Throughout its history, chronic myeloid leukemia (CML) has set precedents for cancer research and therapy. These range from the identification of the first specific chromosomal abnormality associated with cancer to the development of imatinib as a specific, targeted therapy for the disease. The successful development of imatinib as a therapeutic agent for CML can be attributed directly to decades of scientific discoveries. These discoveries determined that the BCR-ABL tyrosine kinase is the critical pathogenetic event in CML and an ideal target for therapy. This was confirmed in clinical trials of imatinib, with imatinib significantly improving the long-term survival of patients with CML. Continuing in this tradition of scientific discoveries leading to improved therapies, the understanding of resistance to imatinib has rapidly led to strategies to circumvent resistance. Continued studies of hematologic malignancies will allow this paradigm of targeting molecular pathogenetic events to be applied to many additional hematologic cancers.

The hematologic malignancies have frequently been at the forefront of the development of cancer therapies. Many chemotherapeutic agents were first used in hematologic malignancies and were subsequently found to have activity in solid tumor. Thus, the first chemotherapeutic agent, nitrogen mustard, was initially used in patients with Hodgkin disease and lymphocytic leukemias. Methotrexate and adriamycin were first developed in childhood leukemia. Continuing in this tradition, imatinib, the first tyrosine kinase inhibitor, was developed for chronic myeloid leukemia (CML) and has also been found to be active in gastrointestinal stromal tumors and some rare myeloproliferative neoplasms. Imatinib has also established the paradigm of targeting molecular pathogenetic events in cancer. The discoveries that led to the development of imatinib have paralleled much of the 50-year history of the American Society of Hematology (ASH). This began with the discovery of the Philadelphia chromosome, leading to the identification of the BCR-ABL tyrosine kinase as the causative molecular event of CML, which allowed the development of imatinib as a specific inhibitor of this kinase. Scientific discoveries have continued to be integrated into the clinical trials of imatinib, allowing a rapid understanding of mechanisms of resistance to imatinib, development of new drugs to circumvent resistance, and the successful use of imatinib in other malignancies driven by imatinib-sensitive kinases

Chronic myeloid leukemia: clinical features

CML was first described by two pathologists, Dr Rudolf Virchow and Dr John Hughes Bennett, in 1845.1,2  Although a debate ensued as to whose description was first, in a public lecture, Virchow acknowledged that Bennett, “observed a case of indubitable leukemia some months before I saw my first case.”3(p2) Of note, these first accounts of CML occurred prior to staining methods for blood, which were not developed until the late 1800s. We now know that CML is a clonal hematopoietic stem cell disorder with an annual incidence of 1 to 2 cases per 100 000 per year.

The chronic, or stable phase of CML is characterized by excess numbers of myeloid cells that differentiate and function normally. Between 90% and 95% of patients will be diagnosed in this phase of the disease. Historically, within an average of 4 to 6 years, the disease transformed through an “accelerated phase” to an invariably fatal acute leukemia, also known as blast crisis. Disease progression is likely due to the accumulation of molecular abnormalities that lead to a progressive loss of the capacity for terminal differentiation of the leukemic clone.4  With currently available therapies, however, median survival might extend to 30 years or more and some patients may never transform and may never die of their leukemia.

Historically, therapy for CML was empirically based. During the late 1800s, the mainstay of therapy for CML was Fowler solution, which was developed by Dr Thomas Fowler in the mid- 1700s. The active ingredient in Fowler solution was probably potassium arsenite and there has been a resurgence of interest in the use of arsenic preparations for CML.3,5,6  During the 1900s, radiation, busulfan, hydroxyurea, interferon-α (IFN-α), and stem cell transplantation were developed for other indications, tried broadly, and found to have activity in CML.3  Allogeneic stem cell transplantation is the only proven curative therapy, but is associated with significant morbidity and mortality.7 

Molecular pathogenesis of CML

To move from empiric therapy to rationally designed therapy requires a precise understanding of the molecular pathogenesis of a disease, and the unraveling of the molecular pathogenesis of CML began shortly after ASH was formed. In 1960, Peter Nowell and David Hungerford, working in Philadelphia, described a consistent chromosomal abnormality in patients with CML, an acrocentric chromosome that was thought to be a chromosomal deletion.8  This was the first example of a chromosomal abnormality linked to a specific malignancy. In their seminal article, the authors stated, “the findings suggest a causal relationship between the chromosomal abnormality observed and chronic granulocytic leukemia.”8  This prescient statement was met with skepticism as it was felt that the chromosome abnormality was an associated rather than causative phenomenon. As chromosomal banding techniques improved, it became apparent that the chromosome abnormality was a shortened chromosome 22. Then, in 1973, Dr Janet Rowley determined that the shortened chromosome 22, the so-called Philadelphia (Ph) chromosome, was the product of a reciprocal translocation between the long arms of chromosomes 9 and 22, t(9:22)(q34;q11)9  (Figure 1).

Figure 1

Schematic diagram of the translocation that creates the Philadelphia chromosome. The ABL and BCR genes reside on the long arms of chromosomes 9 and 22, respectively. As a result of the (9;22) translocation, a BCR-ABL gene is formed on the derivative chromosome 22 (Philadelphia chromosome). Illustration by A. Y. Chen.

Figure 1

Schematic diagram of the translocation that creates the Philadelphia chromosome. The ABL and BCR genes reside on the long arms of chromosomes 9 and 22, respectively. As a result of the (9;22) translocation, a BCR-ABL gene is formed on the derivative chromosome 22 (Philadelphia chromosome). Illustration by A. Y. Chen.

Close modal

In the 1970s and 1980s, the investigation of transforming retroviruses led to major breakthroughs in our understanding of human malignancies. These studies revealed that mutations in normal cellular genes could be oncogenic. For example, the Abelson murine leukemia virus, initially described in 1970,10  led to the identification of its transforming gene, v-ABL, and to the cloning of its normal cellular homolog, c-ABL.11-13  By mapping oncogenes to specific chromosomal locations, it was recognized that c-ABL, which normally resides on the long arm of chromosome 9, had been translocated to chromosome 22 in patients with CML.14  As the breakpoints on chromosome 22 clustered in a relatively small region that spanned 5.3 kilobases, this region was named the breakpoint cluster region, or BCR.15,16 

This confluence of the analysis of chromosomal abnormalities and transforming retoviruses allowed investigators to determine the exact molecular consequences of the (9;22) chromosomal translocation (Figure 1). Northern blots using ABL probes demonstrated a larger than normal ABL mRNA in patients with CML.17,18  This was subsequently shown to be a chimeric mRNA that was a fusion of BCR and ABL sequences.19  Similarly, a larger than normal ABL protein with tyrosine kinase activity was detected in CML cells and was identified as the product of the BCR-ABL mRNA.20-22  These discoveries provided an important link between the field of oncogenes and the biochemistry of protein kinases, as it had previously been recognized that v-ABL possessed a novel kinase activity, the ability to phosphorylate tyrosine residues.23,24  BCR-ABL is now known to have elevated tyrosine kinase activity compared with c-ABL, and the kinase activity of BCR-ABL is essential for its ability to transform cells.25,26  The leukemogenicity of BCR-ABL was confirmed in 1990 by John Groffen's laboratory and George Daley and Richard van Etten in David Baltimore's laboratory. These investigators expressed BCR-ABL in animal models and demonstrated that BCR-ABL, as the sole oncogenic event, induced leukemia,27,28  thus establishing BCR-ABL as a leukemic oncogene.

Once identified as the molecular pathogenetic event in CML, significant effort has been placed into understanding the molecular mechanism of action of BCR-ABL through the identification of signaling pathways that are impacted by BCR-ABL tyrosine kinase activity (Figure 2). Numerous substrates and binding partners of BCR-ABL have been identified and current efforts are directed at linking these pathways to the specific pathologic defects that characterize CML.29  These defects include increased proliferation or decreased apoptosis of a hematopoietic stem cell or progenitor cell leading to a massive increase in myeloid cell numbers; premature release of immature myeloid cells into the circulation, postulated to be due to a defect in adherence of myeloid progenitors to marrow stroma; and genetic instability resulting in disease progression. An example of a cellular pathway that links to an increased proliferative rate is activation of the RAS pathway. Protection from programmed cell death may be mediated in part through STAT5 up-regulation of the antiapoptotic molecule BCLXL and phosphorylation of and inactivation of the proapoptotic molecule BAD by AKT.29  CML cells also exhibit reduced adhesion to fibronectin, possibly as a downstream effect of CRKL phosphorylation. Despite the seemingly endless expansion of the list of pathways activated by BCR-ABL and the increasing complexity that is being revealed in these pathways, all of the transforming functions of BCR-ABL are dependent on its tyrosine kinase activity.26 

Figure 2

Signaling pathways impacted by BCR-ABL expression. Note that this is a simplified diagram and that many more associations between BCR-ABL and signaling proteins have been reported. Reprinted from Deininger et al.15 

Figure 2

Signaling pathways impacted by BCR-ABL expression. Note that this is a simplified diagram and that many more associations between BCR-ABL and signaling proteins have been reported. Reprinted from Deininger et al.15 

Close modal

Thus, the period from 1960 to 1990 identified BCR-ABL as an ideal therapeutic target in CML. It is expressed in all patients with CML and it has been shown to be the cause of CML. BCR-ABL functions as a constitutively activated tyrosine kinase and mutagenic analysis has shown that this activity is essential for the transforming function of the protein. For these reasons, an inhibitor of the BCR-ABL kinase would be predicted to be an effective and selective therapeutic agent for CML (Figure 3).

Figure 3

Mechanism of action of imatinib. (A) The constitutively active BCR-ABL tyrosine kinase functions by transferring phosphate from ATP to tyrosine residues on various substrates to cause excess proliferation of myeloid cells characteristic of CML. (B) Imatinib blocks the binding of ATP to the BCR-ABL tyrosine kinase, thus inhibiting kinase activity. Illustration by A. Y. Chen.

Figure 3

Mechanism of action of imatinib. (A) The constitutively active BCR-ABL tyrosine kinase functions by transferring phosphate from ATP to tyrosine residues on various substrates to cause excess proliferation of myeloid cells characteristic of CML. (B) Imatinib blocks the binding of ATP to the BCR-ABL tyrosine kinase, thus inhibiting kinase activity. Illustration by A. Y. Chen.

Close modal

Given the success of imatinib and the enormous interest in protein kinase inhibitors, it is easy to forget the degree of skepticism that kinase inhibitors faced from the scientific community and the pharmaceutical industry in the 1980s and 1990s. Much of this skepticism was due to the prevailing thought that inhibitors of ATP binding would lack sufficient target specificity to be clinically useful. Further, some of the early tyrosine kinase–null animals had an embryonic lethal phenotype that led to the belief that tyrosine kinase inhibitors would be extremely toxic. Lastly, it was viewed that targeting of a single molecular defect would not suffice to treat highly heterogeneous cancers. The concern regarding specificity was addressed in 1988, when Yaish et al published a series of compounds, known as tyrphostins, that demonstrated that specific tyrosine kinase inhibitors could be developed.30  Working independently, scientists at Ciba-Geigy (now Novartis), under the direction of Nicholas (Nick) Lydon and Alex Matter, began a kinase inhibitor program.

The group at Ciba-Geigy established collaborations with two investigators at Dana-Farber Cancer Institute from 1987 to 1990 to assist their project: Dr Thomas (Tom) Roberts, a kinase expert, and Dr Charles Stiles, an expert on the platelet-derived growth factor receptor (PDGFR), which was one of the targets selected for drug development. Critical to the success of the project was the ability to generate sufficient quantities of kinases to analyze their chemical libraries. Helen Piwnica-Worms, working in Dr Roberts' laboratory, expressed kinases in the baculovirus expression system,31  and this technology was transferred to Ciba-Geigy. A second reagent was the phosphotyrosine antibody, 4G10, that I had developed with Deborah Morrison while working in Tom's lab.32  This antibody allowed the detection of the activity of tyrosine kinases, tyrosine phosphorylation, and was essential to detecting the ability of chemical compounds to inhibit tyrosine kinases. In addition, Nick Lydon and I established a working relationship as I was the only oncologist working in Tom's lab. At the time, I was working on a variety of tyrosine kinase projects, but not CML or BCR-ABL. Nevertheless, I advised Nick that BCR-ABL would likely be the first and the best cancer in which to validate the paradigm of kinase inhibition. As Nick had a background working on v-ABL, he immediately recognized the importance of including the ABL kinase in their profiling of compounds undergoing optimization. While the Ciba-Geigy group was working to identify kinase inhibitors, I decided to combine my emerging expertise in tyrosine kinase signaling with my background in oncology and established a collaboration with Dr James Griffin to work on BCR-ABL signaling.

With essential reagents in hand, N. Lydon's group performed high-throughput screens of chemical libraries searching for compounds with kinase inhibitory activity. From this time-consuming approach, a lead compound of the 2-phenylaminopyrimidine class was identified. The activity of the 2-phenylaminopyrimidine series was optimized for various kinases by an extremely talented chemist, Joerg Zimmerman, who synthesized chemically related compounds and analyzed the relationship between their structure and activity.33-35  One compound series, optimized for activity against the PDGFR was also found by Elisabeth Buchdunger at Ciba-Geigy to possess inhibitory activity toward ABL.36 

In 1993, having moved to Oregon Health & Science University and having established various BCR-ABL–driven models, I reconnected with Nick Lydon as I was interested in finding a company with a BCR-ABL kinase inhibitor. By this point, his group had generated the 2-phenylaminopyrimidine series already noted and he sent me a variety of compounds for testing. From our testing, STI571 (Gleevec, Glivec, imatinib) was identified as the compound that was the most specific at killing CML cells.37  Imatinib ultimately emerged as the lead compound for preclinical development based on its selectivity against CML cells in vitro and its druglike attributes, including pharmacokinetic and formulation properties.38 

Preclinical studies

Our initial laboratory studies showed that imatinib had inhibitory activity against ABL and its activated derivatives v-ABL, BCR-ABL, and TEL-ABL.36,37,39  As previously discussed, imatinib was known to inhibit the PDGFR. We also demonstrated potent activity against KIT,40  with minimal activity against a broad range of other tyrosine and serine/threonine kinases, demonstrating that imatinib had a high level of selectivity.41  In cellular experiments, imatinib was shown to selectively suppress the proliferation of BCR-ABL–expressing cells in vitro.37  This selectivity was confirmed using patient cells in colony-forming assays. In these studies, imatinib caused a 92% to 98% decrease in the number of BCR-ABL–positive colonies formed, with minimal inhibition of normal colony formation.37  Lastly, animal studies showed that imatinib treatment led to a dose-dependent inhibition of BCR-ABL–expressing cells with no effects against v-SRC–expressing tumors.37  The first major presentation of this work was at the ASH meeting in December 1995. This was one of the last meetings with Tuesday morning scientific sessions, which were notorious for poor attendance. Despite the typical low turnout, several interested investigators were in attendance and they confirmed and extended our results.39,42,43  This was also a critical meeting for me as I was able to assemble a team of investigators to assist in the planning for clinical trials. This team included Dr John Goldman from the Hammersmith, Dr Charles Sawyers at UCLA, and Dr Moshe Talpaz at M. D. Anderson. All except Dr Goldman participated in the phase 1 study as it was decided to conduct this study only in the United States.

Despite the promising preclinical data, there were still hurdles to overcome before clinical trials commenced. These included concerns about toxicity, whether targeting a single kinase would be an effective anticancer strategy, and most importantly to a large pharmaceutical company, whether they would realize a return on their investment for a small market disease such as CML. At the time, I had patients in my clinic with CML with no effective treatment options remaining and this connection was a crucial turning point. I became their advocate by lobbying my remaining contacts at Novartis, including Alex Matter and Elisabeth Buchdunger, to move this project forward. Ultimately, we prevailed.

Phase 1 clinical trials

A standard dose-escalation phase 1 study of imatinib began in June 1998. The study population consisted of patients with CML in chronic phase, refractory or resistant to IFN-α–based therapy, or intolerant of this drug.44  At later stages of the study, patients with CML in blast crisis and patients with Ph chromosome–positive acute lymphoblastic leukemia were also enrolled.45  Imatinib was well tolerated with the most common side effects including occasional nausea, periorbital edema, and muscle cramps. Despite dose escalation from 25 mg to 1000 mg in 14 cohorts of patients, a maximally tolerated dose could not be defined. Imatinib was administered once daily and pharmacokinetics showed a half-life of 13 to 16 hours.44  At doses of 300 mg and above, significant therapeutic benefits were observed. In chronic phase patients who had failed therapy with IFN-α, 53 of 54 (98%) patients treated at 300 mg and above achieved a complete hematologic response, and with one year of follow-up, only one of these patients relapsed.44  In myeloid blast crisis patients, 21 of 38 (55%) patients treated at doses of 300 mg or more per day responded, with 18% having responses lasting beyond one year.45  The first major international presentation of these phase 1 results were at the plenary session of ASH in December 1999.

Phase 2 studies

These remarkable phase 1 data led to rapidly accruing phase 2 clinical trials. These clinical trials confirmed the results seen in the phase 1 studies and led to FDA approval of imatinib in May 2001, less than 3 years after the start of the phase 1 study (Figure 4). In chronic phase patients who had failed IFN-α therapy, 95% of patients achieved a complete hematologic response and 60%, a major cytogenetic response, defined as a reduction in the percentage of Ph chromosome–positive metaphases to less than 35.46  The estimated rates of freedom from progression to accelerated phase and blastic phase and overall survival at 6 years were 61% and 76%, respectively.49  In accelerated phase and blast crisis patients, the response rates were also quite high, but relapses have been much more common, with the majority of blast crisis patients relapsing during the first year of therapy.47,48 

Figure 4

Phase 2 clinical trials of imatinib for CML. The results of the phase 2 studies are shown in chronic phase patients who previously received interferon therapy, and accelerated phase and blast crisis patients. Results shown are with a median follow-up of up to 30 months and the rate of disease progression is at 24 months.46-48  Complete response (CR) and partial response (PR) for cytogenetic responses include patients with Ph chromosome–positive metaphases of less than or equal to 35%. Illustration by A. Y. Chen.

Figure 4

Phase 2 clinical trials of imatinib for CML. The results of the phase 2 studies are shown in chronic phase patients who previously received interferon therapy, and accelerated phase and blast crisis patients. Results shown are with a median follow-up of up to 30 months and the rate of disease progression is at 24 months.46-48  Complete response (CR) and partial response (PR) for cytogenetic responses include patients with Ph chromosome–positive metaphases of less than or equal to 35%. Illustration by A. Y. Chen.

Close modal

Cumulative experience with imatinib

The experience with imatinib has yielded a wealth of information, including data from a randomized clinical trial with 5 years of follow-up (the International Randomized Study of Interferon and STI571 [IRIS] study),50  a crystal structure of the ABL kinase domain in complex with imatinib,51  and important insights into the mechanisms of imatinib resistance.52,53  The IRIS trial was initiated in June 2000 for patients newly diagnosed with CML in the chronic phase. It reached its accrual goal of more than 1000 patients in less than 7 months.54  Five hundred fifty-three patients were randomized to each of the two treatments, imatinib at 400 mg per day or interferon-α plus Ara-C. There were no significant differences in prognostic features on the two arms. With a median follow-up of 19 months, patients randomized to imatinib had significantly better results than patients treated with interferon-α plus Ara-C in all parameters measured, including rates of complete hematologic response (97% vs 56%, P < .001), major and complete cytogenetic responses (85% and 74% vs 22% and 8%, respectively, P < .001), discontinuation of assigned therapy due to intolerance (3% vs 31%), and progression to accelerated phase or blast crisis (3% vs 8%, P < .001; Figure 5A).54  The substantial superiority of imatinib resulted in study results being disclosed early and most patients being crossed over to the imatinib arm. Accordingly, this study is now a long-term follow-up study of patients who received imatinib as initial therapy.

Figure 5

Phase 3 results of imatinib for newly diagnosed patients with CML. (A) The results shown are for newly diagnosed chronic phase patients with a median follow-up of 18 months. (B) The 5-year survival curve for newly diagnosed patients with chronic-phase CML treated with imatinib. Shown is the overall survival among patients treated with imatinib based on an intention-to-treat analysis.50  Illustration by A. Y. Chen.

Figure 5

Phase 3 results of imatinib for newly diagnosed patients with CML. (A) The results shown are for newly diagnosed chronic phase patients with a median follow-up of 18 months. (B) The 5-year survival curve for newly diagnosed patients with chronic-phase CML treated with imatinib. Shown is the overall survival among patients treated with imatinib based on an intention-to-treat analysis.50  Illustration by A. Y. Chen.

Close modal

In the most recent update of the IRIS study, the overall survival for newly diagnosed chronic phase patients treated with imatinib at 5 years is 89% (Figure 5B).50  An estimated 93% of imatinib-treated patients remain free from disease progression to the accelerated phase or blast crisis.50  An additional 6% of patients have shown some evidence of loss of response to imatinib, but their disease has not progressed to the accelerated phase or blast crisis.50  Most of the side effects of imatinib are mild to moderate, with the most common being edema, muscle cramps, diarrhea, nausea, skin rashes, and myelosuppression. Thus, imatinib therapy has substantially increased survival for patients with CML while simultaneously offering a generally well-tolerated, oral therapy. Although most patients randomized to imatinib achieve a complete cytogenetic response, the majority of these patients have detectable leukemia as analyzed by reverse transcriptase–polymerase chain reaction (RT-PCR) for BCR-ABL.55  Thus, most patients treated with imatinib have persistent disease at the molecular level and most have relapsed if imatinib was discontinued. Interestingly, over the past 4 years, there has been a trend downward in the risk of relapse; if this trend holds, this suggests that CML could be a long-term, controllable illness for the majority of patients.

Although most patients with chronic phase CML treated with imatinib have well-controlled disease, some patients have relapsed and/or progressed to accelerated phase or blast crisis.50  Furthermore, most patients with advanced phases of CML initially respond to imatinib but subsequently relapse.44,47,48  Therefore, the mechanisms of relapse emerged as a major question.

The first insights into relapse mechanisms came from assays that evaluated BCR-ABL kinase inhibition. This is where an understanding of BCR-ABL signaling and incorporation of this knowledge into the clinical trials was incredibly useful. In 1994, my lab and two others published the finding that the major tyrosine phosphorylated protein in CML patients was a 39-kDa adaptor protein CRKL.56-58  With this knowledge, we developed an assay that evaluated the phosphorylation status of CRKL in all patients enrolled in the imatinib clinical trials. Using this assay, Charles Sawyers' laboratory determined that the majority of patients who respond to imatinib and then relapse have reactivation of the BCR-ABL tyrosine kinase.59  Reactivation of the kinase would mean that resistance could be due to mechanisms that either prevent imatinib from reaching the target or render the target insensitive to imatinib. In the former category are mechanisms such as drug efflux or protein binding of imatinib. In the latter category would be mutations of the BCR-ABL kinase that render BCR-ABL insensitive to imatinib or amplification of the BCR-ABL protein.

In the background of this, we had been examining a puzzling finding. Imatinib is a potent inhibitor of ABL but not its most closely related kinase, SRC. We generated numerous single amino acid substitutions in residues in the kinase domains of these two proteins that we thought might allow SRC to be inhibited by imatinib or prevent imatinib from inhibiting ABL, with no success. Our conclusions were that either we needed combinations of amino acid changes or that despite their amino acid homology, the 3-dimensional structures of SRC and ABL differed sufficiently to account for these findings. This latter conclusion was confirmed when John Kuriyan's laboratory crystallized the ABL kinase in complex with imatinib (Figure 6).51  This structure demonstrated that imatinib binds to a unique inactive form of ABL and despite their homology, there were significant structural differences between the inactive form of ABL and SRC.

Figure 6

Ribbon representation of ABL in complex with imatinib and PD180970. Shown is (A) the conformation of ABL (blue) in complex with imatinib (orange), with the activation loop (magenta) in the inactive conformation, and (B) ABL (green) in complex with a dual SRC/ABL inhibitor, PD180970 (red), with the activation loop (magenta) in the active conformation. Figure prepared by Sandra W. Cowan-Jacob based on reported structures.60,61  Reprinted from Deininger et al.41 

Figure 6

Ribbon representation of ABL in complex with imatinib and PD180970. Shown is (A) the conformation of ABL (blue) in complex with imatinib (orange), with the activation loop (magenta) in the inactive conformation, and (B) ABL (green) in complex with a dual SRC/ABL inhibitor, PD180970 (red), with the activation loop (magenta) in the active conformation. Figure prepared by Sandra W. Cowan-Jacob based on reported structures.60,61  Reprinted from Deininger et al.41 

Close modal

The crystal structural data allowed critical contacts points between the ABL kinase and imatinib to be identified. With these data, we made mutations in all of the contact points and found that substitutions at amino acid 315 resulted in an ABL kinase that could not be inhibited by imatinib.62  This work was originally presented at the ASH meeting in December 2000.63  Subsequently, Charles Sawyers' laboratory reported that T315I mutations were present in patients who relapsed on imatinib therapy.59  We now know that at least 50% of patients who relapse on imatinib therapy have BCR-ABL point mutations in at least 40 different amino acids scattered throughout the ABL kinase domain (Figure 7).52,53  These studies of imatinib relapse mechanisms highlight the importance of incorporating scientific studies into clinical trials.

Figure 7

Schematic of point mutations in the ABL kinase domain. Mutations in the ABL kinase domain (amino acids 240 to 500) cluster in 4 distinct regions, the ATP-binding domain (amino acid 248-255, green), mutations of T315 (red), which form a hydrogen bond with imatinib, M351 (turquoise), which interacts with the SH2 domain and participates in autoregulation of kinase activity, and the activation loop (amino acids 379-398, magenta). The vertical lines represent the frequency each amino acid has been found to be mutated as compiled from the literature. Reprinted from Deininger et al.41 

Figure 7

Schematic of point mutations in the ABL kinase domain. Mutations in the ABL kinase domain (amino acids 240 to 500) cluster in 4 distinct regions, the ATP-binding domain (amino acid 248-255, green), mutations of T315 (red), which form a hydrogen bond with imatinib, M351 (turquoise), which interacts with the SH2 domain and participates in autoregulation of kinase activity, and the activation loop (amino acids 379-398, magenta). The vertical lines represent the frequency each amino acid has been found to be mutated as compiled from the literature. Reprinted from Deininger et al.41 

Close modal

In examining the structural basis for imatinib resistance, two main mechanisms have been identified. One obvious set of mutations are those at contact points between the ABL kinase domain and imatinib. Examples of this include T315I and F359V.62  The second category of kinase domain mutations is hypothesized to have conformational effects on the kinase, either favoring the active conformation to which imatinib cannot bind or decreasing the flexibility of the P-loop such that the conformational changes required for imatinib to bind this region cannot be adopted.64  These mutations include those in the P-loop, which bridges the ATP-binding pocket of the kinase domain (M244V, G250E, Q252H, Y253F/H, and E255K/V), and the activation loop (H396R/P).

Much like the original paradigm, whereby an understanding of the molecular pathogenesis of CML led to the development to imatinib, the understanding of the mechanism of resistance to imatinib led to the rapid development of new drugs to circumvent resistance. The information from the crystal structure suggested two approaches for drug design to circumvent resistance. One approach would be to modify imatinib such that it bound more tightly to the ABL kinase. This is exactly what was accomplished by chemists at Novartis in the design of nilotinib.65  In preclinical testing, nilotinib showed 10- to 30-fold increased potency over imatinib against the major resistant mutants, except T315I.65  Nilotinib retains the kinase specificity profile of imatinib, with inhibition confined to ABL kinases, KIT, and PDGFR kinases. As with imatinib, nilotinib binds the kinase domain of ABL in the inactive conformation.65 

A second approach to circumventing resistance relates to a comparison of the structure of ABL and SRC in their active and inactive conformations. Each kinase has a unique inactive conformation but similar active configurations. In 2000, Richard Jove published findings that a compound, PD180970, is capable of inhibiting SRC and ABL.66  We reasoned that such as inhibitor would likely bind the active conformation of ABL and that this inhibitor might be capable of inhibiting several of the imatinib-resistant mutations. A crystal structure of ABL with PD180970 from John Kuriyan's laboratory confirmed the first of these predictions,60  and in vitro studies performed in our lab confirmed the second.67  Although PD180970 was unable to be formulated for clinical utility, scientists at Bristol-Myers-Squibb examined dasatinib, originally synthesized as a SRC family inhibitor, and found that it was also a significantly more potent ABL inhibitor than imatinib.68  Dasatinib was subsequently shown to inhibit all imatinib-resistant mutants except T315I.69  A similar approach was taken by scientists at Wyeth in developing bosutinib.70 

Dasatinib and nilotinib have progressed rapidly through clinical trials, and both are FDA-approved for patients with resistance or intolerance to imatinib. Both show significant activity and good durability of responses in patients with relapsed, chronic-phase disease.71-74  Both dasatinib and nilotinib have been well tolerated, although each has unique side effects. For example, pleural effusions occur in some patients treated with dasatinib, and elevated liver enzymes and prolongation of QT interval corrected for rate (QTc) occur in some patients treated with nilotinib.71-75 

In the clinical trials of dasatinib and nilotinib, one of the most common mechanisms of resistance has been the emergence of clones with the T315I mutation in BCR-ABL.72,74,76-79  Neither nilotinib nor dasatinib inhibit T315I due to direct contacts between this residue and these molecules.65,80  Therefore, T315I remains a recurring problem for all the current ABL kinase inhibitors. Despite this challenge, several preclinical T315I inhibitors have been reported and are advancing to clinical trials.81,82  Preclinical studies have also suggested that combinations of inhibitors could completely circumvent resistance due to mutations.83  Whether combinations are necessary for the majority of patients presenting in chronic phase and the tolerability of combinations remain open questions. As studies define patients with a higher risk of resistance, these patients might be candidates for combination studies. Regardless, persistence of hematopoietic stem cells expressing BCR-ABL is likely to remain an issue,84,85  and studies to define and circumvent persistence will be required to cure patients with CML.

In addition to inhibiting the ABL tyrosine kinase, imatinib inhibits the PDGFR and KIT tyrosine kinases. There are now several other cancers where imatinib has shown clinical benefits that are based on the profile of kinases inhibited by imatinib and an understanding of the genetic defects causing various malignancies. One such disease is gastrointestinal stromal tumor (GIST), which is driven mainly by KIT mutations.86  Data on the use of imatinib in GIST are reviewed elsewhere.87  Imatinib has also shown significant activity in patients with acute lymphoblastic leukemia (ALL) who are BCR-ABL positive, but responses to single-agent therapy are generally transient.88  Recent data from pediatric clinical trials have shown, however, that combinations of imatinib with standard chemotherapy reverse the negative prognosis conveyed by the presence of the Philadelphia chromosome.89 

Translocations involving the PDGFRB gene have been identified in several myeloproliferative and myelodysplastic syndromes. The most common of these translocations, t(5;12)(q33;p13), is seen in a subset of patients with chronic myelomonocytic leukemia (CMML) and results in fusion of the EVT6 (TEL) and PDGFRB genes.90  Patients with CMML containing the (5;12) translocation have been treated with imatinib and significant responses have been observed.91,92 

Another disease caused by KIT mutations is systemic mastocytosis. The majority of tumors have a mutation of aspartic acid 816 to valine (D816V) in the kinase domain of KIT, resulting in activation of KIT.86  Unfortunately, the kinase activity of the D816V mutant isoform is resistant to imatinib,85,86,92-94  probably because this mutation induces conformational changes in the activation loop of KIT that prevent the binding of imatinib. Thus, imatinib is unlikely to be useful in this disorder, but an inhibitor of this KIT mutation would be predicted to be an effective therapy.

An interesting example has been the activity of imatinib in hypereosinophilic syndrome (HES). Imatinib was tried empirically in this disorder and dramatic results were observed.95  This prompted investigations of the molecular basis for imatinib's activity in this disease. Two groups arrived at the conclusion—that an intrachromosomal deletion on chromosome 4 resulted in a fusion between a gene of unknown function, FIP1L1, and a truncated PDGFRA in a large percentage of patients with this disorder.96,97  The resulting FIP1L1-PRGFRA fusion protein is a constitutively activated tyrosine kinase that is imatinib sensitive, thus accounting for the responsiveness of this disease to imatinib. The important message from this example is that careful study of responding patients can yield significant insights into disease pathogenesis.

The clinical trials with imatinib are a dramatic demonstration of the potential of targeting molecular pathogenetic events in a malignancy. As this paradigm is applied to other malignancies, it is worth remembering that BCR-ABL and CML have several features that were critical to the success of this agent. One of these is that BCR-ABL tyrosine kinase activity has clearly been demonstrated to be critical to the pathogenesis of CML. Thus, not only was the target of imatinib known, but the target is a critical factor required for the development of CML. Another important feature is that as with most malignancies, treatment earlier in the course of the disease yields better results. Specifically, the response rate and durability of responses have been greater in chronic phase patients as opposed to blast phase patients. Thus, for maximal utility as a single agent, the identification of crucial, early events in malignant progression is the first step in reproducing the success with imatinib in other malignancies. An equally as important issue is the selection of patients for clinical trials based on the presence of an appropriate target. Again, in the CML experience, patients with activation of BCR-ABL were easily identifiable by the presence of the Ph chromosome. In this regard, as reagents to analyze molecular end points are developed, these same reagents should be useful in identifying appropriate candidates for treatment with a specific agent. When all of these elements are put together, a critical pathogenetic target that is easily identifiable early in the course of the disease, remarkable results with an agent that targets this abnormality can be achieved. The obvious goal is to identify these early pathogenetic events in each malignancy and to develop agents that specifically target these abnormalities.

As we look back over the past 50 years, it is truly remarkable what has been accomplished by the members of ASH in their pursuit of improved treatments for their patients. In my clinic, it is now common for me to see patients with a CML duration of 10 years or more, a finding that previously would have made me question the diagnosis. It is particularly fitting that this commitment to support of basic discoveries into the molecular pathogenesis of hematologic diseases has been translated into such a successful cancer therapy. With the tools we currently have in hand to understand disease pathogenesis, it will be truly exciting to see what will be accomplished in the next several decades.

I gratefully acknowledge the hundreds of people who have participated in this project and assisted me in my career. This includes all of my current and former laboratory staff, my colleagues and mentors at OHSU, the clinical faculty, and our nurses and data managers. I thank my mentors from Dana-Farber Cancer Institute and the dedicated scientific and clinical staff at Novartis, who shepherded imatinib through clinical trials. I also thank the numerous investigators who enrolled patients on our clinical trials. During this time, I have been supported by various funding agencies including the National Cancer Institute, The Leukemia & Lymphoma Society, the Burroughs Wellcome Fund, the T. J. Martell Foundation, the Doris Duke Charitable Foundation, and the Howard Hughes Medical Institute. I am grateful for this support. Lastly, I thank my patients who have gone on this incredible journey with me.

Contribution: B.J.D. wrote the paper.

Conflict-of-interest disclosure: Molecular MD–OHSU and B.J.D. have a financial interest in MolecularMD. Technology used in this research has been licensed to MolecularMD. This potential conflict of interest has been reviewed and managed by the OHSU Conflict of Interest in Research Committee and the Integrity Program Oversight Council.

Novartis & BMS: I (B.J.D.) currently am the principal investigator on several Novartis and Bristol-Myers-Squibb clinical trials. My institution has contracts with these companies to pay for patient costs, nurse and data manager salaries, and institutional overhead. I do not derive salary, nor does my lab receive funds from these contracts.

Correspondence: Brian J. Druker, Howard Hughes Medical Institute, JELD-WEN Chair of Leukemia Research, Oregon Health & Science University Cancer Institute, L592, 3181 SW Sam Jackson Park Road, Portland, OR; e-mail: drukerb@ohsu.edu.

1
Bennett
 
JH
Case of hypertrophy of the spleen and liver in which death took place from suppuration of the blood.
Edinb Med Surg J
1845
, vol. 
64
 (pg. 
413
-
423
)
2
Virchow
 
R
Weisses Blut.
Frorieps Notizen
1845
, vol. 
36
 (pg. 
151
-
156
)
3
Geary
 
CG
The story of chronic myeloid leukaemia.
Br J Haematol
2000
, vol. 
10
 (pg. 
2
-
11
)
4
Calabretta
 
B
Perrotti
 
D
The biology of CML blast crisis.
Blood
2004
, vol. 
103
 (pg. 
4010
-
4022
)
5
Ito
 
K
Bernardi
 
R
Morotti
 
A
, et al. 
PML targeting eradicates quiescent leukaemia-initiating cells.
Nature
2008
, vol. 
453
 (pg. 
1072
-
1078
)
6
O'Dwyer
 
ME
La Rosee
 
P
Nimmanapalli
 
R
Bhalla
 
KN
Druker
 
BJ
Recent advances in Philadelphia chromosome-positive malignancies: the potential role of arsenic trioxide.
Semin Hematol
2002
, vol. 
39
 (pg. 
18
-
21
)
7
Baccarani
 
M
Saglio
 
G
Goldman
 
J
, et al. 
Evolving concepts in the management of chronic myeloid leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet.
Blood
2006
, vol. 
108
 (pg. 
1809
-
1820
)
8
Nowell
 
PC
Hungerford
 
DA
A minute chromosome in human chronic granulocytic leukemia.
Science
1960
, vol. 
132
 pg. 
1497
 
9
Rowley
 
JD
A new consistent abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and giemsa staining.
Nature
1973
, vol. 
243
 (pg. 
290
-
293
)
10
Abelson
 
HT
Rabstein
 
LS
Lymphosarcoma: virus-induced thymic-independent disease in mice.
Cancer Res
1970
, vol. 
30
 (pg. 
2213
-
2222
)
11
Goff
 
SP
Gilboa
 
E
Witte
 
ON
Baltimore
 
D
Structure of the Abelson murine leukemia virus genome and the homologous cellular gene: studies with cloned viral DNA.
Cell
1980
, vol. 
22
 (pg. 
777
-
785
)
12
Reddy
 
EP
Smith
 
MJ
Srinivasan
 
A
Nucleotide sequence of Abelson murine leukemia virus genome: structural similarity of its transforming gene product to other onc gene products with tyrosine-specific kinase activity.
Proc Natl Acad Sci U S A
1983
, vol. 
80
 (pg. 
3623
-
3627
)
13
Wang
 
JY
Ledley
 
F
Goff
 
S
Lee
 
R
Groner
 
Y
Baltimore
 
D
The mouse c-abl locus: molecular cloning and characterization.
Cell
1984
, vol. 
36
 (pg. 
349
-
356
)
14
de Klein
 
A
van Kessel
 
AG
Grosveld
 
G
, et al. 
A cellular oncogene is translocated to the Philadelphia chromosome in chronic myelocytic leukemia.
Nature
1982
, vol. 
300
 (pg. 
765
-
767
)
15
Deininger
 
MW
Goldman
 
JM
Melo
 
JV
The molecular biology of chronic myeloid leukemia.
Blood
2000
, vol. 
96
 (pg. 
3343
-
3356
)
16
Groffen
 
J
Stephenson
 
JR
Heisterkamp
 
N
de Klein
 
A
Bartram
 
CR
Grosveld
 
G
Philadelphia chromosome breakpoints are clustered within a limited region, bcr, on chromosome 22.
Cell
1984
, vol. 
36
 (pg. 
93
-
99
)
17
Collins
 
SJ
Kubonishi
 
I
Miyoshi
 
I
Groudine
 
MT
Altered transcription of the c-abl oncogene in K-562 and other chronic myelogenous leukemia cells.
Science
1984
, vol. 
225
 (pg. 
72
-
74
)
18
Gale
 
RP
Canaani
 
E
An 8-kilobase abl RNA transcript in chronic myelogenous leukemia.
Proc Natl Acad Sci U S A
1984
, vol. 
81
 (pg. 
5648
-
5652
)
19
Shtivelman
 
E
Lifshitz
 
B
Gale
 
RP
Canaani
 
E
Fused transcript of abl and bcr genes in chronic myelogenous leukaemia.
Nature
1985
, vol. 
315
 (pg. 
550
-
554
)
20
Ben-Neriah
 
Y
Daley
 
GQ
Mes-Masson
 
A-M
Witte
 
ON
Baltimore
 
D
The chronic myelogenous leukemia-specific P210 protein is the product of the bcr/abl hybrid gene.
Science
1986
, vol. 
233
 (pg. 
212
-
214
)
21
Davis
 
RL
Konopka
 
JB
Witte
 
ON
Activation of the c-abl oncogene by viral transduction or chromosomal translocation generates altered c-abl proteins with similar in vitro kinase properties.
Mol Cell Biol
1985
, vol. 
5
 (pg. 
204
-
213
)
22
Mes-Masson
 
A-M
McLaughlin
 
J
Daley
 
GQ
Paskind
 
M
Witte
 
ON
Overlapping cDNA clones define the complete coding region for the P210c-abl gene product associated with chronic myelogenous leukemia cells containing the Philadelphia chromosome.
Proc Natl Acad Sci U S A
1986
, vol. 
83
 (pg. 
9768
-
9772
)
23
Sefton
 
BM
Hunter
 
T
Raschke
 
WC
Evidence that the Abelson virus protein functions in vivo as a protein kinase that phosphorylates tyrosine.
Proc Natl Acad Sci U S A
1981
, vol. 
78
 (pg. 
1552
-
1556
)
24
Witte
 
ON
Dasgupta
 
A
Baltimore
 
D
Abelson murine leukaemia virus protein is phosphorylated in vitro to form phosphotyrosine.
Nature
1980
, vol. 
283
 (pg. 
826
-
831
)
25
Konopka
 
JB
Watanabe
 
SM
Witte
 
ON
An alteration of the human c-abl protein in K562 leukemia cells unmasks associated tyrosine kinase activity.
Cell
1984
, vol. 
37
 (pg. 
1035
-
1042
)
26
Lugo
 
TG
Pendergast
 
AM
Muller
 
AJ
Witte
 
ON
Tyrosine kinase activity and transformation potency of bcr-abl oncogene products.
Science
1990
, vol. 
247
 (pg. 
1079
-
1082
)
27
Daley
 
GQ
Van Etten
 
RA
Baltimore
 
D
Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome.
Science
1990
, vol. 
247
 (pg. 
824
-
830
)
28
Heisterkamp
 
N
Jenster
 
G
ten Hoeve
 
J
Zovich
 
D
Pattengale
 
PK
Groffen
 
J
Acute leukaemia in bcr/abl transgenic mice.
Nature
1990
, vol. 
344
 (pg. 
251
-
253
)
29
Melo
 
JV
Deininger
 
MW
Biology of chronic myelogenous leukemia–signaling pathways of initiation and transformation.
Hematol Oncol Clin North Am
2004
, vol. 
18
 (pg. 
545
-
568
vii-viii
30
Yaish
 
P
Gazit
 
A
Gilon
 
C
Levitzki
 
A
Blocking of EGF-dependent cell proliferation by EGF receptor kinase inhibitors.
Science
1988
, vol. 
242
 (pg. 
933
-
935
)
31
Piwnica-Worms
 
H
Williams
 
NG
Cheng
 
SH
Roberts
 
TM
Regulation of pp60c-src and its interaction with polyomavirus middle T antigen in insect cells.
J Virol
1990
, vol. 
64
 (pg. 
61
-
68
)
32
Druker
 
BJ
Mamon
 
HJ
Roberts
 
TM
Oncogenes, growth factors, and signal transduction.
N Engl J Med
1989
, vol. 
321
 (pg. 
1383
-
1391
)
33
Buchdunger
 
E
Zimmermann
 
J
Mett
 
H
, et al. 
Selective inhibition of the platelet-derived growth factor signal transduction pathway by a protein-tyrosine kinase inhibitor of the 2-phenylaminopyrimidine class.
Proc Natl Acad Sci U S A
1995
, vol. 
92
 (pg. 
2558
-
2562
)
34
Zimmermann
 
J
Buchdunger
 
E
Mett
 
H
Meyer
 
T
Lydon
 
NB
Traxler
 
P
Phenylamino-pyrimidine (PAP) derivatives: a new class of potent and highly selective PDGF-receptor autophosphorylation inhibitors.
Bioorg Med Chem Lett
1996
, vol. 
6
 (pg. 
1221
-
1226
)
35
Zimmermann
 
J
Caravatti
 
G
Mett
 
H
, et al. 
Phenylamino-pyrimidine (PAP) derivatives: a new class of potent and selective inhibitors of protein kinase C (PKC).
Arch Pharm
1996
, vol. 
329
 (pg. 
371
-
376
)
36
Buchdunger
 
E
Zimmermann
 
J
Mett
 
H
, et al. 
Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimidine derivative.
Cancer Res
1996
, vol. 
56
 (pg. 
100
-
104
)
37
Druker
 
BJ
Tamura
 
S
Buchdunger
 
E
, et al. 
Effects of a selective inhibitor of the ABL tyrosine kinase on the growth of BCR-ABL positive cells.
Nat Med
1996
, vol. 
2
 (pg. 
561
-
566
)
38
Druker
 
BJ
Lydon
 
NB
Lessons learned from the development of an abl tyrosine kinase inhibitor for chronic myelogenous leukemia.
J Clin Invest
2000
, vol. 
105
 (pg. 
3
-
7
)
39
Carroll
 
M
Ohno-Jones
 
S
Tamura
 
S
, et al. 
CGP 57148, a tyrosine kinase inhibitor, inhibits the growth of cells expressing BCR-ABL, TEL-ABL and TEL-PDGFR fusion proteins.
Blood
1997
, vol. 
90
 (pg. 
4947
-
4952
)
40
Heinrich
 
MC
Griffith
 
DJ
Druker
 
BJ
Wait
 
CL
Ott
 
KA
Zigler
 
AJ
Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor.
Blood
2000
, vol. 
96
 (pg. 
925
-
932
)
41
Deininger
 
M
Buchdunger
 
E
Druker
 
BJ
The development of imatinib as a therapeutic agent for chronic myeloid leukemia.
Blood
2005
, vol. 
105
 (pg. 
2640
-
2653
)
42
Deininger
 
MW
Goldman
 
JM
Lydon
 
N
Melo
 
JV
The tyrosine kinase inhibitor CGP57148B selectively inhibits the growth of BCR-ABL-positive cells.
Blood
1997
, vol. 
90
 (pg. 
3691
-
3698
)
43
Gambacorti-Passerini
 
C
le Coutre
 
P
Mologni
 
L
, et al. 
Inhibition of the ABL kinase activity blocks the proliferation of BCR/ABL+ leukemic cells and induces apoptosis.
Blood Cells Mol Dis
1997
, vol. 
23
 (pg. 
380
-
394
)
44
Druker
 
BJ
Talpaz
 
M
Resta
 
D
, et al. 
Efficacy and safety of a specific inhibitor of the Bcr-Abl tyrosine kinase in chronic myeloid leukemia.
N Engl J Med
2001
, vol. 
344
 (pg. 
1031
-
1037
)
45
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
2001
, vol. 
344
 (pg. 
1038
-
1042
)
46
Kantarjian
 
H
Sawyers
 
C
Hochhaus
 
A
, et al. 
Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia.
New Engl J Med
2002
, vol. 
346
 (pg. 
645
-
652
)
47
Sawyers
 
CL
Hochhaus
 
A
Feldman
 
E
, et al. 
Imatinib induces hematologic and cytogenetic responses in patients with chronic myeloid leukemia in myeloid blast crisis: results of a phase II study.
Blood
2002
, vol. 
99
 (pg. 
3530
-
3539
)
48
Talpaz
 
M
Silver
 
RT
Druker
 
BJ
, et al. 
Imatinib induces durable hematologic and cytogenetic responses in patients with accelerated phase chronic myeloid leukemia: results of a phase 2 study.
Blood
2002
, vol. 
99
 (pg. 
1928
-
1937
)
49
Hochhaus
 
A
Druker
 
B
Sawyers
 
C
, et al. 
Favorable long-term follow-up results over 6 years for response, survival, and safety with imatinib mesylate therapy in chronic-phase chronic myeloid leukemia after failure of interferon-alpha treatment.
Blood
2008
, vol. 
111
 (pg. 
1039
-
1043
)
50
Druker
 
BJ
Guilhot
 
F
O'Brien
 
SG
, et al. 
Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia.
N Engl J Med
2006
, vol. 
355
 (pg. 
2408
-
2417
)
51
Schindler
 
T
Bornmann
 
W
Pellicena
 
P
Miller
 
WT
Clarkson
 
B
Kuriyan
 
J
Structural mechanism for STI-571 inhibition of abelson tyrosine kinase.
Science
2000
, vol. 
289
 (pg. 
1938
-
1942
)
52
O'Hare
 
T
Eide
 
CA
Deininger
 
MW
Bcr-Abl kinase domain mutations, drug resistance, and the road to a cure for chronic myeloid leukemia.
Blood
2007
, vol. 
110
 (pg. 
2242
-
2249
)
53
Shah
 
NP
Sawyers
 
CL
Mechanisms of resistance to STI571 in Philadelphia chromosome-associated leukemias.
Oncogene
2003
, vol. 
22
 (pg. 
7389
-
7395
)
54
O'Brien
 
SG
Guilhot
 
F
Larson
 
RA
, et al. 
Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia.
N Engl J Med
2003
, vol. 
348
 (pg. 
994
-
1004
)
55
Hughes
 
TP
Kaeda
 
J
Branford
 
S
, et al. 
Frequency of major molecular responses to imatinib or interferon alfa plus cytarabine in newly diagnosed chronic myeloid leukemia.
N Engl J Med
2003
, vol. 
349
 (pg. 
1423
-
1432
)
56
Oda
 
T
Heaney
 
C
Hagopian
 
JR
Okuda
 
K
Griffin
 
JD
Druker
 
BJ
Crkl is the major tyrosine-phosphorylated protein in neutrophils from patients with chronic myelogenous leukemia.
J Biol Chem
1994
, vol. 
269
 (pg. 
22925
-
22928
)
57
Nichols
 
GL
Raines
 
MA
Vera
 
JC
Lacomis
 
L
Tempst
 
P
Golde
 
DW
Identification of CRKL as the constitutively phosphorylated 39-kD tyrosine phosphoprotein in chronic myelogenous leukemia cells.
Blood
1994
, vol. 
84
 (pg. 
2912
-
2918
)
58
ten Hoeve
 
J
Arlinghaus
 
RB
Guo
 
JQ
Heisterkamp
 
N
Tyrosine phosphorylation of CRKL in Philadelphia+ leukemia.
Blood
1994
, vol. 
84
 (pg. 
1731
-
1736
)
59
Gorre
 
ME
Mohammed
 
M
Ellwood
 
K
, et al. 
Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification.
Science
2001
, vol. 
293
 (pg. 
876
-
880
)
60
Nagar
 
B
Bornmann
 
WG
Pellicena
 
P
, et al. 
Crystal structures of the kinase domain of c-Abl in complex with the small molecule inhibitors PD173955 and imatinib (STI-571).
Cancer Res
2002
, vol. 
62
 (pg. 
4236
-
4243
)
61
Cowan-Jacob
 
SW
Guez
 
V
Fendrich
 
G
, et al. 
Imatinib (STI571) resistance in chronic myelogenous leukemia: molecular basis of the underlying mechanisms and potential strategies for treatment.
Mini Rev Med Chem
2004
, vol. 
4
 (pg. 
285
-
299
)
62
Corbin
 
AS
Buchdunger
 
E
Pascal
 
F
Druker
 
BJ
Analysis of the structural basis of specificity of inhibition of the Abl kinase by STI571.
J Biol Chem
2002
, vol. 
277
 (pg. 
32214
-
32219
)
63
Corbin
 
AS
Toledo
 
LM
Lydon
 
NB
Buchdunger
 
E
Kuriyan
 
J
Druker
 
BJ
Analysis of the structural basis of specificity of inhibition of the Abl kinase by STI571 [abstract].
Blood
2000
, vol. 
96
 pg. 
470a
 
64
Shah
 
NP
Nicoll
 
JM
Nagar
 
B
, et al. 
Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia.
Cancer Cell
2002
, vol. 
2
 (pg. 
117
-
125
)
65
Weisberg
 
E
Manley
 
PW
Breitenstein
 
W
, et al. 
Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl.
Cancer Cell
2005
, vol. 
7
 (pg. 
129
-
141
)
66
Dorsey
 
JF
Jove
 
R
Kraker
 
AJ
Wu
 
J
The pyrido[2,3-d]pyrimidine derivative PD180970 inhibits p210Bcr-Abl tyrosine kinase and induces apoptosis of K562 leukemic cells.
Cancer Res
2000
, vol. 
60
 (pg. 
3127
-
3131
)
67
La Rosée
 
P
Corbin
 
AS
Stoffregen
 
EP
Deininger
 
MW
Druker
 
BJ
Activity of the Bcr-Abl kinase inhibitor PD180970 against clinically relevant Bcr-Abl isoforms that cause resistance to imatinib mesylate (Gleevec, STI571).
Cancer Res
2002
, vol. 
62
 (pg. 
7149
-
7153
)
68
Lombardo
 
LJ
Lee
 
FY
Chen
 
P
, et al. 
Discovery of N-(2-chloro-6-methyl-phenyl)-2-(6-(4-(2-hydroxyethyl)-piperazin-1-yl)-2-methylpyrimidin-4-ylamino)thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays.
J Med Chem
2004
, vol. 
47
 (pg. 
6658
-
6661
)
69
Shah
 
NP
Tran
 
C
Lee
 
FY
Chen
 
P
Norris
 
D
Sawyers
 
CL
Overriding imatinib resistance with a novel ABL kinase inhibitor.
Science
2004
, vol. 
305
 (pg. 
399
-
401
)
70
Golas
 
JM
Arndt
 
K
Etienne
 
C
, et al. 
SKI-606, a 4-anilino-3-quinolinecarbonitrile dual inhibitor of Src and Abl kinases, is a potent antiproliferative agent against chronic myelogenous leukemia cells in culture and causes regression of K562 xenografts in nude mice.
Cancer Res
2003
, vol. 
63
 (pg. 
375
-
381
)
71
Talpaz
 
M
Shah
 
NP
Kantarjian
 
H
, et al. 
Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias.
N Engl J Med
2006
, vol. 
354
 (pg. 
2531
-
2541
)
72
Hochhaus
 
A
Kantarjian
 
HM
Baccarani
 
M
, et al. 
Dasatinib induces notable hematologic and cytogenetic responses in chronic-phase chronic myeloid leukemia after failure of imatinib therapy.
Blood
2007
, vol. 
109
 (pg. 
2303
-
2309
)
73
Kantarjian
 
H
Giles
 
F
Wunderle
 
L
, et al. 
Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL.
N Engl J Med
2006
, vol. 
354
 (pg. 
2542
-
2551
)
74
Kantarjian
 
HM
Giles
 
F
Gattermann
 
N
, et al. 
Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is effective in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase following imatinib resistance and intolerance.
Blood
2007
, vol. 
110
 (pg. 
3540
-
3546
)
75
Shah
 
NP
Kantarjian
 
HM
Kim
 
DW
, et al. 
Intermittent target inhibition with dasatinib 100 mg once daily preserves efficacy and improves tolerability in imatinib-resistant and -intolerant chronic-phase chronic myeloid leukemia.
J Clin Oncol
2008
, vol. 
26
 (pg. 
3204
-
3212
)
76
le Coutre
 
P
Ottmann
 
OG
Giles
 
F
, et al. 
Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is active in patients with imatinib-resistant or -intolerant accelerated-phase chronic myelogenous leukemia.
Blood
2008
, vol. 
111
 (pg. 
1834
-
1839
)
77
Cortes
 
J
Rousselot
 
P
Kim
 
DW
, et al. 
Dasatinib induces complete hematologic and cytogenetic responses in patients with imatinib-resistant or -intolerant chronic myeloid leukemia in blast crisis.
Blood
2007
, vol. 
109
 (pg. 
3207
-
3213
)
78
Guilhot
 
F
Apperley
 
J
Kim
 
DW
, et al. 
Dasatinib induces significant hematologic and cytogenetic responses in patients with imatinib-resistant or -intolerant chronic myeloid leukemia in accelerated phase.
Blood
2007
, vol. 
109
 (pg. 
4143
-
4150
)
79
Shah
 
NP
Skaggs
 
BJ
Branford
 
S
, et al. 
Sequential ABL kinase inhibitor therapy selects for compound drug-resistant BCR-ABL mutations with altered oncogenic potency.
J Clin Invest
2007
, vol. 
117
 (pg. 
2562
-
2569
)
80
Tokarski
 
JS
Newitt
 
JA
Chang
 
CY
, et al. 
The structure of Dasatinib (BMS-354825) bound to activated ABL kinase domain elucidates its inhibitory activity against imatinib-resistant ABL mutants.
Cancer Res
2006
, vol. 
66
 (pg. 
5790
-
5797
)
81
Giles
 
FJ
Cortes
 
J
Jones
 
D
Bergstrom
 
D
Kantarjian
 
H
Freedman
 
SJ
MK-0457, a novel kinase inhibitor, is active in patients with chronic myeloid leukemia or acute lymphocytic leukemia with the T315I BCR-ABL mutation.
Blood
2007
, vol. 
109
 (pg. 
500
-
502
)
82
Shakespeare
 
WC
Wang
 
F
Xu
 
Q
, et al. 
Orally active inhibitors of the imatinib resistant Bcr-Abl mutant T315I [abstract].
Blood
2006
, vol. 
108
 pg. 
2180a
 
83
O'Hare
 
T
Eide
 
CA
Tyner
 
JW
, et al. 
SGX393 inhibits the CML mutant Bcr-AblT315I and preempts in vitro resistance when combined with nilotinib or dasatinib.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 (pg. 
5507
-
5512
)
84
Elrick
 
LJ
Jorgensen
 
HG
Mountford
 
JC
Holyoake
 
TL
Punish the parent not the progeny.
Blood
2005
, vol. 
105
 (pg. 
1862
-
1866
)
85
O'Hare
 
T
Corbin
 
AS
Druker
 
BJ
Targeted CML therapy: controlling drug resistance, seeking cure.
Curr Opin Genet Dev
2006
, vol. 
16
 (pg. 
92
-
99
)
86
Heinrich
 
MC
Blanke
 
CD
Druker
 
BJ
Corless
 
CL
Inhibition of KIT tyrosine kinase activity: a novel molecular approach to the treatment of KIT-positive malignancies.
J Clin Oncol
2002
, vol. 
20
 (pg. 
1692
-
1703
)
87
Siddiqui
 
MA
Scott
 
LJ
Imatinib: a review of its use in the management of gastrointestinal stromal tumours.
Drugs
2007
, vol. 
67
 (pg. 
805
-
820
)
88
Ottmann
 
OG
Druker
 
BJ
Sawyers
 
CL
, et al. 
A phase 2 study of imatinib in patients with relapsed or refractory Philadelphia chromosome-positive acute lymphoid leukemias.
Blood
2002
, vol. 
100
 (pg. 
1965
-
1971
)
89
Schultz
 
KR
Bowman
 
WP
Slayton
 
W
, et al. 
Improved early event free survival (EFS) in children with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) with intensive imatinib in combination with high dose chemotherapy: children's oncology group (COG) study AALL0031 [abstract].
Blood
2007
, vol. 
110
 pg. 
4a
 
90
Golub
 
TR
Barker
 
GF
Lovett
 
M
Gilliland
 
DG
Fusion of PDGF receptor beta to a novel ets-like gene, tel, in chronic myelomonocytic leukemia with t(5;12) chromosomal translocation.
Cell
1994
, vol. 
77
 (pg. 
307
-
316
)
91
Apperley
 
JF
Gardembas
 
M
Melo
 
JV
, et al. 
Response to imatinib mesylate in patients with chronic myeloproliferative diseases with rearrangements of the platelet-derived growth factor receptor beta.
N Engl J Med
2002
, vol. 
347
 (pg. 
481
-
487
)
92
Magnusson
 
MK
Meade
 
KE
Nakamura
 
R
Barrett
 
J
Dunbar
 
CE
Activity of STI571 in chronic myelomonocytic leukemia with a platelet-derived growth factor beta receptor fusion oncogene.
Blood
2002
, vol. 
100
 (pg. 
1088
-
1091
)
93
Heinrich
 
MC
Wait
 
CL
Yee
 
KWH
Griffith
 
DJ
STI571 inhibits the kinase activity of wild type and juxtamembrane c-kit mutants but not the exon 17 D816V mutation associated with mastocytosis [abstract].
Blood
2000
, vol. 
96
 pg. 
173b
 
94
Ma
 
Y
Zeng
 
S
Metcalfe
 
DD
, et al. 
The c-KIT mutation causing human mastocytosis is resistant to STI571 and other KIT kinase inhibitors: kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinases and those with regulatory-type mutations.
Blood
2002
, vol. 
99
 (pg. 
1741
-
1744
)
95
Gleich
 
GJ
Leiferman
 
KM
Pardanani
 
A
Tefferi
 
A
Butterfield
 
JH
Treatment of hypereosinophilic syndrome with imatinib mesilate.
Lancet
2002
, vol. 
359
 (pg. 
1577
-
1578
)
96
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 hypereosinophilic syndrome.
N Engl J Med
2003
, vol. 
348
 (pg. 
1201
-
1214
)
97
Griffin
 
JH
Leung
 
J
Bruner
 
RJ
Caligiuri
 
MA
Briesewitz
 
R
Discovery of a fusion kinase in EOL-1 cells and idiopathic hypereosinophilic syndrome.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 (pg. 
7830
-
7835
)
Sign in via your Institution