Bortezomib in relapsed or refractory mantle cell lymphoma (MCL): Results of the PINNACLE study

2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 7512-7512 ◽  
Author(s):  
A. Goy ◽  
S. H. Bernstein ◽  
B. S. Kahl ◽  
B. Djulbegovic ◽  
M. J. Robertson ◽  
...  

7512 Background: Bortezomib (VELCADE, Vc), a novel proteasome inhibitor, is approved in relapsed multiple myeloma and has shown activity in MCL in phase 2 NHL studies. Methods: Patients (pts) with relapsed or refractory MCL with a maximum of 2 prior therapies received Vc 1.3mg/m2 i.v. on d 1, 4, 8, and 11 of a 21-d cycle for up to 1y. Full accrual (155 pts) was completed in this multicenter, phase 2 study. Primary endpoint was TTP, secondary endpoints included RR and duration of response (DOR). Response (International Workshop Criteria) was assessed by the investigators and separately by the sponsor using central radiology review. Results: Data are available for 154 pts. Baseline characteristics included median age 65y, 81% male, 28% KPS <90%, 35% LDH > normal, 43% IPI ≥3, 74% stage IV MCL. 90% of pts had prior intensive therapy (e.g. Hyper CVAD, CHOP, EPOCH), 96% had prior rituximab, and 14% had prior stem cell transplant. Median duration of Vc treatment was 4 cycles. 138 pts were evaluable for response. By investigator assessment, RR was 35% (CR + CRu = 8%) and median DOR was 9.2 mo. Median TTP was 5.5 mo (all patients, n = 154). Using central radiology review RR was 31% (CR + CRu = 7%), median DOR was 4.6 mo and median TTP was 4.1 mo. With median follow-up of 10 mo, median survival has not been reached. The most common non-hematologic AEs were fatigue (14% ≥grade 3), GI events (≥grade 3 diarrhea, abdominal pain, and nausea/vomiting in 5%, 4%, and 3%, respectively), and peripheral neuropathies (7% ≥grade 3). Hematologic toxicities were minimal except for transient thrombocytopenia (10% ≥grade 3), as previously seen with Vc. Conclusions: This study confirms the activity of Vc in relapsed/refractory MCL in a multicenter international setting and supports its rapid development as a new treatment for relapsed MCL. Vc is also being studied in the first-line setting in combination with standard chemotherapy. [Table: see text]

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 125-125 ◽  
Author(s):  
Andre Goy ◽  
Steven Bernstein ◽  
Brad Kahl ◽  
Benjamin Djulbegovic ◽  
Michael Robertson ◽  
...  

Abstract Background: We previously reported substantial activity with single-agent bortezomib (VELCADE®; Vc) in patients (pts) with relapsed or refractory MCL in the PINNACLE study (JCO2006;24:4867–74), which resulted in approval of Vc for MCL pts following ≥1 prior therapy. All pts have now completed treatment. Here we report updated time-to-event data in all pts, and by response category, with extended follow-up. Methods: 155 pts (median age 65 yrs; 55%/41%/4% with 1/2/≥3 prior therapies; 77% Stage IV MCL; 55% positive bone marrow) received Vc 1.3 mg/m2 on days 1, 4, 8, and 11 of 21-day cycles; of these, 141 were response-evaluable. Response and progression were determined by modified International Workshop Response Criteria using independent radiology review. Results: After a median follow-up of 26.4 mo, 55 pts (35%) remained in follow-up; 93 (60%) had died, 2 (1%) had withdrawn consent, and 5 (3%) were lost to follow-up. Pts received a median of 4 treatment cycles (range 1–21; 8 in responding pts). Median time to first response was 1.3 months. Median duration of response (DOR) was 9.2 mo in all responders and has not been reached in pts achieving CR/CRu. Median time to progression (TTP), time to next therapy (TTNT; first Vc dose to start of next therapy), and overall survival (OS) are shown in the table for all pts and by response. Survival rate at 12-mo was 69% overall and 91% in responding pts. In pts refractory to their last therapy (no response or response with TTP <6 mo; n=58), median DOR was 5.9 mo, median TTP was 3.9 mo, median TTNT was 4.6 mo, and median survival was 17.3 mo. Safety profile was similar to previously reported; most common grade ≥3 AEs were peripheral neuropathy (13%), fatigue (12%), and thrombocytopenia (11%). The most common AE resulting in Vc discontinuation was peripheral neuropathy (10%). Twelve (8%) pts died on-study, including 5 (3%) considered related to Vc. Conclusions: Vc provides durable responses plus prolonged time off-therapy and survival in responding pts, suggesting substantial clinical benefit in relapsed/refractory MCL. Median TTP, TTNT, and OS (months) in all pts and by response All pts (N=155) Responders (N=45) CR/CRu (N=11) PR (N=34) SD (N=52) PD (N=34) NE, not estimable TTP 6.7 12.4 NE 9.1 6.9 1.2 TTNT 7.4 14.3 23.9 13.3 7.0 2.3 OS 23.5 35.4 36.0 35.1 27.8 13.7


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3602-3602 ◽  
Author(s):  
Christopher Fraser ◽  
Patrick A. Brown ◽  
Gail C. Megason ◽  
Hyo Seop Ahn ◽  
Bin Cho ◽  
...  

Abstract Abstract 3602 Background: Acute leukemias, consisting of acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML), are the most common form of childhood cancers. New treatments are needed for patients whose disease progresses or recurs following established therapies. Bendamustine is an alkylating agent that has demonstrated activity in adults with chronic lymphoblastic leukemia and rituximab-refractory non-Hodgkin lymphoma (NHL). Bendamustine has antileukemic activity in adults with AML and myelodysplastic syndrome; however, there are few data regarding bendamustine in children or in childhood acute leukemia specifically. This study is a single-arm, phase 1/2 dose-escalating trial to determine the recommended phase 2 dose (RP2D), schedule, pharmacokinetics, and safety profile of bendamustine in pediatric patients with relapsed and refractory acute leukemia. Methods: Subjects were children aged 1–20 years with relapsed or refractory ALL or AML and without the opportunity for curative therapy. Acute toxic effects of prior therapy (ended ≥2 weeks prior to first dose of study drug) had resolved to grade 2 or less. Bendamustine was infused IV over 60 minutes on days 1 and 2 of each 21-day cycle, with delays allowed up to 2 weeks for neutrophil and platelet recovery. The starting dose was 90 mg/m2/dose with planned escalation to 120 and 150 mg/m2 in cohorts of 3. The dose of 150 mg/m2 was to be implemented only if 120 mg/m2 was safe, but produced subtherapeutic plasma levels compared with data from adults. In phase 2, patients were enrolled at the RP2D in phase 1. Patients were followed until disease progression, withdrawal due to safety or other reasons, loss to follow-up, or a maximum of 12 cycles. After the end of treatment, patients were evaluated every 3 months for 12 months after the last dose, or until progression, death, or start of new cancer treatment. Overall response rate was assessed in all recipients and defined as complete response (CR) or CR without platelet recovery (CRp). Duration of response was assessed in patients who achieved CR or CRp. Biologic activity (≥ partial response [PR]) was also recorded. Safety assessments included adverse events (AEs), concomitant medication throughout treatment, vital signs, and clinical laboratory values. Results: Eleven patients were treated in phase 1 and 32 patients in phase 2. There were 27 patients with ALL and 16 with AML. Twenty-five patients had received >3 chemotherapy regimens and 20 patients had received prior hematopoietic cell transplant in addition to chemotherapy regimens. In phase 1, 5 patients received 90 mg/m2/dose, and 6 received 120 mg/m2/dose. Because no dose-limiting toxicities were observed and therapeutic levels were obtained at 120 mg/m2, the RP2D was determined to be 120 mg/m2. In phase 2, 32 patients received 120 mg/m2. Responses in patients with ALL included 2 patients with CR at 90 mg/m2, 1 had PR, and 7 had stable disease at 120 mg/m2. Two patients with AML had stable disease at 120 mg/m2. Duration of response ranged from 1–8 months with one patient still in remission after unrelated stem cell transplant. Three deaths due to progressive disease occurred in phase 1 and 13 in phase 2. None were considered treatment-related. Thirty-seven patients had at least one grade 3 AE and 19 had at least one grade 4 AE. Twenty patients had grade 3/4 thrombocytopenia (47%), 20 had grade 3/4 anemia (46.5%), and 15 had grade 3/4 febrile neutropenia (35%). Most frequent grade 3/4 non-hematologic AEs were hyperkalemia (21%), dyspnea (9.3%), and tumor lysis syndrome (9.3%). No patients withdrew due to AEs. Conclusions: In pediatric patients with multiple relapsed and refractory ALL and AML, preliminary data suggest that bendamustine has an acceptable safety profile. The RP2D was established as 120 mg/m2, which is identical to that used to treat adults with rituximab-refractory NHL. Response data for the study population suggest that bendamustine has minimal activity in heavily pretreated patients with relapsed and refractory ALL, but not in AML. Further studies will be required to evaluate the role of this agent in combination with regimens that are the backbone of current leukemia therapy in children. This research was sponsored by and conducted by Cephalon, Inc., Frazer, PA. Disclosures: Off Label Use: Bendamustine is FDA-approved for adults with chronic lymphocytic leukemia or indolent B-cell non-Hodgkin's lymphoma that has progressed during or within six months of treatment with rituximab or a rituximab-containing regimen. Frankel:PPD-PharmacoVigilance: Employment, PPD-PharmacoVigilance received funding from Cephalon, Inc. to perform the described analysis. Bensen-Kennedy:Cephalon, Inc.: Employment during the execution of the study. Munteanu:Cephalon, Inc.: Employment. Weaver:Cephalon, Inc.: Employment.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 920-920 ◽  
Author(s):  
Brad Pohlman ◽  
Ranjana Advani ◽  
Madeleine Duvic ◽  
Kenneth B. Hymes ◽  
Tanin Intragumtornchai ◽  
...  

Abstract Abstract 920 Background: Belinostat is a pan-HDAC inhibitor of the hydroxamate chemical class that is well-tolerated and has shown clinical activity. Methods: Open label, multicenter trial enrolling patients (pts) with peripheral T-cell lymphoma (PTCL) or cutaneous T-cell lymphoma (CTCL) who failed ≥ 1 prior systemic therapy. Pts received 1000 mg/m2 IV belinostat over 30 min on days 1 to 5 of a 3-wk cycle. Primary endpoint was objective response (OR) assessed by IWG criteria for PTCL and by SWAT (cutaneous lesions) and IWG criteria (non-cutaneous lesions) for CTCL. Pruritus in pts with CTCL was assessed using a 10-point scale; relief defined as reduction of pruritus score of ≥ 3 points in pts with baseline score ≥ 3. ECGs were monitored and reviewed centrally (pre-/ post-infusion ECGs on all treatment days in cycle 1, and pre-/ post-infusion ECGs on day 1 of subsequent cycles) to evaluate potential cardiac toxicity. Results: The study enrolled a total of 53 treated pts, including 20 and 29 evaluable pts with a diagnosis of PTCL and CTCL, respectively. The 20 pts with PTCL [10 PTCL-unspecified (PTCL-U), 3 anaplastic large cell lymphoma (ALCL), 3 angioimmunoblastic TCL (AITL), 3 NK/T-cell lymphoma, and 1 subcutaneous panniculitis-like TCL (SPTCL)] had received a median of 3 prior systemic therapies (range 1 – 10), and 40 % of them had stage IV disease. 5/20 (25%) PTCL pts responded with 2 CR (both in patients with PTCL-U) and 3 PR (PTCL-U, AITL, ALCL). The 5 responding pts had a median duration of response of 159+ days (range 1 – 504+). Additionally, SD was observed in 5 pts (2 PTCL-U, 2 NK/T-cell, and 1 ALCL) with median duration of SD of 109+ days (range 80 -185+). The 29 pts with CTCL [15 mycosis fungoides (MF), 7 Sezary syndrome (SS), 5 non MF/SS, 2 unclassified] had received a median of 1 prior skin directed therapies (range 0 – 4) and 3 prior systemic therapies (range 1 – 9), and 55 % of them had stage IV disease. 4/29 (14%) CTCL pts responded with 2 CR (ALCL, MF) and 2 PR (MF, SS). The 4 responding pts had a median duration of response of 273 days (range 48 - 469+). Importantly, time to response was short with a median of 16 days (range 14-35). In addition, SD was observed in 17 pts (10 MF, 3 SS, 2 non MF/SS, 2 unclassified) with current duration of up to 127 days. Pruritus relief (score reduction ≥ 3) was seen in 7 of 14 pts with significant baseline pruritis. Median time to pruritus relief was also short, 16 days (range 7-45). Hematological toxicity was minimal without any grade 4 events (shift from baseline) and only one pt each experiencing grade 3 neutropenia and grade 3 thrombocytopenia, respectively. No grade 3 QTcF prolongation was detected in more than 700 ECGs. Four grade 3/4 drug-related AEs were reported: pruritis, rash/erythema, edema, and adynamic ileus. Conclusions: Belinostat monotherapy is safe, well tolerated, and efficacious in pts with recurrent/refractory T-cell lymphoma with durable remissions in both CTCL and PTCL. These results are the basis for a pivotal study with belinostat monotherapy in pts with PTCL. Disclosures: Advani: Seattle Genetics, Inc.: Research Funding. Duvic:Topotarget: research support for conduct of clinical trial. Fagerberg:TopoTarget A/S: Employment, Equity Ownership. Foss:Eisai : Speakers Bureau.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 9055-9055 ◽  
Author(s):  
Charu Aggarwal ◽  
Mary Weber Redman ◽  
Primo Lara ◽  
Hossein Borghaei ◽  
Philip C. Hoffman ◽  
...  

9055 Background: LungMAP is a National Clinical Trials Network umbrella trial for previously-treated SqNSCLC. S1400D is a phase II biomarker-driven therapeutic sub-study evaluating the FGFR inhibitor AZD4547 in patients (pts) with FGFR positive chemo-refractory SqNSCLC. Methods: Eligible pts had tumor FGFR alteration and/or mutation by next generation sequencing (Foundation Medicine), measurable disease, Zubrod PS 0-2, progression after 1 line of systemic therapy, and adequate end organ function. Receipt of prior immunotherapy was allowed. Eligible pts received AZD4547 80 mg bid orally. Primary endpoint was overall response rate (ORR) by RECIST; secondary endpoints included progression-free survival (PFS) and duration of response (DoR). Originally designed as a randomized trial of AZD4547 versus docetaxel, it was redesigned to be a single arm AZD4547 trial with the emergence of immunotherapy as standard 2ndline therapy. Forty pts were required to rule out an ORR of < = 15% if the true ORR was > 35% (90% power, alpha 0.05). Results: 93 pts (13% of pts screened on S1400) were assigned to S1400D; 43 were enrolled with 28 receiving AZD4547. Pt characteristics: median age 66.3 y (49-88), female (n = 8, 29%), & Caucasian (n = 25; 89%). Biomarker profile: FGFR1 amplification (n = 38; 86%); FGFR3 S249C (n = 4; 9%); FGFR3 amplification (n = 3; 7%); and FGFR3 fusion (n = 2; 5%). Nine pts (26%) had more than one biomarker alteration. The study was closed at interim analysis for futility in October 2016. Treatment related Grade 3 AEs were seen in 5 pts (dyspnea, fatigue, hyponatremia, lung infection & retinopathy); 1 pt had Grade 4 sepsis. There were no Grade 5 AEs. Median follow up among alive pts was 4.3 months (mos). Of 25 response evaluable pts, one with FGFR3 S249C had unconfirmed PR (4%, 95% CI 1-20%) with DoR of 1.5 mos. Median PFS was 2.7 mos (95% CI 1.4 - 4.3 mos). Conclusions: This is the first Phase II trial to evaluate AZD4547 as a targeted approach in pts with previously treated FGFR-altered SqNSCLC. AZD4547 had an acceptable safety profile but minimal activity in this biomarker-enriched cohort. Evaluation of other targeted agents in LUNG-MAP is currently ongoing. Clinical trial information: NCT02965378.


2014 ◽  
Vol 32 (26_suppl) ◽  
pp. 147-147 ◽  
Author(s):  
Peter Kaufman ◽  
Martin Olivo ◽  
Yi He ◽  
Susan McCutcheon ◽  
Linda Vahdat

147 Background: PN is a known adverse effect of various antimicrotubule agents, including eribulin. We report here the incidence and resolution of PN in breast cancer pts treated with eribulin in completed phase 2 and 3 studies, and the efficacy of eribulin in pts with PN in the phase 3 EMBRACE and 301 studies. Methods: In EMBRACE, women had received 2-5 lines of chemotherapy for advanced disease. In this ≥ 3rd-line setting, pts randomized to eribulin mesylate received it at 1.4 mg/m2 iv, days 1 and 8 every 21 days. The dosing schedule was the same in study 301, which involved pts who had received 0-2 prior chemotherapies for advanced disease. Overall survival (OS) and progression-free survival (PFS) were analyzed by stratified log-rank test. Data from EMBRACE and 301 were pooled with data from 2 phase 2 studies to assess time to improvement (a decrease of ≥ 1 grade) and resolution (decrease in grade to 0, 1 or baseline) of grade 3 or 4 PN. Results: In the pooled safety analysis, 7.7% (116/1503) of pts treated with eribulin had grade 3 or 4 PN; 63.8% of these experienced improvement in PN and 50% had resolution. Median time to improvement was 2.1 weeks and to resolution was 7.7 weeks. Characteristics were similar in pts with or without PN in EMBRACE and 301. Those with PN had longer exposure to eribulin vs pts without PN (median exposure [months], study 301: 4.9 vs 3.2; EMBRACE: 4.8 vs 2.9). OS and PFS were significantly longer in pts with PN (Table). Conclusions: Pts who had a favourable therapeutic response to eribulin, received a longer course of treatment and thus had a greater risk of PN. Severity of PN improved in most pts within a short period. Regarding PN, the risk–benefit ratio for eribulin supports treatment. Clinical trial information: NCT00337103, NCT00388726. [Table: see text]


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2265-2265 ◽  
Author(s):  
C. Michel Zwaan ◽  
Carmelo Rizzari ◽  
Francoise Mechinaud ◽  
Donna L Lancaster ◽  
Pamela R. Kearns ◽  
...  

Abstract Abstract 2265 Background: Pediatric relapsed/refractory leukemia portends a poor prognosis and more effective therapies are urgently needed. Dasatinib is a potent oral BCR-ABL inhibitor approved for treating adults with chronic myeloid leukemia (CML) or Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) resistant or intolerant to imatinib. Dasatinib also has activity against SRC-family kinases and KIT. A phase 3 trial of dasatinib vs imatinib in adults with newly diagnosed with CML in chronic phase (CP) showed superior efficacy of dasatinib with good tolerability (Kantarjian et al. NEJM 2010:362:2260). Methods: The CA180018 trial is a component of a European Medicines Agency-approved comprehensive Pediatric Investigation Plan for dasatinib aimed at improving outcomes in pediatric leukemias. This trial is being conducted via the ITCC consortium in 7 countries (15 centers) as a stratified phase 1 dose-escalation study. The primary aim is to establish a safe and effective phase 2 dose of dasatinib in children/adolescents with subtypes of relapsed/refractory leukemia. Secondary objectives include safety, pharmacokinetics (PK), and rates of hematologic, cytogenetic, and molecular response (cytogenetic/molecular responses in Ph+ only). Patients (pts) were stratified into 3 disease strata: Stratum 1, imatinib-resistant/intolerant Ph+ CML-CP; Stratum 2/3, advanced CML resistant to imatinib or Ph+ ALL relapsed/refractory after imatinib or Ph+ AML in ≥2nd relapse (original strata merged by protocol amendment due to slow enrolment); and Stratum 4, ≥2nd relapse of Ph– ALL or AML. Starting doses were 60, 80, 100, and 120 mg/m2 once daily, with dose escalations based on safety and efficacy. Intrapatient dose escalation was allowed for lack of response. Results: The study opened in March 2006 and closed to accrual in October 2009. 58 pts have been treated, of which 50 (86%) completed therapy by data cut-off of May 2010. No pts with Ph+ AML were enrolled. All pts had prior therapy, including imatinib in 59% (all Ph+ pts), anagrelide or hydroxyurea in 22%, interferon in 3%, other chemotherapy in 69%, radiotherapy in 43%, and stem cell transplant in 50%. Median age (yrs) was 11, including 2 pts (3%) aged <2, 32 (55%) aged 2–11, 23 (40%) aged 12–18, and 1 (2%) aged >18. 39 pts (67%) were male. No pt with Ph– AML had a KIT mutation. Median durations of therapy (range) were: Stratum 1, 11.3 mos (2.3–47.9); Stratum 2/3, 3.0 mos (0.5–24.6); and Stratum 4, 1.1 mos (<0.1–3.4). Dasatinib up to 120 mg/m2, including long-term therapy, was well tolerated. Common drug-related toxicities (≥10%) were: nausea (grade 1/2 in 16 pts [28%], grade 3 in 1 [2%]); headache (grade 1/2 in 11 [19%], grade 3 in 2 [3%]); diarrhea (grade 1/2 in 12 [21%]); vomiting (grade 1/2 in 9 [16%], grade 3 in 1 [2%]); rash (grade 1/2 in 9 [16%]); and pain in extremity (grade 1/2 in 6 [10%]). Pleural effusion occurred in 2 pts (3%) at grade 1 and 1 pt (2%) at grade 3. Two dose-limiting toxicities were seen in Stratum 4: grade 4 anaphylaxis 5 h after first dose (60 mg/m2) and grade 3 upper GI bleed on Day 6 (120 mg/m2) in a pt with platelet count of 16×109/L. Maximum tolerated dose has not been established. PK parameters, analyzed in 52 pts to date, showed high interpatient and intrapatient variability. Dasatinib was rapidly absorbed with median time to maximum concentration of 1.0 h irrespective of dose. Mean half-life ranged from 2.1–3.6 h. With dasatinib 60, 80, 100, or 120 mg/m2, area under the curve was 374, 530, 424, and 606 ng.h/mL and maximum concentration was 113, 138, 114, and 183 ng/mL, respectively. Treatment responses were seen in Ph+ pts who received dasatinib 60 or 80 mg/m2. In Stratum 1 (CML-CP; n=17), rates were complete hematologic response (HR) in 16 (94%), complete cytogenetic response (CCyR) in 14 (82%), major molecular response (MMR) in 6 (35%), and complete molecular response in 4 (24%). In Stratum 2/3 (advanced CML/Ph+ ALL; n=17), rates were major HR in 10 (59%), CCyR in 12 (71%), and MMR in 0/2 pts with advanced CML assessed to date. No pt in Stratum 4 responded (Ph– ALL/AML; n=24). Final data will be presented. Conclusions: This trial shows the safety and efficacy of dasatinib in pediatric pts with Ph+ leukemias and supports the feasibility of evaluating new agents in children with rare malignancies through cooperative group efforts. A phase 2 study is underway to further evaluate dasatinib in children/adolescents with Ph+ leukemias, including newly diagnosed CML. Disclosures: Zwaan: Bristol-Myers Squibb: Consultancy. Off Label Use: Dasatinib treatment of pediatric leukemias. Rosenberg: Bristol-Myers Squibb: Employment, Equity Ownership. Herdlicka: Bristol-Myers Squibb: Employment. Derreumaux: Bristol-Myers Squibb: Employment. Agrawal: Bristol-Myers Squibb: Employment.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3482-3482 ◽  
Author(s):  
Selim Corbacioglu ◽  
Enric Carreras ◽  
Dietger W Niederwieser ◽  
Marco Sardella ◽  
Margaret Hoyle ◽  
...  

Abstract Abstract 3482 Introduction: Defibrotide (DF), a characterized mixture of polydisperse oligonucleotides, has endothelial protective properties with profibrinolytic, antithrombotic, anti-inflammatory and anti-adhesive activity. The safety of DF injectable and oral formulations has been previously established in clinical trials including more than 9000 pts in the treatment of vascular diseases (eg peripheral arterial disease, deep vein thrombosis), with related adverse events (AE) reported in 2% of these pts. The protective endothelial effects of DF combined with its low toxicity profile led to investigation of its use in the treatment and prevention of hepatic veno-occlusive disease (VOD) in SCT pts. As pts undergoing SCT are likely to experience SCT-associated toxicities, it was difficult to assess the contribution of DF to observed toxicities in the initial single-arm trials. Chemotherapy-induced thrombocytopenia and endothelial cell injury predispose SCT pts to hemorrhagic and thrombotic complications. Bleeding is common in this population, particularly in pts with severe VOD (sVOD), characterized by multi-organ failure (MOF). We therefore undertook a comprehensive review of safety for DF in this setting. Methods and Results: To date, of 1824 SCT pts (pediatric: 737; adult: 1087) who received DF for the prevention or treatment of VOD/ sVOD, 104 (6%) experienced DF-related AEs. Of these 1824, 428 were enrolled in controlled clinical trials: one randomized phase 2 trial comparing 2 doses of DF and two phase 3 trials: one comparing the toxicity profile of DF to a historical control (HC) for the treatment of sVOD, the other to ‘no prophylaxis’ for the prevention of VOD in children. The phase 2 trial randomized 149 pts with sVOD/MOF to receive DF 25 or 40mg/kg/d (pediatric: 48; adult: 101). Both doses were well tolerated. DF-related AEs leading to treatment discontinuation (DC) occurred in 4% of pts: hemorrhage [gastrointestinal (GI), pulmonary, and diffuse alveolar], hypotension, and abdominal cramping. The percentage of pts with Grade 3–4 AEs was similar in both arms, however, pts randomized to 40 mg/kg/d were reported to have a higher incidence of all severity grade bleeding events than pts randomized to 25mg/kg/d (57% versus 46%). As efficacy parameters (CR rate; survival) were equal between the 2 dose groups, while the safety profile of the 25 mg/kg/day dose was slightly improved, the 25 mg/kg/d dose was chosen for future studies. In the phase 3 study, 102 pts with sVOD/MOF (pediatric: 44; adult: 58) treated with 25 mg/kg/d were compared with 32 matched HC pts. DF was generally well tolerated. DF-related AEs (possibly, probably, or definitely related) of Grade 3–5 severity occurred in 27 pts (26%). The following DF-related AEs led to DC of drug in 18 pts (18%): hemorrhage (pulmonary, GI, cerebral, catheter site, and hemorrhage, site not specified; hematochezia and epistaxis), headache, lethargy, hypotension, subdural hygroma, and TTP. Overall AEs, including hemorrhagic AEs, were similarly reported between the DF and control arms. The most frequent AEs were hypotension (39 vs 50%), diarrhea (25 vs 38%), vomiting (21 vs 28%), nausea (15 vs 34%) and pyrexia (15 vs 34%). The most common hemorrhages were epistaxis (13 vs 16%), pulmonary hemorrhage (12 vs 9%), catheter site hemorrhage (12 vs 3%) and hematuria (9 vs 16%). The phase 3 open-label study randomized pediatric pts to receive DF prophylaxis 25mg/kg/d (n=177) or control (n=176) from pre-SCT conditioning until Day+30 post SCT. Prophylactic DF was well tolerated, resulting in few related AEs (5%), including 1 GI hemorrhage which led to DF DC in 1 pt (<1%). AEs were reported at a similar incidence in the DF and control arms, including hemorrhage; the most common were hemorrhagic cystitis (11 vs 11%), GI hemorrhage (2 vs 4%), and epistaxis (2 vs 2%). Pts in both arms who reached the endpoint of VOD were treated with DF (25mg/kg/d); the incidence of DF related AEs in all pts treated for VOD was 14% (8/57). Conclusion: In the SCT population, DF-related AEs occurred with an increased incidence compared to other indications. The incidence was lowest in VOD prophylaxis pts and highest in pts with sVOD. In controlled SCT studies, DF was well tolerated, with AEs (including hemorrhage) reported with similar frequency to the control. These results indicate that DF can be used without increasing the risk of complications in this population of pts at high risk. Disclosures: Corbacioglu: Gentium: Consultancy, Research Funding. Carreras:Gentium: Consultancy. Niederwieser:Gentium: Honoraria. Sardella:Gentium: Employment. Hoyle:Gentium: Employment. Hume:Gentium: Employment. Massaro:Gentium: Consultancy. Hannah:Gentium: Consultancy. Iacobelli:Gentium: Employment. Richardson:Gentium: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 621-621 ◽  
Author(s):  
Shaji Kumar ◽  
Ian W. Flinn ◽  
Paul G Richardson ◽  
Parameswaran Hari ◽  
Natalie Scott Callander ◽  
...  

Abstract Abstract 621 Background: Two- and three-drug regimens incorporating bortezomib (Velcade®, V), lenalidomide (Revlimid®, R), dexamethasone (D), or cyclophosphamide (C) have been shown to be effective and well tolerated in previously untreated multiple myeloma (MM). Combining all four drugs in a single regimen (VDCR) may further enhance efficacy. Published results from the phase 1 dose-escalation portion of the non-comparative, multi-center EVOLUTION study showed that the VDCR regimen was highly active and generally well tolerated (Kumar et al Leukemia 2010). Here we present updated results from the phase 2 portion of the trial, focusing on efficacy and safety of the VDCR, VDR, and VDC regimens. Methods: Previously untreated patients with measurable disease were randomized to one of four treatment groups receiving up to eight 21-d cycles of VDCR (V 1.3 mg/m2 d 1, 4, 8, 11; D 40 mg d 1, 8, 15; R 15 mg d 1–14; C 500 mg/m2 d 1, 8), VDR (VD as in VDCR but with R 25 mg d 1–14), VDC (VDC as in VDCR), or VDC-mod (as for VDC but with an additional dose of C on d 15) as induction therapy, followed by four 42-d maintenance cycles of V 1.3 mg/m2 (d 1, 8, 15, 22) (all treatment arms). Patients eligible for autologous stem cell transplant (SCT) could undergo stem cell mobilization any time after cycle 2 and SCT any time after cycle 4. The primary endpoint was the combined complete response (CR) + very good partial response (VGPR) rate; secondary endpoints included safety/tolerability, time to response, duration of response, progression-free survival, and rate of minimal residual disease (MRD) negativity. Responses were assessed according to International Myeloma Working Group (IMWG) uniform criteria using an automated computer algorithm. Adverse events (AEs) were graded using the CTCAE v3.0. Results: Patient characteristics were similar among the groups with respect to age, performance status, ISS stage, and proportion of patients with high-risk cytogenetic features. Patients received a median of 5, 6, 6, and 6 treatment cycles in the VDCR, VDR, VDC, and VDC-mod arms, respectively; 65%, 60%, 52%, and 47% of patients had dose reductions of any drug. In the VDCR, VDR, VDC, and VDC-mod arms, respectively, 52%, 62%, 58%, and 65% of patients completed treatment; 31%, 43%, 24%, and 41%, respectively, underwent SCT. In the phase 2 response-evaluable patients (n=132), all treatment regimens showed substantial efficacy, with CR+VGPR rates of 59% (VDCR), 50% (VDR), 41% (VDC), and 59% (VDC-mod) (Table) (includes pre-transplant responses only in SCT patients). Of MRD-assessed patients, 46% (21/46) of those who achieved CR (including sCR) or nCR were MRD-negative; 48% (10/21), 75% (9/12), 0% (0/7) and 33% (2/6) in the VDCR, VDR, VDC and VDC-mod arms, respectively. Median time to first response was similar across arms (range 1.6–1.8 months); median time to best response of CR+VGPR was 4.0 months (VDCR), 3.4 months (VDR), 5.1 months (VDC), and 3.1 months (VDC-mod). Median duration of response has not been reached in any arm to date. All treatment regimens were generally well tolerated. In the VDCR, VDR, VDC, and VDC-mod arms, at least one grade ≥3 AE was observed in 81%, 76%, 79%, and 88% of enrolled patients, respectively; serious AEs were experienced by 42%, 40%, 21%, and 41% of patients, and AEs resulting in study discontinuation were reported for 19%, 17%, 12%, and 6% of patients. The five most common all-grade AEs across all treatment groups were fatigue (range 47–67%), nausea (36–67%), constipation (40–62%), diarrhea NOS (42–65%), and neutropenia (19–52%). The incidence of grade ≥3 (grade ≥2) peripheral neuropathy (PN) was 13% (40%) in the VDCR arm, 14% (45%) VDR, 9% (48%) VDC, and 18% (41%) VDC-mod; there was no grade 4 PN. Rates of grade ≥3 neutropenia/thrombocytopenia were 42%/10% for VDCR, 7%/7% VDR, 36%/12% VDC, and 65%/18% VDC-mod. Conclusions: All regimens appear highly active and generally well tolerated in previously untreated MM patients. The four-drug combination did not result in a substantial increase in response rate and was associated with a modest increase in the incidence of hematologic toxicities. Continuous weekly C in the VDC regimen was associated with high response rates and rapid responses, comparable to the VDR and VDCR arms. Outcome data will be presented following longer follow-up. Disclosures: Kumar: Celgene: Consultancy, Research Funding; Millennium: Consultancy, Research Funding; Merck: Consultancy, Research Funding; Novartis: Research Funding; Genzyme: Consultancy, Research Funding; Cephalon: Research Funding; Bayer: Research Funding. Off Label Use: Lenalidomide for treatment of newly diagnosed myeloma. Flinn:Millennium Pharmaceuticals, Inc.: Research Funding. Richardson:Celgene, Millennium, Novartis, Johnson & Johnson: Membership on an entity's Board of Directors or advisory committees. Hari:Celgene: Research Funding. Callander:Millennium Pharmaceuticals, Inc.: Research Funding. Noga:Amgen: Honoraria, Research Funding, Speakers Bureau; Millennium: Consultancy, Honoraria, Research Funding, Speakers Bureau; Ortho-Centicor: Honoraria, Speakers Bureau; Cephalon: Honoraria, Research Funding, Speakers Bureau; Celgene: Honoraria, Research Funding, Speakers Bureau. Stewart:Millennium Pharmaceuticals, Inc.: Honoraria, Research Funding; Celgene: Honoraria. Rifkin:Millennium Pharmaceuticals, Inc.: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Speakers Bureau; Amgen: Speakers Bureau; Cephalon: Speakers Bureau; Dendreon: Speakers Bureau. Wolf:Millennium Pharmaceuticals: Consultancy, Honoraria, Speakers Bureau; Genentech and Multiple Myeloma Research: Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Speakers Bureau; OrthoBiotech: Honoraria, Speakers Bureau; Celgene: Honoraria, Speakers Bureau. Estevam:Millennium Pharmaceuticals: Employment. Mulligan:Millennium Pharmaceuticals, Inc.: Employment. Shi:Millennium Pharmaceuticals: Employment. Webb:Millennium Pharmaceuticals: Employment.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3662-3662 ◽  
Author(s):  
Myron S. Czuczman ◽  
Dominick Lamonica ◽  
Shari K. Gaylor ◽  
Leonard Bush ◽  
Penny Nadolny ◽  
...  

Abstract Abstract 3662 Background Mantle-cell lymphoma (MCL) is an aggressive non-Hodgkin lymphoma (NHL) subtype, typically presenting as stage IV disease. There is no established standard of care; chemotherapy usually results in a tumor response, but cure is rare and the median survival is 3 to 5 years. Bendamustine, a unique alkylating agent, and rituximab, an anti-CD20 monoclonal antibody, showed respective overall response rates (ORR) of 75% and 92% in 16 and 12 patients with relapsed/refractory MCL in two open-label, phase 2 clinical trials on NHL and MCL. Methods This 24-week, multicenter, phase 2 study included adults with relapsed/refractory, CD20-positive, B-cell MCL (nonblastoid) who received ≤3 prior chemotherapies. Patients received intravenous (IV) bendamustine 90 mg/m2 on days 1 and 2 and IV rituximab 375 mg/m2 on day 1 (except patients with an absolute lymphocyte count >5 × 109/L that in cycle 1, received rituximab across 3 days and bendamustine on days 3 and 4) for six 28-day treatment cycles (up to 8 cycles for those without disease progression [PD] and without a documented complete response [CR]). Patients received analgesics/antipyretics prior to rituximab administration. The primary efficacy measure was ORR (CR + partial response [PR]). Secondary efficacy measures included duration of response, progression-free survival (PFS), overall survival (OS), and rate of conversion from positron emission tomography (PET)-positive to PET-negative disease. Safety measures included adverse events (AEs). Data from treatment cycles 7 to 8 were included only in the safety analysis. Study enrollment was terminated by the sponsor for nonclinical reasons. Data analysis is ongoing, and final data will be presented. Results Of 45 patients, median age was 71 (range 48–88) years. Median number of prior chemotherapies was 1 (range 1–3). Seven patients discontinued study treatment early (2 each during cycles 2 and 3; 1 each during cycles 1, 5, and 7); 2 patients withdrew consent from the study, 2 discontinued due to AEs (thrombocytopenia; back pain; grade 5 myocardial infarction, pneumonia, and respiratory failure), and 3 due to PD. Median treatment duration was 6 (range 1–8) cycles. ORR was 82% (38% CR, 44% PR) (90% CI 70.2%-90.8%); median duration of response was 14.5 months (range 11.3 months to not reached). Median PFS was 16.4 months, with 1-year PFS rate of 62%. Median 3-year OS has not been reached by Kaplan-Meier analysis. Image-based metabolic profiling in analysis of response is ongoing. Eleven patients (24%) had a bendamustine dose reduction, but no patient had a rituximab dose reduction. There were a total of 24 delayed treatment cycles (median 1, range 0–3), 83% of which had ≤21 days of delay. The most common reasons for dose reduction/delay were neutropenia (n=11) and thrombocytopenia (n=8). Seventeen patients had serious AEs; the most frequent were pneumonia (n=3), confusional state (n=2), and pleural effusion (n=2), which were considered unrelated to bendamustine. Grade 3/4 hematologic laboratory toxicities were lymphopenia (n=40), neutropenia (n=20), leukopenia (n=20), thrombocytopenia (n=3), and anemia (n=2). The most common nonhematologic grade 3/4 AEs were hypokalemia and hypotension (n=3 each). There were 8 patients with grade 3/4 infections and infestations (any term). Four patients (9%) had an infusion-related reaction, and 22 patients (49%) received growth factors during treatment. One death occurred during the study due to myocardial infarction, pneumonia, and respiratory failure; this was considered to be unrelated to study treatment. Conclusions Bendamustine plus rituximab showed a high ORR with a long duration of response. It was adequately manageable in patients with relapsed/refractory MCL, with a low rate of infusion-related reaction. Support: Teva Pharmaceutical Industries Ltd. Disclosures: Czuczman: Mundipharma: Honoraria; Genentech: Consultancy. Off Label Use: Bendamustine is FDA-approved for adults with chronic lymphocytic leukemia or indolent B-cell non-Hodgkin's lymphoma that has progressed during or within six months of treatment with rituximab or a rituximab-containing regimen. Lamonica:Teva: Consultancy. Gaylor:Veeda Oncology, which received research funding from Teva Pharmaceuticals: Employment, Research Funding. Bush:Veeda Oncology, which received research funding from Teva Pharmaceuticals: Employment, Research Funding. Nadolny:Veeda Oncology, which received research funding from Teva Pharmaceuticals: Employment, Research Funding. Colborn:Veeda Oncology, which received research funding from Teva Pharmaceuticals: Employment, Research Funding.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e19507-e19507
Author(s):  
Yuqin Song ◽  
Quanli Gao ◽  
Huilai Zhang ◽  
Lei Fan ◽  
Jianfeng Zhou ◽  
...  

e19507 Background: Tislelizumab is a humanized IgG4 monoclonal antibody with high affinity/specificity for programmed cell death protein 1 (PD-1). It was engineered to minimize binding to Fc-γ receptors on macrophages, thereby decreasing antibody-dependent phagocytosis, a potential mechanism of T-cell clearance and resistance to anti–PD-1 therapy. Tislelizumab therapy was highly active in autologous stem cell transplant (ASCT)-failed or ineligible patients with R/R cHL ( Leukemia. 2020;34:533). Here we report results from up to 3 years follow-up. Methods: This asingle-arm, multicenter phase 2 study (NCT03209973) of 200 mg tislelizumab administered intravenously to patients (pts) with R/R cHL every 3 weeks until progressive disease (PD) or unacceptable toxicity. Patients were eligible if they: failed to achieve a response or progressed after ASCT, or: received ≥2 lines of prior systemic chemotherapy for cHL and were ineligible for ASCT. Primary endpoint was overall response rate (ORR) assessed by an independent review committee (IRC) per Lugano criteria ( J Clin Oncol. 2014;32:3059). Secondary endpoints were progression-free survival (PFS), duration of response (DOR), complete response (CR) rate, and time to response (TTR) per IRC, safety, and tolerability. Results: Pts (N=70) from 11 centers in China were enrolled and treated; characteristics have been previously reported. As of the data cutoff date (Nov 2, 2020), median follow-up was 33.8 months (range, 3.4-38.6). Pts still on treatment at the end of study (n=33; 47.1%) entered a long-term extension study. Efficacy data is presented in the Table below. In the 13 pts who received prior ASCT, 11 (84.6%) achieved CR. The most common treatment-emergent adverse events (AEs; ≥30%) were pyrexia (57.1%), upper respiratory tract infection (38.6%), hypothyroidism (37.1%), and increased weight (34.3%). Treatment-related grade ≥3 AEs (≥2 pts) were pneumonitis, hypertension, neutropenia, lipase increased, weight increased, and increased creatine phosphokinase (CPK; 2.9% each). Immune-related AEs were reported in 32 pts (45.7%), with grade ≥3 AEs in 8 pts (11.4%): pneumonitis (4) and skin adverse reactions, nephritis, lipase increased, and blood CPK increased (1 each). AEs led to treatment discontinuation in 6 pts (8.6%). Conclusions: Long-term follow-up of R/R cHL pts treated with tislelizumab further demonstrated the substantial therapeutic activity and continued PFS benefit. There were no new safety concerns identified for long-term treatment with tislelizumab. Clinical trial information: NCT03209973. [Table: see text]


Sign in / Sign up

Export Citation Format

Share Document