A Multicenter Phase II Study Of Bendamustine In Combination With Rituximab In Older Patients With Previously Untreated Diffuse Large B-Cell Lymphoma

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1791-1791 ◽  
Author(s):  
Steven I. Park ◽  
Kristy L. Richards ◽  
Adam S. Asch ◽  
Oludamilola Olajide ◽  
Allison M. Deal ◽  
...  

Abstract Background Bendamustine in combination with rituximab (BR) has shown to be similar, if not superior, in efficacy with more favorable side-effect profiles compared to R-CHOP (rituximab, cyclophosphamide, doxorubicin hydrochloride, vincristine, and prednisone) for front-line treatment of indolent B-cell lymphoma. In addition, bendamustine 120 mg/m2 in combination with rituximab 375 mg/m2 was associated with high response rates and acceptable toxicity even in older patients with relapsed or refractory aggressive B-cell lymphoma. Clinical evaluation of BR is warranted in the front-line setting for DLBCL patients not eligible for anthracyclines or very elderly, for whom the standard therapy has not been established. Methods In this single-arm phase II study, patients with ≥65 years of age who were deemed poor candidates for R-CHOP therapy at the discretion of the treating physician were enrolled through the UNC Cancer Network to determine the efficacy and safety of BR in previously untreated stage II-IV DLBCL (ClinicalTrials.gov #NCT01234467). Patients received bendamustine at a dose of 120 mg/m2 daily on days 1 and 2 of each 21-day cycle along with rituximab on day 1 for up to 8 cycles of therapy. Patients with ECOG score of 3 at baseline were allowed to receive bendamustine at a dose of 90 mg/m2 daily, and the dose would increase to 120 mg/m2 daily if their ECOG score improved to ≤ 2 after 3 cycles of BR. Pre-phase steroid therapy with prednisone 100 mg daily for five days was permitted prior to the initiation of BR in patients with poor functional status at the initial presentation. Results Twenty three patients were enrolled with the majority (83%) having stage III or IV disease at baseline. The median age was 80 years (range 65 – 89), and 78% of patients had IPI score of ≥3. More than half the patients (52%) presented with poor functional status with ECOG score of ≥2 prior to therapy, including six patients with ECOG score of 3. The overall response rate was 93% with a complete response rate of 60% for 15 evaluable patients. The median time to progression (TTP) was 7.4 months. The median survival was 9.9 months (95% CI of 3.6 – 10.9) for all patients, but for patients with ECOG score of ≥2, the median survival was 3.6 months (1.4 – 6.0). Grade 3/4 AEs observed in > 10% of patients were anemia (27%), neutropenia (18%), lymphopenia (68%), thrombocytopenia (18%), and fatigue (14%). Four grade 5 treatment-related AEs, including two cases of pneumonia and two cases of anorexia, were reported. Four deaths were directly related to disease progression during or after treatment. Six patients died of other causes, including cerebral vascular accidents, congestive heart failure, and hip fracture, which were felt to be related to the patients' underlying comorbidities. Overall survival at 12 months was 25% (8.2 – 46.9). Conclusions Combination therapy with BR demonstrates high response rates in older patients who were deemed poor candidates for the standard R-CHOP therapy for treatment of previously untreated DLBCL. However, the survival rates were not considered promising in this older frail patient population. BR, especially with a high dose of bendamustine, should be used with great caution in future clinical trials involving older DLBCL patients with poor functional status. Disclosures: Park: Seattle Genetics, Inc.: Research Funding; TEVA: Research Funding. Off Label Use: bendamustine in previously untreated DLBCL.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3477-3477
Author(s):  
Mengyang Di ◽  
Thomas A Ollila ◽  
Adam J Olszewski

Background: Ibritumomab tiuxetan (IbT) is a radioimmunoconjugate linking rituximab with 90Y isotope. IbT is approved for treatment of relapsed/refractory indolent B-cell lymphomas, or for consolidation after chemotherapy in follicular lymphoma (FL), yet its use has been limited by concern about treatment-related myeloid neoplasms (tMDS/AML: acute myeloid leukemia and myelodysplastic syndrome). The risk of tMDS/AML may differ among older patients who experience significant competing mortality. Our objective was to use population-based data to examine real-life patterns of use of IbT, and associated incidence of tMDS/AML among older patients with B-cell lymphomas. Methods: From the SEER-Medicare registry, which covers about 28% of the United States population, we selected fee-for-service Medicare beneficiaries >60 years [y] old, with B-cell lymphomas diagnosed between 2001-2015, who received systemic therapy. We identified the use of IbT, purine analogues, alkylating agents, and rituximab. We defined tMDS/AML as myeloid neoplasms diagnosed >2 months from the start of first-line therapy for lymphoma, or use of tMDS/AML-directed chemotherapy. We estimated incidence rate and incidence rate ratio (IRR) for tMDS/AML after IbT exposure using a time-split survival model. By matching IbT recipients with non-recipients according to age, sex, lymphoma histology, and observation time from the start of lymphoma therapy, we compared the cumulative incidence function of tMDS/AML using competing risk models stratified by matched groups. We fitted the univariate model, and model adjusting for prior exposure to purine analogues or alkylators. Results: Among 43,945 Medicare beneficiaries (median age of 76 y) treated for diffuse large B-cell (DLBCL, 48%), FL (22%), mantle cell (7%), marginal zone (10%), or unspecified B-cell lymphoma (14%), 428 (1%) received IbT, at median 18 months from first-line chemotherapy (interquartile range [IQR], 8-34). IbT was used most frequently in FL (50%) and DLBCL (23%), and was applied as first-line treatment in 30 patients (7%). With median follow-up of 8.5 y, 618 cases of tMDS/AML were recorded (15 among IbT recipients, and 603 among non-recipients). Median latency from the start of first-line lymphoma therapy to tMDS/AML was 38 months (IQR, 18-65). The cumulative incidence rate of tMDS/AML after IbT exposure was 2.8% at 5y (95%CI, 1.2-4.4) and 3.9% at 10y (95%CI, 1.8-6.0). No tMDS/AML was observed among patients who received IbT as first-line treatment, before chemotherapy exposure. In the time-split survival analysis, the incidence of tMDS/AML was 0.39 per 100 person-years (95%CI, 0.18-0.60) without IbT exposure, and 1.02 (95%CI, 0.56-1.71) after IbT exposure, with an IRR of 3.22 (95%CI, 1.80-5.26). There was no significant heterogeneity in the IRR by lymphoma histology (Mantel-Haenszel test of homogeneity, P=.47). In the matched analysis (Fig A), the cumulative incidence of tMDS/AML was significantly higher among patients receiving IbT (subhazard ratio [SHR], 2.16; 95%CI, 1.32-3.55). However, the association became non-significant after adjusting for prior exposure to myelotoxic chemotherapy (SHR, 1.58; 95%CI, 0.93-2.68). Prior exposures to a purine analogue (SHR 3.83; 95%CI, 2.96-4.95) or an alkylator (SHR, 1.39; 95%CI, 1.11-1.75) remained significantly associated with increased risk of tMDS/AML. Median overall survival after tMDS/AML diagnosis was 0.6y (95%CI, 0.5-0.7), and it did not differ according to IbT exposure status (P=.62; Fig B). Conclusions: The cumulative incidence of secondary tMDS/AML in this large population-based cohort is similar to reports from clinical trials (Czuczman et al. J Clin Oncol. 2007), and lower than in some prior observational series (Epperla N, et al., Br J Haematol 2017). Our results support the use of IbT for older patients with indolent lymphomas. Exposure to IbT was associated with an increased incidence of tMDS/AML, but it was no longer significant after adjustment for prior exposure to myelotoxic chemotherapy. This result suggests that the increased incidence of tMDS/AML is a cumulative effect of IbT with other myelotoxic agents. Caution should thus be exercised when using IbT after exposure to chemotherapy. The fact that no tMDS/AML were observed in patients receiving IbT as first line therapy suggests that earlier use of IbT may be preferable from the point of view of the tMDS/AML risk. Disclosures Olszewski: Genentech: Research Funding; TG Therapeutics: Research Funding; Spectrum Pharmaceuticals: Research Funding; Adaptive Biotechnologies: Research Funding. OffLabel Disclosure: Ibritumumab tiuxetan - for treatment of any B-cell lymphoma in any setting


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4214-4214
Author(s):  
Jean L. Koff ◽  
Ashish Rai ◽  
Christopher Flowers

Abstract Introduction: Severe immune dysregulation as seen in human immunodeficiency virus infection, immunosuppression after solid organ transplant, and autoimmune (AI) disease is known to act as a major risk factor for non-Hodgkin lymphoma (NHL). Recently, the InterLymph Subtypes Project pooled cases and controls to provide well-powered comparisons of risk factors for specific NHL subtypes, such as diffuse large B cell lymphoma (DLBCL), and showed that B cell-activating AI diseases in general (odds ratio 2.4; 95% confidence interval 1.8-3.1) and systemic lupus erythematosus (SLE) and Sjögren's syndrome (SS) in particular were strongly associated with increased DLBCL risk after controlling for all other risk factors (Cerhan J Natl Cancer Inst Monogr. 2014). However, little is known about the demographics or clinical outcomes of DLBCL that arises in the setting of AI disease. We examined the Surveillance, Epidemiology, and End Results (SEER)-Medicare linked database to determine the frequency of common B cell AI diseases among older patients with DLBCL and characterize the patterns of presentation, treatment, and survival for DLBCL patients with concomitant AI disease. Methods: We used the SEER database for patients diagnosed 2002-2009 linked to their Medicare claims data through 2011 to characterize presentation, treatment, and survival patterns in patients with DLBCL, including those with rheumatoid arthritis (RA), SLE, SS, and other B cell-mediated autoimmune diseases as defined by InterLymph criteria (autoimmune hemolytic anemia, Hashimoto's thyroiditis/hypothyroidism, myasthenia gravis; pernicious anemia; Wang Am J Epidemiol. 2015). Patient age, sex, race/ethnicity, region of residence, marital status, year of diagnosis, cause of death, census tract-level characteristics (education, poverty, and metropolitan/urban/rural status), stage, B symptoms, and nodal or extranodal primary site of involvement were identified from the SEER data. Assessments of poor performance status, anemia, the Charlson comorbidity index, survival, and treatment strategies were identified using Medicare inpatient, outpatient and physician claims. Treatments were categorized as: rituximab (R), cyclophosphamide and vincristine (CVP), R-CVP; cyclophosphamide, hydroxydaunorubicin, and vincristine (CHOP), and R-CHOP. Patients who did not receive any DLBCL-directed treatment within 6 months of diagnosis were categorized separately. We examined the baseline clinical characteristics for patients with DLBCL and RA, SLE, SS, or any B cell-mediated autoimmune disease, plotted overall survival and lymphoma-related survival for these groups and compared median survival times. Results: Patient characteristics are summarized in the Table. With the exception that patients with DLBCL and AI disease were more commonly female[KJL1] [CF2] , patients with DLBCL and RA, SLE, SS, or other B cell AI diseases have similar baseline presenting features as other DLBCL patients and received similar first line treatments. There was a trend towards decreased lymphoma-related survival in patients with SLE and DLBCL compared to all other groups, but this difference was not statistically significant in this cohort (Figure) [KJL3] [KJL4] . Conclusions: In this retrospective claims-based cohort of older patients with DLBCL, concomitant AI disease was uncommon and was more likely to occur in female DLBCL patients, which likely reflects the higher incidence of AI disease in women. The possibility of lower lymphoma-related survival for these patients should be explored in future studies. Disclosures Flowers: Millenium/Takeda: Research Funding; NIH: Research Funding; TG Therapeutics: Research Funding; Pharmacyclics, LLC, an AbbVie Company: Research Funding; Mayo Clinic: Research Funding; Infinity: Research Funding; ECOG: Research Funding; Acerta: Research Funding; AbbVie: Research Funding; Roche: Consultancy, Research Funding; Genentech: Consultancy, Research Funding; Gilead: Consultancy, Research Funding.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3571-3571
Author(s):  
Adrian Minson ◽  
Nada Hamad ◽  
Costas K. Yannakou ◽  
Shu Min Wong ◽  
Jason P Butler ◽  
...  

Abstract Background: R-CHOP remains a standard frontline treatment for patients with DLBCL and high-grade B-cell lymphoma (HGBL). A significant proportion of patients will have refractory disease or subsequently relapse, particularly those with high-risk features such as an elevated IPI score or rearrangements of MYC and BCL2 and/or BCL6 (double/triple hit (DH/TH)). This population remains in need of improved induction treatments that can reduce the requirement for subsequent therapies which are associated with significant toxicities and diminishing response rates. Rationale: Glofitamab is a novel full-length bispecific antibody with a unique 2:1 configuration (two CD20 binding domains and one CD3 binding domain). In combination with a single pre-dose of obinutuzumab, glofitamab has demonstrated >70% complete remission in aggressive B-cell lymphoma at the recommended target dose in a phase 1 trial (Carlo-Stella, EHA 2021). Pre-clinical studies suggest that glofitamab's activity is retained in the presence of concomitant cytotoxic and CD20 antibody therapies, making it an attractive agent for combination with R-CHOP-like induction. Polatuzumab vedotin (pola) is an antibody-drug conjugate approved for R/R DLBCL in combination with BR, and is currently in evaluation for the front-line treatment of DLBCL in combination with RCHP in a randomised trial. The safety and preliminary efficacy of glofitamab in combination with R-CHOP, or pola-RCHP as a front-line treatment for high risk DLBCL is being evaluated. Study Design and Methods: This is a parallel-arm phase Ib/II trial. Treatment consists of an initial cycle of R-CHOP, followed by 5 cycles of combination induction treatment and 2 cycles of consolidation glofitamab monotherapy. Key inclusion criteria are: age 18-65 years, a diagnosis of DLBCL or HBGL, high-risk features (IPI ≥3 or NCCN-IPI ≥4 or presence of DH/TH), treatment naïve or after 1 cycle of R-CHOP, ECOG 0-3. The primary endpoint is the safety of the combination and secondary endpoints include complete response rates at interim and end of treatment FDG-PET assessments by Lugano criteria, progression free survival and overall survival. Correlative studies assessing baseline immunologic profiles, tumour phenotype and potential resistance mechanisms are planned. Approximately 40 patients will be treated in each arm across 12-14 Australian sites. The trial commenced recruitment in July 2021 (NCT04914741). The ability to recruit prior to either cycle 1 or cycle 2 allows seamless cross-referral from non-trial sites. Figure 1 Figure 1. Disclosures Minson: Hoffman La Roche: Research Funding; Novartis: Research Funding. Hamad: Novartis: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Seymour: AbbVie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Sunesis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Morphosys: Honoraria, Membership on an entity's Board of Directors or advisory committees; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Mei Pharma: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; F. Hoffmann-La Roche Ltd: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Celgene: Consultancy, Research Funding, Speakers Bureau. Dickinson: Celgene: Research Funding; Amgen: Honoraria; Takeda: Research Funding; MSD: Consultancy, Honoraria, Research Funding, Speakers Bureau; Gilead Sciences: Consultancy, Honoraria, Speakers Bureau; Janssen: Consultancy, Honoraria; Bristol-Myers Squibb: Consultancy, Honoraria; Roche: Consultancy, Honoraria, Other: travel, accommodation, expenses, Research Funding, Speakers Bureau; Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4093-4093 ◽  
Author(s):  
Sattva S Neelapu ◽  
Mohamed A Kharfan-Dabaja ◽  
Olalekan O. Oluwole ◽  
Patel Krish ◽  
Ran Reshef ◽  
...  

Background: Axicabtagene ciloleucel (axi-cel) is an autologous anti-CD19 chimeric antigen receptor (CAR) T cell therapy approved for the treatment of adult patients who have relapsed/refractory large B cell lymphoma (LBCL) and have had ≥ 2 prior systemic therapies. In ZUMA-1, the registrational study of axi-cel in patients with refractory LBCL, the objective response rate was 83% (complete response rate, 58%), with ongoing responses in 39% after a median follow-up of 27.1 months (Locke FL, et al. Lancet Oncol. 2019). Despite the success of axi-cel, approximately 60% of patients have no response or relapse after treatment, indicating that additional strategies are needed for patients with relapsed/refractory LBCL. Preclinical murine studies have shown that rituximab augmented the tumor-suppressing effects of anti-CD19 CAR T cells, and the combination led to higher rates of tumor reduction (Mihara K, et al. Br J Haematol. 2010; Rufener GA, et al. Cancer Immunol Res. 2016). The IMiD® immunomodulatory agent lenalidomide has shown activity in patients with relapsed/refractory diffuse large B cell lymphoma and has also been shown to enhance the antitumor functions of anti-CD19 and anti-CD20 CAR T cells in mice (Otahal P, et al. Oncoimmunology. 2016). In ZUMA-14, the aim is to investigate the efficacy and safety of axi-cel in combination with rituximab or lenalidomide in adult patients with refractory LBCL. Methods: This Phase 2 study (NCT04002401) has a planned enrollment of approximately 60 patients aged ≥ 18 years with refractory LBCL, defined as a response of either progressive disease or stable disease to previous chemotherapy or progressive disease or relapse ≤ 12 months after an autologous stem cell transplant. Patients with prior IMiD® treatment, including lenalidomide, prior CAR T cell therapy, and/or prior CD19-targeted therapy are excluded. After leukapheresis, patients will be assigned 1:1 to receive axi-cel with either rituximab (Cohort 1) or lenalidomide (Cohort 2). Patients will receive lymphodepleting chemotherapy of fludarabine (30 mg/m2) and cyclophosphamide (500 mg/m2) on days -5 to -3 before axi-cel infusion (2 × 106 cells/kg) on Day 0. Cohort 1 will receive rituximab (375 mg/m2) every 28 days starting on Day -5 for a total of 6 doses. Cohort 2 will receive lenalidomide (10 mg) daily starting 7 days after leukapheresis through Day 3 and for 5 additional cycles (20 mg, first 21 days of each 28-day cycle) beginning after axi-cel infusion starting on Day 21. Patients may not receive any therapy other than conditioning therapy and rituximab or lenalidomide, as specified by cohort, between leukapheresis and axi-cel infusion. The primary endpoint is investigator-assessed complete response rate per the Lugano classification (Cheson BD, et al. J Clin Oncol. 2014). Key secondary endpoints include safety, objective response rate, duration of response, progression-free survival, overall survival, and pharmacokinetics (levels of blood CAR T cells over time). Exploratory endpoints for both cohorts include pharmacodynamic assessment of cytokine profiles and the rate of CD19-negative relapses. An additional exploratory endpoint for Cohort 2 is to investigate immunomodulation of the tumor microenvironment, including the number and activation of T cells and natural killer cells. Enrollment is expected to start in September 2019. Disclosures Neelapu: Cell Medica: Consultancy; Pfizer: Consultancy; BMS: Research Funding; Kite, a Gilead Company: Consultancy, Research Funding; Novartis: Consultancy; Precision Biosciences: Consultancy; Acerta: Research Funding; Karus: Research Funding; Incyte: Consultancy; Merck: Consultancy, Research Funding; Unum Therapeutics: Consultancy, Research Funding; Celgene: Consultancy, Research Funding; Cellectis: Research Funding; Allogene: Consultancy; Poseida: Research Funding. Kharfan-Dabaja:Daiichi Sankyo: Consultancy; Pharmacyclics: Consultancy. Oluwole:Pfizer: Consultancy; Spectrum: Consultancy; Gilead Sciences: Consultancy; Bayer: Consultancy. Krish:Celgene: Consultancy, Research Funding, Speakers Bureau; Genetech: Consultancy, Speakers Bureau; Pharmacyclics: Consultancy, Research Funding, Speakers Bureau; Takeda: Research Funding; Curis: Research Funding; MEI Pharma: Research Funding; Bristol Meyers Squibb: Research Funding; Xencor: Research Funding; Roche: Research Funding; AstraZeneca: Consultancy, Research Funding, Speakers Bureau; Sunesis: Consultancy, Research Funding. Reshef:BMS: Consultancy; Shire: Research Funding; Incyte: Consultancy, Research Funding; Kite, a Gilead Company: Consultancy, Honoraria, Research Funding; Atara: Consultancy, Research Funding; Magenta: Consultancy; Pfizer: Consultancy; Pharmacyclics: Consultancy, Research Funding; Celgene: Research Funding. Riedell:Bayer: Honoraria, Speakers Bureau; Kite/Gilead: Honoraria, Research Funding, Speakers Bureau; Verastem: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Research Funding. Stiff:Amgen: Research Funding; Gamida-Cell: Research Funding; Incyte: Research Funding; Cellectar: Research Funding; Unum: Research Funding; Gilead/Kite Pharma: Consultancy, Honoraria, Research Funding. Goyal:Kite, a Gilead Company: Employment. Kawashima:Kite, a Gilead Company: Employment, Equity Ownership. Milletti:Roche: Employment, Equity Ownership, Other: Travel Expenses, Patents & Royalties; Gilead: Employment, Equity Ownership, Other: Travel Expenses, Patents & Royalties; Kite, a Gilead Company: Employment. Oliva:Kite, a Gilead Company: Employment. Sun:Kite, A Gilead Company: Employment. Munoz:Pharmacyclics /Janssen: Consultancy, Research Funding, Speakers Bureau; Bayer: Consultancy, Speakers Bureau; Merck: Consultancy; Kyowa: Consultancy, Honoraria, Speakers Bureau; Seattle Genetics: Consultancy, Honoraria, Research Funding, Speakers Bureau; Celgene/Juno: Consultancy, Research Funding; Genentech: Consultancy, Research Funding, Speakers Bureau; Kite/Gilead: Consultancy, Research Funding, Speakers Bureau; Bristol-Myers Squibb: Consultancy; Alexion: Consultancy; Pfizer: Consultancy; Fosunkite: Speakers Bureau; AstraZeneca: Speakers Bureau; Portola: Research Funding; Incyte: Research Funding. OffLabel Disclosure: ZUMA-14 is a clinical trial evaluating the investigational combination of axicabtagene ciloleucel with either rituximab or lenalidomide in refractory large B cell lymphoma


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1718-1718 ◽  
Author(s):  
Anas Younes ◽  
Gilles Salles ◽  
R. Gregory Bociek ◽  
Giovanni Martinelli ◽  
Dolores Caballero ◽  
...  

Abstract Background: Non-Hodgkin's lymphoma (NHL), which includes diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and mantle cell lymphoma (MCL), is associated with a high unmet need in the relapsed and refractory setting. The PI3K/AKT/mTOR pathway has been shown to play a key role in the pathogenesis of NHL. Overexpression of PIK3CD (encoding the PI3Kd isoform) is common in B-cell malignancies and is therefore widely viewed as a therapeutic target in NHL. PIK3CA and PIK3CB are also expressed in B-cell malignancies, with PIK3CA overexpression seen particularly at relapse, thereby justifying exploration of pan-PI3K inhibitors in the relapsed or refractory setting. Buparlisib (BKM120) is an oral pan-class I PI3K inhibitor which has demonstrated activity in patients with solid tumors as well as in in vitro and in vivo models of hematologic malignancies. Methods: The primary objective of this Phase II study (NCT01693614) was to evaluate the efficacy of buparlisib in three parallel cohorts of adult patients with relapsed or refractory DLBCL, FL, or MCL. Secondary objectives were to evaluate safety and tolerability, progression-free survival, duration of response, and overall survival. Inclusion criteria were: patients with relapsed or refractory disease who have received at least one prior line of therapy; at least one measurable nodal lesion (≥2 cm); ECOG performance status ≤2; and adequate bone marrow and organ function. Patients with DLBCL must have received, or be ineligible for, autologous or allogeneic stem cell transplant. All patients received single-agent buparlisib 100 mg orally QD continuously until progression, intolerance, or patient withdrawal. Tumor response was evaluated by investigators per standard criteria (Cheson, 2007). Results: At data cut-off (June 19, 2014), 64 patients had been enrolled; 26 DLBCL, 24 FL, and 14 MCL. Here, results are presented for the DLBCL cohort only. Updated results from the DLBCL cohort including biomarker analyses and results for the FL cohort will be presented at the meeting. The MCL cohort is currently enrolling. For the DLBCL cohort, median age (range) was 63.5 (28–81) years and 69% were male. The median (range) number of prior therapy regimens was 3 (1–12). Specific prior therapies included: rituximab (n=25, 96%) and bendamustine/carmustine (n=8, 31%); all patients had received prior anthracycline and an alkylating agent (e.g. cyclophosphamide), and 6 (23%) patients had undergone prior stem cell transplantation. The most common (≥15%) AEs (all grades) regardless of causality were hyperglycemia and nausea (39% each), depression (31%), anxiety and fatigue (23% each), vomiting and diarrhea (19% each), and abdominal pain (15%). Alanine transaminase or aspartate transaminase elevations were rare (4% each, with no Grade 3/4 AEs). The most common (>5%) Grade 3/4 AEs regardless of causality were hyperglycemia (23%), and nausea, depression, anxiety, urinary tract infection, and neutropenia (8% each). Six (23%) patients discontinued therapy due to AEs (potentially treatment-related). Seven on-treatment deaths were reported: 6 were disease-related and 1 was suspected to be treatment-related (unexplained death following a gastrointestinal [GI] bleed in a patient with massive GI lymphoma involvement). Overall response rate was 12% (95% CI = 2.4, 30.2) with 3 responses: 1 complete response (4%) and 2 partial responses (8%). Five patients had stable disease (19%). Eight patients (31%) experienced some decrease in tumor burden (Figure 1). At data cut-off, 2 patients demonstrated durable responses, as they were still on study treatment after 9.2 and 7.4 months, respectively. Figure 1 Figure 1. Conclusions: Early results from this Phase II study of the pan-PI3K inhibitor, buparlisib, demonstrate encouraging clinical activity, and a favorable safety profile in heavily pretreated patients with relapsed or refractory DLBCL. These data suggest that targeting all 4 PI3K isoforms in DLBCL is a viable strategy and worthy of further exploration in patients with NHL. Future combination studies with buparlisib will take a mechanism-based approach. Disclosures Younes: Novartis, Curis, J&J: Research Funding; Bayer, BMS, Celgene, Incyte, Janssen R&D: Honoraria; Sanofi, Seattle Genetics, Takeda Millenium: Honoraria. Mukherjee:Novartis Healthcare Pvt. Ltd. India: Employment. Williams:Novartis: Employment. Herbst:Novartis: Employment. Tavorath:Novartis: Employment. Kim:Novartis, Celgene, Takeda: Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 848-848 ◽  
Author(s):  
Nancy L. Bartlett ◽  
Jeff P. Sharman ◽  
Yasuhiro Oki ◽  
Ranjana H. Advani ◽  
Celeste M. Bello ◽  
...  

Abstract Background Brentuximab vedotin (ADCETRIS®) is an anti-CD30 monoclonal antibody conjugated by a protease-cleavable linker to a microtubule-disrupting agent, monomethyl auristatin E. Variable CD30 expression has been demonstrated in several B-cell non-Hodgkin lymphoma (NHL) subtypes such as diffuse large B-cell lymphoma (DLBCL) and primary mediastinal B-cell lymphoma (PMBL). Methods A phase 2, open-label, single-arm study is ongoing to evaluate the antitumor activity of brentuximab vedotin in relapsed or refractory CD30-positive NHL, including B-cell neoplasms (Clinical Trials.gov NCT01421667). CD30 expression is determined by immunohistochemistry per local laboratory; any level of CD30-positive expression is permitted for enrollment. Brentuximab vedotin, 1.8 mg/kg, is administered every 3 weeks by IV infusion. Patients who achieve at least stable disease are eligible to receive continued treatment until disease progression or unacceptable toxicity. The primary efficacy endpoint is objective response rate (ORR) as assessed by the Revised Response Criteria for Malignant Lymphoma (Cheson 2007). Correlation between antitumor activity and quantitative CD30 expression is also being explored. This subset analysis presents interim data for patients with relapsed/refractory Bcell neoplasms. Results Sixty-two B-cell lymphoma patients with variable CD30 expression by central review (range 0–100%) have been enrolled to date. Diagnoses include DLBCL (n=44) and other B-cell neoplasms (n=18) [grey zone lymphoma (n=6), PMBL (n=6), follicular lymphoma (n=3), and post-transplant lymphoproliferative disorder (n=3)]. The median age of all patients was 55 years (range, 16–85 years), and the majority had an ECOG performance status of 0/1 (92%). Patients had received a median of 2 prior therapies (range, 1 to 19); 11 (18%) had received prior stem cell transplant. Forty patients (65%) had primary refractory disease, and 47 patients (76%) were refractory to their most recent prior therapy. Fourteen patients (23%) had never responded to any prior therapy. At the time of this analysis, patients had received a median of 3 cycles of treatment (range, 1–17 cycles), with a median duration of treatment of 10.5 weeks (range, 2.4 to 57.1 weeks). Twelve (19%) patients remain on treatment. Of the 43 efficacy evaluable DLBCL patients, 40% achieved an objective response [7 complete remission (CR), 10 partial remission (PR)]; the median duration of objective response was 36 weeks (range, 0.1+ to 62.3+ weeks). Of the 18 efficacy evaluable patients with other Bcell neoplasms, 22% achieved an objective response: PMBL (1 CR), PTLD (1 CR), and grey zone lymphoma (2 PRs). The median duration of objective response was 21.7 weeks (range, 6.1 to 37.1 weeks). CD30 expression levels for patients with a CR or PR were variable and ranged from <1% to 90%. There was no statistical correlation between CD30 expression and response rate. Treatment-emergent adverse events (AEs) occurring in >20% of patients included fatigue (40%), nausea and neutropenia (37% each), pyrexia (32%), diarrhea (31%), peripheral sensory neuropathy (26%), vomiting (23%), and anemia and constipation (21% each). Peripheral neuropathy events have been primarily Grade 1 or 2. Neutropenia (29%) was the only Grade 3/4 related AE observed in >10% of patients. AEs led to treatment discontinuation in 6 patients; the most common reason was peripheral sensory neuropathy (2 patients). Conclusions In this interim analysis of 62 patients with highly refractory B-cell lymphomas, compelling antitumor activity has been observed with brentuximab vedotin. Forty percent of DLBCL patients achieved an objective response, with median remission duration of >8 months and a notable proportion of complete remissions. No correlation between CD30 expression and response rate has been observed to date. Safety data are consistent with the profile of brentuximab vedotin. This study continues to enroll patients and updated results will be presented at the meeting. Disclosures: Bartlett: Seattle Genetics, Inc.: Advisory/scientific board membership and travel expenses Other, Research Funding. Off Label Use: Brentuximab vedotin is indicated for treatment of patients with Hodgkin lymphoma after failure of autologous stem cell transplant or after failure of at least two prior multi-agent chemotherapy regimens in patients who are not ASCT candidates and for the treatment of patients with systemic anaplastic large cell lymphoma after failure of at least one prior multi-agent chemotherapy regimen. Sharman:Seattle Genetics, Inc.: Research Funding, Travel expenses Other; Genentech: Research Funding; Gilead: Research Funding. Oki:Seattle Genetics, Inc.: Research Funding. Advani:Millennium: Advisory/scientific board membership, Advisory/scientific board membership Other, Research Funding; Seattle Genetics, Inc.: Advisory/scientific board membership Other, Research Funding; Genentech: Membership on an entity’s Board of Directors or advisory committees, Research Funding; Pharmacyclics, Inc.: Research Funding; Janssen R&D: Research Funding; Allos Therapeutics: Research Funding; Celgene: Membership on an entity’s Board of Directors or advisory committees; Abbott: Research Funding. Bello:Seattle Genetics, Inc.: Research Funding, Speakers Bureau; Celgene: Membership on an entity’s Board of Directors or advisory committees; Spectrum pharmaceuticals: Speakers Bureau. Winter:Seattle Genetics, Inc.: Research Funding. Yang:Seattle Genetics, Inc.: Employment, Equity Ownership. Kennedy:Seattle Genetics, Inc.: Employment, Equity Ownership. Jacobsen:Seattle Genetics, Inc.: Advisory/scientific board membership Other, Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3943-3943 ◽  
Author(s):  
Brad Kahl ◽  
Andrew W Roberts ◽  
John F Seymour ◽  
Ranjana H. Advani ◽  
Daniel Oscar Persky ◽  
...  

Abstract Abstract 3943 Background: Navitoclax (ABT-263) is a novel, orally bioavailable, small molecule that binds with high affinity (Ki ≤1nM) to Bcl-2, Bcl-xL, and Bcl-w, promoting apoptosis. Navitoclax shows potent targeted cytotoxicity (EC50 ≤1μM) in vitro against T and B lymphoid malignancies that over-express Bcl-2. As monotherapy in phase 1 trials, oral navitoclax is well-tolerated and with daily dosing has shown activity in patients (pts) with lymphoid malignancies and many pts (response rate approximately 35%) with chronic relapsed or refractory lymphocytic leukemia (CLL). Thrombocytopenia is the dose-limiting toxicity (DLT). Rituximab mono- or combination therapy is an established treatment for pts with indolent, CD20-positive B-cell malignancies. In multiple preclinical models of B-cell lymphoma, the efficacy of rituximab (monotherapy and in combination with chemotherapy) was enhanced by the addition of navitoclax. Methods: This international, phase 1, dose-escalation study employing a modified Fibonacci 3+3 design, assessed the safety and pharmacokinetics (PK), and determined the maximum tolerated dose (MTD) and the recommended phase 2 dose (RPTD) of oral navitoclax added to standard rituximab monotherapy in pts with CD20-positive malignancies. Secondary objectives were evaluation of progression-free survival, response rate, duration of response, and overall survival. Patient eligibility included ≥1 lesion ≥1.5 cm, ECOG score ≤1, and platelet count of ≥100,000/mm3. For all dose cohorts, after a 7–14 day 150 mg/day dose lead-in, navitoclax was given once daily at 200 mg (Cohort 1), 250 mg (Cohort 2), or 325 mg (Cohort 3). At least 3 pts were enrolled in each cohort. At MTD determination, a safety expansion cohort of up to 12 pts was added. Pts proceeded from lead-in to the combination regimen, if the pre-dose platelet count on Lead-in Day 7 was ≥50,000/mm3. Rituximab was given 375 mg/m2 IV once weekly for 4 doses, starting Day 1 of Week 1. A cycle was 28 days of therapy. Patients were allowed to continue on navitoclax therapy for 2 years in the absence of progressive disease or significant toxicity. Safety was assessed by NCI-CTCAE v3.0, and tumor responses by IWG or NCI-WG criteria (for CLL pts) every 2 months by CT or MRI. Results: As of July 2010, 19 pts, median age 58 years (range 45–92), have been enrolled (11 with follicular lymphoma [FL], 3 with CLL/SLL, and 1 each with diffuse large B-cell lymphoma, transformed disease, lymphoplasmacytic lymphoma [LPL], lymphoblastic lymphoma, and Hodgkin lymphoma, respectively); 4 in the 200 mg, 7 in the 250 mg, 3 in the 325 mg, and 5 in the 250 mg expanded safety cohort. The median number of prior therapies was 3. Seventeen pts had navitoclax-related AEs, the most common being diarrhea (11 pts), nausea (11), and fatigue (8). DLTs were Grade 3 diarrhea (1 pt in the 250 mg cohort), Grade 3 fatigue (1 pt in the 325 mg cohort), and Grade 4 thrombocytopenia (1 pt in the 325 mg cohort). MTD was defined as 250 mg. Preliminary antitumor activity and best response data are available for 12 pts. Eight pts responded (ORR 67%) with 4 CRs (all FL), 4 PRs (SLL, FL, CLL, LPL), 1 SD, and 3 PDs. Five other pts are continuing treatment but have not yet reached the first tumor assessment at 12 weeks. One pt discontinued due to disease progression prior to the first tumor assessment, and 1 pt discontinued after 3 days of dosing due to the taste of the oral drug solution. Twelve pts remain on study at a median of 19.4 weeks (4.4–49.1 weeks). Preliminary PK results indicated that navitoclax PK at doses of 200–325 mg in this combination study was comparable to that in the navitoclax monotherapy study. Conclusions: The combination of navitoclax and rituximab is well tolerated and shows encouraging preliminary evidence of activity. The MTD of navitoclax in combination with rituximab is 250 mg. An expanded cohort of pts is being enrolled at 250 mg of navitoclax to further assess tolerability, confirm this dose as the RPTD, and to better define efficacy. Disclosures: Kahl: Abbott: Consultancy, Research Funding. Roberts:Abbott: Research Funding. Seymour:Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Advani:Abbott: Research Funding. Persky:Millennium, Takeda: Consultancy, Research Funding. Yang:Abbott: Employment. Cui:Abbott: Employment. Busman:Abbott: Employment. Krivoshik:Abbott: Employment. Enschede:Abbott: Employment. Humerickhouse:Abbott: Employment.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2719-2719
Author(s):  
Sarit Assouline ◽  
Michael Crump ◽  
Torsten Holm Nielsen ◽  
Miguel Alcaide ◽  
David Macdonald ◽  
...  

Abstract INTRODUCTION: Novel therapies which consider the molecular diversity of diffuse large B cell lymphoma (DLBCL) are needed to salvage patients who fail to respond to standard chemoimmunotherapy. The majority of DLBCL contain mutations in histone modifying enzymes (HME) which suggests that histone deacetylase inhibitors (HDI) should be active. Preclinical data suggest that they also augment the effect of rituximab. METHODS: In this randomized phase II study, we evaluated the response rate and toxicity of panobinostat, a pan-HDI, 30 mg orally 3 times a week, with or without rituximab, in 40 patients with relapsed and refractory DLBCL (median 3 prior regimens, range1-9). Tumors were sequenced for mutations known to be associated with DLBCL, and serial biopsies were analyzed for changes in biomarkers. RESULTS: 21 patients were treated with panobinostat alone and 19 with panobinostat and rituximab. Overall, 12/40 patients (30%) responded to panobinostat (95% confidence interval 16.6%-46.5%) the addition of rituximab did not increase responses. The median duration of response was 14.5 months (95% confidence interval 9.4 to not reached) (Figure). At a median follow up of 17.5 months for responders, 6 of 12 patients have not progressed. Patients with dual expression of MYC and BCL2 (6/27) and those with FAS mutations (5/33) did not respond although responses were seen in 5/10 patients with TP53 mutations. Responses were seen in 8/24 patients with mutations in HME genes. Patients with MEF2B mutations had the greatest odds of response (P<0.05) and a higher number of HME mutations tended to increase the odds of response. Changes in histone H3 acetylation and MYC expression were observed but did not correlate with response. CONCLUSION: Panobinostat induces durable responses in some patients with relapsed DLBCL, and accompanying biomarker analyses identified DLBCL subgroups likely to respond. Figure 1. Progression free survival overall and for responders, N=40 patients. Figure 1. Progression free survival overall and for responders, N=40 patients. Disclosures Assouline: Novartis: Consultancy, Honoraria, Research Funding; Roche: Consultancy, Honoraria, Research Funding; Janssen: Honoraria; Celgene: Consultancy. Off Label Use: panobinostat for the treatment of diffuse large B cell lymhpoma. Crump:Seattle Genetics: Consultancy; Celgene: Consultancy; Roche Canada: Consultancy. Tosikyan:Novartis: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document