Genome Wide Profiling Of Chromosomal Abnormalities In 37 Patients With Monoclonal Gammopathy Of Undetermined Significance (MGUS)

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1874-1874
Author(s):  
Aneta Mikulasova ◽  
Vladimira Vallova ◽  
Jan Smetana ◽  
Henrieta Greslikova ◽  
Renata Kupska ◽  
...  

Abstract Introduction The incidence of clonal DNA copy number alterations (CNAs) in plasma cells (PCs) is considered as one of the most important and independent prognostic factors in patients with multiple myeloma (MM). Also in the premalignancy MGUS, there are specific chromosomal changes in PCs such as del(13)(q14), IGHtranslocation, gain(1)(q21) and hyperdiploidy. However, not much is known about their significance in relation to malignant transformation. Also, MGUS research is further complicated by small number of malignant cells that could be obtained from patients. Aim Array-CGH technique optimization and DNA CNAs analysis in relation to prognosis at genome-wide level in MGUS patients. Material and methods We have analysed 37 MGUS patients (22M/15F; median age 62) using array-CGH, Agilent platforms “Human Genome CGH, 4×44K” (n=10) and “SurePrint G3 CGH+SNP, 4×180K” (n=27). DNA was isolated from separated PCs (using CD138, CD19 and CD56 markers) and amplified by multiple displacement amplification (MDA). Results CNAs were observed in 57% (21/37) MGUS patients. Numerical and structural CNAs were found in 46% (17/37) and 41% (15/37) MGUS patients, respectively. We distinguished two genetic subgroups similar to MM patients: hyperdiploid and non-hyperdiploid. Hyperdiploidy was present in 32% (12/37) MGUS patients. The most frequent whole-chromosome gains were: 9 (83%, 10/12), 19 (83%, 10/12), 3 (75%, 9/12), 11 (67%, 8/12) and 15 (58%, 7/12). Non-hyperdiploidy was detected in 68% (25/37) MGUS patients. In both, hyperdiploid and non-hyperdiploid subgroups we have found loss of chromosomes 13 (25% vs. 20%) and Y (17% vs. 4%). Loss of chromosome 8 was detected only in hyperdiploid subgroup (8%) and losses of chromosomes 21, 22 and X only in non-hyperdiploid subgroup (8%, 4% and 8%, respectively). Structural changes were significantly more often present in hyperdiploid than in non-hyperdiploid patients (67% vs. 28%, p<0.05). We have identified whole-arm chromosome changes 1q gain and 16q loss in both hyperdiploid and non-hyperdiploid patients (17% vs. 12%, 8% vs. 8%, respectively), but 16p gain was seen only in hyperdiploid patients (8%). Segmental chromosome changes were also present in both hyperdiploid and non-hyperdiploid patients (58% vs. 24%). Interestingly, we have detected more segmental losses (15 vs. 8) and gains (6 vs. 2) in hyperdiploid than in non-hyperdiploid patients, but size median of these losses (3.33 Mb vs. 19.6 Mb) and gains (36.1 Mb vs. 63.5 Mb) was smaller in hyperdiploid than in non-hyperdiploid patients. Moreover, we found one MGUS patient who has progressed to MM and required therapy after 6 months from MGUS diagnosis. Genome-wide profile of MGUS patient was especially unique by high number of structural changes (n=11) compared to other 15 MGUS patients with structural CNAs (median 1; 1 – 6). MGUS patient’s profile showed hyperdiploidy (gains of chromosomes 3, 5, 9, 11, 15 and 19), losses of chromosomes 8, 13 and Y, 7 segmental losses in areas 1p34.2-p13.1, 6p23, 6q12-q27, 7q36.3, 12p12.1-p11.23, 12q12, 12q21.2-q23.3 and 4 segmental gains in areas 6p25.3-p23, 6p23-p11.1, 6q11.1-q12, Xq21.33-q28. In addition, gains and losses, which included whole or large parts of chromosomes, showed unusual profile associated with other alterations. These findings have suggested a complex karyotype. Summary In our study we have optimized protocol of array-CGH analysis from amplified DNA and we have used it in first 37 MGUS patients. We found there are various chromosomal changes (numerical, whole-arm and segmental) in more than half of MGUS patients. Similar to MM, hyperdiploid and non-hyperdiploid genetic subgroups were identified. We also described one MGUS case with unique genome-wide profile indicating unfavourable prognosis. Support NT13492, NT13190, NT11154, OPVK CZ.1.07/2.3.00/20.0183, MSM0021622434, GAP304/10/1395 Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5063-5063
Author(s):  
Hossein Mossafa ◽  
Sabine Defasque ◽  
Christine Fourcade ◽  
JeanPierre Hurst ◽  
Bertrand Joly

Abstract Abstract 5063 Introduction, We describe the simultaneous presentation of multiple myeloma (MM) and yeloproliferative disorders (MPD) or lymphoid diseases (LD) at diagnosis. Therapy-related myelodysplasia (tMDS) occurring during the course of MM is generally believed as a result from hematopoietic stem cell-toxic therapies, such as ionizing radiation and alkylating agent-based chemotherapies (melphalan, nitrosoureas).Patients and methods, We study a total of 342 patients (151 F, 191 M; median age 68.1 years; range 42 to 93 Years), diagnosed with MM based on the International Staging System. The basis for inclusion of patients in this study was with previous untreated MM ones. The study was performed in accordance with the declaration of Helsinki. To determine whether chemotherapies for MM factors play the critical role in the development of secondary disease, simultaneously two different cultures were processed, an unstimulated 96 hours culture (U96HC) on whole BM(WBM), a short-time 24 hours culture (ST24HC) after CD138+ plasma cells (PCs) depleted on negative fraction (CD138- cells) of BM and the FISH was investigated on purified CD138+.All samples were enriched in PCs by the Automated Magnetic Cell Sorter (Miltenyi technology)proceeded with anti-CD138 specific antibodies applied. The CD138+ PCs and the CD138- cells were collected in different tubes. The CD138− cells were used for a ST24HC. FISH was performed on the purified CD138+, PCs with a recommended FISH panel (MM International Working Group). Screening was performed systematically for the following unbalanced alterations and reciprocal rearrangements: del(13)(q14)(D13S25), del(17)(p13)(TP53),+3(D3Z), +9(D9Z1), +15(D15Z14), t(4;14)(p16;q32)/IGH-FGFR3, t(11;14)(q13;q32)/IGH-CCND1 (Abbott).After observing the results of U96HC on whole BM (CD138+ and CD138− cells), ST24HC (CD138− cells) and FISH for each patient, two clone cytogenetically were distinct and unrelated chromosomal abnormalities were found in 40 (11.7%) of the 342 MM patients (6 F, 34 M; median age 74 years; range 42 to 87 Years) 34 had a MPD and 6 had a LD. A second immunophenotyping analysis confirmed the presence of those LD/MM simultaneous haematological malignancy. In the cases of the patients with MM/ MPD, the frequency of cytogenetic abnormality unrelated to the myeloma clone was respectively; the 20q deletion, detected for 13 the 34 patients, the 20q- is a sole abnormality for 12 cases and associated with a complex caryotype in 1 case. The trisomy of chromosome +8 was observed in 7 cases, the del(7q) or monosomy 7 in 5 cases, loss of gonosome Y in 4 cases, del(11) for 2 cases, translocation t(9;22) in one case, 5q abnormality in one case and trisomy 9 with JAK2 V617F mutation in one case. For the patients with MM/LD, 5 patients had a trisomy +12 and or trisomy +18 like sole abnormality or associated with others cytogenetics abnormalities and one patient had 6q deletion. Discussion, Whereas in the literature the most common cytogenetic abnormalities typifying MPD after alkylator-based therapy include partial or complete deletions of chromosomes 5, 7, and 20 as well as trisomy 8. In our study we observed those abnormalities with the same frequency for the patients had simultaneous MPD associated in untreated MM at diagnosis. Six patients had simultaneous LD and MM. The marginal zone lymphoma was confirmed for 3 patients. The CC observed a trisomy +12 for those three patients associated with +18 and +19 for 2 cases and del(13) and trisomy 3 for one among them. We demonstrated in untreated MM patients the coexistence of MM and MPD or LD at diagnosis with MPD-type or LD-type chromosome abnormalities within MM signature karyotype. We hence recommend that CC studies, 96 hours WBM, 24 hours on negative fraction CD138− cells and FISH on purified CD138+ PCs, the three should be an integral part of the evaluation of patients with MM at diagnosis into clinical trials using HDT is warranted to determine whether patients who are predisposed to developing tMDS/sAML, they can be identified prospectively. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 87 (1) ◽  
pp. 299-307 ◽  
Author(s):  
J Dierlamm ◽  
S Pittaluga ◽  
I Wlodarska ◽  
M Stul ◽  
J Thomas ◽  
...  

Clinical, histologic, cytogenetic, and molecular genetic data of 31 patients with extranodal, nodal, and splenic marginal zone B-cell lymphoma (MZBCL) are presented. Despite these variable clinical manifestations, a similar spectrum of morphologic features as well as distinctive immunophenotypic findings were noted. In all cases, a monotypic B-cell proliferation consistently negative for CD5, CD10, and CD23 was found expanding the marginal zone of the B follicle with and without colonization of the follicle centers. Clonal chromosomal abnormalities were detected in 23 of the 31 patients. Recurrent aberrations included whole or partial trisomy 3 (18 cases), trisomy 18 (9 cases), and structural rearrangements of chromosome 1 with breakpoints in 1q21 (9 cases) or 1p34 (6 cases), all of which were seen in extranodal, nodal, as well as splenic MZBCL. Abnormalities of the additional chromosome 3, such as +del(3)(p13),+i(3)(q10), or structural changes involving the distal part of the long arm, were evident in 9 of the 18 cases. All recurrent abnormalities were found in MZBCL more frequently than in other histologic entities of B-cell non-Hodgkin's lymphoma (B-NHL). None of the known lymphoma-associated chromosomal changes or rearrangements of the BCL1, BCL2, BCL3, BCL6, and CMYC genes were detected. We conclude that MZBCL represent a distinct entity of B- NHL with characteristic morphologic and immunophenotypic features and particular chromosomal abnormalities, and that a close histogenetic relationship between extranodal, nodal, and splenic MZBCL is likely, although the clinical presentation may vary.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3724-3724
Author(s):  
Andreas Roller ◽  
Simone Weber ◽  
Alexander Kohlmann ◽  
Melanie Zenger ◽  
Marita Staller ◽  
...  

Abstract Background Gains and losses of chromosomal material are frequent in AML and MDS and usually lead to loss or gain of a single copy of a whole chromosome, a chromosome arm or small stretches of the chromosome that may be microscopically invisible. More rarely, amplifications of chromosomal regions (defined as the presence of more than 6 copies of a region per cell) are observed. These supernumerary copies are located either extrachromosomally as small acentric chromosomal structures - so called double-minutes (dmin) - or intrachromosomally as large contiguous stretches of amplified DNA, so called homogeneously staining regions (HSR). Aims Characterize AML and MDS cases with gene amplifications with respect to size, affected genes and accompanying chromosomal abnormalities as well as TP53 status. Patients and Methods 84 AML and 31 MDS cases with cytogenetically visible amplifications were selected for this study. All cases were analyzed by array CGH, chromosome banding analysis, sequencing for TP53 mutations as well as FISH for TP53 deletions. Results The cohort comprised 55 (47.8%) males and 60 (52.2%) females with a median age of 72.0 years (range 38.0 - 90.3 years). A complex karyotype (≥4 aberrations) was present in 92/115 (80.0%) cases (AML=65/84 (77.4%); MDS=27/31 (87.1%)). In total, 385 amplified regions were identified by array CGH. In more detail: 3q26 (AML: n=6; MDS: n=3), 8q24 (AML: n=15; MDS: n=1), 11q21-25 (AML: n=42; MDS: n=13), 13q12 (AML: n=3; MDS: n=1), 13q31 (AML: n=3; MDS: n=2), 19p13 (AML: n=2; MDS: n=4), and 21q21-q22 (AML: n=24; MDS: n=5). The median number of amplified regions was 3 (range 1-18). In 14/115 (12.2%) cases, the amplification was located in dmins (AML: n=11; MDS: n=3) and in 101/115 (87.8%) patients in HSR (AML: n=73; MDS: n=28). In 40 of the latter 101 cases (39.6%) (AML: n=24; MDS: n=16) the amplification was located on a ring chromosome (rc). In patients with complex karyotypes we detected a significantly higher number of amplified regions as compared to non-complex karyotypes (3.5 vs. 2.8; p=0.015). No association between the complexity of the karyotype and the structural type of the amplification (dmin vs rc) was observed. Cases with non-complex karyotypes frequently harbored a 5q deletion (6/23; 26.1%) or chromosome 8 abnormalities (3/23; 13.0%). Within the subgroup of non-complex karyotypes del(5q) cases showed a tendency to a higher number of amplified regions (3.6 vs. 1.9; p=0.140). Further, amplifications of 11q genes were more frequent in complex karyotypes (54.4% vs. 21.7%; p=0.005), whereas 8q amplifications were more frequent in non-complex karyotypes (43.5% vs. 4.4%; p<0.001). We detected a large region on band 11q24, which was amplified in 41/53 (77.4%) cases. This commonly amplified region contains 1,575 genes including the MLL gene. Cases harboring dmins had shorter amplified regions compared to cases with rc (4,428,112.5 bp vs. 18,265,496.9 bp; p=0.028). Moreover, we detected a positive correlation of patients having a rc and gene amplification on chromosome 11q23-25 (p<0.05). On chromosome 3q, 8/9 (88.9%) cases shared a minimal amplified region covering the EVI1 gene. In comparison to samples obtained from healthy donors (n=47), the EVI1 expression was significantly higher in cases with EVI1 amplification (87.4 vs. 0.5; p=0.048). On chromosome 21q the regions of amplifications were heterogeneous. However, we detected a minimal region containing 11 genes including ERG which was amplified in 26/29 (89.7%) patients. ERG expression data was available in 8 cases and was significantly higher compared to a control cohort of AML with normal karyotype (n=331) (729.2 vs. 229.0; p=0.05). On chromosome 8 an amplified region was identified in 15/16 cases. In 14 of these cases (87.5%) the region included MYC. TP53mut were present in 93/115 (80.9%) patients, accompanied by a TP53del in 28/93 (30.1%) cases. Interestingly, cases harboring a TP53mut had more amplified regions compared to TP53wt (3.4 vs. 1.7; p<0.001). Conclusions 1. MLL is the most frequently amplified gene in AML and MDS. 2. Patients with complex karyotypes or TP53mut harbored more amplified regions compared to patients with non-complex karyotypes and TP53wt. 3. Amplifications on 11q were more frequent in complex karyotype whereas gene amplifications on 8q were predominantly observed in non-complex karyotypes. 4. EVI1 and ERG gene amplifications lead to a higher expression of the respective genes. Disclosures: Roller: MLL Munich Leukemia Laboratory: Employment. Weber:MLL Munich Leukemia Laboratory: Employment. Kohlmann:MLL Munich Leukemia Laboratory: Employment. Zenger:MLL Munich Leukemia Laboratory: Employment. Staller:MLL Munich Leukemia Laboratory: Employment. Kern:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1944-1944 ◽  
Author(s):  
Hervé Avet-Loiseau ◽  
Denis Caillot ◽  
Gerald Marit ◽  
Valerie Lauwers-Cances ◽  
Murielle Roussel ◽  
...  

Abstract Abstract 1944 Cytogenetic abnormalities have been associated with specific outcome in myeloma, as in many other hematologic malignancies. Among the large spectrum of chromosomal changes observed in myeloma, at least two of them are clearly associated with a specific poor outcome, both in terms of PFS and OS, i.e., t(4;14) and del(17p). Recent data suggested that bortezomib can at least partially overcome the prognostic value of t(4;14). However, t(4;14) remains a prognostic factor in the IFM series. Deletion 17p is rare (< 10%), but is associated with a very poor outcome, whatever the treatment proposed to these patients. In 2005, the IFM designed a clinical trial aiming to test the role of lenalidomide maintenance until relapse in patients under 65 years of age treated with a VAD or bortezomib/dexamethasone induction, followed by high-dose melphalan. Six hundred and fourteen patients have been enrolled, and the clinical results of this trial are presented in another abstract. Chromosomal data focused on t(4;14) and del(17p) were available for 488 of those patients, as analyzed at diagnosis by FISH on sorted plasma cells. The t(4;14) was observed in 13.3% of the patients and del(17p) (defined by presence in at least 60% of the plasma cells) was present in 6.6% of them, incidences that are in agreement with previous reports. The median PFS (calculated from the date of randomization) for patients with t(4;14) were 15 months and 27 months for patients in the placebo and lenalidomide arms, respectively. The median for patients with del(17p) were 14 months and 29 months for patients in the placebo and lenalidomide arms, respectively. The comparison of PFS in the lenalidomide arm for patients with t(4;14) or del(17p), as compared with those lacking the chromosomal abnormalities shows a significant difference, in favor of the “no abnormality” group. In conclusion, lenalidomide maintenance very significantly improves the PFS of patients with high-risk cytogenetics, albeit these chromosomal changes retain their prognostic value. Disclosures: Facon: celgene: Consultancy, Research Funding; johnson and johnson: Consultancy. Attal:celgene: Consultancy, Research Funding; johnson and johnson: Consultancy, Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2950-2950
Author(s):  
Kai Neben ◽  
Anna Jauch ◽  
Dirk Hose ◽  
Christiane Heiss ◽  
Thomas Hielscher ◽  
...  

Abstract Abstract 2950 Smoldering MM (sMM) is a plasma cell disorder defined by the presence of ≥10% plasma cells in bone marrow and/or a monoclonal protein level of ≥3 g/dl in serum without organ damage. The aim of this retrospective study was to analyze whether genomic abnormalities confer prognostic information in patients with sMM who are at high risk of progression into symptomatic MM. By using fluorescent in situ hybridization (FISH), we analyzed the prognostic value of 14 chromosomal abnormalities and hyper-/non-hyperdiploidy (HD and NHD, respectively) in a series of sMM-patients (n=200). In addition, the most frequent chromosomal aberration was used to determine the percentage of clonal plasma cells (cPC) in the bone marrow. Interphase-FISH-analysis on CD138-enriched plasma cells detected gains of chromosomes 1q21 (31%), 5p15/5q35 (35%), 9q34 (45%), 11q23 (41%), 15q22 (40%), and 19q13 (41%), as well as deletions of chromosomes 8p21 (9%), 13q14 (37%) and 17p13 (7%). Furthermore, the IgH-translocations t(14;16), t(4;14), t(11;14) and IgH-translocations with unknown translocation partner were observed in a frequency of 5%, 10%, 24% and 22%, respectively. The median percentage of cPC was 85.5 (IQR: 62 – 95). For the entire group, the median follow-up time was 27.2 months (range, 18.2 – 33.5). We analyzed the prognostic impact of each chromosomal aberration on time to progression (TTP). Of all chromosomal abnormalities analyzed, only del(8p21) and the percentage of cPC showed a significant impact on TTP. The TTP at 3 years for patients with del(8p21) was 53% versus 73% for those without (p=0.01). An incremental increase of cPC in the bone marrow by 10% was associated with an elevated relative risk to develop a symptomatic MM of 33% (p<0.001). After adjustment of p-values for multiple testing, only the percentage of cPC showed a statistically significant impact on TTP (p=0.02). Our results show that FISH-analysis on CD138-enriched plasma cells is a useful technique in the study of sMM, because it allows myelomatous plasma cells to be discriminated from their normal counterparts. In addition, our findings suggest that the proportion of cPC (analyzed by FISH) rather than single chromosomal abnormalities predict progression from sMM to symptomatic MM. FISH-based information can be obtained easily at the time of diagnosis, which would help to establish an individually adapted follow-up strategy. Aberration yes vs. no N Incidence 3-yr TTP (present vs. absent) Hazard ratio Wald test p-value adjusted del(8p21) 190 9% 53% vs. 73% 2.59 0.1 del(13q14) 200 37% 61% vs. 79% 1.77 0.8 del(17p13) 198 7% 56% vs. 72% 1.95 1 t (4;14) 198 10% 52% vs. 73% 1.68 1 t (11;14) 198 24% 77% vs. 69% 0.51 1 t (14;16) 197 5% 57% vs. 71% 1.59 1 +1q21 197 31% 60% vs. 75% 1.71 1 HD vs. NHD 197 40% 65% vs. 74% 1.67 1 10% increase of cPC 200 – – 1.33 0.02 cPC >95 vs. ≤95% 200 23% vs. 77% 46% vs. 80% 3.84 <0.001 Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4987-4987 ◽  
Author(s):  
Rakesh Verma ◽  
Lalit Kumar ◽  
Ashutosh Halder ◽  
Atul Sharma ◽  
Ritu Gupta ◽  
...  

Abstract Abstract 4987 Purpose: We prospectively studied patients of multiple myeloma (MM) for presence of t(14q32), and del13q14 on FISH and numerical chromosomal abnormalities on conventional cytogenetics(CC). Methods: Between February 2007 and June 2009, 90 previously untreated patients of MM were enrolled, Patient's median age was 55 years (range, 34 to 75 years) and 67 were males (M: F: 2.9: 1). 24 patients had ISS stage I, stage II-46 & 20 patients had stage III. Purified bone marrow plasma cells, using magnetic activated cell sorter with CD138 micro beads were used. Conventional cytogenetics for numerical chromosomal abnormalities and FISH for deletion 13q14 (RB-1) and 14q32 (IgH) translocations was carried out on these purified plasma cells. 65/90 (72%) patients received therapy; using novel agents, n=54 (thalidomide-dexamethasone (n=50), Lenalidomide-dexamethasone (n=4)) and melphalan, prednisolone and thalidomide (MPT, n=11). Response was evaluated post 4 cycles using EBMT response criteria. Results: Metaphases could be obtained on CC in 53/90 patients (58%); with numerical abnormalities in 21: hyperdiploid -16 & hypodiploid in 5. Del 13q14 and t (14q32) by FISH was present in 51 (56%) and 75 (83%) patients, respectively. t (14q32) was present more in patients with ISS-III (p<.008). Presence of del13q14 correlated with BM plasma cells >40% (P<.007) and Hb ≤9.2G/dl (p=.06). Following therapy, 49/65 patients (75.2%) achieved significant response (CR+VGPR+PR). Patients aged ≤55 years (P = 0.07), those with ISS- I (P <0.05), IgG-kappa (P<0.04) and with absence of del13q14 (P = 0.04) responded better. The median overall survival (OS) has not reached yet, median event-free survival (EFS) is 33 months (95% CI 18–48). Estimated OS and EFS at 2 years is 77% and 50 %, respectively. Durie Salmon stage IIIB (HR 3.6) and response to therapy (HR 7.8) were significant predictors of OS. For EFS - del13q14 (HR 2.7, P <0.036), response to therapy (HR 2.7,P<0.01) and use of novel agents (HR 3.1, (P<0.008) were important predictors. Conclusion: Chromosomal abnormalities including IgH translocations and del13q14 with numerical chromosomal abnormalities are common in MM patients at diagnosis and can predict the treatment outcome and survival. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1352-1352 ◽  
Author(s):  
Hitoshi Sano ◽  
Kentaro Ohki ◽  
Myoung-ja Park ◽  
Yusuke Hara ◽  
Norio Shiba ◽  
...  

Abstract Background Granulocyte colony-stimulating factor (G-CSF; CSF3) and its receptor (G-CSFR; CSF3R) control neutrophil production under basal circumstances and during episodes of bacterial infections. Mutations in a region of the CSF3R gene coding for the intracytoplasmic domain of the G-CSFR have been discovered in patients with severe congenital neutropenia (CN) and were initially suggested to be the cause of CN. Mutations in CSF3R gene are regarded as an early marker of malignant transformation in CN. Common genetic abnormalities like acquired clonal cytogenetic alterations or activating RAS mutations have also been observed to be associated with CN-related myelodysplastic syndrome(MDS)/leukemia. In adults, a frequency of CSF3R mutation was common (59%) in patients with chronic neutrophilic leukemia (CNL) and atypical (BCR-ABL1–negative) chronic myeloid leukemia (CML), whereas that was low (1%) in patients with de novo acute myeloid leukemia (AML). Sequence variants that were identified included membrane proximal mutations and a number of different frameshift or nonsense mutations that truncate the cytoplasmic tail of CSF3R. These mutations segregate within two distinct regions of CSF3R and lead to preferential downstream kinase signaling through SRC family-TNK2 or JAK kinases and differential sensitivity to kinase inhibitors. However, little is known about the incidence and prognostic values of CSF3R mutations in pediatric myeloid malignancies. Methods CSF3R mutations (exon14 and 17) in a total of 376 samples of pediatric de novo AML, 40 samples of juvenile myelomonocytic leukemia (JMML), and 20 samples of MDS were analyzed from gDNA or cDNA extracted from diagnostic bone marrow samples using direct sequencing. Moreover, we assessed whether CSF3R mutations overlap with known gene abnormalities, such as FLT3, c-KIT, NPM1 and SETBP-1. Mutational analyses of FLT3, c-KIT, NPM1 and SETBP-1 were also performed in AML samples. Results We identified a CSF3R mutation in 5 of 376 patients (1.3%) with AML, 2 frameshift mutations (E788X) and 3 missense mutations (L777F and T618I). The patients with 3 (L777F and E788X) of these 5 mutations had complex chromosomal abnormalities involved in chromosome 8 and 21 (t(8;21) and ins(21;8); AML1/ETO). The other 2 patients with missense mutations (T618I) had normal karyotype. They presented with a relatively high WBC counts of 100.6×10*9/l and 159.5×10*9/l. Although 2 of 5 these patients relapsed, all of them were salvaged successfully. Two of 5 had c-KIT mutations, while none of them had FLT3, NPM1, and SETBP-1 mutations. No mutations of CSF3R in JMML and MDS patients were found. Conclusions To our knowledge, this is the first report to describe the CSF3R mutation in a pediatric de novo AML patient. In our study, CSF3R mutation is detected in 1.3% in childhood de novo AML, which suggests the involvements of CSF3R mutations in the pathogenesis of pediatric de novo AML, JMML and MDS are extremely rare compared with those in SCN and adult CNL and atypical CML cases. Since CSF3R mutation is rare event, the prognostic values of CSF3R mutations in de novo AML are still unclear. Further data accumulation is necessary. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3727-3727
Author(s):  
Vesna Najfeld ◽  
Joseph Tripodi ◽  
Marina Kremanskaya ◽  
John Mascarenhas ◽  
Ronald Hoffman

Abstract In the past cytogenetic studies of patients with MF were hindered because marrow (BM) specimens were used for karyotyping but frequently could not be obtained due to advanced marrow fibrosis. Since MF is characterized by the constitutive mobilization of immature myeloid cells into the peripheral blood (PB), we compared unstimulated PB specimens with BM specimens to determine their utility in successfully detecting cytogenetic abnormalities in patients with MF. We also simultaneously performed interphase FISH (IFISH) studies in order to determine if IFISH identified additional genomic abnormalities in MF. We evaluated 183 patients who had had successful conventional and molecular cytogenetic analyses. Our myeloproliferative neoplasm ( MPN)-FISH panel consisted of twelve probes (EGR1 at 5q31, D5S23 at 5p15.3, D7Z1 at 7p11-q11, D7S522 at 7q31, D8Z2 at 8p11-q11, CDKN2A at 9p21, CEP9 at 9p11-q11, ATM at 11q22,1, Rb1 at 13q14, and D20S18 at 20q12, CKS1B at 1q21 and CDKN2C at 1p32). BM cytogenetics was studied in 60 pts (33%) and PB cytogenetics were evaluated in 123 pts (67%). Conventional cytogenetics was informative for 96% (123/128) of PB specimens and 97% (60/62) of BM samples. When conventional cytogenetic analysis was compared to IFISH, concordant results were observed in 154 patients (84%). Moreover, conventional karyotyping identified chromosomal abnormalities in an additional 18 patients (10%), which were not targeted by the 12 loci FISH panel. Among these patients, structural abnormalities of chromosome 12 were detected in 6 patients (33%) which represents the most frequent structural abnormality detected in MF and was associated with poor survival. Of the 63 patients with concordant abnormal cytogenetics and FISH the most frequent abnormalities included: del(20)(q11q13) (n=30, 48%), unbalanced 1q translocations resulting in trisomy 1q or duplication of 1q (n=17, 27%), gain of chromosome 8 (n=9, 14%), +8,+9( n=4, 6%), trisomy 9 (n=4, 6%), del(7q)/-7( n=7,11%), deletion 13q (n=9,14%) and complex karyotype (n=16, 25%). The remaining 11pts (6%) had discordant conventional and molecular cytogenetic results and were divided into two categories: A) those with normal cytogenetics (with or without non clonal abnormalities) and an abnormal MPN IFISH panel and B) those with abnormal clonal karyotype with normal lFISH. When compared to BM, PB specimens had a similar rate of abnormal karyotype: 51% in PB vs 48% in BM. Our results unequivocally demonstrated that conventional cytogenetics of MF can be successfully obtained from unstimulated PB specimens in 96% of patients and that analysis of BM does not reveal additional cytogenetic abnormalities. We conclude that FISH analysis has limited value in MF and is only informative for those patients who lack mitotic cells or who are cytogenetically normal, and in these patients, IFISH detects cryptic abnormalities in 4%. Use of PB karyotyping in MF is sufficient to effectively detects clonal hematopoiesis which contributes to prognostic risk stratification and influences therapeutic decision making. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4546-4546 ◽  
Author(s):  
Montserrat Lopez Rubio ◽  
Anna Gaya ◽  
Marta Morado ◽  
Vicente Carrasco ◽  
Ataulfo Gonzalez Fernandez ◽  
...  

Abstract INTRODUCTION PNH clones appear frequently in patients with myelodysplastic syndrome (MDS) and aplastic anemia (AA) which may even evolve into classic PNH. It has been described isolated cases of clonal evolution from AA, through classic PNH and MDS; and at the end, acute leukemia (AL) has rarely been described. These patients show a progressive replacement of the PNH clone by a dysplastic one carrying a monosomy of chromosome 7. OBJECTIVE We designed a retrospective study of patients included in the database of the Spanish PNH Registry in search of patients with a MDS and a PNH clone or those with a classic PNH who evolved into MDS. Our aim was to study the characteristics of patients who showed some of these entities and AA history. RESULTS Our search retrieved 6 patients, of whom 3 were MDS cases with a small PNH clone (Table 1). One of the patients had had a previous classic PNH followed by a MDS. The remaining two patients had been diagnosed with AA, developing classical PNH and MDS later; of these, one (patient 2) died with an acute leukemia. The median follow-up time was 83 (range 56-480) months, while the follow-up time from diagnosis of MDS was 68 (range 6-108) months. The patient characteristics are describe in Table 1. There are two remarkable features, diagnosis of a RAEB-1 in two cases and the frequency of trisomies of chromosome 8 and 21 and deletion of chromosome 7. These chromosomal abnormalities appeared sequentially as MDS evolved in 3 patients. The PNH clone disappeared in those patients who developed a MDS after an AA/classic PNH and persisted in those MDS cases who showed a PNH clone at diagnosis of MDS. Table 1. Patients Characteristics Previous diagnosis Follow-up time MDS type Karyotype Case 1 None 70 RARS + 8 Case 2 AA + PNH 56 RCMD -7 evolution to -7, + 21 Case 3 Classical PNH 83 RAEB-1 + 8 Case 4 None 83 RAEB-1 +21 evolution to +21, +8 Case 5 AA + classical PNH 480 RCMD + 8 evolution to +8, -7 Case 6 None 108 RCMD Normal CONCLUSIONS Patients with a diagnosis of AA, classic PNH or MDS with a PNH clone must be followed up for life, with bone marrow studies with karyotype and measurements of the PNH clone, to rule out the development of other hematological entities. Our series is a small one and only studies with a larger number of patients will allow us to establish definitive conclusions. The inclusion of these patients in the International PNH Registry is strongly advised. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 87 (1) ◽  
pp. 299-307 ◽  
Author(s):  
J Dierlamm ◽  
S Pittaluga ◽  
I Wlodarska ◽  
M Stul ◽  
J Thomas ◽  
...  

Abstract Clinical, histologic, cytogenetic, and molecular genetic data of 31 patients with extranodal, nodal, and splenic marginal zone B-cell lymphoma (MZBCL) are presented. Despite these variable clinical manifestations, a similar spectrum of morphologic features as well as distinctive immunophenotypic findings were noted. In all cases, a monotypic B-cell proliferation consistently negative for CD5, CD10, and CD23 was found expanding the marginal zone of the B follicle with and without colonization of the follicle centers. Clonal chromosomal abnormalities were detected in 23 of the 31 patients. Recurrent aberrations included whole or partial trisomy 3 (18 cases), trisomy 18 (9 cases), and structural rearrangements of chromosome 1 with breakpoints in 1q21 (9 cases) or 1p34 (6 cases), all of which were seen in extranodal, nodal, as well as splenic MZBCL. Abnormalities of the additional chromosome 3, such as +del(3)(p13),+i(3)(q10), or structural changes involving the distal part of the long arm, were evident in 9 of the 18 cases. All recurrent abnormalities were found in MZBCL more frequently than in other histologic entities of B-cell non-Hodgkin's lymphoma (B-NHL). None of the known lymphoma-associated chromosomal changes or rearrangements of the BCL1, BCL2, BCL3, BCL6, and CMYC genes were detected. We conclude that MZBCL represent a distinct entity of B- NHL with characteristic morphologic and immunophenotypic features and particular chromosomal abnormalities, and that a close histogenetic relationship between extranodal, nodal, and splenic MZBCL is likely, although the clinical presentation may vary.


Sign in / Sign up

Export Citation Format

Share Document