Myelodysplastic syndromes (MDS) are a heterogenous

INVITED REVIEW ARTICLE Pediatric Myelodysplastic Syndromes: They Do Exist! Taly Glaubach, MD,*w Lisa J. Robinson, MD,z and Seth J. Corey, MD, MPH*wy ...
0 downloads 2 Views 170KB Size
INVITED REVIEW ARTICLE

Pediatric Myelodysplastic Syndromes: They Do Exist! Taly Glaubach, MD,*w Lisa J. Robinson, MD,z and Seth J. Corey, MD, MPH*wy

Summary: One of the most common hematologic malignancies in adults, myelodysplastic syndrome (MDS) is a heterogenous group of clonal disorders characterized by peripheral cytopenia(s) and normal or hypercellular bone marrow with dysplasia in Z1 blood cell lineages. MDS frequently evolves to secondary acute myeloid leukemia with poor prognosis. Although uncommon among pediatric hematologic malignancies, both de novo and secondary MDS occur in children and may be the first presentation of an inherited bone marrow failure syndrome. Unlike its adult counterpart, pediatric MDS is more frequently associated with hypocellular bone marrow and monosomy 7. Refractory cytopenia is more typical than refractory anemia, as seen in the elderly. Its recognition and management can be quite challenging and requires the expertise of an experienced hematopathologist. In this review, we describe the epidemiology, genetics, and clinical spectrum of pediatric MDS along with its diagnostic and therapeutic challenges. We also compare and contrast pediatric and adult MDS. Key Words: myelodysplastic syndromes, bone marrow failure syndromes, aplastic anemia, myeloid leukemia

(J Pediatr Hematol Oncol 2014;36:1–7)

M

yelodysplastic syndromes (MDS) are a heterogenous group of clonal disorders characterized by peripheral cytopenia(s) and normal or hypercellular bone marrow with dysplasia in Z1 blood cell lineages. Some are well-recognized distinct entities, for example, refractory anemia with ring sideroblasts, del (5q), and therapy related. MDS frequently evolves to secondary acute myeloid leukemia (AML) with poor prognosis. Its pathobiology is still not well understood, which has hampered the advancement of diagnostic and therapeutic strategies. Recent drug approvals for MDS include lenalidomide for del (5q) syndrome and the hypomethylating agents azacitidine and decitabine. Although MDS is one of the most common hematopoietic malignancies in older adults with a median age of 70 years at diagnosis, it is less frequently diagnosed in pediatrics (for a comparison of features see Table 1). In children and adolescents, MDS occurs in both de novo and secondary forms, but often presents a unique diagnostic challenge for the clinician. Secondary MDS follows either after genotoxic Received for publication August 13, 2013; accepted September 10, 2013. From the *Department of Pediatrics, Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago; wRobert H. Lurie Comprehensive Cancer Center, Chicago, IL; zDepartment of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA; and yDepartment of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL. T.G. is supported by the ASH Research Training Award and the MDS Foundation Young Investigator Grant. The remaining authors declare no conflict of interest. Reprints: Seth J. Corey, MD, MPH, Lurie 5-107, Robert H. Lurie Comprehensive Cancer Center, 303 E. Superior Street, Chicago, IL 60611 (e-mail: [email protected]). Copyright r 2013 by Lippincott Williams & Wilkins

J Pediatr Hematol Oncol



therapy or inherited bone marrow failure syndromes (IBMFS). Juvenile myelomonocytic leukemia (JMML) represents a special form of myelodysplastic and myeloproliferative disorder. Pediatric MDS may be underdiagnosed due to lack of validated, pediatric-specific diagnostic criteria or understanding of the natural history of refractory cytopenias. As little is known about the natural history of MDS in pediatrics, the decision when to transplant may be difficult.

EPIDEMIOLOGY Although the true incidence of pediatric MDS has been difficult to ascertain, best estimates come from large population-based studies in Europe and Canada. The estimated incidence is approximately 1.8 to 4 cases per million per year, with the lower end of the range estimated from exclusion of patients with Down syndrome.1,2 Data published from the United Kingdom suggest a lower annual incidence of 0.8 cases per million, but included only primary MDS.3 In contrast, the incidence of AML and ALL is approximately 6 and 42 cases per million, respectively. The median age at presentation of pediatric MDS is 6.8 years and seems to be equally distributed among males and females.1–7 In the only large published series of advanced or high-risk pediatric MDS, the median age was higher at 10.7 years with a 2:1 male predominance.8 Secondary pediatric MDS is strongly associated with IBMFS, such as Fanconi anemia, Shwachman-Diamond syndrome, severe congenital neutropenia, dyskeratosis congenita, and MonoMAC syndrome.1,9–20 Other genetic conditions associated with MDS include paroxysmal nocturnal hemoglobinuria (PNH), monosomy 7 syndrome, Down syndrome, neurofibromatosis, Bloom syndrome, and Li-Fraumeni syndrome.1,9–20 Some will display MDS as the first presenting sign of that disorder. As our recognition and genetic-based diagnosis of these rare genetic disorders advance, it is likely that this number will increase and the corresponding incidence of purported de novo pediatric MDS will decrease, respectively. In addition, acquired severe aplastic anemia (SAA) infrequently evolves into secondary MDS. However, resolving the distinction between SAA and MDS significantly reduces the number of secondary MDS and AML after immunosuppressive therapy (IST) for SAA.21,22

CLINICAL PRESENTATION About 20% of pediatric MDS is found incidentally on routine laboratory evaluation7 or during evaluation for a suspected bone marrow failure syndrome. More commonly, it presents with symptoms related to the cytopenias(s), such as fatigue, fever, infection, and bleeding.7 In contrast to adults, pediatric MDS often presents with bilineage cytopenias and very rarely with isolated anemia (ie, neutropenia and/or thrombocytopenia, with/without macrocytic anemia).7,23 Isolated 5q  or del (5q) syndrome common in

Volume 36, Number 1, January 2014

www.jpho-online.com |

1

J Pediatr Hematol Oncol

Glaubach et al



Volume 36, Number 1, January 2014

TABLE 1. Comparison of Common Features of Adult and Pediatric MDS

Adult MDS

Pediatric MDS

3-5/105 > 20/105 in those >70 y 70 Isolated, transfusion-dependent anemia, with/ without neutropenia and/or thrombocytopenia

Incidence Median age (y) Presentation Etiology Morphologic subtype BM cellularity

Primary (de novo) most common RARS and 5q  most common Hypercellular or normocellular; hypocellular is rare 5/5q > 7/7q , 3q, 20q anomalies Mutations in DNMT3a, ASXL1, TET2, SF3B1, U2AF35; methylation changes

Cytogenetics Genetics Physical findings

None

Therapeutic options

RBC and platelet transfusions Lenalidomide for del (5q) Azacitidine, decitabine HSCT for those with low risk for morbidity and mortality

1.8-4/106 7 Bilineage cytopenias most common; refractory thrombocytopenia >neutropenia and/or anemia Secondary or therapy related most common RCC most common; RARS or 5q  rare Variable; hypocellular is most common Monosomy 7/7q  > trisomy 8 FANC members, SBDS, DKC, TERT, TERC, ELANE, HAX1, WAS, GATA-2 (related to IBMFS) Skeletal, cutaneous, genitourinary, cardiovascular, and gastrointestinal anomalies (related to IBMFS) Monitoring RBC and platelet transfusions HSCT ?Lenalidomide ?Hypomethylating agents ?IST

HSCT indicates hematopoietic stem cell transplant; IBMFS, inherited bone marrow failure syndromes; IST, immunosuppressive therapy; MDS, myelodysplastic syndrome; RARS, refractory anemia with ring sideroblasts; RBC, red blood cells; RCC, refractory cytopenia of childhood; ?, unknown efficacy.

adults, is almost never seen in children.24,25 These findings are collectively reflected in the current World Health Organization (WHO) nomenclature of “refractory cytopenia of childhood” (RCC), as opposed to “refractory anemia.” RCC is the most common subtype of pediatric MDS, accounting for approximately 50% of cases, with the remainder being indistinguishable from “adult-type” refractory anemia with excess blast (RAEB) or RAEBT.3,26,27

GENETICS Clonal analyses of karyotype and gene mutations revealed that pediatric MDS differs from adult MDS. Karyotype abnormalities are common, occurring in 30% to 50% of pediatric MDS,28 with the majority consisting of copy number variations in all or part of the chromosome. In contrast to abnormalities involving chromosome 5 in adult MDS, monosomy 7 is the most common cytogenetic abnormality in pediatric MDS and occurs in about 30% of patients.28 Other less common but recurrent cytogenetic abnormalities are trisomies 8 and 21 (which may be constitutional, mosaic, or somatically acquired), and the loss of part of chromosome 20 (eg, 20q). Complex karyotypes tend to signify a high-risk population.28 Development and refinement of single-nucleotide polymorphism arrays have allowed the identification of additional microdeletions below the resolution of standard cytogenetic assays, and can reveal previously undetectable regions of copy number—neutral loss of heterozygosity, or uniparental disomy. Understanding noncoding regions of DNA, identification of noncoding RNA, and description of global methylation and histone modification changes (epigenetics) are adding to the complexity of an emerging story for MDS in adults. Recurrent mutations affecting genes that regulate

2 | www.jpho-online.com

DNA methylation (DNMT3A, IDH1/IDH2, and TET2), histone function (EZH2 and ASXL1), and splicing machinery (U2AF35, ZRSR2, SRSF2, and SF3B1) have been identified in adult patients with MDS.29–31 Interestingly, these mutations are rare in pediatric de novo or secondary MDS. Several studies have found splicing factor mutations in only 1 of 187 pediatric MDS patients who had RAEBT,32 and in none of 28 patients with MDS,33 after excluding JMML which accounted for an additional 3 of 142 total JMML patients (combined data). However, even in the identified cases, the SRSF2 mutation did not seem to serve as a driver.33 It was always accompanied by other known oncogenic driver mutations (PTPN11 or NRAS), disappeared during disease progression, and was not present at the time of multiple relapses. To date, mutations in genes that regulate DNA methylation and histone function have not yet been established in pediatric MDS. Exactly how these genetic and epigenetic changes lead to disease have yet to be elucidated, but is an area of active research.

Rare Yet Novel MDS Genes and Associated Syndromes (RUNX1, CEBPA, and GATA2) Although very rare, familial forms of nonsyndromic MDS and AML have been described and 2 genes identified: RUNX1/AML1 in familial platelet disorder with a predisposition to acute myelogenous leukemia and CEBPA in familial AML.34,35 Recently, germline missense mutations in GATA2 were identified in 4 distinct families with multigenerational early-onset MDS/AML in an autosomal dominant manner.36 These mutations result in a dominantnegative loss of function through disruption of protein interactions necessary for DNA binding and activation of target genes. In these patients, the MDS subtype and clinical course were variable. r

2013 Lippincott Williams & Wilkins

J Pediatr Hematol Oncol



Volume 36, Number 1, January 2014

Pediatric Myelodysplastic Syndromes

In addition, recurrent germline mutations in GATA2 have also been described in syndromic forms of familial and sporadic MDS. The MonoMAC syndrome is an immunodeficiency syndrome characterized by marked monocytopenia, and B-cell and NK-cell lymphopenia, with resultant susceptibility to mycobacterial infections, human papillomavirus, and other opportunistic viral and fungal infections. The constellation of features also includes a predisposition to MDS/AML, and/or pulmonary alveolar proteinosis.14,37,38 In Emberger syndrome, GATA2 mutations are associated with lymphedema and MDS/AML.39 As more patients are sequenced for GATA2 mutations, we will learn more about the range of GATA2 mutations and their associated MDS phenotypes.

DIAGNOSTIC CHALLENGE Consideration of pediatric MDS should occur when thrombocytopenia, neutropenia, and/or anemia has persisted for 3 months in an otherwise healthy child or adolescent. Closer inspection might reveal the physical anomalies associated with Fanconi anemia, ShwachmanDiamond syndrome, or dyskeratosis congenita. The presence of warts might suggest Mono-Mac syndrome. Subtle clues also include the presence of macrocytic anemia with normal vitamin B12 and folate levels. Dysplastic granules or abnormal nuclear segmentation in circulating neutrophils would also raise suspicion. Diagnosis depends greatly on bone marrow aspirate and biopsy. MDS is largely a diagnosis of morphology and thus subject to interpretation, interobserver variability in diagnosis, and sampling error if significant hypocellularity exists. Morphologic evaluations of peripheral blood and of bone marrow are important components in the evaluation of pediatric myelodysplasia. The overall spectrum of morphologic change found in pediatric MDS is similar to that of adult MDS, but the frequency and clinical significance of specific abnormalities can differ. Blast cell numbers are a critical morphologic parameter for identifying and classifying MDS. For both adults and children, a blast percentage of at least 5% (but 6

1

1.5

2-5 Good 8-10 50-99 < 800

< 50

Good

Intermediate

2

3

5-10

> 10

Intermediate

4

Poor Very poor

3 double including -7/ abnormalities del(7q), complex: 3 abnormalities

Risk Category Very low Low Intermediate High Very high

This table is meant purely as a point of reference for the discussions in the text of this manuscript. We caution its use in pediatric therapeutic or management strategies. From Greenberg et al.55 Adaptations are themselves works protected by copyright. So in order to publish this adaptation, authorization must be obtained both from the owner of the copyright in the original work and from the owner of copyright in the translation or adaptation.

However, more studies are needed to better characterize these patterns in pediatric MDS to assess the clinical utility of these agents as a therapeutic strategy. Furthermore, the likelihood of these agents providing lifelong durability of response will need to be weighed against the curative intent of HSCT.

CONCLUSIONS The incidence of pediatric MDS is underestimated and diagnosis requires index of suspicion and an experienced hematopathologist. All pediatric patients with suspected MDS or aplastic anemia should have a bone marrow evaluation with routine cytogenetics, in addition to FISH for common MDS-associated cytogenetic anomalies as these 2 modalities are complimentary. In addition, these patients should have a workup to rule out occult PNH or IBMFS (Fanconi anemia, Shwachman-Diamond syndrome, dyskeratosis congenita, etc.) that may have MDS or aplastic anemia as its first presentation. The high rates of MDS/AML in associated congenital syndromes, including pediatric IBMFS, may provide meaningful insights into MDS pathogenesis and mechanisms of disease. However, for primary pediatric MDS, because of their infrequency, the lack of consensus on their classification, and the incomplete understanding of their biology and natural history, therapeutic advances have been stymied. We call upon the Children’s Oncology Group, the European Working Group on MDS, and the Japan Aplastic Anemia Study Group to convene and form an international pediatric consortium to study the biology and therapy of primary MDS. REFERENCES 1. Hasle H, Kerndrup G, Jacobsen BB. Childhood myelodysplastic syndrome in Denmark: incidence and predisposing conditions. Leukemia. 1995;9:1569–1572. r

2013 Lippincott Williams & Wilkins

2. Hasle H, Wadsworth LD, Massing BG, et al. A populationbased study of childhood myelodysplastic syndrome in British Columbia, Canada. Br J Haematol. 1999;106:1027–1032. 3. Passmore SJ, Chessells JM, Kempski H, et al. Paediatric myelodysplastic syndromes and juvenile myelomonocytic leukaemia in the UK: a population-based study of incidence and survival. Br J Haematol. 2003;121:758–767. 4. Jackson GH, Carey PJ, Cant AJ, et al. Myelodysplastic syndromes in children. Br J Haematol. 1993;84:185–186. 5. Luna-Fineman S, Shannon KM, Atwater SK, et al. Myelodysplastic and myeloproliferative disorders of childhood: a study of 167 patients. Blood. 1999;93:459–466. 6. Sasaki H, Manabe A, Kojima S, et al. Myelodysplastic syndrome in childhood: a retrospective study of 189 patients in Japan. Leukemia. 2001;15:1713–1720. 7. Kardos G, Baumann I, Passmore SJ, et al. Refractory anemia in childhood: a retrospective analysis of 67 patients with particular reference to monosomy 7. Blood. 2003;102:1997–2003. 8. Strahm B, Nollke P, Zecca M, et al. Hematopoietic stem cell transplantation for advanced myelodysplastic syndrome in children: results of the EWOG-MDS 98 study. Leukemia. 2011;25:455–462. 9. Vlachos A, Rosenberg PS, Atsidaftos E, et al. Incidence of neoplasia in Diamond Blackfan anemia: a report from the Diamond Blackfan Anemia Registry. Blood. 2012;119: 3815–3819. 10. Alter BP, Giri N, Savage SA, et al. Malignancies and survival patterns in the National Cancer Institute inherited bone marrow failure syndromes cohort study. Br J Haematol. 2010;150:179–188. 11. Dale DC, Bolyard AA, Schwinzer BG, et al. The Severe Chronic Neutropenia International Registry: 10-year follow-up report. Support Cancer Ther. 2006;3:220–231. 12. Rosenberg PS, Alter BP, Bolyard AA, et al. The incidence of leukemia and mortality from sepsis in patients with severe congenital neutropenia receiving long-term G-CSF therapy. Blood. 2006;107:4628–4635. 13. Savage SA, Alter BP. Dyskeratosis congenita. Hematol Oncol Clin North Am. 2009;23:215–231.

www.jpho-online.com |

5

Glaubach et al

J Pediatr Hematol Oncol

14. Vinh DC, Patel SY, Uzel G, et al. Autosomal dominant and sporadic monocytopenia with susceptibility to mycobacteria, fungi, papillomaviruses, and myelodysplasia. Blood. 2010;115: 1519–1529. 15. Narendran A, Hawkins LM, Ganjavi H, et al. Characterization of bone marrow stromal abnormalities in a patient with constitutional trisomy 8 mosaicism and myelodysplastic syndrome. Pediatr Hematol Oncol. 2004;21:209–221. 16. Hasle H, Clausen N, Pedersen B, et al. Myelodysplastic syndrome in a child with constitutional trisomy 8 mosaicism and normal phenotype. Cancer Genet Cytogenet. 1995;79: 79–81. 17. Maris JM, Wiersma SR, Mahgoub N, et al. Monosomy 7 myelodysplastic syndrome and other second malignant neoplasms in children with neurofibromatosis type 1. Cancer. 1997;79:1438–1446. 18. Lange BJ, Kobrinsky N, Barnard DR, et al. Distinctive demography, biology, and outcome of acute myeloid leukemia and myelodysplastic syndrome in children with Down syndrome: Children’s Cancer Group Studies 2861 and 2891. Blood. 1998;91:608–615. 19. Ross JA, Spector LG, Robison LL, et al. Epidemiology of leukemia in children with Down syndrome. Pediatric Blood Cancer. 2005;44:8–12. 20. Seif AE. Pediatric leukemia predisposition syndromes: clues to understanding leukemogenesis. Cancer Genet. 2011;204:227–244. 21. Baumann I, Fuhrer M, Behrendt S, et al. Morphological differentiation of severe aplastic anaemia from hypocellular refractory cytopenia of childhood: reproducibility of histopathological diagnostic criteria. Histopathology. 2012;61:10–17. 22. Baumann I, Niemeyer C, Fu¨hrer M, et al. 378 Morphological differentiation of hypocellular refractory cytopenia of childhood and severe aplastic anemia and clinical outcome. Leukemia Res. 2011;35:S151. 23. Hasle H, Niemeyer CM, Chessells JM, et al. A pediatric approach to the WHO classification of myelodysplastic and myeloproliferative diseases. Leukemia. 2003;17:277–282. 24. Niemeyer CM, Baumann I. Myelodysplastic syndrome in children and adolescents. Semin Hematol. 2008;45:60–70. 25. Pitman SD, Victorio A, Rowsell E, et al. 5q  syndrome in a child with slowly progressive pancytopenia: a case report and review of the literature. J Pediatr Hematol Oncol. 2006;28: 115–119. 26. Pui CH, Schrappe M, Ribeiro RC, et al. Childhood and adolescent lymphoid and myeloid leukemia. Hematology Am Soc Hematol Educ Program. 2004;2004:118–145. 27. Stary J, Baumann I, Creutzig U, et al. Getting the numbers straight in pediatric MDS: distribution of subtypes after exclusion of down syndrome. Pediatric Blood Cancer. 2008;50:435–436. 28. Gohring G, Michalova K, Beverloo HB, et al. Complex karyotype newly defined: the strongest prognostic factor in advanced childhood myelodysplastic syndrome. Blood. 2010; 116:3766–3769. 29. Yoshida K, Sanada M, Shiraishi Y, et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature. 2011;478:64–69. 30. Shih AH, Abdel-Wahab O, Patel JP, et al. The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer. 2012;12:599–612. 31. Przychodzen B, Jerez A, Guinta K, et al. Patterns of missplicing due to somatic U2AF1 mutations in myeloid neoplasms. Blood. 2013;122:999–1006. 32. Hirabayashi S, Flotho C, Moetter J, et al. Spliceosomal gene aberrations are rare, coexist with oncogenic mutations, and are unlikely to exert a driver effect in childhood MDS and JMML. Blood. 2012;119:e96–e99. 33. Takita J, Yoshida K, Sanada M, et al. Novel splicing-factor mutations in juvenile myelomonocytic leukemia. Leukemia. 2012;26:1879–1881.

34. Owen C, Barnett M, Fitzgibbon J. Familial myelodysplasia and acute myeloid leukaemia—a review. Br J Haematol. 2008;140:123–132. 35. Liew E, Owen C. Familial myelodysplastic syndromes: a review of the literature. Haematologica. 2011;96:1536–1542. 36. Hahn CN, Chong CE, Carmichael CL, et al. Heritable GATA2 mutations associated with familial myelodysplastic syndrome and acute myeloid leukemia. Nat Genet. 2011;43:1012–1017. 37. Hsu AP, Sampaio EP, Khan J, et al. Mutations in GATA2 are associated with the autosomal dominant and sporadic monocytopenia and mycobacterial infection (MonoMAC) syndrome. Blood. 2011;118:2653–2655. 38. Dickinson RE, Griffin H, Bigley V, et al. Exome sequencing identifies GATA-2 mutation as the cause of dendritic cell, monocyte, B and NK lymphoid deficiency. Blood. 2011; 118:2656–2658. 39. Ostergaard P, Simpson MA, Connell FC, et al. Mutations in GATA2 cause primary lymphedema associated with a predisposition to acute myeloid leukemia (Emberger syndrome). Nat Genet. 2011;43:929–931. 40. Baumann I, Niemeyer CM, Bennett JM, et al. Childhood myelodysplastic syndrome. In: Swerdlow SH, Campo E, Harris NL, et al, eds. World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2009:104–107. 41. Germing U, Strupp C, Kuendgen A, et al. Prospective validation of the WHO proposals for the classification of myelodysplastic syndromes. Haematologica. 2006;91: 1596–1604. 42. Rosati S, Mick R, Xu F, et al. Refractory cytopenia with multilineage dysplasia: further characterization of an “unclassifiable” myelodysplastic syndrome. Leukemia. 1996; 10:20–26. 43. Cantu Rajnoldi A, Fenu S, Kerndrup G, et al. Evaluation of dysplastic features in myelodysplastic syndromes: experience from the morphology group of the European Working Group of MDS in Childhood (EWOG-MDS). Ann Hematol. 2005; 84:429–433. 44. Verburgh E, Achten R, Louw VJ, et al. A new disease categorization of low-grade myelodysplastic syndromes based on the expression of cytopenia and dysplasia in one versus more than one lineage improves on the WHO classification. Leukemia. 2007;21:668–677. 45. Mueller BU, Tannenbaum S, Pizzo PA. Bone marrow aspirates and biopsies in children with human immunodeficiency virus infection. J Pediatr Hematol Oncol. 1996;18:266–271. 46. Acharya S, Bussel JB. Hematologic toxicity of sodium valproate. J Pediatr Hematol Oncol. 2000;22:62–65. 47. Bottom KS, Adams DM, Mann KP, et al. Trilineage hematopoietic toxicity associated with valproic acid therapy. J Pediatr Hematol Oncol. 1997;19:73–76. 48. Yetgin S, Ozen S, Saatci U, et al. Myelodysplastic features in juvenile rheumatoid arthritis. Am J Hematol. 1997;54: 166–169. 49. Huff JD, Keung YK, Thakuri M, et al. Copper deficiency causes reversible myelodysplasia. Am J Hematol. 2007;82: 625–630. 50. Angotti LB, Post GR, Robinson NS, et al. Pancytopenia with myelodysplasia due to copper deficiency. Pediatric Blood Cancer. 2008;51:693–695. 51. van den Heuvel-Eibrink MM. Paroxysmal nocturnal hemoglobinuria in children. Paediatr Drugs. 2007;9:11–16. 52. Naseem S, Varma N, Trehan A. Primary/de novo paroxysmal nocturnal hemoglobinuria in a child from north India: a case report with review of literature. J Pediatr Hematol Oncol. 2009;31:274–276. 53. van den Heuvel-Eibrink MM, Bredius RG, te Winkel ML, et al. Childhood paroxysmal nocturnal haemoglobinuria (PNH), a report of 11 cases in the Netherlands. Br J Haematol. 2005;128:571–577. 54. Orazi A, Albitar M, Heerema NA, et al. Hypoplastic myelodysplastic syndromes can be distinguished from acquired

6 | www.jpho-online.com

r



Volume 36, Number 1, January 2014

2013 Lippincott Williams & Wilkins

J Pediatr Hematol Oncol

55. 56.

57. 58.

59. 60.

61.

62.

r

Volume 36, Number 1, January 2014

Pediatric Myelodysplastic Syndromes

aplastic anemia by CD34 and PCNA immunostaining of bone marrow biopsy specimens. Am J Clin Pathol. 1997;107: 268–274. Greenberg PL, Tuechler H, Schanz J, et al. Revised International Prognostic Scoring System (IPSS-R) for myelodysplastic syndrome. Blood. 2012;120:2454–2465. Hasle H, Baumann I, Bergstrasser E, et al. The International Prognostic Scoring System (IPSS) for childhood myelodysplastic syndrome (MDS) and juvenile myelomonocytic leukemia (JMML). Leukemia. 2004;18:2008–2014. Stary J, Locatelli F, Niemeyer CM. Stem cell transplantation for aplastic anemia and myelodysplastic syndrome. Bone Marrow Transplant. 2005;35:S13–S16. Smith AR, Christiansen EC, Wagner JE, et al. Early hematopoietic stem cell transplant is associated with favorable outcomes in children with MDS. Pediatric Blood Cancer. 2013;60:705–710. Olnes MJ, Sloand EM. Targeting immune dysregulation in myelodysplastic syndromes. JAMA. 2011;305:814–819. Lim ZY, Killick S, Germing U, et al. Low IPSS score and bone marrow hypocellularity in MDS patients predict hematological responses to antithymocyte globulin. Leukemia. 2007;21: 1436–1441. Saunthararajah Y, Nakamura R, Nam JM, et al. HLA-DR15 (DR2) is overrepresented in myelodysplastic syndrome and aplastic anemia and predicts a response to immunosuppression in myelodysplastic syndrome. Blood. 2002;100:1570–1574. Sloand EM, Wu CO, Greenberg P, et al. Factors affecting response and survival in patients with myelodysplasia treated

with immunosuppressive therapy. J Clin Oncol. 2008;26: 2505–2511. Yoshimi A, Baumann I, Fu¨hrer M, et al. Immunosuppressive therapy with anti-thymocyte globulin and cyclosporine A in selected children with hypoplastic refractory cytopenia. Haematologica. 2007;92:397–400. Hasegawa D, Manabe A, Yagasaki H, et al. Treatment of children with refractory anemia: the Japanese Childhood MDS Study Group trial (MDS99). Pediatric Blood Cancer. 2009; 53:1011–1015. Aalbers AM, van der Velden VHJ, Yoshimi A, et al. The clinical relevance of minor paroxysmal nocturnal hemoglobinuria clones in refractory cytopenia of childhood: a prospective study by EWOG-MDS. Leukemia. 2013. [Epub ahead of print]. George RE, Lahti JM, Adamson PC, et al. Phase I study of decitabine with doxorubicin and cyclophosphamide in children with neuroblastoma and other solid tumors: a Children’s Oncology Group study. Pediatric Blood Cancer. 2010;55: 629–638. Furlan I, Batz C, Flotho C, et al. Intriguing response to azacitidine in a patient with juvenile myelomonocytic leukemia and monosomy 7. Blood. 2009;113:2867–2868. Vidal DO, Paixao VA, Brait M, et al. Aberrant methylation in pediatric myelodysplastic syndrome. Leuk Res. 2007;31: 175–181. Olk-Batz C, Poetsch AR, Nollke P, et al. Aberrant DNA methylation characterizes juvenile myelomonocytic leukemia with poor outcome. Blood. 2011;117:4871–4880.



2013 Lippincott Williams & Wilkins

63.

64.

65.

66.

67.

68.

69.

www.jpho-online.com |

7

Suggest Documents