scholarly journals Comparative Analysis between RQ-PCR, Digital-Droplet-PCR and Next-Generation-Sequencing (NGS) of Immunoglobulin/T-Cell Receptor Gene Rearrangements to Monitor Minimal Residual Disease in Adult Acute Lymphoblastic Leukemia Patients

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2828-2828 ◽  
Author(s):  
Irene Della Starza ◽  
Lucia Anna De Novi ◽  
Alessandra Santoro ◽  
Salemi Domenico ◽  
Marzia Cavalli ◽  
...  

Abstract Background. Minimal residual disease (MRD) is the strongest prognostic factor in both children and adults with acute lymphoblastic leukemia (ALL). Currently, it is most widely monitored by molecular methods based on real-time-quantitative-PCR (RQ-PCR). Digital-droplet-PCR (ddPCR) and next-generation-sequencing (NGS) represent advanced tools that have the potential to overcome some limitations of standard approaches and potentially provide additional benefits. We analyzed adult ALL follow-up (FU) samples by RQ-PCR, ddPCR and NGS in order to better define the discriminating power of these novel methods. Patients and Methods. Thirty adult ALL patients enrolled in the GIMEMA LAL 1913 protocol and their 83 FU bone marrow (BM) samples were studied. All patients received homogeneous induction/early consolidation chemotherapy, with concurrent MRD analysis at four time-points, to optimize risk classification and support risk/MRD-oriented therapy. RQ-PCR analyses followed the EuroMRD Consortium guidelines (van der Velden, 2007), ddPCR was performed as published (Della Starza, 2016; Cavalli, 2017) and NGS, as previously described (Faham, 2012; Kotrova M, 2017). Results. By MRD RQ-PCR analysis, 19/83 samples were positive and quantifiable (Q), 9/83 were positive not-quantifiable (PNQ) and 55/83 were negative (NEG). By MRD ddPCR analysis, 27/83 samples were Q, 1/83 sample was PNQ and 55/83 proved NEG. Comparing the results of the two methods, we observed that MRD detection was concordantly positive or negative in 81% (67/83) of FU samples, while 19% (16/83) samples were classified as discordant. Most of the discordances occurred in samples with low levels of disease, i.e. PNQ or NEG: 9/83 were RQ-PCR PNQ, 4 of which were Q by ddPCR and 5 were ddPCR NEG. In the remaining 7 discordant FU samples, 5 were RQ-PCR NEG/ddPCR Q, 1 sample was RQ-PCR Q /ddPCR NEG and 1 sample was RQ-PCR NEG/ddPCR PNQ. The use of ddPCR significantly reduced the proportion of PNQ samples if compared to RQ-PCR - 1/83 (3%) vs 9/83 (15%) - respectively (p=0.0179), increasing the proportion of Q samples: 27/83 (33%) vs 19/83 (23%). It is worth noting that ddPCR also quantified the levels of disease in 9% (5/55) of samples, that were RQ-PCR NEG (Table 1). MRD analysis was also performed by NGS in 41 samples from 15 patients: 18/41 samples proved Q and 23/41 were NEG. Comparing the MRD detection obtained by both ddPCR and NGS, we observed a concordant result in 98% (40/41) of samples; only 1 sample was ddPCR NEG and NGS Q with a MRD level of 1x10-5. The concordance between RQ-PCR and NGS was 78% (32/41 samples). Moreover, among these 41 samples 9 (from 7 patients) were discordant between RQ-PCR and ddPCR in the first comparative analysis: in 4 RQ-PCR-NEG FU samples, 3 were Q by both ddPCR and NGS, 1 was ddPCR NEG and NGS Q, with a MRD level of 10- 5; 1 subsequent relapse was observed; 4 FU samples that were RQ-PCR-PNQ/ddPCR-Q, were Q also by NGS; 1 subsequent relapse was observed. Finally, 1 RQ-PCR-PNQ sample was negative by both ddPCR and NGS, and no recurrence has so far been observed. Moreover, in the cohort of samples analyzed only by RQ-PCR and ddPCR, in 1 RQ-PCR NEG/ddPCR Q sample a relapse was observed, while the only case that was RQ-PCR Q/ddPCR NEG has so far not relapsed. Notably, 2 of the 3 relapses were documented in patients who were, at decisional treatment TPs, RQ-PCR PNQ or NEG and ddPCR/NGS Q. Conclusions. When MRD levels are very low, it can be difficult to dissect if the not-quantifiable signal observed by PCR is due to few residual leukemic cells or to a non-specific amplification of normal DNA. The superior sensitivity and accuracy of ddPCR and NGS could be instrumental to univocally define these samples, which presently represent a problematic gray area in the clinical practice of MRD-driven protocols and might be associated with clinical relapse: indeed, among 83 FU samples analyzed we observed 3 relapses, whose FU samples were classified as PNQ or NEG by RQ-PCR, but proved Q by ddPCR and/or NGS. At variance, no relapses were recorded in patients whose FU samples were defined RQ-PCR-PNQ, but proved ddPCR/NGS NEG. Moreover, in 2/3 relapsed cases the change of MRD status (PNQ or NEG vs Q) could have led to a switch in risk classification and therefore in a treatment change. Further studies with a larger number of discrepant cases and a longer FU time will allow to conclusively define the clinical application and implication of these new methods. Disclosures Chiaretti: Shire: Consultancy; Pfuzer: Consultancy; Amgen: Consultancy; Incyte: Consultancy. Foà:NOVARTIS: Speakers Bureau; ROCHE: Other: ADVISORY BOARD, Speakers Bureau; CELTRION: Other: ADVISORY BOARD; ABBVIE: Other: ADVISORY BOARD, Speakers Bureau; CELGENE: Other: ADVISORY BOARD, Speakers Bureau; JANSSEN: Other: ADVISORY BOARD, Speakers Bureau; INCYTE: Other: ADVISORY BOARD; AMGEN: Other: ADVISORY BOARD; GILEAD: Speakers Bureau.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1544-1544 ◽  
Author(s):  
Irene Della Starza ◽  
Vittorio Nunes ◽  
Federica Lovisa ◽  
Daniela Silvestri ◽  
Marzia Cavalli ◽  
...  

Abstract Introduction. Minimal residual disease (MRD) is the most powerful prognostic factor in acute lymphoblastic leukemia (ALL). Currently, real-time quantitative PCR (RQ-PCR) is the most widely used molecular method for MRD assessment, rigorously standardized within the EuroMRD consortium. According to the EuroMRD guidelines (Van der Velden et al. Leukemia 2007), a non-negligible fraction of patients with very low MRD levels are classified as positive not-quantifiable (PNQ), a definition that may result problematic in the clinical practice. Digital-droplet-PCR (ddPCR) allows an absolute quantification without the need of a standard curve and has the potential to overcome some limitations of RQ-PCR. High degrees of efficiency, sensitivity and accuracy have been reported for ddPCR compared to RQ-PCR, but no established guidelines for ddPCR MRD analysis and interpretation have so far been defined and its ability to correctly evaluate very low MRD levels is still under investigation. In the present study, we assessed MRD by ddPCR in pediatric ALL cases classified as PNQ and/or negative by RQ-PCR at days +33 and/or +78 of the AIEOP-BFM ALL 2000 trial, to evaluate the potential of ddPCR for low MRD quantification and patients' risk stratification. Patients and Methods. A total of 211 pediatric ALL patients enrolled in the AIEOP-BFM ALL 2000 trial were included in the study. We analyzed 124 B-lineage ALL patients defined as intermediate risk (IR) who had high positive MRD at day +33 and at day +78 were either PNQ (n=45, Slow Early Responders (SER)) or negative (n=79). A case-control design was applied to 36 B- and T-lineage relapsed ALL patients (cases) who at day +33 had PNQ MRD (n=12, IR) or were negative (n=24, standard risk (SR)) and to matched controls (21 and 30 patients who did not present a relapse). ddPCR analysis was performed as previously published (Della Starza et al, BJH 174, 541-9, 2016), by using 1.5 μg and 3.0 μg DNA of the follow-up samples. In the absence of an international consensus, data have been analyzed using two alternative guidelines; results are reported according to Della Starza et al (BJH 2016). Results. Among 45 SER patients, ddPCR performed on 1.5 μg DNA of PNQ samples at day +78 revealed that 13 were quantifiable (Q), 16 PNQ and 16 negative (NEG) . When 3.0 μg of DNA were used (41/45 samples due to material availability), 12 were Q, 19 PNQ and 10 NEG. Event-free survival (EFS) curves are shown in Fig. 1a. Among the 79 patients with high positive MRD at day +33 but who were negative at day +78, ddPCR on 1.5 μg DNA of day +78 identified 5 as Q, 17 PNQ and 57 NEG. When 3.0 μg DNA was used (77/79 samples), 9 patients were Q, 27 PNQ and 41 NEG. EFS curves are reported in Fig. 1b. When ddPCR was applied to 33 PNQ samples at day +33, 2 were Q, 9 PNQ and 22 NEG; when using 3.0 μg of DNA, 1 was Q, 15 were PNQ and 17 NEG. EFS curves are shown in Fig. 1c. Lastly, ddPCR on 1.5 μg of day +33 DNA of 54 SR patients showed 5 PNQ and 49 NEG, whilst by using 3.0 μg on 53 sample, 7 were PNQ and 46 NEG. Conclusions. Our data demonstrate that ddPCR is a very promising tool for the evaluation of MRD in ALL cases with very low or negative RQ-PCR MRD results. In particular, among SER patients most relapses occurred in cases with quantifiable ddPCR MRD at day +78, while patients with negative or PNQ MRD by ddPCR at day +78 had a better outcome. Based on these results, high-risk treatment could be offered only to ddPCR quantifiable cases. Among patients with highly positive MRD at day +33 and negative at day +78, the small number of cases with quantifiable disease by ddPCR at present does not allow to establish the impact of quantification; consistently with SER patients, the outcome was similar for patients with negative or PNQ MRD by ddPCR at day +78. Similarly, among patients with PNQ MRD by RQ-PCR at day +33, a similar outcome was observed for cases negative or PNQ by ddPCR. Lastly, in most SR patients ddPCR confirmed the negative results of RQ-PCR at day +33, associated with an extremely good kinetics of disease reduction, independently of the MRD PCR method. Overall, our data indicate that ddPCR is as sensitive as RQ-PCR and can provide a potentially more accurate prognostic stratification for cases defined as PNQ MRD by RQ-PCR, in view of its ability to quantify without a standard curve. The application of ddPCR in a prospective clinical protocol with international guidelines is needed to define whether it can result in an overall improvement of pediatric ALL patients' stratification and outcome. Disclosures Foà: AMGEN: Other: ADVISORY BOARD; JANSSEN: Other: ADVISORY BOARD, Speakers Bureau; CELTRION: Other: ADVISORY BOARD; INCYTE: Other: ADVISORY BOARD; ABBVIE: Other: ADVISORY BOARD, Speakers Bureau; ROCHE: Other: ADVISORY BOARD, Speakers Bureau; NOVARTIS: Speakers Bureau; CELGENE: Other: ADVISORY BOARD, Speakers Bureau; GILEAD: Speakers Bureau.


2019 ◽  
Vol 20 (12) ◽  
pp. 2929 ◽  
Author(s):  
Nicoletta Coccaro ◽  
Luisa Anelli ◽  
Antonella Zagaria ◽  
Giorgina Specchia ◽  
Francesco Albano

Acute lymphoblastic leukemia (ALL) is the most common childhood cancer and accounts for about a quarter of adult acute leukemias, and features different outcomes depending on the age of onset. Improvements in ALL genomic analysis achieved thanks to the implementation of next-generation sequencing (NGS) have led to the recent discovery of several novel molecular entities and to a deeper understanding of the existing ones. The purpose of our review is to report the most recent discoveries obtained by NGS studies for ALL diagnosis, risk stratification, and treatment planning. We also report the first efforts at NGS use for minimal residual disease (MRD) assessment, and early studies on the application of third generation sequencing in cancer research. Lastly, we consider the need for the integration of NGS analyses in clinical practice for genomic patients profiling from the personalized medicine perspective.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2916-2916 ◽  
Author(s):  
Nicholas J. Short ◽  
Hagop M. Kantarjian ◽  
Jeffrey L. Jorgensen ◽  
Farhad Ravandi ◽  
Musa Yilmaz ◽  
...  

Abstract Background: Minimal residual disease (MRD) assessment by multiparameter flow cytometry (MFC) is prognostic for survival in newly diagnosed patients (pts) with acute lymphoblastic leukemia (ALL). The significance of achieving MRD negativity in the relapsed/refractory setting is less clear. Methods: Between 6/2010 and 5/2015, we identified 130 adult pts with relapsed/refractory B-cell ALL treated at our institution with either inotuzumab ozogamicin (n=75), blinatumomab (n=20) or mini-hyper-CVD plus inotuzumab ozogamicin (HCVD+InO; n=35) in either salvage 1 (S1; n=68) or salvage 2 (S2; n=62). MRD by MFC was assessed on remission bone marrow specimens at the time of achievement of CR/CRp/CRi. The MRD assay used a 15-marker, 6-color panel with a sensitivity of ≤0.01%. Results: Of the initial 130 pts, 78 (60%) achieved morphological response with a median time to response of 30 days (range, 13-99 days) and are the subject of this analysis. Of the 78 responding pts, 41 (53%) received inotuzumab, 11 (14%) blinatumomab, and 26 (33%) HCVD+ino. 46 pts (59%) were in S1 and 32 (41%) in S2. The median number of cycles to best response was 1 (range, 1-3). MRD negativity was achieved in 41 pts (53%). MRD negativity rates for pts in CR, CRp, and CRi were 57%, 53%, and 16%, respectively. Among pts who achieved remission, MRD negativity was achieved in 17 pts (41%) with inotuzumab, 8 (73%) with blinatumomab, and 16 (62%) with HCVD+InO (P=0.10). 26 pts (57%) in S1 and 15 (47%) in S2 became MRD-negative (P=0.40). The median follow-up duration was 27 months (range, 6-55 months). The median event-free survival (EFS) was 12 months in pts who achieved MRD negativity vs. 6 months in those who remained MRD-positive (P=0.09). The median overall survival (OS) was 17 months versus 9 months, respectively (P=0.18). Among pts in S1, achieving MRD negativity was associated with a longer EFS (median 18 months versus 7 months; 2-year EFS rate 46% versus 17%; P=0.06; Figure 1A) and OS (median 27 months versus 9 months; 2-year OS 52% versus 36%; P=0.15; Figure 1B). EFS and OS were similar in S2 regardless of MRD response. As expected, among pts who achieved MRD negativity, those in S1 had longer EFS (median 18 months vs. 5 months; P=0.001) and OS (median 27 months vs. 7 months; P=0.01) compared to those in S2. In contrast, for pts who remained MRD-positive, EFS and OS were similar regardless of salvage status (P=0.41 and P=0.39, respectively). In a 2-month landmark analysis of 64 pts, survival >2 years was observed in all groups of pts regardless of salvage treatment, salvage status or MRD status. 42 (66%) of the pts in this analysis underwent allogeneic stem cell transplantation (alloSCT). EFS and OS did not significantly differ between pts who did or did not undergo alloSCT, although a clear trend for improved long-term survival with alloSCT was observed. Among pts who achieved MRD negativity, the median EFS was 17 months and 12 months, and 2-year EFS rates were 46% and 28% for pts who underwent alloSCT vs. those who did not (P=0.24). The median OS was 24 months and 23 months, and 2-year OS rates were 55% and 46%, respectively (P=0.41). Pts who achieved MRD negativity after S1 treatment and then underwent alloSCT had the best outcomes. Of the 22 pts who achieved MRD negativity after S1 treatment, the median EFS for pts who underwent alloSCT (n=14) compared to those who did not (n=8) was not reached vs. 18 months, and the median OS was not reached vs. 27 months, respectively (P=0.28 for both). Among the 14 pts who achieved MRD negativity after S1 treatment and subsequently underwent alloSCT, 10 (71%) are still alive with a median follow-up of 24 months (range, 5-55 months). Conclusions: In patients with relapsed/refractory ALL, achievement of MRD negativity is associated with improved outcomes. Patients with relapsed/refractory ALL who achieve MRD negativity in S1 can achieve excellent long-term survival, especially if alloSCT is performed. Disclosures O'Brien: Pharmacyclics, LLC, an AbbVie Company: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria. Cortes:ARIAD: Consultancy, Research Funding; BMS: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Teva: Research Funding. DiNardo:Daiichi Sankyo: Other: advisory board, Research Funding; Novartis: Other: advisory board, Research Funding; Abbvie: Research Funding; Celgene: Research Funding; Agios: Other: advisory board, Research Funding. Jain:Genentech: Research Funding; Incyte: Research Funding; BMS: Research Funding; Celgene: Research Funding; Infinity: Research Funding; Pharmacyclics: Consultancy, Honoraria, Research Funding; Servier: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Novimmune: Consultancy, Honoraria; Abbvie: Research Funding; Seattle Genetics: Research Funding; ADC Therapeutics: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria, Research Funding. Konopleva:Cellectis: Research Funding; Calithera: Research Funding. Jabbour:ARIAD: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Novartis: Research Funding; BMS: Consultancy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5201-5201
Author(s):  
Vasudha Rao ◽  
Katherine Clesham ◽  
Jack Luke Bartram ◽  
Phil Ancliff ◽  
Sara Ghorashian ◽  
...  

Abstract Introduction: A recent phase I/II study of blinatumomab in children with relapsed/refractory B cell acute lymphoblastic leukaemia (B-ALL), found 39-51% achieved complete, often MRD negative, remission after two cycles of therapy. Higher responses were observed in patients with less than 50% bone marrow blasts, compared to greater than 50% (55.6% vs 32.7% (95% CI 30.8-78.5 and 20.3-47.1 respectively)1. Furthermore, an adult study showed complete minimal residual disease (MRD) response rates of 78% when blinatumomab was used to treat MRD-positive ALL in haematological remission 2. Hence response rates (and toxicity) to blinatumomab are highly correlated with pre-treatment disease burden. We present preliminary data showing an excellent response to blinatumomab in children and young adults with resistant B-ALL who had persistent MRD, or after debulking chemotherapy to achieve a partial remission. Methods: Eleven patients were identified through a national survey. The mean age of patients was 10 years (range 0.7-22 years, 3 infants and 1 Down syndrome ALL). All patients had B-lineage ALL which was CD19 positive. None had active CNS disease at the point of receiving blinatumomab. Prior to administration of blinatumomab, all patients either had persistent MRD following several courses of intensive chemotherapy or received debulking chemotherapy for heavier marrow infiltrates. Pre-blinatumomab chemotherapy to which the patients had failed to obtain an adequate MRD response included UKALL 2011 Regimens A or C, UKALL R3, Interfant 06 or NOPHO high risk blocks. Patients received 5-15 µg/m2 of blinatumomab for 1-2 cycles prior to definitive therapy. Results: Pre-blinatumomab, all patients except one were in morphological remission with MRD measurable by PCR (0.003-1%), the remaining patient had 9% marrow disease by morphology. After 1-2 cycles of blinatumomab all patients had negative MRD when measured by flow cytometry and/or by PCR, giving a 100% response rate. This was followed by Haemopoietic stem cell transplant (HSCT) in nine patients and the remaining two are awaiting transplant. Further data on patient characteristics, CNS status, relapse and survival outcome are being collected and will be presented at the meeting. Minimal toxicity was observed; of the seven patients in whom toxicity data were available, three had grade 1 CRS, which resolved spontaneously without interruption of therapy or treatment with corticosteroids or Tocilizumab. One patient reported grade 1 neurotoxicity. This preliminary UK experience demonstrates that excellent MRD response is observed with minimal toxicity in children and young adults who receive blinatumomab for persistent MRD or after debulking chemotherapy. This provided a bridge to transplant in patients who would otherwise not have benefited from the procedure because of persistent MRD. We are planning to extend these observations by undertaking a study of this strategy in first high-risk B-ALL relapse. Stackelberg Av, Locatelli F, Zugmaier G, et al. Phase I/Phase II Study of Blinatumomab in Pediatric Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia. Journal of Clinical Oncology. 2016;34(36):4381-4389. Gokbuget N, Dombret H, Bonifacio M, et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood. 2018;131(14):1522-1531. Disclosures Ghorashian: Celgene: Other: travel support; Novartis: Honoraria. Marks:Novartis: Consultancy; Pfizer: Consultancy; Amgen: Consultancy. Vora:Amgen: Other: Advisory board; Novartis: Other: Advisory board; Jazz: Other: Advisory board; Medac: Other: Advisory board; Pfizer: Other: Advisory board.


Sign in / Sign up

Export Citation Format

Share Document