FDA工艺验证指南

FDA工艺验证指南
FDA工艺验证指南

GUIDELINE

ON GENERAL PRINCIPLES OF ROCESS V ALIDATION

May, 1987

Prepared by: Center for Drugs and Biologics and

Center for Devices and Radiological Health

Food and Drug Administration

Maintained by: Division of Manufacturing and Product Quality (HFN-320)

Office of Compliance

Center for Drugs and Biologics

Food and Drug Administration

5600 Fishers Lane

Rockville, Maryland 20857

General Principles of Process V alidation May 1987

GENERAL PRINCIPLES OF PROCESS V ALIDA TION

I. PURPOSE

This guideline outlines general principles that FDA considers to be acceptable elements of process validation for the preparation of human and animal drug products and medical devices.

II. SCOPE

This guideline is issued under Section 10.90 (21 CFR 10.90) and is applicable to the manufacture of pharmaceuticals and medical devices. It states principles and practices of general applicability that are not legal requirements but are acceptable to the FDA. A person may rely upon this guideline with the assurance of its acceptability to FDA, or may follow different procedures. When different procedures are used, a person may, but is not required to, discuss the matter in advance with FDA to prevent the expenditure of money and effort on activities that may later be determined to be unacceptable. In short, this guideline lists principles and practices which are acceptable to the FDA for the process validation of drug products and medical devices; it does not list the principles and practices that must, in all instances, be used to comply with law.

This guideline may be amended from time to time. Interested persons are invited to submit comments on this document and any subsequent revisions. Written comments should be submitted to the Dockets Management Branch (HFA-305), Food and Drug Administration, Room 4-62, 5600 Fishers Lane, Rockville,

Maryland 20857. Received comments may be seen in that office between 9\a.m. and 4\p.m., Monday through Friday.

III. INTRODUCTION

Process validation is a requirement of the Current Good Manufacturing Practices Regulations for Finished Pharmaceuticals, 21 CFR Parts 210 and 211, and of the Good Manufacturing Practice Regulations for Medical Devices, 21 CFR Part 820, and therefore, is applicable to the manufacture of pharamaceuticals and medical devices.

Several firms have asked FDA for specific guidance on what FDA expects firms to do to assure compliance with the requirements for process validation. This guideline discusses process validation elements and concepts that are considered by FDA as acceptable parts of a validation program. The constituents of validation presented in this document are not intended to be all-inclusive. FDA recognizes that, because of the great variety of medical products (drug products and medical devices), processes and manufacturing facilities, it is not possible to state in one document all of the specific validation elements that are applicable. Several broad concepts, however, have general applicability which manufacturers can use successfully as a guide in validating a manufacturing process. Although the particular requirements of process validation will vary according to such factors as the nature of the medical product (e.g., sterile vs non-sterile) and the complexity of the process, the broad concepts stated in this document have general applicability and provide an acceptable framework for building a comprehensive approach to process validation.

Definitions

Installation qualification - Establishing confidence that process equipment and ancillary systems are capable of consistently operating within established limits and tolerances.

Process performance qualification - Establishing confidence that the process is effective and reproducible.

Product performance qualification - Establishing confidence through appropriate testing that the finished product produced by a specified process meets all release requirements for functionality and safety.

Prospective validation - V alidation conducted prior to the distribution of either a new product, or product made under a revised manufacturing process, where the revisions may affect the product's characteristics.

Retrospective validation - V alidation of a process for a product already in distribution based upon accumulated production, testing and control data.

V alidation - Establishing documented evidence which provides a high degree of assurance that a specific process will consistently produce a product meeting its pre-determined specifications and quality attributes.

V alidation protocol - A written plan stating how validation will be conducted, including test parameters, product characteristics, production equipment, and decision points on what constitutes acceptable test results.

Worst case - A set of conditions encompassing upper and lower processing limits and circumstances, including those within standard operating procedures, which pose the greatest chance of process or product failure when compared to ideal conditions. Such conditions do not necessarily induce product or process failure.

IV. GENERAL CONCEPTS

Assurance of product quality is derived from careful attention to a number of factors including selection of quality parts and materials, adequate product and process design, control of the process, and in-process and end-product testing. Due to the complexity of today's medical products, routine end-product testing alone often is not sufficient to assure product quality for several reasons. Some end-product tests have limited sensitivity.1 In some cases, destructive testing would be required to show that the manufacturing process was adequate, and in other situations end-product testing does not reveal all variations that may occur in the product that may impact on safety and effectiveness.2

The basic principles of quality assurance have as their goal the production of articles that are fit for their intended use. These

1 For example, USP XXI states: "No sampling plan for applying sterility tests to a specified proportion of discrete units selected from a sterilization load is capable of demonstrating with complete assurance that all of the untested units are in fact sterile."

2 As an example, in one instance a visual inspection failed to detect a defective structural weld which resulted in the failure of an infant warmer. The defect could only have been detected by using destructive testing or expensive test equipment.

principles may be stated as follows:

(1)quality, safety, and effectiveness must be designed and built into the product;

(2)quality cannot be inspected or tested into the finished product;

(3)and (3) each step of the manufacturing process must be controlled to maximize the probability that

the finished product meets all quality and design specifications. Process validation is a key

element in assuring that these quality assurance goals are met.

It is through careful design and validation of both the process and process controls that a manufacturer can establish a high degree of confidence that all manufactured units from successive lots will be acceptable. Successfully validating a process may reduce the dependence upon intensive in-process and finished product testing.

It should be noted that in most all cases, end-product testing plays a major role in assuring that quality assurance goals are met; i.e., validation and end-product testing are not mutually exclusive.

The FDA defines process validation as follows:

Process validation is establishing documented evidence which provides a high degree of assurance that a specific process will consistently produce a product meeting its pre-determined specifications and quality characteristics.

It is important that the manufacturer prepare a written validation protocol which specifies the procedures (and tests) to be conducted and the data to be collected. The purpose for which data are collected must be clear, the data must reflect facts and be collected carefully and accurately. The protocol should specify a

sufficient number of replicate process runs to demonstrate reproducibility and provide an accurate measure of variability among successive runs. The test conditions for these runs should encompass upper and lower processing limits and circumstances, including those within standard operating procedures, which pose the greatest chance of process or product failure compared to ideal conditions; such conditions have become widely known as "worst case" conditions. (They are sometimes called "most appropriate challenge" conditions.) V alidation documentation should include evidence of the suitability of materials and the performance and reliability of equipment and systems.

Key process variables should be monitored and documented. Analysis of the data collected from monitoring will establish the variability of process parameters for individual runs and will establish whether or not the equipment and process controls are adequate to assure that product specifications are met.

Finished product and in-process test data can be of value in process validation, particularly in those situations where quality attributes and variabilities can be readily measured. Where finished (or in-process) testing cannot adequately measure certain attributes, process validation should be derived primarily from qualification of each system used in production and from consideration of the interaction of the various systems.

V. CGMP REGULA TIONS FOR FINISHED PHARMACEUTICALS

Process validation is required, in both general and specific terms, by the Current Good Manufacturing Practice Regulations for Finished Pharmaceuticals, 21 CFR Parts 210 and 211. Examples of such requirements are listed below for informational purposes, and are not all-inc lusive.

A requirement for process validation is set forth in general terms in section\211.100 -- Written procedures; deviations -- which states, in part:

"There shall be written procedures for production and process control designed to assure that the drug products have the identity, strength, quality, and purity they purport or are represented to possess."

Several sections of the CGMP regulations state validation requirements in more specific terms. Excerpts from some of these sections are:

Section 211.110, Sampling and testing of in-process materials and drug products.

(a) "....control procedures shall be established to monitor the output and V ALIDA TE the performance of those manufacturing processes that may be responsible for causing variability in the characteristics of in-process material and the drug product." (emphasis added) Section 211.113, Control of Microbiological Contamination.

(b) "Appropriate written procedures, designed to prevent microbiological contamination of drug products purporting to besterile, shall be established and followed. Such proceduresshall include V ALIDA TION of any sterilization process."(emphasis added)

VI. GMP REGULA TION FOR MEDICAL DEVICES

Process validation is required by the medical device GMP Regulations, 21 CFR Part\820. Section 820.5 requires every finished device manufacturer to:

"...prepare and implement a quality assurance program that is appropriate to the specific device

manufactured..."

Section 820.3(n) defines quality assurance as:

"...all activities necessary to verify confidence in the quality of the process used to manufacture a finished device."

When applicable to a specific process, process validation is an essential element in establishing confidence that a process will consistently produce a product meeting the designed quality characteristics.

A generally stated requirement for process validation is contained in section\820.100:

"Written manufacturing specifications and processing procedures shall be established, implemented, and controlled to assure that the device conforms to its original design or any approved changes in that design."

V alidation is an essential element in the establishment and implementation of a process procedure, as well as in determining what process controls are required in order to assure conformance to specifications.

Section 820.100(a)(1) states:

"...control measures shall be established to assure that the design basis for the device, components and packaging is correctly translated into approved specifications."

V alidation is an essential control for assuring that the specifications for the device and manufacturing process are adequate to produce a device that will conform to the approveddesign characteristics.

VII. PRELIMINARY CONSIDERA TIONS

A manufacturer should evaluate all factors that affect product quality when designing and undertaking a process validation study. These factors may vary considerably among different products and manufacturing technologies and could include, for example, component specifications, air and water handling systems, environmental controls, equipment functions, and process control operations. No single approach to process validation will be appropriate and complete in all cases; however, the following quality activities should be undertaken in most situations.

During the research and development (R&D) phase, the desired product should be carefully defined in terms of its characteristics, such as physical, chemical, electrical and performance characteristics.3 It is important to translate the product characteristics into specifications as a basis for description and control of the product.

Documentation of changes made during development provide traceability which can later be used to pinpoint solutions to future problems.

The product's end use should be a determining factor in the development of product (and component) characteristics and specifications. All pertinent aspects of the product which impact on safety and effectiveness should be considered. These aspects

For example, in the case of a compressed tablet, physical characteristics would include size, weight, hardness, and freedom from defects, such as capping and splitting. Chemical characteristics would include quantitative formulation/potency; performance characteristics may include bioavailability (reflected by

disintegration and dissolution). In the case of blood tubing, physical attributes would include internal and external diameters, length and color. Chemical characteristics would include raw material formulation. Mechanical properties would include hardness and tensile strength; performance characteristics would include biocompatibility and durability.

include performance, reliability and stability. Acceptable ranges or limits should be established for each characteristic to set up allowable variations.4 These ranges should be expressed in readily measurable terms.

The validity of acceptance specifications should be verified through testing and challenge of the product on a sound scientific basis during the initial development and production phase.

Once a specification is demonstrated as acceptable it is important that any changes to the specification be made in accordance with documented change control procedures.

VIII. ELEMENTS OF PROCESS V ALIDA TION

A. Prospective V alidation

Prospective validation includes those considerations that should be made before an entirely new product is introduced by a firm or when there is a change in the manufacturing process which may affect the product's characteristics, such as uniformity and identity. The following are considered as key elements of prospective validation.

4 For example, in order to assure that an oral, ophthalmic, or parenteral solution has an acceptable pH, a specification may be established by which a lot is released only if it has been shown to have a pH within a narrow established range. For a device, a specification for the electrical resistance of a pacemaker lead would be established so that the lead would be acceptable only if the resistance was within a specified range.

1. Equipment and Process

The equipment and process(es) should be designed and/or selected so that product specifications are consistently achieved. This should be done with the participation of all appropriate groups that are concerned with assuring a quality product, e.g., engineering design, production operations, and quality assurance personnel.

a. Equipment: Installation Qualification Installation qualification studies establish confidence that the process equipment and ancillary systems are capable of consistently operating within established limits and tolerances. After process equipment is designed or selected, it should be evaluated and tested to verify that it is capable of operating satisfactorily within the operating limits required by the process.5 This phase of validation includes examination of equipment design; determination of calibration, maintenance, and adjustment requirements; and identifying critical equipment features that could affect the process and product. Information obtained from these studies should be used to establish written procedures covering equipment calibration, maintenance, monitoring, and control.

5 Examples of equipment performance characteristics which may be measured include temperature and pressure of injection molding machines, uniformity of speed for mixers, temperature, speed and pressure for packaging machines, and temperature and pressure of sterilization chambers.

In assessing the suitability of a given piece of equipment, it is usually insufficient to rely solely upon the representations of the equipment supplier, or upon experience in producing some other product.6 Sound theoretical and practical engineering principles and considerations are a first step in the assessment.

It is important that equipment qualification simulate actual production conditions, including those which are "worst case" situations.

6 The importance of assessing equipment suitability based upon how it will be used to attain desired product attributes is illustrated in the case of deionizers used to produce Purified Water, USP. In one case, a firm used such water to make a topical drug product solution which, in view of its intended use, should have been free from objectionable microorganisms. However, the product was found to be contaminated with a pathogenic microorganism. The apparent cause of the problem was failure to assess the performance of the deionizer from a microbiological standpoint. It is fairly well recognized that the deionizers are prone to build-up of microorganisms--especially if the flow rates are low and the deionizers are not recharged and sanitized at suitable intervals. Therefore, these factors should have been considered. In this case, however, the firm relied upon the representations of the equipment itself, namely the "recharge" (i.e., conductivity) indicator, to signal the time for regeneration and cleaning. Considering the desired product characteristics, the firm should have determined the need for such procedures based upon pre-use testing, taking into account such factors as the length of time the equipment could produce deionized water of acceptable quality, flow rate, temperature, raw water quality, frequency of use, and surface area of deionizing resins.

Tests and challenges should be repeated a sufficient number of times to assure reliable and meaningful results. All acceptance criteria must be met during the test or challenge. If any test or challenge shows that the equipment does not perform within its specifications, an evaluation should be performed to identify the cause of the failure. Corrections should be made and additional test runs performed, as needed, to verify that the equipment performs within specifications. The observed variability of the equipment between and within runs can be used as a basis for determining the total number of trials selected for the subsequent performance qualification studies of the process.7

Once the equipment configuration and performance characteristics are established and qualified, they should be documented. The installation qualification should include a review of pertinent maintenance procedures, repair parts lists, and calibration methods for each piece of equipment. The objective is to assure that all repairs can be performed in such a way that will not affect the

7 For example, the AAMI Guideline for Industrial Ethylene Oxide Sterilization of Medical Devices approved 2 December 1981, states: "The performance qualification should include a minimum of 3 successful, planned qualification runs, in which all of the acceptance criteria are met.....(5.3.1.2.).

characteristics of material processed after the repair. In addition, special post-repair cleaning and calibration requirements should be developed to prevent inadvertent manufacture a of non-conforming product. Planning during the qualification phase can prevent confusion during emergency repairs which could lead to use of the wrong replacement part.

b. Process: Performance Qualification The purpose of performance qualification is to provide rigorous testing to demonstrate the effectiveness and reproducibility of the process. In entering the performance

qualification phase of validation, it is understood that the process specifications have been established and essentially proven acceptable through laboratory or other trial methods and that the equipment has been judged acceptable on the basis of suitable installation studies.

Each process should be defined and described with sufficient specificity so that employees understand what is required.

Parts of the process which may vary so as to affect important product quality should be challenged.8

In challenging a process to assess its adequacy, it is important that challenge conditions simulate those that will be encountered during actual production, including "worst case" conditions. The challenges should be repeated enough times to assure that the results are meaningful and consistent.

8 For example, in electroplating the metal case of an implantable pacemaker, the significant process steps to define, describe, and challenge include establishment and control of current density and temperature values for assuring adequate composition of electrolyte and for assuring cleanliness of the metal to be plated. In the production of parenteral solutions by aseptic filling, the significant aseptic filling process steps to define and challenge should include the sterilization and depyrogenation of containers/closures, sterilization of solutions, filling equipment and product contact surfaces, and the filling and closing of containers.

Each specific manufacturing process should be appropriately qualified and validated. There is an inherent danger in relying on what are perceived to be similarities between products, processes, and equipment without appropriate challenge.9

c. Product: Performance Qualification For purposes of this guideline, product performance qualification activities apply only to medical devices. These steps should be viewed as pre-production quality assurance activities.

9 For example, in the production of a compressed tablet, a firm may switch from one type of granulation blender to another with the erroneous assumption that both types have similar performance characteristics, and, therefore, granulation mixing times and procedures need not be altered. However, if the blenders are substantially different, use of the new blender with procedures used for the previous blender may result in a granulation with poor content uniformity. This, in turn, may lead to tablets having significantly differing potencies. This situation may be averted if the quality assurance system detects the equipment change in the first place, challenges the blender performance, precipitates a revalidation of the process, and initiates appropriate changes. In this example, revalidation comprises installation qualification of the new equipment and performance qualification of the process intended for use in the new blender.

Before reaching the conclusion that a process has been successfully validated, it is necessary to demonstrate that the specified process has not adversely affected the finished product. Where possible, product performance qualification testing should include performance testing under conditions that simulate actual use. Product performance qualification testing should be conducted using product manufactured from the same type of production equipment, methods and procedures that will be used for routine production. Otherwise, the qualified product may not be representative of production units and cannot be used as evidence that the manufacturing process will produce a product that meets the pre-determined specifications and quality

attributes.

10 For example, a manufacturer of heart valves received complaints that the valve-support structure was fracturing under use. Investigation by the manufacturer revealed that all material and dimensional specifications had been met but the production machining process created microscopic scratches on the valve supporting wireform. These scratches caused metal fatigue and subsequent fracture. Comprehensive fatigue testing of production units under simulated use conditions could have detec ted the process deficiency.

In another example, a manufacturer recalled insulin syringes because of complaints that the needles were clogged. Investigation revealed that the needles were clogged by silicone oil which was employed as a lubricant during manufacturing. Investigation further revealed that the method used to extract the silicone oil was only partially effective. Although visual inspection of the syringes seemed to support that the cleaning method was effective, actual use proved otherwise.

After actual production units have sucessfully passed product performance qualification, a formal technical review should be conducted and should include:

Comparison of the approved product specifications and the actual qualified product.

Determination of the validity of test methods used to determine compliance with the approved specifications.

Determination of the adequacy of the specification change control program.

2. System to Assure Timely Revalidation There should be a quality assurance system in place which requires revalidation whenever there are changes in packaging, formulation, equipment, or processes which could impact on product effectiveness or product characteristics, and whenever there are changes in product characteristics. Furthermore, when a change is made in raw material supplier, the manufacturer should consider subtle, potentially adverse differences in the raw material characteristics. A determination of adverse differences in raw material indicates a need to revalidate the process.

One way of detecting the kind of changes that should initiate revalidation is the use of tests and methods of analysis which are capable of measuring characteristics which may vary. Such tests and methods usually yield specific results which go beyond the mere pass/fail basis, thereby detecting variations within product and process specifications and allowing determination of whether a process is slipping out of control.

The quality assurance procedures should establish the circumstances under which revalidation is required. These may be based upon equipment, process, and product performance observed during the initial validation challenge studies. It is desirable to designate individuals who have the responsibility to review product, process, equipment and personnel changes to determine if and when revalidation is warranted.

The extent of revalidation will depend upon the nature of the changes and how they impact upon different aspects of production that had previously been validated. It may not be necessary to revalidate a process from scratch merely because a given circumstance has changed. However, it is important to carefully assess the nature of the change to determine potential ripple effects and what needs to be considered as part of revalidation.

3. Documentation It is essential that the validation program is documented and that the documentation is properly maintained. Approval and release of the process for use in routine manufacturing should be based upon a review of all the validation documentation, including data from the equipment qualification, process performance qualification, and product/package testing to ensure compatibility with the process.

For routine production, it is important to adequately record process details (e.g., time, temperature, equipment used) and to record any changes which have occurred. A maintenance log can be useful in performing failure investigations concerning a specific manufacturing lot. V alidation data (along with specific test data) may also determine expected variance in product or equipment characteristics.

B. Retrospective Process V alidation In some cases a product may have been on the market without sufficient premarket process validation. In these cases, it may be possible to validate, in some measure, the adequacy of the process by examination of accumulated test data on the product and records of the manufacturing procedures used.

Retrospective validation can also be useful to augment initial premarket prospective validation for new products or changed processes. In such cases, preliminary prospective validation should have been sufficient to warrant product marketing. As additional data is gathered on production lots, such data can be used to build confidence in the adequacy of the process.Conversely, such data may indicate a declining confidence in the process and a commensurate need for corrective changes.

Test data may be useful only if the methods and results are adequately specific. As with prospective validation, it may be insufficient to assess the process solely on the basis of lot by lot conformance to specifications if test results are merely expressed in terms of pass/fail. Specific results, on the other hand, can be statistically analyzed and a determination can be made of what variance in data can be expected. It is important to maintain records which describe the operating characteristics of the process, e.g., time, temperature, humidity, and equipment settings.11 Whenever test data are used to demonstrate conformance to specifications, it is important that the test methodology be qualified to assure that test results are objective and accurate.

11 For example, sterilizer time and temperature data collected on recording equipment found to be accurate and precise could establish that process parameters had been reliably delivered to previously processed loads.

A retrospective qualification of the equipment could be performed to demonstrate that the recorded data represented conditions that were uniform throughout the chamber and that product load configurations, personnel practices, initial temperature, and other variables had been adequately controlled during the earlier runs.

IX. ACCEPTABILITY OF PRODUCT TESTING

In some cases, a drug product or medical device may be manufactured individually or on a one-time basis. The concept of prospective or retrospective validation as it relates to those situations may have limited applicability, and data obtained during the manufacturing and assembly process may be used in conjunction with product testing to demonstrate that the instant run yielded a finished product meeting all of its specifications and quality characteristics. Such evaluation of data and product testing would be expected to be much more extensive than the usual situation where more reliance would be placed on prospective

validation.

copyright 1995 by Jason Kaye and Craig Brozefsky

美国FDA分析方法验证指南中英文对照

I. INTRODUCTION This guidance provides recommendations to applicants on submitting analytical procedures, validation data, and samples to support the documentation of the identity, strength, quality, purity, and potency of drug substances and drug products. 1. 绪论 本指南旨在为申请者提供建议,以帮助其提交分析方法,方法验证资料和样品用于支持原料药和制剂的认定,剂量,质量,纯度和效力方面的文件。 This guidance is intended to assist applicants in assembling information, submitting samples, and presenting data to support analytical methodologies. The recommendations apply to drug substances and drug products covered in new drug applications (NDAs), abbreviated new drug applications (ANDAs), biologics license applications (BLAs), product license applications (PLAs), and supplements to these applications. 本指南旨在帮助申请者收集资料,递交样品并资料以支持分析方法。这些建议适用于NDA,ANDA,BLA,PLA及其它们的补充中所涉及的原料药和制剂。 The principles also apply to drug substances and drug products covered in Type II drug master files (DMFs). If a different approach is chosen, the applicant is encouraged to discuss the matter in advance with the center with product jurisdiction to prevent the expenditure of resources on preparing a submission that may later be determined to be unacceptable. 这些原则同样适用于二类DMF所涉及的原料药和制剂。如果使用了其它方法,鼓励申请者事先和FDA药品评审中心的官员进行讨论,以免出现这种情况,那就是花了人力物力所准备起来的递交资料后来发现是不可用的。 The principles of methods validation described in this guidance apply to all types of analytical procedures. However, the specific recommendations in this guidance may not be applicable to certain unique analytical procedures for products such as biological, biotechnological, botanical, or radiopharmaceutical drugs. 本指南中所述的分析方法验证的原则适用于各种类型的分析方法。但是,本指南中特定的建议可能不适用于有些产品所用的特殊分析方法,如生物药,生物技术药,植物药或放射性药物等。 For example, many bioassays are based on animal challenge models, 39 immunogenicity assessments, or other immunoassays that have unique features that should be considered when submitting analytical procedure and methods validation information. 比如说,许多生物分析是建立在动物挑战模式,免疫原性评估或其它有着独特特性的免疫分析基础上的,在递交分析方法和分析方法验证资料时需考虑这些独特的性质。Furthermore, specific recommendations for biological and immunochemical tests that may be necessary for characterization and quality control of many drug substances and drug products are beyond the scope of this guidance document. 而且,许多原料药和制剂的界定和质量控制所需的生物和免疫化学检测并不在本指南的范围之内。 Although this guidance does not specifically address the submission of analytical procedures and validation data for raw materials, intermediates, excipients, container closure components, and other materials used in the production of drug

201507fda行业指南:分析方法验证(中英文)(下).doc

201507 FDA行业指南:分析方法验证(中英文)(下) VII. STATISTICAL ANALYSIS AND MODELS 统计学分析和模型 A. Statistics 统计学 Statistical analysis of validation data can be used to evaluate validation characteristics against predetermined acceptance criteria. All statistical procedures and parameters used in the analysis of the data should be based on sound principles and appropriate for the intended evaluation. Several statistical methods are useful for assessing validation characteristics, for example, an analysis of variance (ANOVA) to assess regression analysis R (correlation coefficient) and R squared (coefficient of determination) or linear regression to measure linearity. Many statistical methods used for assessing validation characteristics rely on population normality, and it is important to determine whether or not to reject this assumption. There are many techniques, such as histograms, normality tests, and probability plots that can be used to evaluate the observed distribution. It may be appropriate to transform the data to better fit the normal distribution or apply distribution-free (nonparametric) approaches when the observed data are

(食品安全管理)FDA清洗验证检查指南

FDA清洗验证检查指南 I.引言 自FDA各种文件(包括化学原料药检查指南、生物技术检查指南)首次提出这个问题之后,清洗过程的验证已经引发了很多讨论。FDA的文件明确指出要求对清洗过程进行验证。 本指南讨论了各种可接受(或不可接受)的验证方法,从而使FDA的检查具有一致性。但必须清楚地认识到:与其他工艺验证一样,清洗验证方法也不止一种。所有过程验证的检查标准是:检查其科学数据能否证明系统稳定一致地达到预期目的,系统结果稳定地符合预先制定的标准。 本指南仅适用于设备化学残留物的清洗验证。 II.背景 对于FDA而言,使用设备前进行清洗不是什么新要求。1963年GMP法(133.4)规定“设备应处于清洁、有序的状态”。1978年的cGMP中规定了设备清洗的章节(211.67)。要求清洗设备的主要目的还是防止污染或混料。由于设备清洗维护不当或防尘管理不当,FDA检查官曾十分注意检查卫生状况。过去FDA总是更注意检查青霉素类与非青霉素类药物之间的交叉污染、药品与甾类物质或激素之间的交叉污染问题。在过去二十年间,因实际或潜在的青霉素交叉污染问题已从市场上撤回了大量的药品。 另一个事件使FDA对交叉污染问题日益重视,即1988年从市场上撤回了消胆胺成品制剂,原因是规程不当。生产该制剂的原料药受到了农业杀虫剂生产中少量中间体和降解物质的污染。造成交叉污染的主要原因使用了回收溶媒。而回收溶媒受到了污染,原因是对溶媒桶的重复使用缺少监控。贮存杀虫剂产生的回收溶媒桶又重复地用于贮存该药品生产中的回收溶媒。而工厂没有对这些溶媒桶进行有效的监控,没有对其中的溶媒进行有效的检验,也没有对桶的清洗规程进行验证。 被杀虫剂污染的部份化学原料药运到了另一地点的第二家工厂生产制剂,使该工厂的流化床干燥器中物料袋受到了杀虫剂的污染,料造成各批产品受到污染,而该工厂根本就没有生产杀虫剂。

美国FDA分析方法验证指南中英文对照

美国FDA分析方法验证指南中英文对照 美国FDA分析方法验证指南中英文对 照 I. INTRODUCTION This guidance provides recommendations to applicants on submitting analytical procedures, validation data, and samples to support the documentation of the identity, strength, quality, purity, and potency of drug substances and drug products. 1. 绪论 本指南旨在为申请者提供建议,以帮助其提交分析方法,方法验证资料和样品用 于支持原料药和制剂的认定,剂量,质量,纯度和效力方面的文件。 This guidance is intended to assist applicants in assembling information, submitting samples, and presenting data to support analytical methodologies. The recommendations apply to drug substances and drug products covered in new drug applications (NDAs), abbreviated new drug applications (ANDAs), biologics license applications (BLAs), product license applications (PLAs), and supplements to these applications. 本指南旨在帮助申请者收集资料,递交样品并资料以支持分析方法。这些建议适 用于NDA,ANDA,BLA,PLA及其它们的补充中所涉及的原料药和制剂。 The principles also apply to drug substances and drug products covered in Type II drug master files (DMFs). If a different approach is chosen, the applicant is encouraged to discuss the matter in advance with

美国FDA药物分析程序及方法验证指导原则(中文版)

药品及生物制品的分析方法和方法验证指导原则 目录 1.介绍...................... (1) 2.背景..................... .. (2) 3.分析方法开发. ..................... . (3) 4.分析程序内容.............................................. ......... ..................................... .. 3 A.原则/范围 (4) B.仪器/设备............................................. . (4) C.操作参数.............................................. .. (4) D.试剂/标准............................................. . (4) E.样品制备.............................................. .. (4) F.标准对照品溶液的制备............................................ .. (5) G.步骤......... ....................................... (5) H.系统适应性..... (5) I.计算 (5) J.数据报告 (5) 5.参考标准和教材............................................ (6) 6分析方法验证用于新药,仿制药,生物制品和DMF (6) A.非药典分析方法............................................. (6) B.验证特征 (7) C.药典分析方法............................................. .. (8) 7.统计分析和模型 (8) A.统计 (8) B.模型 (8) 8.生命周期管理分析程序 (9) A.重新验证 (9) B.分析方法的可比性研究............................................ . (10) 1.另一种分析方法............................................... .. (10) 2.分析方法转移的研究 (11) C.报告上市后变更已批准的新药,仿制药,或生物制品 (11) 9.美国FDA方法验证............................................... . (12) 10.参考文献

美国FDA分析方法验证指南中文译稿[1]

1 II. 背景 (2) III. 分析方法的类型 (3) A. 法定分析方法 (3) B. 可选择分析方法 (3) 3 C. 稳定性指示分析 (3) IV. 对照品…………………………………………………………………………… 4 A. 对照品的类型 (4) B. 分析报告单 (4) C. 对照品的界定 (4) V. IND 中的分析方法验证 (6) VI. NDA, ANDA, BLA 和PLA 中分析方法验证的内容和格式 (6) A. 原则 (6) B. 取样 (7) C. 仪器和仪器参数 (7) D. 试剂 (7) E. 系统适应性实验 (7) F. 对照品的制备 (7) G. 样品的制备 (8) H. 分析方法 (8) L. 计算 (8) J. 结果报告 (8) VII. NDA,ANDA,BLA 和PLA 中的分析方法验证 (9) A.非法定分析方法 (9) 1.验证项目 (9) 2. 其它分析方法验证信息 (10) a. 耐用性 (11) b. 强降解实验 (11) c. 仪器输出/原始资料 (11) 3.各类检测的建议验证项目 (13) B.法定分析方法 (15) VIII. 统计分析………………………………………………………………………… 15 A. 总则 (15)

C. 统计 (16) IX. 再验证 (16) X. 分析方法验证技术包:内容和过程…………………………………………… 17 A. 分析方法验证技术包 (17) B. 样品的选择和运输 (18) C. 各方责任 (19) XI. 方法……………………………………………………………………………… 20 A. 高效液相色谱(HPLC) (20) B. 气相色谱(GC) (22) C. 分光光度法,光谱学,光谱法和相关的物理方法 (23) D. 毛细管电泳 (23) E. 旋光度 (24) F. 粒径相关的分析方法 (25) G. 溶出度 (26) H. 其它仪器分析方法 (27) 附件A:NDA,ANDA,BLA 和PLA 申请的内容 (28) 附件B:分析方法验证的问题和延误 (29) 参考文献…………………………………………………………………………………… 30 术语表……………………………………………………………………………………… 32 This guidance provides recommendations to applicants on submitting analytical procedures, validation data, and samples to support the documentation of the identity, strength, quality, purity, and potency of drug substances and drug products. 1. 绪论 本指南旨在为申请者提供建议,以帮助其提交分析方法,方法验证资料和样品用于支持 原料药和制剂的认定,剂量,质量,纯度和效力方面的文件。 This guidance is intended to assist applicants in assembling information, submitting samples, and presenting data to support analytical methodologies. The recommendations apply to drug substances and drug products covered in new drug applications (NDAs), abbreviated new drug applications (ANDAs), biologics license applications (BLAs), product license applications (PLAs), and supplements to these applications.

2011版FDA最新工艺验证指南

FDA工艺验证指南新旧版透彻比较解读 【整理者提醒】 1-左侧文本为2011年1月最新修订版本,右侧文本为2008年11月草案版本。 2-蓝色文本为修订后文本或者新增加文本。 3-下划线文本是比旧版本增加的部分内容。 4-删除线文本表示该部分存在于旧版本中,在新版本中删除。 5-注释前面加【注释】2字注明。 6-Zhulikou431关于FDA2008年11月草案彻底解读版本可以在丁香园论坛搜索到,欢迎下载阅读、讨论。 7-不得用于商业用途,转载请注明丁香园信息。 8-增加了新旧版本的中文译文。 9-欢迎各位朋友提出宝贵建议,联系邮箱zhulikou431@https://www.360docs.net/doc/588996141.html,. Guidance for Industry Process Validation: General Principles and Practices Final Version January 2011 Draft 2008 I. INTRODUCTION I. INTRODUCTION 简介

This guidance outlines the general principles and approaches that FDA considers appropriate elements of process validation for the manufacture of human and animal drug and biological products, including active pharmaceutical ingredients (APIs or drug substances), collectively referred to in this guidance as drugs or products. This guidance incorporates principles and approaches that all manufacturers can use to validate manufacturing processes. 本指南概括了一般的原则与方法,这些原则与方法是FDA 认为进行工艺验证的恰当要素,这些工艺被用于生产人用药、动物用药以及生物制品,包括活性药物成分(API 或药用物质),在本指南中以上统称为药品或产品。本指南整合了一般的原则和方法,所有的生产企业都可以将这些原则和方法应用于生产工艺的验证。 This guidance outlines the general principles and approaches that FDA considers to be appropriate elements of process validation for the manufacture of human and animal drug and biological products, including active pharmaceutical ingredients (API or drug substance), collectively referred to in this guidance as drugs or products. This guidance incorporates principles and approaches that all manufacturers can use in validating a manufacturing process. 本指南概括了一般的原则与方法,这些原则与方法是FDA 认为进行工艺验证的恰当要素,这些工艺被用于生产人用药、动物用药以及生物制品,包括活性药物成分(API 或药用物质),在本指南中以上统称为药品或产品。本指南整合了一般的原则和方法,所有的生产企业都可以将这些原则和方法应用于生产工艺的验证。 This guidance aligns process validation activities with a product lifecycle concept and with existing FDA guidance, including the FDA/International Conference on Harmonisation (ICH) guidances for industry, Q8(R2) Pharmaceutical Development, Q9 Quality Risk Management, and Q10 Pharmaceutical Quality System.2 Although this guidance does not repeat the concepts and principles explained in those guidances, FDA encourages the use of modern pharmaceutical development concepts, quality risk man- agement, and quality systems at all stages of the manufacturing process lifecycle. 本指南将工艺验证活动与产品生命周期概念以及现有FDA 相关指南进行了协调;这些存在的FDA指南包括FDA/ICH指南,Q8(R2)药品研发指南、Q9质量风险管理指南和Q10制药质量体系指南。尽管本指南没有重复上述指南解释的概念和This guidance aligns process validation activities with the product lifecycle concept and with existing FDA guidance.2 2 See the FDA/International Conference on Harmonisation (ICH) guidances for industry: Q8 Pharmaceutical Development, Q9 Quality Risk Management, and when finalized, Q10 Pharmaceutical Quality System (a notice of availability for the May 2007 ICH draft guidance, Q10 Pharmaceutical Quality System, published in the Federal Register on July 13, 2007 (72 FR 38604)). We update guidance documents periodically. To make sure you have the most recent version of a guidance, check the CDER guidance page at https://www.360docs.net/doc/588996141.html,/cder/guidance/index.htm, the CBER guidance page at

FDA分析方法验证指南

(中英对照)美国FDA 分析方法验证指南

目 录 1.绪论 (4) II.背景 (5) III.分析方法的类型 (7) A. 法定分析方法 (7) B. 替代分析方法 (7) C. 稳定性指示分析 (8) IV 标准品 (8) A.标准品的类型 (8) B.分析报告单 (9) C.标准品的界定 (9) V.IND中的分析方法验证 (11) VI.NDA,ANDA,BLA和PLA中分析方法的内容和格式 (12) A.基本方法 (12) B.取样 (12) C.仪器和仪器参数 (12) D.试剂 (13) E.系统适应性实验 (13) F.标准品的制备 (14) H.操作过程 (14) J.计算 (14) K.结果报告 (14) VII.NDA,ANDA,BLA和PLA中的分析方法验证 (16) A.非药典分析方法 (16) 1)验证项目 (16) 2)其它分析方法验证信息 (17) a.耐用性 (18) b.强降解实验 (19) c.仪器输出/原始资料 (19)

3)各类检测的推荐验证项目 (21) B.药典分析方法 (24) VIII.统计分析 (25) A.基本原则 (25) B.对比研究 (25) C.统计 (25) IX.再验证 (26) X.分析方法验证资料:内容和数据处理 (27) A.分析方法验证资料 (27) B.样品的选择和运输 (29) C.各方职责 (30) XI.方法学 (32) A.高效液相色谱(HPLC) (32) B.气相色谱(GC) (35) C.分光光度法,光谱法和相关的物理方法 (37) D.毛细管电泳(CE) (37) E.旋光度 (39) F.粒径分析相关的分析方法 (40) G.溶出度 (41) H.其它仪器分析方法 (43) 附录A NDA, ANDA, BLA 和PLA申请的内容 (44) 附录B 析方法验证的问题和延误 (45) 参考文献 (46) 术语表 (48)

FDA指南中文版

目录表 I. 导言 (1) II. 背景 (2) III. 分析方法的类型 (3) A. 法定分析方法 (3) B. 可选择分析方法 (3) 3 C. 稳定性指示分析 (3) IV. 对照品 (4) A. 对照品的类型 (4) B. 分析报告单 (4) C. 对照品的界定 (4) V. IND中的分析方法验证 (6) VI. NDA, ANDA, BLA 和PLA中分析方法验证的内容和格式 (6) A. 原则 (6) B. 取样 (7) C. 仪器和仪器参数 (7) D. 试剂 (7) E. 系统适应性实验 (7) F. 对照品的制备 (7) G. 样品的制备 (8) H. 分析方法 (8) L. 计算 (8) J. 结果报告 (8) VII. NDA,ANDA,BLA和PLA中的分析方法验证 (9) A. 非法定分析方法 (9) 1.验证项目 (9) 2. 其它分析方法验证信息 (10) a. 耐用性 (11)

b. 强降解实验 (11) c. 仪器输出/原始资料 (11) 3.各类检测的建议验证项目 (13) B. 法定分析方法 (15) VIII. 统计分析 (15) A. 总则 (15) B. 比较研究 (16) C. 统计 (16) IX. 再验证 (16) X. 分析方法验证技术包:内容和过程 (17) A. 分析方法验证技术包 (17) B. 样品的选择和运输 (18) C. 各方责任 (19) XI. 方法 (20) A. 高效液相色谱(HPLC) (20) B. 气相色谱(GC) (22) C. 分光光度法,光谱学,光谱法和相关的物理方法 (23) D. 毛细管电泳 (23) E. 旋光度 (24) F. 粒径相关的分析方法 (25) G. 溶出度 (26) H. 其它仪器分析方法 (27) 附件 A:NDA,ANDA,BLA和PLA申请的内容 (28) 附件 B:分析方法验证的问题和延误 (29) 参考文献 (30) 术语表 (32) I. INTRODUCTION This guidance provides recommendations to applicants on submitting analytical

色谱方法验证指南-FDA

色谱方法验证审评指南 色谱方法通常用于原料、药物、药物制剂和生物体液中化合物的定性和定量。涉及的成分包括手性的或非手性的药物、过程杂质、残留溶媒、附加剂如防腐剂、分解产物、从容器和密闭包装或制造过程中带入的可提取和可过滤的杂质、植物药中的农药和代谢物等。 试验方法的目的是得到可信赖的和准确的数据,无论是用于验收、出厂、稳定性或药物动力学研究。得到的数据用于药品开发或批准后的定性和定量,试验包括原料的验收、药物和药物制剂的出厂、过程检验(In- process testing)的质量保证和失效期的建立。 方法的验证是由药品的开发者或使用者来检验其方法是否达到预期的可靠性、准确度和精密度的过程。得到的数据成为方法的验证资料的一部分交给CDER.。 方法的验证对于完成机构满足档案要求不是一次性的,开发者和使用者都应验证其方法的耐用度或耐久性(ruggedness or robustness.),其他的分析者、用其它相当的仪器,在其它的日期或地点,在药品生产期限(有效期)全过程,方法都应能够重现。如果产生数据的方法是可靠的,那么所得到的验收、出厂、稳定性或药物动力学的数据就是可信赖的。验证的过程和方法的设计应在开发过程中重要的数据产生之前,如果方法改变了,还应该再验证。 . 色谱类型 色谱是一种技术,通过该技术,样品中的组分载入液相或气相中,通过在固定相上由吸附—解吸附来完成。 A. 高效液相色谱 (HPLC) HPLC分离是基于在样品在流动相液体和固定相之间的不同分配。一般地说HPLC大体分为以下几种(未考虑其重要性顺序) 1. 手性液相色谱 2. 离子交换色谱 3. 离子对/亲和色谱 4. 正相色谱 5. 反相色谱 6. 分子排阻色谱 1. 手性液相色谱 分离光学异构体可在手性固定相上,用衍生化试剂或在非手性固定相上用流动相添加剂形成非对对映体来实现。用作杂质试验方法时,如果光学异构体杂质在光学异构体药物之前洗脱,要增加灵敏度。 2. 离子交换色谱 分离基于荷电功能团,样品负离子(X - )为阴离子,样品正离子((X + )为阳离子,一般用pH程序洗脱。 3. 离子对/亲和色谱 分离基于与目标样品的专一的化学相互作用。更普遍的反相型用缓冲液和加入的对离子(与被分离的样品荷相反电荷)分离。分离受pH、离子强度、温度、浓度和共存的有机溶剂类型的影响。亲和色谱,一般用于大分子,使用配合体(共价结合在固体基质上的生物活性分子),与其同类的抗原(分析介质)反应,生成可逆转的复合物,通过改变缓冲条件洗脱。 4. 正相色谱 正相色谱为用有机溶剂为流动相和极性的固定相。此时较小极性的组分比较大极性组分更快地洗脱。 5. 反相色谱 报给CDER的最通常的实验方法是反相HPLC方法,最通常用紫外检测器。 反相色谱,一种键合相的色谱技术,用水作基本溶剂,选择性也受溶剂强度、柱温和pH的影响,一般来说较大极性比较小极性组分洗脱更快。

FDA药品及生物制品的分析方法和方法验证指导原则草案2014中文

FDA药品及生物制品的分析方法和方法验证指导原则草案 前言 本指导原则草案,定稿后,将代表美国食品和药物管理局(FDA)目前关于这个话题目前的想法。它不会创造或赋予或任何人的任何权利,不约束FDA 或公众。您可以使用另一种方法,如果该方法符合适用的法律和法规的要求。如果你想讨论一个替代方法,请与FDA工作人员负责实施本指南。如果你不能确定适当的FDA 工作人员,请拨打本指南的标题页上所列的电话号码。 介绍: 该修订指南草案将取代行业2000 年的指导分析方法和方法验证草案,并最终确定后,也将取代1987 年美国FDA 行业指南《提交的样品和分析数据的方法验证》。该草案提供了有关申请人如何提交分析程序和方法验证数据来支持说明原料药和制剂具有强度、质量、纯度和效用的文件。它会帮你收集信息和现有数据来支持你的分析方法。该指导原则适用于原料药和制剂产品涵盖新药申请(NDA),简化新药申请(仿制药),生物制品许可申请(BLA),以及这些申请的补充申请。在这个修订草案指导原则也适用于原料药和制剂产品涵盖二类药物主文件(DMFs)。 该修订指南草案补充了国际协调会议(ICH)Q2(R1)指导原则《分析程序的验证:开发和验证的分析方法Q2(R1)和方法的文本。 该修订指南草案不涉及研究性新药申请(IND)方法验证,但研究者在准备研究性新药申请时应考虑该指导原则中的建议。研究性新药申请需要在研究的每个阶段有足够的信息,以确保正确鉴别性,质量,纯度,强度和/或效力。对分析方法和方法验证的信息量将随研究中不同阶段而变化。有关分析程序和需提交的阶段方法验证资料方面的指导意见的研究中,申请者可以参考FDA 的指导原则《Ⅰ期研究药物的IND 的内容和格式,包括性状、疗效和生物技术衍生产品》。 一般考虑在第三阶段的研究进行之前,分析方法和分析方法验证(例如,生物测定)是在FDA 行业指导原则《人类药物和生物制剂、化学、制造、控制信息会议》。 该修订指南草案不涉及生物和免疫化学检测的表征和许多原料药和制剂产 品质量控制的具体方法验证的建议。例如,一些基于动物模型的生物测定,并且免疫原性评估或其它免疫测定具有独特的特征,应开发和验证过程中予以考虑。 此外,需要对现有的分析方法再验证时可能需要在制造过程中产品的生命周期的变化予以考虑。有关适当的验证方法的分析程序或者提交本指南中未提及的信息的问题,您应该向用FDA 产品质量评审人员咨询。 如果您选择了与本指导草案中不同的方式,我们建议您在提交申请前与相应的FDA 产品质量评审人员讨论。 FDA 的指南文件,包括本指导原则,不具有法律强制性的责任。相反,指南描述的是FDA 对某个主题目前的想法,并应仅作为建议,除非有明确的法律或法规要求的引用。使用“应该”这个词在FDA 指南意味着什么建议或推荐,但不是必需的。 II. BACKGROUND背景 每个NDA 和ANDA 都必需包括必要的分析程序,以确保原料药和制剂的鉴别,强度,质量,纯度和效果.每个BLA 必须包括完整的制造方法描述,包括能够确保产品身份、质量、安全、纯度和有效的分析程序。数据必须能够用于建立满足精度和可靠性标准的分析方法并适合与拟定目的.对于BLAs 及补充补充,分析方法和方法验证是许可证申请或补充申请必须提交的一部分,并通过美国FDA质量评审小组进行评估。 分析方法和验证资料应当按照ICH M2 eCTD 的相应部分提交。 当一个分析程序作为NDA,ANDA 或者BLA 的一部分被批准时,它就变成了FDA 获批药品

分析方法验证指导原则-FDA

Guidance for Industry Bioanalytical Method Validation U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Veterinary Medicine (CVM) May 2001 BP

Guidance for Industry Bioanalytical Method Validation Additional copies are available from: Drug Information Branch (HFD-210) Center for Drug Evaluation and Research (CDER) 5600 Fishers Lane, Rockville, MD 20857 (Tel) 301-827-4573 Internet at https://www.360docs.net/doc/588996141.html,/cder/guidance/index.htm or Communications Staff (HFV-12) Center for Veterinary Medicine (CVM) 7500 Standish Place, Rockville, MD 20855 (Tel) 301–594-1755 Internet at https://www.360docs.net/doc/588996141.html,/cvm U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Veterinary Medicine (CVM) May 2001 BP

10美国FDA分析方法验证指南中文译稿

美国FDA分析方法验证指南中文译稿 I. INTRODUCTION This guidance provides recommendations to applicants on submitting analytical procedures, validation data, and samples to support the documentation of the identity, strength, quality, purity, and potency of drug substances and drug products. 1. 绪论 本指南旨在为申请者提供建议,以帮助其提交分析方法,方法验证资料和样品用于支持原料药和制剂的认定,剂量,质量,纯度和效力方面的文件。 This guidance is intended to assist applicants in assembling information, submitting samples, and presentin g data to support analytical methodologies. The recommendations apply to drug substances and drug prod ucts covered in new drug applications (NDAs), abbreviated new drug applications (ANDAs), biologics lic ense applications (BLAs), product license applications (PLAs), and supplements to these applications. 本指南旨在帮助申请者收集资料,递交样品并资料以支持分析方法。这些建议适用于NDA,ANDA,BLA,PLA及其它们的补充中所涉及的原料药和制剂。 The principles also apply to drug substances and drug products covered in Type II drug master files (D MFs). If a different approach is chosen, the applicant is encouraged to discuss the matter in advance wit h the center with product jurisdiction to prevent the expenditure of resources on preparing a submission that may later be determined to be unacceptable. 这些原则同样适用于二类DMF所涉及的原料药和制剂。如果使用了其它方法,鼓励申请者事先和FDA药品评审中心的官员进行讨论,以免出现这种情况,那就是花了人力物力所准备起来的递交资料后来发现是不可用的。 The principles of methods validation described in this guidance apply to all types of analytical procedures. However, the specific recommendations in this guidance may not be applicable to certain unique analytic al procedures for products such as biological, biotechnological, botanical, or radiopharmaceutical drugs. 本指南中所述的分析方法验证的原则适用于各种类型的分析方法。但是,本指南中特定的建议可能不适用于有些产品所用的特殊分析方法,如生物药,生物技术药,植物药或放射性药物等。 For example, many bioassays are based on animal challenge models, 39 immunogenicity assessments, or other immunoassays that have unique features that should be considered when submitting analytical procedure and methods validation information. 比如说,许多生物分析是建立在动物挑战模式,免疫原性评估或其它有着独特特性的免疫分析基础上的,在递交分析方法和分析方法验证资料时需考虑这些独特的性质。

相关文档
最新文档