1
|
He J, Munir F, Catueno S, Connors JS, Gibson A, Robusto L, McCall D, Nunez C, Roth M, Tewari P, Garces S, Cuglievan B, Garcia MB. Biological Markers of High-Risk Childhood Acute Lymphoblastic Leukemia. Cancers (Basel) 2024; 16:858. [PMID: 38473221 PMCID: PMC10930495 DOI: 10.3390/cancers16050858] [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: 01/21/2024] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 03/14/2024] Open
Abstract
Childhood acute lymphoblastic leukemia (ALL) has witnessed substantial improvements in prognosis; however, a subset of patients classified as high-risk continues to face higher rates of relapse and increased mortality. While the National Cancer Institute (NCI) criteria have traditionally guided risk stratification based on initial clinical information, recent advances highlight the pivotal role of biological markers in shaping the prognosis of childhood ALL. This review delves into the emerging understanding of high-risk childhood ALL, focusing on molecular, cytogenetic, and immunophenotypic markers. These markers not only contribute to unraveling the underlying mechanisms of the disease, but also shed light on specific clinical patterns that dictate prognosis. The paradigm shift in treatment strategies, exemplified by the success of tyrosine kinase inhibitors in Philadelphia chromosome-positive leukemia, underscores the importance of recognizing and targeting precise risk factors. Through a comprehensive exploration of high-risk childhood ALL characteristics, this review aims to enhance our comprehension of the disease, offering insights into its molecular landscape and clinical intricacies in the hope of contributing to future targeted and tailored therapies.
Collapse
Affiliation(s)
- Jiasen He
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Faryal Munir
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Samanta Catueno
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Jeremy S. Connors
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Amber Gibson
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Lindsay Robusto
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - David McCall
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Cesar Nunez
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Michael Roth
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Priti Tewari
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Sofia Garces
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Miriam B. Garcia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| |
Collapse
|
2
|
A novel heptasomy 21 associated with complete loss of heterozygosity and loss of function RUNX1 mutation in acute myeloid leukemia. Cancer Genet 2022; 266-267:69-73. [DOI: 10.1016/j.cancergen.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 06/07/2022] [Accepted: 07/01/2022] [Indexed: 11/17/2022]
|
3
|
Gonzales F, Barthélémy A, Peyrouze P, Fenwarth L, Preudhomme C, Duployez N, Cheok MH. Targeting RUNX1 in acute myeloid leukemia: preclinical innovations and therapeutic implications. Expert Opin Ther Targets 2021; 25:299-309. [PMID: 33906574 DOI: 10.1080/14728222.2021.1915991] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Introduction: RUNX1 is an essential transcription factor for normal and malignant hematopoiesis. RUNX1 forms a heterodimeric complex with CBFB. Germline mutations and somatic alterations (i.e. translocations, mutations and abnormal expression) are frequently associated with acute myeloid leukemia (AML) with RUNX1 mutations conferring unfavorable prognosis. Therefore, RUNX1 constitutes a potential innovative and interesting therapeutic target. In this review, we discuss recent therapeutic advances of RUNX1 targeting in AML.Areas covered: Firstly, we cover the clinical basis for RUNX1 targeting. We have subdivided recent therapeutic approaches either by common biochemical pathways or by similar pharmacological targets. Genome editing of RUNX1 induces anti-leukemic effects; however, off-target events prohibit clinical use. Several molecules inhibit the interaction between RUNX1/CBFB and control AML development and progression. BET protein antagonists target RUNX1 (i.e. specific BET inhibitors, BRD4 shRNRA, proteolysis targeting chimeras (PROTAC) or expression-mimickers). All these molecules improve survival in mutant RUNX1 AML preclinical models.Expert opinion: Some of these novel molecules have shown encouraging anti-leukemic potency at the preclinical stage. A better understanding of RUNX1 function in AML development and progression and its key downstream pathways, may result in more precise and more efficient RUNX1 targeting therapies.
Collapse
Affiliation(s)
- Fanny Gonzales
- Factors of Leukemic cell Persistence, Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, Canther, Lille, France.,Pediatric Hematology Department, University Hospital of Lille, Lille, France
| | - Adeline Barthélémy
- Factors of Leukemic cell Persistence, Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, Canther, Lille, France
| | - Pauline Peyrouze
- Factors of Leukemic cell Persistence, Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, Canther, Lille, France
| | - Laurène Fenwarth
- Factors of Leukemic cell Persistence, Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, Canther, Lille, France.,Laboratory of Hematology, CHU Lille, Lille, France
| | - Claude Preudhomme
- Factors of Leukemic cell Persistence, Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, Canther, Lille, France.,Laboratory of Hematology, CHU Lille, Lille, France
| | - Nicolas Duployez
- Factors of Leukemic cell Persistence, Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, Canther, Lille, France.,Laboratory of Hematology, CHU Lille, Lille, France
| | - Meyling H Cheok
- Factors of Leukemic cell Persistence, Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, Canther, Lille, France
| |
Collapse
|
4
|
Jain H, Shetty D, Roy Moulik N, Narula G, Subramanian PG, Banavali S. A novel case of intrachromosomal amplification and insertion of RUNX1 on derivative chromosome 2 in pediatric AML. Cancer Genet 2021; 254-255:65-69. [PMID: 33647813 DOI: 10.1016/j.cancergen.2021.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/15/2021] [Accepted: 02/08/2021] [Indexed: 11/18/2022]
Abstract
Intrachromosomal amplification of RUNX1 gene on chromosome 21 (iAMP21) is a rare occurrence in acute myeloid leukemia (AML). Herein, we describe a case of AML with amplification of RUNX1 and its insertion on chromosome 2 detected by conventional karyotyping and confirmed by metaphase FISH. A six-year-old female was diagnosed as acute myeloid leukemia with monocytic differentiation. The patient's bone marrow revealed 74% blasts which were MPO negative. Conventional karyotyping revealed a complex karyotype, with rearrangements in chromosomes 1, 2, 7, 8 and hsr(21). FISH on interphase cells with LSI RUNX1-RUNX1T1 dual colour dual fusion translocation probe showed 6-7 copies of RUNX1 signal. Metaphase FISH with LSI RUNX1-RUNX1T1 probe confirmed amplification of RUNX1 and insertion of amplified RUNX1 sequences on long arm of chromosome 2. Induction chemotherapy was initiated, however, the patient died within one month of diagnosis suggesting poor outcome associated with this novel finding. Insertion of amplified RUNX1 on another chromosome has not yet been reported so far.
Collapse
Affiliation(s)
- Hemani Jain
- Cancer Cytogenetics Department, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Sector-22, Kharghar, Navi Mumbai 410210, India.
| | - Dhanlaxmi Shetty
- Cancer Cytogenetics Department, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Sector-22, Kharghar, Navi Mumbai 410210, India.
| | - Nirmalya Roy Moulik
- Department of Pediatric Oncology, Tata Memorial Hospital, Dr. E. Borges Road, Parel. Mumbai 400012, India; Cancer Cytogenetics Department, Room No. 6, CCE building, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Sector-22, Kharghar, Navi Mumbai 410210 India.
| | - Gaurav Narula
- Department of Pediatric Oncology, Tata Memorial Hospital, Dr. E. Borges Road, Parel. Mumbai 400012, India; Cancer Cytogenetics Department, Room No. 6, CCE building, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Sector-22, Kharghar, Navi Mumbai 410210 India.
| | - P G Subramanian
- Hematopathology Department, CCE building, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Sector-22, Kharghar, Navi Mumbai 410210, India; Cancer Cytogenetics Department, Room No. 6, CCE building, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Sector-22, Kharghar, Navi Mumbai 410210 India.
| | - Sripad Banavali
- Department of Pediatric Oncology, Tata Memorial Hospital, Dr. E. Borges Road, Parel. Mumbai 400012, India; Cancer Cytogenetics Department, Room No. 6, CCE building, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Sector-22, Kharghar, Navi Mumbai 410210 India.
| |
Collapse
|
5
|
iAMP21 in acute myeloid leukemia is associated with complex karyotype, TP53 mutation and dismal outcome. Mod Pathol 2020; 33:1389-1397. [PMID: 32034282 DOI: 10.1038/s41379-020-0494-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/12/2022]
Abstract
Acute myeloid leukemia (AML) with intrachromosomal amplification of chromosome 21 (iAMP21) is rare and has not been well characterized. We report 13 patients, 7 men and 6 women, with a median age of 65 years. Eleven patients presented with AML with myelodysplasia-related changes, and two patients had therapy-related AML. Cytopenias were detected in all patients (11 pancytopenia and two bi-lineage cytopenia). Myelodysplastic changes were observed in all 11 patients with adequate cells to evaluate. Myelofibrosis was present in ten patients. All patients had a complex karyotype, including abnormalities of chromosomes 5, 7, 17, and hsr(21)(q22), and ten patients showed TP53 deletion and/or mutation. Eleven patients received AML-based chemotherapy, one of whom also received hematopoietic stem cell transplant. By the end of the last follow-up, eight patients died with median survival of 3.2 months, four patients were alive with persistent AML, and one was in complete remission. The median overall survival was 6 months for all patients. We conclude that AML with iAMP21 is often associated with cytopenias, myelodysplasia, a complex karyotype, TP53 mutation/deletion, and a poor prognosis despite current therapies.
Collapse
|
6
|
Laying the foundation for genomically-based risk assessment in chronic myeloid leukemia. Leukemia 2019; 33:1835-1850. [PMID: 31209280 DOI: 10.1038/s41375-019-0512-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 12/16/2022]
Abstract
Outcomes for patients with chronic myeloid leukemia (CML) have substantially improved due to advances in drug development and rational treatment intervention strategies. Despite these significant advances there are still unanswered questions on patient management regarding how to more reliably predict treatment failure at the time of diagnosis and how to select frontline tyrosine kinase inhibitor (TKI) therapy for optimal outcome. The BCR-ABL1 transcript level at diagnosis has no established prognostic impact and cannot guide frontline TKI selection. BCR-ABL1 mutations are detected in ~50% of TKI resistant patients but are rarely responsible for primary resistance. Other resistance mechanisms are largely uncharacterized and there are no other routine molecular testing strategies to facilitate the evaluation and further stratification of TKI resistance. Advances in next-generation sequencing technology has aided the management of a growing number of other malignancies, enabling the incorporation of somatic mutation profiles in diagnosis, classification, and prognostication. A largely unexplored area in CML research is whether expanded genomic analysis at diagnosis, resistance, and disease transformation can enhance patient management decisions, as has occurred for other cancers. The aim of this article is to review publications that reported mutated cancer-associated genes in CML patients at various disease phases. We discuss the frequency and type of such variants at initial diagnosis and at the time of treatment failure and transformation. Current limitations in the evaluation of mutants and recommendations for future reporting are outlined. The collective evaluation of mutational studies over more than a decade suggests a limited set of cancer-associated genes are indeed recurrently mutated in CML and some at a relatively high frequency. Genomic studies have the potential to lay the foundation for improved diagnostic risk classification according to clinical and genomic risk, and to enable more precise early identification of TKI resistance.
Collapse
|
7
|
Wu F, Song T, Yao Y, Song Y. Thermodynamic investigation of DNA-binding affinity of wild-type and mutant transcription factor RUNX1. PLoS One 2019; 14:e0216203. [PMID: 31048839 PMCID: PMC6497270 DOI: 10.1371/journal.pone.0216203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/16/2019] [Indexed: 11/29/2022] Open
Abstract
Transcription factor RUNX1 and its binding partner CBFβ play a critical role in gene regulation for hematopoiesis. Mutations of RUNX1 cause ~10% of acute myeloid leukemia (AML) with a particularly poor prognosis. The current paradigm for the leukemogenesis is that insufficient activity of wild-type (WT) RUNX1 impairs hematopoietic differentiation. The majority of mutant RUNX1 proteins lose the DNA-binding affinity and inhibit WT RUNX1 by depletion of CBFβ. Here, isothermal titration calorimetry (ITC) was used to quantitatively study the interactions of WT and three clinical mutant RUNX1, CBFβ and DNA. Our data show that the binding of RUNX1 to DNA is enthalpy-driven, and the affinity decreases in the order of WT > S114L > R139Q >> K83E, which support previous observations and conclusion. To find potentially beneficial RUNX1 mutations that could increase the overall RUNX1 activity, K83R and H179K mutations of RUNX1 were designed, using structure-based computational modeling, and found to possess significantly higher DNA-binding affinities than does WT RUNX1. K83R and H179K mutant RUNX1 could therefore be protein-based RUNX1 activators.
Collapse
Affiliation(s)
- Fangrui Wu
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Tidie Song
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Yuan Yao
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Yongcheng Song
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
8
|
Assessing copy number abnormalities and copy-neutral loss-of-heterozygosity across the genome as best practice in diagnostic evaluation of acute myeloid leukemia: An evidence-based review from the cancer genomics consortium (CGC) myeloid neoplasms working group. Cancer Genet 2018; 228-229:218-235. [DOI: 10.1016/j.cancergen.2018.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 12/19/2022]
|
9
|
Richter L, Wang Y, Hyde RK. Targeting binding partners of the CBFβ-SMMHC fusion protein for the treatment of inversion 16 acute myeloid leukemia. Oncotarget 2018; 7:66255-66266. [PMID: 27542261 PMCID: PMC5323231 DOI: 10.18632/oncotarget.11357] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/09/2016] [Indexed: 11/25/2022] Open
Abstract
Inversion of chromosome 16 (inv(16)) generates the CBFβ-SMMHC fusion protein and is found in nearly all patients with acute myeloid leukemia subtype M4 with Eosinophilia (M4Eo). Expression of CBFβ-SMMHC is causative for leukemia development, but the molecular mechanisms underlying its activity are unclear. Recently, there have been important advances in defining the role of CBFβ-SMMHC and its binding partners, the transcription factor RUNX1 and the histone deacetylase HDAC8. Importantly, initial trials demonstrate that small molecules targeting these binding partners are effective against CBFβ-SMMHC induced leukemia. This review will discuss recent advances in defining the mechanism of CBFβ-SMMHC activity, as well as efforts to develop new therapies for inv(16) AML.
Collapse
Affiliation(s)
- Lisa Richter
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yiqian Wang
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - R Katherine Hyde
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
10
|
Inokuchi K, Nakayama K, Tauchi T, Takaku T, Yokose N, Yamaguchi H, Kumagai T, Komatsu N, Ohyashiki K. Therapeutic effects of tyrosine kinase inhibitors and subtypes of BCR-ABL1 transcripts in Japanese chronic myeloid leukemia patients with three-way chromosomal translocations. Leuk Res 2018; 65:74-79. [PMID: 29310021 DOI: 10.1016/j.leukres.2018.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/30/2017] [Accepted: 01/02/2018] [Indexed: 10/18/2022]
Abstract
We analyzed the clinical responses to thyrosine kinase inhibitors (TKIs) and the molecular and cytogenetic characteristics of 18 chronic myeloid leukemia (CML) patients with 3-way chromosomal translocations. The patients were 14 men and 4 women, aged 23-75 years (median 57 years). The Sokal risk was low in 12 patients, intermediate in 4 patients, and high in 2 patients. Newly identified translocation breakpoints were seen in 7 of the 18 patients. Three patients had the same breakpoints of t(9;22;11)(q34;q11.2;q23). The best responses to TKIs were partial cytogenic response (PCyR) in 2 patients, complete cytogenic response (CCyR) in 3 patients, molecular response (MR) 3.0 in 7 patients, MR 4.0 in 3 patients, and MR 4.5 or higher in 3 patients. A total of 66.7% of patients did not achieve MR 4.0 or higher. In 3 patients in whom TKIs resulted in MR 4.5 or higher for more than 2 years, TKI treatment was discontinued. However, all of them exhibited a loss of MR3.0, at 2, 6, and 20 months after the discontinuation of treatment, respectively, and TKI treatment needed to be restarted. According to Kaplan-Meier survival curve analysis, the overall survival (OS) was 100 months in 56% of the patients. The 60-months cumulative incidences of CCyR, MR3.0, MR4.0 and MR4.5 were 88.9%, 72.2%, 33.3%, and 16.7%, respectively. In the 11 analyzable patients, the BCR-ABL1 mRNA subtype was e14a2 type in 4 patients and e13a2 type in 7 patients.
Collapse
Affiliation(s)
- Koiti Inokuchi
- Department of Hematology, Nippon Medical School, Tokyo, Japan.
| | | | - Tetsuzo Tauchi
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| | - Tomoiku Takaku
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Norio Yokose
- Department of Hematology, Chiba Hokusoh Hospital, Nippon Medical School, Japan
| | | | - Takashi Kumagai
- Department of Hematology, Ome Municipal General Hospital, Tokyo, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazuma Ohyashiki
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
11
|
Wang L, Wu XY, Jin RM, Zhang BY, Qiu YN. Trisomy 21 with t(5; 11) chromosomal translocation as new unfavorable cytogenetic abnormalities in pediatric acute myeloid leukemia type M2: One case report of nine-year follow-up and literature review. Curr Med Sci 2017; 37:807-810. [PMID: 29058300 DOI: 10.1007/s11596-017-1809-6] [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: 08/01/2017] [Revised: 09/15/2017] [Indexed: 11/29/2022]
Abstract
We report one case of pediatric acute myeloid leukemia type 2 (AML-M2) who presented with karyotypic aberration of trisomy 21 with the t(5;11) chromosomal translocation. The patient achieved complete remission after two cycles of chemotherapy of daunorubicin, cytarabine and etoposide. Then, follow-up cytogenetic analysis from bone marrow cell cultures demonstrated a normal karyotype of 46, XY. After 9 years, the patient relapsed and the karyotypic abnormalities of trisomy 21 with t(5;11) reappeared. It was concluded that trisomy 21 with t(5; 11) is a new unfavorable cytogenetic aberration in AML-M2.
Collapse
Affiliation(s)
- Lin Wang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao-Yan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Run-Ming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bing-Yu Zhang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yi-Ning Qiu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
12
|
Sabatino R, Aquino G, Pinto A, Piris MA, Marra L, Napolitano M, De Chiara A, Franco R. B-cell lymphoblastic lymphoma presenting as solitary temporal mass with amplification of AML1/RUNX1: case report. Hematol Oncol 2017; 35:380-384. [PMID: 28933515 DOI: 10.1002/hon.2269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 09/18/2015] [Accepted: 09/27/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Rocco Sabatino
- Pathology Unit, Istituto Nazionale Tumori " Fondazione 'G. Pascale", Napoli, Italy
| | - Gabriella Aquino
- Pathology Unit, Istituto Nazionale Tumori " Fondazione 'G. Pascale", Napoli, Italy
| | - Antonio Pinto
- Haematology- Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori Fondazione " G. Pascale", Napoli, Italy
| | - Miguel Angel Piris
- Pathology Unit, Hospital Universitario Marques de Valdecilla IDIVAL, Santander, Spain
| | - Laura Marra
- Pathology Unit, Istituto Nazionale Tumori " Fondazione 'G. Pascale", Napoli, Italy
| | - Maria Napolitano
- Immunology- Oncology Unit, Istituto Nazionale Tumori Fondazione " G. Pascale", Napoli, Italy
| | | | - Renato Franco
- Pathology Unit, Istituto Nazionale Tumori " Fondazione 'G. Pascale", Napoli, Italy
| |
Collapse
|
13
|
Clinical Relevance of RUNX1 and CBFB Alterations in Acute Myeloid Leukemia and Other Hematological Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 962:175-199. [PMID: 28299658 DOI: 10.1007/978-981-10-3233-2_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The translocation t(8;21), leading to a fusion between the RUNX1 gene and the RUNX1T1 locus, was the first chromosomal translocation identified in cancer. Since the first description of this balanced rearrangement in a patient with acute myeloid leukemia (AML) in 1973, RUNX1 translocations and point mutations have been found in various myeloid and lymphoid neoplasms. In this chapter, we summarize the currently available data on the clinical relevance of core binding factor gene alterations in hematological disorders. In the first section, we discuss the prognostic implications of the core binding factor translocations RUNX1-RUNX1T1 and CBFB-MYH11 in AML patients. We provide an overview of the cooperating genetic events in patients with CBF-rearranged AML and their clinical implications, and review current treatment approaches for CBF AML and the utility of minimal residual disease monitoring. In the next sections, we summarize the available data on rare RUNX1 rearrangements in various hematologic neoplasms and the role of RUNX1 translocations in therapy-related myeloid neoplasia. The final three sections of the chapter cover the spectrum and clinical significance of RUNX1 point mutations in AML and myelodysplastic syndromes, in familial platelet disorder with associated myeloid malignancy, and in acute lymphoblastic leukemia.
Collapse
|
14
|
Duployez N, Lejeune S, Renneville A, Preudhomme C. Myelodysplastic syndromes and acute leukemia with genetic predispositions: a new challenge for hematologists. Expert Rev Hematol 2016; 9:1189-1202. [DOI: 10.1080/17474086.2016.1257936] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Mutations of myelodysplastic syndromes (MDS): An update. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 769:47-62. [DOI: 10.1016/j.mrrev.2016.04.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/11/2016] [Indexed: 01/08/2023]
|
16
|
Burillo-Sanz S, Vargas MT, Morales-Camacho RM, Caballero-Velázquez T, Sánchez J, García-Lozano JR, Pérez de Soto I, Prats-Martín C, Bernal R, Pérez-Simón JA. RUNX1
amplification in AML with myelodysplasia-related changes and ring 21 chromosomes. Hematol Oncol 2016; 35:894-899. [DOI: 10.1002/hon.2287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 01/25/2016] [Accepted: 01/30/2016] [Indexed: 12/31/2022]
Affiliation(s)
- S Burillo-Sanz
- Servicio de Inmunología; Hospital Universitario Virgen del Rocío; Seville Spain
| | - MT Vargas
- Department of Hematology; Instituto de Biomedicina de Sevilla (IBIS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla; Seville Spain
| | - RM Morales-Camacho
- Department of Hematology; Instituto de Biomedicina de Sevilla (IBIS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla; Seville Spain
| | - T Caballero-Velázquez
- Department of Hematology; Instituto de Biomedicina de Sevilla (IBIS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla; Seville Spain
| | - J Sánchez
- Department of Genetics; Reproduction and Fetal Medicine; Seville Spain
- Centre of Biomedical Network Research on Rare Diseases (CIBERER); Seville Spain
| | - JR García-Lozano
- Servicio de Inmunología; Hospital Universitario Virgen del Rocío; Seville Spain
| | - I Pérez de Soto
- Department of Hematology; Instituto de Biomedicina de Sevilla (IBIS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla; Seville Spain
| | - C Prats-Martín
- Department of Hematology; Instituto de Biomedicina de Sevilla (IBIS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla; Seville Spain
| | - R Bernal
- Department of Hematology; Instituto de Biomedicina de Sevilla (IBIS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla; Seville Spain
| | - JA Pérez-Simón
- Department of Hematology; Instituto de Biomedicina de Sevilla (IBIS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla; Seville Spain
| |
Collapse
|
17
|
Hirade T, Abe M, Onishi C, Taketani T, Yamaguchi S, Fukuda S. Internal tandem duplication of FLT3 deregulates proliferation and differentiation and confers resistance to the FLT3 inhibitor AC220 by Up-regulating RUNX1 expression in hematopoietic cells. Int J Hematol 2015; 103:95-106. [PMID: 26590920 DOI: 10.1007/s12185-015-1908-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 11/06/2015] [Accepted: 11/10/2015] [Indexed: 10/22/2022]
Abstract
Internal tandem duplication in the FLT3 gene (FLT3/ITD), which is found in patients with acute myeloid leukemia (AML), causes resistance to FLT3 inhibitors. We found that RUNX1, a transcription factor that regulates normal hematopoiesis, is up-regulated in patients with FLT3/ITD(+) AML. While RUNX1 can function as a tumor suppressor, recent data have shown that RUNX1 is required for AML cell survival. In the present study, we investigated the functional role of RUNX1 in FLT3/ITD signaling. FLT3/ITD induced growth factor-independent proliferation and impaired G-CSF mediated myeloid differentiation in 32D hematopoietic cells, coincident with up-regulation of RUNX1 expression. Silencing of RUNX1 expression significantly decreased proliferation and secondary colony formation, and partially abrogated the impaired myeloid differentiation of FLT3/ITD(+) 32D cells. Although the number of FLT3/ITD(+) 32D cells declined after incubation with the FLT3/ITD inhibitor AC220, the cells became refractory to AC220, concomitant with up-regulation of RUNX1. Silencing of RUNX1 abrogated the emergence and proliferation of AC220-resistant FLT3/ITD(+) 32D cells in the presence of AC220. Our data indicate that FLT3/ITD deregulates cell proliferation and differentiation and confers resistance to AC220 by up-regulating RUNX1 expression. These findings suggest an oncogenic role for RUNX1 in FLT3/ITD(+) cells and that inhibition of RUNX1 function represents a potential therapeutic strategy in patients with refractory FLT3/ITD(+) AML.
Collapse
Affiliation(s)
- Tomohiro Hirade
- Department of Pediatrics, Shimane University School of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan.
| | - Mariko Abe
- Department of Pediatrics, Shimane University School of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Chie Onishi
- Department of Oncology/Hematology, Shimane University School of Medicine, Izumo, Japan
| | - Takeshi Taketani
- Department of Pediatrics, Shimane University School of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan.,Division of Blood Transfusion, Shimane University School of Medicine, Izumo, Japan
| | - Seiji Yamaguchi
- Department of Pediatrics, Shimane University School of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Seiji Fukuda
- Department of Pediatrics, Shimane University School of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan.
| |
Collapse
|
18
|
de Souza Carrocini GC, Venancio LPR, Bonini-Domingos CR. Screening of Transcription Factors Involved in Fetal Hemoglobin Regulation Using Phylogenetic Footprinting. Evol Bioinform Online 2015; 11:239-44. [PMID: 26543346 PMCID: PMC4624090 DOI: 10.4137/ebo.s15364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/23/2022] Open
Abstract
Fetal hemoglobin (Hb F) is an important genetic modulator of the beta-hemoglobinopathies. The regulation of Hb F levels is influenced by transcription factors. We used phylogenetic footprinting to screen transcription factors that have binding sites in HBG1 and HBG2 genes’ noncoding regions in order to know the genetic determinants of the Hb F expression. Our analysis showed 354 conserved motifs in the noncoding regions of HBG1 gene and 231 motifs in the HBG2 gene between the analyzed species. Of these motifs, 13 showed relation to Hb F regulation: cell division cycle-5 (CDC5), myelo-blastosis viral oncogene homolog (c-MYB), transcription factor CP2 (TFCP2), GATA binding protein 1 (GATA-1), GATA binding protein 2 (GATA-2), nuclear factor erythroid 2 (NF-E2), nuclear transcription factor Y (NF-Y), runt-related transcription factor 1 (RUNX-1), T-cell acute lymphocytic leukemia 1 (TAL-1), YY1 transcription factor (YY1), beta protein 1 (BP1), chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII), and paired box 1 (PAX-1). The last three motifs were conserved only in the noncoding regions of the HBG1 gene. The understanding of genetic elements involved in the maintenance of high Hb F levels may provide new efficient therapeutic strategies in the beta-hemoglobinopathies treatment, promoting reduction in clinical complications of these genetic disorders.
Collapse
Affiliation(s)
- Gisele Cristine de Souza Carrocini
- Laboratory of Hemoglobin and Genetics of Hematologic Diseases, Department of Biology, São Paulo State University - UNESP/IBILCE, São José do Rio Preto, São Paulo, Brazil
| | - Larissa Paola Rodrigues Venancio
- Laboratory of Hemoglobin and Genetics of Hematologic Diseases, Department of Biology, São Paulo State University - UNESP/IBILCE, São José do Rio Preto, São Paulo, Brazil
| | - Claudia Regina Bonini-Domingos
- Laboratory of Hemoglobin and Genetics of Hematologic Diseases, Department of Biology, São Paulo State University - UNESP/IBILCE, São José do Rio Preto, São Paulo, Brazil
| |
Collapse
|
19
|
Touaoussa A, Elhmadi K, El Youssi H, Moncef H, Hassani MA. [Acute myelomonocytic leukemia eosinophilic revealed by acute pancreatitis]. Pan Afr Med J 2015; 20:112. [PMID: 26090060 PMCID: PMC4458318 DOI: 10.11604/pamj.2015.20.112.6024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 01/20/2015] [Indexed: 12/14/2022] Open
Abstract
La leucémie aigüe myélomonocytaire à composante éosinophile (LAM4eo) est une hémopathie maligne rare, caractérisée par une prolifération blastique myéloïde avec présence d'une composante monocytaire et des éosinophiles anormaux. Elle est associée à l'inversion du chromosome 16, parfois à sa variante la translocation (16;16). Nous rapportons un cas de LAM4eo chez un patient de sexe masculin âgé de 51 ans, découverte au décours d'un bilan paraclinique pour pancréatite aiguë (PA) confirmée par la TDM. L'hémogramme a montré une hyperleucocytose à 123G/l faite de 60% de blastes, une monocytose à 5 G/L ainsi qu'une lignée éosinophile dystrophique. Le myélogramme a objectivé l'infiltration de la moelle par une population blastique estimée à 61% d'expression hétérogène à la Cytométrie de flux: Des blastes très immatures exprimant fortement les marqueurs CD117 et CD34; des blastes prédominants qui expriment les antigènes CD33, CD13, CD65 (myéloblastes). Et une partie des blastes, était positive pour le CD14, CD4, CD11c (monoblastes). Après une exploration étiologique approfondie n'ayant pas pu trouver un lien de cause à effet, l'association entre les deux pathologies a été considérée comme fortuite et la pancréatite a été rattachée à la prise de paroxétine. Le patient a été mis alors en condition, et a été traité par chimiothérapie avec bonne évolution clinique et biologique.
Collapse
Affiliation(s)
- Aziz Touaoussa
- Laboratoire Central d'Analyses Médicales, Laboratoire d'Hématologie, CHU Hassan II, Atlas Fès, Maroc
| | - Khalid Elhmadi
- Service de Laboratoire d'Hématologie, de l'Hopital My Ismail de Meknes, Maroc
| | - Hind El Youssi
- Laboratoire Central d'Analyses Médicales, Laboratoire d'Hématologie, CHU Hassan II, Atlas Fès, Maroc
| | | | - Moncef Amrani Hassani
- Laboratoire Central d'Analyses Médicales, Laboratoire d'Hématologie, CHU Hassan II, Atlas Fès, Maroc
| |
Collapse
|
20
|
Angelova S, Spassov B, Nikolova V, Christov I, Tzvetkov N, Simeonova M. Is amplification of c-MYC, MLL and RUNX1 genes in AML and MDS patients with trisomy 8, 11 and 21 a factor for a clonal evolution in the karyotype? CYTOL GENET+ 2015. [DOI: 10.3103/s0095452715030032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Blood Spotlight on iAMP21 acute lymphoblastic leukemia (ALL), a high-risk pediatric disease. Blood 2015; 125:1383-6. [PMID: 25608562 DOI: 10.1182/blood-2014-08-569228] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intrachromosomal amplification of chromosome 21 (iAMP21) defines a distinct cytogenetic subgroup of childhood B-cell precursor acute lymphoblastic leukemia. Breakage-fusion-bridge cycles followed by chromothripsis and other complex structural rearrangements of chromosome 21 underlie the mechanism giving rise to iAMP21. Patients with iAMP21 are older (median age 9 years), with a low white cell count. They have a high relapse rate when treated as standard risk. Recent studies have shown improved outcome on intensive therapy. Molecular targets for therapy are being sought.
Collapse
|
22
|
Solh M, Yohe S, Weisdorf D, Ustun C. Core-binding factor acute myeloid leukemia: Heterogeneity, monitoring, and therapy. Am J Hematol 2014; 89:1121-31. [PMID: 25088818 DOI: 10.1002/ajh.23821] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 07/25/2014] [Accepted: 07/30/2014] [Indexed: 11/06/2022]
Abstract
Core binding factor acute myelogenous leukemia (CBF AML) constitutes 15% of adult AML and carries an overall good prognosis. CBF AML encodes two recurrent cytogentic abnormalities referred to as t(8;21) and inv (16). The two CBF AML entities are usually grouped together but there is a considerable clinical, pathologic and molecular heterogeneity within this group of diseases. Recent and ongoing studies are addressing the molecular heterogeneity, minimal residual disease and targeted therapies to improve the outcome of CBF AML. In this article, we present a comprehensive review about CBF AML with emphasis on molecular heterogeneity and new therapeutic options.
Collapse
Affiliation(s)
- Melhem Solh
- Department of Medicine, Florida Center for Cellular Therapy; University of Central Florida; Orlando Florida
- Department of Medicine; University of Central Florida; Orlando Florida
| | - Sophia Yohe
- Department of Pathology and Laboratory Medicine; University of Minnesota; Minneapolis Minnesota
| | - Daniel Weisdorf
- Department of Medicine; Division of Hematology, Oncology and Transplantation, University of Minnesota; Minneapolis Minnesota
| | - Celalettin Ustun
- Department of Medicine; Division of Hematology, Oncology and Transplantation, University of Minnesota; Minneapolis Minnesota
| |
Collapse
|
23
|
Al-Kzayer LFY, Sakashita K, Al-Jadiry MF, Al-Hadad SA, Uyen LTN, Liu T, Matsuda K, Abdulkadhim JMH, Al-Shujairi TA, Matti ZIIK, Hasan JG, Al-Abdullah HMS, Inoshita T, Kamata M, Sughayer MA, Madanat FF, Koike K. Frequent coexistence of RAS mutations in RUNX1-mutated acute myeloid leukemia in Arab Asian children. Pediatr Blood Cancer 2014; 61:1980-5. [PMID: 25066867 DOI: 10.1002/pbc.25151] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 05/13/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND RUNX1 mutation plays an important role in adult leukemic transformation. However, its contribution to the development of childhood leukemia remains unclear. In the present study, we analyzed point mutations of RUNX1 gene in children and adolescents with acute myeloid leukemia (AML) from Iraq and Jordan. PROCEDURE Bone marrow and/or peripheral blood samples were collected from 178 patients of Arab Asian ethnicity (aged ≤17 years) newly diagnosed with AML: 145 samples from Iraq and 33 samples from Jordan. Direct DNA sequencing was performed on six genes including RUNX1 gene (exons 3-8). RESULTS RUNX1 point mutations were identified in 10 (5.6%) of 178 patients. One patient possessed biallelic mutations of RUNX1 gene. C-terminal area was the predominant site of RUNX1 mutations (eight in C-terminal and two in N-terminal). Patients with RUNX1 mutations were significantly older than those with wild-type of the gene. Additionally, AML M0 subtype was more frequently found in patients with RUNX1 mutations. Both RUNX1 mutations and RAS mutations were identified in 4 of 10 children. Three patients with RUNX1 mutation had FLT3-ITD. On the other hand, 36 (21.4%) and 25 (14.9%) of 168 patients with wild-type of the gene had a RAS mutation and FLT3-ITD, respectively. Eight of 10 patients with RUNX1 mutations died of hematological relapse. CONCLUSION The incidence of RUNX1 mutations in Arab Asian children and adolescents with AML was 5.6%. Further studies are required to clarify whether RAS mutations contribute to the development of pediatric AML associated with RUNX1 mutations.
Collapse
|
24
|
Piazza F, Semenzato G. Molecular therapeutic approaches to acute myeloid leukemia: targeting aberrant chromatin dynamics and signal transduction. Expert Rev Anticancer Ther 2014; 4:387-400. [PMID: 15161438 DOI: 10.1586/14737140.4.3.387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acute myeloid leukemia research and clinical management have greatly benefited from the achievements in molecular biology regarding the identification of the underlying pathogenetic mechanisms of transformation and resistance to therapy. In particular, two categories of alterations, the aberrant activity of transcription/chromatin-remodeling factors and the deregulated activation of signal transduction pathways, have been demonstrated to play a pivotal role in leukemic cell differentiation, proliferation and resistance to apoptosis. These molecular lesions have proven to be suitable therapeutic targets in acute promyelocytic leukemia and chronic myeloid leukemia and are now also seen as therapeutic targets for a wider group of leukemic disorders. The development of novel drugs such as histone deacetylase inhibitors, demethylating agents and inhibitors of receptor tyrosine kinases may potentially benefit acute myeloid leukemia patients.
Collapse
Affiliation(s)
- Francesco Piazza
- Padova University School of Medicine, Venetian Institute of Molecular Medicine, Unit of Hematological Malignancies, via Orus 2 35129 Padova, Italy.
| | | |
Collapse
|
25
|
RUNX1 mutation associated with clonal evolution in relapsed pediatric acute myeloid leukemia with t(16;21)(p11;q22). Int J Hematol 2013; 99:169-74. [DOI: 10.1007/s12185-013-1495-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 12/09/2013] [Accepted: 12/10/2013] [Indexed: 12/13/2022]
|
26
|
Kulasekararaj AG, Mohamedali AM, Mufti GJ. Recent advances in understanding the molecular pathogenesis of myelodysplastic syndromes. Br J Haematol 2013; 162:587-605. [PMID: 23869491 DOI: 10.1111/bjh.12435] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The advent of novel genomic sequencing technologies has aided the identification of somatically acquired genetic abnormalities up to 80% of myelodysplastic syndrome (MDS) patients. Novel recurrent genetic mutations in pathways such as RNA splicing, DNA methylation and histone modification and cohesion complexes, underscore the molecular heterogeneity seen in this clinically varied disease. Functional studies to establish a causative link between genomic aberrations and MDS biogenesis are still in their infancy. The deluge of this molecular information, once validated on a larger cohort, will be incorporated into prognostic systems and clinical practise, and also hopefully aid in MDS therapeutics, especially in guiding targeted therapy.
Collapse
Affiliation(s)
- Austin G Kulasekararaj
- Department of Haematological Medicine, King's College London School of Medicine, London, UK
| | | | | |
Collapse
|
27
|
Koh CP, Wang CQ, Ng CEL, Ito Y, Araki M, Tergaonkar V, Huang G, Osato M. RUNX1 meets MLL: epigenetic regulation of hematopoiesis by two leukemia genes. Leukemia 2013; 27:1793-802. [PMID: 23817177 DOI: 10.1038/leu.2013.200] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/26/2013] [Accepted: 06/26/2013] [Indexed: 01/05/2023]
Abstract
A broad range of human leukemias carries RUNX1 and MLL genetic alterations. Despite such widespread involvements, the relationship between RUNX1 and MLL has never been appreciated. Recently, we showed that RUNX1 physically and functionally interacts with MLL, thereby regulating the epigenetic status of critical cis-regulatory elements for hematopoietic genes. This newly unveiled interaction between the two most prevalent leukemia genes has solved a long-standing conundrum: leukemia-associated RUNX1 N-terminal point mutants that exhibit no obvious functional abnormalities in classical assays for the assessment of transcriptional activities. These mutants turned out to be defective in MLL interaction and subsequent epigenetic modifications that can be examined by the histone-modification status of cis-regulatory elements in the target genes. RUNX1/MLL binding confirms the importance of RUNX1 function as an epigenetic regulator. Recent studies employing next-generation sequencing on human hematological malignancies identified a plethora of mutations in epigenetic regulator genes. These new findings would enhance our understanding on the mechanistic basis for leukemia development and may provide a novel direction for therapeutic applications. This review summarizes the current knowledge about the epigenetic regulation of normal and malignant hematopoiesis by RUNX1 and MLL.
Collapse
Affiliation(s)
- C P Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Keita M, Bachvarova M, Morin C, Plante M, Gregoire J, Renaud MC, Sebastianelli A, Trinh XB, Bachvarov D. The RUNX1 transcription factor is expressed in serous epithelial ovarian carcinoma and contributes to cell proliferation, migration and invasion. Cell Cycle 2013; 12:972-86. [PMID: 23442798 DOI: 10.4161/cc.23963] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Previously, we have identified the RUNX1 gene as hypomethylated and overexpressed in post-chemotherapy (CT) primary cultures derived from epithelial ovarian cancer (EOC) patients, when compared with primary cultures derived from matched primary (prior to CT) tumors. Here we show that RUNX1 displays a trend of hypomethylation, although not significant, in omental metastases compared with primary EOC tumors. Surprisingly, RUNX1 displayed significantly higher expression not only in metastatic tissue, but also in high-grade primary tumors and even in low malignant potential tumors. The RUNX1 expression levels were almost identical in primary tumors and omental metastases, suggesting that RUNX1 hypomethylation might have a limited impact on its overexpression in advanced (metastatic) stage of the disease. Knockdown of the RUNX1 expression in EOC cells led to sharp decrease of cell proliferation and induced G 1 cell cycle arrest. Moreover, RUNX1 suppression significantly inhibited EOC cell migration and invasion. Gene expression profiling and consecutive network and pathway analyses confirmed these findings, as numerous genes and pathways known previously to be implicated in ovarian tumorigenesis, including EOC tumor invasion and metastasis, were found to be downregulated upon RUNX1 suppression, while a number of pro-apoptotic genes and some EOC tumor suppressor genes were induced. Taken together, our data are indicative for a strong oncogenic potential of the RUNX1 gene in EOC progression and suggest that RUNX1 might be a novel EOC therapeutic target. Further studies are needed to more completely elucidate the functional implications of RUNX1 and other members of the RUNX gene family in ovarian tumorigenesis.
Collapse
Affiliation(s)
- Mamadou Keita
- Department of Molecular Medicine, Laval University, Québec, QC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wu D, Ozaki T, Yoshihara Y, Kubo N, Nakagawara A. Runt-related transcription factor 1 (RUNX1) stimulates tumor suppressor p53 protein in response to DNA damage through complex formation and acetylation. J Biol Chem 2012; 288:1353-64. [PMID: 23148227 DOI: 10.1074/jbc.m112.402594] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Representative tumor suppressor p53 plays a critical role in the regulation of proper DNA damage response. In this study, we have found for the first time that Runt-related transcription factor 1 (RUNX1) contributes to p53-dependent DNA damage response. Upon adriamycin (ADR) exposure, p53 as well as RUNX1 were strongly induced in p53-proficient HCT116 and U2OS cells, which were closely associated with significant transactivation of p53 target genes, such as p21(WAF)(1), BAX, NOXA, and PUMA. RUNX1 was exclusively expressed in the cell nucleus and formed a complex with p53 in response to ADR. Chromatin immunoprecipitation assay demonstrated that p53 together with RUNX1 are efficiently recruited onto p53 target gene promoters following ADR exposure, indicating that RUNX1 is involved in p53-mediated transcriptional regulation. Indeed, forced expression of RUNX1 stimulated the transcriptional activity of p53 in response to ADR. Consistent with these observations, knockdown of RUNX1 attenuated ADR-mediated induction of p53 target genes and suppressed ADR-dependent apoptosis. Furthermore, RUNX1 was associated with p300 histone acetyltransferase, and ADR-dependent acetylation of p53 at Lys-373/382 was markedly inhibited in RUNX1 knockdown cells. In addition, knockdown of RUNX1 resulted in a significant decrease in the amount of p53-p300 complex following ADR exposure. Taken together, our present results strongly suggest that RUNX1 is required for the stimulation of p53 in response to DNA damage and also provide novel insight into understanding the molecular mechanisms behind p53-dependent DNA damage response.
Collapse
Affiliation(s)
- Dan Wu
- Laboratory of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, 666-2 Nitona, Chiba 260-8717, Japan
| | | | | | | | | |
Collapse
|
30
|
Mullighan CG. Molecular genetics of B-precursor acute lymphoblastic leukemia. J Clin Invest 2012; 122:3407-15. [PMID: 23023711 DOI: 10.1172/jci61203] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
B-precursor acute lymphoblastic leukemia (B-ALL) is the most common childhood tumor and the leading cause of cancer-related death in children and young adults. The majority of B-ALL cases are aneuploid or harbor recurring structural chromosomal rearrangements that are important initiating events in leukemogenesis but are insufficient to explain the biology and heterogeneity of disease. Recent studies have used microarrays and sequencing to comprehensively identify all somatic genetic alterations in acute lymphoblastic leukemia (ALL). These studies have identified cryptic or submicroscopic genetic alterations that define new ALL subtypes, cooperate with known chromosomal rearrangements, and influence prognosis. This article reviews these advances, discusses results from ongoing second-generation sequencing studies of ALL, and highlights challenges and opportunities for future genetic profiling approaches.
Collapse
Affiliation(s)
- Charles G Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, USA.
| |
Collapse
|
31
|
Qin S, Zhou HX. PI 2PE: A Suite of Web Servers for Predictions Ranging From Protein Structure to Binding Kinetics. Biophys Rev 2012; 5:41-46. [PMID: 23526172 DOI: 10.1007/s12551-012-0086-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
PI2PE (http://pipe.sc.fsu.edu) is a suite of four web servers for predicting a variety of folding- and binding-related properties of proteins. These include the solvent accessibility of amino acids upon protein folding, the amino acids forming the interfaces of protein-protein and protein-nucleic acid complexes, and the binding rate constants of these complexes. Three of the servers debuted in 2007, and have garnered ~2,500 unique users and finished over 30,000 jobs. The functionalities of these servers are now enhanced, and a new sever, for predicting the binding rate constants, is added. Together, these web servers form a pipeline from protein sequence to tertiary structure, then to quaternary structure, and finally to binding kinetics.
Collapse
Affiliation(s)
- Sanbo Qin
- Department of Physics and Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306, USA
| | | |
Collapse
|
32
|
Montero-Ruíz O, Alcántara-Ortigoza MA, Betancourt M, Juárez-Velázquez R, González-Márquez H, Pérez-Vera P. Expression of RUNX1 isoforms and its target gene BLK in childhood acute lymphoblastic leukemia. Leuk Res 2012; 36:1105-11. [PMID: 22748822 DOI: 10.1016/j.leukres.2012.05.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 05/16/2012] [Accepted: 05/22/2012] [Indexed: 12/11/2022]
Abstract
Bone marrow samples from children with acute lymphoblastic leukemia were analyzed for the expression of RUNX1a/b/c isoforms. Obtained patterns were associated with genetic abnormalities and the expression of the RUNX1 regulated gene BLK. RUNX1c was present in all patients, but the expected over-expression of RUNX1a was not observed. Over-expression of total RUNT domain isoforms was detected in patients with extra RUNX1 copies, and unexpectedly, in those with t(4;11). Only expression of the total RUNT domain-containing isoforms and BLK presented positive correlation. Results suggest a more complex role of RUNX1 in leukemogenesis than the proposed antagonism between the isoforms.
Collapse
Affiliation(s)
- Oreth Montero-Ruíz
- Laboratorio de Cultivo de Tejidos, Instituto Nacional de Pediatría, México, DF, Mexico
| | | | | | | | | | | |
Collapse
|
33
|
Markova EN, Kantidze OL, Razin SV. Transcriptional regulation and spatial organisation of the human AML1/RUNX1 gene. J Cell Biochem 2011; 112:1997-2005. [PMID: 21445863 DOI: 10.1002/jcb.23117] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The transcription factor RUNX1 is a key regulator of haematopoiesis in vertebrates. In humans, the 260-kb long gene coding for this transcription factor is located on chromosome 21. This gene is transcribed from two alternative promoters that are commonly referred to as the distal and the proximal promoters. In model experiments, these two promoters were found to be active in cells of different lineages, although RUNX1 is preferentially expressed in haematopoietic cells. In the present study, we attempted to identify the regulatory elements that could guide tissue-specific expression of the RUNX1 gene. Two such regulatory elements were found within the RUNX1 gene. One of these elements, located within intron 1, is a haematopoietic-specific enhancer. The second regulatory element, located within intron 5.2, contributes to the formation of an active chromatin hub, which integrates the above-mentioned enhancer and the P1 and P2 promoters.
Collapse
Affiliation(s)
- Elena N Markova
- Laboratory of Structural and Functional Organization of Chromosomes, Institute of Gene Biology RAS, Moscow, Russia
| | | | | |
Collapse
|
34
|
Inoue KI, Ito Y. Neuroblastoma cell proliferation is sensitive to changes in levels of RUNX1 and RUNX3 protein. Gene 2011; 487:151-5. [DOI: 10.1016/j.gene.2011.05.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 05/16/2011] [Accepted: 05/17/2011] [Indexed: 12/12/2022]
|
35
|
The ability of MLL to bind RUNX1 and methylate H3K4 at PU.1 regulatory regions is impaired by MDS/AML-associated RUNX1/AML1 mutations. Blood 2011; 118:6544-52. [PMID: 22012064 DOI: 10.1182/blood-2010-11-317909] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mixed-lineage leukemia (MLL) H3K4 methyltransferase protein, and the heterodimeric RUNX1/CBFβ transcription factor complex, are critical for definitive and adult hematopoiesis, and both are frequently targeted in human acute leukemia. We identified a physical and functional interaction between RUNX1 (AML1) and MLL and show that both are required to maintain the histone lysine 4 trimethyl mark (H3K4me3) at 2 critical regulatory regions of the AML1 target gene PU.1. Similar to CBFβ, we show that MLL binds to AML1 abrogating its proteasome-dependent degradation. Furthermore, a subset of previously uncharacterized frame-shift and missense mutations at the N terminus of AML1, found in MDS and AML patients, impairs its interaction with MLL, resulting in loss of the H3K4me3 mark within PU.1 regulatory regions, and decreased PU.1 expression. The interaction between MLL and AML1 provides a mechanism for the sequence-specific binding of MLL to DNA, and identifies RUNX1 target genes as potential effectors of MLL function.
Collapse
|
36
|
Cytoplasmic nucleophosmin (cNPM) in acute myeloid leukaemia: Relation to disease characteristics. ALEXANDRIA JOURNAL OF MEDICINE 2011. [DOI: 10.1016/j.ajme.2011.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
37
|
Hyde RK, Liu PP. RUNX1 repression-independent mechanisms of leukemogenesis by fusion genes CBFB-MYH11 and AML1-ETO (RUNX1-RUNX1T1). J Cell Biochem 2010; 110:1039-45. [PMID: 20589720 DOI: 10.1002/jcb.22596] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The core binding factor (CBF) acute myeloid leukemias (AMLs) are a prognostically distinct subgroup that includes patients with the inv(16) and t(8:21) chromosomal rearrangements. Both of these rearrangements result in the formation of fusion proteins, CBFB-MYH11 and AML1-ETO, respectively, that involve members of the CBF family of transcription factors. It has been proposed that both of these fusion proteins function primarily by dominantly repressing normal CBF transcription. However, recent reports have indicted that additional, CBF-repression independent activities may be equally important during leukemogenesis. This article will focus on these recent advances.
Collapse
Affiliation(s)
- R Katherine Hyde
- Oncogenesis and Development Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
38
|
Xu X, Zhao Y, Xu M, Dai Q, Meng W, Yang J, Qin R. Activation of Notch signal pathway is associated with a poorer prognosis in acute myeloid leukemia. Med Oncol 2010; 28 Suppl 1:S483-9. [DOI: 10.1007/s12032-010-9667-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Accepted: 08/18/2010] [Indexed: 10/19/2022]
|
39
|
Jekarl DW, Kim M, Lim J, Kim Y, Han K, Lee AW, Kim HJ, Min WS. CD56 antigen expression and hemophagocytosis of leukemic cells in acute myeloid leukemia with t(16;21)(p11;q22). Int J Hematol 2010; 92:306-13. [DOI: 10.1007/s12185-010-0650-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 07/12/2010] [Accepted: 07/14/2010] [Indexed: 11/29/2022]
|
40
|
Ahmad F, Mandava S, Das BR. Analysis of FLT3-ITD and FLT3-Asp835 mutations in de novo acute myeloid leukemia: evaluation of incidence, distribution pattern, correlation with cytogenetics and characterization of internal tandem duplication from Indian population. Cancer Invest 2010; 28:63-73. [PMID: 19995225 DOI: 10.3109/07357900903095649] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mutation of the FMS-like tyrosine kinase 3 (FLT3) gene in Indian population remains unclear till date. Here, we found FLT3-ITD mutations in 19.1%, FLT3-Asp835 mutations in 4.7%, and dual mutations in 4.2%, accounting for overall mutation in 28% of acute myeloid leukemia (AML) patients. FLT3 mutation was more prevalent in APL than non-APL patients (32.2% vs 26.3%), adults tend to show higher incidence than children (30.6% vs 18.2%, p = .1), and were significantly associated with normal karyotype, high WBCs, with no specific distribution in FAB subtypes. Notably, FLT3 mutation was present in 50% of patients with NPM1-Mt, when compared to only 22.6% of patients with NPM1-wt (p < .001). Sequence analyses of internal tandem duplications (ITDs) revealed that duplications were mostly restricted to JM domain (3 to 165 nucleotides). Interestingly, 92.3% cases showed duplication of at least one amino acid (AA) within the stretch Y589 to K602 that includes the two SH2-binding motifs. Analysis of frequency of single AA in the duplicated region revealed that E598 was the most frequently duplicated single AA in 72%, followed by R595 (69.2%), and Y599 (66.7%). Finally, three types of point mutations were identified, including D835Y, D835H, and D835A.
Collapse
Affiliation(s)
- Firoz Ahmad
- Research and Development Division, Super Religare Laboratories Ltd (Formerly SRL Ranbaxy Ltd), Plot No.124, 17th Street, MIDC, Andheri (E), Mumbai, India
| | | | | |
Collapse
|
41
|
Gocek E, Kiełbiński M, Baurska H, Haus O, Kutner A, Marcinkowska E. Different susceptibilities to 1,25-dihydroxyvitamin D3-induced differentiation of AML cells carrying various mutations. Leuk Res 2009; 34:649-57. [PMID: 19880182 DOI: 10.1016/j.leukres.2009.10.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 09/12/2009] [Accepted: 10/04/2009] [Indexed: 11/16/2022]
Abstract
This study was designed to compare the differentiation-inducing potential of 1,25-dihydroxyvitamin D(3) (1,25D) with some analogs (VDAs) in a panel of acute myeloid leukemia (AML) cell lines and in blast cells isolated from patients with AML. Of the cell lines studied, HL60 proved to be the most sensitive to each of the differentiation-inducing agents when compared to THP-1, NB-4 and U-937 cell lines. Three of the VDAs tested (PRI-1906, PRI-2191 and PRI-2201) were similarly effective as 1,25D in all the cell lines tested. However, blast cells from AML showed a varying sensitivity towards 1,25D. For example, blast cells isolated from patients in which the whole or part of chromosome 7 was deleted were extremely sensitive to 1,25D and its analogs. In contrast, 1,25D failed to increase the expression of differentiation markers in blast cells isolated from patients carrying activating mutations in Flt3 gene. Since, the expression of vitamin D receptor (VDR) in cells with Flt3 mutations was increased to the same extent as in other AML cells this suggests that failure of these cells to differentiate lies downstream of the receptor. That blast cells with different cytogenetic abnormalities have dissimilar responses to 1,25D and its analogs, may have implications in the use of 1,25D as a 'differentiation therapy' for myeloid leukemias. The analog PRI-2191 (tacalcitol) was found to be the most potent in inducing patient's cells differentiation.
Collapse
Affiliation(s)
- Elzbieta Gocek
- Department of Biotechnology, University of Wrocław, Tamka 2, 50-137 Wrocław, Poland
| | | | | | | | | | | |
Collapse
|
42
|
Slovak ML, O'Donnell M, Smith DD, Gaal K. Does MDS with der(1;7)(q10;p10) constitute a distinct risk group? A retrospective single institutional analysis of clinical/pathologic features compared to -7/del(7q) MDS. ACTA ACUST UNITED AC 2009; 193:78-85. [PMID: 19665067 DOI: 10.1016/j.cancergencyto.2009.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 04/07/2009] [Indexed: 10/20/2022]
Abstract
The der(1;7)(q10;p10) aberration is observed in about 1-3% of the myelodysplastic syndromes (MDS) and less commonly in acute myeloid leukemia (AML) and the myeloproliferative disorders. This unbalanced translocation is considered a "variant" of the del(7q)/-7 subgroup and has been assigned a poor risk karyotype score in the MDS International Prognostic Scoring System (IPSS). Recent reports suggest der(1;7) MDS should be considered a discrete MDS subgroup with an intermediate, not poor, karyotype score. At the City of Hope, we compared the clinical-pathologic features of 12 der(1;7) MDS patients to 51 MDS patients with del(7q) (n=10) or -7 (n=41), selected for a similar frequency of secondary aberrations. The der(1;7) patients showed older age at diagnosis, lower platelet counts, less trilineage dysplasia, and lower blast counts. The der(1;7) patients did not differ from del(7q)/-7 patients in subtypes of MDS by World Health Organization, French-American-British classifications, or bone marrow cellularity. Neither the proportion of therapy-related MDS nor the transformation to AML differed significantly among the three subgroups. Five-year survival rates for der(1;7), del(7q), and -7 (44.4, 32.0, and 23.6%, respectively) did not differ significantly (P=0.94). While der(1;7) MDS is associated with some clinically distinctive features, reassignment of risk category based on these data would be premature.
Collapse
Affiliation(s)
- Marilyn L Slovak
- Cytogenetics Laboratory, City of Hope, 1500 E. Duarte Road, Duarte, CA 91010, USA.
| | | | | | | |
Collapse
|
43
|
Bae SY, Kim JS, Han EA, Lim CS, Ryeu BJ, Lee KN, Yoon SY, Cho Y, Kim YK, Lee CK. ConcurrentMYCandMLLamplification on dmin and hsr in acute myeloid leukemia. Leuk Lymphoma 2009; 49:1823-5. [DOI: 10.1080/10428190802216715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
44
|
Mutations ofNPM1gene inde novoacute myeloid leukaemia: determination of incidence, distribution pattern and identification of two novel mutations in Indian population. Hematol Oncol 2009; 27:90-7. [PMID: 19365794 DOI: 10.1002/hon.883] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
45
|
High frequency of RUNX1 biallelic alteration in acute myeloid leukemia secondary to familial platelet disorder. Blood 2009; 113:5583-7. [PMID: 19357396 DOI: 10.1182/blood-2008-07-168260] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Familial platelet disorder (FPD), a rare autosomal dominant disorder characterized by quantitative and qualitative platelet abnormalities, is considered as a model of genetic predisposition to acute myeloid leukemia (AML). So far, monoallelic RUNX1 germline mutations have been found in 19 of 20 families with reported FPD, and the analysis of blast cells from only 5 patients at acute leukemia (AL) stage has shown no additional RUNX1 abnormality. Here, we performed RUNX1 analysis at constitutional and somatic levels in 8 persons with FPD who developed AL from 4 independent families. In addition to the germline RUNX1 mutation, we identified a second RUNX1 alteration in 6 AML cases (acquired point mutations in 4 cases and duplication of the altered RUNX1 allele associated with acquired trisomy 21 in 2 other cases). Although haploinsufficiency of RUNX1 causes FPD, our findings suggest that a second genetic event involving RUNX1 is often associated with progression to AML.
Collapse
|
46
|
Kuo YH, Zaidi SK, Gornostaeva S, Komori T, Stein GS, Castilla LH. Runx2 induces acute myeloid leukemia in cooperation with Cbfbeta-SMMHC in mice. Blood 2009; 113:3323-32. [PMID: 19179305 PMCID: PMC2665897 DOI: 10.1182/blood-2008-06-162248] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Accepted: 01/03/2009] [Indexed: 12/29/2022] Open
Abstract
The core-binding factor (CBF) is a master regulator of developmental and differentiation programs, and CBF alterations are frequently associated with acute leukemia. The role of the CBF member RUNX2 in hematopoiesis is poorly understood. Genetic evidence suggests that deregulation of Runx2 may cause myeloid leukemia in mice expressing the fusion oncogene Cbfb-MYH11. In this study, we show that sustained expression of Runx2 modulates Cbfbeta-smooth muscle myosin heavy chain (SMMHC)-mediated myeloid leukemia development. Expression of Runx2 is high in the hematopoietic stem cell compartment and decreases during myeloid differentiation. Sustained Runx2 expression hinders myeloid progenitor differentiation capacity and represses expression of CBF targets Csf1R, Mpo, Cebpd, the cell cycle inhibitor Cdkn1a, and myeloid markers Cebpa and Gfi1. In addition, full-length Runx2 cooperates with Cbfbeta-SMMHC in leukemia development in transplantation assays. Furthermore, we show that the nuclear matrix-targeting signal and DNA-binding runt-homology domain of Runx2 are essential for its leukemogenic activity. Conversely, Runx2 haplo-insufficiency delays the onset and reduces the incidence of acute myeloid leukemia. Together, these results indicate that Runx2 is expressed in the stem cell compartment, interferes with differentiation and represses CBF targets in the myeloid compartment, and modulates the leukemogenic function of Cbfbeta-SMMHC in mouse leukemia.
Collapse
MESH Headings
- Animals
- Bone Marrow/metabolism
- Bone Marrow/physiology
- Cell Differentiation/genetics
- Cell Transformation, Neoplastic/genetics
- Cells, Cultured
- Core Binding Factor Alpha 1 Subunit/genetics
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/physiology
- Down-Regulation/genetics
- Hematopoiesis/genetics
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/physiology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Mice
- Mice, Transgenic
- Models, Biological
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/physiology
- Survival Analysis
Collapse
Affiliation(s)
- Ya-Huei Kuo
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | |
Collapse
|
47
|
Ayala RM, Martínez-López J, Albízua E, Diez A, Gilsanz F. Clinical significance of Gata-1, Gata-2, EKLF, and c-MPL expression in acute myeloid leukemia. Am J Hematol 2009; 84:79-86. [PMID: 19097174 DOI: 10.1002/ajh.21332] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The aim of this study was to evaluate the biological correlation and prognostic impact of Gata-1, Gata-2, EKLF, and c-MPL transcript level in a group of 41 acute myeloid leukemia (AML) patients. Gata-1 overexpression was related to advanced age and a low percentage of bone marrow blasts and was associated with the expression of CD34 antigen and lymphoid T markers. The negative impact of Gata-1 expression on the probability of achieving complete remission has been confirmed. Gata-2 overexpression was associated with a low percentage of blasts in BM and males. Expression of c-MPL was associated with CD34+ AML and M2 FAB AML subtype. A higher expression of EKLF was found in secondary AML versus primary AML. Nevertheless, patients expressing EKLF had a longer overall survival and event free survival than those patients that did not express EKLF. Our study has identified expression of EKLF as a factor with a favorable impact on prognosis in AML.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Bone Marrow/pathology
- Chromosome Aberrations
- Disease-Free Survival
- Erythropoiesis/genetics
- GATA1 Transcription Factor/analysis
- GATA1 Transcription Factor/physiology
- GATA2 Transcription Factor/analysis
- GATA2 Transcription Factor/physiology
- Gene Expression Regulation, Neoplastic
- Humans
- Kruppel-Like Transcription Factors/analysis
- Kruppel-Like Transcription Factors/physiology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Middle Aged
- Neoplasm Proteins/analysis
- Neoplasm Proteins/physiology
- Neoplasms, Second Primary/genetics
- Neoplasms, Second Primary/metabolism
- Neoplasms, Second Primary/mortality
- Neoplasms, Second Primary/pathology
- Prognosis
- Receptors, Thrombopoietin/analysis
- Receptors, Thrombopoietin/physiology
- Survival Analysis
- Young Adult
Collapse
Affiliation(s)
- Rosa M Ayala
- Servicio de Hematologia, Hospital Universitario 12 de Octubre, Madrid, España.
| | | | | | | | | |
Collapse
|
48
|
Moosavi SA, Sanchez J, Adeyinka A. Marker chromosomes are a significant mechanism of high-level RUNX1 gene amplification in hematologic malignancies. ACTA ACUST UNITED AC 2009; 189:24-8. [DOI: 10.1016/j.cancergencyto.2008.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Accepted: 10/08/2008] [Indexed: 10/21/2022]
|
49
|
Deluche L, Joha S, Corm S, Daudignon A, Geffroy S, Quief S, Villenet C, Kerckaert JP, Laï JL, Preudhomme C, Roche-Lestienne C. Cryptic and partial deletions of PRDM16 and RUNX1 without t(1;21)(p36;q22) and/or RUNX1-PRDM16 fusion in a case of progressive chronic myeloid leukemia: a complex chromosomal rearrangement of underestimated frequency in disease progression? Genes Chromosomes Cancer 2008; 47:1110-7. [PMID: 18767145 DOI: 10.1002/gcc.20611] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disorder characterized by the presence in leukemic stem cells of the Philadelphia chromosome (Ph) and the formation of the BCR-ABL1 fusion. Untreated, the disease progresses to accelerate phase and blast crisis in which hematopoietic differentiation has become arrested. CML progression is frequently associated with cytogenetic evidence of clonal evolution, defined as additional chromosomal aberrations. We here report a CML resistant to tyrosine kinase inhibitors that rapidly progressed to blastic phase. At this time, array CGH performed on CD34(+) cells revealed cryptic partial deletions of both PRDM16 and RUNX1 and duplication of the der(21) chromosome. These genomic rearrangements were confirmed by FISH with probes targeting the deletion on chromosome 21 (24 kb), and with BAC probes flanking the deletion on 1p36 (220 kb). However, no cryptic t(1;21)(p36;q22) and/or RUNX1-PRDM16 were detected, suggesting that these deletions are the residual hallmarks of a more complex mechanism of chromosomal rearrangement, as indicated by the additional inversion of the region bounded by 1p36.32 and 1p36.12 breaks. At the molecular level, these abnormalities lead to the overexpression of the PR-domain negative oncogenic isoform of PRDM16, associated with two deleted copies within the runt domain of C-teminal aberrant RUNX1. These events are not detectable by conventional cytogenetic and molecular strategies, and may be of underestimated frequency in disease progression.
Collapse
Affiliation(s)
- Lauréline Deluche
- Cancer Research Institute of Lille, JP Aubert Center, Inserm Unit 837, Lille, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Pais AP, Amare Kadam PS, Raje GC, Banavali S, Parikh P, Kurkure P, Arora B, Gujral S, Kumar SA, Badrinath Y. RUNX1 aberrations in ETV6/RUNX1-positive and ETV6/RUNX1-negative patients: its hemato-pathological and prognostic significance in a large cohort (619 cases) of ALL. Pediatr Hematol Oncol 2008; 25:582-97. [PMID: 18728978 DOI: 10.1080/08880010802237450] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A large-cohort study (619) of acute lymphoblastic leukemia (ALL) revealed an ETV6/RUNX1 (previously known as TEL/AML1) incidence of 18% in pediatric B-cell precussor ALL, indicating no geographical heterogeinity. Association of CD34-negative phenotype, peak incidence in the 3- to 7-year age group, and a comparatively low frequency of ETV6 homologue loss in ETV6/RUNX1-positive cases were distinct findings in this series. Additional genetic changes, such as ETV6 loss, extra RUNX1, ETV6/RUNX1 duplication, and MLL aberrations in the ETV6/RUNX1-positive group, supported the hypothesis of the ETV6/RUNX1 leukemogenic model that these secondary changes are necessary for leukemogenesis rather than progression of disease. This study disclosed RUNX1 alterations in the ETV6/RUNX1-negative group of BCP-ALL that encourages the investigation of RUNX1 at a large scale with longer follow-up, which will focus on the prognostic importance and the underlying biology of disease.
Collapse
Affiliation(s)
- Anurita Peter Pais
- Cancer Cytogenetics Laboratory, Parel, Tata Memorial Hospital, Mumbai, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|