Safety, Tolerability and Activity of Ofatumumab, Bendamustine and Dexamethasone Combination As First-Line Treatment of Mantle-Cell Lymphoma in the Elderly: A Multicenter Study

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1647-1647 ◽  
Author(s):  
Michele Magni ◽  
Massimo Di Nicola ◽  
Carmelo Carlo-Stella ◽  
Liliana Devizzi ◽  
Anna Guidetti ◽  
...  

Abstract Abstract 1647 Background and Aims: Mantle cell lymphoma (MCL) is the aggressive subtype of non Hodgkin's lymphoma with the poorest long-term survival probability. There is no consensus for its treatment: in most cases, R-CHOP immuno-chemotherapy, with its 34% complete remission (CR) rate, is still considered the standard of care. Very encouraging results have been reported using more aggressive induction treatments as first-line therapy but these have a role only in the initial management of clinically fit patients less than 60–65 years of age. Since the median age at diagnosis is 63 years and intensive treatment tend to be poorly tolerated by elderly patients, a frontline therapy that can be administered to this category of patients is needed. The main objective of the present study was to design a well tolerated regimen, applicable to the vast majority of patients over 60 years with MCL, while improving the limited efficacy of R-CHOP immunochemotherapy. To achieve this goal, we have used two novel drugs, i.e. ofatumumab and bendamustine. Ofatumumab is a fully human monoclonal antibody targeting a unique epitope on the CD20 molecule: its superior complement-dependent cytotoxicity compared to rituximab may translate into longer duration of treatment response. Bendamustine is a unique agent with both nitrogen mustard group and a benzimidazole ring: when given to relapsed or refractory MCL patients in combination with rituximab, it has shown a very favourable toxicity profile, as well as considerable activity. Methods: Previously untreated patients with MCL, any stage (patients with disease limited only to the bone marrow were excluded), aged ≥ 60 years, were enrolled into a therapeutic protocol consisting of six cycles of immunochemotherapy as follows: ofatumumab 1000 mg i.v. on day 1 (300 mg only for the first cycle); bendamustine 120 mg/sm i.v. on day 2 and 3; dexamethasone 40 mg i.v. on day 1 to 4. Each cycle of therapy was administered every 21 days. Hematopoietic growth factors were used in case of neutropenia. Results: As of July 31, 2011, 36 of the 50 planned patients have been enrolled and 19 have received the whole treatment. Cyclin D1 immunohistochemistry was positive in 18 patients (95%), thus confirming the diagnosis, while in the patient found negative at cyclin D1 analysis, Sox11 was used to confirm MCL; 1 patient (5%) had blastoid histology. Median age was 69 years (range 60–81), stage at diagnosis was IV in 95% of patients; MIPI score was low in 8 (42%), intermediate in 8 (42%) and high in 3 patients (16%); there were 15 male (79%) and 4 female patients (21%). Bone marrow (BM) involvement was detected by immunohistochemistry in 15 (79%) patients and by polymerase chain reaction (PCR) in all patients. Involvement of peripheral blood was detected by flow-cytometry in 12 cases (63%), by PCR in 18 cases (94%), while no data were available in the remaining case. A molecular probe was available for all patients: bcl1/IGH rearrangement was detected by PCR in 11 patients (58%), and an IGH allele-specific oriented primer was designed for the remaining 8 patients (42%). Most adverse events (AEs) were mild or moderate in severity. The most common grade ≥ 3 treatment-related AE were neutropenia (10.5%), febrile neutropenia (10.5%), anemia (5.2%) and thrombocytopenia (5.2%). Two patients (10.5%) experienced tumor lysis syndrome: one was complicated by acute renal failure and the other by disseminated intravascular coagulation; both recovered with adequate therapy. Infectious complications consisted of one episode of pneumonia and one herpes zoster reactivation: both cases were resolved with antibiotic and antiviral therapy, respectively. Of note, 10 patients (52%) experienced cytomegalovirus reactivation, detected as pp65 antigen immunofluorescence, without evidence of infection: all these patients received anti-CMV therapy with negativization of the assay. Among the 19 patients evaluable for response, overall response rate was 94%, with CR in 17 patients, partial remission in one patient and progressive disease after the second cycle in one patient; 15 out of 15 patients evaluable for minimal residual disease in the BM showed molecular remission. Conclusions: Chemotherapy with bendamustine and ofatumumab appears generally safe and well tolerated to date in MCL patients aged ≥ 65 years requiring treatment. Preliminary data about efficacy are encouraging: accrual is ongoing for further evaluation. Disclosures: Off Label Use: ofatumumab, in label for CLL.

1997 ◽  
Vol 108 (3) ◽  
pp. 302-307 ◽  
Author(s):  
Mohammad A. Vasef ◽  
L. Jeffrey Medeiros ◽  
Chae Koo ◽  
Althea Mccourty ◽  
Russell K. Brynes

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4099-4099
Author(s):  
Yannick LE Bris ◽  
Olivier Theisen ◽  
Anne Moreau ◽  
Florence Magrangeas ◽  
Danielle Canioni ◽  
...  

Abstract Background Mantle cell lymphoma (MCL) is a lymphoproliferative disorder characterized by the t(11;14) balanced translocation, involving on chromosome 11 the gene encoding cyclin D1, CCND1. The enhancer of IGH, on chromosome 11, induces aberrant Cyclin D1 overexpression. A few reports have shown that the CCND1 oncogene locus is a recurrently amplified region in MCL. However, the prognostic value of this copy number abnormality (CNA) is not known. The incidence and clinical impact of CCND1 CNA were investigated on diagnostic samples from patients enrolled in the first-line randomized controlled trial LyMa (S. Le Gouill et al. NEJM 2017). Patients and methods A series of 100 lymph node biopsies performed at diagnosis for patients enrolled in the LyMa trial (n=299) was selected. After DNA extraction, CNAs were investigated in these samples using the innovative Oncoscan® SNP-array technique adapted to analyze highly degraded DNA extracted from formalin-fixed paraffin-embedded (FFPE) tissues. Ninety-four samples were informative for CNAs. CCND1 gains were controlled by fluorescence in situ hybridization on interphase nuclei, following standard procedures using LSI IGH/CCND1 XT Dual color, dual fusion translocation probes (Abbott Molecular, Des Plaines, IL). Progression-free and overall survivals were calculated from the date of inclusion until relapse/death or last news and death or last news respectively. Cumulative incidence of relapse (CIR) was estimated from the date of inclusion until the date of relapse or last news Results Amplifications of a large portion of 11q, beginning at the t(11;14) breakpoint, were observed with Oncoscan® in 7 patients. All these cases with a gain of 11q13-14 (CCND1) also had a gain of the 14q32 locus (IGH) suggesting amplification of the rearranged IGH-CCND1 region of t(11;14). This was confirmed by FISH analysis which disclosed three different configurations. The first was the classical CCND1-IGH rearrangement (CCND1 on normal chromosome 11, IGH on normal chromosome 14 and two fusion signals from the t(11;14)(CCND1-IGH)). The second configuration associated CCND1-IGH fusion signals and a gain of both CCND1 and IGH signals. The third configuration was a duplication of a CCND1-IGH fusion signals (3 signals). Both in SNP and in FISH, the amplification was always identified as sub-clonal, concerning only part of the cells. In some patients, different configurations coexisted. Compared to other patients of the series, those with amplification of IGH and CCND1 regions had a higher-risk bio-MIPI (11q13-14, p=0.015; 14q32, p=0.004). Patients with large gains at 11q13-14 (CCND1 N=7) had poorer median PFS (18 months vs not reached (NR); p=0.004), OS (35 months vs NR; p=0.01) and CIR (33 months vs not NR; p=0.004). The same was observed for patients with gains at 14q32 (IGH; N=8), with significantly different median PFS (23 months vs NR; p<0.001), OS (38 months vs NR; p=0.001) and CIR (28 months vs NR; p<0.001). Conclusion Gains of the IGH-CCND1 rearrangement or involved genes appear to be a potential new biomarker predictive of poor response to first line immunochemotherapy in young MCL patients. Disclosures Hermine: Novartis: Research Funding; Hybrigenics: Research Funding; AB Science: Consultancy, Equity Ownership, Honoraria, Research Funding; Erythec: Research Funding; Celgene Corporation: Research Funding.


2021 ◽  
Vol 4 (Supplement_1) ◽  
pp. 138-140
Author(s):  
K Donaldson ◽  
S Nassiri ◽  
D Chahal ◽  
M F Byrne

Abstract Background Mantle cell lymphoma (MCL) is an aggressive subtype of B-cell non-Hodgkin lymphoma (NHL), often diagnosed at later stages with secondary gastrointestinal (GI) involvement. Primary GI MCL is rare and is not often discussed in the literature. Aims To increase awareness of a rare condition that is likely to be encountered but can be challenging to diagnose. Methods Case report and review of the literature. Results Case Report A 78-year-old man with multiple untreated vascular risk factors including atrial fibrillation and type 2 diabetes presented with acute onset left hemiplegia, dysarthria, and imaging consistent with a left pontine stroke. As part of his workup he underwent a CT abdomen/pelvis identifying an 11 x 5 cm intraluminal mass in the transverse colon. Previous screening colonoscopies, for family history of colon cancer, were notable for tubular adenomas without high-grade dysplasia at 13, 12, 10, 7, and 2 years prior to admission. The patient had 16 pounds of weight loss without other constitutional symptoms, change in bowel habits or evidence of GI bleeding. Bloodwork was notable for microcytic anemia (Hemoglobin 91 g/L, MCV 75 fL), from a normal baseline one year prior, without other cytopenias. C-reactive protein (44 mg/L) and GGT (164 U/L) were elevated. Other liver enzymes, lactate dehydrogenase, and electrolytes were normal. Colonoscopy revealed numerous polypoid lesions throughout the entire colon and a large non-obstructive mass with submucosal appearance in the transverse colon. Biopsies were taken from the large mass and one of the smaller polypoid lesions. Histology showed a sheet-like infiltrate of small lymphocytes within the lamina propria. Immunohistochemical staining was positive for CD20, BCL2, Cyclin D1, equivocal for CD5, and negative for BCL6 and CD3. Ki67 index approached 30%. A diagnosis of colonic MCL was made. Literature Review Primary MCL of the GI tract is rare, accounting for only 1 to 4% of all GI malignancies. There is a male and Caucasian predominance with a median age of 68 years at diagnosis. Presenting complaints may include abdominal pain, anorexia, and GI bleeding. Typical endoscopic features are small nodular or polypoid tumors, between 2mm and 2 cm in size, along one or more segments of the GI tract referred to as multiple lymphomatous polyposis (MLP). A single colonic mass is infrequently seen, highlighting the importance of endoscopy for diagnosis, as subtle findings may be missed on radiographic evaluation. Biopsies for immunohistochemistry are essential to distinguish MCL from other NHLs, as almost all cases express cyclin D1. Despite aggressive immunochemotherapy, prognosis is often poor due to MCL’s rapid progression and early relapse. Conclusions Primary GI MCL is a rare entity. Awareness is essential as evaluation and management differ from lymphoma at other sites, and other GI malignancies. Funding Agencies None


2003 ◽  
Vol 34 (8) ◽  
pp. 789-791 ◽  
Author(s):  
Andre A Schenka ◽  
Randy D Gascoyne ◽  
Eliane Duchayne ◽  
Georges Delsol ◽  
Pierre Brousset

2001 ◽  
Vol 23 (5) ◽  
pp. 470-476 ◽  
Author(s):  
Brent R. Moody ◽  
Nancy L. Bartlett ◽  
David W. George ◽  
Caroline R. Price ◽  
Wayne A. Breer ◽  
...  

2017 ◽  
Vol 8 (8) ◽  
pp. 223-234 ◽  
Author(s):  
Bita Fakhri ◽  
Brad Kahl

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.


2017 ◽  
Vol 53 (3) ◽  
pp. 347-351
Author(s):  
Umberto Falcone ◽  
Haiyan Jiang ◽  
Shaheena Bashir ◽  
Richard Tsang ◽  
Vishal Kukreti ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5263-5263
Author(s):  
Karin Hohloch ◽  
Christine Windemuth-Kieselbach ◽  
Pier Luigi Zinzani ◽  
Roberto E. Cacchione ◽  
Wojciech Jurczak ◽  
...  

To assess the efficacy of radioimmunotherapy (RIT) with 90yttrium-ibrutinib-tiuxetan (90Y-IT) in mantle cell lymphoma, data from 90 patients registered in the RIT Network with a median follow-up (FU) of 5.5 years after RIT were evaluated. 90Y-IT was given as first-line therapy in 45 (50%) (consolidation 44 pts., primary therapy 1 pt.) and at relapse in 45 (50%) patients (consolidation 24 pts., recurrence 12 pts., therapy refractory 3 pts., conditioning 2 pts., other 4 pts.). As a first-line treatment, 30 patients (pts.) (67%) achieved CR, 10 pts. (22%) PR%., 1 pt. (2%) PD, and for 4 pts. (9%) no response data was available. At relapse, CR was achieved in 17 pts. (38%), PR in 6 pts. (13%), SD in 2 pts. (4%), and 6 pts. (13%) had PD, while the response was not documented for 14 pts. (31%). After a median FU of 5.5 years, median PFS for all patients was 2.11 (95%CI: 1.03-2.32) years, and median OS was 4.05 (95%CI 2.79-7.21) years. Eleven pts. (12.2%) developed second malignancy. In conclusion, this is the largest report of MCL pts. treated with 90Y-IT to date. 90Y-IT was most often used as consolidation after first- and second-line chemotherapy and may improve the results achieved using chemoimmunotherapy alone. However, the results are less encouraging compared to treatment with small molecules such as ibrutinib. Disclosures Zinzani: TG Therapeutics: Honoraria, Speakers Bureau; Kyowa Kirin: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Roche: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Portola: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Immune Design: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Sandoz: Membership on an entity's Board of Directors or advisory committees; Servier: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen-Cilag: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celltrion: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Verastem: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; MSD: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Eusapharma: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Sanofi: Consultancy. Jurczak:Sandoz: Membership on an entity's Board of Directors or advisory committees, Research Funding; Loxo: Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Research Funding; Roche: Research Funding; AstraZeneca: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Research Funding; Bayer: Research Funding; Gilead: Research Funding; MorphoSys: Research Funding; Incyte: Research Funding; Novo Nordisk: Research Funding; Servier: Research Funding; TG Therapeutics: Research Funding; Celtrion: Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding. Truemper:Seattle Genetics, Inc.: Research Funding; Takeda: Consultancy, Research Funding; Roche: Research Funding; Nordic Nanovector: Consultancy; Mundipharma: Research Funding; Janssen Oncology: Consultancy. Scholz:Janssen-Cilag: Consultancy; Hexal: Consultancy; Takeda: Consultancy; Novartis: Consultancy; Celgene: Consultancy; Pfizer: Speakers Bureau; Roche: Consultancy; GILEAD: Consultancy, Speakers Bureau; Daiichi Sankio: Consultancy. OffLabel Disclosure: Yttrium 90 (90Y) Ibritumomab Tiuxetan (Zevalin) is approved for treatment of patients with relapsed follicular lymphoma and as consolidation therapy after chemo(immuno)therapy of patients with follicular lymphoma.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 36-36
Author(s):  
Mats Jerkeman ◽  
Martin Hutchings ◽  
Riikka Räty ◽  
Karin Fahl Wader ◽  
Anna Laurell ◽  
...  

Introduction: In spite of improvements in treatment of mantle cell lymphoma (MCL), this is still considered an incurable lymphoma entity, and the majority of patients eventually relapse. Ibrutinib is a very active agent in MCL, but in vitro has been shown to partially antagonize the activity of rituximab, by suppression of NK cell activity and subsequent ADCC. Lenalidomide, on the other hand, improves rituximab-induced ADCC. In this multi-centre open-label phase II trial, we evaluated safety and efficacy of this triplet combination in patients with relapsed or refractory MCL. Methods: Patients with MCL, relapsing after or refractory to at least one rituximab-containing chemotherapy regimen, WHO PS 0-3, and measurable disease were eligible. The primary endpoint was maximal overall response rate (ORR) measured with CT and PET/CT. Minimal residual disease (MRD) monitoring by PCR was performed during follow-up, according to EuroMRD criteria. Ion Torrent sequencing of the most frequently mutated genes in MCL was performed on frozen tumor cells from bone marrow at time of relapse. Health-related quality of life was assessed by the EORTC-QLQ C30 questionnaire before and during treatment. Treatment schedule: Induction phase: Up to twelve 28-day cycles with: Lenalidomide 15 mg p o daily, days 1-21, Ibrutinib 560 mg p o days 1-28, Rituximab 375 mg/m2 i v day 1 in cycle 1, then 1400 mg s c (or 375 mg/m2i v) days 8, 15 and 22 in cycle 1, then day 1 in cycles 3, 5, 7, 9 and 11. Maintenance phase: For patients in CR, PR or SD, not in need of other treatment, given until progression, cycle duration 56 days. Ibrutinib: 560 mg p o days 1-56, 2. Rituximab 1400 mg s c (or 375 mg/m2i v) day 1 of each cycle. Results: Accrual of 50 pts was completed in June 2016, at 10 centres in Sweden, Norway, Denmark and Finland. The median age was 69.5 years, with a median MIPI score of 6.2. Patients had received a median of two previous regimens, four had progressed after single agent ibrutinib, and three had received prior allo-SCT. A TP53 mutation was detected in 11 of 49 evaluable cases (22%), 8 cases were of blastoid/pleomorphic histology, and 22 of 40 evaluable cases had a Ki67 &gt;30%. Treatment emergent-AEs of any grade in ≥20% of patients were rash (24%) and fatigue (20%). Five pts (10%) experienced rash grade 3, mainly during cycle 1. Hematological toxicity was generally of low grade, apart from grade 3-4 neutropenia in 5 patients. One patient died due to possible treatment-related toxicity (septic shock). In total, 27 patients achieved CR (54%) and 10 PR (20%). Among evaluable patients with a TP53 mutation, blastoid/pleomorphic histology or Ki67 &gt;30%, the CR rates were 7/11 (64%), 15/8 (62%) and 11/22 (50%), respectively. After a median follow-up of 40 months, the median PFS is 18 months (95% CI 6.5-28), and median OS 47 months (95% CI 30-64). Patients with a detectable TP53 mutation at relapse (n=11) had a median PFS of 13 months (95% CI 4.2-21), whereas pts without a TP53 mutation had a median PFS of 34 months (95% CI 8.3-60). Of the 28 patients evaluable for MRD at 6 months, 15/27 (56%) patients achieved molecular remission in blood and 12/28 (43%) in bone marrow. After 12 months, MRD-negativity in BM was 68% (13/19). Out of 4 patients with TP53-mutated MCL, 2 were MRD-negative in BM after 12 months, as well as 2 out of 4 patients with blastoid/pleomorphic histology. By self-reported HRQOL, a lower level of emotional functioning (EF), as well as a higher level of pain (PA) at baseline, was associated with inferior PFS. In addition, low EF was associated with inferior OS. By a Cox regression multivariable analysis, including MIPI, TP53, histology, Ki67, EF and PA, only MIPI was prognostic for PFS or OS with this regimen. Conclusions: The combination of ibrutinib, lenalidomide and rituximab has been shown to be an active and well tolerated regimen in this cohort of high risk R/R MCL, associated with a high rate of molecular remission. The activity in TP53 mutated MCL is lower than in unmutated disease, but this regimen may still serve as an option for a bridge to an allogeneic transplantation or CAR-T therapy in this category of patients. Disclosures Jerkeman: Roche: Research Funding; Abbvie: Research Funding; Celgene: Research Funding; Janssen: Research Funding; Gilead: Research Funding. Hutchings:Genmab: Honoraria; Genmab: Consultancy; Takeda: Consultancy; Roche: Research Funding; Celgene: Research Funding; Daiichi: Research Funding; Sankyo: Research Funding; Genmab: Research Funding; Janssen: Research Funding; Novartis: Research Funding; Sanofi: Research Funding; Takeda: Research Funding; Roche: Honoraria; Roche: Consultancy; Takeda: Honoraria.


Sign in / Sign up

Export Citation Format

Share Document