51
|
Lenalidomide maintenance after autologous haematopoietic stem-cell transplantation in mantle cell lymphoma: results of a Fondazione Italiana Linfomi (FIL) multicentre, randomised, phase 3 trial. LANCET HAEMATOLOGY 2020; 8:e34-e44. [PMID: 33357480 DOI: 10.1016/s2352-3026(20)30358-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Fit patients with mantle cell lymphoma aged 18-65 years are usually given cytarabine and rituximab-based induction regimens followed by autologous haematopoetic stem-cell transplantation (HSCT). We investigated whether post-autologous HSCT maintenance with lenalidomide improves progression-free survival in this population. METHODS This open-label, randomised, multicentre, phase 3 trial was done at 49 haematology and oncology units in Italy and Portugal. Eligible patients had Ann Arbor stage III or IV treatment-naive mantle cell lymphoma (or stage II plus bulky disease [≥5 cm] or B symptoms), and had evidence of cyclin D1 overexpression or the translocation t(11;14)(q13;q32). Patients were aged 18-59 years with Eastern Cooperative Oncology Group (ECOG) performance status 0-3, or aged 60-65 years with ECOG 0-2. After an optional prephase with vincristine and steroids (intravenous vincristine 1·4 mg/m2 on day 1, oral prednisone 100 mg [total dose] on days 1-5), patients were given three courses of R-CHOP (21-day cycle, intravenous rituximab 375 mg/m2 on day 1; intravenous doxorubicin 50 mg/m2, vincristine 1·4 mg/m2, and cyclophosphamide 750 mg/m2 on day 2; oral prednisone 100 mg/m2 on day 2-6). Patients then received one cycle of high-dose CTX (intravenous cyclophosphamide 4 g/m2 on day 1, intravenous rituximab 375 mg/m2 on day 4). After restaging, patients received two cycles of R-HD-cytarabine (high-dose intravenous cytarabine 2 g/m2 every 12 h on days 1-3, intravenous rituximab 375 mg/m2 on days 4 and 10). Patients with complete remission or partial remission proceeded to autologous HSCT and responding patients (complete remission or partial remission) with haematological recovery were randomly assigned (1:1) to receive 24 courses of oral lenalidomide maintenance (15 mg per day for patients with platelets >100 × 109 cells per L or 10 mg per day for platelets 60-100 × 109 cells per L, days 1-21 every 28 days) for 24 months, or observation. The primary endpoint was progression-free survival, measured in the randomised population. This study is registered with EudraCT (2009-012807-25) and ClinicalTrials.gov (NCT02354313). FINDINGS Between May 4, 2010, and Aug 24, 2015, 303 patients were screened for inclusion and 300 patients were enrolled (median age 57 years, IQR 51-62; 235 [78%] male). 95 patients were excluded before randomisation, mostly due to disease progression, adverse events, and inadequate recovery. 104 patients were randomly assigned to the lenalidomide maintenance group and 101 patients to the observation group. 11 (11%) of 104 patients assigned to lenalidomide did not start treatment (3 withdrew, 6 adverse events or protocol breach, 2 lost to follow-up). At a median follow-up of 38 months after randomisation (IQR 24-50), 3-year progression-free survival was 80% (95% CI 70-87) in the lenalidomide group versus 64% (53-73) in the observation group (log-rank test p=0·012; hazard ratio 0·51, 95% CI 0·30-0·87). 41 (39%) of 104 patients discontinued lenalidomide for reasons including death or progression. Treatment-related deaths were recorded in two (2%) of 93 patients in the lenalidomide group (1 pneumonia, 1 thrombotic thrombocytopenic purpura), and one (1%) of 101 in the observation group (pneumonia). 59 (63%) of 93 patients in the lenalidomide group had grade 3-4 haematological adverse events versus 12 (12%) of 101 patients in the observation group (p<0·0001). 29 (31%) of 93 patients in the lenalidomide group and eight (8%) of 101 patients in the observation group had grade 3-4 non-haematological adverse events (p<0·0001), of which infections were the most common.Serious adverse events were reported in 22 (24%) of 93 patients in the lenalidomide group and five (5%) of 101 patients in the observation group. Pneumonia and other infections were the most common serious adverse events. INTERPRETATION Despite non-negligibile toxicity, lenalidomide after autologous HSCT improved progression-free survival in patients with mantle cell lymphoma, highlighting the role of maintenance in mantle cell lymphoma. FUNDING Fondazione Italiana Linfomi and Celgene.
Collapse
|
52
|
Bhella S, Varela NP, Aw A, Bredeson C, Cheung M, Crump M, Fraser G, Sajkowski S, Kouroukis T. First-line therapy, autologous stem-cell transplantation, and post-transplantation maintenance in the management of newly diagnosed mantle cell lymphoma. Curr Oncol 2020; 27:e632-e644. [PMID: 33380879 PMCID: PMC7755434 DOI: 10.3747/co.27.7053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background In Ontario, no clearly defined standard of care for the management of mantle cell lymphoma (mcl) has been developed, and substantial variability from centre to centre is evident. This guidance document was prompted by the need to harmonize practice in Ontario with respect to first-line, conditioning, and post-transplantation maintenance therapy for patients newly diagnosed with transplantation-eligible mcl. Methods The medline and embase databases were systematically searched from January 2013 to January 2020 for evidence, and the best available evidence was used to draft recommendations relevant to first-line therapy, autologous stem-cell transplantation, and post-transplantation maintenance in the management of transplantation-eligible newly diagnosed mcl. Final approval of this guidance document was obtained from the Stem Cell Transplant Advisory Committee. Recommendations These recommendations apply to all cases of transplantation-eligible newly diagnosed mcl:■ Alternating cycles of r-chop (rituximab plus cyclophosphamide-doxorubicin-vincristine-prednisolone) and r-dhap [rituximab plus dexamethasone-high-dose cytarabine-cisplatin] is the recommended first-line treatment for symptomatic patients newly diagnosed with mcl before autologous stem-cell transplantation (asct).■ Rituximab plus hyperfractionated cyclophosphamide-vincristine-doxorubicin-dexamethasone (r-hypercvad), alternating with methotrexate and cytarabine, is not recommended for the treatment of patients with newly diagnosed mcl.■ beam (carmustine-etoposide-cytarabine-melphalan), beac (carmustine-etoposide-cytarabine-cyclophosphamide), and total-body irradiation-based regimens are reasonable conditioning options for patients with mcl who have responded to first-line therapy and who are undergoing asct.■ Maintenance therapy with rituximab is recommended for patients with newly diagnosed mcl who have undergone asct.
Collapse
Affiliation(s)
- S Bhella
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON
| | - N P Varela
- Program in Evidence-Based Care, Ontario Health (Cancer Care Ontario), and Department of Oncology, McMaster University, Hamilton, ON
| | - A Aw
- Division of Hematology, The Ottawa Hospital, Ottawa, ON
| | - C Bredeson
- Malignant Hematology and Stem Cell Transplantation, The Ottawa Hospital, Ottawa, ON
| | - M Cheung
- Division of Medical Oncology and Hematology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON
| | - M Crump
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON
| | - G Fraser
- Division of Malignant Hematology, Juravinski Cancer Centre, and Department of Oncology, McMaster University, Hamilton, ON
| | | | - T Kouroukis
- Division of Malignant Hematology, Juravinski Cancer Centre, and Department of Oncology, McMaster University, Hamilton, ON
| |
Collapse
|
53
|
Outcomes in first relapsed-refractory younger patients with mantle cell lymphoma: results from the MANTLE-FIRST study. Leukemia 2020; 35:787-795. [PMID: 32782382 DOI: 10.1038/s41375-020-01013-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/23/2020] [Accepted: 07/30/2020] [Indexed: 11/08/2022]
Abstract
Patients with mantle cell lymphoma (MCL) that fail induction treatment represent a difficult-to-treat population, where no standard therapy exists. We evaluated outcomes in patients with first relapsed-refractory (r/r) MCL after upfront high dose cytarabine including standard regimens. Overall survival (OS-2) and progression-free survival (PFS-2) were estimated from the time of salvage therapy. The previously described threshold of 24 months was used to define patients as early- or late-progressors (POD). Overall, 261 r/r MCL patients were included. Second-line regimens consisted of rituximab-bendamustine (R-B, 21%), R-B and cytarabine (R-BAC, 29%), ibrutinib (19%), and others (31%). The four groups were balanced in terms of clinicopathological features. Adjusting for age and early/late-POD, patients treated with R-BAC had significantly higher complete remission (63%) than comparators. Overall, Ibrutinib and R-BAC were associated with improved median PFS-2 [24 and 25 months, respectively], compared to R-B (13) or others (7). In patients with early-POD (n = 127), ibrutinib was associated with inferior risk of death than comparators (HR 2.41 for R-B, 2.17 for others, 2.78 for R-BAC). In patients with late-POD (n = 134), no significant differences were observed between ibrutinib and bendamustine-based treatments. Ibrutinib was associated with improved outcome in early-POD patients.
Collapse
|
54
|
Rodgers TD, Friedberg JW. Key Clinical and Translational Research Questions to Address Unmet Needs in Mantle Cell Lymphoma. Hematol Oncol Clin North Am 2020; 34:983-996. [PMID: 32861291 DOI: 10.1016/j.hoc.2020.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Survival for patients with mantle cell lymphoma has improved dramatically over the last 2 decades owing to a better understanding of disease biology and the development of more effective treatment regimens for patients with untreated and relapsed disease. With these advancements, we are now poised to ask questions that challenge old treatment strategies, use new technologies, and improve our understanding of disease heterogeneity. This article focuses on questions that we believe will drive the future of mantle cell lymphoma treatment. Although not an exhaustive list, we review current literature, ongoing studies, and provide expert opinion on future trial design.
Collapse
Affiliation(s)
- Thomas D Rodgers
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, 601 Elmwood Avenue, Box 704, Rochester, NY 14642, USA.
| | - Jonathan W Friedberg
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, 601 Elmwood Avenue, Box 704, Rochester, NY 14642, USA
| |
Collapse
|
55
|
Guy D, Kahl BS. Initial and Consolidation Therapy for Younger Patients with Mantle Cell Lymphoma. Hematol Oncol Clin North Am 2020; 34:861-870. [PMID: 32861283 DOI: 10.1016/j.hoc.2020.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mantle cell lymphoma is an incurable B-cell malignancy. Treatment of young fit patients is particularly challenging, because careful consideration should be made when building a long-term treatment strategy that would provide longer remissions and increase patients' quality of life. Most young fit patients achieve long remissions with a combination of immunochemotherapy containing rituximab and high-dose cytarabine, followed by high-dose chemotherapy and autologous stem-cell transplantation. The addition of maintenance therapy with rituximab following autologous stem-cell transplantation prolongs the time to relapse and increases overall survival. Despite an intensive approach, late relapses are common and are usually treated with novel agents.
Collapse
Affiliation(s)
- Daniel Guy
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8056-29, St Louis, MO 63108, USA
| | - Brad S Kahl
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8056-29, St Louis, MO 63108, USA.
| |
Collapse
|
56
|
Abstract
PURPOSE OF REVIEW Mantle cell lymphoma is a CD5+ non-Hodgkin lymphoma associated with suboptimal outcome. Young, fit patients are generally offered intensive induction followed by autologous hematopoietic cell transplantation (AHCT) in first remission. Some patients may not benefit from this strategy. RECENT FINDINGS Recent studies have investigated the role of AHCT in the modern era. First, an analysis of the National Cancer Database demonstrated improved progression-free survival (PFS) for consolidative AHCT. Second, a multi-center study associated consolidative AHCT with improved PFS even after propensity-weighted analysis. Improved overall survival (OS) for certain subgroups was suggested. Third, patients with p53 mutations derive little benefit from AHCT. Finally, retrospective series suggest certain high-risk patients may be considered for allogenic HCT. AHCT consolidation in first remission is associated with improved PFS even after adjustment for disease severity. An overall survival benefit has not been definitively shown. Patients with p53 mutations should be treated with novel agents.
Collapse
|
57
|
Abstract
Mantle cell lymphoma (MCL) is a rare, B cell non-Hodgkin's lymphoma with highly heterogeneous clinical presentation and aggressiveness. First-line treatment consists of intensive chemotherapy with autologous stem cell transplant for the fit, transplant eligible patients, or less intensive chemotherapy for the less fit (and transplant-ineligible) patients. Patients eventually relapse with a progressive clinical course. Numerous therapeutic approaches have emerged over the last few years which have significantly changed the treatment landscape of MCL. These therapies consist of targeted approaches such as BTK and BCL2 inhibitors that provide durable therapeutic responses. However, the optimum combination and sequencing of these therapies is unclear and is currently investigated in several ongoing studies. Furthermore, cellular therapies such as chimeric antigen receptor (CAR) T cells and bispecific T cell engager (BiTe) antibodies have shown impressive results and will likely shape treatment approaches in relapsed MCL, especially after failure with BTK inhibitors. Herein, we provide a comprehensive review of past and ongoing studies that will likely significantly impact our approach to MCL treatment in both the frontline (for transplant eligible and ineligible patients) as well as in the relapsed setting. We present the most up to date results from these studies as well as perspectives on future studies in MCL.
Collapse
Affiliation(s)
- Walter Hanel
- Division of Hematology, Department of Medicine, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210 USA
| | - Narendranath Epperla
- Division of Hematology, Department of Medicine, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
58
|
Wang YH, Yu SC, Ko BS, Yang YT, Yao M, Tang JL, Huang TC. Correlative analysis of overall survival with clinical characteristics in 127 patients with mantle cell lymphoma: a multi-institutional cohort in Taiwan. Int J Hematol 2020; 112:385-394. [PMID: 32519171 DOI: 10.1007/s12185-020-02903-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/26/2020] [Accepted: 05/29/2020] [Indexed: 11/26/2022]
Abstract
Mantle cell lymphoma (MCL) is a B-cell non-Hodgkin lymphoma often with extranodal involvement at diagnosis, and yet how this feature correlates with survival awaits elucidation. To address this issue, a correlative analysis between clinical features of 127 MCL patients and their overall survival (OS) was conducted. In this cohort, the median age at MCL diagnosis was 62 years and 81% were males. Eighty-four percent of patients were Ann Arbor stage 4, and 15% were blastoid variants. In patients with gastrointestinal MCL, approximately 40% had gastric involvement. In treatment, CHOP-based induction chemotherapy was given to 61.1% of patients. One-third of patients undertook autologous stem cell transplant (SCT), and 4.7% had allogeneic SCT. The median OS was 82 months and well-stratified in MIPI risk groups. In the multivariate analysis for OS, blastoid variants and gastric involvement were both independent risk factors whereas auto-SCT had a protective effect. Overall, this study corroborated with the current understandings and international therapeutic standards for MCL. Auto-SCT associated with a better OS while allo-SCT remained an option for blastoid variants and those who failed Auto-SCT. Interestingly, patients with gastric involvement tended to have worse survival, a finding that spawns more studies to investigate the mechanism.
Collapse
Affiliation(s)
- Yu-Hung Wang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan S. Rd., Taipei City, 10002, Taiwan
| | - Shan-Chi Yu
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Bor-Sheng Ko
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan S. Rd., Taipei City, 10002, Taiwan
- Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Yi-Tsung Yang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Ming Yao
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan S. Rd., Taipei City, 10002, Taiwan
| | - Jih-Luh Tang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan S. Rd., Taipei City, 10002, Taiwan
- Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Tai-Chung Huang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan S. Rd., Taipei City, 10002, Taiwan.
| |
Collapse
|
59
|
Wu A, Graf SA, Burwick N, Grim JE, Dong ZM, Richard RE, Chauncey TR. Mantle cell lymphoma relapsed after autologous stem cell transplantation: a single-center experience. Blood Res 2020; 55:57-61. [PMID: 32269976 PMCID: PMC7106113 DOI: 10.5045/br.2020.55.1.57] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/10/2020] [Accepted: 03/07/2020] [Indexed: 01/29/2023] Open
Abstract
Background Autologous stem cell transplantation (autoSCT) can extend remission of mantle cell lymphoma (MCL), but the management of subsequent relapse is challenging. Methods We examined consecutive patients with MCL who underwent autoSCT at Veterans Affairs Puget Sound Health Care System between 2009 and 2017 (N=37). Results Ten patients experienced disease progression after autoSCT and were included in this analysis. Median progression free survival after autoSCT was 1.8 years (range, 0.3-7.1) and median overall survival after progression was only 0.7 years (range, 0.1 to not reached). The 3 patients who survived more than 1 year after progression were treated with ibrutinib. Conclusion Our findings suggest that ibrutinib can achieve relatively prolonged control of MCL progressing after autoSCT.
Collapse
Affiliation(s)
- Aaron Wu
- Department of Hospital and Specialty Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Solomon A Graf
- Department of Hospital and Specialty Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Oncology, University of Washington Medicine, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nicholas Burwick
- Department of Hospital and Specialty Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Hematology, University of Washington Medicine, Seattle, WA, USA
| | - Jonathan E Grim
- Department of Hospital and Specialty Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Oncology, University of Washington Medicine, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Zhao Ming Dong
- Department of Hospital and Specialty Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Pathology, University of Washington Medicine, Seattle, WA, USA
| | - Robert E Richard
- Department of Hospital and Specialty Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Hematology, University of Washington Medicine, Seattle, WA, USA
| | - Thomas R Chauncey
- Department of Hospital and Specialty Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Oncology, University of Washington Medicine, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
60
|
Hill BT, Rybicki LA, Urban TA, Lucena M, Jagadeesh D, Gerds AT, Dean RM, Sobecks RM, Pohlman B, Bolwell B, Kalaycio ME, Hamilton BK, Copelan EA, Majhail NS. Therapeutic Dose Monitoring of Busulfan Is Associated with Reduced Risk of Relapse in Non-Hodgkin Lymphoma Patients Undergoing Autologous Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 26:262-271. [PMID: 31610237 DOI: 10.1016/j.bbmt.2019.09.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/10/2019] [Accepted: 09/28/2019] [Indexed: 11/28/2022]
Abstract
Optimal administration of busulfan (Bu) is hampered by variable and unpredictable drug metabolism in individual patients. At our institution, Bu was previously administered with fixed weight-based dosing (WBD) in combination with cyclophosphamide (Cy) and etoposide (E) for patients with non-Hodgkin lymphoma (NHL) undergoing autologous stem cell transplantation (ASCT). In 2014, we adopted real-time pharmacokinetic (PK)-guided therapeutic drug monitoring (TDM) of Bu for all NHL patients undergoing Bu-containing ASCT. Here we compare outcomes of NHL patients who underwent ASCT with Bu/Cy/E using WBD and those who did so using TDM of Bu. We studied 336 consecutive adult NHL patients who underwent ASCT with Bu/Cy/E using WBD from January 2007 to December 2013 (n = 258) or TDM from May 2014 to December 2017 (n = 78), excluding patients with mantle cell lymphoma. Clinical outcomes, including relapse, nonrelapse mortality (NRM), progression-free survival (PFS), and overall survival (OS), hepatotoxicity and pulmonary toxicity were compared in the 2 groups. To adjust for differences in baseline characteristics between the groups, propensity-matched cohorts of WBD and TDM patients were also studied. After the first dose of Bu, the dose was increased in 36% of the patients and decreased in 41%. Changes in pulmonary and liver function from baseline to transplantation were not different between the 2 groups, although these changes showed significantly less variability with TDM than with WBD. Relapse was significantly lower and PFS was improved with TDM; 2-year estimates were 19% for TDM and 38% for WBD for relapse (P = .004) and 69% and 55%, respectively, for PFS (P = .038). No significant between-group differences in NRM or OS were seen. In multivariable analysis, TDM remained prognostic for lower risk of relapse (hazard ratio [HR], .52; 95% confidence interval [CI], .30 to .89; P = .018), but did not remain prognostic for PFS (HR, .74; 95% CI, .48 to 1.16; P = .19). Propensity-matched cohorts displayed similar patterns of outcomes. In subset analysis based on disease status at ASCT, TDM was associated with less relapse and better PFS than WBD for patients who underwent transplantation in less than complete remission (CR) compared with those who underwent transplantation in CR. Compared with WBD, PK-directed TDM of Bu reduces the incidence of relapse when used in combination with Cy and E for patients with NHL undergoing ASCT, particularly for patients in less than CR. These data support the continued use of personalized PK-guided dosing for all NHL patients undergoing ASCT with Bu-containing preparative regimens.
Collapse
Affiliation(s)
- Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina.
| | - Lisa A Rybicki
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | | | - Mariana Lucena
- Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio
| | - Deepa Jagadeesh
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | - Aaron T Gerds
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | - Robert M Dean
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | - Ronald M Sobecks
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | - Brad Pohlman
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | - Brian Bolwell
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | - Matt E Kalaycio
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | - Betty K Hamilton
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | - Edward A Copelan
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio; Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio; Levine Cancer Institute, Charlotte, North Carolina
| | - Navneet S Majhail
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
61
|
Klener P. Advances in Molecular Biology and Targeted Therapy of Mantle Cell Lymphoma. Int J Mol Sci 2019; 20:ijms20184417. [PMID: 31500350 PMCID: PMC6770169 DOI: 10.3390/ijms20184417] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a heterogeneous malignancy with a broad spectrum of clinical behavior from indolent to highly aggressive cases. Despite the fact that MCL remains in most cases incurable by currently applied immunochemotherapy, our increasing knowledge on the biology of MCL in the last two decades has led to the design, testing, and approval of several innovative agents that dramatically changed the treatment landscape for MCL patients. Most importantly, the implementation of new drugs and novel treatment algorithms into clinical practice has successfully translated into improved outcomes of MCL patients not only in the clinical trials, but also in real life. This review focuses on recent advances in our understanding of the pathogenesis of MCL, and provides a brief survey of currently used treatment options with special focus on mode of action of selected innovative anti-lymphoma molecules. Finally, it outlines future perspectives of patient management with progressive shift from generally applied immunotherapy toward risk-stratified, patient-tailored protocols that would implement innovative agents and/or procedures with the ultimate goal to eradicate the lymphoma and cure the patient.
Collapse
Affiliation(s)
- Pavel Klener
- First Dept. of Medicine-Hematology, General University Hospital in Prague, 128 08 Prague, Czech Republic.
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, 128 53 Prague, Czech Republic.
| |
Collapse
|
62
|
Tang C, Kuruvilla J. Optimal management of mantle cell lymphoma in the primary setting. Expert Rev Hematol 2019; 12:715-721. [PMID: 31268728 DOI: 10.1080/17474086.2019.1639501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: The management of mantle cell lymphoma (MCL) has significantly improved since the use of intensified induction and autologous stem cell transplant consolidation. Evolving developments in minimal residual disease detection and novel agent therapy are now challenging this frontline treatment paradigm. Areas covered: This review discusses both the established role of induction and transplant consolidation in MCL, followed by evolving concepts in the use of novel agents in the frontline setting, and the use of minimal residual disease as a driver of MCL management. Expert opinion: In an era of novel agents and improved biologic understanding of MCL, our goal for frontline management should evolve toward personalized therapy for individual patients to maximize efficacy and survival whilst minimizing treatment-related toxicities.
Collapse
Affiliation(s)
- Catherine Tang
- Division of Medical Oncology & Hematology, Princess Margaret Hospital , Toronto , Canada.,Department of Medicine, University of Toronto , Toronto , Canada
| | - John Kuruvilla
- Division of Medical Oncology & Hematology, Princess Margaret Hospital , Toronto , Canada.,Department of Medicine, University of Toronto , Toronto , Canada
| |
Collapse
|
63
|
Abstract
Mantle Cell Lymphoma is a rare and generally aggressive form of non Hodgkin lymphoma. Our understanding of the pathophysiology of this disease is improving and whilst risk factors are understood, treatments are not yet tailored towards these. The treatment algorithm in the front line is well established for older and younger patients and observation is the norm for a subset of patients although these are not well characterised as yet. In the relapse setting the role of novel agents, especially the BTK inhibitors is becoming established and combination approaches look promising. Trials are beginning to challenge the role of chemotherapy against the novel agents especially as part of front line therapy. As a consequence it is likely we will see a paradigm shift in the management of this disease in the next few years.
Collapse
Affiliation(s)
- Simon Rule
- Department of Haematology, Derriford Hospital, Plymouth University Medical School, Plymouth, PL6 8DH, UK
| |
Collapse
|
64
|
Jain P, Wang M. Mantle cell lymphoma: 2019 update on the diagnosis, pathogenesis, prognostication, and management. Am J Hematol 2019; 94:710-725. [PMID: 30963600 DOI: 10.1002/ajh.25487] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 12/16/2022]
Abstract
Unprecedented advances in our understanding of the pathobiology, prognostication, and therapeutic options in mantle cell lymphoma (MCL) have taken place in the last few years. Heterogeneity in the clinical course of MCL-indolent vs aggressive-is further delineated by a correlation with the mutational status of the variable region of immunoglobulin heavy chain, methylation status, and SOX-11 expression. Cyclin-D1 negative MCL, in situ MCL neoplasia, and impact of the karyotype on prognosis are distinguished. Apart from Ki-67% and morphology pattern (classic vs blastoid/pleomorphic), the proliferation gene signature has helped to further refine prognostication. Studies focusing on mutational dynamics and clonal evolution on Bruton's tyrosine kinase (BTK) inhibitors (ibrutinib, acalabrutinib) and/or Bcl2 antagonists (venetoclax) have further clarified the prognostic impact of somatic mutations in TP53, BIRC3, CDKN2A, MAP3K14, NOTCH2, NSD2, and SMARCA4 genes. In therapy, long-term follow-up on chemo-immunotherapy studies has demonstrated durable remissions in some patients; however, long-term toxicities, especially from second cancers, are a serious concern with chemotherapy. The therapeutic options in MCL are constantly evolving, with dramatic responses from nonchemotherapeutic agents (ibrutinib, acalabrutinib, and venetoclax). Chimeric antigen receptor therapy and combinations of nonchemotherapeutic agents are actively being studied and our focus is shifting toward making the treatment of MCL chemotherapy-free. Still, MCL remains incurable. The following aspects of MCL continue to pose a challenge: disease transformation, role of the cytokine-microenvironmental milieu, incorporation of positron emission tomography-computerized tomography imaging, minimal residual disease in the prognosis, circulating tumor DNA testing for clonal evolution, predicting resistance to BTK inhibitors, and optimal management of patients who progress on BTK/Bcl2 inhibitors. Next-generation clinical trials should incorporate nonchemotherapeutic agents and personalize the treatment based upon the genomic profile of individual patient. Recent advances in the field of MCL are reviewed.
Collapse
Affiliation(s)
- Preetesh Jain
- Division of Cancer Medicine, Department of Lymphoma/MyelomaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Michael Wang
- Division of Cancer Medicine, Department of Lymphoma/MyelomaThe University of Texas MD Anderson Cancer Center Houston Texas
| |
Collapse
|
65
|
Ng ZY, Bishton M, Ritchie D, Campbell R, Gilbertson M, Hill K, Ratnasingam S, Schwarer A, Manos K, Shorten S, Ng M, Nelson N, Xin L, De Mel Widanalage S, Sunny T, Purtill D, Poon M, Johnston A, Cochrane T, Lee HP, Hapgood G, Tam C, Opat S, Hawkes E, Seymour J, Cheah CY. A multicenter retrospective comparison of induction chemoimmunotherapy regimens on outcomes in transplant-eligible patients with previously untreated mantle cell lymphoma. Hematol Oncol 2019; 37:253-260. [PMID: 30983008 DOI: 10.1002/hon.2618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/11/2019] [Indexed: 11/11/2022]
Abstract
Mantle cell lymphoma (MCL) is an uncommon and typically aggressive form of lymphoma. Although often initially chemosensitive, relapse is common. Several induction and conditioning regimens are used in transplant-eligible patients, and the optimal approach remains unknown. We performed an international, retrospective study of transplant-eligible patients to assess impact of induction chemoimmunotherapy and conditioning regimens on clinical outcomes. We identified 228 patients meeting inclusion criteria. Baseline characteristics were similar among the induction groups except for some variation in age. The type of induction chemoimmunotherapy received did not influence overall response rates (ORRs) (0.43), progression-free survival (PFS) (P > .67), or overall survival (OS) (P > .35) on multivariate analysis (PFS and OS). Delivery of autologous stem cell transplant (ASCT) was associated with favorable PFS and OS (0.01) on univariate analysis only; this benefit was not seen on multivariate analysis-PFS (0.36) and OS (0.21). Compared with busulfan and melphalan (BuMel), the use of the carmustine, etoposide, cytarabine, melphalan (BEAM)-conditioning regimen was associated with inferior PFS (HR = 2.0 [95% CI 1.1-3.6], 0.02) but not OS (HR = 1.1 [95% CI 0.5-2.3], 0.81) on univariate analysis only. Within the limits of a retrospective study and modest power for some comparisons, type of induction therapy did not influence ORR, PFS, or OS for transplant-eligible patients with MCL. International efforts are required to perform randomized clinical trials evaluating chemoimmunotherapy induction regimens.
Collapse
Affiliation(s)
- Zi Yun Ng
- Department of Haematology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Mark Bishton
- Department of Haematology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - David Ritchie
- Department of Haematology, Royal Melbourne Hospital & Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Robert Campbell
- Department of Medical Oncology and Clinical Haematology, Olivia Newton John Cancer Research and Wellness Centre, Austin Health, Heidelberg, Victoria, Australia
| | - Michael Gilbertson
- Clinical Haematology, Monash Health and Monash University, Clayton, Victoria, Australia
| | - Kate Hill
- Cancer Care Services, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Sumita Ratnasingam
- Clinical Haematology, Monash Health and Monash University, Clayton, Victoria, Australia
| | - Anthony Schwarer
- Department of Medical Oncology, Eastern Health, Box Hill, Victoria, Australia
| | - Kate Manos
- Department of Haematology, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Sophie Shorten
- Department of Haematology, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Melissa Ng
- Department of Haematology, Gold Coast University Hospital, Southport, Queensland, Australia
| | - Niles Nelson
- Department of Haematology, Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Liu Xin
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore
| | - Sanjay De Mel Widanalage
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore
| | - Tenny Sunny
- Department of Haematology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Duncan Purtill
- Department of Haematology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Michelle Poon
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore
| | - Anna Johnston
- Department of Haematology, Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Tara Cochrane
- Department of Haematology, Gold Coast University Hospital, Southport, Queensland, Australia
| | - Hui-Peng Lee
- Department of Haematology, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Greg Hapgood
- Cancer Care Services, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Constantine Tam
- Department of Haematology, Royal Melbourne Hospital & Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia.,Department of Haematology, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Stephen Opat
- Clinical Haematology, Monash Health and Monash University, Clayton, Victoria, Australia
| | - Eliza Hawkes
- Department of Medical Oncology and Clinical Haematology, Olivia Newton John Cancer Research and Wellness Centre, Austin Health, Heidelberg, Victoria, Australia.,Department of Medical Oncology, Eastern Health, Box Hill, Victoria, Australia
| | - John Seymour
- Department of Haematology, Royal Melbourne Hospital & Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Chan Yoon Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
66
|
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
|