scholarly journals Rearranged Ig Heavy Chain DNA Is Detectable in Cell-Free Blood Samples of Patients With B-Cell Neoplasia

Blood ◽  
1997 ◽  
Vol 90 (12) ◽  
pp. 4953-4960 ◽  
Author(s):  
N. Frickhofen ◽  
E. Müller ◽  
M. Sandherr ◽  
T. Binder ◽  
M. Bangerter ◽  
...  

Tumor-derived DNA has been shown in various cell-free body fluids. In this study, soluble tumor-derived DNA was analyzed in serum and plasma samples of patients with B-cell malignancies. DNA was extracted from tumor cell specimens as well as serum and plasma samples collected from 110 patients with non-Hodgkin's lymphoma and acute B-precursor lymphoblastic leukemia and was subjected to polymerase chain reaction (PCR) analysis for rearranged immunoglobulin heavy chain DNA. In 54% of serum or plasma samples analyzed at different times before and during treatment, clonal DNA from a rearranged immunoglobulin heavy chain locus was detectable. When examined at diagnosis and before any treatment, clonotypic DNA was found in serum or plasma of 86% of the patients. Serum or plasma from patients with systemic or bulky disease was uniformly PCR positive, whereas clonotypic DNA was also recovered from the serum or plasma from the majority of patients with limited disease stages. Degradation of clonal DNA by nucleases in vitro was shown to be one cause of false-negative PCR results. This technical drawback can be relieved by adding a nuclease inhibitor like EDTA, ie, by using plasma instead of serum for PCR analysis. Treatment of patients with cytotoxic drugs was followed by rapid clearance of DNA from the peripheral blood, suggesting that soluble tumor-derived DNA might be associated with viable and proliferating tumor cells. Follow-up studies showed a close correlation of persisting soluble tumor-derived DNA with resistant disease or early relapse. In summary, these data suggest that tumor-derived DNA can be detected in serum or plasma of the majority of patients with B-cell malignancies and that testing of serum or plasma for tumor-associated DNA may be a novel parameter for monitoring response to treatment.

Blood ◽  
1997 ◽  
Vol 90 (12) ◽  
pp. 4953-4960 ◽  
Author(s):  
N. Frickhofen ◽  
E. Müller ◽  
M. Sandherr ◽  
T. Binder ◽  
M. Bangerter ◽  
...  

Abstract Tumor-derived DNA has been shown in various cell-free body fluids. In this study, soluble tumor-derived DNA was analyzed in serum and plasma samples of patients with B-cell malignancies. DNA was extracted from tumor cell specimens as well as serum and plasma samples collected from 110 patients with non-Hodgkin's lymphoma and acute B-precursor lymphoblastic leukemia and was subjected to polymerase chain reaction (PCR) analysis for rearranged immunoglobulin heavy chain DNA. In 54% of serum or plasma samples analyzed at different times before and during treatment, clonal DNA from a rearranged immunoglobulin heavy chain locus was detectable. When examined at diagnosis and before any treatment, clonotypic DNA was found in serum or plasma of 86% of the patients. Serum or plasma from patients with systemic or bulky disease was uniformly PCR positive, whereas clonotypic DNA was also recovered from the serum or plasma from the majority of patients with limited disease stages. Degradation of clonal DNA by nucleases in vitro was shown to be one cause of false-negative PCR results. This technical drawback can be relieved by adding a nuclease inhibitor like EDTA, ie, by using plasma instead of serum for PCR analysis. Treatment of patients with cytotoxic drugs was followed by rapid clearance of DNA from the peripheral blood, suggesting that soluble tumor-derived DNA might be associated with viable and proliferating tumor cells. Follow-up studies showed a close correlation of persisting soluble tumor-derived DNA with resistant disease or early relapse. In summary, these data suggest that tumor-derived DNA can be detected in serum or plasma of the majority of patients with B-cell malignancies and that testing of serum or plasma for tumor-associated DNA may be a novel parameter for monitoring response to treatment.


Blood ◽  
2000 ◽  
Vol 95 (8) ◽  
pp. 2651-2658 ◽  
Author(s):  
John W. Donovan ◽  
Marco Ladetto ◽  
Guangyong Zou ◽  
Donna Neuberg ◽  
Christina Poor ◽  
...  

Tumor-related immunoglobulin heavy-chain (IgH) rearrangements are markers for polymerase chain reaction (PCR) detection of minimal residual disease (MRD) in B-cell malignancies. Nested PCR with patient IgH allele-specific oligonucleotide primers can detect 1 tumor cell in 104 to 106 normal cells. In childhood acute lymphoblastic leukemia (ALL), persistence of PCR-detectable disease is associated with increased risk of relapse. The clinical significance of qualitative PCR data can be limited, however, because patients can harbor detectable MRD for prolonged periods without relapse. Recent studies indicate that a quantitative rise in tumor burden identifies patients who are at high risk for relapse. Therefore, an efficient and reliable PCR method for MRD quantification is needed for ALL patients. We have developed a real-time PCR method to quantify MRD with IgH VH gene family consensus fluorogenically labeled probes. With this method, a small number of probes can be used to quantify MRD in a large number of different patients. The assay was found to be both accurate and reproducible over a wide range and capable of detecting approximately 1 tumor cell in 5 × 104 normal cells. We demonstrate that this methodology can discriminate between patients with persistence of MRD who relapse and those who do not. This technique is generally applicable to B-cell malignancies and is currently being used to quantify MRD in a number of prospective clinical studies at our institution.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1555-1555
Author(s):  
Michaela Kotrova ◽  
Monika Szczepanowski ◽  
Henrik Knecht ◽  
Christoph Faul ◽  
Mustafa Kondakci ◽  
...  

Abstract Background: High throughput sequencing of immunoglobuline heavy chain gene rearrangements (IGH) recently demonstrated that B-cell precursor ALL (BCP-ALL) is a highly oligoclonal disease in children (Theunissen, Hematologica, 2017), however, little is known about the degree of oligoclonality in adults with BCP-ALL. Furthermore, no data exist on potential changes of the subclonal composition during therapy. Therefore, we aimed to monitor the clonal architecture of the leukemic blasts before and after Rituximab containing induction treatment in the context of the current German Multicentric Acute Lymphoblastic Leukemia (GMALL) 08/2013 trial. Materials & Methods: We identified and studied complete and incomplete immunoglobulin (IG) heavy chain rearrangements in 100ng of bone marrow (BM) DNA at diagnosis (dx) and 500ng BM DNA after Rituximab-containing induction I in 19 patients with BCP-ALL. We employed IGH-VJ FR1 and IGH-DJ NGS assays developed within the EuroClonality-NGS Consortium (www.euroclonalityngs.org), performing next generation sequencing on Illumina MiSeq. We analysed data with the ARResT/Interrogate bioinformatics platform (Bystry, Bioinformatics, 2017), which is also able to isolate and use the DN-J stem of V/DJ junctions to link clonally related rearrangements. Clones with abundance ≥5 %, and all subclones carrying the same DN-J stem as the dominant clones were considered as leukemia-associated and studied further. Employing the sequence of the DN-J stem, we could also link related complete and incomplete rearrangements, even though those are amplified in two separate PCRs. Results: At dx, 18/19 patients carried at least 1 IGH rearrangement with abundance ≥ 5%, 10 patients carried 2 or more. In all 18 patients with dominant markers detected at dx, subclones with abundance <5% carrying the same DN-J stem as the dominant clone were present, indicating oligoclonality and clonal evolution. On average, 53.1 (range 0-295) (sub)clones per DN-J stem were detected at dx, and 32.7 (range 0-238) after treatment. Next, we compared the kinetics of all (sub)clones with abundance ≥1% in at least one of the time-points. Of the 18 patients, 6 only had subclones with abundance <1%, and we did not investigate the kinetics of such subclones. In another 11 patients (Fig. 1, Pt. 1-11), all sub(clones) had the same kinetics, with no clone gaining predominance over time. In 1 patient (Fig. 1, Pt. 12), 3 (sub)clones which were present at dx disappeared and a new subclone appeared after treatment. This patient had a pro-B immunophenotype, where oligoclonality and clonal instability are well known phenomena (Szczepanski, Leukemia, 2001). Conclusions: It has recently been shown that ALL is a highly oligoclonal disease in children (Theunissen, Hematologica, 2017), and our study extends this finding to adults with BCP-ALL. We furthermore demonstrate that the subclonal composition remains stable in the majority of patient during induction chemoimmunotherapy. The fact that the response to treatment is generally consistent among different (sub)clones has important implications for MRD quantification as it reassures the usage of the dominant clonal IG gene rearrangement for MRD monitoring in ALL. However, also significant changes of the clonal composition may occur in BCP-ALL as exemplified in one patient of our cohort. Further investigations are necessary to elucidate factors that influence subclonal heterogeneity in response to treatment. Disclosures Viardot: Pfizer: Consultancy, Honoraria; Gilead Kite: Consultancy, Honoraria; Roche: Consultancy, Honoraria; Amgen: Consultancy; BMS: Consultancy, Honoraria. Kneba:AbbVie: Consultancy, Honoraria; Roche: Consultancy, Honoraria. Goekbuget:Pfizer: Consultancy, Other: Travel support, Research Funding; Novartis: Consultancy, Other: Travel support, Research Funding; Kite / Gilead: Consultancy; Amgen: Consultancy, Other: Travel support, Research Funding; Celgene: Consultancy. Brüggemann:Incyte: Consultancy; PRMA: Consultancy; Regeneron: Research Funding; Affimed: Research Funding; Pfizer: Speakers Bureau; Amgen: Consultancy, Research Funding, Speakers Bureau; Roche: Speakers Bureau.


Blood ◽  
2012 ◽  
Vol 120 (22) ◽  
pp. 4407-4417 ◽  
Author(s):  
Charles Gawad ◽  
Francois Pepin ◽  
Victoria E. H. Carlton ◽  
Mark Klinger ◽  
Aaron C. Logan ◽  
...  

Abstract The ability to distinguish clonal B-cell populations based on the sequence of their rearranged immunoglobulin heavy chain (IgH) locus is an important tool for diagnosing B-cell neoplasms and monitoring treatment response. Leukemic precursor B cells may continue to undergo recombination of the IgH gene after malignant transformation; however, the magnitude of evolution at the IgH locus is currently unknown. We used next-generation sequencing to characterize the repertoire of IgH sequences in diagnostic samples of 51 children with B precursor acute lymphoblastic leukemia (B-ALL). We identified clonal IgH rearrangements in 43 of 51 (84%) cases and found that the number of evolved IgH sequences per patient ranged dramatically from 0 to 4024. We demonstrate that the evolved IgH sequences are not the result of amplification artifacts and are unique to leukemic precursor B cells. In addition, the evolution often follows an allelic exclusion pattern, where only 1 of 2 rearranged IgH loci exhibit ongoing recombination. Thus, precursor B-cell leukemias maintain evolution at the IgH locus at levels that were previously underappreciated. This finding sheds light on the mechanisms associated with leukemic clonal evolution and may fundamentally change approaches for monitoring minimal residual disease burden.


Sign in / Sign up

Export Citation Format

Share Document