5 research outputs found

    РЕЗУЛЬТАТЫ МНОГОЦЕНТРОВОГО ДВОЙНОГО СЛЕПОГО РАНДОМИЗИРОВАННОГО КЛИНИЧЕСКОГО ИССЛЕДОВАНИЯ ПЕРВОЙ ФАЗЫ ПРЕПАРАТА BCD-022 ПО СРАВНЕНИЮ С ПРЕПАРАТОМ ГЕРЦЕПТИН®, ПРИМЕНЯЕМЫХ В СОЧЕТАНИИ С ПАКЛИТАКСЕЛОМ У БОЛЬНЫХ МЕТАСТАТИЧЕСКИМ РАКОМ МОЛОЧНОЙ ЖЕЛЕЗЫ

    No full text
    Within the framework of multicenter double-blind randomized clinical trial studied pharmacokinetics and safety of BCD-022 (trastuzumab, "Biocad" company, Russia), compared with the drug Herceptin (trastuzumab, F. Hoffmann-La Roche Ltd., Switzerland). Evaluation of the effectiveness was not the aim of the interim analysis, the results of which are shown. BCD-022 and Herceptin were used in combination with paclitaxel in patients with metastatic breast cancer with HER2 overexpressing (HER2 (+), mBC).Methods. The analysis included 46 patients with HER2 (+) metastatic breast cancer (mBC) at the age of 29 to 71 years (22 - in the group of studied drug BCD-022 and 24 - in the Herceptin group). All patients received one course of therapy of BCD-022 or Herceptin 8 mg/kg intravenously and paclitaxel 175 mg/m2 intravenously on day 1 of a three-week course of treatment and continue to receive treatment with the same scheme with the use of trastuzumab 6 mg/kg (mandatory in the study is to conduct 6 courses of therapy). Randomization was carried out in groups in a ratio of 1: 1. The primary endpoint of pharmacokinetics evaluation was the area under the curve "concentration-time» (AUC0-504) of trastuzumab after a single application, the secondary - Cmax, T1 / 2 and Tmax. Safety was assessed based on the incidence of adverse events after the first course of therapy.Results. Haematological toxicity, myalgia and arthralgia were the most frequent adverse events. Most reported adverse events had mild to moderate grade according to CTCA 4.03 and were caused  by effect of myelosuppressive chemotherapy. There were no statistically significant differences in adverse events frequency between the groups. There were 6 serious adverse events: 2 - in the BCD-022 group and 4 - in the Herceptin group. All pharmacokinetic parameters, including the primary endpoint (AUC 0-504) and secondary endpoints (Cmax, T1 / 2 and Tmax), of studied drug BCD-022 and Herceptin had no statistically significant difference.Conclusion. BCD-022 (trastuzumab, "Biocad" company, Russia) regarding to its safety profile and pharmacokinetic properties is fully consistent with the original drug trastuzumab Herceptin (F. Hoffmann-La Roche Ltd., Switzerland) and can be recommended for further clinical study.В рамках многоцентрового двойного слепого рандомизированного клинического исследования была изучена фармакокинетика и безопасность препарата BCD-022 (трастузумаб, ЗАО «БИОКАД», Россия) по сравнению с препаратом Герцептин® (трастузумаб, Ф. Хоффманн-Ля Рош Лтд, Швейцария). Оценка эффективности не входила в задачи промежуточного анализа, результаты которого представлены. BCD-022 и Герцептин® применялись в комбинации с паклитакселом у пациенток метастатическим раком молочной железы с гиперэкспрессией HER2 (HER2 (+) мРМЖ).Методы. В анализ включено 46 больных HER2 (+) мРМЖ в возрасте от 29 до 71 года (22 — в группу исследуемого препарата BCD-022, 24 — в группу Герцептин®). Все пациентки получили 1 курс терапии по схеме BCD-022 или Герцептин® 8 мг/кг внутривенно капельно и паклитаксел 175 мг/м2 внутривенно в 1 день трехнедельного курса и продолжают лечение по той же схеме с использованием трастузумаба в дозе 6 мг/кг (обязательным в исследовании является проведение 6 курсов терапии). Рандомизация в группы производилась в соотношении 1:1. Первичной конечной точкой для оценки фармакокинетики была площадь под кривой «концентрация-время» (AUC0–504) трастузумаба после однократного применения, вторичными — Cmax, T1/2 и Tmax. Безопасность оценивалась на основании частоты нежелательных явлений после первого курса терапии.Результаты. Среди нежелательных явлений наиболее часто встречалась гематологическая токсичность, миалгия, артралгия. Большинство зарегистрированных нежелательных явлений имели легкую и умеренную степень по СТСАЕ 4.03 и были обусловлены проведением миелосупрессивной химиотерапии. Статистически значимых различий между группами не было выявлено ни по одному из нежелательных явлений. Зарегистрировано 6 серьезных нежелательных явлений: 2 — в группе исследуемого препарата BCD-022 и 4 — в группе Герцептин®. Все фармакокинетические показатели, включая первичную конечную точку (AUC0–504) и вторичные конечные точки (Cmax, T1/2 и Tmax), исследуемого препарата BCD-022 и Герцептин® не имели статистически значимых отличий.Заключение. BCD-022 (трастузумаб, ЗАО «БИОКАД», Россия) по своему профилю безопасности и фармакокинетическим свойствам полностью соответствует оригинальному препарату трастузумаба Герцептин® (Ф. Хоффманн-Ля Рош Лтд, Швейцария) и может быть рекомендован для дальнейшего клинического изучения

    Rituximab biosimilar RTXM83 versus reference rituximab in combination with CHOP as first-line treatment for diffuse large B-cell lymphoma: a randomized, double-blind study

    No full text
    This multicenter, double-blind, randomized study compared the efficacy, pharmacokinetics (PKs)/pharmacodynamics (PDs), safety and immunogenicity profile of RTXM83 vs. reference rituximab (R-rituximab), both with CHOP, as first-line treatment of diffuse large B-cell lymphoma (DLBCL). A total of 272 patients <65 years of age, with good prognosis (136 per arm) were randomized (1:1) to receive six cycles of either RTXM83 or R-rituximab. The primary efficacy endpoint was achieved (overall response rate of 83.6% for RTXM83 and 82.9% for R-rituximab) with a difference 0.7% between arms (95%CI: [-8.77% to 10.17%]) fulfilling the predefined non-inferiority margin (-13%). Similar number of patients reported at least one adverse event (AE) (131 per arm) or one serious AE (47 with RTXM83 and 45 with R-rituximab). Anti-drug antibody development was comparable between the arms. PK/PD secondary endpoint results support similarity between the compounds. RTXM83 exhibits non-inferior efficacy and similar safety/immunogenicity to R-rituximab, being an accessible alternative for the treatment of patients with previously untreated DLBCL

    Ibrutinib combined with bendamustine and rituximab compared with placebo, bendamustine, and rituximab for previously treated chronic lymphocytic leukaemia or small lymphocytic lymphoma (HELIOS): a randomised, double-blind, phase 3 study

    No full text
    Background: Most patients with chronic lymphocytic leukaemia or small lymphocytic lymphoma relapse after initial therapy. Bendamustine plus rituximab is often used in the relapsed or refractory setting. We assessed the efficacy and safety of adding ibrutinib, an oral covalent inhibitor of Bruton's tyrosine kinase (BTK), to bendamustine plus rituximab in patients with previously treated chronic lymphocytic leukaemia or small lymphocytic lymphoma. Methods: The HELIOS trial was an international, double-blind, placebo-controlled, phase 3 study in adult patients (>= 18 years of age) who had active chronic lymphocytic leukaemia or small lymphocytic lymphoma with measurable lymph node disease (>1.5 cm) by CT scan, and had relapsed or refractory disease following one or more previous lines of systemic therapy consisting of at least two cycles of a chemotherapy-containing regimen, an Eastern Cooperative Oncology Group (ECOG) performance status of 0-1, and adequate bone marrow, liver, and kidney function. Patients with del(17p) were excluded because of known poor response to bendamustine plus rituximab. Patients who had received previous treatment with ibrutinib or other BTK inhibitors, refractory disease or relapse within 24 months with a previous bendamustine-containing regimen, or haemopoietic stem-cell transplant were also excluded. Patients were randomly assigned (1:1) by a web-based system to receive bendamustine plus rituximab given in cycles of 4 weeks' duration (bendamustine: 70 mg/m.intravenously on days 2-3 in cycle 1, and days 1-2 in cycles 2-6; rituximab: 375 mg/m.on day 1 of cycle 1, and 500 mg/m.on day 1 of cycles 2-6 for a maximum of six cycles) with either ibrutinib (420 mg daily orally) or placebo until disease progression or unacceptable toxicity. Patients were stratified according to whether they were refractory to purine analogues and by number of previous lines of therapy. The primary endpoint was independent review committee (IRC)-assessed progression-free survival. Crossover to ibrutinib was permitted for patients in the placebo group with IRC-confirmed disease progression. Analysis was by intention-to-treat and is continuing for further long-term follow-up. The trial is registered with ClinicalTrials.gov, number NCT01611090. Findings: Between Sept 19, 2012, and Jan 21, 2014, 578 eligible patients were randomly assigned to ibrutinib or placebo in combination with bendamustine plus rituximab (289 in each group). The primary endpoint was met at the preplanned interim analysis (March 10, 2015). At a median follow-up of 17 months (IQR 13.7-20.7), progression-free survival was significantly improved in the ibrutinib group compared with the placebo group (not reached in the ibrutinib group (95% CI not evaluable) vs 13.3 months (11.3-13.9) in the placebo group (hazard ratio [HR] 0.203, 95% CI 0.150-0.276; p<0.0001). IRC-assessed progression-free survival at 18 months was 79% (95% CI 73-83) in the ibrutinib group and 24% (18-31) in the placebo group (HR 0.203, 95% CI 0.150-0.276; p<0.0001). The most frequent all-grade adverse events were neutropenia and nausea. 222 (77%) of 287 patients in the ibrutinib group and 212 (74%) of 287 patients in the placebo group reported grade 3-4 events; the most common grade 3-4 adverse events in both groups were neutropenia (154 [54%] in the ibrutinib group vs 145 [51%] in the placebo group) and thrombocytopenia (43 [15%] in each group). A safety profile similar to that previously reported with ibrutinib and bendamustine plus rituximab individually was noted. Interpretation: In patients eligible for bendamustine plus rituximab, the addition of ibrutinib to this regimen results in significant improvements in outcome with no new safety signals identified from the combination and a manageable safety profile

    Talactoferrin alfa versus placebo in patients with refractory advanced non-small-cell lung cancer (FORTIS-M trial)

    No full text
    Background: Talactoferrin alfa is an oral dendritic cell (DC)-mediated immunotherapy (DCMI). We tested whether talactoferrin was superior to placebo in advanced non-small-cell lung cancer (NSCLC). Patients and methods: An FORTIS-M trial was an international, multicenter, randomized, double-blind comparison of talactoferrin (1.5 g p.o. BID) versus placebo BID, in patients with stage IIIB/IV NSCLC whose disease had failed two or more prior regimens. Treatment was administered for a maximum of five 14-week cycles. The primary efficacy end point was overall survival (OS); secondary end points included 6- and 12-month survival, progression-free survival (PFS), and disease control rate (DCR). Results: Seven hundred and forty-two patients were randomly assigned (2:1) to talactoferrin (497) or placebo (245). The median OS in the intent-to-treat (ITT) population was 7.66 months in the placebo arm and 7.49 months in the talactoferrin arm [hazard ratio (HR), 1.04; 95% CI, 0.873-1.24; P = 0.6602]. The 6-month survival rates were 59.9% (95% CI, 53.4% to 65.8%) and 55.7% (95% CI, 51.1% to 59.9%), respectively. The 12-month survival rates were 32.2% (95% CI, 26.3% to 38.2%) and 30.9% (95% CI, 26.8% to 35%), respectively. The median PFS rates were 1.64 months and 1.68 months, respectively (HR, 0.99; 95% CI, 0.835-1.16; P = 0.8073). The DCRs were 38.4 and 37.6%, respectively [stratified odds ratio (OR), 0.96; 95% CI, 0.698-1.33; P = 0.8336]. The safety profiles were comparable between arms. Conclusions: There was no improvement in efficacy with talactoferrin alfa in patients with advanced NSCLC whose disease had failed two or more previous regimens. © The Author 2013. Published by Oxford University Press on behalf of the European Society for Medical Oncology All rights reserved

    Frontline bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone (VR-CAP) versus rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in transplantation-ineligible patients with newly diagnosed mantle cell lymphoma: final overall survival results of a randomised, open-label, phase 3 study

    No full text
    Background: In the LYM-3002 study, the efficacy and safety of frontline bortezomib plus rituximab, cyclophosphamide, doxorubicin, and prednisone (VR-CAP) and rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) were compared in transplant-ineligible patients with untreated, newly diagnosed, mantle cell lymphoma. We report the final overall survival and safety outcomes for patients in the long-term follow-up phase after the primary progression-free-survival endpoint was met. Methods: LYM-3002 was a randomised, open-label, phase 3 study done at 128 clinical centres in 28 countries in Asia, Europe, North America, and South America. Adult patients with confirmed stage II–IV previously untreated mantle cell lymphoma, Eastern Cooperative Oncology Group performance status score of 2 or less, who were ineligible for bone marrow transplantation, were randomly assigned (1:1) to receive six or eight 21-day cycles of VR-CAP (intravenous rituximab 375 mg/m2, cyclophosphamide 750 mg/m2, doxorubicin 50 mg/m2, and bortezomib 1·3 mg/m2, plus oral prednisone 100 mg/m2) or R-CHOP (intravenous vincristine 1·4 mg/m2 [2 mg maximum], rituximab 375 mg/m2, cyclophosphamide 750 mg/m2, and doxorubicin 50 mg/m2, plus oral prednisone 100 mg/m2). Randomisation was done according to a computer-generated randomisation schedule prepared by the sponsor; permuted blocks central randomisation was used (block size of 4), and was stratified by International Prognostic Index score and disease stage at diagnosis. The primary endpoint of this final analysis was overall survival, which was analysed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT00722137, and is closed to new participants with follow-up completed. Findings: Between May 22, 2008, and Dec 5, 2011, 487 patients were enrolled and randomly assigned. 268 patients (140 in the VR-CAP group and 128 in the R-CHOP group) were included in the follow-up analysis, which included patients with data available after the primary analysis clinical cutoff date of Dec 2, 2013. After median follow-up of 82·0 months (IQR 74·1–94·2), median overall survival was significantly longer in the VR-CAP group than in the R-CHOP group (90·7 months [95% CI 71·4 to not estimable] vs 55·7 months [47·2 to 68·9]; hazard ratio 0·66 [95% CI 0·51–0·85]; p=0·001). Three new adverse events were reported since the primary analysis cutoff (one each of grade 4 lung adenocarcinoma and grade 4 gastric cancer in the VR-CAP group, and one case of grade 2 pneumonia in the R-CHOP group). 103 (42%) of 243 patients in the VR-CAP group, and 138 (57%) of 244 in the R-CHOP group died; the most common cause of death was progressive disease. Interpretations: Compared with R-CHOP, VR-CAP was associated with significantly longer survival, and had a manageable and expected safety profile. Our results support further assessment of VR-CAP in patients with previously untreated mantle cell lymphoma. Funding: Janssen Research &amp; Development
    corecore