Open Access

Chromosome t(7;11)(p15;p15) translocation in acute myeloid leukemia coexisting with multilineage dyspoiesis and mutations in NRAS and WT1: A case report and literature review

  • Authors:
    • Jingke Yang
    • Xiaodong Lyu
    • Xinghu Zhu
    • Xiangguang Meng
    • Wenli Zuo
    • Hao Ai
    • Mei Deng
  • View Affiliations

  • Published online on: March 7, 2017     https://doi.org/10.3892/ol.2017.5823
  • Pages: 3066-3070
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The chromosomal translocation t(7;11)(p15;p15) and the resulting nucleoporin 98-homeobox A9 (NUP98-HOXA9) gene fusion is rare but recurrent genetic abnormity in acute myeloid leukemia (AML). The present study describes a case of AML plus maturation (‑M2) with multilineage dyspoiesis in a 30‑year‑old male in whom a 46,XY,t(7;11)(p15;p15) karyotype was detected through chromosome analysis. Subsequent molecular and sequencing analysis demonstrated a NUP98‑HOXA9 fusion gene with a type I fusion between NUP98 exon 12 and HOXA9 exon 1b, and mutations in neuroblastoma V‑Ras oncogene homolog and Wilms tumor 1. The patient achieved hematological complete remission (CR) following two courses of induction chemotherapy. However, the NUP98‑HOXA9 fusion gene remained detectable during the hematological CR period and following intensive consolidation chemotherapy. The disease relapsed 11 months after diagnosis, and the patient became refractory, with complications from an infection causing eventual mortality. The present case and literature review suggest that patients with AML and t(7;11) may have unique biological and clinical characteristics, and a poor prognosis.

Introduction

Acute myeloid leukemia (AML) is a biologically and clinically heterogeneous disorder, characterized by immature myeloid cell proliferation and bone marrow failure (1). The most common subtype of AML (M0-M7) is M2 under FAB classification (2,3). In total ~52% of adult patients with de novo AML are reported to carry at least one type of chromosomal abnormalities (e.g. translocations or deletions) (3). A number of recurrent chromosomal aberrations and gene mutations involved in hematopoietic proliferation and differentiation have been used as critical risk stratification tools, including chromosomal translocations t(8;21) and t(15;17), and internal tandem duplication of Fms-related tyrosine kinase 3 (FLT3) mutations (3,4). The 5-year overall survival rates for each risk group (favorable, intermediate and adverse-risk) are ~55, ~24 and ~5%, respectively (3). However, prognosis remains markedly different for each risk group (4). Therefore, there is a requirement to identify additional genetic alterations that are associated with prognosis.

The chromosomal translocation t(7;11)(p15;p15) is a rare genetic lesion in AML, which was initially reported in a patient with chronic myeloid leukemia (CML) more than 30 years ago (5). This translocation results in a fusion of the N-terminal portion of nucleoporin 98 (NUP98) with the homeodomain of several homeobox A (HOXA) genes, including HOXA9, HOXA11 and HOXA13; the most common fusion is HOXA9 (68). The association between clinical and biological characteristics from AML patients with this type of fusion remains to be fully elucidated. The present study reports the case of a male patient with AML-M2 and t(7;11) translocation resulting in a NUP98-HOXA9 fusion gene. The patient demonstrated dyspoietic alterations, neuroblastoma V-Ras oncogene homolog (NRAS) and Wilms tumor 1 (WT1) mutations and a poor prognosis.

Case report

A 30-year-old male was admitted to Affiliated Cancer Hospital of Zhengzhou University (Zhengzhou, China) following 2 weeks of pain in each leg in August 2015. The patient presented with pale skin and sternal tenderness. Peripheral blood analysis identified a white blood cell count of 32.2×109/l (normal range, 4–10×109/l), with 44% blasts and 26% monocytes (normal range, 3–8%), a hemoglobin level of 110 g/l (normal range, 120–160 g/l) and a platelet count of 87×109/l (normal range, 100–300×109/l). A bone marrow (BM) smear demonstrated a markedly hypercellular BM, with 71% myeloblasts, and a positive result for myeloperoxidase staining, which suggested an AML-M2 subtype (2). Dyspoietic alterations, including multinucleated erythrocyte precursors and micromegakaryocytes, were identified (Fig. 1). Subsequently, immunophenotyping was performed on a multi-color flow cytometer (BD FACSCalibur; BD Biosciences, Franklin Lakes, NJ, USA) with monoclonal antibodies in 1:1 dilutions, including cluster of differentiation (CD) 34-fluorescein isothiocyanate (FITC) (catalog no. 555821), human leukocyte antigen-antigen D-related (HLA-DR)-allophycocyanin (APC) (catalog no. 340549), CD13-phycoerythrin (PE) (catalog no. 347837), cytoplasmic myeloperoxidase (cMPO)-FITC (catalog no. 340580), CD15-FITC (catalog no. 332778), CD20-APC (catalog no. 340941) cytoplasmic (c)CD3-APC (catalog no. 340440), CD5-APC (catalog no. 340583), CD11b-APC (catalog no. 340937) and CD3-APC (catalog no. 555335), all of which were purchased from BD Biosciences. Immunophenotyping was also performed with the following antibodies in 1:1 dilutions: CD33-PE (catalog no. 555450; BD Pharmingen, San Diego, CA, USA), CD117-PE (catalog no. 340529; BD Pharmingen), CD56-PE (catalog no. A07788; Beckman Coulter, Inc., Brea, CA, USA), CD7-PE (catalog no. IM1429U; Beckman Coulter, Inc.), CD19-FITC (catalog no. A07768; Beckman Coulter, Inc.), CD4-FITC (catalog no. A07750; Beckman Coulter, Inc.), CD2-FITC (catalog no. A07743; Beckman Coulter, Inc.), CD10-PE (catalog no. A07760; Beckman Coulter, Inc.), CD14-FITC (catalog no. 0645U; Beckman Coulter, Inc.) and cCD79a-PE (catalog no. IM2221; Beckman Coulter, Inc.).

Blasts were positive for CD34, HLA-DR, CD13, CD33, CD117 and cMPO antigens, and negative for CD56, CD7, CD15, CD19, CD4, CD2, CD10, CD20, CD3, CD5, CD11b, CD14, cCD79a and cCD3 antigens. These results confirmed that the blasts were committed to being myeloid precursors.

Chromosome analysis using the R-banding method identified a 46,XY,t(7;11)(p15;p15) karyotype in all 20 metaphase cells (Fig. 2). NUP98-HOXA9 was identified via a screen of leukemia-associated fusion genes using a multi-fusion gene detection system (43 Fusion Gene Screening kit; Shanghai Yuanqi Biopharmaceutical Company, Ltd., Shanghai, China) (9). The copy of the NUP98-HOXA9 fusion transcript was subsequently detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) using Leukemia Related Fusion Gene Fluorescent qPCR Detection kit (Shanghai Shenyou Bio Technology Co., Ltd., Shanghai, China) with a NUP98-HOXA9/Abelson tyrosine-protein kinase 1 (ABL1) ratio of 0.71. The sequences of primers were designed as previously reported (10). PCR was performed on an ABI PRISM 7000 machine (Applied Biosystems; Thermo Fisher Scientific, Inc., Waltham, MA, USA) under the conditions as previously described (11) and quantified using the standard curve method (12). The experiment was repeated independently three times.

To date, four types of fusion transcripts have been reported, which appear to result from alternative splicing of NUP98 and HOXA9 and each fusion is in-frame (10). In this case, detection of the NUP98-HOXA9 breakpoint was performed by RT-PCR and sequencing as previously described (10). Analysis demonstrated a type I fusion transcript, which means that the breakpoint of NUP98-HOXA9 is between NUP98 exon 12 and HOXA9 exon 1b (10) (Fig. 3). Subsequently, 1 point mutation in NRAS and 2 point mutations in WT1 were detected using the next generation sequencing (NGS)-based multigene mutational screening system on an Ion-Torrent Personal Genome machine (Thermo Fisher Scientific, Inc.) (13). The gene screening profile and results are summarized in Table I.

Table I.

Leukemia-associated fusion genes and mutation-screened genes.

Table I.

Leukemia-associated fusion genes and mutation-screened genes.

Fusion and mutation gene screening profile
Fusion genes for screening
  AML1-ETO, AML1-MDS1/EV11, AML1-MTG16, BCR-ABL, CBFβ-MYH11, DEK-CAN, E2A-HLF, E2A-PBX1, ETV6-PDGFRA, FIP1L1-PDGFRA, MLL-AF4, MLL-AF9, NPM-MLF, PML-RARα, PLZF-RARα, SET-CAN, SIL-TAL1, STAT5b-RARα, TEL-ABL, TEL-JAK2, TEL-PDGFRβ, TLS-ERG, MLL-(AF6, AF10, AF17, AF1q, AF1p, AFX, ELL, ENL, SEPT6), NUP98-(HOXA13, HOXA11, HOXA9a, HOXC11, HOXD13, PMX1), (NPM, FIP1L1, PRKAR1A, NUMA1)-RARα
Genes for mutational detection
  AKT3, ASXL1, ATRX, BCOR, CBL, CCND1, CDKN2A, CEBPA, CREBBP, CSF1R, CSF3R, DNMT3A, EP300, ETV6, EZH2, FLT3, GATA2, IDH1, IDH2, IKZF1, JAK3, KIT, KRAS, MLH1, MPL, NOTCH1, NOTCH2, NPM1, NRASb, PHF6, PTPN11, RAD21, RUNX1, SETBP1, SRSF2, STAG2, STAT3, TET2, TP53, U2AF1, WT1c, ZRSR2

a NUP98-HOXA9-positive

b NRAS point mutation (exon 2, c.38G>A, p.Gly13Asp)

c WT1 point mutations (exon 7, c.1109G>C, p.Arg370Pro; exon 7, c.1144_1145insTCGG, p.Ala382fs). NUP98, nucleoporin 98; HOXA13, homeobox A9; NRAS, neuroblastoma rat sarcoma viral oncogene homolog; WT1, Wilms tumor 1.

Following diagnosis, the patient was treated with induction chemotherapy for 7 days using a combination of daunorubicin (80 mg/day for 3 days) and cytosine arabinoside (Ara-c; 200 mg/day for 7 days). After 3 weeks, the patient demonstrated hematological complete remission (CR), with 1.6% myeloblasts in the BM smear. The repeat RT-qPCR revealed a decrease in the NUP98-HOXA9/ABL1 ratio to 0.0076. Subsequently, the patient underwent 4 courses of consolidation chemotherapy with a high-dose Ara-c (HD-Ara-c) regimen (1.5 g/m2 q12 h on days 1, 3 and 5) at the same time as seeking an HLA-matched sibling or unrelated donor for allogenic-hematopoietic stem cell transplantation (HSCT). During this period, repeated BM and fusion examinations were performed 4 times. Hematological CR was observed in the 4 tests, however, the NUP98-HOXA9/ABL1 ratio remained detectable, with a lowest level of 0.0023.

Due to the failure to find an HLA-matched donor, it was suggested that the patient should receive autogenic-HSCT. However, following treatment with a further course of HD-Ara-c chemotherapy (1.5 g/m2 q12 h on days 1, 3 and 5) as a hematopoietic stem cell mobilization method, the disease reoccurred 11 months after the original diagnosis (June 2016). A BM smear indicated 46% myeloblasts, with the presence of micromegakaryocytes. The NUP98-HOXA9/ABL1 ratio was markedly increased to 0.079. Cytogenetic analysis and NGS demonstrated the same 46,XY,t(7;11)(p15;p15) karyotype and NRAS and WT1 mutations as the diagnostic tests, which indicates that there was no genetic evolution of the tumor. The disease became refractory, and the patient contracted severe pneumonia and respiratory failure, which caused mortality 1 month after recurrence.

Discussion

The role of NUP98-HOXA9 in leukemogenesis has been repeatedly demonstrated in the past two decades. The transforming potential for the development of leukemia may require other cooperative factors (10). Using transgenic mouse models, breakpoint cluster region (BCR)/ABL was observed to interact with NUP98-HOXA9 and promote AML development in CML progression (14). Using retroviral insertional mutagenesis, myeloid ecotropic viral integration site 1 homolog (Meis1), dynein axonemal light chain 4, Fc fragment of IgG receptor IIb, Fc receptor-like and Con1 glycoprotein were identified as co-factors that associate with NUP98-HOXA9 in myeloid leukemogenesis (15). The most frequent of these associations was with Meis1, and the interaction between Meis1 and NUP98-HOXA9 reduces the latency of AML development (16). Furthermore, microarray analysis of human CD34-positive hematopoietic cells also identified oncogenes that may potentially associate with the NUP98-HOXA9, including the homeobox transcription factors, FLT3, c-kit proto-oncogene and WT1 (17). The following four gene mutations, FLT3-internal duplications, NRAS, Kirsten rat sarcoma viral oncogene homolog (KRAS) and WT1 have been identified with the NUP98-HOXA9 in a total of 7 patients with AML and t(7;11) (10,18). KRAS and WT1 mutations demonstrated a significantly association with the NUP98-HOXA9 fusion gene (10). In the present study, a novel gene mutation group of NRAS and WT1 mutations was found to coexist with NUP98-HOXA9 at the initial diagnosis and at the relapse. As RAS and WT1 mutations are key components of the proliferative drive of AML (19,20), taken together with previous studies, the present study indicates these gene alterations may cooperate with NUP98-HOXA9 in promoting the development and relapse of leukemia.

Previous studies have established that patients with AML and t(7;11) have distinct epidemic and clinical characteristics. A literature review was performed using the PubMed database to search for the keywords ‘t(7;11)’, ‘acute myeloid leukemia’ (www.ncbi.nlm.nih.gov/pubmed/?term=t(7%3B11)+AND+ acute+myeloid+leukemia). Patient characteristics including gender, age (years) and French-American-British (FAB) classification subtypes (2) were recorded. To date, only 57 adult patients (7,10,18,2134) were recorded with de novo AML in the literature. The majority of the case studies (98.2%, 56/57) originated in Japan or China. The incidence rate of t(7;11) on de novo adult patients with AML was low at 0.68–2.23% (10,18,27,28,30). For these 57 patients, the median onset age was 36 years (range, 31–38 years) and 63.1% (36/57) were female. A total of 54 patients had FAB classification subtypes and 68.5% (37/54) of these patients has an M2-subtype (7,10,18,2134). These characteristics of being predominantly younger, being female and having an M2-subtype were statistically proven in a large sample study by Chou et al (10) by comparing 11 patients carrying t(7;11) translocations with another 482 adult patients with AML. The case identified by the present study was also of a patient of a young age and with an M2 subtype, which is concordant with these previous studies.

In the present study, marked concomitant abnormal myelopoiesis, including dysplastic erythrocytopoiesis and megakaryocytopoiesis, were established from a de novo bone morrow smear. Similarly, previous studies have identified t(7;11) in AML patients to be associated with myelodysplasia (MDS) or myeloid maturation (10,21,27,30,31). A transgenic murine model with the NUP98-HOXA9 fusion gene also acquired myeloproliferative disorder and subsequently developed AML (35). Although it is primarily observed in AML, t(7;11) has also been identified in patients with MDS (36), CML (5,37) and juvenile myelomonocytic leukemia (8). Therefore, the present study is concordant with the hypothesis that t(7;11) may affect multipotent myeloid stem and progenitor cells.

Patients with AML that have t(7;11) exhibit aggressive clinical progression and a poor prognosis. Using multivariate analysis, Chou et al established that this translocation was an independent factor for predicting a decrease in overall, relapse-free and disease-free survival rates (10). The patient presented in the present study achieved a CR following 2 courses of induction chemotherapy, but relapsed and became refractory after a short duration, with 12 months of overall survival. Via the monitoring of NUP98-HOXA9 transcript copies, fusion protein signals were consistently detected throughout treatment courses and after a hematological CR was achieved. This suggested that residual leukemia cells were not eradicated. This is concordant with the majority of cases reported by Chou et al (10), indicating that this fusion gene may promote survival signals and may be a useful prognostic marker for monitoring minimal residual disease.

The present study and literature review demonstrated that patients with AML and t(7;11) translocation are rare, and may have distinct genetic, molecular and clinical characteristics. NRAS and WT1 gene mutations are able to coexist with NUP98-HOXA9 in a number of patients, indicating that there may be an association between NUP98-HOXA9 and leukemogenesis. Patients with AML and t(7;11) are predominantly from Asia, typically young, female and have a FAB M2-subtype, and may present with concomitant BM dyspoiesis. These patients exhibit an aggressive clinical course and have a poor prognosis. Further studies are required with regard to this distinct entity.

Acknowledgements

The present study was supported by the Science and Technology Project of Henan Province (grant no. 162102310338).

References

1 

Saultz JN and Garzon R: Acute myeloid leukemia: A concise review. J Clin Med. 5:pii: E33. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR and Sultan C: Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. Brit J Haemat. 33:451–458. 1976. View Article : Google Scholar : PubMed/NCBI

3 

Byrd JC, Mrózek K, Dodge RK, Carroll AJ, Edwards CG, Arthur DC, Pettenati MJ, Patil SR, Rao KW, Watson MS, et al: Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: Results from Cancer and Leukemia Group B (CALGB 8461). Blood. 100:4325–4336. 2002. View Article : Google Scholar : PubMed/NCBI

4 

De Kouchkovsky I and Abdul-Hay M: Acute myeloid leukemia: A comprehensive review and 2016 update. Blood Cancer J. 6:e4412016. View Article : Google Scholar : PubMed/NCBI

5 

Tomiyasu T, Sasaki M, Kondo K and Okada M: Chromosome banding studies in 106 cases of chronic myelogenous leukemia. Jinrui Idengaku Zasshi. 27:243–258. 1982. View Article : Google Scholar : PubMed/NCBI

6 

Borrow J, Shearman AM, Stanton VP Jr, Becher R, Collins T, Williams AJ, Dubé I, Katz F, Kwong YL, Morris C, et al: The t(7;11)(p15;p15) translocation in acute myeloid leukaemia fuses the genes for nucleoporin NUP98 and class I homeoprotein HOXA9. Nat Genet. 12:159–167. 1996. View Article : Google Scholar : PubMed/NCBI

7 

Taketani T, Taki T, Ono R, Kobayashi Y, Ida K and Hayashi Y: The chromosome translocation t(7;11)(p15;p15) in acute myeloid leukemia results in fusion of the NUP98 gene with a HOXA cluster gene, HOXA13, but not HOXA9. Genes Chromosomes Cancer. 34:437–443. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Mizoguchi Y, Fujita N, Taki T, Hayashi Y and Hamamoto K: Juvenile myelomonocytic leukemia with t(7;11)(p15;p15) and NUP98-HOXA11 fusion. Am J Hematol. 84:295–297. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Zhang J, Mao S, Su S, Wei L, Li T and Liu Y: Detection of concurrent TEL-AML1/TEL-ABL fusion genes in a patient with B-acute lymphoblastic leukemia using a multi-fusion gene qRT-PCR screening method. Pathol Int. 66:475–477. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Chou WC, Chen CY, Hou HA, Lin LI, Tang JL, Yao M, Tsay W, Ko BS, Wu SJ, Huang SY, et al: Acute myeloid leukemia bearing t(7;11)(p15;p15) is a distinct cytogenetic entity with poor outcome and a distinct mutation profile: Comparative analysis of 493 adult patients. Leukemia. 23:1303–1310. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Lyu X, Yang J, Wang X, Hu J, Liu B, Zhao Y, Guo Z, Liu B, Fan R and Song Y: A novel BCR-ABL1 fusion gene identified by next-generation sequencing in chronic myeloid leukemia. Mol Cytogenet. 9:472016. View Article : Google Scholar : PubMed/NCBI

12 

Bookout AL and Mangelsdorf DJ: Quantitative real-time PCR protocol for analysis of nuclear receptor signaling pathways. Nucl Recept Signal. 1:e0122003. View Article : Google Scholar : PubMed/NCBI

13 

Kanagal-Shamanna R, Portier BP, Singh RR, Routbort MJ, Aldape KD, Handal BA, Rahimi H, Reddy NG, Barkoh BA, Mishra BM, et al: Next-generation sequencing-based multi-gene mutation profiling of solid tumors using fine needle aspiration samples: Promises and challenges for routine clinical diagnostics. Mod Pathol. 27:314–327. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Dash AB, Williams IR, Kutok JL, Tomasson MH, Anastasiadou E, Lindahl K, Li S, Van Etten RA, Borrow J, Housman D, et al: A murine model of CML blast crisis induced by cooperation between BCR/ABL and NUP98/HOXA9. Proc Natl Acad Sci USA. 99:7622–7627. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Iwasaki M, Kuwata T, Yamazaki Y, Jenkins NA, Copeland NG, Osato M, Ito Y, Kroon E, Sauvageau G and Nakamura T: Identification of cooperative genes for NUP98-HOXA9 in myeloid leukemogenesis using a mouse model. Blood. 105:784–793. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Kroon E, Krosl J, Thorsteinsdottir U, Baban S, Buchberg AM and Sauvageau G: Hoxa9 transforms primary bone marrow cells through specific collaboration with Meis1a but not Pbx1b. EMBO J. 17:3714–3725. 1998. View Article : Google Scholar : PubMed/NCBI

17 

Takeda A, Goolsby C and Yaseen NR: NUP98-HOXA9 induces long-term proliferation and blocks differentiation of primary human CD34+ hematopoietic cells. Cancer Res. 66:6628–6637. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Wei S, Wang S, Qiu S, Qi J, Mi Y, Lin D, Zhou C, Liu B, Li W, Wang Y, et al: Clinical and laboratory studies of 17 patients with acute myeloid leukemia harboring t(7;11)(p15;p15) translocation. Leuk Res. 37:1010–1015. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Bowen DT, Frew ME, Hills R, Gale RE, Wheatley K, Groves MJ, Langabeer SE, Kottaridis PD, Moorman AV, Burnett AK and Linch DC: RAS mutation in acute myeloid leukemia is associated with distinct cytogenetic subgroups but does not influence outcome in patients younger than 60 years. Blood. 106:2113–2119. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Rampal R and Figueroa ME: Wilms tumor 1 mutations in the pathogenesis of acute myeloid leukemia. Haematologica. 101:672–679. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Sato Y, Abe S, Mise K, Sasaki M, Kamada N, Kouda K, Musashi M, Saburi Y, Horikoshi A and Minami Y: Reciprocal translocation involving the short arms of chromosomes 7 and 11, t(7p-; 11p+), associated with myeloid leukemia with maturation. Blood. 70:1654–1658. 1987.PubMed/NCBI

22 

Mise K, Abe S, Sato Y, Miura Y and Sasaki M: Localization of c-Ha-ras-1 oncogene in the t(7p-; 11p+) abnormality of two cases with myeloid leukemia. Cancer Genet Cytogenet. 29:191–199. 1987. View Article : Google Scholar : PubMed/NCBI

23 

Takahashi E, Kaneko Y, Ishihara T, Minamihisamatsu M, Murata M and Hori T: A new rare distamycin A-inducible fragile site, fra(11)(p15. 1), found in two acute nonlymphocytic leukemia (ANLL) patients with t(7;11)(p15-p13;p15). Hum Genet. 80:124–126. 1988. View Article : Google Scholar : PubMed/NCBI

24 

Morris CM, Fitzgerald PH, Kennedy MA, Hollings PE, Garry M and Corbett GM: HRAS1 and INS genes are relocated but not structurally altered as a result of the t(7;11)(p15;p15) in a clone from a patient with acute myeloid leukaemia (M4). Br J Haematol. 71:481–486. 1989. View Article : Google Scholar : PubMed/NCBI

25 

Fujimura T, Ohyashiki K, Ohyashiki JH, Kawakubo K, Iwabuchi A, Kodama A and Toyama K: Two additional cases of acute myeloid leukemia with t(7;11)(p15;p15) having low neutrophil alkaline phosphatase scores. Cancer Genet Cytogenet. 68:143–146. 1993. View Article : Google Scholar : PubMed/NCBI

26 

Abe A, Tanimoto M, Towatari M, Matsuoka A, Kitaori K, Kato H, Toyozumi H, Takeo T, Adachi K and Emi N: Acute myeloblastic leukemia (M2) with translocation (7;11) followed by marked eosinophilia and additional abnormalities of chromosome 5. Cancer Genet Cytogenet. 83:37–41. 1995. View Article : Google Scholar : PubMed/NCBI

27 

Huang SY, Tang JL, Liang YJ, Wang CH, Chen YC and Tien HF: Clinical, haematological and molecular studies in patients with chromosome translocation t(7;11): A study of four Chinese patients in Taiwan. Br J Haematol. 96:682–687. 1997. View Article : Google Scholar : PubMed/NCBI

28 

Kwong YL and Pang A: Low frequency of rearrangements of the homeobox gene HOXA9/t(7;11) in adult acute myeloid leukemia. Genes Chromosomes Cancer. 25:70–74. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Kawakami K, Miyanishi S, Nishii K, Usui E, Murata T, Shinsato I and Shiku H: A case of acute myeloid leukemia with t(7;11)(p15;p15) mimicking myeloid crisis of chronic myelogenous leukemia. Int J Hematol. 76:80–83. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Kwong YL and Chan TK: Translocation (7;11)(p15;p15) in acute myeloid leukemia M2: Association with trilineage myelodysplasia and giant dysplastic myeloid cells. Am J Hematol. 47:62–64. 1994. View Article : Google Scholar : PubMed/NCBI

31 

Inaba T, Shimazaki C, Yoneyama S, Hirai H, Kikuta T, Sumikuma T, Sudo Y, Yamagata N, Ashihara E, Goto H, et al: t(7;11) and trilineage myelodysplasia in acute myelomonocytic leukemia. Cancer Genet Cytogenet. 86:72–75. 1996. View Article : Google Scholar : PubMed/NCBI

32 

Ohyashiki K: Nonrandom cytogenetic changes in human acute leukemia and their clinical implications. Cancer Genet Cytogenet. 11:453–471. 1984. View Article : Google Scholar : PubMed/NCBI

33 

Okada M, Mizoguchi H, Kubota K and Nomura Y: Quantitative analysis of chromosomal G-bands in human hematopoietic disorders by methotrexate synchronization technique. Cancer Genet Cytogenet. 13:225–237. 1984. View Article : Google Scholar : PubMed/NCBI

34 

Ishihara T, Minamihisamatsu M and Yokoyama Y: Translocation t(7;11)(p13;p15) in acute nonlymphocytic leukemia. Cancer Genet Cytogenet. 19:363–364. 1986. View Article : Google Scholar : PubMed/NCBI

35 

Kroon E, Thorsteinsdottir U, Mayotte N, Nakamura T and Sauvageau G: NUP98-HOXA9 expression in hemopoietic stem cells induces chronic and acute myeloid leukemias in mice. EMBO J. 20:350–361. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Hatano Y, Miura I, Nakamura T, Yamazaki Y, Takahashi N and Miura AB: Molecular heterogeneity of the NUP98/HOXA9 fusion transcript in myelodysplastic syndromes associated with t(7;11)(p15;p15). Br J Haematol. 107:600–604. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Ahuja HG, Popplewell L, Tcheurekdjian L and Slovak ML: NUP98 gene rearrangements and the clonal evolution of chronic myelo-genous leukemia. Genes Chromosomes Cancer. 30:410–415. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang J, Lyu X, Zhu X, Meng X, Zuo W, Ai H and Deng M: Chromosome t(7;11)(p15;p15) translocation in acute myeloid leukemia coexisting with multilineage dyspoiesis and mutations in NRAS and WT1: A case report and literature review. Oncol Lett 13: 3066-3070, 2017
APA
Yang, J., Lyu, X., Zhu, X., Meng, X., Zuo, W., Ai, H., & Deng, M. (2017). Chromosome t(7;11)(p15;p15) translocation in acute myeloid leukemia coexisting with multilineage dyspoiesis and mutations in NRAS and WT1: A case report and literature review. Oncology Letters, 13, 3066-3070. https://doi.org/10.3892/ol.2017.5823
MLA
Yang, J., Lyu, X., Zhu, X., Meng, X., Zuo, W., Ai, H., Deng, M."Chromosome t(7;11)(p15;p15) translocation in acute myeloid leukemia coexisting with multilineage dyspoiesis and mutations in NRAS and WT1: A case report and literature review". Oncology Letters 13.5 (2017): 3066-3070.
Chicago
Yang, J., Lyu, X., Zhu, X., Meng, X., Zuo, W., Ai, H., Deng, M."Chromosome t(7;11)(p15;p15) translocation in acute myeloid leukemia coexisting with multilineage dyspoiesis and mutations in NRAS and WT1: A case report and literature review". Oncology Letters 13, no. 5 (2017): 3066-3070. https://doi.org/10.3892/ol.2017.5823