are src family kinases responsible for imatinib- and dasatinib-resistant chronic myeloid leukemias?

3
Leukemia Research 35 (2011) 27–29 Contents lists available at ScienceDirect Leukemia Research journal homepage: www.elsevier.com/locate/leukres Guest editorial Are SRC family kinases responsible for imatinib- and dasatinib-resistant chronic myeloid leukemias? Keywords: Chronic myeloid leukemia SRC family kinase Imatinib Philadelphia-positive (Ph+) chronic myeloid leukemia (CML) is a myeloproliferative disease caused by the BCR-ABL fusion gene. The BCR-ABL fusion gene encodes a constitutively activated tyro- sine kinase and is the product of a well-defined translocation event between the ABL kinase gene on chromosomes 9 and the BCR gene on chromosome 22 [1]. More than 90% CML patients start with a chronic phase, progress to an accelerated phase and even- tually develop “blast crisis”, which resembles an acute leukemia [1]. Ten years ago, allogeneic bone marrow transplantation (BMT) was the recommended treatment for newly diagnosed patients with CML and was regarded as a ‘curative’ therapy for CML [2]. Its major drawback relates to the age and availabilities of donors [3]. A great effort has been put to find a new way to overcome this limitation. With the success of “targeted” tyrosine kinase inhibitor (TKI) therapy in BCR-ABL-driven CML, the BCR-ABL kinase inhibitor imatinib mesylate (Gleevec/Glivec, formerly STI571; Novartis) has become the first-line therapy for CML. Imatinib competitively binds to the ATP binding site of BCR-ABL, locks it in a closed confor- mation, and therefore inhibits tyrosine kinase activity [4]. After 5 years of imatinib treatment, the rate of complete cytogentic response among patients has been reported to be 87%, while only 7% of patients progress to accelerated-phase CML or blast crisis [5]. Despite this response, imatinib does not cure CML and resistance to imatinib eventually develops. One major reason for the resis- tance is intractable point mutations in the BCR-ABL kinase domain like T315I [6]. In addition to resistance, it is now understood that BCR-ABL expressing leukemia stem cells (LSCs) are insensitive to imatinib treatment [7,8]. Although imatinib can effectively inhibit BCR-ABL kinase activ- ity and prolong the survival of CML patients, it cannot eradicate LSCs and therefore cure the disease. To resolve the resistance of LSCs to kinase inhibitors and fully understand how BCR-ABL sig- nals in these LSCs, research has been undertaken to identify unique molecular pathways that are dependent on or independent of BCR- ABL kinase activity. Several have been identified to play important roles in CML development and specifically LSC survival, includ- ing the Wnt/ˇ-catenin, Hedgehog, Alox5, Pten and FoxO pathways [9–14] (Fig. 1). These new approaches also provide some potential targets for curative therapy. Arachidonate 5-lipoxygenase (Alox5) gene is a critical regulator for LSCs in BCR-ABL-induced CML. In the absence of ALOX5, BCR-ABL failed to induce CML in a mouse model for human CML. Similarly, treatment of these CML mice with the ALOX5 (5-LO) inhibitor zileuton impaired the function of LSCs and prolonged the survival of treated mice. These results demonstrated that a specific target gene can be found in LSCs and its inhibition can completely disrupt the function of LSCs [11]. A Phase I/II study to evaluate the safety of zileuton in combination with imatinib in CML patients has been initiated and is now recruiting patients (ClinicalTrials.gov Identifier: NCT01130688). Besides these unique signal pathways, SRC family kinases (SFKs) have also been shown to be functionally important in CML cells. SFKs are a group of structurally related non-receptor protein tyro- sine kinases, containing four similar SRC homology domains [15]. Members of this kinase family include BLK, FGR, FYN, HCK, LYN, LCK, c-SRC, YRK and YES [15]. Among them, HCK and LY N are known to interact with BCR-ABL [16]. HCK binds directly to BCR-ABL through SH domains and kinase domain as well as the C-terminal tail, involving kinase-dependent and -independent components. The interaction with HCK or other SRC family members is required for transformation of the myeloid leukemia cell line by BCR- ABL [17]. LYN co-immunoprecipitates with BCR-ABL and regulates BCR-ABL function through SH2-containing tyrosine phosphotase-1 (SHP-1) [18]. HCK and LYN are activated in CML cells in a manner that is apparently kinase-independent, as it is not inhibited by the BCR- ABL kinase inhibitor imatinib. The function of SRC family kinases in CML is also supported by the recent findings that SFKs: FGR, LYN and HCK are required for the proliferation of BCR-ABL-expressing pre-B-cells and that these SFKs remain active following imatinib inhibition of BCR-ABL kinase activity in leukemia cells [19,20]. In this issue of Leukemia Research, Hayette et al. present evidence defining the frequency with which SFKs are involved in imatinib- and dasatinib-resistant CML patients [21]. They identified a high frequency of SFKs involvement in tyrosine kinase inhibitor-resistant CML and noted in particular elevated SFKs activities in patients with progressed disease and blast crisis. In this retrospective study, four categories of imatinib- and dasatinib-treated patients (imatinib resistant/dasatinib- responsive; dasatinib-resistant; blast crisis or CML progression and T315I or F317L mutated patient receiving omacetaxine treatment) were included. Transcript levels of HCK, LYN and another SFK func- tionally related gene BTK were screened in a large population of CML patients. Additionally and more importantly, protein expres- sion of SFKs was analyzed in 46 patients (146 total follow-up samples). SFKs activation was elevated in more than 50% of resis- 0145-2126/$ – see front matter. Published by Elsevier Ltd. doi:10.1016/j.leukres.2010.07.032

Upload: yaoyu-chen

Post on 26-Jun-2016

217 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Are SRC family kinases responsible for imatinib- and dasatinib-resistant chronic myeloid leukemias?

G

Am

KCSI

aTsbgwt[wwI[l(ibtm5r7DttlBi

iLLnmAri[t

0d

Leukemia Research 35 (2011) 27–29

Contents lists available at ScienceDirect

Leukemia Research

journa l homepage: www.e lsev ier .com/ locate / leukres

uest editorial

re SRC family kinases responsible for imatinib- and dasatinib-resistant chronicyeloid leukemias?

eywords:hronic myeloid leukemiaRC family kinasematinib

Philadelphia-positive (Ph+) chronic myeloid leukemia (CML) ismyeloproliferative disease caused by the BCR-ABL fusion gene.

he BCR-ABL fusion gene encodes a constitutively activated tyro-ine kinase and is the product of a well-defined translocation eventetween the ABL kinase gene on chromosomes 9 and the BCRene on chromosome 22 [1]. More than 90% CML patients startith a chronic phase, progress to an accelerated phase and even-

ually develop “blast crisis”, which resembles an acute leukemia1]. Ten years ago, allogeneic bone marrow transplantation (BMT)as the recommended treatment for newly diagnosed patientsith CML and was regarded as a ‘curative’ therapy for CML [2].

ts major drawback relates to the age and availabilities of donors3]. A great effort has been put to find a new way to overcome thisimitation. With the success of “targeted” tyrosine kinase inhibitorTKI) therapy in BCR-ABL-driven CML, the BCR-ABL kinase inhibitormatinib mesylate (Gleevec/Glivec, formerly STI571; Novartis) hasecome the first-line therapy for CML. Imatinib competitively bindso the ATP binding site of BCR-ABL, locks it in a closed confor-

ation, and therefore inhibits tyrosine kinase activity [4]. Afteryears of imatinib treatment, the rate of complete cytogentic

esponse among patients has been reported to be 87%, while only% of patients progress to accelerated-phase CML or blast crisis [5].espite this response, imatinib does not cure CML and resistance

o imatinib eventually develops. One major reason for the resis-ance is intractable point mutations in the BCR-ABL kinase domainike T315I [6]. In addition to resistance, it is now understood thatCR-ABL expressing leukemia stem cells (LSCs) are insensitive to

matinib treatment [7,8].Although imatinib can effectively inhibit BCR-ABL kinase activ-

ty and prolong the survival of CML patients, it cannot eradicateSCs and therefore cure the disease. To resolve the resistance ofSCs to kinase inhibitors and fully understand how BCR-ABL sig-als in these LSCs, research has been undertaken to identify uniqueolecular pathways that are dependent on or independent of BCR-

BL kinase activity. Several have been identified to play importantoles in CML development and specifically LSC survival, includ-ng the Wnt/ˇ-catenin, Hedgehog, Alox5, Pten and FoxO pathways9–14] (Fig. 1). These new approaches also provide some potentialargets for curative therapy. Arachidonate 5-lipoxygenase (Alox5)

145-2126/$ – see front matter. Published by Elsevier Ltd.oi:10.1016/j.leukres.2010.07.032

gene is a critical regulator for LSCs in BCR-ABL-induced CML. In theabsence of ALOX5, BCR-ABL failed to induce CML in a mouse modelfor human CML. Similarly, treatment of these CML mice with theALOX5 (5-LO) inhibitor zileuton impaired the function of LSCs andprolonged the survival of treated mice. These results demonstratedthat a specific target gene can be found in LSCs and its inhibitioncan completely disrupt the function of LSCs [11]. A Phase I/II studyto evaluate the safety of zileuton in combination with imatinibin CML patients has been initiated and is now recruiting patients(ClinicalTrials.gov Identifier: NCT01130688).

Besides these unique signal pathways, SRC family kinases (SFKs)have also been shown to be functionally important in CML cells.SFKs are a group of structurally related non-receptor protein tyro-sine kinases, containing four similar SRC homology domains [15].Members of this kinase family include BLK, FGR, FYN, HCK, LYN, LCK,c-SRC, YRK and YES [15]. Among them, HCK and LY N are known tointeract with BCR-ABL [16]. HCK binds directly to BCR-ABL throughSH domains and kinase domain as well as the C-terminal tail,involving kinase-dependent and -independent components. Theinteraction with HCK or other SRC family members is required fortransformation of the myeloid leukemia cell line by BCR- ABL [17].LYN co-immunoprecipitates with BCR-ABL and regulates BCR-ABLfunction through SH2-containing tyrosine phosphotase-1 (SHP-1)[18]. HCK and LYN are activated in CML cells in a manner that isapparently kinase-independent, as it is not inhibited by the BCR-ABL kinase inhibitor imatinib. The function of SRC family kinasesin CML is also supported by the recent findings that SFKs: FGR, LYNand HCK are required for the proliferation of BCR-ABL-expressingpre-B-cells and that these SFKs remain active following imatinibinhibition of BCR-ABL kinase activity in leukemia cells [19,20].

In this issue of Leukemia Research, Hayette et al. presentevidence defining the frequency with which SFKs are involvedin imatinib- and dasatinib-resistant CML patients [21]. Theyidentified a high frequency of SFKs involvement in tyrosinekinase inhibitor-resistant CML and noted in particular elevatedSFKs activities in patients with progressed disease and blastcrisis. In this retrospective study, four categories of imatinib-and dasatinib-treated patients (imatinib resistant/dasatinib-responsive; dasatinib-resistant; blast crisis or CML progression andT315I or F317L mutated patient receiving omacetaxine treatment)were included. Transcript levels of HCK, LYN and another SFK func-

tionally related gene BTK were screened in a large population ofCML patients. Additionally and more importantly, protein expres-sion of SFKs was analyzed in 46 patients (146 total follow-upsamples). SFKs activation was elevated in more than 50% of resis-
Page 2: Are SRC family kinases responsible for imatinib- and dasatinib-resistant chronic myeloid leukemias?

28 Guest editorial / Leukemia Research 35 (2011) 27–29

ende

twsmt

metercispiipcpaoe

C

A

R

[

[

[

[

[

[

[

[

[

[

[

[

Fig. 1. Signal pathways dependent or indep

ant CML patients, and this increase more significantly correlatesith disease progression. Interestingly, activation of SFKs expres-

ion is not likely caused by BCR-ABL mutation, as four BCR-ABLutated patients showed concomitant SFKs activation similar to

hat seen in patients expressing a wild-type BCR-ABL.Hayette et al. also followed five patients possessing BCR-ABL

utations who were receiving omacetaxine treatment [21]. Omac-taxine (also known as homoharringtonine) targets leukemic cellshrough a mechanism of action that differs from TKIs [22]. Omac-taxine has shown promising clinical activity in CML patientsesistant to imatinib or other TKIs and is currently in a phase IIIlinic trial in United States [22]. It functions as a protein synthesisnhibitor and can inhibit ABL, HSP90 and MCL-1 protein expres-ion in CML cells [23]. SFKs were also shown to be activated inatients with BCR-ABL mutation under omacetaxine treatment, but

n one patient resistant to omacetaxine treatment, SFKs remainednactivated. The SFKs activation after omacetaxine treatment mayrovide insights into the possible mechanisms by which the drugannot eradicate leukemia cells and also suggests some unknownathways are being activated. These findings indicate that SFKsre potential therapeutic targets of imatinib-, dasatinib- ormacetaxine-resistant CML and also are suggestive of a new strat-gy that may effectively target leukemia cells and thus cure CML.

onflicts of interest

The author stated no conflict of interests.

cknowledgements

No support for this work.

eferences

[1] Faderl S, Talpaz M, Estrov Z, Kantarjian HM. Chronic myelogenous leukemia:biology and therapy. Ann Intern Med 1999;131:207–19.

[2] Mughal TI, Yong A, Szydlo RM, Dazzi F, Olavarria E, van Rhee F, et al. Molec-ular studies in patients with chronic myeloid leukaemia in remission 5 yearsafter allogeneic stem cell transplant define the risk of subsequent relapse. Br JHaematol 2001;115:569–74.

[3] Chen Y, Peng C, Sullivan C, Li D, Li S. Critical molecular pathways in cancer stemcells of chronic myeloid leukemia. Leukemia 2010. Jun 24 [Epub ahead of print].

[4] Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford JM, et al. Activity ofa specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronicmyeloid leukemia and acute lymphoblastic leukemia with the Philadelphiachromosome. N Engl J Med 2001;344:1038–42.

[

[

nt on BCR-ABL kinase activity in CML cells.

[5] Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian H, Gattermann N,et al. Five-year follow-up of patients receiving imatinib for chronic myeloidleukemia. N Engl J Med 2006;355:2408–17.

[6] Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao PN, et al. Clini-cal resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation oramplification. Science 2001;293:876–80.

[7] Graham SM, Jorgensen HG, Allan E, Pearson C, Alcorn MJ, Richmond L,et al. Primitive, quiescent, Philadelphia-positive stem cells from patientswith chronic myeloid leukemia are insensitive to STI571 in vitro. Blood2002;99:319–25.

[8] Bhatia R, Holtz M, Niu N, Gray R, Snyder DS, Sawyers CL, et al. Persistenceof malignant hematopoietic progenitors in chronic myelogenous leukemiapatients in complete cytogenetic remission following imatinib mesylate treat-ment. Blood 2003;101:4701–7.

[9] Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM, et al. Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell2007;12:528–41.

10] Zhao C, Chen A, Jamieson CH, Fereshteh M, Abrahamsson A, Blum J, et al. Hedge-hog signalling is essential for maintenance of cancer stem cells in myeloidleukaemia. Nature 2009;458:776–9.

11] Chen Y, Hu Y, Zhang H, Peng C, Li S. Loss of the Alox5 gene impairs leukemiastem cells and prevents chronic myeloid leukemia. Nat Genet 2009;41:783–92.

12] Peng C, Chen Y, Yang Z, Zhang H, Osterby L, Rosmarin AG, et al. PTEN is a tumorsuppressor in CML stem cells and BCR-ABL-induced leukemias in mice. Blood2010;115:626–35.

13] Naka K, Hoshii T, Muraguchi T, Tadokoro Y, Ooshio T, Kondo Y, et al. TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloidleukaemia. Nature 2010;463:676–80.

14] Hu Y, Chen Y, Douglas L, Li S. beta-Catenin is essential for survival of leukemicstem cells insensitive to kinase inhibition in mice with BCR-ABL-inducedchronic myeloid leukemia. Leukemia 2009;23:109–16.

15] Lowell CA, Soriano P. Knockouts of Src-family kinases: stiff bones, wimpy Tcells, and bad memories. Genes Dev 1996;10:1845–57.

16] Danhauser-Riedl S, Warmuth M, Druker BJ, Emmerich B, Hallek M. Activationof Src kinases p53/56lyn and p59hck by p210bcr/abl in myeloid cells. CancerRes 1996;56:3589–96.

17] Lionberger JM, Wilson MB, Smithgall TE. Transformation of myeloid leukemiacells to cytokine independence by Bcr-Abl is suppressed by kinase-defectiveHck. J Biol Chem 2000;275:18581–5.

18] Daigle I, Yousefi S, Colonna M, Green DR, Simon HU. Death receptors bind SHP-1 and block cytokine-induced anti-apoptotic signaling in neutrophils. Nat Med2002;8:61–7.

19] Hu Y, Liu Y, Pelletier S, Buchdunger E, Warmuth M, Fabbro D, et al. Require-ment of Src kinases Lyn, Hck and Fgr for BCR-ABL1-induced B-lymphoblasticleukemia but not chronic myeloid leukemia. Nat Genet 2004;36:453–61.

20] Hu Y, Swerdlow S, Duffy TM, Weinmann R, Lee FY, Li S. Targeting multiple kinasepathways in leukemic progenitors and stem cells is essential for improvedtreatment of Ph+ leukemia in mice. Proc Natl Acad Sci USA 2006;103:16870–5.

21] Hayette S, Chabane K, Michallet M, Michallat E, Cony-Makhoul P, Salesse S, et al.Longitudinal studies of SRC family kinases in imatinib- and dasatinib-resistantchronic myelogenous leukemia patients. Leuk Res 2011;35:38–43.

22] Kantarjian HM, Talpaz M, Santini V, Murgo A, Cheson B, O’Brien SM.Homoharringtonine: history, current research, and future direction. Cancer2001;92:1591–605.

23] Chen Y, Hu Y, Michaels S, Segal D, Brown D, Li S. Inhibitory effects of omacetax-ine on leukemic stem cells and BCR-ABL-induced chronic myeloid leukemiaand acute lymphoblastic leukemia in mice. Leukemia 2009;23:1146–54.

Page 3: Are SRC family kinases responsible for imatinib- and dasatinib-resistant chronic myeloid leukemias?

ia Res

Guest editorial / Leukem

Yaoyu Chen ∗

Department of Medicine, University of MassachusettsMedical School, Lazare Research Building 280J,

364 Plantation Street, Worcester,MA 01605, USA

earch 35 (2011) 27–29 29

∗ Tel.: +1 08 856 4157, fax: +1 508 856 6797.E-mail address: [email protected]

14 July 2010Available online 19 August 2010