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
|
Barraclough A, Tang C, Lasica M, Smyth E, Cirillo M, Mutsando H, Cheah CY, Ku M. Diagnosis and management of mantle cell lymphoma: a consensus practice statement from the Australasian Lymphoma Alliance. Intern Med J 2025; 55:117-129. [PMID: 39578957 DOI: 10.1111/imj.16561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 10/13/2024] [Indexed: 11/24/2024]
Abstract
Mantle cell lymphoma (MCL) is a clinically heterogeneous B-cell neoplasm with unique clinicopathological features, accounting for 5% of all non-Hodgkin lymphoma. Although for many chemoimmunotherapy can lead to durable remissions, those with poor baseline prognostic factors, namely blastoid morphology, TP53 aberrancy and Ki67 >30%, will have less durable responses to conventional therapies. With this in mind, clinical trials have focused on novel targeted therapies to improve outcomes. This review details the recent advances in the understanding of MCL biology and outlines the recommended diagnostic strategies and evidence-based approaches to treatment.
Collapse
Affiliation(s)
- Allison Barraclough
- Department of Haematology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Catherine Tang
- Department of Haematology, Gosford Hospital, Gosford, New South Wales, Australia
- School of Medicine and Public Health, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Masa Lasica
- Department of Haematology, St Vincent's Hospital Melbourne, Melbourne, Victoria, Australia
| | - Elizabeth Smyth
- Department of Haematology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Melita Cirillo
- Department of Haematology, Royal Perth Hospital, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Howard Mutsando
- Cancer Services, Toowoomba Hospital, Toowoomba, Queensland, Australia
- Toowoomba Rural Clinical School, University of Queensland, Toowoomba, Queensland, Australia
| | - Chan Y Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Matthew Ku
- Department of Haematology, St Vincent's Hospital Melbourne, Melbourne, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Hua F, Hu Y, He GC, Lai SH, He Y, Zhang S, Deng Y, Han Y, Liu XD, Yang K, Zhong HX, Xiao J, Zheng ZZ, Yi H. Case report: TP53 c.848G>A germline mutation as a possible screening target at initial diagnosis for acute lymphoblastic leukemia. Hematology 2024; 29:2377860. [PMID: 39007733 DOI: 10.1080/16078454.2024.2377860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUD Li-Fraumeni syndrome is a hereditary tumor syndrome characterized by an elevated risk of malignancy, particularly acute lymphoblastic leukemia (ALL), which can be caused by the heterozygous germline mutation. TP53 gene germline mutation is considered a potential risk factor and crucial prognostic parameter for acute leukemia development and diagnosis, but rarely occurs in adults, and its specific pathogenic significance in acute leukemia is unclear. CASE PRESENTATION We describes a case of a 45-year-old woman diagnosed with ALL. Whole-exome sequencing approach identified one of the TP53 germline mutations from her bone marrow sample with possible pathogenic significance, c.848G>A (p.Arg283His) heterozygous missense mutation located on exon 8, which was further verified in her hair, oral mucous and nail samples. Family pedigree screening revealed that the same TP53 genetic variant was present in the patient's father and non-donor son, whereas not in the donor. Digital PCR observed that this point mutation frequency dropped post-transplantation but remained low during maintenance therapy when the patient was leukemia-free. CONCLUSION This suspected Li-Fraumeni syndrome case report with a likely pathogenic heterozygous TP53 variant expands the cancer genetic spectrum. Screening her family members for mutations facilitates identifying the optimal relative donor and avoids unnecessary treatment by monitoring TP53 germline mutations for minimal residual disease following hematopoietic stem cell transplantation. Its potential roles in hematological malignant tumor development and clinical pathogenic implications necessitate further probing.
Collapse
Affiliation(s)
- Fang Hua
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, People's Republic of China
- Department of Hematology, Zigong First People's Hospital, Zigong, People's Republic of China
| | - Yue Hu
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, People's Republic of China
- North Sichuan Medical College Graduate School, Nanchong, People's Republic of China
| | - Guang-Cui He
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, People's Republic of China
| | - Si-Han Lai
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, People's Republic of China
| | - Ying He
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, People's Republic of China
| | - Shan Zhang
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, People's Republic of China
| | - Yan Deng
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, People's Republic of China
| | - Ying Han
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, People's Republic of China
| | - Xiao-Dong Liu
- Department of Hematology, Zigong First People's Hospital, Zigong, People's Republic of China
| | - Kun Yang
- Department of Hematology, Zigong First People's Hospital, Zigong, People's Republic of China
| | - Hui-Xiu Zhong
- Department of Laboratory Medicine, Zigong First People's Hospital, Zigong, People's Republic of China
| | - Jian Xiao
- Department of Hematology, Zigong First People's Hospital, Zigong, People's Republic of China
| | - Zhong-Zheng Zheng
- Shanghai Tissuebank Biotechnology Co., Ltd, Shanghai, People's Republic of China
| | - Hai Yi
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, People's Republic of China
| |
Collapse
|
4
|
Ahmadi SE, Rahimian E, Rahimi S, Zarandi B, Bahraini M, Soleymani M, Safdari SM, Shabannezhad A, Jaafari N, Safa M. From regulation to deregulation of p53 in hematologic malignancies: implications for diagnosis, prognosis and therapy. Biomark Res 2024; 12:137. [PMID: 39538363 PMCID: PMC11565275 DOI: 10.1186/s40364-024-00676-9] [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: 07/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The p53 protein, encoded by the TP53 gene, serves as a critical tumor suppressor, playing a vital role in maintaining genomic stability and regulating cellular responses to stress. Dysregulation of p53 is frequently observed in hematological malignancies, significantly impacting disease progression and patient outcomes. This review aims to examine the regulatory mechanisms of p53, the implications of TP53 mutations in various hematological cancers, and emerging therapeutic strategies targeting p53. We conducted a comprehensive literature review to synthesize recent findings related to p53's multifaceted role in hematologic cancers, focusing on its regulatory pathways and therapeutic potential. TP53 mutations in hematological malignancies often lead to treatment resistance and poor prognosis. Current therapeutic strategies, including p53 reactivation and gene therapy, show promise in improving treatment outcomes. Understanding the intricacies of p53 regulation and the consequences of its mutations is essential for developing effective diagnostic and therapeutic strategies in hematological malignancies, ultimately enhancing patient care and survival.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Rahimian
- Department of Medical Translational Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Bahraini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maral Soleymani
- Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Mehrab Safdari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ashkan Shabannezhad
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Jaafari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
de Haan LM, de Groen RAL, de Groot FA, Noordenbos T, van Wezel T, van Eijk R, Ruano D, Diepstra A, Koens L, Nicolae-Cristea A, Hartog WCED, Terpstra V, Ahsmann E, Dekker TJA, Sijs-Szabo A, Veelken H, Cleven AHG, Jansen PM, Vermaat JSP. Real-world routine diagnostic molecular analysis for TP53 mutational status is recommended over p53 immunohistochemistry in B-cell lymphomas. Virchows Arch 2024; 485:643-654. [PMID: 37851120 PMCID: PMC11522076 DOI: 10.1007/s00428-023-03676-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/19/2023]
Abstract
Previous studies in patients with mature B-cell lymphomas (MBCL) have shown that pathogenic TP53 aberrations are associated with inferior chemotherapeutic efficacy and survival outcomes. In solid malignancies, p53 immunohistochemistry is commonly used as a surrogate marker to assess TP53 mutations, but this correlation is not yet well-established in lymphomas. This study evaluated the accuracy of p53 immunohistochemistry as a surrogate marker for TP53 mutational analysis in a large real-world patient cohort of 354 MBCL patients within routine diagnostic practice. For each case, p53 IHC was assigned to one of three categories: wild type (staining 1-50% of tumor cells with variable nuclear staining), abnormal complete absence or abnormal overexpression (strong and diffuse staining > 50% of tumor cells). Pathogenic variants of TP53 were identified with a targeted next generation sequencing (tNGS) panel. Wild type p53 expression was observed in 267 cases (75.4%), complete absence in twenty cases (5.7%) and the overexpression pattern in 67 cases (18.9%). tNGS identified a pathogenic TP53 mutation in 102 patients (29%). The overall accuracy of p53 IHC was 84.5% (95% CI 80.3-88.1), with a robust specificity of 92.1% (95% CI 88.0- 95.1), but a low sensitivity of 65.7% (95% CI 55.7-74.8). These results suggest that the performance of p53 IHC is insufficient as a surrogate marker for TP53 mutations in our real-world routine diagnostic workup of MBCL patients. By using p53 immunohistochemistry alone, there is a significant risk a TP53 mutation will be missed, resulting in misevaluation of a high-risk patient. Therefore, molecular analysis is recommended in all MBCL patients, especially for further development of risk-directed therapies based on TP53 mutation status.
Collapse
Affiliation(s)
- Lorraine M de Haan
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. box 9600, 2300RC, Leiden, The Netherlands.
| | - Ruben A L de Groen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fleur A de Groot
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Troy Noordenbos
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. box 9600, 2300RC, Leiden, The Netherlands
| | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. box 9600, 2300RC, Leiden, The Netherlands
| | - Ronald van Eijk
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. box 9600, 2300RC, Leiden, The Netherlands
| | - Dina Ruano
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. box 9600, 2300RC, Leiden, The Netherlands
| | - Arjan Diepstra
- Department of Pathology, University Medical Center Groningen, Groningen, The Netherlands
| | - Lianne Koens
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | | | - Valeska Terpstra
- Department of Pathology, Haaglanden Medical Centrum, The Hague, The Netherlands
| | - Els Ahsmann
- Department of Pathology, Groene Hart Ziekenhuis, Gouda, The Netherlands
| | - Tim J A Dekker
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Aniko Sijs-Szabo
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hendrik Veelken
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arjen H G Cleven
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. box 9600, 2300RC, Leiden, The Netherlands
- Department of Pathology, University Medical Center Groningen, Groningen, The Netherlands
| | - Patty M Jansen
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. box 9600, 2300RC, Leiden, The Netherlands
| | - Joost S P Vermaat
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
6
|
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
|
7
|
Eyre TA, Bishton MJ, McCulloch R, O'Reilly M, Sanderson R, Menon G, Iyengar S, Lewis D, Lambert J, Linton KM, McKay P. Diagnosis and management of mantle cell lymphoma: A British Society for Haematology Guideline. Br J Haematol 2024; 204:108-126. [PMID: 37880821 DOI: 10.1111/bjh.19131] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 10/27/2023]
Affiliation(s)
- Toby A Eyre
- Department of Haematology, Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Mark J Bishton
- Department of Haematology, Nottingham University Hospitals NHS Foundation Trust, Oxford, UK
- Translational Medical Sciences, University of Nottingham, Nottingham, UK
| | - Rory McCulloch
- Department of Haematology, Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| | - Maeve O'Reilly
- Department of Haematology, University College London Hospitals, London, UK
| | - Robin Sanderson
- Department of Haematology, King's College Hospital, London, UK
| | - Geetha Menon
- Department of Cellular Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Sunil Iyengar
- Department of Haematology, The Royal Marsden Hospital, London, UK
| | - David Lewis
- Department of Haematology, Derriford Hospital, Plymouth, UK
| | - Jonathan Lambert
- Department of Haematology, University College London Hospitals, London, UK
| | - Kim M Linton
- Department of Haematology, The Christie NHS Foundation Trust, Manchester, UK
| | - Pamela McKay
- Beatson West of Scotland Cancer Centre, Glasgow, UK
| |
Collapse
|
8
|
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
|
9
|
Lew TE, Cliff ERS, Dickinson M, Tam CS, Seymour JF, Blombery P, Bajel A, Ritchie D, Khot A. Allogeneic stem cell transplantation achieves long-term remissions in mantle cell lymphoma, including in TP53-mutated disease. Leuk Lymphoma 2023; 64:1792-1800. [PMID: 37531077 DOI: 10.1080/10428194.2023.2241095] [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: 05/18/2023] [Accepted: 07/13/2023] [Indexed: 08/03/2023]
Abstract
Cytarabine-containing chemoimmunotherapy followed by autologous transplantation and rituximab maintenance achieves durable remissions for most patients with mantle cell lymphoma (MCL). However, patients with TP53-mutated disease have poor outcomes with standard approaches. We previously reported that allogeneic stem cell transplantation (alloSCT) achieved durable remissions in MCL, however follow-up among patients with TP53-mutated disease was limited. Here we report extended follow-up of the overall cohort (n = 36) and TP53-mutated subset (n = 13) (median follow-up 10.8 and 4.2 years, respectively). Estimated overall survival was 56% at 10 years for the overall cohort and 59% at 4 years for the TP53-mutated subset. Among patients with TP53-mutated disease, no relapses occurred beyond 6 months post-transplant. Survival after post-alloSCT disease relapse was poor (median 2.1 years). These data confirm that alloSCT can be curative in MCL, including patients with TP53-mutated disease, and should be considered for earlier utilization in this subgroup for whom conventional chemoimmunotherapy is ineffective.
Collapse
Affiliation(s)
- Thomas E Lew
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Edward R Scheffer Cliff
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Program on Regulation, Therapeutics and Law, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Dickinson
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Constantine S Tam
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
- Alfred Hospital and Monash University, Melbourne, Australia
| | - John F Seymour
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Piers Blombery
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Ashish Bajel
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - David Ritchie
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Amit Khot
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
10
|
Raghani NR, Shah DD, Shah TS, Chorawala MR, Patel RB. Combating relapsed and refractory Mantle cell lymphoma with novel therapeutic armamentarium: Recent advances and clinical prospects. Crit Rev Oncol Hematol 2023; 190:104085. [PMID: 37536448 DOI: 10.1016/j.critrevonc.2023.104085] [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: 02/14/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023] Open
Abstract
Mantle cell lymphoma (MCL) is a rare, aggressive subtype of non-Hodgkin's lymphoma (NHL), accounting for 5% of all cases. Due to its virulence factor, it is an incurable disease and keeps relapsing despite an intensive treatment regimen. Advancements in research and drug discovery have shifted the treatment strategy from conventional chemotherapy to targeted agents and immunotherapies. The establishment of the role of Bruton tyrosine kinase led to the development of ibrutinib, a first-generation BTK inhibitor, and its successors. A conditioning regimen based immunotherapeutic agent like ibritumumob, has also demonstrated a viable response with a favorable toxicity profile. Brexucabtagene Autoleucel, the only approved CAR T-cell therapy, has proven advantageous for relapsed/refractory MCL in both children and adults. This article reviews certain therapies that could help update the current approach and summarizes a few miscellaneous agents, which, seldom studied in trials, could alleviate the regression observed in traditional therapies. DATA AVAILABILITY: The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Neha R Raghani
- Department of Pharmacology and Pharmacy practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Disha D Shah
- Department of Pharmacology and Pharmacy practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Tithi S Shah
- Department of Pharmacology and Pharmacy practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Rakesh B Patel
- Department of Internal Medicine, Division of Hematology and Oncology, UI Carver College of Medicine: The University of Iowa Roy J and Lucille A Carver College of Medicine, 375 Newton Rd, Iowa City, IA 52242, USA.
| |
Collapse
|
11
|
Hill HA, Jain P, Ok CY, Sasaki K, Chen H, Wang ML, Chen K. Integrative Prognostic Machine Learning Models in Mantle Cell Lymphoma. CANCER RESEARCH COMMUNICATIONS 2023; 3:1435-1446. [PMID: 37538987 PMCID: PMC10395375 DOI: 10.1158/2767-9764.crc-23-0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/17/2023] [Accepted: 06/27/2023] [Indexed: 08/05/2023]
Abstract
Patients with mantle cell lymphoma (MCL), an incurable B-cell malignancy, benefit from accurate pretreatment disease stratification. We curated an extensive database of 862 patients diagnosed between 2014 and 2022. A machine learning (ML) gradient-boosted model incorporated baseline features from clinicopathologic, cytogenetic, and genomic data with high predictive power discriminating between patients with indolent or responsive MCL and those with aggressive disease (AUC ROC = 0.83). In addition, we utilized the gradient-boosted framework as a robust feature selection method for multivariate logistic and survival modeling. The best ML models incorporated features from clinical and genomic data types highlighting the need for correlative molecular studies in precision oncology. As proof of concept, we launched our most accurate and practical models using an application interface, which has potential for clinical implementation. We designated the 20-feature ML model-based index the "integrative MIPI" or iMIPI and a similar 10-feature ML index the "integrative simplified MIPI" or iMIPI-s. The top 10 baseline prognostic features represented in the iMIPI-s are: lactase dehydrogenase (LDH), Ki-67%, platelet count, bone marrow involvement percentage, hemoglobin levels, the total number of observed somatic mutations, TP53 mutational status, Eastern Cooperative Oncology Group performance level, beta-2 microglobulin, and morphology. Our findings emphasize that prognostic applications and indices should include molecular features, especially TP53 mutational status. This work demonstrates the clinical utility of complex ML models and provides further evidence for existing prognostic markers in MCL. Significance Our model is the first to integrate a dynamic algorithm with multiple clinical and molecular features, allowing for accurate predictions of MCL disease outcomes in a large patient cohort.
Collapse
Affiliation(s)
- Holly A. Hill
- Department of Bioinformatics and Computational Biology, Division of Quantitative Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas
| | - Preetesh Jain
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chi Young Ok
- Department of Hematopathology, Division of Pathology-Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Koji Sasaki
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Han Chen
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Michael L. Wang
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, Division of Quantitative Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Wilson MR, Barrett A, Cheah CY, Eyre TA. How I manage mantle cell lymphoma: indolent versus aggressive disease. Br J Haematol 2023; 201:185-198. [PMID: 36807902 DOI: 10.1111/bjh.18697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/23/2023]
Abstract
Mantle cell lymphoma (MCL) is a mature B-cell lymphoma with a variable clinical course and historically poor prognosis. Management is challenging in part due to the heterogeneity of the disease course, with indolent and aggressive subtypes now well recognised. Indolent MCL is often characterised by a leukaemic presentation, SOX11 negativity and low proliferation index (Ki-67). Aggressive MCL is characterised by rapid onset widespread lymphadenopathy, extra-nodal involvement, blastoid or pleomorphic histology and high Ki-67. Tumour protein p53 (TP53) aberrations in aggressive MCL are recognised with clear negative impact on survival. Until recently, trials have not addressed these specific subtypes separately. With the increasing availability of targeted novel agents and cellular therapies, the treatment landscape is constantly evolving. In this review, we describe the clinical presentation, biological factors, and specific management considerations of both indolent and aggressive MCL and discuss current and potential future evidence which may help move to a more personalised approach.
Collapse
Affiliation(s)
| | - Aisling Barrett
- Haematology and Cancer Centre, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, UK
| | - Chan Yoon Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,Division of Internal Medicine, University of Western Australia, Perth, Australia
| | - Toby A Eyre
- Haematology and Cancer Centre, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, UK
| |
Collapse
|
15
|
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
|
16
|
Nolan J, Murphy C, Dinneen K, Lee G, Higgins E, Bacon L, O'Brien D, Flavin R, Quinn F, Vandenberghe E. p53 immunohistochemistry must be confirmed by TP53 next generation sequencing for accurate risk stratification of patients with mantle cell lymphoma. Leuk Lymphoma 2022; 63:3504-3507. [PMID: 36059262 DOI: 10.1080/10428194.2022.2118529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- James Nolan
- Department of Haematology, St James' Hospital, Dublin 8, Ireland.,Department of Haematology, Trinity College, Dublin 2, Ireland
| | - Catherine Murphy
- Department of Haematology, St James' Hospital, Dublin 8, Ireland
| | - Kate Dinneen
- Department of Haematology, St James' Hospital, Dublin 8, Ireland
| | - Greg Lee
- Department of Haematology, St James' Hospital, Dublin 8, Ireland
| | | | - Larry Bacon
- Department of Haematology, St James' Hospital, Dublin 8, Ireland
| | - David O'Brien
- Department of Haematology, St James' Hospital, Dublin 8, Ireland
| | - Richard Flavin
- Department of Histopathology, St James' Hospital, Dublin 8, Ireland.,Department of Histopathology, Trinity College, Dublin 2, Ireland
| | - Fiona Quinn
- Cancer Molecular Diagnostics Laboratory, St James' Hospital, Dublin 8, Ireland
| | - Elisabeth Vandenberghe
- Department of Haematology, St James' Hospital, Dublin 8, Ireland.,Department of Haematology, Trinity College, Dublin 2, Ireland
| |
Collapse
|
17
|
de Leval L, Alizadeh AA, Bergsagel PL, Campo E, Davies A, Dogan A, Fitzgibbon J, Horwitz SM, Melnick AM, Morice WG, Morin RD, Nadel B, Pileri SA, Rosenquist R, Rossi D, Salaverria I, Steidl C, Treon SP, Zelenetz AD, Advani RH, Allen CE, Ansell SM, Chan WC, Cook JR, Cook LB, d’Amore F, Dirnhofer S, Dreyling M, Dunleavy K, Feldman AL, Fend F, Gaulard P, Ghia P, Gribben JG, Hermine O, Hodson DJ, Hsi ED, Inghirami G, Jaffe ES, Karube K, Kataoka K, Klapper W, Kim WS, King RL, Ko YH, LaCasce AS, Lenz G, Martin-Subero JI, Piris MA, Pittaluga S, Pasqualucci L, Quintanilla-Martinez L, Rodig SJ, Rosenwald A, Salles GA, San-Miguel J, Savage KJ, Sehn LH, Semenzato G, Staudt LM, Swerdlow SH, Tam CS, Trotman J, Vose JM, Weigert O, Wilson WH, Winter JN, Wu CJ, Zinzani PL, Zucca E, Bagg A, Scott DW. Genomic profiling for clinical decision making in lymphoid neoplasms. Blood 2022; 140:2193-2227. [PMID: 36001803 PMCID: PMC9837456 DOI: 10.1182/blood.2022015854] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/15/2022] [Indexed: 01/28/2023] Open
Abstract
With the introduction of large-scale molecular profiling methods and high-throughput sequencing technologies, the genomic features of most lymphoid neoplasms have been characterized at an unprecedented scale. Although the principles for the classification and diagnosis of these disorders, founded on a multidimensional definition of disease entities, have been consolidated over the past 25 years, novel genomic data have markedly enhanced our understanding of lymphomagenesis and enriched the description of disease entities at the molecular level. Yet, the current diagnosis of lymphoid tumors is largely based on morphological assessment and immunophenotyping, with only few entities being defined by genomic criteria. This paper, which accompanies the International Consensus Classification of mature lymphoid neoplasms, will address how established assays and newly developed technologies for molecular testing already complement clinical diagnoses and provide a novel lens on disease classification. More specifically, their contributions to diagnosis refinement, risk stratification, and therapy prediction will be considered for the main categories of lymphoid neoplasms. The potential of whole-genome sequencing, circulating tumor DNA analyses, single-cell analyses, and epigenetic profiling will be discussed because these will likely become important future tools for implementing precision medicine approaches in clinical decision making for patients with lymphoid malignancies.
Collapse
Affiliation(s)
- Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Ash A. Alizadeh
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University, Stanford, CA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA
| | - P. Leif Bergsagel
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Phoenix, AZ
| | - Elias Campo
- Haematopathology Section, Hospital Clínic, Institut d'Investigaciones Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Andrew Davies
- Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
| | - Ahmet Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jude Fitzgibbon
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Steven M. Horwitz
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ari M. Melnick
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - William G. Morice
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Ryan D. Morin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
- BC Cancer Centre for Lymphoid Cancer, Vancouver, BC, Canada
| | - Bertrand Nadel
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Stefano A. Pileri
- Haematopathology Division, IRCCS, Istituto Europeo di Oncologia, IEO, Milan, Italy
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Solna, Sweden
| | - Davide Rossi
- Institute of Oncology Research and Oncology Institute of Southern Switzerland, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Itziar Salaverria
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Christian Steidl
- Centre for Lymphoid Cancer, BC Cancer and University of British Columbia, Vancouver, Canada
| | | | - Andrew D. Zelenetz
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Ranjana H. Advani
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
| | - Carl E. Allen
- Division of Pediatric Hematology-Oncology, Baylor College of Medicine, Houston, TX
| | | | - Wing C. Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| | - James R. Cook
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Lucy B. Cook
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Francesco d’Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Stefan Dirnhofer
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Kieron Dunleavy
- Division of Hematology and Oncology, Georgetown Lombardi Comprehensive Cancer Centre, Georgetown University Hospital, Washington, DC
| | - Andrew L. Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Falko Fend
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Philippe Gaulard
- Department of Pathology, University Hospital Henri Mondor, AP-HP, Créteil, France
- Faculty of Medicine, IMRB, INSERM U955, University of Paris-Est Créteil, Créteil, France
| | - Paolo Ghia
- Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy
| | - John G. Gribben
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Olivier Hermine
- Service D’hématologie, Hôpital Universitaire Necker, Université René Descartes, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Daniel J. Hodson
- Wellcome MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Eric D. Hsi
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Elaine S. Jaffe
- Hematopathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kennosuke Karube
- Department of Pathology and Laboratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keisuke Kataoka
- Division of Molecular Oncology, National Cancer Center Research Institute, Toyko, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Wolfram Klapper
- Hematopathology Section and Lymph Node Registry, Department of Pathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Won Seog Kim
- Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Rebecca L. King
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Young H. Ko
- Department of Pathology, Cheju Halla General Hospital, Jeju, Korea
| | | | - Georg Lenz
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - José I. Martin-Subero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Miguel A. Piris
- Department of Pathology, Jiménez Díaz Foundation University Hospital, CIBERONC, Madrid, Spain
| | - Stefania Pittaluga
- Hematopathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Columbia University, New York, NY
- Department of Pathology & Cell Biology, Columbia University, New York, NY
- The Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Scott J. Rodig
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | | | - Gilles A. Salles
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jesus San-Miguel
- Clínica Universidad de Navarra, Navarra, Cancer Center of University of Navarra, Cima Universidad de NavarraI, Instituto de Investigacion Sanitaria de Navarra, Centro de Investigación Biomédica en Red de Céncer, Pamplona, Spain
| | - Kerry J. Savage
- Centre for Lymphoid Cancer, BC Cancer and University of British Columbia, Vancouver, Canada
| | - Laurie H. Sehn
- Centre for Lymphoid Cancer, BC Cancer and University of British Columbia, Vancouver, Canada
| | - Gianpietro Semenzato
- Department of Medicine, University of Padua and Veneto Institute of Molecular Medicine, Padova, Italy
| | - Louis M. Staudt
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Steven H. Swerdlow
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Judith Trotman
- Haematology Department, Concord Repatriation General Hospital, Sydney, Australia
| | - Julie M. Vose
- Department of Internal Medicine, Division of Hematology-Oncology, University of Nebraska Medical Center, Omaha, NE
| | - Oliver Weigert
- Department of Medicine III, LMU Hospital, Munich, Germany
| | - Wyndham H. Wilson
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jane N. Winter
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Pier L. Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna Istitudo di Ematologia “Seràgnoli” and Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale Università di Bologna, Bologna, Italy
| | - Emanuele Zucca
- Institute of Oncology Research and Oncology Institute of Southern Switzerland, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David W. Scott
- Centre for Lymphoid Cancer, BC Cancer and University of British Columbia, Vancouver, Canada
| |
Collapse
|
18
|
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
|
19
|
Tam CS, Gregory GP, Ku M, Fleming S, Handunnetti SM, Lee D, Walker P, Perkins A, Lew TE, Sirdesai S, Chua CC, Gilbertson M, Lasica M, Anderson MA, Renwick W, Grigg A, Patil S, Opat S, Friebe A, Cooke R, De Boer J, Spencer A, Ritchie D, Agarwal R, Blombery P. Recommendation for TP53 mutation testing in newly diagnosed mantle cell lymphoma: a statement from working groups sponsored by the Victorian Comprehensive Cancer Centre. Intern Med J 2022; 52:1286-1287. [PMID: 35879233 DOI: 10.1111/imj.15851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Constantine S Tam
- Alfred Hospital, Melbourne, Victoria, Australia.,Monash University, Melbourne, Victoria, Australia.,VCCC Alliance, Melbourne, Victoria, Australia
| | - Gareth P Gregory
- Monash University, Melbourne, Victoria, Australia.,Monash Haematology, Monash Health, Melbourne, Victoria, Australia
| | - Matthew Ku
- Department of Haematology, St Vincent's Hospital, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia
| | - Shaun Fleming
- Alfred Hospital, Melbourne, Victoria, Australia.,Monash University, Melbourne, Victoria, Australia
| | - Sasanka M Handunnetti
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Denise Lee
- Department of Haematology, Box Hill Hospital, Melbourne, Victoria, Australia
| | - Patricia Walker
- Alfred Hospital, Melbourne, Victoria, Australia.,Department of Clinical Haematology, Peninsula Health, Melbourne, Victoria, Australia.,Peninsula Private Hospital, Melbourne, Victoria, Australia
| | - Andrew Perkins
- Alfred Hospital, Melbourne, Victoria, Australia.,Monash University, Melbourne, Victoria, Australia
| | - Thomas E Lew
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Shreerang Sirdesai
- Department of Haematology, University Hospital Geelong, Geelong, Victoria, Australia
| | - Chong Chyn Chua
- Alfred Hospital, Melbourne, Victoria, Australia.,Department of Clinical Haematology, Northern Health, Melbourne, Victoria, Australia
| | - Michael Gilbertson
- Monash University, Melbourne, Victoria, Australia.,Monash Haematology, Monash Health, Melbourne, Victoria, Australia.,Department of Haematology and Oncology, Western Health, Melbourne, Victoria, Australia
| | - Masa Lasica
- Department of Haematology, St Vincent's Hospital, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia
| | - Mary Ann Anderson
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - William Renwick
- University of Melbourne, Melbourne, Victoria, Australia.,Department of Haematology and Oncology, Western Health, Melbourne, Victoria, Australia
| | - Andrew Grigg
- Department of Clinical Haematology, Austin Hospital, Melbourne, Victoria, Australia
| | - Sush Patil
- Alfred Hospital, Melbourne, Victoria, Australia.,Monash University, Melbourne, Victoria, Australia
| | - Stephen Opat
- Monash University, Melbourne, Victoria, Australia.,Monash Haematology, Monash Health, Melbourne, Victoria, Australia
| | - Adam Friebe
- Department of Haematology, University Hospital Geelong, Geelong, Victoria, Australia
| | - Rachel Cooke
- Department of Clinical Haematology, Northern Health, Melbourne, Victoria, Australia
| | | | - Andrew Spencer
- Alfred Hospital, Melbourne, Victoria, Australia.,Monash University, Melbourne, Victoria, Australia
| | - David Ritchie
- University of Melbourne, Melbourne, Victoria, Australia.,Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Rishu Agarwal
- Department of Clinical Haematology, Austin Hospital, Melbourne, Victoria, Australia
| | - Piers Blombery
- University of Melbourne, Melbourne, Victoria, Australia.,Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Burkart M, Karmali R. Relapsed/Refractory Mantle Cell Lymphoma: Beyond BTK Inhibitors. J Pers Med 2022; 12:376. [PMID: 35330376 PMCID: PMC8954159 DOI: 10.3390/jpm12030376] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 01/27/2023] Open
Abstract
Mantle cell lymphoma (MCL) is a rare mature B-cell non-Hodgkin lymphoma (B-NHL) with historically poor outcomes. Virtually all patients will eventually experience refractory or relapsed (R/R) disease, with a virulent course of resistance and serial relapses, making treatment challenging. The available therapies for R/R MCL are not curative with conventional therapy, their goal being to palliate and prolong survival. A variety of agents approved for R/R MCL, including Bruton's tyrosine kinase inhibitors (BTKi), changed the treatment landscape of R/R MCL. In the pre-BTKi era, the median progression-free survival (PFS) in R/R disease was 4-9 months. With the introduction of ibrutinib, the median PFS improved to 13-14.6 months. Despite these impressive results, the duration of response is limited, and resistance to BTKi inevitably develops in a subset of patients. Outcomes after progression on BTKi are extremely poor, with a median overall survival (OS) of 6 to 10 months. Certain therapies, such as chimeric antigen receptor (CAR) T cells, have shown promising results after BTKi failure. The preferred combination and sequencing of therapies beyond BTKi remain unestablished and are currently being investigated. In this review, we describe the current evidence for the available treatment of R/R MCL after progression on BTKi.
Collapse
Affiliation(s)
- Madelyn Burkart
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA;
| | - Reem Karmali
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA;
| |
Collapse
|
21
|
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: 49] [Impact Index Per Article: 16.3] [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
|
22
|
Yi S, Yan Y, Jin M, Bhattacharya S, Wang Y, Wu Y, Yang L, Gine E, Clot G, Chen L, Yu Y, Zou D, Wang J, Phan AT, Cui R, Li F, Sun Q, Zhai Q, Wang T, Yu Z, Liu L, Liu W, Lyv R, Sui W, Huang W, Xiong W, Wang H, Li C, Xiao Z, Hao M, Wang J, Cheng T, Bea S, Herrera AF, Danilov A, Campo E, Ngo VN, Qiu L, Wang L. Genomic and transcriptomic profiling reveals distinct molecular subsets associated with outcomes in mantle cell lymphoma. J Clin Invest 2022; 132:e153283. [PMID: 34882582 PMCID: PMC8803323 DOI: 10.1172/jci153283] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/02/2021] [Indexed: 11/17/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a phenotypically and genetically heterogeneous malignancy in which the genetic alterations determining clinical indications are not fully understood. Here, we performed a comprehensive whole-exome sequencing analysis of 152 primary samples derived from 134 MCL patients, including longitudinal samples from 16 patients and matched RNA-Seq data from 48 samples. We classified MCL into 4 robust clusters (C1-C4). C1 featured mutated immunoglobulin heavy variable (IGHV), CCND1 mutation, amp(11q13), and active B cell receptor (BCR) signaling. C2 was enriched with del(11q)/ATM mutations and upregulation of NF-κB and DNA repair pathways. C3 was characterized by mutations in SP140, NOTCH1, and NSD2, with downregulation of BCR signaling and MYC targets. C4 harbored del(17p)/TP53 mutations, del(13q), and del(9p), and active MYC pathway and hyperproliferation signatures. Patients in these 4 clusters had distinct outcomes (5-year overall survival [OS] rates for C1-C4 were 100%, 56.7%, 48.7%, and 14.2%, respectively). We also inferred the temporal order of genetic events and studied clonal evolution of 16 patients before treatment and at progression/relapse. Eleven of these samples showed drastic clonal evolution that was associated with inferior survival, while the other samples showed modest or no evolution. Our study thus identifies genetic subsets that clinically define this malignancy and delineates clonal evolution patterns and their impact on clinical outcomes.
Collapse
Affiliation(s)
- Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yuting Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Monrovia, California, USA
| | - Meiling Jin
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Monrovia, California, USA
| | - Supriyo Bhattacharya
- Division of Translational Bioinformatics, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Irwindale, California, USA
| | - Yi Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yiming Wu
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Monrovia, California, USA
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Monrovia, California, USA
| | - Eva Gine
- Lymphoid Neoplasm Program, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hematology Department, Hospital Clínic, Departament d’Anatomia Patològica, Universitat de Barcelona, Barcelona, Spain
| | - Guillem Clot
- Lymphoid Neoplasm Program, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hematology Department, Hospital Clínic, Departament d’Anatomia Patològica, Universitat de Barcelona, Barcelona, Spain
| | - Lu Chen
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Ying Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - An T. Phan
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Monrovia, California, USA
| | - Rui Cui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Department of Hematology, Tianjin First Center Hospital, Tianjin, China
| | - Fei Li
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Qi Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qiongli Zhai
- Department of Pathology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Tingyu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zhen Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lanting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Rui Lyv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenyang Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenjie Xiong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Huijun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Chengwen Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zhijian Xiao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Silvia Bea
- Lymphoid Neoplasm Program, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hematology Department, Hospital Clínic, Departament d’Anatomia Patològica, Universitat de Barcelona, Barcelona, Spain
| | - Alex F. Herrera
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Alexey Danilov
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Elias Campo
- Lymphoid Neoplasm Program, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hematology Department, Hospital Clínic, Departament d’Anatomia Patològica, Universitat de Barcelona, Barcelona, Spain
| | - Vu N. Ngo
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Monrovia, California, USA
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Lili Wang
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Monrovia, California, USA
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| |
Collapse
|
23
|
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
|
24
|
Chiappella A, Diop F, Agostinelli C, Novo M, Nassi L, Evangelista A, Ciccone G, Di Rocco A, Martelli M, Melle F, Moia R, Motta G, Righi S, Santambrogio E, Tucci A, Balzarotti M, Ladetto M, Pileri SA, Gaidano G, Vitolo U. Prognostic impact of
TP53
mutation in newly diagnosed diffuse large B‐cell lymphoma patients treated in the FIL‐DLCL04 trial. Br J Haematol 2021; 196:1184-1193. [DOI: 10.1111/bjh.17971] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Annalisa Chiappella
- Hematology Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di Torino TorinoItaly
| | - Fary Diop
- Division of Hematology Department of Translational Medicine Università del Piemonte Orientale NovaraItaly
| | | | - Mattia Novo
- Multidisciplinary Oncology Outpatient Clinic Candiolo Cancer Institute FPO‐IRCCS TorinoItaly
| | - Luca Nassi
- Division of Hematology Department of Translational Medicine Università del Piemonte Orientale NovaraItaly
| | - Andrea Evangelista
- Unit of Clinical Epidemiology and CPO Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di Torino TorinoItaly
| | - Giovannino Ciccone
- Unit of Clinical Epidemiology and CPO Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di Torino TorinoItaly
| | - Alice Di Rocco
- Department of Traslational and Precision Medicine Università La Sapienza RomaItaly
| | - Maurizio Martelli
- Department of Traslational and Precision Medicine Università La Sapienza RomaItaly
| | - Federica Melle
- Haematopathology Division, IRCCS Istituto Europeo di Oncologia IEO MilanoItaly
| | - Riccardo Moia
- Division of Hematology Department of Translational Medicine Università del Piemonte Orientale NovaraItaly
| | - Giovanna Motta
- Haematopathology Division, IRCCS Istituto Europeo di Oncologia IEO MilanoItaly
| | - Simona Righi
- Pathology Unit Università degli Studi di Bologna BolognaItaly
| | | | | | - Monica Balzarotti
- Unit of Hematology Humanitas Clinical and Research Center RozzanoItaly
| | - Marco Ladetto
- Hematology Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo Alessandria Italy
| | - Stefano A. Pileri
- Haematopathology Division, IRCCS Istituto Europeo di Oncologia IEO MilanoItaly
| | - Gianluca Gaidano
- Division of Hematology Department of Translational Medicine Università del Piemonte Orientale NovaraItaly
| | - Umberto Vitolo
- Multidisciplinary Oncology Outpatient Clinic Candiolo Cancer Institute FPO‐IRCCS TorinoItaly
| | | |
Collapse
|
25
|
Lew TE, Cliff ERS, Dickinson M, Tam CS, Seymour JF, Blombery P, Bajel A, Ritchie D, Khot A. T-cell replete allogeneic stem cell transplant for mantle cell lymphoma achieves durable disease control, including against TP53-mutated disease. Bone Marrow Transplant 2021; 56:2857-2859. [PMID: 34285380 DOI: 10.1038/s41409-021-01418-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Thomas E Lew
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia. .,Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
| | - Edward R S Cliff
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Michael Dickinson
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Constantine S Tam
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - John F Seymour
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Piers Blombery
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia.,Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Ashish Bajel
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - David Ritchie
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Amit Khot
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
26
|
Jing C, Zheng Y, Feng Y, Cao X, Xu C. Prognostic significance of p53, Sox11, and Pax5 co-expression in mantle cell lymphoma. Sci Rep 2021; 11:11896. [PMID: 34099776 PMCID: PMC8185106 DOI: 10.1038/s41598-021-91433-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 05/24/2021] [Indexed: 02/05/2023] Open
Abstract
Mantle cell lymphoma (MCL) is a relatively rare subtype of non-Hodgkin’s lymphoma. To identify molecular biomarkers in MCL, we performed immunohistochemistry tissue arrays using biopsies from 64 MCL patients diagnosed in West China Hospital from 2012 to 2016. TP53 mutation status in those patients was also examined by sequencing. The sequencing results showed TP53 mutations were highly heterogeneous in MCL. We identified four novel TP53 mutations in MCL: P151R, G199R, V218E, and G325R. The MCL patients with TP53 mutations had inferior progression-free survival (PFS, p = 0.002) and overall survival (OS, p = 0.011). Tissue array results showed the expression of p53, Sox11, or Pax5 alone did not correlate with the patient PFS and OS. However, the MCL patients with triple-positive expression of p53/Sox11/Pax5 had inferior PFS (p = 0.008) and OS (p = 0.002). Such risk stratification was independent to the mantle cell lymphoma international prognostic index (MIPI), Ki-67 value, and TP53 mutation status of the patients. The triple-positive patients might represent a subtype of high-risk MCL. Our findings might indicate a novel way to stratify MCL and predict patients’ prognosis.
Collapse
Affiliation(s)
- Caixia Jing
- Department of Hematology/Hematology Research Laboratory, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China.,Department of Hematology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhuan Zheng
- Department of Hematology/Hematology Research Laboratory, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China.,State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Feng
- Department of Hematology/Hematology Research Laboratory, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China
| | - Xia Cao
- Department of Hematology/Hematology Research Laboratory, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China
| | - Caigang Xu
- Department of Hematology/Hematology Research Laboratory, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China.
| |
Collapse
|
27
|
Genomic profiles and clinical outcomes of de novo blastoid/pleomorphic MCL are distinct from those of transformed MCL. Blood Adv 2021; 4:1038-1050. [PMID: 32191807 DOI: 10.1182/bloodadvances.2019001396] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 01/27/2020] [Indexed: 01/03/2023] Open
Abstract
Blastoid and pleomorphic mantle cell lymphomas (MCLs) are variants of aggressive histology MCL (AH-MCL). AH-MCL can arise de novo (AH-DN) or transform from prior classic variant MCL (AH-t). This study is the first integrated analysis of clinical and genomic characteristics of AH-MCL. Patient characteristics were collected from diagnosis (AH-DN) and at transformation (AH-t). Survival after initial diagnosis (AH-DN) and after transformation (AH-t) was calculated. Regression tree analysis was performed to evaluate prognostic variables and in univariate and multivariate analyses for survival. Whole-exome sequencing was performed in evaluable biopsy specimens. We identified 183 patients with AH-MCL (108 were AH-DN, and 75 were AH-t; 152 were blastoid, and 31 were pleomorphic). Median survival was 33 months (48 and 14 months for AH-DN and AH-t, respectively; P = .001). Factors associated with inferior survival were age (≥72 years), AH-t category, Ki-67 ≥50% and poor performance status. AH-t had a significantly higher degree of aneuploidy compared with AH-DN. Transformed MCL patients exhibited KMT2B mutations. AH-MCL patients with Ki-67 ≥50% had exclusive mutations in CCND1, NOTCH1, TP53, SPEN, SMARCA4, RANBP2, KMT2C, NOTCH2, NOTCH3, and NSD2 compared with low Ki-67 (<50%). AH-t patients have poor outcomes and distinct genomic profile. This is the first study to report that AH-MCL patients with high Ki-67 (≥50%) exhibit a distinct mutation profile and very poor survival.
Collapse
|
28
|
Chen L, Mu W, Gu J, Xiao M, Huang L, Zheng M, Li C, Xiao Y, Zhou J, Long X. TP53-Mutated Circulating Tumor DNA for Disease Monitoring in Lymphoma Patients after CAR T Cell Therapy. DIAGNOSTICS (BASEL, SWITZERLAND) 2021; 11:diagnostics11050844. [PMID: 34066756 PMCID: PMC8151854 DOI: 10.3390/diagnostics11050844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/03/2022]
Abstract
Chimeric antigen receptor T (CAR T) cell immunotherapy has shown remarkable efficacy in non-Hodgkin’s lymphoma (NHL) patients. Minimal residual disease (MRD) monitoring in NHL is essential after CAR T cell therapy, which can be achieved by monitoring circulating tumor DNA (ctDNA). The mutation of TP53 in NHL has been suggested to be associated with a poor prognosis. However, whether TP53-mutated ctDNA can be used as a biomarker remains undetermined. In this study, a total of 40 patients with mutated TP53 who received CAR T cell treatment were analyzed, and specific probes targeting 29 different TP53 mutation sites in the 40 patients were designed and verified. Then, the presence of TP53-mutated ctDNA in longitudinal plasma samples was tracked by droplet digital PCR. Patients were stratified into two groups, favorable or unfavorable, based on their highest ctDNA level using a MAF cutoff of 3.15% according to the ROC curve. The unfavorable group had significantly worse PFS than the favorable group (p < 0.001). Our results suggest that patients with mutated TP53 with a favorable ctDNA profile in the first trimester have better prognostic outcomes than patients with an unfavorable profile, and ctDNA can be a reliable predictor of the subsequent clinical outcome.
Collapse
|
29
|
Khanlari M, Wang SA, Fowler NH, Tang G, Saluja K, Muzzafar T, Medeiros LJ, Thakral B. Concurrent TP53 Mutation and Deletion in Refractory Low-grade Follicular Lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:e626-e629. [PMID: 33867306 DOI: 10.1016/j.clml.2021.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/08/2021] [Accepted: 03/14/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Mahsa Khanlari
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX
| | - Sa A Wang
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX
| | - Nathan H Fowler
- Department of Lymphoma and Myeloma Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX
| | - Guilin Tang
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX
| | - Karan Saluja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, TX
| | - Tariq Muzzafar
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX
| | | | - Beenu Thakral
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
30
|
Assis-Mendonça GR, Fattori A, Rocha RM, Lourenço GJ, Delamain MT, Nonogaki S, de Lima VCC, Colleoni GWB, de Souza CA, Soares FA, Lima CSP, Vassallo J. Single nucleotide variants in immune-response genes and the tumor microenvironment composition predict progression of mantle cell lymphoma. BMC Cancer 2021; 21:209. [PMID: 33648463 PMCID: PMC7919095 DOI: 10.1186/s12885-021-07891-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 02/08/2021] [Indexed: 11/20/2022] Open
Abstract
Background There is evidence to consider that the tumor microenvironment (TME) composition associates with antitumor immune response, and may predict the outcome of various non-Hodgkin lymphoma subtypes. However, in the case of mantle cell lymphoma (MCL), a rare and aggressive disease, there is lacking a detailed study of the TME components, as well as an integrative approach among them in patients’ samples. Also, from the genetic point of view, it is known that single nucleotide variants (SNVs) in immune-response genes are among important regulators of immunity. At present, it is uncertain whether SNVs in candidate immune-response genes and the TME composition are able to alter the prognosis in MCL. Methods We assessed a detailed TME composition in 88 MCL biopsies using immunohistochemistry, which was automatically analyzed by pixel counting (Aperio system). We also genotyped SNVs located in candidate immune-response genes (IL12A, IL2, IL10, TGFB1, TGFBR1, TGFBR2, IL17A, IL17F) in 95 MCL patients. We tested whether the SNVs could modulate the respective protein expression and TME composition in the tumor compartment. Finally, we proposed survival models in rituximab-treated patients, considering immunohistochemical and SNV models. Results High FOXP3/CD3 ratios (p = 0.001), high IL17A levels (p = 0.003) and low IL2 levels (p = 0.03) were individual immunohistochemical predictors of poorer survival. A principal component, comprising high quantities of macrophages and high Ki-67 index, also worsened outcome (p = 0.02). In the SNV model, the CC haplotype of IL10 (p < 0.01), the GG genotype of IL2 rs2069762 (p = 0.02) and the AA+AG genotypes of TGFBR2 rs3087465 (p < 0.01) were independent predictors of outcome. Finally, the GG genotype of TGFB1 rs6957 associated with lower tumor TGFβ levels (p = 0.03) and less CD163+ macrophages (p = 0.01), but did not modulate patients’ survival. Conclusions Our results indicate that the TME composition has relevant biological roles in MCL. In this setting, immunohistochemical detection of T-reg cells, IL17A and IL2, coupled with SNV genotyping in IL10, TGFBR2 and IL2, may represent novel prognostic factors in this disease, following future validations. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07891-9.
Collapse
Affiliation(s)
- Guilherme Rossi Assis-Mendonça
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Distrito de Barão Geraldo, Campinas, SP, Brazil.
| | - André Fattori
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Rafael Malagoli Rocha
- Molecular Gynecology Laboratory, Department of Gynecology, Federal University of São Paulo, São Paulo, Brazil
| | - Gustavo Jacob Lourenço
- Laboratory of Cancer Genetics, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | | | - Suely Nonogaki
- Instituto Adolfo Lutz, Secretaria de Estado da Saúde, São Paulo, SP, Brazil
| | | | | | - Cármino Antonio de Souza
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | | | - Carmen Silvia Passos Lima
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil.,Laboratory of Cancer Genetics, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - José Vassallo
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Distrito de Barão Geraldo, Campinas, SP, Brazil.,Rede D'Or Hospitals Network - Pathology Division, São Paulo, SP, Brazil.,Laboratory of Investigative and Molecular Pathology (LIP), CIPED, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
Macon WR. Computational histopathology and deep transfer learning: characterizing the molecular basis of tumor morphology. J Hematop 2020. [DOI: 10.1007/s12308-020-00425-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
34
|
Buege MJ, Kumar A, Dixon BN, Tang LA, Pak T, Orozco J, Peterson TJ, Maples KT. Management of Mantle Cell Lymphoma in the Era of Novel Oral Agents. Ann Pharmacother 2020; 54:879-898. [PMID: 32079411 PMCID: PMC8330616 DOI: 10.1177/1060028020909117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Objectives: To discuss (1) recent and emerging data for pharmacological management of untreated and relapsed/refractory (R/R) mantle cell lymphoma (MCL) with agents approved in the United States, (2) important considerations for toxicity monitoring and management, and (3) preliminary data and ongoing studies for agents in MCL-specific clinical trials. Data Sources: PubMed/MEDLINE, EMBASE, Google Scholar, product labeling, National Comprehensive Cancer Network, American Cancer Society, and ClinicalTrials.gov were searched for studies published between January 1, 2017, and January 31, 2020, and key historical trials. Study Selection and Data Extraction: Relevant studies conducted in humans and selected supporting preclinical data were reviewed. Data Synthesis: MCL is a rare but usually aggressive non-Hodgkin lymphoma that most commonly affects the older population. Traditionally, the treatment of MCL has been determined based on transplant eligibility. Newer data suggest that more tolerable frontline therapy may produce outcomes similar to intensive historical induction regimens, possibly precluding fewer patients from autologous stem cell transplant and producing better long-term outcomes in transplant-ineligible patients. In the R/R setting, novel regimens are improving outcomes and changing the landscape of treatment. Relevance to Patient Care and Clinical Practice: This review summarizes and discusses recent and emerging data for management of newly diagnosed and R/R MCL; key supportive care considerations for agents are also discussed. Conclusions: Recent study results are changing management of MCL. Although these data have complicated the picture of regimen selection, increasingly effective and tolerable therapy and additional anticipated data point to a brighter future for patients with MCL.
Collapse
Affiliation(s)
| | - Anita Kumar
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Laura A Tang
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Terry Pak
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Tim J Peterson
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
35
|
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
|
36
|
Pararajalingam P, Coyle KM, Arthur SE, Thomas N, Alcaide M, Meissner B, Boyle M, Qureshi Q, Grande BM, Rushton C, Slack GW, Mungall AJ, Tam CS, Agarwal R, Dawson SJ, Lenz G, Balasubramanian S, Gascoyne RD, Steidl C, Connors J, Villa D, Audas TE, Marra MA, Johnson NA, Scott DW, Morin RD. Coding and noncoding drivers of mantle cell lymphoma identified through exome and genome sequencing. Blood 2020; 136:572-584. [PMID: 32160292 PMCID: PMC7440974 DOI: 10.1182/blood.2019002385] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 02/20/2020] [Indexed: 12/11/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an uncommon B-cell non-Hodgkin lymphoma (NHL) that is incurable with standard therapies. The genetic drivers of this cancer have not been firmly established, and the features that contribute to differences in clinical course remain limited. To extend our understanding of the biological pathways involved in this malignancy, we performed a large-scale genomic analysis of MCL using data from 51 exomes and 34 genomes alongside previously published exome cohorts. To confirm our findings, we resequenced the genes identified in the exome cohort in 191 MCL tumors, each having clinical follow-up data. We confirmed the prognostic association of TP53 and NOTCH1 mutations. Our sequencing revealed novel recurrent noncoding mutations surrounding a single exon of the HNRNPH1gene. In RNA-seq data from 103 of these cases, MCL tumors with these mutations had a distinct imbalance of HNRNPH1 isoforms. This altered splicing of HNRNPH1 was associated with inferior outcomes in MCL and showed a significant increase in protein expression by immunohistochemistry. We describe a functional role for these recurrent noncoding mutations in disrupting an autoregulatory feedback mechanism, thereby deregulating HNRNPH1 protein expression. Taken together, these data strongly imply a role for aberrant regulation of messenger RNA processing in MCL pathobiology.
Collapse
Affiliation(s)
- Prasath Pararajalingam
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Krysta M Coyle
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Sarah E Arthur
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Nicole Thomas
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Miguel Alcaide
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Barbara Meissner
- BC Cancer Centre for Lymphoid Cancer and
- BC Cancer Research Centre, Vancouver, BC, Canada
| | - Merrill Boyle
- BC Cancer Centre for Lymphoid Cancer and
- BC Cancer Research Centre, Vancouver, BC, Canada
| | - Quratulain Qureshi
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Bruno M Grande
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Christopher Rushton
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Graham W Slack
- BC Cancer Centre for Lymphoid Cancer and
- BC Cancer Research Centre, Vancouver, BC, Canada
| | | | - Constantine S Tam
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- University of Melbourne, Melbourne, VIC, Australia
| | - Rishu Agarwal
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- University of Melbourne, Melbourne, VIC, Australia
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | | | - Randy D Gascoyne
- BC Cancer Centre for Lymphoid Cancer and
- BC Cancer Research Centre, Vancouver, BC, Canada
| | - Christian Steidl
- BC Cancer Centre for Lymphoid Cancer and
- BC Cancer Research Centre, Vancouver, BC, Canada
| | - Joseph Connors
- BC Cancer Centre for Lymphoid Cancer and
- BC Cancer Research Centre, Vancouver, BC, Canada
| | - Diego Villa
- BC Cancer Centre for Lymphoid Cancer and
- BC Cancer Research Centre, Vancouver, BC, Canada
| | - Timothy E Audas
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Marco A Marra
- BC Cancer Centre for Lymphoid Cancer and
- BC Cancer Research Centre, Vancouver, BC, Canada
| | | | - David W Scott
- BC Cancer Centre for Lymphoid Cancer and
- BC Cancer Research Centre, Vancouver, BC, Canada
| | - Ryan D Morin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Michael Smith Genome Sciences Centre, Vancouver, BC, Canada
| |
Collapse
|
37
|
Hill HA, Qi X, Jain P, Nomie K, Wang Y, Zhou S, Wang ML. Genetic mutations and features of mantle cell lymphoma: a systematic review and meta-analysis. Blood Adv 2020; 4:2927-2938. [PMID: 32598477 PMCID: PMC7362354 DOI: 10.1182/bloodadvances.2019001350] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an incurable rare subtype of non-Hodgkin lymphoma and is subject to relapse and therapeutic resistance. Molecular aberrations in MCL affect pathogenesis, prognosis, and therapeutic response. In this systematic review, we searched 3 databases and selected 32 articles that described mutations in MCL patients. We then conducted a meta-analysis using a Bayesian multiregression model to analyze patient-level data in 2127 MCL patients, including prevalence of mutations. In tumor or bone marrow samples taken at diagnosis or baseline, ATM was the most frequently mutated gene (43.5%) followed by TP53 (26.8%), CDKN2A (23.9%), and CCND1 (20.2%). Aberrations were also detected in IGH (38.4%) and MYC (20.8%), primarily through cytogenetic methods. Other common baseline mutations were NSD2 (15.0%), KMT2A (8.9%), S1PR1 (8.6%), and CARD11 (8.5%). Our data also show a change in mutational status from baseline samples to samples at disease progression and present mutations of interest in MCL that should be considered for future analysis. The genes with the highest mutational frequency difference (>5%) are TP53, ATM, KMT2A, MAP3K14, BTK, TRAF2, CHD2, TLR2, ARID2, RIMS2, NOTCH2, TET2, SPEN, NSD2, CARD11, CCND1, SP140, CDKN2A, and S1PR1. These findings provide a summary of the mutational landscape of MCL. The genes with the highest change in mutation frequency should be included in targeted next-generation sequencing panels for future studies. These findings also highlight the need for analysis of serial samples in MCL. Patient-level data of prevalent mutations in MCL provide additional evidence emphasizing molecular variability in advancing precision medicine initiatives in MCL.
Collapse
Affiliation(s)
| | - Xinyue Qi
- Department of Biostatistics, MD Anderson Cancer Center, University of Texas, Houston, TX
| | | | | | - Yucai Wang
- Department of Hematology, Mayo Clinic, Rochester, MN; and
| | - Shouhao Zhou
- Department of Public Health Sciences, Pennsylvania State College of Medicine, Hershey, PA
| | | |
Collapse
|
38
|
Cortelazzo S, Ponzoni M, Ferreri AJM, Dreyling M. Mantle cell lymphoma. Crit Rev Oncol Hematol 2020; 153:103038. [PMID: 32739830 DOI: 10.1016/j.critrevonc.2020.103038] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 06/29/2019] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
MCL is a well-characterized generally aggressive lymphoma with a poor prognosis. However, patients with a more indolent disease have been reported in whom the initiation of therapy can be delayed without any consequence for the survival. In 2017 the World Health Organization updated the classification of MCL describing two main subtypes with specific molecular characteristics and clinical features, classical and indolent leukaemic nonnodal MCL. Recent research results suggested an improving outcome of this neoplasm. The addition of rituximab to conventional chemotherapy has increased overall response rates, but it did not improve overall survival compared to chemotherapy alone. The use of intensive frontline therapies including rituximab and consolidation with autologous stem cell transplantation ameliorated response rate and prolonged progression-free survival in young fit patients, but any impact on survival remains to be proven. Furthermore, the optimal timing, cytoreductive regimen and conditioning regimen, and the clinical implications of achieving a disease remission even at molecular level remain to be elucidated. The development of targeted therapies as the consequence of better understanding of pathogenetic pathways in MCL might improve the outcome of conventional chemotherapy and spare the toxicity of intense therapy in most patients. Cases not eligible for intensive regimens, may be considered for less demanding therapies, such as the combination of rituximab either with CHOP or with purine analogues, or bendamustine. Allogeneic SCT can be an effective option for relapsed disease in patients who are fit enough and have a compatible donor. Maintenance rituximab may be considered after response to immunochemotherapy as the first-line strategy in a wide range of patients. Finally, since the optimal approach to the management of MCL is still evolving, it is critical that these patients are enrolled in clinical trials to identify the better treatment options.
Collapse
Affiliation(s)
| | - Maurilio Ponzoni
- Pathology Unit, San Raffaele Scientific Institute, Milan, Italy; Unit of Lymphoid Malignancies, San Raffaele Scientific Institute, Milan, Italy
| | - Andrés J M Ferreri
- Unit of Lymphoid Malignancies, San Raffaele Scientific Institute, Milan, Italy; Medical Oncology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Martin Dreyling
- Medizinische Klinik III der Universität München-Grosshadern, München, Germany
| |
Collapse
|
39
|
Le Bris Y, Magrangeas F, Moreau A, Chiron D, Guérin-Charbonnel C, Theisen O, Pichon O, Canioni D, Burroni B, Maisonneuve H, Thieblemont C, Oberic L, Gyan E, Pellat-Deceunynck C, Hermine O, Delfau-Larue MH, Tessoulin B, Béné MC, Minvielle S, Le Gouill S. Whole genome copy number analysis in search of new prognostic biomarkers in first line treatment of mantle cell lymphoma. A study by the LYSA group. Hematol Oncol 2020; 38:446-455. [PMID: 32472610 DOI: 10.1002/hon.2750] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/11/2020] [Accepted: 05/24/2020] [Indexed: 11/09/2022]
Abstract
Mantle cell lymphoma (MCL) is a lymphoproliferative disorder characterized by the t(11;14)(q13;q32) CCND1/IGH translocation. This lymphoma is however extremely heterogeneous in terms of molecular alterations. Moreover, the course of the disease can vary greatly between indolent forms with slow progression and aggressive conditions rapidly pejorative. The identification of early markers allowing to predict individual patients outcome has however been unsuccessful so far. The LyMa trial treated homogeneously a cohort of young MCL patients. This appeared as a good opportunity to search for biomarkers of response to therapy. DNA extracted from diagnostic paraffin-embedded lymph node biopsies from 100 patients with newly diagnosed MCL, homogeneously treated in this prospective clinical trial, were investigated for copy number alterations and copy neutral loss of heterozygosity using the Oncoscan SNP-array scanning the whole genome. An independent confirmatory cohort was used to strengthen the possibly relevant anomalies observed. Here we describe the recurrent anomalies identified with this technique. Deletions of 17p(TP53) and 9p(CDKN2A) were more frequent in refractory or early relapsing patients (10%), but had no significant impact in univariate analysis on progression-free (PFS) or overall survival (OS). Regardless of the presence of TP53 or CDKN2A deletions, gains in 7p22 (8,5%) were associated with better PFS in univariate but not in multivariate analysis including MCL International Prognostic Index and treatment. Gains of 11q(CCDN1), suggesting gains of the CCND1/IGH fusion, were associated with worse OS and PFS in univariate and multivariate analyses. This worse prognosis impact was confirmed by FISH in an independent confirmatory cohort. This work, using a whole genome approach, confirms the broad genomic landscape of MCL and shows that gains of the CCND1/IGH fusion can be considered as a new prognostic structural variant. Genomic abnormalities of prognostic impact could be useful to strengthen or de-escalate treatment schedules or choosing targeted therapies or CART-cells.
Collapse
Affiliation(s)
- Yannick Le Bris
- Hematology Biology Department, Nantes University Hospital, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Florence Magrangeas
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Anne Moreau
- Pathology Department Nantes University Hospital, now in Centre Hospitalier Départemental de Vendée, La Roche sur Yon, France
| | - David Chiron
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Catherine Guérin-Charbonnel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Institut de Cancérologie de l'Ouest, U892, Saint-Herblain, France
| | - Olivier Theisen
- Hematology Biology Department, Nantes University Hospital, Nantes, France
| | - Olivier Pichon
- Genetic Department, Nantes University Hospital, Nantes, France
| | | | - Barbara Burroni
- Pathology Department, Cochin University Hospital, Paris, France
| | - Hervé Maisonneuve
- Hematology Clinic, Centre Hospitalier Départemental de Vendée, La Roche sur Yon, France
| | | | - Lucie Oberic
- Clinical Hematology Department, IUCT Oncopole, Toulouse University Hospital, Toulouse, France
| | - Emmanuel Gyan
- Clinical Hematology Department, Tours University Hospital, Tours, France
| | | | - Olivier Hermine
- Clinical Hematology Department, Necker University Hospital, Paris, France
| | | | - Benoît Tessoulin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Clinical Hematology Department, Nantes University Hospital, Nantes, France
| | - Marie-Christine Béné
- Hematology Biology Department, Nantes University Hospital, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Stéphane Minvielle
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Steven Le Gouill
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Clinical Hematology Department, Nantes University Hospital, Nantes, France
| |
Collapse
|
40
|
Song Y, Zhou K, Zou D, Zhou J, Hu J, Yang H, Zhang H, Ji J, Xu W, Jin J, Lv F, Feng R, Gao S, Guo H, Zhou L, Elstrom R, Huang J, Novotny W, Wei R, Zhu J. Treatment of Patients with Relapsed or Refractory Mantle–Cell Lymphoma with Zanubrutinib, a Selective Inhibitor of Bruton's Tyrosine Kinase. Clin Cancer Res 2020; 26:4216-4224. [PMID: 32461234 DOI: 10.1158/1078-0432.ccr-19-3703] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/02/2020] [Accepted: 05/19/2020] [Indexed: 11/16/2022]
Affiliation(s)
- Yuqin Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute (Beijing Cancer Hospital), Beijing, China
| | - Keshu Zhou
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Tianjin, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Wuhan, China
| | - Jianda Hu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Haiyan Yang
- Department of Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Huilai Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jie Ji
- Department of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jie Jin
- Department of Hematology, the First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China
| | - Fangfang Lv
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ru Feng
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Sujun Gao
- Department of Hematology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Haiyi Guo
- BeiGene (Shanghai) Co., Shanghai, China
| | - Lei Zhou
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | | | - Jane Huang
- BeiGene USA, Inc., San Mateo, California
| | | | - Rachel Wei
- BeiGene USA, Inc., San Mateo, California
| | - Jun Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute (Beijing Cancer Hospital), Beijing, China.
| |
Collapse
|
41
|
Non-Nodal CD5-Negative Mantle Cell Lymphoma with Secondary TP53 Deletion. Case Rep Hematol 2020; 2020:9185432. [PMID: 32257467 PMCID: PMC7106885 DOI: 10.1155/2020/9185432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/22/2020] [Accepted: 02/20/2020] [Indexed: 01/24/2023] Open
Abstract
Mantle cell lymphoma is a non-Hodgkin lymphoproliferative neoplasm with several clinical and morphologic variants linked, primarily, through genetic derangement of the cyclin D1 locus. Aberrant phenotypes have been described, though prognostic data in such cohorts are limited due to a paucity of cases. We report a case of mantle cell lymphoma with non-nodal clinical presentation, aberrant loss of CD5 expression, and concomitant cytogenetic deletion of 17p. While non-nodal disease is often associated with an improved prognosis in mantle cell lymphoma, this 67-year-old patient experienced a more challenging clinical course with a poor initial response to chemotherapy. Therefore, this case may represent a type of non-nodal mantle cell lymphoma with a prognosis similar to that of classical cases due to the additional phenotypic and genetic alterations found in this patient.
Collapse
|
42
|
Esmeray E, Küçük C. Genetic alterations in B cell lymphoma subtypes as potential biomarkers for noninvasive diagnosis, prognosis, therapy, and disease monitoring. ACTA ACUST UNITED AC 2020; 44:1-14. [PMID: 32123491 PMCID: PMC7049453 DOI: 10.3906/biy-1908-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neoplastic transformation of germinal center B (GCB) cells may give rise to a variety of different B cell lymphoma subtypes, most of which show substantial heterogeneity in terms of genetic alterations and clinical features. The mutations observed in cancer-related genes in GCB cells are related to abnormalities in the immunogenetic mechanisms associated with germinal center reaction. Recent studies have rapidly identified genomic alterations in B cell lymphomas that may be useful for better subclassification, noninvasive diagnosis, and prediction of response to therapy. The WHO recognizes different lymphoma subsets classified within 2 major categories of B cell lymphoma: Hodgkin’s lymphoma (HL) and B cell non-Hodgkin’s lymphoma (NHL), each with distinct genetic aberrations, including chromosomal translocations, copy number abnormalities, or point mutations. Next-generation sequencing-based technologies have allowed cancer researchers to identify somatic mutations and gene expression signatures at a rapid pace so that novel diagnostic or prognostic biomarkers, as well as therapeutic targets, can be discovered much faster than before. Indeed, deep sequencing studies have recently revealed that lymphoma-specific somatic mutations may be detected in cell-free circulating DNA obtained from the peripheral blood of B cell lymphoma patients, suggesting the possibility of minimally invasive diagnosis, monitoring, and predicting response to therapy of B cell lymphoma patients. In this study, the current status of the recurrent genetic aberrations observed during diagnosis and/or relapse in HL and the major subtypes of B cell NHL (i.e. diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma, and Burkitt lymphoma) are discussed to shed light on their potential use as noninvasive diagnostic or prognostic biomarkers and to reveal their role in lymphomagenesis as a target in therapy for newly diagnosed and chemotherapy-resistant cases.
Collapse
Affiliation(s)
- Esra Esmeray
- İzmir Biomedicine and Genome Center, İzmir Turkey.,İzmir International Biomedicine and Genome Institute, Dokuz Eylül University, İzmir Turkey
| | - Can Küçük
- İzmir Biomedicine and Genome Center, İzmir Turkey.,İzmir International Biomedicine and Genome Institute, Dokuz Eylül University, İzmir Turkey.,Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, İzmir Turkey
| |
Collapse
|
43
|
Ferrero S, Rossi D, Rinaldi A, Bruscaggin A, Spina V, Eskelund CW, Evangelista A, Moia R, Kwee I, Dahl C, Di Rocco A, Stefoni V, Diop F, Favini C, Ghione P, Mahmoud AM, Schipani M, Kolstad A, Barbero D, Novero D, Paulli M, Zamò A, Jerkeman M, da Silva MG, Santoro A, Molinari A, Ferreri A, Grønbæk K, Piccin A, Cortelazzo S, Bertoni F, Ladetto M, Gaidano G. KMT2D mutations and TP53 disruptions are poor prognostic biomarkers in mantle cell lymphoma receiving high-dose therapy: a FIL study. Haematologica 2019; 105:1604-1612. [PMID: 31537689 PMCID: PMC7271566 DOI: 10.3324/haematol.2018.214056] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 09/19/2019] [Indexed: 11/16/2022] Open
Abstract
In recent years, the outcome of mantle cell lymphoma (MCL) has improved, especially in younger patients, receiving cytarabine-containing chemoimmunotherapy and autologous stem cell transplantation. Nevertheless, a proportion of MCL patients still experience early failure. To identify biomarkers anticipating failure of intensive chemotherapy in MCL, we performed target resequencing and DNA profiling of purified tumor samples collected from patients enrolled in the prospective FIL-MCL0208 phase 3 trial (high-dose chemoimmunotherapy followed by autologous transplantation and randomized lenalidomide maintenance). Mutations of KMT2D and disruption of TP53 by deletion or mutation associated with an increased risk of progression and death, both in univariate and multivariate analysis. By adding KMT2D mutations and TP53 disruption to the MIPI-c backbone, we derived a new prognostic index, the “MIPI-genetic” (“MIPI- g”). The “MIPI-g” improved the model discrimination ability compared to the MIPI-c alone, defining three risk groups: i) low-risk patients (4-year progression free survival and overall survival of 72.0% and 94.5%); ii) inter-mediate-risk patients (4-year progression free survival and overall survival of 42.2% and 65.8%) and iii) high-risk patients (4-year progression free survival and overall survival of 11.5% and 44.9%). Our results: i) confirm that TP53 disruption identifies a high-risk population characterized by poor sensitivity to conventional or intensified chemotherapy; ii) provide the pivotal evidence that patients harboring KMT2D mutations share the same poor outcome as patients harboring TP53 disruption; and iii) allow to develop a tool for the identification of high-risk MCL patients for whom novel therapeutic strategies need to be investigated. (Trial registered at clinicaltrials.gov identifier: NCT02354313).
Collapse
Affiliation(s)
- Simone Ferrero
- Department of Molecular Biotechnologies and Health Sciences - Hematology Division, Università di Torino, Torino, Italy .,Hematology Division, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Davide Rossi
- Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland.,Universita' della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Andrea Rinaldi
- Universita' della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Alessio Bruscaggin
- Universita' della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Valeria Spina
- Universita' della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Christian W Eskelund
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre, Copenhagen, Denmark
| | - Andrea Evangelista
- Clinical Epidemiology, Città della Salute e della Scienza and CPO Piemonte, Torino, Italy
| | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Ivo Kwee
- Universita' della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland.,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.,Dalle Molle Institute for Artificial Intelligence (IDSIA), Manno, Switzerland
| | - Christina Dahl
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Alice Di Rocco
- Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, "Sapienza" University of Rome, Roma, Italy
| | - Vittorio Stefoni
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Fary Diop
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Chiara Favini
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Paola Ghione
- Department of Molecular Biotechnologies and Health Sciences - Hematology Division, Università di Torino, Torino, Italy
| | - Abdurraouf Mokhtar Mahmoud
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Mattia Schipani
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Arne Kolstad
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Daniela Barbero
- Department of Molecular Biotechnologies and Health Sciences - Hematology Division, Università di Torino, Torino, Italy
| | - Domenico Novero
- First Unit of Pathology, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Marco Paulli
- Unit of Anatomic Pathology, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Alberto Zamò
- Department of Oncology, Università di Torino, Torino, Italy.,Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Mats Jerkeman
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Maria Gomes da Silva
- Department of Hematology, Instituto Português de Oncologia de Lisboa, Lisboa, Portugal
| | - Armando Santoro
- Humanitas Cancer Center, Humanitas Clinical and Research Center, Rozzano, Italy
| | | | - Andres Ferreri
- Lymphoma Unit, Department of Onco-Haematology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Kirsten Grønbæk
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre, Copenhagen, Denmark
| | - Andrea Piccin
- Department of Hematology, Ospedale Generale, Bolzano, Italy
| | | | - Francesco Bertoni
- Universita' della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Marco Ladetto
- SC Ematologia, Azienda Ospedaliera Santi Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| |
Collapse
|
44
|
Classification of aggressive and classic mantle cell lymphomas using synchrotron Fourier Transform Infrared microspectroscopy. Sci Rep 2019; 9:12857. [PMID: 31492883 PMCID: PMC6731317 DOI: 10.1038/s41598-019-49326-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/15/2019] [Indexed: 12/21/2022] Open
Abstract
Mantle cell lymphoma (MCL) is regarded as an incurable neoplasm, even to the novel drug strategies. It is known MCL has two morphological variants- classic and aggressive. Aggressive MCL is characterized by a higher mitotic index and proliferation rate, and poor overall survival in comparison to classic subtype. The insight into the detailed biochemical composition of MCL is crucial in the further development of diagnostic and treatment guidelines for MCL patients; therefore Synchrotron radiation Fourier Transform Infrared (S-FTIR) microspectroscopy combined with Principal Component Analysis (PCA) was used. The major spectral differences were observed in proteins and nucleic acids content, revealing a classification scheme of classic and aggressive MCLs. The results obtained suggest that FTIR microspectroscopy has reflected the histopathological discrimination of both MCL subtypes.
Collapse
|
45
|
Tang C, Kuruvilla J. Optimal management of mantle cell lymphoma in the primary setting. Expert Rev Hematol 2019; 12:715-721. [PMID: 31268728 DOI: 10.1080/17474086.2019.1639501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: The management of mantle cell lymphoma (MCL) has significantly improved since the use of intensified induction and autologous stem cell transplant consolidation. Evolving developments in minimal residual disease detection and novel agent therapy are now challenging this frontline treatment paradigm. Areas covered: This review discusses both the established role of induction and transplant consolidation in MCL, followed by evolving concepts in the use of novel agents in the frontline setting, and the use of minimal residual disease as a driver of MCL management. Expert opinion: In an era of novel agents and improved biologic understanding of MCL, our goal for frontline management should evolve toward personalized therapy for individual patients to maximize efficacy and survival whilst minimizing treatment-related toxicities.
Collapse
Affiliation(s)
- Catherine Tang
- Division of Medical Oncology & Hematology, Princess Margaret Hospital , Toronto , Canada.,Department of Medicine, University of Toronto , Toronto , Canada
| | - John Kuruvilla
- Division of Medical Oncology & Hematology, Princess Margaret Hospital , Toronto , Canada.,Department of Medicine, University of Toronto , Toronto , Canada
| |
Collapse
|
46
|
|
47
|
Detection and Correlation of Single and Concomitant TP53, PTEN, and CDKN2A Alterations in Gliomas. Int J Mol Sci 2019; 20:ijms20112658. [PMID: 31151164 PMCID: PMC6600458 DOI: 10.3390/ijms20112658] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/16/2022] Open
Abstract
Gliomas are the most frequent primary tumors of central nervous system and represent a heterogeneous group of tumors that originates from the glial cells. TP53, PTEN, and CDKN2A are important tumor suppressor genes that encode proteins involved in sustaining cellular homeostasis by different signaling pathways. Though genetic alterations in these genes play a significant role in tumorigenesis, few studies are available regarding the incidence and relation of concomitant TP53, PTEN, and CDKN2A alterations in gliomas. The purpose of this study was to evaluate the occurrence of mutation and deletion in these genes, through single-strand conformational polymorphism, array-comparative genomic hybridization, and fluorescence in situ hybridization techniques, in 69 gliomas samples. Molecular results demonstrated a significant higher prevalence of TP53, PTEN, and CDKN2A alterations in astrocytoma than other tumor subtypes, and heterozygous deletion was the most frequent event. In addition, a significant association was observed between TP53 and CDKN2A alterations (p = 0.0424), which tend to coexist in low grade astrocytomas (5/46 cases (10.9%)), suggesting that they are early events in development of these tumors, and PTEN and CDKN2A deletions (p = 0.0022), which occurred concomitantly in 9/50 (18%) patients, with CDKN2A changes preceding PTEN deletions, present preferably in high-grade gliomas.
Collapse
|
48
|
Clinical and functional impact of recurrent S1PR1 mutations in mantle cell lymphoma. Blood Adv 2019; 2:621-625. [PMID: 29549086 DOI: 10.1182/bloodadvances.2017014860] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/13/2018] [Indexed: 11/20/2022] Open
Abstract
Key Points
S1PR1 mutations are present in 7.8% of patients with MCL and are significantly more frequent at relapse. S1PR1 mutations reduce expression of the S1PR1 receptor, which mediates migration towards the tissue-to-blood egress factor S1P in MCL.
Collapse
|
49
|
Lin RJ, Ho C, Hilden PD, Barker JN, Giralt SA, Hamlin PA, Jakubowski AA, Castro-Malaspina HR, Robinson KS, Papadopoulos EB, Perales MA, Sauter CS. Allogeneic haematopoietic cell transplantation impacts on outcomes of mantle cell lymphoma with TP53 alterations. Br J Haematol 2018; 184:1006-1010. [PMID: 30537212 DOI: 10.1111/bjh.15721] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 11/05/2018] [Indexed: 01/26/2023]
Abstract
TP53 alterations portend extremely poor prognosis in patients with mantle cell lymphoma treated with standard treatment modalities. We reviewed outcomes of 42 patients with available TP53 status who had received a reduced-intensity or non-myeloablative allogeneic haematopoietic cell transplant at our institution. We demonstrated a 2-year overall survival and progression-free survival of 78% [95% confidence interval (CI) 60-88] and 61% (95% CI 43-75), respectively. The 2-year cumulative incidences of relapse and non-relapse mortality were 19% and 20%, respectively. Importantly, there is no significant difference among patients with and without TP53 alterations, suggesting for the first time a beneficial treatment modality for these high-risk patients.
Collapse
Affiliation(s)
- Richard J Lin
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caleb Ho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Patrick D Hilden
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juliet N Barker
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Sergio A Giralt
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Paul A Hamlin
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Ann A Jakubowski
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Hugo R Castro-Malaspina
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Kevin S Robinson
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Esperanza B Papadopoulos
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Miguel-Angel Perales
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Craig S Sauter
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
50
|
Agarwal R, Chan YC, Tam CS, Hunter T, Vassiliadis D, Teh CE, Thijssen R, Yeh P, Wong SQ, Ftouni S, Lam EYN, Anderson MA, Pott C, Gilan O, Bell CC, Knezevic K, Blombery P, Rayeroux K, Zordan A, Li J, Huang DCS, Wall M, Seymour JF, Gray DHD, Roberts AW, Dawson MA, Dawson SJ. Dynamic molecular monitoring reveals that SWI–SNF mutations mediate resistance to ibrutinib plus venetoclax in mantle cell lymphoma. Nat Med 2018; 25:119-129. [DOI: 10.1038/s41591-018-0243-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 09/21/2018] [Indexed: 11/09/2022]
|