Bioscience Biotechnology Research Communications

An International  Peer Reviewed Refereed Open Access Journal

P-ISSN: 0974-6455 E-ISSN: 2321-4007

Bioscience Biotechnology Research Communications

An Open Access International Journal

Roopali Fotra,*1 Jyotdeep Kour,1 Deepti Mahajan, 2Parvinder Kumar1 and Manoj K. Dhar1

1Institute of Human Genetics, University of Jammu, Jammu-180006

2Department of Pathology/Hematology, Govt. Medical College, Jammu

Article Publishing History

Received: 06/01/2020

Accepted After Revision: 29/02/2020

ABSTRACT:

Acute myeloid leukemia (AML), a cancer of myeloid cells involves the abnormal proliferation and differentiation of myeloid stem cells. AML accounts for approximately 30% of leukemia’s but >40% of leukemia-related deaths. AML is highly diverse and cytogenetic analysis of metaphase cells reveals that approximately 40-50% of patients with de novo AML have a normal karyotype.. This study demonstrated activating mutations of FLT3 gene due to recent advances in cell and molecular biology have revolutionized our understanding of normal hematopoiesis. Mutations within the FMS-like tyrosine kinase 3 (FLT3) genes represent one of the most frequently identified genetic alterations that disturb intracellular signaling networks with a key role in leukemia pathogenesis. The present study has been designed to  highlight and signify the importance of FLT3 and its related gene mutations involved in the onset and progression of Leukemias. Since mutations in FLT3 gene are one of the most common clinically relevant mutations which are expressed in 90% of leukemic blasts of patients with acute myeloid leukemia. Thus, there is an urgent need for the better understanding of the key genetic mutations involved in disease progression and prognosis. FLT3 testing should be done in parallel with cytogenetic testing and can open new horizons for better diagnosis and better treatment option.

KEYWORDS:

Acute Myeloid Leukemia, FMS-like Tyrosine Kinase, genetic Mutations

Download this article as:

Copy the following to cite this article:

Fotra R, Kour J, Mahajan D, Kumar P, Dhar M. K. FMS-like Tyrosine Kinase 3 (FLT3) Gene as a Significant Biomarker for Acute Myeloid Leukemia. Biosc.Biotech.Res.Comm. 2020;13(1).


Copy the following to cite this URL:

Fotra R, Kour J, Mahajan D, Kumar P, Dhar M. K. FMS-like Tyrosine Kinase 3 (FLT3) Gene as a Significant Biomarker for Acute Myeloid Leukemia. Biosc.Biotech.Res.Comm. 2020;13(1). Available from: https://bit.ly/3aMrRg0

Copyright © Fotra et al., This is an open access article distributed under the terms of the Creative Commons Attribution License (CC-BY) https://creativecommons.org/licenses/by/4.0/, which permits unrestricted use distribution and reproduction in any medium, provide the original author and source are credited.


INTRODUCTION

Leukemias are monoclonal diseases that originate from individual cells in the bone marrow. Leukemia like any other cancer also follows a multistep process which a normal cell must possess or pass through a number of distinct intermediate stages before attaining the status of malignancy. Based on the origin of the predominant cell type (myeloid or lymphoid) and the rate of disease progression (acute or chronic), leukemia is categorized into four major subtypes: acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia (CLL). According to a recent report published by Taisen et al., (2019), the incidences of AML and CML has remained constant prior to 2011 but there has been a sudden increase in the incidence rates of CLL. Acute myeloid leukemia (AML), a cancer of myeloid cells involves the abnormal proliferation and differentiation of myeloid stem cells. It is generally accepted that survival, proliferation and differentiation are the three fundamental cellular processes that define normal hematopoietic cells. In acute myeloid leukemia (AML), a heterogeneous disorder of the hematopoietic progenitor cells, abnormalities have been identified that affect the balance between cell proliferation, survival and differentiation. These abnormalities result in the expansion of an abnormal stem cell clone (Irons et al., 1996 Taisen et al., 2019 Nicholas et al 2020).

AML accounts for approximately 30% of leukemia’s but >40% of leukemia-related deaths. It is the most common acute leukemia in adult population. The median age at diagnosis of AML is approximately 67 years and about 1/3rd of Leukemias among adults have been reported from developed countries. The therapeutic armamentarium of acute myeloid leukemia (AML) has rapidly expanded in the past few years, driven largely by translational research into its genomic landscape and an improved understanding of mechanisms of resistance to conventional therapies (Nicholas et al., 2020). Genetic alterations are the frequent features of all human cancers which include amplification, deletions, rearrangements and point mutations. Thus, genomic investigations of AML have also demonstrated the role of several genes which on recurrent mutations can lead to the new genomic classifications and predictive biomarkers, and new therapeutic targets (Daver et al., 2019 Nicholas et al 2020).

AML is highly diverse and cytogenetic analysis of metaphase cells reveals that approximately 40-50% of patients with de novo AML have a normal karyotype. These patients are classified with an intermediate clinical prognosis because clinically they do not have a reference marker and its biological origin is still unknown. The most frequent abnormalities are translocations t(15;17), t(8;21), inv(16) and a gain of number 8. Recently, with the development of methodologies of massive sequencing, new genetic mutations associated with acute myeloid leukemia have been identified. Some of the identified genes include KIT, FLT3, NPM1, CEBPA, RAS, WT1, BAALC, ERG, MN1, DNMT, TET2, IDH, ASXL1, PTPN11 and CBL. Of all these, WHO highlighting the related mutations in FLT3, NPM1 and CEBPα genes because they are associated with treatment response and progress of this disease (Swerdlow et al., 2008, Takahashi 2011,  Martelli 2013 and Liesveld and  Lichtman, 2016, Rubnitz et al., 2016  Nicholas et al 2020).

A number of genetic mutations, such as point mutations, gene rearrangements, and chromosomal translocations, which are involved in the pathogenesis of leukemia, have been documented. Recent advances in cell and molecular biology have revolutionized our understanding of normal hematopoiesis. Mutations within the FMS-like tyrosine kinase 3 (FLT3) genes represent one of the most frequently identified genetic alterations that disturb intracellular signaling networks with a key role in leukemia pathogenesis. FLT3, a receptor tyrosine kinase (RTK), is a membrane-bound receptor is primarily expressed on committed myeloid and lymphoid progenitors. FLT3 is composed of an immunoglobulin-like extracellular ligand-binding domain, a transmembrane domain, a juxtamembrane dimerization domain and a highly conserved intracellular kinase domain interrupted by a kinase insert. FLT3 belongs to the class III subfamily of RTKs, which include structurally similar members such as c-FMS, c-KIT, and PDGF receptor (Martelli et al, 1996 and Gabbianelli et al., 1995 Liesveld and  Lichtman 2016).

FLT3 is expressed in 90% of leukemic blasts of patients with acute myeloid leukemia (Carow et al.,1996) . Approximately 20–30%ofAMLpatientsharbor a unique feature i.e. Internal Tandem Duplication (ITD) mutation in the FLT3 gene between exons 14 and 15 in the juxta membrane domain, which often results in high blast accounts, increased risk of relapse and decreased survival. FLT3-ITD is especially a frequent feature in patients with normal karyotype, t (15;17) (q22;q12) [PML-RARA] and t(6;9)(p23;q34) [DEK-NUP214]which leads to uncontrolled cellular proliferation, survival, and differentiation through constitutive activation of FLT3, (Meshinchi and Appelbaum (2009).  Stirewalt and Radich 2003, Parcells et al, 2006) .FLT3-ITD occurs in the form of a replicated sequence in the juxta membrane domain and/or TKD1 of the FLT3 receptor and varies in location and length within these domains.

FLT3-ITD (high) is a driver mutation that presents with a high leukemic burden, confers a poor prognosis, and bears a significant negative impact on the management of patients with AML (Ding et al., 2012, Grimwade and Mrozek 2011).  This ITD disrupts the auto inhibitory function of the juxta-membrane domain and results in ligand independent activation of the FLT3 receptor. This leads to a proliferative signal via activation of its downstream effectors (Kiyoi et al., 1998, Levis 2013) . Thus, the ITD leads to gain of function mainly by inducing hyper-responsivity of the FLT3 receptor to FL rather than through auto-activation of the receptor (Griffith et al., 2004), Therefore, the FLT3-ITD mutation directly or indirectly confers a selective advantage to a clone in its microenvironment. About 75% of patients with FLT3ITD–mutated AML at diagnosis continue to have the ITD mutation at relapse (Zheng et al., 2011) suggesting that FLT3-ITD may function as the driver mutation responsible for progressing the disease into overt leukemia.

The second common types of mutations in FLT3 are missense mutations in exon 20 of the activation loop (A-loop) in the tyrosine kinase domain (TKD). Almost all these mutations involve the substitution of an aspartate with a tyrosine at codon 835 (D835Y) by a point mutation (GAT→TAT). Aspartate in the 835 position belongs to the domain DFG (Aspartate-Phenylalanine-Glycine) in the A-loop, playing a critical role in preventing efficient binding of ATP. This type of mutation occurs in approximately 7% of patients with AML (  Yomamoto et al., 2001 Kronke et al., 2013 Liesveld and  Lichtman  (2016).

In recent years, it has been shown that somatic activating mutations of the FLT3 gene are the most common genetic abnormalities in AML and have a significant impact on prognosis. Female patients are affected more frequently and these mutations are also associated with hypercellularity and a higher incidence of recurrence (Abu- Duhier et al., 2001). Routine testing for FLT3 in patients with cytogenetically normal AML had been recommended since at least 2010 (Fenski et al., 2000), which corresponds to the time at which molecular testing was routinely performed in 100% of patients at academic centers but not at community sites. This suggests that there is a lack of awareness about the significance of molecular testing at community sites.

CONCLUSION

Identification of FLT3 mutations in AML has yielded novel approaches to the management of this disease. Over the last decade, the biology and the function of the wild-type and mutated FLT3 receptor have been well characterized. Whether it is through their utility as prognostic factors or their use as a target for directed therapies, FLT3 mutations have provided clinicians with novel therapeutic options for a large subset of AML patients. Identification of FLT3 mutations in AML has raised the potential for its utility as a molecular marker for risk-based therapy and as a target for directed therapy with novel small molecular inhibitors. Subsequent studies identified numerous other potential compounds (MLN518, PKC412, SU5416, SU5614, SU11248, CEP-701, CEP-5214) that also block FLT3 activation. Two compounds (CEP-701 and PKC-412) have shown some therapeutic promise for AML patients with FLT3 mutations. CEP-701 (Lestaurtinib) is an indolo carbazole compound that inhibits auto phosphorylation of the WT and mutant FLT3 receptors (Levis et al., 2005 and Levis et al., 2004, Nicholas et al 2020). Current clinical trials are combining FLT3 inhibitors with conventional chemotherapy in an attempt to increase the cytotoxic effect against leukemia cells and reverse the poor prognosis for AML patients with FLT3 mutations.

Funding Agency:

Authors are extremely thankful to the Science and Engineering Research Board (SERB), DST, Govt. of India for providing financial assistance for carrying out the research work.

REFERENCES

Abu-Duhier F.M, Goodeve A.C, Wilson G.A (2011). Identification of novel FLT-3 Asp835 mutations in adult acute myeloid leukaemia. Br J Haematol, 113, 983-8.

Carow C.E., Levenstein M., Kaufmann S.H., et al (1996). Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias. Blood., 87:1089–1096.

Daver N., Schlenk R.F., Russell N.H and Levis M.J. (2019). ­­­Targeting FLT3 mutations in AML: review of current knowledge and evidence. Leukemia, 33, 299-312.

Ding L., Ley T.J., Larson D.E., Miller C.A., Koboldt D.C., Welch JS, et al. (2012). Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature, 481, 506– 10.

Dohner H., Estey E.H., Amadori S., et al. (2010). Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood, 115, 453–474.

Fenski R., Flesch K., Serve S., et al. (2000). Constitutive activation of FLT3 in acute myeloid leukemia and its consequences for growth of 32D cells. Br J Haematol, 108, 322-30.

Gabbianelli M., Pelosi E., Montesoro E., et al. (1995). Multilevel effects of flt3 ligand on human hematopoiesis: expansion of putative stem cells and proliferation of granulomonocytic progenitors/monocyticprecursors. Blood, 86, 1661.

Griffith J., Black J., Faerman C., et al. (2004). The structural basis for autoinhibition of FLT3 by the juxtamembrane domain. Mol Cell ,13, 169-78.

Grimwade D., and Mrozek K. (2011). Diagnostic and prognostic value of cytogenetics in acute myeloid leukemia. Hematol Oncol Clin North Am. 25, 61.

Irons F G Burmit  R.D, Stillman WS. (1996). The process of leukemogenesis. Environ Health Perspect,104, 1239-46.

Kiyoi H., Towatari M., Yokota S., et al. (1998). Internal tandem duplication of the FLT3 gene is a novel modality of elongation mutation which causes constitutive activation of the product. Leukemia, 12, 1333-7.

Kronke J., Bullinger L., Teleanu V., Tschurtz F., Gaidzik V.I., Kuhn M.W., et al. (2013). Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia. Blood, 122,100–8.

Levis M., Smith B.D., Beran M., et al. (2005). A randomized, open-label study of lestaurtinib (CEP-701), an oral FLT3 inhibitor, administered in sequence with an oral chemotherapy in patients with relapsed AML harboring FLT3 activating mutations: clinical response correlates with successful FLT3 inhibition. Am Soc Hematol Annu Meet Abstracts, 106, 403.

Levis M. (2013). FLT3 mutations in acute myeloid leukemia: What is the best approach in 2013? Hematology Am Soc Hematol. Educ. Program, 220–6.

Levis M., Pham R., Smith B.D., Small D. (2004). In vitro studies of a FLT3 inhibitor combined with chemotherapy: sequence of administration is important in order to achieve synergistic cytotoxic effects. Blood, 104:1145.

Liesveld J.L., Lichtman M.A. (2016). Acute myelogenous leukemia. In: Kaushansky K, Lichtman MA, Prchal JT, et al., editors. Williams Hematology. 9th ed. . United States of America: McGraw-Hill Education.

Löwenberg B.,  Downing J.R.,  Burnett, A. (1999). Acute myeloid leukemia. N. Engl. J. Med., 341, 1051–1062.

Meshinchi S., Appelbaum F.R. (2009).  Structural and functional alterations of FLT3 in acute myeloid leukemia. Clin Cancer Res., 15(13), 4263–9.

Nicholas J Short, Marina Konopleva, Courtney D. Di Nardo and Naval Daver (2020). Advances in the Treatment of Acute Myeloid Leukemia: New Drugs and New Challenges.

Martelli M.P., Sportoletti P., Tiacci E., et al. (2013). Mutational landscape of AML with normal cytogenetics: Biological and clinical implications. Blood Rev., 27(1), 13–22.)

Parcells B.W., Ikeda A.K., Simms-Waldrip T., et al. (2006). FMS-like tyrosine kinase 3 in normal hematopoiesis and acute myeloid leukemia. Stem Cells,  24(5), 174–84.

Rosnet O., Buhring H.J., Marchetto S., et al. (1996). Human FLT3/FLK2 receptor tyrosine kinase is expressed at the surface of normal and malignant hematopoietic cells. Leukemia, 10, 238

Rubnitz J.E., Gibson B., Smith F.O. (2010). Acute myeloid leukemia. Hematol. Oncol. Clin. North Am., 24(1), 35–63

Stirewalt D.L., Radich J.P. (2003). The role of FLT3 in haematopoietic malignancies. Nat. Rev. Cancer., 3(9), 650–65.

Swerdlow S.H., Campo E., Harris N.L., Jaffe E.S., Pileri S.A, Stein H., Thiele J., Vardiman J., (2008) editors. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4a ed. Lyon: International Agency for Research on Cancer (IARC).

Takahashi S. (2011). Current findings for recurring mutations in acute myeloid leukemia. J Hematol Oncol., 2011, 4, 36.

Taisen H., Min L.T., Alison B., & WenYong C. (2019).  An emerging trend of rapid increase of leukemia but not all cancers in the aging population in the United States. Scientific reports, Nature.

Yamamoto Y., Kiyoi H., Nakano Y., et al. (2001) Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood, 97, 2434-9.

Yamamoto, J.F.; Goodman, M.T. (2008). Patterns of leukemia incidence in the United States by subtype and demographic characteristics, 1997–2002. Cancer Causes Control,  379–390.

Zheng R., Bailey E., Nguyen B, et al. (2011). Further activation of FLT3 mutants by FLT3 ligand. Oncogene 2011, 30, 4004-14.