1
|
Deng X, Zhang M, Zhou J, Xiao M. Next-generation sequencing for MRD monitoring in B-lineage malignancies: from bench to bedside. Exp Hematol Oncol 2022; 11:50. [PMID: 36057673 PMCID: PMC9440501 DOI: 10.1186/s40164-022-00300-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/21/2022] [Indexed: 12/02/2022] Open
Abstract
Minimal residual disease (MRD) is considered the strongest relevant predictor of prognosis and an effective decision-making factor during the treatment of hematological malignancies. Remarkable breakthroughs brought about by new strategies, such as epigenetic therapy and chimeric antigen receptor-T (CAR-T) therapy, have led to considerably deeper responses in patients than ever, which presents difficulties with the widely applied gold-standard techniques of MRD monitoring. Urgent demands for novel approaches that are ultrasensitive and provide sufficient information have put a spotlight on high-throughput technologies. Recently, advances in methodology, represented by next-generation sequencing (NGS)-based clonality assays, have proven robust and suggestive in numerous high-quality studies and have been recommended by some international expert groups as disease-monitoring modalities. This review demonstrates the applicability of NGS-based clonality assessment for MRD monitoring of B-cell malignancies by summarizing the oncogenesis of neoplasms and the corresponding status of immunoglobulin (IG) rearrangements. Furthermore, we focused on the performance of NGS-based assays compared with conventional approaches and the interpretation of results, revealing directions for improvement and prospects in clinical practice.
Collapse
Affiliation(s)
- Xinyue Deng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Meilan Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China.
| |
Collapse
|
2
|
Metzner B, Pott C, Müller TH, Casper J, Kimmich C, Petershofen EK, Renzelmann A, Rosien B, Thole R, Voß A, Köhne CH, Wellnitz D. Long-term outcome in patients with follicular lymphoma following high-dose therapy and autologous stem cell transplantation. Eur J Haematol 2021; 107:543-552. [PMID: 34288114 DOI: 10.1111/ejh.13691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To contribute data on long-term outcome and potential curative impact of ASCT in FL, especially following HDT with the BEAM protocol (BCNU, etoposide, cytarabine and melphalan), given very limited data on this topic in the literature. PATIENTS AND METHODS Patients with FL (n = 76) were treated in our institution with HDT and ASCT. In the case of long-term remission (≥8 years), peripheral blood was tested for minimal residual disease by t(14;18)- and IGH-PCR, including the last follow-up. RESULTS 10-year overall survival, progression-free survival, and freedom from progression (FFP) after first-line ASCT (n = 20) were 80%, 60%, and 69%, after second-line ASCT (n = 48, following BEAM) 66%, 38%, and 41%, after third/fourth-line ASCT (n = 8) 33%, 25%, and 25%, respectively. Prognostic factors for FFP were treatment line and FLIPI (Follicular Lymphoma International Prognostic Index). 10-year FFP for second-line ASCT and low-risk FLIPI at relapse was 69%, intermediate-risk 28%, and high-risk 25% (P < .05). 26 patients developed sustained long-term clinical and molecular remissions of up to 27 years. CONCLUSIONS Sustained long-term clinical and molecular complete remissions up to 27 years can be achieved following ASCT (including HDT with BEAM in second treatment line), indicating a potential curative impact of ASCT in FL.
Collapse
Affiliation(s)
- Bernd Metzner
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Christiane Pott
- Department of Medicine II, University of Schleswig-Holstein, Kiel, Germany
| | - Thomas H Müller
- Red Cross Blood Transfusion Service NSTOB, Oldenburg, Germany
| | - Jochen Casper
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Christoph Kimmich
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | | | - Andrea Renzelmann
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Bernd Rosien
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Ruth Thole
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Andreas Voß
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Claus Henning Köhne
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Dominique Wellnitz
- Department of Medicine II, University of Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
3
|
Bachy E, Rufibach K, Parreira J, Launonen A, Nielsen T, Hackshaw A. Phase III Clinical Trials in First-Line Follicular Lymphoma: A Review of Their Design and Interpretation. Adv Ther 2021. [PMID: 34041708 DOI: 10.6084/m9.figshare.14381117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Follicular lymphoma (FL) is one of the most common subtypes of non-Hodgkin lymphoma worldwide. Improved survival outcomes with rituximab-based therapy in clinical trials led to the establishment of rituximab-based immunochemotherapy as standard of care for first-line (1L) treatment of FL. In the GALLIUM trial, obinutuzumab-based immunochemotherapy demonstrated improved progression-free survival (PFS), prolonged time-to-next antilymphoma treatment (TTNT) and comparable overall survival (OS) compared with rituximab-based immunochemotherapy as 1L treatment for FL. Using GALLIUM as an example, this article aims to explain how improved outcomes in 1L treatment of FL have changed the landscape for the design and interpretation of future trials. As approved therapies for 1L FL already achieve good responses, it is becoming more difficult to design trials that demonstrate further treatment benefits with the currently accepted primary endpoints. New endpoints are needed to reflect the long remission times, low relapse rates, and impact of subsequent therapies in FL. PFS is used as a primary efficacy endpoint in registrational clinical trials for indolent malignancies like FL, where improvement in OS is not always observed due to the large number of patients and long study duration required to demonstrate a clear survival benefit. However, there are limitations to using PFS as the primary endpoint. Other potential endpoints, including TTNT, progression of disease within 2 years, response rate, and minimal residual disease status are explored.
Collapse
Affiliation(s)
- Emmanuel Bachy
- Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France.
- Hematology Department, Lyon Sud Hospital, Pierre-Bénite, France.
| | | | | | | | | | | |
Collapse
|
4
|
Bachy E, Rufibach K, Parreira J, Launonen A, Nielsen T, Hackshaw A. Phase III Clinical Trials in First-Line Follicular Lymphoma: A Review of Their Design and Interpretation. Adv Ther 2021; 38:3489-3505. [PMID: 34041708 PMCID: PMC8280048 DOI: 10.1007/s12325-021-01738-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 10/26/2022]
Abstract
Follicular lymphoma (FL) is one of the most common subtypes of non-Hodgkin lymphoma worldwide. Improved survival outcomes with rituximab-based therapy in clinical trials led to the establishment of rituximab-based immunochemotherapy as standard of care for first-line (1L) treatment of FL. In the GALLIUM trial, obinutuzumab-based immunochemotherapy demonstrated improved progression-free survival (PFS), prolonged time-to-next antilymphoma treatment (TTNT) and comparable overall survival (OS) compared with rituximab-based immunochemotherapy as 1L treatment for FL. Using GALLIUM as an example, this article aims to explain how improved outcomes in 1L treatment of FL have changed the landscape for the design and interpretation of future trials. As approved therapies for 1L FL already achieve good responses, it is becoming more difficult to design trials that demonstrate further treatment benefits with the currently accepted primary endpoints. New endpoints are needed to reflect the long remission times, low relapse rates, and impact of subsequent therapies in FL. PFS is used as a primary efficacy endpoint in registrational clinical trials for indolent malignancies like FL, where improvement in OS is not always observed due to the large number of patients and long study duration required to demonstrate a clear survival benefit. However, there are limitations to using PFS as the primary endpoint. Other potential endpoints, including TTNT, progression of disease within 2 years, response rate, and minimal residual disease status are explored.
Collapse
Affiliation(s)
- Emmanuel Bachy
- Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France.
- Hematology Department, Lyon Sud Hospital, Pierre-Bénite, France.
| | | | | | | | | | | |
Collapse
|
5
|
Lenalidomide/rituximab induces high molecular response in untreated follicular lymphoma: LYSA ancillary RELEVANCE study. Blood Adv 2021; 4:3217-3223. [PMID: 32673385 DOI: 10.1182/bloodadvances.2020001955] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Complete molecular response (CMR) after first-line immunochemotherapy reflects treatment efficacy and may predict prognosis in patients with follicular lymphoma (FL). RELEVANCE is the first phase 3 trial comparing the chemotherapy-free regimen lenalidomide/rituximab (R2) vs rituximab/chemotherapy (R-Chemo) in previously untreated FL patients (ClinicalTrials.gov identifier: NCT01650701). The objective of the minimal residual disease (MRD) analysis was to determine the ability of a chemotherapy-free regimen to induce CMR. Of 440 French patients participating in the Lymphoma Study Association (LYSA) RELEVANCE MRD study, all 222 patients with a BIOMED-2-detectable BCL2-JH translocation at diagnosis were analyzed. MRD was quantified by droplet digital polymerase chain reaction with a sensitivity ≤10-4. At week 24 (end of induction treatment), 98% and 78% of patients achieved CMR in peripheral blood (PB) and bone marrow (BM), respectively. Achievement of CMR (in PB and/or BM) had a significant impact on progression-free survival (PFS), with 3-year PFS of 84% and 55% for patients with CMR and detectable MRD, respectively (P = .015). CMR at week 24 was reached more frequently in the R2 arm (105/117; 90%) than in the R-Chemo arm (70/90; 77%) (P = .022). The poor prognostic value in terms of PFS for the persistence of molecular disease was observed irrespective of treatment arm (interaction test, P = .31). In agreement with the clinical results of the RELEVANCE trial, our results show that R2 immunomodulatory treatment in first-line FL can achieve high rates of CMR.
Collapse
|
6
|
Genuardi E, Klous P, Mantoan B, Drandi D, Ferrante M, Cavallo F, Alessandria B, Dogliotti I, Grimaldi D, Ragaini S, Clerico M, Lo Schirico M, Saraci E, Yilmaz M, Zaccaria GM, Cortelazzo S, Vitolo U, Luminari S, Federico M, Boccadoro M, van Min M, Splinter E, Ladetto M, Ferrero S. Targeted locus amplification to detect molecular markers in mantle cell and follicular lymphoma. Hematol Oncol 2021; 39:293-303. [PMID: 33742718 PMCID: PMC8451873 DOI: 10.1002/hon.2864] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/22/2021] [Accepted: 03/06/2021] [Indexed: 11/16/2022]
Abstract
Minimal residual disease (MRD) monitoring by PCR methods is a strong and standardized predictor of clinical outcome in mantle cell lymphoma (MCL) and follicular lymphoma (FL). However, about 20% of MCL and 40% of FL patients lack a reliable molecular marker, being thus not eligible for MRD studies. Recently, targeted locus amplification (TLA), a next‐generation sequencing (NGS) method based on the physical proximity of DNA sequences for target selection, identified novel gene rearrangements in leukemia. The aim of this study was to test TLA in MCL and FL diagnostic samples lacking a classical, PCR‐detectable, t(11; 14) MTC (BCL1/IGH), or t(14; 18) major breakpoint region and minor cluster region (BCL2/IGH) rearrangements. Overall, TLA was performed on 20 MCL bone marrow (BM) or peripheral blood (PB) primary samples and on 20 FL BM, identifying a novel BCL1 or BCL2/IGH breakpoint in 16 MCL and 8 FL patients (80% and 40%, respectively). These new breakpoints (named BCL1‐TLA and BCL2‐TLA) were validated by ASO primers design and compared as MRD markers to classical IGH rearrangements in eight MCL: overall, MRD results by BCL1‐TLA were superimposable (R Pearson = 0.76) to the standardized IGH‐based approach. Moreover, MRD by BCL2‐TLA reached good sensitivity levels also in FL and was predictive of a primary refractory case. In conclusion, this study offers the proof of principle that TLA is a promising and reliable NGS‐based technology for the identification of novel molecular markers, suitable for further MRD analysis in previously not traceable MCL and FL patients.
Collapse
Affiliation(s)
- Elisa Genuardi
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | | | - Barbara Mantoan
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | - Daniela Drandi
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | - Martina Ferrante
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy.,Division of Hematology 1, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
| | - Beatrice Alessandria
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | - Irene Dogliotti
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy.,Division of Hematology 1, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
| | - Daniele Grimaldi
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy.,Division of Hematology 1, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
| | - Simone Ragaini
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy.,Division of Hematology 1, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
| | - Michele Clerico
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy.,Division of Hematology 1, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
| | - Mariella Lo Schirico
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | | | | | - Gian Maria Zaccaria
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | | | - Umberto Vitolo
- Department of Oncology, Division of Hematology, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Stefano Luminari
- Hematology Unit, Azienda USL IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy.,Medical Oncology, CHIMOMO department, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Federico
- Medical Oncology, CHIMOMO department, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Boccadoro
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy.,Division of Hematology 1, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
| | | | | | - Marco Ladetto
- Division of Hematology, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Simone Ferrero
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy.,Division of Hematology 1, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
| |
Collapse
|
7
|
Pott C, Sehn LH, Belada D, Gribben J, Hoster E, Kahl B, Kehden B, Nicolas-Virelizier E, Spielewoy N, Fingerle-Rowson G, Harbron C, Mundt K, Wassner-Fritsch E, Cheson BD. MRD response in relapsed/refractory FL after obinutuzumab plus bendamustine or bendamustine alone in the GADOLIN trial. Leukemia 2019; 34:522-532. [PMID: 31462735 PMCID: PMC7214251 DOI: 10.1038/s41375-019-0559-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/25/2019] [Accepted: 07/15/2019] [Indexed: 11/09/2022]
Abstract
We report assessment of minimal residual disease (MRD) status and its association with outcome in rituximab-refractory follicular lymphoma (FL) in the randomized GADOLIN trial (NCT01059630). Patients received obinutuzumab (G) plus bendamustine (Benda) induction followed by G maintenance, or Benda induction alone. Patients with a clonal marker (t[14;18] translocation and/or immunoglobulin heavy or light chain rearrangement) detected at study screening were assessed for MRD at mid-induction (MI), end of induction (EOI), and every 6–24 months post-EOI/discontinuation by real-time quantitative PCR. At MI, 41/52 (79%) patients receiving G-Benda were MRD-negative vs. 17/36 (47%) patients receiving Benda alone (p = 0.0029). At EOI, 54/63 (86%) patients receiving G-Benda were MRD-negative vs. 30/55 (55%) receiving Benda alone (p = 0.0002). MRD-negative patients at EOI had improved progression-free survival (HR, 0.33, 95% CI, 0.19–0.56, p < 0.0001) and overall survival (HR, 0.39, 95% CI, 0.19–0.78, p = 0.008) vs. MRD-positive patients, and maintained their MRD-negative status for longer if they received G maintenance than if they did not. These results suggest that the addition of G to Benda-based treatment during induction can significantly contribute to the speed and depth of response, and G maintenance in MRD-negative patients potentially delays lymphoma regrowth.
Collapse
Affiliation(s)
| | - Laurie H Sehn
- British Columbia Cancer Agency and the University of British Columbia, Vancouver, BC, Canada
| | - David Belada
- Department of Internal Medicine-Haematology, Charles University, Hospital and Faculty of Medicine, Hradec Králové, Czech Republic
| | | | - Eva Hoster
- Hospital of the Ludwig-Maximilians University, Munich, Germany
| | - Brad Kahl
- Washington University School of Medicine, St Louis, MO, USA
| | - Britta Kehden
- University Hospital Schleswig-Holstein, Kiel, Germany
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Della Starza I, Cavalli M, De Novi LA, Genuardi E, Mantoan B, Drandi D, Barbero D, Ciabatti E, Grassi S, Gazzola A, Mannu C, Agostinelli C, Piccaluga PP, Bomben R, Degan M, Gattei V, Guarini A, Foà R, Galimberti S, Ladetto M, Ferrero S, Del Giudice I. Minimal residual disease (MRD) in non‐Hodgkin lymphomas: Interlaboratory reproducibility on marrow samples with very low levels of disease within the FIL (Fondazione Italiana Linfomi) MRD Network. Hematol Oncol 2019; 37:368-374. [PMID: 31325190 DOI: 10.1002/hon.2652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/08/2019] [Accepted: 07/11/2019] [Indexed: 01/08/2023]
Abstract
In 2009, the four laboratories of the Fondazione Italiana Linfomi (FIL) minimal residual disease (MRD) Network started a collaborative effort to harmonize and standardize their methodologies at the national level, performing quality control (QC) rounds for follicular lymphoma (FL) and mantle cell lymphoma (MCL) MRD assessment. In 16 QC rounds between 2010 and 2017, the four laboratories received 208 bone marrow (BM) samples (126 FL; 82 MCL); 187 were analyzed, according to the EuroMRD Consortium guidelines, by both nested (NEST) polymerase chain reaction (PCR) and real-time quantitative (RQ) PCR for BCL2/IGH MBR or IGHV rearrangements. Here, we aimed at analyzing the samples that challenged the interlaboratory reproducibility and data interpretation. Overall, 156/187 BM samples (83%) were concordantly classified as NEST+/RQ+ or NEST-/RQ- by all the four laboratories. The remaining 31 samples (17%) resulted alternatively positive and negative in the interlaboratory evaluations, independently of the method and the type of rearrangement, and were defined "borderline" (brd) samples: 12 proved NEST brd/RQ brd, 7 NEST-/RQ brd, 10 NEST brd/RQ positive not quantifiable (PNQ), and 2 NEST brd/RQ-. Results did not change even increasing the number of replicates/sample. In 6/31 brd samples, droplet digital PCR (ddPCR) was tested and showed no interlaboratory discordance. Despite the high interlaboratory reproducibility in the MRD analysis obtained and maintained by the QC round strategy, samples with the lowest MRD levels can still represent a challenge: 17% (31/187) of our samples showed discordant results in interlaboratory assessments, with 6.4% (12/187) remained brd even applying the two methods. Thus, although representing a minority, brd samples are still problematic, especially when a clinically oriented interpretation of MRD results is required. Alternative, novel methods such as ddPCR and next-generation sequencing have the potential to overcome the current limitations.
Collapse
Affiliation(s)
- Irene Della Starza
- 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
| | - Elisa Genuardi
- Department of Molecular Biotechnologies and Health Sciences, Division of Hematology University of Torino Turin Italy
| | - Barbara Mantoan
- Department of Molecular Biotechnologies and Health Sciences, Division of Hematology University of Torino Turin Italy
| | - Daniela Drandi
- Department of Molecular Biotechnologies and Health Sciences, Division of Hematology University of Torino Turin Italy
| | - Daniela Barbero
- Department of Molecular Biotechnologies and Health Sciences, Division of Hematology University of Torino Turin Italy
| | - Elena Ciabatti
- Division of Hematology, Department of Oncology Santa Chiara Hospital Pisa Italy
| | - Susanna Grassi
- Department of Medical Biotechnologies University of Siena Siena Italy
| | - Anna Gazzola
- Hematopathology Section, Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola‐Malpighi Hospital University of Bologna Bologna Italy
| | - Claudia Mannu
- Hematopathology Section, Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola‐Malpighi Hospital University of Bologna Bologna Italy
| | - Claudio Agostinelli
- Hematopathology Section, Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola‐Malpighi Hospital University of Bologna Bologna Italy
| | - Pier Paolo Piccaluga
- Hematopathology Section, Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola‐Malpighi Hospital University of Bologna Bologna Italy
| | - Riccardo Bomben
- Clinical and Experimental Onco‐Haematology Unit Centro di Riferimento Oncologico, I.R.C.C.S. Aviano Italy
| | - Massimo Degan
- Clinical and Experimental Onco‐Haematology Unit Centro di Riferimento Oncologico, I.R.C.C.S. Aviano Italy
| | - Valter Gattei
- Clinical and Experimental Onco‐Haematology Unit Centro di Riferimento Oncologico, I.R.C.C.S. Aviano 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
| | - Sara Galimberti
- Division of Hematology, Department of Oncology Santa Chiara Hospital Pisa Italy
| | - Marco Ladetto
- Division of Hematology Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo Alessandria Italy
| | - Simone Ferrero
- Department of Molecular Biotechnologies and Health Sciences, Division of Hematology University of Torino Turin Italy
| | - Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine “Sapienza” University of Rome Rome Italy
| | | |
Collapse
|
9
|
Sorigue M, Sancho JM. Current prognostic and predictive factors in follicular lymphoma. Ann Hematol 2017; 97:209-227. [PMID: 29032510 DOI: 10.1007/s00277-017-3154-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 10/09/2017] [Indexed: 12/27/2022]
Abstract
Follicular lymphoma (FL) is generally considered an indolent disorder. With modern day treatments, long remissions are often achieved both in the front-line and relapsed setting. However, a subset of patients has a more aggressive course and a worse outcome. Their identification is the main purpose of modern day prognostic tools. In this review, we attempt to summarize the evidence concerning prognostic and predictive factors in FL, including (1) pre-treatment factors, from baseline clinical characteristics and imaging tests to histological grade, the microenvironment and genomic abnormalities; (2) post-treatment factors, i.e., depth of response, measured both by imaging tests and minimal residual disease; (3) factors at relapse and duration of response; and (4) prognostic factors in histological transformation. We conclude that, despite the existence of numerous tools, the availability of some of them is still limited; they generally suffer from notable downsides, and most have unproven predictive value, thus having scarce bearing on the choice of regimen at present. However, with the technological and scientific developments of the last few years, the potential for these prognostic factors is promising, particularly in combination, which will probably, in time, help guide therapeutic decisions.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols
- Bone Marrow/drug effects
- Bone Marrow/metabolism
- Bone Marrow/pathology
- Chromosomes, Human, Pair 14/chemistry
- Chromosomes, Human, Pair 18/chemistry
- Clinical Trials as Topic
- Disease-Free Survival
- Humans
- Lymphoma, Follicular/diagnostic imaging
- Lymphoma, Follicular/drug therapy
- Lymphoma, Follicular/genetics
- Lymphoma, Follicular/mortality
- Mutation
- Neoplasm Grading
- Neoplasm, Residual/diagnostic imaging
- Neoplasm, Residual/drug therapy
- Neoplasm, Residual/genetics
- Neoplasm, Residual/mortality
- Positron-Emission Tomography
- Prognosis
- Recurrence
- Risk Factors
- Translocation, Genetic
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Marc Sorigue
- Department of Hematology, ICO-Hospital Germans Trias i Pujol, Institut de Recerca Josep Carreras, Universitat Autònoma de Barcelona, Ctra. Canyet, 08916, Badalona, Spain.
| | - Juan-Manuel Sancho
- Department of Hematology, ICO-Hospital Germans Trias i Pujol, Institut de Recerca Josep Carreras, Universitat Autònoma de Barcelona, Ctra. Canyet, 08916, Badalona, Spain
| |
Collapse
|
10
|
Ladetto M, Buske C, Hutchings M, Dreyling M, Gaidano G, Le Gouill S, Luminari S, Pott C, Zamò A, Zucca E. ESMO consensus conference on malignant lymphoma: general perspectives and recommendations for prognostic tools in mature B-cell lymphomas and chronic lymphocytic leukaemia. Ann Oncol 2016; 27:2149-2160. [PMID: 27701070 DOI: 10.1093/annonc/mdw419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/11/2016] [Accepted: 08/23/2016] [Indexed: 01/02/2023] Open
Abstract
The European Society for Medical Oncology (ESMO) consensus conference on mature B-cell lymphomas and chronic lymphocytic leukaemia (CLL) was held on 20 June 2015 in Lugano, Switzerland, and included a multidisciplinary panel of 25 leading experts. The aim of the conference was to develop recommendations on critical subjects difficult to consider in detail in the ESMO Clinical Practice Guidelines. The following areas were identified: (i) the elderly patient, (ii) prognostic factors suitable for clinical use and (iii) the 'ultra-high-risk' group. Before the conference, the expert panel was divided into three working groups; each group focused on one of these areas in order to address four clinically relevant questions relating to that topic. All relevant scientific literature, as identified by the experts, was reviewed in advance. During the consensus conference, each working group developed recommendations to address each of the four questions assigned to their group. These recommendations were then presented to the entire panel and a consensus was reached. This manuscript presents recommendations dedicated to the second area of interest, i.e. prognostic factors suitable for clinical use. The four topics [i.e. interim positron emission tomography (PET), TP53 mutations, cell of origin (COO) and minimal residual disease (MRD)] were primarily chosen because of the bulk of available data together with the lack of clear guidance regarding their use in clinical practice and within clinical trials. Results, including a summary of evidence supporting each recommendation, are detailed in this manuscript. The panel acknowledged that detection of TP53 inactivation by deletion or mutation in CLL should be implemented in clinical practice (level of evidence I, strength of recommendation A). Due to their potentially high prognostic value, at least in some lymphoma entities, implementation of interim PET, COO and MRD was highly recommended in the context of clinical trials. All expert panel members approved this final article.
Collapse
Affiliation(s)
- M Ladetto
- Hematology Division, Azienda Ospedaliera Santi Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - C Buske
- Comprehensive Cancer Center Ulm and Department of Internal Medicine III, Institute of Experimental Cancer Research, University Hospital, Ulm, Germany
| | - M Hutchings
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - M Dreyling
- Medizinische Klinik III, Klinikum der Universität München/LMU, Munich, Germany
| | - G Gaidano
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - S Le Gouill
- Clinical Hematology, Centre Hospitalo-Universitaire de Nantes, UMR892 Team 10, CIC Nantes, France
| | - S Luminari
- Hematology, Arcispedale S. Maria Nuova, IRCCS Reggio Emilia.,Department of Diagnostic, Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - C Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - A Zamò
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - E Zucca
- Lymphoma Unit, Oncology Institute of Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | | |
Collapse
|
11
|
Seo JY, Hong J, Chun K, Jeong J, Cho H, Kim KH, Park J, Ahn JY, Park PW, Lee JH. Prognostic significance of PCR-based molecular staging in patients with diffuse large B-cell lymphoma treated with R-CHOP immunochemotherapy. Leuk Lymphoma 2016; 58:357-365. [PMID: 27282995 DOI: 10.1080/10428194.2016.1190967] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The prognostic role of detecting clonal immunoglobulin gene rearrangement (IgR) from bone marrow (BM) aspirates was evaluated by BIOMED-2 PCR in 97 patients with diffuse large B-cell lymphoma (DLBCL) treated with rituximab-CHOP immunochemotherapy. Sixteen (16.5%) patients had BM involvement (BMI) defined by BM biopsy (MOR+) and 39 (40.2%) had positive IgR (PCR+). Patients with MOR + BMI showed inferior event-free survival (EFS) compared to those with MOR-/PCR- (p < 0.001) or those with MOR-/PCR + BMI (p = 0.002), while no significant difference in EFS was observed between patients with MOR-/PCR + and those with MOR-/PCR - BMI (p = 0.497). Use of the BIOMED-2 for PCR resulted in significant increase in detection of BMI. However, the increased sensitivity by PCR did not translate into improved prediction of prognosis, emphasizing the essential role of histopathological review of trephine biopsy for the detection of BMI.
Collapse
Affiliation(s)
- Ja Young Seo
- a Department of Laboratory Medicine, Gachon University Gil Medical Center, Incheon Regional Cancer Center , Gachon University College of Medicine , Incheon , Republic of Korea
| | - Junshik Hong
- b Department of Internal Medicine, Gachon University Gil Medical Center , Incheon Regional Cancer Center, Gachon University College of Medicine , Incheon , Republic of Korea
| | - Kayeong Chun
- b Department of Internal Medicine, Gachon University Gil Medical Center , Incheon Regional Cancer Center, Gachon University College of Medicine , Incheon , Republic of Korea
| | - Jihoon Jeong
- a Department of Laboratory Medicine, Gachon University Gil Medical Center, Incheon Regional Cancer Center , Gachon University College of Medicine , Incheon , Republic of Korea
| | - Hyeongrae Cho
- c Department of Preventive Medicine , Gachon University College of Medicine , Incheon , Republic of Korea
| | - Kyung Hee Kim
- a Department of Laboratory Medicine, Gachon University Gil Medical Center, Incheon Regional Cancer Center , Gachon University College of Medicine , Incheon , Republic of Korea
| | - Jinny Park
- b Department of Internal Medicine, Gachon University Gil Medical Center , Incheon Regional Cancer Center, Gachon University College of Medicine , Incheon , Republic of Korea
| | - Jeong Yeal Ahn
- a Department of Laboratory Medicine, Gachon University Gil Medical Center, Incheon Regional Cancer Center , Gachon University College of Medicine , Incheon , Republic of Korea
| | - Pil-Whan Park
- a Department of Laboratory Medicine, Gachon University Gil Medical Center, Incheon Regional Cancer Center , Gachon University College of Medicine , Incheon , Republic of Korea
| | - Jae Hoon Lee
- a Department of Laboratory Medicine, Gachon University Gil Medical Center, Incheon Regional Cancer Center , Gachon University College of Medicine , Incheon , Republic of Korea
| |
Collapse
|
12
|
Aresu L, Aricò A, Ferraresso S, Martini V, Comazzi S, Riondato F, Giantin M, Dacasto M, Guadagnin E, Frayssinet P, Rouquet N, Drigo M, Marconato L. Minimal residual disease detection by flow cytometry and PARR in lymph node, peripheral blood and bone marrow, following treatment of dogs with diffuse large B-cell lymphoma. Vet J 2014; 200:318-24. [DOI: 10.1016/j.tvjl.2014.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 02/10/2014] [Accepted: 03/01/2014] [Indexed: 10/25/2022]
|
13
|
Sørensen CD, Jørgensen JM, Nederby L, Hokland P, Nyvold CG. Common consensus LNA probe for quantitative PCR assays in cancer: vehicles for minimal residual disease detection in t(11;14) and t(14;18) positive malignant lymphomas. J Immunol Methods 2014; 406:131-6. [PMID: 24631717 DOI: 10.1016/j.jim.2014.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 12/12/2013] [Accepted: 03/04/2014] [Indexed: 12/30/2022]
Abstract
The use of locked nucleic acid (LNA) probes and primers potentially improves sensitivity and specificity of quantitative PCR (qPCR) assays. One area of application is that of minimal residual cancer where PCR techniques have proved to be highly relevant tools in patient follow-up. We present here sensitive and specific consensus qPCR assays for quantification of the malignant lymphoma translocations, t(11;14) and t(14;18), by taking advantage of the thermodynamic properties of LNA. The assays were applied to genomic DNA from patients diagnosed with mantle cell lymphoma (MCL) and follicular lymphoma (FL), respectively. Two consensus forward primers targeting the BCL1 and BCL2 genes were designed together with a common consensus reverse primer and hydrolysis probe, the latter consisting exclusively of LNA, both targeting the J segments of the immunoglobulin heavy chain (IgH) gene. The quantitative range of both assays was 1×10(0) to 5×10(-5), and the sensitivity was 10(-5), without the need for patient-specific primers. Peripheral blood (PB) and bone marrow (BM) samples from 36 patients diagnosed with MCL and nine patients diagnosed with FL were analysed using this novel qPCR approach. The level of minimal residual disease (MRD) using t(11;14) and t(14;18) as genetic targets reflected the clinical status of the patients: low levels of MRD at clinical remission, and increasing levels at disease progression. The present assays could prove as useful tools in lymphoma therapy.
Collapse
Affiliation(s)
- Camilla Darum Sørensen
- Department of Haematology, Aarhus University Hospital, Tage-Hansens Gade 2, 8000 Aarhus C, Denmark
| | - Judit Meszaros Jørgensen
- Department of Haematology, Aarhus University Hospital, Tage-Hansens Gade 2, 8000 Aarhus C, Denmark
| | - Line Nederby
- Department of Haematology, Aarhus University Hospital, Tage-Hansens Gade 2, 8000 Aarhus C, Denmark
| | - Peter Hokland
- Department of Haematology, Aarhus University Hospital, Tage-Hansens Gade 2, 8000 Aarhus C, Denmark
| | | |
Collapse
|