1
|
Jain P, Wang M. High-risk MCL: recognition and treatment. Blood 2025; 145:683-695. [PMID: 39786418 DOI: 10.1182/blood.2023022354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Significant progress in determining the molecular origins and resistance mechanisms of mantle cell lymphoma (MCL) has improved our understanding of the disease's clinical diversity. These factors greatly impact the prognosis of patients with MCL. Given the dynamic alterations in MCL clones and disease evolution, it is crucial to recognize high-risk prognostic factors at diagnosis and relapse. Clinical factors include a high MCL International Prognostic Index score with a high Ki-67 proliferation index, early disease progression within 24 months of first-line treatment, >3 previous lines of therapy at relapse, and an aggressive (blastoid or pleomorphic) histology. Molecular aberrations include dysregulated cyclin D1, an aberrant SOX11-CD70 axis, upregulated Musashi-2, MYC rearrangement, metabolic reprogramming, and epigenetic changes. Other factors that contribute to high-risk MCL include an immune-depleted microenvironment and clone adaptability with complex chromosomal anomalies and somatic mutations in TP53, NSD2, CCND1, CDKN2A, BIRC3, SP140, KMT2D, NFkBIE, SMARCA4, and NOTCH2. Ultra-high-risk MCL is indicated by the coexistence of multiple high-risk prognostic factors in the relapse setting and can portend very short progression-free survival. As MCL treatments advance toward cellular therapies, resistance to anti-CD19 chimeric antigen receptor T-cell therapy is also observed. These findings necessitate revisiting the prognostic impact of high-risk factors, current management strategies, new bi- and trispecific T-cell engagers, combination therapies, novel therapeutic targets, and next-generation clinical trials for patients with high-risk MCL. This article provides a comprehensive update on recognizing and managing high-risk MCL and encompass current practices and future directions.
Collapse
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
2
|
Sawalha Y, Maddocks K. Your chemo is no good here: management of high-risk MCL. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:34-41. [PMID: 39644043 DOI: 10.1182/hematology.2024000658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Historically considered a lymphoma with limited treatment options and poor outcomes, the treatment landscape in mantle cell lymphoma (MCL) has evolved remarkably in the last decade. Chemoimmunotherapy (CIT) remains the primary frontline treatment for most patients with MCL, typically with an intensive approach in younger and fit patients. The role of consolidative autologous stem cell transplantation remains controversial, with recent data further questioning its benefit. Novel agents have shown promising results in recent frontline clinical trials and challenge the current paradigm in MCL, particularly in high-risk patients who generally have poor outcomes with CIT. Risk stratification is key to incorporating novel agents in the frontline treatment of MCL, identifying patients who do not benefit from or could be spared CIT, guiding treatment intensity and duration, and improving overall outcomes, including safety and quality of life. The MCL International Prognostic Index and Ki-67 play an important role in identifying patients with high-risk MCL. TP53 aberrations, particularly mutations, currently identify patients with the highest risk, limited benefit from CIT, and greatest need for novel therapies. Other genetic aberrations and biological clusters are being identified but currently have limited clinical utility.
Collapse
Affiliation(s)
- Yazeed Sawalha
- Department of Internal Medicine, Division of Hematology, Arthur G. James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Kami Maddocks
- Department of Internal Medicine, Division of Hematology, Arthur G. James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
3
|
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
|
4
|
López C, Silkenstedt E, Dreyling M, Beà S. Biological and clinical determinants shaping heterogeneity in mantle cell lymphoma. Blood Adv 2024; 8:3652-3664. [PMID: 38748869 PMCID: PMC11284685 DOI: 10.1182/bloodadvances.2023011763] [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: 02/23/2024] [Accepted: 05/03/2024] [Indexed: 07/12/2024] Open
Abstract
ABSTRACT Mantle cell lymphoma (MCL) is an uncommon mature B-cell lymphoma that presents a clinical spectrum ranging from indolent to aggressive disease, with challenges in disease management and prognostication. MCL is characterized by significant genomic instability, affecting various cellular processes, including cell cycle regulation, cell survival, DNA damage response and telomere maintenance, NOTCH and NF-κB/ B-cell receptor pathways, and chromatin modification. Recent molecular and next-generation sequencing studies unveiled a broad genetic diversity among the 2 molecular subsets, conventional MCL (cMCL) and leukemic nonnodal MCL (nnMCL), which may partially explain their clinical heterogeneity. Some asymptomatic and genetically stable nnMCL not requiring treatment at diagnosis may eventually progress clinically. Overall, the high proliferation of tumor cells, blastoid morphology, TP53 and/or CDKN2A/B inactivation, and high genetic complexity influence treatment outcome in cases treated with standard regimens. Emerging targeted and immunotherapeutic strategies are promising for refractory or relapsed cases and a few genetic and nongenetic determinants of refractoriness have been reported. This review summarizes the recent advances in MCL biology, focusing on molecular insights, prognostic markers, and novel therapeutic approaches.
Collapse
Affiliation(s)
- Cristina López
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Departament de Fonaments Clínics, Universitat de Barcelona, Barcelona, Spain
| | - Elisabeth Silkenstedt
- Department of Medicine III, Ludwig-Maximilians-University Munich University Hospital, Munich, Germany
| | - Martin Dreyling
- Department of Medicine III, Ludwig-Maximilians-University Munich University Hospital, Munich, Germany
| | - Sílvia Beà
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Departament de Fonaments Clínics, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Yang P, Liu SZ, Li CY, Zhang WL, Wang J, Chen YT, Li S, Liu CL, Liu H, Cai QQ, Zhang W, Jing HM. Genetic and prognostic analysis of blastoid and pleomorphic mantle cell lymphoma: a multicenter analysis in China. Ann Hematol 2024; 103:2381-2391. [PMID: 38165416 DOI: 10.1007/s00277-023-05597-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Blastoid or pleomorphic mantle cell lymphoma (B/P-MCL) is characterized by high invasiveness and unfavorable outcomes, which is still a challenge for treating MCL. This retrospective study was performed to comprehensively analyze the clinical, genomic characteristics and treatment options of patients with B/PMCL from multicenter in China. Data were obtained from 693 patients with B/PMCL from three centers in China between April 1999 and December 2019. Seventy-four patients with BMCL (n = 43) or PMCL (n = 31) were included in the analysis. The median age of the cohort was 60.0 years with a male-to-female ratio of 2.89:1. The 3-year progression-free survival (PFS) and overall survival (OS) rates were 44.1% and 46.0%, respectively. Mutations of TP53, ATM, NOTCH1, NOTCH2, NSD2, SMARCA4, CREBBP, KMT2D, FAT1, and TRAF2 genes were the most common genetic changes in B/P-MCL. Progression of disease within 12 months (POD12) could independently predict the poor prognosis of patients with blastoid and pleomorphic variants. Patients with POD12 carried a distinct mutation profile (TP53, SMARCA4, NSD2, NOTCH2, KMT2D, PTPRD, CREBBP, and CDKN2A mutations) compared to patients with non-POD12. First-line high-dose cytosine arabinoside exposure obtained survival benefits in these populations, and BTKi combination therapy as the front-line treatment had somewhat improvement in survival with no significant difference in the statistic. In conclusion, B/P-MCL had inferior outcomes and a distinct genomic profile. Patients with POD12 displayed a distinct mutation profile and a poor prognosis. New therapeutic drugs and clinical trials for B/P-MCL need to be further explored.
Collapse
Affiliation(s)
- Ping Yang
- Department of Hematology, Peking University Third Hospital, No. 49 Huayuan N Rd Haidian District, Beijing, China
| | - Shuo-Zi Liu
- Department of Hematology, Peking University Third Hospital, No. 49 Huayuan N Rd Haidian District, Beijing, China
| | - Chun-Yuan Li
- Department of Hematology, Peking University Third Hospital, No. 49 Huayuan N Rd Haidian District, Beijing, China
| | - Wei-Long Zhang
- Department of Hematology, Peking University Third Hospital, No. 49 Huayuan N Rd Haidian District, Beijing, China
| | - Jing Wang
- Department of Hematology, Peking University Third Hospital, No. 49 Huayuan N Rd Haidian District, Beijing, China
| | - Ying-Tong Chen
- Department of Hematology, Peking University Third Hospital, No. 49 Huayuan N Rd Haidian District, Beijing, China
| | - Sen Li
- Department of Hematology, Peking University Third Hospital, No. 49 Huayuan N Rd Haidian District, Beijing, China
| | - Cui-Ling Liu
- Department of Hematology, Peking University Third Hospital, No. 49 Huayuan N Rd Haidian District, Beijing, China
| | - Hui Liu
- Department of Hematology, Beijing Hospital, Beijing, China
| | - Qing-Qing Cai
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wei Zhang
- Department of Hematology, Peking Union Medical College Hospital, Beijing, China
| | - Hong-Mei Jing
- Department of Hematology, Peking University Third Hospital, No. 49 Huayuan N Rd Haidian District, Beijing, China.
| |
Collapse
|
6
|
Ware AD, Davis K, Xian RR. Molecular Pathology of Mature Lymphoid Malignancies. Clin Lab Med 2024; 44:355-376. [PMID: 38821649 DOI: 10.1016/j.cll.2023.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Lymphoid malignancies are a broad and heterogeneous group of neoplasms. In the past decade, the genetic landscape of these tumors has been explored and cataloged in fine detail offering a glimpse into the mechanisms of lymphomagenesis and new opportunities to translate these findings into patient management. A myriad of studies have demonstrated both distinctive and overlapping molecular and chromosomal abnormalities that have influenced the diagnosis and classification of lymphoma, disease prognosis, and treatment selection.
Collapse
Affiliation(s)
- Alisha D Ware
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27514, USA
| | - Katelynn Davis
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rena R Xian
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27514, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Johns Hopkins School of Medicine, 1812 Ashland Avenue, Suite 200, Baltimore, MD 21205, USA.
| |
Collapse
|
7
|
Carras S, Torroja A, Emadali A, Montaut E, Daguindau N, Tempescul A, Moreau A, Tchernonog E, Schmitt A, Houot R, Dartigeas C, Barbieux S, Corm S, Banos A, Fouillet L, Dupuis J, Macro M, Fleury J, Jardin F, Sarkozy C, Damaj G, Feugier P, Fornecker LM, Chabrot C, Dorvaux V, Bouabdallah K, Amorim S, Garidi R, Voillat L, Joly B, Morineau N, Moles MP, Zerazhi H, Fontan J, Arkam Y, Alexis M, Delwail V, Vilque JP, Ysebaert L, Burroni B, Callanan M, Le Gouill S, Gressin R. Long-term analysis of the RiBVD phase II trial reveals the unfavorable impact of TP53 mutations and hypoalbuminemia in older adults with mantle cell lymphoma; for the LYSA group. Haematologica 2024; 109:1857-1865. [PMID: 38031755 PMCID: PMC11141646 DOI: 10.3324/haematol.2023.283724] [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: 06/29/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023] Open
Abstract
Between 2011 and 2012, a phase II trial evaluated the use of the RiBVD (rituximab, bendamustine, velcade and dexamethasone) combination as first-line treatment for mantle cell lymphoma (MCL) patients over the age of 65. We have now re-examined the classic prognostic factors, adding an assessment of TP53 mutation status. Patients (N=74; median age 73 years) were treated with the RiBVD combination. Median progression-free survival (mPFS) was 79 months and median overall survival (mOS) was 111 months. TP53 mutation status was available for 54/74 (73%) patients. TP53 mutations (TP53mt) were found in 12 patients (22.2%). In multivariate analysis, among the prognostic factors (PF) evaluated, only TP53mt and an albumin level (Alb) 3.6 g/dL were independently associated with a shorter mPFS. A hazard ratio (HR) of 3.16 (1.3-9.9, P=0.014) was obtained for TP53mt versus TP53 wild-type (wt), and 3.6 (1.39-9.5, P=0.009) for Alb <3.6 g/dL versus Alb ≥3.6 g/dL. In terms of mOS, multivariate analysis identified three PF: TP53mt (HR: 5.9 [1.77-19.5, P=0.004]), Alb <3.6 g/dL (HR: 5.2 [1.46- 18.5, P=0.011]), and ECOG=2 (HR: 3.7 [1.31-10.6, P=0.014]). Finally, a score combining TP53 status and Alb distinguished three populations based on the presence of 0, 1, or 2 PF. For these populations, mPFS was 7.8 years, 28 months, and 2.5 months, respectively. Our prolonged follow-up confirmed the efficacy of the RiBVD regimen, comparing it favorably to other regimens. TP53mt and hypoalbuminemia emerge as strong PF that can be easily integrated into prognostic scores for older adult patients with MCL.
Collapse
Affiliation(s)
- Sylvain Carras
- Université Grenoble Alpes, University Hospital, Institute For Advanced Biosciences (INSERM U1209, CNRSéUMR 5309, UGA), Molecular Biology Department, Grenoble
- Université Grenoble Alpes, University Hospital, Institute For Advanced Biosciences (INSERM U1209, CNRS UMR 5309, UGA), Oncohematology Department, Grenoble
| | - Alexia Torroja
- Université Grenoble Alpes, University Hospital, Institute For Advanced Biosciences (INSERM U1209, CNRS UMR 5309, UGA), Oncohematology Department, Grenoble
| | - Anouk Emadali
- Université Grenoble Alpes, University Hospital, Institute for Advanced Biosciences (INSERM U1209, CNRS UMR 5309, UGA), Research & Innovation Unit, Grenoble
| | - Emilie Montaut
- Université Grenoble Alpes, University Hospital, Institute for Advanced Biosciences (INSERM U1209, CNRS UMR 5309, UGA), Research & Innovation Unit, Grenoble
| | | | | | - Anne Moreau
- Pathology Department, University Hospital, Nantes
| | | | - Anna Schmitt
- Hematology Department, Cancerology Institute Bergonie, Bordeaux
| | - Roch Houot
- Hematology Department, University Hospital, Rennes
| | | | | | - Selim Corm
- Hematology Department, Chambery Hospital, Chambery
| | - Anne Banos
- Hematology Department, Bayonne Cote Basque Hospital, Bayonne
| | | | - Jehan Dupuis
- Lymphoid Malignancies Unit, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil
| | | | - Joel Fleury
- Hematology Department, Cancerology Institute, Clermont-Ferrand
| | | | | | - Ghandi Damaj
- Hematology Department, University Hospital, Caen
| | | | | | - Cecile Chabrot
- Hematology Department, University Hospital, Clermont-Ferrand
| | | | | | - Sandy Amorim
- Hematology & Cellular Therapy Department, Hospital Saint Vincent de Paul, Université Catholique de Lille, Lille
| | - Reda Garidi
- Hematology Department, Hospital Saint Quentin, Saint Quentin
| | - Laurent Voillat
- Hematology Department, Hospital Chalon sur Saone, Chalon sur Saone
| | - Bertrand Joly
- Hematology Department, Corbeil Hospital, Corbeil-Essonnes
| | - Nadine Morineau
- Hematology Department, Hospital La Roche Sur Yon, La Roche Sur Yon
| | | | | | - Jean Fontan
- Hematology Department, University Hospital, Besançon
| | - Yazid Arkam
- Hematology Department, Mulhouse Hospital, Mulhouse
| | - Magda Alexis
- Hematology Department, Orleans Hospital, Orleans
| | - Vincent Delwail
- Onco-Hematology Department, University Hospital Poitiers and INSERM, CIC 1402, University of Poitiers, Poitiers
| | | | - Loic Ysebaert
- Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse
| | - Barbara Burroni
- Assistance Publique – Hôpitaux de Paris (APHP), Hôpital Cochin, Department of Pathology, Centre de Recherche des Cordeliers, Sorbonne University, Inserm, UMRS 1138, Université Paris Cité, F-75006 Paris
| | - Mary Callanan
- Unit For Innovation in Genetics and Epigenetics and Oncology, Dijon University Hospital, Dijon, France
| | - Steven Le Gouill
- Institut Curie, Paris and Paris Saint Quentin University, UVSQ, Paris
| | - Rémy Gressin
- Université Grenoble Alpes, University Hospital, Institute For Advanced Biosciences (INSERM U1209, CNRS UMR 5309, UGA), Oncohematology Department, Grenoble
| |
Collapse
|
8
|
Liang Y, Wei X, Yue PJ, Zhang HC, Li ZN, Wang XX, Sun YY, Fu WN. MYCT1 inhibits hematopoiesis in diffuse large B-cell lymphoma by suppressing RUNX1 transcription. Cell Mol Biol Lett 2024; 29:5. [PMID: 38172714 PMCID: PMC10763471 DOI: 10.1186/s11658-023-00522-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The abnormality of chromosomal karyotype is one factor causing poor prognosis of lymphoma. In the analysis of abnormal karyotype of lymphoma patients, three smallest overlap regions were found, in which MYCT1 was located. MYCT1 is the first tumor suppressor gene cloned by our research team, but its studies relating to the occurrence and development of lymphoma have not been reported. METHODS R banding analyses were employed to screen the abnormality of chromosomal karyotype in clinical specimen and MYCT1 over-expression cell lines. FISH was to monitor MYCT1 copy number aberration. RT-PCR and Western blot were to detect the mRNA and protein levels of the MYCT1 and RUNX1 genes, respectively. The MYCT1 and RUNX1 protein levels in clinical specimen were evaluated by immunohistochemical DAB staining. The interaction between MYCT1 and MAX proteins was identified via Co-IP and IF. The binding of MAX on the promoter of the RUNX1 gene was detected by ChIP and Dual-luciferase reporter assay, respectively. Flow cytometry and CCK-8 assay were to explore the effects of MYCT1 and RUNX1 on the cell cycle and proliferation, respectively. RESULTS MYCT1 was located in one of three smallest overlap regions of diffuse large B-cell lymphoma, it altered chromosomal instability of diffuse large B-cell lymphoma cells. MYCT1 negatively correlated with RUNX1 in lymphoma tissues of the patients. MAX directly promoted the RUNX1 gene transcription by binding to its promoter region. MYCT1 may represses RUNX1 transcription by binding MAX in diffuse large B-cell lymphoma cells. MYCT1 binding to MAX probably suppressed RUNX1 transcription, leading to the inhibition of proliferation and cell cycle of the diffuse large B-cell lymphoma cells. CONCLUSION This study finds that there is a MYCT1-MAX-RUNX1 signaling pathway in diffuse large B-cell lymphoma. And the study provides clues and basis for the in-depth studies of MYCT1 in the diagnosis, treatment and prognosis of lymphoma.
Collapse
Affiliation(s)
- Ying Liang
- Department of Medical Genetics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, People's Republic of China
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Xin Wei
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Peng-Jie Yue
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - He-Cheng Zhang
- Department of Medical Genetics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, People's Republic of China
| | - Zhen-Ning Li
- Department of Oromaxillofacial-Head and Neck Surgery, Liaoning Province Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, People's Republic of China
| | - Xiao-Xue Wang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Yuan-Yuan Sun
- Department of Medical Genetics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, People's Republic of China.
| | - Wei-Neng Fu
- Department of Medical Genetics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, People's Republic of China.
| |
Collapse
|
9
|
Ramsower CA, Rosenthal A, Robetorye RS, Mwangi R, Maurer M, Villa D, McDonnell T, Feldman A, Cohen JB, Habermann T, Campo E, Clot G, Bühler MM, Kulis M, Martin-Subero JI, Giné E, Cook JR, Hill B, Raess PW, Beiske KH, Reichart A, Hartmann S, Holte H, Scott D, Rimsza L. Evaluation of clinical parameters and biomarkers in older, untreated mantle cell lymphoma patients receiving bendamustine-rituximab. Br J Haematol 2024; 204:160-170. [PMID: 37881141 PMCID: PMC11315408 DOI: 10.1111/bjh.19153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/27/2023]
Abstract
Mantle cell lymphoma (MCL) is clinically and biologically heterogeneous. While various prognostic features have been proposed, none currently impact therapy selection, particularly in older patients, for whom treatment is primarily dictated by age and comorbidities. Herein, we undertook a comprehensive comparison of clinicopathological features in a cohort of patients 60 years and older, uniformly treated with bendamustine and rituximab, with a median survival of >8 years. The strongest prognostic indicators in this cohort were a high-risk call by a simplified MCL international prognostic index (s-MIPI) (HR: 3.32, 95% CI: 1.65-6.68 compared to low risk), a high-risk call by MCL35 (HR: 10.34, 95% CI: 2.37-45.20 compared to low risk) and blastoid cytology (HR: 4.21, 95% CR: 1.92-9.22 compared to classic). Patients called high risk by both the s-MIPI and MCL35 had the most dismal prognosis (HR: 11.58, 95% CI: 4.10-32.72), while those with high risk by either had a moderate but clinically relevant prognosis (HR: 2.95, 95% CI: 1.49-5.82). A robust assay to assess proliferation, such as MCL35, along with stringent guidelines for cytological evaluation of MCL, in combination with MIPI, may be a strong path to risk-stratify older MCL patients in future clinical trials.
Collapse
Affiliation(s)
| | - Allison Rosenthal
- Division of Hematology and Medical Oncology, Mayo Clinic, Arizona, Phoenix, USA
| | - Ryan S Robetorye
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, Arizona, USA
| | - Raphael Mwangi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Diego Villa
- Division of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Tim McDonnell
- Department of Hematopathology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew Feldman
- Division of Hematopathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jonathon B Cohen
- Department of Hematology and Medical Oncology, Emory University-Winship Cancer Institute, Atlanta, Georgia, USA
| | | | - Elias Campo
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
- Laboratory of Pathology, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain
| | - Guillem Clot
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
| | - Marco M Bühler
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zürich, Switzerland
| | - Marta Kulis
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
| | - Jose Ignacio Martin-Subero
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, ICREA, Barcelona, Spain
| | - Eva Giné
- Department of Hematology, Hospital Clinic of the University of Barcelona, Barcelona, Spain
| | - James R Cook
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Brian Hill
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Philipp W Raess
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Klaus H Beiske
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Alexander Reichart
- Hematology and Oncology, Medical Office of Dres. Brudler/Reichart, Ausburg, Germany
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt am Main, Frankfurt, Germany
| | - Harald Holte
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Center for B Cell Malignancies, Oslo, Norway
| | - David Scott
- Department of Lymphoid Cancer Research, BC Cancer Centre, Vancouver, British Columbia, Canada
| | - Lisa Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, Arizona, USA
| |
Collapse
|
10
|
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: 4] [Impact Index Per Article: 4.0] [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
|
11
|
Gordon MJ, Bond DA, Kittai AS, Amirmokhtari N, Steinbrunner A, Huffman A, Orellana-Noia V, Shouse G, Cohen JB, Phillips T, Danilov AV. TRES, a validated three-factor comorbidity score, is associated with survival in older patients with mantle cell lymphoma. Haematologica 2023; 108:3110-3114. [PMID: 37226703 PMCID: PMC10620592 DOI: 10.3324/haematol.2023.283074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/06/2023] [Indexed: 05/26/2023] Open
Affiliation(s)
- Max J Gordon
- University of Texas M.D. Anderson Cancer Center, Houston, TX.
| | - David A Bond
- Ohio State University, James Cancer Center, Columbus, OH
| | - Adam S Kittai
- Ohio State University, James Cancer Center, Columbus, OH
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Lefebvre C, Veronese L, Nadal N, Gaillard JB, Penther D, Daudignon A, Chauzeix J, Nguyen-Khac F, Chapiro E. Cytogenetics in the management of mature B-cell non-Hodgkin lymphomas: Guidelines from the Groupe Francophone de Cytogénétique Hematologique (GFCH). Curr Res Transl Med 2023; 71:103425. [PMID: 38016420 DOI: 10.1016/j.retram.2023.103425] [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: 07/03/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/30/2023]
Abstract
Non-Hodgkin lymphomas (NHL) consist of a wide range of clinically, phenotypically and genetically distinct neoplasms. The accurate diagnosis of mature B-cell non-Hodgkin lymphoma relies on a multidisciplinary approach that integrates morphological, phenotypical and genetic characteristics together with clinical features. Cytogenetic analyses remain an essential part of the diagnostic workup for mature B-cell lymphomas. Karyotyping is particularly useful to identify hallmark translocations, typical cytogenetic signatures as well as complex karyotypes, all bringing valuable diagnostic and/or prognostic information. Besides the well-known recurrent chromosomal abnormalities such as, for example, t(14;18)(q32;q21)/IGH::BCL2 in follicular lymphoma, recent evidences support a prognostic significance of complex karyotype in mantle cell lymphoma and Waldenström macroglobulinemia. Fluorescence In Situ Hybridization is also a key analysis playing a central role in disease identification, especially in genetically-defined entities, but also in predicting transformation risk or prognostication. This can be exemplified by the pivotal role of MYC, BCL2 and/or BCL6 rearrangements in the diagnostic of aggressive or large B-cell lymphomas. This work relies on the World Health Organization and the International Consensus Classification of hematolymphoid tumors together with the recent cytogenetic advances. Here, we review the various chromosomal abnormalities that delineate well-established mature B-cell non-Hodgkin lymphoma entities as well as newly recognized genetic subtypes and provide cytogenetic guidelines for the diagnostic management of mature B-cell lymphomas.
Collapse
Affiliation(s)
- C Lefebvre
- Unité de Génétique des Hémopathies, Service d'Hématologie Biologique, CHU Grenoble Alpes, Grenoble, France.
| | - L Veronese
- Service de Cytogénétique Médicale, CHU Estaing, 1 place Lucie et Raymond Aubrac, 63003 Clermont-Ferrand; EA7453 CHELTER, Université Clermont Auvergne, France
| | - N Nadal
- Service de génétique chromosomique et moléculaire, CHU Dijon, Dijon, France
| | - J-B Gaillard
- Unité de Génétique Chromosomique, Service de Génétique moléculaire et cytogénomique, CHU Montpellier, Montpellier, France
| | - D Penther
- Laboratoire de Génétique Oncologique, Centre Henri Becquerel, Rouen, France
| | - A Daudignon
- Laboratoire de Génétique Médicale - Hôpital Jeanne de Flandre - CHRU de Lille, France
| | - J Chauzeix
- Service d'Hématologie biologique CHU de Limoges - CRIBL, UMR CNRS 7276/INSERM 1262, Limoges, France
| | - F Nguyen-Khac
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS_1138, Drug Resistance in Hematological Malignancies Team, F-75006 Paris, France; Sorbonne Université, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Service d'Hématologie Biologique, F-75013 Paris, France
| | - E Chapiro
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS_1138, Drug Resistance in Hematological Malignancies Team, F-75006 Paris, France; Sorbonne Université, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Service d'Hématologie Biologique, F-75013 Paris, France
| |
Collapse
|
13
|
de Pádua Covas Lage LA, Elias MDV, Reichert CO, Culler HF, de Freitas FA, de Oliveira Costa R, Rocha V, da Siqueira SAC, Pereira J. Up-Front ASCT Overcomes the Survival Benefit Provided by HDAC-Based Induction Regimens in Mantle Cell Lymphoma: Data from a Real-Life and Long-Term Cohort. Cancers (Basel) 2023; 15:4759. [PMID: 37835453 PMCID: PMC10571660 DOI: 10.3390/cancers15194759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Mantle cell lymphoma (MCL) is a rare malignancy with heterogeneous behavior. Despite the therapeutic advances recently achieved, MCL remains incurable. Currently, the standard of care for young and fit patients involves induction immunochemotherapy followed by up-front autologous stem cell transplantation (ASCT). However, the role of more intensive induction regimens, such as those based on high doses of cytarabine (HDAC), remains controversial in the management of ASCT-eligible patients. METHODS This retrospective, observational, and single-center study involved 165 MCL patients treated at the largest oncology center in Latin America from 2010 to 2022. We aimed to assess outcomes, determine survival predictors, and compare responses between different primary therapeutic strategies, with a focus on assessing the impact of HDAC-based regimens on outcomes in ASCT-eligible patients. RESULTS The median age at diagnosis was 65 years (38-89 years), and 73.9% were male. More than 90% of the cases had a classic nodal form (cnMCL), 76.4% had BM infiltration, and 56.4% presented splenomegaly. Bulky ≥ 7 cm, B-symptoms, ECOG ≥ 2, and advanced-stage III/IV were observed in 32.7%, 64.8%, 32.1%, and 95.8%, respectively. Sixty-four percent of patients were categorized as having high-risk MIPI. With a median follow-up of 71.1 months, the estimated 2-year OS and EFS were 64.1% and 31.8%, respectively. Patients treated with (R)-HDAC-based regimens had a higher ORR (85.9% vs. 65.7%, p = 0.007) compared to those receiving (R)-CHOP, as well as lower POD-24 rates (61.9% vs. 80.4%, p = 0.043) and lower mortality (43.9% vs. 68.6%, p = 0.004). However, intensified induction regimens with (R)-HDAC were not associated with a real OS benefit in MCL patients undergoing up-front consolidation with ASCT (2-year OS: 88.7% vs. 78.8%, p = 0.289). Up-front ASCT was independently associated with increased OS (p < 0.001), EFS (p = 0.005), and lower POD-24 rates (p < 0.001) in MCL. Additionally, CNS infiltration, TLS, hypoalbuminemia, and the absence of remission after induction were predictors of poor OS. CONCLUSIONS In the largest Latin American cohort of MCL patients, we confirmed the OS benefit promoted by up-front consolidation with ASCT in young and fit patients, regardless of the intensity of the immunochemotherapy regimen used in the pre-ASCT induction. Although HDAC-based regimens were not associated with an unequivocal increase in OS for ASCT-eligible patients, it was associated with higher ORR and lower rates of early relapses for the whole cohort.
Collapse
Affiliation(s)
- Luís Alberto de Pádua Covas Lage
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
| | - Marcela do Vale Elias
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
| | - Cadiele Oliana Reichert
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
| | - Hebert Fabrício Culler
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
| | - Fábio Alessandro de Freitas
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
| | - Renata de Oliveira Costa
- Department of Hematology and Hemotherapy, Faculty of Medical Sciences of Santos (FCMS), Santos 01238-010, SP, Brazil
- Hospital Alemão Osvaldo Cruz (HAOC), São Paulo 01323-020, SP, Brazil
| | - Vanderson Rocha
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
- Fundação Pró-Sangue, Blood Bank of São Paulo, São Paulo 05403-000, SP, Brazil
- Department of Hematology, Churchill Hospital, Oxford University, Oxford OX3 7LE, UK
| | | | - Juliana Pereira
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
- Hospital Alemão Osvaldo Cruz (HAOC), São Paulo 01323-020, SP, Brazil
| |
Collapse
|
14
|
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
|
15
|
Gaudio F, Dicataldo M, Di Giovanni F, Cazzato G, d'Amati A, Perrone T, Masciopinto P, Laddaga FE, Musto P, Maiorano E, Ingravallo G. Prognostic Role of CDKN2A Deletion and p53 Expression and Association With MIPIb in Mantle Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:599-605. [PMID: 37147150 DOI: 10.1016/j.clml.2023.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/30/2023] [Accepted: 04/16/2023] [Indexed: 05/07/2023]
Abstract
INTRODUCTION Mantle cell lymphoma (MCL) is a rare type of lymphoma, which despite improvements in therapies in recent decades, remains an incurable disease. There is currently no reliable marker of chemoresistance available. In this study, we investigated the prognostic role of MIPIb and the association with biological markers including SOX11, p53 expression, Ki-67, and CDKN2A. MATERIALS AND METHODS This retrospective study was focused on 23 patients with newly diagnosed classical MCL, treated at the University Hospital of Bari (Italy) between January 2006 and June 2019. RESULTS We identified a MIPIb value ≥ 5.4440 as a prognostic parameter that correlates with p53 expression and CDKN2A deletion. We also observed that patients with p53 overexpression had a significantly higher MIPIb (5.52 ± 0.53) which in 80% of patients had a value higher than 5.4440. On the other hand, CDKN2A deletion was found more frequently (75%) associated with MIPIb ≥5.4440. Only the CDKN2A deletion was associated with a higher proliferation index, with 66.7% of samples having Ki67 ≥30%. From the survival analysis we found that patients with p53 overexpression and CDKN2A deletion have a significantly worse prognosis with a median overall survival of 50 (P = .012) and 52 months (P = .018), respectively. CONCLUSION p53 expression and CDKN2A deletion represent a reliable pretreatment prognostic factor that identifies patients who do not benefit from currently used immunochemotherapy-based therapies and who are candidates for diversified treatments with the aim of improving prognosis. The MIPIb represents a prognostic index that correlates well with these biological alterations and can be used in clinical practice as their surrogate.
Collapse
Affiliation(s)
- Francesco Gaudio
- Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, Bari, Italy.
| | - Michele Dicataldo
- Unit of Hematology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Gerardo Cazzato
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), "Aldo Moro" University School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), "Aldo Moro" University School of Medicine, Bari, Italy
| | - Tommasina Perrone
- Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, Bari, Italy
| | | | | | - Pellegrino Musto
- Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, Bari, Italy; Section of Hematology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), "Aldo Moro" University School of Medicine, Bari, Italy
| | - Eugenio Maiorano
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), "Aldo Moro" University School of Medicine, Bari, Italy
| | - Giuseppe Ingravallo
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), "Aldo Moro" University School of Medicine, Bari, Italy.
| |
Collapse
|
16
|
Epperla N, Switchenko J, 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, Bond DA, Hamadani M, Fenske TS, Martin P, Malecek MK, Kahl BS, Flowers CR, Link BK, Kaplan LD, Inwards DJ, Feldman AL, Hsi ED, Maddocks K, Blum KA, Bartlett NL, Cerhan JR, Leonard JP, Habermann TM, Maurer MJ, Cohen JB. Impact of diagnosis to treatment interval in patients with newly diagnosed mantle cell lymphoma. Blood Adv 2023; 7:2287-2296. [PMID: 36516079 PMCID: PMC10225877 DOI: 10.1182/bloodadvances.2022009225] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 12/15/2022] Open
Abstract
The prognostic relevance of diagnosis to treatment interval (DTI) in patients with newly diagnosed mantle cell lymphoma (MCL) is unknown. Hence, we sought to evaluate the impact of DTI on outcomes in MCL using 3 large datasets (1) the University of Iowa/Mayo Clinic Specialized Program of Research Excellence Molecular Epidemiology Resource, (2) patients enrolled in the ALL Age Asthma Cohort/CALGB 50403, and (3) a multisitecohort of patients with MCL. Patients were a priori divided into 2 groups, 0 to 14 days (short DTI) and 15 to 60 days (long DTI). The patients in whom observation was deemed appropriate were excluded. One thousand ninety-seven patients newly diagnosed with MCL and available DTI were included in the study. The majority (73%) had long DTI (n=797). Patients with short DTI had worse eastern cooperative oncology group performance status (ECOG PS ≥2), higher lactate dehydrogenase, bone marrow involvement, more frequent B symptoms, higher MCL International Prognostic Index (MIPI ≥6.2), and were less likely to receive intensive induction therapy than long DTI group. The median progression-free survival (2.5 years vs 4.8 years, p<0.0001) and overall survival (7.8 years vs. 11.8 years, p<0.0001) were significantly inferior in the short DTI group than the long DTI cohort and remained significant for progression-free survival and overall survival in multivariable analysis. We show that the DTI is an important prognostic factor in patients newly diagnosed with MCL and is strongly associated with adverse clinical factors and poor outcomes. DTI should be reported in all the patients newly diagnosed with MCL who are enrolling in clinical trials and steps must be taken to ensure selection bias is avoided.
Collapse
Affiliation(s)
- Narendranath Epperla
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, OH
| | | | | | - James N. Gerson
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stefan K. Barta
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Max J. Gordon
- Department of Medicine, MD Anderson Cancer Center, Houston, TX
| | | | - Natalie S. Grover
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Stephanie Mathews
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Madelyn Burkart
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Reem Karmali
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Yazeed Sawalha
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, OH
| | | | - Nilanjan Ghosh
- Department of Medicine, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Steven I. Park
- Department of Medicine, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - David A. Bond
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, OH
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Timothy S. Fenske
- BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Peter Martin
- Department of Medicine, Weill Cornell Medical College, Manhattan, NY
| | - Mary-Kate Malecek
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Brad S. Kahl
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | | | - Brian K. Link
- Department of Medicine, University of Iowa, Iowa City, IA
| | | | | | | | - Eric D. Hsi
- Department of Medicine, Wake Forest University, Winston-Salem, NC
| | - Kami Maddocks
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, OH
| | | | - Nancy L. Bartlett
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | | | - John P. Leonard
- Department of Medicine, Weill Cornell Medical College, Manhattan, NY
| | | | | | | |
Collapse
|
17
|
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
|
18
|
Wang Y, Jain P, Locke FL, Maurer MJ, Frank MJ, Munoz JL, Dahiya S, Beitinjaneh AM, Jacobs MT, Mcguirk JP, Vose JM, Goy A, Andreadis C, Hill BT, Dorritie KA, Oluwole OO, Deol A, Paludo J, Shah B, Wang T, Banerjee R, Miklos DB, Rapoport AP, Lekakis L, Ghobadi A, Neelapu SS, Lin Y, Wang ML, Jain MD. Brexucabtagene Autoleucel for Relapsed or Refractory Mantle Cell Lymphoma in Standard-of-Care Practice: Results From the US Lymphoma CAR T Consortium. J Clin Oncol 2023; 41:2594-2606. [PMID: 36753699 PMCID: PMC10489553 DOI: 10.1200/jco.22.01797] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/02/2022] [Accepted: 12/15/2022] [Indexed: 02/10/2023] Open
Abstract
PURPOSE Brexucabtagene autoleucel (brexu-cel) is an autologous CD19-directed chimeric antigen receptor (CAR) T-cell therapy approved for relapsed/refractory mantle cell lymphoma (MCL). This therapy was approved on the basis of the single-arm phase II ZUMA-2 trial, which showed best overall and complete response rates of 91% and 68%, respectively. We report clinical outcomes with brexu-cel in the standard-of-care setting for the approved indication. PATIENTS AND METHODS Patients who underwent leukapheresis between August 1, 2020 and December 31, 2021, at 16 US institutions, with an intent to manufacture commercial brexu-cel for relapsed/refractory MCL, were included. Patient data were collected for analyses of responses, outcomes, and toxicities as per standard guidelines. RESULTS Of 189 patients who underwent leukapheresis, 168 (89%) received brexu-cel infusion. Of leukapheresed patients, 79% would not have met ZUMA-2 eligibility criteria. Best overall and complete response rates were 90% and 82%, respectively. At a median follow-up of 14.3 months after infusion, the estimates for 6- and 12-month progression-free survival (PFS) were 69% (95% CI, 61 to 75) and 59% (95% CI, 51 to 66), respectively. The nonrelapse mortality was 9.1% at 1 year, primarily because of infections. Grade 3 or higher cytokine release syndrome and neurotoxicity occurred in 8% and 32%, respectively. In univariable analysis, high-risk simplified MCL international prognostic index, high Ki-67, TP53 aberration, complex karyotype, and blastoid/pleomorphic variant were associated with shorter PFS after brexu-cel infusion. Patients with recent bendamustine exposure (within 24 months before leukapheresis) had shorter PFS and overall survival after leukapheresis in intention-to-treat univariable analysis. CONCLUSION In the standard-of-care setting, the efficacy and toxicity of brexu-cel were consistent with those reported in the ZUMA-2 trial. Tumor-intrinsic features of MCL, and possibly recent bendamustine exposure, may be associated with inferior efficacy outcomes.
Collapse
Affiliation(s)
| | - Preetesh Jain
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | - Saurabh Dahiya
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Amer M. Beitinjaneh
- University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL
| | - Miriam T. Jacobs
- Washington University School of Medicine, Siteman Cancer Center, St Louis, MO
| | | | - Julie M. Vose
- University of Nebraska Medical Center, Buffett Cancer Center, Omaha, NE
| | - Andre Goy
- John Theurer Cancer Center, Hackensack Meridian Health, Hackensack, NJ
| | | | | | | | | | - Abhinav Deol
- Wayne State University, Karmanos Cancer Institute, Detroit, MI
| | | | | | - Trent Wang
- University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL
| | - Rahul Banerjee
- University of California San Francisco, San Francisco, CA
| | | | - Aaron P. Rapoport
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Lazaros Lekakis
- University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL
| | - Armin Ghobadi
- Washington University School of Medicine, Siteman Cancer Center, St Louis, MO
| | | | - Yi Lin
- Mayo Clinic, Rochester, MN
| | - Michael L. Wang
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
19
|
Lew TE, Minson A, Dickinson M, Handunnetti SM, Blombery P, Khot A, Anderson MA, Ritchie D, Tam CS, Seymour JF. Treatment approaches for patients with TP53-mutated mantle cell lymphoma. Lancet Haematol 2023; 10:e142-e154. [PMID: 36725119 DOI: 10.1016/s2352-3026(22)00355-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 02/01/2023]
Abstract
Mantle cell lymphoma is an uncommon subtype of lymphoma characterised by clinical and biological heterogeneity. Although most patients with mantle cell lymphoma have durable responses after chemoimmunotherapy, there is a need to prospectively identify high-risk subsets of patients for whom disease control with standard chemotherapy will be short lived. Among the available prognostic factors, TP53 mutations are uniquely informative owing to their strong association with early disease progression and death among patients receiving conventional chemoimmunotherapy, with the highest negative prognostic value compared with other established risk indicators, including the mantle cell lymphoma international prognostic index, histological features, elevated Ki-67, and other genetic lesions. The poor outcomes for patients with TP53-mutated mantle cell lymphoma receiving chemoimmunotherapy and second-line Bruton tyrosine kinase inhibitors represent an urgent need for alternative approaches. In this Review, we synthesise the available data to inform the management of this high-risk subset of patients and present a treatment strategy prioritising clinical trials and early use of cellular therapies.
Collapse
Affiliation(s)
- Thomas E Lew
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia; Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Adrian Minson
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Michael Dickinson
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Sasanka M Handunnetti
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Piers Blombery
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia; Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Amit Khot
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Mary Ann Anderson
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia; Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - David Ritchie
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Constantine S Tam
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia; Department of Haematology, The Alfred Hospital, Melbourne, VIC, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - John F Seymour
- Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
20
|
Romancik JT, Gerber DG, Zhuang T, Cohen JB. SOHO State of the Art Updates and Next Questions: Managing Relapsed Mantle Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:557-565. [PMID: 35123927 DOI: 10.1016/j.clml.2022.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/20/2021] [Accepted: 01/08/2022] [Indexed: 10/19/2022]
Abstract
Mantle cell lymphoma (MCL) is a rare subtype of B-cell non-Hodgkin lymphoma i.e., incurable with current therapies. While some patients experience prolonged remissions following initial therapy, most will have a relapsing-remitting course requiring several lines of treatment over the course of their disease. Several targeted therapies are now available to treat patients with relapsed MCL. The Bruton's tyrosine kinase (BTK) inhibitors, including ibrutinib, acalabrutinib, and zanubrutinib, are highly active in MCL and currently approved for treating patients with relapsed disease. Bortezomib and lenalidomide are available as monotherapy or in combination with other agents. Venetoclax is active and can be considered for use in relapsed MCL, although it is not currently approved by regulatory agencies. Chimeric antigen receptor T-cell (CAR-T) therapy with brexucabtagene autoleucel yields high response rates and is now approved for patients with relapsed MCL. Allogeneic stem cell transplant remains an option for a small subset of medically fit and motivated patients who have progressed through multiple lines of therapy, although its use is limited by substantial toxicity. There is currently no standard approach to sequencing therapies for patients with relapsed MCL, and the ability to utilize disease biologic and clinical characteristics to guide treatment decisions in this setting remains limited. In this review, we summarize the current evidence to guide the management of patients with relapsed MCL, review emerging agents and combination therapies that are under investigation, and outline our current treatment approach for these patients.
Collapse
Affiliation(s)
- Jason T Romancik
- Department of Hematology and Medical Oncology; Winship Cancer Institute at Emory University, Atlanta, GA
| | - Drew G Gerber
- Department of Hematology and Medical Oncology; Winship Cancer Institute at Emory University, Atlanta, GA
| | - Tony Zhuang
- Department of Hematology and Medical Oncology; Winship Cancer Institute at Emory University, Atlanta, GA
| | - Jonathon B Cohen
- Department of Hematology and Medical Oncology; Winship Cancer Institute at Emory University, Atlanta, GA.
| |
Collapse
|
21
|
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
|
22
|
Jain P, Wang ML. Mantle cell lymphoma in 2022-A comprehensive update on molecular pathogenesis, risk stratification, clinical approach, and current and novel treatments. Am J Hematol 2022; 97:638-656. [PMID: 35266562 DOI: 10.1002/ajh.26523] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 12/21/2022]
Abstract
The field of mantle cell lymphoma (MCL) has witnessed remarkable progress due to relentless advances in molecular pathogenesis, prognostication, and newer treatments. MCL consists of a spectrum of clinical subtypes. Rarely, atypical cyclin D1-negative MCL and in situ MCL neoplasia are identified. Prognostication of MCL is further refined by identifying somatic mutations (such as TP53, NSD2, KMT2D), methylation status, chromatin organization pattern, SOX-11 expression, minimal residual disease (MRD), and genomic clusters. Lymphoid tissue microenvironment studies demonstrated the role of B-cell receptor signaling, nuclear factor kappa B (NF-kB), colony-stimulating factor (CSF)-1, the CD70-SOX-11 axis. Molecular mechanism of resistance, mutation dynamics, and pathogenic pathways (B-cell receptor (BCR), oxidative phosphorylation, and MYC) were identified in mediating resistance to various treatments (bruton tyrosine kinase (BTK) inhibitors [ibrutinib, acalabrutinib]. Treatment options range from conventional chemoimmunotherapy and stem cell transplantation (SCT) to targeted therapies against BTK (covalent and noncovalent), Bcl2, ROR1, cellular therapy such as anti-CD19 chimeric antigen receptor therapy (CAR-T), and most recently bispecific antibodies against CD19 and CD20. MCL patients frequently relapse. Complex pathogenesis and the management of patients with progression after treatment with BTK/Bcl2 inhibitors and CAR-T (triple-resistant MCL) remain a challenge. Next-generation clinical trials incorporating newer agents and concurrent translational and molecular investigations are ongoing.
Collapse
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma. Mantle cell lymphoma center of excellence The University of Texas MD Anderson Cancer Center Houston Texas USA
| | - Michael L. Wang
- Department of Lymphoma/Myeloma. Mantle cell lymphoma center of excellence The University of Texas MD Anderson Cancer Center Houston Texas USA
| |
Collapse
|
23
|
Nguyen-Khac F, Bidet A, Daudignon A, Lafage-Pochitaloff M, Ameye G, Bilhou-Nabéra C, Chapiro E, Collonge-Rame MA, Cuccuini W, Douet-Guilbert N, Eclache V, Luquet I, Michaux L, Nadal N, Penther D, Quilichini B, Terre C, Lefebvre C, Troadec MB, Véronèse L. The complex karyotype in hematological malignancies: a comprehensive overview by the Francophone Group of Hematological Cytogenetics (GFCH). Leukemia 2022; 36:1451-1466. [DOI: 10.1038/s41375-022-01561-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/16/2022]
|
24
|
Chen W, Liu H, Wang P, Li G. Clinical and biological features of mantle cell lymphoma patients with co-expression of CD10 and BCL-6: a retrospective study. Transl Cancer Res 2022; 10:4786-4793. [PMID: 35116331 PMCID: PMC8798977 DOI: 10.21037/tcr-21-882] [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: 05/22/2021] [Accepted: 09/03/2021] [Indexed: 11/13/2022]
Abstract
Background Mantle cell lymphoma (MCL) with co-expression of CD10 and BCL-6 was scarcely reported, and its biological features were largely remained unknown. Thus, this study aimed to describe the clinical and biological features, as well as outcome of MCL patients with co-expression of CD10 and BCL-6. Methods A total of 104 cases of MCL who were admitted to our hospital between January 2011 and October 2018 were recruited. Those patients were diagnosed according to the 2016 World Health Organization (WHO) classification system for tumors of the hematopoietic and lymphoid tissues, in compliance with the results of cytomorphology and immunohistochemical analysis. Patients were followed up through telephone interviews, medical records. Differences in age, gender, leukocyte count, lactate dehydrogenase (LDH) level, beta-2 microglobulin (β2-MG) level and results of immunohistochemistry were analyzed. Then the event-free survival (EFS) rate and overall survival (OS) rate were performed by the Kaplan-Meier method and log-rank test. Results The results showed that, in total, 5 patients had superficial lymph node enlargement, 2 patients suffered from abdominal discomfort, and 1 patient’s red blood cell (RBC) count was abnormal at the time of diagnosis. All patients were in stage IV, 75% had bone marrow involvement, and 75% and 100% of patients had elevated levels of LDH and β2-MG, respectively. Three classic genes and five variants were involved in the 8 patients. MCL patients with the co-expression of CD10 and BCL-6 had higher Ki-67 index, white blood cell (WBC) count, LDH level, and β2-MG level than those of without co-expression of CD10 and BCL-6 (P=0.025, 0.038, 0.015, and 0.021, respectively). Besides, MCL patients with CD10 and BCL-6 co-expression had shorter OS and EFS (χ2=6.401 and 5.975; P=0.011 and 0.015, respectively), indicating patients’ susceptibility to get complex karyotype and TP53 abnormality. Conclusions MCL patients with co-expression of CD10 and BCL-6 were more likely to have bone marrow involvement, higher Ki-67 index, increased WBC count, and elevated levels of LDH and β2-MG at the time of diagnosis, then might has complex cytogenetic and poor prognosis.
Collapse
Affiliation(s)
- Wanzi Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hangmin Liu
- Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Pengcheng Wang
- Department of pathology, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guoping Li
- Department of pathology, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
25
|
Eyre TA, Cheah CY, Wang ML. Therapeutic options for relapsed/refractory mantle cell lymphoma. Blood 2022; 139:666-677. [PMID: 34679161 PMCID: PMC9710495 DOI: 10.1182/blood.2021013326] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/20/2021] [Indexed: 02/05/2023] Open
Abstract
Mantle cell lymphoma (MCL) is an uncommon subtype of non-Hodgkin lymphoma in which immunochemotherapy, with or without high-dose therapy, and autologous stem cell transplantation remain standard frontline therapies. Despite their clear efficacy, patients inevitably relapse and require subsequent therapy. In this review, we discuss the key therapeutic approaches in the management of relapsed MCL, covering in depth the data supporting the use of covalent Bruton tyrosine kinase (BTK) inhibitors at first or subsequent relapse. We describe the outcomes of patients progressing through BTK inhibitors and discuss the mechanisms of covalent BTKi resistance and treatment options after covalent treatment with BTKi. Options in this setting may depend on treatment availability, patient's and physician's preference, and the patient's age and comorbidity status. We discuss the rapid recent development of anti-CD19 chimeric antigen receptor T-cell therapy, as well as the utility of allogenic stem cell transplantation and novel therapies, such as noncovalent, reversible BTK inhibitors; ROR1 antibody drug conjugates; and bispecific antibodies.
Collapse
Affiliation(s)
- Toby A. Eyre
- Haematology and Cancer Centre, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Chan Y. Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Medical School, University of Western Australia, Perth, WA, Australia
| | - Michael L. Wang
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
26
|
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
|
27
|
Bond DA, Switchenko JM, Villa D, Maddocks K, Churnetski M, Gerrie AS, Goyal S, Shanmugasundaram K, Calzada O, Kolla B, Bachanova V, Gerson JN, Barta SK, Hill BT, Sawalha Y, Martin P, Maldonado E, Gordon M, Danilov AV, Grover NS, Mathews S, Burkart M, Karmali R, Ghosh N, Park SI, Epperla N, Badar T, Guo J, Hamadani M, Fenske TS, Malecek MK, Kahl BS, Flowers CR, Blum KA, Cohen JB. 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] [MESH Headings] [Grants] [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
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
Affiliation(s)
- David A. Bond
- Division of Hematology, The Ohio State University, Columbus, OH
| | - Jeffrey M. Switchenko
- Winship Cancer Institute, Biostatistics and Bioinformatics, Emory University, Atlanta, GA
| | - Diego Villa
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BC, Canada
- Division of Medical Oncology, University of British Columbia, Vancouver, BC, Canada
| | - Kami Maddocks
- Division of Hematology, The Ohio State University, Columbus, OH
| | - Michael Churnetski
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Alina S. Gerrie
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BC, Canada
- Division of Medical Oncology, University of British Columbia, Vancouver, BC, Canada
| | - Subir Goyal
- Winship Cancer Institute, Biostatistics and Bioinformatics, Emory University, Atlanta, GA
| | - Krithika Shanmugasundaram
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Oscar Calzada
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Bhaskar Kolla
- Division of Hematology Oncology and Transplantation, University of Minnesota, Minneapolis, MN
| | - Veronika Bachanova
- Division of Hematology Oncology and Transplantation, University of Minnesota, Minneapolis, MN
| | - James N. Gerson
- Division of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stefan K. Barta
- Division of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Brian T. Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute,Cleveland Clinic, Cleveland, OH
| | - Yazeed Sawalha
- Division of Hematology, The Ohio State University, Columbus, OH
- Department of Hematology and Medical Oncology, Taussig Cancer Institute,Cleveland Clinic, Cleveland, OH
| | - Peter Martin
- Division of Hematology and Oncology, Weill Cornell Medical Center/New York Presbyterian Hospital, New York, NY
| | - Edward Maldonado
- Knight Cancer Institute, Oregon Health Sciences University, Portland, OR
| | - Max Gordon
- Knight Cancer Institute, Oregon Health Sciences University, Portland, OR
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Alexey V. Danilov
- Knight Cancer Institute, Oregon Health Sciences University, Portland, OR
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA
| | - Natalie S. Grover
- Division of Hematology, University of North Carolina, Chapel Hill, NC
| | - Stephanie Mathews
- Division of Hematology, University of North Carolina, Chapel Hill, NC
| | - Madelyn Burkart
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Division of Hematology, Northwestern University, Chicago, IL
| | - Reem Karmali
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Division of Hematology, Northwestern University, Chicago, IL
| | | | | | | | - Talha Badar
- BMT Cellular Therapy Program, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI; and
| | - Jin Guo
- Division of Hematology and Oncology, Weill Cornell Medical Center/New York Presbyterian Hospital, New York, NY
| | - Mehdi Hamadani
- BMT Cellular Therapy Program, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI; and
| | - Timothy S. Fenske
- BMT Cellular Therapy Program, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI; and
| | | | - Brad S. Kahl
- Department of Medicine, Washington University, St Louis, MO
| | - Christopher R. Flowers
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Kristie A. Blum
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Jonathon B. Cohen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| |
Collapse
|
28
|
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
|
29
|
Ware AD, Davis K, Xian RR. Molecular Pathology of Mature Lymphoid Malignancies. Surg Pathol Clin 2021; 14:529-547. [PMID: 34373101 DOI: 10.1016/j.path.2021.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lymphoid malignancies are a broad and heterogeneous group of neoplasms. In the past decade, the genetic landscape of these tumors has been explored and cataloged in fine detail offering a glimpse into the mechanisms of lymphomagenesis and new opportunities to translate these findings into patient management. A myriad of studies have demonstrated both distinctive and overlapping molecular and chromosomal abnormalities that have influenced the diagnosis and classification of lymphoma, disease prognosis, and treatment selection.
Collapse
Affiliation(s)
- Alisha D Ware
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Katelynn Davis
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Rena R Xian
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Johns Hopkins School of Medicine, 1812 Ashland Avenue, Suite 200, Baltimore, MD 21205, USA.
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Abstract
PURPOSE OF REVIEW Mantle cell lymphoma (MCL) is a heterogenous disease with a variety of morphologic and genetic features, some of which are associated with high risk disease. Here we critically analyze the current state of the understanding of MCL's biology and its implications in therapy, with a focus on chemotherapy-free and targeted therapy regimens. RECENT FINDINGS Mantle cell lymphoma (MCL) is a rare subtype of non-Hodgkin's lymphoma, defined by a hallmark chromosomal translocation t(11;14) which leads to constitutive expression of cyclin D1. Recent discoveries in the biology of MCL have identified a number of factors, including TP53 mutations and complex karyotype, that lead to unresponsiveness to traditional chemoimmunotherapy and poor outcomes. Bruton tyrosine kinase inhibitors, BH3-mimetics and other novel agents thwart survival of the neoplastic B-cells in a manner independent of high-risk mutations and have shown promising activity in relapsed/refractory MCL. These therapies are being investigated in the frontline setting, while optimal responses to chemotherapy-free regimens, particularly in high-risk disease, might require combination approaches. High-risk MCL does not respond well to chemoimmunotherapy. Targeted agents are highly active in the relapsed refractory setting and show promise in high-risk disease. Novel approaches may soon replace the current standard of care in both relapsed and frontline settings.
Collapse
|
33
|
Jain P, Dreyling M, Seymour JF, Wang M. High-Risk Mantle Cell Lymphoma: Definition, Current Challenges, and Management. J Clin Oncol 2020; 38:4302-4316. [DOI: 10.1200/jco.20.02287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Martin Dreyling
- Medizinische Klinik III, Ludwig Maximilian University Klinikum München, München, Germany
| | - John F. Seymour
- Peter MacCallum Cancer Center, Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
34
|
Danilov AV, Persky DO. Incorporating acalabrutinib, a selective next-generation Bruton tyrosine kinase inhibitor, into clinical practice for the treatment of haematological malignancies. Br J Haematol 2020; 193:15-25. [PMID: 33216986 DOI: 10.1111/bjh.17184] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 01/15/2023]
Abstract
Greater understanding of the mechanisms involved in the disease progression of haematological malignancies has led to the introduction of novel targeted therapies with reduced toxicity compared with chemotherapy-based regimens, which has expanded the treatment options for patients with mantle cell lymphoma (MCL) and chronic lymphocytic leukaemia/small lymphocytic lymphoma (CLL/SLL). Ibrutinib is a first-in-class Bruton tyrosine kinase (BTK) inhibitor indicated for the treatment of patients with CLL/SLL or relapsed/refractory MCL. However, next-generation BTK inhibitors have been developed with improved specificity and the potential to reduce the off-target toxicity observed with ibrutinib. Acalabrutinib is a highly selective, next-generation BTK inhibitor, which was granted accelerated approval by the US Food and Drug Administration in 2017 for the treatment of adult patients with MCL who have received at least one prior therapy. In November 2019, it was also granted approval for the treatment of adult patients with CLL/SLL on the basis of two phase 3 clinical trials. This review describes the current understanding of acalabrutinib according to clinical study data for the treatment of MCL and CLL/SLL and shares recommendations from our practice on how it should be used when treating patients in the clinic, including dosing, administration and management of adverse events.
Collapse
|
35
|
Jain P, Nastoupil L, Westin J, Lee HJ, Navsaria L, Steiner RE, Ahmed S, Moghrabi O, Oriabure O, Chen W, Badillo M, Flowers CR, Wang ML. Outcomes and management of patients with mantle cell lymphoma after progression on brexucabtagene autoleucel therapy. Br J Haematol 2020; 192:e38-e42. [PMID: 33152104 DOI: 10.1111/bjh.17197] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Loretta Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason Westin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hun Ju Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lucy Navsaria
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raphael E Steiner
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sairah Ahmed
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Omar Moghrabi
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Onyeka Oriabure
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendy Chen
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria Badillo
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher R Flowers
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael L Wang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
36
|
Abstract
Blastoid and pleomorphic mantle cell lymphoma (MCL) are among the worst prognostic, aggressive histology, high-risk variants of MCL, and, in this article, they are presented as blastoid MCL. Blastoid MCL have not been systematically studied, probably due to their rarity. De novo blastoid MCLs have superior outcomes compared with transformed MCL. Compared with classic MCL, extranodal involvement (mainly skin, central nervous system), frequent relapses, and inferior responses to conventional chemoimmunotherapy, BTK inhibitors and venetoclax are frequent in blastoid MCL. KTE-X19 induces excellent response in blastoid MCL. Combinations with novel agents are actively investigated. This article presents a comprehensive review on blastoid MCL in 2020.
Collapse
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, TX 77030, USA
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, TX 77030, USA.
| |
Collapse
|
37
|
Abstract
Mantle cell lymphoma (MCL) is a biologically heterogeneous disease, and patients may experience a clinical course ranging from indolent to very aggressive. Observational studies suggest that a subset of patients can be safely observed for a period of months to years from initial diagnosis without adversely impacting their outcomes. However, identification of candidates for the "watch and wait" approach remains challenging because selection criteria are not well-defined. Studies that prospectively stratify patients on the basis of MCL biology and disease risk will be informative, and patients with indolent MCL may be ideal candidates for frontline clinical trials exploring novel therapies.
Collapse
Affiliation(s)
- Christina Lee
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 7th Floor, New York, NY 10065, USA
| | - Peter Martin
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 7th Floor, New York, NY 10065, USA.
| |
Collapse
|
38
|
Klener P. Advances in Molecular Biology and Targeted Therapy of Mantle Cell Lymphoma. Int J Mol Sci 2019; 20:ijms20184417. [PMID: 31500350 PMCID: PMC6770169 DOI: 10.3390/ijms20184417] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a heterogeneous malignancy with a broad spectrum of clinical behavior from indolent to highly aggressive cases. Despite the fact that MCL remains in most cases incurable by currently applied immunochemotherapy, our increasing knowledge on the biology of MCL in the last two decades has led to the design, testing, and approval of several innovative agents that dramatically changed the treatment landscape for MCL patients. Most importantly, the implementation of new drugs and novel treatment algorithms into clinical practice has successfully translated into improved outcomes of MCL patients not only in the clinical trials, but also in real life. This review focuses on recent advances in our understanding of the pathogenesis of MCL, and provides a brief survey of currently used treatment options with special focus on mode of action of selected innovative anti-lymphoma molecules. Finally, it outlines future perspectives of patient management with progressive shift from generally applied immunotherapy toward risk-stratified, patient-tailored protocols that would implement innovative agents and/or procedures with the ultimate goal to eradicate the lymphoma and cure the patient.
Collapse
Affiliation(s)
- Pavel Klener
- First Dept. of Medicine-Hematology, General University Hospital in Prague, 128 08 Prague, Czech Republic.
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, 128 53 Prague, Czech Republic.
| |
Collapse
|
39
|
Jain P, Wang M. Mantle cell lymphoma: 2019 update on the diagnosis, pathogenesis, prognostication, and management. Am J Hematol 2019; 94:710-725. [PMID: 30963600 DOI: 10.1002/ajh.25487] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 12/16/2022]
Abstract
Unprecedented advances in our understanding of the pathobiology, prognostication, and therapeutic options in mantle cell lymphoma (MCL) have taken place in the last few years. Heterogeneity in the clinical course of MCL-indolent vs aggressive-is further delineated by a correlation with the mutational status of the variable region of immunoglobulin heavy chain, methylation status, and SOX-11 expression. Cyclin-D1 negative MCL, in situ MCL neoplasia, and impact of the karyotype on prognosis are distinguished. Apart from Ki-67% and morphology pattern (classic vs blastoid/pleomorphic), the proliferation gene signature has helped to further refine prognostication. Studies focusing on mutational dynamics and clonal evolution on Bruton's tyrosine kinase (BTK) inhibitors (ibrutinib, acalabrutinib) and/or Bcl2 antagonists (venetoclax) have further clarified the prognostic impact of somatic mutations in TP53, BIRC3, CDKN2A, MAP3K14, NOTCH2, NSD2, and SMARCA4 genes. In therapy, long-term follow-up on chemo-immunotherapy studies has demonstrated durable remissions in some patients; however, long-term toxicities, especially from second cancers, are a serious concern with chemotherapy. The therapeutic options in MCL are constantly evolving, with dramatic responses from nonchemotherapeutic agents (ibrutinib, acalabrutinib, and venetoclax). Chimeric antigen receptor therapy and combinations of nonchemotherapeutic agents are actively being studied and our focus is shifting toward making the treatment of MCL chemotherapy-free. Still, MCL remains incurable. The following aspects of MCL continue to pose a challenge: disease transformation, role of the cytokine-microenvironmental milieu, incorporation of positron emission tomography-computerized tomography imaging, minimal residual disease in the prognosis, circulating tumor DNA testing for clonal evolution, predicting resistance to BTK inhibitors, and optimal management of patients who progress on BTK/Bcl2 inhibitors. Next-generation clinical trials should incorporate nonchemotherapeutic agents and personalize the treatment based upon the genomic profile of individual patient. Recent advances in the field of MCL are reviewed.
Collapse
Affiliation(s)
- Preetesh Jain
- Division of Cancer Medicine, Department of Lymphoma/MyelomaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Michael Wang
- Division of Cancer Medicine, Department of Lymphoma/MyelomaThe University of Texas MD Anderson Cancer Center Houston Texas
| |
Collapse
|
40
|
Affiliation(s)
- I Brian Greenwell
- a Division of Hematology and Medical Oncology , Medical University of South Carolina Hollings Cancer Center , Charleston , SC , USA
| | - Jonathon B Cohen
- b Department of Hematology and Medical Oncology , Emory University Winship Cancer Institute , Atlanta , GA , USA
| |
Collapse
|
41
|
Obr A, Procházka V, Jirkuvová A, Urbánková H, Kriegova E, Schneiderová P, Vatolíková M, Papajík T. TP53 Mutation and Complex Karyotype Portends a Dismal Prognosis in Patients With Mantle Cell Lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:762-768. [PMID: 30146365 DOI: 10.1016/j.clml.2018.07.282] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/27/2018] [Accepted: 07/09/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND TP53 mutation (TP53mut) and a complex karyotype (CK) were shown to be predictors of poor outcome in mantle-cell lymphoma (MCL). In this study we examined the combined effect of both of these risk factors. PATIENTS AND METHODS Patients diagnosed with MCL between January 2000 and December 2014 (n = 74) were evaluated. Forty-eight of them had available material for TP53 and cytogenetic examination. We analyzed the prognostic effect of combined TP53mut and CK in the cohort of patients treated with rituximab-containing therapy. RESULTS Three-year (3-y) overall survival (OS) and 3-y progression-free survival (PFS) in CK patients were shorter compared with non-CK (P = .001 for OS; P = .02 for PFS). TP53mut was a predictor of shorter survival compared with TP53 wild type (OS and PFS; P < .001). The incidence of TP53mut was not significantly associated with CK (P = .240). CK and TP53mut were predictors of inferior PFS and OS independent of age and Mantle-Cell Lymphoma International Prognostic Index, with hazard ratios of 2.35 (P = .024), 4.50 (P < .001) for PFS and 4.31 (P < .001), 5.46 (P < .001) for OS analysis in the CK and TP53mut groups, respectively. The combination of TP53mut and CK status stratified the patients into 3 prognostic groups (P < .001) with the worst outcome in patients with CK and TP53mut. CONCLUSION TP53 mutation and CK occurred independently and patients harboring both had a dismal prognosis. The study suggests the importance of molecular cytogenetics and examination of the TP53mut status to be performed simultaneously before treatment.
Collapse
Affiliation(s)
- Aleš Obr
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Vít Procházka
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic.
| | - Andrea Jirkuvová
- Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Helena Urbánková
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Eva Kriegova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Petra Schneiderová
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Michaela Vatolíková
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Tomáš Papajík
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| |
Collapse
|