Open Access

Coexistence of p210BCR‑ABL and CBFβ‑MYH11 fusion genes in myeloid leukemia: A report of 4 cases

  • Authors:
    • Yuan‑Yuan Wang
    • Wen‑Jing Ding
    • Feng Jiang
    • Zi‑Xing Chen
    • Jian‑Nong Cen
    • Xiao‑Fei Qi
    • Jian‑Ying Liang
    • Dan‑Dan Liu
    • Jin‑Lan Pan
    • Su‑Ning Chen
  • View Affiliations

  • Published online on: August 24, 2017     https://doi.org/10.3892/ol.2017.6812
  • Pages: 5171-5178
  • Copyright: © Wang 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

Numerous acquired molecular and cytogenetic abnormalities are strongly associated with hematological malignancies. The breakpoint cluster region‑ABL proto‑oncogene 1 (BCR‑ABL) rearrangement leads to a p210 chimeric protein in typical chronic myeloid leukemia (CML), whereas 17‑25% of patients with acute lymphocytic leukemia and 0.9‑3% patients with de novo acute myeloid leukemia (AML) carry a p190BCR‑ABL fusion protein. Cases of patients with AML/CML carrying two specific primary molecular changes, BCR‑ABL and core binding factor‑β‑myosin heavy chain 11 (CBFβ‑MYH11) fusion genes have been rarely reported. The present study aimed to understand the nature and mechanism of this particular type of leukemia through case reports and literature review. A total of four patients who were diagnosed as AML/CML with BCR‑ABL and CBFβ‑MYH11 fusion genes in the First Affiliated Hospital of Soochow University (Suzhou, China) between January 2004 and December 2012 were examined. Morphological analysis of bone marrow cells, flow cytometry, quantitative polymerase chain reactionof p210BCR‑ABL and CBFβ‑MYH11 transcripts as well as cytogenetic and fluorescence in situ hybridization analyses were performed. A total of 4 patients who exhibited fusion of p210BCR‑ABL and CBFβ‑MYH11 were identified. A single patient (case 1) was first diagnosed CML‑acute phase (AP), which progressed rapidly to CML‑blast crisis (BC), and three patients (cases 2, 3 and 4) were diagnosed with AML with bone marrow eosinophilia at first presentation with no evidence of previous onset of CML. All cases achieve dremission following conventional chemotherapy/hematological stem cell transplantation combined with the inhibitor of tyrosine kinase (TKI) maintenance therapy. The patients with CML carrying and expressing BCR‑ABL and CBFβ‑MYH11 fusion genes appeared more likely to rapidly progress to AP or BC. Therefore, the product of the CBFβ‑MYH11 fusion gene may serve an important role in the transformation of CML. The co‑expression of p210BCR‑ABL and CBFβ‑MYH11 fusion genes in myeloid leukemia may be a molecular event occurring not only during the development of CML, but also in AML.

Introduction

Numerous acquired molecular abnormalities and cytogenetic abnormalities are strongly associated with hematological malignancies (1). Core binding factors (CBFs) are a family of heterodimeric transcriptional regulators containing CBFβ (2). The inversion of chromosome 16, fusion of CBFβ gene at 16q22 with the smooth muscle myosin heavy chain (MYH11) gene at 16p13 region, forms the CBFβ-MYH11 fusion gene, inhibiting the differentiation of hematopoietic cells by altering transcriptional regulation (3,4). Inv(16)(p13q22) or t(16;16)(p13q22) is a recurrent genetic abnormality, which fuses the core binding factor-β (CBFβ) gene to the myosin heavy chain 11 (MYH11) gene. Inv(16)(p13q22) or t(16;16)(p13q22) has been recognized as a feature of acute myelomonocytic leukemia with abnormal eosinophils by the 2008 World Health Organization classification and was designated as M4EO in the French-America-British cooperative group classification (5). Patients with inv(16)(p13q22) or t(16;16)(p13q22) mutations have been identified to exhibit a more favorable clinical course, an improved response to chemotherapy/stem cell transplantation, and longer remission and survival in this AML subtype compared to those without inv(16)(p13q22) or t(16;16)(p13q22)(2). First described by Nowell and Hungerford as a minute chromosome in patients with granulocytic leukemia, the Philadelphia (Ph) chromosome is a result of reciprocal translocation involving the long arms of chromosomes 9 and 22 (6,7). This translocation results in the formation of a hybrid breakpoint cluster region-ABL proto-oncogene 1 (BCR-ABL) gene, which codes for a protein with enhanced tyrosine kinase activity (8). It has been demonstrated that the BCR-ABL fusion gene is the hallmark of typical chronic myeloid leukemia (CML). Cases of CML were additionally classified as chronic phase (CP), accelerated phase (AP) or blast crisis (BC) according to criteria of WHO classification (9). The BCR-ABL rearrangement leads to a p210 chimeric protein in typical CML, whereas 17–25% of patients with acute lymphocytic leukemia and 0.9–3% patients with de novo acute myeloid leukemia (AML) possess a p190BCR-ABL fusion protein (1,10). The present study aimed to understand the nature and mechanism of this particular type of leukemia through case reports and literature review. A total of 4 patients who were diagnosed as AML/CML with BCR-ABL and CBFβ-MYH11 fusion genes at the First Affiliated Hospital of Soochow University between January 2004 and December 2012 were examined, with a detectable protein product of p210BCR-ABL, were included in the present study. The cases described in the present report may assist in understanding the entity and mechanism of this particular type of leukemia.

Case report

The clinical files of the First Affiliated Hospital of Soochow University were examined for patients who were diagnosed with acute/chronic leukemia with BCR-ABL and/or CBFβ-MYH11 fusion genes during January 2004 to December 2012. A total of four patients were identified with the CBFβ-MYH11 fusion gene concomitant with BCR-ABL expression. The patient samples were taken at different time points: Case 1 was taken in June 2010, case 2 in August 2010, case 3 in September 2011 and case 4 in September 2010. Bone marrow (BM) aspirates were collected into syringes containing media supplemented with heparin. Therefore, the present report summarizes the clinical and laboratory features of four patients with the coexistence of BCR-ABL and CBFβ-MYH11 fusion genes, including three males and one female with a median age of 29 years (range, 18–40; Tables I and II). All patients exhibited constitutional symptoms, including progressive fatigue, localized or diffuse pain and low-grade fever. Physical examination revealed splenomegaly in cases 1 and 4 and scattered petechiae and ecchymosis on the trunk and lower extremities in cases 3 and 4. The peripheral blood count revealed anemia and thrombocytopenia in three cases. Morphology and cytochemical studies were performed on cells from the bone marrow aspirate smears stained with Wright's stain (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) for 15 min and myeloperoxidase respectively. For myeloperoxidase staining, slides had 10–15 drops 0.3% benzidine ethanol solution added to them, after 1 min, 10–15 drops of H2O2 solution were added for 5 min. Slides were rinsed and Wright's stain was added for 30 min, followed by another rinse (benzidine ethanol solutionand H2O2 solutionwere purchased from Shanghai Sun Biotech Co., Ltd., Shanghai, China). Images were captured using an optical microscope (OLYMPUS CX-31; Olympus Corporation, Tokyo, Japan) at magnifications, ×100 and ×1,000. Based on the morphology of the BM cells (Fig. 1), three cases were diagnosed with AML-M4 subtype with an abnormal eosinophil component (M4EO; cases 2, 3 and 4), and case 1 was diagnosed with CML-BC. As summarized in Table I, the percentage of blasts in bone marrow samples was 10–49%, and the eosinophil counts ranged from 4–14%. Immunophenotyping, the blasts of all patients were positive for cluster of differentiation (CD)13, CD33, CD117, CD34 and negative for B and T cell markers. The multiplex polymerase chain reaction (PCR) detected BCR-ABL and CBFβ-MYH11 transcripts in all 4 cases. Bone marrow mononuclear cells (BMNCs) were separated from bone marrow of patients by Ficoll-Hypaque (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Total RNA was extracted from BMNCs using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.). The RNA was pretreated with DNase and used for cDNA synthesis with random hexamers (Invitrogen; Thermo Fisher Scientific, Inc.). The primers were synthesized to order by Sangon Biotech Co., Ltd. (Shanghai, China) with the following sequences: BCL-ABL F, 5′-AGCATTCCGCTGACCATCA-3′; BCL-ABL R, 5′-ACTCAGACCCTGAGGCTCAAAG-3′; BCL-ABL P, 5′-6-carboxyfluorescein (6-FAM)-AAGCCCTTCAGCGGCCAGTAGCAT-carboxytetramethylrhodamine (TAMRA)-3′. CBFβ/MYH11 F, 5′-CATTAGCACAACAGGCCTTTGA-3′; CBFβ/MYH11 R, 5′-AGGGCCCGCTTGGACTT-3; CBFβ/MYH11 P, 5′-6-FAM-TCGCGTGTCCTTCTCCGAGCCT-TAMRA-3′. The quantitative (q)PCR conditions were as follows: 2 min at 50°C for denaturation, 10 min at 95°C, 15 sec at 95°C and 60 sec at 60°C, followed by 40 cycles of amplification. The relative quantification of CBFβ-MYH11 and BCR-ABL fusion genes was performed by the 2−ΔΔCq method (11). The reverse transcription (RT)-qPCR results demonstrated that the copy numbers of CBFβ-MYH11 and BCR-ABL fusion genes were >5,000 copies/104 ABL copies in three patients (cases 2, 3 and 4). By contrast, in case 1 only the BCR-ABL transcript was detected at similar levels (>5,000 copies/104 ABL copies). Cytogenetic analysis was performed on of the BM cells after 24 h culturing by the RBG (R-band by Brdu using Giemsa) technique (12). At least 10 metaphases were examined for each case. Karyotypes were described according to the standard 2009 International System for Human Cytogenetic Nomenclature criteria (13,14).

Table I.

Summary of clinical features and morphology data of bone marrow samples.

Table I.

Summary of clinical features and morphology data of bone marrow samples.

Peripheral bloodBone marrow, %


Case no.SexAge, yearsSymptomsSignsWBC, ×109/lPLT, ×109/lHB, g/lBlastEosinophilsInitial diagnosisOutcome
1M40Body painSplenomegaly191.8389.0119.010.05.5CML-BCSuccumbed to disease
2M36NASplenomegaly   50.6   50.0   60.049.0NAAML-M4EOSuccumbed to disease
3F18FeverPetechiae, ecchymosis   72.2   22.0   61.046.04.0AML-M4EOSuccumbed to disease
4M31FatigueSplenomegaly, petechiae, ecchymosis   67.5   21.0   99.043.014.0AML-M4EOAlive

[i] NA, not available; M, male; F, female; WBC, white blood cell count; PLT, platelet count; HB, hemoglobin; AML-M4EO, AML with bone marrow eosinophilia; CML-BC; chronic myeloid leukemia-blast crisis.

Table II.

Summary of the molecular and cytogenetic findings.

Table II.

Summary of the molecular and cytogenetic findings.

Case no.Multiplex PCRqPCR (copies/104 ABL copies)FISH (%)Cytogenetic data
1 p210BCR-ABL CBFβ-MYH11BCR-ABL (9958)BCR-ABL(94) CBFβ-MYH11 (47)46,X,der(Y),t(Y;1) (q12;q23), t(9;22)(q34;q11),t(9;11)(p22,q23)[4]/47,idem,+18[3]/ 51, idem,+4,+8,+12, +21,+mar?der(16) inv(16) (p13;q22)[4]/46,XY[2]
2 p210BCR-ABL CBFβ-MYH11 BCR-ABL(8158) CBFβ-MYH11(5610)BCR-ABL(92) CBFβ-MYH11(95) 45,X,-Y,t(9;22)t(16;16)(p13;q22)[1]/46,idem,+ph[9]
3 p210BCR-ABL CBFβ-MYH11 BCR-ABL(9496) CBFβ-MYH11 (12602)BCR-ABL(85) CBFβ-MYH11(91) 46,XX,t(9;22)(q34;q11) inv(16)(p13;q22)[9]/46,XX[2]
4 p210BCR-ABL CBFβ-MYH11 BCR-ABL(5399) CBFβ-MYH11(10473)BCR-ABL(46) CBFβ-MYH11(89) 48,XY,der(8)t(8;10)(p23;q25), der(10),t(8;10),t(10;16)(p13;q22), der(16),inv(16)(p13;q22)t(10;16)[4]/46,XY,idem,t(9;22)(q34;q11)

[i] qPCR, quantitative polymerase chain reaction, CBFβ-MYH11, core binding factor-β-myosin heavy chain 11; BCR-ABL, breakpoint cluster region-ABL proto-oncogene 1.

Fluorescence in situ hybridization (FISH) analysis was performed on interphase nuclei and metaphases of the BM cells to confirm the presence of the BCR-ABL and CBFβ-MYH11-fused genes. The BCR-ABL dual-color, double fusion translocation probes and the CBFβ dual color break-apart rearrangement probes were used in the FISH analysis according to the manufacturer's protocol (Beijing GP Medical Technologies, Ltd., Beijing, China). Bone marrow cells were cultured in RPMI-1640 medium (Hyclone; GE Healthcare Life Sciences, Logan, UT, USA) supplemented with 10% bovine fetal serum (Bovogen Biologicals, Keilor East, VIC, Australia). The specimens were placed on the slide, precipitated in ethanol, then ageing in 2X 0.3 M trisodium citrate, (SSC; pH 7.0) for 30 min at 37°C. Samples were then dehydrated using increasing gradients (70, 85 and 100%) of ethanol at room temperature for 2 min, and then dried at room temperature. A total of 5 µg probes were mixed with the denatured control probe, denatured for 5 min at 75°C, then hybridized overnight at 37°C. Slides were then washed in 0.4X SSC at 72°C for 2 min and rinsed in 2X SSC at room temperature for 2 min, then in 70% ethanol for 2 min. Finally, slides were counterstained with 1 mg/ml 10 µl DAPI for 15 min. Slides were then screened using an fluorescence microscope DMRB (OLYMPUS BX-51; Olympus Corporation, Tokyo, Japan) at magnification, ×1,000. A minimum of 500 interphase cells were observed. Images were captured and analyzed using the VideoTesT (VT-FISH2.1, Nature Gene Corporation, Medford, NJ, USA). FISH analysis also demonstrated the rearrangement of CBFβ-MYH11 and BCR-ABL fusion signals in all cases, and the percentage of the two fusion genes was >45% (Fig. 2). Cytogenetic analysis of all patients revealed inv(16)(p13q22) or t(16;16)(p13q22) and Philadelphia chromosome (Fig. 3). In addition, expression of trisomy 8 was detected in case 1, and the loss of Y chromosome was detected in case 2. Based on physical examinations, hematological data, BM morphology and cytogenetic and molecular analyses, a single patient (case 1) was first diagnosed with CML-acute phase (AP), which rapidly progressed to CML-BC, and three patients (cases 2, 3 and 4) were diagnosed with AML-M4EO at first presentation with no evidence of previous onset of CML. Patients were treated with various therapeutics, including idarubicin (cases 1–4), cytarabine (cases 1–4), daunorubicin (cases 2 and 3) and tyrosine kinase inhibitors (TKI; cases 1 and 4). A total of three patients achieved complete remission following first round of chemotherapy (cases 1, 3 and 4). Case 2 succumbed to respiratory failure following a pulmonary infection. Following the fourth round of chemotherapy, case 3 abandoned treatment. A total of two patients received unrelated donor peripheral hematopoietic stem cell and autologous peripheral hematopoietic stem cell (case 4) transplantations. Case 1 succumbed to Graft vs. host disease (GVHD), and case 4 was in good condition for 24 months following transplantation. The abnormal eosinophils observed in case 1 and case 2 presented as excessive eosinophils in BM samples. In all cases, the blasts were of myeloid lineage and there was evidence of monocytic differentiation, as demonstrated by immunophenotypic analysis, in cases 3 and 4. Peripheral blood monocytosis was exhibited in all cases. Patient data and protocol were approved by the Ethics Committee at the First Affiliated Hospital of Soochow University. Informed written consent was obtained from each patient.

Discussion

The present report describes four patients with CBFβ-MYH11 and BCR-ABL fusion genes. The inv(16) genetic abnormality was detected in case 1 at the time of blastic transformation. Although case 2 was diagnosed with AML-M4EO based on the BM cell morphology, the patient (case 2) should have been diagnosed CML-BP for the following reasons: i) The patient displayed clinical characteristic of CML, including leukocytosis and massive splenomegaly; ii) the Ph chromosome, which typically occur in CML-BC, was detected in case 2; the rearrangement of CBFβ in addition to BCR-ABL had been detected by FISH in the BM samples of case 3 and 4. Additionally, karyotype analysis of the BM samples from case 4 at diagnosis also confirmed that the chromosome translocation inv(16) coexisted with the Phchromosome, suggesting that the translocation of inv(16) occured following the Ph chromosome translocation, therefore this case should have been diagnosed as de novo Ph(+) AML-M4EO.

The morphological features of CML-BC with inv(16) in the present study closely resembled those of AML-M4EO, as described in the WHO classification (15). To the best of our knowledge, the coexistence of the BCR-ABL and CBFβ-MYH11 fusion genes are very rare in CML and de novo AML, with only 34 cases having been identified at present (Tables III and IV) (1,1634), including the cases described in the present study. Extramedullary disease occurs in a wide age spectrum, ranging from 9 to 78 years (17). CBFβ-MYH11 and BCR-ABL affects males and females, with an increased risk in males (17). It is suggested that males may be more likely to exhibit the simultaneous expression of CBFβ-MYH11 and BCR-ABL compared with females.

Table III.

Summary of identified patients with acute myeloid leukemia with BCR-ABL and core binding factor-β-myosin heavy chain 11.

Table III.

Summary of identified patients with acute myeloid leukemia with BCR-ABL and core binding factor-β-myosin heavy chain 11.

Author, yearAgeSexBlasts, % (bone marrow)BCR-ABL statusCytogenetic findingsClinical outcome, time from diagnosis (months)(Refs.)
Preudhomme et al, 199264M32p190 46,XY,inv(16)(p13q22),t(9;22)(q34;q11)[30]Alive, 12(18)
Siddiqui et al, 200223M21ND 46,XY,t(9;22)(q34;q11.2)inv(16)(p13q22)Alive, 36(19)
Li and Hayhoe, 198839M>30ND 46,XY(9%)/47,XY.-18.+22,inv(16)(p13q22),del(20)(p12p13) del(20)(q12q13),t(9;22)(q34;q11),der(16)t(16;?18) (q24;q21),+mar(91%)Alive, 38(20)
Wu et al, 200644M44.5p190 46,XY,t(9;22)(q34.1;q11.2)inv(16)(p13.1q22)Succumbed to disease(16)
Miura et al, 199440M36M-bcr(−) 46,XY,inv(16)(p13q22)[17]/46,XY,idem,t(9;22)(q34;q11)[3]Alive, 27(21)
Secker-Walker et al, 1992  9FNDp190 46,XX,inv(16)(p13q22)[21]/46,XX,t(9;22)(q34;q11)inv(16) (p13q22)[8]/ 46,XX[10]Molecular remission, 1 month(22)
Svaldi et al, 200140FNAp190 46,XX,inv(16)[4]/46,idem(9,22)[18]NA(23)
Tirado et al, 201013M92ND 46,XY,inv(16)(p13.1q22)[2]/46,idem,del(7) (q22q32)[16]/46,idem,t(9;22;19)Alive, 10(24)
Cividin et al, 200438F41p190 46,XX[22]/46,XX,inv(16)(p13q22)[1]/46,XX,idem,t(9;22) (q34;q11)[25]/46,XX,t(2;9;22)(q32;q34;q11),inv(16)(p13q22)[23]Alive, 12(1)
Roth et al, 201130F  9p190 46,XX,t(9;22;17;19)(q34;q11.2;q25;p13.1),inv(16)(p13q22)[19]Alive, 80(25)
Roth et al, 201135M48p190 46,XY,der(16)inv(16)(p13q22)del(16)(p11.2p13.1)[2]/46,XY,idem,t(9;22)(q34;q11.2)[18]Alive, 17(25)
Bustamante et al, 201249M10-15p190 46,XX,inv(16)(p13.1q22)[5]/46,idem,t(9;22)(q34;q11.2)NA(26)

[i] Breakpoint cluster region-ABL proto-oncogene 1; M, male; F, female; ND, not done; NA, not available.

Table IV.

Summary of identified patients withchronic myeloid leukemia with BCR-ABL and core binding factor-β-myosin heavy chain 11.

Table IV.

Summary of identified patients withchronic myeloid leukemia with BCR-ABL and core binding factor-β-myosin heavy chain 11.

Author, yearAgeSexBlasts, % (bone marrow)BCR-ABL statusCytogenetic findingsClinical outcome, time from diagnosis(Refs.)
Wu et al, 200633M27p210 46,XY,t(9;22)(q34.1;q11.2)[18]/46,XY,idem,inv(16) (p13.1q22)[2]Succumbed following allo-BMT(16)
Wu et al, 200641M63p210 46,XY,t(9;22)(q34.1;q11.2),inv(16)(p13.1q22)[20]Alive, 4 years(16)
Wu et al, 200662F52p210 46,XX,t(9;22)(q34.1;q11.2,inv(16)(p13.1q22)[20]Succumbed to disease, 24 months(16)
Wu et al, 200621M  2p210 46,XY,t(9;22)(q34.1;q11.2)[17]/46,idem,inv(16) (p11.2q22)[3]Alive, 6 years(16)
Wu et al, 200644M10p210 46,XY,t(9;22)(q34.1;q11.2)[90%]46,XY,inv(16) (p13q22),t(9;22)(q34.1;q11.2)[1] 46,XY,t(6;16)(q22;q22),t(9;22)(q34.1;q11.2)[1]Succumbed to disease, 24 months(16)
Merzianu et al, 200543F30p210 46,XX,t(9;22)(q34;q11.2),inv(16)(p13q22)[20]Succumbed to disease, 3 months(27)
Merzianu et al, 200561F20p210 46,XX,t(9;22)(q34;q11.2)[3]/46,XX,t(9;22)(q34; q11.2),inv(16)(p13q22)[7]/ 47,XX,+8,t(9;22) (q34; q11.2)[2]/47,XX,+8,t(9;22) (q34;q11.2), inv(16) (p13q22)[4]/ 46,X,add(X)(p22.3),t(9;22) (q34;q11.2),del(12)(p11.2),inv(16)(p13q22)[1]/ 47,XX,t(9;22)(q34;q11.2),inv(16)(p13q22), +der(2)t(9;22)[1]/46,XX[2]Succumbed to disease, 7 months(27)
Merzianu et al, 200547M40p210 46,XY,t(9;22)(q34;q11),inv(16)(p13q22)[25]Succumbed to disease, 1 month(27)
Merzianu et al, 200536F70p2102 46,XX,t(9;22)(q34;q11.2),inv(16)(p13q22)[20]Succumbed to disease, 1 month(27)
Merzianu et al, 200548M20p210 46,XY,t(9;22)(q34;q11.2),inv(16)(p13q22)[6]/46,XY[14]Alive, 7 months(27)
Tsuboi et al, 200244M59p210 47,XY,t(9;22)(q34;q11)inv(16)(p13q22),+der(22) t(9;22)/48,idem,+8Succumbed to disease, 3 months(28)
Myint et al, 199729M90p210 46,XX,t(9;22)inv(16)(p13;q22)Succumbed to disease, 0 months(29)
Evers et al, 199239M25p210 46,XY,-2,-9,-12,-21,+der(9)t(9;?;22),+der(12) t(12;?2)(q24?p22),inv(16)Succumbed to disease, 10 months(30)
Asou et al, 199251M10p210 46,XY,t(9;22)(q34;q11),inv(16)(p13q22)Succumbed to disease, 3 months(31)
Enright et al, 199278M19p210 46,XY,t(9;22)(q34;q11.2),inv(16)(p13q22)Succumbed to disease, 3 months(32)
Heim et al, 199221M70p210 45,X,-Y,t(9;22)(q34.1;q11.2),inv(16)(p13.1q22)Alive, 30 months(33)
Colovic et al, 199858M18p210 46,XY,t(9;22)(q34.1;q11.2),inv(16)(p13.1q22)Alive, 10.5 years(34)
Ninomiya et al, 201163M31p190 46,XX,t(9;22)(q34;q11.2),inv(16)(p13.1q22)Succumbed to disease, 7 months following BMT(17)

[i] BCR-ABL, breakpoint cluster region-ABL proto-oncogene 1; M, male; F, female; BMT, bone marrow transplantation.

It is well-known that the patients with CML-BC exhibit a poor clinical outcome. By contrast, patients with Ph(+) AML with CBFβ-MYH11(+) exhibit a relatively favorable prognosis, similar to AML with CBFβ-MYH11(+) alone (16,17). Notably, the outcomes of the patients of the present study were consistent with these data. As initially diagnosed with CML-AP, case 1 was first treated with imatinib (400 mg/day orally), but was shortly changed to chemotherapy with cytarabine (100 mg/m2/day, days 1–7, intravenous drip) and idarubicin (8 mg/m2/day, days 1–3, intravenous drip) for 2 cycles, in combination with imatinibas the patient rapidly progressed to CML-BC. Although bone marrow and blood remission was achieved, the patient quickly relapsed and the treatment course was complicated by the lung infection. The patient underwent unrelated donor-allogeneic hematopoietic stem cell transplantation in November 2011. However, 15 months following transplantation, the patient succumbed to serious GVHD. Following the standard dose of induced chemotherapy, case 3 and 4 attained complete remission. Subsequently, case 3 received autologous-peripheral blood stem cell transplantation in June 2011 and remained in complete remission for 32 months following diagnosis.

Based on the data of the patient with CML in the present study, and previous studies (35), it is suggested that patients with CML carrying and expressing the BCR-ABL and CBFβ-MYH11 fusion genes appear more likely to rapidly progress to AP or BC compared with patients with CML without the co-existence of these two genes. The product of the CBFβ-MYH11 fusion gene may have served an important role in the transformation of CML, and the monocytes and eosinophils may be derived from common leukemic progenitors affected by the product of the CBFβ-MYH11.

p210BCR-ABL is the hallmark of CML, as identified in case 1 and case 2. However, it has also been previously demonstrated that an additional rearrangement to the classical p190BCR-ABL may be identified in AML (36). It appears that t(9;22)(q34;q11) with a p210BCR-ABL rearrangement may be a molecular event occurring not only during the development of CML, but also in AML following the Ph chromosomal anomaly. From the results of the previously described patients and the patients with AML with p210BCR-ABL in the present study, the TKI imatinib used in maintenance therapy was able to diminish the detectable levels of BCR-ABL transcripts, suggesting that in such cases, imatinib may be an effective treatment.

As these patients are so rare, an appropriate diagnosis and the standard treatment for them are currently not available. As the leukemic cell populations in the majority of patients are composed of diverse clones with heterogeneous phenotypes, the possibility that the p210BCR-ABL rearrangement and CBFβ-MYH11 may reside in different subclones in these patients cannot be excluded. These different subclones would subsequently evolve during the clinical course, depending on their response to treatment. Therefore, more careful karyotyping and FISH analysis are required to dissect the sequential molecular events occurring in a singular clone or in various subclones in these patients, in order to more precisely define this subtype of leukemia.

In conclusion, the present report demonstrated that t(9;22)(q34;q11) with a p210BCR-ABL rearrangement may be a molecular event occurring not only during the development of CML but also in AML following a primary specific chromosomal anomaly. Co-expression of p210BCR-ABL and CBFβ-MYH11 fusion genes may be a molecular event in various types of myeloid leukemia. The patients with CML carrying and expressing BCR-ABL and CBFβ-MYH11 fusion genes appear more likely to progress rapidly to AP or BC compared with patients with CML without the co-existence of these two genes; The product of the CBFβ-MYH11 fusion gene may serve an important role in the transformation of CML.

Acknowledgements

The present study was supported by grants from the Jiangsu Provincial Special Program of Medical Science (grant no. BL2012005), National Key Scientific Projects of China (grant no. 2011CB933501), National Scientific Foundation of China (grant no. 81070402), Jiangsu Province's Key Medical Center (grant no. ZX201102), National Public Health Grand Research Foundation (grant no. 201202017) and the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD).

References

1 

Cividin M, Brizard F, Sorel N, Renaud M, Guilhot F and Brizard A: p190(BCR-ABL) rearrangement as a secondary change in a case of acutenmyelo-monocytic leukemia with inv(16)(p13q22). Leuk Res. 28:97–99. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Delaunay J, Vey N, Leblanc T, Fenaux P, Rigal-Huguet F, Witz F, Lamy T, Auvrignon A, Blaise D, Pigneux A, et al: Prognosis of inv(16)/t(16;16) acute myeloid leukemia (AML): a survey of 110 cases from the French AML Intergroup. Blood. 102:462–469. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Liu P, Tarlé SA, Hajra A, Claxton DF, Marlton P, Freedman M, Siciliano MJ and Collins FS: Fusion between transcription factor CBF beta/PEBP2 beta and a myosin heavy chain in acute myeloid leukemia. Science. 261:1041–1044. 1993. View Article : Google Scholar : PubMed/NCBI

4 

Kundu M and Liu PP: Function of the inv(16) fusion gene CBFB-MYH11. Current Opinion in Hematology. 8:2012001. View Article : Google Scholar : PubMed/NCBI

5 

Swerdlow S, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J and Vardiman JW: WHO classification of tumors of hematopoietic and lymphoid tissues. 4th. IARC WHO Classification of Tumours; Lyon: 2008

6 

Nowell PC and Hungerford DA: A minute chromosome in human chronic granulocytic leukemia. Science. 142:14971960.

7 

Rowley JD: Letter: a new consistent chromosomal abnormality in chronic myelogenous leukemia identified by quinacrine fluorescence and Giemsa staining. Nature. 243:290–293. 1973. View Article : Google Scholar : PubMed/NCBI

8 

Faderl S, Talpaz M, Kantarjian HM and Estrov Z: Should polymerase chain reaction analysis to detect minimal residual disease in patients with chronic myelogenous leukemia be used in clinical decision making? Blood. 93:2755–2759. 1999.PubMed/NCBI

9 

World Health Organization Classification of Tumours: Pathology and genetics of tumours of haematopoietic and lymphoid tissues. Jaffe ES, Harris NL, Stein H and Vardiman JW: IARC WHO Classification of Tumours; Lyon: pp. 3522001

10 

Keung YK, Beaty M, Powell BL, Molnar I, Buss D and Pettenati M: Philadelphia chromosome positive myelodysplastic syndrome and acute myeloid leukemia-retrospective study and review of literature. Leuk Res. 28:579–586. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Cheung SW, Crane JP and Burgess AC: High resolution R banding in amniotic fluid cells using the BrdU-Hoechst 33258-Giemsa (RBG) technique. Human Genetics. 69:86–87. 1985. View Article : Google Scholar : PubMed/NCBI

13 

Shaffer LG, Slovak ML and Campbell LJ: International Standing Committee on Human Cytogenetic Nomenclature: ISCN 2009: An international system for human cytogenetic nomenclature. Shaffer LG, Slovak ML and Campbell LJ: Karger Publishers; Switzerland: 2009

14 

Li X, Li X, Xie W, Hu Y, Li J, Du W, Liu W, Li H, Chen X, Zhang L, et al: Comprehensive profile of cytogenetics in 2308 Chinese children and adults with de novo acute myeloid leukemia. Blood Cells Mol Dis. 49:107–113. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Popescu R, Dăscălescu A, Dănăilă C, Ghiorghiu D, Zlei M, Ivanov A, Sireteanu A, Gorduza EV and Azoicăi D: Co-expression of the CBFβ-MYH11 and BCR-ABL fusion genes in chronic myeloid leukaemia. Revista Romana De Medicina De Laborator. 23:221–230. 2015. View Article : Google Scholar

16 

Wu Y, Slovak ML, Snyder DS and Arber DA: Coexistence of inversion 16 and the philadelphia chromosome in acute and chronic myeloid leukemias: Report of six cases and review of literature. Am J ClinPathol. 125:260–266. 2006.

17 

Ninomiya S, Kanemura N, Tsurumi H, Kasahara S, Hara T, Yamada T and Moriwaki H: Coexistence of inversion 16 and the Philadelphia chromosome comprising P190 BCR/ABL in chronic myeloid leukemia blast crisis. Int J Hematol. 93:806–810. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Preudhomme C, Lai JL, Plantier I, Demory JL, Zandecki M and Fenaux P: Cytogenetic and molecular remission in a case of acute myeloid leukemia (AML) with inversion of chromosome 16 (inv(16)) and Philadelphia chromosome (Ph). Br J Haematol. 82:623–626. 1992. View Article : Google Scholar : PubMed/NCBI

19 

Siddiqui AD, Sheikh ZS, Liu D and Seiter K: Coexistence of inversion 16 and the Philadelphia chromosome in patients with acute myelogenous leukemia. Leuk Lymphoma. 43:1137–1140. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Li YS and Hayhoe FG: Ph1 chromosome positive acute myelomonocytic leukemia with inverted chromosome 16. Br J Haematol. 69:5761988. View Article : Google Scholar : PubMed/NCBI

21 

Miura I, Takatsu H, Yamaguchi A, Hashimoto K, Nimura T, Nishinari T, Niitsu H and Miura AB: Standard Ph chromosome t(9;22)(q34;q11), as an additional change in a patient with acute myelomonocytic leukemia (M4Eo) associated with inv(16)(p13q22). AM J Haematol. 45:94–96. 1994. View Article : Google Scholar

22 

Secker-Walker LM, Morgan GJ, Min T, Swansbury GJ, Craig J, Yamada T, Desalvo L, Medina JW, Chowdhury V, Donahue RP, et al: Inversion of chromosome 16 with the Philadelphia chromosome in acute myelomonocytic leukemia with eosinophilia: report of two cases. Cancer Genet Cytogenet. 58:29–34. 1992. View Article : Google Scholar : PubMed/NCBI

23 

Svaldi M, Lanthaler A, Venturi R, Coser P and Mitterer M: Simultaneous occurrence of bcr-abl and inv16 in a case of M1 acute myeloid leukemia. Leukemia. 15:695–699. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Tirado CA, Valdez F, Klesse L, Karandikar NJ, Uddin N, Arbini A, Fustino N, Collins R, Patel S, Smart RL, et al: Acute myeloid leukemia with inv(16) with CBFBMYH11, 3′CBFB deletion, variant t(9;22) with BCR-ABL1 and del(7)(q22q32) in a pediatric patient: case report and literature review. Cancer Genet Cytogenet. 200:54–59. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Roth CG, Contis L, Gupta S, Agha M and Safyan E: De novo acute myeloid leukemia with Philadelphia chromosome (BCR-ABL)and inversion 16(CBFβ-MYH11): report of two cases and review of the literature. Leukemia & Lymphoma. 52:531–535. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Bustamante D, Chan KR, Czuchlewski DR and Saadi AA: Patterns of BCR breakpoints in patients with coexisting inv(16)(p13.1q22) and t(9;22)(q34;q11.2). Int J Hematol. 95:324–326. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Merzianu M, Medeiros LJ, Cortes J, Yin C, Lin P, Jones D, Glassman A, Kantarjian H and Huh Y: inv(16)(p13q22) in Chronic myelogenous leukemia in blast phase: A clinicopathologic, cytogenetic and molecular study of five cases. Am J Clin Pathol. 124:807–814. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Tsuboi K, Komatsu H, Miwa H, Iida S, Banno S, Wakita A, Nitta M and Ueda R: Lymphoid blastic crisis of chronic myelogenous leukemia with inv(16)(p13;q22). Leuk Res. 26:771–774. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Myint H, Ross FM, Hall JL and Hamblin TJ: Early transformation to acute myeloblastic leukemia with the acquisition of inv(16) in Ph positive chronic granulocytic leukemia. Leuk Res. 21:473–474. 1997. View Article : Google Scholar : PubMed/NCBI

30 

Evers JP, Bagg A, Himoe E, Zwiebel JA and Jacobson RJ: Temporal association of marrow eosinophilia with inversion of chromosome 16 in recurrent blast crises of chronic myelogenous leukemia. Cancer Genet Cytogenet. 62:134–139. 1992. View Article : Google Scholar : PubMed/NCBI

31 

Asou N, Sanada I, Tanaka K, Hidaka M, Suzushima H, Matsuzaki H, Kawano F and Takatsuki K: Inversion of chromosome 16 and bone marrow eosinophilia in a myelomonocytic transformation of chronic myeloid leukemia. Cancer Genet Cytogenet. 61:197–200. 1992. View Article : Google Scholar : PubMed/NCBI

32 

Enright H, Weisdorf D, Peterson L, Rydell RE, Kaplan ME and Arthur DC: Inversion of chromosome 16 and dysplastic eosinophils in accelerated phase of chronic myeloid leukemia. Leukemia. 6:381–384. 1992.PubMed/NCBI

33 

Heim S, Christensen BE, Fioretos T, Sørensen AG and Pedersen NT: Acute myelomonocytic leukemia with inv(16)(p13q22) complicating Philadelphia chromosome positive chronic myeloid leukemia. Cancer Genet Cytogenet. 59:35–38. 1992. View Article : Google Scholar : PubMed/NCBI

34 

Colovic M, Jankovic G, Bila J, Djordjevic V and Wiernik PH: Inversion of chromosome 16 in accelerated phase of chronic myeloid leukemia: report of a case and review of the literature. Med Oncol. 15:199–201. 1998. View Article : Google Scholar : PubMed/NCBI

35 

Yin CC, Jones D, Cortes JE, Kantarjian H and Merzianu M: Medeiros LJ and Bueso-Ramos CE: Type B core binding factor β/smooth muscle myosin heavy chain fusion transcript in myeloid blast phase of chronic myeloid leukemia: Correlation with nuclear and cytoplasmic localization of the fusion protein. Clinical Leukemia. 2:257–260. 2008. View Article : Google Scholar

36 

Dai HP, Xue YQ, Wu LL, Pan JL, Gong YL, Wu YF, Zhang J, Wu DP and Chen SN: p210BCR-ABLas a secondary change in a patient with acute myelomonocytic leukemia(M4Eo) with inv(16). Int J Hematol. 96:814–817. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 14 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
Wang YY, Ding WJ, Jiang F, Chen ZX, Cen JN, Qi XF, Liang JY, Liu DD, Pan JL, Chen SN, Chen SN, et al: Coexistence of p210BCR‑ABL and CBFβ‑MYH11 fusion genes in myeloid leukemia: A report of 4 cases. Oncol Lett 14: 5171-5178, 2017
APA
Wang, Y., Ding, W., Jiang, F., Chen, Z., Cen, J., Qi, X. ... Chen, S. (2017). Coexistence of p210BCR‑ABL and CBFβ‑MYH11 fusion genes in myeloid leukemia: A report of 4 cases. Oncology Letters, 14, 5171-5178. https://doi.org/10.3892/ol.2017.6812
MLA
Wang, Y., Ding, W., Jiang, F., Chen, Z., Cen, J., Qi, X., Liang, J., Liu, D., Pan, J., Chen, S."Coexistence of p210BCR‑ABL and CBFβ‑MYH11 fusion genes in myeloid leukemia: A report of 4 cases". Oncology Letters 14.5 (2017): 5171-5178.
Chicago
Wang, Y., Ding, W., Jiang, F., Chen, Z., Cen, J., Qi, X., Liang, J., Liu, D., Pan, J., Chen, S."Coexistence of p210BCR‑ABL and CBFβ‑MYH11 fusion genes in myeloid leukemia: A report of 4 cases". Oncology Letters 14, no. 5 (2017): 5171-5178. https://doi.org/10.3892/ol.2017.6812