生物类似药ch COriginal and Biosimilar Epoetin Productsonsistency of 质量分析Quality and Batch-to-Bat
天津大学制药工程专业 考研《制药工艺学》中英文对译
天津⼤学《制药⼯艺学》中英⽂对译pharmaceutical technology制药⼯工艺学pharmaceutical pipeline制药链pharmacopoeia药典。
Roswell Park Memorial Instirute RPMI good manufacturing practices for drugs GMP制药⾏行行业medicines,drugs药品traditional Chinese medicines中药natural medicines天然药物chemical drugs化学药物biologics,biologic products⽣生物制品generics,generic drugs仿制药物me-too-drug仿制药biosimilars⽣生物类似药biotechnology⽣生物技术.Food and druge administration FDA biotechnology pharmaceutical,biopharmaceutical⽣生物制药nucleotide核苷酸nucleoside核⽢甘enzyme酶enzyme inhibitor酶抑制剂immunomodulator免疫调节剂penicillin⻘青霉素antibody engineering抗体⼯工程inducer诱导剂precursor前体prodrug前药transformation遗传转化.conversion⽣生物转化fermentation发酵.strain breeding菌种选育separation and purification分离纯化和提纯.cell growth phase/fermentationproduct synthesis phase/product secretion phase.Murashige&Skoog MScell autolysis phase/fermentation anaphase.generic通⽤用药物metabolism代谢.substrate培养基质primary/secondary metabolism初级/次级代谢.specific growth rate⽐比⽣生⻓长速率lag/log/decline/stationary/death phase延滞期/对数⽣生⻓长期/减数期/稳定期/死亡期coupling model⽣生⻓长与⽣生产偶联型.PEG聚⼄乙⼆二醇semi-coupling model⽣生⻓长与⽣生产半偶联型.starter culture培养物non-coupling model⽣生⻓长与⽣生产⾮非偶联型.storage保存protoplast fusion原⽣生质体融合.DMSO⼆二甲基亚砜master stock/cell bank MSB/MCB主菌种库.glycerol⽢甘油working stock/cell bank WSB/WCB⼯工作菌种库.Streptomyces链霉菌quality control QC质量量控制.cholramphenicol氯霉素China Center for Type Culture Collection CCTCC中国典型培养物保藏中⼼心China General Microbiological Culture Collection Center CGMCC中国普通微⽣生物保藏管理理中⼼心China Center of Industrial Culture Collection CICC中国⼯工业微⽣生物菌种保藏管理理中⼼心National Center for Medical Culture Collection(Bacteria)CMCC中国医学微⽣生物菌种保藏中⼼心America Type Culture Collection ATCCEuropean Collection of Cell Culture ECACCInstiture for Fermentation,Osaka IFONational Collection of Type Culture mNCTCmedium培养基.carbon source碳源nitrogen source氮源.mineral salt⽆无机盐macroelement⼤大量量元素.trace element microelement微量量元素growth factor⽣生⻓长因⼦子.precursor前体accelerant促进剂.fed medium补料料培养基agar琼脂粉.contaminated microbe杂菌contamination污染.phage噬菌体disinfection消毒.sterilization灭菌pathogen病原微⽣生物.filtration sterilization过滤灭菌filter过滤介质VVM空⽓气流量量(单位时间单位体积内通⼊入的标准状况下的空⽓气体积)primary culture原代培养.passage culture传代培养solid surface culture固体表⾯面培养.liquid submerged culture液体深层培养immobilized culture固定化培养.high cell density culture⾼高密度培养intermittent opration间歇式操作.discontinuous operation不不连续培养semi-continuous operation半连续培养.batch operation分批式操作fed batch operation补料料-分批式(流加)操作.chemostat恒化器器MPa罐压.dissolved oxygen DO溶解氧cell concentration菌体浓度.fermentation heat发酵热production heat产⽣生热.loss heat散失热biological heat⽣生物热.agitation heat搅拌热evaporation heat蒸发热.sensible heat显热radiant heat辐射热.oxygen supply供氧oxygen consumption耗氧.dissolved oxygen coefficient溶解氧系数oxygen transfer rate OTR氧传递速率.oxygen uptake rate OUR摄氧速率ventilation通⽓气.respiratory intensity呼吸强度oxygen saturation concentration氧饱和浓度.respiratory quotient RQ呼吸熵critical oxygen concentration临界氧浓度.fill补料料withdraw放料料.foam泡沫defoaming agent消沫剂.surfactant表⾯面活性剂dispersant分散剂.emulsifier乳化剂inertcarrier惰性载体.antibiotic抗⽣生素carbenicillin羧苄⻘青霉素/⻘青霉素G6-aminopenicillanic acid6-APA6-氨基⻘青霉烷酸.cephalosporin C CPC头孢菌素C erythromycin红霉素.amino acid氨基酸hybridomn杂交瘤.vitamin维⽣生素recombinant DNA technology重组DNA技术.recombinant DNA products rDNA制品plasmid质粒.replicon复制⼦子promoter启动⼦子.terminator终⽌止⼦子multiple cloning site MCS多克隆隆位点.transferability转移性incompatibility不不相容性.cloning vector克隆隆载体expression vector表达载体.shuttle vector穿梭载体intergration vector整合载体.inclusion body包涵体yeast酵⺟母.genetic engineering strain基因⼯工程菌yeast intergration plasmid YIP酵⺟母整合载体yeast episomal plasmid YEP酵⺟母附加载体yeast centromere plasmid YCP酵⺟母着丝粒载体centromere sequence CEN着丝粒序列列autonomously replicating sequences ARS⾃自主复制序列列yeast replicating plasmid YRP酵⺟母复制质粒polymerase chain reaction PCR聚合酶链式反应reverse transcription PCR RT-PCR反转录PCRcomplementary DNA cDNAavian myeloblastosis virus AMV禽源成髓细胞瘤病毒moloney murine leukemia virus MMLV⿏鼠源败⾎血病毒莫勒勒尼株diethyl pyrocarbonate DEPC焦碳酸⼆二⼄乙酯.denaturation变性annelling退⽕火.extension链延伸restriction endonuclease限制性核酸内切酶.ligase连接酶recombinant重组⼦子.interferon IFN⼲干扰素recombinant human interferon rhIFNtricarboxylic acid cycle TCA循环三羧酸循环pentose phosphate pathway PPP磷酸戊糖途径.glycosylation糖基化apoptosis凋亡.diploid cell⼆二倍体primary cell原代细胞.passage cell传代细胞immortal cell永久细胞系.Chinese hamster ovary CHO中国仓⿏鼠卵卵巢DHFR⼆二氢叶酸还原酶.methotrexate MTX甲氨蝶呤baby hamster kidney BHK幼仓⿏鼠肾脏dicistron双顺反⼦子long terminal repeat sequences LTRS逆转录病毒的⻓长末端重复序列列cytomegalovirus CMV⼈人巨噬病毒.ubiquitin泛素蛋⽩白bovine growth hormone.BGH⽜牛⽣生⻓长素.toppoisomerase拓拓扑异构酶internal ribosome entry site IRES核糖体进⼊入位点.serum⾎血清minimum essential medium MEM basal medium Eagle’s BME Dulbecco’s modified Eagle’s medium DMEMGlasgow’s modified Eagle’s medium GMEMJoklik’s Park Memorial Eagle’s medium JMEMRoswell Park Memorial Institute RPMIserum-free medium SEM⽆无⾎血清培养基.buffer solution缓冲液balance saline solution BSS平衡盐溶液monolayer anchorage-dependent culture单层贴壁培养.suspension culture悬浮培养microcarrier微载体.microencapsulation method微囊法phosphonate buffer solution PBS磷酸盐缓冲液.scale-down缩⼩小erythropoietin EPO红细胞⽣生成素.luria bertani LB recombinant human erythropoietin rhEPO重组⼈人红细胞⽣生成素synthon合成⼦子.synthetic equivalent合成等价物protocol solvent质⼦子性溶剂.micronization微晶化catalyst催化剂.phase transfer catalyst相转移催化剂TEBAC三⼄乙基苄基氯化铵Mokosza催化剂TO/CMAC三⾟辛基甲基氯化铵Starks催化剂.Brandstrom催化剂四丁基硫酸氢铵chirality⼿手性.enantiomers对应异构体configuration构型.chiral drug⼿手性药物enantiomeric excesses对映体过量量e.e.%.restrosynthesis追溯求源法resolution拆分.omeprazole奥美拉唑paclitaxel,Taxol紫杉醇.cephalosporin C CPC头孢菌素7-aminocephalosporanic acid7-ACA7-氨基头孢烷酸.cefalexin头孢氨苄tetrahydrofuran THF四氢呋喃.quality by design QbD质量量源于设计process analysis technology PAT过程分析技术quality target product profile QTPP⽬目标产品质量量概况critical material attribute CMA关键物料料属性critical process parameter CPP关键⼯工艺参数normal operation range NOR正常操作区间proven acceptable range PAR可接受的区间critical quality attribute CQA关键质量量属性bioreactor⽣生物反应器器key process parameter KPP重要⼯工艺参数.fermenter发酵罐complete stirred tank reactor CSTR全混流反应器器.yield得率piston fluid reactor PFR平推流反应器器.titer效价stirred tank reactor STR搅拌罐.scale-up放⼤大fixed bed reactor固定化床反应器器.draft tube导流筒packed bed reactor PBR填充床反应器器.bubble column⿎鼓泡塔fluidized bed reactor FBR流化床反应器器.air-lift reactor⽓气升式反应器器disk and turbine impeller涡流式搅拌桨.process validation⼯工艺验证marine style impeller推进式搅拌桨.process design⼯工艺设计process mass intensity PMI过程质量量强度.process qualification⼯工艺确认reaction mass efficiency RME反应质量量效率standard operation procedure SOP标准操作规程.continued process verification⼯工艺核实biochemical oxygen demand BOD⽣生化需氧量量.total nitrogen TN总氮chemical oxygen demand COD化学需氧量量.suspended subatance SS悬浮物mixed liquor suspended solids MLSS混合液悬浮固体total organic carbon TOC总有机碳.sludge volume SV污泥泥沉降⽐比sludge volume index SVI污泥泥指数piping&instrument diagram PID⼯工艺控制流程图。
常规诊疗条件下比较依那西普生物类似药(益赛普)与阿达木单抗、英夫利西单抗治疗RA的临床疗效
常规诊疗条件下比较依那西普生物类似药(益赛普)与阿达木单抗、英夫利西单抗治疗RA的临床疗效PresentID: SAT0360原文TANAR- A ETANERCEPT BIOSIMILAR IS AS EFFECTIVE AS ADALIMUMAB AND INFLIXIMAB IN ACOHORT OF REAL-LIFE OF PATIENTS WITH RHEUMATOID ARTHRITISP. Santos-Moreno1,*, G.Saavedra-Martinez2, L. Villarreal3, D. Gomez1, J. Bello-Gualtero1, V. Giraldo4,P. Martinez4, A. Sanchez4, M. Sanchez4, E. Uribe4, M. Boon41Rheumatology, 2Epidemiology,3Psychology, 4Internal medicine, Biomab, Center For Rheumatoid Arthritis,Bogota, Bogota, ColombiaBackground: Clinical response in patients with rheumatoid arthritis (RA) using biologics is well-known. However, there is no direct comparison between biologics in cohorts of patients with RA inreal-life settings, which could have implications in treatment decisions andhealth economics.Objectives: The aim of this study was to describe a direct comparison in effectiveness between two classical antiTNF biologics (Adalimumab, Infliximab) and one Etanercept biosimilar in patients with long-standing RA in a cohort of real-life.Methods: A descriptive cross-sectionalstudy was performed. Were included 158 patients with at least 6 visits torheumatologist in last 24 months in a specialized in RA center. Clinical follow-up was designed by the authors according to DAS28 as follows: every 3-5weeks (DAS28 > 5.1), every 7-9 weeks (DAS28 ≥ 3.1 and ≤ 5.1), and every11-13 weeks (DAS28 < 3.1). Therapy had to be adjusted with DAS28 > 3.2 unless patient′s conditions don’t permit it; we considered this follow-up type as implementation of a T2T strategy. We divided patients in two groups:remission-low disease activity (Rem/LDA) patients and moderate-severe diseaseactivity (MDA/ SDA) patients and the aim of the study was to look at what percentage of patients who were MDA/SDA disease activity reached a low disease activity or remission. 158 patients with RA and using Adalimumab, Infliximab and Etanerceptbiosimilar (Etanar? CP Guojian Pharmaceutical Co Ltd, China) were involved. The Etanercept biosimilar was approved for using in Colombiasince 2007. Descriptive epidemiology was done, the medians were analyzed usingt-Student assuming normality for DAS28 distribution and disease activity was analyzed using Pearson′s statistics.Results: 158 patients were included inthis study, 125 (79.1%) women and 33 (20.9%) men. Average age was 59 +/- 10 y/owith disease duration of 11 years (0.5-47). 158 patients with diagnosis of RA using Adalimumab, Etanercept and Infliximab were involved: Adalimumab 61 (38.6%), Etanercept 25 mg 62 (39.2%), and Infliximab35 (22.2%). At 24 months was observed an increase in percentage of patients in remission and a decrease in percentage of patients in MDA/SDA disease activity statistically significant. for Adalimumab at beginning DAS28-3.6 and 24months later 2.6; for Etanercept biosimilar at beginning DAS28-3.6 and 24months later 2.6 and for Infliximab at beginnngDAS28-3.6 and 24 months later 2.6.There were not statistically significant differences between analyzed biologics. On the other hand, there were fewer adverse events with Etanercept-biosimilar than Adalimumab and Infliximab; it was statistically significant.Conclusions: This study shows that the Etanercept biosimilar is as effective as 2 othertraditional anti-TNF biological for disease activity control in patients with rheumatoid arthritis in a real-life setting with fewer adverse events, which could have implications in treatment decisions and health economics. On the other hand the study proves effectiveness of implementation of a T2T strategyin patients with RA.译文背景:生物制剂治疗RA的临床疗效众所周知。
便宜又好用的肿瘤生物仿制药,你了解有多少?
便宜又好用的肿瘤生物仿制药,你了解有多少?生物仿制药不是假药,而是实实在在的真药和好药。
文丨维生素C来源丨医学界肿瘤频道提到生物仿制药,大家可能嗤之以鼻。
在我国,一提“仿制”两个字,人们往往联想到山寨、盗版、非法等字眼。
而国际上的生物仿制药(biosimilar)却是指对原研专利生物药在其专利失去的市场独占权法律保护后,进行的合法仿制。
从成分上来讲,仿制药与品牌药相差无几,是货真价实的真药。
我们先来了解一下概念:原研药是指原创性的新药,在全球最先提出申请,并获得专利保护的药品,一般有20年的保护期,在保护期内,其他企业不得仿制。
要经过对成千上万种化合物进行层层筛选、严格的临床前和临床试验,才得以获准上市。
一般情况下,原研药需要科研人员花费十几年左右的研发时间和数十亿美元的研发经费。
新药刚上市的时候,都伴随着专利保护和品牌,因此新药又叫“专利药”或者“品牌药”。
关于生物仿制药是指是与已批准的生物原研药相似的一种生物药(包括疫苗、血液及血液成分、体细胞、基因治疗、组织和重组治疗性蛋白等)。
它与原研药具有相同的活性成分,在剂量、剂型、给药途径、安全性和有效性、质量、治疗作用以及适应证上没有显著差异的一种仿制品。
具有降低医疗支出、提高药品可及性、提升医疗服务水平等重要经济和社会效益的作用。
生物仿制药的批准基于它与已经获批的生物制品有高度相似的数据,并且在安全性、纯度和效力方面没有临床意义上的差异。
所以,单从药效上来说,仿制药肯定不是假药,而是实实在在的真药和好药。
美国食品药品监督管理局(FDA)批生物仿制药主要看以下几点是否与原研药相同。
包括:作用机理、给药途径、剂型、剂量规格、生产设备。
为促进我国生物制药产业的健康、有序发展,国家药监局及时组织药品审评中心等技术部门,在借鉴世界卫生组织和国内外相关指导原则及国际生物类似药成功研发案例的基础上,结合我国生物药研发的实际情况和具体国情,在2015年2月制订发布了《生物类似药研发与评价技术指导原则(试行)》。
阿达木生物类似药临床研究设计要点考虑(征求意见稿)
阿达木单抗注射液生物类似药临床试验设计考虑要点(征求意见稿)一、概述阿达木单抗(Adalimumab)系在中国仓鼠卵巢细胞中表达的重组全人源化肿瘤坏死因子α(Tumor Necrosis Factor, TNFα)单克隆抗体注射液,由美国雅培公司研发上市,商品名为:修美乐(Humira®)。
阿达木单抗在美国和欧盟已获批多个适应症[1,2],2010年首次获准进口中国。
目前在中国批准用于:①对改善病情抗风湿药(DMARDs),包括甲氨蝶呤疗效不佳的成年中重度活动性类风湿关节炎患者(RA);②常规治疗效果不佳的成年重度活动性强直性脊柱炎患者(AS);和③需要进行系统治疗或光疗,并且对其它系统治疗(包括环孢素、甲氨蝶呤或光化学疗法)不敏感,或具有禁忌症,或不能耐受的成年中重度慢性斑块状银屑病患者(PsO)[3](见表1)。
阿达木单抗注射液原研产品美国专利已于2016年到期,欧洲专利2018年即将到期[4],国内外制药企业已纷纷加入其生物类似药的研发。
Amgen研发的ABP501(Amgevita®)、BI研发的BI695501(Cyltezo®)和Samsung Bioepis研发的SB5(Imraldi®)均已作为阿达木单抗的生物类似药在欧盟获批上市[5-7],其中的ABP501和BI695501在美国也已按生物类似药获准上市[8,9]。
本文在原国家食品药品监督管理总局已发布的《生物类似药研发与评价技术指导原则(试行)》[10](以下简称“指导原则”)基础上,结合阿达木单抗的特点,重点探讨当前普遍关注的临床研究策略和临床试验设计问题,以期为国内阿达木单抗生物类似药的临床研发提供参考。
二、阿达木单抗生物类似药临床研究策略根据《指导原则》,生物类似药研发总体思路是通过系统的比对试验证明候选药与原研药的相似性为基础,支持其安全性、有效性和质量可控。
依据逐步递进的原则,分阶段进行药学、非临床、临床比对研究。
注射用曲妥珠单抗生物类似药临床研究设计及审评考虑要点(
注射用曲妥珠单抗生物类似药临床研究设计及审评考虑要点(征求意见稿)一、前言曲妥珠单抗(Trastuzumab)是由瑞士罗氏公司研发的一种重组DNA衍生的人源化单克隆抗体,含人IgG1亚型框架,互补决定区源自鼠抗p185 HER2 抗体,能够特异性地作用于人表皮生长因子受体-2(HER2)的细胞外部位第IV亚区,竞争性阻断人体表皮生长因子与HER2的结合,从而抑制肿瘤细胞的生长。
罗氏公司的注射用曲妥珠单抗(Herceptin®,赫赛汀®)最早于1998年9月25日获得美国FDA批准上市,2002年进口中国,目前获批的适应症为:单药用于治疗HER2阳性转移性乳腺癌;联合紫杉醇或者多西他赛用于HER2阳性转移性乳腺癌;HER2阳性的可手术乳腺癌患者的辅助治疗;HER2阳性转移性胃癌。
在欧盟,还获批了早期乳腺癌新辅助治疗的适应症。
赫赛汀®在美国、欧盟及中国获准上市的适应症见表1。
曲妥珠单抗在欧盟的专利已于2014年7月到期,美国专利也将于2019年6月到期,其生物类似药的研发成为热点,目前印度(Hertraz, Mylan)、韩国(Herzuma, Celltrion)和俄罗斯(HERtiCAD, Biocad)各有一个生物类似药上市。
本文在CFDA已发布的《生物类似药研发与评价技术指导原则(试行)》(后简写为《指导原则》)基础上,结合该品种的特点,对曲妥珠单抗生物类似药的临床研究策略和方案设计要点进行探讨,以期为曲妥珠生物类似药的研发相关人员提供参考。
二、曲妥珠单抗生物类似药临床研究策略根据《指导原则》,生物类似药研发总体思路是以比对试验证明其与参照药的相似性为基础,支持其安全、有效和质量可控。
采用逐步递进的顺序,分阶段开展药学、非临床、临床比对试验。
根据前期比对试验结果设计后续比对试验研究的内容。
根据前期药学和药理毒理比对试验结果,曲妥珠单抗生物类似药的临床研发可能会存在以下两种情况:1、药学和药理毒理试验证明候选药与参照药相似,按照生物类似药的路径开展药代动力学比对试验和临床安全有效性比对试验。
托珠单抗注射液生物类似药临床试验指导原则(征求意见稿)
1托珠单抗注射液生物类似药临床试验指导原则2(征求意见稿)34一、前言5托珠单抗注射液(Tocilizumab)由罗氏公司研发,采用哺乳动物6细胞(CHO)表达的抗人白介素6受体单克隆抗体制剂,商品名为:7雅美罗®/Actemra®。
通过阻断白介素6与可溶性及膜结合的白介素6 8受体结合,抑制白介素6的信号转导,从而减少病理性炎症反应。
托9珠单抗自2009年2月起陆续在欧盟、美国、日本等多个国家和地区10获准上市,获批的适应症包括:成人类风湿关节炎(RA),多关节型11幼年特发性关节炎(pJIA)、全身型幼年特发性关节炎(sJIA)、巨细12胞动脉炎(GCA)和细胞因子释放综合征(CRS)等。
目前,托珠单13抗在我国获批的适应症包括RA和sJIA[1]。
14托珠单抗注射液原研产品序列专利已到期[2],国内外众多制药企15业纷纷加入其生物类似药的研发过程中。
为了更好地推动生物类似药16的开发,在原国家食品药品监督管理总局已发布的《生物类似药研发17与评价技术指导原则(试行)》[3]基础上,我们结合该品种的特点及18研发企业相关问题的沟通交流情况,讨论形成了托珠单抗生物类似药19临床试验研究设计要点,以期为业界提供参考。
20二、托珠单抗生物类似药临床研究总体要求21原则上,药代动力学比对试验需要进行1项健康受试者单次给药22药代动力学生物等效性研究,验证候选药与原研药PK特征的相似性。
1临床比对研究建议选择原研进口获批RA适应症人群,与原研药进行21项“头对头”比较的临床等效性研究以支持其按生物类似药注册上3市。
4三、临床研究设计考虑要点5生物类似药临床比对研究设计应当以证明候选药与原研药的相6似性为目的,进行科学合理的研究设计。
7(一)健康受试者药代动力学比对研究8试验设计:参照一般生物等效性研究的设计,结合托珠单抗生物9类似药半衰期较长(稳态浓度下,每四周给药一次,4mg/kg时为11 10天,8 mg/kg时为13天),具有免疫原性等特点,建议采用随机、双11盲、平行对照、单次给药的试验设计。
新型非格司亭生物仿制药 Nivestim(TM) 获准在欧洲用于预防因化疗导致的发热性嗜中性白血球减少症.docx
新型非格司亭生物仿制药Nivestim(TM) 获准在欧洲用于预防因化疗导致的发热性嗜中性白血球减少症- Hospira 的非格司亭Nivestim(TM) 已经获得欧盟委员会(EC) 的批准,用于预防发热性嗜中性白血球减少症(FN) 和化疗后嗜中性白血球减少症(CIN) 引起的存活期的缩短- Nivestim 是一种新型非格司亭,该药集给药便利性、便于储藏性和安全性与一体- 嗜中性白血球减少症是由癌症化疗引起的最严重的血液中毒症,可导致化疗剂量相对常规疗程的减少和/或推迟(1)英格兰LEAMINGTON SPA 2010年6月10日电/美通社亚洲/ --Hospira 今天宣布,欧盟委员会已经批准将Nivestim(TM)(非格司亭)用于预防发热性嗜中性白血球减少症,这种病症是由癌症化疗引起的最严重的血液中毒症(1)。
目前Nivestim 已在欧盟各国获得营销授权。
Nivestim 预计将可降低嗜中性白血球减少症的治疗成本。
德国Freiburg University Medical Center(弗赖堡大学医学中心)内科医学副教授Cornelius Waller 博士表示:“Nivestim 的获批为医疗卫生专业人员和患者带来了切实的好处。
因癌症化疗导致的嗜中性白血球减少症可导致患者无法完成全部的化学疗程。
Nivestim 为医疗卫生专业人员提供了一种具有成本效益且易于使用的选择,帮助患者坚持到底。
”Nivestim 是Hospira 的第二种生物仿制药。
该公司的促红细胞生成素生物仿制药Retacrit(TM) 目前在17个欧洲国家销售。
Hospira 是首个在欧洲销售生物仿制药的美国公司。
该公司的生物仿制药产品线还包括非格司亭的长效版聚乙二醇非格司亭,是该行业最大的产品线之一。
Hospira 首席商务官Ron Squarer 表示:“作为Hospira 扩大生物仿制药产品组合持久承诺的一部分,我们很自豪地宣布,Nivestim 已经获得欧盟委员会的批准。
国家食品药品安全专业技术人员培训考试-美国药事法规
药品注册专员岗位知识(美国药事法规部分)满分:100得分:84.0一、美国药品监管机制单选题(共12题,共24.0分)1. 从一开始,美国的药品法的每一次重大发展几乎都以患者生命为代价,有一个法律例外:A.1938年新版《FFDCA》。
B.1962年《Kefauver Harris Amendment》。
C.1984年《Hatch Waxman Act》。
D.2013年《Compounding Quality Act》。
2. FDA的公开执法信息包括A.现场核查,产品召回,不批准上市。
B.警告信,业内除名,罚款入狱。
C.药品安全警告,违法广告警告信,定期执法报告。
D.短缺药品名录,警告信,消费者预警报告。
3. 美国药品法发展的里程碑节点对药品上市提出的最低要求是A.1906年以后要求上市药品必须符合质量可控、安全、有效的标准。
B.1938年以后要求上市药品必须符合质量可控、安全、有效的标准。
C.1962年以后要求上市药品必须符合质量可控、安全、有效的标准。
D.2004年以后要求上市药品必须符合质量可控、安全、有效的标准。
4. 药品作为特殊民用消费品的原因是A.药品与每个人的生老病死息息相关。
B.药品上市必须符合质量标准。
C.药品是非天然产品,必须被批准才能上市。
D.符合安全、有效、质量可靠标准的药品就能被批准上市。
5. 505(b)2指的是A.仿制药,以ANDA申报。
B.原创药,以NDA申报。
C.原创药的简约版,以NDA申报,但是要和仿制药一起排队。
D.既不是仿制药也不是原创药,以505(b)2申报。
6. 特殊试验设计方案(SPA)是A.在任何原创新药开发的任何时期都可以申请。
B.在任何新药开发的特殊重要阶段都可以申请,需要等FDA的批准。
C.需要等FDA的批准才能实施,太麻烦花时间,所以最好不要申请。
D.如果基于人道主义的原则,需要实施不以人为受试者的三期有效性试验,应当申请。
7. 21 CFR Part 11是对电子记录和电子签字的法规,规定了A.电子数据无论出处必须可靠。
生物仿制药,相似但不相同(插件)
权威机构对生物仿制药的态度
“我们不能确定生物仿制药与原研药是可互换的, 这需要在全国范围 内进行讨论” – EMEA Executive Director Thomas Lönngren 2006
由于生物仿制药与标准生物制剂在医学上相似但又不完全相同,对患 者是否使用生物仿制药或标准生物制剂应该听取权威医学药学专业人 士的建议。 – London, 19 April 2007 Doc. Ref. EMEA/74562/2006 法国、西班牙等多个欧洲国家不允许生物仿制药代替生物制剂 生物仿制药不同于传统低分子仿制药物,目前用于批准仿制药的规则 ( “本质相似”(EU)和“生物等效性”(USA)法规)不适用于 生物仿制药 – EMEA、FDA
生物仿制药,相似但不相同
生物制剂不同于化学药物:不能准确复制
传统药物:容易精确复制 拥有精确的化学分子 式和分子结构 生物制剂:不能精确复制 拥蛋白质独特的多维结构→复杂 的作用方式目前无法准确复制 作为生物制剂,即使分子式相同, 且通过相同的细胞或微生物生产, 也可能导致不同的疗效和安全性
J Endocrinol Invest. 2008, 31: 479-488.
生物仿制品与原研生物药存在多方面差别
以欧洲重组人促红细胞生成素的质量规范为标准,对来自巴西、韩国、 印度等13个国家的31种重组人促红细胞生成素的生物仿制品进行同于和原研生物药疗效、安全性相同的药物使用 • 生物仿制药需要对药效和安全性更严格的监控 •一种甘精胰岛素的仿制药由于无法提供免疫原性数据,在哥伦比亚上市申请被退回
1. Singh AK. Gaps in the quality and potential safety of biosimilar epoetins in the developing world: an international survey (Abstract THPO972). Paper presented at the American Society of Nephrology Renal Week 2006; Nov 16, 2006; San Diego.
英国生物类似药医保及临床使用管理措施分析
广东药科大学学报Journal of Guangdong Pharmaceutical UniversityJan.2021,37(1)收稿日期:2020‐11‐02基金项目:国家社会科学基金重大项目(15ZDB167);2019年江苏省研究生实践创新计划(SJCX19_0164)作者简介:陈童,女,在读硕士研究生,主要从事医药政策与法规研究,Email:*******************通信作者:邵蓉,女,教授,博士生导师,主要从事医药政策与法规研究,Email:*******************。
英国生物类似药医保及临床使用管理措施分析陈童,蒋蓉,邵蓉(中国药科大学国家药物政策与医药产业经济研究中心,江苏南京211198)摘要:目的介绍英国生物类似药医保准入、定价管理和使用管理方面的政策措施,为推动我国生物类似药的发展提供借鉴。
方法通过文献研究和检索英国相关部门官方网站,梳理英国生物类似药医保及临床使用管理措施。
结果一般情况下,生物类似药在英国不需要额外进行卫生技术评估,通过协商定价确定其报销价格。
英国已建立生物类似药可互换性的规定,通过药物警戒确保生物类似药的安全使用,并采取相关措施鼓励生物类似药的使用。
结论我国已有8款生物类似药获批上市,但在医保准入、支付管理和临床使用方面仍未建立管理体系,建议我国建立完善的生物类似药医保准入体系,制定科学、合理的医保支付政策并采取措施推动生物类似药的临床使用。
关键词:英国;生物类似药;医保准入;定价;临床使用中图分类号:R951文献标识码:A文章编号:2096‐3653(2021)01‐0110‐05DOI:10.16809/j/cnki.2096‐3653.2020110101Analysis of medical insurance and clinical use management measures of biosimilars in the UKCHEN Tong,JIANG Rong,SHAO Rong *(The Research Center of National Drug Policy &Ecosystem,China Pharmaceutical University,Nanjing211198,China )*Corresponding author Email:*******************Abstract:Objective To introduce the policy measures in the medical insurance access,pricing management and use management of biosimilars in the UK,and provide a reference for promoting the development of biosimilars in China.Methods Through literature research and searching the official websites of relevant departments in the UK,this paper analyzes the medical insurance and clinical usemanagement measures of biosimilars in the UK.Results Generally,biosimilars do not require additional health technology assessment in the UK,and their reimbursement price is determined through negotiation.The UK has established regulations on the interchangeability of biosimilars,ensured the safety of biosimilars through pharmacovigilance,and has taken relevant measures to encourage the use of biosimilars.Conclusion China has approved 8biosimilars for marketing,but there is still no management system in terms of medical insurance access,payment and clinical use.It is recommendedthat China establish a complete biosimilar medical insurance access system,formulate scientific and reasonable medical insurance payment policies and take measures to promote the clinical use of biosimilars.Key words:UK;biosimilars;medical insurance access ;pricing;clinical use第1期陈童,等.英国生物类似药医保及临床使用管理措施分析世界卫生组织(World Health Organization,WHO)将生物类似药定义为与一种已批准的参比生物治疗产品在质量、安全性和效力方面均相似的生物治疗产品[1]。
神州细胞专题研究-十数年磨一剑中国首个重组八因子重磅上市
神州细胞专题研究-十数年磨一剑中国首个重组八因子重磅上市神州细胞专题研究-十数年磨一剑中国首个重组八因子重磅上市一、全球生物工程探路者,在刚需、重磅、工艺高壁垒药中厚积薄发19年技术创新与积累,国际一流水平的全产业链生物工程平台现有产品与主要管线:药效确定而合成壁垒高的生物大分子,重组蛋白。
神州细胞,2007年成立于北京,研发与生产生物药,主要是:有较高生产与工艺壁垒的单抗、重组蛋白与创新疫苗。
公司目前已有9个生物药在临床与上市申请阶段。
主要的治疗领域,涉及肿瘤、自身免疫、眼科及罕见病。
生物大分子药物,活细胞生产的复杂生物制剂,生产技术造就天壤之别不论是全球唯一进入人体临床的十四价HPV蛋白亚单位疫苗,还是重组蛋白药物重组人凝血八因子,亦或是利妥昔单抗或阿达木单抗这类全球重磅的生物类似物,神州细胞只研发生产一种产品:高壁垒重组蛋白,属于生物大分子。
生物大分子药物,原研的称为生物药(Biologics),仿制的称为生物类似物(Biosimilar),而不是小分子药物的仿制药(Generic)。
一个生物类似物,是基于表明它是高度相似于FDA批准的生物制品(被称为批准的基准产品)并且没有临床上从基准产品的安全性和有效性方面的有意义的差异。
生物类似物中,允许存在临床非活性成分的微小差异。
生物类似物Biosimilar与小分子仿制药Generic最大的差别在于,前者是通过培养活细胞来生产的,过程复杂,地球上没有两个生物类似物是一模一样的,而仿制药是确定一样的分子结构。
不同企业的生物类似物之间的生产成本、产能、生物活性可能相去甚远。
相对于小分子药物,生物类似物的开发,需要大量时间和资金的投入,并且Quality-in-Design(质量源于设计),大分子药物的质量从生产的专业基础设施的设计就开始了。
这也是为什么,看似国内生物类似物研发药企总是慢于资本市场期待的快节奏,因为这是一个需要经历一砖一瓦、每个环节不断试错与优化之后才能继续前行推进的相对漫长的探索过程。
生物类似药的研发与临床试验进展
生物类似药的研发与临床试验进展研究方案:生物类似药的研发与临床试验进展一、引言生物类似药(Biosimilar)是指与已上市的原研药(Reference Drug)在结构、质量、功效、安全性等方面高度相似的药物。
随着原研药的专利保护期限逐渐到期,生物类似药的研发与临床试验越来越受到重视。
本研究旨在对生物类似药的研发与临床试验进展进行探索,并提出有关改进现有研究方法和数据分析的新观点和方法。
二、研究方法1. 文献研究方法通过对国内外相关研究文献的检索和梳理,系统性地了解生物类似药的研发与临床试验的最新进展和现有问题。
尤其需要关注国内外监管机构对生物类似药研发与临床试验的最新、标准和要求。
2. 实验室方法选择一种针对特定疾病的生物类似药作为研究对象,比如生物类似肿瘤治疗药物。
使用现有的生物学、生化学和生物技术实验方法对该生物类似药的质量、结构及其作用机制进行研究。
包括:(1)对生物类似药的质量特性进行分析,如高效液相色谱法(HPLC)、氨基酸序列分析、质谱等。
(2)通过体外实验,在细胞水平上研究生物类似药的生物活性、靶向性、毒性和代谢特性。
(3)在动物模型上进行体内实验,评估生物类似药的治疗效果、安全性和体内动力学特性。
3. 临床试验方法基于前期实验室研究的结果,将生物类似药的临床试验分为3个阶段:(1)I期临床试验:主要评估生物类似药的安全性和耐受性,使用小样本量的健康志愿者进行临床试验。
(2)II期临床试验:进一步确定生物类似药的剂量、给药方案和疗效,试验对象为特定疾病的患者,进行较大规模的临床试验。
(3)III期临床试验:进行大规模临床试验,比较生物类似药和原研药的疗效和安全性,为上市申请提供足够的证据。
三、实验设计1. 实验室实验设计(1)选择一种生物类似药作为研究对象,设计并优化相应的实验方案。
(2)根据生物类似药的特性和目标疾病的特点,确定实验所需的细胞系、动物模型和评估指标。
(3)设置试验组和对照组,对生物类似药的药效和安全性进行评估。
关于生物仿制药Biosimilarproducts药学研究问题的思考.doc
关于生物仿制药(Biosimilar products)药学研究问题的思考与小分子的化学药品不同,生物制品的分子量较大且结构复杂,产品质量尤其是生物学活性易受各种因素影响且不太稳定,再加之目前可行的分析方法有限,因此难以通过有限的比较研究来完全确认不同企业产品之间的一致性。
基于生物制品的上述特性,目前国内外药品监管部门均认为,对传统的仿制药品(generic drug)的评价理念和标准并不适用于仿制性的生物制品,不同企业生产的同名的生物制品只能达到相似而非一致,因此对于仿制性的生物制品一般不称为generic drug,而称作Biosimilar products或Follow-on Biologics,而且在进行此类生物制品的研发时需进行非常广泛的比较性研究,包括质量、非临床以及临床试验等,以充分确认其安全性和有效性。
鉴于目前国内对于此类生物制品尚无法定而统一的称谓,因此为简化起见,本文暂称之为生物仿制药。
以下笔者将就此类生物制品药学研究的意义以及国内外相关技术要求等进行简要回顾,并在此基础上提出一些对于生物仿制药药学研究问题的个人观点和看法。
一、关于生物仿制药药学研究的总体考虑药学研究(包括生产和质量研究等)是药物研发的基础性工作。
对于生物仿制药来讲,药学研究不但是为了达到产品质量可控的目的,而且通过对生物仿制药与已上市药物质量的比较,可以对仿制品的安全有效性进行初步的判断,并据此确定后续研发阶段的研究内容,可以说药学研究结果在相当大的程度上决定了对于后续非临床和临床试验研究的技术要求。
因此,任何企业如果进行生物仿制药的研发并期望减少耗资巨大的非临床和临床试验,则应高度重视药学研究工作,并以之作为整体研发工作的重要基础。
二、国内外生物仿制药药学申报资料要求简介就药学方面的技术资料要求来讲,生物仿制药的研究内容与其他生物产品包括创新性产品并无很大不同,只是更加强调与已上市产品的比较性研究。
在这一点上国内外的相关法规和技术指南是基本一致的。
RDPAC 生物类似物建议书 biosimilars position paper-CN
RDPAC关于生物类似物的建议书目录术语及名词解释 (3)序言 (6)1.背景 (7)1.1生物制品的独特性 (7)1.2国外生物类似物的法规现状 (8)1.3中国生物制品的法规现状 (9)1.4建立明确的生物类似物审批途径的必要性和重要性 (10)2.RDPAC的建议 (12)2.1注册分类 (14)2.2加强药物警戒系统 (14)2.3数据保护、专利链接和监测期 (15)2.4总体的注册要求 (16)2.4.1治疗性生物参比制剂(RBP)的选择 (16)2.4.2生产工艺 (17)2.4.3可比性与生物相似性 (17)2.4.4全面(Comprehensive)的质量比较 (18)2.4.5非临床研究考虑 (20)2.4.6临床相似性的分步(Step-Wise)评价方法 (21)2.4.7标签说明书 (24)2.4.8替换 (25)3.结论 (26)缩写RBP:治疗性生物参比制剂SBP:治疗性生物类似物CMC:化学、生产与控制PK:药代动力学PD:药效学SEI:战略新兴产业CFDA:国家食品药品监督管理总局术语及名词解释以下为本建议书中使用的术语。
这些术语在其他文件中可能有不同的含义。
生物技术生物技术是为了获得所需产品或提供服务而对生物体及其组分进行改造的方法和技术。
1970年代出现的所谓“重组DNA技术”将“分子”生物技术带入到一个新阶段,极大促进了不同领域知识的革新。
重组DNA技术是利用生物系统DNA改造制备治疗性蛋白药物的快捷方法。
现今生物技术药大多采用重组DNA技术生产。
这意味着可通过对生物体进行遗传改造来生产所需蛋白质。
1生物药/生物制品生物药或生物制品是通过生物技术(重组DNA技术)或使用生物体(即生物合成,相对于化学合成)制得的药物。
生物类似物生物类似物(Biosimiliar)是一种含有参比制剂活性物质的生物制品,这种参比制剂是基于完整申报资料获准上市且专利和其他专有权已到期的生物制品。
国内外生物类似药研究指导原则要点之比较
国内外生物类似药研究指导原则要点之比较随着药物研发技术的不断进步,国内外的医药界均开始从传统的仿制药转向生物类似药(Biosimilars)的研发,以提高药物研发的效率。
生物类似药是指临床上不同的原料和生产工艺,但具有相同的来源,以及近似的结构、功能和性能的药物。
这种创新的药物正在引起全球医药界的广泛关注。
随着生物类似药在全球市场的快速增长,不同国家均对该新型药物在研发、注册、分销、使用和监管等方面制定了严格的指导原则。
特别是在研发阶段,为了确保创新药物的安全性、有效性和质量,各国的法律法规和指导原则均有所不同。
为了加深我们对国内外指导原则的认识,本文将比较国内外指导原则,以更好地实施Biosimilars 研发和使用。
首先,有关Biosimilars研发指导原则的详细介绍。
在国外,欧洲药品管理局(EMA)出台了第四代药品技术质量指导原则,指出,Biosimilar药品的研发、生产、测试、标准定义和证明原理均需要与传统仿制药不同,因为原料和原理不同。
以及,采用比较和分析的方法来证明两者的差异,以确定Biosimilar药物的质量、安全性和有效性。
此外,美国食品药品管理局(FDA)采取了更为灵活的原则,以致于比较证明原理轻易实现。
FDA鼓励企业根据药品特性和协商协定,利用其他方法来比较原料和原理的差异,以证明Biosimilars的安全性和有效性。
在国内,国家药品监督管理局(NMPA)发布了《生物类似药原料药研发规范(2010年版)》,《国家药典委员会申报条件与批准程序的指导原则》,《生物类似药研发技术文件修订指导原则》等,在原料、技术、测试等方面,对生物类似药研发给出了要求。
更重要的是,为了确保Biosimilars的安全性和有效性,NMPA要求药企提供与原料药对比的实验数据,以证明新药的安全性和有效性。
综上所述,由于不同国家生物类似药研发指导原则差异很大,因此,有必要进一步加深对国内外指导原则的认识,以确保所开发的生物类似药符合国外的立法和国内的标准。
2022年浙江省执业药师继续教育试题+答案生物类似药:现状与思(生物类似药:现状与思考课程答题测试)
2022年浙江省执业药师继续教育试题+答案生物类似药:现状与思考课程答题测试1、生物类似药的英文名为 ( )A.BiosimilarB.Biological analoguesC.BiogenericD.Follow-on biologics2、生物类似药是与已批准的生物原研药相似的一种生物药,但不包括( )A.疫苗B.血液及血液成分C.组织和治疗性小分子化合物等D.体细胞E.基因治疗3、国家食品药品监督管理局药品审评中心(CDE)在其发布的《生物类似药研发与评价技术指导原则(试行)》文件中首次将biosimilar 称为( )A.生物仿制药B.生物相似药C.生物近似药D.生物类似药4、国家食品药品监督管理局药品审评中心(CDE)发布的《生物类似药研发与评价技术指导原则(试行)》的发布时间是( )A.2015年B.2016年C.2017年D.2018年5、CDE发布的《生物类似药研发与评价技术指导原则》中提到,生物类似药是与已获准上市的参照药具有相似性的治疗性生物制品,相似性不包括。
( )A.经济性B.质量C.安全性D.有效性6、生物类似药候选药物的()原则上应与原研药(参照药)相同A.一级结构B.二级结构C.三级结构D.四级结构7.在原研药()到期之后,生物类似药方可获得审批。
A.专利申请B.专利授权C.专利批准D.专利保护8.( )率先提出了“生物类似药”的概念及相关技术要求A.FDAB.NMPAC.EMAD.日本9.单抗生物类似药的研发采用( )的策略,分阶段开展药学、药理毒理和临床比对试验,以证明单抗候选药在质量、安全性和有效性方面与原研药不存在临床意义上的差异。
A.同步进行B.逐步递进C.序贯进行D.阶段完成10、《生物类似药研发与评价技术指导原则》明确适用范围为( )A.体细胞产品B.重组蛋白质制品C.基因治疗D.聚乙二醇等修饰的产品及抗体偶联药物类产品11、《生物类似药研发与评价技术指导原则》中参照药的定义如下:在生物类似药研发过程中与之进行比对试验研究用的产品,包括生产用的或由成品中提取的活性成分,通常为()A.市场销量最好的产品B.原研产品C.其他生物类似药D.标准品12、研发和评价的基本原则()A.比对原则B.逐步递进原则C.一致性原则D.以上都是13、对于药学研究和评价,以下描述不正确的是()A.选择有代表性的批次进行比对试验,对样品质量的批间差异进行分析,检测候选药与参照药之间可能存在的差异B.评估每一个质量特性与临床效果的相关性,并设立判定相似性的限度范围C.对特性分析的比对试验研究结果综合评判时,应根据各质量特性与临床效果相关的程度确定评判相似性的权重,并设定标准 D.对理化特性、生物学活性、纯度和杂质、免疫学特性进行比对研究,对抗体类的产品,无需对其Fab、Fc段的功能进行比对试验研究。
生物类似药
谢谢观看
总的来说,生物药的生产对于其生产条件的要求远比化学药苛刻,当然生产成本也更高,而且生物药的临床前 和临床阶段的研发成本也更高。
各国法规比较
各国法规比较
新出台的指导原则代表了我国在推动生物类似药研发上的显著进展。在中国,患者有望能够获得更多的经济 实惠的生物制药。在生物类似药法规建立的初期阶段,这部指导原则给生物制药公司提供了一个很好的起点,帮 助它们取舍中国的生物类似药审批流程或国外审批流程。
对于生物类似药生产商而言,由于知识产权保护等多种原因,原研药公司所采用的生产工艺甚至是所采用的细 胞系都会不清楚,这就更导致生物类似药与原研药不会一样。另外,对于生物药而言,其生产及流通过程更加复杂, 要求也更高,有许多步骤,细胞培养的条件(温度和营养)、产品的加工、纯化、储存和包装等各个环节都会影响产 品的生产,整个过程中的微小差别都可能会对最终产品的质量、纯度、生物特性以及临床效果产生较大影响。正由 于上述种种原因,虽然化学仿制药的英文是generic drug,但是生物类似药并非是biogeneric,而是biosimilar, 因为生物类似药只可能与原研药“相似”(similar),绝不可能一样。
与美国和欧盟的监管当局相似,我国CDE也希望制药公司提交生物类似药的详细结构和功能特性,以便与参 照药进行比较。另外,CDE还采取了与发达国家相似的渐进式方法,通过比较药理学数据、非临床研究和临床研 究以评价相似性。
尽管该指导原则与美国和欧盟的相关法规在技术审查原则上有不少相似,但它们之间还是存在着显着的差异。
欧盟EMA:“A biological medicine that is developed to be similar to an existing biological medicine(the‘reference medicine’).When approved,a biosimilar’s variability and any differences between it and its reference medicine will have been shown not to affect safety or effectiveness.”(试译:与已经存在的生物药(即:参比药)类似的生物药。在批准时,该生物类 似药自身的可变性以及与参比药的任何不同之处均应被证明不影响仿制药的安全性和有效性。
我院贝伐珠单抗生物类似药与原研药相关不良反应回顾性分析
我院贝伐珠单抗生物类似药与原研药相关不良反应回顾性分析Δ丁年羊1*,李莉2,方攀奇1,徐思露1,赵敏1,燕丹1 #(1.江苏省肿瘤医院/南京医科大学附属肿瘤医院/江苏省肿瘤防治研究所药学部,南京 210009;2.南京市食品药品监督检验院,南京 211198)中图分类号 R969.3文献标志码 A 文章编号 1001-0408(2024)04-0472-04DOI 10.6039/j.issn.1001-0408.2024.04.17摘要目的分析某院贝伐珠单抗生物类似药与原研药相关药物不良反应(ADR)的发生情况,为临床合理用药提供数据支持。
方法对江苏省肿瘤医院2022年1-12月上报的贝伐珠单抗生物类似药和原研药相关ADR报告进行回顾性分析。
结果我院使用贝伐珠单抗的患者共6818人次,上报ADR报告136份,贝伐珠单抗生物类似药的ADR发生率显著高于原研药(2.18% vs.0.71%,P=0.004)。
ADR报告中,治疗方案以贝伐珠单抗与其他肿瘤治疗药物的联合治疗方案为主(129人次);痊愈和好转的患者有118人次;一般ADR报告108份,严重ADR报告28份;ADR主要累及系统/器官以心血管系统为主,贝伐珠单抗生物类似药与原研药引起的高血压/血压升高、白细胞/血小板降低、腹泻和发热发生率比较,差异均无统计学意义。
结论贝伐珠单抗生物类似药的相关ADR发生率明显高于原研药,但ADR临床表现无明显差异,临床医生可以根据患者及其家属意愿选择使用。
关键词贝伐珠单抗;生物类似药;原研药;药物不良反应;合理使用Retrospective analysis of adverse drug reactions of bevacizumab biosimilar and original drug in our hospitalDING Nianyang1,LI Li2,FANG Panqi1,XU Silu1,ZHAO Min1,YAN Dan1(1. Dept. of Pharmacy,Jiangsu Cancer Hospital/The Affiliated Cancer Hospital of Nanjing Medical University/Jiangsu Institute of Cancer Research,Nanjing 210009, China;2. Nanjing Institute for Food and Drug Control, Nanjing 211198, China)ABSTRACT OBJECTIVE To analyze the occurrence of adverse drug reactions (ADR)between bevacizumab biosimilars and original drugs,and to provide data support for rational use of drugs in clinical.METHODS ADR reports of bevacizumab biosimilars and original drugs reported by Jiangsu Cancer Hospital from January to December 2022were retrospectively analyzed. RESULTS A total of 6818patients were treated with bevacizumab,and 136 ADR patients were reported. The incidence of ADR caused by bevacizumab biosimilars was higher than original drugs (2.18%vs. 0.71%,P=0.004). In ADR reports,the main treatment plan was bevacizumab combined with other tumor drugs (129patients);118patients were cured and improved;there were 108 general reports and 28 serious reports; the main system/organ involved in ADR was the cardiovascular system; there were no statistical significance in the incidence rates of hypertension/blood pressure increase,leukocyte/platelet decrease,diarrhea and fever caused by bevacizumab biosimilars and original drugs.CONCLUSIONS The incidence of ADR related to bevacizumab biosimilars is significantly higher than that of the original drugs,but there is no significant difference in the clinical manifestation of ADR. Clinicians can use bevacizumab biosimilars or original drugs based on the willingness of patients and their families. KEYWORDS bevacizumab; biosimilars; original drugs; adverse drug reactions; reasonable use贝伐珠单抗是由中国仓鼠卵巢细胞表达的特异性靶向游离血管内皮生长因子(vascular endothelial growth factor,VEGF)的重组人源化免疫球蛋白G1单克隆抗体,其能通过阻断游离VEGF与其受体结合,抑制肿瘤组织血管生成而发挥抗肿瘤作用。
美国和欧洲生物类似药的发展_英文_
美国和欧洲生物类似药的发展Richard Markus1,Victor Fung1,SundarRamanan2,Diana Landa1,Jennifer Liu1,Primal Kaur1(1.Biosimilars Development,Amgen Inc.,Thousand Oaks,CA91320,USA;2.Global BiosimilarsRegulatory Policy,Amgen Inc.,Thousand Oaks,CA91320,USA)摘要:生物类似药(biosimilars)是与已批准上市的生物制品高度相似的药物,与小分子仿制药物(generics)不同,生物类似药并不是他们参比制品的精确复制品。
尽管高度相似,生物类似药在某些方面仍然可能与参比制品不同,生物类似药间也会互不相同。
生物制品不仅由于其复杂的性质和生产过程,还由于存在独特的免疫原性和活性的安全隐患,给研发和监管带来了相当大的挑战。
欧洲药品管理局和美国食品药品管理局的指导原则建议采用“证据链完备性”(totality-of-evidence)的方法全面覆盖生物类似药物开发的各个步骤,包括分析表征化、结构相似性和功能等效性等的证据。
这种“证据链完备性”是生物类似药整个研发过程中其余工作的基石,包括必须的动物试验研究、人体药代动力学/药效学研究,以及至少1个临床研究,以证实生物类似药功效等效,并且免疫原性或安全性风险没有增加。
临床研究应选择敏感人群来进行试验,以便发现任何有临床意义的差异。
工艺稳定质量恒定的生物类似药的研发需要丰富的经验和专业技能,只有这样才能够确保患者得到良好治疗。
关键词:生物类似药的研发;监管指导原则;欧洲药品管理局指导原则;美国食品药品管理局指导原则;证据链完备性;生物相似性中图分类号:R915.2文献标识码:A文章编号:0254-1793(2015)05-0777-11doi:10.16155/j.0254-1793.2015.05.03Development of biosimilars in the United States and the European Union Richard Markus1,Victor Fung1,SundarRamanan2,Diana Landa1,Jennifer Liu1,Primal Kaur1(1.Biosimilars Development,Amgen Inc.,Thousand Oaks,CA91320,USA;2.Global BiosimilarsRegulatory Policy,Amgen Inc.,Thousand Oaks,CA91320,USA)Abstract:Biosimilars are highly similar versions of approved branded biologics;unlike generics,they are not exact replicas of their reference molecules.Despite being highly similar,biosimilars are expected to be potentially differ-ent in some aspects from the reference,suggesting that biosimilar products will also differ from each other.The de-velopment and regulation of recombinant biologics presents considerable challenges due not only to their complex nature and production process but also to specific safety concerns linked to immunogenicity and activity.Both the European Medicines Agency and United States Food and Drug Administration guidelines recommend a totality-of -evidence approach focused on stepwise development of biosimilars that involves analytical characterization and demonstration of structural similarity and functional equivalence.This forms the cornerstone of the rest of the de-velopment program,including the need for animal studies,human pharmacokinetics/pharmacodynamics studies,and finally at least one clinical study to confirm equivalent efficacy and the absence of increased immunogenicity or safety risk.The clinical study(ies)should be performed in sensitive populations to allow detection of any clini-cally meaningful differences.Considerable experience and expertise is required for the development of a robust bi-osimilar that can be reproduced with predefined and established quality characteristics to ensure that patients re-ceive high-quality therapies.Keywords:biosimilars development;regulatory guidance;EMA guidelines;FDA guidelines;totality of evidence;bio-the first author E-mail:rmarkus@amgen.comsimilarityCLC number:R915.2Document code:A Article ID:0254-1793(2015)05-0777-11 doi:10.16155/j.0254-1793.2015.05.031IntroductionBiologics,including genetically engineered recom-binant proteins and monoclonal antibodies,are biologi-cal medicines derived from genetically modified living organisms and represent a large proportion of newly ap-proved therapies for several conditions including chron-ic inflammatory diseases and cancer.These medicines have been a pivotal innovation by the pharmaceutical industry,successfully addressing previously unmet ther-apeutic needs.Since their introduction,biologics have become increasingly significant not only in terms of new product development and clinical use but also with re-spect to healthcare expenditures[1].With the expiration of patents on several biologics and more patent expirations on the way,there has been considerable focus on the development of highly similar versions of approved branded biologics;these are re-ferred to as biosimilars.The European Medicines Agen-cy(EMA)[2]defines a biosimilar as a biological medi-cine that is similar to another biological medicine that has already been authorized for use in terms of quality characteristics,biological activity,safety,and efficacy,based on a comprehensive comparability exercise.Ac-cording to the United States Food and Drug Administra-tion(US FDA)definition[3],a product is a biosimilar if data from analytical,animal,and clinical studies show the product to be“highly similar”to the reference bio-logic product,notwithstanding minor differences in clin-ically inactive components,and if there are no clinically meaningful differences in terms of safety,purity,and potency.Cost pressures facing both public and third-party payers,along with the desire to increase healthcare ac-cess,have created a demand for biosimilars,which are expected to be cheaper therapeutic alternatives to bran-ded biologics[1].To improve access,the US congress passed the Biologics Price Competition and Innovation (BPCI)Act of2009,authorizing the US FDA to oversee an abbreviated and expedited pathway[351(k)path-way]for the approval of biologics that are highly similar to already approved products[4].The European Union (EU)has been ahead in developing guidelines for the development and approval of biosimilars,with the first biosimilar approval in2006for human growth hor-mone.2Biosimilars are different from generic products Biosimilars are not analogous to generics as they are not“copies”of originator biological agents.Ac-cording to the EMA[5],a generic drug is“a medicine that contains the same active pharmaceutical ingredient as the reference medicine,”and is“used at the same doses to treat the same diseases.”The US FDA[6]de-fines a generic drug as the“same”as the branded drug in dosage form,strength,route of administration,quali-ty,performance,and intended use.Biosimilars,on the other hand,as previously stated,are“similar”or “highly similar”but not the“same”as or“identical”to the branded products.This is interpreted as biosimi-lars having the same amino acid sequence as the origi-nator product,and high similarity with regard to addi-tional features such as glycosylation and post-transla-tional modifications.The complexity of biologics necessitates a dis-tinction of biosimilars from generic drugs.Small-molecule generic products contain an active pharma-ceutical ingredient that is the same as that of their reference drug.Further,small-molecule drugs have well-defined chemical structures and are generally manufactured through chemical synthesis,whereas biologics such as recombinant proteins are large,complex heterogeneous molecules.In contrast to small-molecule drugs,the manufacturing processes for biologics involve living systems such as microbial and animal cells.These living systems are sensitive to the manufacturing process.Hence,each biosimilar is expected to differ from the originator as well as from other biosimilars;all are unique but related mol-ecules[7-8].Tab.1shows the differences in regulato-ry data requirements for an originator product,a ge-neric,and a biosimilar.Tab.1Differences in regulatory requirements for innovatorcompounds,generics,and biosimilarsNew chemical entity or originator biologic Generic BiosimilarQuality Full process and product characterization Full process and product characterizationComparison with reference drugFull process and product characterizationComparison with reference biologicPreclinical Full preclinical program N/A Abbreviated program based on complexity and residual uncertainty from qualityClinicalPhase1Bioequivalence onlyPhase1PK equivalencePD equivalence(dose-response)if marker available Phase2N/A N/APhase3in all indications N/A Phase3in one representative indication*Risk management plan**Yes Yes Pharmacovigilance Yes YesN/A=not applicable;PK=pharmacokinetics;PD=pharmacodynamics.*If the mechanism of action is the same.**Requirement for the European Union only.The challenge of producing biosimilars is quite different from reproducing a small molecule to develop a generic drug through synthetic chemistry.The design and development of a successful biosimilar requires in-depth understanding of the structure and function of the reference(innovator/originator)molecule in order to evaluate which analytical differences may translate into clinically meaningful differences.Biologics,on av-erage,are100-1000times larger than small-molecule drugs,folding into elaborate three-dimensional struc-tures that determine their function.Proposed biosimilars are expected to encode the same primary amino acid sequence as the reference product.However,modifica-tions will arise as a result of cellular activities such as low level of replication errors in the DNA encoding the protein sequence and amino acid misincorporation dur-ing translation,N-or C-terminal truncations,and criti-cal post-translational modifications such as glycosyla-tion during cell culture.This makes it impossible to produce an exact replica of the originator.A unique cell line is required for every proposed biosimilar;even when the same recombinant DNA sequence is used,the composition of the final product is influenced by a vari-ety of factors including the cell line,culture conditions,post-translational modifications,purification methods,formulation and storage conditions,and container-clo-sure systems.Manufacturers of originator products may use proprietary growth and purification conditions,and therefore,knowledge of the exact DNA sequence or o-riginator host cell line is not sufficient to produce the same biologic product.Subtle differences between a proposed biosimilar and the reference biologic can af-fect efficacy,safety,as well as immunogenicity[9-11].Many therapeutic proteins are glycosylated.Glyco-sylation is important because it can influence the bio-logical activity of a protein through various mecha-nisms;for example,glycosylation can affect half-life by influencing the active clearance of a protein.Fc glyco-sylation plays a role in effector functions such as anti-body-dependent cellular cytotoxicity(ADCC)for thera-peutic antibodies[10,12-13].For example,rituximab,an anti-CD20monoclonal antibody produced from Chinese hamster ovary(CHO)cells with a relatively high level of glycosylation,has been found to be several-fold less cytotoxic in vitro than another anti-CD20monoclonal antibody synthesized with fewer glycan residues from a rat cell line[14].In addition to differences from cell lines,differences in raw materials,processes and manu-facturing,equipment,and quality control standards will also exist in every biosimilar molecule.As a result,un-like generic drugs,biosimilars are unique molecules that differ not only from the reference molecule but also from each other.Schellekens[15]cautioned that,“even if the biosimilar product has the same gene sequence,vector,host cell line,culture conditions and purification methods as the originator product,it can still substan-tially differ in its biological and clinical properties.”Hence,evaluation of these potential differences and thorough understanding of the correlation between structural differences and biological function,as well as the potential clinical impact of these differences is criti-cal to the development of biosimilars,which are expec-ted to have similar safety and efficacy profiles as their reference products.3Development of biosimilarsDemonstration of biosimilarity is a stepwise exer-cise that includes in vitro analytical testing,nonclinical comparative pharmacology testing,and one or more clinical trials.The Quality by Design(QbD)approach is used in the development of biosimilars,where product quality is defined by the reference(originator)product/ molecule.Prior to being approved,a biosimilar must demonstrate similarity to its reference product in terms of quality characteristics,biological activity,safety,and efficacy.A key component of the biosimilarity exercise,assuming the molecule demonstrates high analytical similarity including equivalent functions,is an acceler-ated clinical trial program in which the pharmacokinet-ics,efficacy,safety,and immunogenicity of the biosimi-lar are compared to that of the reference product.3.1Quality by designBiosimilars must be systematically engineered to match the reference molecule in structure and function.Process optimization toward similarity and precise con-trol during manufacturing to maintain similarity is im-portant for the quality of biosimilars.Development of a biosimilar begins with transfection of a cell line with a DNA vector encoding the product;however,starting with the correct amino acid sequence does not guaran-tee success.Quality by Design(QbD)strategies are es-sential for matching the biosimilar molecule with the reference molecule to achieve high similarity of the complex features and to ensure quality.QbD requires a thorough understanding of the product and its manufacturing process[9,16].The prod-uct and process knowledge include an understanding of the effect of process,raw materials,as well as the equipment on product quality attributes.Fortunately,a thorough understanding of the product is made possible through extensive analytical and functional character-ization of the originator product.This information can be used to identify critical quality attributes that ensure a highly similar biosimilar molecule.Sponsors of bio-similar products will consider all relevant characteris-tics of the proposed molecule such as the primary,sec-ondary,tertiary,and higher-order structure;post-trans-lational modifications;and biological activities.Risk as-sessment tools enable linking and ranking quality at-tributes to product safety and efficacy;this is augmen-ted by historical and cumulative knowledge about the desired quality attributes for the biosimilar candidate gained from an understanding of the reference product.After the quality attributes are identified,the combina-tion of process conditions(design space)that maximizes the probability of matching the proposed biosimilar to the reference molecule is chosen.For example,to de-velop a biosimilar to a recombinant monoclonal anti-body that has ADCC activity,knowledge of the involved oligosaccharides is used to make an appropriate choice of cell type and host,as well as for selection of the final manufacturing clone and process.This task is compli-cated by the added challenge of having to match multi-ple quality attributes important for safety and efficacy (i.e.,matching all functions).On the basis of this in-formation,media and production format are selected,and cell culture and purification processes are opti-mized to match multiple key attributes.Formulation is then confirmed and modified if required to ensure ap-propriate stability and robustness.Another key advantage of product development by QbD is the ability to link process,raw material,equip-ment,and operations parameters to quality attributes.This knowledge drives decisions on cell hosts,media types,and process formats.The process operating pa-rameters such as pH and temperature can be optimized to match critical quality attributes.With this in-depth understanding,sources of variability can be controlled to ensure consistent high-quality biosimilar products.Processing conditions can influence product quality;for example,to optimize immunoglobulin gamma(IgG)gly-cosylation,the design of experiments(DOE)concept could be utilized to conduct experiments during up-stream and downstream process development to define operating ranges and to identify failure modes.Processsteps and/or raw materials found to have a significant impact on the particular glycan would then undergo fur-ther evaluation and characterization.In the QbD para-digm,processes may be modified to avoid edges of fail-ure.The long-term acceptance and success of biosimi-lar products relies on robust standards for quality,safe-ty,and efficacy.Meeting these standards begins with the intentional science-based design for analytical and functional similarity.Advances in cell culture engineer-ing,state-of-the-art bioprocessing,and high-resolu-tion analytics have contributed to the ability to devel-op,design,and manufacture molecules that are highly similar to the originator biologic products.3.2Analytical similarityThe complexity of biological molecules influences the analytical studies required to demonstrate similari-ty.Proteins can differ in primary sequence,modifica-tions to amino acids,and higher-order structure.A meaningful assessment of analytical similarity requires extensive and robust comparative studies using state-of-the-art analytical techniques.The capability of ana-lytical methods in regards to their resolution and relia-bility,which directly influences the quality of the re-sults demonstrating high similarity of the biosimilar candidate molecule to the originator,also needs to be established.Frequently,orthogonal methods or tech-niques are used to elucidate more definitely any struc-tural and functional differences between the biosimilar molecule and the originator.A comprehensive and well-designed analytical similarity assessment demonstrating that comparative results lie within the prespecified equivalence window can significantly reduce the residual uncertainty of bi-osimilarity.Physicochemical and biological properties should be demonstrated to be highly similar between the proposed biosimilar and reference molecule.This assessment typically comprises a series of comparative studies between the proposed biosimilar and the refer-ence molecule,and examines product quality attributes in multiple analytical disciplines.Product variants and process-related impurity profiles are a consequence of the different manufacturing processes used to produce the biosimilar.The identity and quantity of product-related impurities may change over the course of the product shelf-life;hence,these need to be evaluated with consideration of sample age at the time of testing,and any differences should be reassessed for lack of clinically meaningful impact.Fig.1depicts the various physicochemical and biological properties that should be considered for detailed characterization of any bio-logic.Examination of multiple batches of innovator products on the market and the proposed biosimilar product manufactured using representative process is necessary to understand the process variability of the two products side-by-side.Biosimilarity is established against the reference molecule profiles;the FDA and EMA guidelines recommend analytical studies to serve as the foundation for establishing similarity to the reference molecule.High degree of analytical similarity,with special emphasis on matching all bio-logical functions,provides justification for the re-duced regulatory requirements with respect to pre-clinical and clinical studies,which further facilitates the overall abbreviated approval process for biosimi-lars.When following the stepwise approach to biosimi-lar development,the first step is to establish analytical similarity between the proposed biosimilar and the ref-erence molecule.This focuses on the establishment of an equivalence window using reference molecule char-acterization parameters,followed by a comparative as-sessment between the biosimilar candidate and the ref-erence molecule.The next step is the assessment of bi-ological similarity,e.g.,effector function,which in-volves demonstrating the functional irrelevance of any analytical differences between the proposed biosimilar and the reference molecule.After analytical and func-tional similarity has been established,pharmacokinet-ic/pharmacodynamic equivalence is determined,with the aim of showing same exposure with the same dose of the proposed biosimilar as compared with the refer-ence molecule.These results,as a whole,are then used in planning the clinical study to confirm that there are no clinically meaningful differences between the proposed biosimilar and the reference molecule and to establish that there is no increased risk of im-munogenicity.Reproduced with permission from ICH.org Fig.1Physicochemical and biological characteristics of biosimilarsAnalytical similarity assessment is a repetitive and iterative process conducted throughout biosimilar prod-uct development ,with the goal to increase knowledge and confidence of the analytical similarity of the bio-similar molecule with the reference molecule.The ana-lytical assessment includes nonclinical characterization such as physicochemical properties ,impurity profile ,and stability as well as preclinical characterization suchas functional properties and animal studies [2-3,7,9].3.2.1Physicochemical characterizationPhysicochemical properties :These include ,among others ,primary ,secondary ,tertiary ,and higher -order structure ;disulfide pairing ;and post-translational modifi-cations.Analytical techniques for primary structure in-clude mass spectroscopy ,peptide mapping ,glycan profi-ling ,isoelectrical focusing ,and amino acid analysis.For higher -order structure ,the commonly used analytical techniques are Fourier transform infrared spectroscopy ,circular dichroism ,and differential scanning calorimetry.Impurity profile :Both product-and process-relat-ed impurities need to be identified and compared with the reference product.Product -related impurities in-clude the presence of undesirable species due to insta-bility ,mistranslation ,and /or mutation-related biochem-ical or chemical interactions or alterations of tertiary and quaternary structure or post-translational modifica-tions.Analytical techniques routinely used to assess product-related impurities include size-exclusion chro-matography ,ion-exchange chromatography ,electropho-resis ,and particle size determination.Process-relatedimpurities may arise from host-cell interactions or rea-gents used during production of the biologic agent.Re-sidual host-cell protein and DNA should be monitored to ensure that there is no impact on product safety and immunogenicity.Stability :Stability of the final product is an impor-tant part of characterization ;a proposed biosimilar should have comparable chemical and physical stability with the reference molecule /finished product.Per the draft FDA guidelines [3],stability studies should be performed under accelerated and stressed conditions such as temperature ,light ,and humidity to identify degradation pathways and provide parameters related to process improvement and storage conditions.3.2.2Functional characterizationFunctional properties :These include determination of the biological activity of the proposed biosimilar and known mechanism (s )of action of the reference mole-cule.If the molecule contains multiple functional do-mains (e.g.,monoclonal antibody ),the binding affinity and specificity at individual domains and the combined biological functions should be compared with those of the reference molecule.The biological functions should include ligand or receptor binding (e.g.,Fc γRIII ,FcRn ),cell -based assays (e.g.,antibody dependent cell-mediated cytotoxicity [ADCC ],complement -de-pendent cytotoxicity [CDC ],potency ),and other tests to demonstrate equivalent in vitro biological activity.3.3Preclinical considerationsThese may include in vivo pharmacology (pharma-cokinetics/pharmacodynamics),toxicology,and/or im-mune response assays.Animal studies may not always be feasible or warranted due to limitations of animal models.Animal studies can serve different purposes,de-pending on the molecule in development and the avail-ability of relevant species.Toxicology studies can be conducted if a relevant species is available;these can remove some uncertainty before testing the proposed bi-osimilar in humans.However,based on the confidence in analytical and in vitro pharmacological similarity,use of animals could be minimized.Therefore,informed and stepwise development is important for reduction in the use of animals as well as reduction in the risk for hu-man subjects.For example,an important step in evi-dence generation may be a repeat-dose toxicology stud-y without the need for recovery animals,and testing a high dose level previously tested by the innovator to de-termine similar known toxicities and any potential un-expected toxicity.Ideally,this can be done in a single gender to minimize the use of animals if there are no questions of gender-specific toxicity[17].In vivo animal models can also be informative in understanding dose-response efficacy;as such studies can often not be conducted in humans for ethical issues associated with treating patients at subtherapeutic do-ses.The dose-response relationship is informative for assessing the equivalent activity of the molecule,partic-ularly when doses on the steep part of the dose-re-sponse curve can be tested.In particular,this may be important for the development of oncology products for which there are no pharmacodynamic markers,and may provide confidence in equivalent antitumor activity,thereby increasing the totality of evidence and reducing the residual uncertainty of differences in the clinical benefit of survival.In such situations,animal models with human tumors can be used to evaluate multiple dose levels of the proposed biosimilar in direct compar-ison with the reference molecule to demonstrate equiva-lent antitumor activity prior to testing in humans.3.4Clinical considerationsThe goal of the clinical development program for a biosimilar is to demonstrate the absence of any clinical-ly meaningful difference relative to the reference mole-cule.The extent of the clinical program depends on the degree of similarity demonstrated in preclinical testing,including structural,functional,and animal studies.Ac-cording to the US FDA draft guidelines[6],clinical effi-cacy studies may not be necessary if there are no resid-ual uncertainties of the biosimilarity of the proposed bi-osimilar molecule and the reference molecule based on structural and functional characterization,animal tes-ting,and human pharmacokinetic/pharmacodynamic data.However,human safety and immunogenicity stud-ies would still need to supplement the overall evidence since these cannot be predicted outside of a clinical study in an informative population with appropriate du-ration of exposure and follow-up and sensitive assays.Moreover,for many monoclonal antibodies,clinical tri-als would likely be obligatory because pharmacodyna-mic efficacy markers do not exist for these therapies.Comparative clinical efficacy and safety studies will al-so likely be mandatory for other large,structurally com-plex,heterogeneous biologics(such as fusion proteins and monoclonal antibodies)to confirm comparable effi-cacy and minimize the risk of adverse outcomes.It is recommended that clinical studies be performed using manufacturing scale batches and the final formulation intended for marketing[2,6,18].3.4.1Human pharmacologyHuman pharmacokinetic and/or pharmacodynamic studies are fundamental components in supporting bio-similarity.After physicochemical,structural,functional,and in vivo nonclinical similarity has been established,the proposed biosimilar candidate can proceed to clini-cal development[2,6].This usually starts with a phase1 human pharmacokinetic study designed to establish bio-equivalence with the reference product.The general standard to establish bioequivalence within the prespec-ified acceptance range,with the90%confidence inter-val being within80%-125%for overall exposure(e.g.,serum concentration over time and area under the serum concentration-time curve)[19],provides evidence that the proposed biosimilar product has equivalent ex-posure as the reference product.This is critical to the abbreviated development program for biosimilars,be-cause when combined with having the same analytical/ functional activity,it allows one to skip phase2studiesas it supports the assumption that the clinical dose is known.Human pharmacodynamic studies should dem-onstrate a similar effect on a clinically relevant measure related to mechanism of action and clinical effective-ness,if such a marker is available.These studies are most compelling when the relationship between expo-sure and effect can be demonstrated on the steep part of the dose-response curve and when tested with more than one dose level.3.4.2Efficacy and safetyClinical development for biosimilars typically moves directly to pivotal studies after human pharmaco-kinetics and/or pharmacodynamics has been estab-lished.These studies should be performed in popula-tions that are sensitive enough to detect clinically meaningful differences between the proposed biosimilar candidate and the reference product[6].While efficacy trials include safety evaluations within statistical power constraints,clinical studies should be designed to dem-onstrate that the proposed product has neither de-creased nor increased efficacy compared to the refer-ence product.The most straightforward design is one in which the null hypothesis,based on a prespecified e-quivalence margin,is a two one-sided tests procedure that demonstrate that the proposed biosimilar is neither inferior or superior to the reference product[20].The margins should be scientifically justified and adequate to enable detection of clinically meaningful differences in effectiveness between the proposed biosimilar and reference molecules.The efficacy endpoint can be that of clinical benefit,or alternatively,a meaningful surro-gate for efficacy of the molecule.Ideally,safety is as-sessed in the same study as efficacy,but it is more im-portant that safety be assessed in a sensitive popula-tion.Generally,this may be a population for which the test article is used as monotherapy.The use of surrogate endpoints can also play a key role in the development of biosimilars.For example,while overall survival is considered a gold standard for proving clinical benefit in oncology,it is often not a practical endpoint.Moreover,it is not necessary for bio-similars to re-establish clinical benefit per se;instead,the endpoint needs to be sensitive enough to detect a difference in activity if it exists.Overall response rate and/or complete response could be suitable end-points[21].3.4.3ImmunogenicityThe FDA guidance explicitly mentions that,“im-munogenicity remains a critical factor when assessing biosimilarity,”and the FDA will evaluate immunoge-nicity in a risk-based manner[4,6].Most biopharmaceu-ticals induce immune responses,which in many cases do not have clinically relevant consequences.The im-mune response can include development of antidrug an-tibodies that may either bind to the drug with no conse-quence or reduce its effectiveness,or neutralizing anti-bodies that eliminate activity.The most severe circum-stance is the cross-reaction of antidrug antibodies with an endogenous protein,eliminating its critical function and potentially causing harm.The extent of immunogenicity can vary due to changes in the manufacturing procedures of the same biosimilar or among different manufacturers of biosimi-lars and/or the reference.This risk is apparent from ca-ses of pure red cell aplasia(PRCA)in patients receiv-ing a brand of erythropoietin approved in Europe[22],which were traced to a minor change in the production process.Immune responses may affect both the safety and effectiveness of the product by altering pharmaco-kinetics,inducing anaphylaxis,or promoting develop-ment of neutralizing antibodies.Thus,establishing an immune response that is no worse than that known for the reference product is a key element in the demon-stration of biosimilarity.At least one clinical study that includes a comparison of immunogenicity with sensitive assays and in a sensitive population(immune-compe-tent where relevant)would be expected in the market-ing application.3.4.4ExtrapolationExtrapolation to approved indications other than those studied in support of the proposed biosimilar prod-uct is an important consideration.If equivalence trials were to be performed in each indication,the development program for a biosimilar would effectively negate the con-cept of an abbreviated approval pathway based on develo-ping a highly similar molecule of a reference product with an established risk-benefit profile[20].Both EMA and FDA draft guidelines state that extrapolation is allowable。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
Research ArticleQuality and Batch-to-Batch Consistency of Original and Biosimilar Epoetin ProductsLiem Andhyk Halim 1,Vera Brinks 1,Wim Jiskoot 2,Stefan Romeijn 2,Rob Haselberg 3,Chris Burns 4,Meenu Wadhwa 4,Huub Schellekens 1,*1Department of Pharmaceutics,Utrecht Institute for Pharmaceutical Sciences (UIPS),Utrecht University,3584CG Utrecht,the Netherlands 2Division of Drug Delivery Technology,Leiden Academic Centre for Drug Research,Leiden University,2300RA Leiden,the Netherlands 3Division of BioAnalytical Chemistry,AIMMS research group BioMolecular Analysis,VU University Amsterdam,1081HV Amsterdam,the Netherlands 4Biotherapeutics Group,National Institute for Biological Standards and Control,Hertfordshire,EN63QG,UKa r t i c l e i n f oArticle history:Available online xxxKeywords:recombinant human erythropoietin biosimilarimmunogenicityphysicochemical properties protein characterizationa b s t r a c tComprehensive physicochemical characterization and biological assays are essential parts in assessing quality attributes of biologicals.Here,we compared the quality of different marketed recombinant hu-man erythropoietin (epoetin)products:originators,Eprex and NeoRecormon as well as 2biosimilars,Retacrit and Binocrit.In addition,assessment of batch-to-batch variability was included by collecting 2or more batches of each mon assays which included sodium dodecyl sulfate e polyacrylamide gel electrophoresis,high-performance size-exclusion chromatography,asymmetrical flow field e flow fractionation,capillary zone electrophoresis,and potency testing were used.Of the tested products and among batches of single products,variations in epoetin content,isoform pro files,and potency were found.Ultimately,this study demonstrated the high quality of epoetin products with some degree of variation among products and batches,con firming the “similar but not identical ”paradigm of biologicals.©2016American Pharmacists Association ®.Published by Elsevier Inc.All rights reserved.IntroductionSince the 1980s,the advent of recombinant DNA technology has enabled the development of many innovative recombinant human therapeutic proteins.1,2These products have enabled the treatment of a variety of diseases and have become the fastest growing class of therapeutics.Recombinant human erythropoietin (epoetin)was one of the first authorized recombinant proteins on the market.It is mainly used for the treatment of anemia in patients with chronic kidney disease and cancer.3,4Severe side effects,such as thromboembolic processes and antibody-associated pure red cell aplasia (PRCA)are rare.PRCA may occur if epoetin-induced antibodies are able to neutralize the native endogenous erythropoietin.5,6Epoetin shares its factors for immunogenicity with nearly all therapeutic proteins.The exact mechanisms underlying immunogenicity are still not fullyunderstood.Multiple factors including product-related factors (formulation,contaminants,glycosylation and impurities),storage and handling,route of administration,and patient characteristics play a role in this.7,8Since 2006,the loss of patent and data protection has allowed the introduction of generic versions of therapeutic proteins such as somatropin,filgrastim,and epoetin.However,the generic regula-tory route used for small molecules cannot be used for proteins.Owing to their inherent variability,complexity,and heterogeneity,it is impossible to establish that 2protein products are identical.9,10Individual protein products themselves also demonstrate micro-heterogeneity and batch-to-batch variability so cannot be identical to themselves.Therefore,regulatory frameworks have been established throughout the world requiring an extensive compar-ison in quality,ef ficacy,and safety to show similarity between the original product and the intended copy.11,12If the criteria are met,the duplicate product can be marketed as a biosimilar.As we had access to 4marketed epoetin products,2originators,Eprex and NeoRecormon,and 2biosimilars,Retacrit and Binocrit,we performed quality assessment for these products.Eprex (epoetin alfa)and NeoRecormon (epoetin beta)have been reported to differ in their isoform compositions and biological properties onThis article contains supplementary material available from the authors by request or via the Internet at /10.1016/j.xphs.2015.10.019.*Correspondence to:Huub Schellekens (Telephone:þ31(0)302537306;Fax:þ31(0)302537839).E-mail address:h.schellekens@uu.nl (H.Schellekens).Contents lists available at ScienceDirectJournal of Pharmaceutical Sciencesjournal ho mep age:www.jp harmsci.org/10.1016/j.xphs.2015.10.0190022-3549/©2016American Pharmacists Association ®.Published by Elsevier Inc.All rights reserved.Journal of Pharmaceutical Sciences xxx (2016)1e 9account of the use of different CHO cells strain.13Meanwhile,the quality assessment of Retacrit and Binocrit to their reference product,Eprex,has been shown elsewhere to have slight variation in their quality attributes.14,15Besides quality,batch consistency is also considered important for biologicals.Although a few studies have looked into batch-to-batch variability of an individual epoetin brand,13,16,17there has been no published study on batch-to-batch consistency of multiple epoetin brands marketed in Europe.As we also had the possibility to collect multiple batches from these4epoetin prod-ucts,this comparability study is feasible as a follow-up to a study we published earlier.14Materials and MethodsEpoetin ProductsAll epoetin products(see Table1for an overview)were either obtained from local pharmacies in the Netherlands or provided by Hospira and Sandoz.They were received in the original prefilled syringes and stored as stated on the product specification.As an internal reference standard,epoetin-biological reference prepara-tion(BRP)batch3(EDQM,Strasbourg,France)was included in every experiment to validate the method as recommended in the European Pharmacopeia(Ph.Eur.)monograph on Erythropoietin concentrated solution.18It contains equals parts of epoetin alfa and beta.19Before every test,visual inspection was performed for the potential presence of visible particles.All products remained clear and colorless.In all cases,products were tested within their shelf lives.Sodium Dodecyl Sulfate e Polyacrylamide Gel Electrophoresis The epoetin products were loaded on5%polyacrylamide gel (stacking section)and separated on15%polyacrylamide gel (running section)under nonreducing conditions as previously described by Brinks et al.14Unless indicated otherwise,all materials were obtained from Bio-Rad Laboratories B.V.(Veenendaal,the Netherlands).In short,loading solutions of all epoetin products included24m L of undiluted products and6m L of5Âsample buffer (containing Tris-HCl pH6.8,glycerol,sodium dodecyl sulfate and bromophenol blue).Two micrograms of epoetin-BRP batch3were included on each gel.Before loading,all samples were incubated either at95 C,70 C, or room temperature(±25 C)for10min to facilitate protein unfolding.PageRuler™Prestained Protein Ladder,10-180kDa (Life Technologies,Bleiswijk,the Netherlands)was used as a reference for molecular weight in all cases.Separation was per-formed on Mini-PROTEAN®II Electrophoresis Cell with the following running conditions:30min at70V,followed by60min at 150V.Protein bands were visualized by silver staining method as described by Brinks et al.14High-Performance Size-Exclusion ChromatographyDuring the course of this study,the collection of multiple batches of each epoetin product was rather difficult.Hence, epoetin products were obtained at different time points.Reta-crit and NeoRecormon were obtained back in2010.Subse-quently,Binocrit and Eprex were obtained in early and late 2014,respectively.As there was an urge to analyze unexpired products,high-performance size-exclusion chromatography (HP-SEC)wasfirst performed on a Waters2695Separations Module connected to a Waters2487Dual l Absorbance Detector(Waters Corporation,Milford,MA)for thefirst2 products.The machine was then no longer available,and we had to switch to an Agilent1200HPLC system(Agilent Technologies,Palo Alto,CA)combined with a Wyatt Eclipse (Wyatt Technology Europe GmbH,Dernbach,Germany)to analyze the later products.On both machines,a Tricorn™high-performance Superdex 20010/300GL column(GE Healthcare,Little Chalfont,Buck-inghamshire,United Kingdom)was installed.Auto sampler (Agilent)temperature was set at4 C,and each time,100m L of undiluted product were injected.The eluent was14.4g/L Na2HPO4.2H2O(Sigma-Aldrich,Zwijndrecht,the Netherlands), 0.2g/L KH2PO4,and23.4g/L NaCl(Merck,Darmstadt,Germany)at pH7.4andfiltered through a0.2-m mfilter(Sartorius Stedim, G€o ttingen,Germany).Separation took place at aflow rate of0.5mL/min for60min at 30 C.Absorbance was recorded at280nm and analyzed using either Empower2software version6.20.00.00or Astra software version5.3.4.20.A DAWN®HELEOS™18-angle laser light scattering (MALLS)was part of the Agilent system,therefore allowing estimation of the average molecular weight of eluting compounds. Alternatively,proteins with different molecular weights,(1)lyso-zyme,(2)trypsin,(3)ovalbumin,(4)albumin,and(5)holo-transferrin,were used on the Waters system as calibration standards for molecular weight estimation.All proteins were purchased from Sigma-Aldrich.Subsequently,the protein content was determined from the UV signal at280nm using Beer-Lambert law.For all epoetins, a molar extinction coefficient of22,600MÀ1cmÀ1was used.20Table1List of All Epoetin ProductsBrand Name(INN)Lot Number Declared Potency ExcipientsEprex(epoetin alfa)DDS5L00DGS4W00DHS5T00DIS3M004000IU/0.4mL Sodium dihydrogen phosphate dihydrate,disodium phosphate dihydrate,sodium chloride, glycine,polysorbate80Binocrit(epoetin alfa)45011273041234121110,000IU/1.0mL8000IU/0.8mLSodium dihydrogen phosphate dihydrate,disodium phosphate dihydrate,sodium chloride,glycine,polysorbate80Retacrit(epoetin zeta)8K058L88M072C99F081G99M108N910,000IU/1.0mL Disodium phosphate dihydrate,sodium dihydrogen phosphate dihydrate,sodium chloride, calcium chloride dihydrate,polysorbate20,glycine,leucine,isoleucine,threonine,glutamic acid,phenylalanineNeoRecormon(epoetin beta)H0002H01H0003H0130,000IU/0.6mL Urea,sodium chloride,polysorbate20,sodium dihydrogen phosphate dihydrate,disodium phosphate dodecahydrate,calcium chloride dihydrate,glycine,l-leucine,l-isoleucine,l-threonine,l-glutamic acid,l-phenylalanineL.A.Halim et al./Journal of Pharmaceutical Sciences xxx(2016)1e92One-IU epoetin was set to8.4and8.3ng epoetin protein for epoetin alfa21,22/zeta23and beta,24respectively.Asymmetrical Flow Field e Flow FractionationAsymmetricalflowfield eflow fractionation was performed on an Agilent1200HPLC system(Agilent Technologies)with degasser, cooled auto sampler,and a UV(280nm)and afluorescence de-tector.It was combined with a Wyatt Eclipse(Wyatt Technology Europe GmbH,Dernbach,Germany)and a DAWN®HELEOS™18-angle laser light scattering(MALLS)detector(Wyatt Technology Europe GmbH).Fifty microliters of each undiluted formulation were injected through a350-m m thick,medium wide-spaced in a small channel with a Nadir5-kDa cutoff regenerated cellulose membrane(Wyatt Technology Europe GmbH).The same mobile phase buffer was used as in HP-SEC but wasfiltered through a0.1-m m cellulose nitrate Whatman™filter(GE Healthcare Life Sciences, Pittsburgh,KS).The detectorflow and the focusflow were set to1 and 1.5mL/min,respectively.The Eclipse elution settings are summarized in Table2.Calculation of the molecular weight from the MALLS and UV signals was performed by the Astra software version 5.3.4.20.Protein content was determined as described before.Enzyme-Linked Immunosorbent AssayEpoetin was identified with the Quantikine IVD Human Epoetin ELISA(R&D Systems Europe,Abingdon,Oxon,United Kingdom) according to the manufacturer's instructions.After fractionation by HP-SEC as described by Hermeling et al.,2520m L of each fraction was added to a well containing100m L of assay diluent buffer and 80m L of specimen diluent buffer,provided by the kit Chromogen, was left to react for20min before it was stopped by the addition of acid.The plate was immediately read on an Infinite®M1000PRO microplate reader(Tecan,Giessen,the Netherlands)at450and 600nm(reference wavelength).In each plate,both recombinant human epoetin provided in the kit and Eprex were used as the standards.Capillary Zone ElectrophoresisThe isoform distribution of different epoetin products was assessed by capillary zone electrophoresis(CZE)according to the Ph.Eur.monograph on Erythropoietin concentrated solution.18 Binocrit was analyzed on a7100CE System equipped with a photodiode array detector and ChemStation software from Agilent Technologies(Wilmington,DE);the remaining epoetin products were analyzed on a ProteomeLab™PA800or a PA800Plus Phar-maceutical Analysis System coupled to UV detector and operated with32Karat software from Beckman Coulter(Brea,CA).Epoetin internal reference standard and products were pretreated by direct loading to either Nanosep®(Pall Corporation,Ann Arbor,MI)or Amicon®Ultra(Sigma-Aldrich)centrifugal devices,both with a molecular weight cutoff value of10kDa as described in the Ph.Eur.monograph.Retentates were aliquoted and stored atÀ80 C until just before separation.An uncoated50m m inner diameter fused-silica capillary with an effective length of100cm(Polymicro Technologies,Phoenix,AZ) was used for separation.The CZE buffer consisted of0.01-M tricine (Acros Organics,Geel,Belgium),0.01-M sodium chloride(Merck), 0.01-M sodium acetate(Merck),7-M urea(Amresco,Solon,OH)and 25-mM putrescine(Sigma-Aldrich),pH5.55adjusted with50%(v/v) glacial acetic acid at30 C andfiltered through Minisart®0.45-m mfilter(Sartorius Stedim).The preconditioning of the capil-lary and between-run rinsing was performed,adapting either Ph. Eur.monograph or Zhang et al.26In both methods,epoetin internal reference standard and products were injected hydrodynamically at 0.7psi for40s with a separation voltage of143V/cm.The UV detectorset at214nm was operated at2Hz.The isoform distribution was assessed from3or more independent runs of every batch.In Vitro BioassayAn in vitro bioassay was performed using the erythropoietin-dependent subline UT-7/EPO derived from a human eryth-roleukemia.27Cells were maintained in Iscove's modified Dulbec-co's medium containing10%heat-inactivated fetal calf serum supplemented with L-glutamine(2mM),penicillin(50U/mL), streptomycin(0.05mg/mL),and0.2IU/mL epoetin.Cells were subcultured every2-3days and split1:5when they had reached a cell density of2-5Â105cells/mL.Two-fold dilutions of the epoetin samples ranging from0.1 IU/mL to0.00078IU/mL were incubated with UT-7/EPO cells at a density of0.5Â104cells/well.The plates were incubated at37 C, 5%CO2for48h,and3H-thymidine(thymidine[methyl-3H]1mCi [37MBq]/mL,PerkinElmer,Beaconsfield,United Kingdom) 0.5m Ci/well,diluted in assay medium,added for the last4h of the incubation period.The cells were harvested onto glassfiberfilter mats using a micro96harvester(Molecular Devices,Wokingham, United Kingdom)and the radioactivity incorporated into DNA estimated by scintillation counting using a2450MicroBeta2scin-tillation counter(PerkinElmer,Waltham,MA).Bioactivity estimates of the different preparations were derived relative to the epoetin standard(Third WHO International Standard[third WHO IS]forTable2AF4Elution Program SettingsStep Start(min)Duration(min)Crossflow(mL/min)Elution02 1.8Focus21 1.5Focusþinjection32 1.5Elution510 1.8Elution15100Figure1.SDS-PAGE of all epoetin products under nonreducing condition.Roman number represents the different batches of each products.S is epoetin-BRP,and RT stands for room temperature.Different intensities are most likely due to gel-to-gel variation.L.A.Halim et al./Journal of Pharmaceutical Sciences xxx(2016)1e93erythropoietin,recombinant,for bioassay,11/170available from NIBSC,United Kingdom).In Vivo Potency TestOn account of ethical considerations in the use of animals,only selected epoetin products and batches were assessed for potency in normocythemic mice by measuring the stimulation of reticulocyte production according to the Ph.Eur.monograph for erythropoietin-concentrated solution.18BALB/c female mice received the third WHO IS for erythropoietin and the epoetin products,diluted into a high,middle,and low dose,subcutaneously.Each dilution group consisted of6animals weighing between16and23g.Mice were kept for5days,and blood was withdrawn from the orbital sinus before culling by cervical dislocation.At the end of the assay, reticulocyte concentration as a percentage of total erythrocyte concentration was determined.Potency estimates for the epoetin products were calculated relative to the epoetin standard,byfitting a parallel-line model comparing assay response to log concentra-tion using CombiStats version5.0(1999-2013EDQM/Council of Europe).Assay validity was assessed by analysis of variance with nonlinearity and nonparallelism considered significant at the1% level(p<0.01).Duplicate potency estimates from independent bioassays were combined using CombiStats version5.0.ResultsSodium Dodecyl Sulfate e Polyacrylamide Gel Electrophoresis Possible proteinaceous impurities in the products were checked with sodium dodecyl sulfate e polyacrylamide gel electrophoresis under nonreducing conditions.In all tested products,a single broad band of epoetin was apparent on silver staining(Fig.1),corre-sponding in position and intensity to the single band of the epoetin-BRP batch3.Neither higher molecular weight species nor fragments were found in any batch.In addition,different sample preparations,that is,incubation at either95 C,70 C,or room temperature(25 C),did not induce aggregation and/or degrada-tion.Faint bands identified in the Retacrit and NeoRecormon samples at43kDa and higher were likely due to overloading of prestained protein marker.High-Performance Size-Exclusion ChromatographyHP-SEC was used to characterize soluble aggregates and to quantify epoetin.As demonstrated in Figure2,the main epoetin peak of Eprex and Binocrit(panels a and b)separated on an Agilent HPLC system was detected at29.9and30.1min,respectively.The monomer identities of both epoetin alfa products showed compa-rable average molecular weight close to the theoretical value of 30.4kDa,as estimated by MALLS(Supplementary Fig.1,filled bars).28The additional peaks before the main epoetin peak are probably related to the use of polysorbate80(PS80)as a stabilizer. Thisfinding has also been reported by Hermeling et al.25and will be discussed further below.On a Waters system,the main epoetin peak eluted at29.3and 28.2min for Retacrit and NeoRecormon,respectively(Figs.2b and 2c).Although Retacrit eluted about1min later,its average molec-ular weight was found to be similar to that of NeoRecormon,as estimated by the calibration standards of protein with known molecular weight(Supplementary Fig.1,empty bars).Here,3 possible explanations are suggested.First,the method variability of using proteins with known molecular weight to estimate the molecular weight of epoetin might be a factor.Hence,difference of 1min in elution time cannot be precisely measured in terms of molecular weight.It may also explain the larger estimated average molecular weight of epoetin monomer(57.1kDa)than the theo-retical value(30.4kDa).Second,the difference in elution timebutFigure2.HP-SEC chromatograms of(a)Eprex,(b)Binocrit,(c)Retacrit,and(d)NeoRecormon.The inset is a zoom of the chromatograms of the area between15and30min. The different colors represent the batches of single product.L.A.Halim et al./Journal of Pharmaceutical Sciences xxx(2016)1e94similar estimated molecular weight suggests possible different hydrophilic interactions between the column materials and the 2types of epoetin.Third,differences in glycosylation pattern may lead to differences in the amount of bound water and hence the hydrodynamic volume of the proteins.To study whether the high e molecular weight (HMW)peak was due to PS80or epoetin oligomers or both,(1)0.3mg/mL of PS80,(2)Eprex,and (3)Eprex spiked with 0.3mg/mL of PS80were applied onto the column.As shown in Figure 3a ,the retention times of PS80(peak 1)and Eprex HMW (peak 2)partly overlap.When spiking Eprex with 0.3mg/mL of PS80,the shoulder of Eprex HMW increased signi ficantly and the HMW peak itself shifted to a slightly longer retention time (compare peak 2and peak 3).Conversely,the PS80peak was slightly shifted in the presence of epoetin (compare peak 1and peak 3).These results indicate that epoetin affects the elution behavior of PS80,vice versa ,which compromises an accu-rate assessment of aggregate content.In an attempt to overcome this,we added an equal concentra-tion of PS80to the mobile phase buffer as present in Eprex (0.3mg/mL),with the intention to avoid any PS80signals.25However,although the concept worked for placebo formulation and for the Eprex formulation,the peak became smaller,and for the latter sample,a negative peak appeared (Fig.3b ),again indicating that epoetin and PS80mutually in fluence each other's elution behavior.These data show that it is impossible to accurately determine the amount of epoetin within the HMW species peak.Therefore,as adapted from Hermeling et al.,25fractions (250m L)of Eprex (DDSL500)were collected from 10min until 35min,and theepoetin content therein was assessed by enzyme-linked immuno-sorbent assay.As shown in Figure 4,there was no detectable epoetin dimer or oligomer in the region where HMW species eluted (15-25min).This implies that the different elution behavior of PS80alone,PS80in Eprex formulation,and Eprex spiked with PS80was not solely in fluenced by epoetin.Instead,buffer components or denatured protein could also affect the in fluence behavior of PS80.29Furthermore,the detected HMW species might as well consist of denatured protein which was not detected by the antibodies.The determination of epoetin monomer content in all batches is summarized in Table 3.As expected,a higher EPO monomer con-tent was found in NeoRecormon than that in the other epoetin products,in line with the higher potency as declared on the label.The epoetin monomer content of Binocrit and Retacrit was ~3%and ~14%,respectively,less than that of Eprex.Content discrepancies between batches of 1brand were also apparent.In Eprex,batch DDSL500contained 2%more monomeric epoetin than in DGS4W00.Batch-to-batch variation was found to be the highest in NeoRecormon (~12%).Asymmetrical Flow Field e Flow FractionationAsymmetrical flow field e flow fractionation (AF4)was used as an orthogonal method to HP-SEC for the separationandFigure 3.Zoomed HP-SEC chromatograms of (i)PS80,(ii)Eprex,and (iii)Eprex spiked with 0.3mg/mL PS80in (a)absence or (b)presence of 0.3mg/mL PS80in the mobilephase.Figure 4.Chromatograms of Eprex batch DIS3M00(solid line)and 0.3mg/mL of PS80(dashed line)on Superdex 200column (left y -axis)and results from epoetin speci fic enzyme-linked immunosorbent assay (dotted line)on Eprex column fractions (right y -axis).Table 3Comparison of Content of the 4Epoetin Products Tested Brand NameLotNumber Declared Content (IU/mL)Content UV280(IU/mL)HP-SEC AF4EprexDDS5L0010,0009963±29747±38DGS4W009770±359480±189DHS5T009875±1209614±76DIS3M009825±1009587±38Mean 9858±809607±105Retacrit 8K058L810,0008014±639275±6368M072C98496±5210,059±7009F081G98480±819468±3699M108N98808±4410,450±587Mean 8450±629812±586Binocrit 73041210,0009553±711,008±74245011219400±N.D.10,185±5913412119803±3410,817±265Mean10,767±2711,940±636NeoRecormon H0002H0150,00047,784±35852,158±139H0003H0153,841±9258,582±569Mean50,813±26155,370±414N.D.,not determined.L.A.Halim et al./Journal of Pharmaceutical Sciences xxx (2016)1e 95Figure 5.AF4elugrams of the 4epoetin products (a)Eprex,(b)Binocrit,(c)Retacrit,and (d)NeoRecormon.The inset is a zoom into the elugrams in the area between 10and 25min.The different colors represent the batches of singleproduct.Figure 6.Representative CE-UV analysis of the 4epoetin products.Each product is represented by 1repetition of 1batch.L.A.Halim et al./Journal of Pharmaceutical Sciences xxx (2016)1e 96quanti fication of various sizes of protein monomer and aggregates.As shown in Figure 5,2or more distinct peaks were detected in all epoetin products.The elution time of epoetin monomer slightly differed between products.Monomers of Eprex and Binocrit,which are epoetin alfa,were detected at between 8.6and 8.8min.Monomers of Retacrit (epoetin zeta)and NeoRecormon (epoetin beta)were eluted slightly later at ~8.9-9.2min.As in HP-SEC,differences in elution time between epoetin products were not re flected in the average molecular weight as estimated by MALLS (Supplementary Fig.1).It shows that epoetin possibly interacts with the cellulose membrane.30The peak eluting at ~11min in both epoetin alfa products (Figs.5a and 5b )is likely related to PS80.As shown in Supplementary Figure 2,the peak of PS80alone also has the same elution time.For Retacrit and NeoRecormon (Figs.5c and 5d ),which contain PS20instead of PS80,this particular peak was absent.Peaks eluting earlier than epoetin monomer (<8min)are likely due to the excipients (listed in Table 1).The peaks eluting when the cross flow was stopped (>15min)may be due to larger impurities.However,because small peaks with similar retention times were also observed when injecting placebo for-mulations (results not shown),it is also possible that they may result,at least in part,from contamination of the AF4channel and tubings.In line with the results obtained with HP-SEC,the highest epoetin monomer content was found in NeoRecormon.In addition,the highest content differences between batches ~13%were found in NeoRecormon.The variation between Eprex batches (~3%)was lower than that of other tested products.In contrast,Eprex con-tained the least epoetin monomer content among all epoetin products.These results clearly indicate variation in content deter-mination between HP-SEC and AF4most likely due to different adsorption which hinders the full mass recovery of the injected protein.30Capillary Zone ElectrophoresisMultiple isoforms of epoetin were detected on separation by CZE by an adapted Ph.Eur.method.Owing to the use of different instrumentation,some migration time differences were observed.To correct for this,the time scale of the electropherograms was converted to effective mobility.This conversion enabled good inter-and intraproduct comparison.In all products,the effective mobility of isoforms was observed between 0.001and 0.003cm 2V À1min À1(Fig.6)indicating reproducible migration behavior.Eprex and Binocrit (epoetin alfa)consisted of 6isoforms,whereas Retacrit (epoetin zeta)contained an additional isoform.Neo-Recormon,which is an epoetin beta,contained 8isoforms,similarFigure 7.Relative isoform distribution of (a)different batches of Eprex (n ¼3)and (b)different epoetin products,namely Eprex (n ¼12),Binocrit (n ¼8),Retacrit (n ¼13),and NeoRecormon (n ¼10).The area inside black box represents the acceptance criteria based on Ph.Eur.monograph on Erythropoietin concentrated solution.Error bar indicates standard deviation.Table 4Weighted Mean Potencies (IU/mL)of 4Epoetin Products With Upper and Lower 95%Con fidence Limits ProductBatchDeclared Potency (IU/mL)Epoetin Content as IU/mL (95%Fiducial Limits)In Vitro /In VivoIn Vitro Potency (IU/mL)In Vivo Potency (IU/mL)EprexDDS5L0010,00012,500(12,100-13,000)N.D.N.D.DGS4W00N.D.N.D.N.D.DHS5T00N.D.N.D.N.D.DIS3M0013,100(12,700-13,500)N.D.N.D.Retacrit 8K058L810,0008610(7880-9410)9920(8234-11,951)0.878M072C99190(8300-10,200)N.D.N.D.9F081G99440(9000-9900)N.D.N.D.9M108N99040(8680-9400)11,886(9834-14,365)0.76Binocrit 73041210,00015,300(14,300-16,400)9395(8190-10,777) 1.63450112115,200(14,300-16,400)8015(6828-9408) 1.9034121115,700(14,900-16,600)8544(7573-9640)1.84NeoRecormon H0002H0150,00054,000(52,500-55,500)49,483(42,123-58,128) 1.09H0003H0157,500(55,500-59,500)50,965(43,401-59,847)1.13N.D.,not determined.L.A.Halim et al./Journal of Pharmaceutical Sciences xxx (2016)1e 97。