Loading

Journal of Cancer Biology and Research

Tyrosine Kinase Inhibitors in Myeloid Leukemia Therapy - Molecular Mechanisms and Future Challenges

Review Article | Open Access Issue 2373-9436
Article DOI :

  • 1. Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
  • 2. Shrewsbury High School, Shrewsbury, Massachusetts, USA
  • 3. Foxborough High School, Foxborough, Massachusetts, USA
  • 4. #Contributed Equally to the Manuscript
+ Show More - Show Less
Corresponding Authors
Shaoguang Li, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
ABBREVIATIONS

PTKs: Protein Tyrosine Kinases; CML: Chronic Myeloid Leukemia; AML: Acute Myeloid Leukemia; TKI: Tyrosine Kinase Inhibitor; FLT3: FMS-like tyrosine kinase 3; HSC: Hematopoietic Stem Cell; LSC: Leukemia Stem Cell; OS: Overall Survival; EFS: Event-Free Survival; CR: Complete Remission.

INTRODUCTION

Protein tyrosine kinases (PTKs) are key regulators for many developmental signaling pathways and are involved in various biological processes [1,2]. They comprise a family of enzymes that catalyze phosphorylation of specific tyrosine residues on target proteins, subsequently leading to conformational changes and activation (or inhibition) of target proteins. Since the first PTK, v-Src encoded by the Rous sarcoma virus oncogene, was identified in 1978, about 90 PTKs have been identified. These PTKs are primarily classified as either RTKs (RTK) [e.g. epidermal growth factor receptors (EGFR), FMS-like tyrosine kinase 3 (FLT3), AXL] or nonRTKs [such as ABL and SRC]. Given the essential roles of PTKs in regulating cellular survival, growth and differentiation, their activities are tightly controlled under physiological conditions. On the other hand, oncogenic mutations of some PTKs constitutively activate their functions, resulting in abnormal cellular growth and differentiation and malignant cell transformation [1]. Thus, inhibition of kinase activities of these PTKs provides an effective strategy for treating many types of cancers associated with abberant PTK activation. In fact, a number of target-specific tyrosine kinase inhibitors (TKIs) have been developed, and often function as homologs of adenosine triphosphate (ATP) to compete for ATP-binding sites, leading to a blockade of kinase activities of both PTKs and donwstream signaling cascades. Importantly, TKIs exhibit significant advantages over chemotherapy and provide a targeted therapy for various types of cancers, including hematologic malignancies.

Hematologic malignancies are a major group of human cancers and include leukemia, lymphoma, and multiple myeloma. Hematologic malignancies are highly heterogeneous in molecular characteristics, thereby bringing huge challenges in clinical management of these diseases. The development of TKIs has significantly improved disease outcomes. Chronic myeloid leukemia (CML), is a myeloproliferative hematopoietic stem cell (HSC) disease that is believed to be derived from an abnormal HSC harboring the Philadelphia (Ph) chromosome [3-5], which is generated by a reciprocal translocation between chromosome 9 and 22 [t(9; 22)(q34; q11)]. The resulting chimeric BCR-ABL fusion oncogene encodes the BCR-ABL oncoprotein, which has constitutive tyrosine kinase activity [3, 6]. BCR-ABL activates numerous downstream pathways that drive CML development by increasing proliferation and reducing apoptosis of BCRABL-expressing cells [7]. The chimeric BCR-ABL oncoprotein constitutes the molecular basis of CML and provided the foundation for the development of imatinib, one of the most effective TKIs in treating human cancers. In fact, the development of TKIs for CML therapy has served as a paradigm for drug development and ignited an era in developing TKIs in the oncology field. Acute myeloid leukemia (AML) is also a heterogeneous clonal disorder and is characterized by uncontrolled expansion and blocked differentiation of myeloid cells. Accumulation of genetic alterations in hematopoietic stem/progenitor cells causes aberrant activation of a series of signaling pathways, ultimately resulting in AML development. Approximately 55% of adult AML cases have cytogenetic abnormalities, often in form of the recurrent gene fusions such as CBFB-MYH11 [inv (16)], PMLRARα [t(15;17)], and AML1-ETO [t(8;21)], and about 30% AML patients carry somatic mutations of the tyrosine kinase FLT3 [8,9]. TKIs targeting FLT3 have been used successfully to treat AML. Thus, we will further review the recent advances on the development of TKIs with an emphasis mainly on two tyrosine kinases: BCR-ABL and FLT3.

TKIs for treating CML harboring the BCR-ABL oncogene

The development of TKIs for BCR-ABL: The development of TKIs targeting BCR-ABL revolutionized CML treatment and has dramatically improved disease management and outcomes. Arsenic was used to reduce the leukocyte count prior to the discovery of the Ph chromosome, but failed to prolong CML patient survival. Busulfan and hydroxyurea, conventional cytotoxic chemotherapeutic agents, were also used in controlling elevated white blood cell counts, but they failed to significantly improve the survival of CML patients [10]. In the 1980s, interferon-α (IFNα) was introduced in for treating CML, and induced both disease remission and prolonged survival in approximately one-third of CML patients [11,12]. Since the chimeric BCR-ABL is a tyrosine kinase, it is an ideal molecular target for designing therapeutic chemical inhibitors [13,14]. Imatinib mesylate (also known as Gleevec, STI-571), the first TKI against BCR-ABL, was approved by the U.S. Food and Drug Administration (FDA) in 2001 and, by 2003, became the front-line therapy for CML in chronic phase [15-17]. Imatinib, along with three generations of anti-BCR-ABL TKIs, has been used to treat CML and has brought long-term remission and near-normal life expectancy to the majority of CML patients.

Mechanisms of TKIs for functional inhibition of BCR-ABL: Imatinib was discovered in a time-consuming in vitro screening of inhibitors for protein kinase C [18, 19]. It was originally shown to be an inhibitor of platelet-derived growth factor receptor and v-ABL protein tyrosine kinase, and later was identified as an inhibitor of c-ABL, BCR-ABL, and c-Kit [19]. The overall structure of the c-ABL kinase domain consists of two lobes, a smaller amino-terminal lobe primarily involving in anchoring and orienting ATP, and the carboxyl-terminal lobe responsible for binding the peptide substrate. The catalytic site lies in a cleft between the two lobes. These two lobes can open or close the cleft by moving toward each other. The open form allows ATP to access the catalytic site, while the closed form brings residues into the catalytic site. Thus, blocking the transition between these two forms inhibits kinase activity. Imatinib binds the cleft between these two lobes and competes for the ATP binding site of the ABL kinase by forming hydrogen bonds, keeping the kinase in an inactive conformation [20]. In addition to the ABL kinase, imatinib also inhibits the platelet derived growth factor receptor (PDGFR) and c-Kit tyrosine kinase [21]. The International Randomized Study of Interferon and STI571 (IRIS) demonstrates that imatinib treatment (400 mg per day) as the standard of care for CML results in reliability and supremacy of hematologic and cytogenetic responses in more than 90% of chronic phrase CML patients [22]. An 8-year follow-up of STI571 showed that the estimated event-free survival rate was 81%, and the overall survival (OS) rate was 93% when only considering CML-related deaths. Furthermore, imatinib significantly increased the 10-year survival rate from 10-20% to 80–90% [22]. Without any doubt, the development of TKIs for CML treatment ignited a new era for targeted therapy in oncology.

The next-generation TKIs for BCR-ABL: Currently, TKIs are the standard treatment for all newly diagnosed patients with CML, and imatinib is the first-generation TKI. To overcome imatinib resistance in CML patients, second (dasatinib, nilotinib, bosutinib) and third-generation (ponatinib) TKIs were also developed (Figure 1). Dasatinib is a dual ABL and SRC family kinase (SFK) inhibitor, and was approved by the FDA in 2007 [23]. It binds to the active and inactive forms of BCR-ABL with higher affinity than imatinib. Dasatinib (100mg daily) has achieved a higher response rate than imatinib (400mg daily) in newly-diagnosed CML chronic phase patients [24,25]. In addition, dasatinib is effective against most imatinib-resistant ABL mutations in patients. Nilotinib is another second-generation TKI with 20 to 50-fold higher affinity for the inactive conformation of BCR-ABL [26,27]. Nilotinib is superior to imatinib as a frontline treatment, but it is associated with a higher rate of complications and side effects (pruritus, hyperglycemia and pancreatitis) [28,29]. Bosutinib is a specific inhibitor for BCR-ABL and SFKs, and was approved by the FDA in 2012 for second-line treatment of CML due to its effectiveness against most imatinib-resistant mutations (F317L and Y253H).

Ponatinib is a third-generation TKI approved by the FDA in 2012. It was designed to target the first and second-generation TKI-resistant BCR-ABLT315I mutant [30]. Ponatinib acts through carbon-carbon triple bond between the methylphenyl and purine groups, allowing it to bind to the T315I mutant. Therefore, ponatinib inhibits both wild type and a variety of mutated BCRABL proteins (e.g. M244V, G250E, Q252H, Y253F/H, E255K/V, F317L, M351T, and F359V) [31]. Ponatinib treatment with a low dose (45mg) can result in faster and deeper response [32].

There are factors to consider when selecting between the aforementioned five TKIs for CML patients to ensure optimal duration and life-quality. In general, imatinib is a safe and costeffective TKI for CML patients in chronic phase; patients with high-risk disease are treated with second/third-generation TKIs. Other factors, including the distinct benefits and variable side effects of each TKI, and individual patient features, are also taken into account [29, 33].

TKIs for AML treatment

Dysregulation of multiple tyrosine kinases such as FLT3, KIT, Janus kinase 2 (JAK2) are found in AML. While some TKIs show efficacy in clinical trials, others are still in early stages of development. Regardless, novel therapies targeting these tyrosine kinases represent a promising strategy in AML therapy, although chemotherapy with anthracycline plus cytarabine remains as a conventional AML therapy.

FLT3 tyrosine kinase inhibitors: FLT3 is a receptor tyrosine kinase (RTK) expressed in hematopoietic progenitor cells and plays an important role in hematopoietic development [34-36]. Upon stimulation with FLT3 ligand, FLT3 dimerizes and undergoes autophosphorylation, leading to activation of downstream pathways including PI3K/Akt and Ras signaling pathways [37,38]. High FLT3 expression is detected in most cases of AML and B-acute lymphoblastic leukemia [39]. Approximately 30% AML patients have FLT3 mutations, mostly as internal tandem duplication (FLT3-ITD, 20-25%) as well as point mutation in the tyrosine kinase domain (FLT3-TKD, 5-10%). These alterations cause ligand-independent FLT3 activation, resulting in constitutive stimulation of downstream signaling pathways. FLT3-ITD is associated with poor prognosis for younger patients with de novo AML and normal cytogenetics [40,41]. Therefore, efforts to develop inhibitors targeting FLT3 have led to successive generations of FLT3 inhibitors over the past decade, including the first-generation FLT3 inhibitors tandutinib, sunitinib, lestaurtinib, sorafenib and midostaurin, and the next generation FLT3 inhibitors quizartinib, crenolanib, and gilteritinib. Based on the mechanism of action, these TKIs can also be categorized into: type I TKIs (including midostaurin, gilteritinib and crenolanib) that bind the activation loop or ATPbinding pocket of the FLT3 receptor in the active conformation, and are active against FLT3-ITD and TKD; and type II (including sorafenib and quizartinib) that bind the ATP-binding pocket of FLT3 receptor in the inactive conformation and are active against FLT3-ITD but not TKD. Below, we describe some representative TKIs for FLT3 (Figure 1).

Midostaurin is an oral, first-generation TKI that targets both FLT3-ITD and -TKD mutations, and also shows effectiveness in suppressing PKC, c-Kit, VEGFR, and PDGFR-β. It was approved in 2017 by the FDA for the management of relapsed/refractory AML patients with FLT3 mutations, and was recently accepted as a front-line therapy for newly diagnosed AML patients with FLT3 mutations, systemic mastocytosis, and related disorders due to its inhibitory effect on c-Kit bearing a D816V mutation [42,43]. Based on the RATIFY study, midostaurin combined with standard chemotherapy, followed by the maintenance therapy for 1 year, significantly improved both overall survival (OS) (74.7 months vs. 25.6 months, p = 0.009) and event-free survival (EFS) (8.2 months vs 3.0 months, p = 0.002) but not complete remission (CR) compared to chemotherapy alone [44]. Midostaurin also reduces relapse in FLT3-mutant AML [45]. In a phase II study, midostaurin treatment reduced blast counts 50% or more in 71% patients with R/R AML or high-risk MDS with FLT3 mutations [46].

Sorafenib is an oral multi-kinase inhibitor for RAF-1, VEGFR, c-Kit, PDGFR, ERK and FLT3. Beyond inhibiting kinase activity, sorafenib stimulates IL-15 secretion and increases the survival of AML patients [47]. Moreover, sorafenib also inhibits SRCSTAT3 pathways. Sorafenib monotherapy in R/R AML has marked FLT3-ITD inhibition, good tolerability, and about 11.1% CR rate. Combinations of sorafenib with other agents were also investigated. For instance, azacitidine plus sorafenib showed a CRc of 43% and mOS of 6.2 months in 37 R/R and untreated unfit AML patients (93% were FLT-ITD) [48]. The use of combinations of sorafenib with intensive schemes of induction and consolidation in untreated AML was widely investigated and showed a trend to lower survival in AML with FLT3-ITD [49- 51]. Recently, an open-label, multicenter, randomized phase 3 trial was performed to investigate the efficacy and tolerability of sorafenib maintenance post-transplantation [52]. About 202 AML patients with FLT3-ITD receiving allogeneic HSC transplantation (allo-HSCT) were randomly assigned to sorafenib maintenance (400 mg orally twice daily) or non-maintenance (control) at 30- 60 days post-transplantation; the 1-year cumulative incidence of relapse was 7.0% in the sorafenib group and 24.5% in the control group. Within 210 days post transplantation, sorafenib treatment did not affect adverse events including infections (25% vs 24%), acute graft-versus-host-disease (GVHD; 23% vs 21%), chronic GVHD (18% vs 17%), and hematological toxicity (15% vs 7%). This study indicates that post-transplantation maintenance therapy by sorafenib reduces relapse and is well tolerated in AML with FLT3-ITD. In another randomized, placebo-controlled, double-blind phase II trial (SORMAIN), 83 adult patients with AML with FLT3-ITD in complete hematologic remission after HCT were randomly assigned to receive for 24 months either sorafenib (n = 43) or placebo (n = 40). The 24-month relapsefree survival (RFS) was 85.0% with sorafenib versus 53.3% with placebo [53]. Together, these studies indicate that sorafenib prevents AML relapse after allo-HSCT.

Gilteritinib is an oral potent type I FLT3 inhibitor, and is also effective against AXL. In a phase 3 trial, 371 R/R AML patients with FLT3 mutations were randomly assigned to receive either gilteritinib (120 mg daily) or salvage chemotherapy. The median overall survival (mOS) in the gilteritinib group was significantly longer than that in the chemotherapy group (9.3 months vs. 5.6 months). The median EFS was 2.8 vs 0.7 months, for gilteritinib and chemotherapy groups respectively, and the CR percentage of patients with full or partial hematologic recovery was 34.0% in the gilteritinib group and 15.3% in the chemotherapy group [54]. This indicates that gilteritinib resulted in significantly longer survival and higher remission rate than salvage chemotherapy among patients with relapsed or refractory FLT3-mutated AML [54]. This work led to the approval of gilteritinib by the FDA and EMA for R/R FLT3-mut AML.

Quizartinib is a potent, selective, second-generation FLT3 inhibitor shown to reduce blast number in the bone marrow of a substantial number of patients. A high complete remission response rate and overall survival in R/R AML patients with FLT3-ITD were also observed [55]. Several clinical trials, in which quizartinib was combined with conventional chemotherapy for frontline therapy or as a maintenance therapy following hematopoietic stem cell transplant, are ongoing. Quizartinib exhibited low nanomolar potency, high selectivity, and sustained inhibition of FLT3 kinase activity in leukemic cells of AML patients when compared to sorafenib and other FLT3 inhibitors [56].

FLT3 inhibitors are now important for treating a large subset of high-risk R/R patients, as a frontline for the treatment-naïve, or for maintenance after allo-HSCT. Recent randomized trials of TKIs, including gilteritinib, quizartinib, and sorafenib, predict an even wider use of FLT3 inhibitors in the future.

Tyrosine kinase inhibitors for AXL: The RTK AXL is emerging as a key player in tumor progression and metastasis, and its expression correlates with poor survival in a plethora of cancers. AXL belongs to the TYRO3, AXL, and MERTK (TAM) subfamily of RTKs. TAM family receptors are overexpressed in a wide variety of human cancers in which they provide tumor cells with a survival advantage [57]. TAM RTK activation mechanisms are unique. Growth arrest specific 6 (GAS6) is the ligand of AXL, and also binds to TYRO3 and MERTK. TAM activation stimulates downstream MEK, PI3K/AKT, and ERK pathways, and regulates cytoskeletal function, intracellular signaling, and gene expression (Figure 1) [57].

High AXL and GAS6 expression has been observed in AML and CML, and is associated with poor prognosis in AML [58]. A recent study showed that AXL and GAS6 expression is highly elevated in primitive AML cells, particularly in stem/progenitor cells [59]. In AML cells, RNA m6 A demethylase ALKBH5 affects mRNA stability of RTK AXL in an m6 A-dependent manner [60]. The AXL inhibitor ONO-7475 kills FLT3-mutant AML cells [61]. A potent selective AXL inhibitor, SLC391, was recently developed [59], and showed favorable pharmaceutical properties and efficacy in preclinical patient-derived xenotransplantation (PDX) models of AML. Additionally, AXL inhibition sensitized AML stem/ progenitor cells to treatment with the BCL2 inhibitor venetoclax, accompanied by strong synergistic effects in vitro and in PDX models [59]. A pharmacologic AXL inhibitor, AXL-Fc, inhibited cell growth, induced cell-cycle arrest and apoptosis, and relieved a block in myeloid differentiation of FLT3-ITD+ AML in vitro, suggesting that AXL contributes to the pathogenesis of FLT3-ITD+ AML by regulation of constitutive FLT3 activation [62]. Finally, AXL is required for resistance of FLT3-ITD+ AML cells to the FLT3 inhibitor [63]. Thus, inhibition of AXL activation may overcome resistance to FLT3-targeted therapy in FLT3-ITD+ AML.

Current challenges in treating myeloid leukemias with TKIs

Although some TKIs have achieved a success in induction of clinical remission of leukemias, the development TKI resistance and the existence of resistant leukemia stem cells (LSCs) pose challenges. For instance, discontinuation of TKI treatment results in early molecular relapse in some CML cases, and development of BCR-ABL mutations induces TKI resistance in some CML patients [64]. Also, it has been shown that TKIs are unable to eradicate the primitive quiescent CML LSCs [65-67]. The survival of primitive LSCs is independent of BCR-ABL kinase activity, and other signaling pathways might be responsible for the maintenance and survival of LSCs [68]. Therefore, targeting essential components in LSCs in addition to tarteting BCR-ABL with TKIs should prove more effective in treating myeloid leukemias.

REFERENCES
  1. Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature. 2001; 411: 355-365.
  2. Yang Y, Li S, Wang Y, Zhao Y, Li Q. Protein tyrosine kinase inhibitor resistance in malignant tumors: molecular mechanisms and future perspective. Signal Transduct Target Ther. 2022; 7: 329.
  3. Wong S, Witte ON. The BCR-ABL story: bench to bedside and back. Annu Rev Immunol. 2004; 22: 247-306.
  4. Melo JV, Barnes DJ. Chronic myeloid leukaemia as a model of disease evolution in human cancer. Nat Rev Cancer. 2007; 7: 441-453.
  5. Savona M, Talpaz M. Getting to the stem of chronic myeloid leukaemia. Nat Rev Cancer. 2008; 8: 341-350.
  6. Ren R. Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia. Nat Rev Cancer. 2005; 5: 172-183.
  7. Osman AEG, Deininger MW. Chronic Myeloid Leukemia: Modern therapies, current challenges and future directions. Blood Rev. 2021; 49: 100825.
  8. Nakao M, Yokota S, Iwai T, Kaneko H, Horiike S, Kashima K, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia. 1996; 10: 1911-1918.
  9. Yamamoto Y, Kiyoi H, Nakano Y, Suzuki R, Kodera Y, Miyawaki S, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood. 2001; 97: 2434-2439.
  10. Silver RT, Woolf SH, Hehlmann R, Appelbaum FR, Anderson J, Bennett C, et al. An evidence-based analysis of the effect of busulfan, hydroxyurea, interferon, and allogeneic bone marrow transplantation in treating the chronic phase of chronic myeloid leukemia: developed for the American Society of Hematology. Blood. 1999; 94: 1517-1536.
  11. Hehlmann R, Heimpel H, Hasford J, Kolb HJ, Pralle H, Hossfeld DK, et al. Randomized comparison of interferon-alpha with busulfan and hydroxyurea in chronic myelogenous leukemia. The German CML Study Group. Blood. 1994; 84: 4064-4077.
  12. Hehlmann R, Kister P, Willer A, Simon M, Schenk M, Seifarth W, et al. Therapeutic progress and comparative aspects in chronic myelogenous leukemia (CML): interferon alpha vs. hydroxyurea vs. busulfan and expression of MMTV-related endogenous retroviral sequences in CML. German CML Study Group. Leukemia. 1994; 8: S127-132.
  13. Goldman JM. Chronic myeloid leukemia: a historical perspective. Semin Hematol. 2010; 47: 302-311.
  14. Melo JV. The diversity of BCR-ABL fusion proteins and their relationship to leukemia phenotype. Blood. 1996; 88: 2375-2384.
  15. Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM, Fanning S, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med. 1996; 2: 561-566.
  16. Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford JM, et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N Engl J Med. 2001; 344: 1038-1042.
  17. Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001; 344: 1031-1037.
  18. Druker BJ, Lydon NB. Lessons learned from the development of an abl tyrosine kinase inhibitor for chronic myelogenous leukemia. J Clin Invest. 2000; 105: 3-7.
  19. Capdeville R, Buchdunger E, Zimmermann J, Matter A. Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov. 2002; 1: 493-502.
  20. Schindler T, Bornmann W, Pellicena P, Miller WT, Clarkson B, Kuriyan J. Structural mechanism for STI-571 inhibition of abelson tyrosine kinase. Science. 2000; 289: 1938-1942.
  21. Mukherjee J, Kamnasaran D, Balasubramaniam A, Radovanovic I, Zadeh G, Kiehl TR, et al. Human schwannomas express activated platelet-derived growth factor receptors and c-kit and are growth inhibited by Gleevec (Imatinib Mesylate). Cancer Res. 2009; 69: 5099-5107.
  22. O’Brien SG, Deininger MW. Imatinib in patients with newly diagnosed chronic-phase chronic myeloid leukemia. Semin Hematol. 2003; 40: 26-30.
  23. Quintas-Cardama A, Kantarjian H, Cortes J. Flying under the radar: the new wave of BCR-ABL inhibitors. Nat Rev Drug Discov. 2007; 6: 834-848.
  24. Kantarjian HM, Shah NP, Cortes JE, Baccarani M, Agarwal MB, Undurraga MS, et al. Dasatinib or imatinib in newly diagnosed chronic-phase chronic myeloid leukemia: 2-year follow-up from a randomized phase 3 trials (DASISION). Blood. 2012; 119: 1123-1129.
  25. Jabbour E, Kantarjian HM, Saglio G, Steegmann JL, Shah NP, Boque C, et al. Early response with dasatinib or imatinib in chronic myeloid leukemia: 3-year follow-up from a randomized phase 3 trial (DASISION). Blood. 2014; 123: 494-500.
  26. Weisberg E, Manley PW, Breitenstein W, Bruggen J, Cowan-Jacob SW, Ray A, et al. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer cell. 2005; 7: 129-141.
  27. Jabbour E, Cortes JE, Kantarjian HM. Suboptimal response to or failure of imatinib treatment for chronic myeloid leukemia: what is the optimal strategy? Mayo Clin Proc. 2009; 84: 161-169.
  28. Saglio G, Kim DW, Issaragrisil S, le Coutre P, Etienne G, Lobo C, et al. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med. 2010; 362: 2251-2259.
  29. Jabbour E. Chronic myeloid leukemia: First-line drug of choice. Am J Hematol. 2016; 91: 59-66.
  30. Shamroe CL, Comeau JM. Ponatinib: A new tyrosine kinase inhibitor for the treatment of chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia. Ann Pharmacother. 2013; 47: 1540-1546.
  31. O’Hare T, Shakespeare WC, Zhu X, Eide CA, Rivera VM, Wang F, et al. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer cell. 2009; 16: 401-412.
  32. Cortes JE, Kim DW, Pinilla-Ibarz J, le Coutre P, Paquette R, Chuah C, et al. A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias. N Engl J Med. 2013; 369: 1783-1796.
  33. Larson RA. Is there a best TKI for chronic phase CML Hematology Am Soc Hematol Educ Program. 2015; 2015: 250-256.
  34. Boyer SW, Schroeder AV, Smith-Berdan S, Forsberg EC. All hematopoietic cells develop from hematopoietic stem cells through Flk2/Flt3-positive progenitor cells. Cell Stem Cell. 2011; 9: 64-73.
  35. Gotze KS, Ramirez M, Tabor K, Small D, Matthews W, Civin CI. Flt3high and Flt3low CD34+ progenitor cells isolated from human bone marrow are functionally distinct. Blood. 1998; 91: 1947-1958.
  36. Stirewalt DL, Radich JP. The role of FLT3 in haematopoietic malignancies. Nat Rev Cancer. 2003; 3: 650-665.
  37. Lavagna-Sevenier C, Marchetto S, Birnbaum D, Rosnet O. The CBLrelated protein CBLB participates in FLT3 and interleukin-7 receptor signal transduction in pro-B cells. J Biol Chem. 1998; 273: 14962- 14967.
  38. Lavagna-Sevenier C, Marchetto S, Birnbaum D, Rosnet O. FLT3 signaling in hematopoietic cells involves CBL, SHC and an unknown P115 as prominent tyrosine-phosphorylated substrates. Leukemia. 1998; 12: 301-310.
  39. Rosnet O, Buhring HJ, Marchetto S, Rappold I, Lavagna C, Sainty D, et al. Human FLT3/FLK2 receptor tyrosine kinase is expressed at the surface of normal and malignant hematopoietic cells. Leukemia. 1996; 10: 238-248.
  40. Whitman SP, Archer KJ, Feng L, Baldus C, Becknell B, Carlson BD, et al. Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a cancer and leukemia group B study. Cancer Res. 2001; 61: 7233-7239.
  41. Gale RE, Green C, Allen C, Mead AJ, Burnett AK, Hills RK, et al. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood. 2008; 111: 2776- 2784.
  42. Song MK, Park BB, Uhm JE. Clinical Efficacies of FLT3 Inhibitors in Patients with Acute Myeloid Leukemia. Int J Mol Sci. 2022; 23: 12708.
  43. Gallogly MM, Lazarus HM, Cooper BW. Midostaurin: a novel therapeutic agent for patients with FLT3-mutated acute myeloid leukemia and systemic mastocytosis. Ther Adv Hematol. 2017; 8:245-261.
  44. Ge SS, Liu SB, Xue SL. Developments and challenges of FLT3 inhibitors in acute myeloid leukemia. Front Oncol. 2022; 12: 996438.
  45. Larson RA, Mandrekar SJ, Huebner LJ, Sanford BL, Laumann K, Geyer S, et al. Midostaurin reduces relapse in FLT3-mutant acute myeloid leukemia: the Alliance CALGB 10603/RATIFY trial. Leukemia. 2021; 35: 2539-2551.
  46. Fischer T, Stone RM, Deangelo DJ, Galinsky I, Estey E, Lanza C, et al. Phase IIB trial of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. J Clin Oncol. 2010; 28: 4339-4345.
  47. Mathew NR, Baumgartner F, Braun L, O’Sullivan D, Thomas S, Waterhouse M, et al. Sorafenib promotes graft-versus-leukemia activity in mice and humans through IL-15 production in FLT3-ITDmutant leukemia cells. Nat Med. 2018; 24: 282-291.
  48. Ravandi F, Alattar ML, Grunwald MR, Rudek MA, Rajkhowa T, Richie MA, et al. Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood. 2013; 121: 4655-4662.
  49. Ravandi F, Cortes JE, Jones D, Faderl S, Garcia-Manero G, Konopleva MY, et al. Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia. J Clin Oncol. 2010; 28: 1856-1862.
  50. Serve H, Krug U, Wagner R, Sauerland MC, Heinecke A, Brunnberg U, et al. Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial. J Clin Oncol. 2013; 31: 3110-3118.
  51. Rollig C, Serve H, Huttmann A, Noppeney R, Muller-Tidow C, Krug U, et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015; 16: 1691-1699.
  52. Xuan L, Wang Y, Huang F, Fan Z, Xu Y, Sun J, et al. Sorafenib maintenance in patients with FLT3-ITD acute myeloid leukaemia undergoing allogeneic haematopoietic stem-cell transplantation: an open-label, multicentre, randomised phase 3 trials. Lancet Oncol. 2020; 21: 1201-1212.
  53. Burchert A, Bug G, Fritz LV, Finke J, Stelljes M, Rollig C, et al. Sorafenib Maintenance After Allogeneic Hematopoietic Stem Cell Transplantation for Acute Myeloid Leukemia With FLT3-Internal Tandem Duplication Mutation (SORMAIN). J Clin Oncol. 2020; 38: 2993-3002.
  54. Perl AE, Martinelli G, Cortes JE, Neubauer A, Berman E, Paolini S, et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3- Mutated AML. N Engl J Med. 2019; 381: 1728-1740.
  55. Cortes JE, Kantarjian H, Foran JM, Ghirdaladze D, Zodelava M, Borthakur G, et al. Phase I study of quizartinib administered daily to patients with relapsed or refractory acute myeloid leukemia irrespective of FMS-like tyrosine kinase 3-internal tandem duplication status. J Clin Oncol. 2013; 31: 3681-3687.
  56. Zarrinkar PP, Gunawardane RN, Cramer MD, Gardner MF, Brigham D, Belli B, et al. AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML). Blood. 2009; 114: 2984-2992.
  57. Graham DK, DeRyckere D, Davies KD, Earp HS. The TAM family: phosphatidylserine sensing receptor tyrosine kinases gone awry in cancer. Nat Rev Cancer. 2014; 14: 769-785.
  58. Ben-Batalla I, Schultze A, Wroblewski M, Erdmann R, Heuser M, Waizenegger JS, et al. Axl, a prognostic and therapeutic target in acute myeloid leukemia mediates paracrine crosstalk of leukemia cells with bone marrow stroma. Blood. 2013; 122: 2443-2452.
  59. Niu X, Rothe K, Chen M, Grasedieck S, Li R, Nam SE, et al. Targeting AXL kinase sensitizes leukemic stem and progenitor cells to venetoclax treatment in acute myeloid leukemia. Blood. 2021; 137: 3641-3655.
  60. Wang J, Li Y, Wang P, Han G, Zhang T, Chang J, et al. Leukemogenic Chromatin Alterations Promote AML Leukemia Stem Cells via a KDM4C-ALKBH5-AXL Signaling Axis. Cell Stem Cell. 2020; 27: 81-97
  61. Post SM, Ma H, Malaney P, Zhang X, Aitken MJL, Mak PY, et al. AXL/ MERTK inhibitor ONO-7475 potently synergizes with venetoclax and overcomes venetoclax resistance to kill FLT3-ITD acute myeloid leukemia. Haematologica. 2022; 107: 1311-1322.
  62. Park IK, Mishra A, Chandler J, Whitman SP, Marcucci G, Caligiuri MA. Inhibition of the receptor tyrosine kinase Axl impedes activation of the FLT3 internal tandem duplication in human acute myeloid leukemia: implications for Axl as a potential therapeutic target. Blood. 2013; 121: 2064-2073.
  63. Park IK, Mundy-Bosse B, Whitman SP, Zhang X, Warner SL, Bearss DJ, et al. Receptor tyrosine kinase Axl is required for resistance of leukemic cells to FLT3-targeted therapy in acute myeloid leukemia. Leukemia. 2015; 29: 2382-2389.
  64. O’Hare T, Eide CA, Deininger MW. Bcr-Abl kinase domain mutations, drug resistance, and the road to a cure for chronic myeloid leukemia. Blood. 2007; 110: 2242-2249.
  65. Corbin AS, Agarwal A, Loriaux M, Cortes J, Deininger MW, Druker BJ. Human chronic myeloid leukemia stem cells are insensitive to imatinib despite inhibition of BCR-ABL activity. J Clin Invest. 2011; 121: 396-409.
  66. Chomel JC, Bonnet ML, Sorel N, Bertrand A, Meunier MC, Fichelson S, et al. Leukemic stem cell persistence in chronic myeloid leukemia patients with sustained undetectable molecular residual disease. Blood. 2011; 118: 3657-3660.
  67. Chomel JC, Turhan AG. Chronic myeloid leukemia stem cells in the era of targeted therapies: resistance, persistence and long-term dormancy. Oncotarget. 2011; 2: 713-727.
  68. Zhang H, Peng C, Hu Y, Li H, Sheng Z, Chen Y, et al. The Blk pathway functions as a tumor suppressor in chronic myeloid leukemia stem cells. Nat Genet. 2012; 44: 861-871.
Received : 23 May 2023
Accepted : 27 Apr 2023
Published : 27 Apr 2023
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X