fbpx

אתר הבית של חולי CML

המאמר החדש של 19 מומחים עולמיים לגבי נוהל טיפול בחולי CML- באנגלית


1

EVOLVING CONCEPTS IN THE MANAGEMENT OF CHRONIC MYELOID

LEUKEMIA. RECOMMENDATIONS FROM AN EXPERT PANEL ON BEHALF OF

THE EUROPEAN LEUKEMIANET.

Running title: Management of Chronic Myeloid Leukemia.

Michele Baccarani1, Giuseppe Saglio2, John Goldman3, Andreas Hochhaus4, Bengt Simonsson5,

FrederickAppelbaum6, Jane Apperley7, Francisco Cervantes8, Jorge Cortes9, Michael Deininger10,

Alois Gratwohl11, François Guilhot12, Mary Horowitz13, Timothy Hughes14, Hagop Kantarjian9,

Richard Larson15, Dietger Niederwieser16, Richard Silver17, Rudiger Hehlmann4

Authors institutional affiliations

1. M. Baccarani: Department of Hematology/Oncology “L. and A. Seràgnoli”, University of

Bologna, Bologna, Italy

2. G. Saglio: Universityof Turinat Orbassano, Turin, Italy

3. J. Goldman: Hematology Branch, National Heart, Lung & Blood Institute, NIH, Bethesda, MD,

USA

4. A. Hochhaus, R. Hehlmann: Faculty of Clinical Medicine Mannheim, University of Heidelberg,

Mannheim, Germany

5. B. Simonsson: Department of Hematology, UniversityHospital, Uppsala, Sweden

6. F. Appelbaum: FredHutchinsonCancerResearchCenter, Seattle, WA, USA

7. J. Apperley: Department of Hematology, HammersmithHospital, London, UK

8. F. Cervantes: Hematology Department, Hospital Clinic, IDIBAPS, Universityof Barcelona,

Barcelona, Spain

9. J. Cortes, H. Kantarjian: MD AndersonCancerCenter, Houston, TX, USA

10. M. Deininger: Oregon Health & Science University Cancer Institute, Portland, OR, USA

11. A. Gratwohl: Hematology, UniversityHospital, Basel, Switzerland

12. F. Guilhot: Oncology, Hematology and Cell Therapy, Medical Oncology, EA 3805 and Clinical

Research Centre, CHULa Miletrie,Poitiers, France

13. M. Horowitz: Centre for International Blood and Marrow Transplant Research, MedicalCollege

of Wisconsin, Milwaukee, IL, USA

14. T. Hughes: Instituteof Medicaland Veterinary Science, Adelaide, Australia

15. R. Larson: Universityof Chicago, Chicago, IL, USA

16. D. Niederwieser: Department of Hematology and Oncology, Universityof Leipzig, Leipzig,

Germany

Blood First Edition Paper, prepublished online May 18, 2006; DOI 10.1182/blood-2006-02-005686

Copyright © 2006 American Society of Hematology

2

17. R. Silver: New York Presbyterian-Weill Cornell Medical Center, New York, NY, USA

Work supported by the EU, Sixth Framework Programme, Contract No. LSHC-CT-2004-503216

(European LeukemiaNet)

Word count: Abstract 206

Manuscript 5414

Acknowledgement: The scientific contributions of Professor Jörg Hasford and of many members of

the European LeukemiaNet, Work Package 4, are acknowledged.

The scientific and the technical assistance of Simona Soverini, PhD, Alessandra Dorigo, PhD,

Chiara Ferri, and Katia Vecchi is also kindly acknowledged.

Correspondence:

Michele Baccarani, MD

Department of Hematology-Oncology “L. and A. Seràgnoli”

S.Orsola-MalpighiHospital

Via Massarenti 9

40138 BolognaItaly

Tel: xx39 051 390413 Fax xx39 051 398973

e-mail: [email protected]

3

ABSTRACT

The introduction of imatinib mesylate (IM) has revolutionized the treatment of chronic myeloid

leukemia (CML). Although experience is too limited to permit evidence-based evaluation of

survival, the available data fully justify critical reassessment of CML management. The panel

therefore reviewed treatment of CML since 1998. It confirmed the value of IM (400 mg/day) and of

conventional allogeneic hematopoietic stem cell transplantation (alloHSCT). It recommended that

the preferred initial treatment for most patients newly diagnosed in chronic phase should now be IM

400 mg daily. A dose increase of IM, or alloHSCT, or investigational treatments were

recommended in case of failure and could be considered in case of suboptimal response. Failure

was defined at 3 months (no hematologic response (HR)), 6 months (incomplete HR or no

cytogenetic response (CgR)), 12 months (less than partial CgR (Ph+ >35%)), 18 months (less than

complete CgR), and in case of HR or CgR loss, or appearance of IM-highly-resistant BCR/ABL

mutations. Suboptimal response was defined at 3 months (incomplete HR), 6 months (less than

partial CgR), 12 months (less than complete CgR), 18 months (less than major molecular response

(MMolR)), and in case of MMolR loss, other mutations or other chromosome abnormalities. The

importance of regular monitoring at experienced centers was highlighted.

4

INTRODUCTION

After the initial descriptions of chronic myeloid leukemia (CML) more than 150 years ago, little

meaningful progress was made in its treatment for more than a century. Radiation therapy and

busulfan contributed more to improving quality of life than to prolonging survival. Survival

prolongation was first achieved with hydroxyurea (HU), and much more with allogeneic

hematopoietic stem cell transplantation (alloHSCT) and, later, in a minority of patients with

recombinant interferon-alpha (rIFNá)1. Understanding the pathogenesis of the disease began with

the discovery of the Philadelphia(Ph) chromosome followed by appreciation of its molecular

counterpart, the BCR-ABL fusion gene2,3. Recognition of the tyrosine kinase (TK) activity of the

Bcr-Abl proteins led to the discovery of a new series of compounds targeted against BCR-ABL

encoded proteins, which inhibited the TK activity, thus aborting the signals controlling the leukemic

phenotype4. One of the TK inhibitors, imatinib mesylate (IM) was found to have a high and

relatively specific biochemical activity and an acceptable pharmacokinetic and toxicity profile, and

thus, was rapidly introduced into clinical practice5-7. This resulted in a revolutionary step in the

management of CML and by extension a shift in paradigm for the management of cancer in general.

The most recent comprehensive analysis of CML treatment was an evidence-based guideline

developed in 1998 by an expert panel convened by the American Society of Hematology (ASH),

covering conventional chemotherapy, rIFNá, and alloHSCT8. TK inhibitors were not considered at

that time but were subsequently the subjects of editorials and preliminary reviews7,9-14. Although it

is premature at this time to perform an evidence-based analysis of the effects of IM, the

implications and consequences of the introduction of TK inhibitors are so important that it is not too

early to review the available data and to discuss how the treatment of CML could be managed and

further progress could be pursued, based upon expert opinion. Therefore the European

LeukemiaNet appointed a panel of experts to review the current situation. This report constitutes its

opinion.

5

METHODS

Panel composition

The panel included nineteen members with recognized clinical and research expertise in CML, of

whom ten came from the European Union countries (France, Germany, Italy, Spain, Sweden, and

the United Kingdom), one from Switzerland, seven from the United States of America, and one

from Australia.

Scope of the Review

The first step was to perform a comprehensive and critical review of the literature after 1998 (the

date of the last ASH analysis) was performed. A computerized literature search of the MEDLINE

database was conducted in April 2005 and updated in November 2005. Relevant abstracts presented

at the 2004 and 2005 meetings of the ASH, the American Society of Clinical Oncology, the

European Group for Blood and Marrow Transplantation (EBMT), the European Hematology

Association and the International Society for Experimental Hematology were also reviewed.

Thereafter, the panel met several times to discuss definition, evaluation and monitoring of the

responses, as well as treatment policy. It was agreed that discussion and proposals should be limited

to early chronic phase (ECP) patients not only because the treatment of CML patients in a more

advanced phase is less amenable to generalizations, but also to focus on the importance of a firstline

treatment strategy, late therapeutic interventions being generally less effective.

Definitions

The criteria that we have used to distinguish CP from accelerated phase (AP) are those that have

been used in the most recent treatment reports15-19. These criteria are listed in Table 1, together with

WHO criteria, which differs slightly20. The relative risk (RR) of progression and death in early CP

(ECP) patients may be calculated by using either the Sokal24 or the Hasford25 formulations (Table

2).

6

WHO Other, and this report

– Blast cells in blood or bone marrow

(BM) 10-19%

– Blast cells in blood or BM 15-29%

– Blast cells plus promyelocytes in blood or

BM > 30%, with blast cells < 30%

– Basophils in blood 20% – Basophils in blood 20%

– Persistent thrombocytopenia

(<100×109L) unrelated to therapy

– Persistent thrombocytopenia

(<100×109L) unrelated to therapy

– Thrombocytosis (>1000×109L)

unresponsive to therapy

– (not included)

– Increasing spleen size and increasing

WBC count unresponsive to therapy

(not included)

– Cytogenetic evidence of clonal

evolution (the appearance of an additional

genetic abnormalities that was not present at

the time of diagnosis)

(not included)

TABLE 1 List of the criteria that have been proposed by the World Health Organization

(WHO)15 and of the criteria that have been used in most recent studies15-19 and in this review,

for defining accelerated phase (AP). The definition of chronic phase (CP) implies that none of

these criteria is met. For the definition of blast crisis (BC), the WHO-recommended criteria are the

percentage of blast cells in blood or BM (20%), extramedullary blast proliferation or large foci or

clusters of blasts in the bone marrow biopsy20. In recent treatment reports17,21-23, and in this review,

the criteria for BC were limited to the percent of blast cells in PB or BM (30 rather than 20% as

for WHO), or extramedullary blast involvement. It should be noticed that the introduction of new

treatments could change the boundaries between CP, AP and BC, and modify to some extent the

classical subdivision of CML in three phases.

7

SOKAL(24) HASFORD(25)

Age (years) 0.116 (age – 43.4) 0.666 when age 50

Spleen (cm below costal

margin, max distance)

0.0345 (spleen – 7.51) 0.042 x spleen

Platelet count (׳109/L) 0.188 [(Platelets : 700)2 – 0.563] 1.0956 when platelets 1500

Blood myeloblasts (%) 0.0887 (myeloblasts – 2.10) 0.0584 ׳myeloblasts

Blood basophils (%) Non applicable 0.20399 when basophils > 3%

Blood eosinophils (%) Non applicable 0.0413 ׳eosinophils

RELATIVE RISK EXPONENTIAL OF THE TOTAL TOTAL ׳1000

LOW <0.8 780

INTERMEDIATE 0.8-1.2 781-1480

HIGH >1.2 >1480

TABLE 2 Calculation and definition of disease relative risk (RR). Sokal risk was defined

based on patients treated with conventional chemotherapy24. Hasford risk was defined based on

patients treated with rIFNá-based regimens25. We emphasize that calculation of the risk requires

use of clinical and hematologic data at diagnosis, prior to any treatment.

8

SUMMARY AND UPDATE OF RECOMBINANT INTERFERON-ALPHA (rIFNá)

The superiority of rIFNá-based regimens over conventional chemotherapy was reported previously

in the ASH analysis8 and was confirmed in a subsequent study26. A trial of rIFNá vs a combination

of rIFNá and low-dose arabinosyl cytosine (LDAC)27 partially confirmed an earlier study28,

reporting that the cytogenetic response (CgR) rate was higher with the combination but that overall

survival did not differ. A study testing 3 MIU of IFNá three times a week vs 5 MIU/sqm/day

indicated that the low dose was as effective and better tolerated than the high dose29. The last

updates of the major rIFNá studies reported a 9- or 10-year overall survival (OS) ranging from 27%

to 53%30. In one study of 317 patients who had achieved a CCgR, 50% were still in CCgR and 70%

were alive after 10 years, with a significant difference in OS between low and high Sokal risk

patients (10-year OS 90% vs 40%)31. Residual leukemia was detectable at the molecular level in

almost all these patients. Several studies have provided some insights into the biologic and

molecular bases of the therapeutic effects of rIFNá30 but there have been no new or updated clinical

studies.

9

SUMMARY AND UPDATE OF ALLOGENEIC AND AUTOLOGOUS HEMATOPOIETIC

STEM CELL TRANSPLANTATION (HSCT)

The ASH panel reported that about 50% of the patients submitted to alloHSCT in first CP from a

matched related donor remained alive and leukemia-free after 5 years8. Several subsequent reports

confirmed the data and extended the follow up to 10 years, with an OS of 60% and an event-free

survival (EFS) of 50%32,33, and to 15 years, with an OS of 47%34 and 52%35. In a meta-analysis of

three randomized studies of 316 CP patients, 10-year survival estimates were 63% and 65%36. The

Center for International Blood and Marrow Transplant Research (CIBMTR) reported on 4513

patients, with a median age of 35 years, who were transplanted between 1978 and 199737. OS at 18

years was 50% for 3372 first CP patients and 20% for 1141 non-first CP patients. The cumulative

incidence of relapse at 18 years was 25% for CP patients and 37% for the others. Relapses were

seen up to 21 years after treatment. The longest follow-up of patients transplanted from a matched

related donor is that reported by the EBMT, on 2628 patients transplanted between 1980 and

199038. OS at 20 years was 34% for all patients, 41% for patients transplanted in first CP from an

HLA-identical sibling, and 49% for those who had an EBMT risk score (see below) of 0-1. In

children, 10-year OS estimates were reported to be 65-70%39.

An EBMT survey analyzed 3142 patients submitted to conventional alloHSCT in any phase of

CML and from any donor40. This analysis led to the formulation of a prognostic score subsequently

validated by two other analyses41,42. Depending on the risk score, survival ranged from 72% to 11%

in all patients and from 70% to 25% in the patients who were transplanted in ECP (Table 3).

Progress in molecular DNA typing of HLA alleles, in the management of opportunistic infections

and in supportive care, as well as modifications and improvement of conditioning regimes and

immunosuppresive therapy, have contributed to improved results of alloHSCT, using both family

members and unrelated donors43. For CML patients receiving conventional transplants, the use of

peripheral blood stem cells has not been shown to be better than the use of marrow cells44.

10

Reduced intensity conditioning (RIC) is currently being evaluated for CML45-48. The EBMT has

reported on registry data of 187 patients (median age 50 years) who were submitted to RICalloHSCT,

between 1994 and 2002, mainly from matched related donors49. Three-year OS was 70%

for the patients with an EBMT score of 0-2, 50% for the patients with a score of 3-4, and about 30%

for those with a score 5. The use of RIC may permit transplantation also in older patients, but the

long term impact of these and other experimental procedures of alloHSCT on OS, EFS and quality

of life cannot yet be assessed.

The role of treatment intensification with autologous HSCT (autoHSCT) rescue has been the

subject of a number of studies and reviews covering a period of more than 20 years50. Several

observations suggested that the procedure was useful to achieve more remissions and to prolong

survival. Several randomized studies were initiated but none was completed. A meta-analysis of six

such trials in which patients were randomly allocated to receive autoHSCT or a rIFNá-based

regimen did not show an advantage for autoHSCT51.

11

PROGNOSTIC FACTORS RISK SCORE

Age 0 if < 20 years

1 if 20 – 40 years

2 if > 40 years

Interval from diagnosis to HSCT 0 if 1 year

1 if > 1 year

Disease phase 0 if chronic

1 if accelerated

2 if blastic

Donor-recipient sex match 1 if female donor and male recipient

0 if any other match

Donor type 0 if HLA-identical sib

1 if any other

TOTAL RISK SCORE 5-YEAR OVERALL SURVIVAL

EBMT series

CIBMTR

series, all

patients

CIBMTR

series, ECP

patients

0 – 1 72% 69% 70%

2 62% 63% 67%

3 48% 44% 50%

4 40% 26% 29%

5 – 7 22% 11% 25%

TABLE 3 EBMT transplantation risk score. The table lists the prognostic factors and the

corresponding risk score, and reports five-year overall survival rates, as they were calculated in the

original EBMT (European Group for Blood and Marrow Transplantation) report40 and in the

subsequent CIBMTR (Center for International Blood and Marrow Transplant Research) study41. All

EBMT and CIBMTR patients were treated by conventional alloHSCT procedures between 1989

and 1997. Leukemia free survival (calculated only in the EBMT study) at 5 years was 61% for risk

12

score 0-1, 47% for risk score 2, 37% for risk score 3, 35% for risk score 4, and 19% for risk score 5-

7.

13

SUMMARY AND UPDATE OF IMATINIB (IM) DATA

IM vs rIFNá in ECP

The superiority of IM 400 mg daily over rIFNá and LDAC was established in a prospective

randomized international study of 1106 ECP patients (IRIS study). IM was superior to rIFNá for

efficacy, with a complete hematologic response (CHR) rate of 95% vs 55%, a complete CgR

(CCgR) rate of 76% vs 15% and progression free survival (PFS, survival free from progression to

AP/BC) at 19 months of 97% vs 91% (P < 0.001). It was better also for compliance, toxicity, and

quality of life17,52. As expected, molecular response (MolR) rates were also significantly better, with

an estimated major MolR (MMolR) rate at 12 months of 40% vs 2%53. Since many patients who

had been assigned to rIFNá and LDAC were crossed over to IM, it is difficult to meaningfully

compare the long term results of the two treatment arms. However, two independent retrospective

analyses provided independent confirmation that IM was better than any other non-transplant

treatment54,55. Studies have shown that IM is a cost-effective first line therapy compared to rIFNá56.

Follow-up clinical results in ECP

When IM was given at 400 mg daily for initial treatment of ECP patients, the CHR rate after one

year was 95% and the CCgR rate was 76%17. Of those patients who had achieved a CCgR, a

MMolR was achieved in 57% (40% of the patients who had been assigned to IM)53. The proportion

of MMolR patients was reported at 55% of all patients, after two years57. After 54 months followup,

PFS was 93%, OS was 90%, and survival freedom from progression to AP/BC as well as from

hematologic or cytogenetic relapse was 84%58. Currently, this survival outcome is better than for

14

any other reported treatment. The annual rate of progression to AP/BC appeared to be fairly

constant during the first 4 years of treatment, namely 1.5%, 2.8%, 1.6%, and 0.9% 58.

Clinical results in late chronic phase (LCP), accelerated phase (AP) and blast crisis (BC)

Before IM was initially administered as first line treatment for CML, it was given to patients who

were in CP, but resistant or intolerant to rIFNá or who had been treated with conventional

chemotherapy. These patients are classified as “late CP” (LCP). Four international studies reported

a CCgR rate ranging from 41% to 64% with a 5-year PFS of 69% and a 4-year OS of 86-

88%15,18,19,23,59-61. Moreover, one retrospective analysis found that survival of LCP IM-treated

patients was superior to that of historical controls, even when a CCgR was not achieved62.

For AP patients the best results were achieved at a daily dose of 600 mg, with a CHR rate of 37% ,

a CCgR rate of 19% and a 3-year PFS of 40%17,63. In BC the rate of CHR was about 25% and

several responders achieved also a CCgR, but PFS was short, with a median of 10 months or less,

and only 7% remained alive after 3 years5,21-23, 63, 64.

Molecular response (MolR)

Since the frequency of CCgR is very high in IM treated patients, it is necessary to measure the level

of the BCR-ABL transcripts to determine minimal residual disease (MRD) (FIGURE 1). In about

50% of all patients, corresponding to about 70% of the patients who have achieved a CCgR, a

substantial reduction, commonly referred to as a 3-log reduction from a standard baseline or “major

molecular response” (MMolR), was reported in ECP53,65-67, while in LCP the responses were

consistently lower19,20,67,68. The actual frequency with which no residual BCR-ABL transcripts can

be detected by use of the most sensitive available methods, sometimes imprecisely referred to as

“complete” MolR (CMolR), is very variable, and ranges from 4% to 34%18,19,57,67,69. The rate at

15

which the BCR-ABL transcript levels continue to fall reduces with time57,70,71. This is consistent

with the reports that Ph+ stem cells may be less sensitive to IM than later Ph+ progenitors72-75. The

question of whether the inability to detect BCR-ABL transcripts over the long-term is consonant

with “cure” cannot yet be answered. Some case reports suggest that the disease may recur shortly

after IM discontinuation, so that until more information becomes available IM treatment should not

be discontinued without reasons76-80.

Dose issues

The issue of the optimal dose of IM is not yet settled. In early studies for drug registration the

maximum tolerated dose was not identified. A dose of 300 mg daily was sufficient to achieve a

CHR in almost all LCP patients and at 400 mg daily the blood concentration of IM was consistently

higher than that required to inhibit 50% of Bcr-Abl TK activity in vitro81,82. It was also found that a

daily dose of 600 mg was likely to be more effective than 400 mg for AP/BC patients16,21 and that

increasing the IM dose to 600 or 800 mg could benefit a subgroup of patients with inadequate

response or disease progression83. Since at higher concentrations IM may inhibit more effectively

unmutated Bcr-Abl and some mutants, studies were initiated to test higher doses also in CP. In

patients with both prior hematologic and cytogenetic resistance to 400 mg of IM daily, increasing

the IM dose to 800 mg resulted in a CHR in 65% of patients and a CCgR in 18%84. In LCP patients

who had not received prior IM, 66% achieved a CCgR85. In ECP patients a CCgR was achieved in

90% of patients, with 30% CMolR86. In a multicenter Australian study of IM-naive ECP patients

whose dose was escalated from 600 to 800 mg daily, the CCgR rate and the MMolR rate were 81%

and 53%70,87. These studies had no controls and the median follow up was short (6 to 16 months).

Thus, whether increased doses of IM,compared to standard dose of IM, will achieve an increased

overall number of CCgR and MMolR, or whether these effects will merely occur only earlier,

remains to be determined. Answers are expected from prospective studies that are in progress13,88,89.

16

In contrast no studies have yet explored the response to lower IM doses, probably because the 400

mg dose is usually well tolerated and several reports have discouraged the use of low IM doses

because of the possible development of resistance18,19,58,59,66.

Combination with other drugs

Since rIFNá and AC are effective in the treatment of CML and since their mechanisms of action

differ, the combinations of IM with rIFNá and with AC were the first to be tested. In an exploratory

study of 77 patients, the combination of IM 400 mg daily with pegylated rIFNá2b (PegIntron,

Schering Plough), 50 to 150 ìg weekly, was administered66. The compliance to the combination

was limited, since the median tolerated dose of rIFNá was only 35 ìg/week and 50% of patients

discontinued rIFNá before the end of the first year of treatment; after one year the CCgR and the

MMolR rates were 70% and 48%66. The combination of IM 400 mg with LDAC has been

investigated in 30 ECP patients90; at one year the CCgR rate was 70% , with grade 3 and 4

hematologic toxicity in 53% of patients. Prospective randomized studies of IM alone vs IM in

combination with rIFNá, LDAC and high dose AC are ongoing13,89,91.

Several drugs have been shown to overcome IM resistance or to synergize with IM in preclinical

models, including leptomycin B, proteasome inhibitors, mTOR-inhibitors, arsenic trioxide,

mycophenolic acid, farnesyl-transferase inhibitors, bryostatin, decitabine, histone-deacetylase

inhibitors, homoharringtonine, and phosphoinositol-dependent kinase-1 inhibitors92-106, but results

are still preliminary and limited107-110.

17

Relationship with allogeneic hematopoietic stem cell transplantation (alloHSCT)

Treatment with IM prior to alloHSCT was not reported to be associated with an increase of

transplant related morbidity and mortality111-115. IM was also found to control leukemia in patients

relapsing after alloHSCT116,117. In a multicentric retrospective study of 128 patients, the CCgR rates

were 58% in CP, 48% in AP, and 22% in BC, with molecular negativity in 37%, 33%, and 11% of

cases respectively118. In patients treated in early molecular relapse after alloHSCT, molecular

negativity was reinduced in 15/18 cases78. A synergy of IM with donor lymphocyte infusion has

been suggested119.

Factors affecting drug concentration in target cells

Several factors can influence IM concentration in target cells, including intestinal absorption, liver

metabolism through cytochrome P450 isoenzyme-3A4, plasma binding to á1-acid-glycoprotein,

and the transporters involved in multidrug resistance. P-glycoprotein (Pgp) was found to influence

IM intracellular concentration in some studies120-125 but not in others126,127. Interestingly, some

studies have suggested that Pgp inhibition restored IM sensitivity120,124,125 IM does not cross the

blood brain barrier128 Also, the expression of the organic cation transporter hOCT was reported to

influence intracellular drug concentration123.

Resistance and mutations

Resistance may be multifactorial, including BCR-ABL mutations of the kinase domain interfering

with IM binding, BCR-ABL amplification or overexpression, clonal evolution and decreased IM

biovailability or cell exposure120,130-141. Clonal evolution and mutations (Table 4) are likely to be the

most important factors and are related to each other133,142. The frequency of BCR-ABL mutations in

18

resistant patients was reported to range from 42%139 to 90%133 depending on the methodology of

detection, the definition of resistance and the phase of the disease. Mutations are found more

frequently in AP/BC. In CP patients they are rarer and were identified more frequently in patients

with more than 2-fold increase of the BCR-ABL transcript levels than in those with stable or

decreasing levels143. However, mutant Ph+ sub-clones may remain at low levels, may be transient

or unstable and may not be consistently associated with subsequent relapse144,145. In many cases the

mutations have been detected in samples that were collected during IM treatment, but in several

cases the mutation was also traced back to samples collected before treatment, especially in cases of

AP/BC133,146,147. With more sensitive techniques, mutations were also found in some cases of IMnaive

patients and in patients who were in CCgR147-149. It is important to note that Ph+ primitive

cells have been reported to be less sensitive to IM in vitro and in vivo, to harbor BCR-ABL

mutations even prior to IM exposure and to develop rapidly mutations under IM pressure72,74,147,149-

151. Not all mutations have the same biochemical and clinical properties (Table 4). The T315I

mutation and some mutations affecting the so-called P-loop of BCR-ABL confer a greater level of

resistance, whereas the biochemical resistance of other mutations can be overcome by a dose

increase, and some mutations are functionally irrelevant133,137-140,152,153. Thus, the detection of a

kinase domain mutation must be interpreted within the clinical context.

19

imatinib IC50 (nM)

BCR-ABL

Biochemical Cellular

Wild-type 300 260-500

M244V 380 2000

L248V n.a. 1500

G250E 1000 1350-3900

Q252H n.a. 1200-2800

Y253F >5000 3475

Y253H* >5000 >10000

E255K 2800 4400-8400

P-loop

E255V >5000 >5000

D276G n.a. 1500

T277A n.a. n.a.

F311L 775 480

F311I n.a. n.a.

T315I* >5000 >10000

F317L* 900 810-1500

M343T n.a. n.a.

M351T 820 930

M351V n.a. n.a.

E355D n.a. n.a.

E355G n.a. 400

Catalytic

domain

F359V* 4700 1200

V379I 800 1630

A380T* 340 2450

F382L n.a. n.a.

L387M 1500 1000

L387F n.a. 1100

H396P 340-800 850-4200

Activation loop

H396R 1950 1750

S417Y n.a. n.a.

E459K n.a. n.a.

F486S 1230 2800

Table 4 IC50 values of BCR-ABL mutations observed in patients resistant to IM154. Shaded

boxes highlight residues belonging to the P-loop, catalytic domain and activation loop, as indicated.

Residues marked with an asterisk (*) represent IM contact sites. Other mutations which have not yet

been detected in patients were recovered from in vitro saturation mutagenesis screenings for

mutations conferring resistance to IM or other TK inhibitors. They include: M237I, G250A,

G250V, E255D, A269V, E281K, E282D, K285N, V289S, V299L, T315A, F317C, V338G, Q346H,

S348L, M451L, E352K, E355A, A366D, G398R, G463D, M472I, E494A, with a cellular IC50

20

<1460 nM (that is the mean trough plasma level of IM in patients treated with 400 mg daily), and

E255R, E275K, M278L, E279K, E281K, E292Q, Q300H, F311V, T315S, E316D, G321W,

D325N, A380S, L384M, M388L, E450K, E499K, with a cellular IC50 >1460 nM . IC50 is the

concentration that inhibits by 50% the biochemical TK activity of BCR/ABL and suppresses by

50% the growth of Ph+ cell lines. n.a., not available.

21

Additional chromosome abnormalities (ACA) in Ph+ cells (clonal evolution) and other

chromosome abnormalities (OCA) in Ph- cells

Within the Ph+ clone additional chromosome abnormalities (ACA) can be found in a variable

proportion of metaphases and in a variable number of patients. This phenomenon, also known and

described as clonal evolution, is rare in ECP and becomes more frequent over time and with disease

progression23,134,155-160. A negative relationship of ACA with IM response has been shown,

including a lower CgR rate157, a higher hematologic relapse rate (50% vs 9%)155 and a shorter OS

(75% vs 90% at 2 years)156. Chromosome 9q+ deletions (del9q+) were reported to be associated

with less CHR, less CgR and a shorter PFS in LCP, AP and BC patients in one study161 but not in

another162.

Other chromosome abnormalities (OCA) have been reported in the Ph- cells of about 5% of the

patients who had achieved a CCgR with IM166-170. Many of these patients were in LCP and had been

pretreated with rIFNá based regimens. OCA included trisomy 8 alone in about 50% of such cases,

trisomy 8 with other abnormalities in about 10% of cases, a deletion of chromosome 7 alone or with

other abnormalities in about 15% of cases, and other abnormalities in the remaining cases. The

balance between the Ph+ clone and the Ph- clone with OCA fluctuated depending on IM treatment,

which suppressed Ph+ cells and allowed the Ph- clone with OCA to expand. In some cases Phclone

with ACA were reported to be associated with a myelodysplastic syndrome, mainly in

patients with a deletion of chromosome 7 and/or other complex abnormalities but also in patients

with isolated trisomy 8. It was also reported that many patients remained in complete cytogenetic

and hematologic response after the detection of OCA and that OCA may be transient166,167,169,170 but

the follow-up is still short.

22

Prognostic factors

Two sets of prognostic factors can be considered, namely those that can be identified prior to

treatment (baseline factors) and those that can be identified during the treatment (response-related

factors). The main baseline factors are the phase of disease and the relative risk (RR). Although

different definitions of AP and BC have been used (Table 1), the phase of the disease influences

strongly the response, the duration of the response and OS, with better results in CP than in AP and

in AP than in BC. The RR, either by Sokal’s24 or Hasford’s methods25, predicts the cytogenetic

response to IM 400 mg daily (Table 5) 53,171,172. Moreover, Sokal’s RR has been reported to predict

also MolR and OS. In the IRIS study, the rate of 12-month MMolR among CCgRs was 66%, 45%

and 38% in low, intermediate and high risk patients respectively (P = 0.007)53. The OS at 54

months was 94%, 88% and 81% for low, intermediate and high Sokal risk patients (P <0.001)58.

These risk definitions which were derived from patients treated with conventional chemotherapy or

IFNá, are still useful and should be used until further studies identify and confirm other factors of

possible prognostic relevance, such as genomic profile173-177, genetic polymorphisms178,179, Wilms

tumor gene expression180, total phosphotyrosine levels in CD34+ cells181 and the phosphorylation

level of the adaptor protein Crkl182. In addition, it has been reported that BCR-ABL expression

levels affect the CgR to IM19 and determine the rate of development of resistance to IM141. ACA,

including Ph duplication, and del9q+, are also candidate adverse prognostic factors.

As data from IRIS study are continuously updated58,172,183, early cytogenetic response seems to be

the most important response-related prognostic factor (Table 6). If no CgR is achieved after 3

months, there is still a 50% chance of achieving a CCgR later on. If there is any (even minimal)

CgR after 6 months of treatment, there is still a fair chance of achieving a CCgR later on, but if the

6-month karyotype remains more than 95% Ph+, the probability is only 15%. After 12 months of

treatment, if the CgR is partial the probability of achieving a CCgR at 2 years is still 50%, but if the

23

response is less than partial, this probability becomes less than 20%. The data reported in Table 6

also highlight the relationship between early CCgR and EFS.

The level of MolR was also found to be an important dynamic factor of prognosis. It was reported

that transcript levels after 1 or 2 months of treatment predicted late responses184,185, that a low level

of residual disease was associated with continuous remission68, and that a MMolR after 12 months

of treatment was associated with a better EFS and PFS53,58. A rise of BCR/ABL transcript level has

been consistently associated with mutations or response loss143,186.

24

Complete cytogenetic response

Relative Risk Low Interm High

Italian multicenter study, 77 patients, IM 400 mg, response at

6 months171 70% 41% 8%

International multicenter IRIS study, 383 patients, IM 400 mg

– response at 12 months53 76% 67% 49%

– response at 42 months172 91% 84% 69%

Single-center study, 187 patients, IM 400-800

mg, overall response54

84% 85% 69%

TABLE 5 Cytogenetic response by relative risk. Two independent studies of newly diagnosed,

ECP patients who were treated initially with IM 400 mg daily have shown that the cytogenetic and

the molecular response to that dose of IM was significantly related to Sokal’s risk. In one study171

the relationship was found also using Hasford’s risk. In another study54 the differences were not

significant, but IM dose was higher, 800 mg in 100 patients, 600 mg in 14 patients, and 400 mg in

73 patients. The last update of the IRIS study58 reported OS was also risk related, being 94% for

low risk patients, 88% for intermediate risk patients and 81% for high risk patients (P < 0.001),

after 54 months of therapy.

25

Months on

treatment

Cytogenetic

response

Probability of CCgR

at 2 years

EFS

at 42 months

Partial 90% NA

3 Minor 60% NA

Minimal/None 50% NA

Complete NA

Partial 80%

95%

Minor or Minimal 50%

6

None 15%

75%

Complete NA

12 Partial 50%

90%

Minor/Minimal/None <20% 65%

TABLE 6 – Relationship between the degree of early CgR, the CCgR rate at 2 years, and EFS

at 42 months in IRIS study172,183. From the same study it was reported that after 54 months,

survival free from progression to AP/BC was 97% for the patients with a CCgR at 12 months, 95%

for those with a PCgR and 81% for those who at 12 months had achieved less than a PCgR58. NA =

Not applicable or not available.

26

DEFINING AND MONITORING THE RESPONSE

Hematologic Response (HR) and Cytogenetic Response (CgR)

In almost all recent reports on the treatment of CML, HR and CgR were defined virtually the same

way, and with only minor differences15,17-19,26-29,66. We propose to use the definitions that are listed

in Table 7. We recommend that HR be evaluated every 2 weeks until a CHR has been achieved and

confirmed, and a conventional cytogenetic examination of marrow cells be performed before

treatment, at least every 6 months until a CCgR has been achieved and confirmed, then every 12

months. Once a MMolR has been achieved and confirmed, conventional cytogenetic examination of

marrow cells may be performed less frequently, depending on clinical, hematologic and molecular

findings.

Fluorescence-In-Situ-Hybridization (FISH) on interphase cells has the potential advantage of

evaluating many more cells and of using peripheral blood instead of marrow187,188, but since the

data obtained so far are all based on conventional cytogenetics, we recommend using FISH only

before treatment, to identify cases of Ph-, BCR-ABL+ CML, and those with variant translocations,

Ph amplification or del9q+.

Molecular response (MolR)

The necessity for a quantitative definition of MolR has developed with the introduction of IM,

because with IM most patients achieve a CCgR, so that molecular methods for measuring minimal

residual disease (MRD) are required (Figure 1). The IRIS trial provided evidence for the first time

that a reduction of BCR-ABL transcripts by 3 or more logs below a standard baseline value

correlated with PFS53. The use of the “log reduction” terminology has led to some degree of

27

confusion since it seems to imply that the value is a relative one. For this reason, at a consensus

conference held in Bethesdaunder the auspices of the NIH, it was proposed to move away from the

term “log reduction” and to introduce a standardized numerical International Scale (IS) expressing

the amount of BCR-ABL as a percentage of a control gene and anchored to two “absolute” values

based on validated reference materials (plasmids, lyophilised cells or cell extracts) of known

value189. The first value will be designated 100% on the proposed IS and the second value will

represent a 3-log reduction, i.e. 0.1%. A given laboratory will use the validated reference material to

determine the local value that is equivalent to MMolR as determined in the IRIS trial. By

comparing the value for a 3-log reduction with the value on the internationally agreed scale, each

laboratory can derive a conversion factor which can then be used to express the results in any given

patient on the IS.

In ECP patients, evaluating MRD with real-time quantitative polymerase chain reaction (RQ-PCR)

does not require bone marrow cells. Blood is drawn, e.g. 10 ml, which contains a sufficient amount

of leukocytes for RNA extraction from the whole buffy coat. We propose RQ-PCR on peripheral

blood cells be performed at regular intervals of 3 months, even after RQ-PCR becomes negative.

Assessing the molecular status of a patient is not limited to the evaluation of the level of the BCRABL

transcripts. We propose performing a mutational analysis immediately in any case of

treatment failure or suboptimal response, including a confirmed rise of BCR-ABL transcript level.

We recognize, however, that there is currently no consensus regarding the degree of increase which

should cause concern189 and that there is at present only a limited number of laboratories worldwide

currently performing these analyses.

28

HEMATOLOGIC RESPONSE (HR) CYTOGENETIC RESPONSE (CgR)

MOLECULAR RESPONSE (MolR)

(BCR/ABL to control gene ratio according to

the international scale)

Complete – Platelet < 450×109L

– WBCC < 10 x109L

– Differential without immature

granulocytes and with less than 5%

basophils

– Non palpable spleen

Complete Ph+ 0

Partial Ph+ 1-35%

Minor Ph+ 36-65%

Minimal Ph+ 66-95%

None Ph+ > 95%

“Complete” = transcript non quantifiable

and non detectable

Major 0.10

Check every 2 weeks until a complete response

has been achieved and confirmed, then every 3

months unless otherwise required

Check at least every 6 months until a complete

response has been achieved and confirmed,

hence at least every 12 months

Check every 3 months

Mutational analysis in case of failure,

suboptimal response, or transcript level increase

TABLE 7 Response definition and monitoring. Complete HR (CHR), complete CgR (CCgR) and major MolR (MmolR) should be confirmed in

two subsequent occasions. CgR is evaluated by morphologic cytogenetics of at least 20 marrow metaphases. FISH of peripheral blood cells should

be used only if marrow cells cannot be obtained. MolR is assessed on peripheral blood cells. The international scale for measuring MolR is that

proposed by Hughes et al189.

29

Failure and suboptimal response

The goals of treatment, in order of time and importance, are CHR, CCgR, MMolR, and “complete”

molecular response. Although the time to response may not always affect the prognosis, it is

operationally useful to define at which time point a response may be satisfactory, thus encouraging

continuation of current treatment, or if it is not satisfactory, thus requiring or suggesting a change in

the therapeutic strategy. Based on the available information, as summarized in prior sections, we

propose to define the response to the treatment at different time points as “failure” and

“suboptimal”. In this context “failure” means that continuing IM treatment at the current dose is no

longer appropriate for these patients, who would likely benefit more from other treatments.

“Suboptimal response” means that the patient may still have a substantial benefit from continuing

IM, but that the long-term outcome of the treatment would not likely be as favorable. Moreover, we

propose that some factors should “warn” that standard dose IM treatment may not be the best

choice and that patients with these factors requires a more careful monitoring. The proposed criteria

for failure, suboptimal response and warning are listed in Table 8.

30

TIME FAILURE SUBOPTIMAL RESPONSE WARNINGS

Diagnosis NA NA – High risk

– Del9q+

– Additional chromosome abmormalities

(ACA) in Ph+ cells

3 months – No hematologic response (HR) (stable

disease or disease progression

– Less than Complete HR (CHR)

6 months – Less than Complete HR (CHR)

– No cytogenetic response (CgR) (Ph+

more than 95%)

– Less than Partial CgR (PCgR) (Ph+

more than 35%)

12 months – Less than PCgR

(Ph+ more than 35%)

– Less than Complete CgR (CCgR)

– Less than major MolR (MMolR)

18 months

– Less than CCgR – Less than MMolR

Any time – Loss of CHR (1)

– Loss of CCgR (2)

– Mutation (3)

– ACA in Ph+ cells (4)

– Loss of MMolR (4)

– Mutation (5)

– Any rise in transcript level

– Other chromosome abnormalities in Phcells

(1) To be confirmed on two occasions unless associated with progression to AP/BC (2)

(2) To be confirmed on two occasions, unless associated with CHR loss or progression to AP/BC

(3) High level of insensitivity to IM

(4) To be confirmed on two occasions, unless associated with CHR or CCgR loss

(5) Low level of insensitivity to IM

31

TABLE 8 Operational definition of failure and suboptimal response for previously untreated, ECP, CML patients who are treated with

IM 400 mg daily. Failure implies that the patient should be moved to other treatments whenever available. Suboptimal response implies that the

patient may still have a substantial benefit from continuing IM treatment, but that the long term outcome is not likely to be optimal, so that the

patient becomes eligible for other treatments. Warnings imply that the patient should be monitored very carefully and may become eligible for

other treatments. The same definitions can be used to define the response after IM dose escalation. For risk definitions refer to Table 2. For

mutations refer to Table 4. For the definition of HR, CgR and MolR, refer to Table 7. Abbreviations: ACA = Additional Chromosome

Abnormalities; HR = Hematologic Response; CHR = Complete Hematologic Response; CgR = Cytogenetic Response; PCgR = Partial Cytogenetic

Response; CCgR = Complete Cytogenetic Response; MMolR = Major Molecular Response; NA = Not Applicable.

 

33

TREATMENT POLICY

Standard (non-investigational) treatment of ECP Ph+ CML includes HU, rIFNá±LDAC, IM 400

mg daily and alloHSCT. The superiority of IFNá±LDAC over HU was already demonstrated and

confirmed8,30. The superiority of IM 400 mg over IFNá±LDAC has also been demonstrated17,53.

Standard alloHSCT is a recognized therapeutic procedure achieving long lasting molecular

remissions or cures in about 50% of the patients who are eligible for the procedure, with substantial

differences among recognized risk groups40,41. In countries where IM is available and standard

alloHSCT is feasible we are now in a rather privileged situation to have two potent strategies which

are both established but are neither perfect nor mutually exclusive. IM is preferred as initial

treatment. In a patient with a high disease risk and a low EBMT risk score the choice between IM

and alloHSCT should be discussed, but there is little reason to deny such a patient a trial with IM

since the early response to IM can either reinforce or weaken the indication for alloHSCT.

The motivations for treatments other than IM are intolerance or excess toxicity, failure, suboptimal

response, and “warnings”.

In case of intolerance or excess toxicity, the choices are either alloHSCT or rIFNá±LDAC, which

must be weighed against investigational trials of new agents and should follow the principle of

shared decision-making wherein the patient is explained the risks and rewards of each treatment

decision.

In case of failure (Table 8 ) we propose that the first choice be alloHSCT, or dose-escalation of IM

to 600 or 800 mg daily, provided that the patient tolerated 400 mg and that resistance to IM was not

associated with a BCR-ABL mutation with a high level of insensitivity to IM.

In case of suboptimal response (Table 8) we propose that the first choice be dose-escalation of IM

to 600 or 800 mg daily, provided that the patient tolerated 400 mg. AlloHSCT could be offered to

patients with a low or intermediate EBMT risk score and high RR or other warning features.

34

In patients presenting with “warning” features, standard treatment is still IM 400 mg, but any

“warning” (Table 8) should alert that the patient may become eligible for IM dose escalation, for

alloHSCT, or in selected cases for investigational agents.

There are several other possible scenarios. The first is the patient in whom other treatment options

are not available; in such case the choice would be between continuing IM treatment, if a CHR is

maintained, or to resort to HU. The second scenario is the patient requiring IM dose reduction or

frequent treatment discontinuations. We recommend that the treating physician advise the patient to

adhere to the 400 mg dose insofar as possible; appropriate supportive care should be provided,

including myeloid growth factors and erythropoietin; the response should be monitored frequently.

Monitoring of blood IM concentration is not required, but it would be desirable in case of failure, in

patients who must take drugs interfering with cytochrome P450, and in those who experience a

severe drug-related adverse event.

The proposals and recommendations discussed in this paper focus on ECP patients but sometimes

patients are first diagnosed when initially in AP or BC. There are few data pertaining to treatment

results in these patients. We propose patients in early BC to be treated initially with IM or other TK

inhibitors (based on mutational analysis) and then to proceed to alloHSCT. Since some temporal

latitude exists after the diagnosis of AP, a more prolonged trial with IM is possible.

35

CONCLUSIONS

Progress in drug development, in molecular and cellular biology and in HSCT obliges the medical

community to maintain a critical attitude to the management of Ph+ CML. On the one hand it must

be recognized that the introduction of IM has marked an important and hopefully revolutionary

step, but the long term outcome of this treatment cannot yet be assessed. On the other hand

alloHSCT holds the promise of cure, but with definite toxicity and mortality. At the same time other

TK inhibitors and targeted agents are already in preclinical and clinical evaluation13,154,190-194. The

proposals described in this report have been generated by a panel of experts to strike a balance

between the magic freedom of research in progress and the practice of advising patients and

managing treatment. The proposals concerning treatment policy may be provisional, in the absence

of the evidence that will be provided only by longer follow-up of prospective studies; however, the

recommendations concerning the methods that must be used to evaluate and to monitor the response

are nonetheless cogent. Cytogenetic and molecular monitoring, including mutational analysis, is

expensive and requires appropriate resources and sophisticated facilities. However the cost of

monitoring is negligible by comparison with the cost of treatment, whether it is a targeted agent or

HSCT. Moreover, careful monitoring is required to ensure that an individual patient receives the

proper treatment and to decide if and when a therapy should be changed. Finally, it should be

realized that progress makes treatment more effective but not necessarily easier. Thus the treatment

of Ph+ CML should be provided under the guidance of an experienced center, offering and asking

patients to be registered on investigational studies. This is necessary to ensure that all the data,

clinical and biological, that are urgently required to answer the present questions, are collected and

analysed in an accurate and timely manner, for the benefit of the subsequent patients and for further

progress in the treatment of leukemia.

36

Diagnosis, Pretreament

or Hematologic Relapse

Complete Hematologic Response

Undetectable transcript

(Complete Molecular Response)

Complete Cytogenetic Response

Major Molecular Response

100

10

1

0.1

0.01

0.001

0.0001

BCR-ABL ratio

(according to the International Scale)

1012

1011

1010

109

108

107

106

Number of leukemic cells

37

Figure 1 – Approximate relationship between response, the putative number of leukemic cells

and the level of BCR-ABL transcripts. When a complete cytogenetic response has been achieved,

the (putative) number of residual Ph+ cells can be measured only with quantitative molecular

methods. The figure highlights the importance of molecular methods in the evaluation of the

response to treatment. However, the sensitivity of current methods may vary substantially and in

any case no method can detect the transcript at very low cellular levels. For this reason the term

“complete molecular response” may be misleading, since it might erroneously be interpreted as an

equivalent of complete disease eradication and cure. The term “undetectable BCR-ABL” may better

describe the biological situation.

38

REFERENCES

1. Goldman J. Management of chronic myeloid leukemia. Semin Hematol 2003; 40: 1-103.

2. Sawyers CL. Chronic myeloid leukemia. N Engl J Med. 1999; 340: 1330-1340.

3. Holyoake TL. Recent advances in the molecular and cellular biology of chronic myeloid

leukaemia: lessons to be learned from the laboratory . Br J Haematol. 2001; 113 :11-23.

4. 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; 2: 561-566.

5. Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL

tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001; 344: 1031-1037.

6. 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 Philadelphiachromosome. N Engl J Med. 2001; 344: 1038-1042.

7. Savage DG, Antman KH. Imatinib mesylate–a new oral targeted therapy. N Engl J Med. 2002;

346: 683-693.

8. Silver RT, Woolf SH, Hehlmann R, et al. An Evidence-Based Analysis of the Effect of

Busulfan, Hydroxyurea, Interferon, and Allogeneic Bone Marrow Transplantation in Treating the

Chronic Phase of Chronic Myeloid Leukemia: Developed for the American Society of Hematology.

Blood 1999; 94: 1517-1536.

9. Peggs K, Mackinnon S. Imatinib mesylate–the new gold standard for treatment of chronic

myeloid leukemia. N Engl J Med. 2003;348:1048-1050.

10. Bories D, Devergie A, Gardembas M, et al. Stratégies thérapeutiques et recommandations pour

la prise en charge des patients atteints de leucémie myéloïde chronique. Hématologie 2003; 9: 497-

512.

11. Goldman J, Mahon F, Reiffers J. Imatinib for chronic myeloid leukemia. Semin Hematol. 2003;

40(Suppl 2): 1-113.

12. StoneRM.Optimizing Treatment of Chronic Myeloid Leukemia: A Rational Approach.

Oncologist 2004; 9: 259-270.

13. Hehlmann R, Berger U, Hochhaus A. Chronic myeloid leukemia: a model for oncology. Ann

Hematol. 2005; 84: 487-497.

14. Deininger M, Buchdunger E, Druker BJ. The development of imatinib as a therapeutic agent for

chronic myeloid leukemia. Blood. 2005;105:2640-2653.

15. Kantarjian H, Sawyers C, Hochhaus A, et al. Hematologic and cytogenetic responses to imatinib

mesylate in chronic myelogenous leukemia. N Engl J Med. 2002; 346: 645-652.

39

16. 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; 99: 1928-1937.

17. 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; 348:

994-1004.

18. Kantarjian H, Cortes J, O’Brien S, et al. Long-term survival benefit and improved complete

cytogenetic and molecular response rates with imatinib mesylate in Philadelphiachromosomepositive

chronic-phase chronic myeloid leukemia after failure of interferon-alpha. Blood. 2004;

104: 1979-1988.

19. Rosti G., Martinelli G, Bassi S, et al. Molecular response to imatinib in late chronic phase

chronic myeloid leukemia. Blood. 2004; 103: 2284-2290.

20. Jaffe ES, Harris NL, Stein H, et al. World Health Organization Classification of Tumours.

Pathology and Genetics of Tumours of Hematopoietic and Lymphoid Tissues. IARC Press: Lyon

2001.

21. 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. 2002; 99: 3530-3539.

22. Sureda A, Carrasco M, de Miguel M, et al. Imatinib mesylate as treatment for blastic

transformation of Philadelphiachromosome positive chronic myelogenous leukemia.

Haematologica. 2003; 88: 1213-1220.

23. Lahaye T, Riehm B, Berger U, et al. Response and resistance in 300 patients with BCR-ABLpositive

leukemias treated with imatinib in a single center: a 4.5-year follow-up. Cancer. 2005; 103:

1659-1669.

24. Sokal JE, Cox EB, Baccarani M, et al. Prognostic discrimination in good-risk chronic

granulocytic leukemia. Blood 1984; 63: 789-799.

25. Hasford J, Pfirrmann M, Hehlmann R, et al. A new prognostic score for survival of patients with

chronic myeloid leukemia treated with interferon alfa.

J Natl Cancer Inst. 1998; 90: 850-858.

26. Hehlmann R, Berger U, Pfirrmann M, et al. Randomized comparison of interferon alpha and

hydroxyurea with hydroxyurea monotherapy in chronic myeloid leukemia (CML-study II):

prolongation of survival by the combination of interferon alpha and hydroxyurea. Leukemia. 2003;

17: 1529-1537.

27. Baccarani M, Rosti G, de Vivo A, et al. A randomized study of interferon-alpha versus

interferon-alpha and low-dose arabinosyl cytosine in chronic myeloid leukemia. Blood. 2002; 99:

1527-1535.

28. Guilhot F, Chastang C, Michallet M, et al. Interferon alfa-2b combined with cytarabine versus

interferon alone in chronic myelogenous leukemia. French Chronic Myeloid Leukemia Study

Group. N Engl J Med. 1997; 337: 223-229.

40

29. Kluin-Nelemans H, Buck G, le Cessie S, et al. Randomized comparison of low-dose versus

high-dose interferon-alfa in chronic myeloid leukemia: prospective collaboration of 3 joint trials by

the MRC and HOVON groups. Blood. 2004; 103: 4408-4415.

30. Baccarani M, Russo D, Rosti G, et al. Interferon-alfa for chronic myeloid leukemia. Semin

Hematol. 2003; 40: 22-33.

31. Bonifazi F, de Vivo A, Rosti G, et al. Chronic myeloid leukemia and interferon-alpha: a study

of complete cytogenetic responders. Blood. 2001; 98: 3074-81.

32. ICSG on CML and GITMO. Monitoring treatment and survival in chronic myeloid leukemia. J

Clin Oncol. 1999; 17: 1858-1868.

33. Simonsson B, Öberg G, Björeman M, et al. Intensive treatment and stem cell transplantation in

chronic myelogenous leukemia: long-term follow-up. Acta Haemat. 2005; 113: 155-162.

34. Gratwohl A, Brand R, Apperley J, et al. Graft-versus-host disease and outcome in HLAidentical

sibling transplantations for chronic myeloid leukemia. Blood. 2002; 100: 3877-3886.

35. Robin M, Guardiola P, Devergie A, et al. A 10-year median follow-up study after allogeneic

stem cell transplantation for chronic myeloid leukemia in chronic phase from HLA-identical sibling

donors. Leukemia. 2005; 19: 1613-1620.

36. Socié G, Clift RA, Blaise D, et al. Busulfan plus cyclophosphamide compared with total-body

irradiation plus cyclophosphamide before marrow transplantation for myeloid leukemia: long-term

follow-up of 4 randomized studies. Blood. 2001; 98: 3569-3574.

37. Goldman JM, Rizzo JD, Jabocinski KA, et al. Long term outcome after allogenic stem cell

transplantation for CML. Hematol J. 2004; 5 (Suppl 2): 98, abstract 266.

38. 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.

Haematologica. 2006, in press.

39. Cwynarski K, Roberts IA, Iacobelli S, et al. Stem cell transplantation for chronic myeloid

leukemia in children. Blood. 2003; 102(4): 1224-1231.

40. 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. 1998; 352: 1087-1092.

41. Passweg JR, Walker I, Sobocinski KA, et al. Validation and extension of the EBMT Risk Score

for patients with chronic myeloid leukaemia receiving allogeneic haematopoietic stem cell

transplants. Br J Haematol. 2004;125: 613-620.

42. De Souza CA, Vigorito AC, Ruiz MA, et al. Validation of the EBMT risk score in chronic

myeloid leukemia in Braziland allogeneic transplant outcome. Haematologica. 2005; 90: 232-237.

43. Barrett J. Allogenic stem cell transplantation for chronic myeloid leukemia. Semin Hematol.

2003; 40: 59-71.

41

44. Oehler VG, Radich JP, Storer B, et al. Randomized trial of allogeneic related bone marrow

transplantation versus peripheral blood stem cell transplantation for chronic myeloid leukemia. Biol

Blood Marrow Transplant. 2005; 11: 85-92.

45. Bornhäuser M, Kiehl M, Siegert W, et al. Dose-reduced conditioning for allografting in 44

patients with chronic myeloid leukaemia: a retrospective analysis. Br J Haematol. 2001; 115: 119-

124.

46. 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 2003; 101: 441-445.

47. Weisser M, Schleuning M, Ledderose G, et al. Reduced-intensity conditioning using TBI (8

Gy), fludarabine, cyclophosphamide and ATG in elderly CML patients provides excellent results

especially when performed in the early course of the disease. Bone Marrow Transplant. 2004; 34:

1083-1038.

48. Baron F, Maris MB, Storer BE, et al. HLA-matched unrelated donor hematopoietic cell

transplantation after nonmyeloablative conditioning for patients with chronic myeloid leukemia.

Biol Blood Marrow Transplant. 2005; 11: 272-279.

49. 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. 2005; 106: 2969-2976.

50. Carella AM, Beltrami G, Corsetti MT. Autografting in chronic myeloid leukemia. Semin

Hematol. 2003; 40: 72-78.

51. Richards SM, Apperley J, Carella A, et al. Autografting in chronic myeloid leukaemia: a metaanalysis

of six randomized trials. Haematologica 2005; 90 (suppl 2): 152-153, abstract 0385.

52. Hahn EA, Glendenning GA, Sorensen MV, et al. Quality of life in patients with newly

diagnosed chronic phase chronic myeloid leukemia on imatinib versus interferon alfa plus low-dose

cytarabine: results from the IRIS Study. J Clin Oncol. 2003; 21: 2138-2146.

53. 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;

349: 1421-1432.

54. Kantarjian H, O’Brien S, Cortes J, et al. Imatinib mesylate therapy improves survival in patients

with newly diagnosed Philadelphiachromosome-positive chronic myelogenous leukemia in the

chronic phase: comparison with historic data. Cancer. 2003; 98: 2636-42.

55. Guilhot F, Roy L,Guilhot J, et al. Retrospective comparison of Imatinib versus Interferon plus

Cytarabine for chronic myelogenous leukemia patients in chronic phase. Blood. 2005; 106: 52a,

abstract 165.

56. Reed SD, Anstrom KJ, Ludmer JA, et al. Cost-effectiveness of imatinib versus interferon-alpha

plus low-dose cytarabine for patients with newly diagnosed chronic-phase chronic myeloid

leukemia. Cancer. 2004; 101: 2574-2583.

42

57. Hughes T, Radich J, Kiese B, et al. Long term significance of achieving a major molecular

response for first and second line Imatinib treated chronic phase patients with CML entered in the

IRIS study. Haematologica 2005; 90 (suppl 2): 48, abstract 0118.

58. Simonsson B, on behalf of the IRIS study group. Beneficial effects of cytogenetic and molecular

response on long term outcome in patients with newly diagnosed chronic myeloid leukemia in

chronic phase (CML-CP) treated with Imatinib (IM): update from the IRIS study. Blood. 2005; 106:

52a, abstract 166.

59. Kantarjian H, Talpaz M, O’Brien S, et al. Imatinib mesylate for Philadelphia chromosomepositive,

chronic-phase myeloid leukemia after failure of interferon-alpha: follow-up results. Clin

Cancer Res. 2002; 8: 2177-2187.

60. Cervantes F, Hernández-Boluda JC, Steegmann JL, et al. Imatinib mesylate therapy of chronic

phase chronic myeloid leukemia resistant or intolerant to interferon: results and prognostic factors

for response and progression-free survival in 150 patients. Haematologica. 2003; 88: 1117-1122.

61. Gambacorti C, Talpaz M, Sawyers C, et al. Five year follow-up results of a phase II trial in

patients with late chronic phase chronic myeloid leukemia treated with Imatinib who are

refractory/intolerant of Interferon-á. Blood. 2005; 106: 317a, abstract 1089.

62. Kantarjian H, O’Brien S, Cortes J, et al. Survival advantage with imatinib mesylate therapy in

chronic-phase chronic myelogenous leukemia (CML-CP) after IFN-alpha failure and in late CMLCP,

comparison with historical controls. Clin Cancer Res. 2004; 10: 68-75.

63. Silver RT, Talpaz M, Sawyers CL, et al. Four years of follow-up of 1027 patients with late

chronic phase, accelerated phase, or blast crisis chronic myeloid leukemia treated with Imatinib in

three large phase II trials. Blood. 2004; 104: 11a, abstract 23.

64. Kantarjian H, Cortes J, O’Brien S, et al. Imatinib mesylate (STI571) therapy for Philadelphia

chromosome-positive chronic myelogenous leukemia in blast phase. Blood. 2002; 99: 3547-3553.

65. Müller MC, Gattermann N, Lahaye T, et al. Dynamics of BCR-ABL mRNA expression in firstline

therapy of chronic myelogenous leukemia patients with imatinib or interferon alpha/ara-C.

Leukemia. 2003; 17: 2392-2400

66. Baccarani M, Martinelli G, Rosti G, et al. Imatinib and pegylated human recombinant

interferon-alpha2b in early chronic-phase chronic myeloid leukemia. Blood. 2004; 104: 4245-4251.

67. 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. 2005; 11: 3425-3432.

68. Paschka P, Müller MC, Merx K, et al. Molecular monitoring of response to imatinib (Glivec) in

CML patients pretreated with interferon alpha. Low levels of residual disease are associated with

continuous remission. Leukemia. 2003; 17: 1687-1694.

69. Mueller MC, Paschka P, Ernst T, et al. Long term surveillance of CML patients on Imatinib

therapy. Follow-up of German patients treated within the IRIS trial. Haematologica 2005; 90 (suppl

2): 153, abstract 0387.

43

70. Hughes TP, Branford S, Reynolds J, et al. Maintenance of Imatinib dose intensity in the first six

months of therapy for newly diagnosed patients with CML is predictive of molecular response,

independent of the ability to increase dose at a later point. Blood. 2005; 106: 51a, abstract 164.

71. Goldman JM, Hughes T, Radich J, et al. Continuing reduction in level of residual disease after 4

years in patients with CML in chronic phase responding to first line Imatinib in the IRIS study.

Blood. 2005; 106: 51a, abstract 163.

72. 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.

73. Bhatia R, Holtz M, Niu N, et al. Persistence of malignant hematopoietic progenitors in chronic

myelogenous leukemia patients in complete cytogenetic remission following imatinib mesylate

treatment. Blood. 2003; 101: 4701-4707.

74. Elrick LJ, Jorgensen HG, Mountford JC, et al. Punish the parent not the progeny. Blood. 2005;

105: 1862-1866.

75. Michor F, Hughes TP, Iwasa Y, et al. Dynamics of chronic myeloid leukaemia. Nature. 2005;

435: 1267-1270.

76. Cortes J, O’Brien S, Kantarjian H. Discontinuation of imatinib therapy after achieving a

molecular response. Blood. 2004; 104: 2204-2205.

77. Merante S, Orlandi E, Bernasconi P, et al. Outcome of four patients with chronic myeloid

leukemia after imatinib mesylate discontinuation. Haematologica. 2005; 90: 979-981.

78. 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.

2005; 23: 7583-7593.

79. Daneschnejad S, Lange T, Mueller C, et al. Imatinib for relapsed Philadelphiachromosome

positive chronic myeloid leukaemia after allogeneic haematopoietic cell transplantation. Bone

Marrow Transplant. 2005; 35(suppl 2): P800.

80. Rousselot P, Huguet F, Cayuela JM, et al. Imatinib mesylate discontinuation in patients with

chronic myeloid leukaemia in complete molecular remission for more than two years. Blood. 2005;

106: 321a, abstract 1101.

81. 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. 2004; 22: 935-942.

82. 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. 2005; 60: 35-44.

83. Zonder JA, Pemberton P, Brandt H, et al. The effect of dose increase of imatinib mesylate in

patients with chronic or accelerated phase chronic myelogenous leukemia with inadequate

hematologic or cytogenetic response to initial treatment. Clin Cancer Res. 2003; 9: 2092-2097.

44

84. 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. 2003;

101: 473-475.

85. Cortes J, Giles F, O’Brien S, et al. Result of high-dose imatinib mesylate in patients with

Philadelphiachromosome-positive chronic myeloid leukemia after failure of interferon-alpha.

Blood. 2003; 102: 83-86.

86. Kantarjian H, Talpaz M, O’Brien S, et al. High-dose imatinib mesylate therapy in newly

diagnosed Philadelphiachromosome-positive chronic phase chronic myeloid leukemia. Blood.

2004; 103: 2873-2878.

86. Hughes T, Branford S, Reynolds J, et al. Higher-dose Imatinib (600 mg/day) with selective

intensification in newly diagnosed CML patients in chronic phase; cytogenetic response rates at 12

months are superior to IRIS. Blood. 2004; 104: 286a, abstract 1001.

88. 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)

vs high dose (800 mg daily) in high Sokal risk patients. Blood. 2005; 106: 320a, abstract 1098.

89. Guerci A, Nicolini F, Maloisel F, et al. Randomized comparison of Imatinib with Imatinib

combination therapies in newly diagnosed chronic myelogenous leukemia patients in chronic phase:

design and first interim analysis of a phase II trial from the French CML group. Blood. 2005; 106:

53a, abstract 168.

90. Gardembas M, Rousselot P, Tulliez M, et al. Results of a prospective phase II study combining

Imatinib Mesylate and Cytarabine for the treatment of Philadelphia-positive patients with chronic

myelogenous leukemia in chronic phase. Blood. 2003; 102: 4298-4305.

91. Cortes J, Talpaz M, O’Brien S, et al. A randomized trial of high dose Imatinib Mesylate with or

without Peg-Interferon and GM-CSF as frontline therapy for patients with chronic myeloid

leukemia in early chronic phase. Blood. 2005; 106: 316a, abstract 1084

92. La Rosée P, Johnson K, O’Dwyer ME, et al. In vitro studies of the combination of imatinib

mesylate (Gleevec) and arsenic trioxide (Trisenox) in chronic myelogenous leukemia. Exp

Hematol. 2002; 30: 729-737.

93. Hoover RR, Mahon FX, Melo JV, et al. Overcoming STI571 resistance with the farnesyl

transferase inhibitor SCH66336. Blood. 2002; 100: 1068-1071.

94. Gatto S, Scappini B, Pham L, et al. The proteasome inhibitor PS-341 inhibits growth and

induces apoptosis in Bcr/Abl-positive cell lines sensitive and resistant to imatinib mesylate.

Haematologica. 2003; 88: 853-863.

95. Nimmanapalli R, Fuino L, Bali P, et al. Histone deacetylase inhibitor LAQ824 both lowers

expression and promotes proteasomal degradation of Bcr-Abl and induces apoptosis of imatinib

mesylate-sensitive or -refractory chronic myelogenous leukemia-blast crisis cells. Cancer Res.

2003; 63: 5126-5135.

45

96. La Rosée P, Johnson K, Corbin AS, et al. In vitro efficacy of combined treatment depends on

the underlying mechanism of resistance in imatinib-resistant Bcr-Abl-positive cell lines. Blood.

2004; 103: 208-215.

97. Dai Y, Rahmani M, Pei XY, et al. Bortezomib and flavopiridol interact synergistically to induce

apoptosis in chronic myeloid leukemia cells resistant to imatinib mesylate through both Bcr/Abldependent

and -independent mechanisms. Blood. 2004; 104: 509-518.

98. Jørgensen HG, Allan EK, Graham SM, et al. Lonafarnib reduces the resistance of primitive

quiescent CML cells to imatinib mesylate in vitro. Leukemia. 2005; 19: 1184-1191.

99. Jørgensen HG, Allan EK, Mountford JC, et al. Enhanced CML stem cell elimination in vitro by

bryostatin priming with imatinib mesylate. Exp Hematol. 2005; 33: 1140-1146.

100. Tseng PH, Lin HP, Zhu J, et al. Synergistic interactions between imatinib mesylate and the

novel phosphoinositide-dependent kinase-1 inhibitor OSU-03012 in overcoming imatinib mesylate

resistance. Blood. 2005; 105: 4021-4027.

101. Aloisi A, Di Gregorio S, Stagno F, et al. BCR-ABL nuclear entrapment kills human CML

cells: ex vivo study on 35 patients with the combination of Imatinib Mesylate and Leptomycin B.

Blood. 2006; 107: 1591-1598.

102. Chuah C, Barnes DJ, Kwok M, et al. Zoledronate inhibits proliferation and induces apoptosis

of imatinib-resistant chronic myeloid leukaemia cells. Leukemia. 2005; 19: 1896-1904.

103. Dengler J, von Bubnoff N, Decker T, et al. Combination of imatinib with rapamycin or

RAD001 acts synergistically only in Bcr-Abl-positive cells with moderate resistance to imatinib.

Leukemia. 2005; 19: 1835-1838.

104. Du Y, Wang K, Fang H, et al. Coordination of intrinsic, extrinsic and endoplasmic reticulummediated

apoptosis by imatinib mesylate combined with arsenic trioxide in chronic myeloid

leukemia. Blood 2006; 107: 1582-1590.

105. Gu JJ, SantiagoL, Mitchell BS. Synergy between imatinib and mycophenolic acid in inducing

apoptosis in cell lines expressing Bcr-Abl. Blood. 2005; 105: 3270-3277.

106. Segawa H, Kimura S, Kuroda J, et al. Zoledronate synergises with imatinib mesylate to inhibit

Ph+ primary leukaemic cell growth. Br J Haematol. 2005; 130: 558-560.

107. Cortes J, O’Brien S, Verstovsek S, et al. Phase II study of Lonafarnib (SCH66336) in

combinations with Imatinib for patients with chronic myeloid leukemia after failure to Imatinib.

Blood. 2004; 104: 288a, abstract 1009.

108. Cortes J, Garcia-Manero G, O’Brien S, et al. A phase I study of Tipifarnib in combination with

Imatinib Mesylate for patients with chronic myeloid leukemia in chronic phase who failed IM

therapy. Blood. 2004; 104: 289a, abstract 1011.

109. Marin D, Kaeda JS, Andreasson C, et al. Phase I/II trial of adding semisynthetic

homoharringtonine in chronic myeloid leukemia patients who have achieved partial or complete

cytogenetic response on imatinib. Cancer. 2005; 103: 1850-1855.

46

110. Mauro MJ, Deininger MW, Heinrich MD, et al. Arsenic trioxide (Trisenox) in combination

with Imatinib mesylate in patients with Imatinib-resistant chronic myeloid leukemia in chronic

phase: results of a phase I/II study. Haematologica 2005; 90 (suppl 2): 151-152, abstract 0383.

111. Shimoni A, Kröger N, Zander AR, et al. Imatinib mesylate (STI571) in preparation for

allogeneic hematopoietic stem cell transplantation and donor lymphocyte infusions in patients with

Philadelphia-positive acute leukemias. Leukemia. 2003; 17: 290-297.

112. Kim DW, Chung YJ, Lee S, et al. Pretransplant Imatinib can improve the outcome of non

myeloablative stem cell transplantation without increasing the mortality in Philadelphiachromosome

positive chronic myeloid leukemia. Leukemia 2004; 18: 1907-1909.

113. Zaucha JM, Prejzner W, Giebel S, et al. Imatinib therapy prior to myeloablative allogeneic

stem cell transplantation. Bone Marrow Transplant. 2005; 36: 417-424.

114. Bornhäuser M, Kröger N, Schwerdtfeger R, et al. Allogeneic haematopoietic cell

transplantation for chronic myelogenous leukaemia in the era of imatinib: a retrospective

multicentre study. Eur J Haematol. 2006; 76: 9-17.

115. Deininger M, Schleuning M, Greinix H, et al. The effect of prior exposure to imatinib on

transplant-related mortality. Haematologica. 2006; 91:452-459.

116. Kantarjian HM, O’Brien S, Cortes JE, et al. Imatinib mesylate therapy for relapse after

allogeneic stem cell transplantation for chronic myelogenous leukemia. Blood. 2002; 100: 1590-

1595.

117. De Angelo 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. 2004; 10: 5065-5071.

118. 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. 2003; 17: 1707-

1712.

119. Savani BN, Montero A, Kurlander R, et al. Imatinib synergizes with donor lymphocyte

infusions to achieve rapid molecular remission of CML relapsing after allogeneic stem cell

transplantation. Bone Marrow Transplant. 2005; 36: 1009-1015.

120. Mahon FX, Deininger MW, Schultheis B, et al. Selection and characterization of BCR-ABL

positive cell lines with differential sensitivity to the tyrosine kinase inhibitor STI571: diverse

mechanisms of resistance. Blood. 2000; 96: 1070-1079.

121. Mahon FX, Belloc F, Lagarde V, et al. MDR1 gene overexpression confers resistance to

imatinib mesylate in leukemia cell line models. Blood. 2003; 101: 2368-2373.

122. 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.

2004; 18: 401-408.

47

123. Thomas J, Wang L, Clark RE, et al. Active transport of imatinib into and out of cells:

implications for drug resistance. Blood. 2004; 104: 3739-3745.

124. Radujkovic A, Schad M, Topaly J, et al. Synergistic activity of imatinib and 17-AAG in

imatinib-resistant CML cells overexpressing BCR-ABL. Inhibition of P-glycoprotein function by

17-AAG. Leukemia. 2005; 19: 1198-1206.

125. Rumpold H, Wolf AM, Gruenewald K, et al. RNAi-mediated knockdown of P-glycoprotein

using a transposon-based vector system durably restores imatinib sensitivity in imatinib-resistant

CML cell lines. Exp Hematol. 2005; 33: 767-775.

126. Ferrao PT, Frost MJ, Siah SP, et al. Overexpression of P-glycoprotein in K562 cells does not

confer resistance to the growth inhibitory effects of imatinib (STI571) in vitro. Blood. 2003; 102:

4499-4503.

127. Zong Y, Zhou S, Sorrentino BP. Loss of P-glycoprotein expression in hematopoietic stem cells

does not improve responses to imatinib in a murine model of chronic myelogenous leukemia.

Leukemia. 2005; 19: 1590-1596.

128. Neville K, Parise RA, Thompson P, et al. Plasma and cerebrospinal fluid pharmacokinetics of

imatinib after administration to nonhuman primates. Clin Cancer Res. 2004; 10: 2525-2529.

129. Crossman LC, Druker BJ, Deininger MW. hOCT 1 and resistance to imatinib. Blood. 2005;

106: 1133-1134.

130. Weisberg E, Griffin JD. Mechanism of resistance to the ABL tyrosine kinase inhibitor STI571

in BCR/ABL-transformed hematopoietic cell lines. Blood. 2000; 95: 3498-3505.

131. Le Coutre P, Gambacorti-Passerini C. Induction of resistance to the Abelson inhibitor STI571

in human leukemic cells through gene amplification. Blood. 2000; 95: 1758-1766.

132. 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; 293: 876-880.

133. 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; 2: 117-125.

134. Hochhaus A, Kreil S, Corbin AS, et al. Molecular and chromosomal mechanisms of resistance

to imatinib (STI571) therapy. Leukemia. 2002; 16: 2190-2196.

135. Gambacorti-Passerini C, Gunby RH, Piazza R, et al. Molecular mechanisms of resistance to

imatinib in Philadelphia-chromosome-positive leukaemias. Lancet Oncol. 2003; 4: 75-85.

136. Branford S, Rudzki Z, Walsh S, et al. High frequency of point mutations clustered within the

adenosine triphosphate-binding region of BCR/ABL in patients with chronic myeloid leukemia or

Ph-positive acute lymphoblastic leukemia who develop imatinib (STI571) resistance. Blood. 2002;

99: 3472-3475.

137. 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

48

ATP phosphate-binding loop (P-loop) are associated with a poor prognosis. Blood. 2003; 102: 276-

283.

138. Corbin AS, La Rosée P, Stoffregen EP, et al. Several Bcr-Abl kinase domain mutants

associated with imatinib mesylate resistance remain sensitive to imatinib. Blood. 2003; 101: 4611-

4614.

139. Hochhaus A, La Rosée P. Imatinib therapy in chronic myelogenous leukemia: strategies to

avoid and overcome resistance. Leukemia. 2004; 18: 1321-1331.

140. 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. 2005; 23: 4100-4109.

141. Barnes DJ, Palaiologou D, Panousopoulou E, et al. Bcr-Abl expression levels determine the

rate of development of resistance to imatinib mesylate in chronic myeloid leukemia. Cancer Res.

2005; 65: 8912-8919.

142. Al-Ali HK, Heinrich MC, Lange T, et al. High incidence of BCR-ABL kinase domain

mutations and absence of mutations of the PDGFR and KIT activation loops in CML patients with

secondary resistance to imatinib. Hematol J. 2004; 5: 55-60.

143. 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. 2004; 104: 2926-2932.

144. Sherbenou DW, Wong MJ, Humayun A, et al. In chronic myeloid leukemia patients with

complete cytogenetic response to Imatinib, BCR-ABL kinase domain mutations are relatively rare

and not consistently associated with subsequent relapse. Blood. 2005; 106: 131a, abstract 434.

145. Khorashad JS, Anand M, Marin D, et al. The presence of a BCR-ABL mutant allele in CML

does not always explain clinical resistance to Imatinib. Prepublished online. February 9, 2006. DOI

10.1038/sj.leukemia.2404137.

146. Roche-Lestienne C, Soenen-Cornu V, Grardel-Duflos N, et al. Several types of mutations of

the Abl gene can be found in chronic myeloid leukemia patients resistant to STI571, and they can

pre-exist to the onset of treatment. Blood. 2002; 100: 1014-1018.

147. Willis SG, Lange T, Demehri S, et al. High-sensitivity detection of BCR-ABL kinase domain

mutations in imatinib-naive patients: correlation with clonal cytogenetic evolution but not response

to therapy. Blood. 2005; 106: 2128-2137.

148. Roche-Lestienne C, Laï JL, Darré S, et al. A mutation conferring resistance to imatinib at the

time of diagnosis of chronic myelogenous leukemia. N Engl J Med. 2003; 348: 2265-2266.

149. Chu S, Xu H, Shah NP, et al. Detection of BCR-ABL kinase mutations in CD34+ cells from

chronic myelogenous leukemia patients in complete cytogenetic remission on imatinib mesylate

treatment. Blood. 2005; 105: 2093-2098.

49

150. Angstreich GR, Matsui W, Huff CA, et al. Effects of imatinib and interferon on primitive

chronic myeloid leukaemia progenitors. Br J Haematol. 2005; 130: 373-381.

151. Jiang X, Zhao Y, Chan WY, et al. Leukemic stem cells of chronic phase CML patients

consistently display very high BCR-ABL transcript levels and reduced responsiveness to Imatinib

mesylate in addition to generating a rare subset that produces Imatinib mesylate resistant

differentiated progeny. Blood. 2005; 106: 204a, abstract 711.

152. Hochhaus A, Ernst T, Erben P, et al. Long term observation of CML patients after Imatinib

resistance associated with BCR-ABL mutations. Blood. 2005; 106: 316a, abstract 1086.

153. Soverini S, Colarossi S, Gnani A, et al. Frequency, distribution and prognostic value of ABL

kinase domain mutations in different subsets of Philadelphia-positive patients resistant to Imatinib,

by the GIMEMA working party on CML. Blood. 2005;106: 131a, abstract 435

154. Martinelli G, Soverini S, Rosti G, et al. Dual tyrosine kinase inhibitors in chronic myeloid

leukemia. Leukemia. 2005; 19: 1872-1879.

155. O’Dwyer M, Mauro MJ, Kurilik G, et al. The impact of clonal evolution on response to

imatinib mesylate (STI571) in accelerated phase CML. Blood. 2002; 100: 1628-1633.

156. Cortes JE, Talpaz M, Giles F, et al. Prognostic significance of cytogenetic clonal evolution in

patients with chronic myelogenous leukemia on imatinib mesylate therapy. Blood. 2003; 101: 3794-

3800.

157. Schoch C, Haferlach T, Kern W, et al. Occurrence of additional chromosome aberrations in

chronic myeloid leukemia patients treated with imatinib mesylate. Leukemia. 2003; 17: 461-463.

158. Mohamed AN, Pemberton P, Zonder J, et al. The effect of imatinib mesylate on patients with

Philadelphia chromosome-positive chronic myeloid leukemia with secondary chromosomal

aberrations. Clin Cancer Res. 2003; 9: 1333-1337.

159. 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. 2003; 88: 260-267.

160. O’Dwyer M, Mauro MJ, Blasdel C, et al. Clonal evolution and lack of cytogenetic response

are adverse prognostic factors for hematologic relapse of chronic phase CML patients treated with

imatinib mesylate. Blood. 2004; 103: 451-455.

161. Huntly BJ, Guilhot F, Reid AG, et al. Imatinib improves but may not fully reverse the poor

prognosis of patients with CML with derivative chromosome 9 deletions. Blood. 2003; 102: 2205-

2212.

162. Quintas-Cardama A, Kantarjian H, Talpaz M, et al. Imatinib mesylate therapy may overcome

the poor prognostic significance of deletions of derivative chromosome 9 in patients with chronic

myelogenous leukemia. Blood. 2005; 105: 2281-2286.

163. O’Dwyer ME, Gatter KM, Loriaux M, et al. Demonstration of Philadelphia chromosome

negative abnormal clones in patients with chronic myelogenous leukemia during major cytogenetic

responses induced by imatinib mesylate. Leukemia. 2003; 17: 481-487.

50

164. Bumm T, Müller C, Al-Ali HK, et al. Emergence of clonal cytogenetic abnormalities in Phcells

in some CML patients in cytogenetic remission to imatinib but restoration of polyclonal

hematopoiesis in the majority. Blood. 2003; 101: 1941-1949.

165. 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. 2003;

98: 1905-1911.

166. Abruzzese E, Gozzetti A, Zaccaria A, et al. Ph-abnormal clones emerged during Imatinib

therapy: clinical report and clonal analyses on 23 patients from GIMEMA working party in CML

registry. Blood. 2004; 104: 803a, abstract 2936.

167. 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. 2004; 18: 1340-1346.

168. Bacher U, Hochhaus A, Berger U, et al. Clonal aberrations in Philadelphia chromosome

negative hematopoiesis in patients with chronic myeloid leukemia treated with imatinib or

interferon alpha. Leukemia. 2005; 19: 460-463.

169. Deininger MW, Kantarjian H, Byung P, et al. Good prognosis of CML patients with clonal

cytogenetic abnormalities in Ph-negative cells. Blood. 2005; 106: 315a, abstract 1082.

170. Jabbour E, Kantarjian H, O’Brien S, et al. Chromosomal abnormalities in Philadelphia

chromosome – negative metaphases appearing during Imatinib Mesylate therapy in patients with

newly diagnosed chronic myeloid leukemia in chronic phase. Blood. 2005; 106: 317a, abstract

1090.

171. Rosti G, Trabacchi E, Bassi S, et al. Risk and early cytogenetic response to imatinib and

interferon in chronic myeloid leukemia. Haematologica. 2003; 88: 256-259.

172. Guilhot F, on behalf of the IRIS study group. Sustained durability of responses plus high rates

of cytogenetic responses result in long term benefit for newly diagnosed chronic phase chronic

myeloid leukemia treated with Imatinib therapy: update from the IRIS study. Blood. 2004; 104:

10a, abstract 21.

173. Tipping AJ, Deininger MW, Goldman JM, et al. Comparative gene expression profile of

chronic myeloid leukemia cells innately resistant to imatinib mesylate. Exp Hematol. 2003; 31:

1073-1080.

174. McLean LA, Gathmann I, Capdeville R, et al. Pharmacogenomic analysis of cytogenetic

response in chronic myeloid leukemia patients treated with imatinib. Clin Cancer Res. 2004; 10:

155-165.

175. Crossman Lc, Mori M, Hsieh YC, et al. In chronic myeloid leukemia white cells from

cytogenetic responders and non-responders to imatinib have very similar gene expression

signatures. Haematologica. 2005; 90: 459-464.

51

176. Oehler V, Branford S, Pogosova-Agadjanyan E, et al. Gene expression signatures associated

with treatment and resistance to Imatinib mesylate in chronic myeloid leukemia patients. Blood.

2005; 106: 131a, abstract 433.

177. Yong AS, Szydlo RM, Goldman JM et al. Molecular profiling of CD34+ cells identifies low

expression of CD7, along with high expression of proteinase 3 or elastase, as predictors of longer

survival in patients with CML. Blood. 2006; 107: 205-212.

178. Dressman MA, Malinowski R, McLean LA, et al. Correlation of major cytogenetic response

with a pharmacogenetic marker in chronic myeloid leukemia patients treated with imatinib

(STI571). Clin Cancer Res. 2004; 10: 2265-2271.

179. Crossman LC, Loriaux M, Vartanian K, et al. Gene expression profiling of CML CD34+ cells

prior to Imatinib therapy reveals differences between patients with and without subsequent

complete cytogenetic response. Blood. 2005; 106: 330a, abstract 1222.

180. Cilloni D, Messa F, Gottardi E, et al. Sensitivity to imatinib therapy may be predicted by

testing Wilms tumor gene expression and colony growth after a short in vitro incubation. Cancer.

2004; 101: 979-988.

181. Schultheis B, Szydlo R, Mahon FX, et al. Analysis of total phosphotyrosine levels in CD34+

cells from CML patients to predict the response to imatinib mesylate treatment. Blood. 2005; 105:

4893-4894.

182. White D, Saunders V, Lyons AB, et al. In vitro sensitivity to imatinib-induced inhibition of

ABL kinase activity is predictive of molecular response in patients with de novo CML. Blood.

2005; 106: 2520-2526.

183. Druker B, Gathmann I, Bolton AE et al. Probability and impact of obtaining a cytogenetic

response to Imatinib as initial therapy for chronic myeloid leukemia in chronic phase. Blood. 2003;

102: 182a, abstract 634.

184. Merx K, Müller MC, Kreil S, et al. Early reduction of BCR-ABL mRNA transcript levels

predicts cytogenetic response in chronic phase CML patients treated with imatinib after failure of

interferon alpha. Leukemia. 2002; 16: 1579-1583.

185. Wang L, Pearson K, Ferguson JE, et al. The early molecular response to imatinib predicts

cytogenetic and clinical outcome in chronic myeloid leukaemia. Br J Haematol. 2003; 120(6): 990-

999.

186. Clark RE, Knight K, Lucas CM, et al. Consecutive but not isolated BCR-ABL transcript level

rises are predictive of BCR-ABL kinase mutations in chronic myeloid leukemia patients treated by

Imatinib. Exp Hematol. 2005; 33 (suppl 1): 52, abstract 56.

187. Lesser ML, Dewald GW, Sison CP, et al. Correlation of three methods of measuring

cytogenetic response in chronic myelocytic leukemia. Cancer Genet Cytogenet.2002; 137: 79-84.

188. Schoch C, Schnittger S, Bursch S, et al. Comparison of chromosome banding analysis,

interphase- and hypermetaphase-FISH, qualitative and quantitative PCR for diagnosis and for

follow-up in chronic myeloid leukemia: a study on 350 cases. Leukemia. 2002; 16: 53-59.

52

189. Hughes T, Deininger M, 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. Prepublished on line. March 7, 2006. DOI 10/1182/blood-2006-01-0092.

190. Shah NP, Tran C, Lee FY, et al. Overriding Imatinib resistance with a novel ABL kinase

inhibitor. Science 2004; 305: 399-401.

191. Gumireddy K, Baker SJ, CosenzaSC, et al. A non-ATP-competitive inhibitor of BCR-ABL

overrides imatinib resistance. Proc Natl Acad Sci U S A. 2005; 102: 1992-1997.

192. Weisberg E, Manley PW, Breitenstein W, et al. Characterization of AMN107, a selective

inhibitor of native and mutant Bcr-Abl. Cancer Cell. 2005; 7: 129-141.

193. Golemovic M, Verstovsek S, Giles F, et al. AMN107, a novel aminopyrimidine inhibitor of

Bcr-Abl, has in vitro activity against imatinib-resistant chronic myeloid leukemia. Clin Cancer Res.

2005; 11: 4941-4947.

194. O’Hare T, Walters DK, Stoffregen EP, et al. In vitro activity of Bcr-Abl inhibitors AMN107

and BMS-354825 against clinically relevant imatinib-resistant Abl kinase domain mutants. Cancer

Res. 2005; 65: 4500-4505.

1

 

דפים

יצירת קשר

© 2021, כל הזכויות שמורות ל- עמותת חולי CML בישראל