Recent advances in the treatment and understanding of childhood acute lymphoblastic leukaemia

Recent advances in the treatment and understanding of childhood acute lymphoblastic leukaemia

CANCER TREATMENT REVIEWS 2003; 29: 31–44 doi:10.1016/S0305-7372(02)00106-8 ANTI-TUMOUR TREATMENT Recent advances in the treatment and understanding ...

178KB Sizes 1 Downloads 47 Views

CANCER TREATMENT REVIEWS 2003; 29: 31–44 doi:10.1016/S0305-7372(02)00106-8

ANTI-TUMOUR TREATMENT

Recent advances in the treatment and understanding of childhood acute lymphoblastic leukaemia Jeffrey E. Rubnitz and Ching-Hon Pui Department of Haematology-Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA, Department of Paediatrics, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA Clinical trials have advanced the cure rate of childhood acute lymphoblastic leukaemia to near 80%. Treatment response, as measured by minimal residual disease, has allowed us to refine risk classification schemes and better tailor the intensity of therapy for each patient. More complete molecular analysis of leukaemia cells, pharmacodynamic and pharmacogenetic studies, and the development of targeted therapy should ultimately lead to further improvements in treatment. Pharmacogenetic studies should allow treatment refinements that will decrease the risk of complications while maintaining high cure rates. In addition, gene expression profiling may improve the genetic classification of leukaemia and identify clinically important subgroups. It may also lead to the identification of new targets for novel antileukaemic agents. c 2003 Elsevier Science Ltd. All rights reserved. s Key words: Leukaemia; childhood; minimal residual disease; cytogenetics; pharmacogenetics; therapy.

INTRODUCTION Recent clinical trials have achieved 5-year event-free (EFS) survival rates greater than 70% for patients with childhood acute lymphoblastic leukaemia (ALL) (1–4). Our current efforts to improve outcome include the development of more precise risk classification strategies based on the level of minimal residual disease (MRD), the optimization of therapy through pharmacodynamic and pharmacogenomic studies, and the development of more specific therapies. Here, we summarize some of the recent advances in the treatment and understanding of childhood ALL.

Correspondence to: Jeffrey E. Rubnitz, MD, PhD, Department of Haematology-Oncology, St. Jude Children’s Research Hospital, Mail Stop 260 332 N. Lauderdale Street, Memphis, TN 38105-2794, USA. Tel.: +1-901-495-2388; Fax: +1-901-521-9005; E-mail: jeffrey. [email protected]

GENETIC ALTERATIONS IN LEUKAEMIC CELLS Cytogenetic and molecular analyses of leukaemic blasts have enhanced our understanding of the pathogenesis of ALL and have identified therapeutically important subgroups of disease (5,6). To date, specific genetic abnormalities with clinical relevance have been identified in approximately 75% of childhood ALL cases (Table 1). Gene expression profiling will probably identify additional prognostically important genetic subgroups in the near future (7). Recently, microarray analysis of T-cell ALL identified four major genetic subgroups with prognostic significance: the MLL-ENL fusion gene and expression of HOX11 were associated with a favorable outcome, whereas activation of TAL1 or LYL1 conferred a poor prognosis (8). Additionally, HOX11L2 activation also appears to be associated with a dismal outcome in T-cell ALL (8,9). Although the predictive value of genetic abnormalities is superior to that of clinical features, the

c 2003 ELSEVIER SCIENCE LTD. ALL RIGHTS RESERVED. 0305-7372/03/$ - see front matter s

32

J.E. RUBNITZ AND C.-H. PUI

TA B L E 1 Outcome and frequency of genetic subgroups in childhood acute lymphoblastic leukemia Subgroup

Frequency (%)

5-year event-free survival estimate (%)

B cell (rearranged MYC gene) B precursor Hyperdiploid > 50 TEL-AML1 E2A-PBX1 BCR-ABL MLL-AF4 Hypodiploid < 45 MLL rearranged T cell MLL-ENL HOX11 TAL1 LYL1

2–3

75–85

25 22 5 3 2 1 5

80–90 85–90 75–85 20–40 20–35 25–40 30–50

1 3 6–7 1

85–95 80–90 30–40 30–40

prognostic impact of these abnormalities is dependent on the efficacy of the treatment given. In addition, heterogeneity exists even within genetic subgroups, further limiting their usefulness in predicting outcome. For example, although patients with Philadelphia (Ph) chromosome-positive ALL generally have a poor prognosis, about one third can be cured by chemotherapy alone (10). Among patients with Phþ ALL, those who have a low leukocyte count at initial examination and are between 1 and 9 years of age have a favorable outcome (11). Similarly, the t(4;11) confers a poor prognosis overall, but there is heterogeneity within this genetic subgroup as well (12). Approximately 50% of patients who have t(4;11)-positive disease and are older than 1 year at the time of diagnosis are cured by modern therapy; in contrast, less than 20% of infants with the identical translocation are cured (12). Among patients with hypodiploidy (fewer than 45 chromosomes), the outcome worsens as the chromosome number decreases: patients with 24–28 chromosomes have the worst prognosis (13). Other genetic features, such as hyperdiploidy and the TEL-AML1 fusion gene, confer a good prognosis: EFS estimates exceed 80% (14,15). Within the hyperdiploid group, patients with trisomies 4, 10, and 17 have a superior outcome; this finding again demonstrates the heterogeneity of ALL (14,16,17). In contrast, prognostic indicators have not yet been identified in cases of TEL-AML1-positive ALL. However, the components of treatment probably affect the outcome of TEL-AML1-positive ALL; low relapse rates were observed in some protocols that featured intensified L -asparaginase therapy (18–20) but not in others (21–23). Leukaemic blasts that express TEL-AML1 are more sensitive to L -asparaginase

in vitro (24), and treatment regimens that include prolonged exposure to this agent are extremely effective at treating this type of leukaemia (3). When TEL-AML1-positive ALL relapses, the disease is often still sensitive to chemotherapy (21,23,25). Although patients had a favorable outcome even after relapse (5-year EFS and survival estimates, 63% and 82%, respectively), a matched-pair analysis demonstrated that TEL-AML1 expression was not an independent predictor of outcome after adjustments were made for time to relapse and site of relapse (21). In contrast to leukaemic cells from older children, blasts from infants with ALL are resistant to prednisolone and L -asparaginase in vitro, but are more sensitive to cytarabine (26). On the basis of these findings, high-dose cytarabine has been incorporated into the treatment regimen of a current international trial for infant ALL. Similarly, high-dose methotrexate is an important component of the treatment of hyperdiploid ALL, as leukaemic blasts from these cases are more sensitive to methotrexate in vitro (27) and accumulate higher levels of methotrexate polyglutamates (the active metabolites of methotrexate) (28). Patients with T-cell ALL may benefit from very high-dose methotrexate (14), a result consistent with our finding that T-lineage blasts accumulate methotrexate polyglutamates less avidly than do B-lineage blasts and therefore higher serum concentrations of methotrexate are needed for adequate responses in T-cell ALL (29). In addition to adding prognostic information, the analysis of genetic alterations in leukaemic cells has provided insight into the origins of childhood ALL. Genetic studies of identical twins with leukaemia (30,31) and the detection of leukaemia-specific fusion genes (32) or immunoglobulin gene rearrangements (33) in neonatal blood spots have definitely established the prenatal origin of some cases of leukaemia. With regard to the development of TEL-AML1positive ALL, the Greaves laboratory demonstrated that a pair of monozygotic twins who developed TELAML1-positive ALL shared a unique TEL-AML1 genomic breakpoint. The presence of the unique breakpoint in each child suggested that the leukaemic clone originated in one fetus and was transferred to the other fetus transplacentally (34). Because leukaemia did not develop until one was 3 years and 6 months old and the other was 4 years and 10 months old, the investigators hypothesized that secondary mutations were necessary for the full leukaemic phenotype. This hypothesis was supported by the results of a subsequent study that demonstrated the presence of TEL-AML1 fusion sequences in blood spots of 8 of 11 neonates in whom ALL developed later in childhood; such a finding again suggested that a preleukaemic clone containing the fusion

ADVANCES IN TREATMENT OF ALL

develops in utero (32). Analysis of a second set of twins concordant for TEL-AML1-positive leukaemia but discordant for age at the time of diagnosis revealed identical genomic translocations at diagnosis (31). Furthermore, twin B harbored low levels of the TEL-AML1 gene rearrangement when the leukaemia in twin A was diagnosed (9 years before that of twin B); this finding demonstrates that the putative second event required for leukaemic transformation may occur after a variable and protracted latency. Subsequent analysis of a set of triplets demonstrated that all three children harbored identical genomic TEL-AML1 sequences (35). Interestingly, the two monozygotic twins experienced the onset of ALL at the same time (21 months of age), whereas the third sibling at this time had very low levels of the TELAML1 fusion gene in his bone marrow without an increase in leukaemic blasts. At the time of diagnosis, the leukaemic blasts from monozygotic twins harbored different deletions of the TEL allele that had not been translocated. These patients apparently had undergone distinct second events after birth, further supporting the multihit model of leukemogenesis. Recently, Greaves demonstrated that the TEL-AML1 fusion is present in cord blood of about 1% of randomly selected newborns, a frequency about 100-fold higher than that of TEL-AML1-positive ALL (36). This finding also suggests that preleukaemic clones are generated at high frequencies in utero, but that additional environmentally induced or spontaneous genetic alterations lead to the full leukaemic phenotype in a subset of children.

PHARMACODYNAMICS AND PHARMACOGENOMICS OF ALL Host factors such as pharmacodynamics and pharmacogenomics have a significant effect on treatment outcome in ALL (37–39). There is wide interpatient variability in the pharmacokinetics of common antileukaemic drugs used in the treatment of ALL. Therefore, in some patients, relapse may be attributed to inadequate drug exposure rather than drug resistance of leukaemic cells. Low systemic exposure to methotrexate and low dose-intensity of mercaptopurine have each been associated with inferior outcome (40,41). We have also demonstrated that individualized dosing of methotrexate based on pharmacokinetic parameters leads to an improved outcome for children with B-cell precursor ALL (40). We are therefore applying this treatment strategy by targeting systemic methotrexate exposure during consolidation therapy on our current ALL protocol. Because cytochrome P450 (CYP) enzymes are responsible for the oxidative metabolism of many

33

chemotherapeutic agents, administration of anticonvulsants that induce CYP enzymes can increase the systemic clearance of these chemotherapeutic agents. Patients who received phenobarbital, phenytoin, or carbamazepine had a significantly worse outcome than patients who did not receive anticonvulsants (42). For patients who require anticonvulsants, we now use agents, such as gabapentin, that are less likely to induce drug-metabolizing enzymes. In contrast, azole antifungals (e.g., fluconazole and itraconazole) and the macrolide group of antibiotics (e.g., erythromycin, rifampin, and azithromycin) can inhibit CYP enzymes and thereby decrease clearance and increase the exposure to and the toxicity of certain antileukaemic agents (vincristine and anthracyclines) (43). Polymorphisms of genes encoding proteins involved in drug disposition (drug-metabolizing enzymes, drug transporters, and drug receptors) affect the metabolism and therefore the efficacy and toxicity of antileukaemic agents (37,38). Pharmacogenomics – the study of such genes – uses conventional genetic studies as well as newly developed microarray techniques to identify and characterize these polymorphisms. Pharmacogenomic research may ultimately allow us to select drug and dosage based on each patientÕs ability to metabolize, eliminate, and respond to specific drugs. Some important polymorphisms that are relevant to ALL therapy are shown in Table 2. Thiopurine methyltransferase (TPMT) is an enzyme that converts mercaptopurine, thioguanine, and azathioprine into inactive methylated metabolites; this conversion prevents these drugs from becoming thioguanine nucleotides, which are incorporated into DNA and thus induce an antileukaemic effect. TPMT activity is inherited as an autosomal co-dominant trait. Approximately 90% of the population are homozygous for the wild-type allele and have full enzyme activity, about 10% are heterozygous for the polymorphism and have intermediate levels of enzyme activity, while one in 300 persons carries two mutant TPMT alleles and does not express functional TPMT (44–48). Cells of patients with heterozygous TPMT alleles and intermediate levels of enzyme activity accumulate more thioguanine nucleotides after mercaptopurine or thioguanine therapy than do cells of persons homozygous for the wild-type TPMT alleles. Therefore, patients with intermediate levels of TPMT activity require modest reductions in dosage to prevent excessive myelosuppression (49,50). Moreover, patients with a TMPT deficiency due to homozygous mutant alleles accumulate very high levels of thioguanine nucleotides, and these patients experience life-threatening hematopoietic toxicity if they are treated with standard doses of mercaptopurine, azathioprine, or thioguanine; therefore, the dose must be

34

J.E. RUBNITZ AND C.-H. PUI

TA B L E 2 Genetic polymorphisms of enzymes that affect treatment response Gene product

Polymorphism

Drugs affected by polymorphism

Effect of polymorphism

Thiopurine methyltransferase (TPMT) Glutathione S-transferase (GST)

Single-nucleotide polymorphisms that lead to unstable protein Completion deletions or point mutations

Mercaptopurine, azathioprine, thioguanine Alkylating agents, glucocorticoids, epipodophyllotoxins, anthracyclines

Cytochrome P450 3A4 (CYP3A4)

Single-nucleotide polymorphisms that lead to nonfunctional enzyme Point mutations that lead to protein instability Single-nucleotide polymorphisms that lead to nonfunctional enzyme

Vincristine, anthracyclines, epipodophyllotoxins, glucocorticoids Methotrexate

Myelosuppression, increased risk of therapy-related cancer Increased sensitivity to antileukemic drugs, increased risk of primary or therapy-related leukemia Decreased risk of therapy-related leukemia

Methylenetetrahydrofolate reductase (MTHFR) NAD(P)H:quinone oxidoreductase (NQO1)

reduced to 10–15% of conventional doses. Patients who have ALL and at least one mutant TPMT allele tend to respond better to mercaptopurine therapy and have better outcomes than do those who have two wild-type alleles (41). However, patients with at least one mutant TPMT allele are at an increased risk of epipodophyllotoxin-related acute myeloid leukaemia (AML) (51) or irradiation-induced brain tumors (52) in the context of antimetabolite therapy. In fact, AML has been identified in these patients even when their treatment consisted primarily of antimetabolites (53). We now prospectively identify patients with TPMT deficiency and reduce their dosage of mercaptopurine in an effort to reduce toxicity and improve clinical outcome. Glutathione S-transferases (GSTs) detoxify a wide range of xenobiotics, including cancer chemotherapeutic agents, environmental carcinogens, and reactive oxygen products. GSTs are highly polymorphic: approximately 50% of persons have a homozygous deletion of GSTM1, and 25% have a homozygous deletion of GSTT1 (54). In addition, GSTP1 is subject to polymorphisms: persons with GSTP1 Val105 =Val105 have reduced GST activity (55,56). Deficiency of GST has been associated with lower relapse rates in some studies, possibly because of the reduction in inactivation of cytotoxic chemotherapy (56–59). In contrast, GST genotype did not affect outcome in a recent ChildrenÕs Cancer Group analysis (60). GST deficiency has also been associated with increased toxicity of some anticancer agents, and increased expression of GST has been associated with in vitro drug resistance of cancer cell lines (61). In patients who had AML and received intensive chemotherapy, the GSTT1-null genotype was associated with greater toxicity and with death during remission (62,63). Whether this relationship extends to patients who receive intensive chemotherapy for high-risk ALL or

Alkylating agents, mitomycin C

Increased risk of mucositis, decreased risk of primary ALL Increased risk of infant and childhood ALL, adult AML and therapy-related leukemia; worse outcome of ALL

undergo transplantation for very high-risk ALL remains to be studied. GST polymorphisms have also been implicated in the development of de novo ALL (58,64,65) and therapy-related AML (55,66). Cytochrome P450 (CYP) enzymes, which are localized primarily in the liver and gastrointestinal tract, are responsible for the oxidative metabolism of endogenous steroids, hormones, and drugs (38). These enzymes are involved in activation (e.g., epipodophyllotoxins) or inactivation (e.g., vinca alkaloids) of anticancer drugs or in forming metabolites that are toxic to host tissues (67). The CYP3A family, whose two main forms are CYP3A4 and CYP3A5, is responsible for about half of the drug metabolism carried out by CYP enzymes (61,68). Therefore, patients who inherit functional CYP3A4 and CYP3A5 phenotypes catabolize many antileukaemic agents faster than other patients do (61,68). As mentioned above, the induction of CYP3A by phenytoin or phenobarbital may explain the worse clinical outcome of patients who received these agents for seizure control (42), whereas azole antifungal agents and the macrolide group of antibiotics inhibit CYPs and cause increased toxicity (43). Regarding polymorphisms in the CYP genes, patients carrying the CYP3A4*1B variant allele are at a decreased risk of therapy-related AML (69). In addition, a recent study showed that a variant of CYP1A1 is associated with a worse outcome in childhood ALL, especially in patients with MLH1 Ile219 , a variant of a DNA repair enzyme (70). The CYP2E1*5 (71) and the CYP1A1*2A (64) alleles are associated with a greater risk of de novo ALL. Methylenetetrahydrofolate reductase (MTHFR) catalyzes the reduction of 5,10-methylenetetrahydrofolate to 5-methyltetrahydrofolate, thereby maintaining normal concentrations of reduced folates and homocysteine. Two polymorphisms associated with

ADVANCES IN TREATMENT OF ALL

reduced MTHFR activity have been described: substitution of thymine for cytosine at nucleotide 677 (677C > T; C ! T; alanine ! valine) (72,73) and substitution of cytosine for adenine at nucleotide 1298 (1298A > C; A ! C; glutamine ! alanine) (74). Approximately 10% of the population are homozygous for the 677T variant, which encodes an enzyme with approximately 30% of the wild-type activity, and 40% have a heterozygous genotype with enzyme activity that is about 60% of wild-type. Because the active polyglutamate metabolites of methotrexate can inhibit MTHFR, patients with reduced MTHFR activity, such as that caused by the 677C > T genotype, are at an increased risk of methotrexate-related toxicity, including oral mucositis (75,76). This increased risk of toxicity after low-dose methotrexate therapy has only been reported for patients with the MTHFR 677C > T allele; this risk after high-dose methotrexate therapy and leucovorin rescue has not been reported (76). We believe that leucovorin rescue after high-dose methotrexate may attenuate the increased risk of toxicity by providing an exogenous source of reduced folates that compensates for the low folate concentrations in these patients. Recent findings have suggested that folate plays a role in the development of childhood ALL (77) and that the MTHFR polymorphism protects against the development of ALL (78–80). In one study, MTHFR polymorphism was associated with genetically defined subgroups of childhood ALL: a low number of carriers of the 677C > T allele were present among patients with MLL-rearranged leukaemia, and a lower number of patients with the 1298A > C allele were observed in the hyperdiploid ALL subgroup (80). This seemingly paradoxical protective effect may be caused by the increased fidelity of DNA synthesis afforded by the greater availability of the 5,10-methylenetetrahydrofolate (80). NAD(P)H:quinone oxidoreductase (NQO1) is an enzyme that converts toxic benzoquinones to hydroxyl metabolites. Two polymorphisms have been identified: NQO1*2, which results in the absence of enzyme function, and NQO1*3, which causes diminished enzyme function (71). The function of NQO1 suggests that patients with decreased or no enzyme activity are more susceptible to toxic and carcinogenic effects of benzene and certain chemotherapeutic drugs. As predicted, a higher prevalence of the inactivating polymorphisms has been associated with infant or childhood ALL (71,81), adult AML (82), and therapy-related leukaemia (83,84). In one study of childhood ALL, patients with the NQO1*2 variant had a worse treatment outcome. The authors speculated that impaired protection of cells from free radicals contributes to recurrent leukaemia in patients with this variant (70).

35

Polymorphisms of drug targets or drug transporters may also play a role in response to chemotherapy and susceptibility to leukaemia. In this regard, a recent study showed that a triple tandem polymorphism in the promoter of thymidylate synthase, an important target of methotrexate, was associated with a worse outcome in children with ALL (85). Polymorphisms of thymidylate synthase and another folate-metabolizing enzyme (serine hydroxymethyltransferase) are associated with a lower risk of adult ALL (86). Membrane transporters are involved in drug absorption, distribution into tissues, and efflux from cells. The ATP-binding cassette (ABC) family of membrane transporters comprising P-glycoprotein (PGP) and the multidrug resistanceassociated proteins (MRPs) has been recently reviewed (87). Although overexpression of ABC transporters has been associated with resistance to a broad spectrum of antileukaemia agents in cell lines, the clinical relevance of such overexpression in patients with ALL is unknown (61). Similarly, genetic polymorphisms occur in the human ABC transporters (61,87), but the clinical relevance of the polymorphisms is also unclear.

MINIMAL RESIDUAL DISEASE IN ALL Most current trials for childhood ALL attempt to tailor the intensity of therapy to the risk of relapse (6). Risk classification schemes are based on clinical features (e.g., age and leukocyte count at diagnosis), biological features (e.g., phenotype, karyotype, and molecular abnormalities), and response to therapy. Early response to therapy, which is influenced by the sensitivity of the leukaemic cells to chemotherapy as well as host pharmacodynamic and pharmacogenomic characteristics, is probably the best predictor of outcome. In fact, simple morphologic examination of the peripheral blood after 1 week of therapy is a relatively good indicator of ultimate outcome (88,89). Even within high-risk subgroups such as Phþ ALL and infant ALL, measurement of circulating blasts after 1 week of prednisone therapy is prognostically important (90,91). Similarly, morphologic examination of the bone marrow after 1 to 3 weeks of induction therapy is an independent predictor of outcome (92–94). Recently, we demonstrated that the presence of any blasts in the bone marrow after 3 weeks of induction therapy portends a particularly dismal outcome (95). In contrast to morphologic examination, which tends to be subjective and imprecise, minimal residual disease (MRD) assays provide objective and sensitive measurements of low levels of leukaemic cells (96–98). MRD may be assessed by DNA-based

36

polymerase chain reaction (PCR) analysis of clonal antigen receptor gene rearrangements, RNA-based PCR analysis of leukaemia-specific gene fusions, or flow cytometric detection of aberrant immunophenotypes (96–98). Through a series of studies using immunologic detection of leukaemic cells, we have clearly demonstrated the clinical importance of MRD measurement in the treatment of ALL (99– 102). Patients who attained MRD negativity (defined as less than 1 leukaemic cell among 10,000 mononuclear bone marrow cells) had a significantly lower relapse rate than patients who did not achieve this status (99). Sequential monitoring of MRD further improved the predictive value: persistence of MRD at week 14 of continuation therapy was associated with a 70% cumulative risk of relapse (100). We also demonstrated that approximately one half of patients achieved MRD negativity at day 19 of induction and had an exceptionally good prognosis, with a cumulative risk of relapse of only 6% (101). More recently, we determined that peripheral blood, rather than bone marrow, may be used to monitor MRD in patients with T-cell ALL (102). In addition, application of both flow cytometry and PCR testing has allowed us to successfully study 100% of patients with newly diagnosed disease (103). Although a complete review of all MRD studies in ALL is beyond the scope of this article, it should be mentioned that many other investigators have also demonstrated the prognostic importance of MRD in the context of various chemotherapeutic regimens (98). One recent study suggested that the absolute number of leukaemic cells per milliliter of bone marrow may be even more accurate at predicting outcome than relative measurements are (104). We are now using MRD quantitation to tailor the intensity of therapy we deliver to patients treated for ALL on our current clinical trial. We hope that early intensification of therapy will improve the outcome of patients for whom MRD measurements indicate a slow response early in treatment.

RECENT ADVANCES IN THE TREATMENT OF ALL Treatment groups Characterized by MLL gene rearrangements, unique gene expression profiles, and a poor prognosis, infant ALL is usually considered a separate subgroup of childhood ALL (105,106). Leukaemic blasts from infants with ALL are more resistant in vitro to prednisolone and L -asparaginase but are more sensitive to cytarabine than blasts from older children;

J.E. RUBNITZ AND C.-H. PUI

this finding suggests that infants may benefit from treatment with this agent (26). The use of cytarabine in infant ALL is also supported by the excellent results of a Dana–Farber Cancer Institute (DFCI) trial that featured high-dose cytarabine as postremission intensification therapy (107). However, the results of a recent trial in the United Kingdom (Infant 92) were not as encouraging: the 4-year EFS estimate was only 33% (108). Infants with MLL rearrangements who were treated on the Japan Infant Leukaemia Study MLL96 had a remarkably similar outcome, i.e., a 3year EFS estimate of 34% (109). Currently, two large prospective studies of infant ALL are testing the efficacy of additional intensification of high-dose cytarabine and high-dose methotrexate therapy. Although the high relapse rates associated with infant ALL imply that further intensification is needed, several studies have suggested that allogeneic stem cell transplantation does not improve the outcome of these patients (12,108). In fact, among patients with the t(4;11), any type of transplantation was associated with worse disease-free and overall survivals (12). ALL patients older than 1 year of age are generally divided into three categories based on the patientsÕ risk of relapse, although some investigators have defined four risk groups (6,14). Patients who have an excellent prognosis and are sometimes considered to have low-risk disease are those with B-lineage ALL, between 1 and 9 years of age, and have initial leukocyte counts less than 50  109 =L, and either the TEL-AML1 fusion or trisomies 4, 10, and 17. Patients with standard-risk disease have B-lineage a favorable age and leukocyte count, but lack TEL-AML1 or the trisomies. Other patients with B-lineage ALL and those with T-cell ALL (except those whose leukaemic cells overexpress HOX11) are at high risk of relapse and are therefore treated with more intensive chemotherapy regimens. Finally, the very high-risk group consists of a small subset of patients, primarily those whose disease fails to enter remission after standard induction therapy and those with Phþ ALL; these patients are candidates for allogeneic stem cell transplantation (SCT) during first remission (11). Although other features such as male sex (110,111) and black or Hispanic race (112) are also associated with an inferior outcome, these characteristics are generally not included in risk classification schemes; however, some investigators recommend extending continuation treatment for boys.

Remission induction therapy Some remission induction regimens include only three drugs for low-risk patients – a glucocorticoid,

ADVANCES IN TREATMENT OF ALL

vincristine, and L -asparaginase – and reserve the addition of an anthracycline for patients with standard and high-risk disease. Other regimens, however, use four to seven drugs for all patients, in an attempt to induce a faster and deeper reduction in leukaemic cell burden, thereby preventing the emergence of drug resistance. These intensive remission induction regimens appear to have improved the outcome of patients older than 10 years and those with high-risk ALL (17,113). However, intensive induction therapy may not be necessary for patients with low or standard-risk disease, as long as they receive intensification therapy after their disease enters remission (17). In addition, L -asparaginase may not be a necessary component of induction therapy; several trials that included this agent only in the postremission setting achieved excellent remission induction rates and high probability of long-term survival (114,115). The absence of L asparaginase was also associated with fewer thrombotic complications during remission induction therapy. In contrast, glucocorticoids are an essential constituent of induction therapy, although the optimal steroid is unknown. Dexamethasone may provide better leukaemic control and CNS penetration than prednisone (17,116), but in one study dexamethasone was associated with unacceptable toxicity when used in induction therapy (117).

Intensification therapy Clinical trials performed by the ChildrenÕs Cancer Group (CCG) and the Berlin–Frankfurt–Muenster (BFM) study group demonstrated that delayed intensification (also called reinduction) therapy improved the outcome of patients with standard or high-risk ALL (17,113). Additional intensification, designated augmented BFM or double-delayed intensification by the CCG, improved outcome even further (118,119). While this intensification therapy improved the outcome of children younger than 10 years with high-risk ALL and a slow early-response to therapy, it was associated with a high incidence of avascular necrosis of the bone (118). Recently, the results of the AIEOP ALL95 study demonstrated that the addition of two courses of Protocol II improved the outcome of patients who had poor responses to prednisone (120). In contrast, additional blocks of intensification failed to alter EFS estimates on a recent trial in the United Kingdom, possibly because of concomitant decreases in other elements of therapy and in a lack of sustained, or metronomic, therapy (121). Whereas delayed intensification therapy is generally given about 3 months after induction therapy

37

is completed and includes the same or similar drugs as those used in induction therapy, consolidation therapy includes high-dose methotrexate and mercaptopurine given at the time remission is achieved (5,6). Although the optimal dose of methotrexate is still a subject of investigation, 2.5 g=m2 is probably adequate for patients with B-lineage ALL, and 5 g=m2 for T-cell ALL (6,14,113). In addition, we previously showed that dosing based on plasma concentrations of methotrexate rather than simply on body surface area is beneficial for B-lineage cases (40). A third form of intensification therapy consists of the prolonged use of intensive L -asparaginase therapy to continuously deplete serum of asparagine. A Paediatric Oncology (POG) Group study demonstrated that this strategy improved the outcome of patients with T-cell ALL; however, the regimen used in that study also resulted in a higher incidence of treatment-related AML due to the concomitant use of etoposide (122). The prolonged use of L -asparaginase and doxorubicin in DFCI protocols has contributed to excellent overall outcome, especially for patients with TEL-AML1-positive B-lineage ALL and for patients with T-cell ALL (3,115,123). In the most recent DFCI trial, patients who received at least 26 weekly doses of L -asparaginase had a better outcome than those who did not (3). However, the DFCI trial was also associated with subclinical cardiomyopathy, which was not prevented by the use of continuous infusion, rather than bolus, administration of doxorubicin (124). Finally, allogeneic SCT represents the most intensive form of intensification. Patients with Phþ ALL are clearly candidates for allogeneic SCT during first remission; an international collaborative study demonstrated the outcome of this group of patients who received transplants of stem cells from an HLA-matched related donor is superior that of the same group who received only intensive chemotherapy (11). In contrast, allogeneic SCT does not appear to improve the outcome of patients with t(4;11)-positive ALL (12) or of infants with ALL (108).

Continuation therapy Children with B-precursor or T-cell ALL require continuous postremission therapy for optimal success (6,17,113,115). Although effective for other malignancies, high-dose pulse therapy with intermittent rest periods is associated with an inferior outcome in ALL (6,113). In addition, continuation therapy should be prolonged; attempts to shorten therapy to 12 or 18 months have resulted in inferior overall outcomes

38

(113,125). Although a subset of patients may be cured with a shorter duration of therapy, most clinical trials currently prescribe at least 2–2.5 years of therapy for all patients. Furthermore, because the outcome for boys is generally inferior to that of girls (110,111), several studies, including the current St. Jude trial, are examining the benefit of extending continuation therapy to 3 years for boys. Weekly low-dose methotrexate and daily oral mercaptopurine make up the backbone of most continuation regimens for ALL. Although improved outcome has been associated with increasing mercaptopurine dosages to the limits of individual tolerance (as indicated by neutrophil counts) (126), one must not to be too aggressive. The use of mercaptopurine at dosages that cause neutropenia has been associated with increased rates of infection, an inability to deliver all planned therapy, and a higher relapse rate (41,127). As discussed above, the dosage of mercaptopurine may need to be decreased in patients with TPMT deficiency to decrease acute toxicity and the risk of secondary cancers (45,50– 53,128). We now prospectively decrease the mercaptopurine dose slightly in patients with TPMT deficiency to determine whether this approach will improve outcome. The addition of intermittent pulses of steroids and vincristine to the antimetabolite backbone improves outcome and is included in most modern continuation regimens (17,129). Because of better antileukaemic effect, dexamethasone has replaced prednisone as the steroid of choice, but dexamethasone is associated with increased risks of avascular necrosis, osteoporosis, and fractures (130–132).

Therapy for central nervous system leukaemia The treatment of subclinical or overt central nervous system (CNS) leukaemia has made a tremendous impact on the overall cure rates for childhood ALL (133). Certain patients, including those with high leukocytes counts at diagnosis, T-cell disease, or leukaemic blasts in the cerebrospinal fluid (CSF), are at higher risk of CNS relapse and therefore require more intensive therapy directed at the CNS (134). CNS-directed therapy includes cranial irradiation, intrathecal chemotherapy, and systemic chemotherapy with dexamethasone and high-dose methotrexate (1,17,135). Although cranial irradiation is the most effective modality, it is associated with significant morbidity, including neurotoxic effects, endocrinopathy, and second malignant neoplasms (5,6,52,136,137). Therefore, intrathecal therapy has largely replaced cranial irradiation for all patients except those at very high risk of CNS relapse (2,138).

J.E. RUBNITZ AND C.-H. PUI

In fact, three studies omitted cranial irradiation in all patients regardless of their risk classification and still achieved CNS relapse rates of less than 10% (4,139,140). For patients at high risk of CNS relapse, the BFM study group demonstrated that a radiation dose of 12 Gy, when given with effective intrathecal and systemic chemotherapy, provided adequate CNS control (113). Recently, we have demonstrated that for patients in whom growth hormone deficiency occurs as a result of CNS-directed therapy, growth hormone replacement appears to be safe and effective (141). An area of active discussion remains the optimal treatment of patients with low numbers of identifiable leukaemic blasts in the CSF (i.e., CNS2 status, defined as leukaemic blasts present on cytospin, but fewer than 5 leukocytes per microliter of CSF) (142). Previously, we demonstrated that CNS2 status conferred an adverse prognosis among patients treated on St. Jude Total Therapy Studies XI and XII (143), a finding confirmed by the POG (144). However, three other studies did not show an association between CNS2 status and outcome (145–147). This apparent discrepancy may be explained by differences in treatment regimens, including early CNS-directed therapy on the CCG and BFM studies (145,147) that was more intensive than that on Studies XI and XII (143). We intensified intrathecal therapy for CNS2 patients during remission induction and postremission therapy on our subsequent trials Studies XIIIA (1) and XIIIB and have reduced the cumulative incidence of any CNS relapse in CNS2 patients to less than 5% (unpublished data). To this end, recent CCG studies of patients of all risk groups demonstrated that CNS2 patients in the cohort with presenting leukocyte count > 50  109 =L or age > 10 years had an increased risk for both haematological and CNS relapse, as compared to the CNS1 patients (148). Hence, CNS2 patients should at least receive more intensive CNS-directed therapy. A distinct category of CNS status at diagnosis is traumatic lumbar puncture with identifiable blasts. We previously demonstrated an inferior outcome for this group of patients (134), a finding recently confirmed by the BFM study group (147). Possible explanations for this observation include the iatrogenic introduction of blasts into the CSF and decreased efficacy of subsequent intrathecal treatment after a traumatic lumbar puncture (134,142,147). Although intensive intrathecal therapy can abolish the poor prognostic impact of a traumatic lumbar puncture with blasts, it can also adversely affect neuropsychologic and spinal cord function (149–151). To reduce the frequency of traumatic lumbar puncture, we now routinely perform the diagnostic procedure under deep sedation or general anesthesia, and intrathecal chemotherapy is always administered immediately

ADVANCES IN TREATMENT OF ALL

after the collection of CSF. In addition, the most experienced clinicians perform diagnostic lumbar punctures at our institution; this decision is based on our observation that a less-experienced clinician is a risk factor for traumatic lumbar puncture (152). Because a low platelet count (<100  109 =L) is also a risk factor, we now transfuse platelets into all thrombocytopenic patients before they undergo initial puncture. Together, these preventive measures have lowered the frequency of traumatic lumbar puncture with or without blasts at diagnosis to 4% each in our current trial. We previously reported data indicating that lumbar puncture is generally safe in patients with thrombocytopenia (153). Therefore, we still contend that platelet transfusion is not necessary with subsequent lumbar punctures when patients lack circulating blasts, especially during remission induction therapy that includes prednisone, vincristine and L asparaginase, which generally induce a hypercoagulable state (154).

FUTURE DIRECTIONS Current studies of ABC transporters and other potential mechanisms of drug resistance will hopefully lead to therapeutic methods to overcome leukaemic cell resistance to conventional chemotherapy (61,87,155–157). More promising, however, is the development of novel therapies, particularly those targeted to specific abnormalities in the leukaemic cell. Imatinib mesylate (Gleevec), a selective inhibitor of the BCR-ABL tyrosine kinase, induces durable responses in patients with chronic-phase chronic myelogenous leukaemia (CML) (158,159). The inhibitor also has activity in accelerated-phase or blastcrisis CML, although the results are not as long-lived (160–162). Similarly, a recent phase II study demonstrated that imatinib induced complete bone marrow responses in about one third of patients with relapsed or refractory Phþ ALL, but resistant disease developed rapidly (163). Other kinase inhibitors under development include selective inhibitors of FLT3, which is mutated or overexpressed in subsets of ALL and AML (164–166). Gene expression profiling, in which the expression of thousands of genes is analyzed, has the potential to more accurately predict relapse and the development of secondary cancers (7,106). This method may therefore allow the construction of more precise risk classification schemes and the tailoring of the intensity of therapy to a greater extent than is currently possible. More important, gene expression profiling may identify new targets against which novel and specific therapies may be developed.

39

ACKNOWLEDGEMENTS We thank Julia Cay Jones for expert editorial review. This work was supported in part by the Cancer Center Support Grant CA21765 from the National Cancer Institute, a Center of Excellence grant from the State of Tennessee, and by the American Lebanese Syrian Associated Charities (ALSAC). C.-H. Pui is the American Cancer Society – F.M. Kirby Clinical Research Professor.

REFERENCES 1. Pui CH, Mahmoud HH, Rivera GK, Hancock ML, Sandlund JT, Behm FG, et al. Early intensification of intrathecal chemotherapy virtually eliminates central nervous system relapse in children with acute lymphoblastic leukaemia. Blood 1998; 92: 411–415. 2. Schrappe M, Reiter A, Ludwig WD, Harbott J, Zimmermann M, Hiddemann W, et al. Improved outcome in childhood acute lymphoblastic leukaemia despite reduced use of anthracyclines and cranial radiotherapy: results of trial ALL-BFM 90 German–Austrian–Swiss ALL-BFM Study Group. Blood 2000; 95: 3310–3322. 3. Silverman LB, Gelber RD, Dalton VK, Asselin BL, Barr RD, Clavell LA, et al. Improved outcome for children with acute lymphoblastic leukaemia: results of Dana–Farber consortium protocol 91-01. Blood 2001; 97: 1211–1218. 4. Kamps WA, Bokkerink JP, Hakvoort-Cammel FG, Veerman AJ, Weening RS, Van Wering ER, et al. BFM-oriented treatment for children with acute lymphoblastic leukaemia without cranial irradiation and treatment reduction for standard risk patients: results of DCLSG protocol ALL-8 (1991–1996). Leukaemia 2002; 16: 1099–1111. 5. Pui C-H, Evans WE. Acute lymphoblastic leukaemia. N Engl J Med 1998; 339: 605–615. 6. Pui C-H, Campana D, Evans WE. Childhood acute lymphoblastic leukaemia – current status and future perspectives. Lancet Oncol 2001; 2: 597–607. 7. Yeoh E-J, Ross ME, Shurtleff SA, Williams WK, Patel D, Mahfouz R, et al. Classification, subtype discovery, and prediction of outcome in paediatric acute lymphoblastic leukaemia by gene expression profiling. Cancer Cell 2002; 1: 133–143. 8. Ferrando AA, Neuberg DS, Staunton J, Loh ML, Huard C, Raimondi SC, et al. Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukaemia. Cancer Cell 2002; 1: 75–87. 9. Ballerini P, Blaise A, Busson-Le Coniat M, Su XY, ZucmanRossi J, Adam M, et al. HOX11L2 expression defines a clinical subtype of paediatric T-ALL associated with poor prognosis. Blood 2002; 100: 991–997. 10. Ribeiro RC, Broniscer A, Rivera GK, Hancock ML, Raimondi SC, Sandlund JT, et al. Philadelphia chromosome-positive acute lymphoblastic leukaemia in children: durable responses to chemotherapy associated with low inital white blood cell counts. Leukaemia 1997; 11: 1493–1496. 11. Arico M, Valsecchi MG, Camitta B, Schrappe M, Chessells J, Baruchel A, et al. Outcome of treatment in children with Philadelphia chromosome-positive acute lymphoblastic leukaemia. N Engl J Med 2000; 342: 998–1006. 12. Pui CH, Gaynon PS, Boyett JM, Chessells JM, Baruchel A, Kamps W, et al. Outcome of treatment in childhood acute

40

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

27.

J.E. RUBNITZ AND C.-H. PUI lymphoblastic leukaemia with rearrangements of the 11q23 chromosomal region. Lancet 2002; 359: 1909–1915. Heerema NA, Nachman JB, Sather HN, Sensel MG, Lee MK, Hutchinson R, et al. Hypodiploidy with less than 45 chromosomes confers adverse risk in childhood acute lymphoblastic leukaemia: a report from the childrenÕs cancer group. Blood 1999; 94: 4036–4045. Pui CH, Sallan S, Relling MV, Masera G, Evans WE. International childhood acute lymphoblastic leukaemia workshop: Sausalito, CA, 30 November-1 December 2000. Leukaemia 2001; 15: 707–715. Rubnitz JE, Downing JR, Pui CH, Shurtleff SA, Raimondi SC, Evans WE, et al. TEL gene rearrangement in acute lymphoblastic leukaemia: a new genetic marker with prognostic significance. J Clin Oncol 1997; 15: 1150–1157. Maloney KW, Shuster JJ, Murphy S, Pullen J, Camitta BA. Long-term results of treatment studies for childhood acute lymphoblastic leukaemia: paediatric oncology group studies from 1986–1994. Leukaemia 2000; 14: 2276–2285. Gaynon PS, Trigg ME, Heerema NA, Sensel MG, Sather HN, Hammond GD, et al. Children’s cancer group trials in childhood acute lymphoblastic leukaemia: 1983–1995. Leukaemia 2000; 14: 2223–2233. Loh ML, Silverman LB, Young ML, Neuberg D, Golub TR, Sallan SE, et al. Incidence of TEL/AML1 fusion in children with relapsed acute lymphoblastic leukaemia. Blood 1998; 92: 4792–4797. Rubnitz JE, Behm FG, Wichlan D, Ryan C, Sandlund JT, Ribeiro RC, et al. Low frequency of TEL-AML1 in relapsed acute lymphoblastic leukaemia supports a favorable prognosis for this genetic subgroup. Leukaemia 1999; 13: 19– 21. Zuna J, Hrusak O, Kalinova M, Muzikova K, Stary J, Trka J. TEL/AML1 positivity in childhood ALL: average or better prognosis?. Leukaemia 1999; 13: 22–24. Seeger K, Stackelberg AV, Taube T, Buchwald D, Korner G, Suttorp M, et al. Relapse of TEL-AML1–positive acute lymphoblastic leukaemia in childhood: a matched-pair analysis. J Clin Oncol 2001; 19: 3188–3193. Harbott J, Viehmann S, Borkhardt A, Henze G, Lampert F. Incidence of TEL/AML1 fusion gene analyzed consecutively in children with acute lymphoblastic leukaemia in relapse. Blood 1997; 90: 4933–4937. Seeger K, Adams HP, Buchwald D, Beyermann B, Kremens B, Niemeyer C, et al. TEL-AML1 fusion transcript in relapsed childhood acute lymphoblastic leukaemia. The Berlin– Frankfurt–Muenster study group. Blood 1998; 91: 1716–1722. Ramakers-Van Woerden NL, Pieters R, Loonen AH, Hubeek I, van Drunen E, Beverloo HB, et al. TEL/AML1 gene fusion is related to in vitro drug sensitivity for L -asparaginase in childhood acute lymphoblastic leukaemia. Blood 2000; 96: 1094–1099. Seeger K, Buchwald D, Taube T, Peter A, von Stackelberg A, Schmitt G, et al. TEL-AML1 positivity in relapsed B cell precursor acute lymphoblastic leukaemia in childhood. Berlin–Frankfurt–Muenster study group [letter]. Leukaemia 1999; 13: 1469–1470. Pieters R, den Boer ML, Durian M, Janka G, Schmiegelow K, Kaspers GJL, et al. Relation between age, immunophenotype and in vitro drug resistance in 395 children with acute lymphoblastic leukaemia – implications for treatment of infants. Leukaemia 1998; 12: 1344–1348. Kaspers GJ, Smets LA, Pieters R, Van Zantwijk CH, Van Wering ER, Veerman AJ. Favorable prognosis of hyperdiploid common acute lymphoblastic leukaemia may be explained by sensitivity to antimetabolites and other drugs: results of an in vitro study. Blood 1995; 85: 751–756.

28. Synold TW, Relling MV, Boyett JM, Rivera GK, Sandlund JT, Mahmoud H, et al. Blast cell methotrexate-polyglutamate accumulation in vivo differs by lineage, ploidy, and methotrexate dose in acute lymphoblastic leukaemia. J Clin Invest 1994; 94: 1996–2001. 29. Masson E, Relling MV, Synold TW, Liu Q, Schuetz JD, Sandlund JT, et al. Accumulation of methotrexate polyglutamates in lymphoblasts is a determinant of antileukaemic effects in vivo. A rationale for high-dose methotrexate. J Clin Invest 1996; 97: 73–80. 30. Ford AM, Pombo-de-Oliveira MS, McCarthy KP, MacLean JM, Carrico KC, Vincent RF, et al. Monoclonal origin of concordant T-cell malignancy in identical twins. Blood 1997; 89: 281–285. 31. Wiemels JL, Ford AM, Van Wering ER, Postma A, Greaves M. Protracted and variable latency of acute lymphoblastic leukaemia after TEL-AML1 gene fusion in utero. Blood 1999; 94: 1057–1062. 32. Wiemels JL, Cazzaniga G, Daniotti M, Eden OB, Addison GM, Masera G, et al. Prenatal origin of acute lymphoblastic leukaemia in children. Lancet 1999; 354: 1499–1503. 33. Taub JW, Konrad MA, Ge Y, Naber JM, Scott JS, Matherly LH, et al. High frequency of leukaemic clones in newborn screening blood samples of children with B-precursor acute lymphoblastic leukaemia. Blood 2002; 99: 2992–2996. 34. Ford AM, Bennett CA, Price CM, Bruin MC, Van Wering ER, Greaves M. Fetal origins of the TEL-AML1 fusion gene in identical twins with leukaemia. Proc Natl Acad Sci USA 1998; 95: 4584–4588. 35. Maia AT, Ford AM, Jalali GR, Harrison CJ, Taylor GM, Eden OB, et al. Molecular tracking of leukemogenesis in a triplet pregnancy. Blood 2001; 98: 478–482. 36. Mori H, Colman SM, Xiao Z, Ford AM, Healy LE, Donaldson C, et al. Chromosome translocations and covert leukaemic clones are generated during normal fetal development. Proc Natl Acad Sci USA 2002; 99: 8242–8247. 37. Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science 1999; 286: 487–491. 38. Relling MV, Dervieux T. Pharmacogenetics and cancer therapy. Nat Rev Cancer 2001; 1: 99–108. 39. Pui C-H, Relling MV, Evans WE. Role of pharmacodynamics and pharmacogenomics in the treatment of acute lymphoblastic leukaemia. Bailliere’s Best Practices Res Clin Haematol 2002; in press. 40. Evans WE, Relling MV, Rodman JH, Crom WR, Boyett JM, Pui CH. Conventional compared with individualized chemotherapy for childhood acute lymphoblastic leukaemia. N Engl J Med 1998; 338: 499–505. 41. Relling MV, Hancock ML, Boyett JM, Pui CH, Evans WE. Prognostic importance of 6-mercaptopurine dose intensity in acute lymphoblastic leukaemia. Blood 1999; 93: 2817– 2823. 42. Relling MV, Pui CH, Sandlund JT, Rivera GK, Hancock ML, Boyett JM, et al. Adverse effect of anticonvulsants on efficacy of chemotherapy for acute lymphoblastic leukaemia. Lancet 2000; 356: 285–290. 43. Gibbs MA, Thummel KE, Shen DD, Kunze KL. Inhibition of cytochrome P-450 3A (CYP3A) in human intestinal and liver microsomes: comparison of Ki values and impact of CYP3A5 expression. Drug Metab Dispos 1999; 27: 180–187. 44. Yates CR, Krynetski EY, Loennechen T, Fessing MY, Tai HL, Pui CH, et al. Molecular diagnosis of thiopurine Smethyltransferase deficiency: genetic basis for azathioprine and mercaptopurine intolerance. Ann Intern Med 1997; 126: 608–614. 45. McLeod HL, Krynetski EY, Relling MV, Evans WE. Genetic polymorphism of thiopurine methyltransferase and its

ADVANCES IN TREATMENT OF ALL

46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

clinical relevance for childhood acute lymphoblastic leukaemia. Leukaemia 2000; 14: 567–572. Tai HL, Krynetski EY, Schuetz EG, Yanishevski Y, Evans WE. Enhanced proteolysis of thiopurine S-methyltransferase (TPMT) encoded by mutant alleles in humans (TPMT*3A, TPMT*2): mechanisms for the genetic polymorphism of TPMT activity. Proc Natl Acad Sci USA 1997; 94: 6444–6449. McLeod HL, Lin JS, Scott EP, Pui CH, Evans WE. Thiopurine methyltransferase activity in American white subjects and black subjects. Clin Pharmacol Ther 1994; 55: 15–20. Krynetski EY, Evans WE. Genetic polymorphism of thiopurine S-methyltransferase: molecular mechanisms and clinical importance. Pharmacology 2000; 61: 136–146. Relling MV, Hancock ML, Rivera GK, Sandlund JT, Ribeiro RC, Krynetski EY, et al. Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S-methyltransferase gene locus. J Natl Cancer Inst 1999; 91: 2001–2008. Evans WE, Hon YY, Bomgaars L, Coutre S, Holdsworth M, Janco R, et al. Preponderance of thiopurine S-methyltransferase deficiency and heterozygosity among patients intolerant to mercaptopurine or azathioprine. J Clin Oncol 2001; 19: 2293–2301. Relling MV, Yanishevski Y, Nemec J, Evans WE, Boyett JM, Behm FG, et al. Etoposide and antimetabolite pharmacology in patients who develop secondary acute myeloid leukaemia. Leukaemia 1998; 12: 346–352. Relling MV, Rubnitz JE, Rivera GK, Boyett JM, Hancock ML, Felix CA, et al. High incidence of secondary brain tumours after radiotherapy and antimetabolites. Lancet 1999; 354: 34– 39. Bo J, Schroder H, Kristinsson J, Madsen B, Szumlanski C, Weinshilboum R, et al. Possible carcinogenic effect of 6mercaptopurine on bone marrow stem cells: relation to thiopurine metabolism. Cancer 1999; 86: 1080–1086. Chen CL, Liu Q, Relling MV. Simultaneous characterization of glutathione S-transferase M1 and T1 polymorphisms by polymerase chain reaction in American whites and blacks. Pharmacogenetics 1996; 6: 187–191. Allan JM, Wild CP, Rollinson S, Willett EV, Moorman AV, Dovey GJ, et al. Polymorphism in glutathione S-transferase P1 is associated with susceptibility to chemotherapy-induced leukaemia. Proc Natl Acad Sci USA 2001; 98: 11592–11597. Stanulla M, Schrappe M, Brechlin AM, Zimmermann M, Welte K. Polymorphisms within glutathione S-transferase genes (GSTM1, GSTT1, GSTP1) and risk of relapse in childhood B-cell precursor acute lymphoblastic leukaemia: a case-control study. Blood 2000; 95: 1222–1228. Hall AG, Autzen P, Cattan AR, Malcolm AJ, Cole M, Kernahan J, et al. Expression of mu class glutathione Stransferase correlates with event-free survival in childhood acute lymphoblastic leukaemia. Cancer Res 1994; 54: 5251– 5254. Chen C-L, Liu Q, Pui C-H, Rivera GK, Sandlund JT, Ribeiro R, et al. Higher frequency of glutathione S-transferase deletions in black children with acute lymphoblastic leukaemia. Blood 1997; 89: 1701–1707. Anderer G, Schrappe M, Brechlin AM, Lauten M, Muti P, Welte K, et al. Polymorphisms within glutathione S-transferase genes and initial response to glucocorticoids in childhood acute lymphoblastic leukaemia. Pharmacogenetics 2000; 10: 715–726. Davies SM, Bhatia S, Ross JA, Kiffmeyer WR, Gaynon PS, Radloff GA, et al. Glutathione S-transferase genotypes, genetic susceptibility, and outcome of therapy in childhood acute lymphoblastic leukaemia. Blood 2002; 100: 67–71. Brenner TL, Pui CH, Evan WE. Pharmacogenomics of childhood acute lymphoblastic leukaemia. Curr Opin Mol Ther 2001; 3: 567–578.

41 62. Davies SM, Robison LL, Buckley JD, Tjoa T, Woods WG, Radloff GA, et al. Glutathione S-transferase polymorphisms and outcome of chemotherapy in childhood acute myeloid leukaemia. J Clin Oncol 2001; 19: 1279–1287. 63. Naoe T, Tagawa Y, Kiyoi H, Kodera Y, Miyawaki S, Asou N, et al. Prognostic significance of the null genotype of glutathione S-transferase-T1 in patients with acute myeloid leukaemia: increased early death after chemotherapy. Leukaemia 2002; 16: 203–208. 64. Krajinovic M, Labuda D, Richer C, Karimi S, Sinnett D. Susceptibility to childhood acute lymphoblastic leukaemia: influence of CYP1A1, CYP2D6, GSTM1, and GSTT1 genetic polymorphisms. Blood 1999; 93: 1496–1501. 65. Ida K, Kitabayashi I, Taki T, Taniwaki M, Noro K, Yamamoto M, et al. Adenoviral E1A-associated protein p300 is involved in acute myeloid leukaemia with t(11;22)(q23;q13). Blood 1997; 90: 4699–4704. 66. Woo MH, Shuster JJ, Chen C, Bash RO, Behm FG, Camitta B, et al. Glutathione S-transferase genotypes in children who develop treatment-related acute myeloid malignancies. Leukaemia 2000; 14: 232–237. 67. Danesi R, De Braud F, Fogli S, Di Paolo A, Del Tacca M. Pharmacogenetic determinants of anti-cancer drug activity and toxicity. Trends Pharmacol Sci 2001; 22: 420–426. 68. Kuehl P, Zhang J, Lin Y, Lamba J, Assem M, Schuetz J, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 2001; 27: 383–391. 69. Felix CA, Walker AH, Lange BJ, Williams TM, Winick NJ, Cheung NK, et al. Association of CYP3A4 genotype with treatment-related leukaemia. Proc Natl Acad Sci USA 1998; 95: 13176–13181. 70. Krajinovic M, Labuda D, Mathonnet G, Labuda M, Moghrabi A, Champagne J, et al. Polymorphisms in genes encoding drugs and xenobiotic metabolizing enzymes, DNA repair enzymes, and response to treatment of childhood acute lymphoblastic leukaemia. Clin Cancer Res 2002; 8: 802–810. 71. Krajinovic M, Sinnett H, Richer C, Labuda D, Sinnett D. Role of NQO1, MPO and CYP2E1 genetic polymorphisms in the susceptibility to childhood acute lymphoblastic leukaemia. Int J Cancer 2002; 97: 230–236. 72. Frosst P, Blom HJ, Milos R, Goyette P, Sheppard CA, Matthews RG, et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet 1995; 10: 111–113. 73. Molloy AM, Daly S, Mills JL, Kirke PN, Whitehead AS, Ramsbottom D, et al. Thermolabile variant of 5,10methylenetetrahydrofolate reductase associated with low red-cell folates: implications for folate intake recommendations. Lancet 1997; 349: 1591–1593. 74. van der Put NM, Gabreels F, Stevens EM, Smeitink JA, Trijbels FJ, Eskes TK, et al. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet 1998; 62: 1044–1051. 75. Morgan SL, Baggott JE, Lee JY, Alarcon GS. Folic acid supplementation prevents deficient blood folate levels and hyperhomocysteinemia during longterm, low dose methotrexate therapy for rheumatoid arthritis: implications for cardiovascular disease prevention. J Rheumatol 1998; 25: 441– 446. 76. Ulrich CM, Yasui Y, Storb R, Schubert MM, Wagner JL, Bigler J, et al. Pharmacogenetics of methotrexate: toxicity among marrow transplantation patients varies with the methylenetetrahydrofolate reductase C677T polymorphism. Blood 2001; 98: 231–234. 77. Thompson JR, Gerald PF, Willoughby ML, Armstrong BK. Maternal folate supplementation in pregnancy and

42

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

88.

89.

90.

91.

92.

J.E. RUBNITZ AND C.-H. PUI protection against acute lymphoblastic leukaemia in childhood: a case-control study. Lancet 2001; 358: 1935–1940. Skibola CF, Smith MT, Kane E, Roman E, Rollinson S, Cartwright RA, et al. Polymorphisms in the methylenetetrahydrofolate reductase gene are associated with susceptibility to acute leukaemia in adults. Proc Natl Acad Sci USA 1999; 96: 12810–12815. Franco RF, Simoes BP, Tone LG, Gabellini SM, Zago MA, Falcao RP. The methylenetetrahydrofolate reductase C677T gene polymorphism decreases the risk of childhood acute lymphocytic leukaemia. Br J Haematol 2001; 115: 616–618. Wiemels JL, Smith RN, Taylor GM, Eden OB, Alexander FE, Greaves MF. Methylenetetrahydrofolate reductase (MTHFR) polymorphisms and risk of molecularly defined subtypes of childhood acute leukaemia. Proc Natl Acad Sci USA 2001; 98: 4004–4009. Wiemels JL, Pagnamenta A, Taylor GM, Eden OB, Alexander FE, Greaves MF. A lack of a functional NAD(P)H:quinone oxidoreductase allele is selectively associated with paediatric leukaemias that have MLL fusions United Kingdom childhood cancer study investigators. Cancer Res 1999; 59: 4095–4099. Smith MT, Wang Y, Kane E, Rollinson S, Wiemels JL, Roman E, et al. Low NAD(P)H:quinone oxidoreductase 1 activity is associated with increased risk of acute leukaemia in adults. Blood 2001; 97: 1422–1426. Larson RA, Wang Y, Banerjee M, Wiemels J, Hartford C, Le Beau MM, et al. Prevalence of the inactivating 609C ! T polymorphism in the NAD(P)H:quinone oxidoreductase (NQO1) gene in patients with primary and therapy-related myeloid leukaemia. Blood 1999; 94: 803–807. Naoe T, Takeyama K, Yokozawa T, Kiyoi H, Seto M, Uike N, et al. Analysis of genetic polymorphism in NQO1, GST-M1, GST-T1, and CYP3A4 in 469 Japanese patients with therapyrelated leukaemia/myelodysplastic syndrome and de novo acute myeloid leukaemia. Clin Cancer Res 2000; 6: 4091–4095. Krajinovic M, Costea I, Chiasson S. Polymorphism of the thymidylate synthase gene and outcome of acute lymphoblastic leukaemia. Lancet 2002; 359: 1033–1034. Skibola CF, Smith MT, Hubbard A, Shane B, Roberts AC, Law GR, et al. Polymorphisms in the thymidylate synthase and serine hydroxymethyltransferase genes and risk of adult acute lymphocytic leukaemia. Blood 2002; 99: 3786–3791. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer 2002; 2: 48–58. Riehm H, Reiter A, Schrappe M, Berthold F, Dopfer R, Gerein V, et al. Corticosteroid-dependent reduction of leukocyte count in blood as a prognostic factor in acute lymphoblastic leukaemia in childhood (therapy study ALL-BFM 83). Klin Padiatr 1987; 199: 151–160. Gajjar A, Ribeiro R, Hancock ML, Rivera GK, Mahmoud H, Sandlund JT, et al. Persistence of circulating blasts after 1 week of multiagent chemotherapy confers a poor prognosis in childhood acute lymphoblastic leukaemia. Blood 1995; 86: 1292–1295. Schrappe M, Arico M, Harbott J, Biondi A, Zimmermann M, Conter V, et al. Philadelphia chromosome-positive (Phþ ) childhood acute lymphoblastic leukaemia: good initial steroid response allows early prediction of a favorable treatment outcome. Blood 1998; 92: 2730–2741. Dordelmann M, Reiter A, Borkhardt A, Ludwig WD, Gotz N, Viehmann S, et al. Prednisone response is the strongest predictor of treatment outcome in infant acute lymphoblastic leukaemia. Blood 1999; 94: 1209–1217. Steinherz PG, Gaynon PS, Breneman JC, Cherlow JM, Grossman NJ, Kersey JH, et al. Cytoreduction and prognosis in acute lymphoblastic leukaemia – the

93.

94.

95.

96.

97.

98.

99.

100.

101.

102.

103.

104.

105. 106.

107.

108.

109.

importance of early marrow response: report from the childrenÕs cancer group. J Clin Oncol 1996; 14: 389–398. Gaynon PS, Desai AA, Bostrom BC, Hutchinson RJ, Lange BJ, Nachman JB, et al. Early response to therapy and outcome in childhood acute lymphoblastic leukaemia: a review. Cancer 1997; 80: 1717–1726. Pui CH. Risk assessment in acute lymphoblastic leukaemia: beyond leukaemia cell characteristics. J Pediatr Hematol Oncol 2001; 23: 405–408. Sandlund JT, Harrison PL, Rivera G, Behm FG, Head D, Boyett J, et al. Persistence of lymphoblasts in bone marrow on day 15 and days 22 to 25 of remission induction predicts a dismal treatment outcome in children with acute lymphoblastic leukaemia. Blood 2002; 100: 43–47. Campana D, Coustan-Smith E. Advances in the immunological monitoring of childhood acute lymphoblastic leukaemia. Best Pract Res Clin Haematol 2002; 15: 1–19. Pui CH, Campana D. New definition of remission in childhood acute lymphoblastic leukaemia. Leukaemia 2000; 14: 783–785. Szczepanski T, Orfao A, van DVV, San Miguel JF, van Dongen JJ. Minimal residual disease in leukaemia patients. Lancet Oncol 2001; 2: 409–417. Coustan-Smith E, Behm FG, Sanchez J, Boyett JM, Hancock ML, Raimondi SC, et al. Immunological detection of minimal residual disease in children with acute lymphoblastic leukaemia. Lancet 1998; 351: 550–554. Coustan-Smith E, Sancho J, Hancock ML, Boyett JM, Behm FG, Raimondi SC, et al. Clinical importance of minimal residual disease in childhood acute lymphoblastic leukaemia. Blood 2000; 96: 2691–2696. Coustan-Smith E, Sancho J, Behm FG, Hancock ML, Razzouk BI, Ribeiro RC, et al. Prognostic importance of measuring early clearance of leukaemic cells by flow cytometry in childhood acute lymphoblastic leukaemia. Blood 2002; 100: 52–58. Coustan-Smith E, Sancho J, Hancock ML, Razzouk BI, Ribeiro RC, Rivera GK, et al. Use of peripheral blood instead of bone marrow to monitor residual disease in children with acute lymphoblastic leukaemia. Blood 2002; 100: 2399–2402. Neale GA, Coustan-Smith E, Pan Q, Chen X, Gruhn B, Stow P, et al. Tandem application of flow cytometry and polymerase chain reaction for comprehensive detection of minimal residual disease in childhood acute lymphoblastic leukaemia. Leukaemia 1999; 13: 1221–1226. Dworzak MN, Froschl G, Printz D, Mann G, Potschger U, Muhlegger N, et al. Prognostic significance and modalities of flow cytometric minimal residual disease detection in childhood acute lymphoblastic leukaemia. Blood 2002; 99: 1952–1958. Biondi A, Cimino G, Pieters R, Pui CH. Biological and therapeutic aspects of infant leukaemia. Blood 2000; 96: 24–33. Armstrong SA, Staunton JE, Silverman LB, Pieters R, den Boer ML, Minden MD, et al. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukaemia. Nat Genet 2002; 30: 41–47. Silverman LB, McLean TW, Gelber RD, Donnelly MJ, Gilliland DG, Tarbell NJ, et al. Intensified therapy for infants with acute lymphoblastic leukaemia: results from the Dana–Farber cancer institute consortium. Cancer 1997; 80: 2285–2295. Chessells JM, Harrison CJ, Watson SL, Vora AJ, Richards SM. Treatment of infants with lymphoblastic leukaemia: results of the UK infant protocols 1987–1999. Br J Haematol 2002; 117: 306–314. Isoyama K, Eguchi M, Hibi S, Kinukawa N, Ohkawa H, Kawasaki H, et al. Risk-directed treatment of infant acute

ADVANCES IN TREATMENT OF ALL

110.

111.

112.

113.

114.

115.

116.

117.

118.

119.

120.

121.

122.

123.

lymphoblastic leukaemia based on early assessment of MLL gene status: results of the Japan infant leukaemia study (MLL96). Br J Haematol 2002; 118: 999–1010. Pui C-H, Boyett JM, Relling MV, Harrison PL, Rivera GK, Behm FG, et al. Sex differences in prognosis for children with acute lymphoblastic leukaemia. J Clin Oncol 1999; 17: 818–824. Shuster JJ, Wacker P, Pullen J, Humbert J, Land VJ, Mahoney DHJ, et al. Prognostic significance of sex in childhood Bprecursor acute lymphoblastic leukaemia: a paediatric oncology group study. J Clin Oncol 1998; 16: 2854–2863. Bhatia S, Sather HN, Heerema NA, Trigg ME, Gaynon PS, Robison LL. Racial and ethnic differences in survival of children with acute lymphoblastic leukaemia. Blood 2002; 100: 1957–1964. Schrappe M, Reiter A, Zimmermann M, Harbott J, Ludwig WD, Henze G, et al. Long-term results of four consecutive trials in childhood ALL performed by the ALL-BFM study group from 1981 to 1995. Berlin–Frankfurt–Muenster. Leukaemia 2000; 14: 2205–2222. Harms DO, Janka-Schaub GE. Co-operative study group for childhood acute lymphoblastic leukaemia (COALL): longterm follow-up of trials 82, 85, 89 and 92. Leukaemia 2000; 14: 2234–2239. Silverman LB, Declerck L, Gelber RD, Dalton VK, Asselin BL, Barr RD, et al. Results of Dana–Farber cancer institute consortium protocols for children with newly diagnosed acute lymphoblastic leukaemia (1981–1995). Leukaemia 2000; 14: 2247–2256. Kamps WA, Veerman AJ, Van Wering ER, van Weerden JF, Slater R, Does-van den Berg A. Long-term follow-up of dutch childhood leukaemia study group (DCLSG) protocols for children with acute lymphoblastic leukaemia, 1984–1991. Leukaemia 2000; 14: 2240–2246. Hurwitz CA, Silverman LB, Schorin MA, Clavell LA, Dalton VK, Glick KM, et al. Substituting dexamethasone for prednisone complicates remission induction in children with acute lymphoblastic leukaemia. Cancer 2000; 88: 1964– 1969. Nachman JB, Sather HN, Sensel MG, Trigg ME, Cherlow JM, Lukens JN, et al. Augmented post-induction therapy for children with high-risk acute lymphoblastic leukaemia and a slow response to initial therapy. N Engl J Med 1998; 338: 1663–1671. Lange BJ, Bostrom BC, Cherlow JM, Sensel MG, La MK, Rackoff W, et al. Double-delayed intensification improves event-free survival for children with intermediate-risk acute lymphoblastic leukaemia: a report from the childrenÕs cancer group. Blood 2002; 99: 825–833. Arico M, Valsecchi MG, Conter V, Rizzari C, Pession A, Messina C, et al. Improved outcome in high-risk childhood acute lymphoblastic leukaemia defined by prednisone-poor response treated with double Berlin–Frankfurt–Muenster protocol II. Blood 2002; 100: 420–426. Chessells JM, Harrison G, Richards SM, Gibson BE, Bailey CC, Hill FG, et al. Failure of a new protocol to improve treatment results in paediatric lymphoblastic leukaemia: lessons from the UK Medical Research Council trials UKALL X and UKALL XI. Br J Haematol 2002; 118: 445–455. Amylon MD, Shuster J, Pullen J, Berard C, Link MP, Wharam M, et al. Intensive high-dose asparaginase consolidation improves survival for paediatric patients with T cell acute lymphoblastic leukaemia and advanced stage lymphoblastic lymphoma: a paediatric oncology group study. Leukaemia 1999; 13: 335–342. McLean TW, Ringold S, Neuberg D, Stegmaier K, Tantravahi R, Ritz J, et al. TEL/AML1 dimerizes and is associated with a

43

124.

125.

126.

127.

128.

129.

130.

131.

132.

133. 134.

135.

136.

137.

138. 139.

favorable outcome in childhood acute lymphoblastic leukaemia. Blood 1996; 88: 4252–4258. Lipshultz SE, Giantris AL, Lipsitz SR, Kimball DV, Asselin BL, Barr RD, et al. Doxorubicin administration by continuous infusion is not cardioprotective: the Dana–Farber 91-01 acute lymphoblastic leukaemia protocol. J Clin Oncol 2002; 20: 1677–1682. Toyoda Y, Manabe A, Tsuchida M, Hanada R, Ikuta K, Okimoto Y, et al. Six months of maintenance chemotherapy after intensified treatment for acute lymphoblastic leukaemia of childhood. J Clin Oncol 2000; 18: 1508–1516. Chessells JM, Harrison G, Lilleyman JS, Bailey CC, Richards SM. Continuing (maintenance) therapy in lymphoblastic leukaemia: lessons from MRC UKALL X. Medical research council working party in childhood leukaemia. Br J Haematol 1997; 98: 945–951. Dervieux T, Medard Y, Verpillat P, Guigonis V, Duval M, Lescoeur B, et al. Possible implication of thiopurine Smethyltransferase in occurrence of infectious episodes during maintenance therapy for childhood lymphoblastic leukaemia with mercaptopurine. Leukaemia 2001; 15: 1706– 1712. Fuscoe JC, Knapp GW, Hanley NM, Setzer RW, Sandlund JT, Pui CH, et al. The frequency of illegitimate V(D)J recombinasemediated mutations in children treated with etoposidecontaining antileukaemic therapy. Mutat Res 1998; 419: 107–121. Duration and intensity of maintenance chemotherapy in acute lymphoblastic leukaemia: overview of 42 trials involving 12,000 randomised children. Childhood ALL Collaborative Group. Lancet 1996;347:1783–1788. Mattano Jr LA, Sather HN, Trigg ME, Nachman JB. Osteonecrosis as a complication of treating acute lymphoblastic leukaemia in children: a report from the childrenÕs cancer group. J Clin Oncol 2000; 18: 3262–3272. Ribeiro RC, Fletcher BD, Kennedy W, Harrison PL, Neel MD, Kaste SC, et al. Magnetic resonance imaging detection of avascular necrosis of the bone in children receiving intensive prednisone therapy for acute lymphoblastic leukaemia or non-Hodgkin lymphoma. Leukaemia 2001; 15: 891–897. Strauss AJ, Su JT, Dalton VM, Gelber RD, Sallan SE, Silverman LB. Bony morbidity in children treated for acute lymphoblastic leukaemia. J Clin Oncol 2001; 19: 3066–3072. Pui C-H. Childhood leukaemias. N Engl J Med 1995; 332: 1618–1630. Gajjar A, Harrison PL, Sandlund JT, Rivera GK, Ribeiro RC, Rubnitz JE, et al. Traumatic lumbar puncture at diagnosis adversely affects outcome in childhood acute lymphoblastic leukaemia. Blood 2000; 96: 3381–3384. Eden OB, Harrison G, Richards S, Lilleyman JS, Bailey CC, Chessells JM, et al. Long-term follow-up of the United Kingdom medical research council protocols for childhood acute lymphoblastic leukaemia, 1980–1997. Medical research council childhood leukaemia working party. Leukaemia 2000; 14: 2307–2320. Walter AW, Hancock ML, Pui CH, Hudson MM, Ochs JS, Rivera GK, et al. Secondary brain tumors in children treated for acute lymphoblastic leukaemia at St. Jude childrenÕs research hospital. J Clin Oncol 1998; 16: 3761–3767. Bhatia S, Sather HN, Pabustan OB, Trigg ME, Gaynon PS, Robison LL. Low incidence of second neoplasms among children diagnosed with acute lymphoblastic leukaemia after 1983. Blood 2002; 99: 4257–4264. Pui C-H. Cure of childhood ALL: exacting a lower toll. Blood 2002; 99: 4255. Manera R, Ramirez I, Mullins J, Pinkel D. Pilot studies of species-specific chemotherapy of childhood acute lympho-

44

140.

141.

142. 143.

144.

145.

146.

147.

148.

149.

150.

151.

152.

J.E. RUBNITZ AND C.-H. PUI blastic leukaemia using genotype and immunophenotype. Leukaemia 2000; 14: 1354–1361. Vilmer E, Suciu S, Ferster A, Bertrand Y, Cave H, Thyss A, et al. Long-term results of three randomized trials (58831, 58832, 58881) in childhood acute lymphoblastic leukaemia: a CLCG-EORTC report. Children leukaemia cooperative group. Leukaemia 2000; 14: 2257–2266. Leung W, Rose SR, Zhou Y, Hancock ML, Burstein S, Schriock EA, et al. Outcomes of growth hormone replacement therapy in survivors of childhood acute lymphoblastic leukaemia. J Clin Oncol 2002; 20: 2959–2964. Pui C-H. Toward optimal central-nervous-system treatment in childhood ALL. J Clin Oncol 2002; in press. Mahmoud HH, Rivera GK, Hancock ML, Krance RA, Kun LE, Behm FG, et al. Low leukocyte counts with blast cells in cerebrospinal fluid of children with newly diagnosed acute lymphoblastic leukaemia. N Engl J Med 1993; 329: 314–319. Lauer S, Shuster J, Kirchner P, Kiefer G, Pullen J, Camitta B, et al. Prognostic significance of cerebrospinal fluid (CSF) lymphoblasts (LB) at diagnosis (dx) in children with acute lymphoblastic leukaemia (ALL) (Meeting abstract). Proc Annu Meet Am Soc Clin Oncol 1994; 13: 317. Gilchrist GS, Tubergen DG, Sather HN, Coccia PF, OÕBrien RT, Waskerwitz MJ, et al. Low numbers of CSF blasts at diagnosis do not predict for the development of CNS leukaemia in children with intermediate-risk acute lymphoblastic leukaemia: a childrenÕs cancer group report. J Clin Oncol 1994; 12: 2594–2600. van den BH, Vet R, den Ouden E, Behrendt H. Significance of lymphoblasts in cerebrospinal fluid in newly diagnosed paediatric acute lymphoblastic malignancies with bone marrow involvement: possible benefit of dexamethasone. Med Pediatr Oncol 1995; 25: 22–27. Burger B, Zimmermann M, Mann G, Kuhl J, Loning L, Riehm H, et al. Diagnostic cerebrospinal fluid (CSF) examination in children with acute lymphoblastic leukaemia (ALL): significance of low leukocyte counts with blasts or traumatic lumbar puncture. J Clin Oncol 2002; in press. Nachman J, Cherlow J, Sather HN. Effect of initial central nervous system (CNS) status on event-free (EFS) in children and adolescents with acute lymphoblastic leukaemia (ALL). Med Pediatr Oncol 2002; 39: 277 (abstract). Prassopoulos P, Cavouras D, Golfinopoulos S, Evlogias N, Theodoropoulos V, Panagiotou J. Quantitative assessment of cerebral atrophy during and after treatment in children with acute lymphoblastic leukaemia. Invest Radiol 1996; 31: 749– 754. Vainionpaa L, Kovala T, Tolonen U, Lanning M. Chemotherapy for acute lymphoblastic leukaemia may cause subtle changes of the spinal cord detectable by somatosensory evoked potentials. Med Pediatr Oncol 1997; 28: 41–47. Hill DE, Ciesielski KT, Sethre-Hofstad L, Duncan MH, Lorenzi M. Visual and verbal short-term memory deficits in childhood leukaemia survivors after intrathecal chemotherapy. J Pediatr Psychol 1997; 22: 861–870. Howard SC, Gajjar A, Cheng C, Kritchevsky SB, Somes GW, Harrison PL, et al. Traumatic lumbar puncture in childhood

153.

154.

155. 156.

157. 158.

159.

160.

161.

162.

163.

164.

165.

166.

acute lymphoblastic leukaemia. J Am Med Assoc 2002; in press. Howard SC, Gajjar A, Ribeiro RC, Rivera GK, Rubnitz JE, Sandlund JT, et al. Safety of lumbar puncture for children with acute lymphoblastic leukaemia and thrombocytopenia. J Am Med Assoc 2000; 284: 2222–2224. Pui CH, Jackson CW, Chesney C, Lyles SA, Bowman WP, Abromowitch M, et al. Sequential changes in platelet function and coagulation in leukaemic children treated with L -asparaginase, prednisone, and vincristine. J Clin Oncol 1983; 1: 380–385. Marie JP. Drug resistance in haematologic malignancies. Curr Opin Oncol 2001; 13: 463–469. Zhou S, Schuetz JD, Bunting KD, Colapietro AM, Sampath J, Morris JJ, et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med 2001; 7: 1028–1034. Sorrentino BP. Gene therapy to protect haematopoietic cells from cytotoxic cancer drugs. Nat Rev Cancer 2002; 2: 431–441. Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, et al. Efficacy and safety of a specific inhibitor of the BCRABL tyrosine kinase in chronic myeloid leukaemia. N Engl J Med 2001; 344: 1031–1037. Kantarjian H, Sawyers C, Hochhaus A, Guilhot F, Schiffer C, Gambacorti-Passerini C, et al. Haematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukaemia. N Engl J Med 2002; 346: 645–652. Talpaz M, Silver RT, Druker BJ, Goldman JM, GambacortiPasserini C, Guilhot F, et al. Imatinib induces durable haematologic and cytogenetic responses in patients with accelerated phase chronic myeloid leukaemia: results of a phase 2 study. Blood 2002; 99: 1928–1937. Sawyers CL, Hochhaus A, Feldman E, Goldman JM, Miller CB, Ottmann OG, et al. Imatinib induces haematologic and cytogenetic responses in patients with chronic myelogenous leukaemia in myeloid blast crisis: results of a phase II study. Blood 2002; 99: 3530–3539. Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford JM, et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukaemia and acute lymphoblastic leukaemia with the Philadelphia chromosome. N Engl J Med 2001; 344: 1038–1042. Ottmann OG, Druker BJ, Sawyers CL, Goldman JM, Reiffers J, Silver RT, et al. A phase 2 study of imatinib in patients with relapsed or refractory Philadelphia chromosome-positive acute lymphoid leukaemias. Blood 2002; 100: 1965–1971. Weisberg E, Boulton C, Kelly LM, Manley P, Fabbro D, Meyer T, et al. Inhibition of mutant FLT3 receptors in leukaemia cells by the small molecule tyrosine kinase inhibitor PKC412. Cancer Cell 2002; 1: 433–443. Kelly LM, Yu JC, Boulton CL, Apatira M, Li J, Sullivan CM, et al. CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukaemia (AML). Cancer Cell 2002; 1: 421–432. Levis M, Allebach J, Tse KF, Zheng R, Baldwin BR, Smith BD, et al. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukaemia cells in vitro and in vivo. Blood 2002; 99: 3885– 3891.