chronic myelogenous leukemia

4
CONTEMPO UPDATES LINKING EVIDENCE AND EXPERIENCE Chronic Myelogenous Leukemia Mamta Kalidas, MD Hagop Kantarjian, MD Moshe Talpaz, MD Natural History Chronic myelogenous leukemia (CML) is a disorder of hematopoietic stem cells 1 accounting for 15% of adult leu- kemias. The median age at presenta- tion is between 50 and 60 years; 12% to 30% of patients at diagnosis are older than 60 years. Chronic myelogenous leukemia clas- sically progresses through 3 phases, be- coming more resistant to treatment in each successive phase. The majority of patients present in the chronic phase, which may last 4 to 6 years and is often asymptomatic at diagnosis. In the accel- erated phase, symptoms become worse and immature blasts increase in the pe- ripheral blood. The duration of this phase may last as long as a year. The final and fatal blastic phase (.30% blasts in the bone marrow or peripheral blood) has features of an acute leukemia including fever, weight loss, bleeding, and ane- mia; this phase may last 3 to 6 months. Pathophysiology The Philadelphia (Ph9) chromosome was first described in 1960 in patients with CML as a shortened chromosome that was later discovered to be chromosome 22. 2 In 1973 it was further character- ized as a t(9;22) translocation. 3 Ninety- five percent of patients with CML have this defining Ph9 chromosome from a re- ciprocal translocation t(9;22)(q34;q11). This translocation is also present in 15% to 30% of adult acute lymphoid leuke- mia (ALL) patients and 2% of acute my- eloblastic leukemia patients. 4,5 The con- sequence of this translocation is a fusion of the bcr (breakpoint cluster region) gene on chromosome 22 to the cellular abl (c-abl) gene on chromosome 9. The resulting fusion protein, Bcr-Abl, is a con- stitutively active cytoplasmic tyrosine ki- nase that can vary in size (185 kd-230 kd) and can phosphorylate several sub- strates resulting in the initiation of mul- tiple signal cascades involved in cell growth, differentiation, adhesion, and death 6 (FIGURE 1). Since this tyrosine ki- nase is constitutively active, the cells by- pass regulated growth and undergo a ma- lignant transformation to leukemia. Monitoring With Bcr-Abl Patients with CML currently may be monitored by hematologic, cytogenetic (karyotype), and molecular tests such as FISH (fluorescence in situ hybridiza- tion) and RT-PCR (reverse transcriptase– polymerase chain reaction). Based on these assays, therapy outcome may be defined in terms of hematologic remis- sion (normalization of blood cell counts and spleen size), varying degrees of cyto- genetic response (decline in percentage of Ph9 cells), or a molecular remission (PCR negative). Cytogenetic analysis is tedious, but has become useful in pre- dicting survival outcome. 7-9 Polymerase chain reaction is a highly sensitive assay and can detect a single Ph9 cell in 10 000 to 100 000 normal cells. 10 Bcr-Abl mes- senger RNA (mRNA) transcripts can now be quantitated and followed over time by newly developed PCR assays to predict eventual relapse in patients who have undergone transplantation, 11 although the predictive significance of these assays in nontransplant therapies of CML has not been fully clarified. Evolution of Therapy in CML Prior to the 1950s, splenic irradiation was the mainstay of CML therapy but it had minimal effect on survival (TABLE). 12 In 1953, busulfan was in- troduced for palliation of chronic- phase CML. 14 Ten years later, hydroxy- urea, a ribonucleotide reductase inhibitor, became available 23 and was favored over busulfan because it had fewer adverse effects and improved sur- vival. 15,22 However, since neither drug affected either cytogenetic response or progression to blast crisis, 15,22 more ef- fective therapy was needed. Alloge- neic stem cell transplantation (SCT) for CML was first performed in the 1970s, initially with identical twin trans- plants 24 and subsequently with HLA- matched sibling transplants. 25 Since few patients were eligible for SCT, alterna- tive treatment was desired, and in the early 1980s a-interferon (IFN)–based therapy was introduced. 26 Currently, allogeneic matched sib- ling SCT is favored for young (,30-40 years old) patients in chronic phase who are within 1 year of diagnosis. These pa- tients can have “cure” rates of 50% to 55% at 10 years. 27 For older patients, the transplant-associated mortality rate is 30% to 50%, which is higher than the mortality rate in younger patients. 28,29 Even with the long-term event-free sur- vival rates seen with allogeneic SCT, 10% to 20% of the patients who receive one will experience a disease relapse, which generally occurs within 3 years of trans- See also Patient Page. Author Affiliations: Departments of Bioimmuno- therapy (Drs Kalidas and Talpaz) and Leukemia (Dr Kantarjian), M.D. Anderson Cancer Center, Hous- ton, Tex. Financial Disclosure: Dr Kantarjian received grants or funding from Novartis and Schering Plough; Dr Tal- paz received grants or funding (including lecture spon- sorships) from Novartis, Schering Plough, and Hoff- mann LaRoche. Corresponding Author and Reprints: Moshe Talpaz, MD, Department of Bioimmunotherapy, M.D. Ander- son Cancer Center, 1515 Holcombe Blvd, Box 422, Houston, TX 77030 (e-mail: mtalpaz@mail .mdanderson.org). Contempo Updates Section Editors: Alice T. D. Hughes, MD; Janet M. Torpy, MD, Fishbein Fellows. ©2001 American Medical Association. All rights reserved. (Reprinted) JAMA, August 22/29, 2001—Vol 286, No. 8 895 Downloaded From: http://jama.jamanetwork.com/ by a University Of Connecticut Health Center User on 12/03/2013

Upload: mamta

Post on 19-Dec-2016

215 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Chronic Myelogenous Leukemia

CONTEMPO UPDATESLINKING EVIDENCE AND EXPERIENCE

Chronic Myelogenous LeukemiaMamta Kalidas, MDHagop Kantarjian, MDMoshe Talpaz, MD

Natural HistoryChronic myelogenous leukemia (CML)is a disorder of hematopoietic stemcells1 accounting for 15% of adult leu-kemias. The median age at presenta-tion is between 50 and 60 years; 12%to 30% of patients at diagnosis are olderthan 60 years.

Chronic myelogenous leukemia clas-sically progresses through 3 phases, be-coming more resistant to treatment ineach successive phase. The majority ofpatients present in the chronic phase,which may last 4 to 6 years and is oftenasymptomatic at diagnosis. In the accel-erated phase, symptoms become worseand immature blasts increase in the pe-ripheral blood. The duration of this phasemay last as long as a year. The final andfatal blastic phase (.30% blasts in thebone marrow or peripheral blood) hasfeatures of an acute leukemia includingfever, weight loss, bleeding, and ane-mia; this phase may last 3 to 6 months.

PathophysiologyThe Philadelphia (Ph9) chromosome wasfirst described in 1960 in patients withCML as a shortened chromosome thatwas later discovered to be chromosome22.2 In 1973 it was further character-ized as a t(9;22) translocation.3 Ninety-five percent of patients with CML havethis defining Ph9 chromosome from a re-ciprocal translocation t(9;22)(q34;q11).This translocation is also present in 15%to 30% of adult acute lymphoid leuke-mia (ALL) patients and 2% of acute my-eloblastic leukemia patients.4,5 The con-

sequence of this translocation is a fusionof the bcr (breakpoint cluster region)gene on chromosome 22 to the cellularabl (c-abl) gene on chromosome 9. Theresulting fusionprotein,Bcr-Abl, is a con-stitutively active cytoplasmic tyrosine ki-nase that can vary in size (185 kd-230kd) and can phosphorylate several sub-strates resulting in the initiation of mul-tiple signal cascades involved in cellgrowth, differentiation, adhesion, anddeath6 (FIGURE 1). Since this tyrosine ki-nase is constitutively active, the cells by-pass regulated growth and undergo a ma-lignant transformation to leukemia.

Monitoring With Bcr-AblPatients with CML currently may bemonitored by hematologic, cytogenetic(karyotype), and molecular tests such asFISH (fluorescence in situ hybridiza-tion)andRT-PCR(reverse transcriptase–polymerase chain reaction). Based onthese assays, therapy outcome may bedefined in terms of hematologic remis-sion (normalization of blood cell countsand spleen size), varying degrees of cyto-genetic response (decline in percentageof Ph9 cells), or a molecular remission(PCR negative). Cytogenetic analysis istedious, but has become useful in pre-dicting survival outcome.7-9 Polymerasechain reaction is a highly sensitive assayand can detect a single Ph9 cell in 10000to 100000 normal cells.10 Bcr-Abl mes-sengerRNA(mRNA)transcriptscannowbequantitatedand followedover timebynewly developed PCR assays to predicteventual relapse in patients who haveundergone transplantation,11 althoughthepredictive significanceof theseassaysin nontransplant therapies of CML hasnot been fully clarified.

Evolution of Therapy in CMLPrior to the 1950s, splenic irradiationwas the mainstay of CML therapy but

it had minimal effect on survival(TABLE).12 In 1953, busulfan was in-troduced for palliation of chronic-phase CML.14 Ten years later, hydroxy-urea, a ribonucleotide reductaseinhibitor, became available23 and wasfavored over busulfan because it hadfewer adverse effects and improved sur-vival.15,22 However, since neither drugaffected either cytogenetic response orprogression to blast crisis,15,22 more ef-fective therapy was needed. Alloge-neic stem cell transplantation (SCT) forCML was first performed in the 1970s,initially with identical twin trans-plants24 and subsequently with HLA-matched sibling transplants.25 Since fewpatients were eligible for SCT, alterna-tive treatment was desired, and in theearly 1980s a-interferon (IFN)–basedtherapy was introduced.26

Currently, allogeneic matched sib-ling SCT is favored for young (,30-40years old) patients in chronic phase whoare within 1 year of diagnosis. These pa-tients can have “cure” rates of 50% to55% at 10 years.27 For older patients, thetransplant-associated mortality rate is30% to 50%, which is higher than themortality rate in younger patients.28,29

Even with the long-term event-free sur-vival rates seen with allogeneic SCT, 10%to 20% of the patients who receive onewill experience a disease relapse, whichgenerally occurs within 3 years of trans-

See also Patient Page.

Author Affiliations: Departments of Bioimmuno-therapy (Drs Kalidas and Talpaz) and Leukemia (DrKantarjian), M.D. Anderson Cancer Center, Hous-ton, Tex.Financial Disclosure: Dr Kantarjian received grants orfunding from Novartis and Schering Plough; Dr Tal-paz received grants or funding (including lecture spon-sorships) from Novartis, Schering Plough, and Hoff-mann LaRoche.Corresponding Author and Reprints: Moshe Talpaz,MD, Department of Bioimmunotherapy, M.D. Ander-son Cancer Center, 1515 Holcombe Blvd, Box 422,Houston, TX 77030 (e-mai l : mtalpaz@mai l.mdanderson.org).Contempo Updates Section Editors: Alice T. D.Hughes, MD; Janet M. Torpy, MD, Fishbein Fellows.

©2001 American Medical Association. All rights reserved. (Reprinted) JAMA, August 22/29, 2001—Vol 286, No. 8 895

Downloaded From: http://jama.jamanetwork.com/ by a University Of Connecticut Health Center User on 12/03/2013

Page 2: Chronic Myelogenous Leukemia

plantation.30 Treatment choices in-clude a-IFN, infusion of donor lympho-cytes, or a second SCT. Infusion of donorlymphocytes is most effective when givenwithin the first 2 years of the SCT.31 Lym-phocytes from the original donor aretransfused and induce complete remis-sion through graft-vs-tumor effect in 60%to 80% of patients who relapse with ac-tive chronic-phase CML.31

Although allogeneic SCT from sib-lings is the best chance for cure for CMLpatients in chronic phase, only 15% to20% of patients are candidates for thistreatment given the limitations of age orlack of HLA-matched related donors fail-ing to provide therapeutic solutions forthe majority of patients with CML. Forpatients not eligible for SCT, a-IFN plushydroxyurea and/or cytarabine can beused.32 Combination therapy with a-IFNand cytarabine has a slight survival ad-vantage over a-IFN alone.7,20,33,34

a-Interferon was first used clinicallyin 1983 after reports of activity in CML.26

A single-center study later demon-strated that a-IFN induced complete he-matologic (80%) and complete cytoge-netic (26%) responses in patients withchronic-phase CML21 and similar re-sponses were confirmed by other inves-tigators.9,33-35 Median time to completehematologic remission is 6 to 7 monthsand 12 to 17 months for complete cy-togenetic remission.21 Survival ratesare higher in patients receiving a-IFNthan in patients receiving busulfan orhydroxyurea alone.20,22,34 The most re-markable impact on survival has beendemonstrated among patients withlow-risk disease, where survival rateshave exceeded 9 years.7,19-22,36,37 Further-more, greater than 80% of those whodemonstrate a complete cytogenetic re-sponse to a-IFN are alive for more than10 years.7,21 However, a-IFN therapy isassociated with considerable adverse ef-fects, which include fever, chills, fa-tigue, arthralgias, anorexia, and weightloss, and is discontinued in 4% to 18%of patients due to toxicity.34

Novel TherapiesThe search for more effective treatmenthas continued because of the limita-

Figure 1. Production of the Bcr-Abl Gene and Protein

Chromosome 9 Chromosome 9

Chromosome 22

TRANSFORMED HEMATOPOIETIC PROGENITOR CELL

Philadelphia Chromosome

Translocation

Bcr-Abl Fusion Protein

Bcr-Abl Fusion Protein

Tyrosine Kinase Domain

AblBcr

DNA Binding Sites

IncreasedCell Survival

UnregulatedCell Proliferation

AbnormalCell Adhesion

q34

q11

N C

Bcr-AblFusion Gene

abl Gene

bcr Gene

TranslationBcr-AblmRNA

Transcription of Bcr-Abl Fusion Gene

After the translocation between chromosomes 9 and 22, the Bcr-Abl fusion gene undergoes transcription. Theresulting chimeric Bcr-Abl messenger RNA (mRNA) is then translated into the Bcr-Abl tyrosine kinase protein,which enhances altered cell proliferation, adhesion, and survival.

Table. Evolution of Discoveries and Therapies in Chronic Myelogenous Leukemia*

Era Discovery TreatmentMedian

Survival, mo

1900-1920 Characterization ofdisease morphology

3112

1920-1950 Splenic irradiation 2812,13

1950-1960 Busulfan 35-4514,15

1960 Ph chromosome Hydroxyurea 48-6715-17

1970-present Allogeneic SCT 50%-60%cure rate18†

1980-present Bcr-Abl gene, protein,and cellular signaling

Interferons 55-897,19-22

1998-present STI-571 (specific targetedtherapy)

. . .‡

*SCT indicates stem cell transplant; STI-571, signal transduction inhibitor 571.†Only for patients eligible for SCT.‡Ellipses indicate not yet available.

CHRONIC MYELOGENOUS LEUKEMIA RESEARCH AND TREATMENT

896 JAMA, August 22/29, 2001—Vol 286, No. 8 (Reprinted) ©2001 American Medical Association. All rights reserved.

Downloaded From: http://jama.jamanetwork.com/ by a University Of Connecticut Health Center User on 12/03/2013

Page 3: Chronic Myelogenous Leukemia

tionsofcurrenttherapies—manypatientsare not eligible for a potentially curativeallogeneic SCT, SCT is associated withlife-threatening toxicities, and a-IFN isseldom curative and also associated withmorbidity. In 1996, this effort led to thedevelopment of relatively specific Bcr-Abl tyrosine kinase inhibitors.38 One ofthese, a 2-phenylaminopyrimidine, wasa potent relatively selective inhibitor ofthe Bcr-Abl tyrosine kinase. This com-pound, CGP 57148B (Novartis, Basel,Switzerland),demonstrated inhibitionofproliferation of Bcr-Abl expressing cellsaswell as inhibitionofBcr-Abl (+) tumorformation in profoundly immunodefi-cient mice.38 CGP 57148B (later namedSTI-571, signal transduction inhibitor571) had little in vivo toxicity in ani-mals and did not hinder the growth ofnormal cells.38 Further investigation ofthis compound has revealed that STI-571binds to theadenosinetriphosphate–binding site of Abl, thus inhibiting thephosphorylation of substrates and sub-sequent malignant transformation39

(FIGURE 2). Although STI-571 is rela-tivelyspecific forBcr-Abl tyrosinekinasesand is active against platelet-derivedgrowth factor receptor kinase40 and c-kitreceptor kinase,41 it is inactive against alarge number of other tyrosine kinases.38

Because of the promising in vitro andin vivo data, a phase 1 study of STI-571began in patients with chronic-phaseCML resistant to or intolerant of a-IFN.A total of 53 of 54 patients experiencedcomplete hematologic remissions.42 Cy-togenetic responses, were seen in 54% ofpatients.42 Complete hematologic remis-sions have continued in 51 of the 53 pa-tients after a median follow-up of 265days.42 These results are surprising inlight of the fact that these patients hadlong-standing disease (3 years on aver-age from diagnosis). The adverse ef-fects of STI-571 observed in the study in-cluded nausea, vomiting, and fluidretention; a maximal tolerated dose wasnot reached.40,42 Preliminary data froma phase 2 study of STI-571 evaluating cy-togenetic responses in patients with Ph(+) CML for whom a-IFN had not beensuccessful are also promising. After 6months of therapy, a major cytogenetic

response was observed in 56% of the 290evaluable patients.43 Phase 2 studies thatincluded patients with accelerated-phase CML have demonstrated an over-all hematological response rate of 78%;a complete hematologic remission hasbeen demonstrated in 22 patients.40 STI-571 has also been evaluated in patientswith blastic-phase CML and Ph (+) ALL.Fifty-five percent of patients with CMLin blastic phase demonstrated a re-sponse; 22% of these were complete re-sponses.44 Even though 14 of 20 pa-tients with Ph (+) ALL or lymphoid blastcrisis experienced a response, all but 1have relapsed while receiving therapy.44

A phase 3 trial is under way to investi-gate the use of STI-571 as front-line treat-ment of newly diagnosed patients withCML in chronic phase. It will compareSTI-571 with the combination of a-IFNand cytarabine.

In May 2001, the Food and Drug Ad-ministration approved STI-571 underthe name Gleevec (Novartis, East Ha-nover, NJ) for patients with chronic-

phase CML who are resistant to or in-tolerant of a-IFN as well as for patientswith more advanced stages of CML.

New AgentsA number of novel agents are still un-der investigation for the treatment ofCML. Homoharringtonine, a plant alka-loid, has shown activity in patients in theearly and late chronic phases of CML.45

A potent hypomethylating agent calleddecitabine has also demonstrated activ-ity in patients with CML in acceleratedand blastic phases.46 Of 5 patients withblastic-phase CML treated with troxa-citabine, a cytosine analog, 2 have re-turned to chronic phase.47 Immuno-therapy with specific vaccines is alsounder investigation.48

ConclusionsOver the past century, our understand-ing of CML pathogenesis and therapeu-tic strategies for CML has evolved con-siderably. Chronic myelogenousleukemia is one of the few diseases in

Figure 2. Structure and Mechanism of Action of STI-571

Substrate

Substrate

Malignant Transformation

Bcr-Abl Protein

Bcr-Abl Protein

Phosphorylation

No Phosphorylation

Chemical Structure of STI-571

C29H31N7O • CH4SO3

H

O

CH3

H

ATP

STI-571

ATP

Binds at ATP Binding Site

N

N

N N N

CH3

N

N

A B

A, After adenosine triphosphate (ATP) binds to the Bcr-Abl protein, the cell substrate becomes phosphory-lated and initiates a chain of signal cascades leading to malignant transformation of the cell. B, STI-571 bindsto the Bcr-Abl tyrosine kinase protein at its ATP binding site, thus preventing phosphorylation of the substrateand subsequent malignant transformation.

CHRONIC MYELOGENOUS LEUKEMIA RESEARCH AND TREATMENT

©2001 American Medical Association. All rights reserved. (Reprinted) JAMA, August 22/29, 2001—Vol 286, No. 8 897

Downloaded From: http://jama.jamanetwork.com/ by a University Of Connecticut Health Center User on 12/03/2013

Page 4: Chronic Myelogenous Leukemia

which a specific cytogenetic and mo-lecular abnormality has been impli-cated in the pathogenesis of the disease.The presence of a constitutively activeBcr-Abl tyrosine kinase in the tumor cellshas led to the first targeted moleculartherapy of its kind.This drug has the ad-vantage, compared with traditional an-tineoplastic agents, of being able to tar-get abnormal cells and spare normal cells.

Clinical trials have shown verypromising results of STI-571 in Bcr-Abl (+) leukemias in all phases. Themost durable responses have beenseen in patients with chronic-phaseCML who are resistant to or intolerantof a-IFN, but remarkable responseshave also been seen in accelerated andblastic phases of the disease. The suc-cess of this inhibitor has underscored

the significance of molecular basedtherapy. It may be the first in a seriesof new compounds that operate in asimilar mode of action in a variety ofother malignancies. Additionalresearch should help determine theoptimal role for STI-571, whether it bealone, in combination with a-IFN andcytarabine, or as first-line therapyfor CML.

REFERENCES

1. Fialkow PJ, Jacobson RJ, Papayannopoulou T.Chronic myelocytic leukemia. Am J Med. 1977;63:125-130.2. Nowell PC, Hungerford DA. A minute chromo-somal abnormality in human chronic granulocytic leu-kemia [abstract]. Science. 1960;132:1497.3. Rowley JD. A new consistent chromosomal abnor-mality in chronic myelogenous leukemia identified byquinacrine fluorescence and giemsa staining. Nature.1973;243:290-293.4. Specchia G, Mininni D, Guerrasio A, et al. Ph posi-tive acute lymphoblastic leukemia in adults. Leuk Lym-phoma. 1995;18(suppl 1):37-42.5. Kurzrock R, Gutterman JU, Talpaz M. The molecu-lar genetics of Philadelphia chromosome–positive leu-kemias. N Engl J Med. 1988;319:990-998.6. Sawyers CL. Medical progress. N Engl J Med. 1999;340:1330-1340.7. Italian Cooperative Study Group on Chronic My-eloid Leukemia. Interferon alpha-2a as compared withconventional chemotherapy for the treatment of chronicmyeloid leukemia. N Engl J Med. 1994;330:820-825.8. Talpaz M, Kantarjian H, Kurzrock R, et al. Interferon-alphasustainedcytogenetic responses inchronicmyelog-enous leukemia. Ann Intern Med. 1991;114:532-538.9. MahonFX,FaberesC,PueyoS,etal.Responseatthreemonths is a good predictive factor for newly diagnosedchronic myeloid leukemia patients treated by recombi-nant interferon-alpha. Blood. 1998;92:4059-4065.10. Arthur CK, Apperly JF, Guo AP, et al. Cytoge-netic events after bone marrow transplantation forchronic myeloid leukemia in chronic phase. Blood.1988;71:1179-1186.11. Mughal TI, Yong A, Szydlo RM, et al. RT-PCRstudies in patients with chronic myeloid leukemia (CML)in remission 5 years after allogeneic stem cell trans-plant (SCT) define risk of subsequent relapse [ab-stract]. Blood. 2000;96(suppl 1):141a.12. Minot GR, Buckman TE, Isaacs R. Chronic my-elogenous leukemia. JAMA. 1924;82:1489-1494.13. Medical Research Council’s Working Party forTherapeutic Trials in Leukemia. Chronic granulocyticleukemia. BMJ. 1968;1:201-208.14. Haut A, Abbott WS, Wintrobe MM, CartwrightGE. Busulfan in the treatment of chronic myelocyticleukemia. Blood. 1961;17:1-19.15. Hehlman R, Heimpel H, Hasford J, et al. Random-ized comparison of busulfan and hydroxyurea in chronicmyelogenous leukemia. Blood. 1993;82:398-407.16. Kennedy BJ. The evolution of hydroxyurea therapyin chronic myelogenous leukemia. Semin Oncol. 1992;19(suppl):21-26.17. Benelux CML Study Group. Low dose interferon-alpha 2b combined with hydroxyurea versus hydroxy-urea alone for chronic myelogenous leukemia. BoneMarrow Transplant. 1996;17(suppl 3):S19-S20.18. Savage DG, Goldman JM. Approaches to the treat-

ment of chronic myelogenous leukemia. Int J Hema-tol. 1994;60:1-21.19. Kloke O, Niederle N, Qui MY, et al. Impact of in-terferon alpha-induced cytogenetic improvement onsurvival in chronic myelogenous leukaemia. Br J Hae-matol. 1993;83:399-403.20. Allan NC, Richards SM, Shepard PC. UK MedicalResearch Council randomized, multicentre trial of in-terferon-alpha n1 for chronic myeloid leukaemia. Lan-cet. 1995;345:1392-1397.21. KantarjianHM,SmithTL,O’BrienS,etal.Prolongedsurvival following achievement of cytogenetic responsewith alpha-interferon therapy in chronic myelogenousleukemia. Ann Intern Med. 1995;122:254-261.22. Hehlman R, Heimpel H, Hasford J, et al. Ran-domized comparison of interferon-alpha with busul-fan and hydroxyurea in chronic myelogenous leuke-mia. Blood. 1994;84:4064-4077.23. Kennedy BJ. Hydroxyurea therapy in chronic my-elogenous leukemia. Cancer. 1972;29:1052-1056.24. Fefer A, Cheever MA, Thomas ED, et al. Disap-pearance of Ph1-positive cells in four patients withchronic granulocytic leukemia after chemotherapy, ir-radiation and marrow transplantation from an iden-tical twin. N Engl J Med. 1979;300:333-337.25. Clift RA, Buckner CD, Thomas ED, et al. The treat-ment of chronic granulocytic leukaemia in chronicphase by allogeneic marrow transplantation. Lancet.1982;2:621-622.26. Talpaz M, McCredie KB, Mavligit GM, et al. Leuko-cyte interferon-inducedmyeloidcytoreduction inchronicmyelogenous leukemia. Blood. 1983;62:689-692.27. Horowitz MM, Rowlings PA, Passweg JR. Allo-geneic bone marrow transplantation for CML. BoneMarrow Transplant. 1996;17(suppl 3):S5-S6.28. Gratwohl A, Hermans J, Neiderwieser D, et al. Bonemarrow transplantation for chronic myeloid leuke-mia. Bone Marrow Transplant. 1993;12:509-516.29. Clift RA, Applebaum FR, Thomas ED. Treatmentof chronic myeloid leukemia by marrow transplanta-tion. Blood. 1993;82:1954-1956.30. Hughes TP, Morgan GJ, Martiat P, Goldman JM.Detection of residual leukemia after bone marrowtransplant for chronic myeloid leukemia. Blood. 1991;77:874-878.31. Collins RH, Shpilberg O, Drobyski WR, et al. Do-nor leukocyte infusions in 140 patients with relapsedmalignancy after allogeneic bone marrow transplant.J Clin Oncol. 1997;15:433-444.32. Kantarjian HM, Giles FJ, O’Brien S, et al. Thera-peutic choices in younger patients with chronic my-elogenous leukemia. Cancer. 2000;89:1647-1658.33. Tura S. Cytarabine increases karyotypic responsein alpha-IFN treated chronic myeloid leukemia patients[abstract]. Blood. 1998;92(suppl 1):317a.34. Silver RT, Woolf SH, Hehlmann R, et al. An evi-dence-based analysis of the effect of busulfan, hy-

droxyurea, interferon, and allogeneic bone marrowtransplantation in treating the chronic phase of chronicmyeloid leukemia. Blood. 1999;94:1517-1536.35. Guilhot F, Chastang C, Michallet M, et al. Inter-feron alfa-2b combined with cytarabine versus inter-feron alone in chronic myelogenous leukemia. N EnglJ Med. 1997;337:223-229.36. Sokol J, Cox E, Baccarani M, et al. Prognostic dis-crimination in “good risk” chronic granulocytic leu-kemia. Blood. 1984;63:789-799.37. Kantarjian HM, Giles FJ, O’Brien SM, Talpaz M.Clinical course and therapy of chronic myelogenousleukemia with interferon-alpha and chemotherapy. He-matol Oncol Clin North Am. 1998;12:31-80.38. Druker BJ, Tamura S, Buchdunger E, et al. Effects ofaselectiveinhibitoroftheAbltyrosinekinaseonthegrowthof Bcr-Abl positive cells. Nat Med. 1996;2:561-566.39. Schindler T, Bornmann W, Pellicina P, et al. Struc-tural mechanism for STI-571 inhibition of abelson ty-rosine kinase. Science. 2000;289:1938-1942.40. Talpaz M, Silver RT, Druker B, et al. A phase II studyof STI-571 in adult patients with Philadelphia chromo-some–positive chronic myeloid leukemia in acceler-ated phase [abstract]. Blood. 2000;96(suppl 1):469a.41. Druker BJ, Lydon NB. Lessons learned from the de-velopment of an Abl tyrosine kinase inhibitor for chronicmyelogenous leukemia. J Clin Invest. 2000;105:3-7.42. Druker BJ, Talpaz M, Resta DJ, et al. Efficacy andsafety of a specific inhibitor of the BCR-ABL tyrosinekinase in chronic myeloid leukemia. N Engl J Med.2001;344:1031-1037.43. Kantarjian H, Sawyers C, Hochhaus A, et al. PhaseII study of STI-571, a tyrosine kinase inhibitor, in pa-tients (pts) with resistant or refractory Philadelphia chro-mosome-positive chronic myeloid leukemia (Ph+CML)[abstract]. Blood. 2000;96(suppl 1):470a.44. Druker BJ, Sawyers CL, Kantarjian H, et al. Ac-tivity of a specific inhibitor of the Bcr-Abl tyrosine ki-nase in the blast crisis of chronic myeloid leukemia andacute lymphoblastic leukemia with the Philadelphiachromosome. N Engl J Med. 2001;344:1038-1042.45. O’Brien S, Kantarjian H, Keating M, et al. Homo-harringtonine therapy induces responses in patientswith chronic myelogenous leukemia in late chronicphase. Blood. 1995;86:3322-3326.46. Kantarjian HM, O’Brien SM, Keating M, et al. Re-sults of decitabine therapy in the accelerated and blas-tic phases of chronic myelogenous leukemia. Leuke-mia. 1997;11:1617-1620.47. Giles FJ, Cortes E, Bivine C, et al. Troxacitabine,a novel nucleoside analog is active in acute and chronicmyeloid leukemia [abstract]. American Society of Clini-cal Oncology Program/Proceedings. 2000;19:46a.48. Molldrem JJ, Lee PP, Wang C, et al. Evidence thatspecific T lymphocytes may participate in the elimi-nation of chronic myelogenous leukemia. Nat Med.2000;6:1018-1023.

CHRONIC MYELOGENOUS LEUKEMIA RESEARCH AND TREATMENT

898 JAMA, August 22/29, 2001—Vol 286, No. 8 (Reprinted) ©2001 American Medical Association. All rights reserved.

Downloaded From: http://jama.jamanetwork.com/ by a University Of Connecticut Health Center User on 12/03/2013