1
|
Arai M, Hamad A, Almasry Y, Alamer A, Rasheed W, Aljurf M, El Fakih R. Minimal residual disease testing for classical Hodgkin lymphoma: A comprehensive review. Crit Rev Oncol Hematol 2024; 204:104503. [PMID: 39245298 DOI: 10.1016/j.critrevonc.2024.104503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024] Open
Abstract
Classical Hodgkin lymphoma (cHL) is a common lymphoma that affects young patients. Fortunately, the disease is highly curable as it is susceptible to the currently available treatment modalities. Disease monitoring with Positron Emission Tomography and Computed Tomography (PET/ CT) is an integral part of managing these patients. PET guided protocols are currently used to adjust treatment according to the response. The pivotal idea behind the use of response-adapted approaches is to preserve efficacy while decreasing the toxicity. It also helps to intensify therapy in patients in need because of suboptimal response. However, imaging techniques are limited by their sensitivity and specificity. Minimal Residual Disease (MRD) assessment is a newly emerging concept in many hematologic malignancies. It utilizes various molecular techniques such as polymerase chain reaction (PCR), and next-generation sequencing (NGS) as well as flow cytometry, to detect disease traces. This review looks into MRD detection techniques, its current applications, and the evidence in the literature for its use in cHL.
Collapse
Affiliation(s)
- Momo Arai
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Alaa Hamad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia.
| | - Yazan Almasry
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Abdullah Alamer
- Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Walid Rasheed
- Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Mahmoud Aljurf
- Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Riad El Fakih
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| |
Collapse
|
2
|
Zhou S, Rao J, Ma X, Zeng Y, Xiang X, Li J, Liu H, Lin S, Dong S, Li F, Zhang X, Gao L. Optimized BEAC conditioning regimen improves clinical outcomes of autologous hematopoietic stem cell transplantation in non-Hodgkin lymphomas. Int J Hematol 2024; 120:96-105. [PMID: 38587693 PMCID: PMC11226560 DOI: 10.1007/s12185-024-03755-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024]
Abstract
The conditioning regimen is an important part of autologous hematopoietic stem cell transplantation (ASCT). We explored the efficacy and safety of an optimized BEAC (adjusted-dose, intermediate-dose cytarabine and reduced-dose cyclophosphamide, AD-BEAC) conditioning regimen for non-Hodgkin lymphoma (NHL). A total of 141 NHL patients received AD-BEAC or a standard-dose BEAC (SD-BEAC) conditioning regimen from January 2007 to December 2017, and 104 patients were included in the study after 1:1 propensity matching. The 5-year overall survival (OS) and progression free survival (PFS) rates were significantly higher with AD-BEAC than with SD-BEAC (82.7% vs. 67.3%, P = 0.039; 76.9% vs. 57.7%, P = 0.039). Transplant-related mortality (TRM) was 3.8% in both the AD-BEAC and SD-BEAC groups. The AD-BEAC group had lower incidence of oral ulcers and cardiotoxicity than the SD-BEAC group. An optimized BEAC conditioning regimen is an effective conditioning regimen for ASCT in NHL with acceptable toxicity, that is more effective and safer than a standard BEAC conditioning regimen.
Collapse
Affiliation(s)
- Sha Zhou
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Jun Rao
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiangyu Ma
- Department of Epidemiology, Army Medical University, Chongqing, China
| | - Yunjing Zeng
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Xixi Xiang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Jiali Li
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Hongyun Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Shijia Lin
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Song Dong
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Fu Li
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Li Gao
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China.
| |
Collapse
|
3
|
Kim DH, Siddiqui S, Jain P, Wang M, Thakral B, Li S, Miranda R, Vega F, Medeiros LJ, Ok CY. TP53 mutation is frequent in mantle cell lymphoma with EZH2 expression and have dismal outcome when both are present. Hum Pathol 2024; 146:1-7. [PMID: 38460798 DOI: 10.1016/j.humpath.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
Enhancer of zeste homolog 2 (EZH2) expression is found in about 40% of mantle cell lymphoma (MCL) patients, which is associated with aggressive histology, high Ki-67 proliferation rate, p53 mutant pattern and inferior overall survival (OS). We conducted 11-gene (ATM, BIRC3, CCND1, KMT2C, KMT2D, NOTCH1, NOTCH2, RB1, TP53, TRAF2 and UBR5) next generation sequencing panel to shed more light on MCL with EZH2 expression (EZH2+ MCL). EZH2+ MCL more frequently harbor TP53 mutation compared to EZH2(-) MCL (41.2% vs. 19.1%, respectively, p = 0.045). TP53 mutation and EZH2 expression demonstrated overlapping features including aggressive histology, high Ki-67 proliferation rate and p53 mutant pattern by immunohistochemistry. Comparative analysis disclosed that EZH2 expression correlates with high Ki-67 proliferation rate irrespective of TP53 mutation. Aggressive histology is associated with EZH2 expression or TP53 mutation, possibly via independent mechanisms. p53 mutant pattern is due to TP53 mutation. MCL patients with EZH2 expression or TP53 mutation show inferior outcome and when both are present, patients have dismal outcome.
Collapse
Affiliation(s)
- Do Hwan Kim
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Saima Siddiqui
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Preetesh Jain
- Department of Lymphoma and Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Michael Wang
- Department of Lymphoma and Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Beenu Thakral
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Shaoying Li
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Roberto Miranda
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Francisco Vega
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Chi Young Ok
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Tivey A, Shotton R, Eyre TA, Lewis D, Stanton L, Allchin R, Walter H, Miall F, Zhao R, Santarsieri A, McCulloch R, Bishton M, Beech A, Willimott V, Fowler N, Bedford C, Goddard J, Protheroe S, Everden A, Tucker D, Wright J, Dukka V, Reeve M, Paneesha S, Prahladan M, Hodson A, Qureshi I, Koppana M, Owen M, Ediriwickrema K, Marr H, Wilson J, Lambert J, Wrench D, Burney C, Knott C, Talbot G, Gibb A, Lord A, Jackson B, Stern S, Sutton T, Webb A, Wilson M, Thomas N, Norman J, Davies E, Lowry L, Maddox J, Phillips N, Crosbie N, Flont M, Nga E, Virchis A, Camacho RG, Swe W, Pillai A, Rees C, Bailey J, Jones S, Smith S, Sharpley F, Hildyard C, Mohamedbhai S, Nicholson T, Moule S, Chaturvedi A, Linton K. Ibrutinib as first-line therapy for mantle cell lymphoma: a multicenter, real-world UK study. Blood Adv 2024; 8:1209-1219. [PMID: 38127279 PMCID: PMC10912842 DOI: 10.1182/bloodadvances.2023011152] [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: 07/14/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023] Open
Abstract
ABSTRACT During the COVID-19 pandemic, ibrutinib with or without rituximab was approved in England for initial treatment of mantle cell lymphoma (MCL) instead of immunochemotherapy. Because limited data are available in this setting, we conducted an observational cohort study evaluating safety and efficacy. Adults receiving ibrutinib with or without rituximab for untreated MCL were evaluated for treatment toxicity, response, and survival, including outcomes in high-risk MCL (TP53 mutation/deletion/p53 overexpression, blastoid/pleomorphic, or Ki67 ≥ 30%). A total of 149 patients from 43 participating centers were enrolled: 74.1% male, median age 75 years, 75.2% Eastern Cooperative Oncology Group status of 0 to 1, 36.2% high-risk, and 8.9% autologous transplant candidates. All patients received ≥1 cycle ibrutinib (median, 8 cycles), 39.0% with rituximab. Grade ≥3 toxicity occurred in 20.3%, and 33.8% required dose reductions/delays. At 15.6-month median follow-up, 41.6% discontinued ibrutinib, 8.1% due to toxicity. Of 104 response-assessed patients, overall (ORR) and complete response (CR) rates were 71.2% and 20.2%, respectively. ORR was 77.3% (low risk) vs 59.0% (high risk) (P = .05) and 78.7% (ibrutinib-rituximab) vs 64.9% (ibrutinib; P = .13). Median progression-free survival (PFS) was 26.0 months (all patients); 13.7 months (high risk) vs not reached (NR) (low risk; hazard ratio [HR], 2.19; P = .004). Median overall survival was NR (all); 14.8 months (high risk) vs NR (low risk; HR, 2.36; P = .005). Median post-ibrutinib survival was 1.4 months, longer in 41.9% patients receiving subsequent treatment (median, 8.6 vs 0.6 months; HR, 0.36; P = .002). Ibrutinib with or without rituximab was effective and well tolerated as first-line treatment of MCL, including older and transplant-ineligible patients. PFS and OS were significantly inferior in one-third of patients with high-risk disease and those unsuitable for post-ibrutinib treatment, highlighting the need for novel approaches in these groups.
Collapse
Affiliation(s)
- Ann Tivey
- The University of Manchester, Manchester, United Kingdom
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Rohan Shotton
- The University of Manchester, Manchester, United Kingdom
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Toby A. Eyre
- Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | - David Lewis
- Plymouth Hospitals NHS Trust, Plymouth, United Kingdom
| | | | - Rebecca Allchin
- University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Harriet Walter
- University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Fiona Miall
- University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Rui Zhao
- Torbay Hospital, Torquay, United Kingdom
| | | | - Rory McCulloch
- Gloucestershire Hospitals NHS Foundation Trust, Gloucester, United Kingdom
| | - Mark Bishton
- University of Nottingham, Nottingham, United Kingdom
| | - Amy Beech
- Nottingham University Hospitals, Nottingham, United Kingdom
| | | | - Nicole Fowler
- Royal Cornwall Hospital NHS Trust, Truro, United Kingdom
| | | | - Jack Goddard
- Sheffield Teaching Hospitals, Sheffield, United Kingdom
| | - Sam Protheroe
- Stockport NHS Foundation Trust, Stockport, United Kingdom
| | | | - David Tucker
- Royal Cornwall Hospital NHS Trust, Truro, United Kingdom
| | - Josh Wright
- Sheffield Teaching Hospitals, Sheffield, United Kingdom
| | - Vasavi Dukka
- Stockport NHS Foundation Trust, Stockport, United Kingdom
| | - Miriam Reeve
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Shankara Paneesha
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Mahesh Prahladan
- East Suffolk and North Essex NHS Foundation Trust, Colchester, United Kingdom
| | - Andrew Hodson
- East Suffolk and North Essex NHS Foundation Trust, Colchester, United Kingdom
| | - Iman Qureshi
- University Hospital Coventry and Warwickshire NHS Foundation Trust, Coventry, United Kingdom
| | - Manasvi Koppana
- East Suffolk and North Essex NHS Foundation Trust, Colchester, United Kingdom
| | - Mary Owen
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | | | - Helen Marr
- Newcastle Teaching Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Jamie Wilson
- St Richard's Hospital, Chichester, United Kingdom
| | - Jonathan Lambert
- University College Hospital NHS Foundation Trust, London, United Kingdom
| | - David Wrench
- Guy's and St.Thomas' NHS Foundation Trust, London, United Kingdom
| | - Claire Burney
- University Hospitals Bristol NHS Trust, Bristol, United Kingdom
| | - Chloe Knott
- University Hospitals Bristol NHS Trust, Bristol, United Kingdom
| | - Georgina Talbot
- University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, United Kingdom
| | - Adam Gibb
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | | | - Simon Stern
- Epsom and St Helier University Hospitals NHS Trust, Carshalton, United Kingdom
| | - Taylor Sutton
- Gateshead Health NHS Foundation Trust, Gateshead, United Kingdom
| | - Amy Webb
- Harrogate and District NHS Foundation Trust, Harrogate, United Kingdom
| | - Marketa Wilson
- Harrogate and District NHS Foundation Trust, Harrogate, United Kingdom
| | - Nicky Thomas
- Harrogate and District NHS Foundation Trust, Harrogate, United Kingdom
| | - Jane Norman
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Elizabeth Davies
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Lisa Lowry
- Somerset NHS Foundation Trust, Taunton and Bridgwater, United Kingdom
| | - Jamie Maddox
- South Tees Hospitals NHS Foundation Trust, Middlesborough, United Kingdom
| | - Neil Phillips
- University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, United Kingdom
| | | | - Marcin Flont
- York and Scarborough Teaching Hospitals NHS Foundation, York, United Kingdom
| | - Emma Nga
- Airedale NHS Foundation Trust, Keighley, United Kingdom
| | - Andres Virchis
- The Royal Free London NHS Foundation Trust, London, United Kingdom
| | | | - Wunna Swe
- Calderdale and Huddersfield NHS Foundation Trust, Huddersfield, United Kingdom
| | - Arvind Pillai
- Countess of Chester Hospital NHS Foundation Trust, Chester, United Kingdom
| | - Clare Rees
- Frimley Health NHS Foundation Trust, Frimley, United Kingdom
| | - James Bailey
- Hull University Teaching Hospitals NHS Trust, Hull, United Kingdom
| | - Steve Jones
- Sherwood Forest Hospitals, Nottinghamshire, United Kingdom
| | - Susan Smith
- Sherwood Forest Hospitals, Nottinghamshire, United Kingdom
| | - Faye Sharpley
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Catherine Hildyard
- Milton Keynes University Hospital NHS Foundation Trust, Milton Keynes, United Kingdom
| | - Sajir Mohamedbhai
- University College Hospital NHS Foundation Trust, London, United Kingdom
| | - Toby Nicholson
- St Helens and Knowsley NHS Foundation Trust, Merseyside, United Kingdom
| | - Simon Moule
- Frimley Health NHS Foundation Trust, Frimley, United Kingdom
| | - Anshuman Chaturvedi
- The University of Manchester, Manchester, United Kingdom
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Kim Linton
- The University of Manchester, Manchester, United Kingdom
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
5
|
Wang T, Liu P, Xu L, Gao L, Ni X, Tang G, Chen L, Chen J, Wang L, Wang Y, Fu W, Yue W, Liu N, Li R, Lu G, Luo Y, Yang J. CEAC (oral semustine, etoposide, cytarabine and cyclophosphamide) vs BEAM (carmustine, etoposide, cytarabine, and melphalan) conditioning regimen of autologous stem cell transplantation for diffuse large B-cell lymphoma: a post-hoc, propensity score-matched, cohort study in Chinese patients. Ann Hematol 2024; 103:575-582. [PMID: 37932468 DOI: 10.1007/s00277-023-05513-x] [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: 07/27/2023] [Accepted: 10/14/2023] [Indexed: 11/08/2023]
Abstract
Autologous stem cell transplantation (ASCT) is a salvage therapy for relapsed or refractory diffuse large B-cell lymphoma (DLBCL). We have developed a novel conditioning regimen called CEAC (oral semustine 250 mg/m2 d-6, etoposide 300 mg/m2 d-5 ~ d-2, cytarabine 500 mg/m2 d-5 ~ d-2, and cyclophosphamide 1200 mg/m2 d-5 ~ d-2) In lymphoma patients in China. Here, we conducted a study to compare the conventional BEAM regimen with the CEAC regimen in 110 DLBCL patients. Propensity-score matching was performed in a 1:4 ratio (22 patients received BEAM and 88 received CEAC). Our results showed no significant difference in the overall response rate (95% vs 97%, P = 1.000) and complete response rate (66% vs 73%, P = 0.580) between the two cohorts. The 5-year progression-free survival (PFS), 5-year overall survival (OS), and 5-year cumulative incidence of relapse (CIR) for all patients were 72% (95% CI 62%-82%), 92% (95% CI 86%-97%), and 29% (95% CI 17%-38%), respectively. There was no significant difference in the 5-year PFS (80% vs 70%, P = 0.637), 5-year OS (95% vs 91%, P = 0.496), and 5-year CIR (20% vs 30%, P = 0.733) between cohorts. In terms of safety, the CEAC cohort had a lower incidence rate of grade 1-2 gastrointestinal hemorrhage (P = 0.023) and severe nausea (P = 0.007) compared with the BEAM cohort. In conclusion, the CEAC regimen seems to be a suitable alternative to the BEAM regimen for ASCT in DLBCL patients.
Collapse
Affiliation(s)
- Tao Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ping Liu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Lili Xu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Lei Gao
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xiong Ni
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Gusheng Tang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Li Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Libing Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yang Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Weijia Fu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Wenqin Yue
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Na Liu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ruobing Li
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Guihua Lu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Yanrong Luo
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Jianmin Yang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
6
|
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
|
7
|
Ramsower CA, Rosenthal A, Robetorye RS, Mwangi R, Maurer M, Villa D, McDonnell T, Feldman A, Cohen JB, Habermann T, Campo E, Clot G, Bühler MM, Kulis M, Martin-Subero JI, Giné E, Cook JR, Hill B, Raess PW, Beiske KH, Reichart A, Hartmann S, Holte H, Scott D, Rimsza L. Evaluation of clinical parameters and biomarkers in older, untreated mantle cell lymphoma patients receiving bendamustine-rituximab. Br J Haematol 2024; 204:160-170. [PMID: 37881141 PMCID: PMC11315408 DOI: 10.1111/bjh.19153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/27/2023]
Abstract
Mantle cell lymphoma (MCL) is clinically and biologically heterogeneous. While various prognostic features have been proposed, none currently impact therapy selection, particularly in older patients, for whom treatment is primarily dictated by age and comorbidities. Herein, we undertook a comprehensive comparison of clinicopathological features in a cohort of patients 60 years and older, uniformly treated with bendamustine and rituximab, with a median survival of >8 years. The strongest prognostic indicators in this cohort were a high-risk call by a simplified MCL international prognostic index (s-MIPI) (HR: 3.32, 95% CI: 1.65-6.68 compared to low risk), a high-risk call by MCL35 (HR: 10.34, 95% CI: 2.37-45.20 compared to low risk) and blastoid cytology (HR: 4.21, 95% CR: 1.92-9.22 compared to classic). Patients called high risk by both the s-MIPI and MCL35 had the most dismal prognosis (HR: 11.58, 95% CI: 4.10-32.72), while those with high risk by either had a moderate but clinically relevant prognosis (HR: 2.95, 95% CI: 1.49-5.82). A robust assay to assess proliferation, such as MCL35, along with stringent guidelines for cytological evaluation of MCL, in combination with MIPI, may be a strong path to risk-stratify older MCL patients in future clinical trials.
Collapse
Affiliation(s)
| | - Allison Rosenthal
- Division of Hematology and Medical Oncology, Mayo Clinic, Arizona, Phoenix, USA
| | - Ryan S Robetorye
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, Arizona, USA
| | - Raphael Mwangi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Diego Villa
- Division of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Tim McDonnell
- Department of Hematopathology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew Feldman
- Division of Hematopathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jonathon B Cohen
- Department of Hematology and Medical Oncology, Emory University-Winship Cancer Institute, Atlanta, Georgia, USA
| | | | - Elias Campo
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
- Laboratory of Pathology, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain
| | - Guillem Clot
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
| | - Marco M Bühler
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zürich, Switzerland
| | - Marta Kulis
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
| | - Jose Ignacio Martin-Subero
- Lymphoid Neoplasms Program, Institute for Biomedical Research August Pi I Sunyer, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, ICREA, Barcelona, Spain
| | - Eva Giné
- Department of Hematology, Hospital Clinic of the University of Barcelona, Barcelona, Spain
| | - James R Cook
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Brian Hill
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Philipp W Raess
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Klaus H Beiske
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Alexander Reichart
- Hematology and Oncology, Medical Office of Dres. Brudler/Reichart, Ausburg, Germany
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt am Main, Frankfurt, Germany
| | - Harald Holte
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Center for B Cell Malignancies, Oslo, Norway
| | - David Scott
- Department of Lymphoid Cancer Research, BC Cancer Centre, Vancouver, British Columbia, Canada
| | - Lisa Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, Arizona, USA
| |
Collapse
|
8
|
Karmali R, Abramson JS, Stephens DM, Barnes J, Winter JN, Ma S, Gao J, Kaplan J, Petrich AM, Hochberg E, Takvorian T, Mi X, Nelson V, Gordon LI, Pro B. Ibrutinib maintenance after frontline treatment in patients with mantle cell lymphoma. Blood Adv 2023; 7:7361-7368. [PMID: 37756532 PMCID: PMC10726243 DOI: 10.1182/bloodadvances.2023011271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Maintenance rituximab in mantle cell lymphoma (MCL) has improved survival and supports exploration of maintenance with novel agents. We evaluated the safety and efficacy of ibrutinib maintenance (I-M) after induction in patients with treatment-naive MCL. Patients with MCL with complete response (CR) or partial response to frontline chemoimmunotherapy ± autologous stem cell transplantation (auto-SCT) received I-M 560 mg daily for up to 4 years. Primary objective was 3-year progression-free survival (PFS) rate from initiation of I-M. Minimal residual disease (MRD) assessments by next-generation sequencing (NGS) on peripheral blood were measured before I-M initiation and at 1, 6, and 18 to 24 months after initiation. Among 36 patients, the median age was 60 years (range, 46-90). For frontline treatment, 18 patients (50%) had consolidation with auto-SCT in CR1 before I-M. At median follow-up of 55.7 months, 17 patients (47%) completed full course I-M (median, 37.5 cycles; range, 2-52). The 3-year PFS and overall survival (OS) rates were 94% and 97%, respectively. With prior auto-SCT, 3-year PFS and OS rates were both 100%. The most common treatment-related adverse event with I-M was infection (n = 31; 86%), typically low grade; the most common grade 3/4 toxicities were hematologic. In 22 patients with MRD assessments, all were MRD negative after induction. Six became MRD positive on I-M, with 2 reverting to MRD-negative status with continued I-M, and all maintain radiographic CR with the exception of 1 with disease progression. I-M is feasible in MCL after frontline chemoimmunotherapy with manageable toxicities although significant. Changes in NGS-MRD were noted in limited patients during maintenance with few progression and survival events. This trial was registered at www.clinicaltrials.gov as #NCT02242097.
Collapse
Affiliation(s)
- Reem Karmali
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Jeremy S. Abramson
- Massachusetts General Hospital Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | | | - Jeffrey Barnes
- Massachusetts General Hospital Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Jane N. Winter
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Shuo Ma
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Juehua Gao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jason Kaplan
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Ephraim Hochberg
- Massachusetts General Hospital Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Tak Takvorian
- Massachusetts General Hospital Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Xinlei Mi
- Department of Preventive Medicine–Biostatistics, Northwestern University, Chicago, IL
| | - Valerie Nelson
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Leo I. Gordon
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Barbara Pro
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| |
Collapse
|
9
|
Karolová J, Kazantsev D, Svatoň M, Tušková L, Forsterová K, Maláriková D, Benešová K, Heizer T, Dolníková A, Klánová M, Winkovska L, Svobodová K, Hojný J, Krkavcová E, Froňková E, Zemanová Z, Trněný M, Klener P. Sequencing-based analysis of clonal evolution of 25 mantle cell lymphoma patients at diagnosis and after failure of standard immunochemotherapy. Am J Hematol 2023; 98:1627-1636. [PMID: 37605345 DOI: 10.1002/ajh.27044] [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: 05/18/2023] [Revised: 07/03/2023] [Accepted: 07/16/2023] [Indexed: 08/23/2023]
Abstract
Our knowledge of genetic aberrations, that is, variants and copy number variations (CNVs), associated with mantle cell lymphoma (MCL) relapse remains limited. A cohort of 25 patients with MCL at diagnosis and the first relapse after the failure of standard immunochemotherapy was analyzed using whole-exome sequencing. The most frequent variants at diagnosis and at relapse comprised six genes: TP53, ATM, KMT2D, CCND1, SP140, and LRP1B. The most frequent CNVs at diagnosis and at relapse included TP53 and CDKN2A/B deletions, and PIK3CA amplifications. The mean count of mutations per patient significantly increased at relapse (n = 34) compared to diagnosis (n = 27). The most frequent newly detected variants at relapse, LRP1B gene mutations, correlated with a higher mutational burden. Variant allele frequencies of TP53 variants increased from 0.35 to 0.76 at relapse. The frequency and length of predicted CNVs significantly increased at relapse with CDKN2A/B deletions being the most frequent. Our data suggest, that the resistant MCL clones detected at relapse were already present at diagnosis and were selected by therapy. We observed enrichment of genetic aberrations of DNA damage response pathway (TP53 and CDKN2A/B), and a significant increase in MCL heterogeneity. We identified LRP1B inactivation as a new potential driver of MCL relapse.
Collapse
Affiliation(s)
- J Karolová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine - Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - D Kazantsev
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - M Svatoň
- CLIP - Childhood Leukaemia Investigation Prague, Department of Pediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - L Tušková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - K Forsterová
- First Department of Medicine - Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - D Maláriková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine - Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - K Benešová
- First Department of Medicine - Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - T Heizer
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - A Dolníková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - M Klánová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine - Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - L Winkovska
- CLIP - Childhood Leukaemia Investigation Prague, Department of Pediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - K Svobodová
- Center for Oncocytogenetics, Institute of Medical Biochemistry and Laboratory Diagnostics, Charles University and General University Hospital, Prague, Czech Republic
| | - J Hojný
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - E Krkavcová
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - E Froňková
- CLIP - Childhood Leukaemia Investigation Prague, Department of Pediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Z Zemanová
- Center for Oncocytogenetics, Institute of Medical Biochemistry and Laboratory Diagnostics, Charles University and General University Hospital, Prague, Czech Republic
| | - M Trněný
- First Department of Medicine - Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - P Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine - Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
10
|
Turunen AS, Kuittinen O, Kuitunen H, Vasala K, Penttilä K, Harmanen M, Keskinen L, Mäntymaa P, Pelkonen J, Varmavuo V, Jantunen E, Partanen A. CD34 + Cell Mobilization, Autograft Cellular Composition and Outcome in Mantle Cell Lymphoma Patients. Transfus Med Hemother 2023; 50:428-437. [PMID: 37899989 PMCID: PMC10601603 DOI: 10.1159/000531799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/21/2023] [Indexed: 10/31/2023] Open
Abstract
Backgound Autologous stem cell transplantation (ASCT) is a standard treatment in transplant-eligible mantle cell lymphoma (MCL) patients after first-line chemoimmunotherapy. Study Design and Methods This prospective multicenter study evaluated the impact of CD34+ cell mobilization and graft cellular composition analyzed by flow cytometry on hematologic recovery and outcome in 42 MCL patients. Results During CD34+ cell mobilization, a higher blood CD34+ cell count (>30 × 106/L) was associated with improved overall survival (median not reached [NR] vs. 57 months, p = 0.04). The use of plerixafor did not impact outcome. Higher number of viable cryopreserved graft CD34+ cells (>3.0 × 106/kg) was associated with faster platelet (median 11 vs. 15 days, p = 0.03) and neutrophil (median 9 vs. 10 days, p = 0.02) recovery posttransplant. Very low graft CD3+CD8+ cell count (≤10 × 106/kg) correlated with worse progression-free survival (PFS) (HR 4.136, 95% CI 1.547-11.059, p = 0.005). On the other hand, higher absolute lymphocyte count >2.5 × 109/L at 30 days after ASCT (ALC-30) was linked with better PFS (median NR vs. 99 months, p = 0.045) and overall survival (median NR in either group, p = 0.05). Conclusions Better mobilization capacity and higher graft CD3+CD8+ cell count had a positive prognostic impact in this study, in addition to earlier lymphocyte recovery (ALC-30>2.5 × 106/L). These results need to be validated in another study with a larger patient cohort.
Collapse
Affiliation(s)
| | - Outi Kuittinen
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Oncology, Kuopio University Hospital, Kuopio, Finland
- Cancer Centre, Oulu University Hospital, Oulu, Finland
| | | | - Kaija Vasala
- Department of Oncology, Central Hospital of Central Finland, Jyväskylä, Finland
| | - Karri Penttilä
- Department of Medicine, Savonlinna Central Hospital, Savonlinna, Finland
- Finnish Medicines Agency, Kuopio, Finland
| | - Minna Harmanen
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Leena Keskinen
- Department of Oncology, Tampere University Hospital, Tampere, Finland
| | | | - Jukka Pelkonen
- Laboratory Centre of Eastern Finland, Kuopio, Finland
- Department of Clinical Microbiology, University of Eastern Finland, Kuopio, Finland
| | - Ville Varmavuo
- Department of Medicine, Kymenlaakso Central Hospital, Kotka, Finland
| | - Esa Jantunen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, North Carelia Hospital District, Joensuu, Finland
| | - Anu Partanen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
11
|
de Pádua Covas Lage LA, Elias MDV, Reichert CO, Culler HF, de Freitas FA, de Oliveira Costa R, Rocha V, da Siqueira SAC, Pereira J. Up-Front ASCT Overcomes the Survival Benefit Provided by HDAC-Based Induction Regimens in Mantle Cell Lymphoma: Data from a Real-Life and Long-Term Cohort. Cancers (Basel) 2023; 15:4759. [PMID: 37835453 PMCID: PMC10571660 DOI: 10.3390/cancers15194759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Mantle cell lymphoma (MCL) is a rare malignancy with heterogeneous behavior. Despite the therapeutic advances recently achieved, MCL remains incurable. Currently, the standard of care for young and fit patients involves induction immunochemotherapy followed by up-front autologous stem cell transplantation (ASCT). However, the role of more intensive induction regimens, such as those based on high doses of cytarabine (HDAC), remains controversial in the management of ASCT-eligible patients. METHODS This retrospective, observational, and single-center study involved 165 MCL patients treated at the largest oncology center in Latin America from 2010 to 2022. We aimed to assess outcomes, determine survival predictors, and compare responses between different primary therapeutic strategies, with a focus on assessing the impact of HDAC-based regimens on outcomes in ASCT-eligible patients. RESULTS The median age at diagnosis was 65 years (38-89 years), and 73.9% were male. More than 90% of the cases had a classic nodal form (cnMCL), 76.4% had BM infiltration, and 56.4% presented splenomegaly. Bulky ≥ 7 cm, B-symptoms, ECOG ≥ 2, and advanced-stage III/IV were observed in 32.7%, 64.8%, 32.1%, and 95.8%, respectively. Sixty-four percent of patients were categorized as having high-risk MIPI. With a median follow-up of 71.1 months, the estimated 2-year OS and EFS were 64.1% and 31.8%, respectively. Patients treated with (R)-HDAC-based regimens had a higher ORR (85.9% vs. 65.7%, p = 0.007) compared to those receiving (R)-CHOP, as well as lower POD-24 rates (61.9% vs. 80.4%, p = 0.043) and lower mortality (43.9% vs. 68.6%, p = 0.004). However, intensified induction regimens with (R)-HDAC were not associated with a real OS benefit in MCL patients undergoing up-front consolidation with ASCT (2-year OS: 88.7% vs. 78.8%, p = 0.289). Up-front ASCT was independently associated with increased OS (p < 0.001), EFS (p = 0.005), and lower POD-24 rates (p < 0.001) in MCL. Additionally, CNS infiltration, TLS, hypoalbuminemia, and the absence of remission after induction were predictors of poor OS. CONCLUSIONS In the largest Latin American cohort of MCL patients, we confirmed the OS benefit promoted by up-front consolidation with ASCT in young and fit patients, regardless of the intensity of the immunochemotherapy regimen used in the pre-ASCT induction. Although HDAC-based regimens were not associated with an unequivocal increase in OS for ASCT-eligible patients, it was associated with higher ORR and lower rates of early relapses for the whole cohort.
Collapse
Affiliation(s)
- Luís Alberto de Pádua Covas Lage
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
| | - Marcela do Vale Elias
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
| | - Cadiele Oliana Reichert
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
| | - Hebert Fabrício Culler
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
| | - Fábio Alessandro de Freitas
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
| | - Renata de Oliveira Costa
- Department of Hematology and Hemotherapy, Faculty of Medical Sciences of Santos (FCMS), Santos 01238-010, SP, Brazil
- Hospital Alemão Osvaldo Cruz (HAOC), São Paulo 01323-020, SP, Brazil
| | - Vanderson Rocha
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
- Fundação Pró-Sangue, Blood Bank of São Paulo, São Paulo 05403-000, SP, Brazil
- Department of Hematology, Churchill Hospital, Oxford University, Oxford OX3 7LE, UK
| | | | - Juliana Pereira
- Department of Hematology, Hemotherapy & Cell Therapy, Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil; (M.d.V.E.)
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculty of Medicine, University of São Paulo (USP), São Paulo 05508-080, SP, Brazil
- Hospital Alemão Osvaldo Cruz (HAOC), São Paulo 01323-020, SP, Brazil
| |
Collapse
|
12
|
Patel D, Kahl B. SOHO State of the Art Updates and Next Questions: Tailoring Upfront Therapy in Mantle Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:633-641. [PMID: 37268478 DOI: 10.1016/j.clml.2023.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 06/04/2023]
Abstract
In this article, we will review current strategies for the front-line management of mantle cell lymphoma, an uncommon and biologically and clinically heterogeneous subtype of non-Hodgkin lymphoma that remains incurable with current therapies. Patients invariably relapse with time, and as a result, treatment strategies involve persistent therapy over the course of months to years, including induction, consolidation, and maintenance. Topics discussed include the historical development of various chemoimmunotherapy backbones with continued modifications to maintain and improve efficacy while limiting off-target, off-tumor effects. Chemotherapy-free induction regimens were developed initially for elderly or less fit patients though are now being utilized for younger, transplant-eligible patients due to deeper, more prolonged remission durations with fewer toxicities. The historic paradigm of recommending autologous hematopoietic cell transplant for fit patients in complete or partial remission is now being challenged based in part on ongoing clinical trials in which minimal residual disease directed approaches influence the consolidation strategy for any particular individual. The addition of novel agents, namely first and second generation Bruton tyrosine kinase inhibitors as well as immunomodulatory drugs, BH3 mimetics, and type II glycoengineered anti-CD20 monoclonal antibodies have been tested in various combinations with or without immunochemotherapy. We will attempt to help the reader by systematically explaining and simplifying the various approaches for treating this complicated group of disorders.
Collapse
Affiliation(s)
- Dilan Patel
- Washington University in St Louis School of Medicine, St Louis, Department of Medicine, Division of Oncology, MO
| | - Brad Kahl
- Washington University in St Louis School of Medicine, St Louis, Department of Medicine, Division of Oncology, MO..
| |
Collapse
|
13
|
Scheubeck G, Jiang L, Hermine O, Kluin-Nelemans HC, Schmidt C, Unterhalt M, Rosenwald A, Klapper W, Evangelista A, Ladetto M, Jerkeman M, Ferrero S, Dreyling M, Hoster E. Clinical outcome of Mantle Cell Lymphoma patients with high-risk disease (high-risk MIPI-c or high p53 expression). Leukemia 2023; 37:1887-1894. [PMID: 37495776 PMCID: PMC10457193 DOI: 10.1038/s41375-023-01977-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/28/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
Currently, treatment allocation of patients with Mantle Cell Lymphoma (MCL) is mainly based on age and medical fitness. The combined MCL International Prognostic Index (MIPI-c) allows to predict prognosis using clinical factors (MIPI) and the Ki-67 index. However, high p53 expression as surrogate for TP53 alterations has demonstrated to be an independent predictor for poor outcome. We aimed to define a clear high-risk group based on the combination of MIPI, Ki-67 and p53 expression/TP53 alteration. A total of 684 patients from the prospective European MCL-Younger and MCL-Elderly trials were evaluable. The classification of high-risk disease (HRD) as high-risk MIPI-c or p53 expression >50% versus low-risk disease (LRD) as low, low-intermediate or high-intermediate MIPI-c and p53 expression ≤50% allowed to characterize two distinct groups with highly divergent outcome. Patients with HRD had significantly shorter median failure-free survival (FFS) (1.1 vs. 5.6 years, p < 0.0001) and overall survival (OS) (2.2 vs. 13.2 years, p < 0.0001) compared to those with LRD. These major differences were confirmed in two validation cohorts from the Italian MCL0208 and the Nordic-MCL4 trials. The results suggest that this subset of HRD patients is not sufficiently managed with the current standard treatment and is asking for novel treatment strategies.
Collapse
Affiliation(s)
- Gabriel Scheubeck
- Department of Medicine III, LMU University Hospital, Munich, Germany.
| | - Linmiao Jiang
- Institute for Medical Information Processing, Biometry, and Epidemiology, LMU Munich, Munich, Germany
| | | | - Hanneke C Kluin-Nelemans
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Christian Schmidt
- Department of Medicine III, LMU University Hospital, Munich, Germany
| | - Michael Unterhalt
- Department of Medicine III, LMU University Hospital, Munich, Germany
| | | | - Wolfram Klapper
- Department of Pathology, Hematopathology Section, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Andrea Evangelista
- Unit of Clinical Epidemiology, Azienda Ospedaliera Universitaria Città della Salute e della Scienza and CPO Piemonte, Turin, Italy
| | - Marco Ladetto
- Hematology, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | | | - Simone Ferrero
- Division of Hematology, Department of Molecular Biotechnologies and Health Sciences University of Torino/AOU "Città della Salute e della Scienza di Torino", Turin, Italy
| | - Martin Dreyling
- Department of Medicine III, LMU University Hospital, Munich, Germany
| | - Eva Hoster
- Department of Medicine III, LMU University Hospital, Munich, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology, LMU Munich, Munich, Germany
| |
Collapse
|
14
|
Patel DA, Wan F, Trinkaus K, Guy DG, Edwin N, Watkins M, Bartlett NL, Cashen A, Fehniger TA, Ghobadi A, Shah NM, Kahl BS. Bendamustine/Rituximab Plus Cytarabine/Rituximab, With or Without Acalabrutinib, for the Initial Treatment of Transplant-Eligible Mantle Cell Lymphoma Patients: Pooled Data From Two Pilot Studies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023:S2152-2650(23)00131-3. [PMID: 37183115 DOI: 10.1016/j.clml.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Mantle cell lymphoma (MCL) is a moderately aggressive lymphoma subtype, generally viewed as incurable. For younger, fit patients, the standard of care remains various high-dose cytarabine-based induction regimens followed by autologous hematopoietic cell transplant and 3 years of rituximab maintenance. Despite reasonably good outcomes, with median progression-free survival in the range of 7 to 9 years, most patients eventually relapse, indicating a need to improve the safety and tolerability of remission induction strategies. METHODS Given the impressive activity of bendamustine/rituximab (BR) in older patients with MCL, we developed an induction regimen modeled after the Nordic Regimen but substituted BR in place of R-CHOP. In a second pilot study, we incorporated the second-generation Bruton tyrosine kinase inhibitor (BTKi), acalabrutinib, into the regimen. The primary endpoint of both studies was stem cell mobilization success rate. RESULTS All patients successfully underwent stem cell harvest in both studies. CONCLUSION The experience from our single institution pilot study suggested that sequential rather than alternating BR and cytarabine/rituximab (CR) was easier to administer from the standpoint of toxicities and subsequent dose modifications. Safety and efficacy data from the 2 pilot studies, FitMCL 1.0 and 2.0, were similar. The pilot studies provided preliminary safety data supporting the development of the NCTN trial EA4181, assessing three different induction regimens with or without acalabrutinib.
Collapse
Affiliation(s)
- Dilan A Patel
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Fei Wan
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Kathryn Trinkaus
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Daniel G Guy
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Natasha Edwin
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Marcus Watkins
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Nancy L Bartlett
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Amanda Cashen
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Todd A Fehniger
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Armin Ghobadi
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Neha-Mehta Shah
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Brad S Kahl
- Washington University in St Louis School of Medicine, St Louis, MO, 63110.
| |
Collapse
|
15
|
Harmanen M, Hujo M, Sund R, Sorigue M, Khan M, Prusila R, Klaavuniemi T, Kari E, Jantunen E, Sunela K, Rajamäki A, Alanne E, Kuitunen H, Sancho JM, Jukkola A, Rönkä A, Kuittinen O. Survival of patients with mantle cell lymphoma in the rituximab era: Retrospective binational analysis between 2000 and 2020. Br J Haematol 2022; 201:64-74. [PMID: 36513500 DOI: 10.1111/bjh.18597] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022]
Abstract
Mantle cell lymphoma (MCL) is a rare peripheral B-cell lymphoma characterised by eventual relapse and progression towards a more aggressive disease biology. With the introduction of rituximab- and cytarabine-based immunochemotherapy regimens, the prognosis of the disease has changed dramatically over the last two decades. To assess the real-world survival of patients with MCL, we used a population-based cohort of 564 patients with MCL who were diagnosed and treated between 2000 and 2020. Patient data were collected from seven Finnish treatment centres and one Spanish treatment centre. For the entire patient population, we report a 2-year overall survival (OS) rate of 77%, a 5-year OS of 58%, and a 10-year OS of 32%. The estimated median OS was 80 months after diagnosis. MCL is associated with increased mortality across the entire patient population. Additionally, we assessed the survival of patients after MCL relapse with the aim of establishing a cut-off point of prognostic significance. Based on our statistical analysis of survival after the first relapse, disease progression within 24 months of the initial diagnosis should be considered as a strong indicator of poor prognosis.
Collapse
Affiliation(s)
- Minna Harmanen
- Oncology, Faculty of Health Medicine, School of Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mika Hujo
- Statistics, Faculty of Science and Forestry, School of Computing, University of Eastern Finland, Kuopio, Finland
| | - Reijo Sund
- Faculty of Health Medicine, School of Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Marc Sorigue
- Department of Hematology, ICO-Badalona, Hospital Germans Trias i Pujol, IJC, Badalona, Spain
| | - Madiha Khan
- Oncology, Faculty of Health Medicine, School of Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Roosa Prusila
- Medical Research Centre and Cancer and Translational Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | | | - Esa Kari
- Department of Oncology, Tampere University Hospital, Tampere Cancer Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Esa Jantunen
- Department of Medicine, Hospital District of North Carelia, Institute of Clinical Medicine/Internal Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Kaisa Sunela
- Department of Oncology, Tampere University Hospital, Tampere Cancer Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Aino Rajamäki
- Department of Oncology, Hospital Nova of Central Finland, Jyväskylä, Finland.,Faculty of Health Medicine, School of Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Erika Alanne
- Department of Oncology and Radiotherapy, Turku University Hospital, Western Finland Cancer Centre, Turku, Finland
| | - Hanne Kuitunen
- Medical Research Centre and Cancer and Translational Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Juan-Manuel Sancho
- Department of Hematology, ICO-Badalona, Hospital Germans Trias i Pujol, IJC, Badalona, Spain
| | - Arja Jukkola
- Department of Oncology, Tampere University Hospital, Tampere Cancer Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Aino Rönkä
- Oncology, Faculty of Health Medicine, School of Medicine, Institute of Clinical Medicine, Kuopio University Hospital Department of Oncology, University of Eastern Finland, Kuopio, Finland
| | - Outi Kuittinen
- Oncology, Faculty of Health Medicine, School of Medicine, Institute of Clinical Medicine, Kuopio University Hospital Department of Oncology, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
16
|
Whole Transcriptome Sequencing Reveals Cancer-Related, Prognostically Significant Transcripts and Tumor-Infiltrating Immunocytes in Mantle Cell Lymphoma. Cells 2022; 11:cells11213394. [PMID: 36359790 PMCID: PMC9654955 DOI: 10.3390/cells11213394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/29/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma (NHL) subtype characterized by overexpression of CCND1 and SOX11 genes. It is generally associated with clinically poor outcomes despite recent improvements in therapeutic approaches. The genes associated with the development and prognosis of MCL are still largely unknown. Through whole transcriptome sequencing (WTS), we identified mRNAs, lncRNAs, and alternative transcripts differentially expressed in MCL cases compared with reactive tonsil B-cell subsets. CCND1, VCAM1, and VWF mRNAs, as well as MIR100HG and ROR1-AS1 lncRNAs, were among the top 10 most significantly overexpressed, oncogenesis-related transcripts. Survival analyses with each of the top upregulated transcripts showed that MCL cases with high expression of VWF mRNA and low expression of FTX lncRNA were associated with poor overall survival. Similarly, high expression of MSTRG.153013.3, an overexpressed alternative transcript, was associated with shortened MCL survival. Known tumor suppressor candidates (e.g., PI3KIP1, UBXN) were significantly downregulated in MCL cases. Top differentially expressed protein-coding genes were enriched in signaling pathways related to invasion and metastasis. Survival analyses based on the abundance of tumor-infiltrating immunocytes estimated with CIBERSORTx showed that high ratios of CD8+ T-cells or resting NK cells and low ratios of eosinophils are associated with poor overall survival in diagnostic MCL cases. Integrative analysis of tumor-infiltrating CD8+ T-cell abundance and overexpressed oncogene candidates showed that MCL cases with high ratio CD8+ T-cells and low expression of FTX or PCA3 can potentially predict high-risk MCL patients. WTS results were cross-validated with qRT-PCR of selected transcripts as well as linear correlation analyses. In conclusion, expression levels of oncogenesis-associated transcripts and/or the ratios of microenvironmental immunocytes in MCL tumors may be used to improve prognostication, thereby leading to better patient management and outcomes.
Collapse
|
17
|
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
|
18
|
Affiliation(s)
- James O Armitage
- From the Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska, Omaha (J.O.A.)
| | - Dan L Longo
- From the Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska, Omaha (J.O.A.)
| |
Collapse
|
19
|
Amhaz G, Bazarbachi A, El-Cheikh J. Immunotherapy in indolent Non-Hodgkin's Lymphoma. Leuk Res Rep 2022; 17:100325. [PMID: 35663281 PMCID: PMC9160834 DOI: 10.1016/j.lrr.2022.100325] [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] [Received: 09/20/2021] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 12/01/2022] Open
Abstract
Treatment of non-Hodgkin lymphoma (NHL) in general has improved over the years with the emergence of the monoclonal antibodies (MAB) therapy. NHL is divided into B cell NHL and T cell NHL. Treatment of NHL was based on the subtype of NHL and its staging. NHL is divided into aggressive and indolent NHL (iNHL). Subtypes of iNHL include: Follicular lymphoma (FL), Marginal zone lymphoma (MZL), Chronic lymphocytic leukemia/small-cell lymphocytic lymphoma (CLL/SLL), Gastric mucosa-associated lymphoid tissue (MALT) lymphoma, Lymphoplasmacytic lymphoma, Waldenström macroglobulinemia, Nodal marginal zone lymphoma (NMZL), Splenic marginal zone lymphoma (SMZL). Chemotherapy was the main stay treatment of iNHL until the emergence of Rituximab, anti-CD20 MAB targeting CD-20 surface cell antigens that are present on B-cells lymphoma and not on precursor cells, mainly efficacious in B cell iNHL, It became the mainstay treatment in follicular lymphoma (FL) as a single agent modality or in combination with chemotherapy. The anti-CD20 Rituximab played an important role in the development of the treatment of iNHL to become FDA approved in 1997. It was also proven effective in multiple other types of lymphoma. MAB through targeting the cell surface antigen leads to a direct or immune mediated cytotoxicity. This carries few side effects, including allergic reactions. Other than that, a resistance mechanism to rituximab emerged by inducing a failure in the apoptosis mechanism. Alternative mechanisms of resistance included the presence of soluble antigens that could act by binding to the antibody present before the drug itself can bind the lymphoma cell. Thus, the interest in immunotherapy grew further to explore the possibility of conjugating an immune mediated drug to a radio-sensitizing agent in order to enhance the selectivity of the drug. Here came the development of 90Y-ibritumomab tiuxetan and 131I-tositumomab. After it, humanized anti-CD20 emerged ofatumumab, IMMU106 (veltuzumab) in 2005, and ocrelizumab which are considered as second generation anti-CD20 and 3rd generation anti-CD20 include AME-133v (ocaratuzumab), PRO131921 and GA101 (obinutuzumab). Also multiple other agents emerged targeting different surface cell antigens like CD52 (alemtuzumab), CD22 (unconjugated epratuzumab and calicheamicin conjugated CMC-544 [inotuzumab ozogamicin]), CD80 (galiximab), CD2 (MEDI-507 [siplizumab]), CD30 (SGN-30 and MDX-060 [iratumumab], Brentuximab vedotin), CD40 (SGN-40), and CD79b (Polatuzumab). Other agents include MAB targeting T-Cells like mogamulizumab, Denileukin Diftitox and BiTEs or bispecific T cell engagers like Mosunetuzumab, Glofitamab, and Epcoritamab. Moreover, further studies came up to evaluate the role of immunotherapy in combination chemotherapy as a pathway to evade the resistance mechanisms. Side effects of the treatment were mainly infusion related adverse reactions, myelosuppression in conjugated forms leading to immunosuppression and subsequently to infectious complications. Another important aspect in immunotherapy is the half-lives of the medication which is an important factor that can influence the evaluation of the response. The MAB treatment showed important benefit in the treatment of iNHL and it continuously shows how rapidly it can develop to provide optimum care and benefit to patients with iNHL.
Collapse
Affiliation(s)
- Ghid Amhaz
- Division of hematology-oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Bazarbachi
- Division of hematology-oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jean El-Cheikh
- Division of hematology-oncology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
20
|
Jain P, Wang ML. Mantle cell lymphoma in 2022-A comprehensive update on molecular pathogenesis, risk stratification, clinical approach, and current and novel treatments. Am J Hematol 2022; 97:638-656. [PMID: 35266562 DOI: 10.1002/ajh.26523] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 12/21/2022]
Abstract
The field of mantle cell lymphoma (MCL) has witnessed remarkable progress due to relentless advances in molecular pathogenesis, prognostication, and newer treatments. MCL consists of a spectrum of clinical subtypes. Rarely, atypical cyclin D1-negative MCL and in situ MCL neoplasia are identified. Prognostication of MCL is further refined by identifying somatic mutations (such as TP53, NSD2, KMT2D), methylation status, chromatin organization pattern, SOX-11 expression, minimal residual disease (MRD), and genomic clusters. Lymphoid tissue microenvironment studies demonstrated the role of B-cell receptor signaling, nuclear factor kappa B (NF-kB), colony-stimulating factor (CSF)-1, the CD70-SOX-11 axis. Molecular mechanism of resistance, mutation dynamics, and pathogenic pathways (B-cell receptor (BCR), oxidative phosphorylation, and MYC) were identified in mediating resistance to various treatments (bruton tyrosine kinase (BTK) inhibitors [ibrutinib, acalabrutinib]. Treatment options range from conventional chemoimmunotherapy and stem cell transplantation (SCT) to targeted therapies against BTK (covalent and noncovalent), Bcl2, ROR1, cellular therapy such as anti-CD19 chimeric antigen receptor therapy (CAR-T), and most recently bispecific antibodies against CD19 and CD20. MCL patients frequently relapse. Complex pathogenesis and the management of patients with progression after treatment with BTK/Bcl2 inhibitors and CAR-T (triple-resistant MCL) remain a challenge. Next-generation clinical trials incorporating newer agents and concurrent translational and molecular investigations are ongoing.
Collapse
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma. Mantle cell lymphoma center of excellence The University of Texas MD Anderson Cancer Center Houston Texas USA
| | - Michael L. Wang
- Department of Lymphoma/Myeloma. Mantle cell lymphoma center of excellence The University of Texas MD Anderson Cancer Center Houston Texas USA
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Validated metrics to optimize older adult patient selection for Chimeric Antigen Receptor T-cell therapy (CART) are lacking; however, some preliminary data suggests that geriatric assessments and cumulative illness rating score may be useful tools. In addition, interventions capable of enhancing outcomes in older adults receiving CART have yet to be elucidated. The purpose of this review is to present data extrapolating from other diseases and therapeutic modalities, related to product selection, toxicity mitigation strategies, comprehensive coordinated models of care, and functional optimization of patients. RECENT FINDINGS The most robust data in older adults are among relapsed and refractory (r/r) diffuse large B-cell lymphoma (DLBCL) patients where three products are available with the longest clinical follow up and the most abundant real-world evidence (RWE). Data for the approved CART products for follicular lymphoma (FL) and mantle cell lymphoma (MCL) are relatively new and RWE is lacking in general. Data for CART products in multiple myeloma (MM) and B-cell acute lymphoblastic leukemia (B-ALL) are even more recent, but preliminary data in older adults seem to follow the trend of excellent efficacy in this age group with age-stratified toxicity data limited. Landmark trials and RWE studies indicate that the high response rates of CART for older adult patients, age 65 years and older, are maintained, while toxicity may be amplified. Clinically important toxicities include grade 3 or higher cytokine release syndrome (CRS), neurotoxicity, and infections.
Collapse
|
22
|
Wang YH, Hsieh CY, Hsiao LT, Lin TL, Liu YC, Yao M, Tan TD, Ko BS. Stem cell transplant for mantle cell lymphoma in Taiwan. Sci Rep 2022; 12:5662. [PMID: 35383213 PMCID: PMC8983774 DOI: 10.1038/s41598-022-09539-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/17/2022] [Indexed: 11/09/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a B-cell lymphoma featuring an aggressive course and a progressive relapsing pattern. International guidelines recommend early consolidative autologous stem cell transplant (auto-SCT) for eligible patients while reserving allogeneic SCT (allo-SCT) as therapy for refractory cases. Since data describing the implementation of transplants in the Asian population with MCL are limited, we aimed to analyze post-SCT outcomes of 99 MCL patients from the Taiwan Bone Marrow Transplant Registry database. The median age was 56 years, and 11% of the patients had blastoid variant MCL. Ninety-four patients received auto-SCT, while 13 patients received allo-SCT, eight of which received allo-SCT after failing auto-SCT. Before auto-SCT, 52% of the patients were in their first complete remission (CR1). Overall, 37 patients (39%) relapsed after auto-SCT. The median post-auto-SCT progression-free survival and overall survival (OS) were 43.6 months and not reached, respectively. Blastoid variant MCL, transplant not received in CR1, and disease progression within 12 months post-auto-SCT independently predicted inferior OS in multivariable analysis. The median post-allo-SCT OS was 74 months. Two patients (15%) died of MCL recurrence post-allo-SCT. Three patients with refractory diseases were salvaged with ibrutinib or venetoclax to allo-SCT. Treatment strategies incorporating novel agents warrant further optimization.
Collapse
Affiliation(s)
- Yu-Hung Wang
- Stem Cell and Leukaemia Proteomics Laboratory, University of Manchester, Manchester, UK
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Yun Hsieh
- Division of Hematology and Oncology, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Liang-Tsai Hsiao
- Division of Hematology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tung-Liang Lin
- Division of Hematology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Yi-Chang Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming Yao
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tran-Der Tan
- Division of Hematology and Medical Oncology, Koo Foundation Sun Yat-Sen Cancer Center, No. 125, Lih-Der Road, Pei-Tou District, Taipei, 112, Taiwan.
| | - Bor-Sheng Ko
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Hematological Oncology, National Taiwan University Cancer Center, No. 57, Lane 155, Section 3 of Keelung Rd, Taipei, 100, Taiwan.
| |
Collapse
|
23
|
Evolving frontline immunochemotherapy for mantle cell lymphoma and the impact on survival outcomes. Blood Adv 2022; 6:1350-1360. [PMID: 34662895 PMCID: PMC8864651 DOI: 10.1182/bloodadvances.2021005715] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022] Open
Abstract
Because there have been a dvances in frontline treatment for mantle cell lymphoma (MCL) over the last 2 decades, we sought to characterize the changes in frontline treatment patterns and their association with outcomes. Patients with newly diagnosed MCL from September 2002 through June 2015 were enrolled in a prospective cohort study, and clinical characteristics, treatment, and clinical outcomes were compared between patients diagnosed from 2002 to 2009 (Era 1) compared with 2010 to 2015 (Era 2). Patient age, sex, and simplified MCL International Prognostic Index (sMIPI) score were similar between the 2 groups. In patients age 65 years or younger, there was less use of rituximab plus hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone (R-Hyper-CVAD) (16.1% vs 8.8%) but more use of rituximab plus maximum-strength cyclophosphamide, doxorubicin, vincristine, and prednisone (R-maxi-CHOP) alternating with rituximab plus high-dose cytarabine (R-HiDAC), also known as the Nordic regimen, and R-CHOP alternating with rituximab plus dexamethasone, high-dose cytarabine, and cisplatin (R-DHAP) (1.1% vs 26.4%) and less use of R-CHOP or R-CHOP-like regimens (64.5% vs 35.2%) but more use of R-bendamustine (0% vs 12.1%) in Era 2 (P < .001). These changes were associated with improved event-free survival (EFS; 5-year EFS, 34.3% vs 50.0%; P = .010) and overall survival (OS; 5-year OS, 68.8% vs 81.6%; P = .017) in Era 2. In patients older than age 65 years, there was less use of R-CHOP or R-CHOP-like therapy (39.0% vs 14.3%) and nonstandard systemic therapy (36.6% vs 13.0%) but more use of R-bendamustine (0% vs 49.4%). These changes were associated with a trend for improved EFS (5-year EFS, 25.4% vs 37.5%; P = .051) in Era 2. The shift from R-CHOP or R-CHOP-like regimens to R-bendamustine was associated with improved EFS (5-year EFS, 25.0% vs 44.6%; P = .008) in Era 2. Results from this prospective cohort study provide critical real-world evidence for improved outcomes with evolving frontline patterns of care in patients with MCL.
Collapse
|
24
|
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: 33] [Impact Index Per Article: 11.0] [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
|
25
|
Wu M, Li Y, Huang H, Xu W, Wang Y, Huang H, Zhao W, Liu S, Xu P, Chen Z, Zhu J, Song Y, Ruan J, Wu D. Initial Treatment Patterns and Survival Outcomes of Mantle Cell Lymphoma Patients Managed at Chinese Academic Centers in the Rituximab Era: A Real-World Study. Front Oncol 2022; 11:770988. [PMID: 35059312 PMCID: PMC8763847 DOI: 10.3389/fonc.2021.770988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/06/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose The aim of the study was to delineate the disease characteristics, the initial treatment patterns, and survival in patients with mantle cell lymphoma (MCL) managed in the real world. Methods Data of 518 MCL patients from 5 major Chinese Hematology Centers in the period from 2007 to 2017 were retrospectively analyzed. Results The median age was 58 years. Of the patients, 88.6% had Eastern Cooperative Oncology Group Performance Status (ECOG PS) 0–1 and 80.7% had advanced-stage disease. Ki67 expression was <30% in 39.6% of the patients, and 43.2% of patients were categorized into a low-risk group based on the Mantle Cell Lymphoma International Prognostic Index (MIPI) scoring system. Overall, 73.4% of the patients received rituximab as their first-line therapy. The most commonly used chemotherapy was the CHOP-like (cyclophosphamide, hydroxydaunomycin, oncovin, and prednisone) regimen (45.2%), followed by high-dose cytarabine-containing chemotherapy (31.3%) and bendamustine (3.3%). Of the patients, 13.7% (n = 71) underwent consolidative autologous stem cell transplantation (ASCT), and 19.3% (n = 100) received novel agents containing first-line regimens. With a median follow-up time of 52 months, the 3- and 5-year overall survival (OS) rates were 73.7% and 61.4%, respectively. Age ≤60 years, ECOG PS 0–1, stages I–II, normal lactate dehydrogenase (LDH), absence of bone marrow involvement, Ki67 <30%, and lower-risk IPI/MIPI scores were significantly associated with improved OS (p < 0.05). The inclusion of rituximab improved the 5-year OS, with borderline significance (62.5% vs. 55.2%, p = 0.076). High-dose cytarabine-containing chemotherapy showed significant clinical benefit in 5-year OS (72.1% vs. 55.9%, p = 0.010). Patients with ASCT had better 5-year OS in the younger (≤60 years) age group (87.2% vs. 64.8%, p = 0.002). Conclusion This large retrospective dataset unequivocally confirmed the survival advantage afforded by cytarabine-containing regimen and ASCT in a first-line setting under real-world management in the rituximab era.
Collapse
Affiliation(s)
- Meng Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yun Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Huiqiang Huang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yanyan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiwen Huang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Weili Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Pengpeng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengming Chen
- Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY, United States
| | - Jun Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yuqin Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jia Ruan
- Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY, United States
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
26
|
Is There Still a Role for Transplant for Patients with Mantle Cell Lymphoma (MCL) in the Era of CAR-T Cell Therapy? Curr Treat Options Oncol 2022; 23:1614-1625. [PMID: 36227407 PMCID: PMC9557996 DOI: 10.1007/s11864-022-01020-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2022] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT For years, upfront autologous hematopoietic cell transplant (auto-HCT) has been the standard of care for younger and physically fit mantle cell lymphoma (MCL) patients after chemoimmunotherapy (CIT) induction. Bruton's tyrosine kinase (BTK) inhibitors have proven to be excellent salvage therapies, but their durability remains a question, especially in high-risk (HR) MCL. Allogeneic HCT (allo-HCT) was the only option for long-term remission and possibly cure for MCL relapse after auto-HCT and sometime as upfront consolidation for a young patient with HR MCL (debatable). We have seen a paradigm shift since the FDA approval in July 2020 of the brexucabtagene autoleucel chimeric antigen receptor T (CAR-T) cell therapy for relapsed and refractory (R/R) MCL with an preliminary evidence suggesting CAR-T may overcome known biological risk factors in MCL. Given its safety profile and excellent efficacy, the role of CAR-T among other approved therapies and HCT may need to be better defined. Based on the current evidence, auto-HCT remains a standard frontline consolidation therapy. CAR-T therapy is a preferred option for patients with relapsed/refractory (R/R) MCL, particularly those who failed BTK inhibitors. In certain high-risk MCL patients (such as high ki 67, TP53 alterations, complex karyotype, blastoid morphology, early relapse after initial diagnosis), CAR-T cell therapy may be considered before BTK inhibitors (preferably on a clinical trial). The role of allo-HCT is unclear in the CAR-T era, but remains a viable option for eligible patients who have no access or who have failed CAR-T therapy. Our review discusses current standards and the shifting paradigms in the indications for HCT and the role of CAR-T cell therapy for MCL. Prospective studies tailored based on risk factors are needed to better define the optimal sequences of HCT and cellular therapy and other approved novel therapies.
Collapse
|
27
|
Martinez-Baquero D, Sakhdari A, Mo H, Kim DH, Kanagal-Shamanna R, Li S, Young KH, O'Malley DP, Dogan A, Jain P, Wang ML, McDonnell TJ, Miranda RN, Vega F, Medeiros LJ, Ok CY. EZH2 expression is associated with inferior overall survival in mantle cell lymphoma. Mod Pathol 2021; 34:2183-2191. [PMID: 34376807 PMCID: PMC10563799 DOI: 10.1038/s41379-021-00885-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 11/09/2022]
Abstract
Enhancer of zeste homolog 2 (EZH2) is a catalytic component of the polycomb repressive complex 2 (PRC2) which reduces gene expression via trimethylation of a lysine residue of histone 3 (H3K27me3). Expression of EZH2 has not been assessed systematically in mantle cell lymphoma (MCL). Expression of EZH2 was assessed by immunohistochemistry in 166 patients with MCL. We also assessed other PRC2 components and H3K27me3. Fifty-seven (38%) of MCL patients were positive for EZH2 using 40% cutoff. EZH2 expression was associated with aggressive histologic variants (65% vs. 29%, p < 0.001), high Ki-67 proliferation rate (median, 72% vs. 19%, p < 0.001), and p53 overexpression (43% vs. 2%, p < 0.001). EZH2 expression did not correlate with expression of other PRC2 components (EED and SUZ12), H3K27me3, MHC-I, and MHC-II. Patients with EZH2 expression (EZH2+) had a poorer overall survival (OS) compared with patients without EZH2 expression (EZH2-) (median OS: 3.9 years versus 9.4 years, respectively, p < 0.001). EZH2 expression also predicted a poorer prognosis in MCL patients with classic histology (median OS, 4.6 years for EZH2+ and 9.6 years for EZH2-negative, respectively, p < 0.001) as well as aggressive histology (median OS, 3.7 years for EZH2+ and 7.9 years for EZH2-negative, respectively, p = 0.046). However, EZH2 expression did not independently correlate with overall survival in a multivariate analysis. Gene expression analysis and pathway enrichment analysis demonstrated a significant enrichment in cell cycle and mitotic transition pathways in MCL with EZH2 expression. EZH2 expression detected by immunohistochemistry is present in 38% of MCL cases and it is associated with high proliferation rate, p53 overexpression, aggressive histologic variants, and poorer OS. Based on gene expression profiling data, EZH2 expression could potentiate cell cycle machinery in MCL. These data suggest that assessment of EZH2 expression could be useful to stratify MCL patients into low- and high-risk groups.
Collapse
Affiliation(s)
- Diana Martinez-Baquero
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Department of Pathology, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Ali Sakhdari
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Department of Laboratory Medicine and Pathobiology, University Health Network, The University of Toronto, Toronto, ON, Canada
| | - Huan Mo
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Do Hwan Kim
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Shaoying Li
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Ken H Young
- Division of Hematopathology and Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Dennis P O'Malley
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- NeoGenomics, Aliso Viejo, CA, USA
| | - Ahmet Dogan
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Preetesh Jain
- Department of Lymphoma and Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Michael L Wang
- Department of Lymphoma and Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Timothy J McDonnell
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Francisco Vega
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Chi Young Ok
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
28
|
Tashakori M, Kim DH, Kanagal-Shamanna R, Vega F, Miranda RN, Jain P, Wang M, Medeiros LJ, Ok CY. Mantle cell lymphoma involving tonsils: a clinicopathologic study of 83 cases. Hum Pathol 2021; 118:60-68. [PMID: 34655612 DOI: 10.1016/j.humpath.2021.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 11/26/2022]
Abstract
We report 83 cases of mantle cell lymphoma (MCL) involving the tonsil as initial manifestation (IM). The median age at the time of tonsillar involvement was 58 years (range, 35-79 years). Most (85%) patients presented similar to acute tonsillitis. Lymphadenopathy (84%) and advanced stage of disease (81%) were frequent. With a median follow-up of 6.1 years (range, 0.5-18.4 years), the median overall survival (OS) was 11.3 years for all patients. Cases with classic MCL morphology demonstrated a superior OS (median OS: 11.7 years versus 7.8 years for aggressive morphology, P = 0.0361). Approximately 20% of patients had limited stage of disease, and they had excellent outcomes (median OS: not reached versus 11.3 years for advanced-stage MCL, P = 0.0479). All the patients were alive after a median follow-up of 6.6 years (range, 1-16.2 years). There were no differences in relapse-free survival in morphology and stage (P > 0.05). When tonsils were involved by relapsed MCL, patients less commonly had acute tonsillitis-like symptoms, lymphadenopathy, and advanced stage of disease compared to MCL as IM. Patients in the relapse group had poorer OS than patients in the IM group from the time of tonsillar involvement by MCL to the date of death or last follow-up (7.8 versus 11.7 years, P = 0.003). Compared with a group of 93 patients whose initial biopsy specimen was a lymph node, patients whose initial biopsy specimen was tonsil had similar OS (11.7 versus 8.8 years, P = 0.1764). However, patients with tonsillar MCL more commonly had limited stage disease (19% versus 8%, P = 0.0385) and a low-risk Mantle Cell Lymphoma International Prognostic Index score (71% versus 47%, P = 0.0025).
Collapse
Affiliation(s)
- Mehrnoosh Tashakori
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Do Hwan Kim
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Preetesh Jain
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael Wang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chi Young Ok
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
29
|
Valente VB, de Castro TF, Takamiya AS, Callestini R, Xavier-Junior JCC, Cortopassi GM, Neto SC, Collado FU, Biasoli ÉR, Miyahara GI, Bernabé DG. Blastoid variant of mantle cell lymphoma in palatine tonsil. Oral Oncol 2021; 122:105552. [PMID: 34610523 DOI: 10.1016/j.oraloncology.2021.105552] [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: 09/15/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 11/28/2022]
Abstract
Blastoid variant of mantle cell lymphoma (MCL) is an aggressive and extremely rare malignancy. MCL may be diagnosed in lymph nodes and/or extranodal sites exhibiting a poor prognosis. MCL with primary presentation in palatine tonsils has been rarely reported. Herein, we report the case of a 73-year-old man with a painless nodular mass on the right palatine tonsil. A biopsy was performed, and microscopic analysis revealed a neoplasm composed of small to medium sized lymphocytes with finely dispersed chromatin, roundish nucleus and many mitoses. The tumor cells were positive for CD20 (L26), CD5 (4C7), Cyclin D1 (EP12), Bcl2 (124) and Ki-67 (MIB-1; 90%), and negative for Bcl6 (PG-B6p), MUM1 (MUM1p) and CD3 (Polyclonal). These findings led to the diagnosis of blastoid variant of MCL. Diagnostic workup with computed tomography scan excluded other sites of disease. The patient was treated successfully with cyclophosphamide, doxorubicin, vincristine and prednisolone (mini-CHOP regimen). Although the blastoid variant of MCL is rare, it should be included in the differential diagnosis of rapid-growing masses in the palatine tonsil.
Collapse
Affiliation(s)
- Vitor Bonetti Valente
- Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil
| | - Tamara Fernandes de Castro
- Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil
| | - Aline Satie Takamiya
- Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil
| | - Renata Callestini
- Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil
| | - José Cândido Caldeira Xavier-Junior
- Pathology Institute of Araçatuba, Araçatuba, São Paulo, Brazil; School of Medicine, Centro Universitário Católico Unisalesiano Auxilium, Araçatuba, São Paulo, Brazil
| | | | - Sebastião Conrado Neto
- Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil
| | - Francisco Urbano Collado
- Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil
| | - Éder Ricardo Biasoli
- Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil; Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil
| | - Glauco Issamu Miyahara
- Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil; Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil
| | - Daniel Galera Bernabé
- Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil; Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, 1193 José Bonifácio St, SP 15050-015 Araçatuba, São Paulo, Brazil.
| |
Collapse
|
30
|
Takiar R, Phillips T. Non-chemotherapy Options for Newly Diagnosed Mantle Cell Lymphoma. Curr Treat Options Oncol 2021; 22:98. [PMID: 34524546 DOI: 10.1007/s11864-021-00900-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2021] [Indexed: 12/29/2022]
Abstract
OPINION STATEMENT Mantle cell lymphoma is a rare and incurable non-Hodgkin lymphoma with a heterogenous clinical presentation. Typically, treatment consists of frontline chemoimmunotherapy induction with or without autologous stem cell transplant (ASCT) as consolidation. However, this approach has the propensity to increase short- and long-term toxicities, such as secondary malignancies, without being curative. Genomic profiling of MCL will allow for greater impact of new targeted therapies in the future and may become a helpful tool to guide treatment. Based on the data discussed, use of non-chemotherapy options may become the preferred approach for frontline therapy as opposed to conventional chemotherapy and hematopoietic stem cell transplants.
Collapse
Affiliation(s)
- Radhika Takiar
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Michigan Medicine, 1500 East Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Tycel Phillips
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Michigan Medicine, 1500 East Medical Center Drive, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
31
|
Wullenkord R, Berning P, Niemann AL, Wethmar K, Bergmann S, Lutz M, Schliemann C, Mesters R, Keßler T, Schmitz N, Berdel WE, Lenz G, Stelljes M. The role of autologous stem cell transplantation (ASCT) in aggressive B-cell lymphomas: real-world data from a retrospective single-center analysis. Ann Hematol 2021; 100:2733-2744. [PMID: 34477953 PMCID: PMC8510902 DOI: 10.1007/s00277-021-04650-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/19/2021] [Indexed: 12/01/2022]
Abstract
Patients with high-risk or relapsed aggressive B-cell lymphomas are characterized by poor prognosis. High-dose chemotherapy followed by autologous stem cell transplantation (ASCT) can induce durable remissions in these patients and is potentially curative. Two hundred forty-seven patients with aggressive B-cell lymphomas treated with high-dose chemotherapy and ASCT, either as consolidation after first-line therapy or after salvage therapy for relapsed disease, between 2002 and 2019 at the University Hospital Muenster, were analyzed. The median follow-up of surviving patients was 36 months (range 0–163). Progression-free survival (PFS) and overall survival (OS) after 3 years was 63% and 68%, respectively. After ASCT, 28% of all patients experienced a relapse. The cumulative incidence of non-relapse mortality at day 100 after ASCT was 4%. Multivariate analysis identified remission status at ASCT, age at ASCT, and the numbers of infused CD34+ cells as independent prognostic factors for both PFS and OS. Patients with mantle cell lymphoma (MCL) or primary CNS lymphoma (PCNSL) treated with ASCT in first-line had a superior OS and PFS when compared to patients treated with ASCT in relapsed disease. For patients with diffuse large B-cell lymphoma (DLBCL) and Hodgkin lymphoma (HL), early relapse (< 12 months) after first-line therapy showed a trend towards an inferior PFS and OS. Deaths after ASCT were predominantly caused by lymphoma relapse and/or progression (64%) or due to infections (23%). In conclusion, high-dose chemotherapy followed by ASCT in the era of novel targeted agents remains a feasible and effective approach for patients with high-risk or relapsed aggressive B-cell lymphomas. Remission status and age at ASCT, and the number of infused stem cells were of prognostic relevance.
Collapse
Affiliation(s)
- Ramona Wullenkord
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Philipp Berning
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Anna-Lena Niemann
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Klaus Wethmar
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Sarah Bergmann
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Mathias Lutz
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Christoph Schliemann
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Rolf Mesters
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Torsten Keßler
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Norbert Schmitz
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Matthias Stelljes
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
32
|
Munshi PN, Hamadani M, Kumar A, Dreger P, Friedberg JW, Dreyling M, Kahl B, Jerkeman M, Kharfan-Dabaja MA, Locke FL, Shadman M, Hill BT, Ahmed S, Herrera AF, Sauter CS, Bachanova V, Ghosh N, Lunning M, Kenkre VP, Aljurf M, Wang M, Maddocks KJ, Leonard JP, Kamdar M, Phillips T, Cashen AF, Inwards DJ, Sureda A, Cohen JB, Smith SM, Carlo-Stella C, Savani B, Robinson SP, Fenske TS. American Society of Transplantation and Cellular Therapy, Center of International Blood and Marrow Transplant Research, and European Society for Blood and Marrow Transplantation Clinical Practice Recommendations for Transplantation and Cellular Therapies in Mantle Cell Lymphoma. Transplant Cell Ther 2021; 27:720-728. [PMID: 34452722 PMCID: PMC8447221 DOI: 10.1016/j.jtct.2021.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 11/18/2022]
Abstract
Autologous (auto-) and allogeneic (allo-) hematopoietic cell transplantation (HCT) are accepted treatment modalities in contemporary treatment algorithms for mantle cell lymphoma (MCL). Chimeric antigen receptor (CAR) T cell therapy recently received approval for MCL; however, its exact place and sequence in relation to HCT remain unclear. The American Society of Transplantation and Cellular Therapy, Center of International Blood and Marrow Transplant Research, and the European Society for Blood and Marrow Transplantation jointly convened an expert panel to formulate consensus recommendations for role, timing, and sequencing of auto-HCT, allo-HCT, and CAR T cell therapy for patients with newly diagnosed and relapsed/refractory (R/R) MCL. The RAND-modified Delphi method was used to generate consensus statements. Seventeen consensus statements were generated, with a few key statements as follows: in the first line setting, auto-HCT consolidation represents standard of care in eligible patients, whereas there is no clear role of allo-HCT or CAR T cell therapy outside of clinical trials. In the R/R setting, the preferential option is CAR T cell therapy, especially in patients with MCL failing or intolerant to at least one Bruton's tyrosine kinase inhibitor, while allo-HCT is recommended if CAR T cell therapy fails or is infeasible. Several recommendations were based on expert opinion, where the panel developed consensus statements for important real-world clinical scenarios to guide clinical practice. In the absence of contemporary evidence-based data, the panel found RAND-modified Delphi methodology effective in providing a formal framework for developing consensus recommendations for the timing and sequence of cellular therapies for MCL.
Collapse
Affiliation(s)
- Pashna N Munshi
- Department of Blood and Marrow Transplantation, MedStar Georgetown University Hospital, Washington, DC
| | - Mehdi Hamadani
- Center for International Blood & Marrow Transplant Research and the BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - Ambuj Kumar
- Department of Internal Medicine, Office of Research, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Peter Dreger
- Department of Medicine, University of Heidelberg, Germany
| | | | - Martin Dreyling
- Department of Medicine III, LMU Hospital Munich, Munich, Germany
| | - Brad Kahl
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Mats Jerkeman
- Department of Oncology, Skane University Hospital, Lund University, Lund, Sweden
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapies Program, Mayo Clinic, Jacksonville, Florida
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Mazyar Shadman
- Blood and Marrow Transplantation Program, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Sairah Ahmed
- Department of Lymphoma, Myeloma and Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Craig S Sauter
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Nilanjan Ghosh
- Levine Cancer Institute, Atrium Health, Charlotte, Nnorth Carolina
| | - Matthew Lunning
- Divsion of Hematology and Oncology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Vaishalee P Kenkre
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mahmoud Aljurf
- Blood and Marrow Transplantation Program, King Faisal Specialist Hospital, Riyadh, Saudi Arabia
| | - Michael Wang
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kami J Maddocks
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - John P Leonard
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Manali Kamdar
- Division of Hematology, Hematologic Malignancies and Stem Cell Transplantation, University of Colorado Cancer Center, Denver, Colorado
| | - Tycel Phillips
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Amanda F Cashen
- Division of Oncology, Section of Stem Cell Transplantation, Washington University School of Medicine, St Louis, Missouri
| | | | - Anna Sureda
- Clinical Hematology Department, Catalan Institute of Oncology, Institut d'Investigació Biomèdica de Bellvitge IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | | | - Sonali M Smith
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Carmello Carlo-Stella
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milan, Italy
| | - Bipin Savani
- Blood and Marrow Transplantation Program, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Stephen P Robinson
- University Hospital Bristol NHS Foundation Trust, London, United Kingdom
| | - Timothy S Fenske
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
33
|
Zoellner AK, Unterhalt M, Stilgenbauer S, Hübel K, Thieblemont C, Metzner B, Topp M, Truemper L, Schmidt C, Bouabdallah K, Krauter J, Lenz G, Dürig J, Vergote V, Schäfer-Eckart K, André M, Kluin-Nelemans HC, van Hoof A, Klapper W, Hiddemann W, Dreyling M, Hoster E. Long-term survival of patients with mantle cell lymphoma after autologous haematopoietic stem-cell transplantation in first remission: a post-hoc analysis of an open-label, multicentre, randomised, phase 3 trial. LANCET HAEMATOLOGY 2021; 8:e648-e657. [PMID: 34450102 DOI: 10.1016/s2352-3026(21)00195-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Autologous haematopoietic stem-cell transplantation (HSCT) in first remission is the current standard treatment in fit patients with mantle cell lymphoma. In this long-term follow-up study, we aimed to evaluate the efficacy of autologous HSCT versus interferon alfa maintenance after chemotherapy without or with rituximab in patients with primary advanced-stage mantle cell lymphoma. METHODS We did a post-hoc, long-term analysis of an open-label, multicentre, randomised, phase 3 trial done in 121 participating hospitals or practices across six European countries. Patients who were aged 18-65 years with previously untreated stage III-IV mantle cell lymphoma and an ECOG performance score of 0-2 were eligible for participation. Patients were randomly assigned (1:1) to receive either myeloablative radiochemotherapy (fractionated total body irradiation with 12 Gy/day 6-4 days before autologous HSCT and cyclophosphamide 60 mg/kg per day intravenously 3-2 days before autologous HSCT) followed by autologous HSCT (the autologous HSCT group) or interferon alfa maintenance (the interferon alfa maintenance group; 6 × 106 IU three times a week subcutaneously until progression) after completion of CHOP-like induction therapy (cyclophosphamide 750 mg/m2 intravenously on day 1, doxorubicin 50 mg/m2 intravenously on day 1, vincristine 1·4 mg/m2 [maximum 2 mg] intravenously on day 1, and prednisone 100 mg/m2 orally on days 1-5; repeated every 21 days for up to 6 cycles) without or with rituximab (375 mg/m2 intravenously on day 0 or 1 of each cycle; R-CHOP). The primary outcome was progression-free survival from end of induction until progression or death among patients who had a remission and the secondary outcome was overall survival from the end of induction until death from any cause. We did comparisons of progression-free survival and overall survival according to the intention-to-treat principle between both groups among responding patients and explored efficacy in subgroups according to induction treatment without or with rituximab. Hazard ratios (HRs) were adjusted for the mantle cell lymphoma international prognostic index (MIPI) numerical score, and in the total group also for rituximab use (adjusted HR [aHR]). This trial was started before preregistration was implemented and is therefore not registered, recruitment is closed, and this is the final evaluation. FINDINGS Between Sept 30, 1996, and July 1, 2004, 269 patients were randomly assigned to receive either autologous HSCT or interferon alfa maintenance therapy. The median follow-up was 14 years (IQR 10-16), with the intention-to-treat population consisting of 174 patients (93 [53%] in the autologous HSCT group and 81 [47%] in the interferon alfa maintenance group) who responded to induction therapy. The median age was 55 years (IQR 47-60), and R-CHOP was used in 68 (39%) of 174 patients. The median progression-free survival was 3·3 years (95% CI 2·5-4·3) in the autologous HSCT group versus 1·5 years (1·2-2·0) in the interferon alfa maintenance group (log-rank p<0·0001; aHR 0·50 [95% CI 0·36-0·69]). The median overall survival was 7·5 years (95% CI 5·7-12·0) in the autologous HSCT group versus 4·8 years (4·0-6·6) in the interferon alfa maintenance group (log-rank p=0·019; aHR 0·66 [95% CI 0·46-0·95]). For patients treated without rituximab, the progression-free survival adjusted HR for autologous HSCT versus interferon alfa was 0·40 (0·26-0·61), in comparison to 0·72 (0·42-1·24) for patients treated with rituximab. For overall survival, the adjusted hazard ratio for HSCT versus interferon alfa was 0·52 (0·33-0·82) without rituximab and 1·05 (0·55-1·99) for patients who received rituximab. INTERPRETATION Our results confirm the long-term efficacy of autologous HSCT to treat mantle cell lymphoma established in the pre-rituximab era. The suggested reduced efficacy after immunochemotherapy supports the need for its re-evaluation now that antibody maintenance, high-dose cytarabine, and targeted treatments have changed the standard of care for patients with mantle cell lymphoma. FUNDING Deutsche Krebshilfe, the European Community, and the Bundesministerium für Bildung und Forschung, Kompetenznetz Maligne Lymphome.
Collapse
Affiliation(s)
- Anna-Katharina Zoellner
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Michael Unterhalt
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine I, Saarland University Medical Center, Homburg, Germany; Department of Internal Medicine III, Ulm University, Ulm, Germany
| | - Kai Hübel
- Department of Medicine I, University Hospital of Cologne, Cologne, Germany
| | | | - Bernd Metzner
- Department of Hematology and Oncology, University Hospital Oldenburg, Oldenburg, Germany
| | - Max Topp
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Lorenz Truemper
- Department of Hematology and Oncology, Georg August University, Goettingen, Germany
| | - Christian Schmidt
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Kamal Bouabdallah
- Department of Hematology and Cell Therapy, Haut-Leveque Hospital, Bordeaux, France
| | - Jürgen Krauter
- Department of Hematology and Oncology, Klinikum Braunschweig, Braunschweig, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology and Oncology, University of Münster, Münster, Germany
| | - Jan Dürig
- Department of Hematology, University Medicine Essen, Essen, Germany
| | - Vibeke Vergote
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Kerstin Schäfer-Eckart
- Klinik für Innere Medizin 5, Klinikum Nürnberg, Paracelsus Medizinische Privatuniversität, Nürnberg, Germany
| | - Marc André
- Department of Hematology, CHU UCLouvain Namur, Yvoir, Belgium
| | | | - Achiel van Hoof
- Department of Hematology, A Z St-Jan, Brugge-Oostende, Belgium
| | - Wolfram Klapper
- Hematopathology Section, Christian-Albrechts University, Kiel, Germany
| | - Wolfgang Hiddemann
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Martin Dreyling
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany.
| | - Eva Hoster
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Institute of Medical Information Processing, Biometry and Epidemiology, Ludwig Maximilian University of Munich, Munich, Germany
| | | |
Collapse
|
34
|
Munshi PN, Hamadani M, Kumar A, Dreger P, Friedberg JW, Dreyling M, Kahl B, Jerkeman M, Kharfan-Dabaja MA, Locke FL, Shadman M, Hill BT, Ahmed S, Herrera AF, Sauter CS, Bachanova V, Ghosh N, Lunning M, Kenkre VP, Aljurf M, Wang M, Maddocks KJ, Leonard JP, Kamdar M, Phillips T, Cashen AF, Inwards DJ, Sureda A, Cohen JB, Smith SM, Carlo-Stella C, Savani B, Robinson SP, Fenske TS. ASTCT, CIBMTR, and EBMT clinical practice recommendations for transplant and cellular therapies in mantle cell lymphoma. Bone Marrow Transplant 2021; 56:2911-2921. [PMID: 34413469 PMCID: PMC8639670 DOI: 10.1038/s41409-021-01288-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 11/09/2022]
Abstract
Autologous (auto-) or allogeneic (allo-) hematopoietic cell transplantation (HCT) are accepted treatment modalities for mantle cell lymphoma (MCL). Recently, chimeric antigen receptor (CAR) T-cell therapy received approval for MCL; however, its exact place and sequence in relation to HCT is unclear. The ASTCT, CIBMTR, and the EBMT, jointly convened an expert panel to formulate consensus recommendations for role, timing, and sequencing of auto-, allo-HCT, and CAR T-cell therapy for patients with newly diagnosed and relapsed/refractory (R/R) MCL. The RAND-modified Delphi method was used to generate consensus statements. Seventeen consensus statements were generated; in the first-line setting auto-HCT consolidation represents standard-of-care in eligible patients, whereas there is no clear role of allo-HCT or CAR T-cell therapy, outside of a clinical trial. In the R/R setting, the preferential option is CAR T-cell therapy especially in MCL failing or intolerant to at least one Bruton's tyrosine kinase inhibitor, while allo-HCT is recommended if CAR T-cell therapy has failed or is not feasible. In the absence of contemporary evidence-based data, the panel found RAND-modified Delphi methodology effective in providing a formal framework for developing consensus recommendations for the timing and sequence of cellular therapies for MCL.
Collapse
Affiliation(s)
| | - Mehdi Hamadani
- CIBMTR & BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Ambuj Kumar
- Department of Internal Medicine, Office of Research, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | | | - Martin Dreyling
- Department of Medicine III, LMU Hospital Munich, Munich, Germany
| | - Brad Kahl
- Washington University School of Medicine, St. Louis, MO, USA
| | - Mats Jerkeman
- Department of Oncology, Skane University Hospital, Lund University, Lund, Sweden
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapies Program, Mayo Clinic, Jacksonville, FL, USA
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Mazyar Shadman
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA, USA
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sairah Ahmed
- Department of Lymphoma, Myeloma and Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, USA
| | - Craig S Sauter
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Nilanjan Ghosh
- Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | | | - Vaishalee P Kenkre
- University of Wisconsin, Division of Hematology and Oncology, Madison, WI, USA
| | | | - Michael Wang
- Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kami J Maddocks
- Division of Hematology, Ohio State University, Columbus, OH, USA
| | | | - Manali Kamdar
- Division of Hematology, Hematologic Malignancies and Stem Cell Transplantation, University of Colorado Cancer Center, Denver, CO, USA
| | - Tycel Phillips
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Amanda F Cashen
- Division of Oncology, Section of Stem Cell Transplantation, Washington University School of Medicine, St Louis, MO, USA
| | - David J Inwards
- Division of Hematology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Anna Sureda
- Clinical Hematology Department, Catalan Institute of Oncology, Institut d'Investigació Biomèdica de Bellvitge IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | | | - Sonali M Smith
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Carmello Carlo-Stella
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milan, Italy
| | - Bipin Savani
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Timothy S Fenske
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
35
|
Vorobyev VI, Gemdzhian EG, Fedorova LV, Mikhailova NB, Ilyasov RK, Kaleikina LP, Trubyakova OS, Kaplanov KD, Melnichenko EV, Martynova EV, Yakovleva EP, Li OY, Tarasenko EV, Chumakova EP, Bulieva NB, Nesterova ES, Margolin OV, Zherebtsova VA, Butaev LS, Ptushkin VV. Five year experience in ibrutinib therapy for relapsed and refractory mantle cell lymphoma in real world Russian clinical practice. TERAPEVT ARKH 2021; 93:770-777. [DOI: 10.26442/00403660.2021.07.200930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
Background. Mantle cell lymphoma (MCL) is a rare and clinically aggressive lymphoma subtype. Current approaches have greatly improved patients outcomes, but relapse is inevitable. In phase IIIII clinical trials, ibrutinib has shown significant activity in patients with relapsed or refractory (R/R) MCL.
Aim. To assess efficacy and toxicity of ibrutinib monotherapy in patients with R/R MCL in routine practice outside of clinical trials.
Materials and methods. The study enrolled patients with confirmed R/R MCL who had received at least one line of previous chemotherapy. ECOG 24, cytopenia, infectious complications, hemorrhagic syndrome were not exclusion criteria. Patients received daily oral ibrutinib 560 mg until progression or unacceptable toxicity.
Results. From May 2015 to September 2020 ibrutinib therapy was started in 106 patients with R/R MCL in 16 regions of Russia. The median age was 66 years; ECOG2 18%, blastoid variant (or Ki6740% or WBC50109/l) 43%. The median number of previous treatment lines was 2 (111). The ORR was 78.4% (CRR 27.4%). The median PFS was 13.6 months and OS 23.2 months. In the blastoid group the median PFS was 4.4 months vs 36.5 months in the alternative group (p0.001), the median OS 9.0 vs 41.0 (p=0.001). The median OS of patients after progression on ibrutinib was 3.2 months.
The common complications are hemorrhages (63%), diarrhea (62%), myalgia and muscle cramps (60%), infections (31%), skin and nail toxicity 15%, arrhythmia 8%. None of recipients had to completely discontinue ibrutinib therapy due to complications.
Conclusion. Ibrutinib is effective and well tolerated in routine practice of R/R MCL treatment and our results are consistent with international clinical trials. The favorable toxicity profile and the high response rate made it possible to prescribe ibrutinib in severe somatic status, cytopenia, and even in the presence of infectious complications.
Collapse
|
36
|
Goy A. Exploiting gene mutations and biomarkers to guide treatment recommendations in mantle cell lymphoma. Expert Rev Hematol 2021; 14:927-943. [PMID: 34253131 DOI: 10.1080/17474086.2021.1950529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION While there has been an improvement in the treatment of mantle cell lymphoma (MCL) in both median progression-free survival (PFS; >7-8 years) and overall survival (OS; >10-12 years), patients with high-risk features such as high risk MIPI (mantle cell international prognostic index), high Ki-67 (≥30%), or blastoid variants still carry poor outcome with a median OS of 3 years. Furthermore, patients with high-risk molecular features, such as TP53 mutations, show dismal outcome, with a median OS of 1.8 years, regardless of therapy used. Further studies have led to the development of six novel drugs approved for the treatment of relapse/refractory (R/R) MCL, leading to improved survival even in refractory or high-risk patients. AREAS COVERED This review covers clinical biological and molecular features that impact MCL outcome with current standards. Beyond the recognition of separate subentities, we review how high-risk molecular features have paved the way towards a new paradigm away from chemoimmunotherapy. EXPERT OPINION Progress in novel therapies and in routine diagnostics, particularly next-generation sequencing (NGS), support the development of new treatment strategies, not based on the dose intensity/age dichotomy, which may prevent the need for chemotherapy and improve outcome across MCL including in high-risk subsets.
Collapse
Affiliation(s)
- Andre Goy
- John Theurer Cancer Center, Hackensack University Medical Center, NJ
| |
Collapse
|
37
|
Expression patterns and prognostic potential of circular RNAs in mantle cell lymphoma: a study of younger patients from the MCL2 and MCL3 clinical trials. Leukemia 2021; 36:177-188. [PMID: 34244612 DOI: 10.1038/s41375-021-01311-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/19/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022]
Abstract
Mantle cell lymphoma (MCL) is characterized by marked differences in outcome, emphasizing the need for strong prognostic biomarkers. Here, we explore expression patterns and prognostic relevance of circular RNAs (circRNAs), a group of endogenous non-coding RNA molecules, in MCL. We profiled the circRNA expression landscape using RNA-sequencing and explored the prognostic potential of 40 abundant circRNAs in samples from the Nordic MCL2 and MCL3 clinical trials, using NanoString nCounter Technology. We report a circRNA-based signature (circSCORE) developed in the training cohort MCL2 that is highly predictive of time to progression (TTP) and lymphoma-specific survival (LSS). The dismal outcome observed in the large proportion of patients assigned to the circSCORE high-risk group was confirmed in the independent validation cohort MCL3, both in terms of TTP (HR 3.0; P = 0.0004) and LSS (HR 3.6; P = 0.001). In Cox multiple regression analysis incorporating MIPI, Ki67 index, blastoid morphology and presence of TP53 mutations, circSCORE retained prognostic significance for TTP (HR 3.2; P = 0.01) and LSS (HR 4.6; P = 0.01). In conclusion, circRNAs are promising prognostic biomarkers in MCL and circSCORE improves identification of high-risk disease among younger patients treated with cytarabine-containing chemoimmunotherapy and autologous stem cell transplant.
Collapse
|
38
|
Alderuccio JP, Saul EE, Iyer SG, Reis IM, Alencar AJ, Rosenblatt JD, Lossos IS. R-MACLO-IVAM regimen followed by maintenance therapy induces durable remissions in untreated mantle cell lymphoma - Long term follow up results. Am J Hematol 2021; 96:680-689. [PMID: 33735476 DOI: 10.1002/ajh.26163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/27/2021] [Accepted: 03/15/2021] [Indexed: 11/06/2022]
Abstract
We present long-term combined results of two clinical trials implementing R-MACLO-IVAM induction followed by thalidomide or rituximab maintenance in 44 patients with untreated mantle cell lymphoma (MCL). The first 22 patients (UM-MCL1 ClinicalTrials.gov identifier NCT00450801) received maintenance with thalidomide (200 mg daily until relapse/intolerable toxicity) and a subsequent cohort of 22 patients (UM-MCL2 ClinicalTrials.gov identifier NCT00878254) received rituximab (375 mg/m2 IV weekly × 4, repeated every 6 months for 3 years). Considering all 44 patients, 41 (93.2%) achieved complete response (CR), two (4.5%) partial response (PR), and one (2.3%) was not evaluated for response. With a median follow up of 7.2 years (range < 1 month to 16 years), the 5-year progression-free survival (PFS) was 55.6% (95% CI: 38.9%-69.4%) and median PFS 7.9 years (95% CI: 3.7-11 years). The 5-year OS was 83.3% (95% CI: 68.1%-91.7%) and median OS was not reached. Patients with blastic variant (n = 6) had a 5-year PFS and OS of 20.8% and 60%, respectively. Myelosuppression was the most common adverse event during immunochemotherapy. Long-term treatment-related mortality was 6.8%. Note, R-MACLO-IVAM followed by maintenance therapy is an effective regimen to induce long-term remission in MCL without need for consolidation with ASCT.
Collapse
Affiliation(s)
- Juan P. Alderuccio
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Eduardo E. Saul
- Department of Medicine, Division of Internal Medicine, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Sunil G. Iyer
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Isildinha M. Reis
- Department of Public Health Science, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
- Sylvester Biostatistics and Bioinformatics Shared Resource, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Alvaro J. Alencar
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Joseph D. Rosenblatt
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| | - Izidore S. Lossos
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
- Department of Molecular and Cellular Pharmacology, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
39
|
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
|
40
|
Novel Treatments for Mantle Cell Lymphoma: From Targeted Therapies to CAR T Cells. Drugs 2021; 81:669-684. [PMID: 33783717 DOI: 10.1007/s40265-021-01497-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2021] [Indexed: 10/21/2022]
Abstract
Mantle cell lymphoma is a rare B-cell non-Hodgkin's lymphoma that retains a sobering prognosis despite an extensive research effort. Mantle cell lymphoma remains incurable even with aggressive, and at times toxic, chemoimmunotherapy with early incorporation of autologous stem cell transplantation. Given this, attention has turned to the use of targeted therapies addressing dysregulation of B-cell signaling pathways. Drugs such as immunomodulatory agents, proteasome inhibitors, and Bruton's tyrosine kinase inhibitors have shown success in the relapsed/refractory population, and there is ongoing investigation into the utilization of novel Bruton's tyrosine kinase, B-cell leukemia/lymphoma-2, and spleen tyrosine kinase inhibitors alone or in combination in both the front-line and relapsed settings. Other areas of research in novel immunotherapies include investigations of bispecific T-cell engagers and antibody-drug conjugates. Most recently, chimeric antigen receptor T-cell therapy has been granted US Food and Drug Administration approval as a result of durable remissions even in high-risk patients who have classically done poorly with traditional chemoimmunotherapy. The intent of this article is to review the literature describing these selective therapies and discuss their current and future roles in the treatment of mantle cell lymphoma.
Collapse
|
41
|
Glimelius I, Smedby KE, Albertsson-Lindblad A, Crowther MJ, Eloranta S, Jerkeman M, Weibull CE. Unmarried or less-educated patients with mantle cell lymphoma are less likely to undergo a transplant, leading to lower survival. Blood Adv 2021; 5:1638-1647. [PMID: 33710334 PMCID: PMC7993102 DOI: 10.1182/bloodadvances.2020003645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022] Open
Abstract
It is unknown how many mantle cell lymphoma (MCL) patients undergo consolidation with autologous hematopoietic cell transplantation (AHCT), and the reasons governing the decision, are also unknown. The prognostic impact of omitting AHCT is also understudied. We identified all MCL patients diagnosed from 2000 to 2014, aged 18 to 65 years, in the Swedish Lymphoma Register. Odds ratios (ORs) and 95% confidence intervals (CIs) from logistic regression models were used to compare the likelihood of AHCT within 18 months of diagnosis. All-cause mortality was compared between patients treated with/without AHCT using hazard ratios (HRs) and 95% CIs estimated from Cox regression models. Probabilities of being in each of the following states: alive without AHCT, alive with AHCT, dead before AHCT, and dead after AHCT, were estimated over time from an illness-death model. Among 369 patients, 148 (40%) were not treated with AHCT within 18 months. Compared with married patients, never married and divorced patients had lower likelihood of undergoing AHCT, as had patients with lower educational level, and comorbid patients. Receiving AHCT was associated with reduced all-cause mortality (HR = 0.58, 95% CI: 0.40-0.85). Transplantation-related mortality was low (2%). MCL patients not receiving an AHCT had an increased mortality rate, and furthermore, an undue concern about performing an AHCT in certain societal groups was seen. Improvements in supportive functions potentially increasing the likelihood of tolerating an AHCT and introduction of more tolerable treatments for these groups are needed.
Collapse
Affiliation(s)
- Ingrid Glimelius
- Department of Immunology, Genetics, and Pathology, Clinical and Experimental Oncology, Uppsala University, Uppsala Akademiska Hospital, Uppsala, Sweden
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Karin E Smedby
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Division of Hematology, Department of Medicine Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Michael J Crowther
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom; and
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Eloranta
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Mats Jerkeman
- Department of Oncology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Caroline E Weibull
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
Modified conditioning regimen with idarubicin followed by autologous hematopoietic stem cell transplantation for invasive B-cell non-Hodgkin's lymphoma patients. Sci Rep 2021; 11:4273. [PMID: 33608570 PMCID: PMC7895978 DOI: 10.1038/s41598-021-81944-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 12/28/2020] [Indexed: 12/20/2022] Open
Abstract
High-dose chemotherapy followed by autologous hematopoietic stem cell transplantation (ASCT) is still a consolidation treatment choice for relapsed/refractory B-cell non-Hodgkin’s lymphoma (NHL) patients and some aggressive B-cell NHL as frontline therapy. Due to the shortage of carmustine, we switched to idarubicin-substituted BEAC (IEAC) conditioning regimen. We retrospectively compared the outcomes of 72 aggressive B-cell NHL patients treated with IEAC or BEAC regimens followed by ASCT as upfront consolidative treatment. The median time to neutrophil and platelet reconstitution showed no difference between IEAC and BEAC groups. IEAC regimen was well tolerated without increase of adverse events. Transplant-related mortality didn’t occur. The overall survival (OS) and progression-free survival (PFS) of IEAC group (33 and 23 months) were a little longer than that of BEAC group (30 and 18 months). However, due to the small sample numbers, there’s no significant difference in OS and PFS between IEAC and BEAC group with DLBCL or MCL. Multivariate analysis showed that AnnArbor staging, IPI score, lactate dehydrogenase level, remission of disease, modified regimen were related with PFS and OS. In conclusion, IEAC regimen was well tolerated and replacement with idarubicin could be an alternative when carmustine was not available.
Collapse
|
43
|
Monga N, Tam C, Garside J, Davids MS, Ward K, Quigley J, Parisi L, Tapprich C. Clinical efficacy and safety of first-line treatments in patients with mantle cell lymphoma: A systematic literature review. Crit Rev Oncol Hematol 2020; 158:103212. [PMID: 33383209 DOI: 10.1016/j.critrevonc.2020.103212] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/31/2020] [Accepted: 12/20/2020] [Indexed: 11/24/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a rare form of non-Hodgkin's lymphoma (NHL) with a median overall survival (OS) of approximately 3-5 years. Systematic literature reviews (SLRs) identified efficacy and safety data for first-line therapies, reported in randomised controlled trials (RCTs) and non-randomised interventional studies (NRISs). Nine and 20 independent studies were included in the RCT and NRISs SLRs, respectively. Differences in the regimens and patient outcomes varied according to patient age and suitability for autologous stem cell transplantation (ASCT). In elderly patients ineligible for transplant, OS ranged from 40 months to 69.6 months. In young transplant-eligible patients, OS ranged from 53 months to 152.4 months. Despite the paucity of directly comparable evidence on the efficacy and safety of MCL therapies, these SLRs highlight that MCL remains a difficult NHL subtype to treat, with short survival highlighting the unmet need for newer treatments that improve patient outcomes.
Collapse
Affiliation(s)
| | - Constantine Tam
- St Vincent's Hospital, Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, Australia
| | | | | | - Katherine Ward
- ICON Global Health Economics and Outcomes Research, Abingdon, UK
| | - Joan Quigley
- ICON Global Health Economics and Outcomes Research, Abingdon, UK
| | | | | |
Collapse
|
44
|
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
|
45
|
Lokhande L, Kuci Emruli V, Kolstad A, Hutchings M, Räty R, Jerkeman M, Ek S. Immune-related protein signature in serum stratify relapsed mantle cell lymphoma patients based on risk. BMC Cancer 2020; 20:1202. [PMID: 33287742 PMCID: PMC7720632 DOI: 10.1186/s12885-020-07678-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022] Open
Abstract
Background Response to modern treatment strategies, which combine cytotoxic compounds with immune stimulatory agents and targeted treatment is highly variable among MCL patients. Thus, providing prognostic and predictive markers for risk adapted therapy is warranted and molecular information that can help in patient stratification is a necessity. In relapsed MCL, biopsies are rarely available and molecular information from tumor tissue is often lacking. Today, the main tool to access risk is the MCL international prognostic index (MIPI), which does not include detailed biological information of relevance for different treatment options. To enable continuous monitoring of patients, non-invasive companion diagnostic tools are needed which can further reduce cost and patient distress and enable efficient measurements of biological markers. Methods We have assessed if serum-based protein profiling can identify immune related proteins that stratify relapsed MCL patients based on risk. Overall, 371 scFv targeting 158 proteins were assessed using an antibody microarray platform. We profiled patients (n = 44) who had been treated within the MCL6-Philemon trial combining targeted and immune-modulatory treatment. Results The downstream processing led to the identification of the relapsed immune signature (RIS) consisting of 11 proteins with potential to stratify patients with long and short overall survival (OS). Moreover, in this population, MIPI alone failed to separate high, intermediate and low risk patients, but a combined index based on MIPI together with RIS, MIPIris, showed improved performance and significantly stratified all three risk groups based on OS. Conclusions Our results show that addition of biological parameters to previous prognostic indices improves patient stratification among patients treated with BTK inhibitor triplet combination, particularly, in the identification of an extreme high risk group. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-020-07678-4.
Collapse
Affiliation(s)
| | | | | | | | - Riikka Räty
- Department of Hematology, Helsinki University Central Hospital, Helsinki, Finland
| | - Mats Jerkeman
- Department of Oncology, Lund University, Lund, Sweden
| | - Sara Ek
- Department of Immunotechnology, Lund University, Lund, Sweden.
| |
Collapse
|
46
|
Marques JAS, Ferreira F, Melo DP, Santos M, Vaz RP. Palatine Tonsils Primary Presentation of Blastoid Variant of Mantle Cell Lymphoma: Case Report. Head Neck Pathol 2020; 15:588-592. [PMID: 33091144 PMCID: PMC8134596 DOI: 10.1007/s12105-020-01238-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022]
Abstract
Head and neck lymphomas can present with a wide range of symptoms. Timely and accurate diagnosis is often challenging. The blastoid variant of mantle cell lymphoma (MCL) accounts for less than one-third of all MCL cases. Isolated primary presentation on the palatine tonsils is rare, and prognosis and outcome are seemingly unfavorable. An 81-year-old man presented with persistent odynophagia, dysphagia, and obstructive hypertrophic palatine tonsils with purulent exudate. The signs and symptoms were non-responsive to antibiotic therapy, and the tonsils were biopsied. The cellular morphology, immunophenotype, and genotype supported a diagnosis of the blastoid variant of MCL. After staging, the patient underwent chemotherapy with Rituximab-Bendamustine (R-Benda). The patient is in clinical remission more than two years after therapy. We report an exceedingly rare case of blastoid MCL that is prone to be misdiagnosed as tonsillitis. We review the literature and discuss treatment options of this uncommon malignancy.
Collapse
Affiliation(s)
- Joana A. S. Marques
- Department of Otorhinolaryngology, Centro Hospitalar Universitário S. João, EPE, Porto, Portugal ,Unit of Otorhinolaryngology - Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Fátima Ferreira
- Department of Clinical Haematology, Centro Hospitalar Universitário S. João, EPE, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Daniel P. Melo
- Department of Anatomic Pathology, Centro Hospitalar Universitário S. João, EPE, Porto, Portugal ,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Margarida Santos
- Department of Otorhinolaryngology, Centro Hospitalar Universitário S. João, EPE, Porto, Portugal
| | - Ricardo P. Vaz
- Department of Otorhinolaryngology, Centro Hospitalar Universitário S. João, EPE, Porto, Portugal ,Unit of Anatomy - Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal ,Center for Health Technology and Services Research (CINTESIS), Porto, Portugal
| |
Collapse
|
47
|
Klener P. Mantle cell lymphoma: insights into therapeutic targets at the preclinical level. Expert Opin Ther Targets 2020; 24:1029-1045. [PMID: 32842810 DOI: 10.1080/14728222.2020.1813718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Mantle cell lymphoma (MCL) is a chronically relapsing B-cell non-Hodgkin lymphoma characterized by recurrent molecular-cytogenetic aberrations that lead to deregulation of DNA damage response, cell cycle progression, epigenetics, apoptosis, proliferation, and motility. In the last 10 years, clinical approval of several innovative drugs dramatically changed the landscape of treatment options in the relapsed/refractory (R/R) MCL, which translated into significantly improved survival parameters. AREAS COVERED Here, up-to-date knowledge on the biology of MCL together with currently approved and clinically tested frontline and salvage therapies are reviewed. In addition, novel therapeutic targets in MCL based on the scientific reports published in Pubmed are discussed. EXPERT OPINION Bruton tyrosine-kinase inhibitors, NFkappaB inhibitors, BCL2 inhibitors, and immunomodulary agents in combination with monoclonal antibodies and genotoxic drugs have the potential to induce long-term remissions in majority of newly diagnosed MCL patients. Several other classes of anti-tumor drugs including phosphoinositole-3-kinase, cyclin-dependent kinase or DNA damage response kinase inhibitors have demonstrated promising anti-lymphoma efficacy in R/R MCL. Most importantly, adoptive immunotherapy with genetically modified T-cells carrying chimeric antigen receptor represents a potentially curative treatment approach even in the patients with chemotherapy and ibrutinib-refractory disease.
Collapse
Affiliation(s)
- Pavel Klener
- First Department of Internal Medicine- Hematology, University General Hospital and First Faculty of Medicine, Charles University , Prague, Czech Republic.,Institute of Pathological Physiology, First Faculty of Medicine, Charles University , Prague, Czech Republic
| |
Collapse
|
48
|
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
|
49
|
Guy D, Kahl BS. Initial and Consolidation Therapy for Younger Patients with Mantle Cell Lymphoma. Hematol Oncol Clin North Am 2020; 34:861-870. [PMID: 32861283 DOI: 10.1016/j.hoc.2020.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mantle cell lymphoma is an incurable B-cell malignancy. Treatment of young fit patients is particularly challenging, because careful consideration should be made when building a long-term treatment strategy that would provide longer remissions and increase patients' quality of life. Most young fit patients achieve long remissions with a combination of immunochemotherapy containing rituximab and high-dose cytarabine, followed by high-dose chemotherapy and autologous stem-cell transplantation. The addition of maintenance therapy with rituximab following autologous stem-cell transplantation prolongs the time to relapse and increases overall survival. Despite an intensive approach, late relapses are common and are usually treated with novel agents.
Collapse
Affiliation(s)
- Daniel Guy
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8056-29, St Louis, MO 63108, USA
| | - Brad S Kahl
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8056-29, St Louis, MO 63108, USA.
| |
Collapse
|
50
|
Konjunktivale Malignome. SPEKTRUM DER AUGENHEILKUNDE 2020. [DOI: 10.1007/s00717-020-00450-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|