REVIEW For reprint orders, please contact: [email protected]

Mutations in tyrosine kinase and tyrosine phosphatase and their relevance to the target therapy in hematologic malignancies Ni Zhu‡,1, Haowen Xiao‡,1,2, Li-Mengmeng Wang1, Shan Fu1, Chan Zhao3 & He Huang*,1

ABSTRACT Protein tyrosine kinases and protein tyrosine phosphatases play pivotal roles in regulation of cellular phosphorylation and signal transduction with opposite functions. Accumulating evidences have uncovered the relevance of genetic alterations in these two family members to hematologic malignancies. This review underlines progress in understanding the pathogenesis of these genetic alterations including mutations and aberrant expression and the evolving protein tyrosine kinases and protein tyrosine phosphatases targeted therapeutic strategies in hematologic neoplasms.

The first description of enzymatic phosphorylation of proteins was reported by Eugene P Kennedy nearly 60 years ago [1] , followed by the study of Tony Hunter and colleagues who discovered protein tyrosine kinases (PTKs) in 1979 [2] . For the last three decades, regulation of protein tyrosine phosphorylation has been proved to play important roles in signal transduction of proliferation, differentiation, adhesion, metabolic homeostasis and programmed death of eukaryotic cells [3] . PTKs participate in phosphorylation, which is reversed by protein tyrosine phosphatases (PTPs), namely, dephosphorylation. Dysregulation of protein tyrosine phosphorylation caused by either gene mutations or aberrant expression of PTKs or PTPs, which leads to abnormal biological functions mainly including excessive cell proliferation, impaired cell differentiation and dysregulation of cell apoptosis, has been revealed to be involved in the pathological process of cancer. Depending on the important roles, both PTKs and PTPs play in malignancies, the corresponding target therapy has paved way in treating cancer successfully. Precisely designed kinase inhibitors have been employed in lung cancer [4] , breast cancer [5] , gastrointestinal cancer [6] and leukemia [7] . Advanced understanding of phosphorylation regulation in cancer prompts novel target therapeutic strategies. This review focuses on the genetic alterations including mutations and aberrant expression of PTPs and PTKs and their relevance to the target therapy in hematologic malignancies.

KEYWORDS 

• genetic alterations • hematologic malignancies • protein tyrosine kinases • protein tyrosine phosphatases • target

therapy

Function of PTKs & PTPs PTKs and PTPs regulate downstream signal pathway by phosphorylation and dephosphorylation, which are supposed to be the most important way of intracellular signal regulation, respectively in the opposite directions to achieve physiological or pathological states. The common signal Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China 2 Department of Haematology, Guangzhou Liuhuaqiao Hospital, Guangzhou, Guangdong Province, PR China 3 Department of Paediatrics, Jinhua Central Hospital, Jinhua, Zhejiang Province, PR China *Author for correspondence: Tel.: +86 571 8723 6706; Fax: +86 571 8723 6562; [email protected] ‡ Authors contributed equally 1

10.2217/FON.14.280 © 2015 Future Medicine Ltd

Future Oncol. (2015) 11(4), 659–673

part of

ISSN 1479-6694

659

Review  Zhu, Xiao, Wang, Fu, Zhao & Huang pathways include PI3K and PLC-γ relating to apoptosis. PI3K takes part in resisting apoptosis by phosphorylating AKT in hematologic neoplasms. On the contrary, PLC-γ phosphorylates protein kinase C (PKC), which will finally activate NF-κB that promotes apoptosis. On the other hand, PTKs and PTPs regulate expression of particular genes or proliferation through JAK/STAT and Ras/MEK/ERK pathways. In addition, the regulation of cytoskeleton through β-catenin/E-cadherin pathway is supposed to be related to migration and invasion of cancer cells (Figure 1) [8] . Genetic alterations in PTKs in hematologic malignancies PTKs are known as a large family of enzymes that catalyze the phosphorylation of tyrosine residues. These enzymes play important roles in varieties of cellular activities and vital signal pathways. PTKs are divided into two classes, including receptor tyrosine kinases (RTKs) possessing an N-terminal extracellular domain, a single transmembrane domain and a C-terminal cytoplasmic domain (Figure 2) and non-RTKs, most of which are soluble intracellular proteins without a transmembrane domain [9] . RTKs, a group of high-affinity cell surface receptors for many polypeptide growth factors, PTKs

cytokines and hormones, have already been identified as approximately 20 different classes [10] . Most RTKs function in transmembrane signal process, genetic alterations of which have been reported to be associated with hematopoietic malignancies that have been indicated in Table 1. On the other hand, the non-RTKs (nRTKs) are responsible for signal transduction to the nucleus. nRTKs are cytoplasmic enzymes that can catalyze the phosphate group removing from ATP to a tyrosine residue of a protein, the main function of which is the regulation of the immune system, in activated T and B cells in the immune system [11] . Genetic alterations in the nRTK genes also have been indicated to be relevant to hematopoietic malignancies (Table 2) . ●●FLT-3

FLT-3, which belongs to FMS/KIT family, plays a role in the biology of early hematopoietic progenitor cells and is highly expressed in both acute myelocytic leukemia and acute lymphocytic leukemia [12] . The major somatic mutations include internal tandem duplications (ITDs) that occur in exons 14 and 15 [13] and other point mutations within the activation loop of the tyrosine kinase domain (FLT3/TKD mutations) [14] . ITD mutations are detectable in about 20% of acute myeloid leukemia (AML) patients

PTPs Gene alterations

PI3K

Ras

PLCγ

PKC AKT

β-catenin/E-cadherin

JAK/STAT MEK1/2

MAPK pathway

IKK Cytoskeleton regulation Anti-apoptosis

NF-κB

Apoptosis

ERK1/2

Gene expression

Proliferation

Figure 1. The main signal transduction pathways involved in protein tyrosine kinases and protein tyrosine phosphatases relevant to hematopoietic neoplasms. PTK: Protein tyrosine kinase; PTP: Protein tyrosine phosphatase.

660

Future Oncol. (2015) 11(4)

future science group

Mutations in tyrosine kinase & tyrosine phosphatase 

Extracellular Class III

PDGFR

Class IV

FGFR

Class VII

TRK

Class IX

AXL

TM

Review

Intracellular

Intracellular kinase domain

TM

IgD

LRD

AB

Fn

Figure 2. The structures of protein tyrosine kinases involved in hematopoietic neoplasms. Receptor tyrosine kinases possess an N-terminal extracellular domain, a single transmembrane domain and a C-terminal cytoplasmic domain. AB: Acidic box; FGFR: FGF receptor; FN: Fibronectin III repeat; LRD: Leucine-rich domain; PDGFR: PDGF receptor; TM: Transmembrane domain.

and indicated a poor prognosis [15] . The FLT3 isoforms containing ITD mutations possessed oncogenic potential by downstream constitutively activation of RAS, STAT5 and MAPK pathway [16,17] . The TKD mutations are reported in approximately 7% of patients, which seems to incline to better prognosis [18] . It has been reported that FLT3/ITD+ patients had heavier leukocytosis and higher percentage of BM blast cells at diagnosis. The presence of an FLT3/ITD was the major factor predicting for relapse and disease-free survival and had an adverse effect on event-free survival and overall survival. However, the clinical impact of an FLT3/ITD may also vary between different cytogenetic entities [19] . Another retrospective analysis has revealed that FLT3/ITD adversely affected the outcome of hematopoietic transplantation in the same direction it does after chemotherapy [20] . FLT3/ITD led to increased relapse incidence and was associated with decreased leukemia-free survival. ●●C-KIT (CD117)

Another genetic mutation of RTKs reported is C-KIT (CD117) [21] . C-KIT is expressed in the progenitors of mast cells and is promoted by stem cell factor to regulate hematopoietic stem cells

future science group

and derived mast cells [22,23] . Three major point mutations have been reported including Val560Gly, Asp816-Val and Asp816-Tyr, all of which occur in intracellular TKD of C-KIT [24–26] . There is also a report of exon 8 in-frame deletion plus insertion mutations that involve the loss or replacement of Asp419, a highly conserved codon located in the extracellular domain [27] . The activating KIT mutation in codon 816 is a character of systemic mastocytosis [25,28] . The constitutive activation of KIT kinase plays a pivotal role in the pathogenesis, diagnosis and target therapy of systemic mastocytosis [29] . The activating mutations of C-KIT represent the most frequently mutated target in adult core binding factor AML and indicate higher risk and shorter survival [30,31] . ●●C-FMS

The mutations of C-FMS, which is also named CSF1R are first reported by employing hybridization to allele-specific oligonucleotide probes in AML and myelodysplastic syndrome patients involving codon 969 and 301 [32,33] . However, other studies employing direct sequencing [34] or allele-specific restriction analysis [35] both failed to detect the mutations reported previously. Later Faisel et al. [36] presented mutations involving exons 6 and 9 without the definition of their

www.futuremedicine.com

661

Review  Zhu, Xiao, Wang, Fu, Zhao & Huang Table 1. Receptor protein tyrosine kinases relevant to hematopoietic neoplasms. Receptor PTKs RTK classes

Affected signaling pathways

Major cancer types

Gene alterations

PDGFR-α PDGFR-β C-FMS

III III III

PI3K, ERK1/2, STAT5 PI3K, STAT5, PLC-γ Not clear

CEL CML, CEL, myeloid neoplasm Leukemia cell lines

C-KIT

III

AKT, STAT5, ERK

FLT-3

III

RAS, STAT5, MAPK

Systemic mastocytosis, AML especially CBF-AML AML

FGFR-1 TRK-C AXL

IV III IX

PI3K/AKT, STAT5, PLC-γ, Notch pathway MAPK PI3K/AKT, ERK, NF-κB

SCLL AML CML, AML

Genetic rearrangement Genetic rearrangement Genetic rearrangement, point mutation Point mutations, deletions and overexpression internal tandem duplication, missense point mutation Genetic rearrangement Genetic rearrangement Overexpression

AML: Acute myeloid leukemia; CBF: Core binding factor; CEL: Chronic eosinophilic leukemia; CML: Chronic myeloid leukemia; FGFR: FGF receptor; PDGFR: PDGF receptor; PTK: Protein tyrosine kinase; SCLL: Stem cell leukemia–lymphoma syndrome.

pathological role in leukemogenesis. In addition, the fusion of RBM6 to CSF1R [37] and Y571D mutation [38] was detected in different cell lines. ●●PDGF receptors

Both PDGF receptors (PDGFRs) α and β are involved in mutations relevant to hematopoietic neoplasms. The genetic alteration of PDGFR-β was reported earlier than PDGFR-α with genetic rearrangement in chronic myelomonocytic leukemia with t(5;12) chromosomal translocation [39] . The role of this TEL/PDGFR-β fusion gene in leukemogenesis has been elucidated that the receptor kinase was constitutively activated by enforced dimerization of TEL [40] . Since then, several different fusion genes of PDGFR-β have been reported in succession [41–48] . The ligand binding of PDGFR-β initiates PI3, STAT5 and PLC-γ signal pathways that will ultimately influence gene expression and modulate cell proliferation [49,50] . In addition, the fusion protein is reported to influence apoptosis by antagonizing the effects of TEL and ICSBP on PTPN13 transcription, which increases Fap1-dependent Fas resistance, in a manner that is independent of tyrosine kinase activity [51] . A study of PDGFR-α revealed the fusion with FIP1L1 (Fip1-like1) generated by an interstitial deletion on chromosome 4q12 [52] . The rearrangement is mostly reported in chronic eosinophilic leukemia, which is usually presented as idiopathic hypereosinophilic syndrome, eosinophilia-associated AML and lymphoblastic T-cell lymphoma [53] . The fusion product functions as constitutively activated tyrosine kinase and cooperates with IL-5-dependent signaling in driving abnormal eosinophil development evidenced

662

Future Oncol. (2015) 11(4)

in a mouse model [54] . The study of FIP1L1PDFGR-α expressed in human hematopoietic progenitors indicated a myeloproliferative phenotype via activation of multiple signaling molecules including PI3K, ERK1/2 and STAT5 [55] . Several different types of genetic rearrangements related to PDFGR-α have also been reported [56–59] . ●●FGFR-1

FGFR1, the somatic mutations of which have been specifically associated with stem cell leukemia-lymphoma (SCLL) syndrome that is also known as the 8p11 myeloproliferative syndrome, is one of the FGFR protein family members [60] . Varieties of genes fused to FGFR1 have been reported [61–65] . ZNF198-FGFR1, the most commonly reported rearrangement, induces a myeloproliferative neoplasm (MPN) phenotype in mice as well as activation of the downstream effector molecules PLC-γ, STAT5 and PI3K/AKT via expression in hematopoietic stem cell [66] . In addition, constitutive activation of Notch pathway is also reported to be involved in the etiology of SCLL recently [67] . ●●AXL

The RTK AXL was initially reported with oncogenic potential in chronic myeloid leukemia (CML) and chronic myeloproliferative disease [68] . AXL is overexpressed in AML associated with a poor prognosis and drug resistance by the binding with Gas6, which initiates activation of PI3K/AKT, ERK and NF-κB [69,70] . A recent study identified AXL as a novel PTK in B-cell chronic lymphocytic leukemia that appeared to function as a docking site for multiple non-RTKs and drive leukemic cell survival signals [71] .

future science group

Mutations in tyrosine kinase & tyrosine phosphatase  ●●Eph receptors

The Eph receptors, the largest family of tyrosine kinases, were named after an erythropoietin-producing human hepatocellular carcinoma cell line, from which the first Eph receptor was identified in vitro [72] . Unlike other tyrosine kinases, Eph receptors exhibit the unique way of bidirectional signaling thus playing opposite roles in cellular progress [73] . Eph receptors are quiescent in normal postembryonic tissues thus they are used as markers of malignancies. Eph receptors often play conflicting roles, either as oncogenes or as tumor suppressors, in many different malignancies including solid tumors and hematological neoplasms [74] . Among these receptors, EPHA3 is aberrantly expressed in almost all types of hematological malignancies [75,76] . Low expression of EPHA3 was detected in patients with CML in the chronic phase. However, the expression markedly elevated in patients in the accelerated or blastic phases [77] . The evidence that EPHA3 was expressed on CD34 + CD38-CD123 + cells, which represented a population of leukemia stem cells, while less commonly expressed on other leukemic cell fractions has proposed EPHA3 as a novel target for leukemia stem cells [78] . ●●Anaplastic lymphoma kinase

The RTK, anaplastic lymphoma kinase (ALK), is expressed specifically in the nervous system. Genetic alterations of ALK have been reported in anaplastic large cell lymphoma (ALCL) [79,80] . The translocation creating a fusion gene comprising ALK and nucleophosmin is associated with approximately 60% ALCL patients. Another fusion gene comprising ALK and TPM3 was detected in a smaller fraction of ALCL patients. ●●Non-RTKs

The genetic alterations of non-RTKs involved in hematopoietic neoplasms have been recognized for years. Genetic rearrangement and overexpression of nRTKs are among the most common

Review

alterations. The most comprehensively explored is the fusion of B-cell receptor (BCR) to ABL1, which is detected in all patients with CML and in a significant fraction of Ph-positive patients with acute lymphocytic leukemia [81] . Several variants of the fusion protein have been reported [82] . The other kinase involved in genetic rearrangement is ARG (also named ABL-related gene or ABL2) [83,84] . The genetic arrangement of JAK2 was first detected in a T-cell childhood acute lymphoblastic leukemia patient with t(9;12)(p24;p13) chromosomal translocation [85] , followed by other reports in myeloid leukemia [86] , CML [87] and acute erythroid leukemia [88] . Additionally, a point mutation in JAK2 was discovered in the majority of Philadelphia chromosome-negative MPN patients [89] . This mutation, JAK2V617F, was found in patients with polycythemia vera, essential thrombocythemia and primary myelofibrosis [90] . JAK2V617F induced constitutive activation of downstream signaling through the JAK-STAT, the MAPK and the PI3K/Akt pathways that finally promoted proliferation of affected cells [91] . Genetic alterations in protein tyrosine phosphatases in hematologic malignancies On the contrary, PTPs are a group of enzymes that remove phosphate groups from phosphorylated tyrosine residues on proteins, involved in signal transduction pathways and cellular functions [92] . They are grouped into four families based on the amino acid sequences of their catalytic domains [93] . The most frequently explored are the class I cysteine-based PTPs that comprise classical PTPs, which include receptor PTPs, nonreceptor PTPs and the dual-specific protein phosphatases (DUSPs) characterized with both serine-/threonine- and tyrosine-specific protein phosphatase activities. Genetic alterations of PTPs, which are relevant to hematopoietic neoplasms, are all in class I family (Table 3) .

Table 2. Nonreceptor protein tyrosine kinases relevant to hematopoietic neoplasms. Nonreceptor PTKs

Major cancer types

Gene alterations

LCK ABL1 ARG JAK2

Lymphoblastic leukemia CML, Ph+ ALL AML ALL, CML, AEL

Genetic rearrangement, overexpression Genetic rearrangement Genetic rearrangement Genetic rearrangement, point mutation

AEL: Acute eosinophilic leukemia; ALL: Acute lymphoblastic leukemia; AML: Acute myeloid leukemia; CML: Chronic myeloid leukemia; PTK: Protein tyrosine kinase.

future science group

www.futuremedicine.com

663

Review  Zhu, Xiao, Wang, Fu, Zhao & Huang ●●Nonreceptor tyrosine protein tyrosine

phosphatases

Somatic mutations of PTPN11, which encodes the PTP SHP-2, have been detected mostly in juvenile myelomonocytic leukemia, however, relatively rare in myelodysplastic syndromes and AML [94,95] . Leukemia-associated PTPN11 mutations are missense and are predicted to result in a gain-of-function role of these mutations on SHP-2 catalytic activity by molecular modeling and functional data analysis. Evidence has been found that leukemia-associated PTPN11 mutations induce Ras-ERK pathway, which finally contributes to leukemogenesis [96,97] . A report of focal deletions detected in PTPN2 in a subset of patients with T-cell acute lymphoblastic leukemia provides genetic evidence for a putative tumor suppressor gene of PTPN2 in T-ALL [98] . The overexpression of PTPN22 in CLL is recently reported, which significantly inhibits antigen-induced apoptosis of primary CLL cells by blocking BCR signaling pathways that negatively regulates lymphocyte survival as well as positively regulates the antiapoptotic AKT kinase that provides a powerful survival signal [99] . PTPN7, also named hematopoietic

tyrosine phosphatase (HePTP), was first identified to be expressed exclusively in thymus and spleen [100] . The amplification and overexpression of PTPN7, with the ability to transform NIH 3T3 cells, were suggested as an important cofactor contributing to abnormal myeloid cell growth [101] . The MAP kinase ERK2 is defined as a specific substrate of PTPN7. However, the detailed signal pathway in leukemogenesis has not been defined yet. Interestingly, another study has reported lower expression of PTPN7 in children B-cell lymphoma, which may imply the potential functions of PTPN7 as both oncogene and tumor suppression gene [102] . ●●Receptor protein tyrosine phosphatases

& DUSPs

As to receptor PTPs, the hypermethylation and silencing of PTPRG [103] and PTPRO [104] were detected in cutaneous T-cell lymphoma and chronic lymphocytic leukemia, respectively. It has been elucidated that suppression of PTPRO by promoter methylation could contribute to the augmented phosphorylation and constitutive activity of its substrate BCR/ABL1 [105] . Loss of heterozygosity has been reported to be related

Table 3. Protein tyrosine phosphatases relevant to hematopoietic neoplasms. PTPs

Affected signaling pathways

Major cancer types

Gene alterations

PTPN11

Ras pathway

PTPN13

FAS resistance

PTPN22

AKT, BCR signaling

T-ALL Lymphoma leukemia Leukemia lymphoma Juvenile myelomonocytic leukemia, MDS, AML Multiple lymphoma CLL

Focal deletion Methylation

PTPN7

Not clear Jak/STAT, cytokine signaling pathway Not clear

Cutaneous T-cell lymphoma Lymphoma

Methylation

CLL Non-Hodgkin’s lymphoma

Methylation LOH

Nonreceptor PTPs PTPN2 PTPN6

Amplification downregulation Mutation

Methylation Overexpression

Receptor PTPs PTPRG

Not clear

PTPRK

β-catenin/E-cadherin complexes Bcr-abl1 EGFR endocytosis, dephosphorylation of PDGFR-β

PTPRO PTPRJ

LOH

AML: Acute myeloid leukemia; CLL: Chronic lymphoblastic leukemia; LOH: Loss of heterozygosity; MDS: Myelodysplastic syndrome; PTP: Protein tyrosine phosphatase; T-ALL: T-cell acute lymphoblastic leukemia.

664

Future Oncol. (2015) 11(4)

future science group

Mutations in tyrosine kinase & tyrosine phosphatase  to PTPRK [106] and PTPRJ [107] in lymphoma. Both are presented as putative tumor suppressor genes [108,109] , the inactivation of which induced by loss of heterozygosity, plays roles in neoplasia. The overexpression of PRL-3 [110] , DUSP7 [111] and MTMR6 [112] has been detected in myeloma and leukemia. On the other hand, the downregulation of DUSP2 (PAC-1), which induces constitutive activation of ERK, has been reported in acute leukemia [113] . Novel therapeutic targets to PTKs & PTPs in hematologic malignancies Since the constitutive activation of PTKs or the downstream signal pathways is the common mechanism in neoplasia, drugs inhibiting tyrosine kinases seem to be powerful in clinical therapy. Efforts have been made in exploring TKIs, which have been applied in treatment of hematologic malignancies and have got promising efficacy (Table 4) . ●●BCR-ABL1 inhibitors

Imatinib, which is among the most impressive inhibitors, targets leukemia cells by preventing BCR-ABL1 from phosphorylating subsequent proteins that initiate the signaling pathway necessary for development of disease. The positive detection of BCR-ABL1 in CML cells, which is however absent in normal blood cells, gives opportunities to the impressive effectiveness of imatinib. However, nearly 20% of patients could not achieve a complete cytogenetic response with others acquiring intolerable side effects or drug resistance. The resistance to imatinib prompted the study of mechanism involved and novel approaches to handle the resistance. Studies have provided evidence that the resistance is often associated with the reactivation of BCR-ABL1 signal transduction mainly due to mutations in the kinase domain [114] . Later, evolving generations of inhibitors that own higher potency have been designed including nilotinib and dasatinib [115] . Clinical trials have provided evidence that both nilotinib and dasatinib induce hematologic and cytogenetic responses in patients with CML or Ph-positive ALL resistant to imatinib [116,117] . A Phase III, randomized, open-label, multicenter clinical trial revealed that nilotinib was superior to imatinib in patients with newly diagnosed chronic-phase Ph-positive CML [118] . Similar studies investigating dasatinib also underlined the high efficacy and safety for the first-line treatment of chronic-phase CML [119] . Notably,

future science group

Review

another orally bioavailable dual Src/Abl1 inhibitor, 200-times as potent as imatinib, with minimal inhibitory activity against PDGFR or C-KIT, bosutinib is demonstrated to be active in chronic-phase CML after imatinib and dasatinib and/or nilotinib therapy failure [120] . These novel inhibitors have brought the management of CML patients into a hopeful era. Despite the promising clinical results of advanced TKIs overriding most resistance, none could effectively target the gatekeeper mutation, T315I, which disrupts the binding of imatinib to the ATP-binding pocket of ABL1 [121] . The next generation of TKIs make efforts on overriding the T315I mutation, which may provide opportunities for relapsed patients carrying this mutation. The pan-BCR-ABL1 inhibitor ponatinib has achieved high efficacy in heavily pretreated patients with Ph-positive leukemia with resistance to TKIs, including patients with the BCR-ABL1 T315I mutation, other mutations or no mutations [122] . A recent Phase II trial revealed promising response rates regardless of disease stage or mutation status [123] . Other inhibitors, including DCC-2036 [124] and HG-7-85-01 [125] , are also in research. ●●FLT3 inhibitors

FLT3 inhibitors, another category of comparable TKIs that have been explored passionately currently, mostly inhibit FLT3 activity by competing with ATP for binding to the ATP-binding pocket of the TKD [126] . More than 20 small molecule inhibitors against FLT3 have been reported, eight of which have been evaluated in clinical trials, including tandutinib, sorafenib, sunitinib, midostaurin, lestaurtinib, KW-2449, quizartinib and crenolanib (Table 5) . These clinical trials have demonstrated the potential efficacy of FLT3 inhibitors in AML patients. However, these agents are not specified as first-line treatment yet. Most responses are limited to hematological improvement with no more than 40% complete remission while applying alone. Furthermore, these inhibitors show efficacy in patients without FLT3 mutations, the majority of which is restricted to reduction in blast counts, which is supposed as results of the multikinase activities. Although clinical trials have revealed the initial responses of FLT3 inhibitors in FLT3ITD-positive patients, or even negative patients, subsequent relapse often occurs due to acquisition of secondary FLT3 TKD mutations, mainly

www.futuremedicine.com

665

Review  Zhu, Xiao, Wang, Fu, Zhao & Huang Table 4. Therapeutic targets to protein tyrosine kinases. Tyrosine kinase inhibitors

Target genes

Bosutinib Crizotinib Crenolanib Dasatinib Dovitinib Imatinib Nilotinib Pazopanib Ponatinib Quizartinib Regorafenib Ruxolitinib Sunitinib Sorafenib Tandutinib Vandetanib

SRC-ABL1 ALK FLT3 ABL1, PDGFR,KIT FGFR ABL1, PDGFR, KIT,FLT3 ABL1, PDGFR,KIT PDGFR,KIT FLT3, PDGFR, FGFR,BCR-ABL1 FLT3, PDGFR,KIT PDGFR,KIT JAK2 KIT, PDGFR,C-FMS,FLT3 PDGFR, KIT,FLT3 PDGFR, KIT,FLT3 FGFR

FGFR: FGF receptor; PDGFR: PDGF receptor.

reported at residues D835 and F691, which is paralleled to what happened to BCR-ABL1 inhibitors [137] . Despite the relatively low CR rate, the emergence of relapses has appeared to be a more serious problem. Previous studies have shown that the addition of chemotherapy failed to improve the relapses since no prolonged survival was observed [138] . The emergence of TKD mutations suggests that novel FLT3 inhibitors that harbor the ability to suppress both ITD and TKD mutations may be more powerful. Crenolanib has been demonstrated as a potent inhibitor targeting D835 point mutations both in vitro and in vivo with an ongoing Phase II clinical trial [136] . Interestingly, the study of Zirm et al. revealed that ponatinib, a multikinase inhibitor, effectively induced apoptosis of not only the parental FLT3-ITD cell line but also the murine myeloid cells stably transfected with FLT3-TKD N676D, F691I and G697R mutations [139] , which provided a clue to figure out more candidates against FLT3-ITD and TKD mutations by high-throughput screening. Other strategies to explore efficacious inhibitors focus on targeting FLT3-dependent pathways. For instance, CCT137690, a dual FLT3-Aurora inhibitor, successfully inhibited the growth of FLT3-ITD-D835Y cells that were resistant to AC220 and sorafenib [140] . ●●JAK inhibitors

Due to the discovery of activating mutations in JAK2 and dysregulation of JAK-STAT pathway in hematological malignancies, development of

666

Future Oncol. (2015) 11(4)

JAK inhibitors has been promoted. There are several JAK inhibitors in different stages of clinical trials. Among these inhibitors, ruxolitinib, which was demonstrated to have effects on suppressing hematopoiesis, decreasing inflammatory cytokine levels and relieving splenomegaly, has been approved by the US FDA for treatment of splenomegaly in patients with intermediateand high-risk MPN-associated myelofibrosis (staging system unspecified), and by the EMA for treatment of splenomegaly and/or symptoms in patients with MPN-associated myelofibrosis for all disease stages [141] . However, there were no significant effect on improvement of hematologic parameters or reducing burden of JAK2V617F allele. The predominant effect of ruxolitinib in myelofibrosis was not specific for the neoplastic clones [142] . Studies have reported better survival in patients with MPN-associated myelofibrosis treated with ruxolitinib compared with the best available therapy [143] . Other inhibitors including SAR302503, CYT387, lestaurtinib and pacritinib are also in study. However, the benefits of therapy with JAK inhibitors in MPN are moderate due to the fact that the effects of JAK inhibitors are unrelated to mutation status. The combination with various targeting agents for intervention of multiple levels of the signaling pathway may offer better outcomes. Future studies are needed to better understand the role of JAK mutations and associated downstream signal pathways in order to figure out more effective therapy for further clinical benefits.

future science group

Mutations in tyrosine kinase & tyrosine phosphatase  ●●PTP inhibitors

However, the developing inhibitors of PTPs seem to get frozen up. Although numerous compounds including natural products, small molecules and silencing RNAs are under development [144] , efforts are still needed to relate these compounds to clinical application. Among the drugs currently used that have been reported with PTP inhibitory activity, sodium stibogluconate, which was reported able to inhibit SHP-1, SHP-2 and augment antitumor actions of IFN-α2b [145] , combined with other anticancer agents, has been explored in clinical trials of solid tumors [146,147] . Despite that PTPs have been revealed as putative oncogenes or tumor suppressor genes, the inhibitors of PTPs applied in the treatment of cancer are rarely reported. The less understood mechanisms of PTPs in neoplasia may be one of the reasons, with the problem of effective drug development as another. Excitingly, a novel potent and selective TC-PTP (also known as PTPN2) inhibitor has been developed, which has proposed a new approach for potent, highly selective and cell permeable PTP inhibitory

Review

agents [148] . Since the substrates and downstream regulatory mechanisms have not yet been clarified thoroughly, the inhibitors of PTPs are still far away from clinical therapy. However, one thing we can confirm is that there must be a great potential and clinical application prospect waiting for exploration, which may be a similar situation as the initial study of TKIs. Conclusion & future perspective As mentioned above, both PTKs and PTPs play pivotal roles in regulation of signal pathways, which partly decide cellular fate. Due to the important status in cellular functions, the genetic alterations of PTKs or PTPs are among the reasons of carcinogenesis. As the detecting techniques evolve, the majority have been reported nowadays. We present the alterations relevant to hematologic malignancies including mutation, overexpression, deletion, rearrangement and methylation that have been covered. The types of mutation, relevance to clinical diseases and impact on prognosis have been discussed detailedly. We have also discussed the potential therapeutic targets of PTKs and PTPs.

Table 5. The clinical trials of FLT3 inhibitors. Study (year)

Inhibitor

Phase

Dose

Drug combination

Total patients

ITD

TKD mutation

Outcome

Ref.

DeAngelo et al. Tandutinib (2006) Ravandi et al. Sorafenib (2013) Inaba et al.   (2011)

I

150–700 mg twice daily 400 mg twice daily

None

40

8

1

[127]

5-azacytidine

43

40

0

12

5

0

Fiedler et al.(2005)

Sunitinib

I

15

2

2

CRi: 25%; PR: 75%

[130]

Stone et al. (2012)

Midostaurin

Ib

40

9

4

CR: 80%

[131]

Fischer et al. (2010)

 

IIb

Clofarabine and cytarabine 50–75 mg daily, days None 1–28; 80 mg twice daily, days 29–58 50 mg twice daily Daunorubicin and cytarabine 50 or 100 mg twice None daily

Antileukemic effect: 2 of 8 CRi: 27%, CR: 16%, PR: 3% CRi: 17%, CR: 50% PR: 8%

26

9

HI: 39%, PR: 1%, BR: 53%

[132]

Knapper et al. (2006) Pratz et al. (2009) Cortes et al. (2013)  Galanis et al. (2014) 

Lestaurtinib

II

2

3

BR: 30%

[133]

KW-2449

I

Quizartinib Crenolanib

II I

200 mg, 150 mg twice daily

[128] [129]

None None

11

11

0

BR: 45%

[134]

I

60–80 mg twice daily 25–500 mg twice daily 12–450 mg daily

92 (including MDS) 27

None

76

17

0

CR (of any type): 13%, PR: 17%

[135]

II

100 mg every 8 h

None

Not available yet

[136]

BR: Blast response; CR: Complete remission; CRi: CR with incomplete hematological recovery; HI: Hematological improvement; ITD: Internal tandem duplication; MDS: Myelodysplastic syndrome; PR: Partial remission; TKD: Tyrosine kinase domain.

future science group

www.futuremedicine.com

667

Review  Zhu, Xiao, Wang, Fu, Zhao & Huang The inhibitors of BCR-ABL1 and FLT3 have been employed clinically for years with inspiring prospects. As expected, the appearance of resistance has limited the scope of application, however, has prompted the evolution of novel targeted drugs. Despite that both BCR-ABL1 and FLT3 inhibitors have shown bright prospects although resistance still exists, the majority of potential therapeutic targets have not been explored yet. Experts have made efforts on clarifying the pivotal roles of different PTKs or PTPs. Much of the information has been uncovered on their oncogenic properties, which could confirm their potential as targets in clinical practice. Although the benefits of inhibiting oncogenic enzymes have been mainly discussed, efforts are encouraged to make on exploring particular drugs owning the ability to activate those enzymes that have putative tumor suppressor functions. There are still unknown mutations or other alterations unexplored. With several decades of research, the remaining challenge refers to the biological roles of PTKs and PTPs in positive or inhibitory functions during the development of neoplasms. Identifying the biological context and signal network of PTKs and PTPs is

essential for future directions, as well as uncovering the disease-related mutations, identifying their substrates and characterizing other pharmacologically relevant pathways. The distance of biological research to clinical application is shortening by the identification of regulatory mechanisms and their potential relevance to cancer. Certainly, exciting new generation of drugs will finally enrich the antitumor therapies. Financial & competing interests disclosure This work was funded in part by the Key Project of the National Natural Science Foundation of China (81230014), the National High Technology Research and Development Program of China (2012AA020905), the National Natural Science Foundation of China (81100387, 81170501), the Major Technology Program (Key Social Development) of the Science Technology Department of Zhejiang Province (2012C13021-1) and the Major Social Development Program of Science Technology Department of Jinhua(2011-3-004). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.

EXECUTIVE SUMMARY Protein tyrosine kinases & protein tyrosine phosphatases ●●

Protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) are both important regulators of signal

transduction in eukaryotic cells. Dysregulation of protein tyrosine phosphorylation caused by genetic alterations of PTKs or PTPs has been revealed to be involved in the pathological process of cancer. Genetic alterations in PTKs in hematologic malignancies ●●

PTKs include receptor tyrosine kinases and nonreceptor tyrosine kinases. Genetic alterations of PTKs have been reported to be associated with hematopoietic malignancies and be valuable in predicting the prognosis.

Genetic alterations in PTPs in hematologic malignancies ●●

Genetic alterations of PTPs that are relevant to hematopoietic neoplasms are all in Class I family, including receptor PTPs, nonreceptor PTPs and the dual-specific protein phosphatases.

Novel therapeutic targets to PTKs & PTPs in hematologic malignancies ●●

Tyrosine kinase inhibitors, such as BCR-ABL1 inhibitors and FLT3 inhibitors, have been applied in treatment of

hematologic malignancies and have shown promising efficacy. However, the inhibitors of PTPs applied in the treatment of cancer are rarely reported. Conclusion & future perspective ●●

The mutations of PTKs and PTPs related to hematologic malignancies have impacts on the carcinogenesis and prognosis. Novel targeted drugs to PTKs and PTPs are promising therapeutic strategies.

●●

Identifying the biological signal network of PTKs and PTPs, uncovering the disease-related mutations and

characterizing other pharmacologically relevant pathways are the remaining challenges, which will provide clues to exploration of target therapies.

668

Future Oncol. (2015) 11(4)

future science group

Mutations in tyrosine kinase & tyrosine phosphatase  References Papers of special note have been highlighted as: • of interest 1

Burnett G, Kennedy EP. The enzymatic phosphorylation of proteins. J. Biol. Chem. 211(2), 969–980 (1954).



The first description of enzymatic phosphorylation of proteins.

2

Eckhart W, Hutchinson MA, Hunter T. An activity phosphorylating tyrosine in polyoma T antigen immunoprecipitates. Cell 18(4), 925–933 (1979).

3

Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 100(1), 57–70 (2000).

4

Jang SH. Long term therapeutic plan for patients with non-small cell lung cancer harboring EGFR mutation. Tuberc. Respir. Dis. (Seoul) 76(1), 8–14 (2014).

5

Davoli A, Hocevar BA, Brown TL. Progression and treatment of HER2-positive breast cancer. Cancer Chemother. Pharmacol. 65(4), 611–623 (2010).

preferentially seen in acute myeloid leukemia and myelodysplastic syndrome among various hematological malignancies: a study on a large series of patients and cell lines. Leukemia 11(10), 1605–1609 (1997). 14 Yamamoto Y, Kiyoi H, Nakano Y et al.

Activating mutation of D835 within the activation loop of FLT3 in human hemato­ logic malignancies. Blood 97(8), 2434–2439 (2001). 15 Thiede C, Steudel C, Mohr B et al. Analysis

of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood 99(12), 4326–4335 (2002). 16 Mizuki M, Fenski R, Halfter H et al. Flt3

mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways. Blood 96(12), 3907–3914 (2000). 17 Hayakawa F, Towatari M, Kiyoi H et al.

Tandem-duplicated Flt3 constitutively activates STAT5 and MAP kinase and introduces autonomous cell growth in IL-3-dependent cell lines. Oncogene 19(5), 624–631 (2000).

6

Overton LC, Heinrich MC. Regorafenib for treatment of advanced gastrointestinal stromal tumors. Expert Opin. Pharmacother. 15(4), 549–558 (2014).

7

Pfeifer H, Wassmann B, Pavlova A et al. Kinase domain mutations of BCR-ABL frequently precede imatinib-based therapy and give rise to relapse in patients with de novo Philadelphia-positive acute lympho­ blastic leukemia (Ph+ ALL). Blood 110(2), 727–734 (2007).

18 Mead AJ, Linch DC, Hills RK et al. FLT3

Novellino L, De Filippo A, Deho P et al. PTPRK negatively regulates transcriptional activity of wild type and mutated oncogenic beta-catenin and affects membrane distribution of beta-catenin/E-cadherin complexes in cancer cells. Cell. Signal. 20(5), 872–883 (2008).

19 Kottaridis PD, Gale RE, Frew ME et al. The

8

9

Hubbard SR, Till JH. Protein tyrosine kinase structure and function. Annu. Rev. Biochem. 69, 373–398 (2000).

10 Robinson DR, Wu YM, Lin SF. The protein

tyrosine kinase family of the human genome. Oncogene 19(49), 5548–5557 (2000). 11 Filippakopoulos P, Müller S, Knapp S. SH2

domains: modulators of nonreceptor tyrosine kinase activity. Curr. Opin. Struct. Biol. 19(6), 643–649 (2009). 12 Birg F, Courcoul M, Rosnet O et al.

Expression of the FMS/KIT-like gene FLT3 in human acute leukemias of the myeloid and lymphoid lineages. Blood 80(10), 2584–2593 (1992). 13 Yokota S, Kiyoi H, Nakao M et al. Internal

tandem duplication of the FLT3 gene is

future science group

tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia. Blood 110(4), 1262–1270 (2007). presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML10 and 12 trials. Blood 98(6), 1752–1759 (2001). 20 Brunet S, Labopin M, Esteve J et al. Impact

of FLT3 internal tandem duplication on the outcome of related and unrelated hemato­ poietic transplantation for adult acute myeloid leukemia in first remission: a retrospective analysis. J. Clin. Oncol. 30(7), 735–741 (2012). 21 Sperr WR, Walchshofer S, Horny HP et al.

Systemic mastocytosis associated with acute myeloid leukaemia: report of two cases and detection of the c-kit mutation Asp-816 to Val. Br. J. Haematol. 103(3), 740–749 (1998). 22 Ratajczak MZ, Luger SM, DeRiel K et al.

Role of the KIT protooncogene in normal and malignant human hematopoiesis. Proc. Natl Acad. Sci. USA 89(5), 1710–1714 (1992).

Review

23 Valent P, Spanblöchl E, Sperr WR et al.

Induction of differentiation of human mast cells from bone marrow and peripheral blood mononuclear cells by recombinant human stem cell factor/kit-ligand in long termculture. Blood 80(9), 2237–2245 (1992). 24 Furitsu T, Tsujimura T, Tono T et al.

Identification of mutations in the coding sequence of the protooncogene c-kit in a human mast cell leukemia cell line causing ligand independent activation of c-kit product. J. Clin. Invest. 92(4), 1736–1744 (1993). 25 Longley BJ, Tyrrell L, Lu SZ et al. Somatic

c-kit activating mutation in urticaria pigmentosa and aggressive mastocytosis: establishment of clonality in a human mast cell neoplasm. Nat. Genet. 12(3), 312–314 (1996). 26 Beghini A, Cairoli R, Morra E, Larizza L.

In vivo differentiation of mast cells from acute myeloid leukemia blasts carrying a novel activating ligand-independent C-kit mutation. Blood Cells Mol. Dis. 24(2), 262–270 (1998). 27 Gari M, Goodeve A, Wilson G et al. C-kit

proto-oncogene exon 8 in-frame deletion plus insertion mutations in acute myeloid leukaemia. Br. J. Haematol. 105(4), 894–900 (1999). 28 Longley BJ, Metcalfe DD, Tharp M et al.

Activating and dominant inactivating c-KIT catalytic domain mutations in distinct clinical forms of human mastocytosis. Proc. Natl Acad. Sci. USA. 96, 1609–1614 (1999). 29 Pardanani A. Systemic mastocytosis in adults:

2012 update on diagnosis, risk stratification, and management. Am. J. Hematol. 87(4), 401–411 (2012). 30 Beghini A, Ripamonti CB, Cairoli R et al.

KIT activating mutations: incidence in adult and pediatric acute myeloid leukemia, and identification of an internal tandem duplication. Haematologica 89(8), 920–925 (2004). 31 Schnittger S, Kohl TM, Haferlach T et al.

KIT-D816 mutations in AML1-ETO positive AML are associated with impaired event-free and overall survival. Blood 107(5), 1791–1799 (2006). 32 Ridge SA, Worwood M, Oscier D et al. FMS

mutations in myelodysplastic, leukemic, and normal subjects. Proc. Natl Acad. Sci. USA 87(4), 1377–1380 (1990). 33 Tobal K, Pagliuca A, Bhatt B et al. Mutation

of the human FMS gene (M-CSF receptor) in myelodysplastic syndromes and acute myeloid leukemia. Leukemia 4(7), 486–489 (1990). 34 Shepherd L, Cameron C, Graham C et al.

Direct sequencing of the N-ras, c-FMS and

www.futuremedicine.com

669

Review  Zhu, Xiao, Wang, Fu, Zhao & Huang p53 loci for transforming mutations in myelomonocytic leukaemia. Blood 76(Suppl. 1), 245a (1990). 35 Springall F, O’Mara S, Shounan Y et al. c-fms

point mutations in acute myeloid leukaemia: fact or fiction? Leukemia 7(7), 978–985 (1993). 36 Abu-Duhier FM, Goodeve AC, Wilson GA

et al. c-FMS mutational analysis in acute myeloid leukaemia. Br. J. Haematol. 123(4), 749–750 (2003). 37 Gu TL, Mercher T, Tyner JW et al. A novel

fusion of RBM6 to CSF1R in acute mega­k aryo­blastic leukemia. Blood 110(1), 323–333 (2007). 38 Chase A, Schultheis B, Kreil S et al. Imatinib

sensitivity as a consequence of a CSF1RY571D mutation and CSF1/CSF1R signaling abnormalities in the cell line GDM1. Leukemia 23(2), 358–364 (2009). 39 Golub TR, Barker GF, Lovett M,

Gilliland DG. Fusion of PDGF receptor b to a novel ets-like gene, tel, in chronic myelomonocytic leukaemia with t(5;12) chromosomal translocation. Cell 77(2), 307–316 (1994). 40 Carroll M, Tomasson MH, Barker GF et al.

The TEL/platelet-derived growth factor beta receptor (PDGF beta R) fusion in chronic myelomonocytic leukemia is a transforming protein that self-associates and activates PDGF beta R kinase-dependent signaling pathways. Proc. Natl Acad. Sci. USA 93(25), 14845–14850 (1996). 41 Abe A, Emi N, Tanimoto M et al. Fusion of

the platelet-derived growth factor receptor beta to a novel gene CEV14 in acute myelogenous leukemia after clonal evolution. Blood 90(11), 4271–4277 (1997). 42 Ross TS, Bernard OA, Berger R,

Gilliland DG. Fusion of Huntingtin interacting protein 1 to platelet-derived growth factor beta receptor (PDGFbetaR) in chronic myelomonocytic leukemia with t(5;7) (q33;q11.2). Blood 91(12), 4419–4426 (1998). 43 Magnusson MK, Meade KE, Brown KE et al.

Rabaptin-5 is a novel fusion partner to platelet-derived growth factor beta receptor in chronic myelomonocytic leukemia. Blood 98(8), 2518–2525 (2001). 44 Schwaller J, Anastasiadou E, Cain D et al.

H4(D10S170), a gene frequently rearranged in papillary thyroid carcinoma, is fused to the platelet-derived growth factor receptor beta gene in atypical chronic myeloid leukemia with t(5;10)(q33;q22). Blood 97(12), 3910–3918 (2001).

45 Vizmanos JL, Novo FJ, Román JP et al. NIN,

a gene encoding a CEP110-like centrosomal protein, is fused to PDGFRB in a patient with a t(5;14)(q33;q24) and an imatinibresponsive myeloproliferative disorder. Cancer Res. 64(8), 2673–2676 (2004). 46 Morerio C, Acquila M, Rosanda C et al.

HCMOGT-1 is a novel fusion partner to PDGFRB in juvenile myelomonocytic leukemia with t(5;17)(q33;p11.2). Cancer Res. 64(8), 2649–2651 (2004). 47 Grand FH, Burgstaller S, Kühr T et al.

p53-Binding protein 1 is fused to the platelet-derived growth factor receptor beta in a patient with a t(5;15)(q33;q22) and an imatinib-responsive eosinophilic myelo­ proliferative disorder. Cancer Res. 64(20), 7216–7219 (2004). 48 Levine RL, Wadleigh M, Sternberg DW et al.

KIAA1509 is a novel PDGFRB fusion partner in imatinib-responsive myeloproliferative disease associated with a t(5;14)(q33;q32). Leukemia 19(1), 27–30 (2005). 49 Dierov J, Xu Q, Dierova R, Carroll M.

TEL/platelet-derived growth factor receptor beta activates phosphatidylinositol 3 (PI3) kinase and requires PI3 kinase to regulate the cell cycle. Blood 99(5), 1758–1765 (2002). 50 Sternberg DW, Tomasson MH, Carroll M

et al. The TEL/PDGFβR fusion in chronic myelomonocytic leukemia signals through STAT5-dependent and STAT5-independent pathways. Blood 98(12), 3390–3397 (2001). 51 Huang W, Hu L, Bei L et al. The leukemia-

associated fusion protein Tel-platelet-derived growth factor receptor β (Tel-PdgfRβ) inhibits transcriptional repression of PTPN13 gene by interferon consensus sequence binding protein (Icsbp). J. Biol. Chem. 287(11), 8110–8125 (2012). 52 Cools J, DeAngelo DJ, Gotlib J et al.

A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereo­ sinophilic syndrome. N. Engl. J. Med. 348(13), 1201–1214 (2003). 53 Metzgeroth G, Walz C, Score J et al. Recurrent

finding of the FIP1L1–PDGFRA fusion gene in eosinophilia-associated acute myeloid leukemia and lymphoblastic T-cell lymphoma. Leukemia 21(6), 1183–1188 (2007). 54 Yamada Y, Rothenberg ME, Lee AW et al.

The FIP1L1–PDGFRA fusion gene cooperates with IL-5 to induce murine hypereosinophilic syndrome (HES)/chronic eosinophilic leukemia (CEL)-like disease. Blood 107(10), 4071–4079 (2006). 55 Buitenhuis M, Verhagen LP, Cools J, Coffer PJ.

Molecular mechanisms underlying

670

Future Oncol. (2015) 11(4)

FIP1L1–PDGFRA-mediated myelo­proliferation. Cancer Res. 67(8), 3759–3766 (2007). 56 Baxter EJ, Hochhaus A, Bolufer P et al. The

t(4;22)(q12;q11) in atypical chronic myeloid leukaemia fuses BCR to PDGFRA. Hum. Mol. Genet. 11(12), 1391–1397 (2002). 57 Walz C, Curtis C, Schnittger S et al.

Transient response to imatinib in a chronic eosinophilic leukemia associated with ins(9;4) (q33;q12q25) and a CDK5RAP2–PDGFRA fusion gene. Genes Chromosomes Cancer 45(10), 950–956 (2006). 58 Score J, Curtis C, Waghorn K et al.

Identification of a novel imatinib responsive KIF5B–PDGFRA fusion gene following screening for PDGFRA overexpression in patients with hypereosinophilia. Leukemia 20(5), 827–832 (2006). 59 Curtis CE, Grand FH, Musto P et al. Two

novel imatinib-responsive PDGFRA fusion genes in chronic eosinophilic leukaemia. Br. J. Haematol. 138(1), 77–81 (2007). 60 Xiao S, Nalabolu SR, Aster JC et al. FGFR1 is

fused with a novel zinc-finger gene, ZNF198, in the t(8;13) leukaemia/lymphoma syndrome. Nat. Genet. 18(1), 84–87 (1998). 61 Popovici C, Zhang B, Grégoire MJ et al. The

t(6;8)(q27;p11) translocation in a stem cell myeloproliferative disorder fuses a novel gene, FOP, to fibroblast growth factor receptor 1. Blood 93(4), 1381–1389 (1999). 62 Guasch G, Mack GJ, Popovici C et al. FGFR1

is fused to the centrosome-associated proteinCEP110 in the 8p12 stem cell myeloproliferative disorder with t(8;9) (p12;q33). Blood 95(5), 1788–1796 (2000). 63 Grand EK, Grand FH, Chase AJ et al.

Identification of a novel gene, FGFR1OP2, fused to FGFR1 in 8p11 myeloproliferative syndrome. Genes Chromosomes Cancer 40(1), 78–83 (2004). 64 Walz C, Chase A, Schoch C et al. The t(8;17)

(p11;q23) in the 8p11 myeloproliferative syndrome fuses MYO18A to FGFR1. Leukemia 19(6), 1005–1009 (2005). 65 Belloni E, Trubia M, Gasparini P et al. 8p11

myeloproliferative syndrome with a novel t(7;8) translocation leading to fusion of the FGFR1 and TIF1 genes. Genes Chromosomes Cancer 42(3), 320–325 (2005). 66 Chen J, Deangelo DJ, Kutok JL et al. PKC412

inhibits the zinc finger 198-fibroblast growth factor receptor 1 fusion tyrosine kinase and is active in treatment of stem cell myelo­ proliferative disorder. Proc. Natl Acad. Sci. USA 101(40), 14479–14484 (2004). 67 Ren M, Cowell JK. Constitutive Notch

pathway activation in murine ZMYM2-

future science group

Mutations in tyrosine kinase & tyrosine phosphatase  FGFR1-induced T-cell lymphomas associated with atypical myeloproliferative disease. Blood 117(25), 6837–6847 (2011). 68 Janssen JW, Schulz AS, Steenvoorden AC

et al. A novel putative tyrosine kinase receptor with oncogenic potential. Oncogene 6(11), 2113–2120 (1991). 69 Rochlitz C, Lohri A, Bacchi M et al. Axl

expression is associated with adverse prognosis and with expression of Bcl-2 and CD34 in de novo acute myeloid leukemia (AML): results from a multicenter trial of the Swiss Group for Clinical Cancer Research (SAKK). Leukemia 13(9), 1352–1358 (1999). 70 Hong CC, Lay JD, Huang JS et al. Receptor

tyrosine kinase AXL is induced by chemo­ therapy drugs and overexpression of AXL confers drug resistance in acute myeloid leukemia. Cancer Lett. 268(2), 314–324 (2008). 71 Ghosh AK, Secreto C, Boysen J et al. The

novel receptor tyrosine kinase Axl is constitutively active in B-cell chronic lymphocytic leukemia and acts as a docking site of nonreceptor kinases: implications for therapy. Blood 117(6), 1928–1937 (2011). 72 Hirai H, Maru Y, Hagiwara K et al. A novel

putative tyrosine kinase receptor encoded by the eph gene. Science 238(4834), 1717–1720 (1987). 73 Pasquale EB. Eph-ephrin bidirectional

signaling in physiology and disease. Cell 133(1), 38–52 (2008). 74 Haldimann M, Custer D, Munarini N et al.

Deregulated ephrin-B2 expression in the mammary gland interferes with the development of both the glandular epithelium and vasculature and promotes metastasis formation. Int. J. Oncol. 35(3), 525–536 (2009). 75 Guan M, Liu L, Zhao X et al. Copy number

variations of EphA3 are associated with multiple types of hematologic malignancies. Clin. Lymphoma Myeloma. Leuk. 11(1), 50–53 (2011). 76 Shapiro VS, Mollenauer MN, Weiss A.

Endogenous CD28 expressed on myeloma. cells up-regulates interleukin-8 production: implications for multiple myeloma. progression. Blood 98(1), 187–193 (2001). 77 Nair JR, Carlson LM, Koorella C et al. CD28

expressed on malignant plasma cells. induces a prosurvival and immunosuppressive microenvironment. J. Immunol. 187(3), 1243–1253 (2011). 78 Palath V, Vekhande R, Baer M et al. A

recombinant antibody to EphA3 with pro-apoptotic and enhanced ADCC activity

future science group

against various hematologic malignancies shows selective inhibition of colony formation from long-term culture-initiating cells (LTC-ICs) in primary leukemia samples. Blood (ASH Annual Meeting Abstracts) 116, 2897 (2010). 79 Morris SW, Kirstein MN, Valentine MB et al.

Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin’s lymphoma. Science 263(5151), 1281–1284 (1994). 80 Duyster J, Bai RY, Morris SW. Translocations

involving anaplastic lymphoma kinase (ALK). Oncogene 20(40), 5623–5637 (2001). 81 Druker BJ, Sawyers CL, Kantarjian H 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. 344(14), 1038–1042 (2001). 82 Melo JV. The diversity of BCR-ABL fusion

proteins and their relationship to leukemia phenotype. Blood 88(7), 2375–2384 (1996). 83 Cazzaniga G, Tosi S, Aloisi A et al. The

tyrosine kinase abl-related gene ARG is fused to ETV6 in an AML-M4Eo patient with a t(1;12)(q25;p13): molecular cloning of both reciprocal transcripts. Blood 94(12), 4370–4373 (1999). 84 Iijima Y, Ito T, Oikawa T et al. A new ETV6/

TEL partner gene, ARG (ABL-related gene or ABL2), identified in an AML-M3 cell line with a t(1;12)(q25;p13) translocation. Blood 95(6), 2126–2131 (2000). 85 Lacronique V, Boureux A, Valle VD et al.

A TEL-JAK2 fusion protein with constitutive kinase activity in human leukemia. Science 278(5341), 1309–1312 (1997). 86 Peeters P, Raynaud SD, Cools J et al. Fusion

of TEL, the ETS-variant gene 6 (ETV6 ), to the receptor-associated kinase JAK2 as a result of t(9;12) in a lymphoid and t(9;15;12) in a myeloid leukemia. Blood 90(7), 2535–2540 (1997). 87 Griesinger F, Hennig H, Hillmer F et al.

A BCR–JAK2 fusion gene as the result of a t(9;22)(p24;q11.2) translocation in a patient with a clinically typical chronic myeloid leukemia. Genes Chromosomes Cancer 44(3), 329–333 (2005). 88 Murati A, Gelsi-Boyer V, Adélaïde J et al.

PCM1-JAK2 fusion in myeloproliferative disorders and acute erythroid leukemia with t (8;9) translocation. Leukemia 19(9), 1692–1696 (2005). 89 Kralovics R, Passamonti F, Buser AS et al.

A gain-of-function mutation of JAK2 in

Review

myeloproliferative disorders. N. Engl. J. Med. 352, 1779–1790 (2005). 90 Levine RL, Wadleigh M, Cools J et al.

Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 7, 387–397 (2005). 91 Oku S, Takenaka K, Kuriyama T et al. JAK2

V617F uses distinct signalling pathways to induce cell proliferation and neutrophil activation. Br. J. Haematol. 150, 334–344 (2010). 92 Denu JM, Dixon JE. Protein tyrosine

phosphatases: mechanisms of catalysis and regulation. Curr. Opin. Chem. Biol. 2(5), 633–641 (1998). 93 Alonso A, Sasin J, Bottini N et al. Protein

tyrosine phosphatases in the human genome. Cell 117(6), 699–711 (2004). 94 Tartaglia M, Niemeyer CM, Fragale A et al.

Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nat. Genet. 34(2), 148–150 (2003). 95 Tartaglia M, Martinelli S, Cazzaniga G et al.

Genetic evidence for lineage-related and differentiation stage-related contribution of somatic PTPN11 mutations to leukemogenesis in childhood acute leukemia. Blood 104(2), 307–313 (2004). 96 Chan RJ, Leedy MB, Munugalavadla V et al.

Human somatic PTPN11 mutations induce hematopoietic-cell hypersensitivity to granulocyte-macrophage colony-stimulating factor. Blood 105(9), 3737–3742 (2005). 97 Huang W, Saberwal G, Horvath E et al.

Leukemia-associated, constitutively active mutants of SHP2 protein tyrosine phosphatase inhibit NF1 transcriptional activation by the interferon consensus sequence binding protein. Mol. Cell. Biol. 26(17), 6311–6332 (2006). 98 Kleppe M, Lahortiga I, El Chaar T et al.

Deletion of the protein tyrosine phosphatase gene PTPN2 in T-cell acute lymphoblastic leukemia. Nat. Genet. 42(6), 530–535 (2010). 99 Negro R, Gobessi S, Longo PG et al.

Overexpression of the autoimmunityassociated phosphatase PTPN22 promotes survival of antigen-stimulated CLL cells by selectively activating AKT. Blood 119(26), 6278–6287 (2012). 100 Zanke B, Suzuki H, Kishihara K et al.

Cloning and expression of an inducible lymphoid-specific, protein tyrosine phosphatase (HePTPase). Eur. J. Immunol. 22(1), 235–239 (1992).

www.futuremedicine.com

671

Review  Zhu, Xiao, Wang, Fu, Zhao & Huang 101 Pettiford SM, Herbst R. The MAP-kinase

ERK2 is a specific substrate of the protein tyrosine phosphatase HePTP. Oncogene 19(7), 858–869 (2000).

leukemia and other malignant cells. Oncogene 22(48), 7649–7660 (2003). The resistance of B-CLL cells to DNA damage-induced apoptosis defined by DNA microarrays. Blood 101(11), 4598–4606 (2003). Constitutive activation of extracellular signal-regulated kinase in human acute leukemias: combined role of activation of MEK, hyperexpression of extracellular signal-regulated kinase, and downregulation of a phosphatase, PAC1. Blood 93(11), 3893–3899 (1999).

105 Motiwala T, Majumder S, Ghoshal K et al.

PTPROt inactivates the oncogenic fusion protein BCR/ABL and suppresses transformation of K562 cells. J. Biol. Chem. 284(1), 455–464 (2009). 106 Nakamura M, Kishi M, Sakaki T et al. Novel

tumor suppressor loci on 6q22-23 in primary central nervous system lymphomas. Cancer Res. 63(4), 737–741 (2003). 107 Aya-Bonilla C, Green MR, Camilleri E et al.

High-resolution loss of heterozygosity screening implicates PTPRJ as a potential tumor suppressor gene that affects susceptibility to Non-Hodgkin’s lymphoma. Genes Chromosomes Cancer 52(5), 467–479 (2013). 108 Novellino L, De Filippo A, Deho P et al.

PTPRK negatively regulates transcriptional activity of wild type and mutated oncogenic beta-catenin and affects membrane distribution of beta-catenin/E-cadherin complexes in cancer cells. Cell. Signal. 20(5), 872–883 (2008). 109 Tarcic G, Boguslavsky SK, Wakim J et al.

An unbiased screen identifies DEP-1 tumor suppressor as a phosphatase controlling EGFR endocytosis. Curr. Biol. 19(21), 1788–1798 (2009). 110 Fagerli UM, Holt RU, Holien T et al.

Overexpression and involvement in migration by the metastasis-associated phosphatase PRL-3 in human myeloma cells. Blood 111(2), 806–815 (2008). 111 Levy-Nissenbaum O, Sagi-Assif O, Kapon D

et al. Dual-specificity phosphatase Pyst2-L is constitutively highly expressed in myeloid

672

Conformational control inhibition of the BCR-ABL1 tyrosine kinase, including the gatekeeper T315I mutant, by the switchcontrol inhibitor DCC-2036. Cancer Cell 19(4), 556–568 (2011). 125 Weisberg E, Deng X, Choi HG et al.

Beneficial effects of combining a type II ATP competitive inhibitor with an allosteric competitive inhibitor of BCR-ABL for the treatment of imatinib-sensitive and imatinib-resistant CML. Leukemia 24(7), 1375–1378 (2010).

114 Gorre ME, Mohammed M, Ellwood K et al.

Clinical resistance to STI-571 cancer therapy caused by BCR–ABL gene mutation or amplification. Science 293(5531), 876–880 (2001).

104 Motiwala T, Majumder S, Kutay H et al.

Methylation and silencing of protein tyrosine phosphatase receptor type O in chronic lymphocytic leukemia. Clin. Cancer Res. 13(11), 3174–3181 (2007).

124 Chan WW, Wise SC, Kaufman MD et al.

113 Kim SC, Hahn JS, Min YH et al.

103 van Doorn R, Zoutman WH, Dijkman R

et al. Epigenetic profiling of cutaneous T-cell lymphoma: promoter hypermethylation of multiple tumor suppressor genes including BCL7a, PTPRG, and p73. J. Clin. Oncol. 23(17), 3886–3896 (2005).

phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias. N. Engl. J. Med. 369(19), 1783–1796 (2013).

112 Vallat L, Magdelénat H, Merle-Béral H et al.

102 Fridberg M, Kjellström S, Anagnostaki L

et al. Immunohistochemical analyses of phosphatases in childhood B-cell lymphoma: lower expression of PTEN and HePTP and higher number of positive cells for nuclear SHP2 in B-cell lymphoma cases compared with controls. Pediatr. Hematol. Oncol. 25(6), 528–540 (2008).

123 Cortes JE, Kim DW, Pinilla-Ibarz J et al. A



This report revealed mechanisms of imatinib resistance and suggested a strategy for identifying inhibitors to overcome resistance.

115 O’Hare T, Walters DK, Stoffregen EP et al.

In vitro activity of Bcr-Abl inhibitors AMN107 and BMS-354825 against clinically relevant imatinib-resistant Abl kinase domain mutants. Cancer Res. 65(11), 4500–4505 (2005). 116 Kantarjian H, Giles F, Wunderle L et al.

Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL. N. Engl. J. Med. 354(24), 2542–2551 (2006). 117 Talpaz M, Shah NP, Kantarjian H et al.

Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N. Engl. J. Med. 354(24), 2531–2541 (2006). 118 Saglio G, Kim DW, Issaragrisil S et al.

Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N. Engl. J. Med. 362(24), 2251–2259 (2010). 119 Kantarjian H, Shah NP, Hochhaus A et al.

Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N. Engl. J. Med. 362(24), 2260–2270 (2010). 120 Khoury HJ, Cortes JE, Kantarjian HM et al.

Bosutinib is active in chronic phase chronic myeloid leukemia after imatinib and dasatinib and/or nilotinib therapy failure. Blood 119(15), 3403–3412 (2012). 121 Azam M, Seeliger MA, Gray NS et al.

Activation of tyrosine kinases by mutation of the gatekeeper threonine. Nat. Struct. Mol. Biol. 15(10), 1109–1118 (2008). 122 Cortes JE, Kantarjian H, Shah NP et al.

Ponatinib in refractory Philadelphia chromosome-positive leukemias. N. Engl. J. Med. 367(22), 2075–2088 (2012).

Future Oncol. (2015) 11(4)

126 Knapper S. The clinical development of FLT3

inhibitors in acute myeloid leukemia. Expert Opin. Investig. Drugs 20(10), 1377–1395 (2011). •

A review of the first decade of clinical trials of FLT3-targeted tyrosine kinase inhibitors.

127 DeAngelo DJ, Stone RM, Heaney ML et al.

Phase 1 clinical results with tandutinib (MLN518), a novel FLT3 antagonist, in patients with acute myelogenous leukemia or high-risk myelodysplastic syndrome: safety, pharmacokinetics, and pharmacodynamics. Blood 108(12), 3674–3681 (2006). 128 Ravandi F, Alattar ML, Grunwald MR et al.

Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood 121(23), 4655–4662 (2013). 129 Inaba H, Rubnitz JE, Coustan-Smith E et al.

Phase I pharmacokinetic and pharmaco­ dynamic study of the multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in pediatric relapsed/refractory leukemia. J. Clin. Oncol. 29(24), 3293–3300 (2011). 130 Fiedler W, Serve H, Döhner H et al.

A Phase 1 study of SU11248 in the treatment of patients with refractory or resistant acute myeloid leukemia (AML) or not amenable to conventional therapy for the disease. Blood 105(3), 986–993 (2005). 131 Stone RM, Fischer T, Paquette R et al.

Phase IB study of the FLT3 kinase inhibitor midostaurin with chemotherapy in younger newly diagnosed adult patients with acute myeloid leukemia. Leukemia 26(9), 2061–2068 (2012). 132 Fischer T, Stone RM, Deangelo DJ et al.

Phase IIB trial of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor

future science group

Mutations in tyrosine kinase & tyrosine phosphatase  (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. 28(28), 4339–4345 (2010). 133 Knapper S, Burnett AK, Littlewood T et al.

A Phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood 108(10), 3262–3270 (2006). 134 Pratz KW, Cortes J, Roboz GJ et al.

A pharmacodynamic study of the FLT3 inhibitor KW-2449 yields insight into the basis for clinical response. Blood 113(17), 3938–3946 (2009). 135 Cortes JE, Kantarjian H, Foran JM 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. 31(29), 3681–3687 (2013). 136 Galanis A, Ma H, Rajkhowa T et al.

Crenolanib is a potent inhibitor of FLT3 with activity against resistance-conferring point mutants. Blood 123(1), 94–100 (2014). 137 Smith CC, Wang Q, Chin CS et al.

Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia. Nature 485(7397), 260–263 (2012).

future science group



Provided evidence for the mechanism of relapse after treatment with FLT3 inhibitors.

144 Heneberg P. Use of protein tyrosine

phosphatase inhibitors as promising targeted therapeutic drugs. Curr. Med. Chem. 16(6), 706–733 (2009).

138 Levis M, Ravandi F, Wang ES et al. Results

from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse. Blood 117(12), 3294–3301 (2011).



Anticancer activity of sodium stibogluconate in synergy with IFNs. J. Immunol. 169(10), 5978–5985 (2002). 146 Naing A, Reuben JM, Camacho LH et al.

Phase I dose escalation study of sodium stibogluconate (SSG), a protein tyrosine phosphatase inhibitor, combined with interferon alpha for patients with solid tumors. J. Cancer 2, 81–89 (2011).

140 Moore AS, Faisal A, Gonzalez de Castro D

et al. Selective FLT3 inhibition of FLT3ITD+ acute myeloid leukaemia resulting in secondary D835Y mutation: a model for emerging clinical resistance patterns. Leukemia 26(7), 1462–1470 (2012).

147 Yi T, Elson P, Mitsuhashi M et al.

Phosphatase inhibitor, sodium stibogluconate, in combination with interferon (IFN) alpha 2b: Phase I trials to identify pharmaco­ dynamic and clinical effects. Oncotarget 2(12), 1155–1164 (2011).

141 Harrison C, Kiladjian JJ, Al-Ali HK et al.

JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N. Engl. J. Med. 366(9), 787–798 (2012).

148 Zhang S, Chen L, Luo Y et al. Acquisition of

142 Vannucchi AM. How do JAK2-inhibitors work

a potent and selective TC-PTP inhibitor via a stepwise fluorophore-tagged combinatorial synthesis and screening strategy. J. Am. Chem. Soc. 13(36), 13072–13079 (2009).

in myelofibrosis: an alternative hypothesis. Leuk. Res. 33(12), 1581–1583 (2009). 143 Cervantes F, Vannucchi AM, Kiladjian JJ

et al. Three-year efficacy, safety, and survival findings from COMFORT-II, a Phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood 122(25), 4047–4053 (2013).

A review summarized inhibitors of activity or expression of the particular classical PTPs.

145 Yi T, Pathak MK, Lindner DJ et al.

139 Zirm E, Spies-Weisshart B, Heidel F et al.

Ponatinib may overcome resistance of FLT3-ITD harbouring additional point mutations, notably the previously refractory F691I mutation. Br. J. Haematol. 157(4), 483–492 (2012).

Review



Proposed a new approach for potent, highly selective and cell permeable PTP inhibitory agents.

www.futuremedicine.com

673

Mutations in tyrosine kinase and tyrosine phosphatase and their relevance to the target therapy in hematologic malignancies.

Protein tyrosine kinases and protein tyrosine phosphatases play pivotal roles in regulation of cellular phosphorylation and signal transduction with o...
2MB Sizes 0 Downloads 6 Views