1
|
Malinverni C, Bernardelli A, Glimelius I, Mirandola M, Smedby KE, Tisi MC, Giné E, Albertsson-Lindblad A, Marin-Niebla A, Di Rocco A, Moita F, Sciarra R, Bašić-Kinda S, Hess G, Ohler A, Eskelund CW, Re A, Ferrarini I, Kolstad A, Räty R, Quaglia FM, Eyre TA, Scapinello G, Stefani PM, Morello L, Nassi L, Hohaus S, Ragaini S, Zilioli VR, Bruna R, Cocito F, Arcari A, Jerkeman M, Visco C. Outcomes of younger patients with mantle cell lymphoma experiencing late relapse (>24 months): the LATE-POD study. Blood 2024; 144:1001-1009. [PMID: 38754055 DOI: 10.1182/blood.2023023525] [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: 12/06/2023] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/18/2024] Open
Abstract
ABSTRACT Patients with mantle cell lymphoma (MCL) who experience first relapse/refractoriness can be categorized into early or late progression-of-disease (POD) groups, with a threshold of 24 months from MCL diagnosis. Bruton tyrosine kinase inhibitors (BTKi) are the established standard treatment at first relapse, but their effectiveness compared with chemoimmunotherapy (CIT) in late-POD patients remains unknown. In this international, observational cohort study, we evaluated outcomes among patients at first, late POD beyond 24 months. The primary objective was progression-free survival from the time of second-line therapy (PFS-2) of BTKi vs CIT. Overall, 385 late-POD patients were included from 10 countries. Their median age was 59 years (range, 19-70), and 77% were male. Median follow-up from the time of second-line therapy was 53 months (range, 12-144). Overall, 114 patients had second-line BTKi, whereas 271 had CIT, consisting of rituximab-bendamustine (R-B; n = 101), R-B and cytarabine (R-BAC; n = 70), or other regimens (mostly cyclophosphamide-hydroxydaunorubicin-vincristine-prednisone]- or platinum-based; n = 100). The 2 groups were balanced in clinicopathological features and median time to first relapse. Overall, BTKi was associated with significantly prolonged median PFS-2 than CIT (not reached [NR] vs 26 months, respectively; P = .0003) and overall survival (NR and 56 months, respectively; P = .03). Multivariate analyses showed that BTKi was associated with lower risk of death than R-B and other regimens (hazard ratio, 0.41 for R-B and 0.46 for others), but similar to R-BAC. These results may establish BTKi as the preferable second-line approach in patients with BTKi-naïve MCL.
Collapse
Affiliation(s)
- Chiara Malinverni
- Department of Engineering for Innovation Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Andrea Bernardelli
- Department of Engineering for Innovation Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Ingrid Glimelius
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | - Massimo Mirandola
- Epidemiology Unit, Division of Infectious Diseases, Department of Medicine, World Health Organization Collaborating Centre for Sexual Health and Vulnerable Populations, Verona University Hospital, Verona, Italy
- School of Health Sciences, University of Brighton, Brighton, United Kingdom
| | - Karin E Smedby
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Chiara Tisi
- Department of Hematology and Cell Therapy, San Bortolo Hospital, Vicenza, Italy
| | - Eva Giné
- Department of Hematology, Hospital Clinic of Barcelona, Barcelona, Spain
| | | | - Ana Marin-Niebla
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Alice Di Rocco
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Filipa Moita
- Department of Hematology, Instituto Português de Oncologia de Lisboa, Lisbon, Portugal
| | - Roberta Sciarra
- Division of Hematology, Fondazione IRCSS Policlinico San Matteo, Pavia, Italy
| | | | - Georg Hess
- Department of Hematology, Oncology, and Pneumology, Comprehensive Cancer Center, Johannes Gutenberg University, Mainz, Germany
| | - Anke Ohler
- Department of Hematology, Oncology, and Pneumology, Comprehensive Cancer Center, Johannes Gutenberg University, Mainz, Germany
| | | | - Alessandro Re
- Division of Hematology, Spedali Civili di Brescia, Brescia, Italy
| | - Isacco Ferrarini
- Department of Engineering for Innovation Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Arne Kolstad
- Department of Oncology, Sykehuset Innlandet, Lillehammer, Norway
| | - Riikka Räty
- Department of Hematology, Helsinki University Hospital, Helsinki, Finland
| | - Francesca Maria Quaglia
- Department of Engineering for Innovation Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Toby A Eyre
- Department of Clinical Hematology, Oxford University Hospitals National Health Service Foundation Trust, Oxford, United Kingdom
| | - Greta Scapinello
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Padua, Padua, Italy
| | | | - Lucia Morello
- Department of Medical Oncology and Hematology, Humanitas Clinical and Research Center, Milan, Italy
| | - Luca Nassi
- Division of Hematology, Careggi Hospital and University of Florence, Florence, Italy
| | - Stefan Hohaus
- Department of Radiological and Hematological Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Simone Ragaini
- Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | | | - Riccardo Bruna
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont Amedeo Avogadro, Novara, Italy
| | | | - Annalisa Arcari
- Division of Hematology, Ospedale Guglielmo da Saliceto, Piacenza, Italy
| | - Mats Jerkeman
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Carlo Visco
- Department of Engineering for Innovation Medicine, Section of Hematology, University of Verona, Verona, Italy
| |
Collapse
|
2
|
Ip A, Della Pia A, Goy AH. SOHO State of the Art Updates and Next Questions: Treatment Evolution of Mantle Cell Lymphoma: Navigating the Different Entities and Biological Heterogeneity of Mantle Cell Lymphoma in 2024. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:491-505. [PMID: 38493059 DOI: 10.1016/j.clml.2024.02.010] [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: 12/12/2023] [Revised: 02/12/2024] [Accepted: 02/18/2024] [Indexed: 03/18/2024]
Abstract
Progress in mantle cell lymphoma (MCL) has led to significant improvement in outcomes of patients even in the real world (RW) setting albeit to a lesser degree. In parallel to the demonstration of benefit using combination therapy with rituximab plus high-dose cytarabine (R-AraC) as well as dose intensive therapy-autologous stem cell transplantation (DIT-ASCT) consolidation and maintenance, it became clear over the last 2 decades that MCL is a highly heterogenous disease at the molecular level, explaining differences observed in clinical behavior and response to therapy. While clinical prognostic factors and models have helped stratify patients with distinct outcomes, they failed to help guide therapy. The identification of molecular high-risk (HR) features, in particular, but not only, p53 aberrations (including mutations and deletions [del]), as well as complex karyotype (CK), has allowed to identify subsets of patients with poorer outcomes (median overall survival [OS] <2 years) regardless of conventional therapies used. The constant pattern of relapse seen in MCL has fueled sustained and productive efforts, with 7 novel agents approved in the United States (US), showing high and durable efficacy even in HR and chemo-refractory patients and likely curing a subset of patients in the relapsed or refractory (R/R) setting. Progress in diagnostics, in particular next-generation sequencing (NGS), which is accessible in routine practice nowadays, can help recognize patients with HR features, well beyond MIPI or Ki-67 prognostication, although the impact on decision making is still unclear. The era of integrating novel agents into our prior standard of care (SOC) has begun with a confirmed benefit, for example, ibrutinib (Ib) in the TRIANGLE study, defining the first new potential SOC in younger patients in over 30 years. Expanding on novel agents, either in combination, sequentially or to replace chemotherapy altogether, using biological doublets or triplets has led to a median progression-free survival (PFS) in excess of 72 months, certainly competitive with prior SOC and will continue to reshape the management of MCL patients. Achieving minimal residual disease negative (MRD-ve) status is becoming a new endpoint in MCL, and customizing maintenance and/or de-escalation/consolidation strategies is within reach, although it will require prospective, built-in MRD-based approaches, with the goal of eliminating subclinical disease and not simply delaying time to relapse. Taking into account the biological diversity of MCL is now feasible in routine clinical practice and has already helped recognize what not to do for HR patients (i.e., avoid intensive induction chemotherapy and/or ASCT for p53 mutated patients) as well as identify promising novel options. Ongoing and future work will help expand on these dedicated approaches, to further improve the management and outcomes of all MCL patients.
Collapse
Affiliation(s)
- Andrew Ip
- Lymphoma Division, John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ
| | - Alexandra Della Pia
- Lymphoma Division, John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ
| | - Andre H Goy
- Lymphoma Division, John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ.
| |
Collapse
|
3
|
Epstein-Peterson ZD, Drill E, Aypar U, Batlevi CL, Caron P, Dogan A, Drullinsky P, Gerecitano J, Hamlin PA, Ho C, Jacob A, Joseph A, Laraque L, Matasar MJ, Moskowitz AJ, Moskowitz CH, Mullins C, Owens C, Salles G, Schöder H, Straus DJ, Younes A, Zelenetz AD, Kumar A. Immunochemotherapy plus lenalidomide for high-risk mantle cell lymphoma with measurable residual disease evaluation. Haematologica 2024; 109:1149-1162. [PMID: 37646671 PMCID: PMC10985438 DOI: 10.3324/haematol.2023.282898] [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: 02/07/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
Chemoimmunotherapy followed by consolidative high-dose therapy with autologous stem cell rescue was a standard upfront treatment for fit patients with mantle cell lymphoma (MCL) in first remission; however, treatment paradigms are evolving in the era of novel therapies. Lenalidomide is an immunomodulatory agent with known efficacy in treating MCL. We conducted a single-center, investigator-initiated, phase II study of immunochemotherapy incorporating lenalidomide, without autologous stem cell transplant consolidation, enriching for patients with high-risk MCL (clinicaltrials gov. Identifier: NCT02633137). Patients received four cycles of lenalidomide-R-CHOP, two cycles of R-HiDAC, and six cycles of R-lenalidomide. The primary endpoint was rate of 3-year progression-free survival. We measured measurable residual disease (MRD) using a next-generation sequencing-based assay after each phase of treatment and at 6 months following end-oftreatment. We enrolled 49 patients of which 47 were response evaluable. By intent-to-treat, rates of overall and complete response were equivalent at 88% (43/49), one patient with stable disease, and two patients had disease progression during study; 3-year progression-free survival was 63% (primary endpoint not met) and differed by TP53 status (78% wild-type vs. 38% ALT; P=0.043). MRD status was prognostic and predicted long-term outcomes following R-HiDAC and at 6 months following end-of-treatment. In a high-dose therapy-sparing, intensive approach, we achieved favorable outcomes in TP53- wild-type MCL, including high-risk cases. We confirmed that sequential MRD assessment is a powerful prognostic tool in patients with MCL.
Collapse
Affiliation(s)
- Zachary D Epstein-Peterson
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Esther Drill
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center
| | - Umut Aypar
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Connie Lee Batlevi
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Philip Caron
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Ahmet Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Pamela Drullinsky
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - John Gerecitano
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Paul A Hamlin
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Caleb Ho
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | | | - Ashlee Joseph
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Leana Laraque
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Matthew J Matasar
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Alison J Moskowitz
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Craig H Moskowitz
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | | - Colette Owens
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Gilles Salles
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David J Straus
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Anas Younes
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Andrew D Zelenetz
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Anita Kumar
- Lymphoma Service, Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center.
| |
Collapse
|
4
|
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
|
5
|
Lu T, Zhang J, McCracken JM, Young KH. Recent advances in genomics and therapeutics in mantle cell lymphoma. Cancer Treat Rev 2024; 122:102651. [PMID: 37976759 DOI: 10.1016/j.ctrv.2023.102651] [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: 09/15/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Over the past decades, significant strides have been made in understanding the pathobiology, prognosis, and treatment options for mantle cell lymphoma (MCL). The heterogeneity observed in MCL's biology, genomics, and clinical manifestations, including indolent and aggressive forms, is intricately linked to factors such as the mutational status of the variable region of the immunoglobulin heavy chain gene, epigenetic profiling, and Sox11 expression. Several intriguing subtypes of MCL, such as Cyclin D1-negative MCL, in situ mantle cell neoplasm, CCND1/IGH FISH-negative MCL, and the impact of karyotypic complexity on prognosis, have been explored. Notably, recent immunochemotherapy regimens have yielded long-lasting remissions in select patients. The therapeutic landscape for MCL is continuously evolving, with a shift towards nonchemotherapeutic agents like ibrutinib, acalabrutinib, and venetoclax. The introduction of BTK inhibitors has brought about a transformative change in MCL treatment. Nevertheless, the challenge of resistance to BTK inhibitors persists, prompting ongoing efforts to discover strategies for overcoming this resistance. These strategies encompass non-covalent BTK inhibitors, immunomodulatory agents, BCL2 inhibitors, and CAR-T cell therapy, either as standalone treatments or in combination regimens. Furthermore, developing novel drugs holds promise for further improving the survival of patients with relapsed or refractory MCL. In this comprehensive review, we methodically encapsulate MCL's clinical and pathological attributes and the factors influencing prognosis. We also undertake an in-depth examination of stratified treatment alternatives. We investigate conceivable resistance mechanisms in MCL from a genetic standpoint and offer precise insights into various therapeutic approaches for relapsed or refractory MCL.
Collapse
Affiliation(s)
- Tingxun Lu
- Division of Hematopathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Jie Zhang
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Jenna M McCracken
- Division of Hematopathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ken H Young
- Division of Hematopathology, Duke University Medical Center, Durham, NC 27710, USA; Duke Cancer Institute, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
6
|
Gerson JN, Handorf E, Villa D, Gerrie AS, Chapani P, Li S, Medeiros LJ, Wang M, Cohen JB, Churnetski M, Hill BT, Sawalha Y, Hernandez-Ilizaliturri FJ, Kothari S, Vose JM, Bast M, Fenske T, Rao Gari SN, Maddocks KJ, Bond D, Bachanova V, Kolla B, Chavez J, Shah B, Lansigan F, Burns T, Donovan AM, Wagner-Johnston N, Messmer M, Mehta A, Anderson JK, Reddy N, Kovach AE, Landsburg DJ, Glenn M, Inwards DJ, Ristow K, Karmali R, Kaplan JB, Caimi PF, Rajguru S, Evens A, Klein A, Umyarova E, Pulluri B, Amengual JE, Lue JK, Diefenbach C, Fisher RI, Barta SK. Outcomes of patients with blastoid and pleomorphic variant mantle cell lymphoma. Blood Adv 2023; 7:7393-7401. [PMID: 37874912 PMCID: PMC10758713 DOI: 10.1182/bloodadvances.2023010757] [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: 05/16/2023] [Revised: 08/17/2023] [Accepted: 09/20/2023] [Indexed: 10/26/2023] Open
Abstract
Mantle cell lymphoma (MCL) is a B-cell non-Hodgkin lymphoma; data indicate that blastoid and pleomorphic variants have a poor prognosis. We report characteristics and outcomes of patients with blastoid/pleomorphic variants of MCL. We retrospectively studied adults with newly diagnosed MCL treated from 2000 to 2015. Primary objectives were to describe progression-free survival (PFS) and overall survival (OS). Secondary objectives included characterization of patient characteristics and treatments. Of the 1029 patients with MCL studied, a total of 207 neoplasms were blastoid or pleomorphic variants. Median follow-up period was 82 months (range, 0.1-174 months); median PFS was 38 months (95% confidence interval [CI], 28-66) and OS was 68 months (95% CI, 45-96). Factors associated with PFS were receipt of consolidative autologous hematopoietic transplantation (auto-HCT; hazard ratio [HR], 0.52; 95% CI, 0.31-0.80; P < .05), MCL International Prognostic Index (MIPI) intermediate (HR, 2.3; 95% CI, 1.2-4.3; P < .02) and high (HR, 3.8; 95% CI, 2.0-7.4; P < .01) scores, and complete response to induction (HR, 0.29 (95% CI, 0.17-0.51). Receipt of auto-HCT was not associated with OS (HR, 0.69; 95% CI, 0.41-1.16; P = .16) but was associated with MIPI intermediate (HR, 5.7; 95% CI, 2.5-13.2; P < .01) and high (HR, 10.8; 95% CI, 4.7-24.9; P < .01) scores. We report outcomes in a large cohort of patients with blastoid/pleomorphic variant MCL. For eligible patients, receipt of auto-HCT after induction was associated with improved PFS but not OS. Higher MIPI score and auto-HCT ineligibility were associated with worse survival.
Collapse
Affiliation(s)
- James N. Gerson
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA
| | - Elizabeth Handorf
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Diego Villa
- BC Cancer Centre for Lymphoid Cancer, Vancouver, CA
| | | | - Parv Chapani
- BC Cancer Centre for Lymphoid Cancer, Vancouver, CA
| | | | | | | | - Jonathon B. Cohen
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA
| | - Michael Churnetski
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA
| | | | | | | | | | | | - Martin Bast
- University of Nebraska Cancer Center, Omaha, NE
| | - Timothy Fenske
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI
| | | | | | - David Bond
- Division of Hematology, Ohio State University, Columbus, OH
| | - Veronika Bachanova
- Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN
| | - Bhaskar Kolla
- Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN
| | | | | | | | - Timothy Burns
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | | | | | - Marcus Messmer
- Hematologic Malignancies Division, Johns Hopkins University, Baltimore, MD
| | | | | | | | | | - Daniel J. Landsburg
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA
| | | | | | - Kay Ristow
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Reem Karmali
- Division of Hematology and Oncology, Northwestern University, Evanston, IL
| | - Jason B. Kaplan
- Division of Hematology and Oncology, Northwestern University, Evanston, IL
| | - Paolo F. Caimi
- Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH
| | - Saurabh Rajguru
- Division of Hematology, Medical Oncology and Palliative Care, University of Wisconsin, Madison, WI
| | - Andrew Evens
- Division of Hematology/Oncology, Tufts University, Boston, MA
| | - Andreas Klein
- Division of Hematology/Oncology, Tufts University, Boston, MA
| | - Elvira Umyarova
- Division of Hematology and Oncology, University of Vermont, Burlington, VT
| | - Bhargavi Pulluri
- Division of Hematology and Oncology, University of Vermont, Burlington, VT
| | | | - Jennifer K. Lue
- Division of Hematology and Oncology, Columbia University, New York, NY
| | | | - Richard I. Fisher
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Stefan K. Barta
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
7
|
Karmali R, Abramson JS, Stephens DM, Barnes J, Winter JN, Ma S, Gao J, Kaplan J, Petrich AM, Hochberg E, Takvorian T, Mi X, Nelson V, Gordon LI, Pro B. Ibrutinib maintenance after frontline treatment in patients with mantle cell lymphoma. Blood Adv 2023; 7:7361-7368. [PMID: 37756532 PMCID: PMC10726243 DOI: 10.1182/bloodadvances.2023011271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Maintenance rituximab in mantle cell lymphoma (MCL) has improved survival and supports exploration of maintenance with novel agents. We evaluated the safety and efficacy of ibrutinib maintenance (I-M) after induction in patients with treatment-naive MCL. Patients with MCL with complete response (CR) or partial response to frontline chemoimmunotherapy ± autologous stem cell transplantation (auto-SCT) received I-M 560 mg daily for up to 4 years. Primary objective was 3-year progression-free survival (PFS) rate from initiation of I-M. Minimal residual disease (MRD) assessments by next-generation sequencing (NGS) on peripheral blood were measured before I-M initiation and at 1, 6, and 18 to 24 months after initiation. Among 36 patients, the median age was 60 years (range, 46-90). For frontline treatment, 18 patients (50%) had consolidation with auto-SCT in CR1 before I-M. At median follow-up of 55.7 months, 17 patients (47%) completed full course I-M (median, 37.5 cycles; range, 2-52). The 3-year PFS and overall survival (OS) rates were 94% and 97%, respectively. With prior auto-SCT, 3-year PFS and OS rates were both 100%. The most common treatment-related adverse event with I-M was infection (n = 31; 86%), typically low grade; the most common grade 3/4 toxicities were hematologic. In 22 patients with MRD assessments, all were MRD negative after induction. Six became MRD positive on I-M, with 2 reverting to MRD-negative status with continued I-M, and all maintain radiographic CR with the exception of 1 with disease progression. I-M is feasible in MCL after frontline chemoimmunotherapy with manageable toxicities although significant. Changes in NGS-MRD were noted in limited patients during maintenance with few progression and survival events. This trial was registered at www.clinicaltrials.gov as #NCT02242097.
Collapse
Affiliation(s)
- Reem Karmali
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Jeremy S. Abramson
- Massachusetts General Hospital Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | | | - Jeffrey Barnes
- Massachusetts General Hospital Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Jane N. Winter
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Shuo Ma
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Juehua Gao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jason Kaplan
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Ephraim Hochberg
- Massachusetts General Hospital Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Tak Takvorian
- Massachusetts General Hospital Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Xinlei Mi
- Department of Preventive Medicine–Biostatistics, Northwestern University, Chicago, IL
| | - Valerie Nelson
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Leo I. Gordon
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Barbara Pro
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| |
Collapse
|
8
|
Bock AM, Gile JJ, Larson MC, Poonsombudlert K, Tawfiq RK, Maliske S, Maurer MJ, Kabat BF, Paludo J, Inwards DJ, Ayyappan S, Link BK, Ansell SM, Habermann TM, Witzig TE, Nowakowski GS, Cerhan JR, Farooq U, Wang Y. Evolving treatment patterns and improved outcomes in relapsed/refractory mantle cell lymphoma: a prospective cohort study. Blood Cancer J 2023; 13:169. [PMID: 37957158 PMCID: PMC10643454 DOI: 10.1038/s41408-023-00942-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/10/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Over the last two decades, the frontline therapy for mantle cell lymphoma (MCL) has evolved. However, the impact of subsequent lines of therapy on survival outcomes has not been well characterized. In this study, we investigated the treatment patterns and survival outcomes in patients with relapsed/refractory (R/R) MCL treated with second-line (2 L) therapy. Adult patients with newly diagnosed MCL from 2002 to 2015 were enrolled in a prospective cohort study. Clinical characteristics, 2 L treatment details, and outcomes were compared between patients who received 2 L treatment between 2003-2009 (Era 1), 2010-2014 (Era 2), and 2015-2021 (Era 3). 2 L treatment was heterogenous in all eras, and there was a substantial shift in the pattern of 2 L therapy over time. The estimated 2-year EFS rate was 21% (95% CI, 13-35), 40% (95% CI, 30-53), and 51% (95% CI, 37-68) in Era 1-3 respectively, and the 5-year OS rate was 31% (95% CI, 21-45), 37% (95% CI, 27-50), and 67% (95% CI, 54-83) in Era 1-3, respectively. These results provide real-world evidence on evolving treatment patterns of 2 L therapy based on the era of relapse. The changes in 2 L treatment correlated with improved EFS and OS, suggesting that treatment advances are associated with improved outcomes in patients with R/R MCL.
Collapse
Affiliation(s)
- Allison M Bock
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute/University of Utah, Salt Lake City, UT, USA
| | - Jennifer J Gile
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Melissa C Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Seth Maliske
- Division of Hematology, Oncology, and Bone and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
- Sanford Health System, Fargo, ND, USA
| | - Matthew J Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Brian F Kabat
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jonas Paludo
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - David J Inwards
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sabarish Ayyappan
- Division of Hematology, Oncology, and Bone and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Brian K Link
- Division of Hematology, Oncology, and Bone and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Stephen M Ansell
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Thomas M Habermann
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Thomas E Witzig
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - James R Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Umar Farooq
- Division of Hematology, Oncology, and Bone and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Yucai Wang
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Turunen AS, Kuittinen O, Kuitunen H, Vasala K, Penttilä K, Harmanen M, Keskinen L, Mäntymaa P, Pelkonen J, Varmavuo V, Jantunen E, Partanen A. CD34 + Cell Mobilization, Autograft Cellular Composition and Outcome in Mantle Cell Lymphoma Patients. Transfus Med Hemother 2023; 50:428-437. [PMID: 37899989 PMCID: PMC10601603 DOI: 10.1159/000531799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/21/2023] [Indexed: 10/31/2023] Open
Abstract
Backgound Autologous stem cell transplantation (ASCT) is a standard treatment in transplant-eligible mantle cell lymphoma (MCL) patients after first-line chemoimmunotherapy. Study Design and Methods This prospective multicenter study evaluated the impact of CD34+ cell mobilization and graft cellular composition analyzed by flow cytometry on hematologic recovery and outcome in 42 MCL patients. Results During CD34+ cell mobilization, a higher blood CD34+ cell count (>30 × 106/L) was associated with improved overall survival (median not reached [NR] vs. 57 months, p = 0.04). The use of plerixafor did not impact outcome. Higher number of viable cryopreserved graft CD34+ cells (>3.0 × 106/kg) was associated with faster platelet (median 11 vs. 15 days, p = 0.03) and neutrophil (median 9 vs. 10 days, p = 0.02) recovery posttransplant. Very low graft CD3+CD8+ cell count (≤10 × 106/kg) correlated with worse progression-free survival (PFS) (HR 4.136, 95% CI 1.547-11.059, p = 0.005). On the other hand, higher absolute lymphocyte count >2.5 × 109/L at 30 days after ASCT (ALC-30) was linked with better PFS (median NR vs. 99 months, p = 0.045) and overall survival (median NR in either group, p = 0.05). Conclusions Better mobilization capacity and higher graft CD3+CD8+ cell count had a positive prognostic impact in this study, in addition to earlier lymphocyte recovery (ALC-30>2.5 × 106/L). These results need to be validated in another study with a larger patient cohort.
Collapse
Affiliation(s)
| | - Outi Kuittinen
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Oncology, Kuopio University Hospital, Kuopio, Finland
- Cancer Centre, Oulu University Hospital, Oulu, Finland
| | | | - Kaija Vasala
- Department of Oncology, Central Hospital of Central Finland, Jyväskylä, Finland
| | - Karri Penttilä
- Department of Medicine, Savonlinna Central Hospital, Savonlinna, Finland
- Finnish Medicines Agency, Kuopio, Finland
| | - Minna Harmanen
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Leena Keskinen
- Department of Oncology, Tampere University Hospital, Tampere, Finland
| | | | - Jukka Pelkonen
- Laboratory Centre of Eastern Finland, Kuopio, Finland
- Department of Clinical Microbiology, University of Eastern Finland, Kuopio, Finland
| | - Ville Varmavuo
- Department of Medicine, Kymenlaakso Central Hospital, Kotka, Finland
| | - Esa Jantunen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, North Carelia Hospital District, Joensuu, Finland
| | - Anu Partanen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
10
|
Patel D, Kahl B. SOHO State of the Art Updates and Next Questions: Tailoring Upfront Therapy in Mantle Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:633-641. [PMID: 37268478 DOI: 10.1016/j.clml.2023.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 06/04/2023]
Abstract
In this article, we will review current strategies for the front-line management of mantle cell lymphoma, an uncommon and biologically and clinically heterogeneous subtype of non-Hodgkin lymphoma that remains incurable with current therapies. Patients invariably relapse with time, and as a result, treatment strategies involve persistent therapy over the course of months to years, including induction, consolidation, and maintenance. Topics discussed include the historical development of various chemoimmunotherapy backbones with continued modifications to maintain and improve efficacy while limiting off-target, off-tumor effects. Chemotherapy-free induction regimens were developed initially for elderly or less fit patients though are now being utilized for younger, transplant-eligible patients due to deeper, more prolonged remission durations with fewer toxicities. The historic paradigm of recommending autologous hematopoietic cell transplant for fit patients in complete or partial remission is now being challenged based in part on ongoing clinical trials in which minimal residual disease directed approaches influence the consolidation strategy for any particular individual. The addition of novel agents, namely first and second generation Bruton tyrosine kinase inhibitors as well as immunomodulatory drugs, BH3 mimetics, and type II glycoengineered anti-CD20 monoclonal antibodies have been tested in various combinations with or without immunochemotherapy. We will attempt to help the reader by systematically explaining and simplifying the various approaches for treating this complicated group of disorders.
Collapse
Affiliation(s)
- Dilan Patel
- Washington University in St Louis School of Medicine, St Louis, Department of Medicine, Division of Oncology, MO
| | - Brad Kahl
- Washington University in St Louis School of Medicine, St Louis, Department of Medicine, Division of Oncology, MO..
| |
Collapse
|
11
|
Yang P, Cai Q, Zhang W, Liu S, Liu H, Sun X, Dong Y, Xiao X, Wang J, Li Z, Huang W, Li L, Bao H, Yang W, Wang Y, Wang S, He J, Li X, Liu A, Jing H. Real-world treatment and outcome patterns of patients with mantle cell lymphoma in China: A large, multicenter retrospective analysis. Cancer Med 2023; 12:13204-13216. [PMID: 37148540 PMCID: PMC10315753 DOI: 10.1002/cam4.6009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/04/2023] [Accepted: 04/15/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Mantle cell lymphoma (MCL) is an uncommon heterogeneous subtype of B cell non-Hodgkin lymphoma, and clinical features in MCL appear regional characteristics. MCL treatment opinions are not uniform between countries or regions within Asia and China, and Asian patient-specific data for MCL treatment are fewer. The study aims to explore the clinical characteristics, treatment patterns and prognosis of MCL patients in China. METHODS A total of 805 patients diagnosed with MCL between April 1999 and December 2019 at 19 comprehensive hospitals in China were included in this retrospective analysis. Kaplan-Meier method coupled with the log-rank test was used for univariate analysis, and COX proportional hazards model was used for multivariate analysis (MVA). p < 0.05 was consided statistically significant. All outputs were produced using R version 4.1.0. RESULTS The median age of the cohort was 60.0 years with a male-to-female ratio of 3.36:1. Five-year progression-free survival (PFS) and overall survival (OS) rates were 30.9% and 65.0%, respectively. High-intermediate/high-risk group according to MIPI-c, without high-dose cytarabine, lack of Auto-SCT as consolidation and maintenance treatment and SD/PD in initial treatment remained statistically relevant to poor PFS on MVA, and ki67 ≥50%, B symptoms, high-intermediate/high risk group according to MIPI-c, without high-dose cytarabine, lack of maintenance treatment, SD/PD in initial treatment and relapse/refractory state were independently associated with poorer OS on MVA. CONCLUSIONS First-line high dose cytarabine exposure, auto-SCT as consolidation therapy obtained survival benefits in Chinese population. Our study further confirmed the value of maintenance treatment and explored the application of new drug treatment and bendamustine in R/R MCL patients.
Collapse
Affiliation(s)
- Ping Yang
- Department of HematologyPeking University Third HospitalBeijingChina
| | - Qing‐qing Cai
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Wei Zhang
- Department of HematologyPeking Union Medical College HospitalBeijingChina
| | - Shuo‐zi Liu
- Department of HematologyPeking University Third HospitalBeijingChina
| | - Hui Liu
- Department of HematologyBeijing HospitalBeijingChina
| | - Xiu‐hua Sun
- Department of Medical OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Yu‐jun Dong
- Department of HematologyPeking University First HospitalBeijingChina
| | - Xiu‐bin Xiao
- Senior Department of HematologyThe 5th Medical Center of PLA General HospitalBeijingChina
| | - Jing‐wen Wang
- Department of HematologyBeijing Tongren HospitalBeijingChina
| | - Zhen‐ling Li
- Department of HematologyChina‐Japan Friendship HospitalBeijingChina
| | - Wen‐rong Huang
- Senior Department of HematologyThe 5th Medical Center of PLA General HospitalBeijingChina
| | - Li‐hong Li
- Department of HematologyBeijing Tsinghua Changgung HospitalBeijingChina
| | - Hui‐zheng Bao
- Department of Medical OncologyJilin Cancer HospitalChangchunChina
| | - Wei Yang
- Department of HematologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Ya‐lan Wang
- Department of Medical OncologyBaotou Cancer HospitalBaotouChina
| | - Shu‐ye Wang
- Department of HematologyThe First Hospital of Harbin Medical UniversityHarbinChina
| | - Juan He
- Department of HematologyThe First Hospital of China Medical UniversityShenyangChina
| | - Xiao‐ling Li
- Department of Medical OncologyLiaoning Cancer Hospital & InstituteShenyangChina
| | - Ai‐chun Liu
- Department of Hematology and Lymphatic MedicineHarbin Medical University Cancer HospitalHarbinChina
| | - Hong‐mei Jing
- Department of HematologyPeking University Third HospitalBeijingChina
| |
Collapse
|
12
|
Late effects in patients with mantle cell lymphoma treated with or without autologous stem cell transplantation. Blood Adv 2023; 7:866-874. [PMID: 35973196 PMCID: PMC10018432 DOI: 10.1182/bloodadvances.2022007241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/09/2022] [Accepted: 07/18/2022] [Indexed: 11/20/2022] Open
Abstract
Studies on late effects in patients with mantle cell lymphoma (MCL) are becoming increasingly important as survival is improving, and novel targeted drugs are being introduced. However, knowledge about late effects is limited. The aim of this population-based study was to describe the magnitude and panorama of late effects among patients treated with or without high-dose chemotherapy with autologous stem cell transplantation (HD-ASCT). The study cohort included all patients with MCL, recorded in the Swedish Lymphoma Register, aged 18 to 69 years, diagnosed between 2000 and 2014 (N = 620; treated with HD-ASCT, n = 247) and 1:10 matched healthy comparators. Patients and comparators were followed up via the National Patient Register and Cause of Death Register, from 12 months after diagnosis or matching to December 2017. Incidence rate ratios of the numbers of outpatient visits, hospitalizations, and bed days were estimated using negative binomial regression models. In relation to the matched comparators, the rate of specialist and hospital visits was significantly higher among patients with MCL. Patients with MCL had especially high relative risks of infectious, respiratory, and blood disorders. Within this observation period, no difference in the rate of these complications, including secondary neoplasms, was observed between patients treated with and without HD-ASCT. Most of the patients died from their lymphoma and not from another cause or treatment complication. Taken together, our results imply that most of the posttreatment health care needs are related to the lymphoma disease itself, thus, indicating the need for more efficient treatment options.
Collapse
|
13
|
Kröger K, Siats J, Kerkhoff A, Lenz G, Stelljes M, Eich HT, Reinartz G. Long-Term Survival of Patients with Mantle Cell Lymphoma after Total Body Irradiation, High-Dose Chemotherapy and Stem Cell Transplantation: A Monocenter Study. Cancers (Basel) 2023; 15:983. [PMID: 36765940 PMCID: PMC9913511 DOI: 10.3390/cancers15030983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 02/09/2023] Open
Abstract
INTRODUCTION In patients with mantle cell lymphoma (MCL), long-term remissions can be achieved by stem cell transplantation (SCT). Different conditioning treatment protocols exist with or without total body irradiation (TBI). There are few data published on the role of TBI before autologous stem cell transplantation (autoSCT) or allogenic stem cell transplantation (alloSCT). We report on the long-term survival data of patients treated by TBI prior to autologous or allogenic SCT at our center. PATIENTS In a retrospective analysis, the data of patients treated at the University Hospital of Muenster from May 2004 to February 2015 were collected and evaluated. For the analysis, all data of patients who were histopathologically diagnosed with MCL and underwent TBI prior to stem cell transplantation (SCT) were evaluated. RESULTS A total of 22 patients (19 men and 3 women) were treated with a TBI-based conditioning prior to SCT. The median age at initial diagnosis was 57.5 years (38-65 years). Seventeen patients had Ann Arbor stage IV, two patients had Ann Arbor stage III, and three patients Ann Arbor stage II disease. AutoSCT was performed in 19 patients and alloSCT was performed in 3 patients. In 18 patients, autoSCT was applied as part of first-line therapy, and in one patient after relapse. Two patients received alloSCT after relapse of MCL, and one patient received alloSCT during first-line therapy after an inadequate treatment response. TBI was performed in 12 patients with 10 Gy and in 6 patients with 12 Gy, these patients subsequently received autoSCT. In the group of four patients who received TBI with four Gy, four patients subsequently received alloSCT and one patient received autoSCT. Median overall survival after autoSCT and previous TBI was 11.4 years (142 months). In total, 11 out of 19 patients treated with autoSCT lived longer than 6.8 years (82-202 months). After alloSCT and previous TBI, the median overall survival was 3.25 years (14-59 months). CONCLUSIONS A large proportion of patients with advanced MCL survived remarkably longer than 11.4 years after high-dose chemotherapy, TBI, and SCT. The present results of multimodal treatment support the published reports that TBI-based high-dose therapy followed by autoSCT is highly effective in this prognostically unfavorable disease situation.
Collapse
Affiliation(s)
- Kai Kröger
- Department of Radiation Oncology, University Hospital of Muenster, Albert-Schweitzer Campus 1, Building 1A, 48419 Muenster, Germany
| | - Jan Siats
- Department of Radiation Oncology, University Hospital of Muenster, Albert-Schweitzer Campus 1, Building 1A, 48419 Muenster, Germany
| | - Andrea Kerkhoff
- Bone Marrow Transplantation Unit, Department of Hematology and Oncology, University Hospital of Muenster, Albert-Schweitzer Campus 1, Building 1A, 48419 Muenster, Germany
| | - Georg Lenz
- Bone Marrow Transplantation Unit, Department of Hematology and Oncology, University Hospital of Muenster, Albert-Schweitzer Campus 1, Building 1A, 48419 Muenster, Germany
| | - Matthias Stelljes
- Bone Marrow Transplantation Unit, Department of Hematology and Oncology, University Hospital of Muenster, Albert-Schweitzer Campus 1, Building 1A, 48419 Muenster, Germany
| | - Hans Theodor Eich
- Department of Radiation Oncology, University Hospital of Muenster, Albert-Schweitzer Campus 1, Building 1A, 48419 Muenster, Germany
| | - Gabriele Reinartz
- Department of Radiation Oncology, University Hospital of Muenster, Albert-Schweitzer Campus 1, Building 1A, 48419 Muenster, Germany
| |
Collapse
|
14
|
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
|
15
|
Martin P, Cohen JB, Wang M, Kumar A, Hill B, Villa D, Switchenko JM, Kahl B, Maddocks K, Grover NS, Qi K, Parisi L, Daly K, Zhu A, Salles G. Treatment Outcomes and Roles of Transplantation and Maintenance Rituximab in Patients With Previously Untreated Mantle Cell Lymphoma: Results From Large Real-World Cohorts. J Clin Oncol 2023; 41:541-554. [PMID: 35763708 PMCID: PMC9870229 DOI: 10.1200/jco.21.02698] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
PURPOSE Commonly used first-line (1L) treatments for mantle cell lymphoma include high-dose cytarabine-based induction followed by autologous stem-cell transplant (ASCT) for younger patients and several chemoimmunotherapy regimens for older patients. Continuous debates exist on the role of ASCT in younger patients and maintenance rituximab (MR) after bendamustine plus rituximab (BR). METHODS Retrospective data from 4,216 patients with mantle cell lymphoma in the Flatiron Health electronic record-derived deidentified database diagnosed between 2011 and 2021, mostly in US community oncology settings, were evaluated for treatment patterns and outcomes. The efficacy findings with ASCT and MR were validated in an independent cohort of 1,168 patients from 12 academic centers. RESULTS Among 3,614 patients with documented 1L treatment, BR was the most used. Among 1,265 patients age < 65 years, 30.5% received cytarabine-based induction and 23.5% received ASCT. There was no significant association between ASCT and real-world time to next treatment (hazard ratio [HR], 0.84; 95% CI, 0.68 to 1.03; P = .10) or overall survival (HR, 0.86; 95% CI, 0.63 to 1.18; P = .4) among ASCT-eligible patients. Among MR-eligible patients, MR after BR versus BR alone was associated with a longer real-world time to next treatment (HR, 1.96; 95% CI, 1.61 to 2.38; P < .001) and overall survival (HR, 1.51; 95% CI, 1.19 to 1.92; P < .001). The efficacy findings were consistent in the validation cohort. CONCLUSION In this large cohort of patients treated primarily in the US community setting, only one in four young patients received cytarabine or ASCT consolidation, suggesting the need to develop treatments that can be delivered effectively in routine clinical practice. Together with the validation cohort, data support future clinical trials exploring regimens without ASCT consolidation in young patients, whereas MR should be considered for patients after 1L BR and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone.
Collapse
Affiliation(s)
- Peter Martin
- Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY
| | - Jonathon B Cohen
- Department of Hematology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anita Kumar
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Brian Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Diego Villa
- BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, British Columbia, Canada
| | - Jeffrey M Switchenko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Brad Kahl
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO
| | - Kami Maddocks
- Arthur G James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Natalie S Grover
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Keqin Qi
- Janssen Research and Development, Titusville, NJ
| | - Lori Parisi
- Janssen Research and Development, Oncology, Raritan, NJ
| | | | - Angeline Zhu
- Janssen Research and Development, Oncology, Raritan, NJ
| | - Gilles Salles
- Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
16
|
Fenske TS. Frontline Therapy in Mantle Cell Lymphoma: When Clinical Trial and Real-World Data Collide. J Clin Oncol 2023; 41:452-459. [PMID: 36170622 DOI: 10.1200/jco.22.01661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice.A large number of frontline treatment options exist for mantle cell lymphoma (MCL), making selection of therapy a challenge for the clinician. In this Oncology Grand Rounds article, the case of a 73-year-old woman with MCL who attained remission with bendamustine and rituximab is presented. The relevant literature regarding frontline therapy is then reviewed, with particular focus on selection of induction regimen and the potential roles for autologous transplantation and/or rituximab maintenance. This literature primarily consists of prospective phase 2 and phase 3 clinical trials; however, added to this literature now is a growing body of large retrospective real-world cohorts, such as the new analysis by Martin et al,35 the manuscript that accompanies this Oncology Grand Rounds article. In some cases, the real-world evidence is at odds with data from prospective clinical trials, such as regarding the role of rituximab maintenance after bendamustine plus rituximab induction. These important new real-world data are put into context of an ever-changing treatment landscape, in hopes of aiding clinicians in frontline treatment selection for patients with MCL.
Collapse
|
17
|
Albanyan O, Alkassis S, Kim S, Kin A, Alavi A, Ayash L, Ratanatharathorn V, Modi D, Uberti JP, Deol A. Impact of Pre-transplant Induction Therapy on Outcomes of Patients Who Undergo Autologous Stem Cell Transplantation for Mantle Cell Lymphoma in First Complete Remission. Hematol Oncol Stem Cell Ther 2023; 16:154-157. [PMID: 34425096 DOI: 10.1016/j.hemonc.2021.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 01/31/2023] Open
Abstract
Mantle cell lymphoma is a rare subtype of non-Hodgkin's lymphoma with poor prognosis and continue to be challenging to treat. The choice of first line induction regimen remains a topic of debate due paucity of clinical trials. We retrospectively evaluated 66 patients diagnosed with mantle cell lymphoma who achieved first complete response after induction chemotherapy followed by autologous stem cell transplant. Treatment groups were divided into low-intensity versus high-intensity regimens. Our data showed the intensity of induction regimen does not impact posttransplant outcomes of mantle cell lymphoma who underwent autologous stem cell transplant in first complete response.
Collapse
Affiliation(s)
- Omar Albanyan
- Department of Oncology, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Samer Alkassis
- Department of Internal Medicine, Wayne State University, Detroit, MI, USA
| | - Seongho Kim
- Biostatistics Core, Department of Oncology, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Andrew Kin
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Asif Alavi
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Lois Ayash
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Voravit Ratanatharathorn
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Dipenkumar Modi
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Joseph P Uberti
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Abhinav Deol
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| |
Collapse
|
18
|
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: 1] [Impact Index Per Article: 0.3] [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
|
19
|
Al-Mansour M. Treatment Landscape of Relapsed/Refractory Mantle Cell Lymphoma: An Updated Review. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e1019-e1031. [PMID: 36068158 DOI: 10.1016/j.clml.2022.07.017] [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: 05/30/2022] [Revised: 07/22/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Mantle cell lymphoma (MCL) accounts for nearly 2-6% of all non-Hodgkin lymphoma (NHL) cases, with a steady incidence increase over the past few decades. Although many patients achieve an adequate response to the upfront treatment, the short duration of remission with rapid relapse is challenging during MCL management. In this regard, there is no consensus on the best treatment options for relapsed/refractory (R/R) disease, and the international guidelines demonstrate wide variations in the recommended approaches. The last decade has witnessed the introduction of new agents in the treatment landscape of R/R MCL. Since the introduction of Bruton's tyrosine kinase (BTK) inhibitors, the treatment algorithm and response of R/R MCL patients have dramatically changed. Nevertheless, BTK resistance is common, necessitating further investigations to develop novel agents with a more durable response. Novel agents targeting the B-cell receptor (BCR) signaling have exhibited clinical activity and a well-tolerable safety profile. However, as the responses to these novel agents are still modest in most clinical trials, combination strategies were investigated in pre-clinical and early clinical settings to determine whether the combination of novel agents would exhibit a better durable response than single agents. In this report, we provide an updated literature review that covers recent clinical data about the safety and efficacy of novel therapies for the management of R/R MCL.
Collapse
Affiliation(s)
- Mubarak Al-Mansour
- Adult Medical Oncology, Princess Noorah Oncology Center, Jeddah, Saudi Arabia; College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.
| |
Collapse
|
20
|
Treatment of Mantle Cell Lymphoma in the Frontline Setting: Are We Ready for a Risk-Adapted Approach? J Pers Med 2022; 12:jpm12071134. [PMID: 35887631 PMCID: PMC9324979 DOI: 10.3390/jpm12071134] [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: 05/16/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 12/29/2022] Open
Abstract
Mantle cell lymphoma (MCL), a type of B-cell non-Hodgkin lymphoma characterized by the t(11;14)(q13q32) translocation, is a clinically heterogenous disease which can range from indolent to highly aggressive. Numerous prognostic factors have been identified, including blastoid histology, the Mantle Cell Lymphoma International Prognostic Index (MIPI) score, high proliferation index, p53 deletions and/or mutations, complex karyotype, minimal residual disease, and several others. However, using these prognostic factors to guide treatment selection has largely remained elusive. Given the heterogeneous behavior of this disease and varying patient characteristics, we suggest that the time has come for a more risk-adapted approach to this disease. In this article, we review the numerous prognostic factors that have been described for MCL, both at the time of diagnosis and following first-line treatment. We then propose a risk-adapted approach to first-line therapy for MCL, which would reserve intensive therapy for the highest risk patients and spare others excessive toxicity.
Collapse
|
21
|
Affiliation(s)
- James O Armitage
- From the Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska, Omaha (J.O.A.)
| | - Dan L Longo
- From the Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska, Omaha (J.O.A.)
| |
Collapse
|
22
|
Liu H, Shi X, Fang H, Cao L, Miao Y, Zhao X, Wu W, Xu W, Li J, Fan L. First-Line Autologous Stem Cell Transplantation for Mantle Cell Lymphoma: A Systematic Analysis and Treatment Recommendation. Front Oncol 2022; 12:881346. [PMID: 35646653 PMCID: PMC9130771 DOI: 10.3389/fonc.2022.881346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background In the era of immunotherapy, autologous stem cell transplantation (ASCT) in first-line therapy in patients with mantle cell lymphoma (MCL) has been a controversial topic. This report aimed to explore the association between ASCT and MCL survival through a systematic review with meta-analysis. Methods We performed a systematic search of original articles published from inception to September 2021 using PubMed, MEDLINE, Embase, and Cochrane Library databases. Results We included studies that compared ASCT with non-ASCT consolidation in newly diagnosed transplant-eligible MCL. The endpoints were progression-free survival (PFS) and overall survival (OS). There were seven eligible studies (one randomized clinical trial, one prospective cohort study, and five observational studies) published between 2012 and 2021, in which the total number of participants was 3,271. In the non-intensive induction subgroup, patients with ASCT experienced a significant PFS but no OS benefit compared with those without ASCT. In the intensive induction subgroup, the PFS benefit from ASCT still existed but largely attenuated; no OS benefit was observed though only one study was suitable for evaluation. When compared to the rituximab maintenance arm, ASCT had a worse PFS and OS. Conclusions In the rituximab plus HiDAC era, the benefit of ASCT as a component of first-line treatment has been weakened. First-line maintenance strategy instead of ASCT seems worth exploring .
Collapse
Affiliation(s)
- Hailing Liu
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China.,Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Xiao Shi
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China.,Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Huizi Fang
- Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China
| | - Lei Cao
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China.,Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Yi Miao
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China.,Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaoli Zhao
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China.,Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Wei Wu
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China.,Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Wei Xu
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China.,Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Jianyong Li
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China.,Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Lei Fan
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China.,Nanjing Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Pukou CLL Center, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
23
|
Tarockoff M, Gonzalez T, Ivanov S, Sandoval-Sus J. Mantle Cell Lymphoma: the Role of Risk-Adapted Therapy and Treatment of Relapsed Disease. Curr Oncol Rep 2022; 24:1313-1326. [PMID: 35639332 DOI: 10.1007/s11912-022-01297-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW In this review, the current treatment strategies are recapped, evolving agents are discussed, and we provide guidance in treating R/R MCL. RECENT FINDINGS There has been an advancement in treatment using targeted therapy, cellular therapies including chimeric antigen receptor (CAR) T cell therapy and hematopoietic stem cell transplantation (HSCT) and novel therapeutic agents including non-covalent BTKis, bispecific antibodies, and antibody-drug conjugates for treatment of refractory and relapsed mantle cell lymphoma. Mantle cell lymphoma (MCL) is a mature B-cell lymphoma that is associated with a poor prognosis. Current treatments include immunochemotherapy, chemotherapy and autologous stem cell transplantation (SCT) which place patients in remission but result in relapse. Chemoimmunotherapy uses chemotherapeutic agents paired with rituximab in patients who have chemo-sensitive disease with prolonged remission of at least > 2 years and/or have contraindications to chemotherapy that serve as bridges to more definitive treatment. Additional therapies including proteosome inhibitor-based therapies and immunomodulators, like bortezomib and lenalidomide, can be used as single agents or in combination with others. Bruton's tyrosine kinase (BTK) inhibitors including ibrutinib, acalaburtinib, and zanubrutinib have also been proven effective for the treatment of (R/R) disease. Another agent is Venetoclax, a robust drug that can be used in MCL after progression or intolerance to BTKi. Newer advances in the management of MCL have led to the utilization of cellular therapies including chimeric antigen receptor (CAR) T cell therapy and SCT that are options for healthy young (< 65 years old) who have progressed through several lines of therapies. With progression of disease, mutations are acquired that cause therapy resistance. Novel therapeutic agents such as non-covalent BTKis, bispecific antibodies, and antibody-drug conjugates are paving the way for advancements in treatment for R/R MCL. R/R MCL is a complex disease with many therapeutic options none of which has been proven superior in head-to-head comparison. In this review, the current treatment strategies are recapped, evolving agents are discussed, and we provide guidance in treating R/R MCL.
Collapse
Affiliation(s)
- Meri Tarockoff
- Department of Hematology and Oncology, Memorial Healthcare System, Hollywood, USA
| | - Teresita Gonzalez
- Department of Hematology and Oncology, Memorial Healthcare System, Hollywood, USA
| | - Stanislav Ivanov
- Department of Hematology and Oncology, Memorial Healthcare System, Hollywood, USA
| | - Jose Sandoval-Sus
- Department of Hematology and Oncology, Memorial Healthcare System, Hollywood, USA. .,Moffitt Malignant Hematology & Cellular Therapy at Memorial Healthcare System Memorial Cancer Institute, 603 N. Flamingo Rd., Suite 151, Pembroke Pines, FL, 33028, USA.
| |
Collapse
|
24
|
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: 59] [Impact Index Per Article: 19.7] [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
|
25
|
Mehraj U, Mushtaq U, Mir MA, Saleem A, Macha MA, Lone MN, Hamid A, Zargar MA, Ahmad SM, Wani NA. Chemokines in Triple-Negative Breast Cancer Heterogeneity: New Challenges for Clinical Implications. Semin Cancer Biol 2022; 86:769-783. [PMID: 35278636 DOI: 10.1016/j.semcancer.2022.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022]
Abstract
Tumor heterogeneity is a hallmark of cancer and one of the primary causes of resistance to therapies. Triple-negative breast cancer (TNBC), which accounts for 15% to 20% of all breast cancers and is the most aggressive subtype, is very diverse, connected to metastatic potential and response to therapy. It is a very diverse disease at the molecular, pathologic, and clinical levels. TNBC is substantially more likely to recur and has a worse overall survival rate following diagnosis than other breast cancer subtypes. Chemokines, low molecular weight proteins that stimulate chemotaxis, have been shown to control the cues responsible for TNBC heterogeneity. In this review, we have focused on tumor heterogeneity and the role of chemokines in modulating tumor heterogeneity, since this is the most critical issue in treating TNBC. Additionally, we examined numerous cues mediated by chemokine networks that contribute to the heterogeneity of TNBC. Recent developments in our knowledge of the chemokine networks that regulate TNBC heterogeneity may pave the door for developing difficult-to-treat TNBC treatment options.
Collapse
Affiliation(s)
- Umar Mehraj
- Department of Bioresources, School of Life Sciences, University of Kashmir, Srinagar, Jammu & Kashmir India
| | - Umer Mushtaq
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Manzoor A Mir
- Department of Bioresources, School of Life Sciences, University of Kashmir, Srinagar, Jammu & Kashmir India
| | - Afnan Saleem
- Division of Animal Biotechnology Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Sher-e- Kashmir University of Agricultural Sciences and Technology-Kashmir, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science & Technology Awantipora, Jammu & Kashmir, India
| | - Mohammad Nadeem Lone
- Department of Chemistry, School of Physical & Chemical Sciences, Central University of Kashmir, Ganderbal J & K, India
| | - Abid Hamid
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Mohammed A Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Syed Mudasir Ahmad
- Division of Animal Biotechnology Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Sher-e- Kashmir University of Agricultural Sciences and Technology-Kashmir, India
| | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India.
| |
Collapse
|
26
|
Pu JJ, Savani M, Huang N, Epner EM. Mantle cell lymphoma management trends and novel agents: where are we going? Ther Adv Hematol 2022; 13:20406207221080743. [PMID: 35237397 PMCID: PMC8882940 DOI: 10.1177/20406207221080743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 01/31/2022] [Indexed: 11/20/2022] Open
Abstract
The heterogeneity in disease pathology, the unpredictability in disease
prognosis, and the variability in response to therapy make mantle cell lymphoma
(MCL) a focus of novel therapeutic development. MCL is characterized by
dysregulated expression of cyclin D1 through a chromosome
t(11;14) translocation. MCL international prognostic index
(MIPI), ki-67 proliferation index, and TP53
mutation status are currently utilized for prognostication. With advances in
pharmacokinetic analysis and drug discovery, treatment strategy has evolved from
chemotherapy to combination of targeted, epigenetic, and immune therapies. In
this review, we discuss investigational and newly approved treatment approaches.
In a short time, the US Food and Drug Administration (FDA) has approved five
agents for the treatment of MCL: lenalidomide, an immunomodulatory agent;
bortezomib, a proteasome inhibitor; and ibrutinib, acalabrutinib, and
zanubrutinib, all Bruton kinase inhibitors. Epigenetic agents (e.g. cladribine
and vorinostat), mammalian target of rapamycin (mTOR) inhibitors (e.g.
temsirolimus and everolimus), and monoclonal antibodies and/or antibody-drug
conjugates (e.g. obinutuzumab, polatuzumab, and ublituximab) are promising
therapeutic agents currently under clinical trial investigation. Most recently,
chimeric antigen receptor (CAR)-T cell therapy and bispecific T-cell engager
(BiTE) therapy even open a new venue for MCL treatment. However, due to its
intricate pathology nature and high relapse incidence, there are still unmet
needs in developing optimal therapeutic strategies for both frontline and
relapsed/refractory settings. The ultimate goal is to develop innovative
personalized combination therapy approaches for the purpose of delivering
precision medicine to cure this disease.
Collapse
Affiliation(s)
- Jeffrey J. Pu
- University of Arizona Cancer Center, 1515 N Campbell Avenue, Room #1968C, Tucson, AZ 85724, USA
| | - Malvi Savani
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Nick Huang
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Elliot M. Epner
- Penn State Hershey Cancer Institute, 100 University Drive, Hershey, PA, USA
| |
Collapse
|
27
|
Evolving frontline immunochemotherapy for mantle cell lymphoma and the impact on survival outcomes. Blood Adv 2022; 6:1350-1360. [PMID: 34662895 PMCID: PMC8864651 DOI: 10.1182/bloodadvances.2021005715] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022] Open
Abstract
Because there have been a dvances in frontline treatment for mantle cell lymphoma (MCL) over the last 2 decades, we sought to characterize the changes in frontline treatment patterns and their association with outcomes. Patients with newly diagnosed MCL from September 2002 through June 2015 were enrolled in a prospective cohort study, and clinical characteristics, treatment, and clinical outcomes were compared between patients diagnosed from 2002 to 2009 (Era 1) compared with 2010 to 2015 (Era 2). Patient age, sex, and simplified MCL International Prognostic Index (sMIPI) score were similar between the 2 groups. In patients age 65 years or younger, there was less use of rituximab plus hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone (R-Hyper-CVAD) (16.1% vs 8.8%) but more use of rituximab plus maximum-strength cyclophosphamide, doxorubicin, vincristine, and prednisone (R-maxi-CHOP) alternating with rituximab plus high-dose cytarabine (R-HiDAC), also known as the Nordic regimen, and R-CHOP alternating with rituximab plus dexamethasone, high-dose cytarabine, and cisplatin (R-DHAP) (1.1% vs 26.4%) and less use of R-CHOP or R-CHOP-like regimens (64.5% vs 35.2%) but more use of R-bendamustine (0% vs 12.1%) in Era 2 (P < .001). These changes were associated with improved event-free survival (EFS; 5-year EFS, 34.3% vs 50.0%; P = .010) and overall survival (OS; 5-year OS, 68.8% vs 81.6%; P = .017) in Era 2. In patients older than age 65 years, there was less use of R-CHOP or R-CHOP-like therapy (39.0% vs 14.3%) and nonstandard systemic therapy (36.6% vs 13.0%) but more use of R-bendamustine (0% vs 49.4%). These changes were associated with a trend for improved EFS (5-year EFS, 25.4% vs 37.5%; P = .051) in Era 2. The shift from R-CHOP or R-CHOP-like regimens to R-bendamustine was associated with improved EFS (5-year EFS, 25.0% vs 44.6%; P = .008) in Era 2. Results from this prospective cohort study provide critical real-world evidence for improved outcomes with evolving frontline patterns of care in patients with MCL.
Collapse
|
28
|
Yanada M, Yamamoto K. Hematopoietic cell transplantation for mantle cell lymphoma. Int J Hematol 2022; 115:301-309. [DOI: 10.1007/s12185-022-03294-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 11/28/2022]
|
29
|
Wu M, Li Y, Huang H, Xu W, Wang Y, Huang H, Zhao W, Liu S, Xu P, Chen Z, Zhu J, Song Y, Ruan J, Wu D. Initial Treatment Patterns and Survival Outcomes of Mantle Cell Lymphoma Patients Managed at Chinese Academic Centers in the Rituximab Era: A Real-World Study. Front Oncol 2022; 11:770988. [PMID: 35059312 PMCID: PMC8763847 DOI: 10.3389/fonc.2021.770988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/06/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose The aim of the study was to delineate the disease characteristics, the initial treatment patterns, and survival in patients with mantle cell lymphoma (MCL) managed in the real world. Methods Data of 518 MCL patients from 5 major Chinese Hematology Centers in the period from 2007 to 2017 were retrospectively analyzed. Results The median age was 58 years. Of the patients, 88.6% had Eastern Cooperative Oncology Group Performance Status (ECOG PS) 0–1 and 80.7% had advanced-stage disease. Ki67 expression was <30% in 39.6% of the patients, and 43.2% of patients were categorized into a low-risk group based on the Mantle Cell Lymphoma International Prognostic Index (MIPI) scoring system. Overall, 73.4% of the patients received rituximab as their first-line therapy. The most commonly used chemotherapy was the CHOP-like (cyclophosphamide, hydroxydaunomycin, oncovin, and prednisone) regimen (45.2%), followed by high-dose cytarabine-containing chemotherapy (31.3%) and bendamustine (3.3%). Of the patients, 13.7% (n = 71) underwent consolidative autologous stem cell transplantation (ASCT), and 19.3% (n = 100) received novel agents containing first-line regimens. With a median follow-up time of 52 months, the 3- and 5-year overall survival (OS) rates were 73.7% and 61.4%, respectively. Age ≤60 years, ECOG PS 0–1, stages I–II, normal lactate dehydrogenase (LDH), absence of bone marrow involvement, Ki67 <30%, and lower-risk IPI/MIPI scores were significantly associated with improved OS (p < 0.05). The inclusion of rituximab improved the 5-year OS, with borderline significance (62.5% vs. 55.2%, p = 0.076). High-dose cytarabine-containing chemotherapy showed significant clinical benefit in 5-year OS (72.1% vs. 55.9%, p = 0.010). Patients with ASCT had better 5-year OS in the younger (≤60 years) age group (87.2% vs. 64.8%, p = 0.002). Conclusion This large retrospective dataset unequivocally confirmed the survival advantage afforded by cytarabine-containing regimen and ASCT in a first-line setting under real-world management in the rituximab era.
Collapse
Affiliation(s)
- Meng Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yun Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Huiqiang Huang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yanyan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiwen Huang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Weili Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Pengpeng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengming Chen
- Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY, United States
| | - Jun Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yuqin Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jia Ruan
- Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY, United States
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
30
|
Is There Still a Role for Transplant for Patients with Mantle Cell Lymphoma (MCL) in the Era of CAR-T Cell Therapy? Curr Treat Options Oncol 2022; 23:1614-1625. [PMID: 36227407 PMCID: PMC9557996 DOI: 10.1007/s11864-022-01020-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2022] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT For years, upfront autologous hematopoietic cell transplant (auto-HCT) has been the standard of care for younger and physically fit mantle cell lymphoma (MCL) patients after chemoimmunotherapy (CIT) induction. Bruton's tyrosine kinase (BTK) inhibitors have proven to be excellent salvage therapies, but their durability remains a question, especially in high-risk (HR) MCL. Allogeneic HCT (allo-HCT) was the only option for long-term remission and possibly cure for MCL relapse after auto-HCT and sometime as upfront consolidation for a young patient with HR MCL (debatable). We have seen a paradigm shift since the FDA approval in July 2020 of the brexucabtagene autoleucel chimeric antigen receptor T (CAR-T) cell therapy for relapsed and refractory (R/R) MCL with an preliminary evidence suggesting CAR-T may overcome known biological risk factors in MCL. Given its safety profile and excellent efficacy, the role of CAR-T among other approved therapies and HCT may need to be better defined. Based on the current evidence, auto-HCT remains a standard frontline consolidation therapy. CAR-T therapy is a preferred option for patients with relapsed/refractory (R/R) MCL, particularly those who failed BTK inhibitors. In certain high-risk MCL patients (such as high ki 67, TP53 alterations, complex karyotype, blastoid morphology, early relapse after initial diagnosis), CAR-T cell therapy may be considered before BTK inhibitors (preferably on a clinical trial). The role of allo-HCT is unclear in the CAR-T era, but remains a viable option for eligible patients who have no access or who have failed CAR-T therapy. Our review discusses current standards and the shifting paradigms in the indications for HCT and the role of CAR-T cell therapy for MCL. Prospective studies tailored based on risk factors are needed to better define the optimal sequences of HCT and cellular therapy and other approved novel therapies.
Collapse
|
31
|
Karmali R, Switchenko JM, Goyal S, Shanmugasundaram K, Churnetski MC, Kolla B, Bachanova V, Gerson JN, Barta SK, Gordon MJ, Danilov AV, Grover NS, Epperla N, Mathews S, Burkart M, Sawalha Y, Hill BT, Ghosh N, Park SI, Bond DA, Maddocks KJ, Badar T, Fenske TS, Hamadani M, Guo J, Malecek M, Kahl BS, Martin P, Blum KA, Flowers CR, Cohen JB. Multi-center analysis of practice patterns and outcomes of younger and older patients with mantle cell lymphoma in the rituximab era. Am J Hematol 2021; 96:1374-1384. [PMID: 34324220 DOI: 10.1002/ajh.26306] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022]
Abstract
Clinical outcomes and predictors of survival in patients with newly diagnosed mantle cell lymphoma (MCL) treated in the rituximab era (2000-2015) at 12 US academic centers were assessed to identify determinants of survival across age groups. Objectives were to characterize and compare practice patterns, outcomes and prognostic factors for survival in younger patients (age < 65) and older patients (age ≥ 65 years). Among 1162 patients included, 697 were younger and 465 were older. In younger patients, 2-year progression free survival (PFS) and overall survival (OS) rates were 79% and 92% respectively; blastoid histology, ECOG ≥ 2, and lack of maintenance rituximab (MR) remained statistically relevant to poor OS on univariate analysis (UVA) and multivariate analysis (MVA). In older patients, 2-year PFS and OS rates were 67% and 86% respectively; lack of maintenance rituximab remained significantly associated with inferior PFS and OS on UVA and MVA (p < 0.001). Two-year PFS rates were 79%, and 67% and 2-year OS rates were 92% and 86% for ages < 65 and ≥ 65 respectively (p < 0.001). First-line high-dose cytarabine exposure and/or MR lessened the negative impact of age on survival. Taken collectively, survival outcomes for older patients remain inferior to those of younger patients in the rituximab era. However, maintenance rituximab and potentially high-dose cytarabine-based induction can mitigate the negative impact of age on survival.
Collapse
Affiliation(s)
- Reem Karmali
- Northwestern University Feinberg School of Medicine; Robert H. Lurie Comprehensive Cancer Center Chicago Illinois USA
| | | | - Subir Goyal
- Department of Biostatistics and Bioinformatics Emory University Atlanta Georgia USA
| | | | | | | | | | | | - Stefan K. Barta
- Fox Chase Cancer Center Philadelphia Pennsylvania USA
- University of Pennsylvania Philadelphia Pennsylvania USA
| | - Max J. Gordon
- Knight Cancer Institute Oregon Health & Science University Portland Oregon USA
| | - Alexey V. Danilov
- Knight Cancer Institute Oregon Health & Science University Portland Oregon USA
| | - Natalie S. Grover
- University of North Carolina, Chapel Hill, Lineberger Comprehensive Cancer Center Chapel Hill North Carolina USA
| | | | - Stephanie Mathews
- University of North Carolina, Chapel Hill, Lineberger Comprehensive Cancer Center Chapel Hill North Carolina USA
| | - Madelyn Burkart
- Northwestern University Feinberg School of Medicine; Robert H. Lurie Comprehensive Cancer Center Chicago Illinois USA
| | - Yazeed Sawalha
- The Ohio State University Columbus Ohio USA
- Cleveland Clinic Cleveland Ohio USA
| | | | - Nilanjan Ghosh
- Levine Cancer Institute, Atrium Health Charlotte North Carolina USA
| | - Steven I. Park
- Levine Cancer Institute, Atrium Health Charlotte North Carolina USA
| | | | | | - Talha Badar
- Medical College of Wisconsin Milwaukee Wisconsin USA
| | | | | | - Jin Guo
- Weill Cornell Medical College Manhattan New York USA
| | - Mary Malecek
- Washington University in St. Louis St Louis Missouri USA
| | - Brad S. Kahl
- Washington University in St. Louis St Louis Missouri USA
| | - Peter Martin
- Weill Cornell Medical College Manhattan New York USA
| | - Kristie A. Blum
- Winship Cancer Institute of Emory University Atlanta Georgia USA
- The Ohio State University Columbus Ohio USA
| | | | | |
Collapse
|
32
|
Riedell PA, Hamadani M, Ahn KW, Litovich C, Murthy GSG, Locke FL, Brunstein CG, Merryman RW, Stiff PJ, Pawarode A, Nishihori T, Kharfan-Dabaja MA, Herrera AF, Sauter CS, Smith SM. Outcomes and Utilization Trends of Front-Line Autologous Hematopoietic Cell Transplantation for Mantle Cell Lymphoma. Transplant Cell Ther 2021; 27:911.e1-911.e7. [PMID: 34450333 PMCID: PMC8556305 DOI: 10.1016/j.jtct.2021.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
Although autologous hematopoietic cell transplantation (auto-HCT) has become a common practice for eligible patients in the front-line setting with mantle cell lymphoma (MCL), there are limited data regarding trends in auto-HCT utilization and associated outcomes. This study used the Center for International Blood and Marrow Transplant Research (CIBMTR) database to evaluate survival outcomes and auto-HCT utilization in adults age ≥18 years who underwent auto-HCT within 12 months of diagnosis of MCL between January 2000 and December 2018. The 19-year period from 2000 to 2018 was divided into 4 separate intervals-2000 to 2004, 2005 to 2009, 2010 to 2014, and 2015 to 2018-and encompassed 5082 patients. To evaluate transplantation utilization patterns, we combined MCL incidence derived from the SEER 21 database with CIBMTR- reported auto-HCT activity within 12 months of diagnosis of MCL. Primary outcomes included overall survival (OS) along with the auto-HCT utilization rate. The cumulative incidence of nonrelapse mortality at 1 year decreased from 7% in the earliest cohort (2000 to 2004) to 2% in the latest cohort (2015 to 2018). Mirroring this trend, OS outcomes improved continually with time, with a 3-year OS of 72% in the earliest cohort improving to 86% in the latest cohort. In addition, we noted an increase in auto-HCT utilization from 2001 to 2018, particularly in patients age ≤65 years. This large retrospective analysis highlights trends in auto-HCT utilization and outcomes in patients with MCL and emphasizes the need to optimize pretransplantation and post-transplantation treatment strategies to enhance survival outcomes.
Collapse
Affiliation(s)
- Peter A Riedell
- Division of Hematology and Oncology, University of Chicago Medicine, Chicago, Illinois
| | - Mehdi Hamadani
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - Kwang W Ahn
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Carlos Litovich
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Claudio G Brunstein
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Reid W Merryman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Masachusetts
| | - Patrick J Stiff
- Division of Hematology-Oncology, Department of Medicine, Loyola University Stritch School of Medicine, Maywood, Illinois
| | - Attaphol Pawarode
- Blood and Marrow Transplantation Program, Division of Hematology/Oncology, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Craig S Sauter
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sonali M Smith
- Division of Hematology and Oncology, University of Chicago Medicine, Chicago, Illinois
| |
Collapse
|
33
|
Sawalha Y, Radivoyevitch T, Jia X, Tullio K, Dean RM, Pohlman B, Hill BT, Kalaycio M, Majhail NS, Jagadeesh D. The impact of socioeconomic disparities on the use of upfront autologous stem cell transplantation for mantle cell lymphoma. Leuk Lymphoma 2021; 63:335-343. [PMID: 34521300 DOI: 10.1080/10428194.2021.1978085] [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] [Indexed: 10/20/2022]
Abstract
Using the National Cancer Database, we identified 10,290 patients with newly diagnosed mantle cell lymphoma (MCL) treated with chemotherapy with or without upfront autologous stem cell transplantation (ASCT). Only 17% of patients underwent ASCT. Patients who underwent ASCT were younger and more likely to have lower comorbidity scores, private insurance, higher income and education, and treatment received at an academic facility. On multivariable analysis, age, comorbidity index, insurance type, the transition of care, facility type, distance to facility, and diagnosis year were predictive for ASCT use. ASCT use was associated with improved 5-year overall survival in younger (82% vs. 64%, p < .001) and older (70% vs. 40%, p < .001) patients, which was retained in the matched propensity score and 12-month analyses. Female gender, the diagnosis year ≥2009, private insurance, higher income, and education were associated with superior survival, whereas Black race and higher comorbidities predicted inferior survival.
Collapse
Affiliation(s)
- Yazeed Sawalha
- Arthur G. James Comprehensive Cancer Center, Department of Internal Medicine, Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tomas Radivoyevitch
- Quantitative Health Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Xuefei Jia
- Quantitative Health Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Katherine Tullio
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Robert M Dean
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Brad Pohlman
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Brian T Hill
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Matt Kalaycio
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Navneet S Majhail
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Deepa Jagadeesh
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
34
|
Early Relapse Identifies MCL patients with Inferior Survival after Intensive or Less Intensive Frontline Therapy. Blood Adv 2021; 5:5179-5189. [PMID: 34516611 PMCID: PMC9153047 DOI: 10.1182/bloodadvances.2021004765] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/20/2021] [Indexed: 11/20/2022] Open
Abstract
POD<24 months was associated with inferior survival among patients with MCL after both intensive and less intensive frontline treatment. Patients with POD<6 months or progressive disease after frontline treatment had the highest risk of early mortality.
Although an expanding array of effective treatments has resulted in recent improvement in survival of patients with mantle cell lymphoma (MCL), outcomes remain heterogeneous, and identification of prognostic factors remains a priority. We assessed the prognostic impact of time to progression of disease (POD) after first-line therapy among 455 patients with relapsed MCL. Patients were categorized by duration of first remission as PRF/POD6, defined as progressive disease during induction or POD within 6 months of diagnosis (n = 65; 14%); POD6-24, defined as POD between 6 and 24 months after diagnosis (n = 153; 34%); and POD>24, defined as POD >24 months after diagnosis (n = 237; 53%). The median overall survival from POD (OS2) was 1.3 years (95% confidence interval [CI], 0.9-2.4) for patients with PRF/POD6, 3 years (95% CI, 2-6.8) for those with POD6-24, and 8 years (95% CI, 6.2-NR) for those with POD>24. Median OS2 was inferior in patients with early POD (defined as PRF/POD6 or POD6-24) after both intensive and less intensive frontline treatment. The prognostic performance of time until POD was replicated in an independent cohort of 245 patients with relapsed MCL, with median OS2 of 0.3 years (95% CI, 0.1-0.5) for PRF/POD6, 0.8 years (95% CI, 0.6-0.9) for POD6-24, and 2.4 years (95% CI 2.1-2.7) for POD>24. Early POD is associated with inferior OS2 in patients with relapsed MCL, identifying a high-risk population for future prospective studies.
Collapse
|
35
|
Munshi PN, Hamadani M, Kumar A, Dreger P, Friedberg JW, Dreyling M, Kahl B, Jerkeman M, Kharfan-Dabaja MA, Locke FL, Shadman M, Hill BT, Ahmed S, Herrera AF, Sauter CS, Bachanova V, Ghosh N, Lunning M, Kenkre VP, Aljurf M, Wang M, Maddocks KJ, Leonard JP, Kamdar M, Phillips T, Cashen AF, Inwards DJ, Sureda A, Cohen JB, Smith SM, Carlo-Stella C, Savani B, Robinson SP, Fenske TS. American Society of Transplantation and Cellular Therapy, Center of International Blood and Marrow Transplant Research, and European Society for Blood and Marrow Transplantation Clinical Practice Recommendations for Transplantation and Cellular Therapies in Mantle Cell Lymphoma. Transplant Cell Ther 2021; 27:720-728. [PMID: 34452722 PMCID: PMC8447221 DOI: 10.1016/j.jtct.2021.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 11/18/2022]
Abstract
Autologous (auto-) and allogeneic (allo-) hematopoietic cell transplantation (HCT) are accepted treatment modalities in contemporary treatment algorithms for mantle cell lymphoma (MCL). Chimeric antigen receptor (CAR) T cell therapy recently received approval for MCL; however, its exact place and sequence in relation to HCT remain unclear. The American Society of Transplantation and Cellular Therapy, Center of International Blood and Marrow Transplant Research, and the European Society for Blood and Marrow Transplantation jointly convened an expert panel to formulate consensus recommendations for role, timing, and sequencing of auto-HCT, allo-HCT, and CAR T cell therapy for patients with newly diagnosed and relapsed/refractory (R/R) MCL. The RAND-modified Delphi method was used to generate consensus statements. Seventeen consensus statements were generated, with a few key statements as follows: in the first line setting, auto-HCT consolidation represents standard of care in eligible patients, whereas there is no clear role of allo-HCT or CAR T cell therapy outside of clinical trials. In the R/R setting, the preferential option is CAR T cell therapy, especially in patients with MCL failing or intolerant to at least one Bruton's tyrosine kinase inhibitor, while allo-HCT is recommended if CAR T cell therapy fails or is infeasible. Several recommendations were based on expert opinion, where the panel developed consensus statements for important real-world clinical scenarios to guide clinical practice. In the absence of contemporary evidence-based data, the panel found RAND-modified Delphi methodology effective in providing a formal framework for developing consensus recommendations for the timing and sequence of cellular therapies for MCL.
Collapse
Affiliation(s)
- Pashna N Munshi
- Department of Blood and Marrow Transplantation, MedStar Georgetown University Hospital, Washington, DC
| | - Mehdi Hamadani
- Center for International Blood & Marrow Transplant Research and the BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - Ambuj Kumar
- Department of Internal Medicine, Office of Research, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Peter Dreger
- Department of Medicine, University of Heidelberg, Germany
| | | | - Martin Dreyling
- Department of Medicine III, LMU Hospital Munich, Munich, Germany
| | - Brad Kahl
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Mats Jerkeman
- Department of Oncology, Skane University Hospital, Lund University, Lund, Sweden
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapies Program, Mayo Clinic, Jacksonville, Florida
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Mazyar Shadman
- Blood and Marrow Transplantation Program, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Sairah Ahmed
- Department of Lymphoma, Myeloma and Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Craig S Sauter
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Nilanjan Ghosh
- Levine Cancer Institute, Atrium Health, Charlotte, Nnorth Carolina
| | - Matthew Lunning
- Divsion of Hematology and Oncology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Vaishalee P Kenkre
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mahmoud Aljurf
- Blood and Marrow Transplantation Program, King Faisal Specialist Hospital, Riyadh, Saudi Arabia
| | - Michael Wang
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kami J Maddocks
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - John P Leonard
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Manali Kamdar
- Division of Hematology, Hematologic Malignancies and Stem Cell Transplantation, University of Colorado Cancer Center, Denver, Colorado
| | - Tycel Phillips
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Amanda F Cashen
- Division of Oncology, Section of Stem Cell Transplantation, Washington University School of Medicine, St Louis, Missouri
| | | | - Anna Sureda
- Clinical Hematology Department, Catalan Institute of Oncology, Institut d'Investigació Biomèdica de Bellvitge IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | | | - Sonali M Smith
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Carmello Carlo-Stella
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milan, Italy
| | - Bipin Savani
- Blood and Marrow Transplantation Program, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Stephen P Robinson
- University Hospital Bristol NHS Foundation Trust, London, United Kingdom
| | - Timothy S Fenske
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
36
|
Zoellner AK, Unterhalt M, Stilgenbauer S, Hübel K, Thieblemont C, Metzner B, Topp M, Truemper L, Schmidt C, Bouabdallah K, Krauter J, Lenz G, Dürig J, Vergote V, Schäfer-Eckart K, André M, Kluin-Nelemans HC, van Hoof A, Klapper W, Hiddemann W, Dreyling M, Hoster E. Long-term survival of patients with mantle cell lymphoma after autologous haematopoietic stem-cell transplantation in first remission: a post-hoc analysis of an open-label, multicentre, randomised, phase 3 trial. LANCET HAEMATOLOGY 2021; 8:e648-e657. [PMID: 34450102 DOI: 10.1016/s2352-3026(21)00195-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Autologous haematopoietic stem-cell transplantation (HSCT) in first remission is the current standard treatment in fit patients with mantle cell lymphoma. In this long-term follow-up study, we aimed to evaluate the efficacy of autologous HSCT versus interferon alfa maintenance after chemotherapy without or with rituximab in patients with primary advanced-stage mantle cell lymphoma. METHODS We did a post-hoc, long-term analysis of an open-label, multicentre, randomised, phase 3 trial done in 121 participating hospitals or practices across six European countries. Patients who were aged 18-65 years with previously untreated stage III-IV mantle cell lymphoma and an ECOG performance score of 0-2 were eligible for participation. Patients were randomly assigned (1:1) to receive either myeloablative radiochemotherapy (fractionated total body irradiation with 12 Gy/day 6-4 days before autologous HSCT and cyclophosphamide 60 mg/kg per day intravenously 3-2 days before autologous HSCT) followed by autologous HSCT (the autologous HSCT group) or interferon alfa maintenance (the interferon alfa maintenance group; 6 × 106 IU three times a week subcutaneously until progression) after completion of CHOP-like induction therapy (cyclophosphamide 750 mg/m2 intravenously on day 1, doxorubicin 50 mg/m2 intravenously on day 1, vincristine 1·4 mg/m2 [maximum 2 mg] intravenously on day 1, and prednisone 100 mg/m2 orally on days 1-5; repeated every 21 days for up to 6 cycles) without or with rituximab (375 mg/m2 intravenously on day 0 or 1 of each cycle; R-CHOP). The primary outcome was progression-free survival from end of induction until progression or death among patients who had a remission and the secondary outcome was overall survival from the end of induction until death from any cause. We did comparisons of progression-free survival and overall survival according to the intention-to-treat principle between both groups among responding patients and explored efficacy in subgroups according to induction treatment without or with rituximab. Hazard ratios (HRs) were adjusted for the mantle cell lymphoma international prognostic index (MIPI) numerical score, and in the total group also for rituximab use (adjusted HR [aHR]). This trial was started before preregistration was implemented and is therefore not registered, recruitment is closed, and this is the final evaluation. FINDINGS Between Sept 30, 1996, and July 1, 2004, 269 patients were randomly assigned to receive either autologous HSCT or interferon alfa maintenance therapy. The median follow-up was 14 years (IQR 10-16), with the intention-to-treat population consisting of 174 patients (93 [53%] in the autologous HSCT group and 81 [47%] in the interferon alfa maintenance group) who responded to induction therapy. The median age was 55 years (IQR 47-60), and R-CHOP was used in 68 (39%) of 174 patients. The median progression-free survival was 3·3 years (95% CI 2·5-4·3) in the autologous HSCT group versus 1·5 years (1·2-2·0) in the interferon alfa maintenance group (log-rank p<0·0001; aHR 0·50 [95% CI 0·36-0·69]). The median overall survival was 7·5 years (95% CI 5·7-12·0) in the autologous HSCT group versus 4·8 years (4·0-6·6) in the interferon alfa maintenance group (log-rank p=0·019; aHR 0·66 [95% CI 0·46-0·95]). For patients treated without rituximab, the progression-free survival adjusted HR for autologous HSCT versus interferon alfa was 0·40 (0·26-0·61), in comparison to 0·72 (0·42-1·24) for patients treated with rituximab. For overall survival, the adjusted hazard ratio for HSCT versus interferon alfa was 0·52 (0·33-0·82) without rituximab and 1·05 (0·55-1·99) for patients who received rituximab. INTERPRETATION Our results confirm the long-term efficacy of autologous HSCT to treat mantle cell lymphoma established in the pre-rituximab era. The suggested reduced efficacy after immunochemotherapy supports the need for its re-evaluation now that antibody maintenance, high-dose cytarabine, and targeted treatments have changed the standard of care for patients with mantle cell lymphoma. FUNDING Deutsche Krebshilfe, the European Community, and the Bundesministerium für Bildung und Forschung, Kompetenznetz Maligne Lymphome.
Collapse
Affiliation(s)
- Anna-Katharina Zoellner
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Michael Unterhalt
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine I, Saarland University Medical Center, Homburg, Germany; Department of Internal Medicine III, Ulm University, Ulm, Germany
| | - Kai Hübel
- Department of Medicine I, University Hospital of Cologne, Cologne, Germany
| | | | - Bernd Metzner
- Department of Hematology and Oncology, University Hospital Oldenburg, Oldenburg, Germany
| | - Max Topp
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Lorenz Truemper
- Department of Hematology and Oncology, Georg August University, Goettingen, Germany
| | - Christian Schmidt
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Kamal Bouabdallah
- Department of Hematology and Cell Therapy, Haut-Leveque Hospital, Bordeaux, France
| | - Jürgen Krauter
- Department of Hematology and Oncology, Klinikum Braunschweig, Braunschweig, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology and Oncology, University of Münster, Münster, Germany
| | - Jan Dürig
- Department of Hematology, University Medicine Essen, Essen, Germany
| | - Vibeke Vergote
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Kerstin Schäfer-Eckart
- Klinik für Innere Medizin 5, Klinikum Nürnberg, Paracelsus Medizinische Privatuniversität, Nürnberg, Germany
| | - Marc André
- Department of Hematology, CHU UCLouvain Namur, Yvoir, Belgium
| | | | - Achiel van Hoof
- Department of Hematology, A Z St-Jan, Brugge-Oostende, Belgium
| | - Wolfram Klapper
- Hematopathology Section, Christian-Albrechts University, Kiel, Germany
| | - Wolfgang Hiddemann
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Martin Dreyling
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany.
| | - Eva Hoster
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Institute of Medical Information Processing, Biometry and Epidemiology, Ludwig Maximilian University of Munich, Munich, Germany
| | | |
Collapse
|
37
|
Munshi PN, Hamadani M, Kumar A, Dreger P, Friedberg JW, Dreyling M, Kahl B, Jerkeman M, Kharfan-Dabaja MA, Locke FL, Shadman M, Hill BT, Ahmed S, Herrera AF, Sauter CS, Bachanova V, Ghosh N, Lunning M, Kenkre VP, Aljurf M, Wang M, Maddocks KJ, Leonard JP, Kamdar M, Phillips T, Cashen AF, Inwards DJ, Sureda A, Cohen JB, Smith SM, Carlo-Stella C, Savani B, Robinson SP, Fenske TS. ASTCT, CIBMTR, and EBMT clinical practice recommendations for transplant and cellular therapies in mantle cell lymphoma. Bone Marrow Transplant 2021; 56:2911-2921. [PMID: 34413469 PMCID: PMC8639670 DOI: 10.1038/s41409-021-01288-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 11/09/2022]
Abstract
Autologous (auto-) or allogeneic (allo-) hematopoietic cell transplantation (HCT) are accepted treatment modalities for mantle cell lymphoma (MCL). Recently, chimeric antigen receptor (CAR) T-cell therapy received approval for MCL; however, its exact place and sequence in relation to HCT is unclear. The ASTCT, CIBMTR, and the EBMT, jointly convened an expert panel to formulate consensus recommendations for role, timing, and sequencing of auto-, allo-HCT, and CAR T-cell therapy for patients with newly diagnosed and relapsed/refractory (R/R) MCL. The RAND-modified Delphi method was used to generate consensus statements. Seventeen consensus statements were generated; in the first-line setting auto-HCT consolidation represents standard-of-care in eligible patients, whereas there is no clear role of allo-HCT or CAR T-cell therapy, outside of a clinical trial. In the R/R setting, the preferential option is CAR T-cell therapy especially in MCL failing or intolerant to at least one Bruton's tyrosine kinase inhibitor, while allo-HCT is recommended if CAR T-cell therapy has failed or is not feasible. In the absence of contemporary evidence-based data, the panel found RAND-modified Delphi methodology effective in providing a formal framework for developing consensus recommendations for the timing and sequence of cellular therapies for MCL.
Collapse
Affiliation(s)
| | - Mehdi Hamadani
- CIBMTR & BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Ambuj Kumar
- Department of Internal Medicine, Office of Research, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | | | - Martin Dreyling
- Department of Medicine III, LMU Hospital Munich, Munich, Germany
| | - Brad Kahl
- Washington University School of Medicine, St. Louis, MO, USA
| | - Mats Jerkeman
- Department of Oncology, Skane University Hospital, Lund University, Lund, Sweden
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapies Program, Mayo Clinic, Jacksonville, FL, USA
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Mazyar Shadman
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA, USA
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sairah Ahmed
- Department of Lymphoma, Myeloma and Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, USA
| | - Craig S Sauter
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Nilanjan Ghosh
- Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | | | - Vaishalee P Kenkre
- University of Wisconsin, Division of Hematology and Oncology, Madison, WI, USA
| | | | - Michael Wang
- Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kami J Maddocks
- Division of Hematology, Ohio State University, Columbus, OH, USA
| | | | - Manali Kamdar
- Division of Hematology, Hematologic Malignancies and Stem Cell Transplantation, University of Colorado Cancer Center, Denver, CO, USA
| | - Tycel Phillips
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Amanda F Cashen
- Division of Oncology, Section of Stem Cell Transplantation, Washington University School of Medicine, St Louis, MO, USA
| | - David J Inwards
- Division of Hematology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Anna Sureda
- Clinical Hematology Department, Catalan Institute of Oncology, Institut d'Investigació Biomèdica de Bellvitge IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | | | - Sonali M Smith
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Carmello Carlo-Stella
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milan, Italy
| | - Bipin Savani
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Timothy S Fenske
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
38
|
Jatiani SS, Christie S, Leshchenko VV, Jain R, Kapoor A, Bisignano P, Lee C, Kaniskan HÜ, Edwards D, Meng F, Laganà A, Youssef Y, Wiestner A, Alinari L, Jin J, Filizola M, Aggarwal AK, Parekh S. SOX11 Inhibitors Are Cytotoxic in Mantle Cell Lymphoma. Clin Cancer Res 2021; 27:4652-4663. [PMID: 34158358 PMCID: PMC8364871 DOI: 10.1158/1078-0432.ccr-20-5039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/19/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Mantle cell lymphoma (MCL) is a fatal subtype of non-Hodgkin lymphoma. SOX11 transcription factor is overexpressed in the majority of nodal MCL. We have previously reported that B cell-specific overexpression of SOX11 promotes MCL pathogenesis via critically increasing BCR signaling in vivo. SOX11 is an attractive target for MCL therapy; however, no small-molecule inhibitor of SOX11 has been identified to date. Although transcription factors are generally considered undruggable, the ability of SOX11 to bind to the minor groove of DNA led us to hypothesize that there may exist cavities at the protein-DNA interface that are amenable to targeting by small molecules. EXPERIMENTAL DESIGN Using a combination of in silico predictions and experimental validations, we report here the discovery of three structurally related compounds (SOX11i) that bind SOX11, perturb its interaction with DNA, and effect SOX11-specific anti-MCL cytotoxicity. RESULTS We find mechanistic validation of on-target activity of these SOX11i in the inhibition of BCR signaling and the transcriptional modulation of SOX11 target genes, specifically, in SOX11-expressing MCL cells. One of the three SOX11i exhibits relatively superior in vitro activity and displays cytotoxic synergy with ibrutinib in SOX11-expressing MCL cells. Importantly, this SOX11i induces cytotoxicity specifically in SOX11-positive ibrutinib-resistant MCL patient samples and inhibits Bruton tyrosine kinase phosphorylation in a xenograft mouse model derived from one of these subjects. CONCLUSIONS Taken together, our results provide a foundation for therapeutically targeting SOX11 in MCL by a novel class of small molecules.
Collapse
Affiliation(s)
- Shashidhar S Jatiani
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Stephanie Christie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Violetta V Leshchenko
- Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rinku Jain
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Abhijeet Kapoor
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Paola Bisignano
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Clement Lee
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - H Ümit Kaniskan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Donna Edwards
- Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fanye Meng
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alessandro Laganà
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Youssef Youssef
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Lapo Alinari
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Jian Jin
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aneel K Aggarwal
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Samir Parekh
- Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
39
|
Goy A. Exploiting gene mutations and biomarkers to guide treatment recommendations in mantle cell lymphoma. Expert Rev Hematol 2021; 14:927-943. [PMID: 34253131 DOI: 10.1080/17474086.2021.1950529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION While there has been an improvement in the treatment of mantle cell lymphoma (MCL) in both median progression-free survival (PFS; >7-8 years) and overall survival (OS; >10-12 years), patients with high-risk features such as high risk MIPI (mantle cell international prognostic index), high Ki-67 (≥30%), or blastoid variants still carry poor outcome with a median OS of 3 years. Furthermore, patients with high-risk molecular features, such as TP53 mutations, show dismal outcome, with a median OS of 1.8 years, regardless of therapy used. Further studies have led to the development of six novel drugs approved for the treatment of relapse/refractory (R/R) MCL, leading to improved survival even in refractory or high-risk patients. AREAS COVERED This review covers clinical biological and molecular features that impact MCL outcome with current standards. Beyond the recognition of separate subentities, we review how high-risk molecular features have paved the way towards a new paradigm away from chemoimmunotherapy. EXPERT OPINION Progress in novel therapies and in routine diagnostics, particularly next-generation sequencing (NGS), support the development of new treatment strategies, not based on the dose intensity/age dichotomy, which may prevent the need for chemotherapy and improve outcome across MCL including in high-risk subsets.
Collapse
Affiliation(s)
- Andre Goy
- John Theurer Cancer Center, Hackensack University Medical Center, NJ
| |
Collapse
|
40
|
Upfront intensive chemo-immunotherapy with autograft in 199 adult mantle cell lymphoma patients: prolonged survival and cure potentiality at long term. Bone Marrow Transplant 2021; 56:2606-2609. [PMID: 34234297 PMCID: PMC8486659 DOI: 10.1038/s41409-021-01391-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 06/12/2021] [Accepted: 06/22/2021] [Indexed: 11/09/2022]
|
41
|
Shanmugasundaram K, Goyal S, Switchenko J, Calzada O, Churnetski MC, Kolla B, Bachanova V, Gerson JN, Barta SK, Gordon MJ, Danilov AV, Grover NS, Mathews S, Burkart M, Karmali R, Sawalha Y, Hill BT, Ghosh N, Park SI, Epperla N, Bond DA, Badar T, Blum KA, Hamadani M, Fenske TS, Malecek M, Kahl BS, Martin P, Guo J, Flowers CR, Cohen JB. Intensive induction regimens after deferring initial therapy for mantle cell lymphoma are not associated with improved survival. Eur J Haematol 2021; 107:301-310. [PMID: 33973276 DOI: 10.1111/ejh.13649] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 01/05/2023]
Abstract
INTRODUCTION While most patients with mantle cell lymphoma (MCL) receive therapy shortly after diagnosis, a subset of patients with indolent-behaving disease can safely defer treatment. In this subgroup, we evaluated the importance of treatment intensity in patients with MCL who defer initial therapy. METHODS Out of 1134 patients with MCL from 12 academic centers, we analyzed 219 patients who initiated therapy at least 90 days after diagnosis. Patients who received induction with high-dose cytarabine and/or autologous stem cell transplantation (ASCT) in first remission were considered to have received intensive therapy (n = 88) while all other approaches were non-intensive (n = 131). RESULTS There was no difference in progression-free (PFS; P = .224) or overall survival (OS; P = .167) in deferred patients who received non-intensive vs. intensive therapy. Additionally, univariate and multivariate Cox proportional hazards models were performed for PFS and OS. Treatment at an academic center (HR 0.43, P = .015) was associated with improved OS in both univariate and multivariate models, while intensity of treatment was not associated with improved OS in either model. CONCLUSIONS These results indicate that intensified initial treatment is not associated with improved survival after deferring initial therapy, although prospective studies are needed to determine which of these patients with MCL may benefit from intensive therapy.
Collapse
Affiliation(s)
| | - Subir Goyal
- Winship Cancer Institute, Biostatistics and Bioinformatics, Emory University, Atlanta, GA, US
| | - Jeffery Switchenko
- Winship Cancer Institute, Biostatistics and Bioinformatics, Emory University, Atlanta, GA, US
| | - Oscar Calzada
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University Hospital, Atlanta, GA, US
| | - Michael C Churnetski
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University Hospital, Atlanta, GA, US
| | - Bhaskar Kolla
- Division of Hematology Oncology and Transplantation, University of Minnesota, Minneapolis, MN, US
| | - Veronika Bachanova
- Division of Hematology Oncology and Transplantation, University of Minnesota, Minneapolis, MN, US
| | - James N Gerson
- Hematology and Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, US
| | - Stefan K Barta
- Hematology and Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, US
| | - Max J Gordon
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, US
| | - Alexey V Danilov
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, US
| | - Natalie S Grover
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, US
| | - Stephanie Mathews
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, US
| | - Madelyn Burkart
- Division of Hematology, Northwestern University, Chicago, IL, US
| | - Reem Karmali
- Division of Hematology, Northwestern University, Chicago, IL, US
| | - Yazeed Sawalha
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, US
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, US
| | | | | | - Narendranath Epperla
- Division of Hematology, The Ohio State University Cancer Center, Columbus, OH, US.,Mayo Clinic, Jacksonville, FL, US
| | - David A Bond
- Division of Hematology, The Ohio State University Cancer Center, Columbus, OH, US
| | | | - Kristie A Blum
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University Hospital, Atlanta, GA, US
| | - Mehdi Hamadani
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, US
| | - Timothy S Fenske
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, US
| | - Mary Malecek
- Siteman Cancer Center, Washington University, St. Louis, MO, US
| | - Brad S Kahl
- Siteman Cancer Center, Washington University, St. Louis, MO, US
| | | | - Jin Guo
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Christopher R Flowers
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Jonathon B Cohen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University Hospital, Atlanta, GA, US
| |
Collapse
|
42
|
Alderuccio JP, Saul EE, Iyer SG, Reis IM, Alencar AJ, Rosenblatt JD, Lossos IS. R-MACLO-IVAM regimen followed by maintenance therapy induces durable remissions in untreated mantle cell lymphoma - Long term follow up results. Am J Hematol 2021; 96:680-689. [PMID: 33735476 DOI: 10.1002/ajh.26163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/27/2021] [Accepted: 03/15/2021] [Indexed: 11/06/2022]
Abstract
We present long-term combined results of two clinical trials implementing R-MACLO-IVAM induction followed by thalidomide or rituximab maintenance in 44 patients with untreated mantle cell lymphoma (MCL). The first 22 patients (UM-MCL1 ClinicalTrials.gov identifier NCT00450801) received maintenance with thalidomide (200 mg daily until relapse/intolerable toxicity) and a subsequent cohort of 22 patients (UM-MCL2 ClinicalTrials.gov identifier NCT00878254) received rituximab (375 mg/m2 IV weekly × 4, repeated every 6 months for 3 years). Considering all 44 patients, 41 (93.2%) achieved complete response (CR), two (4.5%) partial response (PR), and one (2.3%) was not evaluated for response. With a median follow up of 7.2 years (range < 1 month to 16 years), the 5-year progression-free survival (PFS) was 55.6% (95% CI: 38.9%-69.4%) and median PFS 7.9 years (95% CI: 3.7-11 years). The 5-year OS was 83.3% (95% CI: 68.1%-91.7%) and median OS was not reached. Patients with blastic variant (n = 6) had a 5-year PFS and OS of 20.8% and 60%, respectively. Myelosuppression was the most common adverse event during immunochemotherapy. Long-term treatment-related mortality was 6.8%. Note, R-MACLO-IVAM followed by maintenance therapy is an effective regimen to induce long-term remission in MCL without need for consolidation with ASCT.
Collapse
Affiliation(s)
- Juan P. Alderuccio
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Eduardo E. Saul
- Department of Medicine, Division of Internal Medicine, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Sunil G. Iyer
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Isildinha M. Reis
- Department of Public Health Science, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
- Sylvester Biostatistics and Bioinformatics Shared Resource, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Alvaro J. Alencar
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Joseph D. Rosenblatt
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Izidore S. Lossos
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
- Department of Molecular and Cellular Pharmacology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
43
|
Yang X, Khoo LP, Chang EWY, Yang VS, Poon E, Somasundaram N, Farid M, Tang TPL, Tao M, Lim ST, Chan JY. Treatment patterns and outcomes of older patients with mantle cell lymphoma in an Asian population. BMC Cancer 2021; 21:566. [PMID: 34001056 PMCID: PMC8130422 DOI: 10.1186/s12885-021-08326-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 05/09/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Significant progress has been made in the treatment outcomes of mantle cell lymphoma (MCL) since the introduction of cytarabine and rituximab in modern regimens. However, older patients may not readily tolerate these agents nor derive benefit. We investigated the impact of age on treatment patterns and clinical outcomes of MCL patients in an Asian population. METHODS A retrospective study was conducted on patients (n = 66) diagnosed with MCL at the National Cancer Centre Singapore between 1998 and 2018. The median follow-up duration was 40 months. Survival analyses were performed using the Kaplan-Meier method and multivariate Cox proportional models. RESULTS The median age of the cohort was 59 years (range, 26-84), with a male predominance (73%). The majority (86%) had advanced stage 3-4 disease at diagnosis. Compared with younger patients, older patients aged ≥60 years (n = 32; 48.5%) presented more frequently with B-symptoms (75% vs 38%, p = 0.0028), anaemia (75% vs 35%, p = 0.0013), and carried higher prognostic risk scores (sMIPI high risk 84% vs 56%, p = 0.016). Non-cytarabine-based induction chemotherapy was more commonly administered in older patients (76% vs 32%, p = 0.0012). The 5-year overall survival (OS) and progression-free survival (PFS) was 68 and 25% respectively. In a multivariable model, older age (HR 3.42, 95%CI 1.48-7.92, p = 0.004) and anemia (HR 2.56, 95%CI 1.10-5.96, p = 0.029) were independently associated with poorer OS while older age (HR 2.24, 95%CI 1.21-4.14, p = 0.010) and hypoalbuminemia (HR 2.20, 95%CI 1.17-4.13, p = 0.014) were independently associated with poorer PFS. In an exploratory analysis, maintenance rituximab following induction chemotherapy improved PFS in younger patients, with median PFS of 131 months and 45 months with or without maintenance therapy respectively (HR 0.39, 95%CI 0.16-0.93, p = 0.035). In contrast, no survival benefit was observed in older patients. CONCLUSIONS We demonstrated in our analysis that older patients with MCL may harbor adverse clinical features and may not derive benefit from maintenance rituximab, highlighting the need for further research in this area of need.
Collapse
Affiliation(s)
- Xinyi Yang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Lay Poh Khoo
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore
| | - Esther Wei Yin Chang
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore
- SingHealth Duke-NUS Blood Cancer Centre, Singapore City, Singapore
| | - Valerie Shiwen Yang
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore
- SingHealth Duke-NUS Blood Cancer Centre, Singapore City, Singapore
- Institute of Molecular and Cell Biology, Singapore City, Singapore
| | - Eileen Poon
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore
- SingHealth Duke-NUS Blood Cancer Centre, Singapore City, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore City, Singapore
| | - Nagavalli Somasundaram
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore
- SingHealth Duke-NUS Blood Cancer Centre, Singapore City, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore City, Singapore
| | - Mohamad Farid
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore
- SingHealth Duke-NUS Blood Cancer Centre, Singapore City, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore City, Singapore
| | - Tiffany Pooi Ling Tang
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore
- SingHealth Duke-NUS Blood Cancer Centre, Singapore City, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore City, Singapore
| | - Miriam Tao
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore
- SingHealth Duke-NUS Blood Cancer Centre, Singapore City, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore City, Singapore
| | - Soon Thye Lim
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore.
- SingHealth Duke-NUS Blood Cancer Centre, Singapore City, Singapore.
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore City, Singapore.
| | - Jason Yongsheng Chan
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore City, 169610, Singapore.
- SingHealth Duke-NUS Blood Cancer Centre, Singapore City, Singapore.
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore City, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore City, Singapore.
| |
Collapse
|
44
|
Bendamustine and rituximab as induction therapy in both transplant-eligible and -ineligible patients with mantle cell lymphoma. Blood Adv 2021; 4:3486-3494. [PMID: 32735654 DOI: 10.1182/bloodadvances.2020002068] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/23/2020] [Indexed: 12/29/2022] Open
Abstract
Rituximab-containing chemotherapy regimens constitute standard first-line therapy for mantle cell lymphoma (MCL). Since June 2013, 190 patients ≥18 years of age with MCL in British Columbia have been treated with bendamustine and rituximab (BR). The overall response rate to BR was 88% (54% complete response). Of these, 61 of 89 patients (69%) aged ≤65 years received autologous stem cell transplantation and 141 of 190 patients (74%) from the entire cohort received maintenance rituximab. Twenty-three patients (12%) had progressive disease, associated with high risk per the Mantle Cell Lymphoma International Prognostic Index (MIPI), Ki-67 ≥50%, and blastoid/pleomorphic histology. Outcomes were compared with a historical cohort of 248 patients treated with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP; January 2003 to May 2013). Treatment with BR was associated with significant improvements in progression-free survival (PFS), but not overall survival (OS), compared with R-CHOP in the whole cohort (3-year PFS, 66% BR vs 51% R-CHOP, P = .003; 3-year OS, 73% BR vs 66% R-CHOP, P = .054) and in those >65 years of age (3-year PFS, 56% BR vs 35% R-CHOP, P = .001; 3-year OS, 64% BR vs 55% R-CHOP, P = .063). Outcomes in transplanted patients were not statistically significantly different compared with R-CHOP (3-year PFS, 85% BR vs 76% R-CHOP, P = .135; 3-year OS, 90% BR vs 88% R-CHOP, P = .305), although in multivariate analyses, treatment with BR was associated with improved PFS (hazard ratio, 0.40 [95% confidence interval, 0.17-0.94]; P = .036) but not OS. BR is an effective first-line option for most patients with MCL, however, outcomes are suboptimal for those with high-risk features and further studies integrating novel agents are warranted.
Collapse
|
45
|
Novel Treatments for Mantle Cell Lymphoma: From Targeted Therapies to CAR T Cells. Drugs 2021; 81:669-684. [PMID: 33783717 DOI: 10.1007/s40265-021-01497-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2021] [Indexed: 10/21/2022]
Abstract
Mantle cell lymphoma is a rare B-cell non-Hodgkin's lymphoma that retains a sobering prognosis despite an extensive research effort. Mantle cell lymphoma remains incurable even with aggressive, and at times toxic, chemoimmunotherapy with early incorporation of autologous stem cell transplantation. Given this, attention has turned to the use of targeted therapies addressing dysregulation of B-cell signaling pathways. Drugs such as immunomodulatory agents, proteasome inhibitors, and Bruton's tyrosine kinase inhibitors have shown success in the relapsed/refractory population, and there is ongoing investigation into the utilization of novel Bruton's tyrosine kinase, B-cell leukemia/lymphoma-2, and spleen tyrosine kinase inhibitors alone or in combination in both the front-line and relapsed settings. Other areas of research in novel immunotherapies include investigations of bispecific T-cell engagers and antibody-drug conjugates. Most recently, chimeric antigen receptor T-cell therapy has been granted US Food and Drug Administration approval as a result of durable remissions even in high-risk patients who have classically done poorly with traditional chemoimmunotherapy. The intent of this article is to review the literature describing these selective therapies and discuss their current and future roles in the treatment of mantle cell lymphoma.
Collapse
|
46
|
Glimelius I, Smedby KE, Albertsson-Lindblad A, Crowther MJ, Eloranta S, Jerkeman M, Weibull CE. Unmarried or less-educated patients with mantle cell lymphoma are less likely to undergo a transplant, leading to lower survival. Blood Adv 2021; 5:1638-1647. [PMID: 33710334 PMCID: PMC7993102 DOI: 10.1182/bloodadvances.2020003645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022] Open
Abstract
It is unknown how many mantle cell lymphoma (MCL) patients undergo consolidation with autologous hematopoietic cell transplantation (AHCT), and the reasons governing the decision, are also unknown. The prognostic impact of omitting AHCT is also understudied. We identified all MCL patients diagnosed from 2000 to 2014, aged 18 to 65 years, in the Swedish Lymphoma Register. Odds ratios (ORs) and 95% confidence intervals (CIs) from logistic regression models were used to compare the likelihood of AHCT within 18 months of diagnosis. All-cause mortality was compared between patients treated with/without AHCT using hazard ratios (HRs) and 95% CIs estimated from Cox regression models. Probabilities of being in each of the following states: alive without AHCT, alive with AHCT, dead before AHCT, and dead after AHCT, were estimated over time from an illness-death model. Among 369 patients, 148 (40%) were not treated with AHCT within 18 months. Compared with married patients, never married and divorced patients had lower likelihood of undergoing AHCT, as had patients with lower educational level, and comorbid patients. Receiving AHCT was associated with reduced all-cause mortality (HR = 0.58, 95% CI: 0.40-0.85). Transplantation-related mortality was low (2%). MCL patients not receiving an AHCT had an increased mortality rate, and furthermore, an undue concern about performing an AHCT in certain societal groups was seen. Improvements in supportive functions potentially increasing the likelihood of tolerating an AHCT and introduction of more tolerable treatments for these groups are needed.
Collapse
Affiliation(s)
- Ingrid Glimelius
- Department of Immunology, Genetics, and Pathology, Clinical and Experimental Oncology, Uppsala University, Uppsala Akademiska Hospital, Uppsala, Sweden
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Karin E Smedby
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Division of Hematology, Department of Medicine Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Michael J Crowther
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom; and
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Eloranta
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Mats Jerkeman
- Department of Oncology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Caroline E Weibull
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
47
|
Mian A, Hill BT. Brexucabtagene autoleucel for the treatment of relapsed/refractory mantle cell lymphoma. Expert Opin Biol Ther 2021; 21:435-441. [PMID: 33566715 DOI: 10.1080/14712598.2021.1889510] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: The therapeutic options for mantle cell lymphoma (MCL) include traditional chemo-immunotherapy for newly diagnosed cases, and targeted treatments including the bruton tyrosine kinase inhibitors in the relapsed/refractory (R/R) disease setting. The advent of commercially available chimeric antigen receptor (CAR) T-cell therapy in the last three years has dramatically improved the outcomes of patients with R/R large B-cell lymphoma.Areas covered: This review is an in-depth evaluation and appraisal of brexucabtagene autoleucel (brexu-cel), the first anti-CD19 CAR T-cell therapy to be approved for patients with R/R MCL, after the results of a Phase II (ZUMA-2) trial.Expert opinion: In the absence of head-to-head comparison studies with Btk inhibitors, up-front use of brexu-cel in patients with high-risk MCL and poor prognostic features may be advantageous, possibly even before exposure to Btk inhibitor, and further study of this approach is warranted. While data on long-term outcomes of CAR T-cell therapy in MCL patients are needed, brexu-cel has shown remarkable clinical activity and its regulatory approval has immediate practice-changing implications in this highly aggressive malignancy.
Collapse
Affiliation(s)
- Agrima Mian
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Brian T Hill
- Lymphoid Malignancies Program, Staff Physician, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland, OH, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
48
|
Rodrigues JM, Nikkarinen A, Hollander P, Weibull CE, Räty R, Kolstad A, Amini RM, Porwit A, Jerkeman M, Ek S, Glimelius I. Infiltration of CD163-, PD-L1- and FoxP3-positive cells adversely affects outcome in patients with mantle cell lymphoma independent of established risk factors. Br J Haematol 2021; 193:520-531. [PMID: 33686666 DOI: 10.1111/bjh.17366] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/23/2020] [Accepted: 01/06/2021] [Indexed: 01/08/2023]
Abstract
We characterised patients with mantle cell lymphoma (MCL) with poor prognosis based on differences in immune infiltration. Different expressions of the tumour cell markers Cyclin D1 and sex-determining region Y-box transcription factor 11 (SOX11), and the immune markers cluster of differentiation 3 (CD3), CD4, CD8, CD25, forkhead box protein P3 (FoxP3), T-box transcription factor TBX21 (T-bet), programmed cell death protein 1 (PD-1), programmed-death ligand 1 (PD-L1) and CD163 were investigated for all-cause mortality in 282 patients with MCL and time-to-progression (TTP) in 106 clinical trial patients. With increasing age, a significantly lower infiltration of CD3+ T lymphocytes was seen. T-cell infiltration was independent of cellular tumour antigen p53 (p53) expression, Ki-67, morphology and frequency of tumour cells. The all-cause mortality was higher in patients with PD-L1-expression above cut-off [hazard ratio (HR) 1·97, 95% confidence interval (CI) 1·18-3·25, adjusted for sex and MCL International Prognostic Index (MIPI)] and a higher frequency of CD163+ cells (continuously, HR 1·51, 95% CI 1·03-2·23, adjusting for age, sex, morphology, Ki-67 and p53). In patients treated within the Nordic Lymphoma Group MCL2/3 trials, TTP was shorter in patients with a higher frequency of FoxP3+ cells (HR 3·22, 95% CI 1·40-7·43) and CD163+ cells (HR 6·09, 95% CI 1·84-20·21), independent of sex and MIPI. When combined a higher frequency of CD163+ macrophages and PD-L1+ cells or high CD163+ macrophages and FoxP3+ regulatory T cells indicated worse outcome independent of established risk factors. The T-cell infiltrate was in turn independent of molecular characteristics of the malignant cells and decreased with age.
Collapse
Affiliation(s)
| | - Anna Nikkarinen
- Department of Immunology, Genetics and Pathology, Clinical and Experimental Oncology, Uppsala University, Uppsala, Sweden
| | - Peter Hollander
- Department of Immunology, Genetics and Pathology, Clinical and Experimental Oncology, Uppsala University, Uppsala, Sweden
| | - Caroline E Weibull
- Department of Medicine, Division of Clinical Epidemiology, Karolinska Institutet, Stockholm, Sweden
| | - Riikka Räty
- Department of Hematology, Helsinki University Hospital, Helsinki, Finland
| | - Arne Kolstad
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Rose-Marie Amini
- Department of Immunology, Genetics and Pathology, Clinical and Experimental Oncology, Uppsala University, Uppsala, Sweden
| | - Anna Porwit
- Department of Clinical Sciences, Oncology and Pathology, Lund University, Lund, Sweden
| | - Mats Jerkeman
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Sara Ek
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Ingrid Glimelius
- Department of Immunology, Genetics and Pathology, Clinical and Experimental Oncology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
49
|
Invited Review: Will Consolidation with ASCT Be a Thing of the Past for MCL and PTCL? Curr Hematol Malig Rep 2021; 16:82-88. [PMID: 33646524 DOI: 10.1007/s11899-021-00609-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE OF REVIEW The treatment landscape of mantle cell (MCL) and peripheral T-cell lymphomas (PTCL) is rapidly changing; however, despite improvement in patients' survival, they still remain a largely incurable diseases. Treatment choice is dependent on patient factors, prior therapy, remission duration, and candidacy for stem cell transplantation (SCT). There are subsets of high-risk patients who do not benefit substantially from autologous SCT (ASCT) and for whom alternative targeted approaches are being examined. Here, we critically analyze the actual role of ASCT in PTCL and MCL. RECENT FINDINGS Research in areas of maintenance therapy and minimal residual disease is ongoing to identify MCL patients who may not require ASCT for durable response. Moreover, there are subsets of high-risk MCL patients who do not benefit substantially from ASCT and for whom alternative, targeted approaches are being examined. Much less clear evidence exists regarding the impact of consolidative ASCT in PTCL, mainly for the heterogeneity of these lymphomas: it is still controversial whether patients who achieved a complete response significantly take advantage of this procedure over active surveillance only. Several clinical and biologic markers are available to predict prognosis; however, despite improvements in outcomes, standard therapeutic approaches have not been able to overcome high-risk disease features for PTCL and MCL. Thus, the need of ASCT for these diseases is still matter of debate among hematologists.
Collapse
|
50
|
Cellular Therapies for Mantle Cell Lymphoma. Transplant Cell Ther 2021; 27:363-370. [PMID: 33965173 DOI: 10.1016/j.jtct.2021.01.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/15/2021] [Accepted: 01/31/2021] [Indexed: 11/24/2022]
Abstract
Mantle cell lymphoma (MCL) is a subtype of B cell non-Hodgkin lymphoma characterized by a heterogeneous clinical presentation. Patients who demonstrate an objective response to induction therapy(ies) and are eligible for intensive therapies are offered an autologous hematopoietic cell transplant (HCT) as front-line consolidation followed by rituximab maintenance. Allogeneic HCT is an option for younger and fit patients with high-risk disease or in patients who have relapsed after autologous HCT. Recent advances in T cell engineering brought chimeric antigen receptor T cell (CAR T) therapy from the bench to the bedside, with brexucabtagene autoleucel being the first CAR T product approved by the US Food and Drug Administration for use in relapsed/refractory MCL. In this comprehensive review, we summarize the literature on available cellular therapies for MCL and present a treatment algorithm that incorporates HCT, autologous or allogeneic, and CAR T therapies.
Collapse
|