文档库 最新最全的文档下载
当前位置:文档库 › 分析方法验证指导原则-FDA

分析方法验证指导原则-FDA

分析方法验证指导原则-FDA
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.wendangku.net/doc/9515132338.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.wendangku.net/doc/9515132338.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

Table of Contents

I.INTRODUCTION (1)

II.BACKGROUND (1)

A.F ULL V ALIDATION (2)

B.P ARTIAL V ALIDATION (2)

C.C ROSS-V ALIDATION (3)

III.REFERENCE STANDARD (4)

IV.METHOD DEVELOPMENT: CHEMICAL ASSAY (4)

A.S ELECTIVITY (4)

B.A CCURACY, P RECISION, AND R ECOVERY (5)

C.C ALIBRATION/S TANDARD C URVE (5)

D.S TABILITY (6)

E.P RINCIPLES OF B IOANALYTICAL M ETHOD V ALIDATION AND E STABLISHMENT (8)

F.S PECIFIC R ECOMMENDATIONS FOR M ETHOD V ALIDATION (10)

V.METHOD DEVELOPMENT: MICROBIOLOGICAL AND LIGAND-BINDING ASSAYS (11)

A.S ELECTIVITY I SSUES (11)

B.Q UANTIFICATION I SSUES (12)

VI.APPLICATION OF VALIDATED METHOD TO ROUTINE DRUG ANALYSIS (13)

A CCEPTANCE C RITERIA FOR THE R UN (15)

VII.DOCUMENTATION (16)

A.S UMMARY I NFORMATION (16)

B.D OCUMENTATION FOR M ETHOD E STABLISHMENT (17)

C.A PPLICATION TO R OUTINE D RUG A NALYSIS (17)

D.O THER I NFORMATION (19)

GLOSSARY (20)

GUIDANCE FOR INDUSTRY1

Bioanalytical Method Validation

I.INTRODUCTION

This guidance provides assistance to sponsors of investigational new drug applications (INDs), new drug applications (NDAs), abbreviated new drug applications (ANDAs), and supplements in developing bioanalytical method validation information used in human clinical pharmacology, bioavailability (BA), and bioequivalence (BE) studies requiring pharmacokinetic (PK) evaluation. This guidance also applies to bioanalytical methods used for non-human pharmacology/toxicology studies and preclinical studies. For studies related to the veterinary drug approval process, this guidance applies only to blood and urine BA, BE, and PK studies.

The information in this guidance generally applies to bioanalytical procedures such as gas chromatography (GC), high-pressure liquid chromatography (LC), combined GC and LC mass spectrometric (MS) procedures such as LC-MS, LC-MS-MS, GC-MS, and GC-MS-MS performed for the quantitative determination of drugs and/or metabolites in biological matrices

such as blood, serum, plasma, or urine. This guidance also applies to other bioanalytical methods, such as immunological and microbiological procedures, and to other biological matrices, such as tissue and skin samples.

This guidance provides general recommendations for bioanalytical method validation. The recommendations can be adjusted or modified depending on the specific type of analytical method used. II.BACKGROUND

1 This guidance has been prepared by the Biopharmaceutics Coordinating Committee in the Center for Drug Evaluation and Research (CDER) in cooperation with the Center for Veterinary Medicine (CVM) at the Food and Drug Administration.

This guidance has been developed based on the deliberations of two workshops: (1) Analytical Methods Validation: Bioavailability, Bioequivalence, and Pharmacokinetic Studies (held on December 3B5, 19902 ) and (2) Bioanalytical Methods Validation C A Revisit With a Decade of Progress (held on January 12B14, 20003).

Selective and sensitive analytical methods for the quantitative evaluation of drugs and their metabolites (analytes) are critical for the successful conduct of preclinical and/or biopharmaceutics and clinical pharmacology studies. Bioanalytical method validation includes all of the procedures that demonstrate that a particular method used for quantitative measurement of analytes in a given biological matrix, such as blood, plasma, serum, or urine, is reliable and reproducible for the intended use. The fundamental parameters for this validation include (1) accuracy, (2) precision, (3) selectivity, (4) sensitivity, (5) reproducibility, and (6) stability. Validation involves documenting, through the use of specific laboratory investigations, that the performance characteristics of the method are suitable and reliable for the intended analytical applications. The acceptability of analytical data corresponds directly to the criteria used to validate the method.

Published methods of analysis are often modified to suit the requirements of the laboratory performing the assay. These modifications should be validated to ensure suitable performance of the analytical method. When changes are made to a previously validated method, the analyst should exercise judgment as to how much additional validation is needed. During the course of a typical drug development program, a defined bioanalytical method undergoes many modifications. The evolutionary changes to support specific studies and different levels of validation demonstrate the validity of an assay’s performance. Different types and levels of validation are defined and characterized as follows:

A.Full Validation

?Full validation is important when developing and implementing a bioanalytical method for the first time.

?Full validation is important for a new drug entity.

? A full validation of the revised assay is important if metabolites are added to an existing assay for quantification.

B.Partial Validation

Partial validations are modifications of already validated bioanalytical methods. Partial validation can range from as little as one intra-assay accuracy and precision determination to a nearly full

2 Workshop Report: Shah, V.P. et al., Pharmaceutical Research: 1992; 9:588-592.

3 Workshop Report: Shah, V.P. et al., Pharmaceutical Research: 2000; 17:in press.

validation. Typical bioanalytical method changes that fall into this category include, but are not limited to:

?Bioanalytical method transfers between laboratories or analysts

?Change in analytical methodology (e.g., change in detection systems)

?Change in anticoagulant in harvesting biological fluid

?Change in matrix within species (e.g., human plasma to human urine)

?Change in sample processing procedures

?Change in species within matrix (e.g., rat plasma to mouse plasma)

?Change in relevant concentration range

?Changes in instruments and/or software platforms

?Limited sample volume (e.g., pediatric study)

?Rare matrices

?Selectivity demonstration of an analyte in the presence of concomitant medications

?Selectivity demonstration of an analyte in the presence of specific metabolites

C.Cross-Validation

Cross-validation is a comparison of validation parameters when two or more bioanalytical methods are used to generate data within the same study or across different studies. An example of cross-validation would be a situation where an original validated bioanalytical method serves as the

reference and the revised bioanalytical method is the comparator. The comparisons should be done both ways.

When sample analyses within a single study are conducted at more than one site or more than one laboratory, cross-validation with spiked matrix standards and subject samples should be conducted at each site or laboratory to establish interlaboratory reliability. Cross-validation should also be considered when data generated using different analytical techniques (e.g., LC-MS-MS vs.

ELISA4) in different studies are included in a regulatory submission.

All modifications should be assessed to determine the recommended degree of validation. The analytical laboratory conducting pharmacology/toxicology and other preclinical studies for regulatory submissions should adhere to FDA=s Good Laboratory Practices (GLPs)5 (21 CFR part 58) and to sound principles of quality assurance throughout the testing process. The bioanalytical method for human BA, BE, PK, and drug interaction studies must meet the criteria in 21 CFR 320.29. The analytical laboratory should have a written set of standard operating procedures (SOPs) to ensure a complete system of quality control and assurance. The SOPs should cover all aspects of analysis from the time the sample is collected and reaches the laboratory until the results of the analysis are reported. The SOPs also should include record keeping, security and chain of sample custody

4 Enzyme linked immune sorbent assay

5 For the Center for Veterinary Medicine, all bioequivalence studies are subject to Good Laboratory Practices.

(accountability systems that ensure integrity of test articles), sample preparation, and analytical tools such as methods, reagents, equipment, instrumentation, and procedures for quality control and verification of results.

The process by which a specific bioanalytical method is developed, validated, and used in routine sample analysis can be divided into (1) reference standard preparation, (2) bioanalytical method development and establishment of assay procedure, and (3) application of validated bioanalytical method to routine drug analysis and acceptance criteria for the analytical run and/or batch. These three processes are described in the following sections of this guidance.

III.REFERENCE STANDARD

Analysis of drugs and their metabolites in a biological matrix is carried out using samples spiked with calibration (reference) standards and using quality control (QC) samples. The purity of the reference standard used to prepare spiked samples can affect study data. For this reason, an authenticated analytical reference standard of known identity and purity should be used to prepare solutions of known concentrations. If possible, the reference standard should be identical to the analyte. When this is not possible, an established chemical form (free base or acid, salt or ester) of known purity can be used. Three types of reference standards are usually used: (1) certified reference standards (e.g., USP compendial standards); (2) commercially supplied reference standards obtained from a reputable commercial source; and/or (3) other materials of documented purity custom-synthesized by an analytical laboratory or other noncommercial establishment. The source and lot number, expiration date, certificates of analyses when available, and/or internally or externally generated evidence of identity and purity should be furnished for each reference standard.

IV.METHOD DEVELOPMENT: CHEMICAL ASSAY

The method development and establishment phase defines the chemical assay. The fundamental parameters for a bioanalytical method validation are accuracy, precision, selectivity, sensitivity, reproducibility, and stability. Measurements for each analyte in the biological matrix should be validated. In addition, the stability of the analyte in spiked samples should be determined. Typical method development and establishment for a bioanalytical method include determination of (1) selectivity, (2) accuracy, precision, recovery, (3) calibration curve, and (4) stability of analyte in spiked samples.

A.Selectivity

Selectivity is the ability of an analytical method to differentiate and quantify the analyte in the

presence of other components in the sample. For selectivity, analyses of blank samples of the

appropriate biological matrix (plasma, urine, or other matrix) should be obtained from at least

six sources. Each blank sample should be tested for interference, and selectivity should be ensured at the lower limit of quantification (LLOQ).

Potential interfering substances in a biological matrix include endogenous matrix components, metabolites, decomposition products, and in the actual study, concomitant medication and other exogenous xenobiotics. If the method is intended to quantify more than one analyte, each analyte should be tested to ensure that there is no interference.

B.Accuracy, Precision, and Recovery

The accuracy of an analytical method describes the closeness of mean test results obtained by the method to the true value (concentration) of the analyte. Accuracy is determined by replicate analysis of samples containing known amounts of the analyte. Accuracy should be measured using a minimum of five determinations per concentration. A minimum of three concentrations in the range of expected concentrations is recommended. The mean value should be within 15% of the actual value except at LLOQ, where it should not deviate by more than 20%. The deviation of the mean from the true value serves as the measure of accuracy.

The precision of an analytical method describes the closeness of individual measures of an analyte when the procedure is applied repeatedly to multiple aliquots of a single homogeneous volume of biological matrix. Precision should be measured using a minimum of five determinations per concentration. A minimum of three concentrations in the range of expected concentrations is recommended. The precision determined at each concentration level should not exceed 15% of the coefficient of variation (CV) except for the LLOQ, where it should not exceed 20% of the CV. Precision is further subdivided into within-run, intra-batch precision or repeatability, which assesses precision during a single analytical run, and between-run, inter-batch precision or repeatability, which measures precision with time, and may involve different analysts, equipment, reagents, and laboratories.

The recovery of an analyte in an assay is the detector response obtained from an amount of the analyte added to and extracted from the biological matrix, compared to the detector response obtained for the true concentration of the pure authentic standard. Recovery pertains to the extraction efficiency of an analytical method within the limits of variability. Recovery of the analyte need not be 100%, but the extent of recovery of an analyte and of the internal standard should be consistent, precise, and reproducible. Recovery experiments should be performed by comparing the analytical results for extracted samples at three concentrations (low, medium, and high) with unextracted standards that represent 100% recovery.

C.Calibration/Standard Curve

A calibration (standard) curve is the relationship between instrument response and known concentrations of the analyte. A calibration curve should be generated for each analyte in the

sample. A sufficient number of standards should be used to adequately define the relationship between concentration and response. A calibration curve should be prepared in the same biological matrix as the samples in the intended study by spiking the matrix with known concentrations of the analyte. The number of standards used in constructing a calibration curve will be a function of the anticipated range of analytical values and the nature of the

analyte/response relationship. Concentrations of standards should be chosen on the basis of the concentration range expected in a particular study. A calibration curve should consist of a blank sample (matrix sample processed without internal standard), a zero sample (matrix sample processed with internal standard), and six to eight non-zero samples covering the expected range, including LLOQ.

1.Lower Limit of Quantification (LLOQ)

The lowest standard on the calibration curve should be accepted as the limit of

quantification if the following conditions are met:

C The analyte response at the LLOQ should be at least 5 times the response

compared to blank response.

C Analyte peak (response) should be identifiable, discrete, and reproducible with

a precision of 20% and accuracy of 80-120%.

2.Calibration Curve/Standard Curve/Concentration-Response

The simplest model that adequately describes the concentration-response relationship

should be used. Selection of weighting and use of a complex regression equation should be justified. The following conditions should be met in developing a calibration curve:

C#20% deviation of the LLOQ from nominal concentration

C#15% deviation of standards other than LLOQ from nominal concentration

At least four out of six non-zero standards should meet the above criteria, including the LLOQ and the calibration standard at the highest concentration. Excluding the

standards should not change the model used.

D.Stability

Drug stability in a biological fluid is a function of the storage conditions, the chemical properties of the drug, the matrix, and the container system. The stability of an analyte in a particular matrix and container system is relevant only to that matrix and container system and should not be extrapolated to other matrices and container systems. Stability procedures should evaluate the stability of the analytes during sample collection and handling, after long-term (frozen at the

intended storage temperature) and short-term (bench top, room temperature) storage, and after going through freeze and thaw cycles and the analytical process. Conditions used in stability experiments should reflect situations likely to be encountered during actual sample handling and analysis. The procedure should also include an evaluation of analyte stability in stock solution.

All stability determinations should use a set of samples prepared from a freshly made stock solution of the analyte in the appropriate analyte-free, interference-free biological matrix. Stock solutions of the analyte for stability evaluation should be prepared in an appropriate solvent at known concentrations.

1.Freeze and Thaw Stability

Analyte stability should be determined after three freeze and thaw cycles. At least three aliquots at each of the low and high concentrations should be stored at the intended

storage temperature for 24 hours and thawed unassisted at room temperature. When

completely thawed, the samples should be refrozen for 12 to 24 hours under the same

conditions. The freeze–thaw cycle should be repeated two more times, then analyzed

on the third cycle. If an analyte is unstable at the intended storage temperature, the

stability sample should be frozen at -700C during the three freeze and thaw cycles.

2.Short-Term Temperature Stability

Three aliquots of each of the low and high concentrations should be thawed at room

temperature and kept at this temperature from 4 to 24 hours (based on the expected

duration that samples will be maintained at room temperature in the intended study) and analyzed.

3.Long-Term Stability

The storage time in a long-term stability evaluation should exceed the time between the date of first sample collection and the date of last sample analysis. Long-term stability

should be determined by storing at least three aliquots of each of the low and high

concentrations under the same conditions as the study samples. The volume of samples should be sufficient for analysis on three separate occasions. The concentrations of all

the stability samples should be compared to the mean of back-calculated values for the standards at the appropriate concentrations from the first day of long-term stability

testing.

4.Stock Solution Stability

The stability of stock solutions of drug and the internal standard should be evaluated at room temperature for at least 6 hours. If the stock solutions are refrigerated or frozen

for the relevant period, the stability should be documented. After completion of the

desired storage time, the stability should be tested by comparing the instrument

response with that of freshly prepared solutions.

5.Post-Preparative Stability

The stability of processed samples, including the resident time in the autosampler, should be determined. The stability of the drug and the internal standard should be assessed

over the anticipated run time for the batch size in validation samples by determining

concentrations on the basis of original calibration standards.

Although the traditional approach of comparing analytical results for stored samples with those for freshly prepared samples has been referred to in this guidance, other statistical approaches based on confidence limits for evaluation of an analyte=s stability in a

biological matrix can be used. SOPs should clearly describe the statistical method and

rules used. Additional validation may include investigation of samples from dosed

subjects.

E.Principles of Bioanalytical Method Validation and Establishment

?The fundamental parameters to ensure the acceptability of the performance of a bioanalytical method validation are accuracy, precision, selectivity, sensitivity,

reproducibility, and stability.

? A specific, detailed description of the bioanalytical method should be written. This can be in the form of a protocol, study plan, report, and/or SOP.

?Each step in the method should be investigated to determine the extent to which environmental, matrix, material, or procedural variables can affect the estimation of analyte in the matrix from the time of collection of the material up to and including the time of

analysis.

?It may be important to consider the variability of the matrix due to the physiological nature of the sample. In the case of LC-MS-MS-based procedures, appropriate steps should be taken to ensure the lack of matrix effects throughout the application of the method,

especially if the nature of the matrix changes from the matrix used during method validation.? A bioanalytical method should be validated for the intended use or application. All experiments used to make claims or draw conclusions about the validity of the method

should be presented in a report (method validation report).

?Whenever possible, the same biological matrix as the matrix in the intended samples should be used for validation purposes. (For tissues of limited availability, such as bone marrow, physiologically appropriate proxy matrices can be substituted.)

?The stability of the analyte (drug and/or metabolite) in the matrix during the collection process and the sample storage period should be assessed, preferably prior to sample

analysis.

?For compounds with potentially labile metabolites, the stability of analyte in matrix from dosed subjects (or species) should be confirmed.

?The accuracy, precision, reproducibility, response function, and selectivity of the method for endogenous substances, metabolites, and known degradation products should be

established for the biological matrix. For selectivity, there should be evidence that the

substance being quantified is the intended analyte.

?The concentration range over which the analyte will be determined should be defined in the bioanalytical method, based on evaluation of actual standard samples over the range,

including their statistical variation. This defines the standard curve.

? A sufficient number of standards should be used to adequately define the relationship between concentration and response. The relationship between response and concentration should be demonstrated to be continuous and reproducible. The number of standards used should be a function of the dynamic range and nature of the concentration-response

relationship. In many cases, six to eight concentrations (excluding blank values) can define the standard curve. More standard concentrations may be recommended for nonlinear than for linear relationships.

?The ability to dilute samples originally above the upper limit of the standard curve should be demonstrated by accuracy and precision parameters in the validation.

?In consideration of high throughput analyses, including but not limited to multiplexing, multicolumn, and parallel systems, sufficient QC samples should be used to ensure control of the assay. The number of QC samples to ensure proper control of the assay should be determined based on the run size. The placement of QC samples should be judiciously considered in the run.

?For a bioanalytical method to be considered valid, specific acceptance criteria should be set in advance and achieved for accuracy and precision for the validation of QC samples over the range of the standards.

F.Specific Recommendations for Method Validation

?The matrix-based standard curve should consist of a minimum of six standard points, excluding blanks, using single or replicate samples. The standard curve should cover the entire range of expected concentrations.

?Standard curve fitting is determined by applying the simplest model that adequately describes the concentration-response relationship using appropriate weighting and statistical tests for goodness of fit.

?LLOQ is the lowest concentration of the standard curve that can be measured with acceptable accuracy and precision. The LLOQ should be established using at least five samples independent of standards and determining the coefficient of variation and/or

appropriate confidence interval. The LLOQ should serve as the lowest concentration on the standard curve and should not be confused with the limit of detection and/or the low QC sample. The highest standard will define the upper limit of quantification (ULOQ) of an analytical method.

?For validation of the bioanalytical method, accuracy and precision should be determined using a minimum of five determinations per concentration level (excluding blank samples).

The mean value should be within ±15% of the theoretical value, except at LLOQ, where it should not deviate by more than ±20%. The precision around the mean value should not exceed 15% of the CV, except for LLOQ, where it should not exceed 20% of the CV.

Other methods of assessing accuracy and precision that meet these limits may be equally acceptable.

?The accuracy and precision with which known concentrations of analyte in biological matrix can be determined should be demonstrated. This can be accomplished by analysis of

replicate sets of analyte samples of known concentrations C QC samples C from an

equivalent biological matrix. At a minimum, three concentrations representing the entire range of the standard curve should be studied: one within 3x the lower limit of quantification (LLOQ) (low QC sample), one near the center (middle QC), and one near the upper

boundary of the standard curve (high QC).

?Reported method validation data and the determination of accuracy and precision should include all outliers; however, calculations of accuracy and precision excluding values that are statistically determined as outliers can also be reported.

?The stability of the analyte in biological matrix at intended storage temperatures should be established. The influence of freeze-thaw cycles (a minimum of three cycles at two

concentrations in triplicate) should be studied.

?The stability of the analyte in matrix at ambient temperature should be evaluated over a time period equal to the typical sample preparation, sample handling, and analytical run times.

?Reinjection reproducibility should be evaluated to determine if an analytical run could be reanalyzed in the case of instrument failure.

?The specificity of the assay methodology should be established using a minimum of six independent sources of the same matrix. For hyphenated mass spectrometry-based

methods, however, testing six independent matrices for interference may not be important.

In the case of LC-MS and LC-MS-MS-based procedures, matrix effects should be

investigated to ensure that precision, selectivity, and sensitivity will not be compromised.

Method selectivity should be evaluated during method development and throughout method

validation and can continue throughout application of the method to actual study samples.

?Acceptance/rejection criteria for spiked, matrix-based calibration standards and validation QC samples should be based on the nominal (theoretical) concentration of analytes.

Specific criteria can be set up in advance and achieved for accuracy and precision over the

range of the standards, if so desired.

V.METHOD DEVELOPMENT: MICROBIOLOGICAL AND LIGAND-BINDING ASSAYS

Many of the bioanalytical validation parameters and principles discussed above are also applicable to microbiological and ligand-binding assays. However, these assays possess some unique characteristics that should be considered during method validation.

A.Selectivity Issues

As with chromatographic methods, microbiological and ligand-binding assays should be shown to be selective for the analyte. The following recommendations for dealing with two selectivity issues should be considered:

1.Interference From Substances Physiochemically Similar to the Analyte

?Cross-reactivity of metabolites, concomitant medications, or endogenous

compounds should be evaluated individually and in combination with the analyte

of interest.

?When possible, the immunoassay should be compared with a validated reference method (such as LC-MS) using incurred samples and predetermined criteria for

agreement of accuracy of immunoassay and reference method.

?The dilutional linearity to the reference standard should be assessed using study (incurred) samples.

?Selectivity may be improved for some analytes by incorporation of separation steps prior to immunoassay.

2.Matrix effects Unrelated to the Analyte

?The standard curve in biological fluids should be compared with standard in buffer to detect matrix effects.

?Parallelism of diluted study samples should be evaluated with diluted standards to detect matrix effects.

?Nonspecific binding should be determined.

B.Quantification Issues

Microbiological and imunoassay standard curves are inherently nonlinear and, in general, more concentration points may be recommended to define the fit over the standard curve range than for chemical assays. In addition to their nonlinear characteristics, the response-error relationship for immunoassay standard curves is a nonconstant function of the mean response (heteroscadisticity). For these reasons, a minimum of six non-zero calibrator concentrations, run in duplicate, is recommended. The concentration-response relationship is most often fitted to a 4- or 5-parameter logistic model, although others may be used with suitable validation. The use of anchoring points in the asymptotic high- and low-concentration ends of the standard curve may improve the overall curve fit. Generally, these anchoring points will be at concentrations that are below the established LLOQ and above the established ULOQ. Whenever possible, calibrators should be prepared in the same matrix as the study samples or in an alternate matrix of equivalent performance. Both ULOQ and LLOQ should be defined by acceptable accuracy, precision, or confidence interval criteria based on the study requirements. For all assays the key factor is the accuracy of the reported results. This accuracy can be improved by the use of replicate samples. In the case where replicate samples should be measured during the validation to improve accuracy, the same procedure should be followed as for unknown samples.

The following recommendations apply to quantification issues:

?If separation is used prior to assay for study samples but not for standards, it is important to establish recovery and use it in determining results. Possible approaches to assess efficiency

and reproducibility of recovery are (1) the use of radiolabeled tracer analyte (quantity too

small to affect the assay), (2) the advance establishment of reproducible recovery, (3) the

use of an internal standard that is not recognized by the antibody but can be measured by

another technique.

?Key reagents, such as antibody, tracer, reference standard, and matrix should be characterized appropriately and stored under defined conditions.

?Assessments of analyte stability should be conducted in true study matrix (e.g., should not use a matrix stripped to remove endogenous interferences).

?Acceptance criteria: At least 67% (4 out of 6) of QC samples should be within 15% of their respective nominal value, 33% of the QC samples (not all replicates at the same

concentration) may be outside 15% of nominal value. In certain situations, wider

acceptance criteria may be justified.

?Assay reoptimization or validation may be important when there are changes in key reagents, as follows:

Labeled analyte (tracer)

?Binding should be reoptimized.

?Performance should be verified with standard curve and QCs.

Antibody

?Key cross-reactivities should be checked.

?Tracer experiments above should be repeated.

Matrix

?Tracer experiments above should be repeated.

Method development experiments should include a minimum of six runs conducted over several days, with at least four concentrations (LLOQ, low, medium, and high) analyzed in duplicate in each run. VI.APPLICATION OF VALIDATED METHOD TO ROUTINE DRUG ANALYSIS Assays of all samples of an analyte in a biological matrix should be completed within the time period for which stability data are available. In general, biological samples can be analyzed with a single determination without duplicate or replicate analysis if the assay method has acceptable variability as defined by validation data. This is true for procedures where precision and accuracy variabilities routinely fall within acceptable tolerance limits. For a difficult procedure with a labile analyte where high precision and accuracy specifications may be difficult to achieve, duplicate or even triplicate analyses can be performed for a better estimate of analyte.

A calibration curve should be generated for each analyte to assay samples in each analytical run and should be used to calculate the concentration of the analyte in the unknown samples in the run. The spiked samples can contain more than one analyte. An analytical run can consist of QC samples, calibration standards, and either (1) all the processed samples to be analyzed as one batch or (2) a batch composed of processed unknown samples of one or more volunteers in a study. The calibration (standard) curve should cover the expected unknown sample concentration range in addition to a calibrator sample at LLOQ. Estimation of concentration in unknown samples by extrapolation of standard curves below LLOQ or above the highest standard is not recommended. Instead, the standard curve should be redefined or samples with higher concentration should be diluted and reassayed. It is preferable to analyze all study samples from a subject in a single run.

Once the analytical method has been validated for routine use, its accuracy and precision should be monitored regularly to ensure that the method continues to perform satisfactorily. To achieve this objective, a number of QC samples prepared separately should be analyzed with processed test samples at intervals based on the total number of samples. The QC samples in duplicate at three concentrations (one near the LLOQ (i.e., #3 x LLOQ), one in midrange, and one close to the high end of the range) should be incorporated in each assay run. The number of QC samples (in multiples of three) will depend on the total number of samples in the run. The results of the QC samples provide the basis of accepting or rejecting the run. At least four of every six QC samples should be within "15% of their respective nominal value. Two of the six QC samples may be outside the "15% of their respective nominal value, but not both at the same concentration.

The following recommendations should be noted in applying a bioanalytical method to routine drug analysis:

? A matrix-based standard curve should consist of a minimum of six standard points, excluding blanks (either single or replicate), covering the entire range.

?Response Function: Typically, the same curve fitting, weighting, and goodness of fit determined during prestudy validation should be used for the standard curve within the

study. Response function is determined by appropriate statistical tests based on the actual

standard points during each run in the validation. Changes in the response function

relationship between prestudy validation and routine run validation indicate potential

problems.

?The QC samples should be used to accept or reject the run. These QC samples are matrix spiked with analyte.

?System suitability: Based on the analyte and technique, a specific SOP (or sample) should be identified to ensure optimum operation of the system used.

?Any required sample dilutions should use like matrix (e.g., human to human) obviating the need to incorporate actual within-study dilution matrix QC samples.

?Repeat Analysis: It is important to establish an SOP or guideline for repeat analysis and acceptance criteria. This SOP or guideline should explain the reasons for repeating sample analysis. Reasons for repeat analyses could include repeat analysis of clinical or preclinical samples for regulatory purposes, inconsistent replicate analysis, samples outside of the assay range, sample processing errors, equipment failure, poor chromatography, and inconsistent pharmacokinetic data. Reassays should be done in triplicate if sample volume allows. The rationale for the repeat analysis and the reporting of the repeat analysis should be clearly documented.

?Sample Data Reintegration: An SOP or guideline for sample data reintegration should be established. This SOP or guideline should explain the reasons for reintegration and how the reintegration is to be performed. The rationale for the reintegration should be clearly

described and documented. Original and reintegration data should be reported.

Acceptance Criteria for the Run

The following acceptance criteria should be considered for accepting the analytical run:

?Standards and QC samples can be prepared from the same spiking stock solution, provided the solution stability and accuracy have been verified. A single source of matrix may also be used, provided selectivity has been verified.

?Standard curve samples, blanks, QCs, and study samples can be arranged as considered appropriate within the run.

?Placement of standards and QC samples within a run should be designed to detect assay drift over the run.

?Matrix-based standard calibration samples: 75%, or a minimum of six standards, when back-calculated (including ULOQ) should fall within ±15%, except for LLOQ, when it should be ±20% of the nominal value. Values falling outside these limits can be discarded, provided they do not change the established model.

?Acceptance criteria for accuracy and precision as outlined in section IV.F, “Specific Recommendation for Method Validation,” should be provided for both the intra-day and intra-run experiment.

?Quality Control Samples: Quality control samples replicated (at least once) at a minimum of three concentrations (one within 3x of the LLOQ (low QC), one in the midrange (middle

QC), and one approaching the high end of the range (high QC)) should be incorporated into

each run. The results of the QC samples provide the basis of accepting or rejecting the run.

At least 67% (four out of six) of the QC samples should be within 15% of their respective

nominal (theoretical) values; 33% of the QC samples (not all replicates at the same

concentration) can be outside the ±15% of the nominal value. A confidence interval

approach yielding comparable accuracy and precision is an appropriate alternative.

The minimum number of samples (in multiples of three) should be at least 5% of the number of unknown samples or six total QCs, whichever is greater.

?Samples involving multiple analytes should not be rejected based on the data from one analyte failing the acceptance criteria.

?The data from rejected runs need not be documented, but the fact that a run was rejected and the reason for failure should be recorded.

VII.DOCUMENTATION

The validity of an analytical method should be established and verified by laboratory studies, and documentation of successful completion of such studies should be provided in the assay validation report. General and specific SOPs and good record keeping are an essential part of a validated analytical method. The data generated for bioanalytical method establishment and the QCs should be documented and available for data audit and inspection. Documentation for submission to the Agency should include (1) summary information, (2) method development and establishment, (3) bioanalytical reports of the application of any methods to routine sample analysis, and (4) other information applicable to method development and establishment and/or to routine sample analysis.

A.Summary Information

Summary information should include:

?Summary table of validation reports, including analytical method validation, partial revalidation, and cross-validation reports. The table should be in chronological sequence,

and include assay method identification code, type of assay, and the reason for the new

method or additional validation (e.g., to lower the limit of quantitation).

?Summary table with a list, by protocol, of assay methods used. The protocol number, protocol title, assay type, assay method identification code, and bioanalytic report code

should be provided.

? A summary table allowing cross-referencing of multiple identification codes should be provided (e.g., when an assay has different codes for the assay method, validation reports,

and bioanalytical reports, especially when the sponsor and a contract laboratory assign different codes).

B.Documentation for Method Establishment

Documentation for method development and establishment should include:

?An operational description of the analytical method

?Evidence of purity and identity of drug standards, metabolite standards, and internal standards used in validation experiments

? A description of stability studies and supporting data

? A description of experiments conducted to determine accuracy, precision, recovery, selectivity, limit of quantification, calibration curve (equations and weighting functions used, if any), and relevant data obtained from these studies

?Documentation of intra- and inter-assay precision and accuracy

?In NDA submissions, information about cross-validation study data, if applicable ?Legible annotated chromatograms or mass spectrograms, if applicable

?Any deviations from SOPs, protocols, or GLPs (if applicable), and justifications for deviations

C.Application to Routine Drug Analysis

Documentation of the application of validated bioanalytical methods to routine drug analysis should include:

?Evidence of purity and identity of drug standards, metabolite standards, and internal standards used during routine analyses

?Summary tables containing information on sample processing and storage. Tables should include sample identification, collection dates, storage prior to shipment, information on shipment batch, and storage prior to analysis. Information should include dates, times,

sample condition, and any deviation from protocols.

?Summary tables of analytical runs of clinical or preclinical samples. Information should include assay run identification, date and time of analysis, assay method, analysts, start and

20.化学药物质量控制分析方法验证技术指导原则 2005年颁布

指导原则编号: 【H】G P H 5-1 化学药物质量控制分析方法验证 技术指导原则 二○○五年三月

目 录 一、概述 (1) 二、方法验证的一般原则 (2) 三、方法验证涉及到的三个主要方面 (2) (一)需要验证的检测项目 (2) (二)分析方法 (3) (三)验证内容 (3) 四、方法验证的具体内容 (3) (一)专属性 (3) 1、鉴别反应 (4) 2、杂质检查 (4) 3、含量测定 (4) (二)线性 (5) (三)范围 (5) 1、含量测定 (6) 2、制剂含量均匀度 (6) 3、溶出度或释放度 (6) 4、杂质 (6) (四)准确度 (6) 1、含量测定 (7) 2、杂质定量试验 (7) (五)精密度 (7) 1、重复性 (8) 2、中间精密度 (8) 3、重现性 (8)

(六)检测限 (8) 1、直观法 (8) 2、信噪比法 (9) (七)定量限 (9) 1、直观法 (9) 2、信噪比法 (9) (八)耐用性 (10) (九)系统适用性试验 (10) 五、方法再验证 (11) 六、方法验证的评价 (12) (一)有关方法验证评价的一般考虑 (12) (二)方法验证的整体性和系统性 (12) 七、参考文献 (13) 八、著者 (13)

化学药物质量控制分析方法验证技术指导原则 一、概述 保证药品安全、有效、质量可控是药品研发和评价应遵循的基本原则,其中,对药品进行质量控制是保证药品安全有效的基础和前提。为达到控制质量的目的,需要多角度、多层面来控制药品质量,也就是说要对药物进行多个项目测试,来全面考察药品质量。一般地,每一测试项目可选用不同的分析方法,为使测试结果准确、可靠,必须对所采用的分析方法的科学性、准确性和可行性进行验证,以充分表明分析方法符合测试项目的目的和要求,这就是通常所说的对方法进行验证。 方法验证的目的是判断采用的分析方法是否科学、合理,是否能有效控制药品的内在质量。从本质上讲,方法验证就是根据检测项目的要求,预先设置一定的验证内容,并通过设计合理的试验来验证所采用的分析方法能否符合检测项目的要求。 方法验证在分析方法建立过程中具有重要的作用,并成为质量研究和质量控制的组成部分。只有经过验证的分析方法才能用于控制药品质量,因此方法验证是制订质量标准的基础。方法验证是药物研究过程中的重要内容。 本指导原则重点探讨方法验证的本质,将分析方法验证的要求与所要达到的目的结合起来进行系统和规律性的阐述,重点阐述如何科学合理地进行论证方案的设计。 本指导原则主要包括方法验证的一般原则、方法验证涉及的三个主要方

药品微生物检验替代方法验证指导原则

药品微生物检验替代方法验证指导原则 本指导原则是为所采用的试验方法能否替代药典规定的方法用于药品微生物的检验提供指导。 随着微生物学的迅速发展,制药领域不断引入了一些新的微生物检验技术,大体可分为三类:(1)基于微生物生长信息的检验技术,如生物发光技术、电化学技术、比浊法等;(2)直接测定被测介质中活微生物的检验技术,如固相细胞技术法、流式细胞计数法等;(3)基于微生物细胞所含有特定组成成分的分析技术,如脂肪酸测定技术、核酸扩增技术、基因指纹分析技术等。这些方法与传统检查方法比较,或简便快速,或具有实时或近实时监控的潜力,使生产早期采取纠正措施及监控和指导优良生产成为可能,同时新技术的使用也促进了生产成本降低及检验水平的提高。 在控制药品微生物质量中,微生物实验室出于各种原因如成本、生产量、快速简便及提高药品质量等需要而采用非药典规定的检验方法(即替代方法)时,应进行替代方法的验证,确认其应用效果优于或等同于药典的方法。 微生物检验的类型及验证参数 药品微生物检验方法主要分两种类型:定性试验和定量试验。定性试验就是测定样品中是否存在活的微生物,如无菌检查及控制菌检査。定量试验就是测定样品中存在的微生物数量,如菌落计数试验。 由于生物试验的特殊性,如微生物检验方法中的抽样误差、稀释误差、操作误差、培养误差和计数误差都会对检验结果造成影响,因此,药品质量标准分析方法验证指导原则(附录XIX A)不完全适宜于微生物替代方法的验证。药品微生物检验替代方法的验证参数见表1。 表1 不同微生物检验类型验证参数 注: 尽管替代方法的验证参数与药品质量标准分析方法验证参数有相似之处,但是其具体的内容是依据微生物检验特点而设立的。替代方法验证的实验结果需进行统计分析,当替代方法属于定性检验时,一般采用非参数的统计技术;当替代方法属于定量检验时,需要采用参数统计技术。 进行微生物替代方法的验证时,若替代方法只是针对药典方法中的某一环节进行技术修改,此时,需要验证的对象仅是该项替代技术而不是整个检验方法。如无菌试验若改为使用含培养基的过滤器,然后通过适宜的技术确认活的微生物存在,那么,验证时仅需验证所用的微生物回收系统而不是整个无菌试验方法。 替代方法验证的一般要求 在开展替代方法对样品检验的适用性验证前,有必要对替代方法有一个全面的了解。首先,所选用的替代方法应具备必要的方法适用性证据,表明在不含样品的情况下,替代方法

液相色谱分析方法建立

一. 方法建立的步骤 二.开始前应知道 1. 样品的性质 在开始方法建立之前,我们应该检查自己对样品的了解程度,并明确分离目标。 表 1 有关样品组分和性质的重要信息 所含化合物的数目 化合物的化学结构(官能团) 化合物的分子量 化合物的pKa值 化合物的UV光谱图 化合物在样品中的浓度范围 样品的溶解度 样品的化学成分能够为选择HPLC分离的最佳初始条件提供有价值的线索根据已知的样品信息,HPLC方法建立有两种不甚相同的模式。一种模式依据样品的“化学性质”选择最佳初始条件,色谱工作者需很大程度依赖于过去的经验(如类似结构化合物的分离)和/或用文献资料补充现有信息而另一种模式则直接开始色谱分离,而对样品的性质不大注意这两种HPLC的方法建立模式可分别称为理沦型与经验型初始分离一旦开始,可以根据类似的思路(理论的与经验的)选择进一步的实验。 2.分离的目的 HPLC分离的目的必须十分明确,下面的问题在建立方法之初就应确定:(1)主要目的是什么?定量或定性,还是定性、定量同时做?; (2)是否有必要解析出样品的所有成分?譬如可能有必要分离出产品中的所有降解物或杂质,以使含量测定结果更加可靠,但却没必要将它们彼此完全分开。(3)如要求定量分析,准确度与精密度需多大?样品主要成分的精密度通常能达到±1—2%,特别是不需样品预处理的情况。 (4)特殊化合物可能会以不同的样品形式出现(如:原料药,一种或多种形态,环保样品等)。是否需要一种以上的HPLC方法?单一方法分离不同形态样品是否理想? (5)一次将分析多少样品?当必须同时处理大量样品时,运行时间将变得非常重要。 有时甚至为了缩短运行时间而以牺牲样品分离度作代价,如缩短柱长或加快流速。当一次分析的样品数目超过10个,运行时间一般应控制在20min以内。(6)将要使用该方法的实验室中,有哪些HPLC设备?色谱柱能否恒温系统能否做梯度洗脱?该方法是否可在不同设计与生产的设备上运行? 方法建立实验开始之前,应明确对方法的这些要求。 三. 样品的预处理和检测 1. 预处理:样品来源形式不同,可能以如下形式出现:

生物样品定量分析方法验证指导原则

9012 生物样品定量分析方法验证指导原则
1. 范围
准确测定生物基质(如全血、血清、血浆、尿)中的药物浓度,对于药物和 制剂研发非常重要。这些数据可被用于支持药品的安全性和有效性,或根据毒动 学、药动学和生物等效性试验的结果做出关键性决定。因此,必须完整地验证和 记录应用的生物分析方法,以获得可靠的结果。
本指导原则提供生物分析方法验证的要求,也涉及非临床或临床试验样品实 际分析的基本要求,以及何时可以使用部分验证或交叉验证,来替代完整验证。
生物样品定量分析方法验证和试验样品分析应符合本指导原则的技术要求。 应该在相应的生物样品分析中遵守 GLP 原则或 GCP 原则。
2. 生物分析方法验证
2.1 分析方法的完整验证
分析方法验证的主要目的是,证明特定方法对于测定在某种生物基质中分析 物浓度的可靠性。此外,方法验证应采用与试验样品相同的抗凝剂。一般应对每 个物种和每种基质进行完整验证。当难于获得相同的基质时,可以采用适当基质 替代,但要说明理由。
一个生物分析方法的主要特征包括:选择性、定量下限、响应函数和校正范 围(标准曲线性能)、准确度、精密度、基质效应、分析物在生物基质以及溶液 中储存和处理全过程中的稳定性。
有时可能需要测定多个分析物。这可能涉及两种不同的药物,也可能涉及一 个母体药物及其代谢物,或一个药物的对映体或异构体。在这些情况下,验证和 分析的原则适用于所有涉及的分析物。
对照标准物质 在方法验证中,含有分析物对照标准物质的溶液将被加入到空白生物基质 中。此外,色谱方法通常使用适当的内标。 应该从可追溯的来源获得对照标准物质。应该科学论证对照标准物质的适用 性。分析证书应该确认对照标准物质的纯度,并提供储存条件、失效日期和批号。 对于内标,只要能证明其适用性即可,例如显示该物质本身或其相关的任何杂质 不产生干扰。 当在生物分析方法中使用质谱检测时,推荐尽可能使用稳定同位素标记的内 标。它们必须具有足够高的同位素纯度,并且不发生同位素交换反应,以避免结 果的偏差。
1

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

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

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

方法学验证指导原则

一、准确度 准确度系指采用该方法测定的结果与真实值或参考值接近的程度,一般用回收率(%)表示。准确度应在规定的范围内测定。 1.化学药含量测定方法的准确度 原料药采用对照品进行测定,或用本法所得结果与已知准确度的另一个方法测定的结果进行比较。制剂可在处方量空白辅料中,加入已知量被测物对照品进行测定。如不能得到制剂辅料的全部组分,可向待测制剂中加人已知量的被测物对照品进行测定,或用所建立方法的测定结果与已知准确度的另一种方法测定结果进行比较。准确度也可由所测定的精密度、线性和专属性推算出来。 2.化学药杂质定量测定的准确度 可向原料药或制剂处方量空白辅料中加人已知量杂质进行测定。如不能得到杂质或降解产物对照品,可用所建立方法测定的结果与另一成熟的方法进行比较,如药典标准方法或经过验证的方法。在不能测得杂质或降解产物的校正因子或不能测得对主成分的相对校正因子的情况下,可用不加校正因子的主成分自身对照法计算杂质含量。应明确表明单个杂质和杂质总量相当于主成分的重量比(%) 或面积比(% )。 3.中药化学成分测定方法的准确度 可用对照品进行加样回收率测定,即向已知被测成分含量的供试品中再精密加人一定量的被测成分对照品,依法测定。用实测值与供试品中含有量之差,除以加入对照品量计算回收率。在加样回收试验中须注意对照品的加人量与供试品中被测成分含有量之和必须在标准曲线线性范围之内;加入对照品的量要适当,过小则引起较大的相对误差,过大则干扰成分相对减少,真实性差。 回收率:%= (C - A ) /S X 100% 式中:A为供试品所含被测成分量;B 为加入对照品量; C 为实测值。 4.校正因子的准确度 对色谱方法而言,绝对(或定量)校正因子是指单位面积的色谱峰代表的待测物质的量。待测定物质与所选定的参照物质的绝对校正因子之比,即为相对校正因子。相对校正因子计算法常应用于化学药有关物质的测定、中药材及其复方制剂中多指标成分的测定。校正因子的表示方法很多,本指导原则中的校正因

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

化学药物质量控制分析方法验证技术指导原则

化学药物质量控制分析方法验证技术指导原则【】HGPH 5-1指导原则编号: 化学药物质量控制分析方法验证 技术指导原则 二??四年十一月 目录 一、概 述 ..................................................................... ............................................ 1 二、方法验证的一般原 则 ..................................................................... ................ 2 三、方法验证涉及到的三个主要方 面 (2) ,一,需要验证的检测项 目 ..................................................................... . (2) ,二,分析方 法 ..................................................................... .. (3) ,三,验证内 容 ..................................................................... .......................... 3 四、方法验证的具体内 容 ..................................................................... . (3)

,一,专属 性 ..................................................................... (3) 1、鉴别反 应 ..................................................................... (3) 2、杂质检 查 ..................................................................... (4) 3、含量测 定 ..................................................................... (4) ,二,线 性 ..................................................................... . (5) ,三,范 围 ..................................................................... . (5) 1、含量测 定 ..................................................................... (5) 2、制剂含量均匀度...................................................................... (5)

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.

有关物质分析方法验证技术指导原则

摘要:本文介绍了在对有关物质检查所用的分析方法进行方法学验证时,各项指标的可接受标准,以利于判断该分析方法的可行性。 关键词:有关物质检查分析方法验证可接收标准 药品中的有关物质泛指在药品的生产与储存过程中产生的工艺杂质或降解产物。由于这些有关物质的存在会影响到药品的纯度,进而可能会产生毒副作用,所以有关物质的控制是药品研发的一个重要方面,也是我们在药品审评中一直重点关注的要点之一。而要对有关物质进行严格的控制,就离不开专属性强、灵敏度高的分析方法,这就涉及到分析方法的筛选与验证。从现有的申报资料看,药品研发单位已基本上意识到分析方法验证的重要性,但是对验证时各具体指标是否可行尚没有一个明确的可接受标准,从而难以对验证结果进行评判。为解决这一问题,本文结合国外一些大型药品研发企业在此方面的要求,提出了在对有关物质检查方法进行验证时的可接受标准,供国内的药品研发单位在进行研究时参考。 1.准确度 该指标主要是通过回收率来反映。验证时一般要求根据有关物质的定量限与质量标准中该杂质的限度分别配制三个浓度的供试品溶液各三份(例如某杂质的限度为0.2%,则可分别配制该杂质浓度为0.1%、0.2%和0.3%的杂质溶液),分别测定其含量,将实测值与理论值比较,计算回收率,并计算9个回收率数据的相对标准差

(RSD)。 该项目的可接受的标准为:各浓度下的平均回收率均应在80%-120%之间,如杂质的浓度为定量限,则该浓度下的平均回收率可放宽至70%-130%,相对标准差应不大于10%。 2.线性 线性一般通过线性回归方程的形式来表示。具体的验证方法为:在定量限至一定的浓度范围内配制6份浓度不同的供试液,分别测定该杂质峰的面积,计算相应的含量。以含量为横坐标(X),峰面积为纵坐标(Y),进行线性回归分析。 可接受的标准为:回归线的相关系数(R)不得小于0.990,Y轴截距应在100%响应值的25%以内,响应因子的相对标准差应不大于10%。 3.精密度 1)重复性 配制6份杂质浓度(一般为0.1%)相同的供试品溶液,由一个分析人员在尽可能相同的条件下进行测试,所得6份供试液含量的相对标准差应不大于15%。 2)中间精密度 配制6份杂质浓度(一般为0.1%)相同的供试品溶液,分别由两个分析人员使用不同的仪器与试剂进行测试,所得12个含量数据的相对标准差应不大于20%。 4.专属性 可接受的标准为:空白对照应无干扰,该杂质峰与其它峰应能完

液相色谱仪的原理和分析方法

液相色谱仪的原理及分析方法 高效液相色谱法是在经典色谱法的基础上,引用了气相色谱的理论,在技术上,流动相改为高压输送(最高输送压力可达4.9′107Pa);色谱柱是以特殊的方法用小粒径的填料填充而成,从而使柱效大大高于经典液相色谱(每米塔板数可达几万或几十万);同时柱后连有高灵敏度的检测器,可对流出物进行连续检测。 特点: 1.高压:液相色谱法以液体为流动相(称为载液),液体流经色谱柱,受到阻力较大,为了迅速地通过色谱柱,必须对载液施加高压。一般可达150~350×105Pa。 2. 高速:流动相在柱内的流速较经典色谱快得多,一般可达1~10ml/min。高效液相色谱法所需的分析时间较之经典液相色谱法少得多,一般少于1h 。 3. 高效:近来研究出许多新型固定相,使分离效率大大提高。 4.高灵敏度:高效液相色谱已广泛采用高灵敏度的检测器,进一步提高了分析的灵敏度。如荧光检测器灵敏度可达10-11g。另外,用样量小,一般几个微升。 5.适应范围宽:气相色谱法与高效液相色谱法的比较:气相色谱法虽具有分离能力好,灵敏度高,分析速度快,操作方便等优点,但是受技术条件的限制,沸点太高的物质或热稳定性差的物质都难于应用气相色谱法进行分析。而高效液相色谱法,只要求试样能制成溶液,而不需要气化,因此不受试样挥发性的限制。对于高沸点、热稳定性差、相对分子量大(大于400 以上)的有机物(这些物质几乎占有机物总数的75% ~80% )原则上都可应用高效液相色谱法来进行分离、分析。据统计,在已知化合物中,能用气相色谱分析的约占20%,而能用液相色谱分析的约占70~80%。 高效液相色谱按其固定相的性质可分为高效凝胶色谱、疏水性高效液相色谱、反相高效液相色谱、高效离子交换液相色谱、高效亲和液相色谱以及高效聚焦液相色谱等类型。用不同类型的高效液相色谱分离或分析各种化合物的原理基本上与相对应的普通液相层析的原理相似。其不同之处是高效液相色谱灵敏、快速、分辨率高、重复性好,且须在色谱仪中进行。 高效液相色谱法的主要类型及其分离原理

化学药物质量控制分析方法验证技术指导原则

指导原则编号: 【H】G P H 5 -1 化学药物质量控制分析方法验证技术指导原则 (第二稿) 二OO四年三月十九日

目录 一、概述 (4) 二、方法验证的一般原则 (5) 三、方法验证涉及到的三个要素 (5) 1、需要验证的检测项目 (5) 2、分析方法 (6) 3、验证内容 (7) 四、方法验证的具体内容 (7) (一)专属性 (7) 1、鉴别反应 (7) 2、杂质检查 (7) 3、含量测定 (8) (二)线性 (8) (三)范围 (9) 1、含量测定 (9) 2、制剂含量均匀度 (9) 3、溶出度或释放度 (9) 4、杂质 (10) (四)准确度 (10) 1、含量测定 (10) 2、杂质定量试验 (10)

(五)精密度 (11) 1、重复性 (11) 2、中间精密度 (12) 3、重现性 (12) (六)检测限 (12) 1、直观法 (12) 2、信噪比法 (12) (七)定量限 (13) 1、直观法 (13) 2、信噪比法 (13) (八)耐用性 (14) (九)系统适用性试验 (14) 五、方法再验证 (15) 六、方法验证的评价 (16) 1、有关方法验证评价的一般考虑 (16) 2、方法验证的整体性和系统性 (16) 七、参考文献 (17) 八、起草说明 (18) 九、著者 (20)

质量控制分析方法验证的技术指导原则 一、概述 保证药品安全、有效、质量可控是药品研发和评价应遵循的基本原则,其中,对药品进行质量控制是保证药品安全有效的基础和前提。为达到控制质量的目的,需要多角度、多层面来控制产品质量,也就是说要对药物进行多个项目测试,来全面考察产品质量。一般地,每一测试项目可选用不同的分析方法,为使测试结果准确、可靠,必须对所采用的分析方法的科学性、准确性和可行性进行验证,以充分表明分析方法符合测试项目的目的和要求,这就是通常所说的对方法进行验证。 方法验证的目的是判断采用的分析方法是否科学、合理,是否能有效控制产品的内在质量。从本质上讲,方法验证就是根据检测项目的要求,预先设置一定的验证内容,并通过设计合理的试验来验证所采用的分析方法是否满足检测项目的要求。 方法验证在分析方法建立过程中具有重要的作用,并成为质量研究和质量控制的组成部分。只有经过验证的分析方法才能用于控制产品质量,因此方法验证是制订质量标准的基础。方法验证是药物研究过程中的重要内容。 本指导原则重点探讨方法验证的本质,将分析方法验证的要求与所要达到的目的结合起来进行系统和规律性的阐述,重点阐述如何科学合理地进行论证方案的设计。

高效液相色谱法的计算方法

高效液相色谱法的计算方法 高效液相色谱法是用高压输液泵将具有不同极性的单一溶剂或不同比例的混合溶剂、缓冲液等流动相泵入装有固定相的色谱柱,经进样阀注入供试品,由流动相带入柱内,在柱内各成分被分离后,依次进入检测器,色谱信号由记录仪或积分仪记录。 1、对仪器的一般要求 所用的仪器为高效液相色谱仪。色谱柱的填料和流动相的组分应按各品种项下的规定。常用的色谱柱填料有硅胶和化学键合硅胶。后者以十八烷基硅烷键合硅胶最为常用,辛基键合硅胶次之,氰基或氨基键合硅胶也有使用;离子交换填料,用于离子交换色谱;凝胶或玻璃微球等,用于分子排阻色谱等。注样量一般为数微升。除另有规定外,柱温为室温,检测器为紫外吸收检测器。 在用紫外吸收检测器时,所用流动相应符合紫外分光光度法(附录ⅣA)项下对溶剂的要求。 正文中各品种项下规定的条件除固定相种类、流动相组分、检测器类型不得任意改变外,其余如色谱柱内径、长度、固定相牌号、载体粒度、流动相流速、混合流动相各组分的比例、柱温、进样量、检测器的灵敏度等,均可适当改变,以适应具体品种并达到系统适用性试验的要求。一般色谱图约于20分钟内记录完毕。 2、系统适用性试验 按各品种项下要求对仪器进行适用性试验,即用规定的对照品对仪器进行试验和调整,应达到规定的要求;或规定分析状态下色谱柱的最小理论板数、分离度和拖尾因子。 (1)色谱柱的理论板数(N,用于定量表示色谱柱的分离效率,简称柱效)。 在选定的条件下,注入供试品溶液或各品种项下规定的内标物质溶液,记录色谱图,量出供试品主成分或内标物质峰的保留时间tR(以分钟或长度计,下同,但应取相同单位)和半高峰宽(W h/2),按n=5.54(t R/Wh/2)2计算色谱柱的理论板数,如果测得理论板数低于各品种项下规定的最小理论板数,应改变色谱柱的某些条件(如柱长、载体性能、色谱柱充填的优劣等),使理论板数达到要求。 (2) 分离度(R)

药品质量标准分析方法验证指导原则样本

药品质量标准分析方法验证指导原则 《中国药典》 药品质量标准分析方法验证的目的是证明采用的方法适合于相应检测要求。在建立药品质量标准时, 分析方法需经验证; 在药品生产工艺变更、制剂的组分变更、原分析方法进行修订时, 则质量标准分析方法也需进行验证。方法验证理由、过程和结果均应记载在药品质量标准起草说明或修订说明中。生物制品质量控制中采用的方法包括理化分析方法和生物学测定方法, 其中理化分析方法的验证原则与化学药品基本相同, 因此可参照本指导原则进行, 但在进行具体验证时还需要结合生物制品的特点考虑; 相对于理化分析方法而言, 生物学测定方法存在更多的影响因素, 因此本指导原则不涉及生物学测定方法验证的内容。 验证的分析项目有: 鉴别试验、限量或定量检查、原料药或制剂中有效成分含量测定, 以及制剂中其它成分( 如防腐剂等, 中药中其它残留物、添加剂等) 的测定。药品溶出度、释放度等检查中, 其溶出量等的测定方法也应进行必要验证。 验证指标有: 准确度、精密度( 包括重复性、中间精密度和重现性) 、专属性、检测限、定量限、线性、范围和耐用性。在分析方法验证中, 须采用标准物质进行试验。由于分析方法具有各自的特点, 并随分析对象而变化, 因此需要视具体方法拟订验证的指标。表1中列出的分析项目和相应的验证指标可供参考。

一、准确度 准确度系指采用该方法测定的结果与真实值或参考值接近的程度, 一般用回收率( %) 表示。准确度应在规定的范围内测定。 1.化学药含量测定方法的准确度 原料药采用对照品进行测定,或用本法所得结果与已知准 确度的另一个方法测定的结果进行比较。制剂可在处方量空白辅料中, 加入已知量被测物对照品进行测定。如不能得到制剂辅料的全部组分, 可向待测制剂中加入已知量的被测物对照品进行测定, 或用所建立方法的测定结果与已知准确度的另一种方法测定结果进行比较。 准确度也可由所测定的精密度、线性和专属性推算出来。 2.化学药杂质定量测定的准确度 可向原料药或制剂处方量空白辅料中加入已知量杂质进行测定。如不能得到杂质或降解产物对照品, 可用所建立方法测定的结果与另一成熟的方法进行比较, 如药典标准方法或经过验证的方法。在不能测得杂质或降解产物的校正因子或不能测得对主成分的相对校正因子的情况下, 可用不加校正因子的主成分自身对照法计算杂质含量。应明确表明单个杂质和杂质总量相当于主成分的重量比( %) 或面积比( %) 。 3.中药化学成分测定方法的准确度

相关文档
相关文档 最新文档