Prognostic and diagnostic value of Ewing family of tumors (EFTs)-associated fusion transcripts detected in peripheral blood specimens.

2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 10538-10538
Author(s):  
Joanna Przybyl ◽  
Hanna Kosela ◽  
Katarzyna Wiater ◽  
Konrad Ptaszynski ◽  
Anna Szumera-Cieckiewicz ◽  
...  

10538 Background: EFTs are characterized by chromosomal translocations leading to formation of oncogenic EWSR1-FLI1 fusion gene in 85-90% of cases. The aim of the study was to detect circulating tumor cells (CTCs) carrying EWSR1-FLI1 fusion transcript in peripheral blood and assess their added value to standard diagnostic procedures and utility as a prognostic marker in EFTs. Methods: 10mL of whole blood was collected from 35 untreated adult EFTs patients at the diagnosis (period: 2008-2011, median age 27 years) and 13 healthy controls. 13 patients presented metastatic disease (M1) at the diagnosis. Nested RT-PCR was applied in triplicate for the detection of EWSR1-FLI1 transcript. Blood specimen was regarded CTC-positive when at least 2 out of 3 nested RT-PCR assays were positive and results were confirmed by sequencing. FISH assay for EWSR1 rearrangement was performed on FFPE tumor tissue in 28 available cases. Median follow-up was 16 months. Results: EWSR1-FLI1 transcript was detected in peripheral blood of 71.4% (n=25) of patients. FISH assay was positive for EWRS1 rearrangement in 58% of cases. In 10 patients, where FISH assay could not be performed due to insufficient quality or lack of material, nested RT-PCR provided confirmation of immunopathological diagnosis. Specificity of RT-PCR blood test in healthy control was 91.4% (12/13 negative; p=0.0001). Median overall survival (OS) was 18 months. CTC-positive patients showed the trend for longer OS than CTC-negative patients (1-year OS: 89% vs. 58%; p=0.07), but longer follow-up is needed. Conclusions: Our results show that the fusion transcript detection in peripheral blood specimens may be a useful additional test to the standard clinicopathological diagnosis of EFTs. High detection rate of EWSR1-FLI1 transcript in peripheral blood of EFTs patients at the diagnosis may imply EFTs as a systemic disease with clinically evident metastases or micrometastases at presentation. Prognostic significance of CTCs in EFTs warrants further studies.

2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e22046-e22046
Author(s):  
Joanna Przybyl ◽  
Maria Debiec-Rychter ◽  
Tomasz Switaj ◽  
Hanna Melania Kosela ◽  
Slawomir Falkowski ◽  
...  

e22046 Background: EFTs are characterized by oncogenic EWSR1-FLI1 fusion gene, which can be detected in 85-90% of tumors and in 20-70% of peripheral blood specimens. It has been previously reported that downregulation of CDH2 (encoding N-cadherin) and overexpression of CDT2 [also known as DTL, encoding denticleless homolog (Drosophila)]in EFTs tumors are associated with poor prognosis. The aim of the study was to evaluate the expression levels of these markers in the circulating tumor cells and assess their utility as prognostic markers in EFTs. Methods: 10mL of PB was collected from 24 untreated adult EFTs patients at the diagnosis (period: 2009-2011, median age 30 years). 9 blood specimens from healthy individuals, and 5 untreated frozen EFTs tumor samples were used as controls. 8 EFTs patients presented metastatic disease (M1) at the diagnosis and 5 patients died during the follow-up period. Median follow-up was 22 months. Quantitative reverse transcription PCR (qRT-PCR) for CDH2 and CDT2 expression were performed in triplicate using the TaqMan Gene Expression Assays (Applied Biosystems). Mean expression level of 2 reference genes PSMC4 and EIF2B1 served as endogenous control. The fold change was calculated using ddCt method with pooled healthy individuals as calibrator. Results: At least 2-fold decrease (range 2.11-16.35) of CDH2 expression level in PB has been observed in 79% (n=19) of EFTs patients (p=0.04) as compared to healthy control. In general, CDH2 expression level in PB of EFTs patients was 3.08-fold lower compared with healthy individuals (p=0.07). All of the patients who had M1 at diagnosis (n=8) and who died during the follow-up (n=5) had CDH2 gene underexpression. CDT2 overexpression (FC range 3.28-6.97) has been observed in 17% (n=4) of EFTs patients (p=0.01). Three of these patients had M1 at diagnosis and two of these patients died during the follow-up. PB specimens presenting CDT2 overexpression clustered together with the tumor specimens. Conclusions: Abnormalities in the expression of CDH2 and CDT2 genes in PB specimens are associated with aggressive clinical course of EFTs. Diagnostic and prognostic significance of these biomarkers warrants further studies.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2340-2340
Author(s):  
Katerina Krejcikova ◽  
Katerina Muzikova ◽  
Eva Fronkova ◽  
Marketa Kalinova ◽  
Leona Reznickova ◽  
...  

Abstract Leukemias with the t(9;22) translocation resulting in BCR/ABL fusion protein expression comprise 3–5% of childhood ALL. Despite modern therapeutic regimens, their prognosis is inferior. Minimal residual disease (MRD) based on leukemia-specific immunoglobulin (Ig) and T-cell receptor (TCR) gene rearrangements has become a tool influencing clinical decisions in many therapeutic trials for childhood ALL. The presence of BCR/ABL fusion gene offers a possibility of the fusion transcript detection - a faster and cheaper alternative to Ig/TCR-based MRD monitoring. Up to now, no direct comparison based on a sufficient number of samples has been done. We analyzed 350 follow-up samples from 16 children (aged 4–17 years) with BCR/ABL-positive ALL by Ig/TCR-based real-time quantitative PCR (RQ-PCR) and by reverse-transcriptase (RT) RQ-PCR for BCR/ABL transcripts. Beta-2 microglobulin housekeeping gene was used for cDNA quality normalization. WBC, age, immunophenotype and blast proportion in the bone marrow (BM) and peripheral blood (PB) showed no relation to the initial BCR/ABL level. All children expressed m-BCR/ABL transcript at the time of diagnosis; 3 of 16 children expressed both m-BCR/ABL and M-BCR/ABL transcripts representing the p190 and p210 variant of BCR/ABL protein, respectively. The expression levels of m-BCR/ABL in diagnostic samples differed up to 3 logs, being the lowest in patients expressing both variants of the fusion gene. In 38 samples from those patients, M-BCR/ABL expression was generally higher than m-BCR/ABL expression, being negative by m-BCR/ABL and positive by M-BCR/ABL in 13 samples. For further analysis we used the higher value of m- and M-BCR/ABL as the BCR/ABL MRD level. For the comparison with Ig/TCR-based method, MRD levels in follow-up samples were related to the expression levels in diagnostic samples, which were set to 1. In total, 133 (38%) and 127 (36%) samples were negative and positive by both methods, respectively. The quantitative levels differed by more than 1 log in 46 (36%) double-positive samples, being underestimated by Ig/TCR method in 25 cases and by m-BCR/ABL quantification in 21 cases. With the same sensitivity of both methods we found significantly more false-negative samples by Ig/TCR approach (70 samples) compared to BCR/ABL quantification (20 samples). Altogether, we tested 219 bone marrow (BM), 130 peripheral blood (PB) and 1 cerebrospinal fluid samples. The PB samples showed significantly worse correlation between the two methods compared to BM (p=0.02). Interestingly, some patients had higher MRD levels in PB compared to BM as shown by corresponding BM and PB samples. Our data suggest that BCR/ABL-positive childhood ALL is a biologically heterogeneous group. We show that all diagnostic samples should be screened for the simultaneous m- and M- BCR/ABL expression to avoid false-negativity when using m-BCR/ABL quantification only. In our hands, the quantification of BCR/ABL transcripts appears to be a more reliable method than the generally accepted Ig/TCR-based MRD monitoring as the number of false-negative samples by BCR/ABL quantification is significantly lower. This contention is further supported by our pilot data on transplanted patients where BCR/ABL positivity preceding transplantation seems to be a better predictor of subsequent relapse than Ig/TCR approach. Support: MSM0021620813, MZ00064203 and 62/2004 GAUK CR. KK and KM contributed equally to this work.


Blood ◽  
2003 ◽  
Vol 102 (8) ◽  
pp. 2746-2755 ◽  
Author(s):  
Susanne Schnittger ◽  
Martin Weisser ◽  
Claudia Schoch ◽  
Wolfgang Hiddemann ◽  
Torsten Haferlach ◽  
...  

Abstract To evaluate the prognostic significance of quantitative PML-RARA, AML1-ETO, and CBFB-MYH11 fusion transcript expression, real-time polymerase chain reaction was used to analyze bone marrow samples of 349 such patients at diagnosis and 522 samples of 142 patients also during therapy (total analyses, n = 859; median number of follow-up samples, 4/patient; median duration of assessment, 12 months). Lower expression levels at diagnosis correlated with better overall and event-free survival in all 3 leukemia subtypes. By combining the median expression ratio after consolidation therapy and the 75th percentile of the expression ratio at diagnosis, a new score was established that separates a group with 100% EFS from a significantly worse group (P < .0001) in each of the 3 acute myeloid leukemia subgroups. Eight patients showed increasing levels of expression during follow-up and all had relapse. In conclusion, patients at high risk for treatment failure can be identified by high levels of fusion gene expression at diagnosis or less than 3 logs of tumor reduction during the first 3 to 4 months of therapy. By combining the transcription ratios at these 2 checkpoints, a new powerful prognostic score has been established.


Blood ◽  
1997 ◽  
Vol 90 (3) ◽  
pp. 1321-1325 ◽  
Author(s):  
Giovanna Meloni ◽  
Daniela Diverio ◽  
Marco Vignetti ◽  
Giuseppe Avvisati ◽  
Saveria Capria ◽  
...  

Abstract Reverse-transcription polymerase chain reaction (RT-PCR) of the PML/RARα fusion gene may predict relapse in acute promyelocytic leukemia (APL) patients in hematologic complete remission (CR). We have prospectively studied by RT-PCR 15 PML/RARα+ APL patients undergoing autologous bone marrow transplantation (ABMT) in second CR. The median time of first CR duration was 12 months (range, 6 to 40). All patients were reinduced with all-trans retinoic acid (ATRA), followed in 12 of 15 cases by mitoxantrone and Ara-C as consolidation. Fourteen patients received the BAVC (BCNU, Ara-C, m-AMSA, and VP-16) schedule as conditioning regimen. Unpurged marrows were collected immediately before conditioning treatment, analyzed by RT-PCR, and reinfused at median of 2 months (range, 2 to 7) from the achievement of second CR. Seven patients were PCR+ and eight PCR− for PML/RARα in their pretransplant marrows. All seven patients of the former group remained PCR+ during the follow-up and relapsed at a median time of 5 months (range, 2 to 9) from ABMT and 9 months (range, 4 to 14) from second CR. Of the eight PCR− patients, all remained PCR− during the follow-up controls. One patient relapsed at 10 months from ABMT, one died of a secondary (PML/RARα−) leukemia, and six are in hematologic and molecular remission at a median time of 28 months (range, 15 to 60) after ABMT and 32 months (range, 17 to 62) from second CR. Our results indicate that, in APL patients in second CR, ABMT with PML/RARα− marrow cells is likely to result in prolonged clinical and molecular remissions. Conversely, patients who test PCR+ after reinduction necessitate the use of alternative aggressive approaches, including unrelated allogeneic transplant.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1096-1096
Author(s):  
Amos Toren ◽  
Rachel Rothman ◽  
Bella Bielorai ◽  
Malka Reichart ◽  
Ninette Amariglio ◽  
...  

Abstract The TEL/AML1 fusion gene is the most common gene rearrangement in pediatric acute lymphoblastic leukemia (ALL). Although considered to be a low risk leukemia it has a 20% risk of late relapse. The coexistence of different sub clones at diagnosis, based on polymerase chain reaction (PCR) studies of Ig/TCR gene rearrangement, was recently reported in this subtype of ALL. Their different response to chemotherapy may explain the emergence of certain sub clones at relapse, and may serve as a marker for minimal residual disease follow-up. Several chromosomal rearrangements such as t(9;22), t(8;21), inv(16) and rearrangements of the MLL gene are frequently associated with submicroscopic deletions and some of them have prognostic significance. Such deletions were not reported in t(12;21) positive ALL. Bone marrow cells from 76 pediatric patients with ALL at diagnosis were analyzed for the presence of the TEL/AML1 fusion gene by interphase fluorescence in situ hybridization (FISH). We used a new system of combined analysis enabling a very large-scale study of the cells of interest with regard to morphology, FISH and immunophenotyping. Fourteen patients were positive for the translocation. Four of them had several sub clones associated with various combinations of additional chromosomal abnormalities. The most striking was an atypical and unexpected hybridization pattern consistent with a submicroscopic deletion of the 5′ region of the AML1 breakpoint (intron2) not previously reported. We describe the use of a larger probe for AML1 (AML1/ETO) to exclude the possibility of insertion of TEL into the AML1 region without breakage and to reduce the false positivity due to optical fusion. This may enable a better monitoring of minimal residual disease in cases with submicroscopic deletion. All patients had some sub-clones with TEL deletion. Other abnormalities included trisomy and tetrasomy 21 as well as double TEL-AML1 fusion. The analysis of numerous sub-clones at presentation in these patients suggests clonal evolution at an early stage of the disease. These sub-clones may have different sensitivities to chemotherapy, and some of them may reappear at relapse. The frequency of AML1 deletion in t(12;21) in addition to other chromosomal abnormalities, is unknown. The involvement of these findings in the generation of leukemic sub clones, their prognostic significance and role in minimal residual disease follow-up deserves further studies in a large number of patients and a longer follow-up.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2989-2989
Author(s):  
Granada Perea ◽  
Adriana Lasa ◽  
Anna Aventin ◽  
Alicia Domingo ◽  
Neus Villamor ◽  
...  

Abstract Objectives: To analyze MRD in 65 patients (pts) with good prognosis AML: 30 t(8;21) and 35 inv(16), using both FC and RT-PCR, and to investigate the prognostic value of MRD in the pts outcome. Methods: MRD was monitored in CR pts (n=55) by FC in 101 follow-up samples obtained after various cycles of treatment, as follows: 40 post-induction (ind), 30 post-intensification (int) and 31 at the end of treatment (ttm), and by RT-PCR in 76 samples: 31, 23 and 22, respectively. In 35 pts the two techniques were applied at the same time of the ttm. MRD by FC was assessed using fixed combinations of three monoclonal antibodies. AML1/ETO and CBFb/MYH11 were analyzed following the BIOMED protocol. Results: Twenty-seven percent (n=15) of CR pts relapsed: 6 with t(8;21) and 9 with inv(16). The mean MRD by FC was 1.1% after ind, 0.2% after int and 0.1% at the end of ttm. At the end of ttm, the MRD detected by FC in relapsed and not relapsed pts were significativaly different: 0.3% vs 0.08% (p=0.002). By RT-PCR, the mean of fusion transcript copies/ablx104 differed between relapsed and nonrelapsed pts: 2385 vs 122 (p=0.001) after ind, 56 vs 7.6 after int (p=0.0001) and 75 vs 3.3 (p=0.0001) at the end of ttm. Relapses were more commonly observed in those pts with FC MRD level >0.1% at the end of ttm than in pts with ≤0.1%: 50% vs 12% (p=ns); likewise, using RT-PCR, a cutoff level of >10 copies at the end of ttm correlated with high risk of relapse: 80% of pts with RT-PCR >10 relapsed compared to 12% of pts with levels <10 (p=0.009). The overall survival (OS) probability was 86% for pts with CF MRD ≤0.1 at the end of ttm and 0% for pts with MRD >0.1 (p=0.1) and the leukemia free survival (LFS) was 78% and 44%, respectively (p=0.05). For pts with RT-PCR ≤10 at the end of ttm, the OS was 100% and for pts with RT-PCR >10 it was 30% (p=0.007) and the LFS was 87% and 20%, respectively (p=0.001). MRD was identified after ind in 55% of relapsed pts and at the end of ttm in 83% of relapsed pts. Only 1 pt (1/13) with FC MRD <0.1 and RT-PCR <10 at the end of ttm relapsed. For patients in complete remission, the mean copy level of chimeric transcript was higher for pts with t(8;21) than for those with inv(16): 30.2 vs 17.4 (p=0.0001). Comments: In tandem analysis of MRD by FC and RT-PCR could improve MRD detection in AML pts.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4130-4130 ◽  
Author(s):  
Sabine Strehl ◽  
Margit König ◽  
Katharina Spath ◽  
Markus Pisecker ◽  
Georg Mann

Abstract T-cell acute lymphoblastic lymphoma/leukemia is frequently associated with recurrent genetic aberrations that result in the deregulation of transcription factors. In this respect, BCL11B plays a key role in the differentiation and survival during T-cell development. The 3′-located regulatory elements of BCL11B are juxtaposed to TLX3 by a cryptic t(5;14)(q35;q32) in approximately 20% of childhood T-ALL, which leads to inappropriate expression of TLX3. BCL11B can also fuse to TRDC through an inv(14)(q11.2q32.31) resulting in the expression of a BCL11B-TRDC fusion transcript in the absence of wild-type BCL11B. Moreover, a t(6;14) involving BCL11B and the 6q26 region has been described. We have identified a novel BCL11B rearrangement in a case of childhood T-cell lymphoblastic lymphoma. Cytogenetics detected a t(14;17)(q32;q21) and subsequent FISH analysis using BCL11B-spanning and BCL11B 3′-breakpoint-cluster-region flanking BAC clones revealed that BCL11B itself was not disrupted. However, a translocation breakpoint downstream of the BCL11B was observed suggesting the activation of a juxtaposed gene usually residing at 17q by the transcriptional regulatory elements of BCL11B. To narrow down the breakpoint at 17q a FISH-based chromosome-walking strategy using a set of chromosome 17q-specific BACs was employed. A BAC clone encompassing - from centromere to telomere - the genes RAB5C (a member of the RAS oncogene family), KCNH4 (potassium voltage-gated channel, subfamily H (eag-related), member 4), HCRT (hypocretin (orexin) neuropeptide precursor), GHDC (GH3 domain containing; LGP1), STAT5B (signal transducer and activator of transcription 5B), and the 5′-end of STAT5A showed a split signal indicating that one of these genes was juxataposed to the BCL11B enhancer. RAB5C, KCNH4, GHDC, and STAT5B are transcribed in a telomere-centromere orientation, whereas STAT5A shows the opposite transcriptional direction. Together with the FISH pattern observed these data suggested that STAT5A was the most likely candidate gene that might be inappropriately expressed via the regulatory elements of BCL11B. However, semi-quantitative expression analysis showed that neither STAT5A nor STAT5B were significantly upregulated in the affected lymph node as compared to normal bone marrow, peripheral blood, and thymus. In fact, compared to the expression levels in the other tissues STAT5A seemed to be expressed at lower levels. Thus, also the expression levels of RAB5C, KCNH4, and GHDC were analyzed. KCNH4 expression was almost undetectable in bone marrow, peripheral blood, and thymus and for all three genes no elevated expression was observed in the T-cell lymphoma. Owing to the unchanged expression of these genes also the transcription level of STAT3, which is localized further distal to the breakpoint determined by FISH was analyzed, and similar to STAT5A showed lower expression. However, depletion of STATs usually results in reduced cell viability and apoptosis. Together, our data suggest several scenarios: rearrangements of the region containing the remote enhancer of BCL11B are not necessarily accompanied by high expression of a gene juxtaposed into the close vicinity, expression levels of the juxtaposed gene may be just modulated rather than strongly enhanced, the presence of a more complex translocation undetectable by cytogenetics that results in the overexpression of a gene not obviously affected by the translocation or the generation of a fusion gene.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1755-1755
Author(s):  
Grzegorz Helbig ◽  
Malgorzata Calbecka ◽  
Justyna Gajkowska ◽  
Andrzej Moskwa ◽  
Alina Urbanowicz ◽  
...  

Abstract Background. A small proportion of patients with hypereosinophilic syndrome (HES) demonstrate the presence of an interstitial deletion in chromosome 4 leading to the creation of the imatinib-responsive fusion gene- FIP1L1-PDGFRA (F/P). Recently, we showed that a single weekly dose of imatinib is sufficient to induce and maintain remission of chronic eosinophilic leukemia (CEL) with detectable F/P transcript. Here, we present data from 12 patients CEL and HES, 11 of which were F/P positive, who achieved a rapid complete haematologic remission (CHR) with daily imatinib treatment and remission was then maintained with a weekly imatinib schedule. Design and methods. Twelve patients, 11 out of 12 with detectable F/P were treated with imatinib at the initial doses varies between 100–400mg. There were 10 male and 2 female with a median age of 57 years (19–80). Median time to start imatinib was 23 months (1–204 months). The imatinib dose was de-escalated while patients remained in haematologic remission. As a response maintenance, once weekly imatinib was established in all cases. Results. All studied patients achieved a complete haematologic remission (CHR) and 100% of cases with detectable F/P fusion gene before imatinib, demonstrated a molecular remission determined by reverse transcriptase polymerase chain reaction (RT-PCR) analysis. The breakpoints occured within exon 12 of PDGFRA whereas breakpoints dispersed across the FIP1L1 locus occuring between exons 10 and 13. Median time to achieve CHR was 13 days (4–90), and median time to molecular remission was 9 months (4–24). As a remission maintenance, imatinib doses were set at 100mg weekly in 9 pts and 200mg weekly in 3. With a median follow-up of 21 months (8–49 months) all pts remain in CHR. The FIP1L1-PDGFRA is undetectable in 11 patients by RT-PCR. Conclusions. Imatinib at weekly dosage may induce and maintain remission in patient with CEL expressing F/P fusion gene. This strategy appears to be safe and cost savings.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2529-2529
Author(s):  
Marketa Zaliova ◽  
Leona Reznickova ◽  
Eva Fronkova ◽  
Katerina Krejcikova ◽  
Katerina Muzikova ◽  
...  

Abstract Minimal residual disease (MRD) monitoring is an essential tool for current leukaemia therapy. The only standard method for MRD monitoring in childhood ALL is the quantitative detection of clonal immunoglobulin (Ig) and T-cell receptor (TCR) genes rearrangements. The quantitative detection of fusion genes or transcripts provides an alternative option for MRD monitoring. We aimed to compare the significance of these MRD methods by parallel monitoring of fusion transcripts/genes and Ig/TCR targets during the follow-up of children from the three most common ALL genotype groups. We analysed 117, 109 and 191 bone marrow samples from 28 TEL/AML1-positive, 7 MLL fusion-positive and 16 BCR/ABL-positive patients, respectively. To keep the comparability of different MRD approaches, we used qPCR detection systems with similar sensitivity (at least 10−4), we adopted ESG-MRD-ALL principles for MRD quantification and we related the MRD level in follow-up samples to the diagnostic level for all MRD methods. We found a very good correlation of fusion transcript- and Ig/TCR-based approaches (R2=0.903) with only 7% of samples differing by more than 1 log in a cohort of TEL/AML1-positive patients. A good correlation was also found between fusion transcript- and Ig/TCR-based MRD in MLL fusion-positive patients (R2=0.8419). Only 10% of samples differed by more than 1 log, being underestimated by Ig/TCR in 4.5% and by MLL-fusion transcript in 5.5%. For the follow-up of MLL-fusion-positive patients we further employed the monitoring of MLL-fusions on genomic level. The MRD based on genomic MLLfusion genes showed a very good correlation with Ig/TCR -based method (R2=0.9124) with only 5% of samples differing by more than 1 log, and it also closely correlated with MLL-fusion transcript levels (R2=0.9195). Strikingly, in BCR/ABL-positive patients we found a limited correlation of fusion transcript-based and Ig/TCR-based MRD (R2=0.6880) with 1/3 (34%) of samples differing by more than 1 log. In contrast to the MLL cases, the underestimation of MRD by individual methods was “asymmetrical”: 8% of the discordant samples had higher MRD measured by Ig/TCR and 26% by BCR/ABL transcript. Despite identical sensitivity of both methods, in 19% of samples the MRD positivity was revealed only by BCR/ABL approach while Ig/TCR approach gave negative results. Detailed analysis showed clinical significance of the discordant BCR/ABL vs. Ig/TCR MRD information. Altogether, 13 relapses occurred during the follow-up of our cohort. We compared number of BCR/ABL and Ig/TCR -positive samples among all BM specimens taken 6 and 12 months before relapse. While the majority of samples preceding relapse were BCR/ABL-positive (14/18 and 22/36 six and twelve months before relapse) only a minority of samples showed Ig/TCR positivity (7/18 and 12/36, respectively). The non-equal distribution of the BCR/ABL and Ig/TCR-positive samples was statistically significant (p=0.04 and p=0.03 for the two time-points, respectively). Our study shows, that in TEL/AML1 and MLL fusion-positive patients, fusion gene/transcript-based MRD monitoring provides information highly concordant to the standard Ig/TCR approach and thus it is useful as a complementary method in patients with absent or inadequate Ig/TCR targets (particularly in MLL cases where clonal Ig/TCR rearrangements are rare). The situation is different in BCR/ABL patients, where the MRD information from both approaches is discordant in a high subset of samples. This result probably reflects the dissimilar biology of this ALL subtype and the fact, that BCR/ABL-positive (prae-)leukaemic stem cell is different and multilineage involvement more common. Thus, in some cases, the fusion transcript monitoring reveals the existing pool of cells that increase the risk of relapse despite the Ig/TCR negativity. We conclude that MRD in all BCR/ABL–positive patients should be monitored not only by the standard Ig/TCR approach but in parallel also by the quantitative fusion transcript-based detection. Support: MSM0021620813, MZO00064203.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4619-4619
Author(s):  
Heidi Mocikova ◽  
Sona Pekova ◽  
Lenka Zejskova ◽  
Martin Spacek ◽  
Tomas Kozak

Abstract Abstract 4619 Background. Expression of cyclin D1 demonstrated by immunohistochemistry is seen in 95% of mantle cell lymphomas and in other lymphoproliferative diseases including 10% of chronic lymphocytic leukemias (CLL). This study analyzed the impact of cyclin D1 positivity on time to treatment (TTT) and overall survival (OS) measured by RT PCR in newly diagnosed CLL patients and correlation with reported prognostic factors. Patients and methods. Level of cyclin D1 (quantitative real-time RT-PCR with a specific TaqMan flurescent hybridization probe) mRNA expression was analysed in 72 samples (57 peripheral blood and 15 bone marrow) from patients with newly diagnosed CLL. Cyclin D1 expression (cut-off according to ROC curve >3 over the threshold expression in healthy donors) was reported as positive. Fisher's exact test was used to analyze the relationship of cyclin D1 positivity and prognostic factors: del17p, del11q, unmutated IgVH, trisomy 12, ZAP 70 and CD38 positivity, elevated B2microglobulin, elevated LDH and lymphocyte doubling time (LDT) <6 months. The comparison of time to treatment (TTT) and prognostic factors with cyclin D1 positivity was calculated via Spearman correlation coefficient. Survival curves were calculated by Kaplan-Meier survival analysis and comparison between subgroups was performed by the log-rank test. Results. Cyclin D1 was positive in 29 (40%) CLL patients. Although cyclin D1 was not statistically significant for TTT (P=0,145), a trend was observed, suggesting a negative prognostic impact of cyclin D1 overexpression in CLL. Following variables correlated significantly with TTT: del17p (P=0.037), del11q (P=0.003), unmutated IgVH (P=0.004), trisomy 12(P=0.024), positive CD38 (P=0.014), elevated B2microglobulin (P<0.001), elevated LDH (P<0.001) and LDT <6 months (P<0.001). Del 17p, del 11q, trisomy 12 and elevated B2microglobulin were independent factors for TTT in the multivariate analysis. None of these factors were significant for overall survival due to the short follow-up. Conclusion. Cyclin D1 measured by RT-PCR from peripheral blood or bone marrow has no statistically singificant impact on TTT or OS, though a trend pointing to cyclin D1 overexpression in CLL as a negative prognostic marker can be suggested. This data should be confirmed on a larger cohort of patients with a longer follow-up. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document