. After 4 weeks of FLT3 TKI treatment with gilteritinib, the leukemic burden had strongly decreased and deep remission was observed. When treatment was discontinued, mice relapsed rapidly. In contrast, Vps34 inhibition strongly decreased the relapse rate, and even more so in association with mobilization by G-CSF and AMD3100. These results demonstrate that remission offers the therapeutic window for a regimen using Vps34 inhibition combined with mobilization to target persistent leukemic stem cells and thus decrease the relapse rate.
Collapse
Affiliation(s)
- Marine Dupont
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
| | - Mathilde Huart
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
| | - Claire Lauvinerie
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
| | - Audrey Bidet
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
- Service d’Hématologie Biologique, CHU Bordeaux, 33000 Bordeaux, France
| | - Amélie Valérie Guitart
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
| | - Arnaud Villacreces
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
| | - Isabelle Vigon
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
| | - Vanessa Desplat
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
| | - Ali El Habhab
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
| | - Arnaud Pigneux
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
- Service d’Hématologie Clinique et Thérapie Cellulaire, CHU Bordeaux, 33000 Bordeaux, France
| | - Zoran Ivanovic
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
- Etablissement Français du Sang Nouvelle Aquitaine, 33035 Bordeaux, France
| | - Philippe Brunet De la Grange
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
- Etablissement Français du Sang Nouvelle Aquitaine, 33035 Bordeaux, France
| | - Pierre-Yves Dumas
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
- Service d’Hématologie Clinique et Thérapie Cellulaire, CHU Bordeaux, 33000 Bordeaux, France
| | - Jean-Max Pasquet
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1312 BRIC, Université de Bordeaux, Bat TP 4e étage, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.D.); (M.H.); (C.L.); (A.B.); (A.V.G.); (A.V.); (I.V.); (V.D.); (A.E.H.); (A.P.); (Z.I.); (P.B.D.l.G.); (P.-Y.D.)
- Correspondence: ; Tel.: +33-07-85-42-59-25
| |
Collapse
|
35
|
Bregante J, Schönbichler A, Pölöske D, Degenfeld-Schonburg L, Monzó Contreras G, Hadzijusufovic E, de Araujo ED, Valent P, Moriggl R, Orlova A. Efficacy and Synergy of Small Molecule Inhibitors Targeting FLT3-ITD + Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:6181. [PMID: 34944800 PMCID: PMC8699584 DOI: 10.3390/cancers13246181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/22/2022] Open
Abstract
Constitutive activation of FLT3 by ITD mutations is one of the most common genetic aberrations in AML, present in ~1/3 of cases. Patients harboring FLT3-ITD display worse clinical outcomes. The integration and advancement of FLT3 TKI in AML treatment provided significant therapeutic improvement. However, due to the emergence of resistance mechanisms, FLT3-ITD+ AML remains a clinical challenge. We performed an unbiased drug screen to identify 18 compounds as particularly efficacious against FLT3-ITD+ AML. Among these, we characterized two investigational compounds, WS6 and ispinesib, and two approved drugs, ponatinib and cabozantinib, in depth. We found that WS6, although not yet investigated in oncology, shows a similar mechanism and potency as ponatinib and cabozantinib. Interestingly, ispinesib and cabozantinib prevent activation of AXL, a key driver and mechanism of drug resistance in FLT3-ITD+ AML patients. We further investigated synergies between the selected compounds and found that combination treatment with ispinesib and cabozantinib or ponatinib shows high synergy in FLT3-ITD+ AML cell lines and patient samples. Together, we suggest WS6, ispinesib, ponatinib and cabozantinib as novel options for targeting FLT3-ITD+ AML. Whether combinatorial tyrosine kinase and kinesin spindle blockade is effective in eradicating neoplastic (stem) cells in FLT3-ITD+ AML remains to be determined in clinical trials.
Collapse
Affiliation(s)
- Javier Bregante
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (J.B.); (A.S.); (D.P.); (G.M.C.); (R.M.)
| | - Anna Schönbichler
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (J.B.); (A.S.); (D.P.); (G.M.C.); (R.M.)
| | - Daniel Pölöske
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (J.B.); (A.S.); (D.P.); (G.M.C.); (R.M.)
| | - Lina Degenfeld-Schonburg
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.); (E.H.); (P.V.)
| | - Garazi Monzó Contreras
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (J.B.); (A.S.); (D.P.); (G.M.C.); (R.M.)
| | - Emir Hadzijusufovic
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.); (E.H.); (P.V.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
- Clinic for Companion Animals and Horses, University Clinic for Small Animals, Internal Medicine Small Animals, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Elvin D. de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada;
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada
| | - Peter Valent
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.); (E.H.); (P.V.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (J.B.); (A.S.); (D.P.); (G.M.C.); (R.M.)
| | - Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (J.B.); (A.S.); (D.P.); (G.M.C.); (R.M.)
| |
Collapse
|
36
|
Beyer M, Henninger SJ, Haehnel PS, Mustafa AHM, Gurdal E, Schubert B, Christmann M, Sellmer A, Mahboobi S, Drube S, Sippl W, Kindler T, Krämer OH. Identification of a highly efficient dual type I/II FMS-like tyrosine kinase inhibitor that disrupts the growth of leukemic cells. Cell Chem Biol 2021; 29:398-411.e4. [PMID: 34762849 DOI: 10.1016/j.chembiol.2021.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/17/2021] [Accepted: 10/21/2021] [Indexed: 12/20/2022]
Abstract
Internal tandem duplications (ITDs) in the FMS-like tyrosine kinase-3 (FLT3) are causally linked to acute myeloid leukemia (AML) with poor prognosis. Available FLT3 inhibitors (FLT3i) preferentially target inactive or active conformations of FLT3. Moreover, they co-target kinases for normal hematopoiesis, are vulnerable to therapy-associated tyrosine kinase domain (TKD) FLT3 mutants, or lack low nanomolar activity. We show that the tyrosine kinase inhibitor marbotinib suppresses the phosphorylation of FLT3-ITD and the growth of permanent and primary AML cells with FLT3-ITD. This also applies to leukemic cells carrying FLT3-ITD/TKD mutants that confer resistance to clinically used FLT3i. Marbotinib shows high selectivity for FLT3 and alters signaling, reminiscent of genetic elimination of FLT3-ITD. Molecular docking shows that marbotinib fits in opposite orientations into inactive and active conformations of FLT3. The water-soluble marbotinib-carbamate significantly prolongs survival of mice with FLT3-driven leukemia. Marbotinib is a nanomolar next-generation FLT3i that represents a hybrid inhibitory principle.
Collapse
Affiliation(s)
- Mandy Beyer
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany
| | - Sven J Henninger
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany
| | - Patricia S Haehnel
- Department of Hematology, Medical Oncology, and Pneumology, University Medical Center, 55131 Mainz, Germany; University Cancer Center, University Medical Center, Mainz, Germany; German Consortia for Translational Cancer Research, 55131 Mainz, Germany
| | - Al-Hassan M Mustafa
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | - Ece Gurdal
- Institute for Pharmacy, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Strasse 3, 06120 Halle (Saale), Germany; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, Atasehir, Istanbul 34755, Turkey
| | - Bastian Schubert
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany
| | - Markus Christmann
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany
| | - Andreas Sellmer
- Institute of Pharmacy, Department of Pharmaceutical/Medicinal Chemistry I, University of Regensburg, 93053 Regensburg, Germany
| | - Siavosh Mahboobi
- Institute of Pharmacy, Department of Pharmaceutical/Medicinal Chemistry I, University of Regensburg, 93053 Regensburg, Germany
| | - Sebastian Drube
- Institute of Immunology, Jena University Hospital, 07743 Jena, Germany
| | - Wolfgang Sippl
- Institute for Pharmacy, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Strasse 3, 06120 Halle (Saale), Germany
| | - Thomas Kindler
- Department of Hematology, Medical Oncology, and Pneumology, University Medical Center, 55131 Mainz, Germany; University Cancer Center, University Medical Center, Mainz, Germany; German Consortia for Translational Cancer Research, 55131 Mainz, Germany
| | - Oliver H Krämer
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany.
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
Kala J, Salman LA, Geara AS, Izzedine H. Nephrotoxicity From Molecularly Targeted Chemotherapeutic Agents. Adv Chronic Kidney Dis 2021; 28:415-428.e1. [PMID: 35190108 DOI: 10.1053/j.ackd.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/16/2021] [Accepted: 09/01/2021] [Indexed: 11/11/2022]
Abstract
The introduction of novel molecularly targeted therapies in the last 2 decades has significantly improved the patient survival compared to standard conventional chemotherapies. However, this improvement has been accompanied by a whole new spectrum of kidney adverse events. Although known as "targeted," many of these agents lack specificity and selectivity, and they have a tendency to inhibit multiple targets including those in the kidneys. Early detection and correct management of kidney toxicities is crucial to preserve kidney functions. The knowledge of these toxicities helps guide optimal and continued utilization of these potent therapies. The incidence, severity, and pattern of nephrotoxicity may vary depending on the respective target of the drug. Here, we review the mechanism of action, clinical findings of kidney adverse events, and their proposed management strategies.
Collapse
|
40
|
Kantarjian HM, Short NJ, Fathi AT, Marcucci G, Ravandi F, Tallman M, Wang ES, Wei AH. Acute Myeloid Leukemia: Historical Perspective and Progress in Research and Therapy Over 5 Decades. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2021; 21:580-597. [PMID: 34176779 PMCID: PMC11938811 DOI: 10.1016/j.clml.2021.05.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/17/2022]
Abstract
With the Food and Drug Administration approval of 9 agents for different acute myeloid leukemia (AML) indications, the prognosis and management of AML is evolving rapidly. Herein, we review the important milestones in the history of AML research and therapy, discuss insights regarding prognostic assessment and prediction of treatment outcome, detail practical supportive care measures, and summarize the current treatment landscape and areas of evolving research.
Collapse
Affiliation(s)
| | - Nicholas J Short
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Amir T Fathi
- Leukemia Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research City of Hope, Duarte, CA, USA
| | - Farhad Ravandi
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Martin Tallman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Eunice S Wang
- Leukemia Service, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Andrew H Wei
- Department of Clinical Hematology, The Alfred Hospital and Monash University, Melbourne, Australia
| |
Collapse
|
41
|
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: 15] [Impact Index Per Article: 3.8] [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
|
42
|
Oliva EN, Ronnebaum SM, Zaidi O, Patel DA, Nehme SA, Chen C, Almeida AM. A systematic literature review of disease burden and clinical efficacy for patients with relapsed or refractory acute myeloid leukemia. AMERICAN JOURNAL OF BLOOD RESEARCH 2021; 11:325-360. [PMID: 34540343 PMCID: PMC8446831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
Acute myeloid leukemia (AML) is a rapidly progressive hematological malignancy that is difficult to cure. The prognosis is poor and treatment options are limited in case of relapse. A comprehensive assessment of current disease burden and the clinical efficacy of non-intensive therapies in this population are lacking. We conducted two systematic literature reviews (SLRs). The first SLR (disease burden) included observational studies reporting the incidence and economic and humanistic burden of relapsed/refractory (RR) AML. The second SLR (clinical efficacy) included clinical trials (phase II or later) reporting remission rates (complete remission [CR] or CR with incomplete hematologic recovery [CRi]) and median overall survival (mOS) in patients with RR AML or patients with de novo AML who are ineligible for intensive chemotherapy. For both SLRs, MEDLINE®/Embase® were searched from January 1, 2008 to January 31, 2020. Clinical trial registries were also searched for the clinical efficacy SLR. After screening, two independent reviewers determined the eligibility for inclusion in the SLRs based on full-text articles. The disease burden SLR identified 130 observational studies. The median cumulative incidence of relapse was 29.4% after stem cell transplant and 46.8% after induction chemotherapy. Total per-patient-per-month costs were $28,148-$29,322; costs and health care resource use were typically higher for RR versus non-RR patients. Patients with RR AML had worse health-related quality of life (HRQoL) scores than patients with de novo AML across multiple instruments, and lower health utility values versus other AML health states (i.e. newly diagnosed, remission, consolidation, and maintenance therapy). The clinical efficacy SLR identified 50 trials (66 total trial arms). CR/CRi rates and mOS have remained relatively stable and low over the last 2 decades. Across all arms, the median rate of CR/CRi was 18.3% and mOS was 6.2 months. In conclusion, a substantial proportion of patients with AML will develop RR AML, which is associated with significant humanistic and economic burden. Existing treatments offer limited efficacy, highlighting the need for more effective non-intensive treatment options.
Collapse
|
43
|
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
|
44
|
Gutjahr JC, Bayer E, Yu X, Laufer JM, Höpner JP, Tesanovic S, Härzschel A, Auer G, Rieß T, Salmhofer A, Szenes E, Haslauer T, Durand-Onayli V, Ramspacher A, Pennisi SP, Artinger M, Zaborsky N, Chigaev A, Aberger F, Neureiter D, Pleyer L, Legler DF, Orian-Rousseau V, Greil R, Hartmann TN. CD44 engagement enhances acute myeloid leukemia cell adhesion to the bone marrow microenvironment by increasing VLA-4 avidity. Haematologica 2021; 106:2102-2113. [PMID: 32616529 PMCID: PMC8327716 DOI: 10.3324/haematol.2019.231944] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Indexed: 12/11/2022] Open
Abstract
Adhesive properties of leukemia cells shape the degree of organ infiltration and the extent of leukocytosis. CD44 and the integrin VLA-4, a CD49d/CD29 heterodimer, are important factors in progenitor cell adhesion in bone marrow. Here, we report their cooperation in acute myeloid leukemia (AML) by a novel non-classical CD44-mediated way of inside-out VLA-4 activation. In primary AML bone marrow samples from patients and the OCI-AML3 cell line, CD44 engagement by hyaluronan induced inside-out activation of VLA-4 resulting in enhanced leukemia cell adhesion on VCAM-1. This was independent of VLA-4 affinity regulation but based on ligand-induced integrin clustering on the cell surface. CD44-induced VLA-4 activation could be inhibited by the Src family kinase inhibitor PP2 and the multikinase inhibitor midostaurin. As a further consequence, the increased adhesion on VCAM-1 allowed AML cells to bind stromal cells strongly. Thereby, the VLA-4/VCAM-1 interaction promoted activation of Akt, MAPK, NF-kB and mTOR signaling and decreased AML cell apoptosis. Collectively, our investigations provide a mechanistic description of an unusual CD44 function in regulating VLA-4 avidity in AML, enhancing AML cell retention in the supportive bone marrow microenvironment.
Collapse
Affiliation(s)
- Julia C Gutjahr
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | - Elisabeth Bayer
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | - Xiaobing Yu
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics
| | - Julia M Laufer
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz
| | - Jan P Höpner
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | | | - Andrea Härzschel
- Department of Internal Medicine I, Medical Center and Faculty of Medicine, University of Freiburg
| | - Georg Auer
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | - Tanja Rieß
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | - Astrid Salmhofer
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | - Eva Szenes
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | - Theresa Haslauer
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | - Valerie Durand-Onayli
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | | | - Sandra P Pennisi
- Department of Internal Medicine I, Medical Center and Faculty of Medicine, University of Freiburg
| | - Marc Artinger
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | - Nadja Zaborsky
- 1Laboratory for Immunological and Molecular Cancer Research
| | | | - Fritz Aberger
- Department Biosciences, Paris-Lodron University of Salzburg
| | | | - Lisa Pleyer
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz
| | | | - Richard Greil
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| | - Tanja N Hartmann
- 3rd Medical Department, SCRI-LIMCR, Paracelsus Medical University, Cancer Cluster Salzburg
| |
Collapse
|
45
|
Sweet K, Bhatnagar B, Döhner H, Donnellan W, Frankfurt O, Heuser M, Kota V, Liu H, Raffoux E, Roboz GJ, Röllig C, Showel MM, Strickland SA, Vives S, Tang S, Unger TJ, Joshi A, Shen Y, Alvarez MJ, Califano A, Crochiere M, Landesman Y, Kauffman M, Shah J, Shacham S, Savona MR, Montesinos P. A 2:1 randomized, open-label, phase II study of selinexor vs. physician's choice in older patients with relapsed or refractory acute myeloid leukemia. Leuk Lymphoma 2021; 62:3192-3203. [PMID: 34323164 DOI: 10.1080/10428194.2021.1950706] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Selinexor, a selective inhibitor of nuclear export, has demonstrated promising activity in patients with acute myeloid leukemia (AML). This randomized, phase II study evaluated selinexor 60 mg twice weekly (n = 118) vs. physician's choice (PC) treatment (n = 57) in patients aged ≥60 years with relapsed/refractory (R/R) AML. The primary outcome was overall survival (OS). Median OS did not differ significantly for selinexor vs. PC (3.2 vs. 5.6 months; HR = 1.18 [95% CI: 0.79-1.75]; p = 0.422). Complete remission (CR) plus CR with incomplete hematologic recovery trending in favor of selinexor occurred in a minority of patients. Selinexor treated patients had an increased incidence of adverse events. The most common grade ≥3 adverse events were thrombocytopenia, febrile neutropenia, anemia, hyponatremia. Despite well-balanced baseline characteristics, there were numerically higher rates of TP53 mutations, prior myelodysplastic syndrome, and lower absolute neutrophil counts in the selinexor group; warranting further investigation of selinexor in more carefully stratified R/R AML patients.Registered trial: NCT02088541.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Susana Vives
- ICO Badalona-Hospital Germans Trias i Pujol, Badalona, Spain
| | | | | | | | - Yao Shen
- DarwinHealth Inc, New York, NY, USA
| | - Mariano J Alvarez
- DarwinHealth Inc, New York, NY, USA.,Columbia University, New York, NY, USA
| | | | | | | | | | - Jatin Shah
- Karyopharm Therapeutics, Newton, MA, USA
| | | | | | - Pau Montesinos
- Departamento de Hematologia, Hospital Universitario y Politécnico La Fe, Valencia, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
46
|
Hogan FL, Williams V, Knapper S. FLT3 Inhibition in Acute Myeloid Leukaemia - Current Knowledge and Future Prospects. Curr Cancer Drug Targets 2021; 20:513-531. [PMID: 32418523 DOI: 10.2174/1570163817666200518075820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/12/2020] [Accepted: 03/29/2020] [Indexed: 12/20/2022]
Abstract
Activating mutations of FMS-like tyrosine kinase 3 (FLT3) are present in 30% of acute myeloid leukaemia (AML) patients at diagnosis and confer an adverse clinical prognosis. Mutated FLT3 has emerged as a viable therapeutic target and a number of FLT3-directed tyrosine kinase inhibitors have progressed through clinical development over the last 10-15 years. The last two years have seen United States Food and Drug Administration (US FDA) approvals of the multi-kinase inhibitor midostaurin for newly-diagnosed FLT3-mutated patients, when used in combination with intensive chemotherapy, and of the more FLT3-selective agent gilteritinib, used as monotherapy, for patients with relapsed or treatment-refractory FLT3-mutated AML. The 'second generation' agents, quizartinib and crenolanib, are also at advanced stages of clinical development. Significant challenges remain in negotiating a variety of potential acquired drug resistance mechanisms and in optimizing sequencing of FLT3 inhibitory drugs with existing and novel treatment approaches in different clinical settings, including frontline therapy, relapsed/refractory disease, and maintenance treatment. In this review, the biology of FLT3, the clinical challenge posed by FLT3-mutated AML, the developmental history of the key FLT3-inhibitory compounds, mechanisms of disease resistance, and the future outlook for this group of agents, including current and planned clinical trials, is discussed.
Collapse
Affiliation(s)
- Francesca L Hogan
- Department of Haematology, University Hospital of Wales, Cardiff, United Kingdom
| | - Victoria Williams
- Department of Haematology, University Hospital of Wales, Cardiff, United Kingdom
| | - Steven Knapper
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
47
|
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: 3] [Impact Index Per Article: 0.8] [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
|
48
|
Daver N, Venugopal S, Ravandi F. FLT3 mutated acute myeloid leukemia: 2021 treatment algorithm. Blood Cancer J 2021; 11:104. [PMID: 34045454 PMCID: PMC8159924 DOI: 10.1038/s41408-021-00495-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/22/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022] Open
Abstract
Approximately 30% of patients with newly diagnosed acute myeloid leukemia (AML) harbor mutations in the fms-like tyrosine kinase 3 (FLT3) gene. While the adverse prognostic impact of FLT3-ITDmut in AML has been clearly proven, the prognostic significance of FLT3-TKDmut remains speculative. Current guidelines recommend rapid molecular testing for FLT3mut at diagnosis and earlier incorporation of targeted agents to achieve deeper remissions and early consideration for allogeneic stem cell transplant (ASCT). Mounting evidence suggests that FLT3mut can emerge at any timepoint in the disease spectrum emphasizing the need for repetitive mutational testing not only at diagnosis but also at each relapse. The approval of multi-kinase FLT3 inhibitor (FLT3i) midostaurin with induction therapy for newly diagnosed FLT3mut AML, and a more specific, potent FLT3i, gilteritinib as monotherapy for relapsed/refractory (R/R) FLT3mut AML have improved outcomes in patients with FLT3mut AML. Nevertheless, the short duration of remission with single-agent FLT3i's in R/R FLT3mut AML in the absence of ASCT, limited options in patients refractory to gilteritinib therapy, and diverse primary and secondary mechanisms of resistance to different FLT3i's remain ongoing challenges that compel the development and rapid implementation of multi-agent combinatorial or sequential therapies for FLT3mut AML.
Collapse
Affiliation(s)
- Naval Daver
- Department of Leukemia, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA.
| | - Sangeetha Venugopal
- Department of Leukemia, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
49
|
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: 79] [Impact Index Per Article: 19.8] [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
|
50
|
A phase 1/2 study of the oral FLT3 inhibitor pexidartinib in relapsed/refractory FLT3-ITD-mutant acute myeloid leukemia. Blood Adv 2021; 4:1711-1721. [PMID: 32330242 DOI: 10.1182/bloodadvances.2020001449] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/25/2020] [Indexed: 01/10/2023] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) tyrosine kinase inhibitors (TKIs) have activity in acute myeloid leukemia (AML) patients with FLT3 internal tandem duplication (ITD) mutations, but efficacy is limited by resistance-conferring kinase domain mutations. This phase 1/2 study evaluated the safety, tolerability, and efficacy of the oral FLT3 inhibitor PLX3397 (pexidartinib), which has activity against the FLT3 TKI-resistant F691L gatekeeper mutation in relapsed/refractory FLT3-ITD-mutant AML. Ninety patients were treated: 34 in dose escalation (part 1) and 56 in dose expansion (part 2). Doses of 800 to 5000 mg per day in divided doses were tested. No maximally tolerated dose was reached. Plasma inhibitory assay demonstrated that patients dosed with ≥3000 mg had sufficient levels of active drug in their trough plasma samples to achieve 95% inhibition of FLT3 phosphorylation in an FLT3-ITD AML cell line. Based on a plateau in drug exposure, the 3000-mg dose was chosen as the recommended phase 2 dose. The most frequently reported treatment-emergent adverse events were diarrhea (50%), fatigue (47%), and nausea (46%). Based on modified response criteria, the overall response rate to pexidartinib among all patients was 21%. Twenty-three percent of patients treated at ≥2000 mg responded. The overall composite complete response rate for the study was 11%. Six patients were successfully bridged to transplantation. Median overall survival (OS) of patients treated in dose expansion was 112 days (90% confidence interval [CI], 77-150 days), and median OS of responders with complete remission with or without recovery of blood counts was 265 days (90% CI, 170-422 days). This trial was registered at www.clinicaltrials.gov as #NCT01349049.
Collapse
|