National Cancer Institute National Cancer Institute
U.S. National Institutes of Health National Cancer Institute
NCI Home Cancer Topics Clinical Trials Cancer Statistics Research & Funding News About NCI
Childhood Acute Myeloid Leukemia/Other Myeloid Malignancies Treatment (PDQ®)
Patient VersionHealth Professional VersionEn españolLast Modified: 08/18/2008



Purpose of This PDQ Summary






General Information






Classification of Pediatric Myeloid Malignancies






Stage Information






Treatment Overview for Acute Myeloid Leukemia






Treatment of Newly Diagnosed Acute Myeloid Leukemia






Postremission Therapy for Acute Myeloid Leukemia







Acute Promyelocytic Leukemia






Children With Down Syndrome






Myelodysplastic Syndromes






Juvenile Myelomonocytic Leukemia






Chronic Myelogenous Leukemia






Recurrent Childhood Acute Myeloid Leukemia






Survivorship and Adverse Late Sequelae






Get More Information From NCI






Changes to This Summary (08/18/2008)






More Information



Page Options
Print This Page  Print This Page
Print This Document  Print Entire Document
View Entire Document  View Entire Document
E-Mail This 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
NCI Highlights
Virtual and Standard Colonoscopy Both Accurate

New Study of Targeted Therapies for Breast Cancer

The Nation's Investment in Cancer Research FY 2009

Cancer Trends Progress Report: 2007 Update

Past Highlights
You CAN Quit Smoking Now!
Acute Promyelocytic Leukemia

Current Clinical Trials

Acute promyelocytic leukemia (APL) is a distinct subtype of acute myeloid leukemia (AML) and is treated differently than other types of AML. The characteristic chromosomal abnormality associated with APL is t(15;17). This translocation involves a breakpoint that includes the retinoid acid receptor and that leads to production of the PML/RARA fusion protein.[1]

Clinically, APL is commonly characterized by a severe coagulopathy often present at the time of diagnosis.[2] Mortality during induction due to bleeding complications is more common in this subtype than other French-American-British classifications. Because of the extremely low incidence of central nervous system disease in patients with APL, a lumbar puncture is not required at the time of diagnosis and prophylactic intrathecal chemotherapy is not administered. Studies have demonstrated that the absence of PML/RARA RNA chimeric transcript expression at the end of therapy, as detected by reverse-transcription–polymerase chain reaction monitoring, predicts a low risk of relapse.[3-5]

Some molecular variants are generally believed to be resistant to treatment with all-trans retinoic acid (ATRA). One such variant, characterized by t(11;17), represents about 0.8% of APL, expresses surface CD56 and has very fine granules compared to t(15;17) APL.[6-8] The t(11;17) variant has been associated with a poor prognosis. Although intensive multiagent chemotherapy regimens are usually used, novel approaches to therapy are needed for this rare but poor-prognosis subgroup of patients. Of interest, though believed to be refractory to ATRA, remissions have been achieved in these patients using ATRA plus granulocyte-macrophage colony-stimulating factor or ATRA plus chemotherapy.[9]

The leukemia cells from patients with APL are especially sensitive to the differentiation-inducing effects of ATRA. The basis for the dramatic efficacy of ATRA against APL is the ability of pharmacologic doses of ATRA to overcome the repression of signaling caused by the PML/RARA fusion protein at physiologic ATRA concentrations. Restoration of signaling leads to differentiation of APL cells and then to postmaturation apoptosis.[10] Most patients with APL achieve a complete remission (CR) when treated with ATRA, though single-agent ATRA is generally not curative.[11,12] A series of randomized clinical trials have defined the benefit for combining ATRA with chemotherapy during induction therapy and also the utility of using ATRA as maintenance therapy.[13-15] For children with APL, survival rates exceeding 80% are now achievable.[16-18]

APL in children is generally similar to APL in adults, though children have a higher incidence of hyperleukocytosis (defined as white blood cell [WBC] count greater than 10 x 109/L) and a higher incidence of the microgranular morphologic subtype.[16-19] Similar to adults, children with WBC count less than 10 x 109/L at diagnosis have significantly better outcome than patients with higher WBC count.[17,18,20] The North American approach to treating children with APL utilizes induction therapy with ATRA, and standard-dose cytarabine and daunorubicin, followed by consolidation therapy with ATRA and daunorubicin (CALGB-C9710). Maintenance therapy, especially for high-risk patients, includes ATRA plus 6-mercaptopurine and methotrexate; this combination showed an advantage over ATRA alone in randomized trials in adults.[13,21] European clinical trials groups (Gruppo Italiano Malattie Ematologiche Maligne dell' Adulto–Associazione Italiana Ematologia ed Ocologia Pediatrica [GIMEMA–AIEOP] and Programa de Estudio y Tratamiento de las Hemopatias Malignas [PETHEMA]) have utilized idarubicin and ATRA without cytarabine for remission induction for children with APL.[17,18] Subsequent therapies for these groups include treatment courses with an anthracycline (idarubicin and mitoxantrone) plus ATRA (PETHEMA) or treatment courses with an anthracycline, ATRA, and other agents (GIMEMA-AIEOP), with both groups utilizing maintenance therapy as described above.[17,18] Because of the positive results of the use of chemotherapy plus ATRA, hematopoietic stem cell transplatation is not recommended in first CR but only following relapse and achievement of a second CR.

Arsenic trioxide has also been identified as an active agent in patients with APL with approximately 85% of patients achieving remission following treatment with this agent in relapse.[22-25] Data are limited on the use of arsenic trioxide in children, though published reports suggest that children with relapsed APL have a response to arsenic trioxide similar to that of adults.[22,24,26,27] A recent study in China randomized adults and older children (14 years or older) with newly diagnosed APL to induction with ATRA alone, arsenic trioxide alone, or a combination of the two, followed by consolidation and maintenance. While the CR rate was high (>90%) in all three groups, the combination group demonstrated greater reduction of PML/RARA transcripts at CR and better disease-free survival (100% of 20 patients) with a median follow-up of 18 months.[25] Although based on relatively small numbers of patients, these preliminary results are intriguing and may serve as the basis for future investigation with arsenic trioxide and ATRA combinations in trials for patients with newly diagnosed APL.[28] Because arsenic trioxide causes Q-T interval prolongation that can lead to life-threatening arrhythmias (e.g., torsades de pointes),[29] it is essential to monitor electrolytes closely in patients receiving arsenic trioxide and to maintain potassium and magnesium values at midnormal ranges.[30]

Current Clinical Trials

Check for U.S. clinical trials from NCI's PDQ Cancer Clinical Trials Registry that are now accepting patients with childhood acute promyelocytic leukemia (M3). 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. Melnick A, Licht JD: Deconstructing a disease: RARalpha, its fusion partners, and their roles in the pathogenesis of acute promyelocytic leukemia. Blood 93 (10): 3167-215, 1999.  [PUBMED Abstract]

  2. Tallman MS, Hakimian D, Kwaan HC, et al.: New insights into the pathogenesis of coagulation dysfunction in acute promyelocytic leukemia. Leuk Lymphoma 11 (1-2): 27-36, 1993.  [PUBMED Abstract]

  3. Gameiro P, Vieira S, Carrara P, et al.: The PML-RAR alpha transcript in long-term follow-up of acute promyelocytic leukemia patients. Haematologica 86 (6): 577-85, 2001.  [PUBMED Abstract]

  4. Jurcic JG, Nimer SD, Scheinberg DA, et al.: Prognostic significance of minimal residual disease detection and PML/RAR-alpha isoform type: long-term follow-up in acute promyelocytic leukemia. Blood 98 (9): 2651-6, 2001.  [PUBMED Abstract]

  5. Hu J, Yu T, Zhao W, et al.: Impact of RT-PCR monitoring on the long-term survival in acute promyelocytic leukemia. Chin Med J (Engl) 113 (10): 899-902, 2000.  [PUBMED Abstract]

  6. Licht JD, Chomienne C, Goy A, et al.: Clinical and molecular characterization of a rare syndrome of acute promyelocytic leukemia associated with translocation (11;17). Blood 85 (4): 1083-94, 1995.  [PUBMED Abstract]

  7. Guidez F, Ivins S, Zhu J, et al.: Reduced retinoic acid-sensitivities of nuclear receptor corepressor binding to PML- and PLZF-RARalpha underlie molecular pathogenesis and treatment of acute promyelocytic leukemia. Blood 91 (8): 2634-42, 1998.  [PUBMED Abstract]

  8. Grimwade D, Biondi A, Mozziconacci MJ, et al.: Characterization of acute promyelocytic leukemia cases lacking the classic t(15;17): results of the European Working Party. Groupe Français de Cytogénétique Hématologique, Groupe de Français d'Hematologie Cellulaire, UK Cancer Cytogenetics Group and BIOMED 1 European Community-Concerted Action "Molecular Cytogenetic Diagnosis in Haematological Malignancies". Blood 96 (4): 1297-308, 2000.  [PUBMED Abstract]

  9. Sirulnik A, Melnick A, Zelent A, et al.: Molecular pathogenesis of acute promyelocytic leukaemia and APL variants. Best Pract Res Clin Haematol 16 (3): 387-408, 2003.  [PUBMED Abstract]

  10. Altucci L, Rossin A, Raffelsberger W, et al.: Retinoic acid-induced apoptosis in leukemia cells is mediated by paracrine action of tumor-selective death ligand TRAIL. Nat Med 7 (6): 680-6, 2001.  [PUBMED Abstract]

  11. Huang ME, Ye YC, Chen SR, et al.: Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia. Blood 72 (2): 567-72, 1988.  [PUBMED Abstract]

  12. Castaigne S, Chomienne C, Daniel MT, et al.: All-trans retinoic acid as a differentiation therapy for acute promyelocytic leukemia. I. Clinical results. Blood 76 (9): 1704-9, 1990.  [PUBMED Abstract]

  13. Fenaux P, Chastang C, Chevret S, et al.: A randomized comparison of all transretinoic acid (ATRA) followed by chemotherapy and ATRA plus chemotherapy and the role of maintenance therapy in newly diagnosed acute promyelocytic leukemia. The European APL Group. Blood 94 (4): 1192-200, 1999.  [PUBMED Abstract]

  14. Fenaux P, Chevret S, Guerci A, et al.: Long-term follow-up confirms the benefit of all-trans retinoic acid in acute promyelocytic leukemia. European APL group. Leukemia 14 (8): 1371-7, 2000.  [PUBMED Abstract]

  15. Tallman MS, Andersen JW, Schiffer CA, et al.: All-trans-retinoic acid in acute promyelocytic leukemia. N Engl J Med 337 (15): 1021-8, 1997.  [PUBMED Abstract]

  16. de Botton S, Coiteux V, Chevret S, et al.: Outcome of childhood acute promyelocytic leukemia with all-trans-retinoic acid and chemotherapy. J Clin Oncol 22 (8): 1404-12, 2004.  [PUBMED Abstract]

  17. Testi AM, Biondi A, Lo Coco F, et al.: GIMEMA-AIEOPAIDA protocol for the treatment of newly diagnosed acute promyelocytic leukemia (APL) in children. Blood 106 (2): 447-53, 2005.  [PUBMED Abstract]

  18. Ortega JJ, Madero L, Martín G, et al.: Treatment with all-trans retinoic acid and anthracycline monochemotherapy for children with acute promyelocytic leukemia: a multicenter study by the PETHEMA Group. J Clin Oncol 23 (30): 7632-40, 2005.  [PUBMED Abstract]

  19. Guglielmi C, Martelli MP, Diverio D, et al.: Immunophenotype of adult and childhood acute promyelocytic leukaemia: correlation with morphology, type of PML gene breakpoint and clinical outcome. A cooperative Italian study on 196 cases. Br J Haematol 102 (4): 1035-41, 1998.  [PUBMED Abstract]

  20. Sanz MA, Lo Coco F, Martín G, et al.: Definition of relapse risk and role of nonanthracycline drugs for consolidation in patients with acute promyelocytic leukemia: a joint study of the PETHEMA and GIMEMA cooperative groups. Blood 96 (4): 1247-53, 2000.  [PUBMED Abstract]

  21. Sanz M, Martínez JA, Barragán E, et al.: All-trans retinoic acid and low-dose chemotherapy for acute promyelocytic leukaemia. Br J Haematol 109 (4): 896-7, 2000.  [PUBMED Abstract]

  22. Soignet SL, Maslak P, Wang ZG, et al.: Complete remission after treatment of acute promyelocytic leukemia with arsenic trioxide. N Engl J Med 339 (19): 1341-8, 1998.  [PUBMED Abstract]

  23. Niu C, Yan H, Yu T, et al.: Studies on treatment of acute promyelocytic leukemia with arsenic trioxide: remission induction, follow-up, and molecular monitoring in 11 newly diagnosed and 47 relapsed acute promyelocytic leukemia patients. Blood 94 (10): 3315-24, 1999.  [PUBMED Abstract]

  24. Shen ZX, Chen GQ, Ni JH, et al.: Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients. Blood 89 (9): 3354-60, 1997.  [PUBMED Abstract]

  25. Shen ZX, Shi ZZ, Fang J, et al.: All-trans retinoic acid/As2O3 combination yields a high quality remission and survival in newly diagnosed acute promyelocytic leukemia. Proc Natl Acad Sci U S A 101 (15): 5328-35, 2004.  [PUBMED Abstract]

  26. Zhang P: The use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia. J Biol Regul Homeost Agents 13 (4): 195-200, 1999 Oct-Dec.  [PUBMED Abstract]

  27. Shigeno K, Naito K, Sahara N, et al.: Arsenic trioxide therapy in relapsed or refractory Japanese patients with acute promyelocytic leukemia: updated outcomes of the phase II study and postremission therapies. Int J Hematol 82 (3): 224-9, 2005.  [PUBMED Abstract]

  28. Wang G, Li W, Cui J, et al.: An efficient therapeutic approach to patients with acute promyelocytic leukemia using a combination of arsenic trioxide with low-dose all-trans retinoic acid. Hematol Oncol 22 (2): 63-71, 2004.  [PUBMED Abstract]

  29. Unnikrishnan D, Dutcher JP, Varshneya N, et al.: Torsades de pointes in 3 patients with leukemia treated with arsenic trioxide. Blood 97 (5): 1514-6, 2001.  [PUBMED Abstract]

  30. Barbey JT: Cardiac toxicity of arsenic trioxide. Blood 98 (5): 1632; discussion 1633-4, 2001.  [PUBMED Abstract]

Back to TopBack 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