Optimal First-Line Treatment of Chronic Myeloid Leukemia: How to Use Imatinib and What Role for Newer Drugs?

News
Article
OncologyONCOLOGY Vol 21 No 6
Volume 21
Issue 6

The development of imatinib mesylate (Gleevec), a tyrosine kinase inhibitor targeted against the causative Bcr-Abl protein in chronic myeloid leukemia (CML), has resulted in hematologic and cytogenetic remissions in all phases of CML. Following imatinib treatment, more than 90% of patients obtain complete hematologic response, and 70% to 80% achieve a complete cytogenetic response. With 5 years of follow-up, the data are very encouraging, exhibiting a major change in the natural history of the disease. The understanding of at least some of the mechanisms of resistance to imatinib has led to a rapid development of new agents that may overcome this resistance. Combination strategies are currently being investigated in preliminary clinical studies and may prove to be useful. Overall, there are an increasing number of treatment options now available for patients with CML.

The development of imatinib mesylate (Gleevec), a tyrosine kinase inhibitor targeted against the causative Bcr-Abl protein in chronic myeloid leukemia (CML), has resulted in hematologic and cytogenetic remissions in all phases of CML. Following imatinib treatment, more than 90% of patients obtain complete hematologic response, and 70% to 80% achieve a complete cytogenetic response. With 5 years of follow-up, the data are very encouraging, exhibiting a major change in the natural history of the disease. The understanding of at least some of the mechanisms of resistance to imatinib has led to a rapid development of new agents that may overcome this resistance. Combination strategies are currently being investigated in preliminary clinical studies and may prove to be useful. Overall, there are an increasing number of treatment options now available for patients with CML.

Chronic myeloid leukemia (CML) is a rare disease. Although its incidence is low, its prevalence is increasing. In the United States, there are approximately 4,500 new cases of CML per year.[1] The annual incidence of CML is 1.6 cases per 100,000 adults.[1] The median age at diagnosis is 55 years. With an estimated survival rate of 90% at 5 years and an annual mortality rate of 2%, the prevalence of CML in 20 years may become 200,000 to 300,000 cases in the United States. CML is characterized by a balanced genetic translocation, involving a fusion of the Abelson oncogene (ABL) from chromosome 9q34 with the breakpoint cluster region (BCR) on chromosome 22q11.2, t(9;22)(q34;q11.2), the Philadelphia chromosome (Ph). The molecular consequence of this translocation is the generation of a BCR-ABL fusion oncogene, which in turn translates into a Bcr-Abl oncoprotein. This most frequently has a molecular weight of 210 kD (p210Bcr/Abl) and has increased tyrosine kinase activity, which is essential to its transforming capability.[2,3]

FIGURE 1


Pathways Activated by Bcr-Abl

Numerous signal transduction pathways, including Ras/Raf/mitogen activated protein kinase (MAPK), phosphatidylinositol 3 kinase, STAT5/Janus kinase, and Myc, are activated by the Bcr-Abl tyrosine kinase (Figure 1).[4,5] Perturbation of these pathways results in uncontrolled cell proliferation and reduced apoptosis. Understanding the CML pathophysiology resulted in the development of novel drugs targeting Bcr-Abl tyrosine kinase and its associated pathways.

The treatment of CML has evolved greatly over the past few years. Imatinib mesylate is now well established as the standard therapy. For many years, stem cell transplantation (SCT) and interferon-alpha were the major therapeutic choices. Long-term survival and possibly cure could be achieved with both of these modalities.[6,7] While SCT is still a valid treatment option for some patients, interferon-alpha has been replaced in CML front-line therapy by imatinib.

Imatinib mesylate is a potent and selective tyrosine kinase inhibitor that has become standard therapy for patients with any stage of CML.[3] A complete cytogenetic response can be achieved in 50% to 60% of patients treated in chronic phase after failure to respond to interferon-alpha[8,9] and in over 80% of those receiving imatinib as first-line therapy.[10,11] Responses are durable in most patients treated in early chronic phase, particularly among those who achieve major molecular responses (eg, ≥ 3-log reduction in transcript levels).[12,13]

Here we will review the current information regarding treatment of patients with newly diagnosed chronic phase CML, issues of imatinib dose schedules, imatinib toxicity and management of adverse events, monitoring of minimal residual disease, and the place of the new tyrosine kinase inhibitors in the front-line therapy arsenal of patients with newly diagnosed CML in chronic phase.

Imatinib: Front-Line Standard of Care in CML

Imatinib is an orally bioavailable 2-phenylaminopyrimidine with targeted inhibitory activity against the constitutively active tyrosine kinase of the Bcr-Abl chimeric fusion protein. Imatinib inhibits other kinases, such as c-kit, platelet-derived growth factor receptor (PDGFR)-alpha and -beta, and Abl-related gene.[14,15] Imatinib has become the standard therapy for CML because of its remarkable activity and mild toxicity profile.

Preclinical studies established that imatinib potently inhibits ABL and the related protein-tyrosine kinase ARG as well as the kinase activity of members of the class III family of receptor tyrosine kinases including KIT, PDGFRs and the macrophage colony-stimulating factor receptor CSF-1R (cFMS).[16-20] Inhibition of BCR-ABL protein-tyrosine kinase activity with imatinib blocks intracellular oncogenic signal transduction pathways.[3,21]

IRIS Trial

The efficacy of imatinib was demonstrated in the phase III International Randomized Study of Interferon and STI571 (IRIS) trial, in which treatment with imatinib at 400 mg daily was compared with combined interferon-alpha/cytarabine in patients with newly diagnosed chronic phase CML (N = 1,106).[22] After a median follow-up of 19 months, imatinib was found to be significantly better than interferon-alpha-based treatment, as shown by rates of complete hematologic response (95% vs 56% of patients; P < .001) and major cytogenetic response (≤ 35% Ph-positive cells in metaphase; 85% vs 22% of patients; P < .001). Major molecular response rates (at 12 months, 40% vs 2%) and progression-free survival were also superior with imatinib.[13,22] Adverse events reported in the imatinib group were generally mild to moderate, and included superficial edema, nausea, muscle cramps, and rashes, and grade 3/4 events were uncommon except for neutropenia (14%) and thrombocytopenia (8%).[22] Adverse event rates of all types were higher in the combination-therapy group.

TABLE 1


Best Responses in Patients Remaining on First-Line Imatinib Therapy: 5-yr Update of the Phase III IRIS Trial

A 5-year update of the IRIS study continued to show positive results (Table 1).[23] A total of 382 patients remained on imatinib front-line therapy. The cumulative best complete hematologic response, major cytogenetic response, and complete cytogenetic response rates were 98%, 92%, and 87%, respectively. The estimated 5-year event-free survival was 83%; only 6.3% of patients progressed to accelerated and blastic phases. The overall annual progression rate has declined to 0.9% in the fifth year of therapy, compared with 1.5%, 4.8%, and 7.5% in the previous 3 years, suggesting that disease progression may be diminished in the following years. The estimated 5-year survival rate was 89%; excluding non-CML deaths, it was 95%. The intensity of the cytogenetic response after 12 and 18 months of imatinib therapy has important implications regarding survival without transformation. The estimated 5-year survival rate in patients not achieving a major cytogenetic response at 12 months was significantly lower (81%) than those who achieved major cytogenetic response (complete, 97%; partial, 93%; P < .001).

FIGURE 2


Survival of Chronic Myeloid Leukemia

At 18 months of therapy, the estimated 5-year survival rate for patients not achieving a complete cytogenetic response was significantly less than for those who achieved a complete cytogenetic response (99% vs 90%; P < .001).[23] The investigators found a continuous improvement in the rate of major molecular response, which rose from 53% at 1 year to 80% at 4 years of therapy (P < .001).[23] This study did not document a survival advantage for imatinib because of the crossover design. Studies comparing the survival of imatinib-treated patients with historical cohorts treated with interferon-alpha-based therapy demonstrated the anticipated survival advantage.[9,24,25] Figure 2 shows the survival of patients treated at M. D. Anderson Cancer Center since 1965 by year of therapy.

Safety Data

Most adverse events with imatinib therapy were mild to moderate in severity. Treatment was discontinued for adverse events in 3.1% of newly diagnosed patients, in 4% of patients in chronic phase after failure of interferon therapy, and in 4% to 5% of patients in accelerated-blastic phase.[10,26-28] The most frequently reported adverse events (all grades) were superficial edema (59%-76%), nausea (47%-73%), muscle cramps (28%-62%), vomiting (21%-58%), diarrhea (39%-57%), musculoskeletal pain (40%-49%), and rash (37%-47%). Severe adverse experiences (grade 3/4) included severe fluid retention (eg, pleural effusion, pulmonary edema, and ascites) in 1% to 6%, superficial edema in 1% to 6%, hemorrhage in 1% to 19%, and musculoskeletal pain in 2% to 9%. Severe fluid retention appeared to be dose-related and was more common in the advanced-phase studies with imatinib dosages of 600 mg/d, and in the elderly.

Grade 3/4 laboratory abnormalities included neutropenia (3%-48%), anemia (< 1%-42%), thrombocytopenia (< 1%-42%), and hepatotoxicity (3%-6%). Treatment was discontinued permanently because of liver function abnormalities in less than 0.5% of patients.

Recently, imatinib was reported to be associated with cardiotoxicity and congestive heart failure, although this toxicity is rare.[29] Among 1,276 patients treated at one institution, 22 patients (1.8%), with a median age of 70 years, were identified as having symptoms that could be attributed to congestive heart failure.[30] At the time these events were reported, 8 were considered possibly or probably related to imatinib. Eighteen patients had previous medical conditions predisposing to cardiac disease: congestive heart failure (6 patients, 27%), diabetes mellitus (6 patients, 27%), hypertension (10 patients, 45%), coronary artery disease (8 patients, 36%), arrhythmia (3 patients, 14%) and cardiomyopathy (1 patient, 5%). Of the 22 patients, 11 continued imatinib therapy with dose adjustments and management for congestive heart failure symptoms with no further complications.

Management of Adverse Events

Myelosuppression, the most common adverse event with imatinib, is managed with dose interruptions and modifications.[31] The use of hematopoietic growth factors (granulocyte colony-stimulating factor [filgrastim, Neupogen] for neutropenia, oprelvekin [interleukin 11, Neumega] for thrombocytopenia, epoetin alfa [Epogen, Procrit] or darbepoetin [Aranesp] for anemia) has been reported to be safe and effective in patients with recurrent or persistent cytopenias.[32-38] Myelosuppression is frequently seen during the first 2 to 3 months of therapy. A brief treatment interruption is often sufficient to allow recovery, and most patients do not require dose reductions.

TABLE 2


Nonhematologic Adverse Events With Imatinib Therapy

Nonhematologic adverse events are relatively common but mild with imatinib, and can be managed easily with supportive care measures (Table 2). Early intervention is important to avoid more significant problems, unnecessary treatment interruptions, and dose reductions.

Imatinib Dose Schedules

The optimal dose of imatinib is not clearly defined. In the dose-finding phase I study, the maximum tolerated dose was not identified. Because of the good responses obtained at doses of 300 to 400 mg daily, particularly in patients in chronic phase, and because the blood concentration of imatinib at 400 mg daily was consistently higher than that required to inhibit 50% of BCR-ABL tyrosine kinase activity in vitro,[39,40] that dose was chosen for subsequent studies. Imatinib at 600 mg daily was more effective than 400 mg for patients in accelerated-blastic phase disease.[27,28] Among patients with CML in late chronic phase in whom interferon-alpha failed, treatment with imatinib at 400 mg twice daily resulted in a complete cytogenetic response rate of 90%, compared with 48% in historical matched controls treated with standard-dose therapy. In addition, 56% of patients had a major molecular response, including 41% with undetectable levels.[41] The toxicity profile was similar to that of imatinib at 400 mg, although there was a higher rate of grade 3/4 myelosuppression.

In patients with both prior hematologic and cytogenetic resistance to 400 mg of imatinib daily, increasing the dose to 800 mg resulted in a complete hematologic response rate of 65% and a complete cytogenetic response rate of 18%.[42] Several phase II studies, using high-dose imatinib in patients with previously untreated CML in chronic phase, have documented complete cytogenetic response rates of up to 95% and higher rates of molecular responses, particularly at the level of a 4-log or greater reduction in transcript levels.[9,43-45]

When compared with historical matched cohorts of patients treated with standard-dose therapy, patients treated with high-dose imatinib had higher rates of complete cytogenetic response (91% vs 76%, P = .002). Moreover, these occurred earlier, with 88% achieving this response after 6 months of therapy vs 56% with standard-dose therapy (P < .00001). The cumulative incidences of major molecular response and complete molecular response were significantly better with high-dose imatinib.[46] Progression-free and transformation-free survivals were significantly better in the high-dose group (P = .02 and .005). Results from ongoing randomized studies will determine whether the positive results obtained with high-dose imatinib will translate into prolonged progression-free and/or overall survival.

Monitoring Imatinib Therapy and Minimal Residual Disease

Given that most patients achieve a complete cytogenetic response with imatinib, achievement of molecular response has become the endpoint of anti-CML strategies and has led to the redefinition of therapeutic goals in CML. The IRIS trial showed that a reduction of BCR-ABL transcript levels by 3 or more logs below a standard baseline value correlated with progression-free survival.[13] In addition, one study reported that achieving a major molecular response within the first 12 months of therapy is predictive of durable cytogenetic remission.[12] However, the lack of consistency in reporting BCR-ABL transcript levels has been a source of debate.

A recent consensual proposal suggests harmonizing the differing methodologies for measuring BCR-ABL transcripts by using a conversion factor, whereby individual laboratories can express BCR-ABL transcript levels using scales that are internationally agreed upon: Results will be converted by comparing analysis of standardized reference samples with a value of 0.1% corresponding to major molecular response in all laboratories.[47] For practical purposes, a major molecular response can be defined as a reduction of BCR-ABL/ABL transcripts to 0.1% or less.[47] After a patient has achieved a complete cytogenetic response, real time polymerase chain reaction (RT-PCR) should be performed every 3 months and a routine cytogenetic analysis of bone marrow, every 12 months. This latter test may permit the detection of clonal evolution and chromosomal abnormalities occurring in normal metaphases.

Significance of Cytogenetic Abnormalities

Following treatment for CML, chromosome abnormalities have been detected in cells that do not contain the Ph chromosome. In most cases, these have followed imatinib treatment,[48-54] although isolated cases have been detected with interferon-alpha treatment.[32,33] Estimates of incidence from retrospective studies (> 100 imatinib-treated patients) range from 1.6% to 6.4%,[27-29,31] with a rate of 3.4% (34/1,001) reported in the largest study to date.[30]

The most commonly reported chromosomal changes associated with imatinib are trisomy 8 and monosomy 7, and changes may occur transiently in some patients. Risk factors identified for Ph-negative abnormalities include previous exposure to cytarabine or idarubicin[26] and shorter interval (≤ 1 year) from disease diagnosis to initiation of imatinib treatment.[33] Chromosomal changes have been detected following both imatinib monotherapy and imatinib treatment after previous therapy.

The prognostic significance of Ph-negative abnormalities remains unclear. Although some investigators have suggested an association between Ph-negative abnormalities and myelodysplasia,[26] this relationship has not been found in other studies.[29-31] In addition, it is not known if chromosome abnormalities are induced by imatinib, or if the disappearance of Ph-positive cells following cytogenetic responses to imatinib enables other chromosomal abnormalities to be observed. One study, however, has demonstrated that imatinib induces chromosome aberrations in vitro in a dose-dependent manner.[34]

Imatinib Resistance and Monitoring of Mutations

Various mechanisms have been proposed to explain resistance to imatinib. A small proportion of cases are thought to result from increased expression of Bcr-Abl kinase through gene amplification.[58,59] Resistance might also result from decreased intracellular drug concentrations caused by drug efflux proteins[60,61] or imatinib binding by plasma proteins.[62,63] Clonal evolution might contribute to imatinib resistance.[59] Lyn and Hck are overexpressed in imatinib-resistant patient isolates and cell lines, suggesting that Src family kinases may be involved in BCR-ABLindependence and progression to imatinib resistance.[64-66] However, approximately 35%-45% of cases of imatinib resistance arise because of mutations in the BCR-ABL kinase domain.[59,67]

Clinically relevant BCR-ABLmutations disrupt critical contact points between imatinib and the Bcr-Abl protein or induce structural alternations that prevent imatinib binding, often by inducing a transition from the inactive to the active conformation of the protein, to which imatinib is unable to bind.[68,69] This illustrates the disadvantages of the high specificity of imatinib for wild-type Bcr-Abl in its inactive conformation. With continued imatinib treatment, resistant mutants are selected and eventually outgrow drug-sensitive leukemic cells. The number of different BCR-ABL mutations identified has steadily increased. Clusters of mutations have been described in different areas of the Bcr-Abl molecule, and include the ATP binding site (P-loop), the imatinib binding site, the activation loop (controlling kinase activation), and the catalytic domain.[68,69] The first mutation to be identified, T315I,[58] is of particular importance, as will be discussed below.

Some BCR-ABL mutations result in a highly resistant phenotype in vitro, whereas others remain relatively sensitive, meaning that resistance can be overcome by increasing the imatinib dose.[59,70-74] Higher rates of mutations have been identified in patients who have received extended treatment, patients who did not achieve a major cytogenetic response, and those with more advanced disease.[75,76] It has also been suggested that mutations in the P-loop are associated with a poor prognosis,[75,77,78] although this has been contradicted in a separate analysis.[76] Kinase domain mutations screening for patients in chronic phase is indicated in cases of hematologic or cytogenetic resistance/relapse,[47] or if there is an increase in BCR-ABL transcript ratio of 1 log or greater.[79] Kinase domain mutations should be investigated in any patients presenting in advanced-phase disease.

Duration of Imatinib Therapy

The optimal duration of imatinib therapy is unknown. The current recommendation is to continue treatment indefinitely unless the patient experiences unacceptable toxicity or treatment failure. There is no evidence to support the concept that imatinib can safely be discontinued even after transcript levels become undetectable. Most patients who have discontinued imatinib therapy have experienced molecular or cytogenetic relapse even after sustained undetectable levels of BCR-ABL for significant periods of time.[80-82] Rousselot et al reported on 12 patients who discontinued imatinib after maintaining undetectable BCR-ABL levels for 24 months or longer; six are still PCR-negative after a median follow-up of 18 months (range: 9-24 months).[83] Of these 12 patients, 10 had been treated with interferon-alpha, suggesting that interferon-alpha immunomodulatory effects may account for the sustained and prolonged molecular responses observed upon therapy discontinuation. It has been suggested that the earliest, probably quiescent, progenitor cells in CML are insensitive to imatinib in vitro.[84] It is conceivable that these progenitors might trigger proliferation of CML once the inhibitory pressure of imatinib is eliminated.

Second-Generation Tyrosine Kinase Inhibitors

TABLE 3


Summary of Outcomes From the Dasatinib Phase II Clinical Development Program

Novel more potent tyrosine kinase inhibitors have been developed to overcome imatinib resistance. Dasatinib (Sprycel), an orally bioavailable dual Bcr-Abl and Src inhibitor, is now approved for the treatment of CML and Ph-positive acute lymphocytic leukemia after imatinib failure.[85] Dasatinib has been clinically assessed in one dose-ranging study and five subsequent studies involving more than 900 imatinib-resistant or -intolerant patients (the START program). Data from the dose-ranging study suggested that dasatinib treatment was associated with a high level of efficacy and durability.[85] Results from the START program demonstrated that treatment with dasatinib at 70 mg twice daily resulted in hematologic and cytogenetic responses across all phases of CML in both imatinib-intolerant and -resistant patients (Table 3).[86-90]

TABLE 4


Summary of Outcomes From Nilotinib Phase II Trials

Nilotinib (Tasigna) is an oral potent and selective Bcr-Abl inhibitor in advanced clinical trials.[91] In a phase I dose-escalation study (n = 119), nilotinib showed activity in patients with imatinib-resistant CML.[91] Nilotinib has also been examined in three ongoing phase II studies conducted in patients with CML and imatinib resistance or intolerance. Data are currently available from 145 patients who received nilotinib at 400 mg twice daily. Nilotinib has demonstrated clinical activity in all phases of CML; complete hematologic responses were recorded in 69%, 16%, and 4% of patients with chronic, accelerated, and blastic phase disease, respectively, and major cytogenetic responses were recorded in 46%, 28%, and 29% of patients (Table 4).[92-94]

Both agents have been investigated in patients with newly diagnosed CML. Preliminary results were recently reported for 14 patients with newly diagnosed CML treated with nilotinib at 400 mg twice daily.[95] A major cytogenetic response was observed in all patients at 3 months (complete in 13 and partial in 1); the complete cytogenetic response rate was 100% in all evaluable patients at 6 months (n = 13) and 9 months (n = 11). Major molecular response rates, at 6 and 9 months, were significantly higher with nilotinib compared to historical data with standard-dose and high-dose imatinib.[95] In another study, 24 patients with newly diagnosed CML in chronic phase were treated with dasatinib at 100 mg daily or 50 mg twice daily. The complete cytogenetic response rates at 6 and 9 months were, respectively, 73% and 95%; more favorable as compared with historical data in patients treated with standard-dose and high-dose imatinib.[96]

Two other novel agents, bosutinib (SKI-606) and INNO-406 (NS-187),[97,98] are being investigated in phase I/II clinical trials. Both compounds will eventually be explored in the front-line setting.

Stem Cell Transplantation

Because of the success of Bcr-Abl inhibitors, SCT has changed from being a preferred first-line therapy to a second- or third-line option. Despite this, SCT remains an important treatment option that offers the potential for disease cure, although this needs to be balanced with potential risks, including graft-vs-host disease, life-threatening infections, and secondary malignancy.[99,100]

Recent reports provide the best estimates of current outcomes following SCT. Data have been analyzed from 131 chronic phase CML patients receiving allogeneic SCT (bone marrow or peripheral blood) from related donors at a single institution in the United States between 1995 and 2000.[101] The estimated probability of disease-free survival at 3 years was 78%, with survival estimated at 86% and relapse estimated at 8%.

The Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT) recently provided updated data.[102] Among all European patients receiving SCT for CML between 2000 and 2003 (n = 3,018), the 2-year survival rate was 61%, transplant-related mortality was 30%, and relapse rate was 22%. However, in patients transplanted in first chronic phase from a human leukocyte antigen (HLA)-identical sibling, 2-year survival was 74%, transplant-related mortality was 22%, and relapse rate was 18%. These data illustrate that outcome following SCT is highly dependent on defined risk factors. In the EBMT study, favorable risk factors were identified as sibling donor, treatment at early disease stage, younger recipient age (< 20 vs 20-40 vs > 40), and less than 12 months from diagnosis to transplantation.[102,103]

If successful, SCT can have long-term results. In a 10-year study of patients receiving allogeneic bone marrow from siblings (46 in first chronic phase, 43 in advanced phases), mean time to hematologic or cytogenetic relapse was 7.7 years, and 46% (13/28) of long-term survivors never relapsed.[104]

Related vs Unrelated Donors

REFERENCE GUIDE

Therapeutic Agents
Mentioned in This Article

Bosutinib (SKI-606)
Cytarabine
Darbepoetin (Aranesp)
Dasatinib (Sprycel)
Epoetin alfa (Epogen, Procrit)
Granulocyte colony-stimulating factor (filgrastim, Neupogen)
Idarubicin
Imatinib mesylate (Gleevec)
INNO-406 (NS-187)
Interferon-alpha
Nilotinib (Tasigna)
Oprelvekin (interleukin 11, Neumega)

Brand names are listed in parentheses only if a drug is not available generically and is marketed as no more than two trademarked or registered products. More familiar alternative generic designations may also be included parenthetically.

Although SCT outcomes are better with related donors, one-half to two-thirds of patients do not have a suitable HLA-matched sibling. This has led to assessment of treatment with unrelated HLA-matched donors.[105] One study has compared results from 2,464 unrelated donor bone marrow transplants with 450 HLA-identical, sibling donor transplants performed at National Marrow Donor Program institutions in the United States between 1988 and 1999.[106] In the unrelated donor group, 63% were matched at HLA-A, -B, and -DRB1 alleles.

Multivariate analysis demonstrated a significantly increased risk of graft failure and acute graft-vs-host disease following transplant from an unrelated as opposed to a related donor. In addition, survival and disease-free survival were significantly worse for unrelated donor recipients. However, for those with chronic phase disease undergoing transplantation within 1 year of diagnosis, the 5-year disease-free survival rate was similar or only slightly inferior. Hematologic relapse was uncommon in both groups and occurred with similar frequency (8%-9%). Several studies have confirmed that complete donor-recipient matching across HLA-A,-B,-C,-DRB1, and -DQB1 alleles can significantly reduce the incidence of graft-vs-host disease and the risk of mortality.[107]

Modifying SCT Outcome

Reduced-intensity conditioning prior to SCT has been used to try and increase tolerability and decrease mortality. This approach takes advantage of a graft-vs-leukemia effect, whereby CML cells are eliminated by donor cells instead of ablative chemotherapy. Although long-term follow-up is not yet available, early results from small studies suggest that rates of survival and disease-free survival may be high.[108,109]

Although donor leukocyte infusions are extremely useful for treating disease relapses in SCT recipients,[110] accumulating evidence demonstrates that imatinib is also useful in this regard.[111-114] In addition, current evidence suggests that prior imatinib treatment does not adversely affect SCT outcome.[115]

Immunotherapy and Residual Disease

The observation of a graft-vs-leukemia effect following SCT suggests that immunotherapy aimed at CML is an approach worth investigating. Because the BCR-ABL fusion protein represents a unique tumor-specific antigen, vaccination using peptides based on the BCR-ABL junction point may be useful, especially in helping to reduce residual disease levels after maximal responses to imatinib have been achieved.[116,117] Two recent clinical studies of vaccination, using peptides derived from the BCR-ABL fusion region, have demonstrated that patients develop a functional immune response to these peptides.[118,119] In one study, vaccination of nine patients who had achieved stable but incomplete cytogenetic responses (median Ph-positive level: 10%) on imatinib resulted in improved cytogenetic responses, including a complete cytogenetic response in five patients. In addition, three patients achieved a complete molecular response.

Conclusions

Imatinib has provided treatment benefits for a large number of patients with CML. Long-term outcomes after 5 years of follow-up are encouraging and suggest a major change in the history of the disease. Adequate management, proper follow-up, and opportune intervention with imatinib dose increases in patients with suboptimal responses can maximize the benefit. The availability of highly potent tyrosine kinase inhibitors has broadened the treatment armamentarium in CML. Long-term treatment of CML may require a combination of tyrosine kinase inhibitors, farnesyl transferase inhibitors, and, possibly, compounds with other mechanisms of action-both conventional and targeted. Vaccines to stimulate patient immunity may control and eliminate residual disease. Overall, treatment options are increasing for patients with CML, and this is likely to continue in the future.

Financial Disclosure:Dr. Cortes receives research grants from Novartis and Bristol-Myers Squibb.

References:

1. Jemal A, Siegel R, Ward E, et al: Cancer statistics, 2006. CA Cancer J Clin 56:106-130, 2006.

2. Faderl S, Talpaz M, Estrov Z, et al: The biology of chronic myeloid leukemia. N Engl J Med 341:164-172, 1999.

3. Goldman JM, Melo JV: Chronic myeloid leukemia-advances in biology and new approaches to treatment. N Engl J Med 349:1451-1464, 2003.

4. Barnes DJ, Melo JV: Cytogenetic and molecular genetic aspects of chronic myeloid leukaemia. Acta Haematol 108:180-202, 2002.

5. Rowley JD: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature 243:290-293, 1973.

6. Thomas ED, Clift RA, Fefer A, et al: Marrow transplantation for the treatment of chronic myelogenous leukemia. Ann Intern Med 104:155-163, 1986.

7. Kantarjian HM, O'Brien S, Cortes J, et al: Complete cytogenetic and molecular responses to interferon-alpha-based therapy for chronic myelogenous leukemia are associated with excellent long-term prognosis. Cancer 97:1033-1041, 2003.

8. Kantarjian H, Sawyers C, Hochhaus A, et al: Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl J Med 346:645-652, 2002.

9. Kantarjian HM, Cortes JE, O'Brien S, et al: Long-term survival benefit and improved complete cytogenetic and molecular response rates with imatinib mesylate in Philadelphia chromosome-positive chronic-phase chronic myeloid leukemia after failure of interferon-{alpha}. Blood 104:1979-1988, 2004.

10. 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 348:994-1004, 2003.

11. Kantarjian H, Talpaz M, O'Brien S, et al: High-dose imatinib mesylate therapy in newly diagnosed Philadelphia chromosome-positive chronic phase chronic myeloid leukemia. Blood 103:2873-2878, 2004.

12. Cortes J, Talpaz M, O'Brien S, et al: Molecular responses in patients with chronic myelogenous leukemia in chronic phase treated with imatinib mesylate. Clin Cancer Res 11:3425-3432, 2005.

13. 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 349:1423-1432, 2003.

14. 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 2:561-566, 1996.

15. Beran M, Cao X, Estrov Z, et al: Selective inhibition of cell proliferation and BCR-ABL phosphorylation in acute lymphoblastic leukemia cells expressing Mr 190,000 BCR-ABL protein by a tyrosine kinase inhibitor (CGP-57148). Clin Cancer Res 4:1661-1672, 1998.

16. 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 56:100-104, 1996.

17. Buchdunger E, Cioffi CL, Law N, et al: Abl protein-tyrosine kinase inhibitor STI571 inhibits in vitro signal transduction mediated by c-kit and platelet-derived growth factor receptors. J Pharmacol Exp Ther 295:139-145, 2000.

18. Okuda K, Weisberg E, Gilliland DG, et al: ARG tyrosine kinase activity is inhibited by STI571. Blood 97:2440-2448, 2001.

19. Heinrich MC, Blanke CD, Druker BJ, et al: Inhibition of KIT tyrosine kinase activity: a novel molecular approach to the treatment of KIT-positive malignancies. J Clin Oncol 20:1692-1703, 2002.

20. Dewar AL, Cambareri AC, Zannettino AC, et al: Macrophage colony stimulating factor receptor, c-fms, is a novel target of imatinib. Blood 105:3127-3132, 2005.

21. Ren R: Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia. Nat Rev Cancer 5:172-183, 2005.

22. 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 348:994-1004, 2003.

23. Druker BJ, Guilhot F, O'Brien S, et al: Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med 355:2408-2417, 2006.

24. Kantarjian HM, O'Brien S, Cortes J, et al: Imatinib mesylate therapy improves survival in patients with newly diagnosed Philadelphia chromosome-positive chronic myelogenous leukemia in the chronic phase. Comparison with historic data. Cancer 98:2636-2642, 2003.

25. Kantarjian HM, Talpaz M, O'Brien S, et al: Survival benefit with imatinib mesylate versus interferon alpha-based regimens in newly diagnosed chronic phase chronic myelogenous leukemia. Blood 108:1835-1840, 2006.

26. Roy L, Guilhot J, Kranhke T, et al: Survival advantage from imatinib compared to the combination interferon-{alpha} plus cytarabine in chronic phase CML: Historical comparison between two phase III trials. Blood 108:1478-1484, 2006.

27. 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 99:1928-1937, 2002.

28. Sawyers CL, Hochhaus A, Feldman E, et al: Imatinib induces hematologic and cytogenetic responses in patients with chronic myelogenous leukemia in myeloid blast crisis: Results of a phase II study. Blood 99:3530-3539, 2002.

29. Kerkela R, Grazette L, Yacobi R, et al: Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med 12:908-916, 2006.

30. Atallah E, Durand JB, Kantarjian H, et al: Congestive heart failure is a rare event in patients receiving imatinib therapy (abstract 246). Blood 108:2006.

31. Deininger MW, O'Brien SG, Ford JM, et al: Practical management of patients with chronic myeloid leukemia receiving imatinib. J Clin Oncol 21:1637-1647, 2003.

32. Quintas-Cardama A, Kantarjian H, O'Brien S, et al: Granulocyte-colony-stimulating factor (filgrastim) may overcome imatinib-induced neutropenia in patients with chronic-phase chronic myelogenous leukemia. Cancer 100:2592-2597, 2004.

33. Mauro MJ, Kurilik G, Balleisen S, et al: Myeloid growth factors for neutropenia during imatinib mesylate therapy for CML: Preliminary evidence of safety and efficacy (abstract). Blood 98:139a, 2001.

34. Marin D, Marktel S, Foot N, et al: G-CSF reverses cytopenia and may increase cytogenetic responses in patients with CML treated with imatinib mesylate (abstract). Blood 100:782a, 2002.

35. Ault P, Kantarjian H, Welch MA, et al: Interleukin 11 may improve thrombocytopenia associated with imatinib mesylate therapy in chronic myelogenous leukemia. Leuk Res 28:613-618, 2004.

36. Cortes J, O'Brien S, Quintas A, et al: Erythropoietin is effective in improving the anemia induced by imatinib mesylate therapy in patients with chronic myeloid leukemia in chronic phase. Cancer 100:2396-2402, 2004.

37. Mauro MJ, Blasdel C, O'Dwyer ME, et al: Erythropoietin for anemia during imatinib mesylate (STI571) therapy for CML: Preliminary evidence of safety and efficacy (abstract 106). Proc Am Soc Clin Oncol 21:27a, 2002.

38. Ault P, Kantarjian H, O'Brien S, et al: Use of darbepoetin alfa for the treatment of anemia occurring during imatinib therapy for CML: Preliminary evidence of safety and efficacy (abstract 2467). Proc Am Soc Clin Oncol 22:613, 2003.

39. Peng B, Hayes M, Resta D, et al: Pharmacokinetics and pharmacodynamics of imatinib in a phase I trial with chronic myeloid leukemia patients. J Clin Oncol 22:935-942, 2004.

40. Schmidli H, Peng B, Riviere GJ, et al: Population pharmacokinetics of imatinib mesylate in patients with chronic-phase chronic myeloid leukaemia: Results of a phase III study. Br J Clin Pharmacol 60:35-44, 2005.

41. Cortes J, Giles F, O'Brien S, et al: Result of high-dose imatinib mesylate in patients with Philadelphia chromosome-positive chronic myeloid leukemia after failure of interferon-á. Blood 102:83-86, 2003.

42. Kantarjian H, Talpaz M, O'Brien S, et al: Dose escalation of imatinib mesylate can overcome resistance to standard-dose therapy in patients with chronic myelogenous leukemia. Blood 101:473-475, 2003.

43. Hughes TP, Branford S, Matthews J, et al: Trial of higher dose imatinib with selective intensification in newly diagnosed CML patients in chronic phase (abstract 95). Blood 102:31a, 2003.

44. Cortes J, Giles F, Salvado A, et al: High-dose (HD) imatinib in patients with previously untreated chronic myeloid leukemia (CML) in early chronic phase (CP): Preliminary results of a multicenter community based trial (abstract 6518). J Clin Oncol 23(16S):564s, 2005.

45. Rosti G, Martinelli G, Castagnetti F, et al: Imatinib 800 mg: preliminary results of a phase II trial of the GIMEMA CML working party in intermediate sokal risk patients and status-of-the-art of an ongoing multinational, prospective randomized trial of imatinib standard dose (400 mg daily) s high dose (800 mg daily) in high Sokal risk patients (abstract 1098). Blood 106:320a, 2005.

46. Aoki E, Kantarjian H, O'Brien S, et al: High-dose (HD) imatinib provides better responses in patients with untreated early chronic phase (CP) chronic myeloid leukemia (CML) (abstract 2143). Blood 108:2006.

47. Hughes TP, Deininger MW, Hochhaus A, et al: Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: review and recommendations for harmonizing current methodology for detecting BCR-ABL transcripts and kinase domain mutations and for expressing results. Blood 108:28-37, 2006.

48. Bumm T, Muller C, Al-Ali HK, et al: Emergence of clonal cytogenetic abnormalities in Ph- cells in some CML patients in cytogenetic remission to imatinib but restoration of polyclonal hematopoiesis in the majority. Blood 101:1941-1949, 2003.

49. Feldman E, Najfeld V, Schuster M, et al: The emergence of Ph-, trisomy -8+ cells in patients with chronic myeloid leukemia treated with imatinib mesylate. Exp Hematol 31:702-707, 2003.

50. Marktel S, Marin D, Foot N, et al: Chronic myeloid leukemia in chronic phase responding to imatinib: The occurrence of additional cytogenetic abnormalities predicts disease progression. Haematologica 88:260-267, 2003.

51. Medina J, Kantarjian H, Talpaz M, et al: Chromosomal abnormalities in Philadelphia chromosome-negative metaphases appearing during imatinib mesylate therapy in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase. Cancer 98:1905-1911, 2003.

52. Terre C, Eclache V, Rousselot P, et al: Report of 34 patients with clonal chromosomal abnormalities in Philadelphia-negative cells during imatinib treatment of Philadelphia-positive chronic myeloid leukemia. Leukemia 18:1340-1346, 2004.

53. Jabbour E, Kantarjian H, O'Brien S, et al: Chromosomal abnormalities in Philadelphia chromosome (Ph)-negative metaphases appearing during imatinib mesylate (IM) therapy in patients (pts) with newly diagnosed chronic myeloid leukemia (CML) in chronic phase (abstract 1090). Blood 106:317a, 2005.

54. Lin Y, Bruyere H, Horsman DE, et al: Philadelphia-negative clonal hematopoiesis following imatinib therapy in patients with chronic myeloid leukemia: A report of nine cases and analysis of predictive factors. Cancer Genet Cytogenet 170:16-23, 2006.

55. Fayad L, Kantarjian H, O'Brien S, et al: Emergence of new clonal abnormalities following interferon-alpha induced complete cytogenetic response in patients with chronic myeloid leukemia: Report of three cases. Leukemia 11:767-771, 1997.

56. Izumi T, Imagawa S, Hatake K, et al: Philadelphia chromosome-negative cells with trisomy 8 after busulfan and interferon treatment of Ph1-positive chronic myelogenous leukemia. Int J Hematol 64:73-77, 1996.

57. Fabarius A, Giehl M, Frank O, et al: Induction of centrosome and chromosome aberrations by imatinib in vitro. Leukemia 19:1573-1578, 2005.

58. Gorre ME, Mohammed M, Ellwood K, et al: Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 293:876-880, 2001.

59. Hochhaus A, Kreil S, Corbin AS, et al: Molecular and chromosomal mechanisms of resistance to imatinib (STI571) therapy. Leukemia 16:2190-2196, 2002.

60. Illmer T, Schaich M, Platzbecker U, et al: P-glycoprotein-mediated drug efflux is a resistance mechanism of chronic myelogenous leukemia cells to treatment with imatinib mesylate. Leukemia 18:401-408, 2004.

61. Thomas J, Wang L, Clark RE, et al: Active transport of imatinib into and out of cells: Implications for drug resistance. Blood 104:3739-3745, 2004.

62. Larghero J, Leguay T, Mourah S, et al: Relationship between elevated levels of the alpha 1 acid glycoprotein in chronic myelogenous leukemia in blast crisis and pharmacological resistance to imatinib (Gleevec) in vitro and in vivo. Biochem Pharmacol 66:1907-1913, 2003.

63. le Coutre P, Kreuzer KA, Na IK, et al: Determination of alpha-1 acid glycoprotein in patients with Ph+ chronic myeloid leukemia during the first 13 weeks of therapy with STI571. Blood Cells Mol Dis 28:75-85, 2002.

64. Lionberger JM, Wilson MB, Smithgall TE: Transformation of myeloid leukemia cells to cytokine independence by Bcr-Abl is suppressed by kinase-defective Hck. J Biol Chem 275:18581-18585, 2000.

65. Donato NJ, Wu JY, Stapley J, et al: BCR-ABL independence and LYN kinase overexpression in chronic myelogenous leukemia cells selected for resistance to STI571. Blood 101:690-698, 2003.

66. Dai Y, Rahmani M, Corey SJ, et al: A Bcr/Abl-independent, Lyn-dependent form of imatinib mesylate (STI-571) resistance is associated with altered expression of Bcl-2. J Biol Chem 279:34227-34239, 2004.

67. Lahaye T, Riehm B, Berger U, et al: Response and resistance in 300 patients with BCR-ABL-positive leukemias treated with imatinib in a single center: A 4.5-year follow-up. Cancer 103:1659-1669, 2005.

68. Deininger M, Buchdunger E, Druker BJ: The development of imatinib as a therapeutic agent for chronic myeloid leukemia. Blood 105:2640-2653, 2005.

69. Litzow MR: Imatinib resistance: Obstacles and opportunities. Arch Pathol Lab Med 130:669-679, 2006.

70. Corbin AS, Buchdunger E, Pascal F, et al: Analysis of the structural basis of specificity of inhibition of the Abl kinase by STI571. J Biol Chem 277:32214-32219, 2002.

71. Corbin AS, La RP, Stoffregen EP, et al: Several Bcr-Abl kinase domain mutants associated with imatinib mesylate resistance remain sensitive to imatinib. Blood 101:4611-4614, 2003.

72. Azam M, Latek RR, Daley GQ: Mechanisms of autoinhibition and STI-571/imatinib resistance revealed by mutagenesis of BCR-ABL. Cell 112:831-843, 2003.

73. 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 2:117-125, 2002.

74. von Bubnoff N, Schneller F, Peschel C, et al: BCR-ABL gene mutations in relation to clinical resistance of Philadelphia-chromosome-positive leukaemia to STI571: A prospective study. Lancet 359:487-491, 2002.

75. Branford S, Rudzki Z, Walsh S, et al: Detection of BCR-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop) are associated with a poor prognosis. Blood 102:276-283, 2003.

76. Jabbour E, Kantarjian H, Jones D, et al: Frequency and clinical significance of BCR-ABL mutations in patients with chronic myeloid leukemia treated with imatinib mesylate. Leukemia 20:1767-1773, 2006.

77. Soverini S, Martinelli G, Rosti G, et al: ABL mutations in late chronic phase chronic myeloid leukemia patients with up-front cytogenetic resistance to imatinib are associated with a greater likelihood of progression to blast crisis and shorter survival: A study by the GIMEMA Working Party on Chronic Myeloid Leukemia. J Clin Oncol 23:4100-4109, 2005.

78. Nicolini FE, Corm S, Le QH, et al: Mutation status and clinical outcome of 89 imatinib mesylate-resistant chronic myelogenous leukemia patients: A retrospective analysis from the French intergroup of CML (Fi(varphi)-LMC GROUP). Leukemia 20:1061-1066, 2006.

79. Branford S, Rudzki Z, Parkinson I, et al: Real-time quantitative PCR analysis can be used as a primary screen to identify patients with CML treated with imatinib who have BCR-ABL kinase domain mutations. Blood 104:2926-2932, 2004.

80. Cortes J, O'Brien S, Kantarjian H: Discontinuation of imatinib therapy after achieving a molecular response. Blood 104:2204-2205, 2004.

81. Mauro MJ, Druker BJ, Maziarz RT: Divergent clinical outcome in two CML patients who discontinued imatinib therapy after achieving a molecular remission. Leukemia Res 28:71-73, 2004.

82. Ghanima W, Kahrs J, Dahl TG 3rd, et al: Sustained cytogenetic response after discontinuation of imatinib mesylate in a patient with chronic myeloid leukaemia. Eur J Haematol 72:441-443, 2004.

83. Rousselot P, Huguet F, Rea D, et al: Imatinib mesylate discontinuation in patients with chronic myelogenous leukaemia in complete molecular remission for more than two years. Blood 109:58-60, 2007.

84. Graham SM, Jorgensen HG, Allan E, et al: Primitive quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro. Blood. 2002; 99:319-325.

85. Talpaz M, Shah NP, Kantarjian H, et al: Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N Engl J Med 354:2531-2541, 2006.

86. Cortes JE, Kim DW, Rosti G, et al: Dasatinib (D) in patients (pts) with chronic myelogenous leukemia (CML) in myeloid blast crisis (MBC) who are imatinib-resistant (IM-R) or IM-intolerant (IM-I): Results of the CA180006 'START-B' study (abstract 6529). J Clin Oncol 24(18S):344s, 2006.

87. Coutre S, Martinelli G, Dombret H, et al: Dasatanib (D) in patients (pts) with chronic myelogenous leukemia (CML) in lymphoid blast crisis (LB-CML) or Philadelphia-chromosome positive acute lymphoblastic leukemia (Ph+ALL) who are imatinib (IM)-resistant (IM-R) or intolerant (IM-I): The CA180015 'START-L' study (abstract 6528). J Clin Oncol 24(18S):344s, 2006.

88. Hochhaus A, Kantarjian H, Baccarani M, et al: Dasatinib in patients with chronic phase chronic myeloid leukemia (CP-CML) who are resistant or intolerant to imatinib: Results of the CA180013 'START-C' Study (abstract 6508). J Clin Oncol 24(18S):339s, 2006.

89. Shah NP, Rousselot P, Pasquini R, et al: Dasatinib (D) vs high dose imatinib (IM) in patients (pts) with chronic phase chronic myeloid leukemia (CP-CML) resistant to imatinib. Results of CA180017 START-R randomized trial (abstract 6507). J Clin Oncol 24(18S):338s, 2006.

90. Talpaz M, Apperley JF, Kim DW, et al: Dasatinib (D) in patients with accelerated phase chronic myeloid leukemia (AP-CML) who are resistant or intolerant to imatinib: Results of the CA180005 'START-A' study. J Clin Oncol 24(18S):343s, 2006

91. Kantarjian H, Giles F, Wunderle L, et al: Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL. N Engl J Med 354:2542-2551, 2006.

92. Giles FJ, Larson R, le Coutre P, et al: A phase II study of AMN107, a novel inhibitor of Bcr-Abl, administered to imatinib-resistant or intolerant patients (pts) with Ph+ chronic myelogenous leukemia (CML) in blast crisis (BC) or relapsed/refractory Ph+ acute lymphoblastic leukemia (ALL) (abstract 6536). J Clin Oncol 24(18S):346s, 2006.

93. Kantarjian HM, Gattermann N, O'Brien SG, et al: A phase II study of AMN107, a novel inhibitor of Bcr-Abl, administered to imatinib resistant and intolerant patients (pts) with chronic myelogenous leukemia (CML) in chronic phase (CP) (abstract 6534). J Clin Oncol 24(18S):345s, 2006.

94. Le Coutre PD, Ottmann O, Gatterman N, et al: A phase II study of AMN107, a novel inhibitor of Bcr-Abl, administered to imatinib-resistant or intolerant patients (pts) with chronic myelogenous leukemia (CML) in accelerated phase (AP) (abstract 6531). J Clin Oncol 24(18S):344s, 2006.

95. Jabbour E, Cortes J, Giles F, et al: Preliminary activity of AMN107, a novel potent oral selective Bcr-Abl tyrosine kinase inhibitor, in newly diagnosed Philadelphia chromosome (Ph)-positive chronic phase chronic myelogenous leukaemia (CML-CP) (abstract 2172). Blood 108:2006.

96. Cortes J, O'Brien S, Jones D, et al: Dasatinib in patients with previously untreated chronic myeloid leukemia in chronic phase (abstract 2161). Blood 108:2006.

97. Cortes J, Kantarjian H, Baccarani M, et al: Phase 1/2 study of SKI-606, a dual inhibitor of Src and Abl kinases, in adults patients with Philadelphia chromosome positive (Ph+) chronic myelogenous leukemia (CML) or acute lymphocytic leukemia (ALL) relapsed refractory or intolerant to imatinib (abstract 168). Blood 108:2006.

98. Naito H, Kimura S, Nakaya Y, et al: In vivo antiproliferative effect of NS-187, a dual Bcr-Abl/Lyn tyrosine kinase inhibitor, on leukemic cells harbouring Abl kinase domain mutations. Leuk Res 30:1443-1446, 2006.

99. Grigg A, Hughes T: Role of allogeneic stem cell transplantation for adult chronic myeloid leukemia in the imatinib era. Biol Blood Marrow Transplant 12:795-807, 2006.

100. Jabbour E, Cortes J, Kantarjian, et al: Allogeneic stem cell transplantation for patients with chronic myeloid leukemia and acute lymphocytic leukemia after Bcr-Abl kinase mutation-related imatinib failure. Blood 108:1421-1423, 2006.

101. Radich JP, Gooley T, Bensinger W, et al: HLA-matched related hematopoietic cell transplantation for chronic-phase CML using a targeted busulfan and cyclophosphamide preparative regimen. Blood 102:31-35, 2003.

102. Gratwohl A, Brand R, Apperley J, et al: Allogeneic hematopoietic stem cell transplantation for chronic myeloid leukemia in Europe 2006: Transplant activity, long-term data and current results. An analysis by the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT). Haematologica 91:513-521, 2006.

103. Gratwohl A, Hermans J, Goldman JM, et al: Risk assessment for patients with chronic myeloid leukaemia before allogeneic blood or marrow transplantation. Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Lancet 352:1087-1092, 1998.

104. Kiss TL, Abdolell M, Jamal N, et al: Long-term medical outcomes and quality-of-life assessment of patients with chronic myeloid leukemia followed at least 10 years after allogeneic bone marrow transplantation. J Clin Oncol 20:2334-2343, 2002.

105. Hansen JA, Gooley TA, Martin PJ, et al: Bone marrow transplants from unrelated donors for patients with chronic myeloid leukemia. N Engl J Med 338:962-968, 1998.

106. Weisdorf DJ, Anasetti C, Antin JH, et al: Allogeneic bone marrow transplantation for chronic myelogenous leukemia: Comparative analysis of unrelated versus matched sibling donor transplantation. Blood 99:1971-1977, 2002.

107. Mickelson EM, Petersdorf EW, Hansen JA: HLA matching and hematopoietic cell transplant outcome. Clin Transpl 263-271, 2002.

108. Or R, Shapira MY, Resnick I, et al: Nonmyeloablative allogeneic stem cell transplantation for the treatment of chronic myeloid leukemia in first chronic phase. Blood 101:441-445, 2003.

109. Crawley C, Szydlo R, Lalancette M, et al: Outcomes of reduced-intensity transplantation for chronic myeloid leukemia: An analysis of prognostic factors from the Chronic Leukemia Working Party of the EBMT. Blood 106:2969-2976, 2005.

110. Gilleece MH, Dazzi F: Donor lymphocyte infusions for patients who relapse after allogeneic stem cell transplantation for chronic myeloid leukaemia. Leuk Lymphoma 44:23-28, 2003.

111. Kantarjian HM, O'Brien S, Cortes JE, et al: Imatinib mesylate therapy for relapse after allogeneic stem cell transplantation for chronic myelogenous leukemia. Blood 100:1590-1595, 2002.

112. DeAngelo DJ, Hochberg EP, Alyea EP, et al: Extended follow-up of patients treated with imatinib mesylate (gleevec) for chronic myelogenous leukemia relapse after allogeneic transplantation: Durable cytogenetic remission and conversion to complete donor chimerism without graft-versus-host disease. Clin Cancer Res 10:5065-5071, 2004.

113. Hess G, Bunjes D, Siegert W, et al: Sustained complete molecular remissions after treatment with imatinib-mesylate in patients with failure after allogeneic stem cell transplantation for chronic myelogenous leukemia: Results of a prospective phase II open-label multicenter study. J Clin Oncol 23:7583-7593, 2005.

114. Olavarria E, Ottmann OG, Deininger M, et al: Response to imatinib in patients who relapse after allogeneic stem cell transplantation for chronic myeloid leukemia. Leukemia 17:1707-1712, 2003.

115. Deininger M, Schleuning M, Greinix H, et al: The effect of prior exposure to imatinib on transplant-related mortality. Haematologica 91:452-459, 2006.

116. Pinilla-Ibarz J, Cathcart K, Scheinberg DA: CML vaccines as a paradigm of the specific immunotherapy of cancer. Blood Rev 14:111-120, 2000.

117. Quintas-Cardama A, Cortes J: Chronic myeloid leukemia: Diagnosis and treatment. Mayo Clin Proc 81:973-988, 2006.

118. Cathcart K, Pinilla-Ibarz J, Korontsvit T, et al: A multivalent bcr-abl fusion peptide vaccination trial in patients with chronic myeloid leukemia. Blood 103:1037-1042, 2004.

119. Bocchia M, Gentili S, Abruzzese E, et al: Effect of a p210 multipeptide vaccine associated with imatinib or interferon in patients with chronic myeloid leukaemia and persistent residual disease: A multicenter observational trial. Lancet 365:657-662, 2005.

Related Videos
Pegulicianine-guided breast cancer surgery may allow practices to de-escalate subsequent radiotherapy, says Barbara Smith, MD, PhD.
Barbara Smith, MD, PhD, spoke about the potential use of pegulicianine-guided breast cancer surgery based on reports from the phase 3 INSITE trial.
Carey Anders, MD, an expert on breast cancer