1
|
Lu T, Zhang J, McCracken JM, Young KH. Recent advances in genomics and therapeutics in mantle cell lymphoma. Cancer Treat Rev 2024; 122:102651. [PMID: 37976759 DOI: 10.1016/j.ctrv.2023.102651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Over the past decades, significant strides have been made in understanding the pathobiology, prognosis, and treatment options for mantle cell lymphoma (MCL). The heterogeneity observed in MCL's biology, genomics, and clinical manifestations, including indolent and aggressive forms, is intricately linked to factors such as the mutational status of the variable region of the immunoglobulin heavy chain gene, epigenetic profiling, and Sox11 expression. Several intriguing subtypes of MCL, such as Cyclin D1-negative MCL, in situ mantle cell neoplasm, CCND1/IGH FISH-negative MCL, and the impact of karyotypic complexity on prognosis, have been explored. Notably, recent immunochemotherapy regimens have yielded long-lasting remissions in select patients. The therapeutic landscape for MCL is continuously evolving, with a shift towards nonchemotherapeutic agents like ibrutinib, acalabrutinib, and venetoclax. The introduction of BTK inhibitors has brought about a transformative change in MCL treatment. Nevertheless, the challenge of resistance to BTK inhibitors persists, prompting ongoing efforts to discover strategies for overcoming this resistance. These strategies encompass non-covalent BTK inhibitors, immunomodulatory agents, BCL2 inhibitors, and CAR-T cell therapy, either as standalone treatments or in combination regimens. Furthermore, developing novel drugs holds promise for further improving the survival of patients with relapsed or refractory MCL. In this comprehensive review, we methodically encapsulate MCL's clinical and pathological attributes and the factors influencing prognosis. We also undertake an in-depth examination of stratified treatment alternatives. We investigate conceivable resistance mechanisms in MCL from a genetic standpoint and offer precise insights into various therapeutic approaches for relapsed or refractory MCL.
Collapse
Affiliation(s)
- Tingxun Lu
- Division of Hematopathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Jie Zhang
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Jenna M McCracken
- Division of Hematopathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ken H Young
- Division of Hematopathology, Duke University Medical Center, Durham, NC 27710, USA; Duke Cancer Institute, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
2
|
Patel DA, Wan F, Trinkaus K, Guy DG, Edwin N, Watkins M, Bartlett NL, Cashen A, Fehniger TA, Ghobadi A, Shah NM, Kahl BS. Bendamustine/Rituximab Plus Cytarabine/Rituximab, With or Without Acalabrutinib, for the Initial Treatment of Transplant-Eligible Mantle Cell Lymphoma Patients: Pooled Data From Two Pilot Studies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023:S2152-2650(23)00131-3. [PMID: 37183115 DOI: 10.1016/j.clml.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Mantle cell lymphoma (MCL) is a moderately aggressive lymphoma subtype, generally viewed as incurable. For younger, fit patients, the standard of care remains various high-dose cytarabine-based induction regimens followed by autologous hematopoietic cell transplant and 3 years of rituximab maintenance. Despite reasonably good outcomes, with median progression-free survival in the range of 7 to 9 years, most patients eventually relapse, indicating a need to improve the safety and tolerability of remission induction strategies. METHODS Given the impressive activity of bendamustine/rituximab (BR) in older patients with MCL, we developed an induction regimen modeled after the Nordic Regimen but substituted BR in place of R-CHOP. In a second pilot study, we incorporated the second-generation Bruton tyrosine kinase inhibitor (BTKi), acalabrutinib, into the regimen. The primary endpoint of both studies was stem cell mobilization success rate. RESULTS All patients successfully underwent stem cell harvest in both studies. CONCLUSION The experience from our single institution pilot study suggested that sequential rather than alternating BR and cytarabine/rituximab (CR) was easier to administer from the standpoint of toxicities and subsequent dose modifications. Safety and efficacy data from the 2 pilot studies, FitMCL 1.0 and 2.0, were similar. The pilot studies provided preliminary safety data supporting the development of the NCTN trial EA4181, assessing three different induction regimens with or without acalabrutinib.
Collapse
Affiliation(s)
- Dilan A Patel
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Fei Wan
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Kathryn Trinkaus
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Daniel G Guy
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Natasha Edwin
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Marcus Watkins
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Nancy L Bartlett
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Amanda Cashen
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Todd A Fehniger
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Armin Ghobadi
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Neha-Mehta Shah
- Washington University in St Louis School of Medicine, St Louis, MO, 63110
| | - Brad S Kahl
- Washington University in St Louis School of Medicine, St Louis, MO, 63110.
| |
Collapse
|
3
|
Huwyler F, Kunz R, Bacher U, Hoffmann M, Novak U, Daskalakis M, Banz Y, Pabst T. Evaluation of Bortezomib-BeEAM (2BeEAM) as Chemotherapy Regimen Prior to ASCT in Patients with Mantle Cell Lymphoma. Cancers (Basel) 2023; 15:cancers15072091. [PMID: 37046753 PMCID: PMC10093600 DOI: 10.3390/cancers15072091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
(1) Background: First-line therapy in fit MCL patients may comprise high-dose chemotherapy (HDCT) with autologous transplantation to consolidate remission before maintenance treatment. However, optimization of HDCT is an unmet clinical need given the substantial relapse rate of first-line treatment, while the use of bortezomib is a promising candidate to be added to standard HDCT. (2) Methods: We analyzed 11 consecutive patients with MCL who received bortezomib added to standard BeEAM (2BeEAM) HDCT at a single academic institution. We assessed safety, feasibility, toxicities, and survival rates. (3) Results: All patients had stage III or IV disease. We found that six patients (55%) developed new or worsening of preexisting peripheral neuropathy following administration of 2BeEAM HDCT. One patient relapsed within the first six months after HDCT, whereas three patients never reached complete remission. After a median follow-up of 22 months, the PFS was 64% and the OS 64% at the last follow-up assessment. At this time, 55% of patients were in CR. (4) Conclusions: The use of bortezomib added to standard BeEAM HDCT is associated with relevant toxicities, particularly with regards to additional neuropathy. Moreover, the anti-lymphoma efficacy of 2BeEAM HDCT appears to be modest; therefore, other therapeutic options should be evaluated for consolidation in this patient group.
Collapse
Affiliation(s)
- Fabrizio Huwyler
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Rebekka Kunz
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Ulrike Bacher
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Michèle Hoffmann
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Urban Novak
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Michael Daskalakis
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Yara Banz
- Institute of Pathology, Inselspital, University of Bern, 3008 Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Correspondence: ; Tel.: +41-31-632-8430; Fax: +41-31-632-3410
| |
Collapse
|
4
|
Fenske TS. Frontline Therapy in Mantle Cell Lymphoma: When Clinical Trial and Real-World Data Collide. J Clin Oncol 2023; 41:452-459. [PMID: 36170622 DOI: 10.1200/jco.22.01661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice.A large number of frontline treatment options exist for mantle cell lymphoma (MCL), making selection of therapy a challenge for the clinician. In this Oncology Grand Rounds article, the case of a 73-year-old woman with MCL who attained remission with bendamustine and rituximab is presented. The relevant literature regarding frontline therapy is then reviewed, with particular focus on selection of induction regimen and the potential roles for autologous transplantation and/or rituximab maintenance. This literature primarily consists of prospective phase 2 and phase 3 clinical trials; however, added to this literature now is a growing body of large retrospective real-world cohorts, such as the new analysis by Martin et al,35 the manuscript that accompanies this Oncology Grand Rounds article. In some cases, the real-world evidence is at odds with data from prospective clinical trials, such as regarding the role of rituximab maintenance after bendamustine plus rituximab induction. These important new real-world data are put into context of an ever-changing treatment landscape, in hopes of aiding clinicians in frontline treatment selection for patients with MCL.
Collapse
|
5
|
Whole Transcriptome Sequencing Reveals Cancer-Related, Prognostically Significant Transcripts and Tumor-Infiltrating Immunocytes in Mantle Cell Lymphoma. Cells 2022; 11:cells11213394. [PMID: 36359790 PMCID: PMC9654955 DOI: 10.3390/cells11213394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/29/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma (NHL) subtype characterized by overexpression of CCND1 and SOX11 genes. It is generally associated with clinically poor outcomes despite recent improvements in therapeutic approaches. The genes associated with the development and prognosis of MCL are still largely unknown. Through whole transcriptome sequencing (WTS), we identified mRNAs, lncRNAs, and alternative transcripts differentially expressed in MCL cases compared with reactive tonsil B-cell subsets. CCND1, VCAM1, and VWF mRNAs, as well as MIR100HG and ROR1-AS1 lncRNAs, were among the top 10 most significantly overexpressed, oncogenesis-related transcripts. Survival analyses with each of the top upregulated transcripts showed that MCL cases with high expression of VWF mRNA and low expression of FTX lncRNA were associated with poor overall survival. Similarly, high expression of MSTRG.153013.3, an overexpressed alternative transcript, was associated with shortened MCL survival. Known tumor suppressor candidates (e.g., PI3KIP1, UBXN) were significantly downregulated in MCL cases. Top differentially expressed protein-coding genes were enriched in signaling pathways related to invasion and metastasis. Survival analyses based on the abundance of tumor-infiltrating immunocytes estimated with CIBERSORTx showed that high ratios of CD8+ T-cells or resting NK cells and low ratios of eosinophils are associated with poor overall survival in diagnostic MCL cases. Integrative analysis of tumor-infiltrating CD8+ T-cell abundance and overexpressed oncogene candidates showed that MCL cases with high ratio CD8+ T-cells and low expression of FTX or PCA3 can potentially predict high-risk MCL patients. WTS results were cross-validated with qRT-PCR of selected transcripts as well as linear correlation analyses. In conclusion, expression levels of oncogenesis-associated transcripts and/or the ratios of microenvironmental immunocytes in MCL tumors may be used to improve prognostication, thereby leading to better patient management and outcomes.
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Validated metrics to optimize older adult patient selection for Chimeric Antigen Receptor T-cell therapy (CART) are lacking; however, some preliminary data suggests that geriatric assessments and cumulative illness rating score may be useful tools. In addition, interventions capable of enhancing outcomes in older adults receiving CART have yet to be elucidated. The purpose of this review is to present data extrapolating from other diseases and therapeutic modalities, related to product selection, toxicity mitigation strategies, comprehensive coordinated models of care, and functional optimization of patients. RECENT FINDINGS The most robust data in older adults are among relapsed and refractory (r/r) diffuse large B-cell lymphoma (DLBCL) patients where three products are available with the longest clinical follow up and the most abundant real-world evidence (RWE). Data for the approved CART products for follicular lymphoma (FL) and mantle cell lymphoma (MCL) are relatively new and RWE is lacking in general. Data for CART products in multiple myeloma (MM) and B-cell acute lymphoblastic leukemia (B-ALL) are even more recent, but preliminary data in older adults seem to follow the trend of excellent efficacy in this age group with age-stratified toxicity data limited. Landmark trials and RWE studies indicate that the high response rates of CART for older adult patients, age 65 years and older, are maintained, while toxicity may be amplified. Clinically important toxicities include grade 3 or higher cytokine release syndrome (CRS), neurotoxicity, and infections.
Collapse
|
7
|
Yanada M, Yamamoto K. Hematopoietic cell transplantation for mantle cell lymphoma. Int J Hematol 2022; 115:301-309. [DOI: 10.1007/s12185-022-03294-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 11/28/2022]
|
8
|
Wang T, Yue W, Tang G, Ye M, Yu J, Liu B, Jiao L, Liu X, Yin S, Chen J, Gao L, Yang J, He M. SAMHD1 Mutations and Expression in Mantle Cell Lymphoma Patients. Front Oncol 2021; 11:763151. [PMID: 34976810 PMCID: PMC8719590 DOI: 10.3389/fonc.2021.763151] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/16/2021] [Indexed: 12/27/2022] Open
Abstract
SAMHD1 (sterile alpha motif domain and histidine-aspartate domain-containing protein 1) is a deoxynucleoside triphosphate triphosphohydrolase regulating innate immune and modulating DNA damage signaling. It plays an important role in the development of some tumors. SAMHD1 was also reported as a barrier to cytarabine, a common chemotherapy drug for mantle cell lymphoma (MCL), and as a biomarker of grim prognosis for acute myelocytic leukemia (AML) patients. However, SAMHD1 expression and function in MCL have not been well-defined. In the present study, we evaluated SAMHD1 expression by immunohistochemistry and its gene structure by Sanger sequencing in MCL. Our results showed that SAMHD1 was positive in 36 (62.1%) patients. Importantly, SAMHD1-positive patients were associated with lower chemotherapy response rate (p = 0.023) and shorter overall survival (p = 0.039) than SAMHD1-negative cases. These results suggest that SAMHD1 is an adverse biomarker for MCL patients, which is due to the high expression of SAMHD1 and rapid cell proliferation. These findings were confirmed in an in vitro study using the siRNA technique. Silencing the SAMHD1 gene in the MCL cell line Jeko-1 significantly decreased cell proliferation and increased cell apoptosis. The MCL cell line with SAMHD1 knockdown showed lower Ki-67 proliferation index, higher caspase-3, and higher sensitivity to cytarabine. Furthermore, for the first time, four previously unreported missense mutations (S302Y, Y432C, E449G, and R451H) in exon 8 and exon 12 of the SAMHD1 gene were discovered by sequencing. The mutations had not been found to corelate with SAMHD1 protein expression detected by immunohistochemistry. The biological functions of this mutated SAMHD1 remain to be investigated.
Collapse
Affiliation(s)
- Tao Wang
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wenqin Yue
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Gusheng Tang
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Mingyu Ye
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jiechen Yu
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Bin Liu
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lijuan Jiao
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xuefei Liu
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Shuyi Yin
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jie Chen
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lei Gao
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jianmin Yang
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Miaoxia He
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
9
|
Riedell PA, Hamadani M, Ahn KW, Litovich C, Murthy GSG, Locke FL, Brunstein CG, Merryman RW, Stiff PJ, Pawarode A, Nishihori T, Kharfan-Dabaja MA, Herrera AF, Sauter CS, Smith SM. Outcomes and Utilization Trends of Front-Line Autologous Hematopoietic Cell Transplantation for Mantle Cell Lymphoma. Transplant Cell Ther 2021; 27:911.e1-911.e7. [PMID: 34450333 PMCID: PMC8556305 DOI: 10.1016/j.jtct.2021.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
Although autologous hematopoietic cell transplantation (auto-HCT) has become a common practice for eligible patients in the front-line setting with mantle cell lymphoma (MCL), there are limited data regarding trends in auto-HCT utilization and associated outcomes. This study used the Center for International Blood and Marrow Transplant Research (CIBMTR) database to evaluate survival outcomes and auto-HCT utilization in adults age ≥18 years who underwent auto-HCT within 12 months of diagnosis of MCL between January 2000 and December 2018. The 19-year period from 2000 to 2018 was divided into 4 separate intervals-2000 to 2004, 2005 to 2009, 2010 to 2014, and 2015 to 2018-and encompassed 5082 patients. To evaluate transplantation utilization patterns, we combined MCL incidence derived from the SEER 21 database with CIBMTR- reported auto-HCT activity within 12 months of diagnosis of MCL. Primary outcomes included overall survival (OS) along with the auto-HCT utilization rate. The cumulative incidence of nonrelapse mortality at 1 year decreased from 7% in the earliest cohort (2000 to 2004) to 2% in the latest cohort (2015 to 2018). Mirroring this trend, OS outcomes improved continually with time, with a 3-year OS of 72% in the earliest cohort improving to 86% in the latest cohort. In addition, we noted an increase in auto-HCT utilization from 2001 to 2018, particularly in patients age ≤65 years. This large retrospective analysis highlights trends in auto-HCT utilization and outcomes in patients with MCL and emphasizes the need to optimize pretransplantation and post-transplantation treatment strategies to enhance survival outcomes.
Collapse
Affiliation(s)
- Peter A Riedell
- Division of Hematology and Oncology, University of Chicago Medicine, Chicago, Illinois
| | - Mehdi Hamadani
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - Kwang W Ahn
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Carlos Litovich
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Claudio G Brunstein
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Reid W Merryman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Masachusetts
| | - Patrick J Stiff
- Division of Hematology-Oncology, Department of Medicine, Loyola University Stritch School of Medicine, Maywood, Illinois
| | - Attaphol Pawarode
- Blood and Marrow Transplantation Program, Division of Hematology/Oncology, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Craig S Sauter
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sonali M Smith
- Division of Hematology and Oncology, University of Chicago Medicine, Chicago, Illinois
| |
Collapse
|
10
|
Riedell PA, Hamadani M, Ahn KW, Litovich C, Brunstein CG, Cashen AF, Cohen JB, Epperla N, Hill BT, Im A, Inwards DJ, Lister J, McCarty JM, Ravi Kiran Pingali S, Shadman M, Shaughnessy P, Solh M, Stiff PJ, Vose JM, Kharfan-Dabaja MA, Herrera AF, Sauter CS, Smith SM. Effect of time to relapse on overall survival in patients with mantle cell lymphoma following autologous haematopoietic cell transplantation. Br J Haematol 2021; 195:757-763. [PMID: 34581433 DOI: 10.1111/bjh.17865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 11/28/2022]
Abstract
In young and fit patients with mantle cell lymphoma (MCL), intensive induction therapy followed by a consolidative autologous haematopoietic cell transplant (autoHCT) is the standard of care in the front-line setting. Recently, time-to-event analysis has emerged as an important risk assessment tool in lymphoma, though its impact in MCL is not well defined. We utilized the Center for International Blood and Marrow Transplant Research database to evaluate the effect of post-autoHCT time to relapse on overall survival (OS) over time in 461 patients who underwent autoHCT within 12 months of MCL diagnosis. On multivariate analysis, the impact of relapse on OS was greatest at the six-month [hazard ratio (HR) = 7·68], 12-month (HR = 6·68), and 18-month (HR = 5·81) landmark timepoints. Using a dynamic landmark model we demonstrate that adjusted OS at five years following each landmark timepoint improved with time for relapsing and non-relapsing patients. Furthermore, early relapse (<18 months) following autoHCT defines a high-risk group with inferior post-relapse OS. This retrospective analysis highlights the impact of time to relapse on OS in MCL patients undergoing up-front autoHCT and emphasizes the need to consider novel therapeutic approaches for patients suffering early relapse.
Collapse
Affiliation(s)
- Peter A Riedell
- Division of Hematology and Oncology, University of Chicago Medicine, Chicago, IL, USA
| | - Mehdi Hamadani
- Department of Medicine, CIBMTR® (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, WI, USA.,BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kwang W Ahn
- Department of Medicine, CIBMTR® (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, WI, USA.,Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Carlos Litovich
- Department of Medicine, CIBMTR® (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, WI, USA
| | - Claudio G Brunstein
- Department of Medicine, Adult Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| | - Amanda F Cashen
- Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jonathon B Cohen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Narendranath Epperla
- Division of Hematology, Department of Medicine, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Annie Im
- Division of Hematology/Oncology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | | | - John Lister
- Division of Hematology and Cellular Therapy, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - John M McCarty
- Bone Marrow Transplant Program, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Mazyar Shadman
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Medical Oncology Division, University of Washington, Seattle, WA, USA
| | - Paul Shaughnessy
- Sarah Cannon Transplant and Cellular Therapy Program Methodist Hospital, San Antonio, TX, USA
| | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, GA, USA
| | - Patrick J Stiff
- Department of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Julie M Vose
- Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Craig S Sauter
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Sonali M Smith
- Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
11
|
Sawalha Y, Radivoyevitch T, Jia X, Tullio K, Dean RM, Pohlman B, Hill BT, Kalaycio M, Majhail NS, Jagadeesh D. The impact of socioeconomic disparities on the use of upfront autologous stem cell transplantation for mantle cell lymphoma. Leuk Lymphoma 2021; 63:335-343. [PMID: 34521300 DOI: 10.1080/10428194.2021.1978085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Using the National Cancer Database, we identified 10,290 patients with newly diagnosed mantle cell lymphoma (MCL) treated with chemotherapy with or without upfront autologous stem cell transplantation (ASCT). Only 17% of patients underwent ASCT. Patients who underwent ASCT were younger and more likely to have lower comorbidity scores, private insurance, higher income and education, and treatment received at an academic facility. On multivariable analysis, age, comorbidity index, insurance type, the transition of care, facility type, distance to facility, and diagnosis year were predictive for ASCT use. ASCT use was associated with improved 5-year overall survival in younger (82% vs. 64%, p < .001) and older (70% vs. 40%, p < .001) patients, which was retained in the matched propensity score and 12-month analyses. Female gender, the diagnosis year ≥2009, private insurance, higher income, and education were associated with superior survival, whereas Black race and higher comorbidities predicted inferior survival.
Collapse
Affiliation(s)
- Yazeed Sawalha
- Arthur G. James Comprehensive Cancer Center, Department of Internal Medicine, Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tomas Radivoyevitch
- Quantitative Health Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Xuefei Jia
- Quantitative Health Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Katherine Tullio
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Robert M Dean
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Brad Pohlman
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Brian T Hill
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Matt Kalaycio
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Navneet S Majhail
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| | - Deepa Jagadeesh
- Taussig Cancer Institute, Department of Medical Oncology and Hematology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
12
|
Munshi PN, Hamadani M, Kumar A, Dreger P, Friedberg JW, Dreyling M, Kahl B, Jerkeman M, Kharfan-Dabaja MA, Locke FL, Shadman M, Hill BT, Ahmed S, Herrera AF, Sauter CS, Bachanova V, Ghosh N, Lunning M, Kenkre VP, Aljurf M, Wang M, Maddocks KJ, Leonard JP, Kamdar M, Phillips T, Cashen AF, Inwards DJ, Sureda A, Cohen JB, Smith SM, Carlo-Stella C, Savani B, Robinson SP, Fenske TS. American Society of Transplantation and Cellular Therapy, Center of International Blood and Marrow Transplant Research, and European Society for Blood and Marrow Transplantation Clinical Practice Recommendations for Transplantation and Cellular Therapies in Mantle Cell Lymphoma. Transplant Cell Ther 2021; 27:720-728. [PMID: 34452722 PMCID: PMC8447221 DOI: 10.1016/j.jtct.2021.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 11/18/2022]
Abstract
Autologous (auto-) and allogeneic (allo-) hematopoietic cell transplantation (HCT) are accepted treatment modalities in contemporary treatment algorithms for mantle cell lymphoma (MCL). Chimeric antigen receptor (CAR) T cell therapy recently received approval for MCL; however, its exact place and sequence in relation to HCT remain unclear. The American Society of Transplantation and Cellular Therapy, Center of International Blood and Marrow Transplant Research, and the European Society for Blood and Marrow Transplantation jointly convened an expert panel to formulate consensus recommendations for role, timing, and sequencing of auto-HCT, allo-HCT, and CAR T cell therapy for patients with newly diagnosed and relapsed/refractory (R/R) MCL. The RAND-modified Delphi method was used to generate consensus statements. Seventeen consensus statements were generated, with a few key statements as follows: in the first line setting, auto-HCT consolidation represents standard of care in eligible patients, whereas there is no clear role of allo-HCT or CAR T cell therapy outside of clinical trials. In the R/R setting, the preferential option is CAR T cell therapy, especially in patients with MCL failing or intolerant to at least one Bruton's tyrosine kinase inhibitor, while allo-HCT is recommended if CAR T cell therapy fails or is infeasible. Several recommendations were based on expert opinion, where the panel developed consensus statements for important real-world clinical scenarios to guide clinical practice. In the absence of contemporary evidence-based data, the panel found RAND-modified Delphi methodology effective in providing a formal framework for developing consensus recommendations for the timing and sequence of cellular therapies for MCL.
Collapse
Affiliation(s)
- Pashna N Munshi
- Department of Blood and Marrow Transplantation, MedStar Georgetown University Hospital, Washington, DC
| | - Mehdi Hamadani
- Center for International Blood & Marrow Transplant Research and the BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - Ambuj Kumar
- Department of Internal Medicine, Office of Research, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Peter Dreger
- Department of Medicine, University of Heidelberg, Germany
| | | | - Martin Dreyling
- Department of Medicine III, LMU Hospital Munich, Munich, Germany
| | - Brad Kahl
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Mats Jerkeman
- Department of Oncology, Skane University Hospital, Lund University, Lund, Sweden
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapies Program, Mayo Clinic, Jacksonville, Florida
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Mazyar Shadman
- Blood and Marrow Transplantation Program, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Sairah Ahmed
- Department of Lymphoma, Myeloma and Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Craig S Sauter
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Nilanjan Ghosh
- Levine Cancer Institute, Atrium Health, Charlotte, Nnorth Carolina
| | - Matthew Lunning
- Divsion of Hematology and Oncology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Vaishalee P Kenkre
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mahmoud Aljurf
- Blood and Marrow Transplantation Program, King Faisal Specialist Hospital, Riyadh, Saudi Arabia
| | - Michael Wang
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kami J Maddocks
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - John P Leonard
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Manali Kamdar
- Division of Hematology, Hematologic Malignancies and Stem Cell Transplantation, University of Colorado Cancer Center, Denver, Colorado
| | - Tycel Phillips
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Amanda F Cashen
- Division of Oncology, Section of Stem Cell Transplantation, Washington University School of Medicine, St Louis, Missouri
| | | | - Anna Sureda
- Clinical Hematology Department, Catalan Institute of Oncology, Institut d'Investigació Biomèdica de Bellvitge IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | | | - Sonali M Smith
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Carmello Carlo-Stella
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milan, Italy
| | - Bipin Savani
- Blood and Marrow Transplantation Program, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Stephen P Robinson
- University Hospital Bristol NHS Foundation Trust, London, United Kingdom
| | - Timothy S Fenske
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
13
|
Munshi PN, Hamadani M, Kumar A, Dreger P, Friedberg JW, Dreyling M, Kahl B, Jerkeman M, Kharfan-Dabaja MA, Locke FL, Shadman M, Hill BT, Ahmed S, Herrera AF, Sauter CS, Bachanova V, Ghosh N, Lunning M, Kenkre VP, Aljurf M, Wang M, Maddocks KJ, Leonard JP, Kamdar M, Phillips T, Cashen AF, Inwards DJ, Sureda A, Cohen JB, Smith SM, Carlo-Stella C, Savani B, Robinson SP, Fenske TS. ASTCT, CIBMTR, and EBMT clinical practice recommendations for transplant and cellular therapies in mantle cell lymphoma. Bone Marrow Transplant 2021; 56:2911-2921. [PMID: 34413469 PMCID: PMC8639670 DOI: 10.1038/s41409-021-01288-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 11/09/2022]
Abstract
Autologous (auto-) or allogeneic (allo-) hematopoietic cell transplantation (HCT) are accepted treatment modalities for mantle cell lymphoma (MCL). Recently, chimeric antigen receptor (CAR) T-cell therapy received approval for MCL; however, its exact place and sequence in relation to HCT is unclear. The ASTCT, CIBMTR, and the EBMT, jointly convened an expert panel to formulate consensus recommendations for role, timing, and sequencing of auto-, allo-HCT, and CAR T-cell therapy for patients with newly diagnosed and relapsed/refractory (R/R) MCL. The RAND-modified Delphi method was used to generate consensus statements. Seventeen consensus statements were generated; in the first-line setting auto-HCT consolidation represents standard-of-care in eligible patients, whereas there is no clear role of allo-HCT or CAR T-cell therapy, outside of a clinical trial. In the R/R setting, the preferential option is CAR T-cell therapy especially in MCL failing or intolerant to at least one Bruton's tyrosine kinase inhibitor, while allo-HCT is recommended if CAR T-cell therapy has failed or is not feasible. In the absence of contemporary evidence-based data, the panel found RAND-modified Delphi methodology effective in providing a formal framework for developing consensus recommendations for the timing and sequence of cellular therapies for MCL.
Collapse
Affiliation(s)
| | - Mehdi Hamadani
- CIBMTR & BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Ambuj Kumar
- Department of Internal Medicine, Office of Research, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | | | - Martin Dreyling
- Department of Medicine III, LMU Hospital Munich, Munich, Germany
| | - Brad Kahl
- Washington University School of Medicine, St. Louis, MO, USA
| | - Mats Jerkeman
- Department of Oncology, Skane University Hospital, Lund University, Lund, Sweden
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapies Program, Mayo Clinic, Jacksonville, FL, USA
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Mazyar Shadman
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA, USA
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sairah Ahmed
- Department of Lymphoma, Myeloma and Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, USA
| | - Craig S Sauter
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Nilanjan Ghosh
- Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | | | - Vaishalee P Kenkre
- University of Wisconsin, Division of Hematology and Oncology, Madison, WI, USA
| | | | - Michael Wang
- Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kami J Maddocks
- Division of Hematology, Ohio State University, Columbus, OH, USA
| | | | - Manali Kamdar
- Division of Hematology, Hematologic Malignancies and Stem Cell Transplantation, University of Colorado Cancer Center, Denver, CO, USA
| | - Tycel Phillips
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Amanda F Cashen
- Division of Oncology, Section of Stem Cell Transplantation, Washington University School of Medicine, St Louis, MO, USA
| | - David J Inwards
- Division of Hematology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Anna Sureda
- Clinical Hematology Department, Catalan Institute of Oncology, Institut d'Investigació Biomèdica de Bellvitge IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | | | - Sonali M Smith
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Carmello Carlo-Stella
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milan, Italy
| | - Bipin Savani
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Timothy S Fenske
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
14
|
Roerden M, Wirths S, Sökler M, Bethge WA, Vogel W, Walz JS. Impact of Mantle Cell Lymphoma Contamination of Autologous Stem Cell Grafts on Outcome after High-Dose Chemotherapy. Cancers (Basel) 2021; 13:cancers13112558. [PMID: 34071000 PMCID: PMC8197101 DOI: 10.3390/cancers13112558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 11/19/2022] Open
Abstract
Simple Summary High-dose chemotherapy followed by autologous hematopoietic stem cell transplantation (Auto-HSCT) is a standard frontline treatment for fit mantle cell lymphoma (MCL) patients. As there is a need for predictive factors to identify patients unlikely to benefit from this therapy, we investigated the prognostic impact of lymphoma cell contamination of autologous stem cell grafts. Analyzing a cohort of 36 MCL patients, we show that lymphoma cell contamination of stem cell grafts is associated with poor outcomes after Auto-HSCT. Its analysis might thus improve risk assessment and enable risk-stratified treatment strategies for MCL patients. Abstract Novel predictive factors are needed to identify mantle cell lymphoma (MCL) patients at increased risk for relapse after high-dose chemotherapy and autologous hematopoietic stem cell transplantation (HDCT/Auto-HSCT). Although bone marrow and peripheral blood involvement is commonly observed in MCL and lymphoma cell contamination of autologous stem cell grafts might facilitate relapse after Auto-HSCT, prevalence and prognostic significance of residual MCL cells in autologous grafts are unknown. We therefore performed a multiparameter flow cytometry (MFC)-based measurable residual disease (MRD) assessment in autologous stem cell grafts and analyzed its association with clinical outcome in an unselected retrospective cohort of 36 MCL patients. MRD was detectable in four (11%) autologous grafts, with MRD levels ranging from 0.002% to 0.2%. Positive graft-MRD was associated with a significantly shorter progression-free and overall survival when compared to graft-MRD negative patients (median 9 vs. 56 months and 25 vs. 132 months, respectively) and predicted early relapse after Auto-HSCT (median time to relapse 9 vs. 44 months). As a predictor of outcome after HDCT/Auto-HSCT, MFC-based assessment of graft-MRD might improve risk stratification and support clinical decision making for risk-oriented treatment strategies in MCL.
Collapse
Affiliation(s)
- Malte Roerden
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- Correspondence:
| | - Stefan Wirths
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Martin Sökler
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Wolfgang A. Bethge
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Wichard Vogel
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
| | - Juliane S. Walz
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; (S.W.); (M.S.); (W.A.B.); (W.V.); (J.S.W.)
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology (IKP) and Robert Bosch Center for Tumor Diseases (RBCT), Auerbachstr. 112, 70376 Stuttgart, Germany
| |
Collapse
|
15
|
Genomic profiles and clinical outcomes of de novo blastoid/pleomorphic MCL are distinct from those of transformed MCL. Blood Adv 2021; 4:1038-1050. [PMID: 32191807 DOI: 10.1182/bloodadvances.2019001396] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 01/27/2020] [Indexed: 01/03/2023] Open
Abstract
Blastoid and pleomorphic mantle cell lymphomas (MCLs) are variants of aggressive histology MCL (AH-MCL). AH-MCL can arise de novo (AH-DN) or transform from prior classic variant MCL (AH-t). This study is the first integrated analysis of clinical and genomic characteristics of AH-MCL. Patient characteristics were collected from diagnosis (AH-DN) and at transformation (AH-t). Survival after initial diagnosis (AH-DN) and after transformation (AH-t) was calculated. Regression tree analysis was performed to evaluate prognostic variables and in univariate and multivariate analyses for survival. Whole-exome sequencing was performed in evaluable biopsy specimens. We identified 183 patients with AH-MCL (108 were AH-DN, and 75 were AH-t; 152 were blastoid, and 31 were pleomorphic). Median survival was 33 months (48 and 14 months for AH-DN and AH-t, respectively; P = .001). Factors associated with inferior survival were age (≥72 years), AH-t category, Ki-67 ≥50% and poor performance status. AH-t had a significantly higher degree of aneuploidy compared with AH-DN. Transformed MCL patients exhibited KMT2B mutations. AH-MCL patients with Ki-67 ≥50% had exclusive mutations in CCND1, NOTCH1, TP53, SPEN, SMARCA4, RANBP2, KMT2C, NOTCH2, NOTCH3, and NSD2 compared with low Ki-67 (<50%). AH-t patients have poor outcomes and distinct genomic profile. This is the first study to report that AH-MCL patients with high Ki-67 (≥50%) exhibit a distinct mutation profile and very poor survival.
Collapse
|
16
|
Modified conditioning regimen with idarubicin followed by autologous hematopoietic stem cell transplantation for invasive B-cell non-Hodgkin's lymphoma patients. Sci Rep 2021; 11:4273. [PMID: 33608570 PMCID: PMC7895978 DOI: 10.1038/s41598-021-81944-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 12/28/2020] [Indexed: 12/20/2022] Open
Abstract
High-dose chemotherapy followed by autologous hematopoietic stem cell transplantation (ASCT) is still a consolidation treatment choice for relapsed/refractory B-cell non-Hodgkin’s lymphoma (NHL) patients and some aggressive B-cell NHL as frontline therapy. Due to the shortage of carmustine, we switched to idarubicin-substituted BEAC (IEAC) conditioning regimen. We retrospectively compared the outcomes of 72 aggressive B-cell NHL patients treated with IEAC or BEAC regimens followed by ASCT as upfront consolidative treatment. The median time to neutrophil and platelet reconstitution showed no difference between IEAC and BEAC groups. IEAC regimen was well tolerated without increase of adverse events. Transplant-related mortality didn’t occur. The overall survival (OS) and progression-free survival (PFS) of IEAC group (33 and 23 months) were a little longer than that of BEAC group (30 and 18 months). However, due to the small sample numbers, there’s no significant difference in OS and PFS between IEAC and BEAC group with DLBCL or MCL. Multivariate analysis showed that AnnArbor staging, IPI score, lactate dehydrogenase level, remission of disease, modified regimen were related with PFS and OS. In conclusion, IEAC regimen was well tolerated and replacement with idarubicin could be an alternative when carmustine was not available.
Collapse
|
17
|
Cellular Therapies for Mantle Cell Lymphoma. Transplant Cell Ther 2021; 27:363-370. [PMID: 33965173 DOI: 10.1016/j.jtct.2021.01.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/15/2021] [Accepted: 01/31/2021] [Indexed: 11/24/2022]
Abstract
Mantle cell lymphoma (MCL) is a subtype of B cell non-Hodgkin lymphoma characterized by a heterogeneous clinical presentation. Patients who demonstrate an objective response to induction therapy(ies) and are eligible for intensive therapies are offered an autologous hematopoietic cell transplant (HCT) as front-line consolidation followed by rituximab maintenance. Allogeneic HCT is an option for younger and fit patients with high-risk disease or in patients who have relapsed after autologous HCT. Recent advances in T cell engineering brought chimeric antigen receptor T cell (CAR T) therapy from the bench to the bedside, with brexucabtagene autoleucel being the first CAR T product approved by the US Food and Drug Administration for use in relapsed/refractory MCL. In this comprehensive review, we summarize the literature on available cellular therapies for MCL and present a treatment algorithm that incorporates HCT, autologous or allogeneic, and CAR T therapies.
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Mantle cell lymphoma (MCL) is a heterogenous disease with a variety of morphologic and genetic features, some of which are associated with high risk disease. Here we critically analyze the current state of the understanding of MCL's biology and its implications in therapy, with a focus on chemotherapy-free and targeted therapy regimens. RECENT FINDINGS Mantle cell lymphoma (MCL) is a rare subtype of non-Hodgkin's lymphoma, defined by a hallmark chromosomal translocation t(11;14) which leads to constitutive expression of cyclin D1. Recent discoveries in the biology of MCL have identified a number of factors, including TP53 mutations and complex karyotype, that lead to unresponsiveness to traditional chemoimmunotherapy and poor outcomes. Bruton tyrosine kinase inhibitors, BH3-mimetics and other novel agents thwart survival of the neoplastic B-cells in a manner independent of high-risk mutations and have shown promising activity in relapsed/refractory MCL. These therapies are being investigated in the frontline setting, while optimal responses to chemotherapy-free regimens, particularly in high-risk disease, might require combination approaches. High-risk MCL does not respond well to chemoimmunotherapy. Targeted agents are highly active in the relapsed refractory setting and show promise in high-risk disease. Novel approaches may soon replace the current standard of care in both relapsed and frontline settings.
Collapse
|
19
|
Monga N, Tam C, Garside J, Davids MS, Ward K, Quigley J, Parisi L, Tapprich C. Clinical efficacy and safety of first-line treatments in patients with mantle cell lymphoma: A systematic literature review. Crit Rev Oncol Hematol 2020; 158:103212. [PMID: 33383209 DOI: 10.1016/j.critrevonc.2020.103212] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/31/2020] [Accepted: 12/20/2020] [Indexed: 11/24/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a rare form of non-Hodgkin's lymphoma (NHL) with a median overall survival (OS) of approximately 3-5 years. Systematic literature reviews (SLRs) identified efficacy and safety data for first-line therapies, reported in randomised controlled trials (RCTs) and non-randomised interventional studies (NRISs). Nine and 20 independent studies were included in the RCT and NRISs SLRs, respectively. Differences in the regimens and patient outcomes varied according to patient age and suitability for autologous stem cell transplantation (ASCT). In elderly patients ineligible for transplant, OS ranged from 40 months to 69.6 months. In young transplant-eligible patients, OS ranged from 53 months to 152.4 months. Despite the paucity of directly comparable evidence on the efficacy and safety of MCL therapies, these SLRs highlight that MCL remains a difficult NHL subtype to treat, with short survival highlighting the unmet need for newer treatments that improve patient outcomes.
Collapse
Affiliation(s)
| | - Constantine Tam
- St Vincent's Hospital, Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, Australia
| | | | | | - Katherine Ward
- ICON Global Health Economics and Outcomes Research, Abingdon, UK
| | - Joan Quigley
- ICON Global Health Economics and Outcomes Research, Abingdon, UK
| | | | | |
Collapse
|
20
|
Jain P, Dreyling M, Seymour JF, Wang M. High-Risk Mantle Cell Lymphoma: Definition, Current Challenges, and Management. J Clin Oncol 2020; 38:4302-4316. [DOI: 10.1200/jco.20.02287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Martin Dreyling
- Medizinische Klinik III, Ludwig Maximilian University Klinikum München, München, Germany
| | - John F. Seymour
- Peter MacCallum Cancer Center, Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
21
|
Abstract
Blastoid and pleomorphic mantle cell lymphoma (MCL) are among the worst prognostic, aggressive histology, high-risk variants of MCL, and, in this article, they are presented as blastoid MCL. Blastoid MCL have not been systematically studied, probably due to their rarity. De novo blastoid MCLs have superior outcomes compared with transformed MCL. Compared with classic MCL, extranodal involvement (mainly skin, central nervous system), frequent relapses, and inferior responses to conventional chemoimmunotherapy, BTK inhibitors and venetoclax are frequent in blastoid MCL. KTE-X19 induces excellent response in blastoid MCL. Combinations with novel agents are actively investigated. This article presents a comprehensive review on blastoid MCL in 2020.
Collapse
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, TX 77030, USA
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, TX 77030, USA.
| |
Collapse
|
22
|
Guy D, Kahl BS. Initial and Consolidation Therapy for Younger Patients with Mantle Cell Lymphoma. Hematol Oncol Clin North Am 2020; 34:861-870. [PMID: 32861283 DOI: 10.1016/j.hoc.2020.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mantle cell lymphoma is an incurable B-cell malignancy. Treatment of young fit patients is particularly challenging, because careful consideration should be made when building a long-term treatment strategy that would provide longer remissions and increase patients' quality of life. Most young fit patients achieve long remissions with a combination of immunochemotherapy containing rituximab and high-dose cytarabine, followed by high-dose chemotherapy and autologous stem-cell transplantation. The addition of maintenance therapy with rituximab following autologous stem-cell transplantation prolongs the time to relapse and increases overall survival. Despite an intensive approach, late relapses are common and are usually treated with novel agents.
Collapse
Affiliation(s)
- Daniel Guy
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8056-29, St Louis, MO 63108, USA
| | - Brad S Kahl
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8056-29, St Louis, MO 63108, USA.
| |
Collapse
|
23
|
Kaplan LD, Maurer MJ, Stock W, Bartlett NL, Fulton N, Pettinger A, Byrd JC, Blum KA, LaCasce AS, Hsi ED, Liu YT, Scott DW, Hurd D, Ruppert AS, Hernandez‐Ilizaliturri F, Leonard JP, Cheson BD. Bortezomib consolidation or maintenance following immunochemotherapy and autologous stem cell transplantation for mantle cell lymphoma: CALGB/Alliance 50403. Am J Hematol 2020; 95:583-593. [PMID: 32170769 DOI: 10.1002/ajh.25783] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 12/25/2022]
Abstract
Immunochemotherapy followed by autologous transplant (ASCT) in CALGB/Alliance 59909 achieved a median progression-free survival (PFS) in mantle cell lymphoma (MCL) of 5 years, but late recurrences occurred. We evaluated tolerability and efficacy of adding post-transplant bortezomib consolidation (BC) or maintenance (BM) to this regimen in CALGB/Alliance 50403, a randomized phase II trial. Following augmented-dose R-CHOP/ methotrexate, high-dose cytarabine-based stem cell mobilization, cyclophosphamide/carmustine/etoposide (CBV) autotransplant, and rituximab, patients were randomized to BC (1.3 mg/m2 IV days 1, 4, 8, 11 of a 3-week cycle for four cycles) or BM (1.6 mg/m2 IV once weekly × 4 every 8 weeks for 18 months) beginning day 90. The primary endpoint was PFS, measured from randomization for each arm. Proliferation signature, Ki67, and postinduction minimal residual disease (MRD) in bone marrow were assessed. Of 151 patients enrolled; 118 (80%) underwent ASCT, and 102 (68%) were randomized. Both arms met the primary endpoint, with median PFS significantly greater than 4 years (P < .001). The 8-year PFS estimates in the BC and BM arms were 54.1% (95% CI 40.9%-71.5%) and 64.4% (95% 51.8%-79.0%), respectively. Progression-free survival was significantly longer for transplanted patients on 50403 compared with those on 59909. Both the PFS and OS were significantly better for those who were MRD-negative post-induction. The high risk proliferation signature was associated with adverse outcome. Both BM and BC were efficacious and tolerable, although toxicity was significant. The comparison between studies 50403 and 59909 with long-term follow up suggests a PFS benefit from the addition of BC or BM post- transplant.
Collapse
Affiliation(s)
- Lawrence D. Kaplan
- Medicine/Hematology‐OncologyUniversity of California, San Francisco San Francisco California USA
| | - Matthew J. Maurer
- Alliance Statistics and Data CenterMayo Clinic Rochester Minnesota USA
| | - Wendy Stock
- Medicine, University of Chicago Comprehensive Cancer Center Chicago Illinois USA
| | - Nancy L. Bartlett
- Siteman Cancer CenterWashington University School of Medicine St Louis Missouri USA
| | - Noreen Fulton
- Medicine, University of Chicago Comprehensive Cancer Center Chicago Illinois USA
| | - Adam Pettinger
- Alliance Statistics and Data CenterMayo Clinic Rochester Minnesota USA
| | - John C. Byrd
- Hematology, The Ohio State University Columbus Ohio USA
| | | | - Ann S. LaCasce
- Medical OncologyDana‐Farber/Partners CancerCare Boston Massachusetts USA
| | - Eric D. Hsi
- Department of Laboratory MedicineCleveland Clinic Cleveland Ohio USA
| | - Yi Tian Liu
- Centre for Lymphoid Cancer, British Columbia Cancer Vancouver British Columbia Canada
| | - David W. Scott
- Centre for Lymphoid Cancer, British Columbia Cancer Vancouver British Columbia Canada
| | - David Hurd
- Hematology‐OncologyWake Forest University Health Sciences Winston‐Salem North Carolina USA
| | - Amy S. Ruppert
- Alliance Statistics and Data CenterThe Ohio State University Columbus Ohio USA
| | | | - John P. Leonard
- Department of MedicineWeill Medical College of Cornell University New York New York USA
| | - Bruce D. Cheson
- Hematology‐OncologyMedStar Georgetown University Hospital Washington District of Columbia USA
| |
Collapse
|
24
|
Bair SM, Brandstadter JD, Ayers EC, Stadtmauer EA. Hematopoietic stem cell transplantation for blood cancers in the era of precision medicine and immunotherapy. Cancer 2020; 126:1837-1855. [PMID: 32073653 DOI: 10.1002/cncr.32659] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/08/2019] [Accepted: 09/19/2019] [Indexed: 01/12/2023]
Abstract
Hematopoietic stem cell transplantation (HCT) has been an integral component in the treatment of many hematologic malignancies. Since the development of HCT nearly 50 years ago, the role of this modality has evolved as newer treatment approaches have been developed and integrated into the standard of care. In the last decade, novel and highly active targeted therapies and immunotherapies have been approved for many hematologic malignancies, raising the question of whether HCT continues to retain its prominent role in the treatment paradigms of various hematologic malignancies. In this review, the authors have described the current role of autologous and allogeneic HCT in the treatment of patients with acute leukemias, aggressive B-cell lymphomas, and multiple myeloma and discussed how novel targeted therapies and immunotherapies have changed the potential need, timing, and goal of HCT in patients with these diseases.
Collapse
Affiliation(s)
- Steven M Bair
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua D Brandstadter
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emily C Ayers
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward A Stadtmauer
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Furlong E, Jensen J, Woodard M, Griffiths K, Knight G, Sturm M, Kerr F, Gough H, Bear N, Carter TL, Cole CH, Kotecha RS, Ramachandran S. Optimized peripheral blood progenitor cell mobilization for autologous hematopoietic cell transplantation in children with high-risk and refractory malignancies. Pediatr Transplant 2020; 24:e13602. [PMID: 31631445 DOI: 10.1111/petr.13602] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/15/2019] [Accepted: 09/19/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Autologous hematopoietic stem cell transplantation (aHSCT) using hematopoietic progenitor cells (HPCs) has become an important therapeutic modality for patients with high-risk malignancies. Current literature on standardized method for HPC apheresis in children is sparse and failure rate reported as high as 30%. PATIENTS/METHODS A retrospective study of 125 pediatric patients with high-risk malignancies undergoing aHSCT in Western Australia between 1997 and 2016 was conducted. RESULTS Mobilization was achieved by means of chemotherapy and granulocyte colony-stimulating factor (G-CSF). Patients underwent apheresis the day after CD34+ counts reached ≥20/µL and an additional dose of G-CSF. Peripheral arterial and intravenous lines were inserted in pediatric intensive care unit under local anesthetic and/or sedation, omitting the need for general anesthesia as well as facilitating an uninterrupted apheresis flow. Larger apheresis total blood volumes were processed in patients weighing ≤20 kg. The minimal dose of ≥2 × 106 CD34+ cells/kg was successfully collected in 98.4% of all patients. The optimal dose of 3-5 × 106 CD34+ cells/kg was collected in 96% of patients scheduled for a single aHSCT, 87.5% for tandem, and 100% for triple aHSCT. All HPC collections were completed in one apheresis session. Mobilization after ≤3 chemotherapy cycles and cycles including cyclophosphamide resulted in a significantly higher yield of CD34+ cells. CONCLUSION Our approach to HPC mobilization by means of chemotherapy and single myeloid growth factor combined with optimal collection timing facilitated by continuous apheresis flow resulted in highly effective HPC harvest in children and adolescents with high-risk cancers.
Collapse
Affiliation(s)
- Eliska Furlong
- Department of Paediatric and Adolescent Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia
| | - Jesper Jensen
- PathWest Laboratory Medicine WA, Perth, WA, Australia
| | - Mark Woodard
- Paediatric Intensive Care Unit, Perth Children's Hospital, Perth, WA, Australia.,School of Nursing, Midwifery and Paramedicine, Curtin University, Perth, WA, Australia
| | - Katherine Griffiths
- Paediatric Intensive Care Unit, Perth Children's Hospital, Perth, WA, Australia
| | - Geoff Knight
- Paediatric Intensive Care Unit, Perth Children's Hospital, Perth, WA, Australia.,Division of Paediatrics, School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Marian Sturm
- Cell and Tissue Therapy, Royal Perth Hospital, Perth, WA, Australia
| | - Fiona Kerr
- Department of Paediatric and Adolescent Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia
| | - Hazel Gough
- Department of Paediatric and Adolescent Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia
| | - Natasha Bear
- Department of Clinical Research and Education, Perth Children's Hospital, Perth, WA, Australia
| | - Tina L Carter
- Department of Paediatric and Adolescent Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia.,PathWest Laboratory Medicine WA, Perth, WA, Australia.,Division of Paediatrics, School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Catherine H Cole
- Division of Paediatrics, School of Medicine, University of Western Australia, Perth, WA, Australia.,Paediatric Haematology and Oncology, Sidra Medicine, Doha, Qatar
| | - Rishi S Kotecha
- Department of Paediatric and Adolescent Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia.,Division of Paediatrics, School of Medicine, University of Western Australia, Perth, WA, Australia.,Division of Children's Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Shanti Ramachandran
- Department of Paediatric and Adolescent Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia.,Division of Paediatrics, School of Medicine, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
26
|
Wang JD, Katz SG, Morgan EA, Yang DT, Pan X, Xu ML. Proapoptotic protein BIM as a novel prognostic marker in mantle cell lymphoma. Hum Pathol 2019; 93:54-64. [PMID: 31425695 PMCID: PMC7038910 DOI: 10.1016/j.humpath.2019.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/31/2019] [Accepted: 08/07/2019] [Indexed: 12/20/2022]
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell lymphoma. Numerous studies have demonstrated many genetic aberrations in MCL in addition to the characteristic t(11:14), including frequent biallelic deletions of Bim, a proapoptotic member of the BCL-2 family. In mice, Bim deletion coupled with cyclin D1 overexpression generates pathologic and molecular features of human MCL. Since the regulation of apoptosis is crucial in MCL pathogenesis, we hypothesize that BIM expression may be associated with tumor cell survival. Clinical data and tissue from 100 nodal MCL cases between 1988 and 2009 were collected from three large academic medical centers. The average patient age of our MCL cohort was 65.5 years old (range, 42-97) with a 2:1 male to female ratio. Immunohistochemistry was performed with a validated anti-BIM antibody. Patients were separated into low and high BIM-expressing categories with a cutoff of 80%. As expected for a proapoptotic tumor suppressor, patients with high BIM expression were less likely to have progressive disease and more likely to have a complete response (P = .022). In addition, high BIM-expressing MCL tumors revealed a trend toward increased overall survival with this trend persisting in sub-analysis of Ann Arbor stages III and IV. No correlation between BIM expression, Ki-67 index, and MIPI score was observed, suggesting a role for BIM as a novel independent prognostic factor. While BIM is only one member of a complex family of apoptosis-regulating proteins, these findings may yield clinically relevant information for the prognosis and therapeutic susceptibility of MCL.
Collapse
Affiliation(s)
- Jeff D Wang
- Department of Pathology, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT 06510.
| | - Samuel G Katz
- Department of Pathology, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT 06510.
| | - Elizabeth A Morgan
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115.
| | - David T Yang
- Department of Pathology, University of Wisconsin Medical Center, Madison, WI 53705-2281.
| | - Xueliang Pan
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210.
| | - Mina L Xu
- Department of Pathology, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT 06510.
| |
Collapse
|
27
|
|
28
|
Dahi PB, Lazarus HM, Sauter CS, Giralt SA. Strategies to improve outcomes of autologous hematopoietic cell transplant in lymphoma. Bone Marrow Transplant 2019; 54:943-960. [PMID: 30390059 PMCID: PMC9062884 DOI: 10.1038/s41409-018-0378-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/05/2018] [Accepted: 09/30/2018] [Indexed: 11/08/2022]
Abstract
High-dose chemotherapy and autologous hematopoietic cell transplantation (HDT-AHCT) remains an effective therapy in lymphoma. Over the past several decades, HDT with BEAM (carmustine, etoposide, cytarabine, and melphalan) and CBV (cyclophosphamide, carmustine, and etoposide) have been the most frequently used preparatory regimens for AHCT in Hodgkin (HL) and non-Hodgkin lymphoma (NHL). This article reviews alternative combination conditioning regimens, as well as novel transplant strategies that have been developed, to reduce transplant-related toxicity while maintaining or improving efficacy. These data demonstrate that incorporation of maintenance therapy posttransplant might be the best way to improve outcomes.
Collapse
Affiliation(s)
- Parastoo B Dahi
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | - Hillard M Lazarus
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Craig S Sauter
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Sergio A Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
29
|
Affiliation(s)
- I Brian Greenwell
- a Division of Hematology and Medical Oncology , Medical University of South Carolina Hollings Cancer Center , Charleston , SC , USA
| | - Jonathon B Cohen
- b Department of Hematology and Medical Oncology , Emory University Winship Cancer Institute , Atlanta , GA , USA
| |
Collapse
|
30
|
Mori S, Patel RD, Ahmad S, Varela J, Smith T, Altoos R, Shen Q, Goldstein SC, Persky DO. Aggressive Leukemic Non-Nodal Mantle Cell Lymphoma With P53 Gene Rearrangement/Mutation is Highly Responsive to Rituximab/Ibrutinib Combination Therapy. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:e93-e97. [DOI: 10.1016/j.clml.2018.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/01/2018] [Accepted: 11/02/2018] [Indexed: 01/03/2023]
|
31
|
Blastoid and pleomorphic mantle cell lymphoma: still a diagnostic and therapeutic challenge! Blood 2018; 132:2722-2729. [DOI: 10.1182/blood-2017-08-737502] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/26/2018] [Indexed: 01/28/2023] Open
Abstract
Abstract
Blastoid mantle cell lymphoma is characterized by highly aggressive features and a dismal clinical course. These blastoid and pleomorphic variants are defined by cytomorphological features, but the criteria are somewhat subjective. The diagnosis may be supported by a high cell proliferation based on the Ki-67 labeling index. Recent analyses have shown that the Ki-67 index overrules the prognostic information derived from the cytology subtypes. Nevertheless, genetic analysis suggests that blastoid and pleomorphic variants are distinct from classical mantle cell lymphoma. In clinical cohorts, the frequency of these subsets varies widely but probably represents ∼10% of all cases. Chemotherapy regimens commonly used in mantle cell lymphoma, such as bendamustine, rarely achieve prolonged remissions when given at the dosage developed for classical variants of the disease. Thus, high-dose cytarabine–containing regimens with high-dose consolidation may be generally recommended based on the more aggressive clinical course in these patients. However, even with these intensified regimens, the long-term outcome seems to be impaired. Thus, especially in this patient subset, allogeneic transplantation may be discussed at an early time point in disease management. Accordingly, targeted approaches are warranted in these patients, but clinical data are scarce. Ibrutinib treatment results in high rates of responses, but the median duration of remission is <6 months. Similarly, lenalidomide and temsirolimus result in only short-term remissions. Novel approaches, such as chimeric antigenic receptor T cells, may have the potential to finally improve the dismal long-term outcome of these patients.
Collapse
|
32
|
Smith SD, Gandhy S, Gopal AK, Reddy P, Shadman M, Till BG, Lynch RC, Kanan S, Cowan A, Low L, Hill BT. Modified VR-CAP, Alternating With Rituximab and High-dose Cytarabine: An Effective Pre-transplant Induction Regimen for Mantle Cell Lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 19:48-52. [PMID: 30409719 DOI: 10.1016/j.clml.2018.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/18/2018] [Accepted: 10/06/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND Initial treatment of mantle cell lymphoma (MCL) incorporating autologous stem cell transplantation affords long-term remissions, but relapses still occur. Optimal pretransplant therapy will afford high complete response rates and not impair stem cell collection. Incorporation of bortezomib represents a natural evolution of pretransplant therapy, given its proven first-line efficacy and minimal impact on stem cell collection. PATIENTS AND METHODS At the University of Washington/Seattle Cancer Care Alliance and the Cleveland Clinic Foundation, we developed modified VR-CAP/R+ara-C (bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone, alternating with rituximab and high-dose cytarabine), for transplant-eligible patients with MCL. This regimen was administered as standard-of-care, pretransplant therapy to consecutive patients with MCL from April 2015 to the present. RESULTS A total of 37 patients were treated with this regimen, including 18 at the University of Washington/Seattle Cancer Care Alliance and 19 at the Cleveland Clinic Foundation. Most patients had intermediate- or high-risk disease by both (mantle-cell lymphoma international prognostic index (MIPI)-B and MIPI-C category. Complete response to induction was achieved in 32 (86%) of 37 evaluable patients; 2 achieved partial response, and 3 had primary refractory disease. Stem cell collection was successful in 1 attempt in 30 of 32 patients. The median follow-up of survivors measured from start of treatment is 17.4 months. Five patients have progressed, and 4 have died (2 owing to lymphoma, 2 from toxicity). CONCLUSION Modified VR-CAP/R+ara-C is feasible pretransplant therapy for patients with MCL and is associated with a high rate of complete response and eligibility for autologous stem cell transplantation.
Collapse
Affiliation(s)
- Stephen D Smith
- Department of Medicine, University of Washington/Seattle Cancer Care Alliance, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA.
| | - Shruti Gandhy
- Taussig Cancer Institute, Hematologic Oncology and Blood Disorders, Cleveland Clinic, Cleveland, OH
| | - Ajay K Gopal
- Department of Medicine, University of Washington/Seattle Cancer Care Alliance, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Prathima Reddy
- CHI Franciscan Health, Franciscan Inpatient Services, Federal Way, WA
| | - Mazyar Shadman
- Department of Medicine, University of Washington/Seattle Cancer Care Alliance, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Brian G Till
- Department of Medicine, University of Washington/Seattle Cancer Care Alliance, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Ryan C Lynch
- Department of Medicine, University of Washington/Seattle Cancer Care Alliance, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Sandra Kanan
- Department of Medicine, University of Washington/Seattle Cancer Care Alliance, Seattle, WA
| | - Andrew Cowan
- Department of Medicine, University of Washington/Seattle Cancer Care Alliance, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Lauren Low
- Department of Medicine, University of Washington/Seattle Cancer Care Alliance, Seattle, WA
| | - Brian T Hill
- Taussig Cancer Institute, Hematologic Oncology and Blood Disorders, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
33
|
Ye H, Desai A, Zeng D, Romaguera J, Wang ML. Frontline Treatment for Older Patients with Mantle Cell Lymphoma. Oncologist 2018; 23:1337-1348. [PMID: 29895632 PMCID: PMC6291324 DOI: 10.1634/theoncologist.2017-0470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 04/05/2018] [Indexed: 11/17/2022] Open
Abstract
The natural history of mantle cell lymphoma (MCL) undergoing chemotherapy is a cyclical pattern of remission followed by recurrence of disease due to acquired chemotherapy resistance. The median age of the occurrence of MCL is 65 years, so half of the newly diagnosed MCL patients are considered "elderly." The tolerance to long-term chemotherapy in elderly patients is decreased; hence, the response to frontline therapy used is of paramount importance. We hope that our review may guide clinicians in treating such populations in a more personalized and evidence-based manner.In the older patients with risk variables, frontline treatment is determined according to different body status of fit, unfit or compromised, and frail. In the fit patients, the pursuit of remission and prolongation of survival might currently justify the use of more intense and toxic therapies. For unfit or compromised older patients, disease control needs to be prioritized, maintaining a balance between the benefits and toxicities of the treatment. For frail patients, tolerance of treatment and minimizing myelotoxicity should be the primary focus. "Chemotherapy-free" regimens are likely to be considered as the first-line strategy for this population. On the other hand, in the older MCL population without risk variables, observation or "watch and wait" can prevent overtreatment. Furthermore, more clinical trials and research studies on novel agents and targeted therapies need to be translated into the general population to provide optimal treatment and to guide personalized treatment. IMPLICATIONS FOR PRACTICE: This review emphasizes the importance of frontline therapies for older MCL patients. MCL patients commonly experience a cyclical pattern of remission followed by recurrence of disease due to acquired chemotherapy resistance. As a special population, elderly patients have various comorbidities and decreased organ function, which may reduce the chances of undergoing treatment for recurrent disease. Thus, this older population of patients with MCL should be treated separately and exceptionally. So far, systematic reviews with regard to frontline treatment for older patients with MCL have not been encountered, but the hope is that this review may guide clinicians in treating such populations in a more personalized and evidence-based manner.
Collapse
Affiliation(s)
- Haige Ye
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Aakash Desai
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas Health Science Center, Houston, Texas, USA
| | - Dongfeng Zeng
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jorge Romaguera
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael L Wang
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
34
|
Hilal T, Wang Z, Almader-Douglas D, Rosenthal A, Reeder CB, Jain T. Rituximab maintenance therapy for mantle cell lymphoma: A systematic review and meta-analysis. Am J Hematol 2018; 93:1220-1226. [PMID: 30033656 DOI: 10.1002/ajh.25226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 01/11/2023]
Abstract
Mantle cell lymphoma is characterized by relapse and progressive disease, despite initial response to chemoimmunotherapy. We conducted a systematic review and meta-analysis to determine the efficacy of rituximab maintenance (RM) therapy in patients with mantle cell lymphoma. We searched PubMed, Embase and Cochrane Central Register of Controlled Trials from database inception through November 1, 2017. Only full-text articles were included. Prespecified data elements were extracted from each trial. Outcomes of interest included progression-free survival (PFS) and overall survival (OS). The overall effect was pooled using the Der Simonian-Laird random effects model. Three randomized controlled trials and four observational studies met our inclusion criteria and were identified in the analyses. Six studies compared RM therapy to observation, and one compared RM therapy to interferon alfa. Meta-analysis evaluating outcomes of patients treated after ASCT revealed that RM improved for both PFS (HR = 0.33, 95% CI = 0.23-0.49) and OS (HR of death = 0.35, 95% CI = 0.17-0.69). A second meta-analysis of studies evaluating outcomes of patients who are ASCT-ineligible treated with anthracycline-based induction therapy revealed that RM improved PFS (HR = 0.38, 95% CI = 0.25-0.58). There is a paucity of data on the role of RM in ASCT-ineligible patients and those with relapsed disease. Overall, RM therapy appears to improve PFS and OS in previously untreated patients with mantle cell lymphoma who undergo induction chemoimmunotherapy followed by ASCT.
Collapse
Affiliation(s)
- Talal Hilal
- Department of Internal Medicine, Division of Hematology/Oncology, Mayo Clinic, Phoenix, Arizona
| | - Zhen Wang
- Evidence-Based Practice Center, Mayo Clinic, Rochester, Minnesota
| | | | - Allison Rosenthal
- Department of Internal Medicine, Division of Hematology/Oncology, Mayo Clinic, Phoenix, Arizona
| | - Craig B Reeder
- Department of Internal Medicine, Division of Hematology/Oncology, Mayo Clinic, Phoenix, Arizona
| | - Tania Jain
- Department of Internal Medicine, Division of Hematology/Oncology, Mayo Clinic, Phoenix, Arizona
| |
Collapse
|
35
|
Ye H, Desai A, Huang S, Jung D, Champlin R, Zeng D, Yan F, Nomie K, Romaguera J, Ahmed M, Wang ML. Paramount therapy for young and fit patients with mantle cell lymphoma: strategies for front-line therapy. J Exp Clin Cancer Res 2018; 37:150. [PMID: 30005678 PMCID: PMC6044039 DOI: 10.1186/s13046-018-0800-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/18/2018] [Indexed: 11/16/2022] Open
Abstract
The natural history of mantle cell lymphoma (MCL) is a continuous process with the vicious cycle of remission and recurrence. Because MCL cells are most vulnerable before their exposure to therapeutic agents, front-line therapy could eliminate MCL cells at the first strike, reduce the chance for secondary resistance, and cause long-term remissions. If optimized, it could become an alternative to cure MCL. The key is the intensity of front-line therapy. Both the Nordic 2 and the MD Anderson Cancer Center HCVAD trials, with follow-up times greater than 10 years, achieved long-term survivals exceeding 10 years. But the Achilles heel in both trials were the severe toxicities, such as secondary malignancies including myelodysplastic syndromes /leukemia. Therefore, intensive therapies can act as a double-edged sword providing long term survival at the cost of severe toxicities. In our opinion, although intensive chemotherapy can cause detrimental side effects, it is indispensable given that we run the risk of sacrificing long-term survivals in these young and fit patients. We must seek for a powerful alternative at the front-line. Furthermore, minimal residual disease negativity should be the optimal therapeutic goal to achieve before and after autologous stem cell transplantation. Some novel therapeutic strategies have shown to improve outcomes, but it is not yet clear as to how these results translate in population. Of note, MCL patients need to be stratified at diagnosis and be provided with different intensities of front-line regimen. In this review, we discuss current strategies for the treatment of young patients with newly diagnosed MCL.
Collapse
Affiliation(s)
- Haige Ye
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Aakash Desai
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
- University of Texas Health Science Centre at Houston, Houston, TX, USA
| | - Shengjian Huang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Dayoung Jung
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Richard Champlin
- Division of Cancer Medicine, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dongfeng Zeng
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Fangfang Yan
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Krystle Nomie
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jorge Romaguera
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Makhdum Ahmed
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Michael L Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
36
|
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: 0.7] [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
|
37
|
McKay P, Leach M, Jackson B, Robinson S, Rule S. Guideline for the management of mantle cell lymphoma. Br J Haematol 2018; 182:46-62. [PMID: 29767454 DOI: 10.1111/bjh.15283] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Pamela McKay
- Department of Haematology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Mike Leach
- Department of Haematology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Bob Jackson
- Department of Pathology, Queen Elizabeth University Hospital, Glasgow, UK
| | - Stephen Robinson
- Department of Haematology, University Hospitals Bristol, Bristol, UK
| | - Simon Rule
- Department of Haematology, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| |
Collapse
|
38
|
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: 1.3] [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
|
39
|
Long-term outcome analysis of reduced-intensity allogeneic stem cell transplantation in patients with mantle cell lymphoma: a retrospective study from the EBMT Lymphoma Working Party. Bone Marrow Transplant 2018; 53:617-624. [PMID: 29335632 DOI: 10.1038/s41409-017-0067-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/13/2017] [Accepted: 10/31/2017] [Indexed: 11/09/2022]
Abstract
Reduced-intensity allogeneic stem cell transplantation (RIST) is usually reserved for patients with mantle cell lymphoma who relapse after an autoSCT. However, the long-term efficacy of RIST and its curative potential have not been clearly demonstrated. We studied the long-term outcome of patients receiving a RIST for MCL as reported to the EBMT. A total of 324 patients, median age 57 years (range 31-70), underwent a RIST between 2000 and 2008; 43% of the patients had received >3 lines of prior therapy, including an autoSCT in 46%. Non-relapse mortality (NRM) was 10% at 100 days and 24% at 1 year and was lower for patients receiving anti-thymocyte globulin (ATG)/ALG (RR 0.59, p = 0.046). After a median follow-up of 72 months (range 3-159), 118 patients relapsed at a median of 8 months post RIST (range 1-117). The cumulative incidence of relapse was 25% and 40% at 1 and 5 years, respectively, and was associated with chemorefractory disease (HR 0.49, p = 0.01) and the use of CAMPATH (HR 2.59, p = 0.0002). The 4-year progression-free survival rate and overall survival rate was 31 and 40%, respectively. RIST results in long-term disease-free survival in about 30% of the patients, including those patients relapsing after a prior autoSCT.
Collapse
|
40
|
|
41
|
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: 1.6] [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
|
42
|
Efficacy of Standard Dose R-CHOP Alternating With R-HDAC Followed by Autologous Hematopoietic Cell Transplantation as Initial Therapy of Mantle Cell Lymphoma, a Single-Institution Experience. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 18:e95-e102. [PMID: 29208403 DOI: 10.1016/j.clml.2017.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/15/2017] [Accepted: 11/08/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Young fit patients with mantle cell lymphoma (MCL) are commonly treated with induction chemotherapy followed by high-dose chemotherapy and autologous hematopoietic cell transplantation (AHCT). Induction regimens with modifications of R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) and/or incorporation of high-dose cytarabine (HDAC) appear more effective than R-CHOP alone. PATIENTS AND METHODS We adopted a modification of the Nordic protocol using standard, rather than higher dose R-CHOP, alternating with HDAC (rituximab plus HDAC), for 3 cycles each or, for patients already treated with R-CHOP alone before referral for AHCT, an additional 2 cycles of rituximab plus HDAC. We herein report our experience with 28 patients treated with this regimen who proceeded to AHCT, and compare their outcomes with patients treated with either standard-dose R-CHOP (n = 38) or R-HCVAD/MA (cyclophosphamide, vincristine, doxorubicin, dexamethasone alternating with methotrexate, and cytarabine; n = 21), before AHCT. RESULTS With a median follow-up duration of 26 months, our data show that this modification of the Nordic regimen is a highly effective pre-AHCT first-line therapy for MCL (3-year progression-free and overall survival rates of 69% and 75%, respectively). CONCLUSION By using a less intense induction, this regimen can serve as a platform for combined use of novel agents, with less risk of additive toxicity.
Collapse
|
43
|
Abstract
In contemporary clinical practice, almost all allogeneic transplantations and autologous transplantations now capitalize on peripheral blood stem cells (PBSCs) as opposed to bone marrow (BM) for the source of stem cells. In this context, granulocyte colony-stimulating factor (G-CSF) plays a pivotal role as the most frequently applied frontline agent for stem cell mobilization. For patients classified as high-risk, chemotherapy based mobilization regimens can be preferred as a first choice and it is notable that this also used for remobilization. Mobilization failure occurs at a rate of 10%-40% with traditional strategies and it typically leads to low-efficiency practices, resource wastage, and delayed in treatment intervention. Notably, however, several factors can impact the effectiveness of CD34+ progenitor cell mobilization, including patient age and medical history (prior chemotherapy or radiotherapy, disease and marrow infiltration at the time of mobilization). In recent years, main (yet largely ineffective) approach was to increase G-CSF dose and add SCF, but novel and promising pathways have been opened up by the synergistic impact of a reversible inhibitor of CXCR4, plerixafor, with G-CSF. The literature shows to its favorable results in upfront and failed mobilizers, and it is necessary to use plerixafor (or equivalent agents) to optimize HSC harvest in poor mobilizers. Different CXCR4 inhibitors, growth hormone, VLA4 inhibitors, and parathormone, have been cited as new agents for mobilization failure in recent years. In view of the above considerations, the purpose of this paper is to examine the mobilization of PBSC while focusing specifically on poor mobilizers.
Collapse
Affiliation(s)
- Sinem Namdaroglu
- Izmir Bozyaka Training and Research Hospital, Department of Hematology, Izmir, Turkey.
| | - Serdal Korkmaz
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology, BMT Unit, Ankara, Turkey
| | - Fevzi Altuntas
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology, BMT Unit, Ankara, Turkey; Yıldırım Beyazıt University, Medical Faculty, Department of Hematology, Ankara, Turkey
| |
Collapse
|
44
|
Autologous hematopoietic cell transplantation of mantle cell lymphoma: emerging trends. Curr Opin Hematol 2017; 24:502-508. [PMID: 28832354 DOI: 10.1097/moh.0000000000000383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The management of mantle cell lymphoma has changed significantly with the adoption of immunochemotherapy and dose intensive treatment strategies in specific patient populations. Randomized controlled trials have established the role of rituximab-based treatments and autologous stem cell transplantation as standards of care. Novel therapeutics are also being integrated into these treatment strategies. RECENT FINDINGS Rituximab-based primary treatment has been shown to significantly improve complete remission rates. The addition of autologous stem cell transplantation has also improved progression-free survival (PFS) although data regarding potential overall survival (OS) benefits are not clear. Complete remission and minimal residual disease (MRD) negative disease states are predictive of outcome. Rituximab maintenance post SCT has also been shown to significantly improves PFS and OS. SUMMARY Current therapeutic standards in mantle cell lymphoma have clearly improved patient outcomes with improvements in remission rates, PFS, and OS. Autologous stem cell transplant (ASCT) as a consolidation strategy of primary treatment has improved outcomes, and the incorporation of novel drugs into frontline therapy may further improve the efficacy of the treatment. MRD-driven strategies may ultimately define appropriate patient subsets towards ASCT or alternative approaches.
Collapse
|
45
|
Staton AD, Langston AA. Autologous Stem Cell Transplant: Still the Standard for Fit Patients With Mantle Cell Lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17S:S96-S99. [PMID: 28760309 DOI: 10.1016/j.clml.2017.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 02/28/2017] [Indexed: 12/22/2022]
Abstract
Mantle cell lymphoma is a relatively rare malignancy, comprising fewer than 10% of all non-Hodgkin lymphomas. It is a heterogeneous disease, and although most patients experience an aggressive clinical course, some have a more indolent disease and may not require immediate therapy. There are currently few reliable prognostic markers, making it difficult to accurately predict which patients require early intensive treatment. We argue that consolidative autologous stem cell transplantation in first remission remains the standard of care for the young and fit patient population, based on long-term data from phase II and III trials demonstrating that early transplantation extends both progression-free and overall survival. Novel targeted agents are currently being investigated in both the upfront and relapse settings, but to date there are few data to suggest durable treatment responses that compare favorably with results of transplantation.
Collapse
Affiliation(s)
- Ashley D Staton
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA
| | - Amelia A Langston
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA.
| |
Collapse
|
46
|
Fakhri B, Kahl B. Current and emerging treatment options for mantle cell lymphoma. Ther Adv Hematol 2017; 8:223-234. [PMID: 28811872 DOI: 10.1177/2040620717719616] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/08/2017] [Indexed: 11/17/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a B-cell non-Hodgkin lymphoma with typically aggressive behavior. The genetic signature is the chromosomal translocation t(11;14)(q13;q32) resulting in overexpression of cyclin D1. Asymptomatic newly diagnosed MCL patients with low tumor burden can be closely observed, deferring therapy to the time of disease progression. Although MCL classically responds to upfront chemotherapy, it remains incurable with standard approaches. For patients in need of frontline therapy, the initial decision is whether to proceed with an intensive treatment strategy or a non-intensive treatment strategy. In general, given the unfavorable risk-benefit profile, older MCL patients should be spared intensive strategies, while younger and fit patients can be considered for intensive strategies. The bendamustine and rituximab (BR) regimen is becoming an increasingly popular treatment option among the elderly population, with improved progression-free survival (PFS) and acceptable side-effect profile. Although rituximab maintenance after R-CHOP improves survival outcomes in elderly patients, no clinical trial to date has shown statistical significance to support the use of rituximab maintenance after BR induction in older patients. In young and fit patients with MCL, an intensive strategy to maximize the length of first remission has emerged as a worldwide standard of care. With current high-dose cytarabine-containing immunochemotherapy regimens followed by autologous stem cell transplantation, the median PFS has exceeded 7 years. In the relapsed or refractory (R/R) setting, reduced intensity conditioning allogeneic hematopoietic stem cell transplantation may offer the highest likelihood of long-term survival in young R/R MCL patients, at the cost of increased risk of non-relapse mortality and chronic graft versus host disease. Novel agents targeting activated pathways in MCL cells, such as bortezomib, lenalidamide, ibrutinib and temsirolimus are now available for the management of R/R disease.
Collapse
Affiliation(s)
- Bita Fakhri
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Brad Kahl
- 660 South Euclid Ave, Campus Box 8056, Saint Louis, MO 63110, USA
| |
Collapse
|
47
|
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
|
48
|
Carnevale J, Rubenstein JL. The Challenge of Primary Central Nervous System Lymphoma. Hematol Oncol Clin North Am 2017; 30:1293-1316. [PMID: 27888882 DOI: 10.1016/j.hoc.2016.07.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Primary central nervous system (CNS) lymphoma is a challenging subtypes of aggressive non-Hodgkin lymphoma. Emerging clinical data suggest that optimized outcomes are achieved with dose-intensive CNS-penetrant chemotherapy and avoiding whole brain radiotherapy. Anti-CD20 antibody-based immunotherapy as a component of high-dose methotrexate-based induction programs may contribute to improved outcomes. An accumulation of insights into the molecular and cellular basis of disease pathogenesis is providing a foundation for the generation of molecular tools to facilitate diagnosis as well as a roadmap for integration of targeted therapy within the developing therapeutic armamentarium for this challenging brain tumor.
Collapse
Affiliation(s)
- Julia Carnevale
- Division of Hematology/Oncology, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143, USA
| | - James L Rubenstein
- Division of Hematology/Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, M1282 Box 1270, San Francisco, CA 94143, USA.
| |
Collapse
|
49
|
Mantle Cell Lymphoma: Contemporary Diagnostic and Treatment Perspectives in the Age of Personalized Medicine. Hematol Oncol Stem Cell Ther 2017; 10:99-115. [PMID: 28404221 DOI: 10.1016/j.hemonc.2017.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 02/01/2017] [Accepted: 02/20/2017] [Indexed: 11/22/2022] Open
Abstract
Mantle cell lymphoma is a clinically heterogeneous disease occurring within a heterogeneous patient population, highlighting a need for personalized therapy to ensure optimal outcomes. It is therefore critical to understand the benefits and risks associated with both intensive and deintensified approaches. In the following review we provide a therapeutic roadmap to strategically guide treatment for newly diagnosed and relapsed/refractory patients highlighting pivotal and recently published results involving known and novel therapies.
Collapse
|
50
|
Kharfan-Dabaja MA, El-Jurdi N, Ayala E, Kanate AS, Savani BN, Hamadani M. Is myeloablative dose intensity necessary in allogeneic hematopoietic cell transplantation for lymphomas? Bone Marrow Transplant 2017; 52:1487-1494. [PMID: 28368373 DOI: 10.1038/bmt.2017.55] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 12/21/2016] [Accepted: 01/08/2017] [Indexed: 12/25/2022]
Abstract
The advent of novel immunotherapy and tyrosine kinase inhibitors has ushered a new era in the treatment of Hodgkin and non-Hodgkin lymphomas. Allogeneic hematopoietic cell transplantation remains, however, a vital component in the management and potential cure of lymphomas, especially in the relapsed setting. Considering the biological and clinical heterogeneity of various subtypes of lymphomas, the optimal intensity of conditioning regimens remains controversial. Reduced intensity conditioning regimens have broadened applicability of the procedure to older and frail patients. Observational studies suggest that although reduced intensity allografting is associated with higher risk of relapse, overall survival is comparable and in some cases even better, than observed with myeloablative regimens. Here, we review the available published data pertaining to allogeneic hematopoietic cell transplantation using reduced intensity or myeloablative conditioning for various lymphoma histologies. Owing to the lack of randomized prospective trials, recommendations are mainly based on registry and single-institution studies. Special emphasis must be given to implementing strategies to prevent relapse when using reduced intensity regimens. Identifying particular patients who may benefit from myeloablative regimens in lymphomas remains to be better defined.
Collapse
Affiliation(s)
- M A Kharfan-Dabaja
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - N El-Jurdi
- Division of Hematology-Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | - E Ayala
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - A S Kanate
- Osborn Hematopoietic Malignancy and Transplantation Program, West Virginia University, Morgantown, WV, USA
| | - B N Savani
- Division of Hematology and Oncology and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Hamadani
- Division of Hematology-Oncology, Department of Internal Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|