1
|
Engelmann R, Böttcher S. Flow Cytometric MRD Detection in Selected Mature B-Cell Malignancies. Methods Mol Biol 2025; 2865:145-188. [PMID: 39424724 DOI: 10.1007/978-1-0716-4188-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
The quantification of submicroscopic minimal residual disease (MRD) after therapy proved to have independent prognostic significance in many mature B-cell malignancies. With the advent of routine benchtop cytometers capable of simultaneously analyzing ≥8 colors and with improved standardization, flow cytometry has become the method of choice for MRD assessments in some lymphoma entities. Herein we describe general aspects of flow cytometric standardization. Chronic lymphocytic leukemia (CLL) and multiple myeloma (MM) are used as examples to explain the technical standardization of flow cytometry for MRD detection according to EuroFlow strategies. MRD data acquisition and detailed analysis in MM and CLL is a particular focus of this chapter.
Collapse
Affiliation(s)
- Robby Engelmann
- Rostock University Medical Center, Division of Internal Medicine, Medical Clinic III - Hematology, Oncology and Palliative Medicine, Special Hematology Laboratory, Rostock, Germany
| | - Sebastian Böttcher
- Rostock University Medical Center, Division of Internal Medicine, Medical Clinic III - Hematology, Oncology and Palliative Medicine, Special Hematology Laboratory, Rostock, Germany.
| |
Collapse
|
2
|
Pott C, Brüggemann M, Ritgen M, van der Velden VHJ, van Dongen JJM. MRD Detection in B-Cell Non-Hodgkin Lymphomas Using Ig Gene Rearrangements and Chromosomal Translocations as Targets for Real-Time Quantitative PCR and ddPCR. Methods Mol Biol 2025; 2865:189-219. [PMID: 39424725 DOI: 10.1007/978-1-0716-4188-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Minimal residual disease (MRD) diagnostics is of high clinical relevance in patients with indolent B-cell non-Hodgkin lymphomas (B-NHL), B-cell chronic lymphocytic leukemia (CLL), and multiple myeloma and serves as a surrogate parameter to evaluate treatment effectiveness and long-term prognosis. Real-time quantitative PCR (RQ-PCR) targeting circulating lymphoma cells is still the gold standard for MRD detection in indolent B-NHL and currently the most sensitive and the most broadly applied method in follicular lymphoma (FL) and mantle cell lymphoma (MCL). Alternatively, droplet digital PCR (ddPCR) can be used for MRD monitoring in multiple myeloma, mantle cell lymphoma, CLL, and FL with comparable sensitivity, accuracy, and reproducibility.The most broadly applicable MRD target in B-NHL is the junctional regions of the rearranged immunoglobulin heavy (IGH) and light chain genes. Complete and incomplete IGH and additionally IG kappa light chain rearrangements can be used as targets for MRD. Next-generation sequencing (NGS) of IG-rearrangements (IG-NGS) as new sequencing-based technology can overcome the limitation of PCR-based approaches and has a potential for higher sensitivity. Chromosomal translocations like the t(14;18)(q32;q21) translocation associated with IGH::BCL2 fusion in FL and t(11;14)(q13;q32) translocation in MCL leading to the IGH::CCND1 fusion can be used as MRD target in selected lymphoma subtypes. In patients with CLL, both flow-cytometry and RQ-PCR are equally suited for MRD assessment as long as a sensitivity of 10-4 is achieved.MRD diagnostics targeting the IG loci is complex and requires extensive knowledge and experience because the junctional regions of each clonal rearranged gene have to be identified before the patient-specific PCR assays can be designed for MRD monitoring. In addition, the presence and load of somatic hypermutation within the rearranged IGH gene occurring during B-cell development of germinal center and post-germinal center B-cell lymphomas may hamper appropriate primer binding leading to false-negative results. The translocations mentioned above have the advantage that consensus forward primers and probes, both placed in the breakpoint regions of chromosome 18 in FL and chromosome 11 in MCL, can be used in combination with a reverse primer placed in the IGH joining region of chromosome 14. PCR-based methods using allele-specific primers can reach a high sensitivity of up to 10-5. This chapter provides all relevant background information and technical aspects for the complete laboratory process from detection of the clonal IG gene rearrangements and the chromosomal translocations at diagnosis to the actual MRD measurements in clinical follow-up samples of B-NHL. However, it should be noted that MRD diagnostics for clinical treatment protocols has to be accompanied by regular international quality control rounds to ensure the reproducibility and reliability of the MRD results. This is available by the EuroMRD network ( https://euromrd.org ), a subgroup of ESHLO ( https://eslho.org ).
Collapse
Affiliation(s)
- Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Monika Brüggemann
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Ritgen
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Jacques J M van Dongen
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC, USAL-CSIC-FICUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
- European Scientific foundation for Laboratory Hemato Oncology (ESLHO), Zutphen, The Netherlands
| |
Collapse
|
3
|
Arai M, Hamad A, Almasry Y, Alamer A, Rasheed W, Aljurf M, El Fakih R. Minimal residual disease testing for classical Hodgkin lymphoma: A comprehensive review. Crit Rev Oncol Hematol 2024; 204:104503. [PMID: 39245298 DOI: 10.1016/j.critrevonc.2024.104503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024] Open
Abstract
Classical Hodgkin lymphoma (cHL) is a common lymphoma that affects young patients. Fortunately, the disease is highly curable as it is susceptible to the currently available treatment modalities. Disease monitoring with Positron Emission Tomography and Computed Tomography (PET/ CT) is an integral part of managing these patients. PET guided protocols are currently used to adjust treatment according to the response. The pivotal idea behind the use of response-adapted approaches is to preserve efficacy while decreasing the toxicity. It also helps to intensify therapy in patients in need because of suboptimal response. However, imaging techniques are limited by their sensitivity and specificity. Minimal Residual Disease (MRD) assessment is a newly emerging concept in many hematologic malignancies. It utilizes various molecular techniques such as polymerase chain reaction (PCR), and next-generation sequencing (NGS) as well as flow cytometry, to detect disease traces. This review looks into MRD detection techniques, its current applications, and the evidence in the literature for its use in cHL.
Collapse
Affiliation(s)
- Momo Arai
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Alaa Hamad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia.
| | - Yazan Almasry
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Abdullah Alamer
- Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Walid Rasheed
- Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Mahmoud Aljurf
- Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Riad El Fakih
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| |
Collapse
|
4
|
Patel DA, Wan F, Trinkaus K, Guy DG, Edwin N, Watkins M, Bartlett NL, Cashen A, Fehniger TA, Ghobadi A, Shah NM, Kahl BS. Bendamustine/Rituximab Plus Cytarabine/Rituximab, With or Without Acalabrutinib, for the Initial Treatment of Transplant-Eligible Mantle Cell Lymphoma Patients: Pooled Data From Two Pilot Studies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023:S2152-2650(23)00131-3. [PMID: 37183115 DOI: 10.1016/j.clml.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Mantle cell lymphoma (MCL) is a moderately aggressive lymphoma subtype, generally viewed as incurable. For younger, fit patients, the standard of care remains various high-dose cytarabine-based induction regimens followed by autologous hematopoietic cell transplant and 3 years of rituximab maintenance. Despite reasonably good outcomes, with median progression-free survival in the range of 7 to 9 years, most patients eventually relapse, indicating a need to improve the safety and tolerability of remission induction strategies. METHODS Given the impressive activity of bendamustine/rituximab (BR) in older patients with MCL, we developed an induction regimen modeled after the Nordic Regimen but substituted BR in place of R-CHOP. In a second pilot study, we incorporated the second-generation Bruton tyrosine kinase inhibitor (BTKi), acalabrutinib, into the regimen. The primary endpoint of both studies was stem cell mobilization success rate. RESULTS All patients successfully underwent stem cell harvest in both studies. CONCLUSION The experience from our single institution pilot study suggested that sequential rather than alternating BR and cytarabine/rituximab (CR) was easier to administer from the standpoint of toxicities and subsequent dose modifications. Safety and efficacy data from the 2 pilot studies, FitMCL 1.0 and 2.0, were similar. The pilot studies provided preliminary safety data supporting the development of the NCTN trial EA4181, assessing three different induction regimens with or without acalabrutinib.
Collapse
Affiliation(s)
- Dilan A Patel
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Fei Wan
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Kathryn Trinkaus
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Daniel G Guy
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Natasha Edwin
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Marcus Watkins
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Nancy L Bartlett
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Amanda Cashen
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Todd A Fehniger
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Armin Ghobadi
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Neha-Mehta Shah
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Brad S Kahl
- Washington University in St Louis School of Medicine, St Louis, MO, 63110.
| |
Collapse
|
5
|
Metzner B, Müller TH, Casper J, Kimmich C, Köhne CH, Petershofen E, Renzelmann A, Thole R, Voss A, Dreyling M, Hoster E, Klapper W, Pott C. Long-term outcome in patients with mantle cell lymphoma following high-dose therapy and autologous stem cell transplantation. Eur J Haematol 2023. [PMID: 37094812 DOI: 10.1111/ejh.13985] [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: 12/07/2022] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND Long-term clinical and molecular remissions in patients with mantle cell lymphoma (MCL) after autologous stem cell transplantation (ASCT) have been evaluated in only a few studies. DESIGN AND METHODS Sixty-five patients with MCL received ASCT (54 first-line ASCT, 10 second-line ASCT, and 1 third-line ASCT). In the case of long-term remission (≥5 years; n = 27), peripheral blood was tested for minimal residual disease (MRD) by t(11;14)- and IGH-PCR at the last follow-up. RESULTS Ten-year overall survival (OS), progression-free survival (PFS), and freedom from progression (FFP) after first-line ASCT were 64%, 52%, and 59% versus after second-line ASCT 50%, 20%, and 20%, respectively. Five-year OS, PFS, and FFP for the first-line cohort were 79%, 63%, and 69%, respectively. Five-year OS, PFS, and FFP after second-line ASCT were 60%, 30%, and 30%, respectively. Treatment-related mortality (3 months after ASCT) was 1.5%. So far 26 patients developed sustained long-term clinical and molecular complete remissions of up to 19 years following ASCT in first treatment line. CONCLUSION Sustained long-term clinical and molecular remissions are achievable following ASCT.
Collapse
Affiliation(s)
- Bernd Metzner
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, 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
| | - Claus-Henning Köhne
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | | | - Andrea Renzelmann
- 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
| | - Martin Dreyling
- Department of Internal Medicine III, University Hospital Munich, Grosshadern, Germany
| | - Eva Hoster
- Department of Internal Medicine III, University Hospital Munich, Grosshadern, Germany
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University of Schleswig-Holstein, Kiel, Germany
| | - Christiane Pott
- Department of Medicine II, University of Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
6
|
Assanto GM, Del Giudice I, Della Starza I, Soscia R, Cavalli M, Cola M, Bellomarino V, Di Trani M, Guarini A, Foà R. Research Topic: Measurable Residual Disease in Hematologic Malignancies. Can digital droplet PCR improve measurable residual disease monitoring in chronic lymphoid malignancies? Front Oncol 2023; 13:1152467. [PMID: 36998457 PMCID: PMC10043164 DOI: 10.3389/fonc.2023.1152467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/01/2023] [Indexed: 03/17/2023] Open
Abstract
Minimal/measurable residual disease (MRD) monitoring is progressively changing the management of hematologic malignancies. The possibility of detecting the persistence/reappearance of disease in patients in apparent clinical remission offers a refined risk stratification and a treatment decision making tool. Several molecular techniques are employed to monitor MRD, from conventional real-time quantitative polymerase chain reaction (RQ-PCR) to next generation sequencing and digital droplet PCR (ddPCR), in different tissues or compartments through the detection of fusion genes, immunoglobulin and T-cell receptor gene rearrangements or disease-specific mutations. RQ-PCR is still the gold standard for MRD analysis despite some limitations. ddPCR, considered the third-generation PCR, yields a direct, absolute, and accurate detection and quantification of low-abundance nucleic acids. In the setting of MRD monitoring it carries the major advantage of not requiring a reference standard curve built with the diagnostic sample dilution and of allowing to reduce the number of samples below the quantitative range. At present, the broad use of ddPCR to monitor MRD in the clinical practice is limited by the lack of international guidelines. Its application within clinical trials is nonetheless progressively growing both in acute lymphoblastic leukemia as well as in chronic lymphocytic leukemia and non-Hodgkin lymphomas. The aim of this review is to summarize the accumulating data on the use of ddPCR for MRD monitoring in chronic lymphoid malignancies and to highlight how this new technique is likely to enter into the clinical practice.
Collapse
Affiliation(s)
| | - Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
- *Correspondence: Ilaria Del Giudice, ; Robin Foà,
| | - Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
- Gruppo Italiano Malattie Ematologiche dell'Adulto (GIMEMA), Fondazione GIMEMA Franco Mandelli Onlus, Rome, Italy
| | - Roberta Soscia
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Marzia Cavalli
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Mattia Cola
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Vittorio Bellomarino
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Mariangela Di Trani
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Anna Guarini
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
- *Correspondence: Ilaria Del Giudice, ; Robin Foà,
| |
Collapse
|
7
|
Qualls D, Kumar A. Personalized approaches for treatment-naïve mantle cell lymphoma. Expert Rev Hematol 2023; 16:95-107. [PMID: 36748785 DOI: 10.1080/17474086.2023.2174516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Mantle cell lymphoma (MCL) can have diverse disease presentations, which vary in aggressiveness and prognosis, and can occur in patients with varying ability to tolerate therapy. Additionally, the options for treatment of MCL have expanded rapidly in the last decade, translating to improved outcome for patients. AREAS COVERED We review the initial evaluation of patients with MCL, identifying disease- and patient-specific prognostic factors, along with personalized therapies for patients with MCL. Specific scenarios include indolent and limited-stage MCL, advanced-stage disease in transplant-eligible and ineligible patients, and high-risk TP53 mutant disease. Ongoing trials and future directions in MCL treatment are also highlighted. EXPERT OPINION Given the wide array of disease and patient presentations with MCL, a personalized therapeutic approach is needed to optimize outcomes. The best therapeutic strategy should incorporate disease prognostic factors, patient status and comorbidities, goals of care, and response to treatment.
Collapse
Affiliation(s)
- David Qualls
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Anita Kumar
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
8
|
Schroers-Martin JG, Alig S, Garofalo A, Tessoulin B, Sugio T, Alizadeh AA. Molecular Monitoring of Lymphomas. ANNUAL REVIEW OF PATHOLOGY 2023; 18:149-180. [PMID: 36130071 DOI: 10.1146/annurev-pathol-050520-044652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Molecular monitoring of tumor-derived alterations has an established role in the surveillance of leukemias, and emerging nucleic acid sequencing technologies are likely to similarly transform the clinical management of lymphomas. Lymphomas are well suited for molecular surveillance due to relatively high cell-free DNA and circulating tumor DNA concentrations, high somatic mutational burden, and the existence of stereotyped variants enabling focused interrogation of recurrently altered regions. Here, we review the clinical scenarios and key technologies applicable for the molecular monitoring of lymphomas, summarizing current evidence in the literature regarding molecular subtyping and classification, evaluation of treatment response, the surveillance of active cellular therapies, and emerging clinical trial strategies.
Collapse
Affiliation(s)
- Joseph G Schroers-Martin
- Department of Medicine, Divisions of Hematology and Oncology, Stanford University Medical Center, Stanford, California, USA;
| | - Stefan Alig
- Department of Medicine, Divisions of Hematology and Oncology, Stanford University Medical Center, Stanford, California, USA;
| | - Andrea Garofalo
- Department of Medicine, Divisions of Hematology and Oncology, Stanford University Medical Center, Stanford, California, USA;
| | - Benoit Tessoulin
- Department of Medicine, Divisions of Hematology and Oncology, Stanford University Medical Center, Stanford, California, USA; .,Current affiliation: Clinical Hematology Department, Nantes University Hospital, Nantes, France
| | - Takeshi Sugio
- Department of Medicine, Divisions of Hematology and Oncology, Stanford University Medical Center, Stanford, California, USA;
| | - Ash A Alizadeh
- Department of Medicine, Divisions of Hematology and Oncology, Stanford University Medical Center, Stanford, California, USA; .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA.,Stanford Cancer Institute, Stanford University, Stanford, California, USA
| |
Collapse
|
9
|
Kumar A, Eyre TA, Lewis KL, Thompson MC, Cheah CY. New Directions for Mantle Cell Lymphoma in 2022. Am Soc Clin Oncol Educ Book 2022; 42:1-15. [PMID: 35561299 DOI: 10.1200/edbk_349509] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mantle cell lymphoma is a rare B-cell non-Hodgkin lymphoma that is clinically and biologically heterogeneous. Risk stratification at the time of diagnosis is critical. One of the most powerful prognostic indices is the Mantle Cell Lymphoma International Prognostic Index-Combined, which integrates an estimate of proliferation (Ki67 index) with the standard Mantle Cell Lymphoma International Prognostic Index clinical factors. In addition, the presence of TP53 mutation is associated with suboptimal response to intensive chemoimmunotherapy and particularly dismal survival outcomes. Given their excellent activity in the relapsed/refractory setting, increasingly, biologically targeted therapeutics-such as covalent Bruton tyrosine kinase inhibitors, lenalidomide, and venetoclax-are being incorporated into "chemotherapy-free" regimens and in combination with established chemoimmunotherapy backbones for treatment-naïve mantle cell lymphoma. In addition, risk-adapted treatment programs are increasingly being studied. These programs tailor treatment according to baseline prognostic factors (e.g., presence of TP53 mutation) and may incorporate biomarkers of response such as minimal residual disease assessment. Although still investigational, these studies present an opportunity to move beyond the biology-agnostic, historical fitness-based treatment selection paradigm and toward a more personalized, tailored treatment approach in mantle cell lymphoma. After Bruton tyrosine kinase inhibitor failure, many promising standard or investigational therapies exist, including CAR T-cell therapy (including brexucabtagene autoleucel and lisocabtagene maraleucel), bispecific antibody therapy targeting CD20-CD3, zilovertamab vedotin (an antibody-drug conjugate that targets ROR1), and the noncovalent Bruton tyrosine kinase inhibitor pirtobrutinib. These new therapies show promising efficacy, even among high-risk patients, and will likely translate to improvements in survival outcomes for patients with progressive mantle cell lymphoma following treatment with a Bruton tyrosine kinase inhibitor.
Collapse
Affiliation(s)
- Anita Kumar
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Toby A Eyre
- Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Katharine L Lewis
- Department of Haematology, Sir Charles Gairdner Hospital, Perth, Australia.,Medical School, University of Western Australia, Perth, Australia
| | | | - Chan Y Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Perth, Australia.,Medical School, University of Western Australia, Perth, Australia
| |
Collapse
|
10
|
Circulating Tumor DNA Predicts Therapeutic Outcome in Mantle Cell Lymphoma. Blood Adv 2022; 6:2667-2680. [PMID: 35143622 PMCID: PMC9043939 DOI: 10.1182/bloodadvances.2021006397] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/02/2022] [Indexed: 11/30/2022] Open
Abstract
Early changes in ctDNA dynamics are prognostic in untreated MCL. Bortezomib maintenance after bortezomib-based induction therapy does not improve outcome in untreated MCL.
Mantle cell lymphoma (MCL) is biologically and clinically heterogeneous and would benefit from prognostic biomarkers to guide management. Circulating tumor DNA (ctDNA) is a novel prognostic biomarker in diffuse large B-cell lymphoma that may have applicability in MCL. We analyzed ctDNA dynamics in previously untreated patients with MCL who received induction therapy with bortezomib and DA-EPOCH-R for 6 cycles followed by random assignment to observation or bortezomib maintenance in responding patients in a prospective phase 2 study. Most patients also underwent initial treatment window of bortezomib alone prior to induction. Serum was collected pretreatment, after the window, after cycles 1 and 2, at the end of induction, and at each follow-up visit along with restaging computed tomography scans. Next-generation sequencing was used to identify and quantify ctDNA encoding the immunoglobulin receptor sequences in serum as markers of minimal residual disease. Fifty-three patients were enrolled, with a median follow-up of 12.7 years. Patients without detectable ctDNA after 2 cycles of induction had longer progression-free survival (PFS) and overall survival (OS) compared with those with detectable ctDNA (median PFS, 2.7 vs 1.8 years; overall P = .005; median OS, 13.8 vs 7.4 years; overall P = .03). Notably, in vivo assessment of ctDNA dynamics during the bortezomib window was not prognostic, and there was no difference in PFS or OS with bortezomib maintenance. ctDNA monitoring after induction showed that molecular relapse preceded clinical relapse in some cases. In conclusion, interim ctDNA negativity strongly correlates with improved survival and supports the investigation of response-adapted strategies. This trial was registered at www.clinicaltrials.gov as #NCT00114738.
Collapse
|
11
|
Elkjær LAL, Cédile O, Hansen MH, Nielsen C, Møller MB, Abildgaard N, Haaber J, Nyvold CG. Exploration of residual disease in stem cell products from mantle cell lymphoma using next-generation sequencing. Leuk Res Rep 2022; 18:100341. [PMID: 36039182 PMCID: PMC9418493 DOI: 10.1016/j.lrr.2022.100341] [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: 01/14/2022] [Revised: 07/01/2022] [Accepted: 08/07/2022] [Indexed: 11/05/2022] Open
Abstract
High-dose chemotherapy followed by autologous stem cell transplantation (ASCT) has become a treatment option for fit patients with mantle cell lymphoma (MCL). However, these patients often relapse within few years, potentially caused by contaminating lymphoma cells within the reinfused stem cell product (SCP). Studies have shown that measurable residual disease, also termed minimal residual disease (MRD), following ASCT predicts shorter survival. Using next-generation sequencing, we explore whether the diagnostic MCL clonotype is present within the infused SCP. MRD was detected in 4/17 of the SCPs, ranging 4–568 clonal cells/100,000 cells. With a median survival of 17 months, 3/4 of patients with MRD+ graft succumbed from MCL relapse versus 2/13 in the MRD– fraction. Patients receiving MRD+ grafts had increased risk of mortality, and thus screening of SCPs may be important for clinical decision-making.
Collapse
|
12
|
Bortezomib in combination with fludarabine plus cyclophosphamide for patients with relapsed or refractory mantle-cell lymphoma: results of the LYM-4003 study. Ann Hematol 2021; 100:2961-2968. [PMID: 34331111 DOI: 10.1007/s00277-021-04619-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 07/18/2021] [Indexed: 10/20/2022]
Abstract
This study aimed to identify the maximum-tolerated dose (MTD) of cyclophosphamide when combined with bortezomib and fludarabine (B-FC) in a phase 1b trial, and to assess the efficacy and safety of this combination in a phase 2 trial in patients with relapsed or refractory MCL (rrMCL). Forty patients were enrolled between April 8, 2011, and October 10, 2015. The MTD of cyclophosphamide was identified to be 250 mg/m2 days 1-2. At a median follow-up of 31.6 months (13.5-47.4), among 32 patients in phase 2, 10 (31%) had a complete response and 13 (41%) had a partial response. The median progression-free survival was 21 months (95% CI 7.3-34.7), and the median overall survival was 32.4 months (95% CI 17.8-47.0). Grade 3-4 hematologic AEs included neutropenia (27%) and thrombocytopenia (39%). The B-FC regimen has satisfactory responses and manageable toxicities in rrMCL patients (ClinicalTrials.gov NCT01322776).
Collapse
|
13
|
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
|
14
|
Roerden M, Wirths S, Sökler M, Bethge WA, Vogel W, Walz JS. Impact of Mantle Cell Lymphoma Contamination of Autologous Stem Cell Grafts on Outcome after High-Dose Chemotherapy. Cancers (Basel) 2021; 13:cancers13112558. [PMID: 34071000 PMCID: PMC8197101 DOI: 10.3390/cancers13112558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 11/19/2022] Open
Abstract
Simple Summary High-dose chemotherapy followed by autologous hematopoietic stem cell transplantation (Auto-HSCT) is a standard frontline treatment for fit mantle cell lymphoma (MCL) patients. As there is a need for predictive factors to identify patients unlikely to benefit from this therapy, we investigated the prognostic impact of lymphoma cell contamination of autologous stem cell grafts. Analyzing a cohort of 36 MCL patients, we show that lymphoma cell contamination of stem cell grafts is associated with poor outcomes after Auto-HSCT. Its analysis might thus improve risk assessment and enable risk-stratified treatment strategies for MCL patients. Abstract Novel predictive factors are needed to identify mantle cell lymphoma (MCL) patients at increased risk for relapse after high-dose chemotherapy and autologous hematopoietic stem cell transplantation (HDCT/Auto-HSCT). Although bone marrow and peripheral blood involvement is commonly observed in MCL and lymphoma cell contamination of autologous stem cell grafts might facilitate relapse after Auto-HSCT, prevalence and prognostic significance of residual MCL cells in autologous grafts are unknown. We therefore performed a multiparameter flow cytometry (MFC)-based measurable residual disease (MRD) assessment in autologous stem cell grafts and analyzed its association with clinical outcome in an unselected retrospective cohort of 36 MCL patients. MRD was detectable in four (11%) autologous grafts, with MRD levels ranging from 0.002% to 0.2%. Positive graft-MRD was associated with a significantly shorter progression-free and overall survival when compared to graft-MRD negative patients (median 9 vs. 56 months and 25 vs. 132 months, respectively) and predicted early relapse after Auto-HSCT (median time to relapse 9 vs. 44 months). As a predictor of outcome after HDCT/Auto-HSCT, MFC-based assessment of graft-MRD might improve risk stratification and support clinical decision making for risk-oriented treatment strategies in MCL.
Collapse
Affiliation(s)
- Malte Roerden
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- Correspondence:
| | - Stefan Wirths
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Martin Sökler
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Wolfgang A. Bethge
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Wichard Vogel
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Juliane S. Walz
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology (IKP) and Robert Bosch Center for Tumor Diseases (RBCT), Auerbachstr. 112, 70376 Stuttgart, Germany
| |
Collapse
|
15
|
Stance of MRD in Non-Hodgkin's Lymphoma and its upsurge in the novel era of cell-free DNA. Clin Transl Oncol 2021; 23:2206-2219. [PMID: 33991328 DOI: 10.1007/s12094-021-02635-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/02/2021] [Indexed: 10/21/2022]
Abstract
Cancer genomics has evolved over the years from understanding the pathogenesis of cancer to screening the future possibilities of cancer occurrence. Understanding the genetic profile of tumors holds a prognostic as well as a predictive value in this era of therapeutic surveillance, molecular remission, and precision medicine. Identifying molecular markers in tumors is the current standard of approach, and requires an efficient combination of an accessible sample type and a profoundly sensitive technique. Liquid biopsy or cell-free DNA has evolved as a novel sample type with promising results in recent years. Although cell-free DNA has significant role in various cancer types, this review focuses on its application in Non-Hodgkin's Lymphoma. Beginning with the current concept and clinical relevance of minimal residual disease in Non-Hodgkin's lymphoma, we discuss the literature on circulating DNA and its evolving application in the realm of cutting-edge technology.
Collapse
|
16
|
Rituximab/bendamustine and rituximab/cytarabine induction therapy for transplant-eligible mantle cell lymphoma. Blood Adv 2021; 4:858-867. [PMID: 32126141 DOI: 10.1182/bloodadvances.2019001355] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/31/2020] [Indexed: 11/20/2022] Open
Abstract
The addition of high-dose cytarabine to rituximab/bendamustine (RB) induction could improve outcomes for transplant-eligible patients with mantle cell lymphoma (MCL). We conducted a pooled analysis of 2 phase 2 trials and an off-trial cohort each testing 3 cycles of RB and 3 cycles of rituximab/high-dose cytarabine (RC) followed by autologous stem cell transplantation (ASCT) among untreated, transplant-eligible patients with MCL. Dana-Farber Cancer Institute (DFCI) and Washington University in St. Louis (WUSTL) led separate phase 2 trials testing sequential and alternating cycles of RB/RC, respectively. Patients treated at DFCI with sequential RB/RC off trial were retrospectively identified. Minimal residual disease (MRD) was assessed in the DFCI trial. A total of 88 patients (23 DFCI trial, 18 WUSTL trial, and 47 off trial) received RB/RC; 92% of patients completed induction, and 84% underwent planned consolidative ASCT. Grade 3 or 4 adverse events among trial patients included lymphopenia (88%), thrombocytopenia (85%), neutropenia (83%), and febrile neutropenia (15%). There were no treatment-related deaths during induction and 2 following ASCT. Among 87 response-evaluable patients, the end-of-induction overall and complete response rates were 97% and 90%, respectively. After a median follow-up of 33 months, 3-year progression-free survival and overall survival were 83% and 92%, respectively. Patients undergoing MRD testing experienced prolonged MRD negativity after ASCT with emergence of MRD occurring in only 1 patient who subsequently relapsed. RB/RC followed by ASCT achieves high rates of durable remissions in transplant-eligible patients with MCL. These trials were registered at www.clinicaltrials.gov as #NCT01661881 (DFCI trial) and #NCT02728531 (WUSTL trial).
Collapse
|
17
|
Wu X, Lu H, Pang T, Li X, Luo H, Tan H, Liu S. Association of minimal residual disease levels with clinical outcomes in patients with mantle cell lymphoma: A meta-analysis. Leuk Res 2021; 108:106605. [PMID: 34090063 DOI: 10.1016/j.leukres.2021.106605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/12/2021] [Accepted: 04/26/2021] [Indexed: 02/08/2023]
Abstract
Some studies have elucidated that Minimal residual disease (MRD) in patient with Mantle Cell Lymphoma (MCL) was a significant prognostic factor, with potential value in assessing overall survival (OS) and progression-free survival (PFS). However, most studies were widely varied in included population, sample sources and MRD detection time points. Some studies even have conflicting results. In view of this, a meta-analysis was performed to evaluate association of MRD levels with clinical outcomes in patients with MCL. We identified 7 included articles, which were published in recent 20 years. Then, we extracted or calculated hazard ratios (HRs) and their 95 % confidence intervals (CIs). Our results reveal that patients with MRD negativity have improved OS (HR = 0.63; 95 % CI: 0.50-0.79) and PFS (HR = 0.40, 95 % CI: 0.21-0.76), comparing with patients with MRD positivity. There are also consistent results in subgroups based on sample sources and MRD detection time points. Our study also demonstrates that MRD level is a strong prognostic factor of clinical outcomes. Thus, MRD is expected to be an effective clinical indicator for assessing prognosis and guide treatment decisions in MCL patients.
Collapse
Affiliation(s)
- Xue Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan Province, China
| | - Hongyu Lu
- School of Medical Technology, Chengdu University of TCM, Chengdu, 611137, Sichuan Province, China; Key Laboratory of Transplant Engineering and Immunology, Regenerative Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Pang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan Province, China
| | - Xue Li
- Department of Laboratory Medicine, Hanyuan People's Hospital, Yuan, 25000, Sichuan, China
| | - Hongzhi Luo
- School of Laboratory Medicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Hong Tan
- Department of General Surgery, Chengdu Integrated TCM&Western Medicine Hospital (Chengdu First People's Hospital), No.18 Vientiane North Road, Hi-tech Zone, Chengdu, 610041, China.
| | - Shan Liu
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No.32 West Second Section First Ring Road, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
18
|
Malecek MK, Mehta-Shah N. Prognosis and risk stratification of peripheral T-cell lymphomas. Semin Hematol 2021; 58:70-77. [PMID: 33906724 DOI: 10.1053/j.seminhematol.2021.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/22/2021] [Accepted: 02/01/2021] [Indexed: 12/31/2022]
Abstract
Peripheral T-cell lymphomas represent a rare heterogeneous group of non-Hodgkin lymphomas with generally worse outcomes with standard chemotherapy compared to B-cell lymphomas. Clinical risk prediction tools at baseline have been shown to be prognostic but generally do not impact clinical decision making. However, improving understanding of the prognostic implications of histology and its molecular underpinnings as well as strategies surrounding the use of CD30 as a predictive biomarker for brentuximab vedotin have led to better understanding of how to risk stratify patients. Baseline, interim, and end of treatment PET/CT as evaluated by the Lugano criteria as well as by baseline metabolic tumor volume have also been shown to be prognostic. The role of minimal residual disease tools such as cell free DNA and T-cell gene receptor sequencing remain active areas of investigation in hopes to develop predictive biomarkers in these rare diseases. This review focuses on strategies used to prognosticate in more common forms of peripheral T-cell lymphoma as well as in extranodal NK/T-cell lymphoma.
Collapse
|
19
|
Zhou Y, Chen H, Tao Y, Zhong Q, Shi Y. Minimal Residual Disease and Survival Outcomes in Patients with Mantle Cell Lymphoma: a systematic review and meta-analysis. J Cancer 2021; 12:553-561. [PMID: 33391451 PMCID: PMC7738989 DOI: 10.7150/jca.51959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/18/2020] [Indexed: 01/07/2023] Open
Abstract
Background: Minimal residual disease (MRD) has shown the prognostic value in mantle cell lymphoma (MCL). To quantify the relationships between progression free survival (PFS) and overall survival (OS) with MRD status in MCL, we conducted this meta-analysis. Methods: We searched databases including Pubmed, Embase, Web of Science and the Cochrane Library up to July 15th, 2020. Data of patients' characteristics, MRD assessment and survival outcomes were extracted and analyzed. Results: Ten articles were included. For the impact of post-induction MRD status on survival outcomes, MRD positive status was associated with worse PFS (HR=1.44; 95%CI 1.27-1.62; P<0.00001) and OS (HR=1.30; 95%CI 1.03-1.64; P=0.03) compared with MRD negative status. Regarding the impact of post-consolidation MRD status on survival outcomes, MRD positivity predicted shorter PFS (HR=1.84; 95%CI 1.49-2.26; P<0.00001) and OS (HR=2.38; 95%CI 1.85-3.06; P<0.00001) than MRD negativity. Conclusions: This study indicated that MRD positivity after induction and consolidation treatments was associated with worse PFS and OS for MCL. MRD-based treatment strategies should be further explored in clinical trials and real-world practice.
Collapse
Affiliation(s)
| | | | | | | | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| |
Collapse
|
20
|
Kambhampati S, Hunter B, Varnavski A, Fakhri B, Kaplan L, Ai WZ, Pampaloni M, Huang CY, Martin T, Damon L, Andreadis CB. Ofatumumab, Etoposide, and Cytarabine Intensive Mobilization Regimen in Patients with High-risk Relapsed/Refractory Diffuse Large B-Cell Lymphoma Undergoing Autologous Stem Cell Transplantation. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 21:246-256.e2. [PMID: 33288485 DOI: 10.1016/j.clml.2020.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND More than one-half of high-risk patients with relapsed/refractory (rr) diffuse large B-cell lymphoma (DLBCL) relapse after autologous hematopoietic cell transplantation (auto-HCT). In this phase II study, we investigate the long-term outcomes of high-risk patients with rrDLBCL receiving intensive consolidation therapy (ICT) with OVA (ofatumumab, etoposide, and high-dose cytarabine) prior to auto-HCT. PATIENTS AND METHODS The primary endpoints were the ability of OVA to mobilize peripheral stem cells and the 2-year progression-free survival (PFS) rate following OVA. Secondary endpoints included safety, 2-year overall survival (OS), impact of cell of origin (COO), and the prognostic utility of next-generation sequencing minimal residual disease (MRD) testing. We simultaneously retrospectively assessed the outcomes of DLBCL patients who underwent ICT with a similar regimen at our institution. RESULTS Twenty-seven patients received salvage chemotherapy, with a response rate of 25% in patients with germinal center B-cell (GCB)-DLBCL versus 92% in patients with non-GCB-DLBCL (P = .003). Nineteen responding patients underwent ICT with OVA (100% successful stem cell mobilization). The 2-year PFS and OS rate was 47% and 59%, respectively, with no difference based on COO. Similar findings were observed when the study and retrospective cohorts were combined. Neutropenia was the most common toxicity (47%). MRD-negative patients at the completion of salvage had a median OS of not reached versus 3.5 months in MRD-positive patients (P = .02). CONCLUSIONS OVA followed by auto-HCT is effective and safe for high-risk rrDLBCL. Patients with GCB-DLBCL had a lower response to salvage chemotherapy, but no difference in outcomes based on COO was seen after auto-HCT. MRD testing in the relapsed setting was predictive of long-term survival.
Collapse
MESH Headings
- Adult
- Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Cytarabine/administration & dosage
- Cytarabine/adverse effects
- Drug Resistance, Neoplasm
- Etoposide/administration & dosage
- Etoposide/adverse effects
- Female
- Germinal Center/pathology
- Hematopoietic Stem Cell Transplantation
- Humans
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Male
- Middle Aged
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/therapy
- Neoplasm, Residual
- Prognosis
- Progression-Free Survival
- Retrospective Studies
- Salvage Therapy/adverse effects
- Salvage Therapy/methods
- Survival Rate
- Transplantation, Autologous/methods
Collapse
Affiliation(s)
- Swetha Kambhampati
- Division of Hematology/Oncology, Department of Medicine, UCSF Medical Center, San Francisco, CA; UCSF Helen Diller Family Comprehensive Cancer Center, UCSF Medical Center, San Francisco, CA
| | - Bradley Hunter
- Department of Hematology, Intermountain Healthcare, Salt Lake City, UT
| | | | - Bita Fakhri
- Division of Hematology/Oncology, Department of Medicine, UCSF Medical Center, San Francisco, CA; UCSF Helen Diller Family Comprehensive Cancer Center, UCSF Medical Center, San Francisco, CA
| | - Lawrence Kaplan
- Division of Hematology/Oncology, Department of Medicine, UCSF Medical Center, San Francisco, CA; UCSF Helen Diller Family Comprehensive Cancer Center, UCSF Medical Center, San Francisco, CA
| | - Weiyun Z Ai
- Division of Hematology/Oncology, Department of Medicine, UCSF Medical Center, San Francisco, CA; UCSF Helen Diller Family Comprehensive Cancer Center, UCSF Medical Center, San Francisco, CA
| | | | - Chiung-Yu Huang
- UCSF Helen Diller Family Comprehensive Cancer Center, UCSF Medical Center, San Francisco, CA; Department of Epidemiology and Biostatistics, UCSF Medical Center, San Francisco, CA
| | - Thomas Martin
- Division of Hematology/Oncology, Department of Medicine, UCSF Medical Center, San Francisco, CA; UCSF Helen Diller Family Comprehensive Cancer Center, UCSF Medical Center, San Francisco, CA
| | - Lloyd Damon
- Division of Hematology/Oncology, Department of Medicine, UCSF Medical Center, San Francisco, CA; UCSF Helen Diller Family Comprehensive Cancer Center, UCSF Medical Center, San Francisco, CA
| | - Charalambos B Andreadis
- Division of Hematology/Oncology, Department of Medicine, UCSF Medical Center, San Francisco, CA; UCSF Helen Diller Family Comprehensive Cancer Center, UCSF Medical Center, San Francisco, CA.
| |
Collapse
|
21
|
Five-year outcomes of the S1106 study of R-hyper-CVAD vs R-bendamustine in transplant-eligible patients with mantle cell lymphoma. Blood Adv 2020; 3:3132-3135. [PMID: 31648328 DOI: 10.1182/bloodadvances.2019000526] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/14/2019] [Indexed: 01/21/2023] Open
Abstract
Key Points
Five-year follow-up of S1106 demonstrates similar efficacy, MRD negativity, and 5-year survival with RH or RB, but RH was more toxic than RB. RB showed excellent efficacy and survival and less toxicity compared with a cytarabine-based regimen in transplant-eligible MCL patients.
Collapse
|
22
|
Rodgers TD, Friedberg JW. Key Clinical and Translational Research Questions to Address Unmet Needs in Mantle Cell Lymphoma. Hematol Oncol Clin North Am 2020; 34:983-996. [PMID: 32861291 DOI: 10.1016/j.hoc.2020.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Survival for patients with mantle cell lymphoma has improved dramatically over the last 2 decades owing to a better understanding of disease biology and the development of more effective treatment regimens for patients with untreated and relapsed disease. With these advancements, we are now poised to ask questions that challenge old treatment strategies, use new technologies, and improve our understanding of disease heterogeneity. This article focuses on questions that we believe will drive the future of mantle cell lymphoma treatment. Although not an exhaustive list, we review current literature, ongoing studies, and provide expert opinion on future trial design.
Collapse
Affiliation(s)
- Thomas D Rodgers
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, 601 Elmwood Avenue, Box 704, Rochester, NY 14642, USA.
| | - Jonathan W Friedberg
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, 601 Elmwood Avenue, Box 704, Rochester, NY 14642, USA
| |
Collapse
|
23
|
Minimal Residual Disease in Mantle Cell Lymphoma: Methods and Clinical Significance. Hematol Oncol Clin North Am 2020; 34:887-901. [PMID: 32861285 DOI: 10.1016/j.hoc.2020.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Several biological and clinical features have been recognized in mantle cell lymphoma (MCL). In recent years, the minimal residual disease (MRD) has been extensively investigated and is now considered as one of the strongest clinical predictors in this lymphoma. This article reviews methods used for the assessment of MRD in MCL and discusses their strengths and weaknesses. In addition, it examines the MRD contribution to the biology knowledge of MCL and the development of effective strategies for its management, including the possibility of personalized treatment based on MRD response.
Collapse
|
24
|
Cortelazzo S, Ponzoni M, Ferreri AJM, Dreyling M. Mantle cell lymphoma. Crit Rev Oncol Hematol 2020; 153:103038. [PMID: 32739830 DOI: 10.1016/j.critrevonc.2020.103038] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 06/29/2019] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
MCL is a well-characterized generally aggressive lymphoma with a poor prognosis. However, patients with a more indolent disease have been reported in whom the initiation of therapy can be delayed without any consequence for the survival. In 2017 the World Health Organization updated the classification of MCL describing two main subtypes with specific molecular characteristics and clinical features, classical and indolent leukaemic nonnodal MCL. Recent research results suggested an improving outcome of this neoplasm. The addition of rituximab to conventional chemotherapy has increased overall response rates, but it did not improve overall survival compared to chemotherapy alone. The use of intensive frontline therapies including rituximab and consolidation with autologous stem cell transplantation ameliorated response rate and prolonged progression-free survival in young fit patients, but any impact on survival remains to be proven. Furthermore, the optimal timing, cytoreductive regimen and conditioning regimen, and the clinical implications of achieving a disease remission even at molecular level remain to be elucidated. The development of targeted therapies as the consequence of better understanding of pathogenetic pathways in MCL might improve the outcome of conventional chemotherapy and spare the toxicity of intense therapy in most patients. Cases not eligible for intensive regimens, may be considered for less demanding therapies, such as the combination of rituximab either with CHOP or with purine analogues, or bendamustine. Allogeneic SCT can be an effective option for relapsed disease in patients who are fit enough and have a compatible donor. Maintenance rituximab may be considered after response to immunochemotherapy as the first-line strategy in a wide range of patients. Finally, since the optimal approach to the management of MCL is still evolving, it is critical that these patients are enrolled in clinical trials to identify the better treatment options.
Collapse
Affiliation(s)
| | - Maurilio Ponzoni
- Pathology Unit, San Raffaele Scientific Institute, Milan, Italy; Unit of Lymphoid Malignancies, San Raffaele Scientific Institute, Milan, Italy
| | - Andrés J M Ferreri
- Unit of Lymphoid Malignancies, San Raffaele Scientific Institute, Milan, Italy; Medical Oncology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Martin Dreyling
- Medizinische Klinik III der Universität München-Grosshadern, München, Germany
| |
Collapse
|
25
|
Galimberti S, Genuardi E, Mazziotta F, Iovino L, Morabito F, Grassi S, Ciabatti E, Guerrini F, Petrini M. The Minimal Residual Disease in Non-Hodgkin's Lymphomas: From the Laboratory to the Clinical Practice. Front Oncol 2019; 9:528. [PMID: 31293969 PMCID: PMC6606710 DOI: 10.3389/fonc.2019.00528] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/31/2019] [Indexed: 01/05/2023] Open
Abstract
Minimal residual disease (MRD) in non-Hodgkin's lymphomas (NHLs) still represents matter of interest and debate: indeed, the new available treatments offer higher rates of complete responses and MRD negativity than in the past, with a positive impact on the long-term survival. Furthermore, the introduction of more sensitive and accurate molecular techniques, such as digital PCR (ddPCR) and the next generation sequencing techniques (NGS), increased the possibility of identifying molecular targets to be followed after therapy (such as rearrangement of immunoglobulins, fusion genes, or mutations). This review focused on how molecular biology can help to detect MRD in different types of NHLs and how MRD can change the clinical practice in 2019. In follicular lymphoma (FL), contamination of the grafts and molecular disease persistence after transplantation represent a negative prognostic factors. The combination of Rituximab or Obinutuzumab with Bendamustine seems to be the most effective way to clear MRD in FL patients receiving chemo-immunotherapy (further studies are in progress), and also 90Yttrium-Ibritumomab-Tiuxetan offers a deep clearance of molecular disease. Finally, molecular MRD can further stratify PET-negative cases, with subjects both PET- and MRD-negative presenting the best outcome. In aggressive lymphomas, MRD has a relevant prognostic power and can represent the platform for immunotherapy (such as CAR-T). In diffuse large B-cell lymphoma (DLBCL), the assessment of MRD in the plasma (where cell-free DNA and exosomes circulate) seems to be more predictive than the bone marrow analysis or peripheral blood mononuclear cells. Finally, NGS technologies could be more useful than the classical "patient allele-specific PCR" because they can identify any possible clone emerging during the treatment or follow-up, even if different from that identified at diagnosis, thus predicting relapse. After all, the present available molecular approaches can move MRD from the bench side to the clinical practice.
Collapse
Affiliation(s)
- Sara Galimberti
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisa Genuardi
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Turin, Italy
| | - Francesco Mazziotta
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,GeNOMEC School of Doctorate, University of Siena, Siena, Italy
| | - Lorenzo Iovino
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Clinical and Translational Sciences School of Doctorate, University of Pisa, Pisa, Italy
| | - Fortunato Morabito
- Hematology Oncology Department, Augusta Victoria Hospital, East Jerusalem, Israel.,Biotechnology Research Unit, Cosenza, Italy
| | - Susanna Grassi
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,GeNOMEC School of Doctorate, University of Siena, Siena, Italy
| | - Elena Ciabatti
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesca Guerrini
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Mario Petrini
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
26
|
Affiliation(s)
- I Brian Greenwell
- a Division of Hematology and Medical Oncology , Medical University of South Carolina Hollings Cancer Center , Charleston , SC , USA
| | - Jonathon B Cohen
- b Department of Hematology and Medical Oncology , Emory University Winship Cancer Institute , Atlanta , GA , USA
| |
Collapse
|
27
|
Pott C, Brüggemann M, Ritgen M, van der Velden VHJ, van Dongen JJM, Kneba M. MRD Detection in B-Cell Non-Hodgkin Lymphomas Using Ig Gene Rearrangements and Chromosomal Translocations as Targets for Real-Time Quantitative PCR. Methods Mol Biol 2019; 1956:199-228. [PMID: 30779036 DOI: 10.1007/978-1-4939-9151-8_9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Minimal residual disease (MRD) diagnostics is of high clinical relevance in patients with indolent B-cell non-Hodgkin lymphomas (B-NHL) and serves as a surrogate parameter to evaluate treatment effectiveness and long-term prognosis. MRD diagnostics performed by real-time quantitative PCR (RQ-PCR) is still the gold standard and currently the most sensitive and the most broadly applied method in follicular lymphoma (FL) and mantle cell lymphoma (MCL). Alternatively, droplet digital PCR (ddPCR) can be used for MRD monitoring in multiple myeloma, mantle cell lymphoma, and follicular lymphoma with comparable sensitivity, accuracy, and reproducibility.The most broadly applicable MRD target in B-NHL is the junctional regions of the rearranged immunoglobulin heavy chain gene (IGHV). Chromosomal translocations like the t(14;18) translocation in FL and t(11;14) translocation in MCL can be used as MRD target in selected lymphoma subtypes. In patients with B-cell chronic lymphocytic leukemia, both flow-cytometry and RQ-PCR are equally suited for MRD assessment as long as a sensitivity of 10-4 shall be achieved.MRD diagnostics targeting the IGHV gene is complex and requires extensive knowledge and experience because the junctional regions of each lymphoma have to be identified before the patient-specific RQ-PCR assays can be designed for MRD monitoring. In addition, the presence and load of somatic hypermutation (SHM) within the rearranged IG heavy variable (IGHV) gene occurring as during B-cell development of germinal center and post-germinal center lymphomas may hamper appropriate primer binding leading to false-negative results. The translocations mentioned above have the advantage that consensus forward primers and probes, both placed in the breakpoint regions of chromosome 18 in FL and chromosome 11 in MCL, can be used in combination with a reverse primer placed in the IGH joining region of chromosome 14. RQ-PCR-based methods can reach a good sensitivity (≤10-4). This chapter provides all relevant background information and technical aspects for the complete laboratory process from detection of the clonal IGHV gene rearrangement and the chromosomal translocations at diagnosis to the actual MRD measurements in clinical follow-up samples of B-NHL. However, it should be noted that MRD diagnostics for clinical treatment protocols has to be accompanied by regular international quality control rounds to ensure the reproducibility and reliability of the MRD results.
Collapse
Affiliation(s)
- Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Monika Brüggemann
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Ritgen
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael Kneba
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
28
|
Abstract
The quantification of submicroscopic minimal residual disease (MRD) after therapy proved to have independent prognostic significance in many mature B-cell malignancies. With the advent of routine benchtop cytometers capable of simultaneously analyzing ≥4 colors and with improved standardization, flow cytometry has become the method of choice for MRD assessments in some lymphoma entities. Herein we describe general aspects of flow cytometric standardization. Chronic lymphocytic leukemia and multiple myeloma (MM) are used as examples to explain the technical standardization of flow cytometry for MRD detection according to EuroFlow strategies. MRD data acquisition and detailed analysis using a newly developed approach (so-called next generation flow, NGF) in MM is a particular focus of this chapter.
Collapse
|
29
|
Ruan J, Martin P, Christos P, Cerchietti L, Tam W, Shah B, Schuster SJ, Rodriguez A, Hyman D, Calvo-Vidal MN, Smith SM, Svoboda J, Furman RR, Coleman M, Leonard JP. Five-year follow-up of lenalidomide plus rituximab as initial treatment of mantle cell lymphoma. Blood 2018; 132:2016-2025. [PMID: 30181173 PMCID: PMC6634960 DOI: 10.1182/blood-2018-07-859769] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/24/2018] [Indexed: 01/30/2023] Open
Abstract
We report 5-year follow-up of a multicenter phase 2 study of lenalidomide plus rituximab (LR) as initial treatment of mantle cell lymphoma (MCL). The regimen includes induction and maintenance with the LR doublet. Treatment was continuous until progression, with optional discontinuation after 3 years. The median age of the 38 participants was 65 years, with MCL international prognostic index scores balanced among low, intermediate, and high risk (34%, 34%, and 32%, respectively). Twenty-seven (75%) of the 36 evaluable patients completed ≥3 years of study treatment. At a median follow-up of 64 months (range, 21-78), the 3-year progression-free survival (PFS) and overall survival (OS) were 80% and 90%, respectively, with 5-year estimated PFS and OS of 64% and 77%, respectively. During maintenance, hematologic adverse events (AEs) included asymptomatic grade 3 or 4 cytopenias (42% neutropenia, 5% thrombocytopenia, 3% anemia) and mostly grade 1 or 2 infections managed in the outpatient setting (45% upper respiratory infection, 21% urinary tract infection, 13% sinusitis, 11% cellulitis, 8% pneumonia). Nonhematologic AEs, such as constitutional and inflammatory symptoms, occurred at reduced frequency and intensity compared with induction. A peripheral blood minimal residual disease (MRD) assay (clonoSEQ) showed MRD-negative complete remission in 8 of 10 subjects who had completed ≥3 years of treatment and with available samples for analysis. With longer follow-up, LR continues to demonstrate durable responses and manageable safety as initial induction and maintenance therapy for MCL (ClinicalTrials.gov NCT01472562).
Collapse
Affiliation(s)
- Jia Ruan
- Division of Hematology and Medical Oncology
| | | | | | | | - Wayne Tam
- Department of Pathology and Laboratory Medicine, Meyer Cancer Center, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY
| | | | - Stephen J Schuster
- University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; and
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gressin R, Daguindau N, Tempescul A, Moreau A, Carras S, Tchernonog E, Schmitt A, Houot R, Dartigeas C, Pignon JM, Corm S, Banos A, Mounier C, Dupuis J, Macro M, Fleury J, Jardin F, Sarkozy C, Damaj G, Feugier P, Fornecker LM, Chabrot C, Dorvaux V, Bouadallah K, Amorin S, Garidi R, Voillat L, Joly B, Celigny PS, Morineau N, Moles MP, Zerazhi H, Fontan J, Arkam Y, Alexis M, Delwail V, Vilque JP, Ysebaert L, Le Gouill S, Callanan MB. A phase 2 study of rituximab, bendamustine, bortezomib and dexamethasone for first-line treatment of older patients with mantle cell lymphoma. Haematologica 2018; 104:138-146. [PMID: 30171024 PMCID: PMC6312036 DOI: 10.3324/haematol.2018.191429] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 08/23/2018] [Indexed: 01/30/2023] Open
Abstract
We present results of a prospective, multicenter, phase II study evaluating rituximab, bendamustine, bortezomib and dexamethasone as first-line treatment for patients with mantle cell lymphoma aged 65 years or older. A total of 74 patients were enrolled (median age, 73 years). Patients received a maximum of six cycles of treatment at 28-day intervals. The primary objective was to achieve an 18-month progression-free survival rate of 65% or higher. Secondary objectives were to evaluate toxicity and the prognostic impact of mantle cell lymphoma prognostic index, Ki67 expression, [18F]fluorodeoxyglucose-positron emission tomography and molecular minimal residual disease, in peripheral blood or bone marrow. With a median follow-up of 52 months, the 24-month progression-free survival rate was 70%, hence the primary objective was reached. After six cycles of treatment, 91% (54/59) of responding patients were analyzed for peripheral blood residual disease and 87% of these (47/54) were negative. Four-year overall survival rates of the patients who did not have or had detectable molecular residual disease in the blood at completion of treatment were 86.6% and 28.6%, respectively (P<0.0001). Neither the mantle cell lymphoma index, nor fluorodeoxyglucose-positron emission tomography nor Ki67 positivity (cut off of ≥30%) showed a prognostic impact for survival. Hematologic grade 3-4 toxicities were mainly neutropenia (51%), thrombocytopenia (35%) and lymphopenia (65%). Grade 3-4 non-hematologic toxicities were mainly fatigue (18.5%), neuropathy (15%) and infections. In conclusion, the tested treatment regimen is active as frontline therapy in older patients with mantle cell lymphoma, with manageable toxicity. Minimal residual disease status after induction could serve as an early predictor of survival in mantle cell lymphoma. ClinicalTrials.gov: NCT 01457144.
Collapse
Affiliation(s)
- Rémy Gressin
- Onco-Hematology Department, Grenoble University Hospital .,INSERM 1209, CNRS UMR 5309, Faculté de Médecine, Université Grenoble-Alpes, Institute for Advanced Biosciences, Grenoble
| | | | | | - Anne Moreau
- Pathology Department, Nantes University Hospital
| | - Sylvain Carras
- Onco-Hematology Department, Grenoble University Hospital
| | | | - Anna Schmitt
- Hematology Department, Cancer Institute Bergonie Bordeaux
| | - Roch Houot
- Hematology Department, Rennes University Hospital
| | | | | | - Selim Corm
- Hematology Department, Chambery Hospital
| | | | | | - Jehan Dupuis
- Lymphoid Malignancies Unit, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil
| | | | - Joel Fleury
- Hematology Department, Clermont-Ferrand Cancer Institute
| | | | - Clementine Sarkozy
- Hematology Department, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud. INSERM 1052
| | - Ghandi Damaj
- Hematology Department, Amiens University Hospital
| | | | | | - Cecile Chabrot
- Hematology Department, University Clermont-Ferrand Hospital
| | | | | | - Sandy Amorin
- Hematology Department, University Hospital Paris Saint-Louis
| | - Reda Garidi
- Hematology Department, Saint Quentin Hospital
| | | | | | | | | | | | | | - Jean Fontan
- Hematology Department, Besançon University Hospital
| | | | | | - Vincent Delwail
- Onco-Hematology Department, University Hospital Poitiers and INSERM, CIC 1402, Poitiers University
| | | | | | | | - Mary B Callanan
- INSERM 1209, CNRS UMR 5309, Faculté de Médecine, Université Grenoble-Alpes, Institute for Advanced Biosciences, Grenoble .,Unit for Innovation in Genetics and Epigenetics in Oncology, Dijon University Hospital, France
| | | |
Collapse
|
31
|
Tam CS, Anderson MA, Pott C, Agarwal R, Handunnetti S, Hicks RJ, Burbury K, Turner G, Di Iulio J, Bressel M, Westerman D, Lade S, Dreyling M, Dawson SJ, Dawson MA, Seymour JF, Roberts AW. Ibrutinib plus Venetoclax for the Treatment of Mantle-Cell Lymphoma. N Engl J Med 2018; 378:1211-1223. [PMID: 29590547 DOI: 10.1056/nejmoa1715519] [Citation(s) in RCA: 300] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Both the BTK inhibitor ibrutinib and the BCL2 inhibitor venetoclax are active as monotherapy in the treatment of mantle-cell lymphoma. Complete response rates of 21% have been observed for each agent when administered as long-term continuous therapy. Preclinical models predict synergy in combination. METHODS We conducted a single-group, phase 2 study of daily oral ibrutinib and venetoclax in patients, as compared with historical controls. Patients commenced ibrutinib monotherapy at a dose of 560 mg per day. After 4 weeks, venetoclax was added in stepwise, weekly increasing doses to 400 mg per day. Both drugs were continued until progression or an unacceptable level of adverse events. The primary end point was the rate of complete response at week 16. Minimal residual disease (MRD) was assessed by flow cytometry in bone marrow and by allele-specific oligonucleotide-polymerase chain reaction (ASO-PCR) in blood. RESULTS The study included 24 patients with relapsed or refractory mantle-cell lymphoma (23 patients) or previously untreated mantle-cell lymphoma (1 patient). Patients were 47 to 81 years of age, and the number of previous treatments ranged from none to six. Half the patients had aberrations of TP53, and 75% had a high-risk prognostic score. The complete response rate according to computed tomography at week 16 was 42%, which was higher than the historical result of 9% at this time point with ibrutinib monotherapy (P<0.001). The rate of complete response as assessed by positron-emission tomography was 62% at week 16 and 71% overall. MRD clearance was confirmed by flow cytometry in 67% of the patients and by ASO-PCR in 38%. In a time-to-event analysis, 78% of the patients with a response were estimated to have an ongoing response at 15 months. The tumor lysis syndrome occurred in 2 patients. Common side effects were generally low grade and included diarrhea (in 83% of the patients), fatigue (in 75%), and nausea or vomiting (in 71%). CONCLUSIONS In this study involving historical controls, dual targeting of BTK and BCL2 with ibrutinib and venetoclax was consistent with improved outcomes in patients with mantle-cell lymphoma who had been predicted to have poor outcomes with current therapy. (Funded by Janssen and others; AIM ClinicalTrials.gov number, NCT02471391 .).
Collapse
Affiliation(s)
- Constantine S Tam
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Mary Ann Anderson
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Christiane Pott
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Rishu Agarwal
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Sasanka Handunnetti
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Rodney J Hicks
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Kate Burbury
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Gillian Turner
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Juliana Di Iulio
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Mathias Bressel
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - David Westerman
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Stephen Lade
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Martin Dreyling
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Sarah-Jane Dawson
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Mark A Dawson
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - John F Seymour
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| | - Andrew W Roberts
- From the Peter MacCallum Cancer Centre, Melbourne, VIC (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., G.T., J.D.I., M.B., D.W., S.L., S.-J.D., M.A.D., J.F.S., A.W.R.), and the Victorian Comprehensive Cancer Centre (C.S.T., M.A.A., R.A., S.H., R.J.H., K.B., D.W., S.-J.D., M.A.D., J.F.S., A.W.R.), the Faculty of Medicine (C.S.T., R.J.H., S.-J.D., M.A.D., J.F.S., A.W.R.) and Centre for Cancer Research (S.-J.D., M.A.D.), University of Melbourne, the Department of Clinical Haematology and Bone Marrow Transplantation, the Royal Melbourne Hospital (C.S.T., M.A.A., K.B., J.F.S., A.W.R.), and the Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research (M.A.A., A.W.R.), Parkville, VIC - all in Australia; and the University Hospital of Schleswig-Holstein, Kiel (C.P.), and Klinikum der Universität, Ludwig-Maximilian University of Munich, Munich (M.D.) - both in Germany
| |
Collapse
|
32
|
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
|
33
|
Cohen JB, Zain JM, Kahl BS. Current Approaches to Mantle Cell Lymphoma: Diagnosis, Prognosis, and Therapies. Am Soc Clin Oncol Educ Book 2017; 37:512-525. [PMID: 28561694 DOI: 10.1200/edbk_175448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mantle cell lymphoma (MCL) is a unique lymphoma subtype, both biologically and clinically. Virtually all cases are characterized by a common genetic lesion, t(11;14), resulting in overexpression of cyclin D1. The clinical course is moderately aggressive, and the disease is considered incurable. Considerable biologic and clinical heterogeneity exists, with some patients experiencing a rapidly progressive course, while others have disease that is readily managed. New tools exist for risk stratification and may allow for a more personalized approach in the future. Landmark studies have been completed in recent years and outcomes appear to be improving. Randomized clinical trials have clarified the role of high-dose cytarabine (Ara-C) for younger patients and have demonstrated a role for maintenance rituximab therapy. Multiple areas of uncertainty remain, however, and are the focus of ongoing research. This review focuses on (1) strategies to differentiate between aggressive and less aggressive cases, (2) understanding who should receive hematopoietic stem cell transplantation, and (3) the role for maintenance therapy in MCL.
Collapse
Affiliation(s)
- Jonathon B Cohen
- From Emory University, Atlanta, GA; City of Hope, Duarte, CA; Washington University School of Medicine, St. Louis, MO
| | - Jasmine M Zain
- From Emory University, Atlanta, GA; City of Hope, Duarte, CA; Washington University School of Medicine, St. Louis, MO
| | - Brad S Kahl
- From Emory University, Atlanta, GA; City of Hope, Duarte, CA; Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
34
|
Herrera AF, Armand P. Minimal Residual Disease Assessment in Lymphoma: Methods and Applications. J Clin Oncol 2017; 35:3877-3887. [PMID: 28933999 PMCID: PMC5707209 DOI: 10.1200/jco.2017.74.5281] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Standard methods for disease response assessment in patients with lymphoma, including positron emission tomography and computed tomography scans, are imperfect. In other hematologic malignancies, particularly leukemias, the ability to detect minimal residual disease (MRD) is increasingly influencing treatment paradigms. However, in many subtypes of lymphoma, the application of MRD assessment techniques, like flow cytometry or polymerase chain reaction-based methods, has been challenging because of the absence of readily detected circulating disease or canonic chromosomal translocations. Newer MRD detection methods that use next-generation sequencing have yielded promising results in a number of lymphoma subtypes, fueling the hope that MRD detection may soon be applicable in clinical practice for most patients with lymphoma. MRD assessment can provide real-time information about tumor burden and response to therapy, noninvasive genomic profiling, and monitoring of clonal dynamics, allowing for many possible applications that could significantly affect the care of patients with lymphoma. Further validation of MRD assessment methods, including the incorporation of MRD assessment into clinical trials in patients with lymphoma, will be critical to determine how best to deploy MRD testing in routine practice and whether MRD assessment can ultimately bring us closer to the goal of personalized lymphoma care. In this review article, we describe the methods available for detecting MRD in patients with lymphoma and their relative advantages and disadvantages. We discuss preliminary results supporting the potential applications for MRD testing in the care of patients with lymphoma and strategies for including MRD assessment in lymphoma clinical trials.
Collapse
Affiliation(s)
- Alex F. Herrera
- Alex F. Herrera, City of Hope National Medical Center, Duarte, CA; and Philippe Armand, Dana-Farber Cancer Institute, Boston, MA
| | - Philippe Armand
- Alex F. Herrera, City of Hope National Medical Center, Duarte, CA; and Philippe Armand, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
35
|
Riedell PA, Bishop MR. Post-autologous transplant maintenance therapies in lymphoma: current state and future directions. Bone Marrow Transplant 2017; 53:11-21. [PMID: 28967896 DOI: 10.1038/bmt.2017.196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 01/11/2023]
Abstract
Disease relapse following high-dose chemotherapy and autologous stem cell transplant (ASCT) remains the principal cause of mortality in patients with relapsed or refractory lymphomas. In an effort to prevent post-ASCT relapse, a number of studies have evaluated the role of maintenance therapy with varying success. In diffuse large B-cell lymphoma, studies evaluating maintenance rituximab (MR) following ASCT failed to demonstrate improved outcomes. In follicular lymphoma, MR was associated with an improvement in PFS; however, no overall survival (OS) benefit was noted. Emerging data evaluating MR in mantle cell lymphoma (MCL) have demonstrated improvements in PFS, although a consistent improvement in OS has yet to be demonstrated. Given the aggressive and incurable nature of MCL, it is prudent for practitioners to weigh the risks and benefits of MR in the post-ASCT setting. Similarly, post-ASCT maintenance therapy with brentuximab vedotin in Hodgkin lymphoma, has led to improved PFS and may be considered in those with a high risk of relapse. Ongoing clinical studies evaluating a multitude of novel maintenance therapies are crucial to the efforts of further defining and optimizing the role of post-transplant maintenance therapy in lymphoma.
Collapse
Affiliation(s)
- P A Riedell
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - M R Bishop
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
36
|
Ferrero S, Dreyling M. Minimal residual disease in mantle cell lymphoma: are we ready for a personalized treatment approach? Haematologica 2017; 102:1133-1136. [PMID: 28655809 PMCID: PMC5566011 DOI: 10.3324/haematol.2017.167627] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Simone Ferrero
- Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Italy
| | - Martin Dreyling
- Department of Medicine III, Hospital of the University LMU München, Germany
| |
Collapse
|
37
|
Martin P, Ghione P, Dreyling M. Mantle cell lymphoma – Current standards of care and future directions. Cancer Treat Rev 2017. [DOI: 10.1016/j.ctrv.2017.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
38
|
High-throughput sequencing for noninvasive disease detection in hematologic malignancies. Blood 2017; 130:440-452. [PMID: 28600337 DOI: 10.1182/blood-2017-03-735639] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/25/2017] [Indexed: 12/20/2022] Open
Abstract
Noninvasive monitoring of minimal residual disease (MRD) has led to significant advances in personalized management of patients with hematologic malignancies. Improved therapeutic options and prolonged survival have further increased the need for sensitive tumor assessment that can inform treatment decisions and patient outcomes. At diagnosis or relapse of most hematologic neoplasms, malignant cells are often easily accessible in the blood as circulating tumor cells (CTCs), making them ideal targets to noninvasively profile the molecular features of each patient. In other cancer types, CTCs are generally rare and noninvasive molecular detection relies on circulating tumor DNA (ctDNA) shed from tumor deposits into circulation. The ability to precisely detect and quantify CTCs and ctDNA could minimize invasive procedures and improve prediction of clinical outcomes. Technical advances in MRD detection methods in recent years have led to reduced costs and increased sensitivity, specificity, and applicability. Among currently available tests, high-throughput sequencing (HTS)-based approaches are increasingly attractive for noninvasive molecular testing. HTS-based methods can simultaneously identify multiple genetic markers with high sensitivity and specificity without individual optimization. In this review, we present an overview of techniques used for noninvasive molecular disease detection in selected myeloid and lymphoid neoplasms, with a focus on the current and future role of HTS-based assays.
Collapse
|
39
|
Kahl BS, Dreyling M, Gordon LI, Quintanilla-Martinez L, Sotomayor EM. Recent advances and future directions in mantle cell lymphoma research: report of the 2016 mantle cell lymphoma consortium workshop. Leuk Lymphoma 2017; 58:1561-1569. [PMID: 28140709 DOI: 10.1080/10428194.2017.1283036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma typically associated with the t(11;14) chromosomal translocation, resulting in overexpression of cyclin D1. Although MCL is associated with clinical heterogeneity, outcomes are generally poor and no standard treatment has been established. However, the recent approval of ibrutinib provides a new therapeutic option. Moreover, recent clinical trials have provided new perspectives on the relative efficacy and safety of various approaches for both transplant-eligible and transplant-ineligible patients. Multiple novel strategies are being evaluated in the treatment of MCL, including both targeted agents and cellular immunotherapies. At the Lymphoma Research Foundation's 12th MCL Workshop, researchers gathered to discuss research findings, clinical trial results, and future directions related to MCL, its biology, and its treatment. This manuscript, which includes a summary of each presentation, aims to review recent findings in MCL research and highlight potential areas for future study.
Collapse
Affiliation(s)
- Brad S Kahl
- a Department of Medicine, Washington University in Saint Louis , St. Louis , MO , USA
| | - Martin Dreyling
- b Department of Medicine, University of Munich-Grosshadern , Munich , Germany
| | - Leo I Gordon
- c Department of Medicine, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center , Chicago , IL , USA
| | | | - Eduardo M Sotomayor
- e Department of Medicine, George Washington University , Washington DC , USA
| |
Collapse
|
40
|
Kolstad A, Pedersen LB, Eskelund CW, Husby S, Grønbæk K, Jerkeman M, Laurell A, Räty R, Elonen E, Andersen NS, Brown PD, Kimby E, Bentzen H, Sundström C, Ehinger M, Karjalainen-Lindsberg ML, Delabie J, Ralfkiær E, Fagerli UM, Nilsson-Ehle H, Lauritzsen GF, Kuittinen O, Niemann C, Geisler CH. Molecular Monitoring after Autologous Stem Cell Transplantation and Preemptive Rituximab Treatment of Molecular Relapse; Results from the Nordic Mantle Cell Lymphoma Studies (MCL2 and MCL3) with Median Follow-Up of 8.5 Years. Biol Blood Marrow Transplant 2016; 23:428-435. [PMID: 28039078 DOI: 10.1016/j.bbmt.2016.12.634] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/23/2016] [Indexed: 11/18/2022]
Abstract
The main objectives of the present study were to monitor minimal residual disease (MRD) in the bone marrow of patients with mantle cell lymphoma (MCL) to predict clinical relapse and guide preemptive treatment with rituximab. Among the patients enrolled in 2 prospective trials by the Nordic Lymphoma Group, 183 who had completed autologous stem cell transplantation (ASCT) and in whom an MRD marker had been obtained were included in our analysis. Fresh samples of bone marrow were analyzed for MRD by a combined standard nested and quantitative real-time PCR assay for Bcl-1/immunoglobulin heavy chain gene (IgH) and clonal IgH rearrangements. Significantly shorter progression-free survival (PFS) and overall survival (OS) was demonstrated for patients who were MRD positive pre-ASCT (54 patients) or in the first analysis post-ASCT (23 patients). The median PFS was only 20 months in those who were MRD-positive in the first sample post-ASCT, compared with 142 months in the MRD-negative group (P < .0001). OS was 75% at 10 years and median not reached in the MRD-negative group, compared with only 35 months in the MRD-positive group (P < .0001). Of the 86 patients (47%) who remained in continuous molecular remission, 73% were still in clinical remission after 10 years. For all patients, the median time from ASCT to first molecular relapse was 55 months, with a continuous occurrence of late molecular relapses. Fifty-eight patients who experienced MRD relapse received rituximab as preemptive treatment on 1 or more occasions, and in this group, the median time from first molecular relapse to clinical relapse was 55 months. In most cases, rituximab converted patients to MRD negativity (87%), but many patients became MRD-positive again later during follow-up (69%). By multivariate analysis, high-risk Mantle Cell Lymphoma International Prognostic Index score and positive MRD status pre-ASCT predicted early molecular relapse. In conclusion, preemptive rituximab treatment converts patients to MRD negativity and likely postpones clinical relapse. Molecular monitoring offers an opportunity to select some patients for therapeutic intervention and to avoid unnecessary treatment in others. MRD-positive patients in the first analysis post-ASCT have a dismal prognosis and thus are in need of novel strategies.
Collapse
Affiliation(s)
- Arne Kolstad
- Department of Oncology, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.
| | - Lone Bredo Pedersen
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian W Eskelund
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Simon Husby
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kirsten Grønbæk
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mats Jerkeman
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Anna Laurell
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Riikka Räty
- Department of Hematology and Oncology, Helsinki University Central Hospital, Helsinki, Finland
| | - Erkki Elonen
- Department of Hematology and Oncology, Helsinki University Central Hospital, Helsinki, Finland
| | | | - Peter deNully Brown
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Eva Kimby
- Department of Hematology, Karolinska Institute, Stockholm, Sweden
| | - Hans Bentzen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Mats Ehinger
- Department of Pathology, Skåne University Hospital, Lund, Sweden
| | | | - Jan Delabie
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Elisabeth Ralfkiær
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Herman Nilsson-Ehle
- Department of Hematology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Outi Kuittinen
- Department of Oncology, Oulu University Hospital, Oulu, Finland
| | - Carsten Niemann
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | |
Collapse
|
41
|
Hoster E, Pott C. Minimal residual disease in mantle cell lymphoma: insights into biology and impact on treatment. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:437-445. [PMID: 27913513 PMCID: PMC6142459 DOI: 10.1182/asheducation-2016.1.437] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Despite the recent substantial improvement of clinical outcome in mantle cell lymphoma (MCL), resistance to immunochemotherapy and common relapses are challenges for long-term tumor control. The assessment of minimal residual disease (MRD) by real-time quantitative polymerase chain reaction has emerged as a widely feasible and standardized tool for direct assessment of therapy-induced reduction of tumor burden and regrowth after cytotoxic treatment in MCL, with much improved sensitivity compared with conventional staging procedures. Several studies have shown that intensification of initial treatment, which has resulted in improved clinical outcome, is immediately reflected in higher molecular remission rates; they have also shown that high-dose consolidation might not be able to compensate for less intensive induction regimens. Persistence or reappearance of MRD in clinical remission proved to be highly predictive for imminent clinical relapse associated with shorter overall survival. Therefore, the investigation of novel MRD-guided treatment strategies aimed at early eradication of MRD and pre-emptive treatment of molecular relapse seems warranted. Furthermore, the integration of MRD assessment into clinical response criteria could result in a more specific and potentially earlier end point for treatment efficacy. New technical developments such as high-throughput sequencing will further enhance the wide applicability of MRD detection in MCL.
Collapse
Affiliation(s)
- Eva Hoster
- Department of Internal Medicine III, University Hospital Munich, and Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-University Munich, Munich, Germany; and
| | - Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
42
|
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
|
43
|
Sato M, Yamazaki J, Goto-Koshino Y, Setoguchi A, Takahashi M, Baba K, Fujino Y, Ohno K, Tsujimoto H. Minimal residual disease in canine lymphoma: An objective marker to assess tumour cell burden in remission. Vet J 2016; 215:38-42. [DOI: 10.1016/j.tvjl.2016.05.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 05/12/2016] [Accepted: 05/24/2016] [Indexed: 10/21/2022]
|
44
|
Hermine O, Hoster E, Walewski J, Bosly A, Stilgenbauer S, Thieblemont C, Szymczyk M, Bouabdallah R, Kneba M, Hallek M, Salles G, Feugier P, Ribrag V, Birkmann J, Forstpointner R, Haioun C, Hänel M, Casasnovas RO, Finke J, Peter N, Bouabdallah K, Sebban C, Fischer T, Dührsen U, Metzner B, Maschmeyer G, Kanz L, Schmidt C, Delarue R, Brousse N, Klapper W, Macintyre E, Delfau-Larue MH, Pott C, Hiddemann W, Unterhalt M, Dreyling M. Addition of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation in patients aged 65 years or younger with mantle cell lymphoma (MCL Younger): a randomised, open-label, phase 3 trial of the European Mantle Cell Lymphoma Network. Lancet 2016; 388:565-75. [PMID: 27313086 DOI: 10.1016/s0140-6736(16)00739-x] [Citation(s) in RCA: 274] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Mantle cell lymphoma is characterised by a poor long-term prognosis. The European Mantle Cell Lymphoma Network aimed to investigate whether the introduction of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation (ASCT) improves outcome. METHODS This randomised, open-label, parallel-group, phase 3 trial was done in 128 haemato-oncological hospital departments or private practices in Germany, France, Belgium, and Poland. Patients aged 65 years or younger with untreated stage II-IV mantle cell lymphoma were centrally randomised (1:1), with computer-assisted random block selection, to receive either six courses of R-CHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone) followed by myeloablative radiochemotherapy and ASCT (control group), or six courses of alternating R-CHOP or R-DHAP (rituximab plus dexamethasone, high-dose cytarabine, and cisplatin) followed by a high-dose cytarabine-containing conditioning regimen and ASCT (cytarabine group). Patients were stratified by study group and international prognostic index. The primary outcome was time to treatment failure from randomisation to stable disease after at least four induction cycles, progression, or death from any cause. Patients with stage II-IV mantle cell lymphoma were included in the primary analysis if treatment was started according to randomisation. For safety analyses, patients were assessed according to the treatment actually started. This study is registered with ClinicalTrials.gov, number NCT00209222. FINDINGS Of 497 patients (median age 55 years [IQR 49-60]) randomised from July 20, 2004, to March 18, 2010, 234 of 249 in the control group and 232 of 248 in the cytarabine group were included in the primary analysis. After a median follow-up of 6.1 years (95% CI 5.4-6.4), time to treatment failure was significantly longer in the cytarabine group (median 9.1 years [95% CI 6.3-not reached], 5 year rate 65% [95% CI 57-71]) than in the control group (3.9 years [3.2-4.4], 40% [33-46]; hazard ratio 0.56; p=0.038). During induction immunochemotherapy, patients who received high-dose cytarabine had increased grade 3 or 4 haematological toxicity (haemoglobin 71 [29%] of 241m vs 19 [8%] of 227 controls; platelets 176 [73%] of 240 vs 21 [9%] of 225), grade 3 or 4 febrile neutropenia (39 [17%] of 230 vs 19 [8%] of 224), and grade 1 or 2 renal toxicity (creatinine 102 [43%] of 236 vs 22 [10%] of 224). The number of ASCT-related deaths was similar (eight [3.4%]) in both groups. INTERPRETATION Immunochemotherapy containing high-dose cytarabine followed by ASCT should be considered standard of care in patients aged 65 years or younger with mantle cell lymphoma. FUNDING European Commission, Lymphoma Research Foundation, and Roche.
Collapse
Affiliation(s)
- Olivier Hermine
- Department of Hematology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, University Paris Descartes, Paris, France; INSERM U1163 and CNRS 8254, Imagine Institute, Université Sorbonne Paris Cité, Paris, France
| | - Eva Hoster
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital Munich, Munich, Germany; Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jan Walewski
- Department of Lymphoid Malignancies, The Maria Skłodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | - André Bosly
- Louvain Catholic University, Namur University Hospital, Namur, Belgium
| | | | | | - Michal Szymczyk
- Department of Lymphoid Malignancies, The Maria Skłodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | - Reda Bouabdallah
- Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Michael Kneba
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
| | - Gilles Salles
- Hospices Civils de Lyon, Université Claude Bernard, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Pierre Feugier
- Nancy University Hospital, Department of Hematology, Nancy, France
| | | | - Josef Birkmann
- Department of Oncology, Paracelsus Medical University, Nuremberg, Germany
| | - Roswitha Forstpointner
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital Munich, Munich, Germany
| | - Corinne Haioun
- Lymphoid Malignancies Unit, Hospital Henri Mondor, Créteil, France
| | - Mathias Hänel
- Department of Internal Medicine III, Klinikum Chemnitz, Germany
| | | | - Jürgen Finke
- Department of Hematology and Oncology, University Hospital Freiburg, Freiburg, Germany
| | - Norma Peter
- Second Medical Department, Carl-Thiem-Klinikum Cottbus, Cottbus, Germany
| | - Kamal Bouabdallah
- Department of Hematology and Cell Therapy, University Hospital of Bordeaux, Pessac, France
| | | | - Thomas Fischer
- Department of Hematology and Oncology, University Hospital Magdeburg, Magdeburg, Germany
| | - Ulrich Dührsen
- Department of Hematology, University Hospital Essen, Essen, Germany
| | - Bernd Metzner
- Department of Hematology and Oncology, University Hospital Oldenburg, Oldenburg, Germany
| | - Georg Maschmeyer
- Department of Hematology, Oncology and Palliative Medicine, Ernst-von-Bergmann-Hospital, Potsdam, Germany
| | - Lothar Kanz
- Department of Medicine II, University of Tübingen, Tübingen, Germany
| | - Christian Schmidt
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital Munich, Munich, Germany
| | - Richard Delarue
- Department of Hematology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, University Paris Descartes, Paris, France
| | - Nicole Brousse
- Department of Pathology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, University Paris Descartes, Paris, France
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section, and Lymph Node Registry, University of Kiel, Kiel, Germany
| | - Elizabeth Macintyre
- Department of Biological Hematology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, University Paris Descartes, Paris, France
| | - Marie-Hélène Delfau-Larue
- Department of Biological Hematology and Immunology, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, INSERM U955, UPEC University, Creteil, France
| | - Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Wolfgang Hiddemann
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital Munich, Munich, Germany
| | - Michael Unterhalt
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital Munich, Munich, Germany
| | - Martin Dreyling
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital Munich, Munich, Germany.
| |
Collapse
|
45
|
Hiyoshi-Kanemoto S, Goto-Koshino Y, Fukushima K, Takahashi M, Kanemoto H, Uchida K, Fujino Y, Ohno K, Tsujimoto H. Detection of circulating tumor cells using GeneScan analysis for antigen receptor gene rearrangements in canine lymphoma patients. J Vet Med Sci 2016; 78:877-81. [PMID: 26888583 PMCID: PMC4905848 DOI: 10.1292/jvms.15-0433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The presence of circulating tumor cells (CTCs) serves as a prognostic marker and
indicator of disease relapse, as well as a means of evaluating treatment efficacy in human
and canine lymphoma patients. As an extension of our previous study for the construction
of clinically useful GeneScan system, we utilized the GeneScan system for detecting CTCs
in canine lymphoma patients. Samples from the primary lesion and peripheral blood
mononuclear cells (PBMCs) were obtained from 32 dogs with lymphoma at initial diagnosis.
All samples were subjected to polymerase chain reaction (PCR) for antigen receptor gene
rearrangements (PARR) followed by GeneScan analysis. Common clonal rearrangements with
identical amplified fragments were detected in both the primary lesion and PBMCs in 19 of
the 32 dogs (59.4%). However, the detection rate of CTCs varied among the anatomical
classification of lymphoma studied. GeneScan analysis following PARR would facilitate
studies on determining the clinical significance of CTCs in canine lymphoma patients.
Collapse
Affiliation(s)
- Saaya Hiyoshi-Kanemoto
- Department of Veterinary Internal Medicine, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cowan AJ, Stevenson PA, Cassaday RD, Graf SA, Fromm JR, Wu D, Holmberg LA, Till BG, Chauncey TR, Smith SD, Philip M, Orozco JJ, Shustov AR, Green DJ, Libby EN, Bensinger WI, Shadman M, Maloney DG, Press OW, Gopal AK. Pretransplantation Minimal Residual Disease Predicts Survival in Patients with Mantle Cell Lymphoma Undergoing Autologous Stem Cell Transplantation in Complete Remission. Biol Blood Marrow Transplant 2016; 22:380-385. [PMID: 26348890 PMCID: PMC4716882 DOI: 10.1016/j.bbmt.2015.08.035] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/31/2015] [Indexed: 01/22/2023]
Abstract
Autologous stem cell transplantation (ASCT) is standard therapy for mantle cell lymphoma (MCL) in remission after induction chemotherapy, with the best results for patients in complete remission (CR). We hypothesized that evaluation of minimal residual disease (MRD) before ASCT could further stratify outcomes for these patients. Patients with MCL who underwent ASCT in clinical CR between 1996 and 2011 with pretransplantation MRD testing were eligible. Presence of a clonal IgH rearrangement, t(11; 14) by PCR or positive flow cytometry from blood or bone marrow, was considered positive. An adjusted proportional hazards model for associations with progression-free (PFS) and overall survival (OS) was performed. Of 75 MCL patients in CR, 8 (11%) were MRD positive. MRD positivity was associated with shorter OS and PFS. The median OS for MRD-negative patients was not reached, with 82% survival at 5 years, whereas for the MRD-positive patients, median OS was 3.01 years (hazard ratio [HR], 4.04; P = .009), with a median follow-up of 5.1 years. The median PFS for MRD-negative patients was not reached with 75% PFS at 5 years, whereas for MRD-positive patients, it was 2.38 years (HR, 3.69; P = .002). MRD positivity is independently associated with poor outcomes after ASCT for MCL patients in CR.
Collapse
Affiliation(s)
- Andrew J Cowan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Philip A Stevenson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Ryan D Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Solomon A Graf
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Jonathan R Fromm
- Marrow Transplant Unit, Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - David Wu
- Marrow Transplant Unit, Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Leona A Holmberg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Thomas R Chauncey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington; VA Puget Sound Health Care System, Seattle, Washington
| | - Stephen D Smith
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Mary Philip
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Johnnie J Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Andrei R Shustov
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Edward N Libby
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - William I Bensinger
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Mazyar Shadman
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - David G Maloney
- Transplantation Biology Department, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Ajay K Gopal
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington.
| |
Collapse
|
47
|
Dreyling M, Ferrero S. The role of targeted treatment in mantle cell lymphoma: is transplant dead or alive? Haematologica 2016; 101:104-14. [PMID: 26830211 PMCID: PMC4938333 DOI: 10.3324/haematol.2014.119115] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/17/2015] [Indexed: 12/16/2022] Open
Abstract
Based on the profound biological insights of the last years into the molecular pathogenesis of mantle cell lymphoma and the clinical introduction of new targeted drugs, with high efficacy and a good safety profile, the therapeutic scenario for this tumor has been shown to be thoroughly favourable. No longer characterized by a uniformly dismal prognosis, mantle cell lymphoma has been revealed as a spectrum of different diseases, ranging from very indolent cases to highly aggressive and refractory ones. Thus, there is an urgent need to adapt therapy to accommodate the diverse presentations of the disease. High-dose chemotherapy, followed by autologous stem cell transplantation is the current standard of care for younger patients, generally providing high responses and long survival rates, but hampered by acute and long-term toxicity. In addition, some patients may be overtreated, while others could benefit from targeted approaches, based on the new, molecular-directed compounds. Such a personalized treatment based on the specific characteristics of individual patients may be guided by validated prognostic tools, such as the Mantle Cell Lymphoma International Prognostic Index and the Ki-67 Proliferative Index, as well as by early predictors of treatment response, like minimal residual disease analysis. Moreover, mutation screening of distinctive genomic alterations may provide new, predictive biomarkers, with an additional impact on clinical practice. Only after tailoring treatment according to the clinical and biological heterogeneity of the disease the role of transplantation and modern therapeutic options will be redefined in mantle cell lymphoma.
Collapse
Affiliation(s)
- Martin Dreyling
- Department of Medicine III, Hospital of the University LMU München, Germany
| | - Simone Ferrero
- Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Italy
| |
Collapse
|
48
|
Cheminant M, Derrieux C, Touzart A, Schmit S, Grenier A, Trinquand A, Delfau-Larue MH, Lhermitte L, Thieblemont C, Ribrag V, Cheze S, Sanhes L, Jardin F, Lefrère F, Delarue R, Hoster E, Dreyling M, Asnafi V, Hermine O, Macintyre E. Minimal residual disease monitoring by 8-color flow cytometry in mantle cell lymphoma: an EU-MCL and LYSA study. Haematologica 2015; 101:336-45. [PMID: 26703963 DOI: 10.3324/haematol.2015.134957] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/18/2015] [Indexed: 11/09/2022] Open
Abstract
Quantification of minimal residual disease may guide therapeutic strategies in mantle cell lymphoma. While multiparameter flow cytometry is used for diagnosis, the gold standard method for minimal residual disease analysis is real-time quantitative polymerase chain reaction (RQ-PCR). In this European Mantle Cell Lymphoma network (EU-MCL) pilot study, we compared flow cytometry with RQ-PCR for minimal residual disease detection. Of 113 patients with at least one minimal residual disease sample, RQ-PCR was applicable in 97 (86%). A total of 284 minimal residual disease samples from 61 patients were analyzed in parallel by flow cytometry and RQ-PCR. A single, 8-color, 10-antibody flow cytometry tube allowed specific minimal residual disease assessment in all patients, with a robust sensitivity of 0.01%. Using this cut-off level, the true-positive-rate of flow cytometry with respect to RQ-PCR was 80%, whereas the true-negative-rate was 92%. As expected, RQ-PCR frequently detected positivity below this 0.01% threshold, which is insufficiently sensitive for prognostic evaluation and would ideally be replaced with robust quantification down to a 0.001% (10-5) threshold. In 10 relapsing patients, the transition from negative to positive by RQ-PCR (median 22.5 months before relapse) nearly always preceded transition by flow cytometry (4.5 months), but transition to RQ-PCR positivity above 0.01% (5 months) was simultaneous. Pre-emptive rituximab treatment of 2 patients at minimal residual disease relapse allowed re-establishment of molecular and phenotypic complete remission. Flow cytometry minimal residual disease is a complementary approach to RQ-PCR and a promising tool in individual mantle cell lymphoma therapeutic management. (clinicaltrials identifiers: 00209209 and 00209222).
Collapse
Affiliation(s)
- Morgane Cheminant
- Biological Hematology, Paris Descartes - Sorbonne Paris Cité University, Institut Necker-Enfants Malades, AP-HP, France Clinical Hematology, Paris Descartes - Sorbonne Paris Cité University, IMAGINE Institut, Necker Hospital, AP-HP, France
| | - Coralie Derrieux
- Biological Hematology, Paris Descartes - Sorbonne Paris Cité University, Institut Necker-Enfants Malades, AP-HP, France
| | - Aurore Touzart
- Biological Hematology, Paris Descartes - Sorbonne Paris Cité University, Institut Necker-Enfants Malades, AP-HP, France
| | - Stéphanie Schmit
- Biological Hematology, Paris Descartes - Sorbonne Paris Cité University, Institut Necker-Enfants Malades, AP-HP, France
| | - Adrien Grenier
- Biological Hematology, Paris Descartes - Sorbonne Paris Cité University, Institut Necker-Enfants Malades, AP-HP, France
| | - Amélie Trinquand
- Biological Hematology, Paris Descartes - Sorbonne Paris Cité University, Institut Necker-Enfants Malades, AP-HP, France
| | | | - Ludovic Lhermitte
- Biological Hematology, Paris Descartes - Sorbonne Paris Cité University, Institut Necker-Enfants Malades, AP-HP, France
| | - Catherine Thieblemont
- Hemato-Oncology, Saint-Louis Hospital, APHP - Paris Diderot - Sorbonne Paris Cité University - INSERM U728 - Institut Universitaire d'Hematologie, France
| | - Vincent Ribrag
- Département de Médecine, Institut Gustave Roussy, Villejuif, France
| | - Stéphane Cheze
- Clinical Hematology, University Hospital of Caen, France
| | | | - Fabrice Jardin
- Clinical Hematology, INSERM U918, IRIB, Centre Henri Becquerel, Rouen, France
| | - François Lefrère
- Clinical Hematology, Paris Descartes - Sorbonne Paris Cité University, IMAGINE Institut, Necker Hospital, AP-HP, France
| | - Richard Delarue
- Clinical Hematology, Paris Descartes - Sorbonne Paris Cité University, IMAGINE Institut, Necker Hospital, AP-HP, France
| | - Eva Hoster
- Institute of Medical Informatics, Biometry, and Epidemiology, University of Munich, Germany Department of Internal Medicine III, University Hospital Munich, Germany
| | - Martin Dreyling
- Department of Internal Medicine III, University Hospital Munich, Germany
| | - Vahid Asnafi
- Biological Hematology, Paris Descartes - Sorbonne Paris Cité University, Institut Necker-Enfants Malades, AP-HP, France
| | - Olivier Hermine
- Clinical Hematology, Paris Descartes - Sorbonne Paris Cité University, IMAGINE Institut, Necker Hospital, AP-HP, France
| | - Elizabeth Macintyre
- Biological Hematology, Paris Descartes - Sorbonne Paris Cité University, Institut Necker-Enfants Malades, AP-HP, France
| |
Collapse
|
49
|
Navarro Matilla B, García-Marco JA. [Mantle cell lymphoma: Towards a personalized therapeutic strategy?]. Med Clin (Barc) 2015; 144:553-9. [PMID: 25023849 DOI: 10.1016/j.medcli.2014.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
Abstract
Mantle cell lymphoma (MCL) is a clinically heterogeneous non-Hodgkin lymphoma with an aggressive clinical behaviour and short survival in some cases and an indolent course in others. Advances in the biology and pathogenesis of MCL have unveiled several genes involved in deregulation of cell cycle checkpoints and the finding of subclonal populations with specific recurrent mutations (p53, ATM, NOTCH2) with an impact on disease progression and refractoriness to treatment. Prognostic stratification helps to distinguish between indolent and aggressive forms of MCL. Currently, younger fit patients benefit from more intensive front line chemotherapy regimens and consolidation with autologous transplantation, while older or frail patients are treated with less intensive regimens and rituximab maintenance. For relapsing disease, the introduction of bortezomib and lenalidomide containing regimens and B-cell receptor pathway inhibitors such as ibrutinib and idelalisib in combination with immunochemotherapy have emerged as therapeutic agents with promising clinical outcomes.
Collapse
Affiliation(s)
- Belén Navarro Matilla
- Servicio de Hematología y Hemoterapia, Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda, Madrid, España
| | - José A García-Marco
- Servicio de Hematología y Hemoterapia, Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda, Madrid, España.
| |
Collapse
|
50
|
Cheminant M, Robinson S, Ribrag V, Le Gouill S, Suarez F, Delarue R, Hermine O. Prognosis and outcome of stem cell transplantation for mantle cell lymphoma. Expert Rev Hematol 2015; 8:493-504. [DOI: 10.1586/17474086.2015.1047759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|