1
|
Lucas BJ, Connors JS, Wang H, Conneely S, Cuglievan B, Garcia MB, Rau RE. Observation and Management of Juvenile Myelomonocytic Leukemia and Noonan Syndrome-Associated Myeloproliferative Disorder: A Real-World Experience. Cancers (Basel) 2024; 16:2749. [PMID: 39123476 PMCID: PMC11311611 DOI: 10.3390/cancers16152749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Juvenile Myelomonocytic Leukemia (JMML) is a rare and clonal hematopoietic disorder of infancy and early childhood with myeloproliferative/myelodysplastic features resulting from germline or somatic mutations in the RAS pathway. Treatment is not uniform, with management varying from observation to stem cell transplant. The aim of our retrospective review is to describe the treatment and outcomes of a cohort of patients with JMML or Noonan Syndrome-associated Myeloproliferative Disorder (NS-MPD) to provide management guidance for this rare and heterogeneous disease. We report on 22 patients with JMML or NS-MPD managed at three institutions in the Texas Medical Center. Of patients with known genetic mutations and cytogenetics, 6 harbored germline mutations, 12 had somatic mutations, and 9 showed cytogenetic abnormalities. Overall, 14/22 patients are alive. Spontaneous clinical remission occurred in one patient with somatic NRAS mutation, as well as two with germline PTPN11 mutations with NS-MPD, and two others with germline PTPN11 mutations and NS-MPD remain under surveillance. Patients with NS-MPD were excluded from treatment analysis as none required chemotherapeutic intervention. All patients (5/5) treated with 5-azacitidine alone and one of the four treated with 6-mercaptopurine monotherapy had a reduction in mutant variant allele frequency. Transformation to acute myeloid leukemia was seen in two patients who both died. Among patients who received transplants, 7/13 are alive, and relapse post-transplant occurred in 3/13 with a median time to relapse of 3.55 months. This report provides insight into therapy responses and long-term outcomes across different genetic subsets of JMML and lends insight into the expected time to spontaneous resolution in patients with NS-MPD with germline PTPN11 mutations.
Collapse
Affiliation(s)
- Bryony J. Lucas
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Cancer and Hematology Center, Houston, TX 77030, USA
| | - Jeremy S. Connors
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Heping Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shannon Conneely
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Cancer and Hematology Center, Houston, TX 77030, USA
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Pediatric Oncology, Children’s Memorial Hermann Hospital, Houston, TX 77030, USA
| | - Miriam B. Garcia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Pediatric Oncology, Children’s Memorial Hermann Hospital, Houston, TX 77030, USA
| | - Rachel E. Rau
- Department of Pediatrics, Ben Towne Center for Childhood Cancer Research, Seattle Children’s Hospital, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
2
|
Yi ES, Kim SK, Ju HY, Lee JW, Cho B, Kim BK, Kang HJ, Baek HJ, Kook H, Yang EJ, Lim YT, Ahn WK, Hahn SM, Park SK, Yoo ES, Yoo KH. Allogeneic hematopoietic cell transplantation in patients with juvenile myelomonocytic leukemia in Korea: a report of the Korean Pediatric Hematology-Oncology Group. Bone Marrow Transplant 2023; 58:20-29. [PMID: 36167906 DOI: 10.1038/s41409-022-01826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/08/2022] [Accepted: 09/05/2022] [Indexed: 01/07/2023]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a life-threatening myeloproliferative neoplasm. This multicenter study evaluated the characteristics, outcomes, and prognostic factors of allogeneic hematopoietic cell transplantation (HCT) in recipients with JMML who were diagnosed between 2000 and 2019 in Korea. Sixty-eight patients were retrospectively enrolled-28 patients (41.2%) received HCT during 2000-2010 and 40 patients (58.8%) during 2011-2020. The proportion of familial mismatched donors increased from 3.6 to 37.5%. The most common conditioning therapy was changed from Busulfan/Cyclophosphamide-based to Busulfan/Fludarabine-based therapy. The 5-year probabilities of event-free survival (EFS) and overall survival (OS) were 52.6% and 62.3%, respectively. The 5-year incidence of transplant-related mortality was 30.1%. Multivariate analysis revealed that the proportion of hemoglobin F ≥ 40%, abnormal cytogenetics, and matched sibling donors were independent risk factors for a higher relapse rate. Patients whose donor chimerism was below 99% had a significantly higher relapse rate. Better OS and lower treatment-related mortality were observed in patients with chronic graft-versus-host disease (GVHD), whereas grade III or IV acute GVHD was associated with worse EFS. In conclusion, the number of transplant increased along with the increase in alternative donor transplants, nevertheless, similar results were maintained. Alternative donor transplantation should be encouraged.
Collapse
Affiliation(s)
- Eun Sang Yi
- Department of Pediatrics, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea.,Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seong Koo Kim
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Wook Lee
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bin Cho
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bo Kyung Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Cancer Research Institute, Wide River Institute of Immunology, Seoul National University Children's Hospital, Seoul, Korea
| | - Hyoung Jin Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Cancer Research Institute, Wide River Institute of Immunology, Seoul National University Children's Hospital, Seoul, Korea
| | - Hee Jo Baek
- Department of Pediatrics, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Hoon Kook
- Department of Pediatrics, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Eu Jeen Yang
- Department of Pediatrics, Pusan National University School of Medicine, Pusan National University Children's Hospital, Yangsan, Korea
| | - Young Tak Lim
- Department of Pediatrics, Pusan National University School of Medicine, Pusan National University Children's Hospital, Yangsan, Korea
| | - Won Kee Ahn
- Department of Pediatrics, Yonsei University College of Medicine, Severance Hospital, Seoul, Korea
| | - Seung Min Hahn
- Department of Pediatrics, Yonsei University College of Medicine, Severance Hospital, Seoul, Korea
| | - Sang Kyu Park
- Department of Pediatrics, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Eun Sun Yoo
- Department of Pediatrics, Ewha Womans University College of Medicine, Ewha Womans University Seoul Hospital, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea. .,Department of Health Science and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea. .,Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea.
| |
Collapse
|
3
|
Li G, Sun Z, Geng L, Wan X, Zhu X, Tang B, Tong J, Yao W, Song K, Qiang P, Zhang L, Zhang X, Zhang S, Liu H. Clinical outcome of cord blood transplantation for nine children with juvenile myelomonocytic leukemia receiving fludarabine-busulfan-cyclophosphamide-based conditioning. Pediatr Transplant 2022; 26:e14181. [PMID: 34747111 DOI: 10.1111/petr.14181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 09/07/2021] [Accepted: 10/13/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Juvenile myelomonocytic leukemia (JMML) is a rare hematological malignancy in young children and can only be cured through the allogeneic stem cell transplantation. PROCEDURE We have retrospectively analyzed the outcomes of nine children with JMML after unrelated cord blood transplantation (UCBT). RESULTS Eight patients who have received a myeloablative conditioning regimen of fludarabine (FLU), busulfan (BU), and cyclophosphamide (CY) have gotten engraftment. None of the nine patients has relapsed following initial UCBT. Six patients are still alive and in complete remission after UCBT with a median observation time of 43 months (range: 10-80 months). CONCLUSIONS This study shows that UCBT with FLU-BU-CY conditioning regimen can represent a suitable option for children with JMML.
Collapse
Affiliation(s)
- Guifang Li
- Division of Life Sciences and Medicine, Department of Geriatrics, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China.,Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Zimin Sun
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China.,Division of Life Sciences and Medicine, Blood and Cell Therapy Institute, University of Science and Technology of China, Hefei, China
| | - Liangquan Geng
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xiang Wan
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xiaoyu Zhu
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China.,Division of Life Sciences and Medicine, Blood and Cell Therapy Institute, University of Science and Technology of China, Hefei, China
| | - Baolin Tang
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Juan Tong
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Wen Yao
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Kaidi Song
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Ping Qiang
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Lei Zhang
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xuhan Zhang
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Shiyang Zhang
- Division of Life Sciences and Medicine, Department of Geriatrics, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Huilan Liu
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China.,Division of Life Sciences and Medicine, Blood and Cell Therapy Institute, University of Science and Technology of China, Hefei, China
| |
Collapse
|
4
|
Wintering A, Dvorak CC, Stieglitz E, Loh ML. Juvenile myelomonocytic leukemia in the molecular era: a clinician's guide to diagnosis, risk stratification, and treatment. Blood Adv 2021; 5:4783-4793. [PMID: 34525182 PMCID: PMC8759142 DOI: 10.1182/bloodadvances.2021005117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/03/2021] [Indexed: 12/03/2022] Open
Abstract
Juvenile myelomonocytic leukemia is an overlapping myeloproliferative and myelodysplastic disorder of early childhood . It is associated with a spectrum of diverse outcomes ranging from spontaneous resolution in rare patients to transformation to acute myeloid leukemia in others that is generally fatal. This unpredictable clinical course, along with initially descriptive diagnostic criteria, led to decades of productive international research. Next-generation sequencing now permits more accurate molecular diagnoses in nearly all patients. However, curative treatment is still reliant on allogeneic hematopoietic cell transplantation for most patients, and additional advances will be required to improve risk stratification algorithms that distinguish those that can be observed expectantly from others who require swift hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Astrid Wintering
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
| | - Christopher C. Dvorak
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Mignon L. Loh
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| |
Collapse
|
5
|
Donor Killer Immunoglobulin Receptor Gene Content and Ligand Matching and Outcomes of Pediatric Patients with Juvenile Myelomonocytic Leukemia Following Unrelated Donor Transplantation. Transplant Cell Ther 2021; 27:926.e1-926.e10. [PMID: 34407489 DOI: 10.1016/j.jtct.2021.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 11/21/2022]
Abstract
Natural killer (NK) cell determinants predict relapse-free survival after allogeneic hematopoietic cell transplantation (HCT) for acute myelogenous leukemia, and previous studies have shown a beneficial graft-versus-leukemia effect in patients with juvenile myelomonocytic leukemia (JMML). However, whether NK cell determinants predict protection against relapse for JMML patients undergoing HCT is unknown. Therefore, we investigated NK cell-related donor and recipient immunogenetics as determinants of HCT outcomes in patients with JMML. Patients with JMML (age 0 to <19 years) who underwent a first allogeneic HCT from an unrelated donor between 2000 and 2017 and had available donor samples from the Center for International Blood and Marrow Transplant Research Repository were included. Donor killer immunoglobulin receptor (KIR) typing was performed on pre-HCT samples. The primary endpoint was disease-free survival (DFS); secondary endpoints included relapse, grade II-IV acute graft versus-host-disease (aGVHD), chronic GVHD (cGVHD), GVHD-free relapse-free survival, transplantation-related mortality, and overall survival (OS). Donor KIR models tested included KIR genotype (AA versus Bx), B content (0-1 versus ≥2), centromeric and telomeric region score (AA versus AB versus BB), B content score (best, better, or neutral), composite score (2 versus 3 versus 4), activating KIR content, and the presence of KIR2DS4. Ligand-ligand and KIR-ligand mismatch effects on outcomes were analyzed in HLA-mismatched donors (≤7/8; n = 74) only. Univariate analyses were performed for primary and secondary outcomes of interest, with a P value <.05 considered significant. One hundred sixty-five patients (113 males), with a median follow-up of 85 months (range, 6 to 216 months) met the study criteria. Of these, 111 underwent an unrelated donor HCT and 54 underwent a UCB HCT. Almost all (n = 161; 98%) received a myeloablative conditioning regimen. After exclusion of recipients of reduced-intensity/nonmyeloablative conditioning regimens and ex vivo T cell-depleted grafts (n = 8), there were 42 AA donors and 115 Bx donors, respectively. Three-year DFS, OS, relapse, and GRFS for the entire cohort were 58% (95% confidence interval [CI], 50% to 66%), 67% (95% CI, 59% to 74%), 26% (95% CI, 19% to 33%), and 27% (95% CI, 19% to 35%), respectively. The cumulative incidence of grade II-IV aGVHD at 100 days was 36% (95% CI, 27% to 44%), and that of cGVHD at 1 year was 23% (95% CI, 17% to 30%). There were no differences between AA donors and Bx donors for any recipient survival outcomes. The risk of grade II-IV aGVHD was lower in patients with donors with a B content score of ≥2 (hazard ratio [HR], 0.46; 95% CI, 0.26 to 0.83; P = .01), an activating KIR content score of >3 (HR, 0.52; 95% CI, 0.29 to 0.95; P = .032), centromeric A/B score (HR, 0.57; 95% CI, 033 to 0.98; P = .041), and telomeric A/B score (HR, 0.58; 95% CI, 0.34 to 1.00; P = .048). To our knowledge, this is the first study analyzing the association of NK cell determinants and outcomes in JMML HCT recipients. This study identifies potential benefits of donor KIR-B genotypes in reducing aGVHD. Our findings warrant further study of the role of NK cells in enhancing the graft-versus-leukemia effect via recognition of JMML blasts.
Collapse
|
6
|
Greenmyer JR, Kohorst M. Pediatric Neoplasms Presenting with Monocytosis. Curr Hematol Malig Rep 2021; 16:235-246. [PMID: 33630234 DOI: 10.1007/s11899-021-00611-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW Juvenile myelomonocytic leukemia (JMML) is a rare but severe pediatric neoplasm with hematopoietic stem cell transplant as its only established curative option. The development of targeted therapeutics for JMML is being guided by an understanding of the pathobiology of this condition. Here, we review JMML with an emphasis on genetics in order to (i) demonstrate the relationship between JMML genotype and clinical phenotype and (ii) explore potential genetic targets of novel JMML therapies. RECENT FINDINGS DNA hypermethylation studies have demonstrated consistently that methylation is related to disease severity. Increasing understanding of methylation in JMML may open the door to novel therapies, such as DNA methyltransferase inhibitors. The PI3K/AKT/MTOR, JAK/STAT, and RAF/MEK/ERK pathways are being investigated as therapeutic targets for JMML. Future therapy for JMML will be driven by an increased understanding of pathobiology. Targeted therapeutic approaches hold potential for improving outcomes in patients with JMML.
Collapse
Affiliation(s)
| | - Mira Kohorst
- Pediatric Hematology and Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
7
|
Lasho T, Patnaik MM. Juvenile myelomonocytic leukemia – A bona fide RASopathy syndrome. Best Pract Res Clin Haematol 2020; 33:101171. [DOI: 10.1016/j.beha.2020.101171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/20/2022]
|
8
|
Ceraulo A, Girard S, Ranchère-Vince D, Marceau A, Marec-Berard P, Renard C, Bertrand Y. Posttransplantation relapse of pediatric chronic myelomonocytic leukemia cured using donor lymphocyte infusion. Pediatr Blood Cancer 2018; 65. [PMID: 28895279 DOI: 10.1002/pbc.26808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/10/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Antony Ceraulo
- Department of Pediatric Oncology and Hematology, Institut d'Hématologie et d'Oncologie Pédiatrique (IHOPe), Lyon, France.,Department of Medicine, University Lyon I, Lyon, France
| | - Sandrine Girard
- Laboratory of Hematology, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Bron, France
| | | | - Alice Marceau
- Department of Biology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Perrine Marec-Berard
- Department of Pediatric Oncology and Hematology, Institut d'Hématologie et d'Oncologie Pédiatrique (IHOPe), Lyon, France
| | - Cécile Renard
- Department of Pediatric Oncology and Hematology, Institut d'Hématologie et d'Oncologie Pédiatrique (IHOPe), Lyon, France
| | - Yves Bertrand
- Department of Pediatric Oncology and Hematology, Institut d'Hématologie et d'Oncologie Pédiatrique (IHOPe), Lyon, France
| |
Collapse
|
9
|
Locatelli F, Algeri M, Merli P, Strocchio L. Novel approaches to diagnosis and treatment of Juvenile Myelomonocytic Leukemia. Expert Rev Hematol 2018; 11:129-143. [DOI: 10.1080/17474086.2018.1421937] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Franco Locatelli
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
- Department of Pediatric Science, University of Pavia, Pavia, Italy
| | - Mattia Algeri
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Pietro Merli
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Luisa Strocchio
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| |
Collapse
|
10
|
Lipka DB, Witte T, Toth R, Yang J, Wiesenfarth M, Nöllke P, Fischer A, Brocks D, Gu Z, Park J, Strahm B, Wlodarski M, Yoshimi A, Claus R, Lübbert M, Busch H, Boerries M, Hartmann M, Schönung M, Kilik U, Langstein J, Wierzbinska JA, Pabst C, Garg S, Catalá A, De Moerloose B, Dworzak M, Hasle H, Locatelli F, Masetti R, Schmugge M, Smith O, Stary J, Ussowicz M, van den Heuvel-Eibrink MM, Assenov Y, Schlesner M, Niemeyer C, Flotho C, Plass C. RAS-pathway mutation patterns define epigenetic subclasses in juvenile myelomonocytic leukemia. Nat Commun 2017; 8:2126. [PMID: 29259247 PMCID: PMC5736667 DOI: 10.1038/s41467-017-02177-w] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 11/13/2017] [Indexed: 01/15/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative disorder of early childhood characterized by mutations activating RAS signaling. Established clinical and genetic markers fail to fully recapitulate the clinical and biological heterogeneity of this disease. Here we report DNA methylome analysis and mutation profiling of 167 JMML samples. We identify three JMML subgroups with unique molecular and clinical characteristics. The high methylation group (HM) is characterized by somatic PTPN11 mutations and poor clinical outcome. The low methylation group is enriched for somatic NRAS and CBL mutations, as well as for Noonan patients, and has a good prognosis. The intermediate methylation group (IM) shows enrichment for monosomy 7 and somatic KRAS mutations. Hypermethylation is associated with repressed chromatin, genes regulated by RAS signaling, frequent co-occurrence of RAS pathway mutations and upregulation of DNMT1 and DNMT3B, suggesting a link between activation of the DNA methylation machinery and mutational patterns in JMML.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Biopsy
- Child
- Child, Preschool
- Chromatin/genetics
- Chromatin/metabolism
- DNA (Cytosine-5-)-Methyltransferase 1/metabolism
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- DNA Methylation
- DNA Mutational Analysis
- Epigenomics
- Female
- Gene Expression Regulation, Leukemic
- Hematopoietic Stem Cell Transplantation
- Humans
- Infant
- Leukemia, Myelomonocytic, Juvenile/genetics
- Leukemia, Myelomonocytic, Juvenile/mortality
- Leukemia, Myelomonocytic, Juvenile/pathology
- Leukemia, Myelomonocytic, Juvenile/therapy
- Male
- Mutation
- Noonan Syndrome/genetics
- Noonan Syndrome/pathology
- Prognosis
- Prospective Studies
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
- Proto-Oncogene Proteins c-cbl
- Proto-Oncogene Proteins p21(ras)/genetics
- Proto-Oncogene Proteins p21(ras)/metabolism
- Signal Transduction/genetics
- Up-Regulation
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
- Daniel B Lipka
- Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany.
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke-University, Leipziger Strasse 44, 39120, Magdeburg, Germany.
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Leipziger Strasse 44, 39120, Magdeburg, Germany.
| | - Tania Witte
- Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
- Cancer Epigenetics Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Reka Toth
- Computational Epigenomics Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Jing Yang
- Division of Theoretical Bioinformatics (B080), German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Manuel Wiesenfarth
- Division of Biostatistics, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Peter Nöllke
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine Medical Center, Faculty of Medicine, University of Freiburg, Heiliggeiststrasse 1, 79106, Freiburg, Germany
| | - Alexandra Fischer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine Medical Center, Faculty of Medicine, University of Freiburg, Heiliggeiststrasse 1, 79106, Freiburg, Germany
| | - David Brocks
- Cancer Epigenetics Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Zuguang Gu
- Division of Theoretical Bioinformatics (B080), German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Jeongbin Park
- Division of Theoretical Bioinformatics (B080), German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Brigitte Strahm
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine Medical Center, Faculty of Medicine, University of Freiburg, Heiliggeiststrasse 1, 79106, Freiburg, Germany
| | - Marcin Wlodarski
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine Medical Center, Faculty of Medicine, University of Freiburg, Heiliggeiststrasse 1, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), 79106, Freiburg, Germany
| | - Ayami Yoshimi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine Medical Center, Faculty of Medicine, University of Freiburg, Heiliggeiststrasse 1, 79106, Freiburg, Germany
| | - Rainer Claus
- Division of Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Michael Lübbert
- Division of Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Hauke Busch
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Stefan-Meier-Strasse 17, 79104, Freiburg, Germany
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Melanie Boerries
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Stefan-Meier-Strasse 17, 79104, Freiburg, Germany
- German Cancer Consortium (DKTK), 79106, Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Mark Hartmann
- Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Maximilian Schönung
- Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Umut Kilik
- Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Jens Langstein
- Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Justyna A Wierzbinska
- Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
- Cancer Epigenetics Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Caroline Pabst
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, INF 410, 69120, Heidelberg, Germany
| | - Swati Garg
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, INF 410, 69120, Heidelberg, Germany
| | - Albert Catalá
- Department of Hematology and Oncology, Hospital Sant Joan de Déu, Passeig de Sant Joan de Déu, 2, 08950, Esplugues de Llobrega, Barcelona, Spain
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Michael Dworzak
- St. Anna Children's Hospital and Children's Cancer Research Institute, Medical University of Vienna, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Henrik Hasle
- Department of Pediatrics, Aarhus University Hospital Skejby, Palle Juul-Jensens Boulevard 82, 8200, Aarhus, Denmark
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Bambino Gesú Children's Hospital, University of Pavia, Piazza S. Onofrio 4, Rome, 00165, Italy
| | - Riccardo Masetti
- Department of Pediatric Oncology and Hematology, University of Bologna, Via Massarenti 11, 40138, Bologna, Italy
| | - Markus Schmugge
- Department of Hematology and Oncology, University Children's Hospital, Steinwiesstrasse 75, 8032, Zurich, Switzerland
| | - Owen Smith
- Department of Paediatric Oncology and Haematology, Our Lady's Children's Hospital Crumlin, Dublin, 12, Ireland
| | - Jan Stary
- Department of Pediatric Hematology and Oncology, Charles University and University Hospital Motol, V Úvalu 84, 150 06, Prague 5, Czech Republic
| | - Marek Ussowicz
- Department of Pediatric Hematology, Oncology and BMT, Wroclaw Medical University, ul. Borowska 213, 50-556, Wroclaw, Poland
| | | | - Yassen Assenov
- Computational Epigenomics Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Matthias Schlesner
- Division of Theoretical Bioinformatics (B080), German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
- Bioinformatics and Omics Data Analytics (B240), German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Charlotte Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine Medical Center, Faculty of Medicine, University of Freiburg, Heiliggeiststrasse 1, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), 79106, Freiburg, Germany
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine Medical Center, Faculty of Medicine, University of Freiburg, Heiliggeiststrasse 1, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), 79106, Freiburg, Germany
| | - Christoph Plass
- Cancer Epigenetics Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany.
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.
| |
Collapse
|
11
|
Paulus S, Koronowska S, Fölster-Holst R. Association Between Juvenile Myelomonocytic Leukemia, Juvenile Xanthogranulomas and Neurofibromatosis Type 1: Case Report and Review of the Literature. Pediatr Dermatol 2017; 34:114-118. [PMID: 28111791 DOI: 10.1111/pde.13064] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The occurrence of juvenile myelomonocytic leukemia (JMML), juvenile xanthogranuloma (JXG), and neurofibromatosis type 1 (NF1) together is relatively rare. Approximately only 20 cases have been reported in the literature. It is debated whether children with NF1 and JXG are at higher risk of developing JMML than children with NF1 alone. We present the case of a boy primarily diagnosed with NF1 with coexisting JXG who developed JMML at the age of 22 months. The clinical course from initial presentation to final diagnosis is detailed and the genetic features and hematologic characteristics are discussed. We report this case to underscore the importance of close monitoring of blood count and strict clinical follow-up in children presenting with concurrent NF1 and JXG and provide a possible explanation for this association.
Collapse
Affiliation(s)
- Samuel Paulus
- Department of Dermatology, University of Kiel, Kiel, Germany
| | | | | |
Collapse
|
12
|
Tüfekçi Ö, Koçak Ü, Kaya Z, Yenicesu İ, Albayrak C, Albayrak D, Yılmaz Bengoa Ş, Patıroğlu T, Karakükçü M, Ünal E, Ünal İnce E, İleri T, Ertem M, Celkan T, Özdemir GN, Sarper N, Kaçar D, Yaralı N, Özbek NY, Küpesiz A, Karapınar T, Vergin C, Çalışkan Ü, Tokgöz H, Sezgin Evim M, Baytan B, Güneş AM, Yılmaz Karapınar D, Karaman S, Uygun V, Karasu G, Yeşilipek MA, Koç A, Erduran E, Atabay B, Öniz H, Ören H. Juvenile Myelomonocytic Leukemia in Turkey: A Retrospective Analysis of Sixty-five Patients. Turk J Haematol 2017; 35:27-34. [PMID: 28179213 PMCID: PMC5843771 DOI: 10.4274/tjh.2017.0021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE This study aimed to define the status of juvenile myelomonocytic leukemia (JMML) patients in Turkey in terms of time of diagnosis, clinical characteristics, mutational studies, clinical course, and treatment strategies. MATERIALS AND METHODS Data including clinical and laboratory characteristics and treatment strategies of JMML patients were collected retrospectively from pediatric hematology-oncology centers in Turkey. RESULTS Sixty-five children with JMML diagnosed between 2002 and 2016 in 18 institutions throughout Turkey were enrolled in the study. The median age at diagnosis was 17 months (min-max: 2-117 months). Splenomegaly was present in 92% of patients at the time of diagnosis. The median white blood cell, monocyte, and platelet counts were 32.9x109/L, 5.4x109/L, and 58.3x109/L, respectively. Monosomy 7 was present in 18% of patients. JMML mutational analysis was performed in 32 of 65 patients (49%) and PTPN11 was the most common mutation. Hematopoietic stem cell transplantation (HSCT) could only be performed in 28 patients (44%), the majority being after the year 2012. The most frequent reason for not performing HSCT was the inability to find a suitable donor. The median time from diagnosis to HSCT was 9 months (min-max: 2-63 months). The 5-year cumulative survival rate was 33% and median estimated survival time was 30±17.4 months (95% CI: 0-64.1) for all patients. Survival time was significantly better in the HSCT group (log-rank p=0.019). Older age at diagnosis (>2 years), platelet count of less than 40x109/L, and PTPN11 mutation were the factors significantly associated with shorter survival time. CONCLUSION Although there has recently been improvement in terms of definitive diagnosis and HSCT in JMML patients, the overall results are not satisfactory and it is necessary to put more effort into this issue in Turkey.
Collapse
Affiliation(s)
- Özlem Tüfekçi
- Dokuz Eylül University Faculty of Medicine, Department of Pediatric Hematology, İzmir, Turkey
| | - Ülker Koçak
- Gazi University Faculty of Medicine, Department of Pediatric Hematology, Ankara, Turkey
| | - Zühre Kaya
- Gazi University Faculty of Medicine, Department of Pediatric Hematology, Ankara, Turkey
| | - İdil Yenicesu
- Gazi University Faculty of Medicine, Department of Pediatric Hematology, Ankara, Turkey
| | - Canan Albayrak
- Ondokuz Mayıs University Faculty of Medicine, Department of Pediatric Hematology, Samsun, Turkey
| | - Davut Albayrak
- Ondokuz Mayıs University Faculty of Medicine, Department of Pediatric Hematology, Samsun, Turkey
| | - Şebnem Yılmaz Bengoa
- Dokuz Eylül University Faculty of Medicine, Department of Pediatric Hematology, İzmir, Turkey
| | - Türkan Patıroğlu
- Erciyes University Faculty of Medicine, Department of Pediatric Hematology and Oncology, Kayseri, Turkey
| | - Musa Karakükçü
- Erciyes University Faculty of Medicine, Department of Pediatric Hematology and Oncology, Kayseri, Turkey
| | - Ekrem Ünal
- Erciyes University Faculty of Medicine, Department of Pediatric Hematology and Oncology, Kayseri, Turkey
| | - Elif Ünal İnce
- Ankara University Faculty of Medicine, Department of Pediatric Hematology and Oncology, Ankara, Turkey
| | - Talia İleri
- Ankara University Faculty of Medicine, Department of Pediatric Hematology and Oncology, Ankara, Turkey
| | - Mehmet Ertem
- Ankara University Faculty of Medicine, Department of Pediatric Hematology and Oncology, Ankara, Turkey
| | - Tiraje Celkan
- İstanbul University Cerrahpaşa Faculty of Medicine, Department of Pediatric Hematology and Oncology, İstanbul, Turkey
| | - Gül Nihal Özdemir
- İstanbul University Cerrahpaşa Faculty of Medicine, Department of Pediatric Hematology and Oncology, İstanbul, Turkey
| | - Nazan Sarper
- Kocaeli University Faculty of Medicine, Department of Pediatric Hematology, Kocaeli, Turkey
| | - Dilek Kaçar
- Ankara Children's Hematology and Oncology Training and Research Hospital, Ankara, Turkey
| | - Neşe Yaralı
- Ankara Children's Hematology and Oncology Training and Research Hospital, Ankara, Turkey
| | - Namık Yaşar Özbek
- Ankara Children's Hematology and Oncology Training and Research Hospital, Ankara, Turkey
| | - Alphan Küpesiz
- Akdeniz University Faculty of Medicine, Department of Pediatric Hematology and Oncology, Antalya, Turkey
| | - Tuba Karapınar
- Dr. Behçet Uz Children Training and Research Hospital, Clinic of Pediatric Hematology and Oncology, İzmir, Turkey
| | - Canan Vergin
- Dr. Behçet Uz Children Training and Research Hospital, Clinic of Pediatric Hematology and Oncology, İzmir, Turkey
| | - Ümran Çalışkan
- Necmettin Erbakan University Meram Faculty of Medicine, Department of Pediatric Hematology, Konya, Turkey
| | - Hüseyin Tokgöz
- Necmettin Erbakan University Meram Faculty of Medicine, Department of Pediatric Hematology, Konya, Turkey
| | - Melike Sezgin Evim
- Uludağ University Faculty of Medicine, Department of Pediatric Hematology, Bursa, Turkey
| | - Birol Baytan
- Uludağ University Faculty of Medicine, Department of Pediatric Hematology, Bursa, Turkey
| | - Adalet Meral Güneş
- Uludağ University Faculty of Medicine, Department of Pediatric Hematology, Bursa, Turkey
| | | | - Serap Karaman
- Şişli Hamidiye Etfal Training and Research Hospital, Clinic of Pediatric Hematology and Oncology, İstanbul, Turkey
| | - Vedat Uygun
- Bahçeşehir University Faculty of Medicine, Department of Pediatric Hematology and Oncology, İstanbul, Turkey
| | - Gülsun Karasu
- Bahçeşehir University Faculty of Medicine, Department of Pediatric Hematology and Oncology, İstanbul, Turkey
| | - Mehmet Akif Yeşilipek
- Bahçeşehir University Faculty of Medicine, Department of Pediatric Hematology and Oncology, İstanbul, Turkey
| | - Ahmet Koç
- Marmara University Faculty of Medicine, Department of Pediatric Hematology and Oncology, İstanbul, Turkey
| | - Erol Erduran
- Karadeniz Technical University Faculty of Medicine, Department of Pediatric Hematology and Oncology, Trabzon, Turkey
| | - Berna Atabay
- Tepecik Training and Research Hospital, Clinic of Pediatric Hematology and Oncology, İzmir, Turkey
| | - Haldun Öniz
- Tepecik Training and Research Hospital, Clinic of Pediatric Hematology and Oncology, İzmir, Turkey
| | - Hale Ören
- Dokuz Eylül University Faculty of Medicine, Department of Pediatric Hematology, İzmir, Turkey
| |
Collapse
|
13
|
Abstract
RAS genes encode a family of 21 kDa proteins that are an essential hub for a number of survival, proliferation, differentiation and senescence pathways. Signaling of the RAS-GTPases through the RAF-MEK-ERK pathway, the first identified mitogen-associated protein kinase (MAPK) cascade is essential in development. A group of genetic syndromes, named "RASopathies", had been identified which are caused by heterozygosity for germline mutations in genes that encode protein components of the RAS/MAPK pathway. Several of these clinically overlapping disorders, including Noonan syndrome, Noonan-like CBL syndrome, Costello syndrome, cardio-facio-cutaneous (CFC) syndrome, neurofibromatosis type I, and Legius syndrome, predispose to cancer and abnormal myelopoiesis in infancy. This review focuses on juvenile myelomonocytic leukemia (JMML), a malignancy of early childhood characterized by initiating germline and/or somatic mutations in five genes of the RAS/MAPK pathway: PTPN11, CBL, NF-1, KRAS and NRAS. Natural courses of these five subtypes differ, although hematopoietic stem cell transplantation remains the only curative therapy option for most children with JMML. With whole-exome sequencing studies revealing few secondary lesions it will be crucial to better understand the RAS/MAPK signaling network with its crosstalks and feed-back loops to carefully design early clinical trials with novel pharmacological agents in this still puzzling leukemia.
Collapse
Affiliation(s)
- Charlotte M Niemeyer
- Department of Pediatric Hematology and Oncology, Universitätsklinikum Freiburg, Germany
| |
Collapse
|
14
|
Stieglitz E, Ward AF, Gerbing RB, Alonzo TA, Arceci RJ, Liu YL, Emanuel PD, Widemann BC, Cheng JW, Jayaprakash N, Balis FM, Castleberry RP, Bunin NJ, Loh ML, Cooper TM. Phase II/III trial of a pre-transplant farnesyl transferase inhibitor in juvenile myelomonocytic leukemia: a report from the Children's Oncology Group. Pediatr Blood Cancer 2015; 62:629-36. [PMID: 25704135 PMCID: PMC4339233 DOI: 10.1002/pbc.25342] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 10/01/2014] [Indexed: 02/04/2023]
Abstract
BACKGROUND Juvenile myelomonocytic leukemia (JMML) is not durably responsive to chemotherapy, and approximately 50% of patients relapse after hematopoietic stem cell transplant (HSCT). Here we report the activity and acute toxicity of the farnesyl transferase inhibitor tipifarnib, the response rate to 13-cis retinoic acid (CRA) in combination with cytoreductive chemotherapy, and survival following HSCT in children with JMML. PROCEDURE Eighty-five patients with newly diagnosed JMML were enrolled on AAML0122 between 2001 and 2006. Forty-seven consented to receive tipifarnib in a phase II window before proceeding to a phase III trial of CRA in combination with fludarabine and cytarabine followed by HSCT and maintenance CRA. Thirty-eight patients enrolled only in the phase III trial. RESULTS Overall response rate was 51% after tipifarnib and 68% after fludarabine/cytarabine/CRA. Tipifarnib did not increase pre-transplant toxicities. Forty-six percent of the 44 patients who received protocol compliant HSCT relapsed. Five-year overall survival was 55 ± 11% and event-free survival was 41 ± 11%, with no significant difference between patients who did or did not receive tipifarnib. CONCLUSIONS Administration of tipifarnib in the window setting followed by HSCT in patients with newly diagnosed JMML was safe and yielded a 51% initial response rate as a single agent, but failed to reduce relapse rates or improve long-term overall survival.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Child
- Child, Preschool
- Cytarabine/administration & dosage
- Disease-Free Survival
- Enzyme Inhibitors/administration & dosage
- Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors
- Female
- Hematopoietic Stem Cell Transplantation
- Humans
- Infant
- Isotretinoin/administration & dosage
- Leukemia, Myelomonocytic, Juvenile/drug therapy
- Leukemia, Myelomonocytic, Juvenile/enzymology
- Leukemia, Myelomonocytic, Juvenile/mortality
- Leukemia, Myelomonocytic, Juvenile/pathology
- Male
- Middle Aged
- Quinolones/administration & dosage
- Survival Rate
- Vidarabine/administration & dosage
- Vidarabine/analogs & derivatives
Collapse
Affiliation(s)
- Elliot Stieglitz
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children’s Hospital, Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Ashley F. Ward
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children’s Hospital, Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | | | | | - Robert J. Arceci
- Ronald A. Matricaria Institute of Molecular Medicine, Phoenix Children’s Hospital, University of Arizona, Phoenix, AZ, USA
| | - Y. Lucy Liu
- University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Peter D. Emanuel
- University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | | | | | - Frank M. Balis
- The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Nancy J. Bunin
- The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mignon L. Loh
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children’s Hospital, Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Todd M. Cooper
- Aflac Cancer and Blood Disorders Center, Emory University School of Medicine/Children’s Healthcare of Atlanta, Atlanta, GA, USA
| |
Collapse
|
15
|
Abstract
Abstract
Juvenile myelomonocytic leukemia (JMML) is a unique, aggressive hematopoietic disorder of infancy/early childhood caused by excessive proliferation of cells of monocytic and granulocytic lineages. Approximately 90% of patients carry either somatic or germline mutations of PTPN-11, K-RAS, N-RAS, CBL, or NF1 in their leukemic cells. These genetic aberrations are largely mutually exclusive and activate the Ras/mitogen-activated protein kinase pathway. Allogeneic hematopoietic stem cell transplantation (HSCT) remains the therapy of choice for most patients with JMML, curing more than 50% of affected children. We recommend that this option be promptly offered to any child with PTPN-11-, K-RAS-, or NF1-mutated JMML and to the majority of those with N-RAS mutations. Because children with CBL mutations and few of those with N-RAS mutations may have spontaneous resolution of hematologic abnormalities, the decision to proceed to transplantation in these patients must be weighed carefully. Disease recurrence remains the main cause of treatment failure after HSCT. A second allograft is recommended if overt JMML relapse occurs after transplantation. Recently, azacytidine, a hypomethylating agent, was reported to induce hematologic/molecular remissions in some children with JMML, and its role in both reducing leukemia burden before HSCT and in nontransplant settings requires further studies.
Collapse
|
16
|
Choudhary D, Sharma SK, Gupta N, Handoo A. Stem cell transplant for juvenile myelomonocytic leukemia and chronic myelomonocytic leukemia. Indian J Hematol Blood Transfus 2014; 30:40-2. [PMID: 25332531 DOI: 10.1007/s12288-013-0233-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/25/2013] [Indexed: 11/25/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) and chronic myelomonocytic leukemia (CMML) are myelodysplastic/myeloproliferative neoplasms associated with poor prognosis. There is no definitive treatment for such patients other than stem cell transplantation, and chemotherapy is not much effective. Timely diagnosis and early referral to the transplant centre is important for the management of these diseases. We report here a case of JMML and another case of CMML who were treated successfully with peripheral blood stem cell transplantation.
Collapse
Affiliation(s)
- Dharma Choudhary
- Bone Marrow Transplant Centre, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Sanjeev Kumar Sharma
- Bone Marrow Transplant Centre, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Nitin Gupta
- Bone Marrow Transplant Centre, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Anil Handoo
- Bone Marrow Transplant Centre, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| |
Collapse
|
17
|
Analysis of risk factors influencing outcomes after cord blood transplantation in children with juvenile myelomonocytic leukemia: a EUROCORD, EBMT, EWOG-MDS, CIBMTR study. Blood 2013; 122:2135-41. [PMID: 23926304 DOI: 10.1182/blood-2013-03-491589] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We retrospectively analyzed 110 patients with juvenile myelomonocytic leukemia, given single-unit, unrelated donor umbilical cord blood transplantation. Median age at diagnosis and at transplantation was 1.4 years (age range, 0.1-6.4 years) and 2.2 years (age range, 0.5-7.4 years), respectively. Before transplantation, 88 patients received chemotherapy; splenectomy was performed in 24 patients. Monosomy of chromosome 7 was the most frequent cytogenetic abnormality, found in 24% of patients. All but 8 patients received myeloablative conditioning; cyclosporine plus steroids was the most common graft-versus-host disease prophylaxis. Sixteen percent of units were HLA-matched with the recipient, whereas 43% and 35% had either 1 or 2 to 3 HLA disparities, respectively. The median number of nucleated cells infused was 7.1 × 10(7)/kg (range, 1.7-27.6 × 10(7)/kg). With a median follow-up of 64 months (range, 14-174 months), the 5-year cumulative incidences of transplantation-related mortality and relapse were 22% and 33%, respectively. The 5-year disease-free survival rate was 44%. In multivariate analysis, factors predicting better disease-free survival were age younger than 1.4 years at diagnosis (hazard ratio [HR], 0.42; P = .005), 0 to 1 HLA disparities in the donor/recipient pair (HR, 0.4; P = .009), and karyotype other than monosomy 7 (HR, 0.5; P = .02). Umbilical cord blood transplantation may cure a relevant proportion of children with juvenile myelomonocytic leukemia. Because disease recurrence remains the major cause of treatment failure, strategies to reduce incidence of relapse are warranted.
Collapse
|
18
|
Ozono S, Inada H, Nakagawa SI, Ueda K, Matsumura H, Kojima S, Koga H, Hashimoto T, Oshima K, Matsuishi T. Juvenile myelomonocytic leukemia characterized by cutaneous lesion containing Langerhans cell histiocytosis-like cells. Int J Hematol 2011; 93:389-393. [DOI: 10.1007/s12185-011-0787-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 02/07/2011] [Accepted: 02/09/2011] [Indexed: 12/23/2022]
|
19
|
Santini V, Alessandrino PE, Angelucci E, Barosi G, Billio A, Di Maio M, Finelli C, Locatelli F, Marchetti M, Morra E, Musto P, Visani G, Tura S. Clinical management of myelodysplastic syndromes: update of SIE, SIES, GITMO practice guidelines. Leuk Res 2010; 34:1576-88. [PMID: 20149927 DOI: 10.1016/j.leukres.2010.01.018] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 01/11/2010] [Accepted: 01/17/2010] [Indexed: 12/13/2022]
Affiliation(s)
- V Santini
- Functional Unit of Haematology, AOU Careggi, University of Florence, Firenze, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Yoshimi A, Kojima S, Hirano N. Juvenile myelomonocytic leukemia: epidemiology, etiopathogenesis, diagnosis, and management considerations. Paediatr Drugs 2010; 12:11-21. [PMID: 20034338 DOI: 10.2165/11316200-000000000-00000] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare hematopoietic malignancy of early childhood with features characteristic of both myelodysplastic and myeloproliferative disorders. Recent studies clearly show that the deregulated activation of the RAS signaling pathway plays a central role in the pathogenesis of JMML. Somatic defects in either RAS, PTPN11 or NF1 genes involved in this pathway are detected in 70-80% of JMML patients, allowing a molecular diagnosis to be made in the majority of cases. Patients with JMML respond poorly to chemotherapy, and the probability of survival without allogeneic hematopoietic stem cell transplantation (HSCT) is less than 10%. Recent studies show that the event-free survival after HSCT is between 24 and 54%, with no difference between transplants using matched family donors and those using unrelated donors. The use of therapies such as intensive chemotherapy and splenectomy prior to HSCT does not improve the outcome. The relapse rate following HSCT is over 30%, which is unacceptably high. Cumulative evidence suggests that a graft-versus-leukemia effect occurs in JMML. Donor leukocyte infusion is not usually successful in JMML, but the outcome of second HSCT is generally favorable. Based on recent advances in the understanding of the pathogenesis of JMML, the development of novel targeted therapies, which might improve the outcome of patients, is keenly awaited.
Collapse
Affiliation(s)
- Ayami Yoshimi
- Department of HSCT Data Management, Nagoya University, Japan.
| | | | | |
Collapse
|
21
|
de Vries ACH, Bredius RGM, Lankester AC, Bierings M, Trebo M, Sedlacek P, Niemeyer CM, Zecca M, Locatelli F, van den Heuvel-Eibrink MM. HLA-identical umbilical cord blood transplantation from a sibling donor in juvenile myelomonocytic leukemia. Haematologica 2008; 94:302-4. [PMID: 19109215 DOI: 10.3324/haematol.2008.000216] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
22
|
Yabe M, Sako M, Yabe H, Osugi Y, Kurosawa H, Nara T, Tokuyama M, Adachi S, Kobayashi C, Yanagimachi M, Ohtsuka Y, Nakazawa Y, Ogawa C, Manabe A, Kojima S, Nakahata T. A conditioning regimen of busulfan, fludarabine, and melphalan for allogeneic stem cell transplantation in children with juvenile myelomonocytic leukemia. Pediatr Transplant 2008; 12:862-7. [PMID: 18397212 DOI: 10.1111/j.1399-3046.2008.00931.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A pilot study was undertaken using a myeloablative conditioning with fludarabine, busulfan, and melphalan to improve the outcome of HSCT in 10 children, aged six months to six yr, with JMML. All patients were conditioned with oral busulfan (560 mg/m(2)), fludarabine (120 mg/m(2)), and melphalan (180-210 mg/m(2)) prior to HSCT, and received stem cells from bone marrow in seven cases, and from cord blood in three cases. Engraftment was documented in eight patients, whereas graft failure occurred in two, one of whom had received HLA-mismatched cord blood and other had received bone marrow from HLA-mismatched mother. Three patients, including two in who graft failure had occurred, relapsed. Five patients developed acute GVHD and two developed chronic GVHD. Seven patients are alive and in remission 27-69 months after transplantation. Thus, our study showed that HSCT following conditioning with fludarabine, busulfan, and melphalan was well tolerated and appeared to be effective for JMML.
Collapse
Affiliation(s)
- Miharu Yabe
- Department of Cell Transplantation, Tokai University School of Medicine, Kanagawa, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Tilak V, Sookmane DD, Gupta V, Shukla J. Myelodysplastic syndrome. Indian J Pediatr 2008; 75:729-32. [PMID: 18716744 DOI: 10.1007/s12098-008-0138-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Accepted: 05/27/2008] [Indexed: 11/28/2022]
Abstract
Pediatric myelodysplastic syndrome (MDS), though rare, constitutes a distinct entity quite different from adult MDS. They have unique clinical features, aggressive clinical course with an overall mean survival of only 9.9 months. A pediatric approach to the WHO classification has become necessary since the WHO classification of MDS has failed to address the uniqueness of pediatric MDS. A new prognostic system also needs to be evolved since the international prognostic system has limited prognostic impact in children. Intensive chemotherapy such as the one used in de novo-acute myeloid leukemia (AML) leads to complete remission in some children and this may be the treatment of choice in pediatric MDS.
Collapse
Affiliation(s)
- V Tilak
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| | | | | | | |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Myelodysplastic and myeloproliferative disorders are clonal myeloid malignancies characterized by the triad of a growth advantage of clonal cells, disturbed differentiation and increased apoptosis. The rarity of these disorders in children and the lack of a widely accepted classification have contributed to the paucity of reports on these malignancies in the pediatric literature. A number of significant advances have been achieved in recent years. The present review will focus on diagnostics and therapy. RECENT FINDINGS International consensus has been achieved on classifying these disorders into three main groups; myelodysplastic syndrome (MDS), myeloid leukemia of Down syndrome (ML-DS) and juvenile myelomonocytic leukemia (JMML). In the last few years we have witnessed important advances, especially regarding the therapy of these disorders, and we have gained insights into the molecular pathogenesis of ML-DS and JMML. SUMMARY Classification of myelodysplastic and myeloproliferative disorders has been facilitated. Chemotherapy regimens for ML-DS have been reduced, resulting in fewer toxic deaths and improved survival. The results of stem-cell transplantation for MDS and JMML have improved. Insight into the molecular mechanisms involved may open new therapeutic avenues.
Collapse
MESH Headings
- Child
- Diagnosis, Differential
- Hematopoietic Stem Cell Transplantation
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/classification
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myelomonocytic, Chronic/classification
- Leukemia, Myelomonocytic, Chronic/diagnosis
- Leukemia, Myelomonocytic, Chronic/therapy
- Myelodysplastic Syndromes/classification
- Myelodysplastic Syndromes/diagnosis
- Myelodysplastic Syndromes/therapy
- Myeloproliferative Disorders/classification
- Myeloproliferative Disorders/diagnosis
- Myeloproliferative Disorders/therapy
- Prognosis
- Transplantation Conditioning
Collapse
Affiliation(s)
- Henrik Hasle
- Department of Pediatrics, Aarhus University Hospital Skejby, Aarhus, Denmark.
| |
Collapse
|
25
|
Yoshimi A, Mohamed M, Bierings M, Urban C, Korthof E, Zecca M, Sykora KW, Duffner U, Trebo M, Matthes-Martin S, Sedlacek P, Klingebiel T, Lang P, Führer M, Claviez A, Wössmann W, Pession A, Arvidson J, O'Marcaigh AS, van den Heuvel-Eibrink MM, Starý J, Hasle H, Nöllke P, Locatelli F, Niemeyer CM. Second allogeneic hematopoietic stem cell transplantation (HSCT) results in outcome similar to that of first HSCT for patients with juvenile myelomonocytic leukemia. Leukemia 2007; 21:556-60. [PMID: 17268527 DOI: 10.1038/sj.leu.2404537] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Sauer M, Zeidler C, Meissner B, Rehe K, Hanke A, Welte K, Lohse P, Sykora KW. Substitution of cyclophosphamide and busulfan by fludarabine, treosulfan and melphalan in a preparative regimen for children and adolescents with Shwachman–Diamond syndrome. Bone Marrow Transplant 2007; 39:143-7. [PMID: 17211437 DOI: 10.1038/sj.bmt.1705553] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the only definitive treatment for severe bone marrow dysfunction and clonal disorders in patients diagnosed with Shwachman-Diamond syndrome (SDS). In an attempt to minimize regimen-related toxicity (RRT), we have initiated a fludarabine/treosulfan/melphalan-based pilot protocol avoiding the combination of busulfan and cyclophosphamide. Median age at transplantation was 9.6 years (range 1.5-17 years). All three patients received conditioning with fludarabine (30 mg/m2/day x 6), treosulfan (12 g/m2/day x 3) and melphalan (140 mg/m2/day x 1). CAMPATH-1H (0.1 mg/kg x 2) was added in two cases, while rabbit ATG (Genzyme; 3 x 2.5 mg/kg) was given to the cord blood recipient. One patient was transplanted with a non-manipulated marrow graft from an HLA-identical sibling, one with a marrow graft from a 10/10 matched unrelated donor, and one with a 9/10 matched unrelated umbilical cord blood (UCB) unit. Mean cell doses given were 3.6 x 10(8) nucleated cells/kg BW for the bone marrow recipients and 4.2 x 10(7) nucleated cells/kg BW for UCB recipient. Overall, two of three patients are alive and display 100% donor chimerism. Acute graft-versus-host disease grade II was seen in one patient, while no GVHD exceeding grade I occurred in the remaining two.
Collapse
Affiliation(s)
- M Sauer
- Department of Pediatric Hematology - Oncology, Hannover Medical University, Hannover, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Stachel DK, Leipold A, Kuhlen M, Gravou-Apostolatou C, Hirv K, Bader P, Niemeyer CM, Beck JD, Holter W. Simultaneous control of third-degree graft-versus-host disease and prevention of recurrence of juvenile myelomonocytic leukemia (JMML) with 6-mercaptopurine following fulminant JMML relapse early after KIR-mismatched bone marrow transplantation. J Pediatr Hematol Oncol 2005; 27:672-4. [PMID: 16344675 DOI: 10.1097/01.mph.0000193471.91690.f4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The authors describe a young boy with juvenile myelomonocytic leukemia (JMML) who relapsed 45 days after HLA and killer immunoglobulin-like receptor (KIR) mismatched unrelated donor bone marrow transplant (MMUD-BMT) and subsequently developed life-threatening graft-versus-host disease (GvHD). Treatment with 6-mercaptopurine (6-MP) appeared to control severe GvHD and possibly prevented recurrence of leukemic relapse.
Collapse
Affiliation(s)
- Daniel K Stachel
- Department of Pediatrics, University of Erlangen-Nurnberg, Erlangen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Korthof ET, Snijder PP, de Graaff AA, Lankester AC, Bredius RGM, Ball LM, Lie JLWT, Vossen JM, Egeler RM. Allogeneic bone marrow transplantation for juvenile myelomonocytic leukemia: a single center experience of 23 patients. Bone Marrow Transplant 2005; 35:455-61. [PMID: 15654356 PMCID: PMC7091614 DOI: 10.1038/sj.bmt.1704778] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a childhood leukemia for which allogeneic BMT is the only curative therapy. At our pediatric stem cell transplantation unit, we performed 26 BMTs in 23 children (age 0.5–12.7 years). Conditioning was CY/TBI based (1980–1996, n=14) or BU/CY/melphalan based (1996–2001, n=9). Donors were HLA-identical siblings (n=11), unrelated volunteers (n=9) or mismatched family members (n=3). A total of 10 patients survive in CR (median follow-up 6.8 years, range 3.1–22.2 years). Relapse or persistent disease was observed in eight and two patients, respectively. Nine of these patients died, one achieved a second remission following acute nonlymphatic leukemia chemotherapy (duration to date 5.3 years). Transplant-related mortality occurred in four patients. Overall survival at 5 and 10 years was 43.5%. Using T-cell-depleted, one-antigen mismatched unrelated donors was the only significant adverse factor associated with relapse in multivariate analysis (P=0.039, hazard ratio 4.9). Together with a trend towards less relapse in patients with graft-versus-host-disease and in patients transplanted with matched unrelated donors, this suggests a graft-versus-leukemia effect of allogeneic BMT in JMML.
Collapse
Affiliation(s)
- E T Korthof
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Yoshimi A, Bader P, Matthes-Martin S, Starý J, Sedlacek P, Duffner U, Klingebiel T, Dilloo D, Holter W, Zintl F, Kremens B, Sykora KW, Urban C, Hasle H, Korthof E, Révész T, Fischer A, Nöllke P, Locatelli F, Niemeyer CM. Donor leukocyte infusion after hematopoietic stem cell transplantation in patients with juvenile myelomonocytic leukemia. Leukemia 2005; 19:971-7. [PMID: 15800672 DOI: 10.1038/sj.leu.2403721] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a clonal myeloproliferative disorder of early childhood. In all, 21 patients with JMML who received donor leukocyte infusion (DLI) after allogeneic hematopoietic stem cell transplantation (HSCT) for either mixed chimerism (MC, n=7) or relapse (n=14) were studied. Six patients had been transplanted from an HLA-matched sibling and 15 from other donors. Six of the 21 patients (MC: 3/7 patients; relapse: 3/14 patients) responded to DLI. Response rate was significantly higher in patients receiving a higher total T-cell dose (> or =1 x 10(7)/kg) and in patients with an abnormal karyotype. None of the six patients receiving DLI from a matched sibling responded. Response was observed in five of six patients who did and in one of 15 children who did not develop acute graft-versus-host disease following DLI (P=0.01). The overall outcome was poor even for the responders. Only one of the responders is alive in remission, two relapsed, and three died of complications. In conclusion, this study shows that some cases of JMML may be sensitive to DLI, this providing evidence for a graft-versus-leukemia effect in JMML. Infusion of a high number of T cells, strategies to reduce toxicity, and cytoreduction prior to DLI may improve the results.
Collapse
Affiliation(s)
- A Yoshimi
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University of Freiburg, Freiburg 79106, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Koyama M, Nakano T, Takeshita Y, Sakata A, Sawada A, Yasui M, Okamura T, Inoue M, Kawa K. Successful treatment of JMML with related bone marrow transplantation after reduced-intensity conditioning. Bone Marrow Transplant 2005; 36:453-4; author reply 454. [PMID: 15968292 DOI: 10.1038/sj.bmt.1705047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
31
|
Yoshimi A, Niemeyer CM, Bohmer V, Duffner U, Strahm B, Kreyenberg H, Dilloo D, Zintl F, Claviez A, Wössmann W, Kremens B, Holter W, Niethammer D, Beck JF, Kontny U, Nöllke P, Klingebiel T, Bader P. Chimaerism analyses and subsequent immunological intervention after stem cell transplantation in patients with juvenile myelomonocytic leukaemia. Br J Haematol 2005; 129:542-9. [PMID: 15877738 DOI: 10.1111/j.1365-2141.2005.05489.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Chimaerism analysis was performed by polymerase chain reaction amplification of short-tandem repeat markers in 30 children following haematopoietic stem cell transplantation for juvenile myelomonocytic leukaemia (JMML). Fourteen patients always had complete chimaerism (CC); one of them relapsed after the discontinuation of the study and 13 continued in complete remission (CR). Mixed chimaerism (MC) was noted in 16 patients. Of those 12 patients demonstrated increasing MC (i-MC); 10 relapsed and two achieved CC following discontinuation of immunosuppressive therapy (IST). Four other patients demonstrating only transient MC are alive in CR. MC with up to 20% of autologous cells could be successfully eradicated without induction of severe graft-versus-host disease when IST was reduced or discontinued directly after the first demonstration of MC. At the same time, MC with up to 10% of autologous cells could disappear without intervention. The interval between MC and relapse ranged from 0-320 d (median 38 d). Donor leucocyte infusion was given to six patients with i-MC, but only one patient responded. Peripheral blood seems as valuable as bone marrow for chimaerism studies. In conclusion, serial quantitative chimaerism studies can identify patients with i-MC who are at high risk for relapse of JMML. Immediate withdrawal of IST is advised in these patients.
Collapse
Affiliation(s)
- Ayami Yoshimi
- Department of Paediatrics and Adolescent Medicine, University of Freiburg, Mathildenstrasse 1, 79106 Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hasegawa D, Manabe A, Kubota T, Kawasaki H, Hirose I, Ohtsuka Y, Tsuruta T, Ebihara Y, Goto YI, Zhao XY, Sakashita K, Koike K, Isomura M, Kojima S, Hoshika A, Tsuji K, Nakahata T. Methylation status of the p15 and p16 genes in paediatric myelodysplastic syndrome and juvenile myelomonocytic leukaemia. Br J Haematol 2005; 128:805-12. [PMID: 15755284 DOI: 10.1111/j.1365-2141.2005.05392.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aberrant DNA methylation is frequently observed in adults with myelodysplastic syndrome (MDS), and is recognized as a critical event in the disease's pathogenesis and progression. This is the first report to investigate the methylation status of p15 and p16, cell cycle regulatory genes, in children with MDS (n = 9) and juvenile myelomonocytic leukaemia (JMML; n = 18) by using a methylation-specific polymerase chain reaction. The frequency of p15 hypermethylation in paediatric MDS was 78% (7/9), which was comparable to that in adult MDS. In contrast, p15 hypermethylation in JMML was a rare event (17%; 3/18). In JMML, clinical and laboratory characteristics including PTPN11 mutations and aberrant colony formation were not different between the three patients with hypermethylated p15 and the others. Aberrant methylation of p16 was not detected in children with either MDS or JMML. Since p15 and p16 genes were unmethylated in two children with JMML, in whom the disease had progressed with an increased number of blasts, a condition referred to as blastic crisis, we infer that the aberrant methylation of these genes is not responsible for the progression of JMML. The results suggest that demethylating agents may be effective in most children with MDS and a few patients with JMML.
Collapse
Affiliation(s)
- Daisuke Hasegawa
- Department of Paediatric Haematology-Oncology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Peters C, Schrauder A, Schrappe M, von Stackelberg A, Stary J, Yaniv I, Gadner H, Klingebiel T. Allogeneic haematopoietic stem cell transplantation in children with acute lymphoblastic leukaemia: the BFM/IBFM/EBMT concepts. Bone Marrow Transplant 2005; 35 Suppl 1:S9-11. [PMID: 15812540 DOI: 10.1038/sj.bmt.1704835] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Children with high risk or relapsed acute lymphoblastic leukaemia (ALL) can benefit from allogeneic haematopoietic stem cell transplantation (SCT). To reduce transplantation-associated complications, the BFM study group, the IBFM study group and the PD-WP-EBMT initiated a prospective cooperative multicentre trail for paediatric ALL patients with an indication for allogeneic stem cell transplantation. Four-digit high-resolution HLA typing for all nonsibling donors, standardised GvHD prophylaxis and therapy, uniform conditioning regimen and minimum standards for supportive care should reduce not only treatment-related mortality but also ameliorate late effects for young patients. Furthermore, the prospective evaluation aims to assess the role of haematopoietic SCT in comparison to chemotherapy to enable valuable treatment recommendations for further decisions.
Collapse
Affiliation(s)
- C Peters
- St Anna Kinderspital, Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Manabe A, Yoshimasu T, Ebihara Y, Yagasaki H, Wada M, Ishikawa K, Hara J, Koike K, Moritake H, Park YD, Tsuji K, Nakahata T. Viral Infections in Juvenile Myelomonocytic Leukemia: Prevalence and Clinical Implications. J Pediatr Hematol Oncol 2004; 26:636-641. [PMID: 27811604 DOI: 10.1097/01.mph.0000140653.50344.5c] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Viral infections may complicate the diagnosis of juvenile myelomonocytic leukemia (JMML) in a substantial proportion of patients, but this possibility has not been tested in a prospective study. The authors therefore measured the cellular expression of the MxA protein, a reliable marker of viral infection, at diagnosis in children with JMML to estimate the prevalence of such infections. METHODS Eighteen children, aged 1 to 69 months, who met the diagnostic criteria of the International JMML Working Group were prospectively studied. MxA expression was assessed by flow cytometric analysis of peripheral blood mononuclear cells stained with an antihuman MxA antibody. All data were obtained through the MDS Committee of the Japanese Society of Pediatric Hematology. RESULTS Twelve patients (67%) had elevated levels of the MxA protein, with rotavirus, RS virus, or CMV infection documented in three of these patients. Although none of the patients had primary Epstein-Barr virus (EBV) infection, reactivation of the virus was strongly suspected in four children, including two with monosomy 7, each having increased levels of MxA. Southern blot analysis revealed monoclonal integration of the EBV genome into bone marrow mononuclear cells from one of these patients. There was no discernible correlation between increases in the marker protein and the presenting features or course of the disease. CONCLUSIONS Viral infection may be present in two thirds of children with newly diagnosed JMML, but it does not constitute a basis for revising clinical management. The possibility that EBV or other viruses contribute to JMML pathogenesis by stimulating pre-exiting malignant clones warrants further investigation.
Collapse
Affiliation(s)
- Atsushi Manabe
- From the *Department of Pediatric Hematology-Oncology, Institute of Medical Science, University of Tokyo, Tokyo; †Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan; ‡Department of Pediatrics, Osaka University, Osaka, Japan; §Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan; ¶Department of Pediatrics, Miyazaki Medical College, Miyazaki, Japan; ∥Department of Pediatrics, Nara Medical University, Nara, Japan; and **Department of Pediatrics, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Valera ET, Latorre MDRD, Mendes WL, Seber A, Lee MLM, de Paula MJA, Loggetto SR, Velloso E, Niero-Melo L, Lopes LF. Treatment of pediatric myelodysplastic syndromes and juvenile myelomonocytic leukemia: the Brazilian experience in the past decade. Leuk Res 2004; 28:933-9. [PMID: 15234570 DOI: 10.1016/j.leukres.2004.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2003] [Revised: 01/13/2004] [Accepted: 01/14/2004] [Indexed: 11/25/2022]
Abstract
BACKGROUND Therapy strategies for myelodysplastic syndromes (MDS) and juvenile myelomonocytic leukemia (JMML) vary considerably. OBJECTIVE To review the treatment of Brazilian children who were diagnosed with MDS or JMML in the past decade and reported to the Brazilian Cooperative Group on Pediatric Myelodysplastic Syndromes (BCG-MDS-PED). RESULTS Of 173 children reported to the BCG-MDS-PED from January 1997 to January 2003 with a suspected diagnosis of MDS or JMML, 91 had the diagnosis confirmed after central review of the bone marrow aspirate and biopsy. Information on previous treatments was available for 78 MDS/JMML patients. Treatment varied from different schedules of low-dose (14%) and standard-dose chemotherapy (50%), granulocyte-colony-stimulating factor (G-CSF 7%), interferon (5%), steroids (2%) and erythropoietin (2%) to allogeneic stem-cell transplantation (SCT) (14%). No survival advantage could be demonstrated based on Hasle's classification or based on treatment. CONCLUSION This report reflects the current practice in treating Brazilian children with MDS/JMML without specific Cooperative Group guidelines. Treatment modalities were very heterogeneous. The strategies for implementing a national protocol should consider international guidelines and focus on local experience and available resources.
Collapse
Affiliation(s)
- Elvis Terci Valera
- Departamento de Puericultura e Pediatria, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes, 3900 Monte Alegre, CEP 14049-900, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Imamura T, Matsuo S, Yoshihara T, Chiyonobu T, Mori K, Ishida H, Nishimura Y, Kasubuchi Y, Naya M, Morimoto A, Hibi S, Imashuku S. Granulocytic Sarcoma Presenting with Severe Adenopathy (Cervical Lymph Nodes, Tonsils, and Adenoids) in a Child with Juvenile Myelomonocytic Leukemia and Successful Treatment with Allogeneic Bone Marrow Transplantation. Int J Hematol 2004; 80:186-9. [PMID: 15481450 DOI: 10.1532/ijh97.04040] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The occurrence of adenopathy in patients with myelodysplastic syndrome-associated extramedullary myeloid cell tumors has rarely been reported. We describe a 7-year-old girl with juvenile myelomonocytic leukemia who showed the novel chromosomal abnormality t(9;12)(p22;q24.1) and who developed severe adenopathy of the cervical lymph nodes, tonsils, and adenoids that was manifested as granulocytic sarcoma. Following chemotherapy, the patient underwent a conditioning regimen of busulfan, cyclophosphamide, and total body irradiation followed by successful allogeneic bone marrow transplantation from her single HLA locus-mismatched mother at 6 months after her diagnosis. The patient continues to be well and in remission 3 years after stem cell transplantation.
Collapse
Affiliation(s)
- Toshihiko Imamura
- Department of Pediatrics, Matsushita Memorial Hospital, Moriguchi, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kalwak K, Wójcik D, Gorczyńska E, Toporski J, Turkiewicz D, Slociak M, Ussowicz M, Pajdosz K, Socha P, Chybicka A. Allogeneic hematopoietic cell transplantation from alternative donors in children with myelodysplastic syndrome: is that an alternative? Transplant Proc 2004; 36:1574-7. [PMID: 15251388 DOI: 10.1016/j.transproceed.2004.05.081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) in children with myelodysplastic syndrome (MDS) remains a challenge due to the toxic conditioning regimens administered to minimize the risk of relapse in the HLA-matched or of graft rejection in the HLA-mismatched settings. In the absence of matched sibling donors, alternative donors such as unrelated and/or partially matched family sources remain risky, yet the only available, options. Herein we report the results of HCT from alternative donors in 14 children with different subtypes of MDS (juvenile myelomonocytic leukemia [JMML] n = 9; myelodysplastic syndrome [MDS] refractory anemia n = 3; MDS refractory anemia with excess of blasts in transformation n = 2) transplanted at our institution. The median time from diagnosis to HCT was 9 months (range 4 to 90 months). The variety of HCT types included: unrelated peripheral blood progenitor cell transplantation (PBPCT) (n = 2), partially matched family donor T-cell-repleted BMT/PBPCT (n = 6), and haploidentical T-cell-depleted PBPCT (n = 6). Five of 14 patients remain alive at 7 to 37 months posttransplant (including two patients after partially matched family donor BMT, two patients after haploidentical T-cell-depleted-PBPCT, and one after unrelated-PBPCT, respectively). The major complications were: primary graft failure in the haploidentical T-cell-depleted-setting or graft-versus-host disease (GvHD) in T-cell-repleted partially matched family or unrelated settings, respectively. Despite the high transplant-related mortality rate in this series, allogeneic HCT from alternative donors remains an interesting solution for children with MDS who lack matched sibling donors. Due to improved immune reconstitution, despite an increased risk of GvHD, T-cell-repleted transplants from single HLA-mismatched family donors remain a valuable option for children without matched donors. Splenectomy prior to HCT may positively affect the posttransplant course in patients with overt splenomegaly for example those afflicted with JMML.
Collapse
Affiliation(s)
- K Kalwak
- Department of Pediatric Hematology/Oncology, Wroclaw University of Medicine, Wroclaw, Poland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Juvenile myelomonocytic leukemia is an aggressive neoplasia of early childhood. Only allogeneic stem cell transplantation (SCT) offers long-term cure. In the absence of an HLA-matched family donor, early SCT from an unrelated donor is the treatment of choice for most children. With clear evidence of a graft-versus-leukemia effect and a high post-transplant relapse rate, the outcome of SCT depends, in part, on the management of immunosuppression during the procedure. The impact of pretransplant cytoreductive treatment, such as intensive chemotherapy, splenectomy, or 13-cis retinoic acid, is unclear. Hypersensitivity for granulocyte-macrophage colony-stimulating factor and pathologic activation of the Ras/MAPK pathway play an important role in the pathophysiology of juvenile myelomonocytic leukemia and provide the opportunity for several novel therapeutic approaches.
Collapse
Affiliation(s)
- Charlotte Marie Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Mathildenstrasse 1, 79106 Freiburg, Germany.
| | | |
Collapse
|
39
|
Chang YH, Jou ST, Lin DT, Lu MY, Lin KH. Differentiating juvenile myelomonocytic leukemia from chronic myeloid leukemia in childhood. J Pediatr Hematol Oncol 2004; 26:236-42. [PMID: 15087951 DOI: 10.1097/00043426-200404000-00005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare clonal myeloproliferative disease of early childhood. To determine the diagnostic features, appropriate treatment, and overall patient survival pertaining to JMML for children, the authors reviewed the clinical data of 16 children with JMML admitted to the National Taiwan University Hospital between 1978 and 2001. Median age at diagnosis was 2.5 years. Fever was the most common symptom at diagnosis. At initial presentation, the mean white blood count and absolute monocyte count were 30 x 10(9)/L and 4.5 x 10(9)/L, respectively. Cytogenetic analysis was performed in 14 patients, and 2 patients (14%) had monosomy 7. Another patient, with normal karyotype at diagnosis, had deletion of 7q22 at the follow-up chromosome study. Forty-seven chronic myeloid leukemia (CML) patients were also diagnosed and followed at the same hospital during the same interval period. The age, leukocyte counts, platelet counts, basophil counts, monocyte percentages on peripheral blood smears, and median survival rate showed significant differences between JMML and CML patients (P < 0.05). The median survival was 10 months and the probability of 10-month survival was 0.38 by Kaplan-Meier analysis for 12 of the 16 JMML patients who did not receive hematopoietic stem cell transplantation (HSCT). Among three patients receiving HSCT, one patient relapsed 9 months after the first HSCT and was treated successfully by a second HSCT from the same sibling donor.
Collapse
MESH Headings
- Age of Onset
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Child
- Child, Preschool
- Diagnosis, Differential
- Female
- Humans
- Infant
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Leukemia, Myelomonocytic, Chronic/diagnosis
- Leukemia, Myelomonocytic, Chronic/drug therapy
- Leukemia, Myelomonocytic, Chronic/mortality
- Male
- Survival Analysis
- Time Factors
- Treatment Outcome
Collapse
Affiliation(s)
- Yu-Hsiang Chang
- Department of Pediatrics, Veterans General Hospital-Kaohsiung, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|
40
|
Fujimaki K, Taguchi J, Fujita H, Hattori M, Yamazaki E, Takahashi N, Fujisawa S, Kanamori H, Maruta A, Ishigatsubo Y. Thiotepa/cyclophosphamide/TBI as a conditioning regimen for allogeneic hematopoietic stem cell transplantation in patients with myelodysplastic syndrome. Bone Marrow Transplant 2004; 33:789-92. [PMID: 15064685 DOI: 10.1038/sj.bmt.1704451] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In all, 18 patients (30-56 years; median 49) with MDS underwent allogeneic HSCT from related (n=12) or unrelated (n=6) donors after a conditioning regimen comprising thiotepa, cyclophosphamide, and TBI. GVHD prophylaxis consisted of cyclosporine (n=15) or tacrolimus (n=3) with short-course methotrexate. Four patients had low-risk disease (refractory anemia or complete remission after chemotherapy) and 14 patients had high-risk disease (RAEB, RAEB-t, or AML). Grade II-IV acute GVHD developed in six patients and chronic GVHD in 10. With a median follow-up of 31 months, the 2-year survival probability is 75% for low-risk patients and 57% for high-risk patients. One patient died of leukemia and six of treatment-related causes. This conditioning regimen requires further study in patients with MDS.
Collapse
Affiliation(s)
- K Fujimaki
- First Department of Internal Medicine, Yokohama City University School of Medicine, Yokohama, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Baker D, Cole C, Price J, Phillips M. Allogeneic bone marrow transplantation in juvenile myelomonocytic leukemia without total body irradiation. J Pediatr Hematol Oncol 2004; 26:200-3. [PMID: 15125615 DOI: 10.1097/00043426-200403000-00012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Allogeneic bone marrow transplantation (BMT) without a total body irradiation (TBI) conditioning regimen was investigated in children with juvenile myelomonocytic leukemia (JMML). Eight consecutive patients with JMML (n = 6) or monosomy 7 (n = 2) underwent BMT at a median age of 20 months. Donor source included fully matched related (n = 3), mismatched related (n = 2), or fully matched unrelated (n = 3). The conditioning regimen included busulfan, cyclophosphamide, and etoposide (VP16) (melphalan was substituted for VP16 in one patient). The first patient in the series underwent TBI. Graft-versus-host disease prophylaxis was with cyclosporin and methotrexate and in vivo T-cell depletion (Campath 1 g) for mismatched and unrelated transplants. Seven and two patients, respectively, received chemotherapy and splenectomy before BMT. At a median follow-up of 48 months after BMT, five patients remained in remission. The overall survival rate was 63% at 5 years. All deaths occurred in patients with refractory disease at the time of BMT. Allogeneic BMT without TBI appears to be effective therapy for JMML and avoids some of the potential late sequelae of TBI in preschool children.
Collapse
Affiliation(s)
- David Baker
- Department of Hematology, Princess Margaret Hospital, GPO Box D184, Perth, Western Australia 6001.
| | | | | | | |
Collapse
|
42
|
Chang YH, Jou ST, Lin DT, Lu MY, Lin KH. Second allogeneic hematopoietic stem cell transplantation for juvenile myelomonocytic leukemia: case report and literature review. J Pediatr Hematol Oncol 2004; 26:190-3. [PMID: 15125612 DOI: 10.1097/00043426-200403000-00009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare clonal myeloproliferative disease in young childhood. Hematopoietic stem cell transplantation (HSCT) is the only way to cure the disease, but relapse after HSCT remains a major cause of treatment failure. A 5-year-old girl with JMML, who had experienced a relapse after the first transplant, did not respond to donor lymphocyte infusion and withdrawal of immune-suppressing agents. She was successfully treated using a second transplant. Detailed reports from the English literature since 1988 relating to a total of 13 JMML patients undergoing a second transplant were reviewed. Seven of the 13 JMML patients (54%) were alive and disease-free, with a median follow-up of 53 months after the second transplant. Within the first 6 months following the initial transplant, 10 JMML patients suffered either autologous recovery (n = 6) or early relapse (n = 4). Seven of the 10 (70%) were alive, with a median survival period of 53 months after the second transplant. Six JMML patients underwent retransplantation within 6 months of the first transplant, with three of these (50%) alive at follow-ups of 24, 57, and 90 months after the second procedure. The authors conclude that a second transplant within 6 months may be worth considering for JMML patients who experience autologous recovery or earlier relapse after the first transplant.
Collapse
Affiliation(s)
- Yu-Hsiang Chang
- Department of Pediatrics, Veterans General Hospital-Kaohsiung, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|
43
|
Kang HJ, Shin HY, Choi HS, Ahn HS. Novel regimen for the treatment of juvenile myelomonocytic leukemia (JMML). Leuk Res 2004; 28:167-70. [PMID: 14654081 DOI: 10.1016/s0145-2126(03)00217-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The effective treatment for juvenile myelomonocytic leukemia (JMML) patients lacking access to stem cell transplantation remains unavailable. Here, we describe a promising result obtained with novel regimen comprised of a combination of chemotherapy and differentiation therapy. Five patients diagnosed as JMML were treated with a standard regimen (cytosine arabinoside (Ara-C) 100mg/m(2) per day continuous infusion (days 0-6), etoposide 100mg/m(2) per day (days 0-4), vincristine 1.5mg/m(2) per d (day 9) and isotretinoin 75-100mg/m(2) per day (days 10-20)). All patients responded to the standard regimen. Three of the five were later treated with salvage a regimen (Ara-C 100mg/m(2) per day continuous infusion (days 0-4), etoposide 100mg/m(2) per day (days 0-4) and Ara-C 15mg/m(2) per day SC (days 6-15)) when immature myeloid cells reappeared in the peripheral blood, the spleen increased or blast crisis occurred. Immature myeloid cells disappeared again after one cycle of salvage regimen in all patients. All patients are alive now with a median follow up duration of 27 months (8-69 months). Although the number of patients enrolled was limited, the standard and salvage regimens were found to be safe and effective alternatives for JMML patients without a matched donor. These regimens also could be used safely before stem cell transplantation.
Collapse
Affiliation(s)
- Hyoung Jin Kang
- Pediatric Oncology Branch, National Cancer Center, Gyenggi-do, Republic of Korea
| | | | | | | |
Collapse
|
44
|
Abstract
Juvenile myelomonocytic leukemia is an aggressive neoplasia of early childhood. Only allogeneic stem cell transplantation (SCT) offers a long-term cure. In the absence of an HLA-matched family donor, early SCT from an unrelated donor will be the treatment of choice for most children. With clear evidence of a graft-versus-leukemia effect and a high post-transplant relapse rate, outcome of SCT will depend, in part, on the management of immunosuppression during the procedure. The impact of pretransplant cytoreductive treatment, such as intensive chemotherapy, splenectomy, or 13-cis retinoic acid, is unclear. Hypersensitivity for granulocyte-macrophage colony-stimulating factor and pathologic activation of the Ras/MAPK pathway play an important role in the pathophysiology of juvenile myelomonocytic leukemia and will provide the opportunity for several novel therapy approaches.
Collapse
Affiliation(s)
- Charlotte Marie Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Mathildenstrasse 1, 79106 Freiburg, Germany.
| | | |
Collapse
|
45
|
Worth A, Rao K, Webb D, Chessells J, Passmore J, Veys P. Successful treatment of juvenile myelomonocytic leukemia relapsing after stem cell transplantation using donor lymphocyte infusion. Blood 2003; 101:1713-4. [PMID: 12393482 DOI: 10.1182/blood-2002-07-2011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare pediatric malignancy. Hematopoietic stem cell transplantation (SCT) is the only curative approach. However, relapse after SCT remains the major cause of treatment failure. Unlike most other pediatric malignancies, JMML may be susceptible to a graft-versus-leukemia (GVL) effect, although, unlike chronic myeloid leukemia, reports of response to donor lymphocyte infusions (DLIs) remain scanty. This is the first report that describes the successful treatment of relapsed JMML with DLI in the absence of further chemotherapy and provides definite proof of a GVL effect in JMML.
Collapse
Affiliation(s)
- Austen Worth
- Great Ormond Street Hospital for Children, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
46
|
Kröger N, Zabelina T, Guardiola P, Runde V, Sierra J, Van Biezen A, Niederwieser D, Zander AR, De Witte T. Allogeneic stem cell transplantation of adult chronic myelomonocytic leukaemia. A report on behalf of the Chronic Leukaemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT). Br J Haematol 2002; 118:67-73. [PMID: 12100129 DOI: 10.1046/j.1365-2141.2002.03552.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We report the results of 50 allogeneic transplantations from related (n = 43) or unrelated (n = 7) donors, performed for chronic myelomonocytic leukaemia (CMML) in 43 European centres. The median age at transplant was 44 years (range 19-61). Eighteen patients had excess blasts ranging from 5% to 30% at the time of transplantation. Two graft failures were observed (4%). Neutrophil (> 0.5 x 109/l) and platelet engraftment (> 50 x 109/l) was reached after a median of 17 d (range 11-38) and 27 d (range 11-48) respectively. Acute graft-versus-host disease (GvHD grade II-IV was seen in 35% of patients, while 20% developed severe-acute GvHD grade III/IV. Twenty-six patients (52%) died of treatment-related causes. After a median follow-up of 40 months (range 11-110), the 5-year-estimated overall survival was 21% (95% CI: 15-27%) and the 5-year-estimated disease-free survival (DFS) was 18% (95% CI: 13-23%). Earlier transplantation in the course of disease, male donor, use of unmanipulated grafts, allogeneic transplantation and occurrence of acute GvHD favoured better DFS, but did not reach statistical significance. The 5-year estimated probability of relapse was 49%. The data showed a trend for a lower relapse probability of acute GvHD grade II-IV (24% vs 54%; P = 0.07), and for a higher relapse rate in patients with T cell-depleted grafts (62% vs 45%), suggesting a 'graft-versus-CMML effect'.
Collapse
|
47
|
Abstract
Myelodysplastic syndromes (MDS) in children constitute a heterogeneous disorder, including "primary" MDS and MDS associated with constitutional disorders or metabolic diseases. The Franco-American-British (FAB) cytological classification for adults can be applied for childhood in 50 to 100% of the cases. The transformation into acute myeloblastic leukemia often occurs, but stabilisation or spontaneous regression of the disease may also be observed. Allogenic bone marrow transplantation is the best curative option when treatment is necessary.
Collapse
Affiliation(s)
- B Bader-Meunier
- Fédération de Pédiatrie, Hôpital de Bicêtre, 78, rue du Général-Leclerc, 94275 Le Kremlin-Bicêtre, France.
| |
Collapse
|
48
|
Manabe A, Okamura J, Yumura-Yagi K, Akiyama Y, Sako M, Uchiyama H, Kojima S, Koike K, Saito T, Nakahata T. Allogeneic hematopoietic stem cell transplantation for 27 children with juvenile myelomonocytic leukemia diagnosed based on the criteria of the International JMML Working Group. Leukemia 2002; 16:645-9. [PMID: 11960345 DOI: 10.1038/sj.leu.2402407] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2001] [Accepted: 11/21/2001] [Indexed: 11/08/2022]
Abstract
Prognostic factors of juvenile myelomonocytic leukemia (JMML) have not been clarified because of its very low incidence and inaccuracy in the diagnosis. The purpose of this study was to evaluate children with JMML given an allogeneic hematopoietic stem cell transplantation (SCT) and the role of different variables potentially influencing outcome in a nationwide survey in Japan based on the newly proposed criteria by the International JMML Working Group. The study patients were 27 children who underwent SCT among 55 JMML patients retrospectively collected in the survey. The source of grafts was HLA-identical siblings in 12 cases, HLA-matched unrelated individuals in 10 and others in five. Total body irradiation was used in 18 cases. Event-free and overall survival (OS) at 4 years after SCT were 54.2 +/- 11.2% (s.e.) and 57.9 +/- 11.0% (s.e.), respectively. Six patients died of relapse and three of complications. Patients with abnormal karyotypes showed a significantly lower OS than those with normal karyotypes (P < 0.001). Patients below 1 year of age showed a significantly higher OS than those of 1 year of age or more (P = 0.02). Patients with grade 0-1 acute graft-versus-host disease (GVHD) or chronic GVHD had a more favorable OS than those without them, although they were not statistically significant (P > 0.05). Other variables studied were not associated with OS. A multivariate analysis of these factors yielded the abnormal karyotype as the only significant risk factor for lower OS (risk ratio: 11.0; 95% CI: 2.7-45.1).
Collapse
Affiliation(s)
- A Manabe
- Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Smith FO, King R, Nelson G, Wagner JE, Robertson KA, Sanders JE, Bunin N, Emaunel PD, Davies SM. Unrelated donor bone marrow transplantation for children with juvenile myelomonocytic leukaemia. Br J Haematol 2002; 116:716-24. [PMID: 11849238 DOI: 10.1046/j.0007-1048.2001.03333.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Children with juvenile myelomonocytic leukaemia (JMML) have a poor outcome, with survival in a minority of patients. The major limitation on success of sibling donor bone marrow transplantation for JMML has been reported to be relapse. A total of 46 children with a diagnosis of JMML underwent unrelated donor marrow (URD) transplantation facilitated by the National Marrow Donor Program. Forty-three of 46 patients had neutrophil engraftment at a median of 20 d post transplant, with platelet recovery in 28 of 40 evaluable patients at a median of 34.5 d. Thirty-two of 44 evaluable patients developed acute graft-versus-host-disease (GVHD) (Grades 2-4) and chronic GVHD developed in 14 of 35 evaluable patients. At a median follow-up of 2.0 years, probabilities of survival and disease-free survival were 42% and 24% respectively. The probability of relapse was 58% at 2 years and represents the major cause of treatment failure. Multivariate analysis revealed that chronic GVHD was associated with reduced relapse [risk ratio 0.20 (95% CI 0.04-1.02, P=0.05)] improved survival [risk ratio 0.13 (95% CI 0.03-0.68, P=0.02)] and event-free survival [risk ratio 0.23 (95% CI 0.06-0.94, P=0.04)]. This study demonstrates that relapse is the major cause of treatment failure in patients with JMML undergoing URD transplantation. With lower relapse observed in patients with chronic GVHD, new treatment strategies that focus on enhancing the graft-versus-leukaemia effect may improve survival.
Collapse
Affiliation(s)
- Franklin O Smith
- Division of Hematology/Oncology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Arceci RJ, Longley BJ, Emanuel PD. Atypical cellular disorders. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2002; 2002:297-314. [PMID: 12446429 DOI: 10.1182/asheducation-2002.1.297] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Atypical cellular disorders are commonly considered part of the gray zone linking oncology to hematology and immunology. Although these disorders are relatively uncommon, they often represent significant clinical problems, provide an opportunity to understand basic disease mechanisms, and serve as model systems for the development of novel targeted therapies. This chapter focuses on such disorders. In Section I, Dr. Arceci discusses the pathogenesis of Langerhans cell histiocytosis (LCH) in terms of the hypothesis that this disorder represents an atypical myeloproliferative syndrome. The clinical manifestations and treatment of LCH in children and adults is discussed along with possible future therapeutic approaches based upon biological considerations. In Section II, Dr. Longley considers the molecular changes in the c-Kit receptor that form the basis of mastocytosis. Based on the location and function of c-Kit mutations, he develops a paradigm for the development of specific, targeted therapies. In Section III, Dr. Emanuel provides a review of the "mixed myeloproliferative and myelodysplastic disorders," including novel therapeutic approaches based on aberrant pathogenetic mechanisms. Taken together, these chapters should provide an overview of the biological basis for these disorders, their clinical manifestations, and new therapeutic approaches.
Collapse
Affiliation(s)
- Robert J Arceci
- Div. of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA
| | | | | |
Collapse
|