bcr-abl variants - biological and clinical aspects

8
Leukemia Research 26 (2002) 713–720 Millennium Review Bcr–Abl variants: biological and clinical aspects Anjali S. Advani a , Ann Marie Pendergast b,a Departments of Hematology and Oncology, Duke University Medical Center, Durham, NC 27710, USA b Department of Pharmacology and Cancer Biology, LSRC Room C233A, La Salle St. Extension, Duke University Medical Center, Durham, NC 27710, USA Received 8 November 2001; accepted 25 November 2001 Abstract Bcr–Abl is an oncogene that arises from fusion of the Bcr gene with the c-Abl proto-oncogene. Three different Bcr–Abl variants can be formed, depending on the amount of Bcr gene included: p185, p210, and p230. The three variants are associated with distinct types of human leukemias. Examination of the signaling pathways differentially regulated by the Bcr–Abl proteins will help us gain better insight into Bcr–Abl mediated leukemogenesis. © 2002 Elsevier Science Ltd. All rights reserved. Keywords: Philadelphia chromosome; Bcr–Abl; Acute lymphocytic leukemia; Chronic myelogenous leukemia; Chronic neutrophilic leukemia 1. Introduction The Philadelphia chromosome (Ph) involves fusion of the breakpoint cluster region (Bcr) gene on chromosome 22 at band q11 with the Abl proto-oncogene on chromosome 9 at band q34 [1]. Three different forms of the Bcr–Abl oncogene exist, p185, p210, and p230, that are associated with distinct types of leukemia. P185 is associated with 20–30% of acute lymphocytic leukemia (ALL), p210 with 90% of chronic myelogenous leukemia (CML), and p230 with a subset of patients with chronic neutrophilic leukemia (CNL) [2]. This review will discuss the biological and clinical aspects of the Bcr–Abl variants. Abbreviations: Ph, Philadelphia chromosome; Bcr, breakpoint cluster region; ALL, acute lymphocytic leukemia; CML, chronic myelogenous leukemia; CNL, chronic neutrophilic leukemia; AUL, acute undifferen- tiated leukemia; AML, acute myelogenous leukemia; Ph+, Philadelphia chromosome positive; CALGB, cancer and leukemia group B; BMT, bone marrow transplant; CML-N, cases of chronic neutrophilic leukemia that are Ph+; CMML, chronic myelomonocytic leukemia; RT-PCR, reverse transcriptase polymerase chain reaction; CFU-erythroid, colony forming unit-erythroid; CFU-GM, colony forming unit-granulocyte macrophage; PCR, polymerase chain reaction; STI-571, signal transduction inhibitor 571; ATP, adenosine triphosphate; mRNA, messenger RNA Corresponding author. Fax: +1-919-681-7148. E-mail address: [email protected] (A.M. Pendergast). 2. Clinical correlates CML typically has a biphasic course that is characterized by chronic and blastic phases [3–6]. The chronic phase is characterized by an expansion of myeloid cells in the periph- eral blood, bone marrow, and spleen. The molecular basis for the myeloid expansion is puzzling as Bcr–Abl transforms the hematopoietic stem cell and is present in all hematopoi- etic elements [3–6]. Approximately 50% of patients in the chronic phase have no symptoms and are diagnosed by rou- tine testing [7,8]. Signs and symptoms, however, can include fatigue, weight loss, abdominal fullness, bleeding, sweats, splenomegaly, and hepatomegaly [7–9]. The blastic phase is thought to occur secondary to addi- tional mutations (see below). Prior to entering blast phase, 75% of patients develop an intervening accelerated phase that is characterized by increased basophilia, blasts, and promyelocytes [7,10]. This usually progresses to blastic phase within 3–18 months [7]. The acute leukemia is ALL in one-third of cases, whereas two-thirds are acute undiffer- entiated leukemia (AUL) or acute myelogenous leukemia (AML) [7,11]. Patients with a lymphoid blastic phase may respond to treatment with ALL regimens; however, the me- dian duration of response is 9–12 months [7,12]. Because the disease is relatively treatment resistant, the median sur- vival is 3 months for myeloid blast crisis and 6 months for lymphoid blast crisis [13–16]. Philadelphia chromosome positive (Ph+) ALL is an acute leukemia, which unlike CML, requires prompt, aggressive 0145-2126/02/$ – see front matter © 2002 Elsevier Science Ltd. All rights reserved. PII:S0145-2126(01)00197-7

Upload: carolina-moraes

Post on 18-Apr-2015

47 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Bcr-Abl Variants - Biological and Clinical Aspects

Leukemia Research 26 (2002) 713–720

Millennium Review

Bcr–Abl variants: biological and clinical aspects

Anjali S. Advania, Ann Marie Pendergastb,∗a Departments of Hematology and Oncology, Duke University Medical Center, Durham, NC 27710, USA

b Department of Pharmacology and Cancer Biology, LSRC Room C233A, La Salle St. Extension,Duke University Medical Center, Durham, NC 27710, USA

Received 8 November 2001; accepted 25 November 2001

Abstract

Bcr–Abl is an oncogene that arises from fusion of theBcr gene with thec-Abl proto-oncogene. Three differentBcr–Abl variants canbe formed, depending on the amount ofBcr gene included:p185, p210, andp230. The three variants are associated with distinct types ofhuman leukemias. Examination of the signaling pathways differentially regulated by the Bcr–Abl proteins will help us gain better insightinto Bcr–Abl mediated leukemogenesis. © 2002 Elsevier Science Ltd. All rights reserved.

Keywords: Philadelphia chromosome;Bcr–Abl; Acute lymphocytic leukemia; Chronic myelogenous leukemia; Chronic neutrophilic leukemia

1. Introduction

The Philadelphia chromosome (Ph) involves fusion of thebreakpoint cluster region (Bcr) gene on chromosome 22 atband q11 with theAbl proto-oncogene on chromosome 9 atband q34 [1]. Three different forms of theBcr–Abl oncogeneexist,p185, p210, andp230, that are associated with distincttypes of leukemia.P185 is associated with 20–30% of acutelymphocytic leukemia (ALL),p210 with 90% of chronicmyelogenous leukemia (CML), andp230 with a subset ofpatients with chronic neutrophilic leukemia (CNL) [2]. Thisreview will discuss the biological and clinical aspects of theBcr–Abl variants.

Abbreviations: Ph, Philadelphia chromosome; Bcr, breakpoint clusterregion; ALL, acute lymphocytic leukemia; CML, chronic myelogenousleukemia; CNL, chronic neutrophilic leukemia; AUL, acute undifferen-tiated leukemia; AML, acute myelogenous leukemia; Ph+, Philadelphiachromosome positive; CALGB, cancer and leukemia group B; BMT, bonemarrow transplant; CML-N, cases of chronic neutrophilic leukemia thatare Ph+; CMML, chronic myelomonocytic leukemia; RT-PCR, reversetranscriptase polymerase chain reaction; CFU-erythroid, colony formingunit-erythroid; CFU-GM, colony forming unit-granulocyte macrophage;PCR, polymerase chain reaction; STI-571, signal transduction inhibitor571; ATP, adenosine triphosphate; mRNA, messenger RNA

∗ Corresponding author. Fax:+1-919-681-7148.E-mail address: [email protected] (A.M. Pendergast).

2. Clinical correlates

CML typically has a biphasic course that is characterizedby chronic and blastic phases [3–6]. The chronic phase ischaracterized by an expansion of myeloid cells in the periph-eral blood, bone marrow, and spleen. The molecular basisfor the myeloid expansion is puzzling as Bcr–Abl transformsthe hematopoietic stem cell and is present in all hematopoi-etic elements [3–6]. Approximately 50% of patients in thechronic phase have no symptoms and are diagnosed by rou-tine testing [7,8]. Signs and symptoms, however, can includefatigue, weight loss, abdominal fullness, bleeding, sweats,splenomegaly, and hepatomegaly [7–9].

The blastic phase is thought to occur secondary to addi-tional mutations (see below). Prior to entering blast phase,75% of patients develop an intervening accelerated phasethat is characterized by increased basophilia, blasts, andpromyelocytes [7,10]. This usually progresses to blasticphase within 3–18 months [7]. The acute leukemia is ALLin one-third of cases, whereas two-thirds are acute undiffer-entiated leukemia (AUL) or acute myelogenous leukemia(AML) [7,11]. Patients with a lymphoid blastic phase mayrespond to treatment with ALL regimens; however, the me-dian duration of response is 9–12 months [7,12]. Becausethe disease is relatively treatment resistant, the median sur-vival is 3 months for myeloid blast crisis and 6 months forlymphoid blast crisis [13–16].

Philadelphia chromosome positive (Ph+) ALL is an acuteleukemia, which unlike CML, requires prompt, aggressive

0145-2126/02/$ – see front matter © 2002 Elsevier Science Ltd. All rights reserved.PII: S0145-2126(01)00197-7

Page 2: Bcr-Abl Variants - Biological and Clinical Aspects

714 A.S. Advani, A.M. Pendergast / Leukemia Research 26 (2002) 713–720

treatment with high dose chemotherapy [17]. Compared toBcr–Abl negative ALL, it tends to have a worse prognosis.In a Cancer and Leukemia Group B (CALGB) study, themedian remission duration for Ph+ ALL was only 8 months[17,18]. Because fewer than 5% of patients are cured bychemotherapy, allogeneic BMT remains the best chance ofcure [19,20]. The reason why p185 tends to be associatedwith acute leukemia is unknown. One hypothesis is that p185is more efficient at promoting secondary events necessaryfor the acute phase disease [21].

Although the majority of Ph+ ALL exhibit a B-lymphoidlineage phenotype, Ph+ ALL may also involve the stemcell, because some are biphenotypic [21,22]. In addition,involvement of the myeloid lineage can be seen in both p185and p210 ALL [21–29].

Finally, p230 is associated with a subset of CNL, whichhas a much more benign course. Bcr–Abl positive CNL wasidentified in patients who showed the presence of aBcr–Able19a2 transcript [30,31]. About 100 cases of Bcr–Abl posi-tive CNL have been reported [30,32,33]. Most cases of CNLare Ph chromosome negative, so these cases have been re-ferred to as CML-N [30]. CNL involves a sustained ma-ture neutrophilic expansion with mild hepatosplenomegaly.In general, there is a low proportion of immature granulo-cytes and milder anemia [34]. Progression to blast crisis isuncommon [30].

Although p185 is typically associated with ALL, p230with CNL, and p210 with CML, there is overlap. P210 oc-curs in 40% of Ph+ ALL, p185 occurs in 2–3% CML, andp230 in some cases of CML [2]. Patients with p185 CMLare thought to have a disease more reminiscent of chronicmyelomonocytic leukemia (CMML), and to have a mono-cytic defect [2,35]. Kantajaran et al. examined 32 patientswith p210 or p185 Ph+ ALL and found no difference inclinico-laboratory, karyotypic, or prognostic implications[13,36]. It is not clear whether intrinisic differences in theactivities of the three Bcr–Abl proteins account for theirassociation with different disease phenotypes or whether

Fig. 1. Structural features of the Bcr–Abl variants. The p185 Bcr–Abl protein is primarily associated with ALL, p210 Bcr–Abl with CML, and p230Bcr–Abl with CNL. The C-terminal (Abl) region of all Bcr–Abl variants contains Src-homology (SH3 and SH2) domains, tyrosine kinase (SH1) domain,proline-rich sequences that contain PXXP motifs for SH3 domain binding, DNA binding domain, actin binding domain, nuclear localization signals (NLS),and nuclear export signal (NES). The N-terminal (Bcr) portion of the protein differs among the variants. The variants include different amounts of Bcrsequence, with p230 including the largest amount of Bcr sequence. All three variants have a dimerization domain (DD) and serine/threonine kinase domain(P-S/T). P210 and p230 both have Dbl and PH domains. In addition, p230 has a calcium/phospholipid binding domain (CalB) and a Gap–Rac domain.

expression of each protein occurs in a distinict hematopoi-etic lineage.

In addition, some patients with CML co-expressp185andp210, based on detection by reverse transcriptase poly-merase chain reaction (RT-PCR) [2,37–39]. High levelsof both transcripts have been found in patients with blas-tic transformation of CML, whereas those with CML inchronic phase being treated with interferon have low orundetectable levels ofp185 [38,39]. This suggests that aquantitative variation of the two transcripts may play a rolein the clinical disease [38].

Different forms of p210 are also produced by alterna-tive splicing, namely,b2a2 andb3a2, with the latter encod-ing a longer transcript. There is some data indicating thatpatients withb3a2 have a better prognosis thanb2a2, ex-hibiting a long chronic phase and better response to inter-feron [2,40–42]. This is interesting, given that the longerBcr–Abl variants (i.e.p230) tend to have a more indolentbiological/clinical phenotype than the shorter variants (i.e.p185). However, more recent prospective studies suggest nodifference in prognosis between patients withb2a2 versusb3a2 transcripts [43–45]. Other data suggest that patientswith b3a2 transcripts have higher platelets than patients withb2a2 transcripts [46,47]. Relevant to this, 9/10 patients withPh+ essential thrombocythemia were found to have theb3a2variant [48–50].

3. An overview: Bcr–Abl structure and signaling

The Bcr–Abl fusion protein acts as an onco-protein byactivating several signaling paths that lead to transfor-mation. Among these are Myc, Ras, c-Raf, MAPK/ERK,SAPK/JNK, Stat, NFKB, PI-3 kinase, and c-Jun [51–61].Inhibition of Ras [62], Raf [63], PI3K [64], Akt [65],Jun [66,67], and Myc [68,69] impairs Bcr–Abl mediatedtransformation. The oncogenic ability of Bcr–Abl requiresderegulated tyrosine kinase activity which leads to recruit-

Page 3: Bcr-Abl Variants - Biological and Clinical Aspects

A.S. Advani, A.M. Pendergast / Leukemia Research 26 (2002) 713–720 715

ment of adaptor molecules, phosphorylation of signalingmolecules, and activation of downstream signaling events[70–73].

Structurally, Bcr–Abl contains multiple domains (Fig. 1).The Abl sequences encode Src-homology (SH3 and SH2)domains, tyrosine kinase domain, DNA-binding domain,actin-binding domain, nuclear localization signals, andnuclear export signal [3,74]. The Bcr region contains acoiled-coil oligomerization domain, serine/threonine kinasedomain, pleckstrin homology (PH) domain, a Dbl/cdc24guanine nucleotide exchange factor homology domain, sev-eral serine/threonine and tyrosine phosphorylation sites, andbinding sites for the Abl SH2 domain, Grb2, and 14-3-3proteins [3,75,76].

The SH2 domain of Bcr–Abl recruits signaling proteinssuch as p62dok, c-Cbl, and Rin1 [3,77–85]. Binding andphosphorylation of these molecules may be functionallyimportant as SH2 mutations in Bcr–Abl affect the courseof disease in biological models [85]. The actin bindingdomain directly links Bcr–Abl to the cytoskeleton, and fa-cilitates tyrosine phosphorylation of cytoskeletal proteins[86].

Expression of the Bcr–Abl kinases upregulates cell pro-liferation [68,69,87–91], decreases apoptosis [87,92–94],increases cytokine-independent growth [4,95,96], decreasesadhesion to the bone marrow stroma [4,87], and producescytoskeletal abnormalities [4,87].

4. Bcr–Abl variants: molecular structure

Bcr–Abl oncogenes differ in the amount of Bcr includedin the fusion protein, and are formed by joining of vari-ous amounts of theBcr gene to the sameAbl sequences.This difference in structure influences the biological andclinical phenotypes associated with theBcr–Abl variants[2]. Splicing at the m, M, or� breakpoints inBcr pro-duces three distinct protein products, namely, p185 (e1a2junction), p210 (b2a2 or b3a2 junction), and p230 (e19a2junction) (Fig. 1) [2,97]. P185 encodes the dimerizationand SH2- binding domains of Bcr [98,99]. P210 has in ad-dition to the domains present in p185, the Bcr PH and Dbldomains [43,100]. Finally, P230 has the largest amount ofBcr sequence, including the calcium/phospholipid bindingdomain and the first third of the domain associated withGTPase activity for p21 (GAP–Rac) [30]. The inclusion ofthe Dbl/PH domains in p210 and p230 may contribute to thegranulocytic differentiation associated with these variants.

5. Bcr–Abl proteins: biological/laboratory correlates

Although the three Bcr–Abl kinases (p185, p210, andp230) are similar in structure, they exhibit distinct proper-ties. When primary mouse bone marrow cells are infectedwith the different Bcr–Abl variants, and cultured in the

presence of cytokines and stroma, p185 cultures differ-entiate into a lymphoid lineage, whereas p210 and p230become myeloid [100]. In vitro, p185 has increased abil-ity to stimulate expansion of lymphoid cells, compared top210 [101]. P230 Bcr–Abl cultures require cytokines foroptimal growth, whereas p185 and p210 Bcr–Abl culturesare cytokine-independent for growth and survival [100].P185 Bcr–Abl has a greater tumorigenic potential thanp210 or p230 Bcr–Abl [100,102]. In soft agar assays, p185is 100-fold more effective than p210 at eliciting transfor-mation [73]. It was reported that SCID mice injected withp185-expressing primary bone marrow cells developedpre-B cell tumors, whereas no tumors developed in miceinjected with p210- or p230-expressing cells [100]. Millerand Pear have also shown a less aggressive leukemia asso-ciated with p230 Bcr–Abl in 5 FU-lethally irradiated mice[100]. All mice injected with the three Bcr–Abl variantsdeveloped a myeloproliferative disease, but there was anincreased latency in p230- injected mice (27 weeks) versus4–5 weeks in the p185- and p210-injected mice [100].

There are several possibilities to explain the different bio-logical effects of the three Bcr–Abl kinases. Li et al. haveshown that p230 exhibits lower intrinsic tyrosine activitythan p185 and p210 [102]. P210 also has been shown to havedecreased tyrosine kinase activity than p185 [73]. In addi-tion, Nishimura et al. have shown that different sets of tyro-sine phosphorylated proteins may exist in p185 versus p210leukemic cell lines [103]. A 36 kDa protein was detected inK562 cells (p210 cell line), but not MR87 (p185 cell line);and a 62 kDa protein was present in MR87 but not in K562[103]. The 62 kDa protein was a Gap-like protein, and is a cy-toskeletal target for tyrosine kinases [103]. Hef1, a memberof the�1 integrin signaling pathway, is tyrosine phosphory-lated in p185, but not p210 [104]. Tyrosine phosphorylationof these proteins, may lead to differential interaction withthe cytoskeleton or altered downstream signaling. AlthoughStat5, Stat1, and Stat3 are constitutively activated by tyro-sine phosphorylation in p185- and p210-expressing cells,p185 but not p210 promotes activation of Stat6 [59]. Addi-tionally, it was recently demonstrated that the SH2 domainis required for efficient induction of CML-like disease inmice by p210, but not p185, further suggesting a differencein signaling between the two variants [85].

It is possible that differences in the content of the Bcrregion itself may lead to distinct clinical disease [105].Additional Bcr sequences, such as Dbl/PH in p210 andGap–Rac in p230 may allow for increased differentiationalong the myeloid lineage [2,105,106]. The Dbl/PH do-mains present in p210 but not p185, may also contributeto the stabilization of actin fibers [107,108]. Dbl is knownto activate Rho, and activated Rho promotes the formationof actin stress fibers [109,110]. It has been reported that aBcr–Abl fusion containing Bcr (amino acids 1–191) can ef-ficiently transform rat 1-myc cells, but fusion of additionalBcr sequences present in p210 causes decreased disorgani-zation of the actin cytoskeleton [107]. Alternatively, it is

Page 4: Bcr-Abl Variants - Biological and Clinical Aspects

716 A.S. Advani, A.M. Pendergast / Leukemia Research 26 (2002) 713–720

possible that Bcr breakpoints that yield p185 may occurpreferentially in immature B cells, and breakpoints thatproduce p210, in hematopoietic stem cells [105]. The latterpoint is supported by data from Li et al. [102]. In a murinebone marrow transplantation model, p185, p210, and p230were all equally potent in inducing a CML-like disease,when 5 FU donors were used. The proviral integrationsite was polyclonal, implicating a multipotential target cell[102]. When no 5 FU was used, a mixture of CML, ALL,and macrophage tumors was observed [102]. P185 in thesemice induced lymphoid leukemias with a shorter latencythan p210 or p230. In addition, lymphoid leukemias wereoligoclonal, suggesting a lineage restricted target cell [102].This data, however, has not been supported by analysis ofpatients with Ph+ ALL. Adult patients with Ph+ ALL havebeen reported to have a multipotential stem cell target basedon examination of metaphases of individual colony form-ing unit-erythroid (CFU-erythroid) and CFU-granulocytemacrophage (CFU-GM) [29,111,112]. This suggests theremay differences in the biology of Bcr–Abl in patients versusanimal models.

6. Molecular changes: blastic phase of CML

CML eventually progresses to a blastic phase, reminis-cent of acute leukemia that is much more treatment resistant[13,113]. The molecular changes that accompany this eventhave been of interest, but are not consistent. Progression toblast crisis has been shown to correlate with the addition ofcytogenetic abnormalities such as 22q−, loss of the Y chro-mosome, isochromosome 17, trisomies-8, -9, -19, an addi-tional Ph chromosome, translocation t(3;21), or inversion ofchromosome 16 [13,114,115]. Specific mutations have beenobserved inRas, p53, Myc, p16, andRb [116–121]. In addi-tion, increased transcription ofBcr–Abl, h-Ras, andc-Mycis detected in many clinical cases of accelerated phase CML[122–130].

In some cases, alterations in known oncogenes/tumorsuppressers correspond to the cytogenetic changes seenabove. For example,p53 is located on chromosome 17,Rbon chromosome 13,c-Myc on chromosome 8,p16 on chro-mosome 9, andAML/EV-1 on chromosomes 3 and 21 [127].In addition, AXL, a putative receptor with tyrosine kinaseactivity, located on chromosome 19, may be involved in theprogression of CML to blastic phase [21,128–130].

7. Current treatment implications

Traditionally, CML in the chronic phase has been treatedwith a combination of hydroxyurea and interferon, with a73% hematologic response rate and 58% rate of cytogeneticremissions [13,131,132]. This has been slightly improvedwith the addition of cytarabine [13]. Despite this, the 5year survival rates are 57% [131,132], so allogeneic bone

marrow transplant (BMT) has been the treatment of choicefor younger patients with an HLA matched identical sib-ling, showing a 70% cure rate [13,133]. However, only aminority of patients are transplanted because of age, or lackof an HLA-matched identical sibling [13]. After transplant,PCR forBcr–Abl has been used to monitor patient’s diseasestatus. Repeated PCR positivity at greater than 6 monthspost-transplant or increasing values based on quantitativePCR tend to correlate with relapse [13,134,135]. Donorlymphocyte infusions have been found to be the most ef-fective form of adoptive salvage immunotherapy in patientswho relapse post-transplant [7,136].

Other treatments for CML have been investigated includ-ing antisense oligonucleotides, immunotherapy, farnesyltransferase inhibitors, and tyrosine kinase inhibitors [87].The tyrosine kinase inhibitors have been of particular interestbecause numerous studies have demonstrated that tyrosinekinase activity is required for the transforming capacity ofBcr–Abl [73,87,137]. In the mid-1990s, signal transductioninhibitor 571 (STI-571) (Gleevec), a phenylaminopyrimidederivative with potent tyrosine kinase inhibition and selec-tivity for Abl, c-Kit, and platelet-derived growth factor wasdeveloped [87,138]. STI-571 binds to a pocket of the cat-alytic domain of the Abl tyrosine kinase and competitivelyinhibits binding of adenosine triphosphate (ATP), therebyresulting in inhibition of autophosphorylation and inhibitionof substrate phosphorylation [139].

Pre-clinically, it was shown that STI-571 caused adose-dependent inhibition of 32D 210 cells injected intomice [140]. Subsequently, a phase 1 trial was carried outwith 54 patients with CML refractory to interferon-�. Ofthese patients, 23/24 patients receiving a dose of 300 mg orhigher underwent a complete hematologic remission, and8/24 had a complete cytogenetic remission [141]. Currently,a phase 3 study is underway comparing STI-571 versusinterferon-� +/− cytarabine as front-line therapy for CML[87].

STI-571 was also used in 33 patients with Ph+ acuteleukemia. Both blast crisis and Ph+ ALL are difficultto treat. Patients with a myeloid blast crisis treated withSTI-571 had a 55% response rate and 22% complete remis-sion rate. Patients with lymphoid blast crisis had an 82%response rate and 55% complete remission rate [142]. Theseresults are surprisingly encouraging, and suggest that eitherthe Bcr–Abl tyrosine kinase activity is necessary for blastictransformation of that other signaling pathways that areactivated during blast phase are inhibited by STI-571 [143].

Despite enthusiasm based on the above results, the emer-gence of STI-571 resistance in patients with acute leukemia,and the isolation of Ph+ leukemia cell lines that are resis-tant to STI-571 highlights the need for combination therapy[144,145]. The molecular mechanism of resistance includesincreasedBcr–Abl messenger RNA (mRNA), increased pro-tein, amplification of the transgene, and point mutationswithin the Abl kinase domain ofBcr–Abl [144–146]. Cur-rently, trials with ST-571 combined with chemotherapy for

Page 5: Bcr-Abl Variants - Biological and Clinical Aspects

A.S. Advani, A.M. Pendergast / Leukemia Research 26 (2002) 713–720 717

Ph+ leukemias are underway. In addition, other moleculartargeted therapies are being examined. Second generationtyrosine kinase inhibitors are being developed for testing[87]. An alternative approach to inhibition of the kinase hasbeen to develop an Abl-targeted tyrosine phosphatase [147].

Because Ras activation is thought to be important inBcr–Abl mediated transformation, farnesyl transferase in-hibitors have also been of particular interest [148]. Thefarnesyl transferase inhibitor, SCHH66336 has been de-signed to block both mutant and wild-type Ras signaling intransformed cells [148]. In a murine ALL model, it has beenshown to revert early signs of leukemia and significantlyprolong survival [148]. Currently, it is being investigated inclinical trials.

The above therapies are illustrative of how understandingthe molecular biology of a disease can contribute to elegantand successful molecular targeted therapies. It is likely thisparadigm will be applied to other diseases.

Acknowledgements

A.S. Advani is supported by National Institutes of HealthTraining Grant 2-T32-HC07057-26. A.M. Pendergast is sup-ported by National Institutes of Health grants CA61033 andCA70940.

References

[1] Rowley JD. A new consistent chromosomal abnormality in chronicmyelogenous leukemia identified by quinacrine fluorescence andGiesma staining. Nature 1973;243:290.

[2] Melo JV. The diversity of BCR–ABL fusion proteins and theirrelationship to leukemia phenotype. Blood 1996;88:2375.

[3] Zhang X, Rong R, Hao SX, Pear WS, Ren R. The SH2 domainof Bcr–Abl is not required to induce a murine myeloproliferativedisease; however, SH2 signaling influences disease latency andphenotype. Blood 2001;97:277.

[4] Sawyers CL. Chronic myeloid leukemia. N Engl J Med 1999;340:1330.

[5] Goldman JM. Chronic myeloid leukemia. Curr Opin Hematol1997;4:277.

[6] Faderl S, Talpaz M, Estrov Z, O’Brien S, Kurzrock R, KantarjianHM. The biology of chronic myeloid leukemia. N Engl J Med1999;341:164.

[7] Faderl S, Kantarjian HM, Talpaz M. Chronic myelogenousleukemia: update on biology and treatment. Oncology 1999;13:169.

[8] Kantarjian HM, Deisseroth A, Kurzrock R, et al. Chronicmyelogenous leukemia: a concise update. Blood 1993;82:691.

[9] Cortes J, Talpaz M, Kantarjian H. Chronic myelogenous leukemia:a review. Am J Med 1996;100:555.

[10] Kantarjian H, Dixon D, Keating MJ, et al. Characteristics ofaccelerated disease in chronic myelogenous leukemia. Cancer1988;61:1441.

[11] Griffin JD, Todd III RF, Ritz J, et al. Differentiation patterns inthe blastic phase of chronic myeloid leukemia. Blood 1983;61:85.

[12] Kantarjian HM, Keating MJ, Talpaz M, et al. Chronic myelogenousleukemia in blast crisis: analysis of 242 patients. Am J Med1987;83:445.

[13] Thijsen S, Schuurhuis G, van Oostveen J, Ossenkoppele G. Chronicmyeloid leukemia from basics to bedside. Leukemia 1999;13:1646.

[14] Derderian PM, Kantarjian HM, Talpaz M, O’Brien S, Cork A, EsteyE, et al. Chronic myelogenous leukemia in the lymphoid blasticphase: characteristics, treatment response, and prognosis. Am J Med1993;94:69.

[15] Kantarjian HM, Talpaz M, Kontoyiannis D, Gutterman J, KeatingMJ, Estey EH, et al. Treatment of chronic myelogenous leukemiain accelerated and blastic phases with daunorubicin, high-dosecytarabine, and granulocyte-macrophage colony-stimulating factor.J Clin Oncol 1992;10:398.

[16] Kantarjian HM, Walters RS, Keating MJ, Talpaz M, AnderssonB, Beran M, et al. Treatment of the blastic phase of chronicmyelogenous leukemia with mitoxantrone and high-dose cytosinearabinoside. Cancer 1988;62:672.

[17] Hoelzer D. Therapy and prognostic factors in adult acutelymphoblastic leukemia. Baillieres Clin Haematol 1994;7:299.

[18] Hooberman A, Westbrook C, Spino C, et al. Clinical significanceof molecular detection of theBCR–ABL fusion gene in acutelymphoblastic leukemia (ALL): a Cancer and Leukemia Group B(CALGB) study. Proc Am Soc Clin Oncol 1991;10:222.

[19] Spencer A, O’Brien SG, Goldman JM. Options for therapy inchronic myeloid leukaemia. Br J Haematol 1995;91:2.

[20] Pui CH, Evans WE. Acute lymphoblastic leukemia. N Engl J Med1998;339:605.

[21] Allen PB, Morgan GJ, Wiedemann LM. Philadelphia chromosome-positive leukemia: the translocated genes and their gene products.Baillieres Clin Haematol 1992;5:897.

[22] Reardon DA, Hanson CA, Roth MS, Castle VP. Lineage switchin Philadelphia chromosome-positive acute lymphoblastic leukemia.Cancer 1994;73:1526.

[23] Abe R, Ishibashi T, Kimura H, et al. The significance of cytogeneticfindings of erythroid colonies derived from a Ph+ ALL patient:fundamental differences between Ph+ ALL and blastic CML.Cancer Genet Cytogenet 1985;18:49.

[24] Kitano K, Sato Y, Suda T, Miura Y. Difference of cell lineageexpression of hematopoietic progenitor cells in Philadelphia-positiveacute lymphoblastic leukemia and chronic myelogenous leukemia.Br J Haematol 1988;70:21.

[25] Secker-Walker LM, Cooke HMG, Browett PJ, et al. VariablePhiladelphia breakpoints and potential lineage restriction ofbcrrearrangement in acute lymphoblastic leukemia. Blood 1988;72:784.

[26] Turhan AG, Eaves CJ, Kalousek DK, et al. Molecular analysisof clonality and bcr rearrangements in Philadelphia chromo-some-positive acute lymphoblastic leukemia. Blood 1988;71:1495.

[27] Craig JM, Hawkins JM, Yamada T. First intron and M-bcrbreakpoints are restricted to the lymphoid lineage in Philadelphiapositive acute lymphoblastic leukemia. Leukemia 1990;4:678.

[28] Dow LW, Tachibana N, Raimondi SC, Lauer SJ, Witte ON, ClarkSS. Comparative biochemical and cytogenetic studies of childhoodacute lymphoblastic leukemia with the Philadelphia chromosomeand other 22q11 variants. Blood 1989;73:1291.

[29] Tachibana N, Raimondi SC, Lauer SJ, et al. Evidence for amultipotential stem cell in some childhood Philadelphia chromo-some—positive acute lymphoblastic leukemias. Blood 1987;70:1458.

[30] Pane F, Frigeri F, Sindona M, Luciano L, Ferrara F, Cimino R, etal. Neutrophilic-chronic myeloid leukemia: a distinct disease witha specific molecular marker BCR/ABL with C3/A2 junction. Blood1996;88:2410.

[31] Tuohy EL. A case of splenomegaly with polymorphonuclearneutrophil hyperleukocytosis. Am J Med Sci 1920;18:160.

[32] Meyer S, Feremans W, Cantiniaux B, Capel P, Huygen R, DicatoM. Successful alpha-2b-interferon therapy for chronic neutrophilicleukemia (letter). Blood 1993;82:1035.

[33] Kwong YL, Cheng G. Clonal nature of chronic neutrophilicleukemia (letter). Blood 1993;82:1035.

[34] Melo JV. Bcr–Abl gene variants. Baillieres Clin Haematol 1997;10:203.

Page 6: Bcr-Abl Variants - Biological and Clinical Aspects

718 A.S. Advani, A.M. Pendergast / Leukemia Research 26 (2002) 713–720

[35] Melo JV, Myint H, Galton DA, Goldman JM. P190 BCR–ABLchronic myeloid leukemia: the missing link with chronic myelo-monocytic leukemia? Leukemia 1994;8:208.

[36] Kantarjian HM, Talpaz M, Dhingra K, Estey E, Keating MJ, Ku S,et al. Significance of the P210 versus P190 molecular abnormalitiesin adults with Philadelphia chromosome-positive acute leukemia.Blood 1991;78:2411.

[37] Saglio G, Pane F, Gottardi E, Frigeri F, Buonaiuto MR, GuerasioA, et al. Consistent amounts of acute leukemia-associatedP190BCR/ABL transcripts are expressed by chronic myelogenousleukemia pateints at diagnosis. Blood 1996;87:1075.

[38] Saglio G, Pane F, Martinelli G, Guerrasio A. BCR/ABL re-arrangement and leukemia phenotype. Leukemia 1999;13(Suppl1):S96.

[39] van Rhee F, Hochhaus A, Lin F, Melo JV, Goldman JM, CrossNCP. P190 BCR–ABL mRNA is expressed at low levels inp210-positive chronic myeloid and acute lymphoblastic leukemias.Blood 1996;87:5213.

[40] Mills KI, MacKenzie ED, Birnie GD. The site of the breakpointwithin the bcr is a prognostic factor in Philadelphia-positive CMLpatients. Blood 1988;72:1237.

[41] Mills KI, Sproul AM, Leibowitz D, Burnett AK. Mappingof breakpoints, and relationship toBCR–ABL RNA expression,in Philadelphia-chromosome-positive chronic myeloid leukemiapatients with a breakpoint around exon 14 (b3) of theBCR gene.Leukemia 1991;5:937.

[42] Elliott SL, Taylor KM, Taylor DL, Rodwell RL, Williams BF,Shuttlewood MM, et al. Cytogenetic response to alpha-interferonis predicted in early chronic phase chronic myeloid leukemia byM-bcr breakpoint location. Leukemia 1995;9:946.

[43] Shepherd P, Suffolk R, Halsey J, Allan N. Analysis of molecularbreakpoint and m-RNA transcripts in a prospective randomizedtrial of interferon in chronic myeloid leukemia: no correlation withclinical features, cytogenetic response, duration of chronic phase,or survival. Br J Haematol 1995;89:546.

[44] Fioretos T, Nilsson PG, Aman P, Heim S, Kirstoffersson U, MalmC, et al. Clinical impact of breakpoint position within M-bcr inchronic myeloid leukemia. Leukemia 1993;7:1225.

[45] Rozman C, Urbano Ispizua A, Cervantes F, Rozman M, ColomerD, Feliz P, et al. Analysis of the clinical relevance of the breakpointlocation within M-bcr and the type of chimeric mRNA in chronicmyelogenous leukemia. Leukemia 1995;9:1104.

[46] Inokuchi K, Inoue T, Tojo A, Futaki M, Miyake K, Yamada T,et al. A possible correlation between the type ofbcr–abl hybridmessenger RNA and platelet count in Philadelphia-positive chronicmyelogenous leukemia. Blood 1991;78:3125.

[47] Ardern JC, Speak J, Hyde K, Hunt LP, Lawson R, Gorst DW, etal. Molecular analysis in chronic granulocytic leukemia: location ofbreakpoints within M-bcr and relationship with presentation plateletcounts. Clin Lab Hematol 1993;15:253.

[48] Martiat P, Ifrah N, Rassool F, Morgan G, Giles F, Gow J, etal. Molecular analysis of Philadelphia positive essential throm-bocythemia. Leukemia 1989;3:563.

[49] Cervantes F, Urbano Ispizua A, Villamor N, Feliu E, Milla F, LopezGuillermo A, et al. Ph-positive chronic myeloid leukemia mimickingessential thrombocythemia and termination into megakaryoblasticblast crisis: report of two cases with molecular studies. Leukemia1993;7:327.

[50] Morris CM, Heisterkamp N, Groffen J, Fitzgerald PH. EntireABL gene is joined with 5′-BCR in some patients withPhiladelphia-positive leukemia. Blood 1991;78:1078.

[51] Cortez D, Reuther G, Pendergast AM. The Bcr–Abl tyrosine kinaseactivates mitogenic signaling pathways and stimulates G1-to-S phasetransition in hematopoietic cells. Oncogene 1997;15:2333.

[52] Pendergast AM, Quilliam LA, Cripe LD, Bassing CH, Dai Z, LiN, et al. BCR–ABL-induced oncogenesis is mediated by directinteraction with the SH2 domain of the GRB-2 adaptor protein.Cell 1993;75:175.

[53] Cortez D, Kadlec L, Pendergast AM. Structural and signalingrequirements for BCR–ABL-mediated transformation and inhibitionof apoptosis. Mol Cell Biol 1995;15:5531.

[54] Salomoni P, Wasik MA, Riedel RF, Reiss K, Choi JK, SkorskiT, et al. Expression of constitutively active Raf-1 in the mito-chondria restores antiapoptotic and leukemogenic potential ofa transformation-deficientBCR/ABL mutant. J Exp Med 1998;187:1995.

[55] Okuda K, Matulonis U, Salgia R, Kanakura Y, Druker B, GriffinJD. Factor independence of human myeloid leukemia cell lines isassociated with increased phosphorylation of the proto-oncogeneRaf-1. Exp Hematol 1994;22:1111.

[56] Burgess GS, Williamson EA, Cripe LD, Litz-Jackson S, Bhatt JA,Stanley K, et al. Regulation of thec-jun gene in p210 BCR–ABLtransformed cells corresponds with activity of JNK, the c-junN-terminal kinase. Blood 1998;92:2450.

[57] Sawyers CL, Callahan W, Witte ON. Dominant negative MYCblocks transformation byABL oncogenes. Cell 1992;70:901.

[58] Shuai K, Halpern J, ten Hoeve J, Rao X, Sawyers CL. Constitutiveactivation of STAT5 by theBCR–ABL oncogene in chronicmyelogenous leukemia. Oncogene 1996;13:247.

[59] Ilaria Jr RL, Van Etten RA. P210 and P190(BCR/ABL) inducethe tyrosine phosphorylation and DNA binding activity of multiplespecific STAT family members. J Biol Chem 1996;271:31704.

[60] Skorski T, Bellacosa A, Nieborowska-Skorska M, Majewski M,Martinez R, Choi JK, et al. Transformation of hematopoietic cellsby BCR/ABL requires activation of a PI-3k/Akt-dependent pathway.EMBO J 1997;16:6151.

[61] Reuther JY, Reuther GW, Cortez D, Pendergast AM, Baldwin JrAS. A requirement for NF-kappaB activation in Bcr–Abl-mediatedtransformation. Genes Dev 1998;12:968.

[62] Sawyers CL, McLaughlin J, Witte ON. Genetic requirement forRas in the transformation of fibroblasts and hematopoietic cells bythe Bcr–Abl oncogene. J Exp Med 1995;181:307.

[63] Okuda K, Matulonis U, Salgia R, Kanakura Y, Druker B, Griffin JD.Factor independence of human myeloid leukemia cells is associatedwith increased phosphorylation of the proto-oncogeneRaf-1. ExpHematol 1994;22:1111.

[64] Skorski T, Kanakaraj P, Nieborowska-Skorska M, et al.Phosphatidylinositol-3 kinase activity is regulated by BCR/ABLand is required for the growth of Philadelphia chromosome-positivecells. Blood 1995;86:726.

[65] Skorski T, Bellacosa A, Nieborowska-Skorska M, et al.Transformation of hematopoietic cells by BCR/ABL requires acti-vation of a PI-3K/Akt-dependent pathway. EMBO J 1997;16:6151.

[66] Raitano AB, Halpern JR, Hambuch TM, Sawyers CL. TheBcr–Ablleukemia oncogene activates Jun kinase and requires Jun fortransformation. Proc Natl Acad Sci USA 1995;92:11746.

[67] Dickens M, Rogers JS, Cavanaugh J, et al. A cytoplasmic inhibitorof the JNK signal transduction pathway. Science 1997;277:693.

[68] Sawyers CL, Callahan W, Witte ON. Dominant negative Myc blockstransformation byABL oncogenes. Cell 1992;70:901–10.

[69] Afar DEH, Goga A, McLaughlin J, Witte ON, Sawyers CL.Differential complementation ofBcr–Abl point mutants withc-Myc.Science 1994;264:424.

[70] Dai Z, Quackenbush RC, Courtney KD, Grove M, Cortez D,Reuther GW, et al. Oncogenic Abl and Src tyrosine kinases elicitthe ubiquitin-dependent degradation of target proteins through aRas-independent pathway. Genes Dev 1998;12:1415.

[71] Evans CA, Owen-Lynch PJ, Whetton AD, Dive C. Activation ofthe Abelson tyrosine kinase activity is associated with suppressionof apoptosis in hemopoietic cells. Cancer Re 1993;53:1735.

[72] Daley GQ, Van Etten RA, Baltimore D. Induction of chronicmyelogenous leukemia in mice by theP210bcr/abl gene of thePhiladelphia chromosome. Science 1990;247:824.

Page 7: Bcr-Abl Variants - Biological and Clinical Aspects

A.S. Advani, A.M. Pendergast / Leukemia Research 26 (2002) 713–720 719

[73] Lugo TG, Pendergast AM, Muller AJ, Witte ON. Tyrosine kinaseactivity and transformation potency ofbcr–abl oncogene products.Science 1990;247:1079.

[74] Wang JY. Abl tyrosine kinase in signal transduction and cell-cycleregulation. Curr Opin Genet Dev 1993;3:35.

[75] Raitano AB, Whang YE, Sawyers CL. Signal transduction bywild-type and leukemogenic Abl proteins. Biochim Biophys Acta1997;1333:F201.

[76] Ghaffari S, Daley GQ, Lodish HF. Growth factor independence andBCR/ABL transformation: promise and pitfalls of murine modelsystems and assays. Leukemia 1999;13:1200.

[77] Pendergast AM, Muller AJ, Havlik MH, Maru Y, Witte ON.BCR sequences essential for transformation by theBCR–ABLoncogene bind to the ABL SH2 regulatory domain in anon-phosphotyrosine-dependent manner. Cell 1991;66:161.

[78] Howell BW, Gertler FB, Cooper JA. Mouse disabled (mDab1): a Srcbinding protein in neuronal development. EMBO J 1997;16:121.

[79] Yamanashi Y, Baltimore D. Identification of the Abl- andrasGAP-associated 62 kDa protein as a docking protein. Dok Cell1997;88:205.

[80] Carpino N, Wisniewski D, Strife A, et al. P62(dok): aconstitutively tyrosine-phosphorylated, GAP-associated protein inchronic myelogenous leukemia progenitor cells. Cell 1997;88:197.

[81] Bhat A, Johnson KJ, Oda T, Corbin AS, Druker BJ. Interactionsof p62(dok) with p210(bcr–abl) and Bcr–Abl associated proteins. JBiol Chem 1998;273:32360.

[82] Bhat A, Kolibaba K, Oda T, Ohno-Jones S, Heaney C, Druker BJ.Interactions of CBL with BCR–ABL and CRKL in BCR–ABL-transformed myeloid cells. J Biol Chem 1997;272:16170.

[83] Afar DE, Han L, McLaughlin J, et al. Regulation of the oncogenicactivity of BCR–ABL by a tightly bound substrate protein RIN1.Immunity 1997;6:773.

[84] Kapeller R, Moriarty A, Strauss A, et al. Tyrosine phosphorylationof tub and its association with Src homology 2 domain-containingproteins implicate tub in intracellular signaling by insulin. J BiolChem 1999;274:24980.

[85] Roumiantsev S, de Aos IE, Varticovski L, Ilaria R, Van Etten RA.The src homology 2 domain of Bcr/Abl is required for efficientinduction of chronic myeloid leukemia-like disease in mice but notfor lymphoid leukemogenesis or activation of phosphatidylinositol3-kinase. Blood 2001;97:4.

[86] Heisterkamp N, Voncken JW, Senadheera D, Gonzalez-Gomez I,Reichert A, Haataja L, et al. Reduced oncogenicity of p190 Bcr/AblF-actin-binding domain mutants. Blood 2000;96:2226.

[87] Jahagirdar BN, Miller JS, Shet A, Verfaillie CM. Novel therapiesfor chronic myelogenous leukemia. Exp Hematol 2001;29:543.

[88] Afar DE, Goga A, Cohen L, et al. Genetic approaches to definingsignaling by the CML-associated tyrosine kinase BCR–ABL. ColdSpring Harbor Symp Quant Biol 1994;59:589.

[89] Afar DE, McLaughlin J, Sherr CJ, Witte ON, Roussel MF. Signalingby ABL oncogenes through cyclin D1. Proc Natl Acad Sci USA1995;92:9540.

[90] Jiang Y, Zhao RC, Verfaillie CM. Abnormal integrin-mediatedregulation of chronic myelogenous leukemia CD34+ cell prolifer-ation: BCR/ABL up-regulates the cyclin-dependent kinase inhibitor,p27Kip, which is relocated to the cell cytoplasm and incapableof regulating cdk2 activity. Proc Natl Acad Sci USA 2000;97:10538.

[91] Sawyers CL. Molecular consequences of the BCR–ABL trans-location in chronic myelogenous leukemia. Leuk Lymphoma1993;11:101.

[92] Bedi A, Zehnbauer BA, Barber JP, Sharkis SJ, Jones RJ. Inhibitionof apoptosis by BCR–ABL in chronic myeloid leukemia. Blood1994;83:2038.

[93] McGahon A, Bissonnette R, Schmitt M, Cotter KM, Green DR,Cotter TG. BCR–ABL maintains resistance of chronic myelogenousleukemia cells to apoptotic cell death. Blood 1994;83:1179.

[94] Cotter TG. BCR–ABL: an anti-apoptosis gene in chronic myelo-genous leukemia. Leuk Lymphoma 1995;18:231.

[95] McLaughlin J, Chianese E, Witte ON. In vitro transformation ofimmature hematopoietic cells by theP210 BCR/ABL oncogeneproduct of the Philadelphia chromosome. Proc Natl Acad Sci USA1987;84:6558.

[96] Gishizky ML, Witte ON. Initiation of dysregulated growth ofmultipotent progenitor cells by bcr–abl in vitro. Science 1992;256:836.

[97] Chissoe SL, Bodenteich A, Wang YF, Wang YP, Burian D, CliftonSW, et al. Sequence and analysis of the humanABL gene, theBCR gene, and regions involved in the Philadelphia chromosomaltranslocation. Genomics 1995;27:67.

[98] Kurzrock R, Shtalrid M, Romero P, et al. A novel c-abl proteinproduct in Philadelphia-positive acute lymphoblastic leukemia.Nature 1987;325:631.

[99] Chan L, Karhi KK, Rayter SI, et al. A novel abl protein expressedin Philadelphia chromosome positive acute lymphoblastic leukemia.Nature 1987;325:635.

[100] Quackenbush RQ, Reuther GW, Miller JP, Courtney KD, Pear WS,Pendergast AM. Analysis of the biologic properties of p230 Bcr–Ablreveals unique and overlapping properties with the oncogenic p185and p210 Bcr–Abl tyrosine kinases. Blood 2000;95:2913.

[101] McLaughlin J, Chianese E, Witte ON. Alternative forms of theBCR–ABL oncogene have quantitatively different potencies forstimulation of immature lymphoid cells. Mol Cell Biol 1989;9:1866.

[102] Li S, Ilaria Jr RL, Million RP, Daley GQ, Van Etten RA. TheP190,P210, andP230 forms of theBCR/ABL oncogene induce a similarchronic myeloid leukemia-like syndrome in mice but have differentlymphoid leukemogenic activity. J Exp Med 1999;189:1399.

[103] Nishimura J, Shibata K, Sadamura S, Takahira H, Hirata J,Umemura T, et al. Differences in tyrosine phosphorylated proteinsbetween cells expressing P210bcr/abl and P190bcr/abl. Int JHematol 1992;55:227.

[104] de Jong R, van Wijk A, Haataja L, Heisterkamp N, Groffen J.BCR/ABL-induced leukemogenesis causes phosphorylation of Hef1and its association with Crkl. J Biol Chem 1997;272:32649.

[105] Okuda K, Golub TR, Gilliland DG, Griffin JD. p210BCR/ABL,p190BCR/ABL, and TEL/ABL activate similar signal transductionpathways in hematopoietic cell lines. Oncogene 1996;13:1147.

[106] Didsbury J, Weber RF, Bokoch GM, Evans T, Snyderman R. Rac,a novel ras-related family of proteins that are botulinum toxinsubstrates. J Biol Chem 1989;264:16378.

[107] McWhirter JR, Wang JY. Effect of Bcr sequences on the cellularfunction of the Bcr–Abl oncoprotein. Oncogene 1997;15:1625.

[108] Daley GQ, Ben-Neriah Y. Implicating thebcr/abl gene in thepathogenesis of Philadelphia chromosome-positive human leukemia.Adv Cancer Res 1991;57:151.

[109] Chuang T-H, Xu X, Kaartinen V, Heisterkamp N, Groffen J,Bokoch G. Abr and Bcr are multifunctional regulators of theRho GTP-binding protein family. Proc Natl Acad Sci USA1995;92:10282.

[110] Ridley AJ, Hall A. The small GTP-binding protein rho regulatesthe assembly of focal adhesions and actin stress fibers in responseto growth factors. Cell 1992;70:389.

[111] Estrov Z, Talpaz M, Kantarjian H, Zipf T, McClain KL, KurzrockR. Heterogeneity in lineage derivation of Philadelphia-positiveacute lymphoblastic leukemia expressing p190 BCR–ABL or p210BCR–ABL: determination by analysis of individual colonies withthe polymerase chain reaction. Cancer Res 1993;53:3289.

[112] Kalousek DK, Dube ID, Eaves CJ, Eaves AC. Cytogenetic studiesof hemopoietic colonies from patients with an initial diagnosis ofacute lymphoblastic leukemia. Br J Hematol 1988;70:5.

[113] Kantarjian HM, Talpaz M. Definition of the accelerated phase ofchronic myelogenous leukemia. J Clin Oncol 1988;6:18.

[114] Nanjangud G, Kadam PR, Saikia T, Bhisey AN, Kumar A, GopalR, et al. Karyotypic findings as an independent prognostic markerin chronic myeloid leukemia blast crisis. Leuk Res 1994;18:385.

Page 8: Bcr-Abl Variants - Biological and Clinical Aspects

720 A.S. Advani, A.M. Pendergast / Leukemia Research 26 (2002) 713–720

[115] Asou N, Sanada I, Tanaka K, Hidaka M, Suzushima H, MatsuzakiH, et al. Inversion of chromosome 16 and bone marrow eosinophiliain a myelomonocytic transformation of chronic myeloid leukemia.Cancer Genet Cytogenet 1992;61:197.

[116] Chopra R, Pu QQ, Elefanty AG. Biology of BCR–ABL. Blood Rev1999;13:211.

[117] Cogswell PC, Morgan R, Dunn M. Mutations in theras proto-oncogenes in chronic myelogenous leukemia: a high frequencyof ras mutations in bcr/abl rearrangement-negative chronicmyelogenous leukemia. Blood 1989;74:2629.

[118] Feinstein E, Cimino G, Gale RP. P53 in chronic myelogenousleukemia in acute phase. Proc Natl Acad Sci USA 1991;88:6293.

[119] Sawyers CL. The role of myc in transformation by BCR–ABL.Leuk Lymphoma 1993;11(Suppl 1):45.

[120] Ahuja H, Bar-Eli M, Arlin Z. The spectrum of molecular alterationsin the evolution of chronic myelocytic leukemia. J Clin Invest1991;87:2042.

[121] Towatari M, Aduchi K, Kato H, Saito H. Absence of the humanretinoblastoma gene product in the megakaryoblastic crisis ofchronic myelogenous leukemia. Blood 1991;78:2178.

[122] Cambier N, Chopra R, Strasser A, Metcalf D, Elefanty AG.BCR–ABL activates pathways mediating cytokine independenceand protection against apoptosis in murine hematopoietic cells in adose-dependent manner. Oncogene 1998;16:335.

[123] Collins SJ, Kubonishi I, Miyoshi I, Groudine MT. Alteredtranscription of thec-abl oncogene in K-562 and other chronicmyelogenous leukemia cells. Science 1984;225:72.

[124] Andrews DF, Collins SJ. Heterogeneity in expression of thebcr–ablfusion transcript in CML blast crisis. Leukemia 1987;1:718.

[125] Gaiger A, Henn T, Horth E, Geissler K, Mitterbauer G,Maier-Dobersberger T, et al. Increase ofbcr–abl chimeric mRNAexpression in tumor cells of patients with chronic myeloid leukemiaprecedes disease progression. Blood 1995;86:2371.

[126] Beck Z, Bacsi A, Kovacs E, Kiss J, Kiss A, Balogh E, et al.Changes in oncogene expression implicated in evolution of chronicgranulocytic leukemia from its chronic phase to acceleration. LeukLymphoma 1998;30:293.

[127] Mitelman F. They cytogenetic scenario of chronic myeloid leukemia.Leuk Lymphoma 1993;11:11.

[128] Liu E, Hjelle B, Bishop JM. Transforming genes in chronicmyelogenous leukemia. Proc Natl Acad Sci USA 1988;85:1952.

[129] O’Bryan JP, Frye RA, Cogswell PC, et al.Axl, a transforming geneisolated from primary human myeloid leukemia cells, encodes anovel receptor tyrosine kinase. Mol Cell Biol 1991;11:501.

[130] Rowley DJ, Testa JR. Chromosome abnormalities in malignanthematologic disease. Adv Cancer Res 1982;36:103.

[131] Mauro MJ, Druker BJ. Chronic myelogenous leukemia. Curr OpinOncol 2001;13:3.

[132] Chronic Myeloid Leukemia Trialists’ Cooperative Group. Inter-feron-alfa versus chemotherapy for chronic myeloid leukemia:a meta-analysis of seven randomized trials. J Natl Cancer Inst1997;89:1616.

[133] Gratwohl A, Hermans J. Allogeneic bone marrow transplantation forchronic myeloid leukemia. Working Party Chronic Leukemia of the

European Group for Blood and Marrow Transplantation (EBMT).Bone Marrow Transplant 1996;17(Suppl 3):S7.

[134] Radich JP, Gehly G, Gooley T, Bryant E, Clift RA, CollinsS, et al. Polymerase chain reaction detection of thebcr–ablfusion transcript after allogeneic marrow transplantation for chronicmeyloid leukemia—results and implications in 346 patients. Blood1995;85:2632.

[135] Lion T, Gaiger A, Henn T, Horth E, Haas OA, Geissler K, et al.Use of quantitative polymerase chain reaction to monitor residualdisease in chronic myelogenous leukemia during treatment withinterferon. Leukemia 1995;9:1353.

[136] Giralt SA, Kolb HJ. Donor lymphocyte infusions. Curr Opin Oncol1996;8:96.

[137] Oda T, Tamura S, Matsuguchi T, Griffin JD, Druker BJ. TheSH2 domain of ABL is not required for factor-independent growthinduced by BCR–ABL in a murine myeloid cell line. Leukemia1995;9:295.

[138] Buchdunger E, Zimmermann J, Mett H, et al. Inhibition ofthe Abl protein-tyrosine kinase in vitro and in vivo by a2-phenylaminopyrimidine derivative. Cancer Res 1996;56:100.

[139] Schindler T, Bornmann W, Pellicena P, Miller WT, Clarkson B,Kuriyan J. Structural mechanism for STI-571 inhibition of abelsontyrosine kinase. Science 2000;289:1938.

[140] Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selectiveinhibitor of the Abl tyrosine kinase on the growth of Bcr–Ablpositive cells. Nat Med 1996;2:561.

[141] Druker BJ, Sawyers CL, Talpaz M. Phase I trial of specificabl tyrosine kinase inhibitor, CGP 57148, in interferon refractorychronic myelogenous leukemia patients. Proc ASCO 1999;8:24a(abstract).

[142] Talpaz M, Sawyers CL, Kantarjian HM, et al. Activity of an ABLspecific tyrosine kinase inhibitor in patients with BCR–ABL positiveacute leukemia, including chronic myelogenous leukemia in blastcrisis. Proc ASCO 2000;19:4a (abstract).

[143] Goldman JM, Melo JV. Targeting the BCR–ABL tyrosine kinase inchronic myeloid leukemia (letter). N Engl J Med 2001;344:1084.

[144] Mahon FX, Deininger MW, Schultheis B, Chabrol J, Reiffers J,Goldman JM, Melo JV, Selection and characterization of BCR–ABLpositive cell lines with differential sensitivity to the tyrosine kinaseinhibitor STI571: diverse mechanisms of resistance. Blood. 2000;96(3):1070–9.

[145] Weisberg E, Griffin JD. Mechanism of resistance to theABL tyrosine kinase inhibitor STI571 in BCR/ABL-transformedhematopoietic cell lines. Blood 2000;95:3498.

[146] Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, RaoPN, et al. Clinical resistance in STI-571 cancer treatment causedby Bcr–Abl gene mutation or amplification. Science 2001;293:876.

[147] Lim Y-M, Wong S, Lau G, Witte ON, Colicelli J. BCR/ABLinhibition by an escort/phosphatase fusion protein. Proceedings ofthe National Academy of Sciences of the USA. Proc Natl AcadSci USA 2000;97:12233.

[148] Peters DG, Hoover RR, Gerlach MJ, Koh EY, Zhang H, Choe K, etal. Activity of the farnesyl protein transferase inhibitor SCH66336against BCR/ABL-induced murine leukemia and primary cells frompatients with chronic myeloid leukemia. Blood 2001;97:1404.