National Cancer Institute
U.S. National Institutes of Health | www.cancer.gov

NCI Home
Cancer Topics
Clinical Trials
Cancer Statistics
Research & Funding
News
About NCI
Childhood Acute Lymphoblastic Leukemia Treatment (PDQ®)
Patient Version   Health Professional Version   En español   Last Modified: 10/31/2008



Purpose of This PDQ Summary






General Information






Cellular Classification and Prognostic Variables






Treatment Option Overview






Untreated Childhood Acute Lymphoblastic Leukemia






Childhood Acute Lymphoblastic Leukemia in Remission






Postinduction Treatment for Childhood Acute Lymphoblastic Leukemia Subgroups






Recurrent Childhood Acute Lymphoblastic Leukemia






Get More Information From NCI






Changes to this Summary (10/31/2008)






More Information



Page Options
Print This Page
Print Entire Document
View Entire Document
E-Mail This Document
Quick Links
Director's Corner

Dictionary of Cancer Terms

NCI Drug Dictionary

Funding Opportunities

NCI Publications

Advisory Boards and Groups

Science Serving People

Español
Quit Smoking Today
NCI Highlights
Report to Nation Finds Declines in Cancer Incidence, Death Rates

High Dose Chemotherapy Prolongs Survival for Leukemia

Prostate Cancer Study Shows No Benefit for Selenium, Vitamin E

The Nation's Investment in Cancer Research FY 2009

Past Highlights
Untreated Childhood Acute Lymphoblastic Leukemia

Induction Chemotherapy
        Options under clinical evaluation for end-induction treatment assignment
Central Nervous System Therapy
        No clinical evidence of CNS involvement at diagnosis
        Clinically evident CNS involvement at diagnosis
Current Clinical Trials



Induction Chemotherapy

Three-drug induction therapy using vincristine, prednisone/dexamethasone, plus L-asparaginase in conjunction with intrathecal therapy (IT), results in complete remission rates of greater than 95%.[1] For patients at high risk of treatment failure, a more intense induction regimen (four or five agents) may result in improved event-free survival (EFS),[2-4] and high-risk patients generally receive induction therapy that includes an anthracycline (e.g., daunomycin) in addition to vincristine, prednisone/dexamethasone, plus L-asparaginase. For patients who are at standard risk or low risk of treatment failure, four-drug induction therapy does not appear necessary for favorable outcome provided that adequate postremission intensification therapy is administered.[2,5,6] Because of the likelihood of increased toxicity with four-drug induction therapy, Children's Oncology Group (COG) protocols for National Cancer Institute (NCI) standard-risk precursor B-cell ALL currently utilize dexamethasone, vincristine, and L-asparaginase. Induction regimens using four or more agents are reserved for higher risk patients.[2,5,7]

Many current regimens utilize dexamethasone instead of prednisone during remission induction and later phases of therapy. The Children's Cancer Group (CCG) conducted a randomized trial comparing dexamethasone and prednisone in standard-risk patients, and reported that dexamethasone was associated with a superior EFS.[8] Results from another randomized trial conducted by the United Kingdom Medical Research Council (MRC) demonstrated that dexamethasone was associated with a more favorable outcome than prednisolone in all patient subgroups.[9] In the MRC trial, patients who received dexamethasone had a significantly lower incidence of both central nervous system (CNS) and non-CNS relapses than patients who received prednisolone.[9] However, a third randomized trial (conducted in Japan) did not confirm a survival advantage with dexamethasone.[10] This discrepant result might have been due to the use of a higher dose of prednisolone during induction therapy, the use of a more intensive backbone chemotherapy regimen, and/or the smaller number of patients included in that trial.

While dexamethasone may be more effective than prednisone, data also suggest that dexamethasone may also be more toxic, especially in the context of more intensive induction regimens. Several reports indicate that dexamethasone may increase the frequency and severity of infections and/or other complications in patients receiving intensive (more than three drugs) induction regimens.[11,12] The Berlin-Frankfurt-Muenster (BFM) group observed increased mortality in adolescent patients who received dexamethasone instead of prednisone during a four-drug induction regimen. However, the MRC group did not observe any toxicity or mortality differences between dexamethasone and prednisone during a four-drug induction regimen.[3] Dexamethasone appears to have a greater suppressive effect on short-term linear growth than prednisone,[13] and has been associated with a higher risk of osteonecrosis, especially in adolescent patients.[14]

Several forms of L-asparaginase are available for use in the treatment of children with ALL, with Eschericia coli (E. coli) L-asparaginase being most commonly used. PEG-L-asparaginase is an alternative form of L-asparaginase in which the E. coli enzyme is modified by the covalent attachment of polyethylene glycol. PEG-L-asparaginase has a much longer serum half-life than native E. coli L-asparaginase, allowing it to produce asparagine depletion with less frequent administration.[15] A single intramuscular dose of PEG-L-asparaginase given in conjunction with vincristine and prednisone during induction therapy appears to have similar activity and toxicity as nine doses of intramuscular E. coli L-asparaginase (three times a week for 3 weeks).[16] In a randomized comparison of PEG-L-asparaginase versus native E. coli asparaginase in which each agent was to be given over a 30-week period following achievement of remission, similar outcome and similar rates of asparaginase-related toxicities were observed for both groups of patients.[17] In another randomized trial in which patients with standard-risk ALL were randomly assigned to receive PEG-L-asparaginase versus native E. coli asparaginase in induction and each of two delayed intensification courses, the use of PEG-L-asparaginase was associated with more rapid blast clearance and a lower incidence of neutralizing antibodies.[16] Current COG protocols use PEG-L-asparaginase during induction for all patients with ALL. Patients with an allergic reaction to PEG-L-asparaginase should be switched to Erwinia L-asparaginase. If Erwinia L-asparaginase is utilized, the shorter half-life of the Erwinia preparation requires more frequent administration and a higher dose. In two studies, patients randomly assigned to receive Erwinia L-asparaginase on the same schedule and dosage as E. coli L-asparaginase had a significantly worse EFS.[18,19]

More than 95% of children with newly diagnosed ALL will achieve a complete remission within the first 4 weeks of treatment. Of the 2% to 4% of patients who fail to achieve complete remission within the first 4 weeks, approximately half will experience a toxic death during the induction phase (usually due to infection) and the other half will have resistant disease (persistent morphologic leukemia).[19-21] Patients who require more than 4 weeks to achieve remission have a poor prognosis and may benefit from an allogeneic stem cell transplant once complete remission is achieved.[22-24] Outcome is also less favorable for patients who demonstrate more than 25% blasts in the bone marrow or persistent blasts in the peripheral blood after 1 week of intensive multiagent induction therapy,[3,25,26] and protocols of the former CCG-based treatment decisions on the day 7 bone marrow response (for high-risk patients) or day 14 bone marrow response (for standard-risk patients).[27]

Options under clinical evaluation for end-induction treatment assignment

The COG is evaluating a new end-induction classification system. Standard-risk patients (aged 1–9.99 years, <50,000 white blood cell count (WBC)/µL, precursor B-cell ALL) receive a three-drug induction regimen with dexamethasone, PEG-L-asparaginase, and vincristine. High-risk precursor B-cell ALL patients receive a four-drug induction regimen including daunomycin and are randomly assigned to receive either dexamethasone for 14 days or prednisone for 28 days. Patients with T-cell ALL receive a four-drug induction regimen with prednisone as the induction steroid. Patients with ALL undergo an evaluation on day 28 that includes a bone marrow biopsy/aspiration for morphology and minimal residual disease (MRD) determinations. On the basis of day 14 marrow morphology, day 28 marrow morphology, and day 28 MRD determination, ALL patients are classified as rapid responders, slow responders, very slow responders, or induction failures. Patients with t(9;22) and hypodiploidy with fewer than 44 chromosomes move to the very high-risk protocol at the end of induction therapy, regardless of response classification. Patients with MLL gene rearrangements who do not show a rapid response also move to the very high-risk treatment protocol. Patients with ALL and slow or rapid response to induction receive consolidation on the appropriate standard or high-risk precursor B-cell ALL or T-cell treatment protocol. Patients with very slow response to induction therapy receive an additional 2 weeks of induction therapy and then have a day 42 bone marrow evaluation for morphology and MRD. Patients with an M1 day 42 marrow and less than 0.1 MRD go onto consolidation therapy; all other patients are considered induction failures and move to the very high-risk treatment protocol.

Table 1. COG End-Induction Response Classification
Early Marrow Morphology  MRD Day 28  Marrow Morphology Day 28  COG Classification 
Day 7 Day 14
M1 - <0.1% M1 Rapid response
M2/M3 M1 <0.1% M1 Rapid response
- M2/M3 <1.0% M1 Slow response
Any Any 0.1%–1.0% M1 Slow response
Any Any >1.0% M1, M2 Very slow response; extended induction
Any Any Any M2 Very slow response; extended induction
Any Any Any M3 Induction failure

The Dana-Farber Cancer Institute (DFCI) ALL Consortium is also testing a new risk classification system for patients with precursor B-cell ALL. All patients are initially risk classified as either standard risk or high risk based upon age, presenting leukocyte count, and presence or absence of CNS disease. At the completion of a five-drug remission induction regimen (4 weeks from diagnosis), the level of MRD is determined. Patients with high MRD (≥0.1%) are classified as very high risk and receive a more intensive postremission consolidation. Patients with low MRD (<0.1%) continue to receive treatment based on their initial risk group classification. The goal of this new classification schema is to determine whether intensification of therapy will improve the outcome of patients with high MRD at the end of remission induction. Patients with T-cell ALL are treated as high risk, regardless of MRD status. All patients with MLL translocations or hypodiploidy (<45 chromosomes) are classified as very high-risk, regardless of MRD status or phenotype. Patients with the Philadelphia chromosome are treated as high risk, but receive an allogeneic stem cell transplant in first remission.

Central Nervous System Therapy

The early institution of adequate CNS therapy is critical for eliminating clinically evident CNS disease at diagnosis and for preventing CNS relapse in patients without overt CNS involvement at diagnosis. IT chemotherapy is usually started at the beginning of induction, intensified during consolidation (4–8 doses of IT given every 2–3 weeks), and often continued throughout maintenance. A current goal of ALL therapy design is to achieve effective CNS therapy while minimizing neurotoxicity. Every patient with ALL receives IT chemotherapy with methotrexate alone or methotrexate with cytarabine plus hydrocortisone.[28] IT methotrexate may also have a significant systemic effect that could result in a decrease in marrow relapse rate.[29] Historically, significant control of bone marrow relapse did not occur until CNS therapy was instituted. Conversely, the type and extent of systemic intensification also appears to influence the efficacy of the CNS therapy. Systemically administered drugs such as dexamethasone, L-asparaginase, high-dose methotrexate, and high-dose cytarabine may provide some degree of CNS protection. For example, in a CCG study of patients with standard-risk ALL, oral dexamethasone decreased the CNS relapse rate by 50% compared with patients receiving oral prednisone (patients received IT methotrexate alone for CNS prophylaxis).[8]

No clinical evidence of CNS involvement at diagnosis

IT chemotherapy may be the sole form of presymptomatic CNS therapy, or it may be combined with systemic moderate-dose to high-dose infusions of methotrexate with leucovorin rescue and/or cranial radiation (12–18 Gy).[30] The DFCI ALL Consortium 95-01 trial randomized lower risk patients to receive either 18 Gy of cranial irradiation with IT chemotherapy or more frequently dosed IT chemotherapy alone (no radiation therapy) and demonstrated no significant difference in EFS.[19] Appropriate systemic therapy combined with IT chemotherapy resulted in CNS relapse rates of less than 5% for children with standard-risk ALL.[5,20,31,32] The question of whether to use triple (methotrexate, prednisone, cytarabine) IT therapy or single (methotrexate) IT therapy in nonirradiated standard-risk patients was studied as a randomized question in the CCG-1952 clinical trial.[33] The results showed an isolated CNS relapse rate of 3.4 ± 1.0% for triple IT therapy and 5.9 ± 1.2% for single IT therapy (P = .004). There were, however, more bone marrow relapses in the group that received the triple IT therapy, leading to a worse overall survival (OS) in this group (90.3 ± 1.5%) compared with the single IT therapy group (94.4 ± 1.1%; P = .01). When the analysis was restricted to patients with precursor B-cell ALL and rapid early response (M1 marrow on day 14), there was no difference between triple and single IT therapy in CNS relapse rate, OS, or EFS.[33] Patients with blasts in the cerebrospinal fluid (CSF) but less than 5 WBC/µL (CNS2) are at increased risk of CNS relapse and may require more intensive IT chemotherapy (but not cranial radiation).[34,35] Data also suggest that a traumatic lumbar puncture with blasts at the time of diagnosis is associated with an increased risk of CNS relapse, however, with more intensive IT chemotherapy, the risk is abrogated.[35,36]

Approximately 5% to 20% of children with ALL receive cranial radiation as part of their CNS-directed therapy, even if they present without CNS involvement at diagnosis, including those with T-cell phenotype and subsets of patients with high-risk precursor B-cell ALL.[37,38] Whether some or all of these patients could be as effectively treated without radiation is controversial and is currently under investigation.[39]

Toxic effects of CNS-directed therapy for childhood ALL can be divided into two broad groups. Acute/subacute toxicities include seizures, stroke, somnolence syndrome, and ascending paralysis. Chronic and late toxicities include meningiomas, leukoencephalopathy and a range of neurocognitive, behavioral, and neuroendocrine disturbances.[40,41]

Long-term deleterious effects of cranial radiation, particularly at doses higher than 18 Gy, have been recognized for years.[42] Children receiving these higher doses of cranial radiation are at significant risk of neurocognitive and neuroendocrine sequelae.[43-47] Young children (i.e., younger than 4 years) are at increased risk of neurocognitive decline and other sequelae following cranial radiation.[48-50] Girls may be at a higher risk of radiation-induced neuropsychologic and neuroendocrine sequelae than boys.[49-51] In general, high-dose methotrexate should not be given following cranial radiation because of the increased risk of neurologic sequelae, including leukoencephalopathy. In addition, radiation has been associated with the development of second neoplasms (many of which are benign or of low malignant potential).[47,52] Attempts at reducing the adverse sequelae of cranial radiation have included lowering the dose and utilizing alternative fractionation schedules. Children receiving 18 Gy of cranial radiation may be at diminished risk of neurologic toxicity compared with those receiving 24 Gy,[53] although neurocognitive and neuroendocrine effects, as well as development of second malignant neoplasms have been noted at this lower dose.[21,48,54] In the German BFM and DFCI ALL Consortium studies, many of the patients treated with cranial radiation receive a dose of only 12 Gy.[21] Longer follow-up is needed to determine whether 12 Gy will be associated with a lower incidence of neurologic sequelae. In a randomized trial, hyperfractionated radiation (at a dose of 18 Gy) did not decrease neurologic late effects when compared with conventionally fractionated radiation, although cognitive function for both groups was not significantly impaired.[55,56][Level of evidence: 1iiC]

The most common toxicity associated with IT chemotherapy alone is seizures. Approximately 5% to 10% of patients with ALL treated with frequent doses of IT chemotherapy will have at least one seizure during therapy.[7] Patients with ALL who develop seizures during the course of treatment and who receive anticonvulsant therapy should not receive phenobarbital or phenytoin as anticonvulsant treatment, as these drugs may increase the clearance of some chemotherapeutic drugs and adversely affect treatment outcome.[57] Valproic acid or gabapentin are alternative anticonvulsants with less enzyme-inducing capabilities.[57] In general, patients who receive IT chemotherapy without cranial radiation as CNS therapy appear to have a low incidence of neurocognitive sequelae, and the deficits that do develop represent relatively modest declines in a limited number of domains of neuropsychological functioning.[58-60] This modest decline is especially seen in young children and girls.[61] Regimens that utilize a biweekly schedule of 12 doses of intravenous high-dose methotrexate with leucovorin rescue and IT chemotherapy in the off-week have been associated with excessive neurologic toxicity.[62] Controversy exists about whether patients who receive dexamethasone are at higher risk for neurocognitive disturbances.[63]

Presymptomatic CNS therapy options under clinical evaluation

The following are examples of national and/or institutional clinical trials that are currently being conducted. Information about ongoing clinical trials is available from the NCI Web site.

  • In the COG-AALL0232 study, patients with high-risk precursor B-cell ALL receive either IT methotrexate alone (rapid early responders) or IT methotrexate with cranial radiation (slow early responders). One goal of this study is to determine whether oral dexamethasone, high-dose methotrexate, or both agents will decrease the incidence of CNS relapse.


  • In the COG-AALL0434 protocol for patients with T-cell ALL, low-risk T-cell patients will be treated without cranial radiation, and intermediate-risk T-cell patients will receive 12 Gy (instead of 18 Gy) cranial radiation. High-risk T-cell patients will continue to receive 18 Gy cranial radiation.


  • A clinical trial at St. Jude Children's Research Hospital is testing whether intensive IT and systemic chemotherapy without radiation can be used for CNS prophylaxis in all patients regardless of initial CNS status.


  • DFCI ALL Consortium Protocol 05-01 is testing whether IT chemotherapy alone can replace cranial radiation in some high-risk patients. Approximately 20% of patients will receive cranial radiation, including B-lineage patients with high presenting leukocyte counts (≥100,000 μ/L), CNS3 disease at diagnosis, or high MRD levels at the end of remission induction, and all T-cell ALL patients. The remaining 80% of patients will receive triple IT chemotherapy, but no radiation. The goal of this treatment schema is to reduce neurotoxicity and other CNS late effects without compromising efficacy by limiting the number of patients exposed to radiation and by lowering the radiation dose (12 Gy instead of 18 Gy) to those still receiving radiation.


Clinically evident CNS involvement at diagnosis

Standard treatment for ALL patients with clinically evident CNS disease (>5 WBC/μL with blasts on cytospin; CNS3) at diagnosis includes IT chemotherapy and cranial radiation (usual dose: 18 Gy). Spinal radiation is no longer used.

Treatment options under clinical evaluation

The following is an example of a national and/or institutional clinical trial that is currently being conducted. Information about ongoing clinical trials is available from the NCI Web site.

  • A clinical trial at St. Jude Children's Research Hospital is testing whether patients with clinically evident CNS disease at diagnosis can be treated with intensive IT and systemic chemotherapy without radiation.


Current Clinical Trials

Check for U.S. clinical trials from NCI's PDQ Cancer Clinical Trials Registry that are now accepting patients with untreated childhood acute lymphoblastic leukemia. The list of clinical trials can be further narrowed by location, drug, intervention, and other criteria.

General information about clinical trials is also available from the NCI Web site.

References

  1. Pui CH, Evans WE: Treatment of acute lymphoblastic leukemia. N Engl J Med 354 (2): 166-78, 2006.  [PUBMED Abstract]

  2. Tubergen DG, Gilchrist GS, O'Brien RT, et al.: Improved outcome with delayed intensification for children with acute lymphoblastic leukemia and intermediate presenting features: a Childrens Cancer Group phase III trial. J Clin Oncol 11 (3): 527-37, 1993.  [PUBMED Abstract]

  3. Gaynon PS, Steinherz PG, Bleyer WA, et al.: Improved therapy for children with acute lymphoblastic leukemia and unfavorable presenting features: a follow-up report of the Childrens Cancer Group Study CCG-106. J Clin Oncol 11 (11): 2234-42, 1993.  [PUBMED Abstract]

  4. LeClerc JM, Billett AL, Gelber RD, et al.: Treatment of childhood acute lymphoblastic leukemia: results of Dana-Farber ALL Consortium Protocol 87-01. J Clin Oncol 20 (1): 237-46, 2002.  [PUBMED Abstract]

  5. Veerman AJ, Hählen K, Kamps WA, et al.: High cure rate with a moderately intensive treatment regimen in non-high-risk childhood acute lymphoblastic leukemia. Results of protocol ALL VI from the Dutch Childhood Leukemia Study Group. J Clin Oncol 14 (3): 911-8, 1996.  [PUBMED Abstract]

  6. Chauvenet AR, Martin PL, Devidas M, et al.: Antimetabolite therapy for lesser-risk B-lineage acute lymphoblastic leukemia of childhood: a report from Children's Oncology Group Study P9201. Blood 110 (4): 1105-11, 2007.  [PUBMED Abstract]

  7. Mahoney DH Jr, Shuster J, Nitschke R, et al.: Intermediate-dose intravenous methotrexate with intravenous mercaptopurine is superior to repetitive low-dose oral methotrexate with intravenous mercaptopurine for children with lower-risk B-lineage acute lymphoblastic leukemia: a Pediatric Oncology Group phase III trial. J Clin Oncol 16 (1): 246-54, 1998.  [PUBMED Abstract]

  8. Bostrom BC, Sensel MR, Sather HN, et al.: Dexamethasone versus prednisone and daily oral versus weekly intravenous mercaptopurine for patients with standard-risk acute lymphoblastic leukemia: a report from the Children's Cancer Group. Blood 101 (10): 3809-17, 2003.  [PUBMED Abstract]

  9. Mitchell CD, Richards SM, Kinsey SE, et al.: Benefit of dexamethasone compared with prednisolone for childhood acute lymphoblastic leukaemia: results of the UK Medical Research Council ALL97 randomized trial. Br J Haematol 129 (6): 734-45, 2005.  [PUBMED Abstract]

  10. Igarashi S, Manabe A, Ohara A, et al.: No advantage of dexamethasone over prednisolone for the outcome of standard- and intermediate-risk childhood acute lymphoblastic leukemia in the Tokyo Children's Cancer Study Group L95-14 protocol. J Clin Oncol 23 (27): 6489-98, 2005.  [PUBMED Abstract]

  11. Hurwitz CA, Silverman LB, Schorin MA, et al.: Substituting dexamethasone for prednisone complicates remission induction in children with acute lymphoblastic leukemia. Cancer 88 (8): 1964-9, 2000.  [PUBMED Abstract]

  12. Belgaumi AF, Al-Bakrah M, Al-Mahr M, et al.: Dexamethasone-associated toxicity during induction chemotherapy for childhood acute lymphoblastic leukemia is augmented by concurrent use of daunomycin. Cancer 97 (11): 2898-903, 2003.  [PUBMED Abstract]

  13. Ahmed SF, Tucker P, Mushtaq T, et al.: Short-term effects on linear growth and bone turnover in children randomized to receive prednisolone or dexamethasone. Clin Endocrinol (Oxf) 57 (2): 185-91, 2002.  [PUBMED Abstract]

  14. Mattano LA Jr, Sather HN, Trigg ME, et al.: Osteonecrosis as a complication of treating acute lymphoblastic leukemia in children: a report from the Children's Cancer Group. J Clin Oncol 18 (18): 3262-72, 2000.  [PUBMED Abstract]

  15. Asselin BL, Whitin JC, Coppola DJ, et al.: Comparative pharmacokinetic studies of three asparaginase preparations. J Clin Oncol 11 (9): 1780-6, 1993.  [PUBMED Abstract]

  16. Avramis VI, Sencer S, Periclou AP, et al.: A randomized comparison of native Escherichia coli asparaginase and polyethylene glycol conjugated asparaginase for treatment of children with newly diagnosed standard-risk acute lymphoblastic leukemia: a Children's Cancer Group study. Blood 99 (6): 1986-94, 2002.  [PUBMED Abstract]

  17. Silverman LB, Gelber RD, Dalton VK, et al.: Improved outcome for children with acute lymphoblastic leukemia: results of Dana-Farber Consortium Protocol 91-01. Blood 97 (5): 1211-8, 2001.  [PUBMED Abstract]

  18. Duval M, Suciu S, Ferster A, et al.: Comparison of Escherichia coli-asparaginase with Erwinia-asparaginase in the treatment of childhood lymphoid malignancies: results of a randomized European Organisation for Research and Treatment of Cancer-Children's Leukemia Group phase 3 trial. Blood 99 (8): 2734-9, 2002.  [PUBMED Abstract]

  19. Moghrabi A, Levy DE, Asselin B, et al.: Results of the Dana-Farber Cancer Institute ALL Consortium Protocol 95-01 for children with acute lymphoblastic leukemia. Blood 109 (3): 896-904, 2007.  [PUBMED Abstract]

  20. Pui CH, Sandlund JT, Pei D, et al.: Improved outcome for children with acute lymphoblastic leukemia: results of Total Therapy Study XIIIB at St Jude Children's Research Hospital. Blood 104 (9): 2690-6, 2004.  [PUBMED Abstract]

  21. Schrappe M, Reiter A, Ludwig WD, et al.: Improved outcome in childhood acute lymphoblastic leukemia despite reduced use of anthracyclines and cranial radiotherapy: results of trial ALL-BFM 90. German-Austrian-Swiss ALL-BFM Study Group. Blood 95 (11): 3310-22, 2000.  [PUBMED Abstract]

  22. Balduzzi A, Valsecchi MG, Uderzo C, et al.: Chemotherapy versus allogeneic transplantation for very-high-risk childhood acute lymphoblastic leukaemia in first complete remission: comparison by genetic randomisation in an international prospective study. Lancet 366 (9486): 635-42, 2005 Aug 20-26.  [PUBMED Abstract]

  23. Silverman LB, Gelber RD, Young ML, et al.: Induction failure in acute lymphoblastic leukemia of childhood. Cancer 85 (6): 1395-404, 1999.  [PUBMED Abstract]

  24. Ogden AK, Pollock BH, Bernstein ML, et al.: Intermediate-dose methotrexate and intravenous 6-mercaptopurine chemotherapy for children with acute lymphoblastic leukemia who did not respond to initial induction therapy. J Pediatr Hematol Oncol 24 (3): 182-7, 2002 Mar-Apr.  [PUBMED Abstract]

  25. Gajjar A, Ribeiro R, Hancock ML, et al.: Persistence of circulating blasts after 1 week of multiagent chemotherapy confers a poor prognosis in childhood acute lymphoblastic leukemia. Blood 86 (4): 1292-5, 1995.  [PUBMED Abstract]

  26. Steinherz PG, Gaynon PS, Breneman JC, et al.: Cytoreduction and prognosis in acute lymphoblastic leukemia--the importance of early marrow response: report from the Childrens Cancer Group. J Clin Oncol 14 (2): 389-98, 1996.  [PUBMED Abstract]

  27. Nachman JB, Sather HN, Sensel MG, et al.: Augmented post-induction therapy for children with high-risk acute lymphoblastic leukemia and a slow response to initial therapy. N Engl J Med 338 (23): 1663-71, 1998.  [PUBMED Abstract]

  28. Pullen J, Boyett J, Shuster J, et al.: Extended triple intrathecal chemotherapy trial for prevention of CNS relapse in good-risk and poor-risk patients with B-progenitor acute lymphoblastic leukemia: a Pediatric Oncology Group study. J Clin Oncol 11 (5): 839-49, 1993.  [PUBMED Abstract]

  29. Thyss A, Suciu S, Bertrand Y, et al.: Systemic effect of intrathecal methotrexate during the initial phase of treatment of childhood acute lymphoblastic leukemia. The European Organization for Research and Treatment of Cancer Children's Leukemia Cooperative Group. J Clin Oncol 15 (5): 1824-30, 1997.  [PUBMED Abstract]

  30. Clarke M, Gaynon P, Hann I, et al.: CNS-directed therapy for childhood acute lymphoblastic leukemia: Childhood ALL Collaborative Group overview of 43 randomized trials. J Clin Oncol 21 (9): 1798-809, 2003.  [PUBMED Abstract]

  31. Tubergen DG, Gilchrist GS, O'Brien RT, et al.: Prevention of CNS disease in intermediate-risk acute lymphoblastic leukemia: comparison of cranial radiation and intrathecal methotrexate and the importance of systemic therapy: a Childrens Cancer Group report. J Clin Oncol 11 (3): 520-6, 1993.  [PUBMED Abstract]

  32. Conter V, Aricò M, Valsecchi MG, et al.: Extended intrathecal methotrexate may replace cranial irradiation for prevention of CNS relapse in children with intermediate-risk acute lymphoblastic leukemia treated with Berlin-Frankfurt-Münster-based intensive chemotherapy. The Associazione Italiana di Ematologia ed Oncologia Pediatrica. J Clin Oncol 13 (10): 2497-502, 1995.  [PUBMED Abstract]

  33. Matloub Y, Lindemulder S, Gaynon PS, et al.: Intrathecal triple therapy decreases central nervous system relapse but fails to improve event-free survival when compared with intrathecal methotrexate: results of the Children's Cancer Group (CCG) 1952 study for standard-risk acute lymphoblastic leukemia, reported by the Children's Oncology Group. Blood 108 (4): 1165-73, 2006.  [PUBMED Abstract]

  34. Mahmoud HH, Rivera GK, Hancock ML, et al.: Low leukocyte counts with blast cells in cerebrospinal fluid of children with newly diagnosed acute lymphoblastic leukemia. N Engl J Med 329 (5): 314-9, 1993.  [PUBMED Abstract]

  35. Bürger B, Zimmermann M, Mann G, et al.: Diagnostic cerebrospinal fluid examination in children with acute lymphoblastic leukemia: significance of low leukocyte counts with blasts or traumatic lumbar puncture. J Clin Oncol 21 (2): 184-8, 2003.  [PUBMED Abstract]

  36. Gajjar A, Harrison PL, Sandlund JT, et al.: Traumatic lumbar puncture at diagnosis adversely affects outcome in childhood acute lymphoblastic leukemia. Blood 96 (10): 3381-4, 2000.  [PUBMED Abstract]

  37. Schrappe M, Reiter A, Riehm H: Prophylaxis and treatment of neoplastic meningeosis in childhood acute lymphoblastic leukemia. J Neurooncol 38 (2-3): 159-65, 1998 Jun-Jul.  [PUBMED Abstract]

  38. Laver JH, Barredo JC, Amylon M, et al.: Effects of cranial radiation in children with high risk T cell acute lymphoblastic leukemia: a Pediatric Oncology Group report. Leukemia 14 (3): 369-73, 2000.  [PUBMED Abstract]

  39. Nathan PC, Maze R, Spiegler B, et al.: CNS-directed therapy in young children with T-lineage acute lymphoblastic leukemia: High-dose methotrexate versus cranial irradiation. Pediatr Blood Cancer 42 (1): 24-9, 2004.  [PUBMED Abstract]

  40. Moore IM, Espy KA, Kaufmann P, et al.: Cognitive consequences and central nervous system injury following treatment for childhood leukemia. Semin Oncol Nurs 16 (4): 279-90; discussion 291-9, 2000.  [PUBMED Abstract]

  41. Goshen Y, Stark B, Kornreich L, et al.: High incidence of meningioma in cranial irradiated survivors of childhood acute lymphoblastic leukemia. Pediatr Blood Cancer 49 (3): 294-7, 2007.  [PUBMED Abstract]

  42. Eiser C, Tillmann V: Learning difficulties in children treated for acute lymphoblastic leukaemia (ALL). Pediatr Rehabil 4 (3): 105-18, 2001 Jul-Sep.  [PUBMED Abstract]

  43. Stubberfield TG, Byrne GC, Jones TW: Growth and growth hormone secretion after treatment for acute lymphoblastic leukemia in childhood. 18-Gy versus 24-Gy cranial irradiation. J Pediatr Hematol Oncol 17 (2): 167-71, 1995.  [PUBMED Abstract]

  44. Rowland JH, Glidewell OJ, Sibley RF, et al.: Effects of different forms of central nervous system prophylaxis on neuropsychologic function in childhood leukemia. J Clin Oncol 2 (12): 1327-35, 1984.  [PUBMED Abstract]

  45. Halberg FE, Kramer JH, Moore IM, et al.: Prophylactic cranial irradiation dose effects on late cognitive function in children treated for acute lymphoblastic leukemia. Int J Radiat Oncol Biol Phys 22 (1): 13-6, 1992.  [PUBMED Abstract]

  46. Hill JM, Kornblith AB, Jones D, et al.: A comparative study of the long term psychosocial functioning of childhood acute lymphoblastic leukemia survivors treated by intrathecal methotrexate with or without cranial radiation. Cancer 82 (1): 208-18, 1998.  [PUBMED Abstract]

  47. Pui CH, Cheng C, Leung W, et al.: Extended follow-up of long-term survivors of childhood acute lymphoblastic leukemia. N Engl J Med 349 (7): 640-9, 2003.  [PUBMED Abstract]

  48. Jankovic M, Brouwers P, Valsecchi MG, et al.: Association of 1800 cGy cranial irradiation with intellectual function in children with acute lymphoblastic leukaemia. ISPACC. International Study Group on Psychosocial Aspects of Childhood Cancer. Lancet 344 (8917): 224-7, 1994.  [PUBMED Abstract]

  49. Sklar C, Mertens A, Walter A, et al.: Final height after treatment for childhood acute lymphoblastic leukemia: comparison of no cranial irradiation with 1800 and 2400 centigrays of cranial irradiation. J Pediatr 123 (1): 59-64, 1993.  [PUBMED Abstract]

  50. Christie D, Leiper AD, Chessells JM, et al.: Intellectual performance after presymptomatic cranial radiotherapy for leukaemia: effects of age and sex. Arch Dis Child 73 (2): 136-40, 1995.  [PUBMED Abstract]

  51. Waber DP, Tarbell NJ, Kahn CM, et al.: The relationship of sex and treatment modality to neuropsychologic outcome in childhood acute lymphoblastic leukemia. J Clin Oncol 10 (5): 810-7, 1992.  [PUBMED Abstract]

  52. Löning L, Zimmermann M, Reiter A, et al.: Secondary neoplasms subsequent to Berlin-Frankfurt-Münster therapy of acute lymphoblastic leukemia in childhood: significantly lower risk without cranial radiotherapy. Blood 95 (9): 2770-5, 2000.  [PUBMED Abstract]

  53. Waber DP, Shapiro BL, Carpentieri SC, et al.: Excellent therapeutic efficacy and minimal late neurotoxicity in children treated with 18 grays of cranial radiation therapy for high-risk acute lymphoblastic leukemia: a 7-year follow-up study of the Dana-Farber Cancer Institute Consortium Protocol 87-01. Cancer 92 (1): 15-22, 2001.  [PUBMED Abstract]

  54. Hata M, Ogino I, Aida N, et al.: Prophylactic cranial irradiation of acute lymphoblastic leukemia in childhood: outcomes of late effects on pituitary function and growth in long-term survivors. Int J Cancer 96 (Suppl): 117-24, 2001.  [PUBMED Abstract]

  55. Waber DP, Silverman LB, Catania L, et al.: Outcomes of a randomized trial of hyperfractionated cranial radiation therapy for treatment of high-risk acute lymphoblastic leukemia: therapeutic efficacy and neurotoxicity. J Clin Oncol 22 (13): 2701-7, 2004.  [PUBMED Abstract]

  56. Waber DP, Turek J, Catania L, et al.: Neuropsychological outcomes from a randomized trial of triple intrathecal chemotherapy compared with 18 Gy cranial radiation as CNS treatment in acute lymphoblastic leukemia: findings from Dana-Farber Cancer Institute ALL Consortium Protocol 95-01. J Clin Oncol 25 (31): 4914-21, 2007.  [PUBMED Abstract]

  57. Relling MV, Pui CH, Sandlund JT, et al.: Adverse effect of anticonvulsants on efficacy of chemotherapy for acute lymphoblastic leukaemia. Lancet 356 (9226): 285-90, 2000.  [PUBMED Abstract]

  58. Jansen NC, Kingma A, Schuitema A, et al.: Post-treatment intellectual functioning in children treated for acute lymphoblastic leukaemia (ALL) with chemotherapy-only: a prospective, sibling-controlled study. Eur J Cancer 42 (16): 2765-72, 2006.  [PUBMED Abstract]

  59. Espy KA, Moore IM, Kaufmann PM, et al.: Chemotherapeutic CNS prophylaxis and neuropsychologic change in children with acute lymphoblastic leukemia: a prospective study. J Pediatr Psychol 26 (1): 1-9, 2001 Jan-Feb.  [PUBMED Abstract]

  60. Copeland DR, Moore BD 3rd, Francis DJ, et al.: Neuropsychologic effects of chemotherapy on children with cancer: a longitudinal study. J Clin Oncol 14 (10): 2826-35, 1996.  [PUBMED Abstract]

  61. von der Weid N, Mosimann I, Hirt A, et al.: Intellectual outcome in children and adolescents with acute lymphoblastic leukaemia treated with chemotherapy alone: age- and sex-related differences. Eur J Cancer 39 (3): 359-65, 2003.  [PUBMED Abstract]

  62. Mahoney DH Jr, Shuster JJ, Nitschke R, et al.: Acute neurotoxicity in children with B-precursor acute lymphoid leukemia: an association with intermediate-dose intravenous methotrexate and intrathecal triple therapy--a Pediatric Oncology Group study. J Clin Oncol 16 (5): 1712-22, 1998.  [PUBMED Abstract]

  63. Waber DP, Carpentieri SC, Klar N, et al.: Cognitive sequelae in children treated for acute lymphoblastic leukemia with dexamethasone or prednisone. J Pediatr Hematol Oncol 22 (3): 206-13, 2000 May-Jun.  [PUBMED Abstract]

Back to Top

< Previous Section  |  Next Section >


A Service of the National Cancer Institute
Department of Health and Human Services National Institutes of Health USA.gov