1
|
Seferna K, Svaton M, Rennerova A, Skotnicova A, Reznickova L, Valova T, Sedlacek P, Riha P, Formankova R, Keslova P, Sramkova L, Stary J, Zuna J, Kolenova A, Salek C, Trka J, Fronkova E. NGS-MRD negativity in post-HSCT ALL spares unnecessary therapeutic interventions triggered by borderline qPCR results without an increase in relapse risk. Hemasphere 2025; 9:e70124. [PMID: 40201744 PMCID: PMC11978274 DOI: 10.1002/hem3.70124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/03/2025] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
Monitoring of minimal residual disease (MRD) after hematopoietic stem cell transplantation (HSCT) in patients with acute lymphoblastic leukemia (ALL) is vital for timely therapeutic intervention planning. However, interpreting low-positive results from the current standard method, quantitative PCR (qPCR) of immunoglobulin and T-cell receptor gene rearrangements (IG/TR), poses challenges due to the risk of false positivity caused by non-specific amplification. We aimed to improve MRD detection specificity using the next-generation amplicon sequencing (NGS) of IG/TR rearrangements for better relapse prediction. In pediatric and young adult ALL patients undergoing sequential post-HSCT MRD monitoring, we prospectively re-tested positive non-quantifiable qPCR results with NGS-MRD using the EuroClonality-NGS approach. We were able to confirm 13 out of 47 (27.7%) qPCR positive results using the more specific NGS-MRD method. Out of 10 patients with at least one MRD positivity confirmed by NGS, six relapsed (60%) 1-3.7 months after testing. Among 25 patients with all NGS-MRD results negative, two relapses occurred (8%) after 5.1 and 12.1 months. One-year RFS was 40% versus 96% and 3-year OS was 33.3% versus 94.4% for the NGS-positive and NGS-negative groups, respectively. The difference was not attributable to a varying rate of therapeutic interventions. Six patients out of 14 who had immunosuppressive treatment tapered or received donor lymphocyte infusion in response to MRD positivity developed significant graft versus host disease, leading to one fatality. This underscores the importance of enhancing the post-HSCT relapse risk prediction accuracy through NGS-MRD testing to avoid unnecessary interventions.
Collapse
Affiliation(s)
- Krystof Seferna
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Michael Svaton
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Andrea Rennerova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Aneta Skotnicova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Leona Reznickova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Tatana Valova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Petr Sedlacek
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Petr Riha
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Renata Formankova
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Petra Keslova
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Lucie Sramkova
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Jan Stary
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Jan Zuna
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Alexandra Kolenova
- Department of Pediatric Hematology and OncologyNational Institute of Children's Diseases and Comenius UniversityBratislavaSlovakia
| | - Cyril Salek
- Institute of Haematology and Blood TransfusionPragueCzechia
- Institute of Clinical and Experimental HaematologyFirst Faculty of Medicine, Charles UniversityPragueCzechia
| | - Jan Trka
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Eva Fronkova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| |
Collapse
|
2
|
Jia MZ, Li WJ, Wang CJ, Zhang Q, Gao C, Huang XT, Zhu T, Zhang RD, Cui L, Li ZG. Tracing back of relapse clones by Ig/TCR gene rearrangements reveals complex patterns of recurrence in pediatric acute lymphoblastic leukemia. Int J Lab Hematol 2023; 45:717-725. [PMID: 37194559 DOI: 10.1111/ijlh.14100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/04/2023] [Indexed: 05/18/2023]
Abstract
INTRODUCTION Relapse remained the major obstacle to improving the prognosis of children with acute lymphoblastic leukemia (ALL). This study aimed to investigate the changing patterns of Ig/TCR gene rearrangements between diagnosis and relapse and the clinical relevance and to explore the mechanism of leukemic relapse. METHODS Clonal Ig/TCR gene rearrangements were screened by multiplex PCR amplification in 85 paired diagnostic and relapse bone marrow (BM) samples from children with ALL. The new rearrangements presented at relapse were quantitatively assessed by the RQ-PCR approach targeting the patient-specific junctional region sequence in 19 diagnostic samples. The relapse clones were further back-traced to diagnostic and follow-up BM samples from 12 patients. RESULTS Comparison of Ig/TCR gene rearrangements between diagnosis and relapse showed that 40 (57.1%) B-ALL and 5 (33.3%) T-ALL patients exhibited a change from diagnosis to relapse, and 25 (35.7%) B-ALL patients acquired new rearrangements at relapse. The new relapse rearrangements were present in 15 of the 19 (78.9%) diagnostic samples as shown by RQ-PCR, with a median level of 5.26 × 10-2 . The levels of minor rearrangements correlated with B immunophenotype, WBC counts, age at diagnosis, and recurrence time. Furthermore, back-tracing rearrangements in 12 patients identified three patterns of relapse clone dynamics, which suggested the recurrence mechanisms not only through clonal selection of pre-existing subclones but also through an ongoing clonal evolution during remission and relapse. CONCLUSION Backtracking Ig/TCR gene rearrangements in relapse clones of pediatric ALL revealed complex patterns of clonal selection and evolution for leukemic relapse.
Collapse
Affiliation(s)
- Ming-Zhu Jia
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Wei-Jing Li
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Chan-Juan Wang
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
| | - Qing Zhang
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Chao Gao
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Xiao-Tong Huang
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Ting Zhu
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Rui-Dong Zhang
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
| | - Lei Cui
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Zhi-Gang Li
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Luib L, Kreyenberg H, Michaelis S, Handgretinger R, Mezger M. Transferring measurable residual disease measurement in pediatric acute lymphoblastic leukemia from quantitative real-time PCR to digital droplet PCR. Pediatr Transplant 2023; 27:e14483. [PMID: 36750990 DOI: 10.1111/petr.14483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/02/2023] [Accepted: 01/23/2023] [Indexed: 02/09/2023]
Abstract
BACKGROUND Since the measurement of measurable residual disease (MRD) is part of clinical routine examination for children affected with acute lymphoblastic leukemia (ALL), continuous efforts are made to improve its method, applicability and accuracy. Whereas quantitative real-time polymerase chain reaction (qPCR) is considered as the gold standard for MRD detection and endowed with international guidelines for implementation and evaluation, these do not yet exist for digital droplet PCR (ddPCR). However, advantages are seen in droplet partitioning for MRD measurement to allow absolute quantification without depending on reference samples. METHODS In this study, 17 MRD targets of nine patients with childhood B-ALL were analyzed with qPCR and ddPCR, respectively. All patients were assigned to high risk group and had hematopoietic stem cell transplantation and CD19 antibody therapy for relapse prevention. Starting with the sequences and guidelines of qPCR and optimizing the protocol for ddPCR, the MRD targets could also be measured precisely with this novel method, using the same primer and probe sets as for qPCR. RESULTS The already established MRD protocol of qPCR could be transferred to ddPCR and all 17 MRD targets were measured in dilution series reaching comparable Limit of detection levels with both PCR methods. CONCLUSIONS With a given qPCR protocol and some experience in conventional MRD monitoring, it is conceivable to transfer the procedure of MRD measurement to ddPCR technology. Our data is in line with other studies which are summarized and discussed here as well to facilitate the transfer of MRD diagnostics to ddPCR.
Collapse
Affiliation(s)
- Luise Luib
- Department of General Paediatrics, Haematology/Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Hermann Kreyenberg
- Clinic for Children and Adolescents, Department for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt/Main, Germany
| | - Sebastian Michaelis
- Department of General Paediatrics, Haematology/Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Rupert Handgretinger
- Department of General Paediatrics, Haematology/Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Markus Mezger
- Department of General Paediatrics, Haematology/Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
4
|
Svaton M, Skotnicova A, Reznickova L, Rennerova A, Valova T, Kotrova M, van der Velden VHJ, Brüggemann M, Darzentas N, Langerak AW, Zuna J, Stary J, Trka J, Fronkova E. NGS better discriminates true MRD positivity for the risk stratification of childhood ALL treated on an MRD-based protocol. Blood 2023; 141:529-533. [PMID: 36240445 PMCID: PMC10651772 DOI: 10.1182/blood.2022017003] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/02/2022] [Accepted: 10/06/2022] [Indexed: 02/07/2023] Open
Abstract
We compared minimal/measurable residual disease (MRD) levels evaluated by routinely used real-time quantitative polymerase chain reaction (qPCR) patient-specific assays and by next-generation sequencing (NGS) approach in 780 immunoglobulin (IG) and T-cell receptor (TR) markers in 432 children with B-cell precursor acute lymphoblastic leukemia treated on the AIEOP-BFM ALL 2009 protocol. Our aim was to compare the MRD-based risk stratification at the end of induction. The results were concordant in 639 of 780 (81.9%) of these markers; 37 of 780 (4.7%) markers were detected only by NGS. In 104 of 780 (13.3%) markers positive only by qPCR, a large fraction (23/104; 22.1%) was detected also by NGS, however, owing to the presence of identical IG/TR rearrangements in unrelated samples, we classified those as nonspecific/false-positive. Risk group stratification based on the MRD results by qPCR and NGS at the end of induction was concordant in 76% of the patients; 19% of the patients would be assigned to a lower risk group by NGS, largely owing to the elimination of false-positive qPCR results, and 5% of patients would be assigned to a higher risk group by NGS. NGS MRD is highly concordant with qPCR while providing more specific results and can be an alternative in the front line of MRD evaluation in forthcoming MRD-based protocols.
Collapse
Affiliation(s)
- Michael Svaton
- CLIP–Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Aneta Skotnicova
- CLIP–Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Leona Reznickova
- CLIP–Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Andrea Rennerova
- CLIP–Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Tatana Valova
- CLIP–Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Michaela Kotrova
- Department of Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Vincent H. J. van der Velden
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Monika Brüggemann
- Department of Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nikos Darzentas
- Department of Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Anton W. Langerak
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jan Zuna
- CLIP–Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan Trka
- CLIP–Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Eva Fronkova
- CLIP–Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
5
|
Della Starza I, De Novi LA, Elia L, Bellomarino V, Beldinanzi M, Soscia R, Cardinali D, Chiaretti S, Guarini A, Foà R. Optimizing Molecular Minimal Residual Disease Analysis in Adult Acute Lymphoblastic Leukemia. Cancers (Basel) 2023; 15:374. [PMID: 36672325 PMCID: PMC9856386 DOI: 10.3390/cancers15020374] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Minimal/measurable residual disease (MRD) evaluation has resulted in a fundamental instrument to guide patient management in acute lymphoblastic leukemia (ALL). From a methodological standpoint, MRD is defined as any approach aimed at detecting and possibly quantifying residual neoplastic cells beyond the sensitivity level of cytomorphology. The molecular methods to study MRD in ALL are polymerase chain reaction (PCR) amplification-based approaches and are the most standardized techniques. However, there are some limitations, and emerging technologies, such as digital droplet PCR (ddPCR) and next-generation sequencing (NGS), seem to have advantages that could improve MRD analysis in ALL patients. Furthermore, other blood components, namely cell-free DNA (cfDNA), appear promising and are also being investigated for their potential role in monitoring tumor burden and response to treatment in hematologic malignancies. Based on the review of the literature and on our own data, we hereby discuss how emerging molecular technologies are helping to refine the molecular monitoring of MRD in ALL and may help to overcome some of the limitations of standard approaches, providing a benefit for the care of patients.
Collapse
Affiliation(s)
- Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
- GIMEMA Foundation, 00182 Rome, Italy
| | - Lucia Anna De Novi
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Loredana Elia
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Vittorio Bellomarino
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Marco Beldinanzi
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Roberta Soscia
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Deborah Cardinali
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Sabina Chiaretti
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Anna Guarini
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| |
Collapse
|
6
|
Kipf E, Schlenker F, Borst N, Fillies M, Kirschner-Schwabe R, Zengerle R, Eckert C, von Stetten F, Lehnert M. Advanced Minimal Residual Disease Monitoring for Acute Lymphoblastic Leukemia with Multiplex Mediator Probe PCR. J Mol Diagn 2021; 24:57-68. [PMID: 34757015 DOI: 10.1016/j.jmoldx.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 09/16/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most frequent malignancy in childhood. Minimal residual disease (MRD) monitoring is an important prognostic factor for treatment response and patient stratification. It uses personalized real-time PCR to measure the amount of cancer cells among normal cells. Due to clonal tumor evolution or secondary rearrangement processes, MRD markers can disappear during treatment, leading to false-negative MRD results and wrong decision-making in personalized treatments. Therefore, monitoring of multiple MRD markers per patient is required. For the first time, the authors present personalized multiplex mediator probe PCR (MP PCR) for MRD monitoring in ALL. These assays can precisely quantify more MRD markers in less sample material. Therefore, clinical outcomes will be less affected by clonal tumor evolution. Personalized duplex MP PCR assays were developed for different genomic MRD markers, including immunoglobulin/T-cell receptor gene rearrangements, gene fusions, and gene deletions. One duplex assay was successfully applied in a prospective patient case and compared with hydrolysis probes. Moreover, the authors increased the multiplex level from duplex to 4-plex and still met the EuroMRD requirements for reliable quantification. In addition, the authors' MRD-MP design guidelines and multiplex workflow facilitate and accelerate MP PCR assay development. This helps the standardization of personal diagnostics.
Collapse
Affiliation(s)
- Elena Kipf
- Hahn-Schickard, Freiburg, Germany; Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | | | - Nadine Borst
- Hahn-Schickard, Freiburg, Germany; Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Marion Fillies
- Department of Pediatric Oncology/Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Renate Kirschner-Schwabe
- Department of Pediatric Oncology/Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roland Zengerle
- Hahn-Schickard, Freiburg, Germany; Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Cornelia Eckert
- Department of Pediatric Oncology/Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix von Stetten
- Hahn-Schickard, Freiburg, Germany; Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | - Michael Lehnert
- Hahn-Schickard, Freiburg, Germany; Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Chen D, Sutton R, Giles J, Venn NC, Huang L, Law T, Subhash VV, Trahair TN, Henderson MJ. Analytical Quality Controls for ddPCR Detection of Minimal Residual Disease in Acute Lymphoblastic Leukemia. Clin Chem 2021; 67:1373-1383. [PMID: 39506256 DOI: 10.1093/clinchem/hvab117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/03/2021] [Indexed: 11/08/2024]
Abstract
BACKGROUND Droplet digital PCR (ddPCR) is a promising technique for absolute quantification of minimal residual disease (MRD) in acute lymphoblastic leukemia (ALL), but there is no comprehensive quality assurance program to enable its application in clinical laboratories. Current guidelines for real-time quantitative PCR (qPCR) assays targeting immunoglobulin/T-cell receptor (Ig/TCR) gene rearrangements needed adaptation for ddPCR to cover droplet generation, intraassay variation, and interassay variation in the absence of standard curves. METHODS Six qPCR MRD assays for Ig/TCR gene rearrangements and a standard albumin control gene assay were migrated to a ddPCR platform and used to test 82 remission samples from 6 patients with ALL. Three analytical quality controls (QC) were developed and evaluated for ddPCR MRD detection. RESULTS Analytical QC for droplet number generation (DN-QC), for albumin ddPCR assay performance (Alb-QC) and for patient-specific marker assay performance (PS-QC) were established with pass/fail limits and corresponding QC rules. Compared to established qPCRs, the ddPCR assays had comparable sensitivity and quantitative range. Overall, there was close agreement (91%) of MRD results between qPCR and ddPCR (κ = 0.86, P < 0.0001) and stronger concordance in 32 quantifiable samples (R2 = 0.97, P < 0.0001). CONCLUSIONS The use of this newly developed quality control system for ddPCR MRD testing avoids the need to repeat standard curves and provides reliable results comparable to standardized qPCR methods for MRD detection in ALL.
Collapse
Affiliation(s)
- Dan Chen
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Rosemary Sutton
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- School of Women's and Children's Health, UNSW Medicine, Randwick, Australia
| | - Jodie Giles
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Nicola C Venn
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Libby Huang
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Tamara Law
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Vinod Vijay Subhash
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Toby N Trahair
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- School of Women's and Children's Health, UNSW Medicine, Randwick, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
| | - Michelle J Henderson
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- School of Women's and Children's Health, UNSW Medicine, Randwick, Australia
| |
Collapse
|
8
|
Correia RP, Bento LC, de Sousa FA, Barroso RDS, Campregher PV, Bacal NS. How I investigate minimal residual disease in acute lymphoblastic leukemia. Int J Lab Hematol 2021; 43:354-363. [PMID: 33423385 DOI: 10.1111/ijlh.13463] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 12/21/2020] [Accepted: 12/27/2020] [Indexed: 10/22/2022]
Abstract
Minimal Residual Disease (MRD) is the most important independent prognostic factor in acute lymphoblastic leukemia (ALL) and refers to the deep level of measurable disease in cases with complete remission by conventional pathologic analysis, especially by cytomorphology. MRD can be detected by multiparametric flow cytometry, molecular approaches such as quantitative polymerase chain reaction for immunoglobulin and T-cell receptor (IG/TR) gene rearrangements or fusion genes transcript, and high-throughput sequencing for IG/TR. Despite the proven clinical usefulness in detecting MRD, these methods have differences in sensitivity, specificity, applicability, turnaround time and cost. Knowing and understanding these differences, as well as the principles and limitations of each technology, is essential to laboratory standardization and correct interpretation of MRD results in line with treatment time points, therapeutic settings, and clinical trials. Here, we review the methodological approaches to measure MRD in ALL and discuss the advantages and limitations of the most commonly used techniques.
Collapse
Affiliation(s)
- Rodolfo P Correia
- Clinical Pathology Laboratory, Flow Cytometry Division, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Laiz C Bento
- Clinical Pathology Laboratory, Flow Cytometry Division, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Flávia A de Sousa
- Clinical Pathology Laboratory, Flow Cytometry Division, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Rodrigo de S Barroso
- Clinical Pathology Laboratory, Flow Cytometry Division, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Paulo V Campregher
- Clinical Pathology Laboratory, Molecular Genetics Division, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Nydia S Bacal
- Clinical Pathology Laboratory, Flow Cytometry Division, Hospital Israelita Albert Einstein, São Paulo, Brazil.,Centro de Hematologia de São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
Abstract
Although new techniques (i.e., droplet digital-PCR, next-generation sequencing, advanced flow cytometry) are being developed, DNA-based allele-specific real-time quantitative (RQ)-PCR is still the gold standard for sensitive and accurate immunoglobulin/T cell receptor (IG/TR)-based minimal residual disease (MRD) monitoring, allowing the detection of up to 1 leukemic cell in 100,000 normal lymphoid cells. We herewith describe the standard PCR procedure which has been developed and standardized (with minor modification in single labs) through the last 20 years of activity of the EuroMRD Consortium, a volunteer activity of expert laboratories that is continuously providing education, standardization, quality control rounds, and guidelines for interpretation of RQ-PCR data.
Collapse
Affiliation(s)
- Giovanni Cazzaniga
- Centro Ricerca Tettamanti, Fondazione Tettamanti, Pediatrics, Monza, Italy.
- Department of Medicine and Surgery, University of Milan Bicocca, Monza, Italy.
| | - Simona Songia
- Centro Ricerca Tettamanti, Fondazione Tettamanti, Pediatrics, Monza, Italy
| | - Andrea Biondi
- Department of Medicine and Surgery, University of Milan Bicocca, Monza, Italy
- Pediatrics, Ospedale San Gerardo/Fondazione MBBM, Italy
| |
Collapse
|
10
|
Kim IS. Minimal residual disease in acute lymphoblastic leukemia: technical aspects and implications for clinical interpretation. Blood Res 2020; 55:S19-S26. [PMID: 32719172 PMCID: PMC7386891 DOI: 10.5045/br.2020.s004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
Minimal residual disease (MRD) monitoring has proven to be one of the fundamental independent prognostic factors for patients with acute lymphoblastic leukemia (ALL). Sequential monitoring of MRD using sensitive and specific methods, such as real-time quantitative polymerase chain reaction (qPCR) or flow cytometry (FCM), has improved the assessment of treatment response and is currently used for therapeutic stratification and early detection. Although both FCM and qPCR yield highly consistent results with sensitivities of 10‒4, each method has several limitations. For example, qPCR is time-consuming and laborious: designing primers that correspond to the immunoglobulin (IG) and T-cell receptor (TCR) gene rearrangements at diagnosis can take 3‒4 weeks. In addition, the evolution of additional clones beyond the first or index clone during therapy cannot be detected, which might lead to false-negative results. FCM requires experienced technicians and sometimes does not achieve a sensitivity of 10‒4. Accordingly, a next generation sequencing (NGS)-based method has been developed in an attempt to overcome these limitations. With the advent of high-throughput NGS technologies, a more in-depth analysis of IG and/or TCR gene rearrangements is now within reach, which impacts all applications of IG/TR analysis. However, standardization, quality control, and validation of this new technology are warranted prior to its incorporation into routine practice.
Collapse
Affiliation(s)
- In-Suk Kim
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| |
Collapse
|
11
|
Brown NA, Elenitoba-Johnson KSJ. Enabling Precision Oncology Through Precision Diagnostics. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2020; 15:97-121. [PMID: 31977297 DOI: 10.1146/annurev-pathmechdis-012418-012735] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genomic testing enables clinical management to be tailored to individual cancer patients based on the molecular alterations present within cancer cells. Genomic sequencing results can be applied to detect and classify cancer, predict prognosis, and target therapies. Next-generation sequencing has revolutionized the field of cancer genomics by enabling rapid and cost-effective sequencing of large portions of the genome. With this technology, precision oncology is quickly becoming a realized paradigm for managing the treatment of cancer patients. However, many challenges must be overcome to efficiently implement the transition of next-generation sequencing from research applications to routine clinical practice, including using specimens commonly available in the clinical setting; determining how to process, store, and manage large amounts of sequencing data; determining how to interpret and prioritize molecular findings; and coordinating health professionals from multiple disciplines.
Collapse
Affiliation(s)
- Noah A Brown
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA;
| | - Kojo S J Elenitoba-Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
12
|
Li Z, Jiang N, Lim EH, Chin WHN, Lu Y, Chiew KH, Kham SKY, Yang W, Quah TC, Lin HP, Tan AM, Ariffin H, Yang JJ, Yeoh AEJ. Identifying IGH disease clones for MRD monitoring in childhood B-cell acute lymphoblastic leukemia using RNA-Seq. Leukemia 2020; 34:2418-2429. [PMID: 32099036 DOI: 10.1038/s41375-020-0774-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 02/07/2020] [Accepted: 02/13/2020] [Indexed: 12/19/2022]
Abstract
Identifying patient-specific clonal IGH/TCR junctional sequences is critical for minimal residual disease (MRD) monitoring. Conventionally these junctional sequences are identified using laborious Sanger sequencing of excised heteroduplex bands. We found that the IGH is highly expressed in our diagnostic B-cell acute lymphoblastic leukemia (B-ALL) samples using RNA-Seq. Therefore, we used RNA-Seq to identify IGH disease clone sequences in 258 childhood B-ALL samples for MRD monitoring. The amount of background IGH rearrangements uncovered by RNA-Seq followed the Zipf's law with IGH disease clones easily identified as outliers. Four hundred and ninety-seven IGH disease clones (median 2, range 0-7 clones/patient) are identified in 90.3% of patients. High hyperdiploid patients have the most IGH disease clones (median 3) while DUX4 subtype has the least (median 1) due to the rearrangements involving the IGH locus. In all, 90.8% of IGH disease clones found by Sanger sequencing are also identified by RNA-Seq. In addition, RNA-Seq identified 43% more IGH disease clones. In 69 patients lacking sensitive IGH targets, targeted NGS IGH MRD showed high correlation (R = 0.93; P = 1.3 × 10-14), better relapse prediction than conventional RQ-PCR MRD using non-IGH targets. In conclusion, RNA-Seq can identify patient-specific clonal IGH junctional sequences for MRD monitoring, adding to its usefulness for molecular diagnosis in childhood B-ALL.
Collapse
Affiliation(s)
- Zhenhua Li
- VIVA-NUS Centre for Translational Research in Acute Leukaemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nan Jiang
- VIVA-NUS Centre for Translational Research in Acute Leukaemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Evelyn Huizi Lim
- VIVA-NUS Centre for Translational Research in Acute Leukaemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Winnie Hui Ni Chin
- VIVA-NUS Centre for Translational Research in Acute Leukaemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yi Lu
- VIVA-NUS Centre for Translational Research in Acute Leukaemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kean Hui Chiew
- VIVA-NUS Centre for Translational Research in Acute Leukaemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shirley Kow Yin Kham
- VIVA-NUS Centre for Translational Research in Acute Leukaemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wentao Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Thuan Chong Quah
- VIVA-NUS Centre for Translational Research in Acute Leukaemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Viva-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
| | - Hai Peng Lin
- Subang Jaya Medical Centre, Subang Jaya, Malaysia
| | - Ah Moy Tan
- Department of Paediatrics, KK Women's & Children's Hospital, Singapore, Singapore
| | - Hany Ariffin
- University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Allen Eng-Juh Yeoh
- VIVA-NUS Centre for Translational Research in Acute Leukaemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Viva-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore. .,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
13
|
Bai Y, Chim CS. Ficolled bone marrow is superior to bone marrow buffy coat for detection of minimal residual disease in multiple myeloma. ACTA ACUST UNITED AC 2019; 24:533-537. [PMID: 31280705 DOI: 10.1080/16078454.2019.1637574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Buffy coat and ficoll of bone marrow (BM) are viable options for the study of minimal residual disease (MRD) in multiple myeloma (MM). As yet, there is no data about the superiority of either sample types. Herein, we aimed to address this issue. METHODS Forty pairs of ficolled BMs and BM buffy coats of 19 MM patients were studied for MRD by allele-specific oligonucleotide real-time quantitative PCR, with patient-specific primers/probes whenever appropriate. RESULTS There were 41 pairs of MRD data for comparison analysis due to one patient with biclonal disease. MRD levels in ficolls and buffy coats were highly concordant (rs = 0.936, P < 0.0001), with 31 (76%) and seven (17%) pairs being concomitantly MRD-positive or -negative. On the other hand, apart from the 16 pairs being both MRD-negative, or -positive but not quantifiable in ficolls and buffy coats, majority (n = 22, 88%) had higher MRD levels in ficolled BMs than BM buffy coats. Furthermore, in 17 pairs, in which MRD was quantifiable in both, MRD levels in ficolled BMs were 3.1 times those of BM buffy coats (median, 567/105 vs. 184/105, P = 0.001). CONCLUSION Taken together, ficolled BM is more sensitive than BM buffy coat for MRD detection in MM, hence should be recommended.
Collapse
Affiliation(s)
- Yinlei Bai
- a Institute for Immunology and School of Medicine , Tsinghua University , Beijing , People's Republic of China
| | - Chor Sang Chim
- b Department of Medicine , The University of Hong Kong , Hong Kong , People's Republic of China
| |
Collapse
|
14
|
Del Giudice I, Raponi S, Della Starza I, De Propris MS, Cavalli M, De Novi LA, Cappelli LV, Ilari C, Cafforio L, Guarini A, Foà R. Minimal Residual Disease in Chronic Lymphocytic Leukemia: A New Goal? Front Oncol 2019; 9:689. [PMID: 31555576 PMCID: PMC6727319 DOI: 10.3389/fonc.2019.00689] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022] Open
Abstract
In chronic lymphocytic leukemia (CLL), there is a growing interest for minimal residual disease (MRD) monitoring, due to the availability of drug combinations capable of unprecedented complete clinical responses. The standardized and most commonly applied methods to assess MRD in CLL are based on flow cytometry (FCM) and, to a lesser extent, real-time quantitative PCR (RQ-PCR) with allele-specific oligonucleotide (ASO) primers of immunoglobulin heavy chain genes (IgH). Promising results are being obtained using droplet digital PCR (ddPCR) and next generation sequencing (NGS)-based approaches, with some advantages and a potential higher sensitivity compared to the standardized methodologies. Plasma cell-free DNA can also be explored as a more precise measure of residual disease from all different compartments, including the lymph nodes. From a clinical point of view, CLL MRD quantification has proven an independent prognostic marker of progression-free survival (PFS) and overall survival (OS) after chemoimmunotherapy as well as after allogeneic transplantation. In the era of mechanism-driven drugs, the paradigms of CLL treatment are being revolutionized, challenging the use of chemoimmunotherapy even in first-line. The continuous administration of ibrutinib single agent has led to prolonged PFS and OS in relapsed/refractory and treatment naïve CLL, including those with TP53 deletion/mutation or unmutated IGHV genes, though the clinical responses are rarely complete. More recently, chemo-free combinations of venetoclax+rituximab, venetoclax+obinutuzumab or ibrutinib+venetoclax have been shown capable of inducing undetectable MRD in the bone marrow, opening the way to protocols exploring a MRD-based duration of treatment, aiming at disease eradication. Thus, beside a durable disease control desirable particularly for older patients and/or for those with comorbidities, a MRD-negative complete remission is becoming a realistic prospect for CLL patients in an attempt to obtain a long-lasting eradication and possibly cure of the disease. Here we discuss the standardized and innovative technical approaches for MRD detection in CLL, the clinical impact of MRD monitoring in chemoimmunotherapy and chemo-free trials and the future clinical implications of MRD monitoring in CLL patients outside of clinical trials.
Collapse
Affiliation(s)
- Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Sara Raponi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.,GIMEMA Foundation, Rome, Italy
| | - Maria Stefania De Propris
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marzia Cavalli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Anna De Novi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Luca Vincenzo Cappelli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Caterina Ilari
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Luciana Cafforio
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Anna Guarini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
15
|
Della Starza I, Chiaretti S, De Propris MS, Elia L, Cavalli M, De Novi LA, Soscia R, Messina M, Vitale A, Guarini A, Foà R. Minimal Residual Disease in Acute Lymphoblastic Leukemia: Technical and Clinical Advances. Front Oncol 2019; 9:726. [PMID: 31448230 PMCID: PMC6692455 DOI: 10.3389/fonc.2019.00726] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/22/2019] [Indexed: 12/28/2022] Open
Abstract
Introduction: Acute lymphoblastic leukemia (ALL) is the first neoplasm where the assessment of early response to therapy by minimal residual disease (MRD) monitoring has proven to be a fundamental tool to guide therapeutic choices. The most standardized methods to study MRD in ALL are multi-parametric flow cytometry (MFC) and polymerase chain reaction (PCR) amplification-based methods. Emerging technologies hold the promise to improve MRD detection in ALL patients. Moreover, novel therapies, such as monoclonal antibodies, bispecific T-cell engagers, and chimeric antigen receptor T cells (CART) represent exciting advancements in the management of B-cell precursor (BCP)-ALL. Aims: Through a review of the literature and in house data, we analyze the current status of MRD assessment in ALL to better understand how some of its limitations could be overcome by emerging molecular technologies. Furthermore, we highlight the future role of MRD monitoring in the context of personalized protocols, taking into account the genetic complexity in ALL. Results and Conclusions: Molecular rearrangements (gene fusions and immunoglobulin and T-cell receptor-IG/TR gene rearrangements) are widely used as targets to detect residual leukemic cells in ALL patients. The advent of novel techniques, namely next generation flow cytometry (NGF), digital-droplet-PCR (ddPCR), and next generation sequencing (NGS) appear important tools to evaluate MRD in ALL, since they have the potential to overcome the limitations of standard approaches. It is likely that in the forthcoming future these techniques will be incorporated in clinical trials, at least at decisional time points. Finally, the advent of new powerful compounds is further increasing MRD negativity rates, with benefits in long-term survival and a potential reduction of therapy-related toxicities. However, the prognostic relevance in the setting of novel immunotherapies still needs to be evaluated.
Collapse
Affiliation(s)
- Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.,GIMEMA Foundation, Rome, Italy
| | - Sabina Chiaretti
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria S De Propris
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Loredana Elia
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marzia Cavalli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia A De Novi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Roberta Soscia
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Monica Messina
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonella Vitale
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Anna Guarini
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
16
|
Rocha JMC, Xavier SG, Souza MEDL, Murao M, de Oliveira BM. Comparison between flow cytometry and standard PCR in the evaluation of MRD in children with acute lymphoblastic leukemia treated with the GBTLI LLA - 2009 protocol. Pediatr Hematol Oncol 2019; 36:287-301. [PMID: 31287348 DOI: 10.1080/08880018.2019.1636168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Minimal residual disease (MRD) monitoring is of prognostic importance in childhood acute lymphoblastic leukemia (ALL). The detection of immunoglobulin and T-cell receptor gene rearrangements by real-time quantitative PCR (RT-PCR) is considered the gold standard for this evaluation. However, more accessible methods also show satisfactory performance. This study aimed to compare MRD analysis by four-color flow cytometry (FC) and qualitative standard PCR on days 35 and 78 of chemotherapy and to correlate these data with patients' clinical characteristics. Forty-two children with a recent diagnosis of ALL, admitted to a public hospital in Brazil for treatment in accordance with the Brazilian Childhood Cooperative Group for ALL Treatment (GBTLI LLA-2009), were included. Bone marrow samples collected at diagnosis and on days 35 and 78 of treatment were analyzed for the immunophenotypic characterization of blasts by FC and for the detection of clonal rearrangements by standard PCR. Paired analyses were performed in 61/68 (89.7%) follow-up samples, with a general agreement of 88.5%. Disagreements were resolved by RT-PCR, which evidenced one false-negative and four false-positive results in FC, as well as two false-negative results in PCR. Among the prognostic factors, a significant association was found only between T-cell lineage and MRD by standard PCR. These results show that FC and standard PCR produce similar results in MRD detection of childhood ALL and that both methodologies may be useful in the monitoring of disease treatment, especially in regions with limited financial resources.
Collapse
Affiliation(s)
| | | | | | - Mitiko Murao
- Federal University of Minas Gerais (UFMG) , Belo Horizonte , MG , Brazil
| | | |
Collapse
|
17
|
Theunissen PMJ, de Bie M, van Zessen D, de Haas V, Stubbs AP, van der Velden VHJ. Next-generation antigen receptor sequencing of paired diagnosis and relapse samples of B-cell acute lymphoblastic leukemia: Clonal evolution and implications for minimal residual disease target selection. Leuk Res 2018; 76:98-104. [PMID: 30389174 DOI: 10.1016/j.leukres.2018.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/01/2018] [Accepted: 10/18/2018] [Indexed: 11/26/2022]
Abstract
Antigen receptor gene rearrangements are frequently applied as molecular targets for detection of minimal residual disease (MRD) in B-cell precursor acute lymphoblastic leukemia patients. Since such targets may be lost at relapse, appropriate selection of antigen receptor genes as MRD-PCR target is critical. Recently, next-generation sequencing (NGS) - much more sensitive and quantitative than classical PCR-heteroduplex approaches - has been introduced for identification of MRD-PCR targets. We evaluated 42 paired diagnosis-relapse samples by NGS (IGH, IGK, TRG, TRD, and TRB) to evaluate clonal evolution patterns and to design an algorithm for selection of antigen receptor gene rearrangements most likely to remain stable at relapse. Overall, only 393 out of 1446 (27%) clonal rearrangements were stable between diagnosis and relapse. If only index clones with a frequency >5% at diagnosis were taken into account, this number increased to 65%; including only index clones with an absolute read count >10,000, indicating truly major clones, further increased the stability to 84%. Over 90% of index clones at relapse were also present as index clone at diagnosis. Our data provide detailed information about the stability of antigen receptor gene rearrangements, based on which we propose an algorithm for selecting stable MRD-PCR targets, successful in >97% of patients.
Collapse
Affiliation(s)
- Prisca M J Theunissen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Maaike de Bie
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - David van Zessen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands; Department of Bioinformatics, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | | | - Andrew P Stubbs
- Department of Bioinformatics, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | | |
Collapse
|
18
|
García-Barchino MJ, Sarasquete ME, Panizo C, Morscio J, Martinez A, Alcoceba M, Fresquet V, Gonzalez-Farre B, Paiva B, Young KH, Robles EF, Roa S, Celay J, Larrayoz M, Rossi D, Gaidano G, Montes-Moreno S, Piris MA, Balanzategui A, Jimenez C, Rodriguez I, Calasanz MJ, Larrayoz MJ, Segura V, Garcia-Muñoz R, Rabasa MP, Yi S, Li J, Zhang M, Xu-Monette ZY, Puig-Moron N, Orfao A, Böttcher S, Hernandez-Rivas JM, Miguel JS, Prosper F, Tousseyn T, Sagaert X, Gonzalez M, Martinez-Climent JA. Richter transformation driven by Epstein-Barr virus reactivation during therapy-related immunosuppression in chronic lymphocytic leukaemia. J Pathol 2018; 245:61-73. [PMID: 29464716 DOI: 10.1002/path.5060] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/19/2018] [Accepted: 02/15/2018] [Indexed: 12/22/2022]
Abstract
The increased risk of Richter transformation (RT) in patients with chronic lymphocytic leukaemia (CLL) due to Epstein-Barr virus (EBV) reactivation during immunosuppressive therapy with fludarabine other targeted agents remains controversial. Among 31 RT cases classified as diffuse large B-cell lymphoma (DLBCL), seven (23%) showed EBV expression. In contrast to EBV- tumours, EBV+ DLBCLs derived predominantly from IGVH-hypermutated CLL, and they also showed CLL-unrelated IGVH sequences more frequently. Intriguingly, despite having different cellular origins, clonally related and unrelated EBV+ DLBCLs shared a previous history of immunosuppressive chemo-immunotherapy, a non-germinal centre DLBCL phenotype, EBV latency programme type II or III, and very short survival. These data suggested that EBV reactivation during therapy-related immunosuppression can transform either CLL cells or non-tumoural B lymphocytes into EBV+ DLBCL. To investigate this hypothesis, xenogeneic transplantation of blood cells from 31 patients with CLL and monoclonal B-cell lymphocytosis (MBL) was performed in Rag2-/- IL2γc-/- mice. Remarkably, the recipients' impaired immunosurveillance favoured the spontaneous outgrowth of EBV+ B-cell clones from 95% of CLL and 64% of MBL patients samples, but not from healthy donors. Eventually, these cells generated monoclonal tumours (mostly CLL-unrelated but also CLL-related), recapitulating the principal features of EBV+ DLBCL in patients. Accordingly, clonally related and unrelated EBV+ DLBCL xenografts showed indistinguishable cellular, virological and molecular features, and synergistically responded to combined inhibition of EBV replication with ganciclovir and B-cell receptor signalling with ibrutinib in vivo. Our study underscores the risk of RT driven by EBV in CLL patients receiving immunosuppressive therapies, and provides the scientific rationale for testing ganciclovir and ibrutinib in EBV+ DLBCL. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Maria J García-Barchino
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain
| | - Maria E Sarasquete
- Department of Haematology, University Hospital, and Institute of Molecular and Cellular Biology of Cancer, CIBERONC, University of Salamanca, Salamanca, Spain
| | - Carlos Panizo
- Department of Haematology, Clinica Universidad de Navarra, CIBERONC, University of Navarra, Pamplona, Spain
| | - Julie Morscio
- KU Leuven, Translational Cell and Tissue Research, Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Antonio Martinez
- Haematopathology Section, Hospital Clinic, Institut d'Investigacions Biomediques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Miguel Alcoceba
- Department of Haematology, University Hospital, and Institute of Molecular and Cellular Biology of Cancer, CIBERONC, University of Salamanca, Salamanca, Spain
| | - Vicente Fresquet
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain
| | - Blanca Gonzalez-Farre
- Haematopathology Section, Hospital Clinic, Institut d'Investigacions Biomediques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Bruno Paiva
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eloy F Robles
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain
| | - Sergio Roa
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain
| | - Jon Celay
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain
| | - Marta Larrayoz
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain
| | - Davide Rossi
- Division of Haematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Gianluca Gaidano
- Division of Haematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Santiago Montes-Moreno
- Department of Pathology, Hospital Universitario and Instituto de Formacion e Investigacion Marques de Valdecilla, Santander, Spain
| | - Miguel A Piris
- Department of Pathology, Hospital Universitario and Instituto de Formacion e Investigacion Marques de Valdecilla, Santander, Spain
| | - Ana Balanzategui
- Department of Haematology, University Hospital, and Institute of Molecular and Cellular Biology of Cancer, CIBERONC, University of Salamanca, Salamanca, Spain
| | - Cristina Jimenez
- Department of Haematology, University Hospital, and Institute of Molecular and Cellular Biology of Cancer, CIBERONC, University of Salamanca, Salamanca, Spain
| | - Idoia Rodriguez
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain
| | - Maria J Calasanz
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain.,Department of Genetics, School of Medicine, University of Navarra, Pamplona, Spain
| | - Maria J Larrayoz
- Department of Genetics, School of Medicine, University of Navarra, Pamplona, Spain
| | - Victor Segura
- Bio-informatics Unit, Department of Genomics and Proteomics, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | - Maria P Rabasa
- Department of Haematology, Hospital San Pedro, Logroño, Spain
| | - Shuhua Yi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianyong Li
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mingzhi Zhang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zijun Y Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Noemi Puig-Moron
- Department of Haematology, University Hospital, and Institute of Molecular and Cellular Biology of Cancer, CIBERONC, University of Salamanca, Salamanca, Spain
| | - Alberto Orfao
- Cancer Research Centre, Institute for Biomedical Research of Salamanca and Department of Medicine and Cytometry Service, CIBERONC, University of Salamanca, Salamanca, Spain
| | - Sebastian Böttcher
- Medical Clinic II, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Jesus M Hernandez-Rivas
- Department of Haematology, University Hospital, and Institute of Molecular and Cellular Biology of Cancer, CIBERONC, University of Salamanca, Salamanca, Spain
| | - Jesus San Miguel
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain.,Department of Haematology, Clinica Universidad de Navarra, CIBERONC, University of Navarra, Pamplona, Spain
| | - Felipe Prosper
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain.,Department of Haematology, Clinica Universidad de Navarra, CIBERONC, University of Navarra, Pamplona, Spain
| | - Thomas Tousseyn
- KU Leuven, Translational Cell and Tissue Research, Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Xavier Sagaert
- KU Leuven, Translational Cell and Tissue Research, Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Marcos Gonzalez
- Department of Haematology, University Hospital, and Institute of Molecular and Cellular Biology of Cancer, CIBERONC, University of Salamanca, Salamanca, Spain
| | - Jose A Martinez-Climent
- Division of Haematological Oncology, Centre for Applied Medical Research (CIMA), CIBERONC, University of Navarra, Pamplona, Spain
| |
Collapse
|
19
|
Lokvenc M, Kalinova M, Forsterova K, Klener P, Trneny M, Fronkova E, Kodet R. Cyclin D1 mRNA as a molecular marker for minimal residual disease monitoring in patients with mantle cell lymphoma. Ann Hematol 2017; 97:467-474. [PMID: 29273915 DOI: 10.1007/s00277-017-3210-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/13/2017] [Indexed: 11/28/2022]
Abstract
Chromosomal translocation t(11;14)(q13;q32) is a characteristic molecular marker of mantle cell lymphoma (MCL) and leads to the fusion of the immunoglobulin heavy chain enhancer-promoter with the cyclin D1 gene. Both aberrant cyclin D1 expression and underlying chromosomal aberration may be used as molecular targets for monitoring minimal residual disease (MRD). The present study aims to assess the usefulness of quantitative cyclin D1 gene expression compared to the standardised but more technologically demanding DNA-based method for immunoglobulin heavy chain (IGH) or t(11;14) clone-specific gene rearrangement quantification in a cohort of bone marrow (BM) and peripheral blood (PB) samples from patients with MCL. We simultaneously evaluated DNA-MRD and cyclin D1 expression levels in 234 samples from 57 patients. We observed that both in DNA-MRD positive and negative BM/PB pairs from the same time points the expression levels of cyclin D1 are lower in PB than in BM (median 19×, BM/PB range 0.41-352). The correlation of cyclin D1 transcript levels with DNA-MRD or with flow cytometry was good only in samples with a very high infiltration. In DNA-MRD-negative BM samples, we observed a significant heterogeneity of cyclin D1 expression (in the range of more than three orders of magnitude). This is in contrast to previous reports demonstrating the usefulness of cyclin D1 for MRD monitoring that did not use DNA-based method as a reference. In PB, the specificity of cyclin D1 expression was better due to a lower physiological background. In conclusion, we show that cyclin D1 is unsuitable for MRD monitoring in BM.
Collapse
Affiliation(s)
- M Lokvenc
- Department of Pathology and Molecular Medicine, University Hospital Motol, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marketa Kalinova
- Department of Pathology and Molecular Medicine, University Hospital Motol, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - K Forsterova
- First Department of Medicine - Department of Haematology, Charles University General Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - P Klener
- First Department of Medicine - Department of Haematology, Charles University General Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - M Trneny
- First Department of Medicine - Department of Haematology, Charles University General Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - E Fronkova
- CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - R Kodet
- Department of Pathology and Molecular Medicine, University Hospital Motol, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
20
|
Bai Y, Orfao A, Chim CS. Molecular detection of minimal residual disease in multiple myeloma. Br J Haematol 2017; 181:11-26. [PMID: 29265356 DOI: 10.1111/bjh.15075] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite the significantly higher complete remission rates and improved survival achieved in the last decade, multiple myeloma (MM) patients continue to relapse due to persistence of minimal residual disease (MRD). Generally, MRD refers to persistence of low levels of disease in the order of one tumour cell in ≥105 normal cells. Currently, molecular and immunophenotypic techniques are employed for MRD detection. This review focuses on MRD detection by molecular techniques, including allele-specific oligonucleotide polymerase chain reaction (ASO-PCR), next-generation sequencing (NGS) and digital PCR (dPCR), in addition to a brief description of, and comparison with, multiparameter flow cytometry. The basic principles, technical advantages and limitations, and the clinical impact of all three molecular techniques are reviewed and compared. They all have a sensitivity of at least 10-5 , among which ASO real-time quantitative PCR is the most well-standardized, and NGS carries the highest sensitivity and applicability, while dPCR is still under investigation. Furthermore, molecular MRD negativity is a favourable prognostic factor for survival of patients with MM. However, several challenges inherent to molecular detection of MRD still remain to be overcome, particularly false negativity and failure to detect extramedullary disease. Finally, detection of MRD from peripheral blood remains challenging.
Collapse
Affiliation(s)
- Yinlei Bai
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Spain
| | - Alberto Orfao
- Department of Medicine, University of Salamanca and University Hospital of Salamanca-IBSAL, Salamanca, Spain
| | - Chor Sang Chim
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Spain
| |
Collapse
|
21
|
Nunes V, Cazzaniga G, Biondi A. An update on PCR use for minimal residual disease monitoring in acute lymphoblastic leukemia. Expert Rev Mol Diagn 2017; 17:953-963. [PMID: 28891364 DOI: 10.1080/14737159.2017.1377073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Acute lymphoblastic leukemia (ALL) is the first neoplasm where the assessment of early response to therapy by minimal residual disease (MRD) monitoring has proven to be a fundamental tool for guiding therapeutic choices. In recent years, thanks to real-time quantitative PCR (qPCR), MRD monitoring has further achieved higher levels of sensitivity and standardization. However, some outstanding issues still remain to be addressed and emerging technologies hold the promise of improving MRD detection in ALL patients. Areas covered: Through a comprehensive review of the literature, we analyze the state-of-the-art of molecular MRD assessment in ALL to better understand how, in the upcoming years, some of its limitations could be tackled by emerging molecular technologies. Furthermore, we highlight the future role of molecular MRD monitoring in the context of personalized protocols, taking into account the growing genetic complexity in ALL. Expert commentary: Although new molecular technologies are promising tools for MRD assessment, qPCR still remains the gold standard for evaluating MRD in ALL. High-throughput sequencing and droplet digital PCR allow to identify new prognostic factors and/or MRD targets at diagnosis and to perform earlier MRD evaluations, thereby optimizing patient stratification and earlier MRD-based clinical intervention to improve ALL patient outcomes.
Collapse
Affiliation(s)
- Vittorio Nunes
- a Tettamanti Research Center, Department of Pediatrics , University of Milano Bicocca , Monza , Italy
| | - Gianni Cazzaniga
- a Tettamanti Research Center, Department of Pediatrics , University of Milano Bicocca , Monza , Italy
| | - A Biondi
- a Tettamanti Research Center, Department of Pediatrics , University of Milano Bicocca , Monza , Italy
| |
Collapse
|
22
|
Kotrova M, Trka J, Kneba M, Brüggemann M. Is Next-Generation Sequencing the way to go for Residual Disease Monitoring in Acute Lymphoblastic Leukemia? Mol Diagn Ther 2017; 21:481-492. [DOI: 10.1007/s40291-017-0277-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
23
|
Monitoring of childhood ALL using BCR-ABL1 genomic breakpoints identifies a subgroup with CML-like biology. Blood 2017; 129:2771-2781. [PMID: 28331056 DOI: 10.1182/blood-2016-11-749978] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/17/2017] [Indexed: 12/20/2022] Open
Abstract
We used the genomic breakpoint between BCR and ABL1 genes for the DNA-based monitoring of minimal residual disease (MRD) in 48 patients with childhood acute lymphoblastic leukemia (ALL). Comparing the results with standard MRD monitoring based on immunoglobulin/T-cell receptor (Ig/TCR) gene rearrangements and with quantification of IKZF1 deletion, we observed very good correlation for the methods in a majority of patients; however, >20% of children (25% [8/32] with minor and 12.5% [1/8] with major-BCR-ABL1 variants in the consecutive cohorts) had significantly (>1 log) higher levels of BCR-ABL1 fusion than Ig/TCR rearrangements and/or IKZF1 deletion. We performed cell sorting of the diagnostic material and assessed the frequency of BCR-ABL1-positive cells in various hematopoietic subpopulations; 12% to 83% of non-ALL B lymphocytes, T cells, and/or myeloid cells harbored the BCR-ABL1 fusion in patients with discrepant MRD results. The multilineage involvement of the BCR-ABL1-positive clone demonstrates that in some patients diagnosed with BCR-ABL1-positive ALL, a multipotent hematopoietic progenitor is affected by the BCR-ABL1 fusion. These patients have BCR-ABL1-positive clonal hematopoiesis resembling a chronic myeloid leukemia (CML)-like disease manifesting in "lymphoid blast crisis." The biological heterogeneity of BCR-ABL1-positive ALL may impact the patient outcomes and optimal treatment (early stem cell transplantation vs long-term administration of tyrosine-kinase inhibitors) as well as on MRD testing. Therefore, we recommend further investigations on CML-like BCR-ABL1-positive ALL.
Collapse
|
24
|
Next-generation sequencing indicates false-positive MRD results and better predicts prognosis after SCT in patients with childhood ALL. Bone Marrow Transplant 2017; 52:962-968. [PMID: 28244980 DOI: 10.1038/bmt.2017.16] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/19/2016] [Accepted: 01/03/2017] [Indexed: 11/09/2022]
Abstract
Minimal residual disease (MRD) monitoring via quantitative PCR (qPCR) detection of Ag receptor gene rearrangements has been the most sensitive method for predicting prognosis and making post-transplant treatment decisions for patients with ALL. Despite the broad clinical usefulness and standardization of this method, we and others have repeatedly reported the possibility of false-positive MRD results caused by massive B-lymphocyte regeneration after stem cell transplantation (SCT). Next-generation sequencing (NGS) enables precise and sensitive detection of multiple Ag receptor rearrangements, thus providing a more specific readout compared to qPCR. We investigated two cohorts of children with ALL who underwent SCT (30 patients and 228 samples). The first cohort consisted of 17 patients who remained in long-term CR after SCT despite having low MRD positivity (<0.01%) at least once during post-SCT monitoring using qPCR. Only one of 27 qPCR-positive samples was confirmed to be positive by NGS. Conversely, 10 of 15 samples with low qPCR-detected MRD positivity from 13 patients who subsequently relapsed were also confirmed to be positive by NGS (P=0.002). These data show that NGS has a better specificity in post-SCT ALL management and indicate that treatment interventions aimed at reverting impending relapse should not be based on qPCR only.
Collapse
|
25
|
Besbes S, Hamadou WS, Boulland ML, Youssef YB, Achour B, Regaieg H, Khelif A, Fest T, Soua Z. Minimal residual disease detection in Tunisian B-acute lymphoblastic leukemia based on immunoglobulin gene rearrangements. ACTA ACUST UNITED AC 2017; 50:e5426. [PMID: 28099581 PMCID: PMC5264541 DOI: 10.1590/1414-431x20165426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 10/11/2016] [Indexed: 11/22/2022]
Abstract
IGH gene rearrangement and IGK-Kde gene deletion can be used as molecular markers for the assessment of B lineage acute lymphoblastic leukemia (B-ALL). Minimal residual disease detected based on those markers is currently the most reliable prognosis factor in B-ALL. The aim of this study was to use clonal IGH/IGK-Kde gene rearrangements to confirm B-ALL diagnosis and to evaluate the treatment outcome of Tunisian leukemic patients by monitoring the minimal residual disease (MRD) after induction chemotherapy. Seventeen consecutive newly diagnosed B-ALL patients were investigated by multiplex PCR assay and real time quantitative PCR according to BIOMED 2 conditions. The vast majority of clonal VH-JH rearrangements included VH3 gene. For IGK deletion, clonal VK1f/6-Kde recombinations were mainly identified. These rearrangements were quantified to follow-up seven B-ALL after induction using patient-specific ASO. Four patients had an undetectable level of MRD with a sensitivity of up to 10-5. This molecular approach allowed identification of prognosis risk group and adequate therapeutic decision. The IGK-Kde and IGH gene rearrangements might be used for diagnosis and MRD monitoring of B-ALL, introduced for the first time in Tunisian laboratories.
Collapse
Affiliation(s)
- S Besbes
- Research Unit 14 ES 19, Department of Biochemistry, Faculty of Medicine, University of Sousse, Sousse, Tunisia
| | - W S Hamadou
- Research Unit 14 ES 19, Department of Biochemistry, Faculty of Medicine, University of Sousse, Sousse, Tunisia
| | - M L Boulland
- Biological Hematology Department, Centre Hospitalier Universitaire Pontchaillou, Rennes, France
| | - Y B Youssef
- Research Unit 14 ES 19, Department of Biochemistry, Faculty of Medicine, University of Sousse, Sousse, Tunisia.,Clinical Hematology Department, Hospital F. Hached, Sousse, Tunisia
| | - B Achour
- Clinical Hematology Department, Hospital F. Hached, Sousse, Tunisia
| | - H Regaieg
- Research Unit 14 ES 19, Department of Biochemistry, Faculty of Medicine, University of Sousse, Sousse, Tunisia.,Clinical Hematology Department, Hospital F. Hached, Sousse, Tunisia
| | - A Khelif
- Research Unit 14 ES 19, Department of Biochemistry, Faculty of Medicine, University of Sousse, Sousse, Tunisia.,Clinical Hematology Department, Hospital F. Hached, Sousse, Tunisia
| | - T Fest
- Biological Hematology Department, Centre Hospitalier Universitaire Pontchaillou, Rennes, France
| | - Z Soua
- Research Unit 14 ES 19, Department of Biochemistry, Faculty of Medicine, University of Sousse, Sousse, Tunisia
| |
Collapse
|
26
|
Bai Y, Wong KY, Fung TK, Chim CS. High applicability of ASO-RQPCR for detection of minimal residual disease in multiple myeloma by entirely patient-specific primers/probes. J Hematol Oncol 2016; 9:107. [PMID: 27724958 PMCID: PMC5057274 DOI: 10.1186/s13045-016-0336-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 10/04/2016] [Indexed: 11/10/2022] Open
Abstract
Allele-specific oligonucleotide real-time quantitative PCR (ASO-RQPCR) is a standardized technique for detection and monitoring of minimal residual disease (MRD) in acute lymphoblastic leukemia (ALL) but not multiple myeloma (MM) due to a low applicability inherent with presence of somatic hypermutation. Herein, by a staged PCR approach and sequencing, clonality and tumor-specific complementarity-determining region 3 (CDR3) sequence were identified in 13/13 MM by sequential PCR of IgH VDJ (n = 10), IgH DJ (n = 2), or IgK VJ (n = 1). Using consensus primers/probes conventionally employed in ALL, ASO-RQPCR worked in three (23.1 %) cases only. Conversely, using entirely patient-specific primers/probes, ASO-RQPCR was applicable in eight (61.5 %) cases with a sensitivity of 5 × 10−4–10−5. Moreover, using standard curves constructed by serial dilution of plasmids cloned with patient-specific CDR3, ASO-RQPCR was successful in 12 (92.3 %) cases with a sensitivity of 10−4–10−5, but not in a case lacking an N region, in which design of a tumor-specific ASO primer was precluded. Finally, in a patient in complete response (CR), further reduction of MRD after autologous stem cell transplantation (ASCT) was demonstrated. In summary, using entirely patient-specific primers/probes, ASO-RQPCR was applicable in >90 % MM patients and enabled detection of dynamic changes of MRD before and after ASCT despite conventional CR.
Collapse
Affiliation(s)
- Yinlei Bai
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kwan Yeung Wong
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Tsz Kin Fung
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chor Sang Chim
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
27
|
Besbes S, Hamadou WS, Boulland ML, Lefranc MP, Ben Youssef Y, Achour B, Khelif A, Fest T, Soua Z. Combined IKZF1 and IG markers as new tools for diagnosis and minimal residual disease assessment in Tunisian B-ALL. Bull Cancer 2016; 103:822-828. [PMID: 27614734 DOI: 10.1016/j.bulcan.2016.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/21/2016] [Accepted: 07/23/2016] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The monitoring of minimal residual disease (MRD) approach in patients diagnosed with B-acute lymphoblastic leukemia (B-ALL) allows an early detection of residual clones inducing relapses and therefore appropriate therapy strategy. The molecular markers may identify and quantify the residual blasts in B-ALL with normal cytology. In this study, we aimed to use combined IKZF1, IGH and IGK immunoglobulin genes for diagnosis and MRD monitoring in B-ALL sample using MLPA, multiplex PCR and real-time quantitative PCR. MATERIAL We showed that multiplex PCR and MLPA are necessary and complementary to detect IKZF1 deletions. RESULTS We have identified at the diagnosis clonal IGH rearrangement (VH3-JH5) and IKZF1 deletion (Δ4-7), which we have used it for MRD evaluation after induction chemotherapy. Despite the absence of chromosome abnormality, the patient may be classified in high-risk group with a relapse rate of residual blasts>10-4 and sensitivity up to 10-5. This molecular approach enabled the patient's stratification, which was overlooked by classical methods. CONCLUSION The combined IKZF1 and immunoglobulin genes will be used as appropriate molecular tools for diagnosis and MRD assessment of B-lineage leukemias and introduced as a routine tests in Tunisian clinical laboratories. They will be useful to stratify patients into risk groups leading to better treatment strategy.
Collapse
Affiliation(s)
- Sawsen Besbes
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia
| | - Walid-Sabri Hamadou
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia
| | | | | | - Yosra Ben Youssef
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia; CHU F. Hached, service d'hématologie clinique, 4000 Sousse, Tunisia
| | - Béchir Achour
- CHU F. Hached, service d'hématologie clinique, 4000 Sousse, Tunisia
| | - Abderrahim Khelif
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia; CHU F. Hached, service d'hématologie clinique, 4000 Sousse, Tunisia
| | - Thierry Fest
- CHU Pontchaillou, service d'hématologie biologique, 35033 Rennes, France
| | - Zohra Soua
- Université de Sousse, faculté de médecine, laboratoire de biochimie, unité de recherche 14 ES 19, 4000 Sousse, Tunisia.
| |
Collapse
|
28
|
Paula FDF, Elói-Santos SM, Xavier SG, Ganazza MA, Jotta PY, Yunes JA, Viana MB, Assumpção JG. Comparison between qualitative and real-time polymerase chain reaction to evaluate minimal residual disease in children with acute lymphoblastic leukemia. Rev Bras Hematol Hemoter 2015; 37:373-80. [PMID: 26670399 PMCID: PMC4678902 DOI: 10.1016/j.bjhh.2015.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/18/2015] [Accepted: 08/14/2015] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Minimal residual disease is an important independent prognostic factor that can identify poor responders among patients with acute lymphoblastic leukemia. OBJECTIVE The aim of this study was to analyze minimal residual disease using immunoglobulin (Ig) and T-cell receptor (TCR) gene rearrangements by conventional polymerase chain reaction followed by homo-heteroduplex analysis and to compare this with real-time polymerase chain reaction at the end of the induction period in children with acute lymphoblastic leukemia. METHODS Seventy-four patients diagnosed with acute lymphoblastic leukemia were enrolled. Minimal residual disease was evaluated by qualitative polymerase chain reaction in 57 and by both tests in 44. The Kaplan-Meier and multivariate Cox methods and the log-rank test were used for statistical analysis. RESULTS Nine patients (15.8%) were positive for minimal residual disease by qualitative polymerase chain reaction and 11 (25%) by real-time polymerase chain reaction considering a cut-off point of 1×10(-3) for precursor B-cell acute lymphoblastic leukemia and 1×10(-2) for T-cell acute lymphoblastic leukemia. Using the qualitative method, the 3.5-year leukemia-free survival was significantly higher in children negative for minimal residual disease compared to those with positive results (84.1%±5.6% versus 41.7%±17.3%, respectively; p-value=0.004). There was no significant association between leukemia-free survival and minimal residual disease by real-time polymerase chain reaction. Minimal residual disease by qualitative polymerase chain reaction was the only variable significantly correlated to leukemia-free survival. CONCLUSION Given the difficulties in the implementation of minimal residual disease monitoring by real-time polymerase chain reaction in most treatment centers in Brazil, the qualitative polymerase chain reaction strategy may be a cost-effective alternative.
Collapse
Affiliation(s)
| | | | | | | | | | - José Andrés Yunes
- Centro Infantil Boldrini, Campinas, SP, Brazil; Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | |
Collapse
|
29
|
Minimal residual disease diagnostics in acute lymphoblastic leukemia: need for sensitive, fast, and standardized technologies. Blood 2015; 125:3996-4009. [PMID: 25999452 DOI: 10.1182/blood-2015-03-580027] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/10/2015] [Indexed: 12/26/2022] Open
Abstract
Monitoring of minimal residual disease (MRD) has become routine clinical practice in frontline treatment of virtually all childhood acute lymphoblastic leukemia (ALL) and in many adult ALL patients. MRD diagnostics has proven to be the strongest prognostic factor, allowing for risk group assignment into different treatment arms, ranging from significant treatment reduction to mild or strong intensification. Also in relapsed ALL patients and patients undergoing stem cell transplantation, MRD diagnostics is guiding treatment decisions. This is also why the efficacy of innovative drugs, such as antibodies and small molecules, are currently being evaluated with MRD diagnostics within clinical trials. In fact, MRD measurements might well be used as a surrogate end point, thereby significantly shortening the follow-up. The MRD techniques need to be sensitive (≤10(-4)), broadly applicable, accurate, reliable, fast, and affordable. Thus far, flow cytometry and polymerase chain reaction (PCR) analysis of rearranged immunoglobulin and T-cell receptor genes (allele-specific oligonucleotide [ASO]-PCR) are claimed to meet these criteria, but classical flow cytometry does not reach a solid 10(-4), whereas classical ASO-PCR is time-consuming and labor intensive. Therefore, 2 high-throughput technologies are being explored, ie, high-throughput sequencing and next-generation (multidimensional) flow cytometry, both evaluating millions of sequences or cells, respectively. Each of them has specific advantages and disadvantages.
Collapse
|
30
|
Ben Lassoued A, Nivaggioni V, Gabert J. Minimal residual disease testing in hematologic malignancies and solid cancer. Expert Rev Mol Diagn 2015; 14:699-712. [PMID: 24938122 DOI: 10.1586/14737159.2014.927311] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Minimal residual disease (MRD) assays are of a great value to assess treatment efficacy and may provide prognostic information. This is particularly relevant in the era of targeted therapy where the introduction of MRD monitoring has fundamentally transformed the way in which cancer patients are managed. While MRD guidelines are well-established for chronic myeloid leukemia, acute promyelocytic leukemia and acute lymphoblastic leukemia, areas for continuing development are available. High level of standardization and regular external quality control rounds and recommendations for data interpretation remain essential to improve MRD monitoring. In this review, we describe the different applications of MRD assays in most frequent hematologic malignancies and solid cancer and provide an overview of the strengths and potential weaknesses of each method.
Collapse
Affiliation(s)
- Amin Ben Lassoued
- Laboratoire de Biochimie et de Biologie Moléculaire, Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital NORD, Marseille, France
| | | | | |
Collapse
|
31
|
Kalinova M, Fronkova E, Klener P, Forsterova K, Lokvenc M, Mejstrikova E, Belada D, Mocikova H, Trneny M, Kodet R, Trka J. The use of formalin-fixed, paraffin-embedded lymph node samples for the detection of minimal residual disease in mantle cell lymphoma. Br J Haematol 2014; 169:145-8. [DOI: 10.1111/bjh.13182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Marketa Kalinova
- Department of Pathology and Molecular Medicine; 2nd Faculty of Medicine; University Hospital Motol; Charles University in Prague; Prague Czech Republic
| | - Eva Fronkova
- CLIP; Department of Paediatric Haematology/Oncology; 2nd Faculty of Medicine; University Hospital Motol Charles University Prague; Prague Czech Republic
| | - Pavel Klener
- First Department of Medicine - Department of Haematology; General University Hospital; First Faculty of Medicine and Charles University in Prague; Prague Czech Republic
| | - Kristina Forsterova
- First Department of Medicine - Department of Haematology; General University Hospital; First Faculty of Medicine and Charles University in Prague; Prague Czech Republic
| | - Milan Lokvenc
- Department of Pathology and Molecular Medicine; 2nd Faculty of Medicine; University Hospital Motol; Charles University in Prague; Prague Czech Republic
| | - Ester Mejstrikova
- CLIP; Department of Paediatric Haematology/Oncology; 2nd Faculty of Medicine; University Hospital Motol Charles University Prague; Prague Czech Republic
| | - David Belada
- Fourth Department of Medicine - Department of Haematology; Hradec Kralove University Hospital; Hradec Králové Czech Republic
| | - Heidi Mocikova
- Department of Haematology; Kralovske Vinohrady University Hospital; Prague Czech Republic
| | - Marek Trneny
- First Department of Medicine - Department of Haematology; General University Hospital; First Faculty of Medicine and Charles University in Prague; Prague Czech Republic
| | - Roman Kodet
- Department of Pathology and Molecular Medicine; 2nd Faculty of Medicine; University Hospital Motol; Charles University in Prague; Prague Czech Republic
| | - Jan Trka
- CLIP; Department of Paediatric Haematology/Oncology; 2nd Faculty of Medicine; University Hospital Motol Charles University Prague; Prague Czech Republic
| |
Collapse
|
32
|
Silvennoinen R, Lundan T, Kairisto V, Pelliniemi TT, Putkonen M, Anttila P, Huotari V, Mäntymaa P, Siitonen S, Uotila L, Penttilä TL, Juvonen V, Selander T, Remes K. Comparative analysis of minimal residual disease detection by multiparameter flow cytometry and enhanced ASO RQ-PCR in multiple myeloma. Blood Cancer J 2014; 4:e250. [PMID: 25303369 PMCID: PMC4220647 DOI: 10.1038/bcj.2014.69] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 08/28/2014] [Indexed: 01/02/2023] Open
Abstract
Multiparameter flow cytometry (MFC) and allele-specific oligonucleotide real-time quantitative PCR (ASO RQ-PCR) are the two most sensitive methods to detect minimal residual disease (MRD) in multiple myeloma (MM). We compared these methods in 129 paired post-therapy samples from 22 unselected, consecutive MM patients in complete/near complete remission. Appropriate immunophenotypic and ASO RQ-PCR-MRD targets could be detected and MRD analyses constructed for all patients. The high PCR coverage could be achieved by gradual widening of the primer sets used for clonality detection. In addition, for 13 (55%) of the patients, reverse orientation of the ASO primer and individual design of the TaqMan probe improved the sensitivity and specificity of ASO RQ-PCR analysis. A significant nonlinear correlation prevailed between MFC-MRD and PCR-MRD when both were positive. Discordance between the methods was found in 32 (35%) paired samples, which were negative by MFC-MRD, but positive by ASO RQ-PCR. The findings suggest that with the described technique, ASO RQ-PCR can be constructed for all patients with MM. ASO RQ-PCR is slightly more sensitive in MRD detection than 6-10-color flow cytometry. Owing to technical demands ASO RQ-PCR could be reserved for patients in immunophenotypic remission, especially in efficacy comparisons between different drugs and treatment modalities.
Collapse
Affiliation(s)
- R Silvennoinen
- 1] Department of Medicine, Tampere University Hospital, Tampere, Finland [2] Department of Medicine and Hematology, Kuopio University Hospital, Kuopio, Finland
| | - T Lundan
- TYKSLAB, Laboratory of Molecular Genetics, Turku University Hospital, Turku, Finland
| | - V Kairisto
- TYKSLAB, Laboratory of Molecular Genetics, Turku University Hospital, Turku, Finland
| | - T-T Pelliniemi
- 1] Fimlab Medical Laboratories Ltd, Tampere University Hospital, Tampere, Finland [2] Department of Clinical Chemistry, Turku University, Turku, Finland
| | - M Putkonen
- Department of Internal Medicine, Turku University Hospital, Turku University, Turku, Finland
| | - P Anttila
- Helsinki University Hospital, Helsinki, Finland
| | - V Huotari
- 1] NordLab Oulu, Oulu University Hospital, Oulu, Finland [2] Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - P Mäntymaa
- Laboratory of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - S Siitonen
- HUSLAB, Laboratory of Clinical Chemistry, Helsinki University Hospital, Helsinki, Finland
| | - L Uotila
- HUSLAB, Laboratory of Clinical Chemistry, Helsinki University Hospital, Helsinki, Finland
| | - T-L Penttilä
- TYKSLAB, Laboratory of Clinical Haematology, Turku University Hospital, Turku, Finland
| | - V Juvonen
- TYKSLAB, Laboratory of Molecular Genetics, Turku University Hospital, Turku, Finland
| | - T Selander
- Science Services Center, Kuopio University Hospital, Kuopio, Finland
| | - K Remes
- Department of Internal Medicine, Turku University Hospital, Turku University, Turku, Finland
| |
Collapse
|
33
|
Clinical significance of minimal residual disease in young adults with standard-risk/Ph-negative precursor B-acute lymphoblastic leukemia: results of prospective study. Med Oncol 2014; 31:938. [PMID: 24692146 DOI: 10.1007/s12032-014-0938-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/21/2014] [Indexed: 01/08/2023]
Abstract
Clinical risk classification is inaccurate in predicting outcome in adult patients with acute lymphoblastic leukemia (ALL), sometimes resulting in patients receiving inappropriate chemotherapy or stem cell transplantation. To identify complementary markers suitable for further treatment stratification in patients with standard-risk (SR)/philadelphia-negative (Ph-negative) precursor B-ALL, we evaluated the predictive value of minimal residual disease (MRD) after induction and consolidation chemotherapy in strictly defined SR/Ph-negative precursor B-ALL patients who were treated with a standard protocol using quantitative real-time polymerase chain reaction with the rearranged immunoglobulin heavy chain gene as a molecular marker. The cytologic complete response (CR) rate was 92.3 % after induction. At this time point, the molecular CR rate was 73.9%. Patients with molecular CR (MolCR) after induction had a significantly higher probability of disease-free survival (DFS; 78.8 vs 30.8%; P = .001) and of overall survival (OS; 82.4 vs 41.7%; P < .0001) compared to patients with molecular failure (MolFail). MRD at end consolidation had the same significance. Quantitative MRD assessment identified patients with MolFail after induction and/or consolidation as a high-risk group, with 3-year DFS and OS rates of 28.6 and 35.7%, respectively. Patients with MolCR after induction and consolidation were classified as low-risk and had 3-year DFS rate of 89.7% and OS rate of 93.3%. Thus, MRD quantification during treatment identified prognostic subgroups within the otherwise homogeneous SR/Ph-negative precursor B-ALL population who may benefit from individualized treatment.
Collapse
|
34
|
Sadri-Ardekani H, Homburg CH, van Capel TMM, van den Berg H, van der Veen F, van der Schoot CE, van Pelt AMM, Repping S. Eliminating acute lymphoblastic leukemia cells from human testicular cell cultures: a pilot study. Fertil Steril 2014; 101:1072-1078.e1. [PMID: 24581582 DOI: 10.1016/j.fertnstert.2014.01.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 01/10/2014] [Accepted: 01/10/2014] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To study whether acute lymphoblastic leukemia (ALL) cells survive in a human testicular cell culture system. DESIGN Experimental laboratory study. SETTING Reproductive biology laboratory, academic medical center. PATIENT(S) Acute lymphoblastic leukemia cells from three patients and testicular cells from three other patients. INTERVENTION(S) Acute lymphoblastic leukemia cells were cultured alone or in combination with testicular cells, at various concentrations, in a system that has recently been developed to propagate human spermatogonial stem cells. MAIN OUTCOME MEASURE(S) Viability of ALL and testicular cells during culture was evaluated by flow cytometry using markers for live/dead cells. Furthermore, the presence of ALL cells among testicular cells was determined by highly sensitive (1:10,000 to 1:100,000 cells) patient-specific antigen-receptor minimal residual disease polymerase chain reaction. The presence of spermatogonia at the end of culture was determined by reverse transcription-polymerase chain reaction for ZBTB16, UCHL1, and GPR125. RESULT(S) The ALL cells cultured separately did not survive beyond 14 days of culture. When cultured together with testicular cells, even at extremely high initial concentrations (40% ALL cells), ALL cells were undetectable beyond 26 days of culture. Reverse transcription-polymerase chain reaction confirmed the presence of spermatogonia at the end of the culture period. CONCLUSION(S) Our pilot study shows that the described testicular cell culture system not only allows for efficient propagation of spermatogonial stem cells but also eliminates contaminating ALL cells.
Collapse
Affiliation(s)
- Hooman Sadri-Ardekani
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Reproductive Biotechnology Research Center, Avicenna Research Institute, The Academic Center for Education, Culture and Research (ACECR), Tehran, Iran.
| | - Christa H Homburg
- Experimental Immunohematology, Sanquin Research at the Central Laboratory of the Netherlands Red Cross Blood Transfusion Service (CLB), Amsterdam, the Netherlands
| | - Toni M M van Capel
- Departments of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Henk van den Berg
- Department of Pediatric Oncology, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Fulco van der Veen
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - C Ellen van der Schoot
- Experimental Immunohematology, Sanquin Research at the Central Laboratory of the Netherlands Red Cross Blood Transfusion Service (CLB), Amsterdam, the Netherlands
| | - Ans M M van Pelt
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| | - Sjoerd Repping
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
35
|
van der Velden VHJ, Noordijk R, Brussee M, Hoogeveen PG, Homburg C, de Haas V, van der Schoot CE, van Dongen JJM. Minimal residual disease diagnostics in acute lymphoblastic leukaemia: impact of primer characteristics and size of junctional regions. Br J Haematol 2013; 164:451-3. [DOI: 10.1111/bjh.12621] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | - Rianne Noordijk
- Department of Immunology; Erasmus MC; University Medical Centre Rotterdam; Rotterdam The Netherlands
| | - Mark Brussee
- Department of Experimental immunohematology; Sanquin; Amsterdam The Netherlands
| | - Patricia G. Hoogeveen
- Department of Immunology; Erasmus MC; University Medical Centre Rotterdam; Rotterdam The Netherlands
| | - Christa Homburg
- Department of Experimental immunohematology; Sanquin; Amsterdam The Netherlands
| | | | | | - Jacques J. M. van Dongen
- Department of Immunology; Erasmus MC; University Medical Centre Rotterdam; Rotterdam The Netherlands
| |
Collapse
|
36
|
Puig N, Sarasquete ME, Balanzategui A, Martínez J, Paiva B, García H, Fumero S, Jiménez C, Alcoceba M, Chillón MC, Sebastián E, Marín L, Montalbán MA, Mateos MV, Oriol A, Palomera L, de la Rubia J, Vidriales MB, Bladé J, Lahuerta JJ, González M, Miguel JFS, García-Sanz R. Critical evaluation of ASO RQ-PCR for minimal residual disease evaluation in multiple myeloma. A comparative analysis with flow cytometry. Leukemia 2013; 28:391-7. [DOI: 10.1038/leu.2013.217] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 06/14/2013] [Accepted: 07/10/2013] [Indexed: 11/09/2022]
|
37
|
Clinical practice guidelines for diagnosis, treatment, and follow-up of patients with mantle cell lymphoma. Recommendations from the GEL/TAMO Spanish Cooperative Group. Ann Hematol 2013; 92:1151-79. [PMID: 23716187 DOI: 10.1007/s00277-013-1783-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 05/02/2013] [Indexed: 12/22/2022]
Abstract
Mantle cell lymphoma (MCL) is considered a distinct type of B-cell lymphoma genetically characterized by the t(11;14) translocation and cyclin D1 overexpression. There is also a small subset of tumors negative for cyclin D1 expression that are morphologically and immunophenotypically indistinguishable from conventional MCL. Although in the last decades, the median overall survival of patients with MCL has improved significantly, it is still considered as one of the poorest prognoses diseases among B-cell lymphomas. Election of treatment for patients with MCL is complex due to the scarcity of solid evidence. Current available data shows that conventional chemotherapy does not yield satisfactory results as in other types of B-cell lymphomas. However, the role of other approaches such as autologous or allogenic stem cell transplantation, immunotherapy, the administration of consolidation or maintenance schedules, or the use of targeted therapies still lack clear indications. In view of this situation, the Spanish Group of Lymphomas/Autologous Bone Marrow Transplantation has conducted a series of reviews on different aspects of MCL, namely its diagnosis, prognosis, first-line and salvage treatment (both in young and elderly patients), new targeted therapies, and detection of minimal residual disease. On the basis of the available evidence, a series of recommendations have been issued with the intention of providing guidance to clinicians on the diagnosis, treatment, and monitoring of patients with MCL.
Collapse
|
38
|
Pagni F, Zannella S, Valenzise V, Cazzaniga G, Isimbaldi G, Castelli A. Occult monoclonal B-cell disorder of hyoid bone. Hematol Oncol 2013; 32:107-9. [PMID: 23696376 DOI: 10.1002/hon.2077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/08/2013] [Accepted: 04/10/2013] [Indexed: 01/14/2023]
Affiliation(s)
- Fabio Pagni
- Department of Pathology, San Gerardo Hospital, University of Milan Bicocca, Monza, Italy.
| | | | | | | | | | | |
Collapse
|
39
|
Fagioli F, Quarello P, Zecca M, Lanino E, Rognoni C, Balduzzi A, Messina C, Favre C, Foà R, Ripaldi M, Rutella S, Basso G, Prete A, Locatelli F. Hematopoietic stem cell transplantation for children with high-risk acute lymphoblastic leukemia in first complete remission: a report from the AIEOP registry. Haematologica 2013; 98:1273-81. [PMID: 23445874 DOI: 10.3324/haematol.2012.079707] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Children with high-risk acute lymphoblastic leukemia in first complete remission can benefit from allogeneic hematopoietic stem cell transplantation. We analyzed the outcome of 211 children with high-risk acute lymphoblastic leukemia in first complete remission who were given an allogeneic transplant between 1990 and 2008; the outcome of patients who, despite having an indication for transplantation and a suitable donor, did not receive the allograft for different reasons in the same time period was not analyzed. Sixty-nine patients (33%) were transplanted between 1990 and 1999, 58 (27%) between 2000 and 2005, and 84 (40%) between 2005 and 2008. A matched family donor was employed in 138 patients (65%) and an unrelated donor in 73 (35%). The 10-year probabilities of overall and disease-free survival were 63.4% and 61%, respectively. The 10-year cumulative incidences of transplantation-related mortality and relapse were 15% and 24%, respectively. After 1999, no differences in either disease-free survival or transplant-related mortality were observed in patients transplanted from unrelated or matched family donors. In multivariate analysis, grade IV acute graft-versus-host disease was an independent factor associated with worse disease-free survival. By contrast, grade I acute graft-versus-host disease and age at diagnosis between 1 and 9 years were favorable prognostic variables. Our study, not intended to evaluate whether transplantation is superior to chemotherapy for children with acute lymphoblastic leukemia in first complete remission and high-risk features, shows that the allograft cured more than 60% of these patients; in the most recent period, the outcome of recipients of grafts from matched family and unrelated donors was comparable.
Collapse
Affiliation(s)
- Franca Fagioli
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Torino.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Clinical drug resistance linked to interconvertible phenotypic and functional states of tumor-propagating cells in multiple myeloma. Blood 2012; 121:318-28. [PMID: 23169779 DOI: 10.1182/blood-2012-06-436220] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The phenotype and function of cells enriched in tumor-propagating activity and their relationship to the phenotypic architecture in multiple myeloma (MM) are controversial. Here, in a cohort of 30 patients, we show that MM composes 4 hierarchically organized, clonally related subpopulations, which, although phenotypically distinct, share the same oncogenic chromosomal abnormalities as well as immunoglobulin heavy chain complementarity region 3 area sequence. Assessed in xenograft assays, myeloma-propagating activity is the exclusive property of a population characterized by its ability for bidirectional transition between the dominant CD19(-)CD138(+) plasma cell (PC) and a low frequency CD19(-)CD138(-) subpopulation (termed Pre-PC); in addition, Pre-PCs are more quiescent and unlike PCs, are primarily localized at extramedullary sites. As shown by gene expression profiling, compared with PCs, Pre-PCs are enriched in epigenetic regulators, suggesting that epigenetic plasticity underpins the phenotypic diversification of myeloma-propagating cells. Prospective assessment in paired, pretreatment, and posttreatment bone marrow samples shows that Pre-PCs are up to 300-fold more drug-resistant than PCs. Thus, clinical drug resistance in MM is linked to reversible, bidirectional phenotypic transition of myeloma-propagating cells. These novel biologic insights have important clinical implications in relation to assessment of minimal residual disease and development of alternative therapeutic strategies in MM.
Collapse
|
41
|
Gao C, Zhao XX, Li WJ, Cui L, Zhao W, Liu SG, Yue ZX, Jiao Y, Wu MY, Li ZG. Clinical features, early treatment responses, and outcomes of pediatric acute lymphoblastic leukemia in China with or without specific fusion transcripts: a single institutional study of 1,004 patients. Am J Hematol 2012; 87:1022-7. [PMID: 22911440 DOI: 10.1002/ajh.23307] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Revised: 06/21/2012] [Accepted: 06/25/2012] [Indexed: 01/22/2023]
Abstract
Acute lymphoblastic leukemia (ALL) with distinct fusion transcripts has unique clinical features. In this study, the incidence, clinical characteristics, early treatment response, and outcomes of 1,004 Chinese pediatric ALLs were analyzed. Patients with TEL-AML1 and E2A-PBX1 fusion genes or other B cell precursor ALLs (BCP-ALL) had favorable clinical features, were sensitive to prednisone, had low minimal residual disease (MRD), and an excellent prognosis, with a 5-year event-free survival (EFS) of 84-92%. T-ALL was associated with a high WBC, increased age, more central nervous system involvement, a poor prednisone response, and high MRD, with a 5-year EFS of 68.4 ± 5.2%. Patients with BCR-ABL and MLL rearrangements usually had adverse clinical presentations and treatment responses, and a dismal prognosis, with 5-year EFS of 27.3 and 57.4%, respectively. We also showed that BCR-ABL and MLL rearrangements, the prednisone response, and MRD were independent prognostic factors. Interestingly, the BCH-2003 protocol resulted in a better outcome for E2A-PBX1(+) patients than the CCLG-2008 protocol. Intermediate and late relapses were more common in TEL-AML1(+) patients and other BCP-ALLs compared with other subgroups (P = 0.018). Therefore, this study suggests that a fusion gene-specific chemotherapy regimen and/or targeted therapy should be developed to improve further the cure rate of pediatric ALL.
Collapse
Affiliation(s)
- Chao Gao
- Hematology Center, Beijing Children's Hospital, Capital Medical University, 56 Nanlishi Road, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Garand R, Beldjord K, Cavé H, Fossat C, Arnoux I, Asnafi V, Bertrand Y, Boulland ML, Brouzes C, Clappier E, Delabesse E, Fest T, Garnache-Ottou F, Huguet F, Jacob MC, Kuhlein E, Marty-Grès S, Plesa A, Robillard N, Roussel M, Tkaczuk J, Dombret H, Macintyre E, Ifrah N, Béné MC, Baruchel A. Flow cytometry and IG/TCR quantitative PCR for minimal residual disease quantitation in acute lymphoblastic leukemia: a French multicenter prospective study on behalf of the FRALLE, EORTC and GRAALL. Leukemia 2012; 27:370-6. [PMID: 23070018 DOI: 10.1038/leu.2012.234] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Minimal residual disease (MRD) quantification is widely used for therapeutic stratification in pediatric acute lymphoblastic leukemia (ALL). A robust, reproducible, sensitivity of at least 0.01% has been achieved for IG/TCR clonal rearrangements using allele-specific quantitative PCR (IG/TCR-QPCR) within the EuroMRD consortium. Whether multiparameter flow cytometry (MFC) can reach such inter-center performance in ALL MRD monitoring remains unclear. In a multicenter study, MRD was measured prospectively on 598 follow-up bone marrow samples from 102 high-risk children and 136 adult ALL patients, using IG/TCR-QPCR and 4/5 color MFC. At diagnosis, all 238 patients (100%) had at least one suitable MRD marker with 0.01% sensitivity, including 205/238 samples (86%) by using IG/TCR-QPCR and 223/238 samples (94%) by using MFC. QPCR and MFC were evaluable in 495/598 (83%) samples. Qualitative results (<0.01% or ≥0.01%) concurred in 96% of samples and overall positivity (including <0.01% and nonquantifiable positivity) was concurrent in 84%. MRD values ≥0.01% correlated highly (r(2)=0.87) and 69% clustered within half-a-log(10). QPCR and MFC can therefore be comparable if properly standardized, and are highly complementary. MFC strategies will benefit from a concerted approach, as does molecular MRD monitoring, and will contribute significantly to the achievement of 100% MRD informativity in adult and pediatric ALL.
Collapse
|
43
|
Tjønnfjord G, Ly B, Johannesen T, Tierens A, Beiske K, Heim S, Jønsson V. Kronisk lymfatisk leukemi i Norge - insidens og prognose ved diagnosetidspunktet. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2012; 132:2056-9. [DOI: 10.4045/tidsskr.11.1349] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
44
|
Stutterheim J, Ichou FA, den Ouden E, Versteeg R, Caron HN, Tytgat GAM, van der Schoot CE. Methylated RASSF1a is the first specific DNA marker for minimal residual disease testing in neuroblastoma. Clin Cancer Res 2011; 18:808-14. [PMID: 22142825 DOI: 10.1158/1078-0432.ccr-11-0849] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE PCR-based detection of minimal residual disease (MRD) in neuroblastoma (NB) is presently based on NB-specific transcripts. However, the expression of these targets varies between patients and upon treatment, and only PHOX2B is truly specific. RASSF1a is methylated (RASSF1a(M)) in NB, and we investigated whether it can serve as a specific and stable DNA MRD marker. PATIENTS AND METHODS The RASSF1a(M)-specific quantitative real-time PCR was tested on control bone marrow (BM; n = 50), on 71 NB tumors, and on 159 clinical BM samples at diagnosis and at follow-up of 77 patients. Results were compared with a panel of RNA markers and correlated with prognosis. RESULTS RASSF1a(M) was present in all stage 4 and 4s tumors (n = 50) and in 86% stages 1 to 3 tumors (n = 21). The level of methylation in stage 4 NB was correlated with overall survival (P = 0.02). RASSF1a(M)-PCR was highly specific (only 1 amplification in 50 control samples tested in triplicate) and had a similar sensitivity as the RNA-based PCRs, as shown on clinical samples. Moreover, RASSF1a(M) enabled accurate quantification without need for the original tumor. CONCLUSIONS RASSF1a(M) is a novel, highly specific DNA marker for MRD detection in NB, equal to PHOX2B in specificity and sensitivity, and better suitable for MRD quantification. We propose to include RASSF1a(M) in further prospective MRD studies in NB alongside RNA MRD markers. In addition, this assay might also be applicable for detection of circulating tumor cells in patients with other cancers withRASSF1a(M) such as breast or lung cancer.
Collapse
Affiliation(s)
- Janine Stutterheim
- Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
45
|
Volejnikova J, Mejstrikova E, Valova T, Reznickova L, Hodonska L, Mihal V, Sterba J, Jabali Y, Prochazkova D, Blazek B, Hak J, Cerna Z, Hrusak O, Stary J, Trka J, Fronkova E. Minimal residual disease in peripheral blood at day 15 identifies a subgroup of childhood B-cell precursor acute lymphoblastic leukemia with superior prognosis. Haematologica 2011; 96:1815-21. [PMID: 21880630 DOI: 10.3324/haematol.2011.042937] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Most minimal residual disease-directed treatment interventions in current treatment protocols for acute lymphoblastic leukemia are based on bone marrow testing, which is a consequence of previous studies showing the superiority of bone marrow over peripheral blood as an investigational material. Those studies typically did not explore the prognostic impact of peripheral blood involvement and lacked samples from very early time points of induction. DESIGN AND METHODS In this study, we employed real-time quantitative polymerase chain reaction analysis to examine minimal residual disease in 398 pairs of blood and bone marrow follow-up samples taken from 95 children with B-cell precursor acute lymphoblastic leukemia treated with the ALL IC-BFM 2002 protocol. RESULTS We confirmed the previously published poor correlation between minimal residual disease in blood and marrow at early treatment time points, with levels in bone marrow being higher than in blood in most samples (median 7.9-fold, range 0.04-8,293-fold). A greater involvement of peripheral blood at diagnosis was associated with a higher white blood cell count at diagnosis (P=0.003) and with enlargement of the spleen (P=0.0004) and liver (P=0.05). At day 15, a level of minimal residual disease in blood lower than 10(-4) was associated with an excellent 5-year relapse-free survival in 78 investigated patients (100% versus 69 ± 7%; P=0.0003). Subgroups defined by the level of minimal residual disease in blood at day 15 (high-risk: ≥ 10(-2), intermediate-risk: <10(-2) and ≥ 10(-4), standard-risk: <10(-4)) partially correlated with bone marrow-based stratification described previously, but the risk groups did not match completely. No other time point analyses were predictive of outcome in peripheral blood, except for a weak association at day 8. CONCLUSIONS Minimal residual disease in peripheral blood at day 15 identified a large group of patients with an excellent prognosis and added prognostic information to the risk stratification based on minimal residual disease at day 33 and week 12.
Collapse
Affiliation(s)
- Jana Volejnikova
- Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Meleshko AN, Savva NN, Fedasenka UU, Romancova AS, Krasko OV, Eckert C, von Stackelberg A, Aleinikova OV. Prognostic value of MRD-dynamics in childhood acute lymphoblastic leukemia treated according to the MB-2002/2008 protocols. Leuk Res 2011; 35:1312-20. [PMID: 21596436 DOI: 10.1016/j.leukres.2011.04.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2010] [Revised: 03/05/2011] [Accepted: 04/12/2011] [Indexed: 10/18/2022]
Abstract
Detection of minimal residual disease (MRD) during the treatment of acute lymphoblastic leukemia (ALL) by RQ-PCR analysis of clonal Ig/TCR rearrangements is used for risk group stratification in European treatment protocols. In Belarus patients with childhood ALL are treated according to ALL-MB protocols, which do not use MRD-based risk stratification. To evaluate the prognostic significance of MRD for ALL-MB-2002/2008 protocols, MRD was quantified by RQ-PCR in 68 ALL patients at four time points: on day 15, on day 36, before and after maintenance therapy (MT). MRD positivity, as well as quantitative level of MRD were analyzed and compared between patients who stayed in remission and relapsed. Relapse-free survival revealed to be significantly associated with MRD levels at different time points. Unfavorable prognosis was shown for MRD≥10(-3) on day 36 (p<0.001), and any positive MRD before (p<0.001) and after (p=0.001) MT. Multivariate Cox regression analysis proved MRD as independent significant prognosis factor at day 36 (p=0.005) and before MT (p=0.001). We conclude, that MRD quantified by RQ-PCR in children with ALL treated with ALL-MB protocols is feasible and independently associated with outcome. MRD may be a suitable parameter for treatment stratification in MB protocols in future.
Collapse
Affiliation(s)
- Alexander N Meleshko
- Belarusian Research Centre for Pediatric Oncology and Hematology, Pos. Lesnoe, Minsk 223040, Belarus.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Thörn I, Forestier E, Botling J, Thuresson B, Wasslavik C, Björklund E, Li A, Lindström-Eriksson E, Malec M, Grönlund E, Torikka K, Heldrup J, Abrahamsson J, Behrendtz M, Söderhäll S, Jacobsson S, Olofsson T, Porwit A, Lönnerholm G, Rosenquist R, Sundström C. Minimal residual disease assessment in childhood acute lymphoblastic leukaemia: a Swedish multi-centre study comparing real-time polymerase chain reaction and multicolour flow cytometry. Br J Haematol 2011; 152:743-53. [PMID: 21250970 DOI: 10.1111/j.1365-2141.2010.08456.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Minimal residual disease (MRD) assessment is a powerful prognostic factor for determining the risk of relapse in childhood acute lymphoblastic leukaemia (ALL). In this Swedish multi-centre study of childhood ALL diagnosed between 2002 and 2006, the MRD levels were analysed in 726 follow-up samples in 228 children using real-time quantitative polymerase chain reaction (RQ-PCR) of rearranged immunoglobulin/T-cell receptor genes and multicolour flow cytometry (FCM). Using an MRD threshold of 0·1%, which was the sensitivity level reached in all analyses, the concordance between RQ-PCR and FCM MRD values at day 29 was 84%. In B-cell precursor ALL, an MRD level of ≥0·1% at day 29 predicted a higher risk of bone marrow relapse (BMR) with both methods, although FCM was a better discriminator. However, considering the higher median MRD values achieved with RQ-PCR, a higher MRD cut-off (≥0·2%) improved the predictive capacity of RQ-PCR. In T-ALL, RQ-PCR was notably superior to FCM in predicting risk of BMR. That notwithstanding, MRD levels of ≥0·1%, detected by either method at day 29, could not predict isolated extramedullary relapse. In conclusion, the concordance between RQ-PCR and FCM was high and hence both methods are valuable clinical tools for identifying childhood ALL cases with increased risk of BMR.
Collapse
Affiliation(s)
- Ingrid Thörn
- Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Putkonen M, Kairisto V, Juvonen V, Pelliniemi TT, Rauhala A, Itälä-Remes M, Remes K. Depth of response assessed by quantitative ASO-PCR predicts the outcome after stem cell transplantation in multiple myeloma. Eur J Haematol 2010; 85:416-23. [PMID: 20722702 DOI: 10.1111/j.1600-0609.2010.01510.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Achievement of complete response (CR) is a new goal of therapy for multiple myeloma (MM). By sensitive methods, the depth of response can be measured even among the patients in CR. We used a sensitive real-time quantitative polymerase chain reaction by allele-specific primers (qASO-PCR) to assess the level of minimal residual disease (MRD) in bone marrow of 37 patients with myeloma who had achieved CR/near-to-CR after autologous or allogeneic stem cell transplantation (SCT). Allele-specific primers could be successfully designed for 86% of patients. Three to six months after autotransplantation, the PCR target was not detectable in 53% of patients (16/30 patients), and the respective figure after allotransplantation was 71% (5/7 patients); the median sensitivity of PCR assay was <0.002%. The proportion of patients without detectable PCR target was 22% of all autotransplanted patients. A threshold level of 0.01% in the qASO-PCR assay 3-6 months after SCT was found to be a useful cut-off limit to divide the patients into two prognostic groups: MRD low/negative vs. MRD high. Low/negative MRD after SCT was a significant predictive factor for the prolongation of progression free (70 vs. 19 months; P = 0.003) and suggestively also for overall survival. We conclude that not only CR but also its depth is important for the long-term outcome in MM.
Collapse
Affiliation(s)
- Mervi Putkonen
- Department of Medicine, Turku University Central Hospital, Vaasa, Finland.
| | | | | | | | | | | | | |
Collapse
|
49
|
Waanders E, van der Velden VHJ, van der Schoot CE, van Leeuwen FN, van Reijmersdal SV, de Haas V, Veerman AJ, van Kessel AG, Hoogerbrugge PM, Kuiper RP, van Dongen JJM. Integrated use of minimal residual disease classification and IKZF1 alteration status accurately predicts 79% of relapses in pediatric acute lymphoblastic leukemia. Leukemia 2010; 25:254-8. [PMID: 21102428 DOI: 10.1038/leu.2010.275] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Response to therapy as determined by minimal residual disease (MRD) is currently used for stratification in treatment protocols for pediatric acute lymphoblastic leukemia (ALL). However, the large MRD-based medium risk group (MRD-M; 50-60% of the patients) harbors many relapses. We analyzed MRD in 131 uniformly treated precursor-B-ALL patients and evaluated whether combined MRD and IKZF1 (Ikaros zinc finger-1) alteration status can improve risk stratification. We confirmed the strong prognostic significance of MRD classification, which was independent of IKZF1 alterations. Notably, 8 of the 11 relapsed cases in the large MRD-M group (n=81; 62%) harbored an IKZF1 alteration. Integration of both MRD and IKZF1 status resulted in a favorable outcome group (n=104; 5 relapses) and a poor outcome group (n=27; 19 relapses), and showed a stronger prognostic value than each of the established risk factors alone (hazard ratio (95%CI): 24.98 (8.29-75.31)). Importantly, whereas MRD and IKZF1 status alone identified only 46 and 54% of the relapses, respectively, their integrated use allowed prediction of 79% of all the relapses with 93% specificity. Because of the unprecedented sensitivity in upfront relapse prediction, the combined parameters have high potential for future risk stratification, particularly for patients originally classified as non-high risk, such as the large group of MRD-M patients.
Collapse
Affiliation(s)
- E Waanders
- Department of Human Genetics, Radboud University Nijmegen Medical Centre, Radboud University Centre of Oncology and Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Combined analysis of minimal residual disease at two time points and its value for risk stratification in childhood B-lineage acute lymphoblastic leukemia. Leuk Res 2010; 34:1314-9. [DOI: 10.1016/j.leukres.2009.11.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 11/16/2009] [Accepted: 11/30/2009] [Indexed: 11/16/2022]
|