scholarly journals RAS/RAF mutations in tumor samples and cell-free DNA from plasma and bone marrow aspirates in multiple myeloma patients

2020 ◽  
Vol 11 (12) ◽  
pp. 3543-3550
Author(s):  
Qian Li ◽  
Helen J Huang ◽  
Jing Ma ◽  
Yafei Wang ◽  
Zeng Cao ◽  
...  
Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5603-5603
Author(s):  
Juan Du ◽  
Jing Lu ◽  
Wanting Qiang ◽  
Lu Li ◽  
Jin Liu ◽  
...  

Abstract Background: Multiple myeloma (MM) is a plasma cell malignancy characterized by complex cytogenetic and molecular abnormalities including translocations involving the immunoglobin heavy chain locus and mutations involving numerous oncogenic signaling pathways. Fluorescence in situ hybridization (FISH) has emerged as the most useful current cytogenetic assessment and provide a new level of insight into the correlation of myeloma prognosis risk model. However, the identification or sorting of malignant cells is required before FISH probes and only involved expansion of the types of probe and number of detectable targets is reached. Cell-free DNA (cfDNA) offers the potential for minimally invasive genome-wide profiling of tumor alterations without tumor biopsy and may be associated with cancer precision medicine and patient prognosis. Methods: In this retrospective cohort study, we identified 37 patients from 9 relapsed/refractory (RR) and 33 newly diagnosed (ND) patients were analyzed for chromosomal copy number imbalance using the ultrasensitive chromosomal aneuploidy detector (UCAD) platform. Results: Chromosome copy number aberration (CNA) were frequently (82.6%, N=46) detected in MM plasma cell free DNA. Applying UCAD to cfDNA, FISH in CD138 purified bone marrow aspirates, and some matched bone marrow biopsies, we find concordance in copy number alterations (~81%) between liquid and tumor biopsies. Significant copy number changes, including 1q gains, 13q deletion and 17p deletion could be found in 57.89%, 54.05%, and 16.67% in plasma of MM, which is higher percentage than FISH assay (46.81%, 28.26%, and 8.89%), respectively. Besides, chromosome 6p and 6q were determined the higher frequency aberration from UCAD. Moreover, a higher frequency of copy number aberrations and variations was detected in RR patients than ND (100% vs 78.4%, respectively), obviously CNAs heterogeneity displayed in advanced disease. In the inconsistent some samples, UCAD from the plasma and bone marrow showed the similar results, which indicated the FISH is underdetermined and insensitivity in some patients' routine inspection. Conclusion: We conclude that cfDNA analysis as an adjunct to BM biopsy represents a noninvasive and broaden the applicability strategy for comprehensive genomic profiling and therapeutic monitoring of MM. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Vol 9 (1) ◽  
Author(s):  
S. Manier ◽  
J. Park ◽  
M. Capelletti ◽  
M. Bustoros ◽  
S. S. Freeman ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2592-2592
Author(s):  
Nicholas J. Short ◽  
Keyur Patel ◽  
Maher Albitar ◽  
Miguel Franquiz ◽  
Rashmi Kanagal-Shamanna ◽  
...  

Background: Circulating cell-free DNA (ccfDNA) is highly fragmented DNA in plasma that is released by normal or tumor cells when they undergo apoptosis or necrosis. ccfDNA allows for non-invasive sampling of somatic genomic alterations and is informative in various solid tumors, including as a marker of measurable residual disease (MRD). We sought to assess the utility of baseline assessment and tracking of leukemia-associated mutations through peripheral blood sampling of ccfDNA in patients (pts) with acute leukemias. Methods: Plasma ccfDNA was isolated and analyzed using a next-generation sequencing (NGS) assay of 275 genes. This NGS analysis is based on Single Primer Extension library preparation with unique molecular identifier (Qiagen, Germantown, MD); a sequence coverage ≥ 100X (after removing duplicates) was required. Amplicon-based NGS was also performed on DNA extracted from the bone marrow (BM) in a CLIA-certified molecular diagnostics laboratory. This BM panel detects mutations in the coding sequence of 28 leukemia-associated genes, with an analytic sensitivity of 5-10%. The ccfDNA panel included all 28 genes evaluated on the BM NGS panel (ABL1, ASXL1, BRAF, DNMT3A, EGFR, EZH2, FLT3, GATA1, GATA2, HRAS, IDH1, IDH2, IKZF1, JAK2, KIT, KRAS, MDM2, MLL, MPL, MYD88, NOTCH1, NPM1, NRAS, PTPN11, RUNX1, TET2, TP53, WT1). Established bioinformatics pipelines were used to identify somatic variants. Results: Twenty-four pts (AML, n=22; ALL, n=2) underwent paired ccfDNA and BM sequencing at diagnosis prior to receiving frontline intensive chemotherapy. For baseline samples, ccfDNA was collected a median of 6 days after BM collection (range, 0-27 days) and a median of 0.5 days after start of induction chemotherapy (range, -7 to 7 days). Eleven pts (46%) also had ccfDNA collected at ≥1 time point during remission. Among the 28 genes of interest, the median number of mutations per pt detected in BM and in ccfDNA was 1 (range, 0-4) for both assays (P=0.39). A total of 40 mutations were detected: 18 mutations (45%) were detected by both methods, 7 (18%) were detected only in ccfDNA, and 15 (38%) were detected only in BM. Time from start of chemotherapy until ccfDNA collection did not appear to impact the concordance of ccfDNA and BM mutation analysis (P=0.87). Among mutations detected by ccfDNA in baseline samples, the median variant allelic frequency (VAF) was 33.7% (range, 2.7-90.8%). Among the 18 overlapping mutations, the concordance of VAF assessment by both methods was high (R2 = 0.849). Mutations detected by only one of the two methods were generally of lower VAF than those detected by both methods, suggesting that either method may miss small subclonal populations. The median VAF of mutations (as measured in ccfDNA) that were detected by both methods was higher than those detected only in ccfDNA (39.8% vs 25.2%, respectively; P=0.04); similarly, the median VAF of mutations (as measured in BM) that were detected by both methods was higher than those detected only in BM (40.2% vs 6.6%; P=0.001). Among the 7 mutations detected only by ccfDNA, ASXL1 was detected in 2 pts, WT1 in 1 pt, IDH1 in 1 pt, and BRAF and two EGFR mutations in 1 pt. Among the 5 pts in whom mutations were detected in ccfDNA but not BM, 2 eventually relapsed. In both pts, the discordant mutation (IDH1 and ASXL1) was detected in the relapse BM, suggesting that these were true mutations that were missed by NGS of the baseline BM. ccfDNA detected leukemia-associated mutations during remission that appeared to herald overt relapse (Figure 1). Two pts with t(8;21) AML developed new RUNX1 mutations detected by ccfDNA while in remission and subsequently relapsed 3 months and 14 months later. In both of these pts, the new RUNX1 mutation was confirmed in the BM at the time of morphological relapse. Another pt with AML had persistent TP53 and TET2 mutations detected by ccfDNA 1 month after allogeneic stem cell transplant and subsequently relapsed 1 month later. Conclusions: This study demonstrates that sequencing of ccfDNA can identify prognostic or targetable mutations not detected by BM NGS. However, true mutations were missed by both ccfDNA and BM analysis, suggesting that these methodologies may be complementary in the assessment and monitoring of pts with leukemia. The use of ccfDNA as a non-invasive method to detect mutations and track MRD in AML and other leukemias should be evaluated in larger, prospective cohorts. Disclosures Short: Takeda Oncology: Consultancy, Research Funding; AstraZeneca: Consultancy; Amgen: Honoraria. Jabbour:Amgen: Consultancy, Research Funding; BMS: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Cyclacel LTD: Research Funding; AbbVie: Consultancy, Research Funding; Adaptive: Consultancy, Research Funding; Takeda: Consultancy, Research Funding. Garcia-Manero:Amphivena: Consultancy, Research Funding; Helsinn: Research Funding; Novartis: Research Funding; AbbVie: Research Funding; Celgene: Consultancy, Research Funding; Astex: Consultancy, Research Funding; Onconova: Research Funding; H3 Biomedicine: Research Funding; Merck: Research Funding. Kantarjian:BMS: Research Funding; Amgen: Honoraria, Research Funding; Agios: Honoraria, Research Funding; Immunogen: Research Funding; Takeda: Honoraria; Novartis: Research Funding; Ariad: Research Funding; Astex: Research Funding; Pfizer: Honoraria, Research Funding; Actinium: Honoraria, Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Research Funding; Jazz Pharma: Research Funding; Cyclacel: Research Funding; AbbVie: Honoraria, Research Funding. Ravandi:Macrogenix: Consultancy, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Xencor: Consultancy, Research Funding; Menarini Ricerche: Research Funding; Cyclacel LTD: Research Funding; Selvita: Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3423-3423 ◽  
Author(s):  
Lenka Kubiczkova-Besse ◽  
Daniela Drandi ◽  
Lenka Sedlarikova ◽  
Stefania Oliva ◽  
Manuela Gambella ◽  
...  

Abstract Background Circulating nucleic acids, such as cell-free DNA (cf-DNA), are becoming a promising minimally-invasive diagnostic tool for cancer detection. Recent studies demonstrated that tumor-derived cf-DNA can be used to monitor tumor burden and response to treatment in patients (pts) with solid tumors as well as hematological malignancies (Dawson et al, 2013, Armand et al, 2013). In this study we investigated the clinical utility of cf-DNA in the monitoring of minimal residual disease (MRD) of pts with multiple myeloma (MM) carrying the tumor specific immunoglobulin (IGH) rearrangement. Methods Cf-DNA was extracted from 1 ml of serum sample from 13 MM patients enrolled in Italian CRD/MEL-200 and EMN-02 protocols. The total amount of cf-DNA was estimated by fluorometric measurement (median 560 ng, range 15-5158 ng) and the length of fragments was evaluated by high sensitivity dsDNA chips (Agilent). Patient specific clonal IGH rearrangement was identified at the time of diagnosis from bone marrow (BM) genomic DNA (gDNA) as previously reported (Ladetto et al, 2000). For each patient, MRD in BM and peripheral blood (PB) was estimated by real time quantitative PCR (qPCR) using ASO-specific primers and the quantification was based on serial 10-fold dilution standard curves from plasmid carrying the patient specific IGH rearrangement. The amount of IGH rearrangement in cf-DNA (cf-IGH) was estimated by qPCR and droplet digital PCR (ddPCR) (Bio-Rad) on diagnostic and follow up samples and was expressed as the amount of copies per 1 µg of total cf-DNA. qPCR and ddPCR results were interpreted according to the Euro-MRD guidelines (van der Velden et al, 2003). Results Overall, 54 cf-DNA samples from MM serum (13 diagnostic, 41 follow-up samples) were analyzed for the presence of patient specific IGH rearrangement. The most abundant fraction of cf-DNA was 180-220bp, than 350-400bp and 700-10000bp (in 100%, 85% and 68% of samples respectively), whereas longer fragments more often appeared in follow-up samples. By qPCR, cf-IGH at diagnosis were observed in 11/13 diagnostic samples. Only 3/13 pts were quantifiable (116, 85, 187 copies/1 µg of cfDNA) and 8/13 pts were positive but not quantifiable (PNQ) cf-IGH. By ddPCR, levels of cf-IGH at diagnosis were observed in 9/13 pts. 6/13 pts were quantifiable (246, 195, 96, 88, 184, 25 copies/1µg of cfDNA), and only 3/13 pts were PNQ. In follow-up samples, levels of cf-IGH were undetectable by qRT-PCR; however in 5 samples they were PNQ by ddPCR. Interestingly, in one available relapse sample, cf-IGH reappeared again to quantifiable level (61 copies by qRT-PCR and 190 copies by ddPCR). The levels of cf-IGH are quantifiable in samples with higher amount of tumor specific IGH rearrangements in BM or PB; however, no association was observed between cf-IGH level at diagnosis and disease burden estimated by the PCs infiltration in BM or the monoclonal immunoglobulin concentration in blood/urine. Conclusions These data show the potential utility of cf-IGH monitoring in MM pts. Although by qPCR, cf-IGH were detected in 11/13 pts, they were quantifiable only in 3/13 pts and ddPCR was more precise as it was able to quantify cf-IGH in 6/13 pts. Since cf-IGH copies were quantifiable only in diagnostic samples and in 1 available sample at the relapse, we conclude that higher amounts of serum are necessary to overcome the limitation of assay sensitivity. Potential advantages and predictive value, for monitoring tumor marker in a non-invasive manner, need to be further validated on larger cohort of samples using increased amount of cf-DNA. Work was supported by IGA grants NT12130, NT14575. This work is funded by a Black Swan Research Initiative grant by the International Myeloma Foundation "Dynamics of microRNA and cell-free DNA profiles during multiple myeloma progression“. Disclosures Boccadoro: Celgene: Honoraria; Janssen: Honoraria; Onyx: Honoraria. Palumbo:Amgen: Consultancy, Honoraria; Bristol-Myers Squibb: Consultancy, Honoraria; Array BioPharma: Honoraria; Genmab A/S: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Janssen-Cilag: Consultancy, Honoraria; Millennium Pharmaceuticals, Inc.: Consultancy, Honoraria; Onyx Pharmaceuticals: Consultancy, Honoraria; Sanofi Aventis: Honoraria.


2002 ◽  
Vol 48 (3) ◽  
pp. 421-427 ◽  
Author(s):  
Yanni YN Lui ◽  
Ki-Wai Chik ◽  
Rossa WK Chiu ◽  
Cheong-Yip Ho ◽  
Christopher WK Lam ◽  
...  

Abstract Background: Despite current interest in the biology and diagnostic applications of cell-free DNA in plasma and serum, the cellular origin of this DNA is poorly understood. We used a sex-mismatched bone marrow transplantation model to study the relative contribution of hematopoietic and nonhematopoietic cells to circulating DNA. Methods: We studied 22 sex-mismatched bone marrow transplantation patients. Paired buffy coat and plasma samples were obtained from all 22 patients. Matching serum samples were also obtained from seven of them. Plasma DNA, serum DNA, and buffy coat were quantified by real-time PCR of the SRY and β-globin gene DNA. To investigate the effects of blood drawing and other preanalytical variables on plasma DNA concentrations, blood samples were also collected from 14 individuals who had not received transplants. The effects of blood sampling by syringe and needle, centrifugation, and time delay in blood processing were studied. Results: The median percentage of Y-chromosome DNA in the plasma in female patients receiving bone marrow from male donors (59.5%) differed significantly (P <0.001) from that in the male patients receiving bone marrow from female donors (6.9%). This indicated that plasma DNA in the bone marrow transplantation recipients was predominantly of donor origin. Compared with paired plasma samples, serum samples had a median 14-fold higher DNA concentration, with the additional DNA being of donor origin. Control experiments indicated that none of the three tested preanalytical variables contributed to a significant change in cell-free DNA concentration. Conclusions: After bone marrow transplantation, the DNA in plasma and serum is predominantly hematopoietic in origin. Apart from the biological implications of this observation, this finding suggests that plasma and serum can be used as alternative materials for the study of postbone marrow transplantation chimerism.


Sign in / Sign up

Export Citation Format

Share Document