1
|
Jain P, Wang M. High-risk MCL: recognition and treatment. Blood 2025; 145:683-695. [PMID: 39786418 DOI: 10.1182/blood.2023022354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Significant progress in determining the molecular origins and resistance mechanisms of mantle cell lymphoma (MCL) has improved our understanding of the disease's clinical diversity. These factors greatly impact the prognosis of patients with MCL. Given the dynamic alterations in MCL clones and disease evolution, it is crucial to recognize high-risk prognostic factors at diagnosis and relapse. Clinical factors include a high MCL International Prognostic Index score with a high Ki-67 proliferation index, early disease progression within 24 months of first-line treatment, >3 previous lines of therapy at relapse, and an aggressive (blastoid or pleomorphic) histology. Molecular aberrations include dysregulated cyclin D1, an aberrant SOX11-CD70 axis, upregulated Musashi-2, MYC rearrangement, metabolic reprogramming, and epigenetic changes. Other factors that contribute to high-risk MCL include an immune-depleted microenvironment and clone adaptability with complex chromosomal anomalies and somatic mutations in TP53, NSD2, CCND1, CDKN2A, BIRC3, SP140, KMT2D, NFkBIE, SMARCA4, and NOTCH2. Ultra-high-risk MCL is indicated by the coexistence of multiple high-risk prognostic factors in the relapse setting and can portend very short progression-free survival. As MCL treatments advance toward cellular therapies, resistance to anti-CD19 chimeric antigen receptor T-cell therapy is also observed. These findings necessitate revisiting the prognostic impact of high-risk factors, current management strategies, new bi- and trispecific T-cell engagers, combination therapies, novel therapeutic targets, and next-generation clinical trials for patients with high-risk MCL. This article provides a comprehensive update on recognizing and managing high-risk MCL and encompass current practices and future directions.
Collapse
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
2
|
Kumar A, Soumerai J, Abramson JS, Barnes JA, Caron P, Chhabra S, Chabowska M, Dogan A, Falchi L, Grieve C, Haydu JE, Johnson PC, Joseph A, Kelly HE, Labarre A, Lue JK, Martignetti R, Mi J, Moskowitz A, Owens C, Plummer S, Puccio M, Salles G, Seshan V, Simkins E, Slupe N, Zhang H, Zelenetz AD. Zanubrutinib, obinutuzumab, and venetoclax for first-line treatment of mantle cell lymphoma with a TP53 mutation. Blood 2025; 145:497-507. [PMID: 39437708 PMCID: PMC11826521 DOI: 10.1182/blood.2024025563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
ABSTRACT TP53-mutant mantle cell lymphoma (MCL) is associated with poor survival outcomes with standard chemoimmunotherapy. We conducted a multicenter, phase 2 study of zanubrutinib, obinutuzumab, and venetoclax (BOVen) in untreated patients with MCL with a TP53 mutation. Patients initially received 160 mg zanubrutinib twice daily and obinutuzumab. Obinutuzumab at a dose of 1000 mg was given on cycle 1 day 1, 8, and 15, and on day 1 of cycles 2 to 8. After 2 cycles, venetoclax was added with weekly dose ramp-up to 400 mg daily. After 24 cycles, if patients were in complete remission with undetectable minimal residual disease (uMRD) using an immunosequencing assay, treatment was discontinued. The primary end point was met if ≥11 patients were progression free at 2 years. The study included 25 patients with untreated MCL with a TP53 mutation. The best overall response rate was 96% (24/25) and the complete response rate was 88% (22/25). Frequency of uMRD at a sensitivity level of 1 × 10-5 and uMRD at a sensitivity level of 1 × 10-6 at cycle 13 was 95% (18/19) and 84% (16/19), respectively. With a median follow-up of 28.2 months, the 2-year progression-free, disease-specific, and overall survival were 72%, 91%, and 76%, respectively. Common side effects were generally low grade and included diarrhea (64%), neutropenia (32%), and infusion-related reactions (24%). BOVen was well tolerated and met its primary efficacy end point in TP53-mutant MCL. These data support its use and ongoing evaluation. This trial was registered at www.ClinicalTrials.gov as #NCT03824483.
Collapse
MESH Headings
- Humans
- Lymphoma, Mantle-Cell/drug therapy
- Lymphoma, Mantle-Cell/genetics
- Lymphoma, Mantle-Cell/mortality
- Sulfonamides/administration & dosage
- Sulfonamides/adverse effects
- Aged
- Female
- Male
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Middle Aged
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/adverse effects
- Tumor Suppressor Protein p53/genetics
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Mutation
- Aged, 80 and over
- Pyrazoles/administration & dosage
- Pyrazoles/adverse effects
- Pyrimidines/administration & dosage
- Pyrimidines/adverse effects
- Piperidines/administration & dosage
- Piperidines/adverse effects
- Adult
Collapse
Affiliation(s)
- Anita Kumar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jacob Soumerai
- Center for Lymphoma, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jeremy S. Abramson
- Center for Lymphoma, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jeffrey A. Barnes
- Center for Lymphoma, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Philip Caron
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Shalini Chhabra
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maria Chabowska
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lorenzo Falchi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Clare Grieve
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - J. Erika Haydu
- Center for Lymphoma, Massachusetts General Hospital Cancer Center, Boston, MA
| | | | - Ashlee Joseph
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hailey E. Kelly
- Center for Lymphoma, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Alyssa Labarre
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Rosalba Martignetti
- Center for Lymphoma, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Joanna Mi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alison Moskowitz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Colette Owens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sean Plummer
- Center for Lymphoma, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Madeline Puccio
- Center for Lymphoma, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Gilles Salles
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Venkatraman Seshan
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elizabeth Simkins
- Center for Lymphoma, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Natalie Slupe
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Honglei Zhang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew D. Zelenetz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
3
|
Barraclough A, Tang C, Lasica M, Smyth E, Cirillo M, Mutsando H, Cheah CY, Ku M. Diagnosis and management of mantle cell lymphoma: a consensus practice statement from the Australasian Lymphoma Alliance. Intern Med J 2025; 55:117-129. [PMID: 39578957 DOI: 10.1111/imj.16561] [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] [Received: 03/02/2024] [Accepted: 10/13/2024] [Indexed: 11/24/2024]
Abstract
Mantle cell lymphoma (MCL) is a clinically heterogeneous B-cell neoplasm with unique clinicopathological features, accounting for 5% of all non-Hodgkin lymphoma. Although for many chemoimmunotherapy can lead to durable remissions, those with poor baseline prognostic factors, namely blastoid morphology, TP53 aberrancy and Ki67 >30%, will have less durable responses to conventional therapies. With this in mind, clinical trials have focused on novel targeted therapies to improve outcomes. This review details the recent advances in the understanding of MCL biology and outlines the recommended diagnostic strategies and evidence-based approaches to treatment.
Collapse
Affiliation(s)
- Allison Barraclough
- Department of Haematology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Catherine Tang
- Department of Haematology, Gosford Hospital, Gosford, New South Wales, Australia
- School of Medicine and Public Health, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Masa Lasica
- Department of Haematology, St Vincent's Hospital Melbourne, Melbourne, Victoria, Australia
| | - Elizabeth Smyth
- Department of Haematology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Melita Cirillo
- Department of Haematology, Royal Perth Hospital, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Howard Mutsando
- Cancer Services, Toowoomba Hospital, Toowoomba, Queensland, Australia
- Toowoomba Rural Clinical School, University of Queensland, Toowoomba, Queensland, Australia
| | - Chan Y Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Matthew Ku
- Department of Haematology, St Vincent's Hospital Melbourne, Melbourne, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Zhang S, Wang X, Yang Z, Ding M, Zhang M, Young KH, Zhang X. Minimal residual disease detection in lymphoma: methods, procedures and clinical significance. Front Immunol 2024; 15:1430070. [PMID: 39188727 PMCID: PMC11345172 DOI: 10.3389/fimmu.2024.1430070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 08/28/2024] Open
Abstract
Lymphoma is a highly heterogeneous lymphohematopoietic tumor. As our understanding of the biological and pathological characteristics of lymphoma improves, we are identifying an increasing number of lymphoma subtypes. Genotyping has enhanced our ability to diagnose, treat, and monitor the prognosis of lymphoma. Despite significant improvements in treatment effectiveness, traditional methods for assessing disease response and monitoring prognosis are imperfect, and there is no significant improvement in overall remission rates for lymphoma patients. Minimal Residual Disease (MRD) is often indicative of refractory disease or early relapse. For lymphoma patients, personalized MRD monitoring techniques offer an efficient means to estimate disease remission levels, predict early relapse risk, and assess the effectiveness of new drug regimens. In this review, we delve into the MRD procedures in lymphoma, including sample selection and requirements, detection methods and their limitations and advantages, result interpretation. Besides, we also introduce the clinical applications of MRD detection in lymphoma.
Collapse
Affiliation(s)
- Sijun Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Xiangyu Wang
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenzhen Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Mengjie Ding
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Ken H. Young
- Division of Hematopathology, Duke University Medicine Center, Duke Cancer Institute, Durham, NC, United States
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Coulter EM, Bewicke-Copley F, Mossner M, Graham TA, Fitzgibbon J, Okosun J. Defining an Optimized Workflow for Enriching and Analyzing Residual Tumor Populations Using Intracellular Markers. J Mol Diagn 2024; 26:245-256. [PMID: 38280422 DOI: 10.1016/j.jmoldx.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/12/2023] [Accepted: 01/11/2024] [Indexed: 01/29/2024] Open
Abstract
Tumor relapse is well recognized to arise from treatment-resistant residual populations. Strategies enriching such populations for in-depth downstream analyses focus on tumor-specific surface markers; however, enrichment using intracellular biomarkers remains challenging. Using B-cell lymphoma as an exemplar, we demonstrate feasibility to enrich B-cell lymphoma 2 (BCL2)high populations, a surrogate marker for t(14;18)+ lymphomas, for use in downstream applications. Different fixation protocols were assessed for impact on antibody expression and RNA integrity; glyoxal fixation demonstrated superior results regarding minimal effects on surface and intracellular expression, and RNA quality, compared with alternative fixatives evaluated. Furthermore, t(14;18)+ B cells were effectively detected using intracellular BCL2 overexpression to facilitate tumor cell enrichment. Tumor cell populations were enriched using the cellenONE F1.4 single-cell sorting platform, which detected and dispensed BCL2high-expressing cells directly into library preparation reagents for transcriptome analyses. Sorted glyoxal-fixed cells generated good quality sequencing libraries, with high concordance between live and fixed single-cell transcriptomic profiles, discriminating cell populations predominantly on B-cell biology. Overall, we successfully developed a proof-of-concept workflow employing a robust cell preparation protocol for intracellular markers combined with cell enrichment using the cellenONE platform, providing an alternative to droplet-based technologies when cellular input is low or requires prior enrichment to detect rare populations. This workflow has wider prognostic and therapeutic potential to study residual cells in a pan-cancer setting.
Collapse
Affiliation(s)
- Eve M Coulter
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Findlay Bewicke-Copley
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Maximilian Mossner
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom; Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Trevor A Graham
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom; Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jude Fitzgibbon
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; AstraZeneca, Waltham, Massachusetts
| | - Jessica Okosun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
6
|
Rezazadeh A, Pruett J, Detzner A, Edwin N, Hamadani M, Shah NN, Fenske TS. Immunoglobulin High Throughput Sequencing (Ig-HTS) Minimal Residual Disease (MRD) Analysis is an Effective Surveillance Tool in Patients With Mantle Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:254-259. [PMID: 38195321 DOI: 10.1016/j.clml.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Mantle cell lymphoma (MCL) accounts for 4% to 6% of B-cell non-Hodgkin lymphoma with historically poor outcomes. With the advent of intensive first-line, targeted, and cellular therapies, outcomes have improved, and initial remission can be 8 to 10 years or longer. As patients experience longer remissions, this raises the question of the optimal surveillance modality. Peripheral blood minimal residual disease (MRD) analysis offers a potential alternative to surveillance imaging that is sensitive, less costly, and eliminates the risk of radiation exposure. MATERIALS AND METHODS The clonoSEQ assay (Adaptive Biotechnologies) is an FDA-cleared commercially available Ig-HTS MRD assay with a sensitivity of 1 cell in 1,000,000. We performed a retrospective analysis of 34 patients from 2015 to 2021, who underwent MRD testing after achieving remission with first-line therapy. RESULTS With a median follow-up of 6.5 years, 10-year progression free survival (PFS) was 60% and 10-year overall survival was 92% of the entire cohort. Among 12 patients who sustained a radiographic relapse, peripheral blood became MRD+ either at or prior to the time of relapse in 11 patients (92%). The first MRD+ test had a lead time of 0 to 24 months (median 34 days) prior to radiographic relapse. Only 1 patient had a MRD- result while being found to have progressive disease on imaging. Among 22 patients who sustained continuous clinical remission, 21 have remained MRD-. Several patients were able to enjoy 2 to 4-year intervals without surveillance imaging. CONCLUSIONS Our data suggest that the clonoSEQ MRD assay is an effective surveillance tool for MCL patients following first-line therapy and is predictive of relapse prior to imaging.
Collapse
Affiliation(s)
- Alexandra Rezazadeh
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Julie Pruett
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Amy Detzner
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI
| | | | - Mehdi Hamadani
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI; BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI
| | - Nirav N Shah
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI; BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI
| | - Timothy S Fenske
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI; BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
7
|
Hoster E, Delfau-Larue MH, Macintyre E, Jiang L, Stilgenbauer S, Vehling-Kaiser U, Salles G, Thieblemont C, Tilly H, Wirths S, Feugier P, Hübel K, Schmidt C, Ribrag V, Kluin-Nelemans JC, Dreyling M, Pott C. Predictive Value of Minimal Residual Disease for Efficacy of Rituximab Maintenance in Mantle Cell Lymphoma: Results From the European Mantle Cell Lymphoma Elderly Trial. J Clin Oncol 2024; 42:538-549. [PMID: 37992261 DOI: 10.1200/jco.23.00899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/17/2023] [Accepted: 09/06/2023] [Indexed: 11/24/2023] Open
Abstract
PURPOSE The outcome of older patients with mantle cell lymphoma (MCL) has improved by the introduction of immunochemotherapy, followed by rituximab (R)-maintenance. Assessment of minimal residual disease (MRD) represents a promising tool for individualized treatment decisions and was a prospectively planned part of the European MCL Elderly trial. We investigated how MRD status influenced the efficacy of R-maintenance and how MRD can enable tailored consolidation strategies. PATIENTS AND METHODS Previously untreated patients with MCL age 60 years or older have been randomly assigned to R versus interferon-alpha maintenance after response to rituximab, fludarabine, cyclophosphamide (R-FC) versus rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone (R-CHOP). MRD monitoring was performed by real-time quantitative polymerase chain reaction (qPCR) following EuroMRD guidelines. RESULTS A qPCR assay with a median sensitivity of 1 × 10-5 could be generated in 80% of 288 patients in an international, multicenter, multilaboratory setting. More extensive tumor dissemination facilitated the identification of a molecular marker. The efficacy of R-maintenance in clinical remission was confirmed for MRD-negative patients at the end of induction in terms of progression-free survival (PFS; hazard ratio [HR], 0.38 [95% CI, 0.21 to 0.63]) and overall survival (OS; HR, 0.37 [95% CI, 0.20 to 0.68]), particularly in R-CHOP-treated patients (PFS-HR, 0.23 [95% CI, 0.10 to 0.52]; OS-HR, 0.19 [95% CI, 0.07 to 0.52]). R-maintenance appeared less effective in MRD-positive patients (PFS-HR, 0.51 [95% CI, 0.26 to 1.02]) overall and after R-CHOP induction (PFS-HR, 0.59 [95% CI, 0.28 to 1.26]). R-FC achieved more frequent and faster MRD clearance compared with R-CHOP. MRD positivity in clinical remission after induction was associated with a short median time to clinical progression of approximately 1-1.7 years. CONCLUSION The results confirm the strong efficacy of R-maintenance in patients who are MRD-negative after induction. Treatment de-escalation for MRD-negative patients is discouraged by our results. More effective consolidation strategies should be explored in MRD-positive patients to improve their long-term prognosis.
Collapse
Affiliation(s)
- Eva Hoster
- Institute for Medical Informatics, Biometry, and Epidemiology (IBE), LMU Munich, Munich, Germany
- Department of Internal Medicine III, University Hospital LMU Munich, Munich, Germany
| | | | - Elizabeth Macintyre
- Laboratory of Onco-Hematology, Université Paris Cité, Institut Necker-Enfants Malades and Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Linmiao Jiang
- Institute for Medical Informatics, Biometry, and Epidemiology (IBE), LMU Munich, Munich, Germany
| | | | | | - Gilles Salles
- Hospices Civils de Lyon, Université Claude Bernard, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Hervé Tilly
- Department of Hematology and U1245, Centre Henri Becquerel, Rouen, France
| | - Stefan Wirths
- Department of Medicine II, University of Tübingen, Tübingen, Germany
| | - Pierre Feugier
- Department of Hematology and INSERM 1256, University of Lorraine, Vandoeuvre les Nancy, France
| | - Kai Hübel
- Klinik I für Innere Medizin, Universität zu Köln, Köln, Germany
| | - Christian Schmidt
- Department of Internal Medicine III, University Hospital LMU Munich, Munich, Germany
| | | | - Johanna C Kluin-Nelemans
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martin Dreyling
- Department of Internal Medicine III, University Hospital LMU Munich, Munich, Germany
| | - Christiane Pott
- Department of Medicine, University Hospital Schleswig-Holstein Campus Kiel/Christian-Albrechts University Kiel, Germany
| |
Collapse
|
8
|
Singh AP, Courville EL. Advances in Monitoring and Prognostication for Lymphoma by Flow Cytometry. Clin Lab Med 2023; 43:351-361. [PMID: 37481316 DOI: 10.1016/j.cll.2023.04.010] [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: 07/24/2023]
Abstract
Flow cytometry (FC) is a well-established method important in the diagnosis and subclassification of lymphoma. In this article, the role of FC in lymphoma prognostication will be explored, and the clinical role for FC minimal/measurable residual disease testing as a monitoring tool for mature lymphoma will be introduced. Potential pitfalls of monitoring for residual/recurrent disease following immunotherapy will be presented.
Collapse
Affiliation(s)
- Amrit P Singh
- Department of Pathology, University of Virginia Health, PO Box 800214, Charlottesville, VA 22908, USA
| | - Elizabeth L Courville
- Department of Pathology, University of Virginia Health, PO Box 800214, Charlottesville, VA 22908, USA.
| |
Collapse
|
9
|
What is the role of up-front autologous stem cell transplantation in mantle cell lymphoma? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:155-162. [PMID: 36485104 PMCID: PMC9820454 DOI: 10.1182/hematology.2022000333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Up-front autologous stem cell transplantation (ASCT) is the established standard of care for younger, transplant-eligible MCL patients and is associated with a prolonged progression-free survival (PFS) benefit. However, there is no randomized controlled trial data, with therapy including rituximab and cytarabine, that has established a PFS and overall survival (OS) benefit with ASCT in the modern era. Multiple retrospective studies have failed to identify an OS benefit associated with ASCT in younger MCL patients. The high-risk patient subgroup with evidence of baseline TP53 mutation has a dismal outcome with intensive chemoimmunotherapy followed by ASCT, thus up-front ASCT is not optimal for this patient subset. Ongoing randomized clinical trials will help to clarify the role of up-front ASCT in the future. For example, the ongoing European MCL Network Triangle study incorporating ibrutinib into chemoimmunotherapy induction and maintenance with and without ASCT will help define the role of ASCT in the era of novel biologically targeted agents (ClinicalTrials.gov identifier: NCT02858258). Additionally, minimal residual disease (MRD) assessment is a powerful prognostic tool in MCL, and the ongoing Eastern Cooperative Oncology Group-American College of Radiology Imaging Network E4151 study is comparing maintenance rituximab alone vs ASCT consolidation in MCL patients who achieve remission and MRD-undetectable status post induction (ClinicalTrials.gov identifier: NCT03267433). ASCT remains a highly efficacious initial therapy for younger MCL patients; however, ultimately the decision to pursue ASCT requires discussion of risks vs benefits, incorporating patient preferences and values.
Collapse
|
10
|
Ferrero S, Grimaldi D, Genuardi E, Drandi D, Zaccaria GM, Alessandria B, Ghislieri M, Ferrante M, Evangelista A, Mantoan B, De Luca G, Stefani PM, Benedetti F, Casaroli I, Zanni M, Castellino C, Pavone V, Petrini M, Re F, Hohaus S, Musuraca G, Cascavilla N, Ghiggi C, Liberati AM, Cortelazzo S, Ladetto M. Punctual and kinetic MRD analysis from the Fondazione Italiana Linfomi MCL0208 phase 3 trial in mantle cell lymphoma. Blood 2022; 140:1378-1389. [PMID: 35737911 PMCID: PMC9507010 DOI: 10.1182/blood.2021014270] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 05/27/2022] [Indexed: 11/20/2022] Open
Abstract
Minimal residual disease (MRD) analysis is a known predictive tool in mantle cell lymphoma (MCL). We describe MRD results from the Fondazione Italiana Linfomi phase 3 MCL0208 prospective clinical trial assessing lenalidomide (LEN) maintenance vs observation after autologous stem cell transplantation (ASCT) in the first prospective comprehensive analysis of different techniques, molecular markers, and tissues (peripheral blood [PB] and bone marrow [BM]), taken at well-defined time points. Among the 300 patients enrolled, a molecular marker was identified in 250 (83%), allowing us to analyze 234 patients and 4351 analytical findings from 10 time points. ASCT induced high rates of molecular remission (91% in PB and 83% in BM, by quantitative real-time polymerase chain reaction [RQ-PCR]). Nevertheless, the number of patients with persistent clinical and molecular remission decreased over time in both arms (up to 30% after 36 months). MRD predicted early progression and long-term outcome, particularly from 6 months after ASCT (6-month time to progression [TTP] hazard ratio [HR], 3.83; P < .001). In single-timepoint analysis, BM outperformed PB, and RQ-PCR was more reliable, while nested PCR appeared applicable to a larger number of patients (234 vs 176). To improve MRD performance, we developed a time-varying kinetic model based on regularly updated MRD results and the MIPI (Mantle Cell Lymphoma International Prognostic Index), showing an area under the ROC (Receiver Operating Characteristic) curve (AUROC) of up to 0.87 using BM. Most notably, PB reached an AUROC of up to 0.81; with kinetic analysis, it was comparable to BM in performance. MRD is a powerful predictor over the entire natural history of MCL and is suitable for models with a continuous adaptation of patient risk. The study can be found in EudraCT N. 2009-012807-25 (https://eudract.ema.europa.eu/).
Collapse
Affiliation(s)
- Simone Ferrero
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
- AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Daniele Grimaldi
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
- Hematology Division, AO S.Croce e Carle, Cuneo, Italy
| | - Elisa Genuardi
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Daniela Drandi
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Gian Maria Zaccaria
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
- Hematology and Cell Therapy Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Beatrice Alessandria
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Marco Ghislieri
- PoliToBIOMed Lab, Politecnico di Torino, Torino, Italy
- Department of Electronics and Telecommunications, Politecnico di Torino, Torino, Italy
| | - Martina Ferrante
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Andrea Evangelista
- Unit of Cancer Epidemiology, CPO, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Barbara Mantoan
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Gabriele De Luca
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | | | - Fabio Benedetti
- Department of Medicine, Section of Hematology and Bone Marrow Transplant Unit, University of Verona, Verona, Italy
| | | | - Manuela Zanni
- Division of Hematology, Antonio e Biagio e Cesare Arrigo Hospital, Alessandria, Italy
| | | | | | | | - Francesca Re
- Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Stefan Hohaus
- Hematology Unit, Università Cattolica S.Cuore; Roma, Italy
| | - Gerardo Musuraca
- Department of Hematology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS
| | - Nicola Cascavilla
- Hematology, Casa Sollievo della Sofferenza IRCCS Hospital, San Giovanni Rotondo, Italy
| | - Chiara Ghiggi
- Department of Hematology, San Martino Hospital and University, Genova, Italy
| | - Anna Marina Liberati
- Department of Hematology, A.O. Santa Maria Terni, University of Perugia, Perugia, Italy; and
| | | | - Marco Ladetto
- Division of Hematology, Antonio e Biagio e Cesare Arrigo Hospital, Alessandria, Italy
| |
Collapse
|
11
|
Jain P, Wang ML. Mantle cell lymphoma in 2022-A comprehensive update on molecular pathogenesis, risk stratification, clinical approach, and current and novel treatments. Am J Hematol 2022; 97:638-656. [PMID: 35266562 DOI: 10.1002/ajh.26523] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 12/21/2022]
Abstract
The field of mantle cell lymphoma (MCL) has witnessed remarkable progress due to relentless advances in molecular pathogenesis, prognostication, and newer treatments. MCL consists of a spectrum of clinical subtypes. Rarely, atypical cyclin D1-negative MCL and in situ MCL neoplasia are identified. Prognostication of MCL is further refined by identifying somatic mutations (such as TP53, NSD2, KMT2D), methylation status, chromatin organization pattern, SOX-11 expression, minimal residual disease (MRD), and genomic clusters. Lymphoid tissue microenvironment studies demonstrated the role of B-cell receptor signaling, nuclear factor kappa B (NF-kB), colony-stimulating factor (CSF)-1, the CD70-SOX-11 axis. Molecular mechanism of resistance, mutation dynamics, and pathogenic pathways (B-cell receptor (BCR), oxidative phosphorylation, and MYC) were identified in mediating resistance to various treatments (bruton tyrosine kinase (BTK) inhibitors [ibrutinib, acalabrutinib]. Treatment options range from conventional chemoimmunotherapy and stem cell transplantation (SCT) to targeted therapies against BTK (covalent and noncovalent), Bcl2, ROR1, cellular therapy such as anti-CD19 chimeric antigen receptor therapy (CAR-T), and most recently bispecific antibodies against CD19 and CD20. MCL patients frequently relapse. Complex pathogenesis and the management of patients with progression after treatment with BTK/Bcl2 inhibitors and CAR-T (triple-resistant MCL) remain a challenge. Next-generation clinical trials incorporating newer agents and concurrent translational and molecular investigations are ongoing.
Collapse
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma. Mantle cell lymphoma center of excellence The University of Texas MD Anderson Cancer Center Houston Texas USA
| | - Michael L. Wang
- Department of Lymphoma/Myeloma. Mantle cell lymphoma center of excellence The University of Texas MD Anderson Cancer Center Houston Texas USA
| |
Collapse
|
12
|
Jain P, Dreyling M, Seymour JF, Wang M. High-Risk Mantle Cell Lymphoma: Definition, Current Challenges, and Management. J Clin Oncol 2020; 38:4302-4316. [DOI: 10.1200/jco.20.02287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Martin Dreyling
- Medizinische Klinik III, Ludwig Maximilian University Klinikum München, München, Germany
| | - John F. Seymour
- Peter MacCallum Cancer Center, Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
13
|
Jung D, Jain P, Yao Y, Wang M. Advances in the assessment of minimal residual disease in mantle cell lymphoma. J Hematol Oncol 2020; 13:127. [PMID: 32972438 PMCID: PMC7513535 DOI: 10.1186/s13045-020-00961-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022] Open
Abstract
The clinical impact of minimal residual disease detection at early time points or during follow-ups has been shown to accurately predict relapses among patients with lymphomas, mainly in follicular and diffuse large B cell lymphoma. The field of minimal residual disease testing in mantle cell lymphoma is still evolving but has great impact in determining the prognosis. Flow cytometry and polymerase chain reaction-based testing are most commonly used methods in practice; however, these methods are not sensitive enough to detect the dynamic changes that underline lymphoma progression. Newer methods using next-generation sequencing, such as ClonoSeq, are being incorporated in clinical trials. Other techniques under evolution include CAPP-seq and anchored multiplex polymerase chain reaction-based methods. This review article aims to provide a comprehensive update on the status of minimal residual disease detection and its prognostic effect in mantle cell patients. The role of circulating tumor DNA-based minimal residual disease detection in lymphomas is also discussed.
Collapse
Affiliation(s)
- Dayoung Jung
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Preetesh Jain
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Hemapathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Yixin Yao
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|