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
|
Bugarin C, Antolini L, Buracchi C, Matarraz S, Coliva TA, Van der Velden VH, Szczepanski T, Da Costa ES, Van der Sluijs A, Novakova M, Mejstrikova E, Nierkens S, De Mello FV, Fernandez P, Aanei C, Sędek Ł, Strocchio L, Masetti R, Sainati L, Philippé J, Valsecchi MG, Locatelli F, Van Dongen JJM, Biondi A, Orfao A, Gaipa G. Phenotypic profiling of CD34 + cells by advanced flow cytometry improves diagnosis of juvenile myelomonocytic leukemia. Haematologica 2024; 109:521-532. [PMID: 37534527 PMCID: PMC10828789 DOI: 10.3324/haematol.2023.282805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023] Open
Abstract
Diagnostic criteria for juvenile myelomonocytic leukemia (JMML) are currently well defined, however in some patients diagnosis still remains a challenge. Flow cytometry is a well established tool for diagnosis and follow-up of hematological malignancies, nevertheless it is not routinely used for JMML diagnosis. Herewith, we characterized the CD34+ hematopoietic precursor cells collected from 31 children with JMML using a combination of standardized EuroFlow antibody panels to assess the ability to discriminate JMML cells from normal/reactive bone marrow cell as controls (n=29) or from cells of children with other hematological diseases mimicking JMML (n=9). CD34+ precursors in JMML showed markedly reduced B-cell and erythroid-committed precursors compared to controls, whereas monocytic and CD7+ lymphoid precursors were significantly expanded. Moreover, aberrant immunophenotypes were consistently present in CD34+ precursors in JMML, while they were virtually absent in controls. Multivariate logistic regression analysis showed that combined assessment of the number of CD34+CD7+ lymphoid precursors and CD34+ aberrant precursors or erythroid precursors had a great potential in discriminating JMMLs versus controls. Importantly our scoring model allowed highly efficient discrimination of truly JMML versus patients with JMML-like diseases. In conclusion, we show for the first time that CD34+ precursors from JMML patients display a unique immunophenotypic profile which might contribute to a fast and accurate diagnosis of JMML worldwide by applying an easy to standardize single eight-color antibody combination.
Collapse
Affiliation(s)
- Cristina Bugarin
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza (MB)
| | - Laura Antolini
- Center of Biostatistics for Clinical Epidemiology, Dipartimento di Medicina e Chirurgia, Università degli Studi Milano-Bicocca, Monza (MB)
| | - Chiara Buracchi
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza (MB)
| | - Sergio Matarraz
- Cancer Research Center (IBMCC-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, CIBERONC and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca
| | | | | | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Medical University of Silesia (SUM), Zabrze
| | | | - Alita Van der Sluijs
- Department of Immunohematology and Blood Transfusion (IHB) Leiden University Medical Center (LUMC), Leiden
| | - Michaela Novakova
- CLIP-Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Ester Mejstrikova
- CLIP-Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Paula Fernandez
- Institute for Laboratory Medicine, Kantonsspital Aarau AG, Aarau
| | - Carmen Aanei
- Hematology Laboratory CHU de Saint-Etienne, Saint-Etienne, Cedex 2
| | - Łukasz Sędek
- Department of Pediatric Hematology and Oncology, Medical University of Silesia (SUM), Zabrze
| | - Luisa Strocchio
- Department of Pediatric Hematology and Oncology IRCCS Ospedale Pediatrico Bambino Gesu', Sapienza University of Rome
| | - Riccardo Masetti
- Pediatric Oncology and Hematology Unit 'Lalla Seràgnoli', IRCCS Azienda Ospedaliero- Universitaria di Bologna, Bologna
| | - Laura Sainati
- Dipartimento di Salute della Donna e del Bambino, Clinica di Oncoematologia Pediatrica, Azienda Ospedale Università di Padova, Padua
| | - Jan Philippé
- Department of Laboratory Medicine, Ghent University Hospital, Ghent
| | - Maria Grazia Valsecchi
- Center of Biostatistics for Clinical Epidemiology, Dipartimento di Medicina e Chirurgia, Università degli Studi Milano-Bicocca, Monza (MB).
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology IRCCS Ospedale Pediatrico Bambino Gesu', Sapienza University of Rome
| | - Jacques J M Van Dongen
- Cancer Research Center (IBMCC-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, CIBERONC and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Department of Immunohematology and Blood Transfusion (IHB) Leiden University Medical Center (LUMC), Leiden
| | - Andrea Biondi
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza (MB), Italy; Dipartimento di Medicina e Chirurgia, Università degli Studi Milano-Bicocca, Monza (MB).
| | - Alberto Orfao
- Cancer Research Center (IBMCC-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, CIBERONC and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca
| | - Giuseppe Gaipa
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza (MB)
| |
Collapse
|
3
|
Yang WY, Liu LP, Liu F, Qi BQ, Chang LX, Zhang L, Chen XJ, Zou Y, Chen YM, Guo Y, Zhu XF. [Clinical features and prognosis of juvenile myelomonocytic leukemia: an analysis of 63 cases]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:265-271. [PMID: 36946161 PMCID: PMC10032072 DOI: 10.7499/j.issn.1008-8830.2209129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
OBJECTIVES To investigate the clinical features of juvenile myelomonocytic leukemia (JMML) and their association with prognosis. METHODS Clinical and prognosis data were collected from the children with JMML who were admitted from January 2008 to December 2016, and the influencing factors for prognosis were analyzed. RESULTS A total of 63 children with JMML were included, with a median age of onset of 25 months and a male/female ratio of 3.2∶1. JMML genetic testing was performed for 54 children, and PTPN11 mutation was the most common mutation and was observed in 23 children (43%), among whom 19 had PTPN11 mutation alone and 4 had compound PTPN11 mutation, followed by NRAS mutation observed in 14 children (26%), among whom 12 had NRAS mutation alone and 2 had compound NRAS mutation. The 5-year overall survival (OS) rate was only 22%±10% in these children with JMML. Of the 63 children, 13 (21%) underwent hematopoietic stem cell transplantation (HSCT). The HSCT group had a significantly higher 5-year OS rate than the non-HSCT group (46%±14% vs 29%±7%, P<0.05). There was no significant difference in the 5-year OS rate between the children without PTPN11 gene mutation and those with PTPN11 gene mutation (30%±14% vs 27%±10%, P>0.05). The Cox proportional-hazards regression model analysis showed that platelet count <40×109/L at diagnosis was an influencing factor for 5-year OS rate in children with JMML (P<0.05). CONCLUSIONS The PTPN11 gene was the most common mutant gene in JMML. Platelet count at diagnosis is associated with the prognosis in children with JMML. HSCT can improve the prognosis of children with JMML.
Collapse
Affiliation(s)
- Wen-Yu Yang
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Li-Peng Liu
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Fang Liu
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Ben-Quan Qi
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Li-Xian Chang
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Li Zhang
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Xiao-Juan Chen
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yao Zou
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yu-Mei Chen
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Ye Guo
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Xiao-Fan Zhu
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
4
|
Yi ES, Baek HJ, Ju HY, Kim SK, Lee JW, Cho B, Kim BK, Kang HJ, Kook H, Yang EJ, Lim YT, Ahn WK, Hahn SM, Park SK, Yoo ES, Yoo KH. Response to chemotherapy in juvenile myelomonocytic leukemia and its clinical implications for survival: A retrospective registry-based study of the Korean Pediatric Hematology-Oncology Group. Leuk Res 2023; 129:107070. [PMID: 37019050 DOI: 10.1016/j.leukres.2023.107070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a life-threatening myeloproliferative neoplasm. The chemotherapeutic effect on survival remains unclear, and feasible standardized response criteria are yet to be established. We aimed to evaluate the chemotherapeutic response and its effect on survival in patients with JMML. A retrospective registry was reviewed for children diagnosed with JMML between 2000 and 2019. Response was assessed according to the criteria proposed by the International JMML Symposium in 2007 (criteria I) and the updated version in 2013 with its modifications (criteria II). A total of 73 patients were included in this study. Complete response (CR) rates were 46.6% and 28.8% using the criteria I and criteria II, respectively. A platelet count ≥ 40 × 109/L at diagnosis was associated with higher CR rates using the criteria II. Patients with criteria I-based CR had a better overall survival (OS) than those without CR (81.1% vs. 49.1% at 5 years). Patients with criteria II-based CR showed better OS (85.7% vs. 55.5% at 5 years) and event-free survival (EFS) (71.1% vs. 44.7% at 5 years) than those without CR. Additionally, a trend toward better EFS was observed in patients with criteria II-based CR than in those with criteria I-based CR but without criteria II-based CR (71.1% vs. 53.8% at 5 years). Chemotherapeutic response is associated with better survival outcomes. Along with splenomegaly, the addition of platelet count recovery, existence of extramedullary leukemic infiltration, and more stringent leukocyte counts to the response criteria allows for a more sensitive prediction of survival outcomes.
Collapse
|
5
|
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
|
6
|
Juvenile Myelomonocytic Leukemia in a Child: A Case Report of Palliative Chemotherapy and Literature Review Applied to Limited Resources Centers. Case Rep Hematol 2022; 2022:1185140. [DOI: 10.1155/2022/1185140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare hematopoietic malignancy in children, with an incidence of 1.2 per million children per year. At this moment, we present a case report and a brief literature review of JMML in a child, primarily focused on its applicability in low-middle income countries. A 3.5-year-old male was referred to our tertiary center due to pallor, enlarging abdomen and neck mass, recurrent fever, and chronic diarrhea. Initial laboratory workup showed hemoglobin of 6.4 g/dl, white blood cell of 315.62 × 103/μL, and platelet of 17 × 103/μL. Blood smears showed 10% suspected blasts, 17% myelocytes, and 17% metamyelocytes with thrombocytopenic crisis. The HbF level was 5.8%. BCR-ABL gene tested negative. The patient was diagnosed with juvenile myelomonocytic leukemia. Considering that HSCT could not be done in our center and lack other financial possibilities to seek treatment abroad, the family agreed to do the palliative treatment. The patient was treated with oral 6-mercaptopurine and subcutaneous cytarabine. Four weeks after receiving 6-mercaptopurine, the white blood cell count decreased to 10.6 × 103/μL and the spleen size was half of the original size. The patient continued chemotherapy until week 15, chemotherapy was stopped, but 16 weeks after the diagnosis of JMML, he developed severe thrombocytopenia, endophthalmitis, and sepsis and passed away. As a conclusion, in JMML cases in developing countries without HSCT, palliative chemotherapy is acceptable, and palliative care is an important aspect.
Collapse
|
7
|
Wintering A, Smith S, Fuh B, Rangaswami A, Dahl G, Chien M, Gruber TA, Dang J, Li LS, Lenzen A, Savelli S, Dvorak CC, Agrawal AK, Stieglitz E. Therapy-related myeloid neoplasms resembling juvenile myelomonocytic leukemia: a case series and review of the literature. Pediatr Blood Cancer 2022; 69:e29499. [PMID: 34939322 PMCID: PMC8957526 DOI: 10.1002/pbc.29499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 11/10/2022]
Abstract
Therapy-related myeloid neoplasms (t-MN) are a distinct subgroup of myeloid malignancies with a poor prognosis that include cases of therapy-related myelodysplastic syndrome (t-MDS), therapy-related myeloproliferative neoplasms (t-MPN) and therapy-related acute myeloid leukemia (t-AML). Here, we report a series of patients with clinical features consistent with juvenile myelomonocytic leukemia (JMML), an overlap syndrome of MDS and myeloproliferative neoplasms that developed after treatment for another malignancy.
Collapse
Affiliation(s)
- Astrid Wintering
- Department of Pediatrics, UCSF Benioff Children’s Hospital San Francisco, University of California San Francisco, San Francisco, CA 94158, USA
| | - Stephen Smith
- Department of Pediatrics, UCSF Benioff Children’s Hospital Oakland, Oakland, CA 94609, USA
| | - Beng Fuh
- Department of Pediatrics, East Carolina University, Greenville, NC 27834, USA
| | - Arun Rangaswami
- Department of Pediatrics, UCSF Benioff Children’s Hospital San Francisco, University of California San Francisco, San Francisco, CA 94158, USA
| | - Gary Dahl
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - May Chien
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Tanja A. Gruber
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Jinjun Dang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Loretta S. Li
- Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Alicia Lenzen
- Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Stephanie Savelli
- Department of Pediatrics, Akron Children’s Hospital, Akron, OH 44308, USA
| | - Christopher C. Dvorak
- Department of Pediatrics, UCSF Benioff Children’s Hospital San Francisco, University of California San Francisco, San Francisco, CA 94158, USA,Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA
| | - Anurag K. Agrawal
- Department of Pediatrics, UCSF Benioff Children’s Hospital Oakland, Oakland, CA 94609, USA
| | - Elliot Stieglitz
- Department of Pediatrics, UCSF Benioff Children’s Hospital San Francisco, University of California San Francisco, San Francisco, CA 94158, USA,Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
8
|
Oliveira AF, Tansini A, Toledo T, Balceiro R, Lee MLM, Villela N, Ikeuty P, Metze K, Lopes LF, Lorand-Metze I. Immunophenotypic changes in juvenile myelomonocytic leukaemia after treatment with hypomethylating agent: Do they help to evaluate dept of response? Br J Haematol 2022; 197:339-348. [PMID: 35187646 DOI: 10.1111/bjh.18089] [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] [Received: 11/03/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 11/29/2022]
Abstract
5-Azacitidine has been used before stem cell transplantation in juvenile myelomonocytic leukaemia (JMML) patients. Recently, we have described immunophenotypic features in JMML at diagnosis. Here, our aim was to examine the changes in the immunophenotypic features during azacitidine treatment, correlating it with clinical response. Patients treated with 5-azacitidine were evaluated at diagnosis and after three and six cycles of medication. Among 32 patients entering the study, 28 patients were examined after three cycles and 25 patients after six. Patients showed a reduction in CD34/CD117+ cells: median 3.35% at diagnosis, 2.8% after three cycles and 1.63% after six. B-cell progenitors were decreased at diagnosis and decreased after treatment. Monocytes decreased: 11.91% to 6.4% and 4.18% respectively. Complete response was associated with increase in classical monocytes. T lymphocytes, reduced at diagnosis, increased in patients responding to 5-azacitidine. Immunophenotypic aberrancies including expression of CD7 in myeloid progenitors remained after treatment. This feature was associated with a worse response to treatment, as well as presence of NF1. Immunophenotyping was feasible in all patients. Clinical response was associated with a decrease of myeloid progenitors and monocytes and a rise in T lymphocytes although phenotypic aberrancies persisted. The largest effect was observed after three cycles.
Collapse
Affiliation(s)
- Anita Frisanco Oliveira
- Barretos Children´s Cancer Hospital, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Myeloproliferative Diseases Committee, Barretos, Brazil
| | - Aline Tansini
- Barretos Children´s Cancer Hospital, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Morfology and Flow Cytometry Committee, Barretos, Brazil
| | - Thais Toledo
- Barretos Children´s Cancer Hospital, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Morfology and Flow Cytometry Committee, Barretos, Brazil
| | | | - Maria Lucia Martino Lee
- Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Myeloproliferative Diseases Committee, Barretos, Brazil
| | - Neysimelia Villela
- Barretos Children´s Cancer Hospital, Hematopoietic Stem Cell Transplantation, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), HSCT Committee, Barretos, Brazil
| | - Patricia Ikeuty
- Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), HSCT Committee, Barretos, Brazil
| | - Konradin Metze
- Department of Pathology, Faculty Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Luiz Fernando Lopes
- Barretos Children´s Cancer Hospital, Barretos, Brazil.,Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Barretos, Brazil
| | - Irene Lorand-Metze
- Brazilian Co-operative Group of Pediatric Myelodysplastic Syndrome (GCB-SMD-PED), Morfology and Flow Cytometry Committee, Barretos, Brazil.,Department of Internal Medicine, Faculty Medical Sciences, State University of Campinas, Campinas, Brazil
| |
Collapse
|
9
|
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
|
10
|
Juvenile myelomonocytic leukemia presenting in an infant with a subdural hematoma. Childs Nerv Syst 2021; 37:2075-2079. [PMID: 33404720 DOI: 10.1007/s00381-020-05013-7] [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: 11/14/2020] [Accepted: 12/09/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Juvenile myelomonocytic leukemia (JMML) is a rare childhood hematopoietic disorder typically presenting with hepatosplenomegaly, lymphadenopathy, pallor, fever, and cutaneous findings. The authors report the first case, to our knowledge, of JMML presenting in a pediatric patient with a subdural hematoma. CASE DESCRIPTION A 7-month old male with recurrent respiratory infections and a low-grade fever presented with a full fontanelle and an increasing head circumference and was found to have chronic bilateral subdural collections. Abusive head trauma, infectious, and coagulopathy workups were unremarkable, and the patient underwent bilateral burr holes for evacuation of the subdural collections. The postoperative course was complicated by the development of thrombocytopenia, anemia, and an acute subdural hemorrhage which required evacuation. Cytologic analysis of the subdural fluid demonstrated atypical cells, which prompted flow cytometric analysis, a bone marrow biopsy, and ultimately a diagnosis of JMML. Following chemotherapy, the patient's counts improved, and he subsequently underwent a hematopoietic stem cell transplant. CONCLUSION Subdural collections may rarely represent the first presenting sign of hematologic malignancies. In patients with a history of recurrent infections and a negative workup for abusive head trauma, clinicians should include neoplastic etiologies in the differential for chronic subdural collections and have a low threshold for fluid analysis.
Collapse
|
11
|
McCullough KB, Kuhn AK, Patnaik MM. Treatment advances for pediatric and adult onset neoplasms with monocytosis. Curr Hematol Malig Rep 2021; 16:256-266. [PMID: 33728588 DOI: 10.1007/s11899-021-00622-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW For decades, the management of chronic myelomonocytic leukemia (CMML) or juvenile myelomonocytic leukemia (JMML) has been largely inextricable from myelodysplastic syndromes (MDS), myeloproliferative neoplasms, and acute myeloid leukemia. Hallmarks of these diseases have been the emergence of unique genomic signatures and discouraging responses to available therapies. Here, we will critically examine the current options for management and review the rapidly developing opportunities based on advances in CMML and JMML disease biology. RECENT FINDINGS Few clinical trials have exclusively been done in CMML, and in JMML, the rarity of the disease limits wide scale participation. Recent case series in JMML suggest that hypomethylating agents (HMAs) are a viable option for bridging to curative intent with allogeneic hematopoietic stem cell transplant or as posttransplant maintenance. Emerging evidence has demonstrated targeting the RAS-pathway via MEK inhibition may also be considered. In CMML, treatment with HMAs is largely derived from data inclusive of MDS patients, including a small number of patients with dysplastic CMML variants. Based on CMML disease biology, additional therapeutic targets being investigated include inhibitors of splicing, CD123/dendritic cell axis, inherent GM-CSF progenitor cell hypersensitivity, and targeting the JAK/STAT pathway. Current evidence is also expanding for oral HMAs. The management of CMML and JMML is rapidly evolving and clinicians must be aware of the genetic landscape and expanding treatment options to ensure these rare populations are afforded therapeutic interventions best suited to their needs.
Collapse
Affiliation(s)
- Kristen B McCullough
- Department of Pharmacy Services, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
| | - Alexis K Kuhn
- Department of Pharmacy Services, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Mrinal M Patnaik
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
12
|
Schönung M, Meyer J, Nöllke P, Olshen AB, Hartmann M, Murakami N, Wakamatsu M, Okuno Y, Plass C, Loh ML, Niemeyer CM, Muramatsu H, Flotho C, Stieglitz E, Lipka DB. International Consensus Definition of DNA Methylation Subgroups in Juvenile Myelomonocytic Leukemia. Clin Cancer Res 2020; 27:158-168. [PMID: 33139265 DOI: 10.1158/1078-0432.ccr-20-3184] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/01/2020] [Accepted: 10/21/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Known clinical and genetic markers have limitations in predicting disease course and outcome in juvenile myelomonocytic leukemia (JMML). DNA methylation patterns in JMML have correlated with outcome across multiple studies, suggesting it as a biomarker to improve patient stratification. However, standardized approaches to classify JMML on the basis of DNA methylation patterns are lacking. We, therefore, sought to define an international consensus for DNA methylation subgroups in JMML and develop classification methods for clinical implementation. EXPERIMENTAL DESIGN Published DNA methylation data from 255 patients with JMML were used to develop and internally validate a classifier model. Accuracy across platforms (EPIC-arrays and MethylSeq) was tested using a technical validation cohort (32 patients). The suitability of both methods for single-patient classification was demonstrated using an independent cohort (47 patients). RESULTS Analysis of pooled, published data established three DNA methylation subgroups as a de facto standard. Unfavorable prognostic parameters (PTPN11 mutation, elevated fetal hemoglobin, and older age) were significantly enriched in the high methylation (HM) subgroup. A classifier was then developed that predicted subgroups with 98% accuracy across different technological platforms. Applying the classifier to an independent validation cohort confirmed an association of HM with secondary mutations, high relapse incidence, and inferior overall survival (OS), while the low methylation subgroup was associated with a favorable disease course. Multivariable analysis established DNA methylation subgroups as the only significant factor predicting OS. CONCLUSIONS This study provides an international consensus definition for DNA methylation subgroups in JMML. We developed and validated methods which will facilitate the design of risk-stratified clinical trials in JMML.
Collapse
Affiliation(s)
- Maximilian Schönung
- Section Translational Cancer Epigenomics, Division Translational Medical Oncology, German Cancer Research Center (DKFZ) & National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Julia Meyer
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, California
| | - Peter Nöllke
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Adam B Olshen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Mark Hartmann
- Section Translational Cancer Epigenomics, Division Translational Medical Oncology, German Cancer Research Center (DKFZ) & National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Norihiro Murakami
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Manabu Wakamatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yusuke Okuno
- Medical Genomics Center, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Christoph Plass
- Division Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Charlotte M Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), partner site Freiburg, Germany
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), partner site Freiburg, Germany
| | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Daniel B Lipka
- Section Translational Cancer Epigenomics, Division Translational Medical Oncology, German Cancer Research Center (DKFZ) & National Center for Tumor Diseases (NCT), Heidelberg, Germany. .,Faculty of Medicine, Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
13
|
Wan Z, Han B. Comparison and Implications of Mutational Profiles of Myelodysplastic Syndromes, Myeloproliferative Neoplasms, and Myelodysplastic/Myeloproliferative Neoplasms: A Meta-Analysis. Front Oncol 2020; 10:579221. [PMID: 33117717 PMCID: PMC7575718 DOI: 10.3389/fonc.2020.579221] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Dysplasia and proliferation are histological properties that can be used to diagnose and categorize myeloid tumors in myelodysplastic syndromes (MDS) and myeloproliferative neoplasms (MPN). However, these conditions are not exclusive, and overlap between them leads to another classification, MDS/MPN. As well as phenotype continuity, these three conditions may have genetic relationships that have not yet been identified. This study aimed to obtain their mutational profiles by meta-analysis and explore possible similarities and differences. We reviewed screening studies of gene mutations, published from January 2000 to March 2020, from PubMed and Web of Science. Fifty-three articles were eligible for the meta-analysis, and at most 9,809 cases were involved for any gene. The top mutant genes and their pooled mutation rates were as follows: SF3B1 (20.2% [95% CI 11.6-30.5%]) in MDS, TET2 (39.2% [95% CI 21.7-52.0%]) in MDS/MPN, and JAK2 (67.9% [95% CI 64.1-71.6%]) in MPN. Subgroup analysis revealed that leukemic transformation-related genes were more commonly mutated in high-risk MDS (MDS with multilineage dysplasia and MDS with excess blasts) than that in other MDS entities. Thirteen genes including ASXL1, U2AF1, SRSF2, SF3B1, and ZRSR2 had significantly higher mutation frequencies in primary myelofibrosis (PMF) compared with essential thrombocythemia and polycythemia vera; this difference distinguished PMF from MPN and likened it to MDS. Chronic myelomonocytic leukemia and atypical chronic myeloid leukemia were similar entities but showed several mutational differences. A heat map demonstrated that juvenile myelomonocytic leukemia and MDS/MPN with ring sideroblasts and thrombocytosis were two distinct entities, whereas MDS/MPN-unclassifiable was closest to high-risk MDS. Such genetic closeness or difference reflected features in the pathogenesis, diagnosis, treatment, and progression of these conditions, and could inspire future genetic studies.
Collapse
Affiliation(s)
- Ziqi Wan
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Bing Han
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
14
|
Frisanco Oliveira A, Tansini A, Toledo TR, Balceiro R, Onofre Vidal D, de Martino Lee ML, Lorand-Metze I, Lopes LF. Immunophenotypic characteristics of juvenile myelomonocytic leukaemia and their relation with the molecular subgroups of the disease. Br J Haematol 2020; 192:129-136. [PMID: 32966606 DOI: 10.1111/bjh.17098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/18/2020] [Indexed: 11/30/2022]
Abstract
The diagnosis of juvenile myelomonocytic leukaemia (JMML) is based on clinical, laboratory and molecular features but immunophenotyping [multiparametric flow cytometry (MFC)] has not been used routinely. In the present study, we describe the flow cytometric features at diagnosis with special attention to the distribution of monocytic subsets and the relation between MFC and molecular subgroups. MFC was performed with an eight-colour platform based on Euroflow. We studied 33 JMML cases. CD34+ /CD117+ /CD13+ cells >2% was found in 25 cases, and 51·5% presented an aberrant expression of CD7. A decrease of CD34+ /CD19+ /CD10+ cells was seen in eight cases and in four they were absent. The granulocytic population had a decreased side scatter in 29 cases. Bone marrow monocytic precursors were increased in 28 patients, with a decrease in classical monocytes (median 80·7%) and increase in CD16+ (intermediate and non-classical). A more pronounced increase in myeloid CD34+ cells was seen in patients with Neurofibromatosis type 1 (NF1) and tyrosine-protein phosphatase non-receptor type 11 (PTPN11), with aberrant CD7 expression in four of six and 10/12 patients respectively. Thus, JMML shows an immunophenotypic profile similar to myelodysplastic syndromes, and a different monocyte subset distribution when compared with chronic MML. MFC proved to be an important diagnostic tool that can help in differential diagnosis with other clonal diseases with monocytosis.
Collapse
Affiliation(s)
- Anita Frisanco Oliveira
- Barretos Children's Cancer Hospital, Barretos, São Paulo, Brazil.,Brazilian Co-operative Study Group for Paediatric Myelodysplastic Syndrome (GCB-SMD-PED) - Morphology and Flow Cytometry Committee, Barretos, São Paulo, Brazil
| | - Aline Tansini
- Barretos Children's Cancer Hospital, Barretos, São Paulo, Brazil.,GCB-SMD-PED - Flow cytometry Committee, Barretos, São Paulo, Brazil
| | - Thais Regina Toledo
- Barretos Children's Cancer Hospital, Barretos, São Paulo, Brazil.,GCB-SMD-PED - Flow cytometry Committee, Barretos, São Paulo, Brazil
| | - Rafael Balceiro
- Barretos Children's Cancer Hospital, Barretos, São Paulo, Brazil.,Brazilian Co-operative Study Group for Paediatric Myelodysplastic Syndrome (GCB-SMD-PED) - Morphology and Flow Cytometry Committee, Barretos, São Paulo, Brazil
| | - Daniel Onofre Vidal
- GCB-SMD-PED - Molecular Biology and Genetic Committee, Barretos, São Paulo, Brazil
| | - Maria Lucia de Martino Lee
- Barretos Children's Cancer Hospital, Barretos, São Paulo, Brazil.,GCB-SMD-PED - Morphology and Myeloproliferative Diseases Committee, Barretos, São Paulo, Brazil
| | - Irene Lorand-Metze
- Brazilian Co-operative Study Group for Paediatric Myelodysplastic Syndrome (GCB-SMD-PED) - Morphology and Flow Cytometry Committee, Barretos, São Paulo, Brazil.,GCB-SMD-PED - Chairman, Barretos, São Paulo, Brazil
| | - Luiz Fernando Lopes
- Barretos Children's Cancer Hospital, Barretos, São Paulo, Brazil.,GCB-SMD-PED - Chairman, Barretos, São Paulo, Brazil
| |
Collapse
|
15
|
Marcu A, Colita A, Radu LE, Jercan CG, Bica AM, Asan M, Coriu D, Tanase AD, Diaconu CC, Mambet C, Botezatu A, Pasca S, Teodorescu P, Anton G, Gurban P, Colita A. Single-Center Experience With Epigenetic Treatment for Juvenile Myelomonocytic Leukemia. Front Oncol 2020; 10:484. [PMID: 32328464 PMCID: PMC7161089 DOI: 10.3389/fonc.2020.00484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Juvenile myelomonocytic leukemia (JMML) is a rare myelodysplastic/myeloproliferative neoplasm diagnosed in young children, characterized by somatic or germline mutations that lead to hyperactive RAS signaling. The only curative option is hematopoietic stem cell transplantation (HSCT). Recent data showing that aberrant DNA methylation plays a significant role in pathogenesis and correlates with clinical risk suggest a possible benefit of hypomethylating agents (HMA) in JMML treatment. Aim: The aim is to report the results of HMA-based therapy with 5-azacytidine (AZA) in three JMML patients treated in a single center, non-participating in EWOG-MDS study. Methods: The diagnosis and treatment response were evaluated according to international consensus criteria. AZA 75 mg/m2 intravenous (i.v.) was administered once daily on days 1-7 of each 28-day cycle. All patients were monitored for hematologic response, spleen size, and evolution of extramedullary disease. Targeted next generation sequencing (NGS) were performed after the 3rd AZA cycle and before SCT to evaluate the molecular alterations and genetic response. Results: Three patients diagnosed with JMML were treated with AZA (off-label indication) in Pediatric Department of Fundeni Clinical Institute, Bucharest, Romania between 2017 and 2019. There were two females and one male with median age 11 months, range 2-16 months. The cytogenetic analysis showed normal karyotype in all patients. Molecular analysis confirmed KRAS G13D mutation in two patients and NRAS G12D mutation in one patient. The clinical evaluation showed important splenomegaly and hepatomegaly in all 3 pts. One patient received AZA for early relapse after haploidentical HSCT and the other two patients received upfront AZA, as bridging therapy before HSCT. After HMA therapy, 2/3 patients achieved clinical partial response (cPR), 1/3 had clinical stable disease (cSD) and all had genetic stable disease (gSD) after 3 cycles and were able to receive the planned HSTC. One patient achieved clinical and genetic complete response before HSCT. During 22 cycles of AZA there were only four adverse events but only one determined dose reduction and treatment delay. Conclusion: Our data show that AZA monotherapy is safe and effective in controlling disease both in upfront and relapsed patients in order to proceed to HSCT.
Collapse
Affiliation(s)
- Andra Marcu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Andrei Colita
- Department of Stem Cell Transplantation, Coltea Hospital, Bucharest, Romania
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Letitia Elena Radu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Cristina Georgiana Jercan
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ana Maria Bica
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Minodora Asan
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Daniel Coriu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Alina Daniela Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Hematology, Titu Maiorescu University of Medicine, Bucharest, Romania
| | - Carmen C. Diaconu
- Cellular and Molecular Pathology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Cristina Mambet
- Cellular and Molecular Pathology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Anca Botezatu
- Molecular Virology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Sergiu Pasca
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania
| | - Gabriela Anton
- Molecular Virology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Petruta Gurban
- Personal Genetics-Medical Genetics Center, Bucharest, Romania
| | - Anca Colita
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
16
|
Marcu A, Colita A, Radu LE, Jercan CG, Bica AM, Asan M, Coriu D, Tanase AD, Diaconu CC, Mambet C, Botezatu A, Pasca S, Teodorescu P, Anton G, Gurban P, Colita A. Single-Center Experience With Epigenetic Treatment for Juvenile Myelomonocytic Leukemia. Front Oncol 2020; 10:484. [PMID: 32328464 PMCID: PMC7161089 DOI: 10.3389/fonc.2020.00484,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/17/2020] [Indexed: 01/27/2025] Open
Abstract
Background: Juvenile myelomonocytic leukemia (JMML) is a rare myelodysplastic/myeloproliferative neoplasm diagnosed in young children, characterized by somatic or germline mutations that lead to hyperactive RAS signaling. The only curative option is hematopoietic stem cell transplantation (HSCT). Recent data showing that aberrant DNA methylation plays a significant role in pathogenesis and correlates with clinical risk suggest a possible benefit of hypomethylating agents (HMA) in JMML treatment. Aim: The aim is to report the results of HMA-based therapy with 5-azacytidine (AZA) in three JMML patients treated in a single center, non-participating in EWOG-MDS study. Methods: The diagnosis and treatment response were evaluated according to international consensus criteria. AZA 75 mg/m2 intravenous (i.v.) was administered once daily on days 1-7 of each 28-day cycle. All patients were monitored for hematologic response, spleen size, and evolution of extramedullary disease. Targeted next generation sequencing (NGS) were performed after the 3rd AZA cycle and before SCT to evaluate the molecular alterations and genetic response. Results: Three patients diagnosed with JMML were treated with AZA (off-label indication) in Pediatric Department of Fundeni Clinical Institute, Bucharest, Romania between 2017 and 2019. There were two females and one male with median age 11 months, range 2-16 months. The cytogenetic analysis showed normal karyotype in all patients. Molecular analysis confirmed KRAS G13D mutation in two patients and NRAS G12D mutation in one patient. The clinical evaluation showed important splenomegaly and hepatomegaly in all 3 pts. One patient received AZA for early relapse after haploidentical HSCT and the other two patients received upfront AZA, as bridging therapy before HSCT. After HMA therapy, 2/3 patients achieved clinical partial response (cPR), 1/3 had clinical stable disease (cSD) and all had genetic stable disease (gSD) after 3 cycles and were able to receive the planned HSTC. One patient achieved clinical and genetic complete response before HSCT. During 22 cycles of AZA there were only four adverse events but only one determined dose reduction and treatment delay. Conclusion: Our data show that AZA monotherapy is safe and effective in controlling disease both in upfront and relapsed patients in order to proceed to HSCT.
Collapse
Affiliation(s)
- Andra Marcu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Andrei Colita
- Department of Stem Cell Transplantation, Coltea Hospital, Bucharest, Romania
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Letitia Elena Radu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Cristina Georgiana Jercan
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ana Maria Bica
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Minodora Asan
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Daniel Coriu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Alina Daniela Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Hematology, Titu Maiorescu University of Medicine, Bucharest, Romania
| | - Carmen C. Diaconu
- Cellular and Molecular Pathology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Cristina Mambet
- Cellular and Molecular Pathology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Anca Botezatu
- Molecular Virology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Sergiu Pasca
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania
| | - Gabriela Anton
- Molecular Virology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Petruta Gurban
- Personal Genetics-Medical Genetics Center, Bucharest, Romania
| | - Anca Colita
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
17
|
Meynier S, Rieux-Laucat F. FAS and RAS related Apoptosis defects: From autoimmunity to leukemia. Immunol Rev 2019; 287:50-61. [PMID: 30565243 DOI: 10.1111/imr.12720] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/09/2018] [Indexed: 02/07/2023]
Abstract
The human adaptive immune system recognizes almost all the pathogens that we encounter and all the tumor antigens that may arise during our lifetime. Primary immunodeficiencies affecting lymphocyte development or function therefore lead to severe infections and tumor susceptibility. Furthermore, the fact that autoimmunity is a frequent feature of primary immunodeficiencies reveals a third function of the adaptive immune system: its self-regulation. Indeed, the generation of a broad repertoire of antigen receptors (via a unique strategy of random somatic rearrangements of gene segments in T cell and B cell receptor loci) inevitably creates receptors with specificity for self-antigens and thus leads to the presence of autoreactive lymphocytes. There are many different mechanisms for controlling the emergence or action of autoreactive lymphocytes, including clonal deletion in the primary lymphoid organs, receptor editing, anergy, suppression of effector lymphocytes by regulatory lymphocytes, and programmed cell death. Here, we review the genetic defects affecting lymphocyte apoptosis and that are associated with lymphoproliferation and autoimmunity, together with the role of somatic mutations and their potential involvement in more common autoimmune diseases.
Collapse
Affiliation(s)
- Sonia Meynier
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Frédéric Rieux-Laucat
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| |
Collapse
|
18
|
Zeng MH, He XL, Yang MH, Zheng MC, Wan WQ, Zou RY, Chen KK. [Clinical and genetic analyses of juvenile myelomonocytic leukemia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:365-369. [PMID: 31014430 PMCID: PMC7389224 DOI: 10.7499/j.issn.1008-8830.2019.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/05/2019] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To study the clinical and genetic features of juvenile myelomonocytic leukemia (JMML) and the association between genotype and prognosis. Methods The clinical data of 15 children who were diagnosed with JMML were collected. Next-generation sequencing was used to detect common gene mutations of JMML. RESULTS The male/female ratio was 6.5:1, and the age of onset was 19 months (range 2-67 months). Of the 15 children, 11 (73%) experienced disease onset before the age of 4 years, with abdominal distension and pyrexia as initial symptoms. All children had hepatosplenomegaly and superficial lymphadenectasis, with a number of peripheral blood mononuclear cells of >1.0×109/L and a percentage of juvenile cells of 1%-7% in peripheral blood smear. The percentage of bone marrow blasts + juvenile cells was <20%, and the percentage of monoblasts + promonocytes was 1%-10%. Of the 15 children, 10 (67%) had a higher level of hemoglobin F than the normal level at the corresponding age, with the highest level of 62.5%. All 15 children had the absence of Philadelphia chromosome, and one child had chromosome 7 deletion. All 15 children had a negative result of BCR/ABL fusion gene detection. PTPN11 gene mutation was found in 5 children (33%), NF1 mutation in 4 children (27%), CBL mutation in 3 children (20%), and RAS mutation in 3 children (20%). No children received regular chemotherapy, and one child underwent hematopoietic stem cell transplantation. The median follow-up time of 15 children was 18 months (range 1-48 months). Among the 15 children, 8 died (among whom 4 had PTPN11 gene mutation, 3 had NF1 mutation, and 1 had RAS mutation) and 7 survived. The children with PTPN11 mutation had the worst prognosis and the highest mortality rate, and those with CBL or NRAS mutation had a relatively good prognosis. The level of hemoglobin F was negatively correlated with survival time (rs=-7.21, P=0.002). CONCLUSIONS In children with JMML, the type of gene mutation is associated with prognosis. The children with PTPN11 mutation often have a poor prognosis, and those with CBL or NRAS mutation have a relatively good prognosis.
Collapse
Affiliation(s)
- Min-Hui Zeng
- Department of Hematology and Oncology, Children's Medical Center, Hunan Provincial People's Hospital/First Hospital of Hunan Normal University, Changsha 410005, China.
| | | | | | | | | | | | | |
Collapse
|
19
|
Guignard N, Roujeau T, Saumet L, Gascou G, Mondain M, Akkari M. Sphenoidal sinogenic extradural empyema associated with juvenile myelomonocytic leukemia. Int J Pediatr Otorhinolaryngol 2018; 115:45-48. [PMID: 30368391 DOI: 10.1016/j.ijporl.2018.09.016] [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: 07/15/2018] [Revised: 09/16/2018] [Accepted: 09/16/2018] [Indexed: 10/28/2022]
Abstract
Intracranial empyema is a rare but serious complication of sinusitis in children. Myelodysplastic/myeloproliferative syndromes (MMS), including juvenile myelomonocytic leukemia (JMML), can lead to immunosuppression, thus favouring infections. We report the case of a sphenoid sinogenic retro-clival extradural empyema in a 14-year-old female patient associated with JMML. Treatment consisted in an endonasal transphenoidal drainage of the empyema associated with intravenous antibiotherapy. The patient was thereafter enrolled in chemotherapeutic treatment with Azacitidine. The disease progressed to blast phase, indicating bone marrow graft. This is the first reported case of an endocranial complication of bacterial sinusitis associated with MMS in a child.
Collapse
Affiliation(s)
- N Guignard
- Department of ENT and Head and Neck Surgery, University Hospital Gui de Chauliac, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, University of Montpellier, France
| | - T Roujeau
- Department of Pediatric Neurosurgery, University Hospital Gui de Chauliac, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, University of Montpellier, France
| | - L Saumet
- Department of Pediatric Oncology, University Hospital Arnaud de Villeneuve, 371 Avenue du Doyen Gaston Giraud, 34295, Montpellier, University of Montpellier, France
| | - G Gascou
- Department of Neuroradiology, University Hospital Gui de Chauliac, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, University of Montpellier, France
| | - M Mondain
- Department of ENT and Head and Neck Surgery, University Hospital Gui de Chauliac, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, University of Montpellier, France
| | - M Akkari
- Department of ENT and Head and Neck Surgery, University Hospital Gui de Chauliac, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, University of Montpellier, France.
| |
Collapse
|
20
|
Cai YL, Zhang JL, Zhu XF. [Advances in the treatment of juvenile myelomonocytic leukemia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:958-963. [PMID: 30477631 PMCID: PMC7389026 DOI: 10.7499/j.issn.1008-8830.2018.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 09/11/2018] [Indexed: 06/09/2023]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare chronic myeloid leukemia in children and has the features of both myelodysplastic syndrome and myeloproliferative neoplasm. It is highly malignant and has a poor treatment outcome. Children with JMML have a poor response to conventional chemotherapy. At present, hematopoietic stem cell transplantation is the only possible cure for this disease. In recent years, significant progress has been made in targeted therapy for mutant genes in the Ras signaling pathway and demethylation treatment of aberrant methylation of polygenic CpG islands. This article reviews the treatment and efficacy evaluation of JMML.
Collapse
Affiliation(s)
- Yu-Li Cai
- Department of Pediatrics, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China.
| | | | | |
Collapse
|