Overview of the Bioequivalence Review
人体生物等效性研究--常见问题及其相关考虑

科
High Permeability
High Permeability
Low Permeability
迈
医
药
三、其他类型的BE豁免
BE豁免--例外
对于副作用太大,无法用健康受试者做高规格制剂 BE试验的药物,若 Low Solubility High Solubility
制剂为含量属于小浓度(当浓度小于 5%),高效能的药品,可以低规 High Permeability High Permeability 格通过BE,则高规格获得BE豁免(需满足上述BE豁免三要素)。 【例】 盐酸普拉克索片BE(治疗特发性帕金森氏症)
4. BCS Ⅲ类药物受试制剂与参比制剂的辅料性质相同、含量相似
泰
科
High Permeability
迈
医
Low Solubility
药
二、基于BCS分类的BE豁免
溶出度:具有快速溶出能力High Permeability
溶解性:在250 mL或更少的介质中溶解 渗透性:体内吸收程度≥85%
基于BCS分类的 BE 豁免不适用于口服缓释(extended release,ER) Low Permeability
Low Permeability
或控释(control release,CR)制剂, ER和CR制剂不得BE豁免; 只有制剂中的API属于BCS I类或BCS III类药物才可以考虑BE豁免。
Low Solubility 糖醇在胃肠道的吸收较差,可增加肠道渗透压,影响胃肠 Low Permeability
道中水分的输送和肠道运药时间,从而影响药物吸收。表 面活性剂、脂肪酸、壳聚糖等辅料可能改变肠道通透性。
《以药动学参数为终点评价指标的化学药物仿制药人体生物等效性研

《以药动学参数为终点评价指标的化学药物仿制药人体生物等效性研究技术指导原则》解读摘要等效性评价是我国仿制药评审的重要环节,目前等效性评价均以2005年颁布的《化学药物制剂人体生物利用度和生物等效性研究技术指导原则》为依据。
2016年3月,国家食品药品监督管理总局发布《以药动学参数为终点评价指标的化学药物仿制药人体生物等效性研究技术指导原则》,本文从分析方法、试验设计、受试者和试验例数、参比制剂等几个方面解读两者的差别。
ABSTRACT Equivalence assessment is an important part of Chinese generic drug evaluation. At present,it is based on“the technical guidelines for the study of the human bioavailability and bioequivalence of chemical pharmaceutical preparations”promulgated in 2005. The technical guideline for the b ioequivalence study of generic chemical drugs with pharmacokinetics parameters as the final evaluation index was issued by China Food and Drug Administration in Mar.,2016. In this paper,the differences between the two guidelines are analyzed from several aspects,such as the analysis method,the experimental design,the number of subjects and test cases,and the reference preparation and so on.KEY WORDS bioequivalence;generic drug;guideline按照我国仿制药审评办法规定,等效性评价是最重要的环节。
欧盟GMP指南中文版

QbR Frequently Asked QuestionsDisclaimer: These are general answers and may not be applicable to every product. Each ANDA is reviewed individually. This document represents the Office of Generic Drugs’s (OGD’s) current thinking on these topics.Format and SubmissionHow should QbR ANDAs be submitted?OGD’s QbR was designed with the expectation that ANDA applications would beorganized according to the Common Technical Document (CTD) format, a submissionformat adopted by multiple regulatory bodies including FDA. Generic firms are strongly recommended to submit their ANDAs in the CTD format (either eCTD or paper) tofacilitate implementation of the QbR. The ANDA Checklist for completeness andacceptability of an application for filing can be found on the OGD web page:/cder/ogd/anda_checklist.pdf .What is a QOS?The Quality Overall Summary (QOS) is the part of the CTD format that provides asummary of the CMC aspects of the application. It is an important tool to make the QbR review process more efficient.How long should a QOS be?OGD believes the CTD guidance1 recommendation of 40 pages to be an appropriatecompromise between level of detail and concision. The CTD guidance recommendation does not include tables and figures.The same information should not be included in multiple locations in the QOS. Instead of repeating information, refer to the first location of the original information in the QOS by CTD section number.Should the QOS be submitted electronically?All applications should include an electronic QOS. For paper submissions, it isrecommended that both an electronic QOS and a paper QOS be included.What file format should be used for the QOS?All applications, both eCTD and paper submissions, should have an electronic QOS. The electronic QOS should be contained in one document. Do not provide separate files foreach section or question.The electronic QOS should be provided as both a pdf and a Microsoft Word file. Microsoft Word files should be readable by Word 2003.1 Guidance for Industry M4Q: The CTD – Quality (August 2001) /cder/guidance/4539Q.htmWhat fonts should be used in the QOS?Because of FDA’s internal data management systems, please use only use these TrueType fonts: Times New Roman, Arial, Courier New. Times New Roman is recommended as the main text font.Should the applicable QbR question be presented within the body of Module 2 of the relevant section, followed by sponsor's answer?Yes, include all the QbR questions without deletion in the QOS.Can the granularity of module 3 be used in module 2?Yes, the granularity can be used for section and subsection headings. However, the QOS should always be submitted as a single file.Can color be used in the QOS?Yes, but sponsors should ensure that the QOS is legible when printed in black and white.Colored text should not be used.Is the QOS format available on OGD webpage and questions therein mandatory to be followed?For an efficient review process, OGD desires all applications to be in a consistent format.See the OGD QbR questions and example QOS:/cder/ogd/QbR-Quality_Overall_Summary_Outline.doc/cder/ogd/OGD_Model_Quality_Overall_ Summary.pdf/cder/ogd/OGD_Model_QOS_IR_Product.pdfFor amendments to applications, should the documentation consist of a revision of the QOS? Would new PD reports be required?The QOS should not be updated after submission of the original ANDA. Any additional data (including any new PD reports) should be provided as a stand alone amendment.Responses to deficiencies should be provided in electronic format as both a pdf andMicrosoft Word file.After January 2007, what will happen to an application that does not have a QOS or contains an incomplete QOS?OGD will contact the sponsor and ask them to provide a QOS. If the sponsor provides the QOS before the application comes up for review, OGD will use the sponsor’s QOS.OGD’s QbR questions represent the current thinking about what information is essential to evaluate an ANDA. Reviewers will use deficiency letters to ask ANDA sponsors thequestions that are not answered in the sponsor’s QOS.In February 2007, 75% of ANDAs submitted contained a QOS.If a question is not applicable to a specific formulation or dosage form, should the question be deleted or unanswered?Sponsors should never delete a QbR question, but instead answer as not applicable, with a brief justification. Please answer all parts of multi-part questions.For sterile injectables, to what extent should sterility assurance be covered in QOS?The current QbR was not intended to cover data recommendations for Sterility Assurance information. In the future, other summaries will cover other disciplines.MAPP 5040.1, effective date 5/24/04, specifies location of the microbiology information in the CTD format.Where in the CTD should an applicant provide comparative dissolution data between the generic and RLD?The comparison between the final ANDA formulation and the RLD should be provided in5.3.1, this comparison should be summarized in the QOS. Comparisons with otherformulations conducted during development should be included in 3.P.2.Is it possible to submit an amendment in CTD format for a product that was already submitted in the old ANDA format?No, all amendments to an application under review should use the same format as theoriginal submission.How is a paper CTD to be paginated?“Page numbering in the CTD format should be at the document level and not at the volume or module level. (The entire submission should never be numbered consecutively by page.) In general, all documents should have page numbers. Since the page numbering is at the document level, there should only be one set of page numbers for each document.”2. For paper submission, tabs locating sections and subsections are useful.For the ANDA submitted as in paper CTD format, can we submit the bioequivalence study report electronically? Or does the Agency require paper copy only?The bioequivalence summary tables should always be provided in electronic format.Will QbR lead to longer review times?Many of the current long review times result from applications that do not completelyaddress all of the review issues and OGD must request additional information through the deficiency process. This iterative process will be reduced with the use of the QbR template.Sponsors that provide a QOS that clearly and completely addresses all the questions in the QbR should find a reduction in the overall review time.Will DMFs for the drug substance be required to be in CTD if the ANDA is in CTD format?No. CTD format DMFs are recommended.What should be included in 3.2.R.1.P.2, Information on Components?COA’s for drug substance, excipients and packaging components used to produce theexhibit batch.2 Submitting Marketing Applications According to the ICH/CTD Format: General Considerations/cder/guidance/4707dft.pdfHow should an ANDA sponsor respond to deficiencies?OGD requests that sponsors provide a copy of the response to deficiencies in electronic format as both a pdf file and a Microsoft Word file.QUALITY OVERALL SUMMARY CONTENT QUESTIONS2.3 Introduction to the Quality Overall SummaryWhat information should be provided in the introduction?Proprietary Name of Drug Product:Non-Proprietary Name of Drug Product:Non-Proprietary Name of Drug Substance:Company Name:Dosage Form:Strength(s):Route of Administration:Proposed Indication(s):Maximum Daily Dose:2.3.S DRUG SUBSTANCEWhat if an ANDA contains two or more active ingredients?Prepare separate 2.3.S sections of the QOS for each API. Label them 2.3.S [API 1] and2.3.S [API 2].What if an ANDA contains two or more suppliers of the same active ingredient?Provide one 2.3.S section. Information that is common between suppliers should not be repeated. Information that is not common between suppliers (e.g. different manufacturing processes) should have separate sections and be labeled accordingly (drug substance,manufacturer 1) and (drug substance, manufacturer 2).Can information in this section be provided by reference to a DMF?See individual questions for details. As a general overview:•Information to be referenced to the DMFo Drug substance structure elucidation;o Drug substance manufacturing process and controls;o Container/closure system used for packaging and storage of the drugsubstance;o Drug substance stability.•Information requested from ANDA Sponsoro Physicochemical properties;o Adequate drug substance specification and test methods including structure confirmation;o Impurity profile in drug substance (process impurity or degradant);o Limits for impurity/residual solvent limits;o Method validation/verification;o Reference standard.2.3.S.1 General InformationWhat are the nomenclature, molecular structure, molecular formula, and molecular weight?What format should be used for this information?Chemical Name:CAS #:USAN:Molecular Structure:Molecular Formula:Molecular Weight:What are the physicochemical properties including physical description, pKa, polymorphism, aqueous solubility (as function of pH), hygroscopicity, melting point, and partition coefficient?What format should be used for this information?Physical Description:pKa:Polymorphism:Solubility Characteristics:Hygroscopicity:Melting Point:Partition Coefficient:Should all of these properties be reported? Even if they are not critical?Report ALL physicochemical properties listed in the question even if they are not critical.If a property is not quantified, explain why, for example: “No pKa because there are no ionizable groups in the chemical structure” or “No melting point because compounddegrades on heating”.What solubility data should be provided?The BCS solubility classification3 of the drug substance should be determined for oral dosage forms.Report aqueous solubility as a function of pH at 37º C in tabular form. Provide actualvalues for the solubility and not descriptive phrases such as “slightly soluble”.3 See BCS guidance /cder/guidance/3618fnl.pdf for definitionSolvent Media and pH Solubility Form I(mg/ml) Solubility Form II(mg/ml)Should pH-solubility profiles be provided for all known polymorphic forms?No, it is essential that the pH-solubility profile be provided for the form present in the drug product. The relative solubility (at one pH) should be provided for any other more stable forms.Physicochemical information such as polymorphic form, pKa, solubility, is usually in the confidential section of DMF. Is reference to a DMF acceptable for this type of information?No, knowledge of API physicochemical properties is crucial to the successful development of a robust formulation and manufacturing process. In view of the critical nature of thisinformation, OGD does not consider simple reference to the DMF to be acceptable.The Guidance for Industry: M4Q: The CTD-Quality Questions and Answers/ Location Issues says only the polymorphic form used in the drug product should be described in S.1 and other known polymorphic forms should be described in S.3. OGD’s examples placed information about all known polymorphic forms in S.1. Where does OGD want this information?This information may be included in either S.1 or in S.3. Wherever presented, list allpolymorphic forms reported in literature and provide brief discussion (i.e., which one is the most stable form) and indicate which form is used for this product.Other polymorph information should be presented by the ANDA applicant as follows: • 2.3.S.3 Characterization: Studies performed (if any) and methods used to identify the potential polymorphic forms of the drug substance. (x-ray, DSC, and literature) • 2.3.S.4 Specification: Justification of whether a polymorph specification is needed and the proposed analytical method• 2.3.P.2.1.1 Pharmaceutical Development –Drug Substance: Studies conducted to evaluate if polymorphic form affects drug product propertiesWhy does OGD need to know the partition coefficient and other physicochemical properties?Physical and chemical properties may affect drug product development, manufacture, or performance.2.3.S.2 ManufactureWho manufactures the drug substance?How should this be answered?Provide the name, address, and responsibility of each manufacturer, including contractor, and each proposed production site or facility involved in manufacturing and testing.Include the DMF number, refer to the Letter of Authorization in the body of data, andidentify the US Agent (if applicable)How do the manufacturing processes and controls ensure consistent production of the drug substance?Can this question be answered by reference to a DMF?Yes. It is preferable to mention the source of the material (synthetic or natural) when both sources are available.The DMF holder’s COA for the batch used to manufacture the exhibit batches should be provided in the body of data at 3.2.S.4.4.If there is no DMF, what information should be provided?A complete description of the manufacturing process and controls used to produce the drugsubstance.2.3.S.3 CharacterizationHow was the drug substance structure elucidated and characterized?Can structure elucidation be answered by reference to a DMF?Yes.What information should be provided for chiral drug substances?When the drug substance contains one or more chiral centers, the applicant should indicate whether it is a racemate or a specific enantiomer.When the drug substance is a specific enantiomer, then tests to identify and/or quantify that enantiomer should be included. Discussion of chirality should include the potential forinterconversion between enantiomers (e.g. racemization/epimerization).How were potential impurities identified and characterized?List related compounds potentially present in the drug substance. Identify impurities bynames, structures, or RRT/HPLC. Under origin, classify impurities as process impurities and/or degradants.Structure Origin ID ChemicalName[SpecifiedImpurity]Is identification of potential impurities needed if there is a USP related substances method?Yes.Can this question be answered by reference to a DMF?The ANDA should include a list of potential impurities and their origins. The methodsused to identify and characterize these impurities can be incorporated by reference to the DMF.According to the CTD guidance, section S.3 should contain a list of potential impurities and the basis for the acceptance criteria for impurities, however in the OGD examples this information was in section S.4. Where should it go?This information may be included in either S.3 or in S.4.2.3.S.4 Control of Drug SubstanceWhat is the drug substance specification? Does it include all the critical drug substance attributes that affect the manufacturing and quality of the drug product?What format should be used for presenting the specification?Include a table of specifications. Include the results for the batch(es) of drug substance used to produce the exhibit batch(es). Identify impurities in a footnote. Test results and acceptance criteria should be provided as numerical values with proper units whenapplicable.Tests Acceptancecriteria AnalyticalprocedureTest results for Lot#AppearanceIdentificationA:B:AssayResidualSolventsSpecified ImpuritiesRC1RC2RC3Any UnspecifiedImpurityTotal Impurities[AdditionalSpecification]*RC 1: [impurity identity]RC 2: [impurity identity]RC 3: [impurity identity]What tests should be included in the drug substance specification?USP drugs must meet the USP monograph requirements, but other tests should be included when appropriate. For USP and non USP drugs, other references (EP, BP, JP, the DMF holder’s specifications, and ICH guidances) can be used to help identify appropriate tests.Only relevant tests should be included in the specification. Justify whether specific tests such as optical rotation, water content, impurities, residual solvents; solid state properties(e.g. polymorphic form, particle size distribution, etc) should be included in thespecification of drug substance or not.Does OGD accept foreign pharmacopeia tests and criteria for drug substances?There are several examples where a drug substance is covered by a monograph in EP or JP, but not in the USP. ANDA and DMF holders can obtain information regardingphysicochemical properties, structure of related impurities, storage conditions, analytical test methods, and reference standards from EP or JP to support their submission to OGD.Although the USP remains our official compendium, we usually accept EP when the drug substance is not in USP (However, a complete validation report for EP methods should be provided in the ANDA).For each test in the specification, is the analytical method(s) suitable for its intended use and, if necessary, validated? What is the justification for the acceptance criterion?What level of detail does OGD expect for the analytical method justifications and validations?Provide a summary of each non-USP method. This can be in a tabular or descriptive form.It should include the critical parameters for the method and system suitability criteria ifapplicable. See an example in section 2.3.P.5 of this document.For each analytical procedure, provide a page number/link to the location of validationinformation in Module 3. For quantitative non-compendial analytical methods, provide a summary table for the method validation. See an example in section 2.3.P.5 of thisdocument.Is validation needed for a USP method?No, but USP methods should be verified and an ANDA sponsor should ensure that theUSP assay method is specific (e.g. main peak can be separated from all process impurities arising from their manufacturing process and from degradation products) and the USPrelated substance method is specific (e.g. all the process impurities and degradants can be separated from each other and also separated from main peak).Is validation needed if the USP method is modified or replaced by an in-house method?Yes. Data supporting the equivalence or superiority of the in-house method should beprovided. In case of a dispute, the USP method will be considered the official method.Is reference to the DMF for drug substance analytical method validations acceptable?No. ANDA sponsors need to either provide full validation reports from the ANDA holder or reference full validation reports from the DMF holder (provided there is a copy of the method validation report in the ANDA and method verification from the ANDA holder). AppearanceIdentityAssayImpurities (Organic impurities)What format should be used for related substances?List related compounds potentially present in the drug substance. (Either here or S.3)Name Structure Origin[SpecifiedImpurity]Provide batch results and justifications for the proposed acceptance criteria. See guidance on ANDA DS impurities4 for acceptable justifications. If the DS is compendial, include the USP limits in the table. If the RLD product is used for justification/qualification, then its results should also be included. If an ICH justification is used, then the calculation of the ICH limits should be explained.To use the ICH limits, determine the Maximum Daily Dose (MDD) indicated in the label and use it to calculate the ICH Thresholds: Reporting Threshold (RT), Identification1. The amount of drug substance administered per day2. Higher reporting thresholds should be scientifically justified3. Lower thresholds can be appropriate if the impurity is unusually toxicSponsors can use the ICH limits to ensure the LOQ for the analytical method is equal or below the RT, establish the limit for “Any Unspecified Impurity” to equal or below the IT, and establish limits for each “Specified Identified Impurity” and each “SpecifiedUnidentified Impurity”5 to equal or below the QT.An impurity must be qualified if a limit is established above the QT. Options forqualification include reference to the specific impurity listed in a USP monograph,comparison to the RLD product, identifying the impurity as a significant metabolite of the drug substance, literature references including other compendial monographs (EP, BP, JP), or conducting a toxicity study.4/cder/guidance/6422dft.pdf5 The ANDA DS guidance states “For unidentified impurities to be listed in the drug substance specification, we recommend that you clearly state the procedure used and assumptions made in establishing the level of the impurity. It is important that unidentified specified impurities be referred to by an appropriate qualitative analytical descriptive label (e.g., unidentified A, unidentified with relative retention of 0.9)”. Q3A(R) states “When identification of an impurity is not feasible, a summary of the laboratory studies demonstrating the unsuccessful effort should be included in the application.”Name DrugSubstance(Lot #)USPLimit forDrugSubstanceRLDDrugProduct(Lot #)ProposedAcceptancecriteriaJustification[Specified Impurity,Identified][BatchResults][BatchResults][SpecifiedImpurity,Unidentified]Any UnspecifiedImpurityTotalImpuritiesInclude the column for RLD drug product only if that data is used to justify the drugsubstance limit (example a process impurity that is also found in the RLD).What is OGD’s policy on genotoxic impurities?FDA is developing a guidance for genotoxic impurities. According to the ICH Q3A lower thresholds are appropriate for impurities that are unusally toxic.If impurities levels for an approved generic drug are higher than the RLD, can the approved generic drug data be used as justification for a higher impurity specification?According to ANDA DP and DS Impurity guidances, any approved drug product can be used to qualify an impurity level. However, the guidances qualify this by later stating “This approved human drug product is generally the reference listed drug (RLD). However, you may also compare the profile to a different drug product with the same route ofadministration and similar characteristics (e.g. tablet versus capsule) if samples of thereference listed drug are unavailable or in the case of an ANDA submitted pursuant to a suitability petition.”What if there are no impurities’ tests found in the USP monograph for a USP drug substance? What should the ANDA sponsor do?Please work with your supplier (DMF Holder) to ensure that potential synthetic process impurities (e.g. isomers (if any), side reaction products), degradation impurities, metalcatalysts, and residual solvents are adequately captured by your impurities test method.There may be information available in published literature as well, regarding potentialimpurities.Can levels of an impurity found in the RLD and identified by RRT be used for qualification?Qualification of a specified unidentified impurity by means of comparative RRT, UVspectra, and mass spectrometry with the RLD may be acceptable. However, the ANDAsponsor should make every attempt to identify the impurity.If levels are higher than in an approved drug product then the sponsor should provide data for qualification of the safety of this impurity at this level.Can a limit from a USP monograph for “any unspecified impurity” be used to justify a limit for “any unspecified impurity” greater than the ICH Q3 identification threshold?No. Any unspecified impurity (any unknown) limit should not exceed ICH Q3A “IT”based on MDD. Non-specific compendial acceptance criteria (e.g. Any Individual Impurity is NMT 0.5%) should not be used for justification of proposed impurity acceptance criteria.However, if the USP limit is less than the ICH threshold, then the USP limit should be used. Can a limit for an identified impurity in the drug substance be qualified with data obtained from RLD drug product samples treated under the stressed conditions?No. Test various samples of marketed drug product over the span of its shelf life (ideally, near the end of shelf-life). Data generated from accelerated or stressed studies of the RLD is considered inappropriate.Impurities (Residual Solvents)Will OGD base residual solvent acceptance limits on ICH limits or process capability?The ICH guidance on residual solvents6 provides safety limits for residual solvents but also indicates that “residual solvents should be removed to the extent possible”. ANDA residual solvent limits should be within the ICH safety limits, but the review of the ANDA includes both of these considerations.OGD generally accepts the ICH limits when they are applied to the drug product.What about solvents that are not listed in Q3C?Levels should be qualified for safety.Impurities (Inorganic impurities)Polymorphic FormWhen is a specification on polymorphic form necessary?See ANDA polymorphism guidance7 for a detailed discussion.Particle SizeWhen is a drug substance particle size specification necessary?A specification should be included when the particle size is critical to either drug productperformance or manufacturing.For example, in a dry blending process, the particle size distribution of the drug substance and excipients may affect the mixing process. For a low solubility drug, the drug substance particle size may have a critical impact on the dissolution of the drug product. For a high solubility drug, particle size is often not critical to product performance.6 /cder/guidance/Q3Cfnl.pdf7/cder/guidance/6154dft.pdfWhat justification is necessary for drug substance particle size specifications?As for other API properties, the specificity and range of acceptance criteria for particle size, and the justification thereof, could vary from none to very tight limits, depending upon the criticality of this property for that drug product.Particle size specifications should be justified based on whether a change in particle sizewill affect the ability to manufacture the product or the final product performance.In general, a sponsor either should demonstrate through mechanistic understanding orempirical experiments how changes in material characteristics such as particle size affecttheir product.In the absence of pharmaceutical development studies, the particle size specificationshould represent the material used to produce the exhibit batch.When should the particle size be specified as distribution [d90,d50,d10] and when is a single point limit appropriate?When critical, a particle size should be specified by the distribution. There may be othersituations when a single point limit can be justified by pharmaceutical development studies.2.3.S.5 Reference StandardsHow were the primary reference standards certified?For non-compendial, in-house reference standards, what type of qualification data is recommended? Will a COA be sufficient?COA should be included in Module 3, along with details of its preparation, qualification,and characterization. This should be summarized in the QOS.In terms of the qualification data that may be requested, it is expected that these reference standards be of the highest possible purity (e.g. may necessitate an additionalrecrystallization beyond those used in the normal manufacturing process of the activeingredient) and be fully characterized (e.g. may necessitate in the qualification reportadditional characterization information such as proof of structure via NMR) beyond theidentification tests that are typically reported in a drug substance COA. StandardLaboratory Practice for preparation of reference standards entails recrystallization toconstant physical measurements or to literature values for the pure material.2.3.S.6 Container Closure SystemWhat container closure is used for packaging and storage of the drug substance?Can this question be answered by reference to a DMF?Yes.。
药物制剂的生物等效性研究

药物制剂的生物等效性研究在现代医学领域中,药物制剂的研发和应用是保障人类健康的重要环节。
而在这一过程中,生物等效性研究具有至关重要的地位。
什么是生物等效性呢?简单来说,生物等效性指的是两种药物制剂在相同的试验条件下,以相同的剂量给予相同的试验对象,其吸收速度和程度在统计学上没有显著差异。
这意味着,尽管两种制剂可能在成分、剂型等方面有所不同,但它们在人体内产生的药效是相似的。
为什么要进行生物等效性研究呢?首先,这是药品审批和监管的重要依据。
新研发的仿制药只有通过生物等效性研究,证明其与原研药具有生物等效性,才能够获得批准上市。
其次,对于患者来说,生物等效性的研究结果能够让他们在使用不同制剂的药物时,获得相似的治疗效果和安全性保障。
再者,从医疗资源的合理利用角度来看,生物等效性研究有助于降低医疗成本,提高药品的可及性。
生物等效性研究的设计和实施需要遵循严格的科学原则和规范。
在研究对象的选择上,通常会选择健康志愿者或者特定疾病患者。
对于一些特殊的药物,如治疗心血管疾病、精神疾病等的药物,可能会更倾向于选择患者作为研究对象,因为他们对于药物的反应更能反映临床实际情况。
研究的给药方式和剂量也是需要精心设计的。
给药方式要尽可能模拟临床实际使用的情况,剂量则要根据药物的特性和治疗需求来确定。
在研究过程中,还需要严格控制各种影响因素,比如饮食、作息、合并用药等,以确保研究结果的准确性和可靠性。
生物等效性研究中,最重要的指标是药代动力学参数。
常见的药代动力学参数包括血药浓度时间曲线下面积(AUC)、达峰浓度(Cmax)和达峰时间(Tmax)等。
通过对这些参数的测定和分析,可以评估药物的吸收、分布、代谢和排泄情况。
在进行生物等效性评价时,通常会采用统计学方法。
一般会设定一个等效性的界值,如果两种制剂的药代动力学参数的差异在这个界值范围内,就认为它们是生物等效的。
然而,需要注意的是,生物等效并不意味着完全相同,而是在一定的可接受范围内相似。
仿制药一致性评价与衡量

仿制药一致性评价定义仿制药一致性评价是指对已经批准上市的仿制药,按与原研药品质量和疗效一致的原则,分期分批进行质量一致性评价,就是仿制药需在质量与药效上达到与原研药一致的水平。
药学研究是指通过体外溶出等分析方法对药物进行药学分析,其目的在于考察制剂的生产工艺及处方是否有需要变更,初步确认制剂与原研药的一致性。
生物等效性(bioequivalency , BE )是指在同样试验条件下试验制剂和对照标准制剂在药物的吸收程度和速皮的统计学差异。
当吸收速度的差别没有临床意艾时,某些药物制剂其吸收程度相同而速度不同也可以认为生物等效。
一站式服务我司作为提供专业的医药科技公司,能够提供包括:1、药学研究(CMC):包括:制剂处方工艺、质量研究(杂质及溶出曲线等)、稳定性考察等完整的药学研究过程2、生物等效性(BE):包括:寻找合作临床机构、招募受试者釆血、生物样品测试及分析、数摇菅理及统计分析等全过程的服务3、需要进行大临床试验的品种,按照2017年已经颁布的指导原则,参照II期、III期临床试验的经验,提供整个临床试验的组织及监查管理服一致性评价(CMC&BE)的主要工作内容第一阶段:项目评估♦项目的市场价值♦竞争品种的多少♦是否有参比制剂♦评估需要的费用和周期♦咨询相关官员与专家♦项目立项确定进行BCS I类或者III类豁免BE的申请:商渗透性的数摒与文献支持材料、体外溶出曲线数据的提供与分析,如果能够满足CFDA的2016年87号文《人体生物等效性试验豁免指导原则》就可以豁免BE研究。
第二阶段:药学研究(CMC)♦参比制剂的选择及备案♦购买参比制剂♦与参比制剂的质量对比(主要包含溶出曲线和杂质)♦药学等效判定♦处方工艺等的二次开发♦溶出曲线的对比♦处方工艺的确定及中试放大♦三批中试产品的工艺验证♦中试样品的质量和参比制剂的一致♦API的溶解性和渗透性研究(限BCS I和BCS川类)♦制剂稳定性和包装考察♦申报资料的撰写及整理,提供原始记录第三阶段:BE研究API的BCS分类属于II和IV的产品必须进行BE研究,不能够豁免;BCSI类或者川类,符合豁免BE的条件可以不进行BE研究,否则就必须进行BE研究。
CMC审评_科学加风险的综合评估_美国FDACDER新药质量审评部简介

41Journal of China Prescription Drug 2009.3 No.84——美国FDA CDER新药质量审评部简介Meheb Nasr 博士 汤玉冰 博士ONDQA持续不断的致力于寻求多种方式以进一步推动科学与风险综合评估举措进入审评规范,改进审评效率,应用最先进的科学手段应对突发事件,以及鼓励药厂运用质量源于设计的原理进行药品质量管理。
除非处方药典药(O T C monograph drugs)及临床试验药(Investigational New Drugs,IND)以外,在美国上市的所有新药都必须遵守美国相关新药申请(New Drug Application)法规,事先接受美国食品药品管理局(US Food and Drug Administration,FDA)的审理并取得其批准。
FDA 下属的化学类药物评价与研究中心(Center for Drug Evaluation and Research,CDER)是其属下的五个中心之一。
此中心的职责为审评并批准申请在美上市的化学类药品及某些生物类药品的安全性、有效性及质量可控性。
新药申请材料中相当一部分涉及药品的药物化学、药品生产以及生产和质量监控(Chemistry,Manufacture and Control,CMC)。
而这部分的审评则为CDER 下属的新药质量评审部(Office of New Drug Quality Assessment,ONDQA)的主要职责,生物药品的这部分则属于生物科技产品部的职责。
需要补充说明的是国际会议协调组织(International Conference Harmonization,I C H )将C M C 部分统称为质量CMC 审评:科学加风险的综合评估专家简介Moheb Nasr博士为本文英文作者,现为美国FDA药物评审与研究中心(CDER)新药质量评审办公室主任。
负责审查药品的化学、生产与控制(CMC)的属性。
盐酸氨基葡萄糖片的相对生物利用度及其生物等效性评价
照品 (质 量 分 数 为 && 5 ’ A , 四川宝光药业股份有 限公司) , 法莫替 丁 (内 标, 质 量 分 数 为 && 5 ) A , 中 国药品生物制品检定所) , 乙腈 (色谱纯, 山东禹王 试剂厂) , 空白血浆 (沈阳中心血站) 。
[B , <] 。 分析方法符合有关国际规范要求
:;<
提取回收率和稳定性考察
取空白血浆 <22 ! 除内标溶液在过滤后加 4, 入外, 按 “1 A L A 1” 条方法制成低、 中、 高 G 个质量浓 度 (氨基葡 萄 糖 血 浆 质 量 浓 度 分 别 为 <2 A 2 、 122 A 2 和 6 222 A 2 ! ・4 5 6 ) 的 样 品, 每一质量浓度进行 L 3 样本分 析。 同 时, 另 取 空 白 血 浆 <22 ! 除标准 4, 系列溶液和内标溶液在过滤后加入外, 按 “1 A L A 1 ” 条方 法 制 成 低、 中、 高 G 个质量浓度 (氨 基 葡 萄 糖 血 浆 质 量 浓 度 分 别 为 ・4 6 222 A 2 ! 3
分别 取 ’ 名 受 试 者 的 空 白 血 浆 #%% ! 按 -, “! " #” 条方法操作 (不 加 内 标 溶 液 ) , 进 样 #% ! -记
C
沈
阳
药
科
大
学
学Байду номын сангаас
报
第 1G 卷
法操作, 进样 <2 ! 记 录 色 谱 图。 以 血 浆 中 氨 基 4, 葡萄糖质量浓 度 为 横 坐 标, 氨基葡萄糖与内标物 最小二乘 的峰面积比值 为 纵 坐 标, 用加权 ( 6 F @1 )
青蒿琥酯片在中国健康成年受试者的生物等效性试验
二、临床部分-入选标准
Second, Clinical Research Unit Inclusion criteria
1、18 ~ 45岁的健康男性成年人,同批受试者年龄相差≤10岁; 1. 18 ~ 45-year-old healthy adult male subjects with the approved age difference ≦ 10year-old. 2、体重≥50kg,体重指数(BMI)在18~25范围内(BMI=体重(kg)/身高2(m2)); 2. Weight ≧ 50kg, body mass index (BMI) within the range of 18 to 25 (BMI = body weight (kg) / height2 (m2)). 3、非过敏体质,无青蒿素类及氨基喹啉类药物或其他药物食物 过敏史; 3. Non-allergic, and not allergic to the study medicines or any other medicines in the same class or food; 4、试验前病史、体检、实验室项目及试验相关各项检查、检测 均正常或无临床意义 的轻度异常,临床研究医生判断认为合格者; 4. Medical history, physical examination, laboratory projects and test related to the inspection are normal or mildly abnormal with no clinical significance, also be judged as qualifier. 5、受试者能够和研究者进行良好的沟通,并且理解和遵守本项研究的各项要求,理 解并签署知情同意书。 5. Written informed consent obtained prior to any screening procedures.
生物等效性试验和等效性判定标准
Copyright ?国家食品药品监督管理总局药品审评中心All Right Reserved.地址:中国北京市海淀区复兴路甲1号邮编:100038总机:传真:备案序号:京ICP备09013725号
此页面上的内容需要较新版本的Adobe Flash Player。
当前位置:科学研究>>电子刊物>>电子刊物详细
发布日期
20110913
栏目
化药药物评价&等效性判定标准
作者
王凌张玉琥
部门
化药药学二部
正文内容
1、生物等效性判定标准设定的背景生物等效性(Bioequivalence,BE)是指生物效应的一致性,主要包括临床应用的安全性与有效性。仿制药的研究开发与临床药品应用的替换,其基本要求都是不同制剂间具有生物等效性。因此,BE试验在药品研发中具有非常重要的地位和作用。药物制剂间的BE评价,虽然可以通过临床对照试验,用临床指标判断两种或两种以上制剂是否具有生物等效性,但临床效应测定结果的影响因素众多、结果变异大、样本量要求大,因此并不是首选的评价方法。目前,国内外最常用的BE评价方法是药动学方法,即采用生物利用度(Bioavailability,BA)指标进行BE评价。通常,BA指制剂中活性成分被吸收的程度和速度。用药动学方法进行BE评价,就是考察药学等效制剂或可替换药品在相同试验条件下,服用相同剂量,其活性成分吸收的程度和速度是否满足预先设定的等效标准。在药动学参数中,表征吸收程度和速度的参数主要是AUC、Tmax和Cmax。因此,用药动学方法评价制剂间是否具有生物等效性,就是以统计学方法评价试验制剂与参比制剂测得的AUC、Tmax和Cmax等指标是否满足预先设定的等效标准。预先设定的等效标准如何,也就成为影响BE评价的关键因素之一。根据临床医生的建议以及FDA以往的经验,对大多药品来说,如果循环系统的药物暴露差别在20%以内,将不会对临床治疗效果产生显著影响。基于此点,FDA设定了试验制剂和参比制剂的药代动力学参数(AUC和Cmax)“差异应小于20%”作为等效性判定标准,具体判定方法为:通过双单侧t检验及(1-2α%)置信区间法,得到两种制剂AUC或Cmax几何均值比值的90%置信区间(Confidence Interval,CI),对于非窄治疗窗的药物,此90% CI必须落在80.00%~125.00%范围内。另外,FDA和EMEA的指导原则还特别强调,此置信区间必须保留两位有效数字,并且不得通过四舍五入的方法,使受试药物BE检验合格,即下限的最低值为80.00%,而上限不得超过125.00%,比如某项生物等效性试验结果为79.96%~110.20%,则判定为生物不等效[1,2]。作为非正态分布的Tmax,则要求用非参数的统计方法证明制剂间差异无统计学意义。2、全球主要国家、组织和机构采用的生物等效性判定标准同FDA要求一致,其他主要国家、地区的药品监管机构(包括欧盟EMEA,日本厚生省)和世界卫生组织(WHO)都以80.00%~125.00%作为AUC和Cmax 90% CI的等效性判定标准[2-4]。在上述机构所制订的指导原则中,对于AUC的等效性判定标准比较严格,通常只能缩小范围(如:针对某些治疗窗窄的药物,EMEA建议可以缩小范围至90.00%~111.11%)[2,3]。相对而言,Cmax的等效性判定标准具有一定的灵活性,比如加拿大药品监管机构(Health Canada)只要求Cmax均值的比值落在80~125%即可[5]。EMEA和WHO则提出,对于某些特殊情况的药物(如高变异药物,即药动学参数的个体内差异在30%以上),可以根据情况适当扩大等效性判定标准的范围[2,3],如EMEA建议对于个体内变异(CVintra)为35%的药物,等效性判定标准可以扩大到77.23%~129.48%,当CVintra为40%时,该范围可扩大至74.62%~134.02%,当CVintra为50%或以上则可以扩大至69.84%~143.19%[2]。但申办方必须提供证据证明,在此判定标准下,不会引起药物安全性问题,并保证药物的临床疗效没有显著差异,即需要证明Cmax差异的增大不会引起不良反应的显著增加,也不会显著影响疗效。此外Cmax等效性判定标准范围的扩大必须在BE试验开始前设定,并提供相应的证据,而不能在试验结束后,根据试验结果更改[2,3]。日本厚生省则建议,如果扩大Cmax的等效性判定标准范围,必须满足以下三个条件:(1)受试者人数不低于20,或在增加受试者人数之后总人数不低于30;(2)Cmax均值的对数差值在log(0.9)~log(1.1)之间;(3)对于体外溶出试验,在任何的试验条件下,当参比制剂体外溶出为30%,50%和80%时,受试制剂和参比制剂溶出度差别都在10%以内[5]。3、我国目前的现行标准和展望我国2005年颁布的《化学药物制剂人体生物利用度和生物等效性研究技术指导原则》中,AUC的90% CI的等效性判定标准和国际标准一致,而Cmax的标准,由于当时技术水平相对较低、临床试验条件等的限制,为方便BE试验的管理和审评,统一设定了较为宽松的等效性判定标准,即70%~143%[6]。近年来,随着我国临床药动学试验水平的进步和制剂研究水平的提升,对于药品质量控制的要求将更加严格,以确保高质量仿制药的开发。因此,参考先进国家与组织的规定,有必要提高Cmax的等效性判定标准要求,即采用80.00%-125.00%作为等效性判定标准。在此标准下,特殊药物,如高变异药物,可以适当扩大等效性判定标准范围,但申办者必须在BE试验前提供相关安全性和临床疗效的证据,以及个体内变异情况的证据,在此基础上重新设定等效性判定标准,如:75.00%-133.00%或者70.00%-143.00%。在试验结束后,即使发现由于个体内差异很大,造成生物不等效,也不能根据结果再次对等效性判定标准的范围进行放大。应当通过扩大受试者人数重新进行临床试验,降低标准偏差,来重新判定生物等效性。4、总结综上,国内BE试验的管理、实施和标准的制定将与国际通用标准进一步接轨,采用更为严格、统一的标准。本文作者就Cmax的判定标准进行了探讨,分析了目前国内外主要国家和地区组织的现行标准,供药品研发和注册申请人参考。
FDA《口服制剂的生物利用度和生物等效性研究一般性考虑》介绍
FDA《口服制剂的生物利用度和生物等效性研究一般性考虑》介绍《口服制剂的生物利用度和生物等效性研究:一般性考虑》是由美国食品和药物管理局(FDA)颁布的一项指南,旨在指导药物研发人员和监管机构评估口服制剂的生物利用度和生物等效性。
本文将对该指南进行详细介绍。
首先,生物利用度是衡量口服制剂在体内吸收程度的指标。
它是通过比较口服给药和静脉给药后药物在体内浓度的变化来评估的。
生物利用度高意味着药物能够更有效地被吸收,从而提高治疗效果。
而生物等效性则是判断不同制剂之间的可互换性的指标。
该指南首先阐述了生物利用度和生物等效性的概念和重要性。
接着,它详细解释了口服制剂的生物利用度评价的原则和方法。
除了传统的生物利用度评估方法,如血药浓度-时间曲线和药物代谢动力学分析,指南还介绍了现代生物技术在生物利用度评估中的应用。
同时,指南还提供了评价食物对生物利用度的影响的指导原则。
然后,指南介绍了生物等效性评价的原则和方法,包括在健康志愿者中进行的生物等效性研究设计和统计分析等方面的要点。
此外,指南还解释了当使用生物等效性研究设计可能存在困难或不切实际时的替代方法。
在指南的最后,它提供了一些特殊情况下的考虑事项和建议,例如对口服溶解速度较慢的药物、控释剂型制剂和具有良好肠黏附性的药物的评价方法。
此外,指南还指出了在特殊人群(如儿童和老年人)中进行生物等效性评价时需要考虑的因素。
《口服制剂的生物利用度和生物等效性研究:一般性考虑》的发布为药物研发人员和监管机构提供了一套科学合理的评价标准和方法,以确保口服制剂的质量和可互换性。
这对于提高药物的疗效和安全性具有重要意义,并有助于加快新药的上市速度。
但需要注意的是,该指南并非硬性要求,而是一份指导性文件,在具体情况下可能需要根据实际情况进行调整和灵活应用。
总之,FDA《口服制剂的生物利用度和生物等效性研究:一般性考虑》为口服制剂的生物利用度和生物等效性评价提供了科学指导,促进了药物的质量和可互换性评估工作。