introduction, objectives, and key requirements...

33
1 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS 1.1 INTRODUCTION 1.1.1 Good Laboratory Practices Good laboratory practices (GLPs 21 CFR PART 58) is a standard by which laboratory studies are designed, implemented, and reported to assure the public that the results are accurate/reliable and the experiment can be reproduced accordingly [1], at any time in the future. In less technical terms, GLP is the cornerstone of all laboratory- based activities in any organization that prides itself on the quality of the work it performs. And, despite its immediate association with the pharmaceutical sector, GLPs can (and should) be applied to virtually all industries in which laboratory work is conducted, including companies involved in drug development, manufacturing, foods, pesticides (agrochemicals), drink production, and engineering testing. In addition, commercial testing laboratories (for toxicology, metabolism, materials, and safety, for example), research establishments, and universities—in fact, all laboratories engaged in product or safety testing or research and development— should adopt and apply the doctrines of GLP. GLP is not a luxury. It is a necessity for any professional laboratory wishing to gain and retain the respect of its employees, clients, regulators, and perhaps most importantly, its competitors. If a company is seen to be applying and adhering to the highest standards of laboratory practice, it will gain significant competitive advantage and will compete successfully for business and recognition within its Regulated Bioanalytical Laboratories: Technical and Regulatory Aspects from Global Perspectives, By Michael Zhou Copyright Ó 2011 John Wiley & Sons, Inc. 1

Upload: doanthuy

Post on 04-Feb-2018

228 views

Category:

Documents


1 download

TRANSCRIPT

1INTRODUCTION, OBJECTIVES,AND KEY REQUIREMENTSFOR GLP REGULATIONS

1.1 INTRODUCTION

1.1.1 Good Laboratory Practices

Good laboratory practices (GLPs 21 CFR PART 58) is a standard by which laboratory

studies are designed, implemented, and reported to assure the public that the results

are accurate/reliable and the experiment can be reproduced accordingly [1], at any

time in the future. In less technical terms, GLP is the cornerstone of all laboratory-

based activities in any organization that prides itself on the quality of the work it

performs. And, despite its immediate association with the pharmaceutical sector,

GLPs can (and should) be applied to virtually all industries in which laboratory work

is conducted, including companies involved in drug development, manufacturing,

foods, pesticides (agrochemicals), drink production, and engineering testing. In

addition, commercial testing laboratories (for toxicology, metabolism, materials,

and safety, for example), research establishments, and universities—in fact, all

laboratories engaged in product or safety testing or research and development—

should adopt and apply the doctrines of GLP.

GLP is not a luxury. It is a necessity for any professional laboratorywishing to gain

and retain the respect of its employees, clients, regulators, and perhaps most

importantly, its competitors. If a company is seen to be applying and adhering to

the highest standards of laboratory practice, it will gain significant competitive

advantage and will compete successfully for business and recognition within its

Regulated Bioanalytical Laboratories: Technical and Regulatory Aspects from Global Perspectives,

By Michael Zhou

Copyright � 2011 John Wiley & Sons, Inc.

1

operational environment. Conversely, without rigidly enforced GLPs, good clinical

practice (GCP) [2], good manufacturing practices (GMPs) [3], or GxPs—a scientific

organizationwill not achieve the commercial success and respect that its products and

personnel deserve.

Published GLP regulations and guidelines have a significant impact on the daily

operations of analytical and/or bioanalytical laboratories. GLP is a regulation that

enhances good analytical practice. Good analytical/bioanalytical practice is impor-

tant, but it is not enough. For example, the laboratory must have a specific

organizational structure and procedures to perform and document laboratory work.

The objective is not only quality of data but also traceability and integrity of data.

However, the biggest difference between GLP and non-GLP work is the type and

amount of documentation. GLP functions as a regulation, which deals with the

specific organizational structure and documents related to laboratory work in order to

maintain integrity and confidentiality of the data. The entire cost of GLP-based work

is about 40% or more additional (from case to case) when compared to non-GLP

operations. For aGLP inspector, it should be possible to look at the documentation and

to easily find out the following:

. Who has done a study

. How the experiment was carried out

. Which procedures have been used, and

. Whether there has been any problem and if so

. How it has been addressed and solved where applicable

And this should not only be possible during and right after the study has been

finished but also 5–10 or more years later.

From worldwide perspectives, good practice rules govern drug/product develop-

ment activities in many parts of theworld.World Health Organization (WHO), which

has published documents on current good manufacturing practices (cGMPs) and

GCPs, has not previously recommended or endorsed any quality standard governing

the nonclinical phases of drug/product development. GLPs are recognized rules

governing the conduct of nonclinical safety studies, ensuring the quality, integrity, and

reliability of their data. To introduce the concepts of GLP to scientists in developing

countries, workshops onGLP have been organized in these regions. As an outcome of

the workshops (industries and regulatory bodies), it became apparent that some

formal guidance would be needed for the successful implementation of the GLP

regulations.

The first scientific working group on GLP issues was convened on November 25,

1999, in Geneva, to discuss quality issues in general and the necessity for a WHO

guidance document on GLP in particular. The working group concluded that it was

important to avoid the coexistence of two GLP standards, the Principles of good

laboratory practice of the Organization for Economic Cooperation and Development

(OECD) [4] being the internationally recognized and accepted standard, and

recommended that theOECDPrinciples be adopted byWHOforResearch&Training

2 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

in Tropical Disease (TDR) as the basis of this guidance document. The experts also

recognized the need to address quality issues in areas other than the strictly regulated

safety studies for regulatory submission, and recommended that some explanation be

included in this guidance document. The working group further recommended that

WHO/TDR should request OECD’s permission to publish the existing OECD GLP

textwith aWHOendorsement, and to supplement it with an explanatory introduction.

Classical drug development (drug life cycle) is characterized by four well-defined

stages as follows:

Stage 1: The first stage, the discovery of potential new drug products, is neither

covered by a regulatory standard, nor are studies demonstrating proof of

concept. This area may well require some international standards or guidance

documents in the future.

Stage 2: The position of GLP studies within the drug development process is

specific to the second stage. These studies are termed “nonclinical” as they are

not performed in human. Their primary purpose is safety testing. Toxicology

and safety pharmacology studies, with a potential extension to pharmacoki-

netics and bioavailability, are those studies where the compliance with GLP is

required, which is the rather restricted scope of GLP.

Stage 3: The third stage, following on from safety studies, encompasses the clinical

studies in human. Here, GCP is the basis for quality standards, ethical conduct,

and regulatory compliance. GCP must be instituted in all clinical trials from

Phase I (to demonstrate tolerance of the test drug and to define human

pharmacokinetics), through Phase II (where the dose–effect relationship is

confirmed), to Phase III (full-scale, often multicenter, clinical efficacy trials in

hundreds and thousands of patients).

Stage 4: The fourth stage is postapproval. Here the drug is registered and available

on the market. However, even after marketing, the use of the drug is monitored

through formalized pharmacovigilance procedures. Any subsequent clinical

trials (Phase IV) must also comply with GCP.

A brief summary of different stages is shown in Table 1.1.

TABLE 1.1 Stages Defined Within Discovery and Development Programs

Stage I Stage II Stage III Stage IV

Establish discovery

assessment of

compounds with

in vitro and/or

in vivo data

(not regulated

under GxP)

Demonstrate

efficacy, identify

side effects

including Tox and

assessment of

pharmacokinetics

(GLP and GCP)

Gain more data on

safety and

effectiveness in

multicenters with

thousands of

patients (GLP,

GCP, cGMP)

Monitor claims or

demonstrate new

indications;

examine special

drug–drug

interactions; assess

pharmacokinetics

(GLP, GCP, cGMP)

INTRODUCTION 3

1.1.2 Bioanalytical Laboratories—Bioanalysis

Bioanalytical laboratories have increasingly become center of excellence and

critically important in data generation for discovery, preclinical and clinical devel-

opment in life science industries. Bioanalysis is a broad term that is derived from

analytical applications to biologicalmaterials (matrices) such as human and/or animal

biological fluids and materials (blood, plasma, serum, urine, feces, tissues, etc.),

biopharmaceutical (peptides, protein, etc.), and biochemistry (DNA, RNA, organo-

nucleotides, etc.). The main focus of this book is within the aspects of liquid

chromatography–tandem mass spectrometry (LC–MS/MS) and to certain extent

of immunochemistry assays—enyzme-linked immunosorbent assays (ELISA) or

ligand-binding assays (LBAs). Bioanalysis is mainly referred to the quantitative

determination of drugs and their metabolites, and other life science products in

various sample matrices. However, it should also apply to qualitative analysis

(identification and elucidations) of drug degradants, metabolites, impurities, and

other analytes of interests. The techniques (chromatographic-based and ligand-

binding-based assays) are used very early in the drug discovery and development

process to provide support to product discovery programs on metabolite fate and

pharmacokinetics of chemicals in living cells and animals. They are referred by FDA

Guidance for Industry Bioanalytical Method Validation for chromatographic-based

and ligand-binding-based assays [5]. Their uses continue throughout the nonclinical

and clinical product development phases into postmarketing support and may

sometimes extend into clinical therapeutic monitoring. Recent developments and

industry trends for rapid sample throughput and data generation are introduced and

discussed in following chapters, together with examples of how these high throughput

needs are met in bioanalysis.

1.1.2.1 High-Throughput Bioanalytical Sample Preparation Methods and

automation strategies are authoritative reference on the current state-of-the-art in

sample preparation techniques for bioanalysis. The following related chapters focus

on high-throughput (rapid productivity) techniques and describe exactly how to

perform and automate these methodologies, including useful strategies for method

development and optimization. A thorough review of the literature is included

describing high-throughput sample preparation techniques: protein removal by

precipitation; equilibrium dialysis and ultrafiltration; liquid–liquid extraction; solid

phase extraction; and various online techniques. A schematic diagram of analytical/

bioanalytical techniques used in automation is shown in Figure 1.1.

Among the sample preparation scheme, protein precipitation (PPT) is the most

commonly used approach for a simple, fast, and unique process of removing

unwanted materials from analyte(s) of interest for analysis or in some case for

further cleanup. High selectivity and sensitivity are also imperative for bioanalytical

laboratories to deal with sample analyses with great demand in method limits of

quantitation (LOQ), wide dynamic range (linearity and range), free of interferences

(specificity and selectivity), and other highly challenging requirements such as

multiple compounds (analytes—parent drugs, prodrugs, and their degradants/

4 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

metabolites), various sample types (matrices), and different analytical techniques

including LC–MS/MS,GC–MS/MS, LC–NMR, ICP–MS, and other advanced hybrid

techniques. In addition to above analytical techniques, immunoassays (ELISA and/or

ligand-binding assays—LBAs or alike) are also widely used within bioanalytical

laboratories, especially in biopharmaceutical and biotechnology industries where

relatively large molecules are dealt such as peptides and proteins as part of

drug development compounds and applying to different therapeutic areas. Rapid

advances in chromatographic as well as ligand-binding assay technologies have

been observed to meet the needs in product research and development processes.

More details of description are elaborated on above analytical and bioanalytical

techniques as powerful methodologies in trace level qualitative and quantitative

analyses.

There have been varieties of separation and detection techniques involved in

analytical and bioanalytical methodologies as indicated in Figure 1.2. More recent

years, biomarker analysis in various therapeutic areas has become incredibly

significant in drug/product development and monitoring programs. Without any

doubt, this has increasingly become part of bioanalytical capabilities. Biomarker

Analytical/Bioanalytical Sample Prep. Chemistry and Techniques

Protein Precipitation Liquid–LiquidExtraction

Solid Phase Extraction

Automation: Liquid Handling Workstations and Robots

TomtecQuadra 96 Plus

Packard MultiProbe IIEx

TecanGenesis Freedom

HamiltonSTAR

FIGURE 1.1 General schematic of analytical/bioanalytical techniques used in laboratory

operations/automation.

Analytical/Bioanalytical Separation/Detection Techniques

Spectroscopy/LBAsSpectrophotometryChromatography

GC, HPLC, CE, UV–VIS, FT-IR, NMR, MS/MS, LBAs, etc.

Small and Large Molecules

Ionic and PolarSpecies

Volatile andNonvolatile

Liquid, Gas, and Solids

FIGURE 1.2 Commonly used techniques in analytical/bioanalytical separation and

detection.

INTRODUCTION 5

measurements now support key decisions throughout the drug development process,

from lead optimization to regulatory approvals. They are essential for documenting

exposure–response relationships, specificity and potency toward themolecular target,

untoward effects, and therapeutic applications. In a broader sense, biomarkers

constitute the basis of clinical pathology and laboratory medicine. The utility of

biomarkers is limited by their specificity and sensitivity toward the drug or disease

process and by their overall variability. Understanding and controlling sources of

variability is not only imperative for delivering high-quality assay results, but

ultimately for controlling the size and expense of research studies. Variability in

biomarker measurements is affected by biological and environmental factors (e.g.,

gender, age, posture, diet, and biorhythms), sample collection factors (e.g., preser-

vatives, transport and storage conditions, and collection technique), and analytical

factors (e.g., purity of reference material, pipetting precision, and antibody speci-

ficity). The quality standards for biomarker assays used in support of nonclinical

safety studies fall under GLP (FDA) regulations, whereas, those assays used to

support human diagnostics and healthcare are established by Clinical Laboratory

Improvement Amendments (CLIAs) and Centers for Medicare &Medicaid Services

(CMSs) regulations and accrediting organizations such as the College of American

Pathologists (CAPs). While most research applications of biomarkers are not

regulated, biomarker laboratories in all settings are adopting similar laboratory

practices in order to deliver high-quality data. Because of the escalation in demand

for biomarker measurements, the highly parallel (multiplexed) assay platforms that

have fueled the rise of genomics will likely evolve into the analytical engines that

drive the biomarker laboratories of tomorrow. The role of biomarkers in drug

discovery and development has gained precedence over the years. As biomarkers

become integrated into drug development and clinical trials, quality assurance and, in

particular, assay validation become essential with the need to establish standardized

guidelines for bioanalytical methods used in biomarker measurements. New bio-

markers can revolutionize both the development and use of therapeutics but are

contingent on the establishment of a concrete validation process that addresses

technology integration and method validation as well as regulatory pathways for

efficient biomarker development. Perspective focuses on the general principles of the

biomarker validation process with an emphasis on assay validation and the collab-

orative efforts undertaken by various sectors to promote the standardization of this

procedure for efficient biomarker development. It is important to point out that

biomarker method validation is distinct from pharmacokinetic validation and routine

laboratory validation. The FDA has issued guidance for industry [5] on bioanalytical

method validation for assays that support pharmacokinetic studies that are specific for

chromatographic and ligand-binding assays, and that are not directly related to the

qualification or validation of biomarker assays.Whereas routine laboratory validation

refers to laboratories that do testing on human specimens for diagnosis, prevention, or

treatment of any disease and falls under the jurisdiction of the Clinical Laboratory

Improvement Amendments of 1988, there is little regulatory guidance on biomarker

assay validation. Hence, a “fit-for-purpose” approach for biomarker method devel-

opment and validation is derived with the idea that assay qualification or validation

6 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

should be tailored to meet the intended purpose of the biomarker study. Numerous

applications using bioanalytical techniques have generated enormous interests and

some case reveal ultimate solutions in drug efficacies and other indications that are

critical to the success in drug/product development and approval processes.

1.1.3 Good Laboratory Practices Versus Bioanalytical Labs/Bioanalysis

Recently, more and more debate and discussion around the connection between GLP

and Bioanalysis are surfaced. It is noted that there is no direct reference from GLP

regulations to bioanalysis. However, it has become common terminology and

acceptance when people refer to GLP–Bioanalysis. In a regulatory term, it may be

referred as regulated bioanalysis to support programs or studies under GLP compli-

ance. There is a misconception in some quarters that GLP is required for the conduct

of clinical studies. This is not correct. The introduction to the OECD Principles of

GLP (and the introduction to the USFDA GLPs in 21 CFR part 58) makes clear that

they apply only to the portions of nonclinical (preclinical) studies. The relevant

documents for clinical studies are the various codes of GC(R)P (e.g., ICH; TGA). The

USFDA and other registration authorities do require a demonstration of the quality of

test data from clinical studies. In the United States, this may well be by means of

conformance with Clinical Laboratories Improvement Act (CLIA) [6]. In Australia,

this is best demonstrated by the testing laboratory’s NATA accreditation (in Medical

Testing, Chemical Testing, etc.)1. Nevertheless, bioanalytical laboratories generate

data in support of clinical studies and ultimately as part of data submissions to

regulatory agencies. More detailed discussions on above techniques and guidelines

are available in respective chapters of this book. The regulatory environment in which

clinical trials are conducted continues to evolve. The changes are generally focused on

requiring more rigorous control within the organizations performing clinical trials in

order to ensure patient safety and the reliability of data produced. The global

acceptance of the ICH Guideline for GCP and the implementation of the European

Union Clinical Trials Directive (2001/20/EC) are two clear examples of such change.

For some years, it has been internationally recognized that clinical laboratories

processing specimens from clinical trials require an appropriate set of standards to

guide good practices. With that aim in mind, the Good Clinical Laboratory Practice

Guidelines [7] were drafted and published in 2003 by a working party of the Clinical

Committee of the British Association of ResearchQuality Assurance (BARQA). This

guidance identifies systems required and procedures to be followed within an

organization conducting analysis of samples from clinical trials in compliance with

the requirements of GCP. It thus provides sponsors, laboratory management, project

managers, clinical research associates (CRAs), and quality assurance personnel with

the framework for a quality system in analysis of clinical trial samples, ensuring GCP

compliance overall of processes and results.

1 The National Association of Testing Authorities (NATA)—Australia’s national laboratory accreditation

authority. NATA accreditation recognizes and promotes facilities competent in specific types of testing,

measurement, inspection, and calibration.

INTRODUCTION 7

1.2 OBJECTIVES AND KEY REQUIREMENTS FOR GLP

REGULATIONS

The ability to provide timely, accurate, and reliable data is essential to the role of

analytical and bioanalytical chemists and is especially true in the discovery, devel-

opment, and manufacture of pharmaceuticals and life science products. Analytical

and bioanalytical data are used to screen potential drug candidates, aid in the

development of drug syntheses, support formulation studies, animal PK/Tox, clinical

safety and efficacy programs, monitor the stability of bulk pharmaceuticals and

formulated products, and test final products for release. The quality of analytical and

bioanalytical data is a key factor in the success of a drug or product development

program. The process of method development and validation has a direct impact on

the quality of these data.

Although a thorough validation cannot rule out some potential problems, the

process ofmethod development and validation should address themost commonones.

Examples of typical problems that can be minimized or avoided are synthesis

impurities that coelute with the analyte peak in an HPLC assay; a particular type

of column that no longer produces the separation needed because the supplier of the

column has changed themanufacturing process; an assaymethod that is transferred to

a second laboratory where they are unable to achieve the same detection limit; and a

quality assurance audit of a validation report that finds no documentation on how the

method was performed during the validation.

Problems increase as additional people, laboratories, and equipment are used

to perform the method. When the method is used in the developer’s laboratory, a

small adjustment can usually be made to make the method work, but the flexibility

to change it is lost once the method is transferred to other laboratories or used for

official product testing. This is especially true in the pharmaceutical and life

science industries, where methods are submitted to regulatory agencies and changes

may require formal approval before they can be implemented for official testing/

intended use. The best way to minimize method problems is to perform adequate

validation experiments during development and establishment. Analysis of chemi-

cals/drugs in the complex environments/matrices in which they occur are carried out

by a vast range of institutions for a variety of purposes, from pharmaceutical and

agrochemical companies to hospital biochemistry labs and industry laboratories, from

environmental monitoring to safety and toxicity testing of new drugs/products. The

range of compounds for analysis is enormous, from naturally occurring compounds

such as vitamins to man-made chemicals from the pharmaceutical and agrochemical

industries. The following chapters offer an integrated, readable reference text

describing the full range of analytical techniques and regulatory requirements

available for such small molecules (mostly) and large molecules in an up-to-date

manner and should be useful and appeal to all involved in the rapidly growing field of

bioanalytical sciences.

. Responsibilities should be defined for the sponsor management, for the study

management, and for the quality assurance unit.

8 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

. All routine work should follow written standard operating procedures (SOPs).

. Facilities such as laboratories should be large enough and have the right

construction/facility to ensure the integrity of a study, for example, to avoid

cross contamination during implementation and processes.

. Test and control articles should have the right quality and instruments should be

calibrated and well maintained.

. People should be trained or otherwise qualified for the job.

. Raw data and other data should be acquired, processed, and archived to ensure

integrity of data.

The main objective is clearly stated within GLP regulations and guidelines—

embodies a set of principles that provides a framework forAquality system concerned

with the organizational process and the conditions underwhich laboratory studies are

planned, performed, monitored, recorded, reported, and archived. These studies are

undertaken to generate data by which the hazards and risks to users, consumers, and

third parties, including the environment, can be assessed for pharmaceuticals,

agrochemicals, veterinary medicines, industrial chemicals, cosmetics, food and feed

additives, and biocides. GLP helps assure regulatory authorities that the data

submitted are a true reflection of the results obtained during the study and can

therefore be relied upon whenmaking risk/safety assessments. GLP regulations were

established by the regulatory bodies to ensure that research submitted to them is not

only properly executed but also documented thoroughly enough so that any scientist/

organization skilled or qualified can follow the documentation and replicate the

results. The level of detail required to achieve this level of documentation is

substantial to ensure the integrity, quality, and accuracy of data for product approval.

Unfortunately most laboratories are in situations where they have had to interpret

the regulations. Procedures have been developed on an ad hoc basis, in isolation, in

response to inspections by both their company’s Quality Assurance Unit (QAU) and

regulatory bodies. Under such duress, many scientists in industry have developed

procedures to validate their instrumentation even though the same approach will

already have been applied at the instrument manufacturer’s sites. SOPs written to

accompany such validation efforts often duplicate extracts from operationmanuals—

why don’t the manufacturers provide the SOPs directly? When it comes to validating

the instrument’s application software, the person responsible has to take the man-

ufacturer’s word for it that the software has been validated and hope that supporting

documents, such as test results and source code are available to regulatory agencies

upon request, as part of basic requirements for GLP quality system and

implementation.

. To assign responsibility for sponsor management, study management, and

quality assurance.

. Standard Operating Procedures must be followed.

. Calibration and maintenance of instruments.

. Right construction of laboratories to maintain integrity of the study.

OBJECTIVES AND KEY REQUIREMENTS FOR GLP REGULATIONS 9

. Raw data should be processed and achieved.

. Employee should be well qualified and trained as per the job assigned.

1.3 FUNDAMENTAL UNDERSTANDING OF GLP REGULATIONS

AND PRINCIPLES

Scientificmeasurements (whether they pertain tomonitoring contaminants and active

ingredients in pharmaceutical products, clinical determinations of diversified func-

tional elements, characterization of forensic evidence, or testing materials for

intermediates and/or final products) are generally recognized as affecting decisions

literally concerned with life and death issues. As personal acknowledgement of

their responsibility, scientists have traditionally adopted sound laboratory practices

directed at assuring the quality of their data. However, until recently these practices

were not consistently adopted, enforced, or audited. Because of some notorious

historic examples where erroneous data have lead to tragic consequences, national

and international agencies have developed guidelines directed at various industries

(food, agriculture, pharmaceutical, clinical, environmental, etc.), which fall in the

general category of GLPs.

Good laboratory practice regulations becamepart of the regulatory landscape in the

latter part of the 1970s in response tomalpractice in R&D activities of pharmaceutical

companies and contract facilities used by them. Themalpractice included some cases

of fraud, but by far the most important aspect of poor practice was the lack of proper

management and organization of studies used to complete regulatory dossiers. The

investigations of the US Food and Drug Administration (FDA) in the toxicology

laboratories in the United States demonstrated a lack of organization and poor

management which, it was decided, could only be dealt with by imposing regulations.

These regulations are the GLP regulations. First the US FDA, then the US Environ-

mental Protection Agency (EPA), instituted GLP regulations, and eventually many

nations of the world followed suit. In 1981, the OECD also published GLP Principles

and these have now dominated the international scene—so far 30 countries (the

member states of the OECD) have signed agreements that make the OECD GLP

Principles binding on them. This effectively makes the OECD Principles an interna-

tional text. The intent ofGLPwas to regulate the practices of scientists working on the

safety testing of prospective drugs. With the obvious potential impact on consumers

andpatients recruited for clinical trials, the safety of drugsbecameakey issue andGLP

was seen as ameans of ensuring that scientists did not invent ormanipulate safety data

and a means of ensuring that GLP compliant studies are properly managed and

conducted. Hence GLP became the champion of the consumer, the regulatory

safeguard, and the guarantee that the safety data were being honestly reported to the

registration or receiving authorities as the basis of a decision whether or not to allow a

newdrug onto themarket. GLPwas imposed on the industry by regulatory authorities,

in the same way as GMP had been before, and GCP was to be afterwards.

Within the United States, federal agencies such as FDA and EPA have produced

documents defining laboratory operational requirements, which must be met so that

10 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

technical data from laboratory studies may be acceptable by those agencies for any

legal or contractual purposes. Laboratories doing business with and/or for these

agenciesmust therefore complywith the specifiedGLP regulations. Not only the issue

of GLP is obviously so crucial to modern laboratory operations, but also most

importantly because good laboratory practice is an essential ingredient for any

professional scientist, this chapter will incorporate many of the principles that are

part of GLP in contemporary laboratories. A brief summary of GLP principles is

described and presented below.

1.3.1 Elements of Good Laboratory Practices

In general, basic elements of GLP may be defined as follows (but not limited to):

. Qualification of test facility management and personnel

. Standard operating procedures

. Quality assurance program

. Qualification of facilities (e.g., bioanalytical/analytical testing facilities)

. Qualification and validation of apparatus (equipment, computers, or comput-

erized systems), materials, and reagents

. Test systems, and test and reference items

. Performance of the study and reporting of study results

. Storage and retention of records and materials

. Documentation and maintenance of records

1.3.1.1 Qualification of Test Facility Management and Personnel The test

facility (TF) management means the person(s) who has (have) the authority and

formal responsibility for the organization and functioning of the TF according to the

GLP regulations. This requires the identification of management and the need of a

job description, qualification background, and training records (CVs or resumes).

The organization has to describe in an ad hoc document the way the TF is structured.

The TF management must ensure the availability of a master schedule, appropriate

facilities, equipment, and materials for the timely and proper conduct of the study.

A statement has to be in place that identifies the individual(s) within the TF by whom

the responsibilities of management are fulfilled.

1.3.1.2 Standard Operating Procedures SOPs provide standard working tools

that can be used to document routine quality system management and technical

activities. The development and use of SOPs are an integral part of a successful quality

system as it provides individuals with the information to perform a job or complete a

project properly, and facilitates consistency in the quality and integrity of a product or

end-result. The term “SOP” may not always be appropriate and terms such as

protocols, instructions, worksheets, and laboratory operating procedures may also

be used. SOPs detail the regularly recurringwork processes that are to be conducted or

FUNDAMENTAL UNDERSTANDING OF GLP REGULATIONS AND PRINCIPLES 11

followed within an organization. They document the way activities are to be

performed to facilitate consistent conformance to technical and quality system

requirements and to support data quality. They may describe, for example, funda-

mental programmatic actions and technical actions such as analytical processes, and

processes for maintaining, calibrating, and using equipment. SOPs are intended to be

specific to the organization or facility whose activities are described and assist that

organization to maintain their quality control and quality assurance processes and

ensure compliance with governmental regulations.

1.3.1.3 Quality Assurance The primary products of any laboratory concerned

with qualitative and quantitative analysis are the analytical data reported for speci-

mens examined by that laboratory. QA for such a laboratory includes all of the

activities associatedwith insuring that chemical and physicalmeasurements aremade

properly, interpreted correctly, and reported with appropriate estimates of error and

confidence levels. QA activities also include thosemaintaining appropriate records of

specimen/sample origins and history (sample tracking), as well as procedures, raw

data, and results associated with each specimen/sample. The various elements of

Quality Assurance are itemized here: (1) SOPs; (2) instrumentation validation; (3)

reagent/materials certification; (4) analyst qualification/certification; (5) lab facilities

qualification/certification; and (6) specimen/sample tracking.

Many volumes could be written regarding each of the QA elements itemized

above. However, a brief discussion is presented here. SOPs are what the name

implies . . . procedures which have been tested and approved for conducting a

particular determination. Often, these procedures will have been evaluated and

published by the regulatory agency involved (e.g., EPA or FDA); these agencies

may not accept analytical data obtained by other procedures for particular analytes.

Within the context of laboratory work, the experimental procedures provided in

LaboratoryManual correspond to the SOPs.Within any commercial laboratory, SOPs

should be either available or developed to acceptable standards, so that any analytical

data collected and reported can be tied according to a documented procedure.

Presumably, this implies that a given determination can be repeated at any later

time, for an identical specimen, using the SOP specified.

1.3.1.4 Qualification/Certification of Laboratory Facilities These are normally

done by some external agency. For example, an analytical and bioanalytical labo-

ratory might be audited by representatives of a federal agency with which they have a

contract. An independent laboratorymight file documentationwith a responsible state

or federal agency. The evaluation is concerned with such issues as space (amount,

quality, and relevance), ventilation, equipment, storage, and hygiene. Routine

chemistry laboratories are generally evaluated by the American Chemical Society,

as part of the process of granting approval for the overall chemistry programpresented

by the college or university. This latter approval process is not as detailed regarding

analytical facilities as the certification processes pursued by agencies, concerned

specifically with quality assurance.

12 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

1.3.1.5 Instrumentation/Apparatus Qualification and Validation It is a process

inherently necessary for any analytical and bioanalytical laboratory. Data produced

by “faulty” instruments may give the appearance of valid data. These events are

particularly difficult to detect with modern computer-controlled systems, which

remove the analyst from the data collection/instrument control functions. Thus, it

is essential that some objective procedures be implemented for continuously asses-

sing the validity of instrumental data. These procedures, when executed on a regular

basis, will establish the continuing acceptable operation of laboratory instruments

within prescribed specifications.

1.3.1.6 Reagent/Materials Certification It is an obvious element of quality

assurance. However, GLP guidelines emphasize that certification must follow

accepted procedures, and must be adequately documented. Moreover, some guide-

lineswill specify that each container for laboratory reagents/materialsmust be labeled

with information related to its certification value, date, and expiration time. This

policy is meant to assure that reagents used are as specified in the SOPs.

1.3.1.7 Qualification/Certification of Analysts (Quality Personnel) This is a

required part of QA. Some acceptable proof of satisfactory training and/or compe-

tence with specific laboratory procedures must be established for each analyst.

Because the American Chemical Society does not currently have a policy regarding

“certification” of chemists or analysts, the requirements for “certification” vary, and

are usually prescribed by the laboratory in question. These standardswould have to be

accepted by any agency or client obtaining results from that laboratory. For routine

laboratory, the requirement for certification as an analyst is satisfactory completion of

the predefined assignments (specified by relevant SOPs). Execution of these basic

procedures will be repeated, if necessary, until satisfactory results are obtained

(evaluated based on analytical accuracy and precision).

1.3.1.8 Specimen/SampleTracking This is an aspect of quality assurance that has

received a great deal of attention with the advent of computer-based Laboratory

Information Management Systems (LIMSs). However, whether done by hand with

paper files, or by computer with modern bar-coding techniques, sample tracking is a

crucial part of quality assurance. The terms “specimen” and “sample” are often used

interchangeably. However, “specimen” usually refers to an item to be characterized

chemically; whereas “sample” usually refers to a finite portion of the specimen,which

is taken for analysis. When the specimen is homogeneous (such as a stable solution),

the sample represents the overall composition of the specimen. However, if specimens

are heterogeneous (e.g., metal alloys, rock, soil, textiles, foods, polymer composites,

vitamin capsules, etc.), then the samples may not represent the overall compositions.

Maintaining the distinction in records of analytical results can be crucial to the

interpretation of data.

Procedures for assuring adequate specimen/sample tracking will vary among

laboratories. The bottom line, however, is that these procedures must maintain the

FUNDAMENTAL UNDERSTANDING OF GLP REGULATIONS AND PRINCIPLES 13

unmistakable connection between a set of analytical data and the specimen and/or

samples from which they were obtained. In addition, the original source of the

specimen/sample(s) must be recorded and likewise unmistakably connected with the

set of analytical data. Finally, in many cases the “chain-of-custody” must be specified

and validated. This is particularly true for forensic samples (related to criminal

prosecution), but can also be essential for many other situations as well. For example,

a pharmaceutical companydeveloping a newproductmay be called upon at some time

to defend their interpretation of clinical trial tests. Such defense may require the

company to establish that specimens collected during these trials could not have been

deliberately tampered. That is, they may have to establish an unbroken chain-of-

custody, which would remove all doubt regarding the integrity of specimens

submitted to sample analysis.

1.3.1.9 Performance of the Study and Reporting of Study Results A critical

process to the success and quality of a study and data generated within a study. For

each study, a written protocol/plan should exist prior to the initiation of the study.

Acceptance criteria should be defined and followed. Deviations (e.g., amendments)

from the study plan and criteria should be justified, described, acknowledged,

approved (if necessary/applicable), and documented during the process/execution.

Upon completion of a study, results should be reported in a timely manner. Study

reports should be prepared in a format that is compliant with GLP requirements

(including GLP compliance statement where applicable) and signed/approved by

Study Director, which is a formal record to confirm that the study was/is conducted

and reported in accordance with GLP, clearly identifying, as appropriate, where the

study deviated from GLP. The GLP compliance statement should not be confused

with the QA statement, also presented in the study report, which is a distinct and

separate record of QA study monitoring.

1.3.1.10 Documentation and Maintenance of Records A central feature of GLP

guidelines is documentation along with the maintenance of records of specimen/

sampleorigins, chain-of-custody, rawanalyticaldata, processedanalyticaldata,SOPs,

instrument validation results, reagent certification results, analyst certification docu-

ments, etc. Maintenance of instrument and reagent certification records provides for

postevaluation of results, even after the passage of several years. Maintenance of all

records specified provides documentation, whichmay be required in the event of legal

challenges due to repercussions of decisions based on the original analytical results.

So important is this record-keeping feature of GLP that many vendors are now

providing many of these capabilities as part of computer packages for operating

modern instruments. For example, many modern computer-based instruments will

provide for the indefinite storage of raw analytical data for specific samples in a

protected (tamper proof) environment. They also provide for maintenance of his-

torical records of control chart data establishing the operational quality of instruments

in any period during which analytical data have been acquired by that instrument.

The length of time over which laboratory records should be maintained will vary

with the situation. However, the general guidelines followed in regulated laboratories

14 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

are to maintain records for at least 5 years. In practice, these records are being

maintained much longer. The development of higher density storage devices for

digitized data is making this kind of record-keeping possible. The increasing

frequency of litigation regarding chemistry-related commercial products is making

this kind of record-keeping essential. Moreover, establishing the integrity of the

stored data is becoming a high-level security issue for companies concerned about

future litigation.

1.3.1.11 Accountability GLP procedures inherently establish accountability for

laboratory results. Analysts, instruments, reagents, and analytical methods cannot

(and should not) maintain the anonymity that might be associated with a lack of GLP

policy. Responsibility for all aspects of the laboratory processes leading to technical

results and conclusions is clearly defined and documented. This situation should place

appropriate pressure on analysts to conduct studies with adequate care and concern.

Moreover, it allows the possibility of identifying more quickly and succinctly the

source(s) of error(s) and taking corrective action to maintain acceptable quality of

laboratory data.

The OCED GLP Principles simply state that the fundamental points of GLP

help the research to perform his/her work in compliance with reestablished plan

and standardized procedures worldwide. The regulations/principles do not con-

cern the scientific or technical content of the research programs. All GLP texts,

whatever their origins or the industry targeted, stress the importance of the

following points:

(1) Resources: Organization, personnel, facilities, and equipment.

(2) Rules: Protocols and written procedures.

(3) Characterization: Test items and test systems.

(4) Documentation: Raw data, final report, and archives.

(5) Quality assurance unit.

The training program of the WHO takes each of these five fundamental points in

turn and explains the rules of GLP in each case. The major points are summarized

here.

(1) Resources (Organization and Personnel): GLP regulations require that the

structure of the research organization and the responsibilities of the research

personnel be clearly defined. GLP also stresses that staffing levels must be

sufficient to perform the tasks required. The qualifications and the training

of staff must also be defined and documented. Facilities and Equipment—

The regulations emphasize the need for sufficient facilities and equipment in

order to perform the studies. All equipment must be in working order. A strict

program of qualification, calibration, and maintenance attains this.

(2) Rules (Protocols and Written Procedures): The main steps of research

studies are described in the study plan or protocol. However, the protocol

FUNDAMENTAL UNDERSTANDING OF GLP REGULATIONS AND PRINCIPLES 15

does not contain all the technical details necessary to exactly repeat the study.

Since being able to repeat studies and obtain similar results is a sine qua non of

mutual acceptance of data (and, indeed, a central tenet in the scientific

method), the routine procedures are described in written SOPs. Laboratories

may also need to standardize certain techniques to facilitate comparison of

results; here again written SOPs are an invaluable tool.

(3) Characterization: In order to perform a study correctly, it is essential to know

as much as possible about the materials used during the study. For studies to

evaluate the properties of pharmaceutical compounds during the preclinical

phase, it is a prerequisite to have details about the test item and about the test

system (often an animal or plant) to which it is administered.

(4) Documentation (Raw Data): All studies generate raw data. These are the

fruits of research and represent the basis for establishing results and arriving at

conclusions. The raw data must also reflect the procedures and conditions of

the study. Final Report—The study report, just like all other aspects of the

study, is the responsibility of the study director. He/she must ensure that the

contents of the report describe the study accurately. The study director is also

responsible for the scientific interpretation of the results. Archives—Storage

of records must ensure safekeeping for many years, coupled with logical and

prompt retrieval.

(5) Quality Assurance: QA as defined by GLP is a team of persons charged with

assuring management that GLP compliance has been attained within the

laboratory. They are organized independently of the operational and study

program, and function as witnesses to the whole preclinical research

process.

1.4 KEY ELEMENTS OF BIOANALYTICAL METHODS VALIDATION

It is apparent that the quality of bioanalytical data is critical to supporting regulatory

filing and approval process. BMV employed for the quantitative determination of

drugs and their metabolites in biological fluids plays a significant role in the

evaluation and interpretation of BA, bioequivalence (BE), PK, and toxicokinetic

(TK) study data. The quality of these studies is directly related to the quality and

integrity of the underlying bioanalytical data. It is therefore important that guiding

principles for the validation of these analytical methods be established and dissem-

inated to the pharmaceutical and life sciences communities.

FDA Bioanalytical Method Validation Guidance for Industry (May 2001) [5]

provides assistance to sponsors of investigational new drug (INDs) applications,

new drug applications (NDAs), abbreviated new drug applications (ANDAs), and

supplements indevelopingbioanalyticalmethodvalidation informationused inhuman

clinical pharmacology,BA,andBEstudies requiringPKevaluation.Theguidancealso

applies tobioanalyticalmethodsused fornonhumanpharmacology/toxicologystudies

16 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

and nonclinical studies. For studies related to theveterinary drug approval process, the

guidance applies only to blood and urine in BA, BE, and PK studies.

The information in the guidance generally applies to bioanalytical procedures such

as gas chromatography (GC), high-performance liquid chromatography (HPLC),

combinedGC and LCmass 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. The

guidance also applies to other bioanalytical methods, such as immunological and

microbiological procedures, and to other biological matrices, such as tissue, skin,

feces, and other samples/specimens. The guidance provides general recommenda-

tions for bioanalytical method validation. The recommendations may be adjusted or

modified depending on the specific type of analytical method for intended use. The

guidance should be an excellent reference to other similar method validation in life

science industries.

Selective and sensitive analytical methods for the quantitative evaluation of drugs

and their metabolites (analytes of interest) are critical for the successful conduct of

nonclinical and/or biopharmaceutics and clinical pharmacology studies. Bioanaly-

tical method validation includes all of the procedures that demonstrate that a

particular method used for quantitativemeasurement of analytes in a given biological

matrix, such as blood, plasma, serum, or urine, is reliable and reproducible for the

intended purpose, scope, and use. The fundamental parameters for validation include:

(1) accuracy, (2) precision, (3) selectivity, (4) sensitivity, (5) reproducibility, (6)

stability, and (7) ruggedness and robustness (not clearly addressed within the

Guidance). 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 or meet 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:

Full Validation: It is important when developing and implementing a bioanaly-

tical method for the first time or is considered to be method official establish-

ment? 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.

Partial Validations: These are modifications of already validated bioanalytical

methods. Partial validation can range from as little as one intra-assay accuracy

KEY ELEMENTS OF BIOANALYTICAL METHODS VALIDATION 17

and precision determination to a nearly full 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; and

. selectivity demonstration of an analyte in the presence of specificmetabolites.

Typical recommendation of Method Partial and Full Validation is given in

Table 1.2.

Cross-Validation: It 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.

TABLE 1.2 Typical Recommendation of Method Partial and Full Validation

Items Full Validation Partial Validation

Different matrices þExtend dynamic range þAdd metabolite(s) þReduce matrix volume þCheck for comed þDifferent anticoagulant þLLOQ þChange analysts þChange instruments þChange extraction (mechanism) þChange detection system þChange chromatography þPlease note that some of the partial validation may be up to a full validation (e.g., anticoagulant with

different types and chromatographic conditions).

18 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

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 consideredwhen data

generated using different analytical techniques (e.g., LC–MS/MS versus LBAs)

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 nonclinical studies for regulatory submissions should adhere to FDA’s GLPs

(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 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 generated

and reported. The SOPs also should include record keeping, security and chain of

sample custody (accountability systems that ensure integrity of test articles), sample

preparation, and analytical tools such as methods, reagents, equipment, instrumen-

tation, and procedures for quality control and verification of results. Here are typical

recommendations of what need to be performed as for a full validation or a partial

validation.

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 sample analysis and

acceptance criteria for the analytical run and/or batch. These three processes are

described below.

1.4.1 Reference Standards

Analysis of drugs and their metabolites in a biological matrix is carried out using

samples spiked with calibration (reference) standards and using 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 noncom-

mercial establishment. The source and lot number, expiration date, certificates of

analyseswhen available, and/or internally or externally generated evidence of identity

and purity should be furnished for each reference standard.

KEY ELEMENTS OF BIOANALYTICAL METHODS VALIDATION 19

1.4.2 Method Development—Chemical/Chromatographic Assay

The method development and establishment phase defines the chemical assay. The

fundamental parameters for a bioanalyticalmethod 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 establish-

ment for a bioanalytical method include determination of (1) selectivity; (2) accuracy,

precision, recovery; (3) calibration curve; and (4) stability of analyte in spiked

samples.

Selectivity/specificity 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 biologicalmatrix (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, concom-

itant medication and other exogenous xenobiotics. If the method is intended to

quantifymore than one analyte, each analyte should be tested to ensure that there is no

interference.

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 concen-

tration. Aminimum 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 bymore than 20%. The deviation of themean from

the true value serves as the measure of accuracy.

Precision of an analyticalmethod describes the closeness of individualmeasures 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 concentra-

tions 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, intrabatch precision, or repeatability,

which assesses precision during a single analytical run, and between-run, interbatch

precision, or repeatability, which measures precision with time, and may involve

different analysts, equipment, reagents, and laboratories.

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

20 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

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.

1.4.3 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 curvewill 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 nonzero samples covering

the expected range, including LLOQ.

(1) Lower Limit of Quantification: The lowest standard on the calibration curve

should be accepted as the limit of quantification if the following conditions

are met: The analyte response at the LLOQ should be at least five times the

response compared to blank response. 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 sim-

plest model that adequately describes the concentration–response relation-

ship 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: 20% deviation of the LLOQ from nominal

concentration; 15% deviation of standards other than LLOQ from nominal

concentration. At least four out of six nonzero 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.

1.4.4 Stability

Drug stability in a biological fluid is a function of the storage conditions, the chemical

properties of the drug, thematrix, and the container system. The stability of an analyte

in a particularmatrix and container system is relevant only to thatmatrix 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

KEY ELEMENTS OF BIOANALYTICAL METHODS VALIDATION 21

collection and handling, after long-term (frozen at the intended storage tempera-

ture) 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.

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 h and

thawed unassisted at room temperature. When completely thawed, the samples

should be refrozen for 12–24 h under the same conditions. The freeze–thaw

cycle should be repeated twomore times, and then analyzed on the third cycle. If

an analyte is unstable at the intended storage temperature, the stability sample

should be frozen at -70�C during the three freeze and thaw cycles.

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 h (based on the expected duration that samples will

be maintained at room temperature in the intended study) and analyzed.

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 or more 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.

Stock Solution Stability: The stability of stock solutions of drug and the internal

standard should be evaluated at room temperature for at least 6 h. 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.

Postpreparative Stability: The stability of processed samples, including the

resident time in the auto-sampler, 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

22 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

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.

1.4.5 Reproducibility

It is part of evaluation for the closeness of the agreement between the results of

successive measurements of the same analyte in identical material made by the same

method under different conditions, for example, different operators and different

laboratories and considerably separated in time. Incurred sample reanalysis (ISR) is

highly recommended for this investigation. Results should be expressed in terms of

the reproducibility standard deviation, the reproducibility coefficient of variation or

the confidence interval of the mean value.

1.4.6 Robustness or Ruggedness

It measures the capacity of a test to remain unaffected by small variations in the

procedures. It is measured by deliberately introducing small changes to the method

and examining the consequences, which is not mentioned within BMV Guidelines,

but it is important to evaluate and establish rugged/robust methods, especially for

pivotal clinical trial sample analyses.

1.5 BASIC 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 (test 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 towhich environmental, matrix,material, or procedural variables can affect

the estimation of analyte in thematrix 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 themethod 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

BASIC PRINCIPLES OF BIOANALYTICAL METHOD VALIDATION AND ESTABLISHMENT 23

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 evaluated and established.

The evaluation of accuracy, precision, reproducibility, response function, and

selectivity of the method for endogenous substances, metabolites, and known

degradation products should be performed and 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 multi-

plexing, 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/batch size. The placement ofQC samples

should be judiciously considered in the run/batch sequences. 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.

1.5.1 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 in support of various studies. Please note

that different dynamic ranges, possibly including dilution evaluation may be neces-

sary to cover various regulated programs/studies including nonclinical toxicology

studies. 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 confi-

dence interval. The LLOQ should serve as the lowest concentration on the standard

24 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

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 (exclud-

ing blank samples). The mean value should be within 15% of the theoretical value,

except at LLOQ, where it should not deviate bymore 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 QC samples from an equivalent biological matrix.

At a minimum, three concentrations representing the entire range of the standard

curve should be studied: one within 3� the LLOQ (low QC sample), one near the

center (middle QC), and one near the upper boundary of the standard curve (high

QC—being approximately 80% of ULOQ).

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 estab-

lished. 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 or even greater

than 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-

basedmethods, however, testing six independentmatrices for interferencemay not be

absolutely 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.

1.5.1.1 Method Development for Microbiological and Ligand-Binding AssaysMany of the bioanalytical validation parameters and principles discussed above are also

applicable tomicrobiological and ligand-binding assays.However, these assays possess

some unique characteristics that should be considered during method validation.

BASIC PRINCIPLES OF BIOANALYTICAL METHOD VALIDATION AND ESTABLISHMENT 25

1.5.1.2 Selectivity Issues Aswith chromatographicmethods,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:

Interference from Substances Physiochemically Similar to the Analyte: Cross-

reactivity of metabolites, concomitant medications, or endogenous compounds

should be evaluated individually and in combinationwith the analyte of interest.

When possible, the immunoassay should be compared with a validated refer-

ence method (such as LC–MS) using incurred samples and predetermined

criteria for agreement of accuracy of immunoassay and reference method. The

dilution linearity to the reference standard should be assessed using study

(incurred) samples. Selectivity may be improved for some analytes by incor-

poration of separation steps prior to immunoassay.

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 where

applicable.

1.5.1.3 Quantification Issues Microbiological and immunoassay standard curves

are inherently nonlinear and, in general, more concentration points may be recom-

mended 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 nonzero calibrator con-

centrations, run in duplicate, are recommended. The concentration–response rela-

tionship is most often fitted to a 4- or 5-parameter logisticmodel, 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 samematrix as the study samples or in an alternatematrix 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; and (3) the use of an internal standard that is not recognized by

26 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

the antibody but can be measured by another technique. Key reagents, such as

antibody, tracer, reference standard, andmatrix 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% (four out of six) 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 (1) labeled analyte (tracer); (2) binding should be reopti-

mized; (3) performance should be verified with standard curve and QCs; and (4)

antibody. Key cross-reactivities should be checked and evaluated. 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.

1.5.1.4 Application of Validated Method to Routine Sample 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 concentration. A typical

flowchart of method development and validation is shown in Figure 1.3.

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 intended 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�LLOQ), one inmidrange, and one close to the high end of the

BASIC PRINCIPLES OF BIOANALYTICAL METHOD VALIDATION AND ESTABLISHMENT 27

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/batch. 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 sample analysis: Amatrix-based standard curve should consist of a

minimum of six standard points, excluding blanks (either single or replicate),

covering the entire range.

ResponseFunction: 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/batch. 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/similar matrix (e.g., human to

human) obviating the need to incorporate actual within-study dilution matrix

QC samples.

FIGURE 1.3 Typical flowchart of method development and validation.

28 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

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 defined or justified and 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. Reintegration data

along with original result(s) should be reported.

1.5.1.5 Documentation The validity of an analytical method should be estab-

lished 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 submis-

sion 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.

1.5.2 Acceptance Criteria for Analytical Run

It is vitally important to establish analytical run (batch) acceptance criteria to ensure

the quality of data for regulated studies. An in-house SOP specifying batch pass/fail

criteria should be written in accordance with FDA BMV Guidelines. The following

acceptance criteria should be considered for accepting an 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 (pooled one)may also be used, provided selectivity has beenverified. 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

(includingULOQ) should fall within 15%, except for LLOQ,when it should bewithin

20% of the nominal value. Values falling outside these limits can be discarded,

provided they do not change the establishedmodel (e.g., regression/weighingmodel).

Acceptance criteria for accuracy and precision should be provided for both the

intraday and intrarun experiments.

BASIC PRINCIPLES OF BIOANALYTICAL METHOD VALIDATION AND ESTABLISHMENT 29

Quality Control Samples: Quality control samples replicated (at least once) at

a minimum of three concentrations (one within 3� 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 explained and

recorded.

1.5.2.1 Reasons for Repeat Analysis

Documentation for Repeat Analyses: Documentation should include the initial

and repeat analysis results, the reported result, assay run identification, the

reason for the repeat analysis, the requestor of the repeat analysis, and the

manager authorizing reanalysis. Repeat analysis of a clinical or preclinical

sample should be performed only under a predefined SOP.

Documentation for Reintegrated Data: Documentation should include the

initial and repeat integration results, the method used for reintegration,

the reported result, assay run identification, the reason for the reintegra-

tion, the requestor of the reintegration, and the manager authorizing

reintegration. Reintegration of a clinical or preclinical sample should be

performed only under a predefined SOP. Deviations from the analysis

protocol or SOP, with reasons and justifications should be documented for

the deviations.

Summary Information Summary information should include the following: sum-

mary tables of validation reports include (but not limited to) 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 should also include a list, by protocol, of assay

methods used and so on. The protocol number, protocol title, assay type, assay

method identification code, and bioanalytical 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 assaymethod, validation

reports, and bioanalytical reports, especially when the sponsor and a contract

laboratory assign different codes).

30 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

Documentation for Method Establishment Documentation for method develop-

ment and establishment should include the following:

(1) An operational description of the analytical method (e.g., lab or test method)

(2) Evidence of purity and identity of drug standards, metabolite standards, and

internal standards used in validation experiments

(3) A description of stability studies and supporting data

(4) 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

(5) Documentation of intra- and interassay precision and accuracy

(6) In NDA submissions, information about cross-validation study data, if

applicable

(7) Legible annotated chromatograms or mass spectrograms, if applicable

(8) Any deviations from SOPs, protocols, or GLPs (if applicable), and justifica-

tions for deviations.

Application to Sample Analysis Documentation of the application of validated

bioanalytical methods to routine drug/sample analysis should include: (1) evidence of

purity and identity of drug standards, metabolite standards, and internal standards

used during routine analyses; (2) summary tables should contain information on

sample processing and storage; (3) tables should also include sample identification,

collection dates, storage prior to shipment, information on shipment batch, and

storage prior to analysis; and (4) information should include dates, times, sample

condition, and any deviation from protocols.

For summary tables of analytical runs of clinical or preclinical samples,

information should include: (1) assay run identification, date and time of analysis,

assay method, analysts, start and stop times, duration, significant equipment and

material changes, and any potential issues or deviation from the establishedmethod;

(2) equations used for back calculation of results; (3) tables of calibration curve data

used in analyzing samples and calibration curve summary data; (4) summary

information on intra- and interassay values of QC samples and data on intra- and

interassay accuracy and precision from calibration curves and QC samples used for

accepting the analytical run; (5) QC graphs and trend analyses in addition to raw

data and summary statistics are encouraged; and (6) data tables from analytical runs

of clinical or preclinical samples. Tables should include (1) assay run identification,

sample identification, raw data and back-calculated results, integration codes, and/

or other reporting codes; and (2) complete serial chromatograms from 5% to 20%

of subjects, with standards and QC samples from those analytical runs. For

pivotal bioequivalence studies for marketing, chromatograms from 20% of serially

selected subjects should be included. In other studies, chromatograms from 5% of

randomly selected subjects in each study should be included. Subjects whose

chromatograms are to be submitted should be defined prior to the analysis of any

clinical samples.

BASIC PRINCIPLES OF BIOANALYTICAL METHOD VALIDATION AND ESTABLISHMENT 31

1.5.2.2 Other Information Other information applicable to both method devel-

opment and establishment and/or to routine sample analysis could include: lists of

abbreviations and any additional codes used, including sample condition codes,

integration codes, and reporting codes.

(1) Reference lists and legible copies of any references

(2) SOPs or protocols covering the following areas:

. Calibration standard acceptance or rejection criteria

. Calibration curve acceptance or rejection criteria

. Quality control sample and assay run acceptance or rejection criteria

Acceptance criteria should be defined for reported values when all unknown

samples are assayed in duplicate. Sample code designations, including clinical or

preclinical sample codes and bioassay sample code should be documented. Infor-

mation such as sample collection, processing, storage, and repeat analyses of samples;

reintegration of samples, and so on should also be included.

Bioanalysis and the production of pharmacokinetic, toxicokinetic, and metabolic

data play a fundamental role in pharmaceutical life sciences research and develop-

ment; therefore, the data must be produced to acceptable scientific standards and

regulatory (GLP) compliance. For this reason and the need to satisfy regulatory

authority requirements, all bioanalytical methods should be properly validated for

intended purposes and uses. It is hoped that these validation guidelines not only have

taken into account the statistical arguments described in the literature but also have

regard to the practicalities of performing bioanalytical method validations for the

pharmaceutical industry in this highly competitive era and that they aid further

standardization in this field.

It is very important to note that the validation of standard or collaboratively tested

methods should not be taken for granted, no matter how impeccable the method’s

pedigree—the laboratory should satisfy itself that the degree of validation of a

particular method is adequate for the required or intended purpose, and that the

laboratory is itself able tomatch any stated performance data. There are two important

requirements in this excerpt:

(1) The standard’s method validation data are adequate and sufficient to meet the

laboratory’s method requirements.

(2) The laboratory must be able to match the performance data as described in the

standard.

The main objectives of GLP regulations/principles and Bioanalytical Method

Validation Guidelines are to help scientists obtain results that are: (1) reliable, (2)

repeatable, (3) auditable, and (4) recognized by scientists worldwide. These may also

enhance the opportunity ofLimitingwaste of resources,Ensuring high quality of data,

Acquiring comparability of results, and Deriving to mutual recognition of scientific

findings worldwide, and ultimately Securing the health and well-being of our

societies, as being LEADS concept per author’s perspectives. The fundamental

32 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

requirements for GLP are to define conditions under which studies are planned,

performed, recorded, monitored, reported, and archived based on study’s designs.

REFERENCES

1. Code of Federal Regulation. Food Drug and Cosmetic Act, 21 CFR, Part 58—Good

laboratory practice for nonclinical laboratory studies, 1978.

2. International Conference on Harmonization (ICH)/World Health Organization (WHO).

Topic E 6 (R1) Guideline for Good Clinical Practice, 1996.

3. Code of Federal Regulation. FoodDrug andCosmetic Act 21 CFR Part 210 and 211 Current

Good Manufacturing Practices, 1995–2007.

4. Organization for EconomicCo-operation andDevelopment. Principles onGoodLaboratory

Practice (as revised in 1997) OECD, Paris, 1998 (Series on principles of GLP and

compliance monitoring, No. 1, ENV/MC/CHEM(98)17).

5. USFDA Guidance for Industry: Bioanalytical Method Validation, 2001.

6. Code of Federal Regulation. Department of Health and Human Services, Centers for

Disease Control and Prevention 21 CFR, Part 493—Clinical Laboratory Improvement Act,

2004.

7. World Health Organization (WHO). Good Clinical Laboratory Practices (GCLP), 2009.

REFERENCES 33