Impact of Minimal Residual Disease after Allografting Detected By JAK2V617F, MPL or Calreticulin in Myelofibrosis Patients

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4343-4343
Author(s):  
Nicolaus Kroeger ◽  
Anita Badbaran ◽  
Tatjana Zabelina ◽  
Maximilian Christopeit ◽  
Christine Wolschke ◽  
...  

Abstract Introduction: Residual minimal disease (MRD) in hematological malignancies has become a valid tool to predict clinical relapse after allogeneic stem cell transplantation (ASCT). Methods and Patients: We screened 154 patients with myelofibrosis who underwent ASCT for molecular residual disease by qPCR or next-generation sequencing (NGS) for JAK2V617F, MPLW515L and MPLW515K or Calreticulin (L367fs*46, and K385fs*47) mutations in peripheral blood (PB) on days +100 and +180 after transplantation. Out of 154 pts, 103 were JAK2V617F (n=101), 31 Calreticulin (CALR), and 4 MPL mutated, while 13 pts were triple negative. 136 pts could be followed after ASCT with one molecular marker. The median age of the pts was 58 years (range, 32-75 y). Patients had either primary myelofibrosis (n= 90), post ET/PV myelofibrosis (n=40), myelofibrosis in acceleration or were transformed to AML (n=6). Conditioning mainly relied on a busulfan-based reduced-intensity regimen. Donor were HLA-identical sibling (n=26), matched unrelated (n=71) or mismatched unrelated (n=39). JAK2V617F, MPL, and CALR mutations were measured by usage of Taqman PCR or in case of CALR Type 2 mutation by digital PCR on day +100 and day +180 from PB as described elsewhere. Results: After a median follow up of 78 months (range, 49-101 months) the 5-year estimated overall survival was 60% (95% CI: 50-70%) and the cumulative incidence of relapse at 5 years was 26% (95% CI: 18-34%) for the entire study population. On days +100 and +180 after transplantation in 27% and 12% of the patients the underlying mutation was still detectable in peripheral blood. The percentage of complete clearance was higher in CALR-mutated patients (96%) in comparison to MPL (75%) and JAKV2617F (70%) mutated pts. Whereas there was a trend for better survival for CALR-mutated patients in comparison to JAK2-mutated patients (71% vs 57%; p=0.48), once a patient achieved molecular remission post-transplant the risk of relapse remained low independently of the underlying mutation. Patients who were alive and without relapse at days +100 or +180 but with still detectable mutation in PB had a significantly higher risk of relapse than those who were molecular negative (62% vs 10%, p<0.001; and 70% vs 10%, p<0.001, respectively). In a multivariate analysis only high-risk disease status (HR 2.5; 95% CI: 1.18-5.25, p=0.016) and detectable MRD at day 180 (HR 8.36, 95% CI: 2.76-25.30, p<0.001) were significant factors for a higher risk of relapse. Conclusions: JAK2V617F, CALR and MPL genetic lesions allow to monitor MRD in around 90% of the myelofibrosis patients after ASCT by qPCR or digital PCR in the PB. Persistence of MRD on days +100 or +180 in peripheral blood can be used to taper immunosuppressive drugs or to apply donor lymphocyte infusion to prevent clinical relapse. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2255-2255
Author(s):  
Carol Moreno ◽  
Neus Villamor ◽  
Dolors Colomer ◽  
Jordi Esteve ◽  
Francesc Bosch ◽  
...  

Abstract Autologous stem cell transplants (auto-SCT) are increasingly performed in patients with CLL. Although this procedure results in a high complete response (CR) rate, most patients eventually relapse. The median time to relapse is around 5 years. Increasing levels of minimal residual disease (MRD), detected by PCR or flow cytometry (FC) are associated with clinical relapse. The early detection of patients likely to relapse shortly after SCT may be useful in the management of these patients. With this background, we analyzed the levels of MRD and its correlation with the risk of relapse and clinical outcome in 19 patients with CLL submitted to auto-SCT. MRD was assessed by FC and quantitative real time PCR of the IgH region using allele-specific oligonucleotides (ASO-PCR) in peripheral blood and/or bone marrow DNA samples obtained before SCT and at different time points thereafter. After SCT, 17 patients achieved CR and 2 partial response. A continuous pattern of relapses was observed and, after a median follow-up of 48 months (range, 11–101), 11/19 patients have progressed. The median number of CLL cells detected prior to SCT was 2.4x10−2 decreasing to 5.31x10−4 at 3–6 months after auto-SCT. No further decrease was observed beyond that point. At 3–6 months after auto-SCT, only 3/17 patients in CR had undetectable levels of disease. Patients with a MRD level >10−3 at this time point (3–6 months after transplant) had a significantly higher risk of progression than those who had less than 10−3 CLL cells. All but one patients with MRD>10−3 have relapsed (7/8) whereas only 4/9 with MRD<10−3 did so. As shown in the figure, median time to progression was significantly shorter in those patients with a higher MRD level (16 vs. 55 months; p=0.003) Figure Figure In conclusion, quantification of MRD within the first 6 months after auto-SCT allows the identification of CLL patients with a high risk of early clinical relapse. These data provide background to investigate whether early treatment, before clinically overt relapse occurs, might be useful in patients with high risk of relapse after SCT.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4672-4672
Author(s):  
Dana Dvorakova ◽  
Zdenek Racil ◽  
Ivo Palasek ◽  
Marketa Protivankova ◽  
Ivana Jeziskova ◽  
...  

Abstract Abstract 4672 Background Mutations within NPM1 gene occurs in about 60% of adult cytogenetic normal AML (CN-AML) and represent the single most frequent molecular aberration in this subgroups of patients. These mutations usually occur at exon 12 and induce most frequently a net insertion of four base pairs. Aims To examine the applicability and sensitivity of DNA-based real-time quantitative polymerase chain reaction (RQ-PCR) with mutation-specific reverse primers and common minor groove binding (MGB) probe and to evaluate whether minimal residual disease levels are of prognostic relevance in CN-AML patients with NPM1 mutations. Methods Patients were treated within different AML trials and follow-up samples of peripheral blood or bone marrow were referred to perform an RQ-PCR. Samples were analysed at diagnosis, during, and after therapy. The NPM1 mutations were A (17 pts), B (1 pt), D (2 pts) and 7 patients with individual rare types. For all cases, levels of minimal residual disease were determined by DNA-based RQ-PCR with mutation-specific reverse primer, one common forward primer and one common MGB probe. The NPM1 mutation value was normalized on the number of albumin gene copies and expressed as the number of NPM1 mutations every 106 genomic equivalents. This assay is highly specific as no wildtype NPM1 could be detected. Maximal reproducible sensitivity was 10 plasmide molecules per reaction. Results A total of 950 samples of bone marrow and/or peripheral blood from 27 patients have been analyzed. Twenty of 27 patients (74%) achieved molecular remission (MR), twenty-six of 27 patients (96%) achieved hematological remission (HR). 6 of 27 (22%) patients achieved HR without MR and one patient failed therapy. 8 of 20 patients (40%) with MR after treatment relapsed at molecular level and except one in all these patients hematological relaps occured (one patient is still in HR with bone marrow blast present, but < 5%). Considering relapsed patients, time from molecular to hematological relapse was 1 to 5 months (median: 3 months). Considering all 14 patients with HR without MR (6 pts) or with molecular relapse (8 pts), in 11 of them hematological relaps occured (79%) and molecular positivity anticipating hematological relaps with median of 3,5 month (1-7 months). 3 of these 14 patients are still in HR. Conclusions Mutations within NPM1 gene are a sensitive marker for monitoring minimal residual disease in CN-AML patients. RQ-PCR using a MGB probe is an efficient approach to long-term follow-up of residual leukemia cells and frequent quantitative monitoring is useful for reliably predicting hematological relapse. Achievement of negativity appears to predict favorable clinical outcome. This work was partially supported by research grant No. MSM0021622430 Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3386-3386
Author(s):  
Yu Wang ◽  
Yanrong Liu ◽  
Yazhen Qin ◽  
Xiao-Jun Huang

Abstract Background Recent results from our prospective multi-center study identified patients with t (8; 21) AML as high-risk according to their MRD status after the second consolidation chemotherapy and allo-HSCT can benefit this part of patients; however, the relapse rate was reported to be 22% even after allo-HSCT for those high-risk patients. To date, there have been no studies to answer the question of whether the specific fusion gene, RUNX1/RUNX1T1 can be used to further distinguish between patients with low and high risks of relapse in allo-HSCT setting, just like the already established standard for MRD measurement in CML and APL. Methods Sixty consecutive AML patients with t (8; 21) and identified as high-risk according to the criteria from our recently published report patients who received allo-HSCT were enrolled between January 2006 and January 2013. Serial MRD monitoring by RQ-PCR post HSCT was done. The impact of MRD monitoring on transplant outcomes was assessed. Results A > 3 log reduction at 1 month after HSCT in RUNX1/RUNX1T1 transcripts from diagnosis, is associated with 2-year CIR of 17% in the 88% of patients achieving it, compared with 43% in the remaining (p=.02). A > 3 log reduction at 2 months after HSCT in transcripts from diagnosis, is associated with a CIR of 9% and LFS of 66% in the 86% of patients achieving it, compared with CIR of 100% and LFS of 0% in the remaining (both p<.001). A > 3 log reduction at 3 months after HSCT in RUNX1/RUNX1T1 transcripts from diagnosis, is associated with a CIR of 11% and LFS of 73% in the 78% of patients achieving it, compared with CIR of 44% and LFS of 0% in the remaining (both p<.001). Of the 60 patients analyzed, 28 patients had positive MRD, occurring at a median of 110 days (30-540 days) after transplant. The predictive value of sequential monitoring could be demonstrated in 8 patients culminating in clinical relapse (representing 73% of all relapsing patients). The median time from MRD positivity in BM to morphological relapse was 95 days (range, 33-140 days). Conclusions A > 3 log reduction at the first 3 months after HSCT in RUNX1/RUNX1T1 transcripts from diagnosis is highly prognostic. We conclude that MRD monitoring by RQ-PCR at regular early time points post HSCT in t (8; 21) AML allows further identification of patients at high risk of relapse even after allo-HSCT and could now be incorporated in clinical trials to evaluate the role of risk directed prophylactic/preemptive therapy. This work was partly supported by The Key Program of National Natural Science Foundation of China £¨Grant No. 81230013£©, Beijing Municipal Science & Technology Commission (No.Z121107002812033) and Bejing Municipal Science & Technology Commission£¨No.Z111107067311070). Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1391-1391
Author(s):  
Taehyung Kim ◽  
Joon Ho Moon ◽  
Jae-Sook Ahn ◽  
Marc S. Tyndel ◽  
Seung-Shin Lee ◽  
...  

Abstract Introduction NGS-based detection of minimal residual disease (MRD) has been successfully demonstrated for its correlation with relapse risk in AML. However, in a subtype of AML, CBF-AML, its clinical relevance of residual allelic burden at complete remission (CR) has not been fully explored. The standard MRD detection method in CBF-AML is quantitative PCR (qPCR). In this study, we aimed to explore applicability and feasibility of NGS-based MRD detection in CBF-AML taking various approaches, rather than simply using residual allele burden at CR. Patients and Methods Fifty-three patients (pts) diagnosed with CBF-AML were enrolled in this study (31 pts with RUNX1-RUNX1T1 and 22 pts with CBFB-MYH11). All 53 patients achieved complete remission (CR). We performed targeted deep sequencing on 84 genes in 106 samples collected at diagnosis and at CR as well as T-cell (n = 53, CD3+) fraction as a control using Illumina Hiseq 2500. Mean on-target coverage for 159 sequenced was 1,572x. The level of RUNX1-RUNX1T1 was measured at diagnosis and at CR for 29 patients using qPCR. Results At diagnosis, 99 mutations from 49 pts (n = 49/53, 92%) were detected, where median number of mutations for 49 pts was 2 (range 1-6). Consistent with previous studies, KIT (36%), NRAS (32%), KRAS (17%), ASXL2 (15%) were commonly mutated. Among mutations detected at diagnosis, cKIT-D816 mutation and mutations in genes in DNA methylation pathway (DNMT3A and TET2) were associated with higher risk of relapse (5.29, [1.89 - 14.87], p = 0.002 and 3.15 [1.07 - 9.26], p= 0.037, respectively). In CR samples, 46 mutations from 32 pts were still detectable (46/99, 46%, mean VAF: 0.60%, range 0.04%-6.28%, Fig A). Only 4 mutations from 2 pts were over 2.5% (2 in TET2, 1 in ASXL1, and 1 in U2AF1). When tracing back at diagnosis, allelic burden of 46 mutations detected at remission were higher than 53 cleared mutations (p < 0.002), indicating clonal mutations are more likely to be detected at CR (Fig A). They were mostly in genes associated with activated signaling (32/46, 70%). When considering complete clearance rate, mutations in genes associated with activated signaling, DNA methylation, and spliceosome tended to be persistent at CR (32/64, 4/5, and 1/1), whereas mutations in cohesin complex and chromatin modifiers were mostly completely cleared (0/8 and 4/13). We then assessed clinical relevance of mutation clearance from various perspectives. We did not find association of mutation clearance at 0.3% (MC03) with OS (p = 0.43) or with relapse risk (p = 0.8, Fig B). Complete mutation clearance also did not show significant association with OS and relapse risk. Among 29 pts with RUNX1-RUNX1T1 with available qPCR data, 20 pts were MRD-positive by qPCR at CR. Nine pts who achieved MRD-negative also achieved MC03 as well. When considering only MRD-positive pts, achievement of MC03 did not affect OS (p = 0.69) and relapse incidence (p = 0.86, Fig C). Lastly, we assessed whether persistence of high risk mutations at CR (cKIT-D816, DNMT3A, and TET2) is associated with higher risk of relapse, but complete clearance of KIT-D816 mutation also did not affect OS and relapse incidence (p = 0.94 and p = 0.40, respectively, Fig D). We were not able to analyze DNMT3A and TET2 mutations as only 1/5 mutation was cleared. Conclusion Current study demonstrates that low residual allelic burden measured by NGS at CR does not provide additional clinically relevant information in addition to baseline mutation profile nor qPCR-based MRD in CBF-AML Figure. Figure. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1738-1738
Author(s):  
Mary Alikian ◽  
Rebecca Ellwood ◽  
Olga Tatarinova ◽  
Graham David Rose ◽  
George Nteliopoulos ◽  
...  

Abstract Background We have previously shown that DNA based digital PCR (dPCR) is a more sensitive approach for monitoring patients with CML in deep molecular remission compared to the 3 alternatives: qPCR, RT-qPCR and RT-dPCR. In this study we compared dPCR and RT-qPCR for their ability to predict molecular relapse in the context of national study of de-escalation of TKI dose followed by cessation of therapy (DESTINY). Methods The DESTINY study recruited 174 patients in major (or deeper) molecular remission, stratified according to whether their molecular remission status at study entry was MR3 or ≥MR4. The standard dose of TKI (imatinib, dasatinib and nilotinib) was halved at study entry and stopped 12 months later provided the patient remained in ≥ MR3. Material from diagnosis, required to clone the patient specific breakpoint for dPCR, was available only in a limited subset of patients. We cloned the breakpoints in 30 patients who reflected the range of outcomes of the study: 4 relapsed within the first 12 months of de-escalation, 12 relapsed 12-24 months from study entry (0-12 months from stopping TKI) and one relapsed 26 months from study entry (14 months from stopping therapy). All 30 patients were in ≥MR4 at study entry, of whom 7, 13 and 10 patients were in MR4, MR4.5 and MR5, respectively. Relapse was defined as loss of MR3. Fusion specific assays were designed and validated for all patients and dPCR was performed using the RainDrop platform (BioRad®). Multiple negative samples were used to establish the true positive quantification threshold per assay, including a pooled DNA samples from healthy controls and from other CML patients. Approximately 80,000 cells were included per reaction. RT-qPCR was performed according to standard protocols and all results expressed on the international scale (IS). Samples were collected at a minimum of 3 specified intervals, study entry, time of stopping TKI (month 12), month 24 and/or at the time of relapse. The log rank test was used to predict the risk of relapse and the Spearman correlation coefficient to compare the sensitivity of the two methods. Results At trial entry, patients were in MR4, MR4.5 or MR5 as defined by RT-qPCR: the depth of remission was not associated with the risk of relapse (log rank test p = 0.18 and 0.07, 0.19, 0.59 for MR4 vs MR4.5, MR4 vs MR5 and MR4.5 vs MR5, respectively). However, dPCR was positive in only 5 of the 30 patients of whom 3/5 (60%) relapsed compared to 14/25 (56%) who were dPCR negative (p=0.03). At 12 months neither the depth of remission by RT-qPCR nor the detection of malignant cells by dPCR, predicted subsequent relapse. In order to account for the fact that both methods were detecting different types of target molecules, the results were treated as a binary outcome (target detected/target not detected). 54 samples (49%) were positive by both methods, 6 (5%) were negative by both, 47 (42%) were positive only by RT-qPCR and 4 (4%) were positive only by dPCR. There was no difference in the predictive value of relapse when both methods were positive or negative. The majority of target molecules were not detected by dPCR when RT-qPCR showed transcript numbers ≤ 7 suggesting the possibility of false positivity and that dPCR is more specific in detecting true positive residual disease. Target molecules were detected by dPCR in all samples defined as > MR3 by RT-qPCR: absolute numbers of target molecules were understandably lower by dPCR (detecting cells) than by RT-qPCR (detecting transcripts). Conclusion Both methods can detect truly negative samples, however, dPCR is more specific in excluding false positivity. Neither dPCR nor RT-qPCR was capable of predicting relapse at the time of study entry. The ability of dPCR to predict loss of MR3 while off-therapy however requires the monitoring of serial time points leading to relapse before a final conclusion could be made. The question that remains posed here is the clinical validity of early residual disease detection given that, in the case of relapse (defined by ≥MR3 by RT-qPCR), all patients regain molecular remission after the TKI is reintroduced as it has been shown by all the STOP clinical trials. Disclosures Milojkovic: Incyte: Honoraria, Speakers Bureau; Novartis: Honoraria, Speakers Bureau; Pfizer: Honoraria, Speakers Bureau; BMS: Honoraria, Speakers Bureau. Clark:Pfizer: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Bristol Myers Squibb: Consultancy, Research Funding; Ariad/Incyte: Consultancy. Apperley:Novartis: Honoraria, Research Funding, Speakers Bureau; BMS: Honoraria, Speakers Bureau; Incyte: Honoraria, Speakers Bureau; Pfizer: Honoraria, Speakers Bureau.


2020 ◽  
Vol 9 (12) ◽  
pp. 3818
Author(s):  
Paula Aguirre-Ruiz ◽  
Beñat Ariceta ◽  
María Cruz Viguria ◽  
María Teresa Zudaire ◽  
Zuriñe Blasco-Iturri ◽  
...  

Patients with myeloid neoplasms who relapsed after allogenic hematopoietic stem cell transplant (HSCT) have poor prognosis. Monitoring of chimerism and specific molecular markers as a surrogate measure of relapse is not always helpful; therefore, improved systems to detect early relapse are needed. We hypothesized that the use of next generation sequencing (NGS) could be a suitable approach for personalized follow-up post-HSCT. To validate our hypothesis, we analyzed by NGS, a retrospective set of peripheral blood (PB) DNA samples previously evaluated by high-sensitive quantitative PCR analysis using insertion/deletion polymorphisms (indel-qPCR) chimerism engraftment. Post-HCST allelic burdens assessed by NGS and chimerism status showed a similar time-course pattern. At time of clinical relapse in 8/12 patients, we detected positive NGS-based minimal residual disease (NGS-MRD). Importantly, in 6/8 patients, we were able to detect NGS-MRD at time points collected prior to clinical relapse. We also confirmed the disappearance of post-HCST allelic burden in non-relapsed patients, indicating true clinical specificity. This study highlights the clinical utility of NGS-based post-HCST monitoring in myeloid neoplasia as a complementary specific analysis to high-sensitive engraftment testing. Overall, NGS-MRD testing in PB is widely applicable for the evaluation of patients following HSCT and highly valuable to personalized early treatment intervention when mixed chimerism is detected.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1384-1384
Author(s):  
Karthik B.K Bommannan ◽  
Man Updesh Singh Sachdeva ◽  
Parveen Bose ◽  
Deepak Bansal ◽  
Ram Kumar Marwaha ◽  
...  

Abstract Introduction Minimal residual disease (MRD) has emerged as an independent prognostic factor for patients of acute lymphoblastic leukemia (ALL). There is a strong correlation between MRD levels in bone marrow and the risk of relapse in childhood & adult leukemias 1, 2. Bone marrow MRD (BM-MRD) level of ≥ 0.01% is considered as positive and a mid-induction MRD of ≥ 1% is associated with high risk of relapse 3. Recently, the concept of peripheral blood MRD (PB-MRD), as a replacement for BM-MRD, has hit the lime light. In pediatric B-ALL, presence of PB-MRD is associated with a high relapse rate in comparison to cases which are PB-MRD negative 4, 5. This study was aimed to compare the levels of mid-induction (day 15) MRD levels in bone marrow and peripheral blood of pediatric B-ALL patients with a hypothesis that PB-MRD levels correlate with BM-MRD levels, and thus can predict BM-MRD levels for further management of the patient. Methods Forty newly diagnosed CD19+CD10+CD34+/- pediatric B-ALL patients under Vincristine, L-Asparaginase and Dexamethasone, were assessed for MRD levels on their paired day 15 PB & BM samples using six colour flow cytometry. With informed consent, both the samples were collected in EDTA vacutainers and lyse-stain-wash technique was used to prepare a single six colour tube comprising of SYTO 13/ CD34PE/ CD20PerCP/ CD19 PECy7/ CD10APC/ CD45APCH7 for each sample. The processed samples were run on BD FACS Canto II with acquisition of 1 million events or till the tubes were empty. Analysis was done using BD FACS Diva software and MRD of ≥ 0.01% was considered positive. Results Among 40 pairs of day 15 PB and BM samples, 25 (62.5%) were BM-MRD positive. Sixteen pairs (40%) had PB-MRD and BM-MRD co-positivity, 9 pairs (22.5%) had isolated BM-MRD positivity and 15 pairs (37.5%) were MRD negative in both PB and BM samples. In other words, among the 25 BM-MRD positive cases, simultaneous PB-MRD was positive in 16 patients (64%) and none of the samples had isolated PB-MRD positivity. Overall analysis of MRD positive cases showed a direct correlation between PB-MRD and BM-MRD (ρ = +0.684, p < 0.000) and BM-MRD levels were 7 times higher than the PB-MRD. In addition, ROC analysis with PB-MRD of ≥ 0.01% as a cut-off, revealed that, the most likelihood of PB-MRD being positive was when BM-MRD was ≥ 0.31%. Conclusions In contrast to the sparsely available literature, our study shows a significant correlation between PB & BM-MRD levels in day 15 paired samples of B-ALL cases. The MRD levels were 7 times higher in BM as compared to PB and PB-MRD was mostly positive with BM-MRD of ≥0.31%. In other words, day 15 PB-MRD positivity indirectly indicates that there is a minimum BM-MRD of 0.31%. Since literature reports prognostic significance of mid-induction BM-MRD at levels ≥1%, on day 15, an assessment of peripheral blood MRD alone, might yield clinically relevant prognostic information. A paired analysis at different time points might also establish a similar correlation as seen in the present study, eliminating the need of BM-MRD during further follow ups of the patient. This will help in avoiding an invasive procedure and improve patient compliance. References 1. Irving J, Jesson J, Virgo P, Case M, Minto L, Eyre L, et al. Establishment and validation of a standard protocol for the detection of minimal residual disease in B lineage childhood acute lymphoblastic leukemia by flow cytometry in a multi-center setting. haematologica. 2009;94(6):870-4. 2. Coustan-Smith E, Sancho J, Behm FG, Hancock ML, Razzouk BI, Ribeiro RC, et al. Prognostic importance of measuring early clearance of leukemic cells by flow cytometry in childhood acute lymphoblastic leukemia. Blood. 2002;100(1):52-8. 3. Basso G, Veltroni M, Valsecchi MG, Dworzak MN, Ratei R, Silvestri D, et al. Risk of relapse of childhood acute lymphoblastic leukemia is predicted by flow cytometric measurement of residual disease on day 15 bone marrow. Journal of Clinical Oncology. 2009;27(31):5168-74. 4. Elain CS, Sancho J, Michael LH, Bassem. Use of peripheral blood instead of bone marrow to monitor residual disease in children with acute lymphoblastic leukemia. Blood. 2002;100 (7):2399-402. 5. Brisco MJ, Sykes PJ, Hughes E, Dolman G, Neoh SH, Peng LM, et al. Monitoring minimal residual disease in peripheral blood in B lineage acute lymphoblastic leukaemia. British journal of haematology. 1997;99(2):314-9. Disclosures: No relevant conflicts of interest to declare.


Hemato ◽  
2022 ◽  
Vol 3 (1) ◽  
pp. 63-81
Author(s):  
Emilie Arnault Carneiro ◽  
Filipa Barahona ◽  
Carolina Pestana ◽  
Cristina João

Multiple myeloma (MM) is the second deadliest hematological cancer. Despite the enormous innovation on MM treatment in the last decades, still 48% of patients die within 5 years after diagnosis. MM diagnosis and therapeutic strategy mainly rely on direct bone marrow (BM) assessment. Given the MM heterogeneity, BM biopsies do not accurately reflect the whole disease status, hampering accurate disease prognosis. Moreover, biopsies are painful and invasive procedures, highlighting the need for non-invasive and more accurate source of biomarkers. Liquid biopsies are promising sources of biomarkers that may overcome these limitations. Peripheral blood carries circulating myeloma components that are being extensively explored since the last few years as an alternative to BM aspirates. These include circulating tumor cells (CTC), cell-free DNA (cfDNA), and extracellular vesicles containing miRNA and proteins. The current review summarizes scientific evidence establishing BM as a gold standard for the diagnosis, prognosis, and evaluation of minimal residual disease. We discuss the last advances regarding cfDNA and CTC biomarkers from peripheral blood in patients with MM as well as the statistical validations. This paper addresses the technological hurdles associated with liquid biopsies and examines the missing steps for their inclusion into the clinical practice.


Sign in / Sign up

Export Citation Format

Share Document