51
|
Pajouheshnia R, Groenwold RHH, Peelen LM, Reitsma JB, Moons KGM. When and how to use data from randomised trials to develop or validate prognostic models. BMJ 2019; 365:l2154. [PMID: 31142454 DOI: 10.1136/bmj.l2154] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Romin Pajouheshnia
- Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, 3508 GA Utrecht, Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Rolf H H Groenwold
- Department of Clinical Epidemiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Linda M Peelen
- Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, 3508 GA Utrecht, Netherlands
| | - Johannes B Reitsma
- Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, 3508 GA Utrecht, Netherlands
- Cochrane Netherlands, Utrecht, Netherlands
| | - Karel G M Moons
- Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, 3508 GA Utrecht, Netherlands
- Cochrane Netherlands, Utrecht, Netherlands
| |
Collapse
|
52
|
Saksena A, Yin CC, Xu J, Li J, Zhou J, Wang SA, Lin P, Tang G, Wang L, Wang M, Miranda RN, Medeiros LJ, Li S. CD23 expression in mantle cell lymphoma is associated with CD200 expression, leukemic non-nodal form, and a better prognosis. Hum Pathol 2019; 89:71-80. [PMID: 31054894 DOI: 10.1016/j.humpath.2019.04.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/11/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022]
Abstract
Mantle cell lymphoma (MCL) is usually CD23 negative, a feature helpful in distinguishing MCL from chronic lymphocytic leukemia/small lymphocytic lymphoma. However, a subset of MCL cases can be CD23+. Limited data are available regarding the clinicopathological features and prognosis of patients with CD23+ MCL. In this study, we reviewed 798 cases of MCL and identified 103 (13%) that were CD23+ by flow cytometry, all of which were positive for cyclin D1 and/or associated with CCND1/IGH. In all cases of CD23+ MCL, CD23 expression was dim partial or dim, unlike moderate to bright CD23 expression observed in chronic lymphocytic leukemia/small lymphocytic lymphoma. The clinicopathological features and outcome of patients with CD23+ MCL were compared with 240 patients with typical MCL negative for CD23. Patients with CD23+ MCL more often had an elevated leukocyte count (33% versus 18%, P = .009), bone marrow involvement (89% versus 78%, P = .02), stage 4 disease (87% versus 77%, P = .03), and a leukemic presentation (42% versus 11%, P = .0001). CD23+ MCL was also more often positive for CD200 (17% versus. 4.6%, P = .0005) and less commonly positive for SOX11 (55% versus. 74%, P = .027). All other clinicopathological features were similar. With similar treatment regimens and observation times, patients with CD23+ MCL had a significant better overall survival (P = .02) and progression-free survival (P = .029). In conclusion, CD23 expression was observed in 13% of MCL cases and is associated with a better prognosis in patients with MCL. CD23 is associated with leukocytosis, a leukemic presentation, bone marrow involvement, CD200 expression, and a lower frequency of SOX11 positivity.
Collapse
Affiliation(s)
- Annapurna Saksena
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Pathology, UT Health San Antonio, TX 78229, USA
| | - C Cameron Yin
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Xu
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jingyi Li
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Hematology/Oncology, Tianjin Hospital, Tianjin 300000, China
| | - Jiehao Zhou
- Department of Pathology, Indiana University, Indianapolis, IN 46202, USA
| | - Sa A Wang
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pei Lin
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guilin Tang
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lifu Wang
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Pathology, Henan Provincial Hospital, Zhengzhou, Henan 450000, China
| | - Michael Wang
- Lymphoma and Myeloma, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Roberto N Miranda
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shaoying Li
- Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
53
|
Iqbal M, Castano YG, Sher T, Kharfan-Dabaja MA. Intraocular involvement of Mantle cell lymphoma: A case report and literature review. Hematol Oncol Stem Cell Ther 2019; 14:147-152. [PMID: 30978309 DOI: 10.1016/j.hemonc.2019.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/07/2019] [Indexed: 11/19/2022] Open
Abstract
Lymphomatous involvement of the ocular adnexal region (OAR) is generally rare; however, it constitutes the most common ocular malignancy. Most cases of ocular adnexal lymphomas are extra nodal marginal zone B cell lymphomas. Mantle cell lymphoma involvement (MCL) of the OAR is a less common presentation compared with other lymphomas and it has been reported anecdotally. Studies have suggested an association of OAR involvement with poor prognosis. Here, we report the case of a 61 year old man who started experiencing conjunctival erythema which was followed by development of bilateral proptosis. The diagnosis of Stage IV Mantle cell lymphoma with OAR involvement was confirmed 5 months after patient onset of symptomatology. He underwent treatment with systemic chemotherapy, which was followed by high-dose chemotherapy and autologous hematopoietic cell transplant. He is currently day +100 from his transplant and is in complete remission. A review of the literature regarding ocular involvement of MCL is also summarized.
Collapse
Affiliation(s)
- Madiha Iqbal
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA.
| | - Yennifer Gil Castano
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Taimur Sher
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
54
|
Kahl BS, Dreyling M, Gordon LI, Martin P, Quintanilla-Martinez L, Sotomayor EM. Recent advances and future directions in mantle cell lymphoma research: report of the 2018 mantle cell lymphoma consortium workshop. Leuk Lymphoma 2019; 60:1853-1865. [PMID: 30696305 DOI: 10.1080/10428194.2019.1571205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma characterized by the t(11;14) chromosomal translocation. This translocation most often results in overexpression of cyclin D1. MCL is clinically heterogeneous, outcomes are generally poor, and no standard treatment has been established. The recent approvals of ibrutinib and acalabrutinib have provided an additional therapeutic option; however, resistance has emerged as a significant issue and presents the need for more detailed studies of resistance mechanisms. Recent clinical trials have provided new perspectives on the relative efficacy and safety of various approaches for both transplant-eligible and transplant-ineligible patients. Multiple novel strategies are being evaluated in the treatment of MCL, including both targeted agents and cellular immunotherapies. At the Lymphoma Research Foundation's 13th MCL Workshop, researchers gathered to discuss research findings, clinical trial results, and future directions related to MCL, its biology, and its treatment. This report, which includes a summary of each presentation, aims to review recent findings in MCL research and highlight potential areas for future study.
Collapse
Affiliation(s)
- Brad S Kahl
- a Washington University School of Medicine , St. Louis , MO , USA
| | - Martin Dreyling
- b Department of Medicine III , University Hospital, LMU Munich , Munchen , Germany
| | - Leo I Gordon
- c Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Peter Martin
- d Weill Cornell Medicine Division of Hematology-Oncology , New York , NY , USA
| | | | | |
Collapse
|
55
|
Gerson JN, Handorf E, Villa D, Gerrie AS, Chapani P, Li S, Medeiros LJ, Wang MI, Cohen JB, Calzada O, Churnetski MC, Hill BT, Sawalha Y, Hernandez-Ilizaliturri FJ, Kothari S, Vose JM, Bast MA, Fenske TS, Narayana Rao Gari S, Maddocks KJ, Bond D, Bachanova V, Kolla B, Chavez J, Shah B, Lansigan F, Burns TF, Donovan AM, Wagner-Johnston N, Messmer M, Mehta A, Anderson JK, Reddy N, Kovach AE, Landsburg DJ, Glenn M, Inwards DJ, Karmali R, Kaplan JB, Caimi PF, Rajguru S, Evens A, Klein A, Umyarova E, Pulluri B, Amengual JE, Lue JK, Diefenbach C, Fisher RI, Barta SK. Survival Outcomes of Younger Patients With Mantle Cell Lymphoma Treated in the Rituximab Era. J Clin Oncol 2019; 37:471-480. [PMID: 30615550 DOI: 10.1200/jco.18.00690] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Mantle cell lymphoma (MCL) is a B-cell lymphoma characterized by cyclin D1 expression. Autologous hematopoietic cell transplantation (AHCT) consolidation after induction chemotherapy is often used for eligible patients; however, the benefit remains uncertain in the rituximab era. Herein we retrospectively assessed the impact of AHCT consolidation on survival in a large cohort of transplantation-eligible patients age 65 years or younger. PATIENTS AND METHODS We retrospectively studied transplantation-eligible adults age 65 years or younger with newly diagnosed MCL treated between 2000 and 2015. The primary objective was to assess for improved progression-free survival (PFS) with AHCT consolidation and secondarily to assess for improved overall survival (OS). Cox multivariable regression analysis and propensity score-weighted (PSW) analysis were performed. RESULTS Data were collected from 25 medical centers for 1,254 patients; 1,029 met inclusion criteria. Median follow-up for the cohort was 76 months. Median PFS and OS were 62 and 139 months, respectively. On unadjusted analysis, AHCT was associated with improved PFS (75 v 44 months with v without AHCT, respectively; P < .01) and OS (147 v 115 months with v without AHCT, respectively; P < .05). On multivariable regression analysis, AHCT was associated with improved PFS (hazard ratio [HR], 0.54; 95% CI, 0.44 to 0.66; P < .01) and a trend toward improved OS (HR, 0.77; 95% CI, 0.59 to 1.01; P = .06). After PSW analysis, AHCT remained associated with improved PFS (HR, 0.70; 95% CI, 0.59 to 0.84; P < .05) but not improved OS (HR, 0.87; 95% CI, 0.69 to 1.1; P = .2). CONCLUSION In this large cohort of younger, transplantation-eligible patients with MCL, AHCT consolidation after induction was associated with significantly improved PFS but not OS after PSW analysis. Within the limitations of a retrospective analysis, our findings suggest that in younger, fit patients, AHCT consolidation may improve PFS.
Collapse
Affiliation(s)
| | | | - Diego Villa
- 2 BC Cancer, Vancouver, British Columbia, Canada
| | | | - Parv Chapani
- 2 BC Cancer, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | - Julie M Vose
- 7 University of Nebraska Cancer Center, Omaha, NE
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Martha Glenn
- 17 Huntsman Cancer Institute, Salt Lake City, UT
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Pott C, Brüggemann M, Ritgen M, van der Velden VHJ, van Dongen JJM, Kneba M. MRD Detection in B-Cell Non-Hodgkin Lymphomas Using Ig Gene Rearrangements and Chromosomal Translocations as Targets for Real-Time Quantitative PCR. Methods Mol Biol 2019; 1956:199-228. [PMID: 30779036 DOI: 10.1007/978-1-4939-9151-8_9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Minimal residual disease (MRD) diagnostics is of high clinical relevance in patients with indolent B-cell non-Hodgkin lymphomas (B-NHL) and serves as a surrogate parameter to evaluate treatment effectiveness and long-term prognosis. MRD diagnostics performed by real-time quantitative PCR (RQ-PCR) is still the gold standard and currently the most sensitive and the most broadly applied method in follicular lymphoma (FL) and mantle cell lymphoma (MCL). Alternatively, droplet digital PCR (ddPCR) can be used for MRD monitoring in multiple myeloma, mantle cell lymphoma, and follicular lymphoma with comparable sensitivity, accuracy, and reproducibility.The most broadly applicable MRD target in B-NHL is the junctional regions of the rearranged immunoglobulin heavy chain gene (IGHV). Chromosomal translocations like the t(14;18) translocation in FL and t(11;14) translocation in MCL can be used as MRD target in selected lymphoma subtypes. In patients with B-cell chronic lymphocytic leukemia, both flow-cytometry and RQ-PCR are equally suited for MRD assessment as long as a sensitivity of 10-4 shall be achieved.MRD diagnostics targeting the IGHV gene is complex and requires extensive knowledge and experience because the junctional regions of each lymphoma have to be identified before the patient-specific RQ-PCR assays can be designed for MRD monitoring. In addition, the presence and load of somatic hypermutation (SHM) within the rearranged IG heavy variable (IGHV) gene occurring as during B-cell development of germinal center and post-germinal center lymphomas may hamper appropriate primer binding leading to false-negative results. The translocations mentioned above have the advantage that consensus forward primers and probes, both placed in the breakpoint regions of chromosome 18 in FL and chromosome 11 in MCL, can be used in combination with a reverse primer placed in the IGH joining region of chromosome 14. RQ-PCR-based methods can reach a good sensitivity (≤10-4). This chapter provides all relevant background information and technical aspects for the complete laboratory process from detection of the clonal IGHV gene rearrangement and the chromosomal translocations at diagnosis to the actual MRD measurements in clinical follow-up samples of B-NHL. However, it should be noted that MRD diagnostics for clinical treatment protocols has to be accompanied by regular international quality control rounds to ensure the reproducibility and reliability of the MRD results.
Collapse
Affiliation(s)
- Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Monika Brüggemann
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Ritgen
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael Kneba
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
57
|
He JS, Chen X, Wei GQ, Sun J, Zheng WY, Shi JM, Wu WJ, Zhao Y, Zheng GF, Huang H, Cai Z. Simplified MIPI-B prognostic stratification method can predict the outcome well-retrospective analysis of clinical characteristics and management of newly-diagnosed mantle cell lymphoma patients from China. Medicine (Baltimore) 2019; 98:e13741. [PMID: 30608386 PMCID: PMC6344161 DOI: 10.1097/md.0000000000013741] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 11/26/2018] [Indexed: 12/20/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an invasive B-cell lymphoma with significant individual differences. Currently, MCL international prognostic index (MIPI) score and tumor cell proliferation index Ki-67 have been proved to be the most important prognostic factors. But the prognostic effect of these factors in Asian population is uncertain. This study aimed to analyze the disease characteristics and prognostic factors of Chinese MCL patients.A total of 83 cases of newly-diagnosed MCL patients diagnosed by the Department of Pathology of our hospital between January 1, 2011, and May 31, 2016, were enrolled. The disease characteristics, treatment effects, and outcomes of the patients were collected and analyzed.According to our analysis, MCL cases accounted for 6.2% of non-Hodgkin lymphoma (NHL) cases and mainly occurred in elderly males. But the proportion of patients at stage IV by Ann Arbor staging system and high-risk group by simplified-MIPI (s-MIPI) were significantly lower than that among European patients. Immunochemotherapy containing rituximab was significantly more effective than chemotherapy (overall response rate, [ORR]: 88.5% vs 65.2%, P = .021) and significantly prolonged patient survival (progression free survival [PFS]: 45.5 m vs 16.2 m, P = .001; overall survival [OS]: 58.3 m vs 22.8 m, P = .001). The multivariate analysis showed that the B symptoms, s-MIPI and administration of immunochemotherapy were independent prognostic factors that affected PFS and OS of the patients. s-MIPI and B symptom make up s-MIPI-B stratification method, by which patients in low-risk group of s-MIPI without B symptom were classified as low-risk, patients in high-risk group of s-MIPI and patients in low-risk group of s-MIPI with B symptom as high-risk, the rest as middle-risk. 3-year PFS of the 3 groups were 74.9%, 43.4% and 16.1%, respectively (P = .001). 3-year OS were 84.4%, 62.2%, 27.6% (P <.001).Chinese MCL was male predominance. We have a minor proportion of late-stage and high-risk patients compared to European patients. Immunochemotherapy was proved to significantly improve the prognosis of MCL patients. B symptoms, s-MIPI, and administration of rituximab independently influenced the outcome. s-MIPI-B prognostic stratification method may better predict the prognosis of Asian MCL patients. Still, further confirmation in larger populations is needed.
Collapse
Affiliation(s)
- Jing-Song He
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| | - Xi Chen
- Lymphoma Department, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Guo-Qing Wei
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| | - Jie Sun
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| | - Wei-Yan Zheng
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| | - Ji-Min Shi
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| | - Wen-Jun Wu
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| | - Yi Zhao
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| | - Gao-Feng Zheng
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| | - He Huang
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| | - Zhen Cai
- The Bone Marrow Transplantation Center & Multiple Myeloma Treatment Center, The First Affiliated Hospital of Medical College, Zhejiang University
| |
Collapse
|
58
|
Robak T, Jin J, Pylypenko H, Verhoef G, Siritanaratkul N, Drach J, Raderer M, Mayer J, Pereira J, Tumyan G, Okamoto R, Nakahara S, Hu P, Appiani C, Nemat S, Cavalli F, Van Hoof A, Sheliga A, Teixeira A, Tomita A, Rocafiguera AO, Suvorov A, Kuzmin A, Khojasteh A, Mezlini A, Golenkov A, Bosly A, Belch A, Van De Velde A, Illes Á, Mukhopadhyay A, Meddeb B, De Prijck B, Garichochea B, Undar B, Gabarrón C, Cao C, Souza C, Farber C, Won Suh C, Burcoveanu CI, Cebotaru CL, Truica CL, Maruyama D, Belada D, Ben Yehuda D, Udovitsa D, Dolores, Morra E, Späth-Schwalbe E, Gonzalez-Barca E, Osmanov E, Capote FJ, Offner F, Cardenas G, Heß G, Manikhas G, Babu G, Rekhtman G, Rossi G, Marques H, Bumbea H, Wang H, Huang H, Choi I, Bulavina I, Lysenko I, Avivi I, Kryachok I, Zaucha JM, Novak J, Díaz J, Demeter J, Alexeeva J, Zhu J, Vilchevskaya K, Ishizawa K, Mauricio K, Tobinai K, Ando K, Abdulkadryrov K, Shih LY, Kuzina L, Gumus M, De Wit M, Capra M, Marques M, Golubeva M, Ojeda-Uribe M, Kyselyova M, Taniwaki M, Federico M, Crump M, Baccarani M, Ogura M, Egyed M, Udvardy M, Kurosawa M, Uike N, Khuageva N, Shpilberg O, Gladkov O, Samoilova O, Serduk O, Santi P, Zachee P, Kaplan P, Stoia R, Gressin R, Arranz R, Greil R, Grosicki S, Cancelado S, Nair S, Le Gouill S, Van Steenweghen S, Yoon SS, Chuncharune S, Scheider T, Shimoyama T, Liu T, Kinoshita T, Uchida T, Bunworasate U, Vitolo U, Pavlov V, Phooshkooru VR, Lima V, Merkulov V, Nawarawong W, Hong X, Ke X, Terui Y, Tee Goh Y, Maeda Y, Shi Y, Dunaev Y, Lorie Y, Wang Z, Shen Z, Borbenyi Z, Gasztonyi Z, Masliak Z. Frontline bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone (VR-CAP) versus rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in transplantation-ineligible patients with newly diagnosed mantle cell lymphoma: final overall survival results of a randomised, open-label, phase 3 study. Lancet Oncol 2018; 19:1449-1458. [DOI: 10.1016/s1470-2045(18)30685-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 10/28/2022]
|
59
|
Holte H, Beiske K, Boyle M, Trøen G, Blaker YN, Myklebust J, Kvaløy S, Rosenwald A, Lingjærde OC, Rimsza LM, Smeland EB, Scott DW, Kolstad A. The MCL35 gene expression proliferation assay predicts high-risk MCL patients in a Norwegian cohort of younger patients given intensive first line therapy. Br J Haematol 2018; 183:225-234. [PMID: 30080252 PMCID: PMC6530911 DOI: 10.1111/bjh.15518] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/19/2018] [Indexed: 11/29/2022]
Abstract
Patients with mantle cell lymphoma (MCL) generally have a dismal prognosis. Intensified induction treatment with rituximab and high dose cytarabine (R_HDAC), and consolidation with high-dose therapy with autologous stem cell support has resulted in 10-year overall survival (OS) higher than 60%. However, the clinical course varies. Diagnostic tools capable of stratifying patients include the MCL International Prognostic Index (MIPI), gene expression-based proliferation signature, Ki-67 proliferation index or tumour cell morphology. Here, we tested the performance of a newly developed Nanostring-based RNA expression-based proliferation assay (MCL35) on formalin-fixed paraffin-embedded tumour tissue from younger patients recruited in or treated according to Nordic MCL protocols compared to the prognosticators listed above. Seventy-four patients were included and the assay performed well in all cases except four, which had inadequate RNA quality. The patients were evenly distributed in the MCL35 low-, intermediate- and high-risk categories. MCL35 low- and intermediate- risk groups had overlapping progression-free survival (PFS), while patients in the high-risk category had significantly inferior PFS. Combining MCL35 with MIPI or the MIPI-C (MIPI with the addition of binary Ki67 score +/-30%) showed a better discrimination than either assessment alone. In conclusion, the MCL35 assay alone or combined with MIPI or MIPI-C scores can identify patients who still have a dismal outcome despite intensified treatment.
Collapse
Affiliation(s)
- Harald Holte
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute for Clinical Medicine, University of Oslo, Norway
| | - Klaus Beiske
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Merrill Boyle
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Gunhild Trøen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Yngvild Nuvin Blaker
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo
| | - June Myklebust
- KG Jebsen Centre for B-cell malignancies, Institute for Clinical Medicine, University of Oslo, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo
| | | | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center (CCC) Mainfranken, Würzburg, Germany
| | | | - Lisa M. Rimsza
- Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Erlend B Smeland
- KG Jebsen Centre for B-cell malignancies, Institute for Clinical Medicine, University of Oslo, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo
| | - David W. Scott
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Arne Kolstad
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Cancer Immuntherapy, Institute for Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
60
|
Condoluci A, Rossi D, Zucca E, Cavalli F. Toward a Risk-Tailored Therapeutic Policy in Mantle Cell Lymphoma. Curr Oncol Rep 2018; 20:79. [PMID: 30132080 DOI: 10.1007/s11912-018-0728-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Mantle cell lymphoma (MCL) prognosis is strictly related to the characteristics of the disease, which can range from very indolent cases to highly aggressive and refractory ones. Here we will review the current knowledge on MCL biomarkers. RECENT FINDINGS Biomarker-informed diagnosis is essential for differentiating MCL from other mature B cell tumors. Diagnosis of MCL relies on the identification of the t(11;14) translocation by FISH or the consequently aberrant expression of cyclin D1 by immunohistochemistry. For the few cases staining negative for cyclin D1, SOX11 may help to define the diagnosis. Prognostic biomarkers have been proposed to stratify MCL patients, including baseline clinical aspects (leukemic non-nodal presentation, in situ presentation, Mantle cell International Prognostic Index-MIPI), pathological aspects (blastoid morphology, Ki-67 proliferation index, SOX11 expression), genetic aspects (immunoglobulin gene mutation status, TP53 deletion or mutation, CDKN2A deletion), and depth of response after treatment (PET imaging, molecular minimal residual disease). Such tools are increasingly used as a guide for therapeutic decisions. Watchful waiting approach is recommended for patients harboring favorable clinico-biological features, such as leukemic non-nodal presentation, low MIPI score, non-blastoid disease, low Ki-67 proliferation rate, mutated immunoglobulin genes, and the lack of SOX11 expression. For patients in need of frontline therapy, the decision of whether to undertake intensive regimens is based upon patient's age and comorbidities. Central nervous system prophylaxis is recommended for cases showing blastoid morphology. The duration of remission is tightly correlated to the depth of response. With the aim of achieving a longer duration of remission and survival, younger patients may pursue more intensive regimens incorporating high-dose cytarabine, followed by myeloablative consolidation chemotherapy, autologous stem cell transplantation, and rituximab maintenance. Older patients could, on the other hand, benefit from lower intensity immunochemotherapy followed or not by a maintenance therapy depending on which frontline regimen is used. Despite the identification of several potential useful biomarkers that may inform the treatment decisions and the design of clinical trials, the treatment choice remains nowadays determined by the patient age and fitness rather than by the individual patient characteristics. Tailoring therapy toward a risk-adapted strategy to accommodate the wide spectrum of disease is an urgent challenge, and clinical trials may explore the feasibility of a biomarker-defined therapeutic policy.
Collapse
Affiliation(s)
- Adalgisa Condoluci
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Via Vela 6, 6500, Bellinzona, Switzerland
| | - Davide Rossi
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Via Vela 6, 6500, Bellinzona, Switzerland
| | - Emanuele Zucca
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Via Vela 6, 6500, Bellinzona, Switzerland
| | - Franco Cavalli
- Institute of Oncology Research (IOR), Via Vela 6, 6500, Bellinzona, Switzerland.
| |
Collapse
|
61
|
Rauert-Wunderlich H, Mottok A, Scott DW, Rimsza LM, Ott G, Klapper W, Unterhalt M, Kluin-Nelemans HC, Hermine O, Hartmann S, Thorns C, Rymkiewicz G, Holte H, Dreyling M, Hoster E, Rosenwald A. Validation of the MCL35 gene expression proliferation assay in randomized trials of the European Mantle Cell Lymphoma Network. Br J Haematol 2018; 184:616-624. [PMID: 30095158 DOI: 10.1111/bjh.15519] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023]
Abstract
Mantle cell lymphoma (MCL) is still considered incurable and the course of the disease is highly variable. Established risk factors include the Mantle Cell Lymphoma International Prognostic Index (MIPI) and the quantification of the proliferation rate of the tumour cells, e.g. by Ki-67 immunohistochemistry. In this study, we aimed to validate the prognostic value of the gene expression-based MCL35 proliferation assay in patient cohorts from randomized trials of the European Mantle Cell Lymphoma Network. Using this assay, we analysed the gene expression proliferation signature in routine diagnostic lymph node specimens from MCL Younger and MCL Elderly trial patients, and the calculated MCL35 score was used to assign MCL patients to low (61%), standard (27%) or high (12%) risk groups with significantly different outcomes. We confirm here in our prospective clinical trial cohort of MCL patients, that the MCL35 assay is strongly prognostic, providing additional information to the Ki-67 index and the MIPI. Thus, this robust assay may assist in making treatment decisions or in devising risk-adapted prospective clinical trials in the future.
Collapse
Affiliation(s)
- Hilka Rauert-Wunderlich
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Centre (CCC) Mainfranken, Würzburg, Germany
| | - Anja Mottok
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Centre (CCC) Mainfranken, Würzburg, Germany
| | - David W Scott
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Lisa M Rimsza
- Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Wolfram Klapper
- Department of Pathology, Haematopathology Section and Lymph Node Registry, University of Kiel, Kiel, Germany
| | - Michael Unterhalt
- Department of Medicine III, University Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Hanneke C Kluin-Nelemans
- Department of Haematology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Olivier Hermine
- Department of Haematology, Necker Hospital, Public Assistance - Hospital of Paris, University Paris Descartes, Paris, France.,Imagine Institute, University of Sorbonne Paris City, Paris, France
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology, Goethe University, Frankfurt am Main, Germany
| | | | - Grzegorz Rymkiewicz
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie Institute-Oncology Centre, Warsaw, Poland
| | - Harald Holte
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Martin Dreyling
- Department of Medicine III, University Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Eva Hoster
- Department of Medicine III, University Hospital of the Ludwig Maximilians University Munich, Munich, Germany.,Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig Maximilians University Munich, Munich, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Centre (CCC) Mainfranken, Würzburg, Germany
| |
Collapse
|
62
|
Abstract
Mantle cell lymphoma (MCL) is a B-cell non-Hodgkin lymphoma with historically poor long-term survival compared with other B-cell malignancies. Treatment strategies for this disease are variable and dependent on symptoms and patient fitness. Despite recent advances, MCL remains incurable and patients with high-risk disease have particularly poor outcomes. This review focuses on recent developments that enhance our understanding of the biology of MCL and new treatment approaches that have led to substantial improvements in clinical outcomes. We will outline induction immuno-chemotherapy and maintenance strategies in transplant-eligible patients. In addition, effective strategies for patients unfit for intensive induction will be discussed, with a particular focus on novel molecular therapies with activity in MCL. Lastly, a number of ongoing clinical trials will be presented; the data from these trials are anticipated to redefine standards of care in the near future.
Collapse
Affiliation(s)
- Michael Schieber
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Leo I Gordon
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Reem Karmali
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
63
|
Evolving treatment strategies in mantle cell lymphoma. Best Pract Res Clin Haematol 2018; 31:270-278. [PMID: 30213396 DOI: 10.1016/j.beha.2018.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 07/06/2018] [Indexed: 12/23/2022]
Abstract
Mantle cell lymphoma is an incurable, moderately aggressive B cell lymphoma. While a small proportion of patients with indolent disease can be managed expectantly, most patients require treatment. The therapeutic approach is driven by physician recommendation, patient choice, age, fitness and comorbidities. Young, fit patients often receive combination chemoimmunotherapy, including high dose cytarabine, with autologous stem cell transplant. Recent data has indicated benefit from maintenance rituximab following autologous stem cell transplant. Ongoing trials are investigating combinations of chemotherapy and targeted agents as well as the role of minimal residual disease guided therapy. Older, less fit patients often receive bendamustine and rituximab or anthracycline based regimens. Maintenance rituximab is typically administered in older MCL patients after anthracycline based chemotherapy although its use after bendamustine based therapy is not supported by current data. Current trials focus on refining this regimen with the addition of targeted agents. In the relapsed and refractory setting, novel agents have demonstrated activity although durability of responses remains unsatisfactory.
Collapse
|
64
|
Jin J, Okamoto R, Yoon SS, Shih LY, Zhu J, Liu T, Hong X, Pei L, Rooney B, van de Velde H, Huang H. Bortezomib-based therapy for transplant-ineligible East Asian patients with newly diagnosed mantle-cell lymphoma. Onco Targets Ther 2018; 11:3869-3882. [PMID: 30013367 PMCID: PMC6039072 DOI: 10.2147/ott.s150339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Introduction This subgroup analysis of the LYM-3002 Phase III study (NCT00722137) investigated whether substituting bortezomib for vincristine in frontline R-CHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone) therapy could improve outcomes in East Asian patients with newly diagnosed mantle-cell lymphoma (MCL). Materials and methods A total of 121 East Asian patients from China, Taiwan, Japan, and the Republic of Korea with stage II–IV MCL who were ineligible or not considered for stem-cell transplantation were enrolled to six to eight 21-day cycles of R-CHOP or VR-CAP (R-CHOP with bortezomib replacing vincristine). Results The primary end point was progression-free survival. After a median follow-up of 42.4 months, median progression-free survival in East Asian patients was 13.9 (R-CHOP) versus 28.6 (VR-CAP) months (HR 0.7, P=0.157; 43% improvement with VR-CAP). Secondary end points (R-CHOP vs VR-CAP), including complete response rate (47% vs 63%), duration of complete response (median 16.6 vs 46.7 months), and treatment-free interval (median 21 vs 46.5 months), were improved with VR-CAP. VR-CAP was associated with increased but manageable toxicity. The most frequent adverse events were hematologic toxicities. Conclusion VR-CAP was effective in East Asian patients with newly diagnosed MCL, and could be considered for patients in whom stem-cell transplantation is not an option.
Collapse
Affiliation(s)
- Jie Jin
- Department of Hematology, The First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China
| | - Rumiko Okamoto
- Department of Chemotherapy, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Lee-Yung Shih
- Division of Hematology-Oncology, Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Jun Zhu
- Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ting Liu
- Division of Hematology, Department of Internal Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaonan Hong
- Lymphoma and GI Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lixia Pei
- Janssen Research & Development, LLC, Raritan, NJ, USA
| | - Brendan Rooney
- Janssen Research & Development, High Wycombe, Buckinghamshire, UK
| | - Helgi van de Velde
- Oncology Clinical Research, Millennium Pharmaceuticals, Inc., Boston, MA, USA
| | - Huiqiang Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China,
| |
Collapse
|
65
|
Agarwal R, Dawson MA, Dreyling M, Tam CS. Understanding resistance mechanisms to BTK and BCL2 inhibitors in mantle cell lymphoma: implications for design of clinical trials. Leuk Lymphoma 2018; 59:2769-2781. [DOI: 10.1080/10428194.2018.1457148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Rishu Agarwal
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mark A. Dawson
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Centre for Cancer Research, University of Melbourne, Parkville, Victoria, Australia
| | - Martin Dreyling
- Department of Medicine III, University Hospital, LMU Munich, Germany
| | - Constantine S. Tam
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Haematology, St Vincent's Hospital, Fitzroy, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
66
|
Jain P, Romaguera J, Srour SA, Lee HJ, Hagemeister F, Westin J, Fayad L, Samaniego F, Badillo M, Zhang L, Nastoupil L, Kanagal-Shamanna R, Fowler N, Wang ML. Four-year follow-up of a single arm, phase II clinical trial of ibrutinib with rituximab (IR) in patients with relapsed/refractory mantle cell lymphoma (MCL). Br J Haematol 2018; 182:404-411. [PMID: 29785709 DOI: 10.1111/bjh.15411] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 04/18/2018] [Indexed: 01/03/2023]
Abstract
Ibrutinib has shown significant activity in patients with relapsed or refractory mantle cell lymphoma (RR-MCL). We report the long-term outcome and safety profile of a single-centre, single arm, open-label, phase 2 study of RR-MCL treated with IR. Overall, the median follow-up time was 47 months (range 1-52 months), median duration on treatment was 16 months (range 1-53 months) and median number of treatment cycles was 17 (range 1-56). Twenty-nine patients (58%) achieved complete remission and of these, 12 patients continue on study. Thirty-eight patients discontinued treatment, 14 due to disease progression (2 transformed). Patients with blastoid morphology, high risk MCL International Prognostic Index score and high Ki67% had inferior survival. The commonest grade 1-2 toxicities were fatigue, diarrhoea, nausea, arthralgias and myalgias. None had long term toxicities. Median progression-free survival was 43 months. Eighteen patients (36%) died (14 deaths were MCL-related). The median overall survival has not been reached. Treatment with IR can provide durable remissions in a subset of patients with RR-MCL, especially those with low Ki67%. The possible benefit of adding other therapies in combination with IR in RR-MCL is under exploration.
Collapse
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jorge Romaguera
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samer A Srour
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hun J Lee
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frederick Hagemeister
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason Westin
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luis Fayad
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Felipe Samaniego
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria Badillo
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liang Zhang
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lorreta Nastoupil
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nathan Fowler
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael L Wang
- Department of Lymphoma and myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
67
|
Awan FT, Jurczak W. Use of acalabrutinib in patients with mantle cell lymphoma. Expert Rev Hematol 2018; 11:495-502. [DOI: 10.1080/17474086.2018.1473030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Farrukh T. Awan
- Department of Internal Medicine, Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Wojciech Jurczak
- Department of Hematology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
68
|
Chukkapalli V, Gordon LI, Venugopal P, Borgia JA, Karmali R. Metabolic changes associated with metformin potentiates Bcl-2 inhibitor, Venetoclax, and CDK9 inhibitor, BAY1143572 and reduces viability of lymphoma cells. Oncotarget 2018; 9:21166-21181. [PMID: 29765528 PMCID: PMC5940409 DOI: 10.18632/oncotarget.24989] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 03/06/2018] [Indexed: 12/21/2022] Open
Abstract
Metformin exerts direct anti-tumor effects by activating AMP-activated protein kinase (AMPK), a major sensor of cellular metabolism in cancer cells. This, in turn, inhibits pro-survival mTOR signaling. Metformin has also been shown to disrupt complex 1 of the mitochondrial electron transport chain. Here, we explored the lymphoma specific anti-tumor effects of metformin using Daudi (Burkitt), SUDHL-4 (germinal center diffuse large B-cell lymphoma; GC DLBCL), Jeko-1 (Mantle-cell lymphoma; MCL) and KPUM-UH1 (double hit DLBCL) cell lines. We demonstrated that metformin as a single agent, especially at high concentrations produced significant reductions in viability and proliferation only in Daudi and SUDHL-4 cell lines with associated alterations in mitochondrial oxidative and glycolytic metabolism. As bcl-2 proteins, cyclin dependent kinases (CDK) and phosphoinositol-3- kinase (PI3K) also influence mitochondrial physiology and metabolism with clear relevance to the pathogenesis of lymphoma, we investigated the potentiating effects of metformin when combined with novel agents Venetoclax (bcl-2 inhibitor), BAY-1143572 (CDK9 inhibitor) and Idelalisib (p110δ- PI3K inhibitor). Co-treating KPUM-UH1 and SUDHL-4 cells with 10 mM of metformin resulted in 1.4 fold and 8.8 fold decreases, respectively, in IC-50 values of Venetoclax. By contrast, 3-fold and 10 fold reduction in IC-50 values of BAY-1143572 in Daudi and Jeko-1 cells respectively was seen in the presence of 10 mM of metformin. No change in IC-50 value for Idelalisib was observed across cell lines. These data suggest that although metformin is not a potent single agent, targeting cancer metabolism with similar but more effective drugs in novel combination with either bcl-2 or CDK9 inhibitors warrants further exploration.
Collapse
Affiliation(s)
- Vineela Chukkapalli
- Departments of Hematology, Oncology and Stem Cell Therapy, Rush University Medical Center, Chicago, IL, USA
| | - Leo I Gordon
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Parameswaran Venugopal
- Departments of Hematology, Oncology and Stem Cell Therapy, Rush University Medical Center, Chicago, IL, USA
| | - Jeffrey A Borgia
- Departments of Pathology and Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Reem Karmali
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
69
|
Greenwell IB, Staton AD, Lee MJ, Switchenko JM, Saxe DF, Maly JJ, Blum KA, Grover NS, Mathews SP, Gordon MJ, Danilov AV, Epperla N, Fenske TS, Hamadani M, Park SI, Flowers CR, Cohen JB. Complex karyotype in patients with mantle cell lymphoma predicts inferior survival and poor response to intensive induction therapy. Cancer 2018; 124:2306-2315. [PMID: 29579328 DOI: 10.1002/cncr.31328] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/16/2018] [Accepted: 02/05/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND Risk stratification of newly diagnosed patients with mantle cell lymphoma (MCL) primarily is based on the MCL International Prognostic Index (MIPI) and Ki-67 proliferative index. Single-center studies have reported inferior outcomes in patients with a complex karyotype (CK), but this remains an area of controversy. METHODS The authors retrospectively reviewed 483 patients from 5 academic centers in the United States and described the effect of a CK on survival outcomes in individuals with MCL. RESULTS A CK was found to be associated with inferior overall survival (OS) (4.5 vs 11.6 years; P<.01) and progression-free survival (PFS) (1.9 vs 4.4 years; P<.01). In patients who underwent high-intensity induction followed by autologous stem cell transplantation (ASCT) in first remission, a CK was associated with poor OS (5.1 vs 11.6 years; P = .04) and PFS (3.6 vs 7.8 years; P<.01). Among patients with a CK, high-intensity induction had no effect on OS (4.5 vs 3.8 years; P = .77) nor PFS (2.3 vs 1.5 years; P = .46). Similarly, ASCT in first remission did not improve PFS (3.5 vs 1.2 years; P = .12) nor OS (5.1 vs 4.0 years; P = .27). On multivariable analyses with Ki-67 and MIPI, only CK was found to be predictive of OS (hazard ratio [HR], 1.98; 95% confidence interval [95% CI], 1.12-3.49 [P = .02]), whereas both CK (HR, 1.91; 95% CI, 1.17-3.12 [P = .01]) and Ki-67 >30% (HR, 1.86; 95% CI, 1.06-3.28 [P = .03]) were associated with inferior PFS. Multivariable analysis did not identify any specific cytogenetic abnormalities associated with inferior survival. CONCLUSIONS CK appears to be independently associated with inferior outcomes in patients with MCL regardless of the intensity of induction therapy and receipt of ASCT. Cytogenetics should be incorporated into the workup of a new diagnosis of MCL and novel therapeutic approaches should be investigated for patients with CK. Cancer 2018;124:2306-15. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- I Brian Greenwell
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Ashley D Staton
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Michael J Lee
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jeffrey M Switchenko
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Debra F Saxe
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Joseph J Maly
- Division of Hematology/Oncology, The Ohio State University James Cancer Hospital, Columbus, Ohio
| | - Kristie A Blum
- Division of Hematology/Oncology, The Ohio State University James Cancer Hospital, Columbus, Ohio
| | - Natalie S Grover
- Division of Hematology/Oncology, University of North Carolina at Chapel Hill Lineberger Cancer Center, Chapel Hill, North Carolina
| | - Stephanie P Mathews
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill Lineberger Cancer Center, Chapel Hill, North Carolina
| | - Max J Gordon
- Department of Medicine, OHSU Knight Cancer Institute, Portland, Oregon
| | - Alexey V Danilov
- Department of Medicine, OHSU Knight Cancer Institute, Portland, Oregon
| | - Narendranath Epperla
- Department of Hematology and Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin
| | - Timothy S Fenske
- Department of Hematology and Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin
| | - Mehdi Hamadani
- Department of Hematology and Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin
| | | | - Christopher R Flowers
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jonathon B Cohen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
70
|
The degree of overlap between the follicular dendritic cell meshwork and tumor cells in mantle cell lymphoma is associated with prognosis. Pathol Res Pract 2018. [PMID: 29525403 DOI: 10.1016/j.prp.2018.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study concerning mantle cell lymphoma (MCL) investigated retrospectively an association between patient prognosis and the percentage of the total number of lymphoma cells found in the follicular dendritic cell (FDC) meshwork, that is, the degree of overlap of lymphoma cells. Two hundred and nine MCL patients were apportioned to grades I-III, in which the CD21-positive FDC meshwork covered ≤50%, 51%-89%, and ≥90% of the tumor area, respectively. Significant differences among the grades (all, P < 0.01) were found in the following: duration of disease (from onset of clinical manifestation to diagnosis); clinical staging; extranodal involvement (non-lymphoid organs); histological subtype; and Ki-67 proliferation index (PI). After removing the aggressive variants, the overall survival rates of grade I (n = 92) and II (n = 57) patients were similar. The overall survival rates of grade III (n = 46) patients differed from that of grade I + II patients (P < 0.01). The grades negatively correlated with the Ki-67 PI value (r = -0.234, P = 0.001). At each grade the OSR of patients with Ki-67 PI ≤30% was similar to that of patients with Ki-67 >30%. In the Ki-67 PI ≤30% group, the OSRs of the patients differed significantly among the grades. In the Ki-67 >30% group the OSRs of the grades were similar. The results of multivariate Cox regression analysis showed that the degree of overlap, age and Ki-67 PI was the independent prognostic factors of the OSRs of MCL patients. Our data suggests that MCL patients in whom there was a high degree of overlap between the FDC meshwork and tumor area have a better clinical prognosis. The degree of overlap correlates well with the Ki-67 PI, which can be used to predict the prognosis of patients.
Collapse
|
71
|
Inamdar AA, Goy A, Ayoub NM, Attia C, Oton L, Taruvai V, Costales M, Lin YT, Pecora A, Suh KS. Mantle cell lymphoma in the era of precision medicine-diagnosis, biomarkers and therapeutic agents. Oncotarget 2018; 7:48692-48731. [PMID: 27119356 PMCID: PMC5217048 DOI: 10.18632/oncotarget.8961] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/10/2016] [Indexed: 12/15/2022] Open
Abstract
Despite advances in the development of clinical agents for treating Mantle Cell Lymphoma (MCL), treatment of MCL remains a challenge due to complexity and frequent relapse associated with MCL. The incorporation of conventional and novel diagnostic approaches such as genomic sequencing have helped improve understanding of the pathogenesis of MCL, and have led to development of specific agents targeting signaling pathways that have recently been shown to be involved in MCL. In this review, we first provide a general overview of MCL and then discuss about the role of biomarkers in the pathogenesis, diagnosis, prognosis, and treatment for MCL. We attempt to discuss major biomarkers for MCL and highlight published and ongoing clinical trials in an effort to evaluate the dominant signaling pathways as drugable targets for treating MCL so as to determine the potential combination of drugs for both untreated and relapse/refractory cases. Our analysis indicates that incorporation of biomarkers is crucial for patient stratification and improve diagnosis and predictability of disease outcome thus help us in designing future precision therapies. The evidence indicates that a combination of conventional chemotherapeutic agents and novel drugs designed to target specific dysregulated signaling pathways can provide the effective therapeutic options for both untreated and relapse/refractory MCL.
Collapse
Affiliation(s)
- Arati A Inamdar
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andre Goy
- Clinical Divisions, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Nehad M Ayoub
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Christen Attia
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Lucia Oton
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Varun Taruvai
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Mark Costales
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Yu-Ting Lin
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andrew Pecora
- Clinical Divisions, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - K Stephen Suh
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
72
|
Karls S, Shah H, Jacene H. PET/CT for Lymphoma Post-therapy Response Assessment in Other Lymphomas, Response Assessment for Autologous Stem Cell Transplant, and Lymphoma Follow-up. Semin Nucl Med 2018; 48:37-49. [DOI: 10.1053/j.semnuclmed.2017.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
73
|
Arcaini L, Lamy T, Walewski J, Belada D, Mayer J, Radford J, Jurczak W, Morschhauser F, Alexeeva J, Rule S, Cabeçadas J, Campo E, Pileri SA, Biyukov T, Patturajan M, Casadebaig Bravo M, Trnĕný M. Prospective subgroup analyses of the randomized MCL-002 (SPRINT) study: lenalidomide versus investigator's choice in relapsed or refractory mantle cell lymphoma. Br J Haematol 2018; 180:224-235. [PMID: 29193019 PMCID: PMC5814930 DOI: 10.1111/bjh.15025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/19/2017] [Indexed: 01/08/2023]
Abstract
In the mantle cell lymphoma (MCL)-002 study, lenalidomide demonstrated significantly improved median progression-free survival (PFS) compared with investigator's choice (IC) in patients with relapsed/refractory MCL. Here we present the long-term follow-up data and results of preplanned subgroup exploratory analyses from MCL-002 to evaluate the potential impact of demographic factors, baseline clinical characteristics and prior therapies on PFS. In MCL-002, patients with relapsed/refractory MCL were randomized 2:1 to receive lenalidomide (25 mg/day orally on days 1-21; 28-day cycles) or single-agent IC therapy (rituximab, gemcitabine, fludarabine, chlorambucil or cytarabine). The intent-to-treat population comprised 254 patients (lenalidomide, n = 170; IC, n = 84). Subgroup analyses of PFS favoured lenalidomide over IC across most characteristics, including risk factors, such as high MCL International Prognostic Index score, age ≥65 years, high lactate dehydrogenase (LDH), stage III/IV disease, high tumour burden, and refractoriness to last prior therapy. By multivariate Cox regression analysis, factors associated with significantly longer PFS (other than lenalidomide treatment) included normal LDH levels (P < 0·001), nonbulky disease (P = 0·045), <3 prior antilymphoma treatments (P = 0·005), and ≥6 months since last prior treatment (P = 0·032). Overall, lenalidomide improved PFS versus single-agent IC therapy in patients with relapsed/refractory MCL, irrespective of many demographic factors, disease characteristics and prior treatment history.
Collapse
Affiliation(s)
- Luca Arcaini
- Department of Molecular MedicineUniversity of PaviaPaviaItaly
- Department of Haematology OncologyFondazione IRCCS Policlinico San MatteoPaviaItaly
| | - Thierry Lamy
- Department of HaematologyHôpital PontchaillouRennesFrance
| | - Jan Walewski
- Department of Lymphoid MalignanciesMaria Sklodowska‐Curie Memorial Institute and Oncology CentreWarsawPoland
| | - David Belada
- Fourth Department of Internal Medicine, HaematologyCharles University Hospital and Faculty of MedicineHradec KrálovéCzech Republic
| | - Jiri Mayer
- Department of Internal Medicine, Haematology and OncologyUniversity Hospital BrnoBrnoCzech Republic
| | - John Radford
- The University of Manchester and the Christie NHS Foundation TrustManchester Academic Health Science CentreManchesterUnited Kingdom
| | - Wojciech Jurczak
- Department of HaematologyJagiellonian University Medical CollegeKrakówPoland
| | | | - Julia Alexeeva
- Department of Haematology ResearchFederal Medical Research CentreSaint PetersburgRussia
| | - Simon Rule
- Department of HaematologyDerriford HospitalPlymouthUnited Kingdom
| | - José Cabeçadas
- Serviço de Anatomia PatológicaInstituto Português de Oncologia de Lisboa Francisco GentilLisboaPortugal
| | - Elias Campo
- Haematopathology Unit, Hospital ClinicUniversity of BarcelonaBarcelonaSpain
| | | | - Tsvetan Biyukov
- Clinical Research and DevelopmentCelgene SàrlBoudrySwitzerland
| | | | | | - Marek Trnĕný
- Department of HaematologyCharles University HospitalPragueCzech Republic
| | | |
Collapse
|
74
|
Cohen JB, Zain JM, Kahl BS. Current Approaches to Mantle Cell Lymphoma: Diagnosis, Prognosis, and Therapies. Am Soc Clin Oncol Educ Book 2017; 37:512-525. [PMID: 28561694 DOI: 10.1200/edbk_175448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mantle cell lymphoma (MCL) is a unique lymphoma subtype, both biologically and clinically. Virtually all cases are characterized by a common genetic lesion, t(11;14), resulting in overexpression of cyclin D1. The clinical course is moderately aggressive, and the disease is considered incurable. Considerable biologic and clinical heterogeneity exists, with some patients experiencing a rapidly progressive course, while others have disease that is readily managed. New tools exist for risk stratification and may allow for a more personalized approach in the future. Landmark studies have been completed in recent years and outcomes appear to be improving. Randomized clinical trials have clarified the role of high-dose cytarabine (Ara-C) for younger patients and have demonstrated a role for maintenance rituximab therapy. Multiple areas of uncertainty remain, however, and are the focus of ongoing research. This review focuses on (1) strategies to differentiate between aggressive and less aggressive cases, (2) understanding who should receive hematopoietic stem cell transplantation, and (3) the role for maintenance therapy in MCL.
Collapse
Affiliation(s)
- Jonathon B Cohen
- From Emory University, Atlanta, GA; City of Hope, Duarte, CA; Washington University School of Medicine, St. Louis, MO
| | - Jasmine M Zain
- From Emory University, Atlanta, GA; City of Hope, Duarte, CA; Washington University School of Medicine, St. Louis, MO
| | - Brad S Kahl
- From Emory University, Atlanta, GA; City of Hope, Duarte, CA; Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
75
|
TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy. Blood 2017; 130:1903-1910. [DOI: 10.1182/blood-2017-04-779736] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022] Open
Abstract
Key Points
The intensified standard-of-care regimens for younger patients with MCL do not overcome the deleterious effects of TP53 mutations. MCLs with TP53 mutations should be considered for alternative frontline treatment.
Collapse
|
76
|
Abstract
There are no widely accepted prognostic indices for extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue (MALT). This study aimed to develop and validate a specific prognostic tool to personalize and optimize treatment of patients with MALT lymphoma. A prognostic index was built by Cox regression (stepwise selection) using data from 401 patients enrolled in the international randomized International Extranodal Lymphoma Study Group 19 (IELSG-19) trial (NCT 00210353). A validation set, including 633 patients, was obtained by merging 3 independent cohorts of MALT lymphoma patients. The 3 individual features maintaining the greatest prognostic significance for event-free survival (EFS, the main endpoint of the IELSG-19 trial) were age ≥70 years (hazard ratio [HR], 1.72; 95% confidence interval [CI], 1.26-2.33), Ann Arbor stage III or IV (HR, 1.79; 95% CI ,1.35-2.38), and an elevated lactate dehydrogenase level (HR, 1.87; 95% CI, 1.27-2.77). The prognostic index (MALT-IPI) constructed using these 3 parameters identified 3 groups: low, intermediate, and high risk (corresponding to the presence of 0, 1, or ≥2 of these factors, respectively). The 5-year EFS rates in the low-, intermediate-, and high-risk groups were 70%, 56%, and 29%, respectively. The MALT-lymphoma International Prognostic Index (MALT-IPI) also significantly discriminated between patients with different progression-free, overall, and cause-specific survival. The prognostic utility was retained in gastric and nongastric lymphomas, in each treatment arm (chlorambucil, rituximab, and rituximab plus chlorambucil), and was confirmed in the validation set. The new index, MALT-IPI, is a simple, accessible, and effective tool to identify MALT lymphoma patients at risk of poor outcomes. It may help define appropriate treatment approaches for individual patients.
Collapse
|
77
|
Gong Y, Zhang X, Chen R, Wei Y, Zou Z, Chen X. Cytoplasmic expression of C-MYC protein is associated with risk stratification of mantle cell lymphoma. PeerJ 2017. [PMID: 28626618 PMCID: PMC5472035 DOI: 10.7717/peerj.3457] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim To investigate the association of C-MYC protein expression and risk stratification in mantle cell lymphoma (MCL), and to evaluate the utility of C-MYC protein as a prognostic biomarker in clinical practice. Methods We conducted immunohistochemical staining of C-MYC, Programmed cell death ligand 1 (PD-L1), CD8, Ki-67, p53 and SRY (sex determining region Y) -11 (SOX11) to investigate their expression in 64 patients with MCL. The staining results and other clinical data were evaluated for their roles in risk stratification of MCL cases using ANOVA, Chi-square, and Spearman’s Rank correlation coefficient analysis. Results Immunohistochemical staining in our study indicated that SOX11, Ki-67 and p53 presented nuclear positivity of tumor cells, CD8 showed membrane positivity in infiltrating T lymphocytes while PD-L1 showed membrane and cytoplasmic positivity mainly in macrophage cells and little in tumor cells. We observed positive staining of C-MYC either in the nucleus or cytoplasm or in both subcellular locations. There were significant differences in cytoplasmic C-MYC expression, Ki-67 proliferative index of tumor cells, and CD8 positive tumor infiltrating lymphocytes (CD8+TIL) among three risk groups (P = 0.000, P = 0.037 and P=0.020, respectively). However, no significant differences existed in the expression of nuclear C-MYC, SOX11, p53, and PD-L1 in MCL patients with low-, intermediate-, and high risks. In addition, patient age and serum LDH level were also significantly different among 3 groups of patients (P = 0.006 and P = 0.000, respectively). Spearman’s rank correlation coefficient analysis indicated that cytoplasmic C-MYC expression, Ki-67 index, age, WBC, as well as LDH level had significantly positive correlations with risk stratification (P = 0.000, 0.015, 0.000, 0.029 and 0.000, respectively), while CD8+TIL in tumor microenvironment negatively correlated with risk stratification of patients (P = 0.006). Patients with increased positive cytoplasmic expression of C-MYC protein and decreased CD8+TIL appeared to be associated with a poor response to chemotherapy, but the correlation was not statistically significant. Conclusion Our study suggested that assessment of cytoplasmic C-MYC overexpression and cytotoxic T lymphocytes (CTLs) by immunohistochemical staining might be helpful for MCL risk stratification and outcome prediction. However, large cohort studies of MCL patients with complete follow up are needed to validate our speculation.
Collapse
Affiliation(s)
- Yi Gong
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China.,Department of Hematology-oncology, Chongqing Cancer Institute/Hospital, Chongqing, China
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Rui Chen
- Department of Pathology, Chongqing Cancer Institute/Hospital, Chongqing, China
| | - Yan Wei
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Zhongmin Zou
- Institute of Toxicology, School of Preventive Medicine, The Third Military Medical University, Chongqing, China
| | - Xinghua Chen
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
78
|
CD5-negative mantle cell lymphoma shows a less aggressive outcome and variable SOX11 staining. J Hematop 2017. [DOI: 10.1007/s12308-017-0292-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
79
|
Abstract
Mantle cell lymphoma is a relatively rare subtype of lymphoma with a great deal of heterogeneity, both clinically and biologically. Since its recognition as a separate entity in the early 1990s though, consistent efforts have led to a significant improvement of overall survival, from a median overall survival of 2.5 years initially to 5-7 years currently. This decades-long and stepwise progress, summarized in the article, definitely accelerated recently, shedding light on a changing paradigm.
Collapse
Affiliation(s)
- Andre Goy
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA.
| |
Collapse
|
80
|
Hu G, Gupta SK, Troska TP, Nair A, Gupta M. Long non-coding RNA profile in mantle cell lymphoma identifies a functional lncRNA ROR1-AS1 associated with EZH2/PRC2 complex. Oncotarget 2017; 8:80223-80234. [PMID: 29113297 PMCID: PMC5655192 DOI: 10.18632/oncotarget.17956] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/07/2017] [Indexed: 12/03/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell lymphoma characterized by rapid disease progression. The needs for new therapeutic strategies for MCL patients call for further understanding on the molecular mechanisms of pathogenesis of MCL. Recently, long noncoding RNAs (lncRNAs) have been recognized as key regulators of gene expression and disease development, however, the role of lncRNAs in non-Hodgkin lymphoma and specifically in MCL is still unknown. Next generation RNA-sequencing was carried out on MCL patient samples along with normal controls and data was analyzed. As a result, several novel lncRNAs were found significantly overexpressed in the MCL samples with lncRNA ROR1-AS1 the most significant one. We cloned the ROR1-AS1 lncRNA in expression vector and ectopically transfected in MCL cell lines. Results showed that overexpression of ROR1-AS1 lncRNA promoted growth of MCL cells while decreased sensitivity to the treatment with drugs ibrutinib and dexamethasone. ROR-AS1 overexpression also decreased the mRNA expression of P16 (P = 0.21), and SOX11 (p = 0.017), without much effect on P53, ATM and P14 mRNA. RNA-immunoprecipitation assays demonstrated high affinity binding of lncRNA ROR1-AS1 with EZH2 and SUZ12 proteins of the polycomb repressive complex-2 (PRC2). Suppressing EZH2 activity with pharmacological inhibitor GSK343 abolished binding of ROR1-AS1 with EZH2. Taken together, this study identified a functional lncRNA ROR-AS1 involved with regulation of gene transcription via associating with PRC2 complex, and may serve as a novel biomarker in MCL patients.
Collapse
Affiliation(s)
- Guangzhen Hu
- Division of Hematology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Shiv K Gupta
- Department of Radiation Oncology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Tammy P Troska
- Division of Hematology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Asha Nair
- Division of Biomedical Statistics and Informatics, Mayo Clinic Rochester, Rochester, MN, USA
| | - Mamta Gupta
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, GW Cancer Center, Washington, DC, USA
| |
Collapse
|
81
|
Scott DW, Abrisqueta P, Wright GW, Slack GW, Mottok A, Villa D, Jares P, Rauert-Wunderlich H, Royo C, Clot G, Pinyol M, Boyle M, Chan FC, Braziel RM, Chan WC, Weisenburger DD, Cook JR, Greiner TC, Fu K, Ott G, Delabie J, Smeland EB, Holte H, Jaffe ES, Steidl C, Connors JM, Gascoyne RD, Rosenwald A, Staudt LM, Campo E, Rimsza LM. New Molecular Assay for the Proliferation Signature in Mantle Cell Lymphoma Applicable to Formalin-Fixed Paraffin-Embedded Biopsies. J Clin Oncol 2017; 35:1668-1677. [PMID: 28291392 DOI: 10.1200/jco.2016.70.7901] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Mantle cell lymphoma is an aggressive B-cell neoplasm that displays heterogeneous outcomes after treatment. In 2003, the Lymphoma/Leukemia Molecular Profiling Project described a powerful biomarker-the proliferation signature-using gene expression in fresh frozen material. Herein, we describe the training and validation of a new assay that measures the proliferation signature in RNA derived from routinely available formalin-fixed paraffin-embedded (FFPE) biopsies. Methods Forty-seven FFPE biopsies were used to train an assay on the NanoString platform, using microarray gene expression data of matched fresh frozen biopsies as a gold standard. The locked assay was applied to pretreatment FFPE lymph node biopsies from an independent cohort of 110 patients uniformly treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. Seventeen biopsies were tested across three laboratories to assess assay reproducibility. Results The MCL35 assay, which contained a 17-gene proliferation signature, yielded gene expression of sufficient quality to assign an assay score and risk group in 108 (98%) of 110 archival FFPE biopsies. The MCL35 assay assigned patients to high-risk (26%), standard-risk (29%), and low-risk (45%) groups, with different lengths of overall survival (OS): a median of 1.1, 2.6, and 8.6 years, respectively (log-rank for trend, P < .001). In multivariable analysis, these risk groups and the Mantle Cell Lymphoma International Prognostic Index were independently associated with OS ( P < .001 for both variables). Concordance of risk assignment across the three independent laboratories was 100%. Conclusion The newly developed and validated MCL35 assay for FFPE biopsies uses the proliferation signature to define groups of patients with significantly different OS independent of the Mantle Cell Lymphoma International Prognostic Index. Importantly, the analytic and clinical validity of this assay defines it as a reliable biomarker to support risk-adapted clinical trials.
Collapse
Affiliation(s)
- David W Scott
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Pau Abrisqueta
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - George W Wright
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Graham W Slack
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Anja Mottok
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Diego Villa
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Pedro Jares
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Hilka Rauert-Wunderlich
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Cristina Royo
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Guillem Clot
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Magda Pinyol
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Merrill Boyle
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Fong Chun Chan
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Rita M Braziel
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Wing C Chan
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Dennis D Weisenburger
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - James R Cook
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Timothy C Greiner
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Kai Fu
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - German Ott
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Jan Delabie
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Erlend B Smeland
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Harald Holte
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Elaine S Jaffe
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Christian Steidl
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Joseph M Connors
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Randy D Gascoyne
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Andreas Rosenwald
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Louis M Staudt
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Elias Campo
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | - Lisa M Rimsza
- David W. Scott, Pau Abrisqueta, Graham W. Slack, Anja Mottok, Diego Villa, Merrill Boyle, Fong Chun Chan, Christian Steidl, Joseph M. Connors, and Randy D. Gascoyne, BC Cancer Agency; Anja Mottok, Christian Steidl, and Randy D. Gascoyne, University of British Columbia, Vancouver, British Columbia; Jan Delabie, University of Toronto, Toronto, Ontario, Canada; Pau Abrisqueta, Vall d'Hebron University Hospital; Pedro Jares, Cristina Royo, Guillem Clot, Magda Pinyol, and Elias Campo, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; George W. Wright, Elaine S. Jaffe, and Louis M. Staudt, National Institutes of Health, Bethesda, MD; Hilka Rauert-Wunderlich and Andreas Rosenwald, University of Würzburg, Würzburg; German Ott, Robert Bosch Hospital and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Rita M. Braziel, Oregon Health & Sciences University, Portland, OR; Wing C. Chan and Dennis D. Weisenburger, City of Hope, Duarte, CA; James R. Cook, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Timothy C. Greiner and Kai Fu, University of Nebraska Medical Center, Omaha, NE; Erlend B. Smeland and Harald Holte, Oslo University Hospital, Oslo, Norway; and Lisa M. Rimsza, Mayo Clinic, Phoenix, AZ
| | | |
Collapse
|
82
|
Verhoef G, Robak T, Huang H, Pylypenko H, Siritanaratkul N, Pereira J, Drach J, Mayer J, Okamoto R, Pei L, Rooney B, Cakana A, van de Velde H, Cavalli F. Association between quality of response and outcomes in patients with newly diagnosed mantle cell lymphoma receiving VR-CAP versus R-CHOP in the phase 3 LYM-3002 study. Haematologica 2017; 102:895-902. [PMID: 28183846 PMCID: PMC5477608 DOI: 10.3324/haematol.2016.152496] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/07/2017] [Indexed: 12/13/2022] Open
Abstract
In the phase 3 LYM-3002 study comparing intravenous VR-CAP with R-CHOP in patients with newly-diagnosed, measurable stage II-IV mantle cell lymphoma, not considered or ineligible for transplant, the median progression-free survival was significantly improved with VR-CAP (24.7 versus 14.4 months with R-CHOP; P<0.001). This post-hoc analysis evaluated the association between the improved outcomes and quality of responses achieved with VR-CAP versus R-CHOP in LYM-3002. Patients were randomized to six to eight 21-day cycles of VR-CAP or R-CHOP. Outcomes included progression-free survival, duration of response (both assessed by an independent review committee), and time to next anti-lymphoma treatment, evaluated by response (complete response/unconfirmed complete response and partial response), MIPI risk status, and maximum reduction of lymph-node measurements expressed as the sum of the product of the diameters. Within each response category, the median progression-free survival was longer for patients given VR-CAP than for those given R-CHOP (complete response/unconfirmed complete response: 40.9 versus 19.8 months; partial response: 17.1 versus 11.7 months, respectively); similarly, the median time to next anti-lymphoma treatment was longer among the patients given VR-CAP than among those treated with R-CHOP (complete response/unconfirmed complete response: not evaluable versus 26.6 months; partial response: 35.3 versus 24.3 months). Within the complete/unconfirmed complete and partial response categories, improvements in progression-free survival, duration of response and time to next anti-lymphoma treatment were more pronounced in patients with low-and intermediate-risk MIPI treated with VR-CAP than with R-CHOP. In each response category, more VR-CAP than R-CHOP patients had a sum of the product of the diameters nadir of 0 during serial radiological assessments. Results of this post-hoc analysis suggest a greater duration and quality of response in patients treated with VR-CAP in comparison with those treated with R-CHOP, with the improvements being more evident in patients with low- and intermediate-risk MIPI. LYM-3002 ClinicalTrials.gov: NCT00722137.
Collapse
Affiliation(s)
| | - Tadeusz Robak
- Medical University of Lodz, Copernicus Memorial Hospital, Poland
| | - Huiqiang Huang
- Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | | | | | - Juliana Pereira
- Hospital das Clinicas da Faculdade de Medicina da USP, São Paolo, Brazil
| | | | - Jiri Mayer
- Masaryk University Hospital Brno, Czech Republic
| | - Rumiko Okamoto
- Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Japan
| | - Lixia Pei
- Janssen Research & Development, LLC, Raritan, NJ, USA
| | - Brendan Rooney
- Janssen Research & Development, High Wycombe, Buckinghamshire, UK
| | - Andrew Cakana
- Janssen Research & Development, High Wycombe, Buckinghamshire, UK
| | | | - Franco Cavalli
- Oncology Institute of Southern Switzerland, Ospedale San Giovanni, Bellinzona, Ticino, Switzerland
| |
Collapse
|
83
|
Spurgeon SE, Till BG, Martin P, Goy AH, Dreyling MP, Gopal AK, LeBlanc M, Leonard JP, Friedberg JW, Baizer L, Little RF, Kahl BS, Smith MR. Recommendations for Clinical Trial Development in Mantle Cell Lymphoma. J Natl Cancer Inst 2016; 109:2758475. [PMID: 28040733 DOI: 10.1093/jnci/djw263] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/24/2016] [Accepted: 10/04/2016] [Indexed: 12/16/2022] Open
Abstract
Mantle cell lymphoma (MCL) comprises around 6% of all non-Hodgkin's lymphoma (NHL) diagnoses. In younger patients, age less than 60 to 65 years, aggressive induction often followed by consolidation with autologous stem cell transplant has suggested improved outcomes in this population. Less intensive therapies in older patients often followed by maintenance have been studied or are under active investigation. However, despite recent advances, MCL remains incurable, with a median overall survival of around five years. Patients with high-risk disease have particularly poor outcomes. Treatment varies widely across institutions, and to date no randomized trials comparing intensive vs less intensive approaches have been reported. Although recent data have highlighted the heterogeneity of MCL outcomes, patient assessment for treatment selection has largely been driven by patient age with little regard to fitness, disease biology, or disease risk. One critical advance is the finding that minimal residual disease status (MRD) after induction correlates with long-term outcomes. As such, its use as a potential end point could inform clinical trial design. In order to more rapidly improve the outcomes of MCL patients, clinical trials are needed that prospectively stratify patients on the basis of MCL biology and disease risk, incorporate novel agents, and use MRD to guide the need for additional therapy.
Collapse
Affiliation(s)
- Stephen E Spurgeon
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Brian G Till
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Peter Martin
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Andre H Goy
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Martin P Dreyling
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Ajay K Gopal
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Michael LeBlanc
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - John P Leonard
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Jonathan W Friedberg
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Lawrence Baizer
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Richard F Little
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Brad S Kahl
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| | - Mitchell R Smith
- Affiliations of authors: Division of Hematology and Medical Oncology, Oregon Health and Science (OHSU) University Knight Cancer Institute, Portland, OR (SES); Clinical Research Division, Fred Hutchinson Cancer Research Center/ Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA (BGT, AKG); Department of Medicine, Weill Cornell Medicine, New York, NY (PM, JPL); John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ (AHG); Department of Medicine III, Klinikum der Universität München, Campus Grosshadern, Munich, Germany (MPD); Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (ML); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester, Rochester, NY (JWF); Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD (LB); HIV and AIDS Malignancy Branch, Center for Cancer Research, and Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD (RFL); Department of Medicine, Oncology Division, Washington University, St. Louis, MO (BSK); Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH (MRS)
| |
Collapse
|
84
|
Kolstad A, Pedersen LB, Eskelund CW, Husby S, Grønbæk K, Jerkeman M, Laurell A, Räty R, Elonen E, Andersen NS, Brown PD, Kimby E, Bentzen H, Sundström C, Ehinger M, Karjalainen-Lindsberg ML, Delabie J, Ralfkiær E, Fagerli UM, Nilsson-Ehle H, Lauritzsen GF, Kuittinen O, Niemann C, Geisler CH. Molecular Monitoring after Autologous Stem Cell Transplantation and Preemptive Rituximab Treatment of Molecular Relapse; Results from the Nordic Mantle Cell Lymphoma Studies (MCL2 and MCL3) with Median Follow-Up of 8.5 Years. Biol Blood Marrow Transplant 2016; 23:428-435. [PMID: 28039078 DOI: 10.1016/j.bbmt.2016.12.634] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/23/2016] [Indexed: 11/18/2022]
Abstract
The main objectives of the present study were to monitor minimal residual disease (MRD) in the bone marrow of patients with mantle cell lymphoma (MCL) to predict clinical relapse and guide preemptive treatment with rituximab. Among the patients enrolled in 2 prospective trials by the Nordic Lymphoma Group, 183 who had completed autologous stem cell transplantation (ASCT) and in whom an MRD marker had been obtained were included in our analysis. Fresh samples of bone marrow were analyzed for MRD by a combined standard nested and quantitative real-time PCR assay for Bcl-1/immunoglobulin heavy chain gene (IgH) and clonal IgH rearrangements. Significantly shorter progression-free survival (PFS) and overall survival (OS) was demonstrated for patients who were MRD positive pre-ASCT (54 patients) or in the first analysis post-ASCT (23 patients). The median PFS was only 20 months in those who were MRD-positive in the first sample post-ASCT, compared with 142 months in the MRD-negative group (P < .0001). OS was 75% at 10 years and median not reached in the MRD-negative group, compared with only 35 months in the MRD-positive group (P < .0001). Of the 86 patients (47%) who remained in continuous molecular remission, 73% were still in clinical remission after 10 years. For all patients, the median time from ASCT to first molecular relapse was 55 months, with a continuous occurrence of late molecular relapses. Fifty-eight patients who experienced MRD relapse received rituximab as preemptive treatment on 1 or more occasions, and in this group, the median time from first molecular relapse to clinical relapse was 55 months. In most cases, rituximab converted patients to MRD negativity (87%), but many patients became MRD-positive again later during follow-up (69%). By multivariate analysis, high-risk Mantle Cell Lymphoma International Prognostic Index score and positive MRD status pre-ASCT predicted early molecular relapse. In conclusion, preemptive rituximab treatment converts patients to MRD negativity and likely postpones clinical relapse. Molecular monitoring offers an opportunity to select some patients for therapeutic intervention and to avoid unnecessary treatment in others. MRD-positive patients in the first analysis post-ASCT have a dismal prognosis and thus are in need of novel strategies.
Collapse
Affiliation(s)
- Arne Kolstad
- Department of Oncology, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.
| | - Lone Bredo Pedersen
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian W Eskelund
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Simon Husby
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kirsten Grønbæk
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mats Jerkeman
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Anna Laurell
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Riikka Räty
- Department of Hematology and Oncology, Helsinki University Central Hospital, Helsinki, Finland
| | - Erkki Elonen
- Department of Hematology and Oncology, Helsinki University Central Hospital, Helsinki, Finland
| | | | - Peter deNully Brown
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Eva Kimby
- Department of Hematology, Karolinska Institute, Stockholm, Sweden
| | - Hans Bentzen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Mats Ehinger
- Department of Pathology, Skåne University Hospital, Lund, Sweden
| | | | - Jan Delabie
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Elisabeth Ralfkiær
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Herman Nilsson-Ehle
- Department of Hematology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Outi Kuittinen
- Department of Oncology, Oulu University Hospital, Oulu, Finland
| | - Carsten Niemann
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | |
Collapse
|
85
|
Mantle cell lymphoma initial therapy with abbreviated R-CHOP followed by 90Y-ibritumomab tiuxetan: 10-year follow-up of the phase 2 ECOG-ACRIN study E1499. Leukemia 2016; 31:517-519. [PMID: 27780968 DOI: 10.1038/leu.2016.305] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
86
|
Cohen JB, Han X, Jemal A, Ward EM, Flowers CR. Deferred therapy is associated with improved overall survival in patients with newly diagnosed mantle cell lymphoma. Cancer 2016; 122:2356-2363. [DOI: 10.1002/cncr.30068] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Jonathon B. Cohen
- Department of Hematology and Medical Oncology; Emory University; Atlanta Georgia
| | | | | | | | | |
Collapse
|
87
|
Eskelund CW, Kolstad A, Jerkeman M, Räty R, Laurell A, Eloranta S, Smedby KE, Husby S, Pedersen LB, Andersen NS, Eriksson M, Kimby E, Bentzen H, Kuittinen O, Lauritzsen GF, Nilsson-Ehle H, Ralfkiaer E, Ehinger M, Sundström C, Delabie J, Karjalainen-Lindsberg ML, Workman CT, Garde C, Elonen E, Brown P, Grønbaek K, Geisler CH. 15-year follow-up of the Second Nordic Mantle Cell Lymphoma trial (MCL2): prolonged remissions without survival plateau. Br J Haematol 2016; 175:410-418. [DOI: 10.1111/bjh.14241] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/25/2016] [Indexed: 11/27/2022]
Affiliation(s)
| | - Arne Kolstad
- Department of Oncology; Oslo University Hospital; Oslo Norway
| | - Mats Jerkeman
- Department of Oncology; Lund University Hospital; Lund Sweden
| | - Riikka Räty
- Department of Haematology; Helsinki University Central Hospital; Helsinki Finland
| | - Anna Laurell
- Department of Oncology; Uppsala University Hospital; Uppsala Sweden
| | - Sandra Eloranta
- Clinical Epidemiology Unit; Department of Medicine Solna; Karolinska Institute; Stockholm Sweden
| | - Karin E. Smedby
- Clinical Epidemiology Unit; Department of Medicine Solna; Karolinska Institute; Stockholm Sweden
| | - Simon Husby
- Department of Haematology; Rigshospitalet; Copenhagen Denmark
| | | | | | - Mikael Eriksson
- Department of Oncology; Lund University Hospital; Lund Sweden
| | - Eva Kimby
- Department of Haematology; Karolinska Institute; Stockholm Sweden
| | - Hans Bentzen
- Department of Haematology; Aarhus University Hospital; Aarhus Denmark
| | - Outi Kuittinen
- Department of Oncology and Radiotherapy; Oulu University Hospital; Oulu Finland
| | | | - Herman Nilsson-Ehle
- Section of Haematology and Coagulation Medicine; Sahlgrenska University Hospital; Gothenburg Sweden
| | | | - Mats Ehinger
- Department of Pathology; Lund University Hospital; Lund Sweden
| | - Christer Sundström
- Department of Genetics and Pathology; Uppsala University Hospital; Uppsala Sweden
| | - Jan Delabie
- Department of Pathology; Oslo University Hospital; Oslo Norway
| | | | - Christopher T. Workman
- Department of Systems Biology; Technical University of Denmark; Lyngby Denmark
- Department of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Christian Garde
- Department of Systems Biology; Technical University of Denmark; Lyngby Denmark
- Department of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Erkki Elonen
- Department of Haematology; Helsinki University Central Hospital; Helsinki Finland
| | - Peter Brown
- Department of Haematology; Rigshospitalet; Copenhagen Denmark
| | | | | |
Collapse
|
88
|
Lenalidomide-bendamustine-rituximab in patients older than 65 years with untreated mantle cell lymphoma. Blood 2016; 128:1814-1820. [PMID: 27354719 DOI: 10.1182/blood-2016-03-704023] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/07/2016] [Indexed: 12/18/2022] Open
Abstract
For elderly patients with mantle cell lymphoma (MCL), there is no defined standard therapy. In this multicenter, open-label phase 1/2 trial, we evaluated the addition of lenalidomide (LEN) to rituximab-bendamustine (R-B) as first-line treatment for elderly patients with MCL. Patients >65 years with untreated MCL, stages II-IV were eligible for inclusion. Primary end points were maximally tolerable dose (MTD) of LEN and progression-free survival (PFS). Patients received 6 cycles every four weeks of L-B-R (L D1-14, B 90 mg/m2 IV, days 1-2 and R 375 mg/m2 IV, day 1) followed by single LEN (days 1-21, every four weeks, cycles 7-13). Fifty-one patients (median age 71 years) were enrolled from 2009 to 2013. In phase 1, the MTD of LEN was defined as 10 mg in cycles 2 through 6, and omitted in cycle 1. After 6 cycles, the complete remission rate (CRR) was 64%, and 36% were MRD negative. At a median follow-up time of 31 months, median PFS was 42 months and 3-year overall survival was 73%. Infection was the most common nonhematologic grade 3 to 5 event and occurred in 21 (42%) patients. Opportunistic infections occurred in 3 patients: 2 Pneumocystis carinii pneumonia and 1 cytomegalovirus retinitis. Second primary malignancies (SPM) were observed in 8 patients (16%). LEN could safely be combined with R-B when added from the second cycle in patients with MCL, and was associated with a high rate of CR and molecular remission. However, we observed a high degree of severe infections and an unexpected high number of SPMs, which may limit its use. This trial is registered at www.Clinicaltrials.gov as #NCT00963534.
Collapse
|
89
|
Liang R, Wang Z, Zhu MN, Hao CX, Zhang N, Wang JH, Zhang T, Yang L, Gu HT, Dong BX, Bai QX, Gao GX, Chen XQ. [Clinical analysis of the character and prognostic factors of 23 cases of mantle cell lymphoma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2016; 37:491-6. [PMID: 27431074 PMCID: PMC7348349 DOI: 10.3760/cma.j.issn.0253-2727.2016.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the clinical characteristics and prognostic factors of mantle cell lymphoma (MCL) patients. METHODS The clinical data of 23 MCL patients were retrospectively analyzed. Immunohistochemical stain was performed to detect the protein expressions of Mcl-1, pNF-κB p65 and 14-3-3ζ of MCL patients to analyze its prognostic factors. RESULTS Among 23 MCL patients, there were 17(73.9%) patients with IPI 0-2 (low risk group) and 6(26.1%) patients with IPI 3-4. Only the rate of 2y-progression-free survival (PFS) of group IPI 0-2 was superior to that of group IPI 3-4 patients (47.1% vs 0, P=0.049); There were 16 (69.5%) patients with MIPI < 5.7, whose rates of overall response rate (ORR), 2y overall survival (OS) and PFS were better than those of the ones with MIPI ≥ 5.7(ORR: 81.3% vs 33.3% P=0.032; OS: 68.8% vs 16.7% P=0.041; PFS: 50% vs 0, P=0.040 respectively). The rates of ORR, 2y-OS and 2y-PFS (100.0%, 80.0% and 70.0%) of patienets received regimen R+CHOP(E) were all superior to those (38.5%, 30.8% and 7.7%) of ones received regimen CHOP(E) (P=0.002, P= 0.024, P=0.003, respectively). Among 12 patients, 2 out of 6 cases with Mcl-1 positive expression achieved good response (CR+PR) and 2y-OS, 1 case 2y-PFS; All 6 cases with Mcl-1 negative expression achieved good response (CR+PR) and 2y-OS, 5 cases 2y-PFS. 3 out of 6 cases with pNF-κB p65 positive expression achieved good response (CR+PR) and 2y-OS, 1 case 2y-PFS; 5 out of 6 cases with pNF-κB p65 negative expression achieved good response (CR+ PR) and 2y-OS/PFS. 5 out of 8 cases with 14-3-3ζ positive expression achieved good response (CR+PR), 4 cases 2y-OS, and 3 cases 2y-PFS. 3 out of 4 cases with 14-3-3ζ negative expression achieved CR, 4 cases 2y-OS, and 3 cases 2y-PFS. CONCLUSION MCL patients had high heterogeneity. MIPI has better prognostic significance than IPI. R+CHOP(E) as first line treatment improved the rates of OS/PFS. The expressions of Mcl-1, pNF-κB p65 and 14-3-3ζ proteins in MCL might be related to prognosis.
Collapse
Affiliation(s)
- R Liang
- Department of Hematology, Xijing Hosptial, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Hambley B, Caimi PF, William BM. Bortezomib for the treatment of mantle cell lymphoma: an update. Ther Adv Hematol 2016; 7:196-208. [PMID: 27493710 DOI: 10.1177/2040620716648566] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Bortezomib is a first in class proteasome inhibitor, initially approved by the US Food and Drug Administration for the treatment of plasma cell myeloma. Bortezomib has been approved for the treatment of relapsed and refractory mantle cell lymphoma (MCL) and, more recently, in the upfront setting as well. Treatment algorithms for MCL have rapidly evolved over the past two decades, and the optimal regimen remains to be defined. The choice of treatment regimen is based on disease risk stratification models, the expected toxicity of antineoplastic agents, the perceived patient ability to tolerate the planned treatments and the availability of novel agents. As new drugs with novel mechanisms of action and variable toxicity profiles come into use, treatment decisions for a given patient have become increasingly complex. This article provides an overview of the evolving use of bortezomib in the rapidly changing management landscape of MCL.
Collapse
Affiliation(s)
- Bryan Hambley
- Department of Medicine, University Hospitals Case Medical Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Paolo F Caimi
- Department of Medicine, University Hospitals Case Medical Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Basem M William
- Department of Internal Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, A352 Starling Loving Hall, 320 West 10th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
91
|
Pathogenic role of B-cell receptor signaling and canonical NF-κB activation in mantle cell lymphoma. Blood 2016; 128:82-92. [PMID: 27127301 DOI: 10.1182/blood-2015-11-681460] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 04/23/2016] [Indexed: 02/06/2023] Open
Abstract
To interrogate signaling pathways activated in mantle cell lymphoma (MCL) in vivo, we contrasted gene expression profiles of 55 tumor samples isolated from blood and lymph nodes from 43 previously untreated patients with active disease. In addition to lymph nodes, MCL often involves blood, bone marrow, and spleen and is incurable for most patients. Recently, the Bruton tyrosine kinase (BTK) inhibitor ibrutinib demonstrated important clinical activity in MCL. However, the role of specific signaling pathways in the lymphomagenesis of MCL and the biologic basis for ibrutinib sensitivity of these tumors are unknown. Here, we demonstrate activation of B-cell receptor (BCR) and canonical NF-κB signaling specifically in MCL cells in the lymph node. Quantification of BCR signaling strength, reflected in the expression of BCR regulated genes, identified a subset of patients with inferior survival after cytotoxic therapy. Tumor proliferation was highest in the lymph node and correlated with the degree of BCR activation. A subset of leukemic tumors showed active BCR and NF-κB signaling apparently independent of microenvironmental support. In one of these samples, we identified a novel somatic mutation in RELA (E39Q). This sample was resistant to ibrutinib-mediated inhibition of NF-κB and apoptosis. In addition, we identified germ line variants in genes encoding regulators of the BCR and NF-κB pathway previously implicated in lymphomagenesis. In conclusion, BCR signaling, activated in the lymph node microenvironment in vivo, appears to promote tumor proliferation and survival and may explain the sensitivity of this lymphoma to BTK inhibitors.
Collapse
|
92
|
Hoster E, Rosenwald A, Berger F, Bernd HW, Hartmann S, Loddenkemper C, Barth TF, Brousse N, Pileri S, Rymkiewicz G, Kodet R, Stilgenbauer S, Forstpointner R, Thieblemont C, Hallek M, Coiffier B, Vehling-Kaiser U, Bouabdallah R, Kanz L, Pfreundschuh M, Schmidt C, Ribrag V, Hiddemann W, Unterhalt M, Kluin-Nelemans JC, Hermine O, Dreyling MH, Klapper W. Prognostic Value of Ki-67 Index, Cytology, and Growth Pattern in Mantle-Cell Lymphoma: Results From Randomized Trials of the European Mantle Cell Lymphoma Network. J Clin Oncol 2016; 34:1386-94. [DOI: 10.1200/jco.2015.63.8387] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Purpose Mantle-cell lymphoma (MCL) is a rather aggressive B-cell malignancy whose considerable variability of individual outcome is associated with clinical characteristics (Mantle Cell Lymphoma International Prognostic Index [MIPI]). The Ki-67 index is a strong independent prognostic factor; however, the biologic MIPI (MIPI-b) distinguishes only two groups, which does not appropriately depict the clinical heterogeneity. By using the cohort from the European MCL Younger and MCL Elderly trials, we aimed to evaluate the additional prognostic impact of cytology and growth pattern and to improve risk stratification with the Ki-67 index and MIPI. Patients and Methods Diagnostic tumor biopsies were reviewed by the European Mantle Cell Lymphoma Pathology Panel to determine Ki-67 index by using published guidelines, cytology, and growth pattern. We evaluated prognostic effects for overall survival (OS) by Cox regression. For the cohort used for MIPI-b development (German Low-Grade Lymphoma Study Group [GLSG] 1996 and GLSG2000), we checked whether the equally weighted combination of Ki-67 index (dichotomized at the validated 30% cutoff) and MIPI risk groups was adequate and compared the prognostic power of this modified combination to MIPI and MIPI-b for the MCL Younger/MCL Elderly cohort. Results The Ki-67 index was assessed in 508 of 832 patients (median age, 62 years). Blastoid cytology was associated with inferior OS independently of MIPI but not independently of the Ki-67 index. Growth pattern was not independently prognostic. The modified combination of the Ki-67 index and MIPI separated four groups with 5-year OS: 85%, 72%, 43%, and 17% (P < .001) and was more discriminative than MIPI and MIPI-b. Conclusion Using the Ki-67 index is superior to using cytology and growth pattern as prognostic factors in MCL. The modified combination of the Ki-67 index and MIPI showed a refined risk stratification, reflecting their strong complementary prognostic effects while integrating the most relevant prognostic factors available in clinical routine.
Collapse
Affiliation(s)
- Eva Hoster
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Andreas Rosenwald
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Françoise Berger
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Heinz-Wolfram Bernd
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Sylvia Hartmann
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Christoph Loddenkemper
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Thomas F.E. Barth
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Nicole Brousse
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Stefano Pileri
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Grzegorz Rymkiewicz
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Roman Kodet
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Stephan Stilgenbauer
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Roswitha Forstpointner
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Catherine Thieblemont
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Michael Hallek
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Bertrand Coiffier
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Ursula Vehling-Kaiser
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Réda Bouabdallah
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Lothar Kanz
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Michael Pfreundschuh
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Christian Schmidt
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Vincent Ribrag
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Wolfgang Hiddemann
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Michael Unterhalt
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Johanna C. Kluin-Nelemans
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Olivier Hermine
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Martin H. Dreyling
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| | - Wolfram Klapper
- Eva Hoster, Roswitha Forstpointner, Christian Schmidt, Wolfgang Hiddemann, Michael Unterhalt, and Martin H. Dreyling, University Hospital Munich; Eva Hoster, University of Munich, Munich; Andreas Rosenwald, University of Würzburg, Würzburg; Heinz-Wolfram Bernd, University Hospital Schleswig-Holstein, Lübeck; Sylvia Hartmann, University Hospital of Frankfurt, Frankfurt am Main; Christoph Loddenkemper, University Hospital Berlin Charité; Christoph Loddenkemper, Pathologie PathoTres, Berlin; Thomas F.E
| |
Collapse
|
93
|
Which Patients With Mantle Cell Lymphoma Do Not Need Aggressive Therapy. Curr Hematol Malig Rep 2016; 11:234-40. [DOI: 10.1007/s11899-016-0324-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
94
|
Trněný M, Lamy T, Walewski J, Belada D, Mayer J, Radford J, Jurczak W, Morschhauser F, Alexeeva J, Rule S, Afanasyev B, Kaplanov K, Thyss A, Kuzmin A, Voloshin S, Kuliczkowski K, Giza A, Milpied N, Stelitano C, Marks R, Trümper L, Biyukov T, Patturajan M, Bravo MLC, Arcaini L. Lenalidomide versus investigator's choice in relapsed or refractory mantle cell lymphoma (MCL-002; SPRINT): a phase 2, randomised, multicentre trial. Lancet Oncol 2016; 17:319-331. [PMID: 26899778 DOI: 10.1016/s1470-2045(15)00559-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/26/2015] [Accepted: 11/26/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND Lenalidomide, an immunomodulatory drug with antineoplastic and antiproliferative effects, showed activity in many single-group studies in relapsed or refractory mantle cell lymphoma. The aim of this randomised study was to examine the efficacy and safety of lenalidomide versus best investigator's choice of single-agent therapy in relapsed or refractory mantle cell lymphoma. METHODS The MCL-002 (SPRINT) study was a randomised, phase 2 study of patients with mantle cell lymphoma aged 18 years or older at 67 clinics and academic centres in 12 countries who relapsed one to three times, had Eastern Cooperative Oncology Group performance status of 0-2, at least one measurable lesion to be eligible, and who were ineligible for intensive chemotherpy or stem-cell transplantation. Using a centralised interactive voice response system, we randomly assigned (2:1) patients in a permuted block size of six to receive lenalidomide (25 mg orally on days 1-21 every 28 days) until progressive disease or intolerability, or single-agent investigator's choice of either rituximab, gemcitabine, fludarabine, chlorambucil, or cytarabine. Randomisation was stratified by time from diagnosis, time from last anti-lymphoma therapy, and previous stem-cell transplantation. Individual treatment assignment between lenalidomide and investigator's choice was open label, but investigators had to register their choice of comparator drug before randomly assigning a patient. Patients who progressed on investigator's choice could cross over to lenalidomide treatment. We present the prespecified primary analysis results in the intention-to-treat population for the primary endpoint of progression-free survival, defined as the time from randomisation to progressive disease or death, whichever occurred first. Patient enrolment is complete, although treatment and collection of additional time-to-event data are ongoing. This study is registered with ClinicalTrials.gov, number NCT00875667. FINDINGS Between April 30, 2009, and March 7, 2013, we enrolled 254 patients in the intention-to-treat population (170 [67%] were randomly assigned to receive lenalidomide, 84 [33%] to receive investigator's choice monotherapy). Patients had a median age of 68·5 years and received a median of two previous regimens. With a median follow-up of 15·9 months (IQR 7·6-31·7), lenalidomide significantly improved progression-free survival compared with investigator's choice (median 8·7 months [95% CI 5·5-12·1] vs 5·2 months [95% CI 3·7-6·9]) with a hazard ratio of 0·61 (95% CI 0·44-0·84; p=0·004). In the 167 patients in the lenalidomide group and 83 patients in the investigator's choice group who received at least one dose of treatment the most common grade 3-4 adverse events included neutropenia (73 [44%] of 167 vs 28 [34%] of 83) without increased risk of infection, thrombocytopenia (30 [18%] vs 23 [28%]), leucopenia (13 [8%] vs nine [11%]), and anaemia (14 [8%] vs six [7%]). INTERPRETATION Patients with relapsed or refractory mantle cell lymphoma ineligible for intensive chemotherapy or stem-cell transplantation have longer progression-free survival, with a manageable safety profile when treated with lenalidomide compared with monotherapy investigator's choice options. FUNDING Celgene Corporation.
Collapse
Affiliation(s)
- Marek Trněný
- Department of Hematology, Charles University Hospital, Prague, Czech Republic.
| | - Thierry Lamy
- Department of Hematology, Rennes University Hospital, Rennes, France
| | - Jan Walewski
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie Memorial Institute and Oncology Centre, Warsaw, Poland
| | - David Belada
- Fourth Department of Internal Medicine-Hematology, Charles University Hospital and Faculty of Medicine, Hradec Králové, Czech Republic
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, Masaryk University Hospital, Brno, Czech Republic
| | - John Radford
- The University of Manchester and The Christie NHS Foundation Trust, Manchester, UK
| | - Wojciech Jurczak
- Department of Haematology, Jagiellonian University, Krakow, Poland
| | - Franck Morschhauser
- Centre Hospitalier Universitaire Régional de Lille, Unité GRITA, Lille, France
| | | | - Simon Rule
- Department of Hematology, Derriford Hospital, Plymouth, UK
| | - Boris Afanasyev
- First Pavlov State Medical University of St Petersburg, St Petersburg, Russia
| | - Kamil Kaplanov
- Volgograd Regional Clinical Oncology Dispensary Number 1, Department of Hematology, Volgograd, Russia
| | - Antoine Thyss
- Medical Oncology Department, Centre Antoine Lacassagne, Nice, France
| | - Alexej Kuzmin
- Republican Clinical Oncology Dispensary, Kazan, Russia
| | - Sergey Voloshin
- Russian Research Institute of Hematology and Transfusion, St Petersburg, Russia
| | - Kazimierz Kuliczkowski
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Agnieszka Giza
- Department of Haematology, Jagiellonian University, Krakow, Poland
| | - Noel Milpied
- Service d'Hématologie et de Thérapie Cellulaire, CHU Haut-Lévêque, Bordeaux, France; University of Bordeaux, Bordeaux, France
| | | | - Reinhard Marks
- Department of Hematology and Oncology, University Medical Center Freiburg, Freiburg, Germany
| | - Lorenz Trümper
- Department of Hematology and Oncology, Universitätsmedizin Göttingen, Georg-August-Universität, Göttingen, Germany
| | | | | | | | - Luca Arcaini
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo & Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | |
Collapse
|
95
|
Dreyling M, Ferrero S. Personalized medicine in lymphoma: is it worthwhile? The mantle cell lymphoma experience. Haematologica 2016; 100:706-8. [PMID: 26034112 DOI: 10.3324/haematol.2015.127472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Martin Dreyling
- Department of Medicine III, Hospital of the University LMU München, Germany
| | - Simone Ferrero
- Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Italy
| | | |
Collapse
|
96
|
Mantle Cell Lymphoma: First-line Therapy in Patients Not Eligible for Stem Cell Transplantation. Curr Treat Options Oncol 2016; 16:29. [PMID: 25975443 DOI: 10.1007/s11864-015-0343-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OPINION STATEMENT Mantle cell lymphoma is a distinct subtype of non-Hodgkin's lymphoma, which has historically been associated with a poor prognosis. It is now recognized as a heterogeneous disease with variable biologic and clinical behavior. Treatment paradigms have evolved along two lines. Younger, fit mantle cell lymphoma (MCL) patients are generally treated with intensive strategies and older less fit patients with non-intensive strategies. Most of the published literature has focused on intensive strategies, which appear to result in more durable remissions, but with an unclear impact on overall survival. The literature is more sparse for the roughly 50% of patients who are not candidates for intensive strategies, and no "standard" approach has been established for this patient population. However, clues are emerging. Randomized clinical trials have (a) established that bendamustine-rituximab (BR) is more efficacious and less toxic than rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP); (b) established that bortezomib should replace vincristine if using an R-CHOP backbone; and (c) established that maintenance rituximab (MR) is beneficial after an R-CHOP induction. In our opinion, it is reasonable to extrapolate the data supporting MR after R-CHOP and apply MR after a BR induction. In our practice, we recommend BR followed by MR for 2 years to MCL patients not eligible for intensive therapy. An ongoing US intergroup trial is testing the addition of bortezomib to the BR backbone and the addition of lenalidomide to MR. This trial may establish a standard of care in the older MCL population. In addition, exciting options for relapsed MCL have emerged in the last few years, with the introduction of the Bruton tyrosine kinase (BTK) inhibitor ibrutinib and the development of the lenalidomide-rituximab combination. In this article, we will discuss the current available options for these older MCL patients and the evidence supporting those options.
Collapse
|
97
|
Cowan AJ, Stevenson PA, Cassaday RD, Graf SA, Fromm JR, Wu D, Holmberg LA, Till BG, Chauncey TR, Smith SD, Philip M, Orozco JJ, Shustov AR, Green DJ, Libby EN, Bensinger WI, Shadman M, Maloney DG, Press OW, Gopal AK. Pretransplantation Minimal Residual Disease Predicts Survival in Patients with Mantle Cell Lymphoma Undergoing Autologous Stem Cell Transplantation in Complete Remission. Biol Blood Marrow Transplant 2016; 22:380-385. [PMID: 26348890 PMCID: PMC4716882 DOI: 10.1016/j.bbmt.2015.08.035] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/31/2015] [Indexed: 01/22/2023]
Abstract
Autologous stem cell transplantation (ASCT) is standard therapy for mantle cell lymphoma (MCL) in remission after induction chemotherapy, with the best results for patients in complete remission (CR). We hypothesized that evaluation of minimal residual disease (MRD) before ASCT could further stratify outcomes for these patients. Patients with MCL who underwent ASCT in clinical CR between 1996 and 2011 with pretransplantation MRD testing were eligible. Presence of a clonal IgH rearrangement, t(11; 14) by PCR or positive flow cytometry from blood or bone marrow, was considered positive. An adjusted proportional hazards model for associations with progression-free (PFS) and overall survival (OS) was performed. Of 75 MCL patients in CR, 8 (11%) were MRD positive. MRD positivity was associated with shorter OS and PFS. The median OS for MRD-negative patients was not reached, with 82% survival at 5 years, whereas for the MRD-positive patients, median OS was 3.01 years (hazard ratio [HR], 4.04; P = .009), with a median follow-up of 5.1 years. The median PFS for MRD-negative patients was not reached with 75% PFS at 5 years, whereas for MRD-positive patients, it was 2.38 years (HR, 3.69; P = .002). MRD positivity is independently associated with poor outcomes after ASCT for MCL patients in CR.
Collapse
Affiliation(s)
- Andrew J Cowan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Philip A Stevenson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Ryan D Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Solomon A Graf
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Jonathan R Fromm
- Marrow Transplant Unit, Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - David Wu
- Marrow Transplant Unit, Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Leona A Holmberg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Thomas R Chauncey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington; VA Puget Sound Health Care System, Seattle, Washington
| | - Stephen D Smith
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Mary Philip
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Johnnie J Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Andrei R Shustov
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Edward N Libby
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - William I Bensinger
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Mazyar Shadman
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - David G Maloney
- Transplantation Biology Department, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Ajay K Gopal
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington.
| |
Collapse
|
98
|
Dreyling M, Ferrero S. The role of targeted treatment in mantle cell lymphoma: is transplant dead or alive? Haematologica 2016; 101:104-14. [PMID: 26830211 PMCID: PMC4938333 DOI: 10.3324/haematol.2014.119115] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/17/2015] [Indexed: 12/16/2022] Open
Abstract
Based on the profound biological insights of the last years into the molecular pathogenesis of mantle cell lymphoma and the clinical introduction of new targeted drugs, with high efficacy and a good safety profile, the therapeutic scenario for this tumor has been shown to be thoroughly favourable. No longer characterized by a uniformly dismal prognosis, mantle cell lymphoma has been revealed as a spectrum of different diseases, ranging from very indolent cases to highly aggressive and refractory ones. Thus, there is an urgent need to adapt therapy to accommodate the diverse presentations of the disease. High-dose chemotherapy, followed by autologous stem cell transplantation is the current standard of care for younger patients, generally providing high responses and long survival rates, but hampered by acute and long-term toxicity. In addition, some patients may be overtreated, while others could benefit from targeted approaches, based on the new, molecular-directed compounds. Such a personalized treatment based on the specific characteristics of individual patients may be guided by validated prognostic tools, such as the Mantle Cell Lymphoma International Prognostic Index and the Ki-67 Proliferative Index, as well as by early predictors of treatment response, like minimal residual disease analysis. Moreover, mutation screening of distinctive genomic alterations may provide new, predictive biomarkers, with an additional impact on clinical practice. Only after tailoring treatment according to the clinical and biological heterogeneity of the disease the role of transplantation and modern therapeutic options will be redefined in mantle cell lymphoma.
Collapse
Affiliation(s)
- Martin Dreyling
- Department of Medicine III, Hospital of the University LMU München, Germany
| | - Simone Ferrero
- Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Italy
| |
Collapse
|
99
|
Total body irradiation after high-dose cytarabine in mantle cell lymphoma: a comparison of Nordic MCL2, HOVON-45, and European MCL Younger trials. Leukemia 2015; 30:1428-30. [PMID: 26598017 DOI: 10.1038/leu.2015.322] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
100
|
Till BG, Li H, Bernstein SH, Fisher RI, Burack WR, Rimsza LM, Floyd JD, DaSilva MA, Moore DF, Pozdnyakova O, Smith SM, LeBlanc M, Friedberg JW. Phase II trial of R-CHOP plus bortezomib induction therapy followed by bortezomib maintenance for newly diagnosed mantle cell lymphoma: SWOG S0601. Br J Haematol 2015; 172:208-18. [PMID: 26492567 DOI: 10.1111/bjh.13818] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/14/2015] [Indexed: 12/01/2022]
Abstract
Bortezomib is active in mantle cell lymphoma (MCL), with approval in upfront and relapsed settings. Given inevitable recurrence following induction chemoimmunotherapy, maintenance approaches are a rational strategy to improve clinical outcomes. We conducted a phase II study to evaluate the safety and efficacy of six cycles of R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone) plus bortezomib (1.3 mg/m2 days 1 and 4 of 21 d cycles) followed by bortezomib maintenance (1.3 mg/m2 days 1, 4, 8, and 11 every 3 months for 2 years). Sixty-five eligible patients were enrolled. The treatment was well tolerated and toxicities were mainly haematological. The rate of grade ≥3 peripheral neuropathy was low (5%). With a median follow-up of 6.8 years, 2-year progression-free survival (PFS) was 62%, and 2-year overall survival (OS) was 85%. At 5 years, PFS was 28% and OS was 66%. MCL International Prognostic Index scores were significantly associated with 2-year PFS, but did not predict long-term (≥5-year) PFS. Baseline Ki-67 index was significantly associated with survival. Combination R-CHOP with bortezomib followed by maintenance bortezomib appears to improve outcomes compared historically with R-CHOP alone, with prolonged remissions in a subset of patients. These results suggest that inclusion of bortezomib with induction chemotherapy and/or maintenance is promising in MCL and warrants further exploration.
Collapse
Affiliation(s)
- Brian G Till
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington Medical Center, Seattle, WA, USA
| | - Hongli Li
- SWOG Statistical Center, Seattle, WA, USA
| | - Steven H Bernstein
- James P. Wilmot Cancer Center, University of Rochester, Rochester, NY, USA
| | - Richard I Fisher
- Fox Chase Cancer Center, Temple University School of Medicine, Philadelphia, PA, USA
| | - W Richard Burack
- James P. Wilmot Cancer Center, University of Rochester, Rochester, NY, USA
| | - Lisa M Rimsza
- Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Justin D Floyd
- Heartland NCORP/Saint Francis Medical Center, Cape Girardeau, MO, USA
| | - Marco A DaSilva
- Southeast Cancer Consortium-Upstate NCORP/Kingsport Hem & Onc Associates, Kingsport, TN, USA
| | | | | | | | | | | |
Collapse
|