Skip page top navigationFDA Logo--links to FDA home page Logo of and Link to start page of Office of Regulatory Affairs, U.S. Food and Drug Administration U.S. Food and Drug Administration Office of Regulatory Affairs HHS Logo and link to Department of Health and Human Services website

FDA Home Page | Compliance | Federal-State | Inspection | Import Program | Science Ref | ORA Search  Vignette Managed Content: Click here for Metadata

Science References
ORA Laboratory Manual
Previous Page| Document TOC| Next Page

DFS Pyramid Logo

Other Laboratory Operations

Food and Drug Administration

DOCUMENT NO.:

III-05

VERSION NO.:1.2

Section 5 - Analysts on Inspection

EFFECTIVE DATE: 10/01/2003 REVISED: 06/06/2008

5.3 Types of Inspections

An establishment inspection is a careful, critical, official examination of a facility to determine its compliance with laws administered by FDA.  Inspections may be used to obtain evidence to support legal action when violations of the law are found or they may be directed to obtain information on new technologies, good commercial practices or data for establishing other regulations, etc.   The kind and type of inspections conducted will normally be defined by a Center program or assignment. There are comprehensive and directed inspections.  Comprehensive inspections direct coverage to everything in the firm subject to FDA jurisdiction while a directed inspection focuses on the areas described in the Center program or assignment.

The types of inspections most analysts find themselves on are directed inspections of food or drug firms.  On occasion, analysts will accompany consumer safety officers on the inspection of cosmetics, medical devices, and animal feed firms.

5.3.1   Food Inspections

The Food, Drug, and Cosmetic Act (FD&C Act) provides protection of the public from products that may be deleterious, are unclean or decomposed, or have been exposed to insanitary conditions that may contaminate the product with filth or may render it injurious to health. A food microbiologist, entomologist, chemist or sanitarian may participate in a team inspection of food manufacturers in order to evaluate and document insanitary conditions (e.g., filth and microbiological contamination), decomposition, adulteration with pesticides and industrial chemicals, or illegal use of color or food additives.

There are specific current Good Manufacturing Practice (cGMP) regulations that must be followed by food processing facilities under 21 CFR Part 110 "Current Good Manufacturing Practice in Manufacturing, Packing or Holding Human Food".  Analysts need to review and become familiar with this section of the CFR.  In addition, analysts need to assess that the water being used in contact with the product (contact water) is safe. Processing water includes water that is used for post-harvest treatment of produce, such as washing, cooling, waxing, and product transport. Water can be a carrier of many microorganisms including pathogenic strains of Escherichia coli, Salmonella, Vibrio cholerae, Shigella, Cryptosporidium parvum, Giardia lamblia, Cyclospora cayetanensis, Toxiplasma gondii, and the Norwalk and hepatitis A viruses. Even small amounts of contamination with some of these organisms can result in foodborne illness. Reusing processing water may result in the build-up of microbial loads, including undesirable pathogens from the crop.

Good Manufacturing Practices (GMPs) for water used for food and food contact surfaces in processing facilities are in Title 21 of the Code of Federal Regulations (CFR), sections 110.37(a) and 110.80(a)(1). 21 CFR 110.19 provides an exemption from the requirements in 21 CFR part 110 for establishments engaged solely in the harvesting, storage, or distribution of raw agricultural commodities.

The analyst needs to evaluate whether food contact services are cleaned and handled properly. Food contact surfaces may be sanitized by a process that is effective in destroying or substantially reducing the numbers of microorganisms of public health concern, as well as other undesirable microorganisms, without adversely affecting the quality of the involved product or its safety for the consumer.

The analyst will also need to assure that in the facility proper precautions are in place to reduce the risk for food contamination or cross contamination, personal protection is being used, proper handling of toxic compounds is being performed, and health conditions and pests are being addressed.

Food sanitation team inspections may also involve an entomologist or food sanitarian. During a team inspection at a food warehouse, the entomologist or food sanitarian may be able to lend expertise in identifying or documenting insect infestation or rodent contamination of food products during warehouse storage. For example, documentation and identification of whole insects, excreta pellets, urine stains, insect damage, and insect and/or rodent gnawing will be needed to support certain regulatory actions.

A microbiological inspection demands a thorough understanding of the critical factors associated with the production and testing of the product being inspected. During the inspection, a microbiologist needs to fully identify the likely sources and possible routes of microbiological contamination. The microbiologist will need to document temperature abuses and delays in processing steps that will affect the product, evaluate microbial testing of the incoming product component(s) or of the finished product(s), and focus on positive findings of pathogenic microorganisms. Determine if equipment is constructed or covered to protect contents from dust and environmental contamination. Determine what equipment is present in the laboratory and if it is usable for the purpose intended. If the firm uses a consulting laboratory, determine what tests are performed and how often. Review laboratory records for the period immediately preceding the inspection.

One of the common types of team inspections a microbiologist may participate in are Low acid canned food or Acidified food manufacturer inspections. The absence of oxygen, normal room temperature storage conditions, moisture and nutrients associated with low-acid foods favors growth of Clostridium botulinum. A failure to either destroy or control (by water activity or acidification) the germination and growth of spores of Clostridium botulinum because of improper manufacture, processing, or packing may result in the production of a toxin which causes the potentially fatal food poisoning known as botulism.

Low-acid canned foods and Acidified foods are subject to all of the requirements under the Federal Food, Drug, and Cosmetic Act and the Fair Packaging and Labeling Act. These laws require that foods be safe, clean, and wholesome, and that labeling be honest and informative. The processing of Low-acid canned foods must comply with the requirements of the Good Manufacturing Practice regulations (21 CFR Part 113). The processing of Acidified foods must comply with the requirements of the Good Manufacturing Practice regulations (21 CFR Part 114).  

Sections 21 CFR 108 & 113 on Thermally Processed Low-Acid Foods Packaged in Hermetically Sealed Containers, and 21 CFR 114 Acidified Foods are of particular importance when conducting these types of inspections. 

Another type of food inspection is a HACCP inspection. FDA's Seafood Hazard Analysis Critical Control Point (HACCP) program is set forth in 21 CFR part 123. These regulations require processors of fish and fishery products to operate preventive control systems for human food safety that incorporate seven principles of HACCP. Processors must, among other things, establish "critical control points" in their operations where they can most effectively maintain the safety of their products, systematically monitor the operation of those critical control points to ensure that they are working as they should, and keep records of the results of that monitoring. Processors also must develop written HACCP plans that describe the details and operation of their HACCP systems. Each processor may tailor its HACCP system to meet its own circumstances.  

The regulations require processors to make their HACCP records and plans "for official review and copying at reasonable times" (§123.9 (c)). Finally, the regulations provide that fish and fishery products are adulterated under section 402(a)(4) of the Federal Food, Drug, and Cosmetic Act if their processor fails to have and implement a HACCP plan when one is called for, or otherwise fails to meet any of the requirements of the regulations, including allowing the official review of records (§123.6(g)). 

Another type of team inspection the analyst may participate in is a High-Risk Food inspection. High-Risk foods are foods that are susceptible to contamination by pathogenic organisms and that are essentially ready-to-eat; that is, they will not undergo a sufficient kill step by the preparer. Microbiologists are often asked to participate in cheese inspections. Some cheeses (primarily soft cheeses) have been linked to foodborne outbreaks and illnesses caused by Salmonella, Listeria monocytogenes, and Escherichia coli contamination. Consumption of Feta and Mexican-style soft cheese has been linked to a high rate of perinatal listeriosis. During the inspection, a microbiologist may be asked to review the testing of these pathogens.

5.3.2  Drug Inspections

There are several different types of drug inspections and each has a different focus.  For example, a cGMP inspection determines if the laboratory is performing analytical testing in accordance with cGMPs. The scope of this type of inspection is broad and usually encompasses all product related operations. Pre-Approval inspections determine if the laboratory is performing analytical testing in accordance with cGMPs, human and animal drug application commitments, and the data submitted in the application is verified against the raw data.  The scope of this type of inspection is narrow, and focuses solely on operations which impact the drug product mentioned in the application. Post Approval Audit inspections provide continuing coverage of approved products.

However, there are areas common to these inspections. As a chemist or microbiologist participating in a team inspection, these common areas may be used as a starting point during the inspection.  

5.3.2.1   General Areas (Chemistry and Microbiology Inspections)

The following areas are typically covered during all pharmaceutical inspections when applicable.  Depending on the assignment, not all of these general areas will be covered.  However, these are considered the backbone of the pharmaceutical laboratory inspections.

Accountability of Raw Materials and/or Samples  

  • Have an employee, preferably the person who actually works in this area and not management, explain the firm's receiving, sampling, assigning samples, and quarantine procedures.
  • Visually examine the receiving area and storage rooms.  Determine if the room is acceptable for the materials in storage or does the climate need to be controlled and if so, how.  Examine the temperature control records.
  • Review all pertinent Standard Operating Procedures (SOP) and compare with actual operations.  Review physical records to determine compliance with the SOPs for this area.

Testing

  • Examine methods with corresponding product specifications to determine acceptability.  If a product specification is in question, discuss it with the review chemist or review microbiologist for the application.  Product specifications are a Center issue and cannot be placed on a FDA-483.
  • With the aide of the Investigator, select a product and lot number(s).  Review all analytical data associated with this product.  This includes raw material testing, in-process testing, finished product testing, and stability testing. Check calculations (or spreadsheets), transcriptions, and reviewer's signatures for errors and discrepancies.
  • Ask for and review the raw data and notebooks associated with each test. Compare the raw data to the summary documents.
  • Review method validation records to determine adequacy (See Method Validation section for guidance).  Determine if the method used in the microbiology or chemistry laboratory is the same as the method that was validated.
  • Compare all analytical results with product specifications. Determine whether raw material or products with non-compliant test results were released, retested, or reworked.
  • Examine written procedures for retesting of failed product (for example, assay failure or sterility failure) and compare with actual laboratory practice.  Evaluate if the written procedure is complete and usable (See Out of Specification/Product Failure section).  
  • Ask to see all initial positive sterility test results. If a manufacturer of aseptically filled products has never found an initial positive sterility result, there may be a testing issue.
  • Microbiological testing may include an identification of colonies or isolates found during the Total Aerobic Plate Count test or enrichment testing. Review these documents.
  • If the method was validated at another site, review the method transfer documents to determine if the transfer was a success (see Method transfer section for guidance).
  • For ancillary systems, select an audit period for evaluation of water system testing and environmental monitoring of controlled areas for sterile products.
  • Review bacterial endotoxin, and bioburden testing data for parenterals, and any objectionable microorganism testing for non-sterile drugs. The amount of testing performed on non-sterile drugs will depend upon the product and its intended use. The significance of microorganisms in non-sterile pharmaceutical products should be evaluated in terms of the use of the product, the nature of the product, and the potential hazard to the user.
  • Media fill, environmental monitoring, sterility test results, and other data should be reviewed to assure the absence of slow growing organisms.
  • Determine if raw materials are periodically retested to assure continued quality.  Review the associated procedures to determine compliance.   
  • Determine if dehydrated media is being used for the preparation of media. Good practice includes the periodic challenge of prepared media.  
  • Review the methods being used for microorganism incubation to determine if they conform to those listed in approved or pending applications. Evaluate the time period used for sterility test sample incubation.

Standards/Controls

  • Visually examine standard/controls storage conditions.
  • Examine how indicator organisms are being stored.
  • Review written procedures to determine how positive and negative controls are prepared. Good practice for such testing includes the use of known terminally sterilized or irradiated samples as a system control. Alternatively, vials or ampoules filled during media fills have also been used.
  • Review the standard written procedures and compare with standard storage conditions and the use logs for compliance.
  • Determine if the firm uses secondary or in-house standards.  Evaluate whether the assay to determine potency and purity is complete and usable.
  • Determine if the secondary or in-house standards are re-assayed periodically.  Does the written procedure address how often this re-assay occurs?  Is there sufficient data to determine if this re-test period is valid? Who performs the re-testing?
  • Determine if the primary standard has expired.
  • Determine how often volumetric solutions are standardized.  Evaluate if this schedule is valid.  Determine if commercially purchased standard solutions are standardized as well. Keep in mind that commercial standards solutions are not primary standards and need to be periodically re-standardized.
  • Review standardization logs or records for completeness and compare with the written procedure for compliance.

Equipment and Facilities

  • Visually examine analytical equipment for proper maintenance and upkeep.  Determine if the calibration/qualification status meets the SOP.
  • Obtain a list of analytical equipment that is in use if possible.
  • Review instrument written procedures and compare with maintenance and calibration/qualification records for compliance.
  • Determine if the calibration/qualification written procedure is valid.  Should the instrument be calibrated/qualified daily, weekly, monthly, quarterly, yearly? Is there data to support this schedule? Does the procedure have specification limits, specific directions, and remedial action directions? 
  • Equipment should be evaluated with its intended use in mind.  For example, dissolution apparatus dedicated to paddles, may not need to be calibrated with both paddles and baskets, and an autoclave used for a specified temperature range may not have to be calibrated at all temperature ranges.
  • For sterile products the USP states, "The facility for sterility testing should be such as to offer no greater a microbial challenge to the articles being tested than that of an aseptic processing production facility". If possible and feasible, the analyst should actually observe how sterility testing is being performed by the laboratory analysts. Proper design would, therefore, include a gowning area and pass-through airlock. Environmental monitoring and gowning should be equivalent to that used for manufacturing product.
  • Begin the inspection with a review of microbiological analyses being conducted and inspect the plates and tubes of media being incubated.
  • Inspect the autoclaves used for the sterilization of media.
  • Inspect ovens used for pyrogenation and washers used for stoppers.

Stability

Drug products are to remain potent throughout their expiry.  Therefore, the firm needs to be able to show that their product still meets assay and other specifications throughout its life.  In order to do this, stability programs are set up and product is tested on a predetermined schedule to assure it is still a quality product.  This is also required by the cGMPs in 21 CFR 211.166(a). GMPs allow accelerated studies to be used to establish a tentative expiration date. However, real time stability studies are conducted at defined temperatures which reflect normal storage conditions.  Stability should cover the physical, chemical, and microbial attributes of the drug substance.  Validated stability indicating analytical procedures are to be used (See Stability Indicating Methods and Preservative Effectiveness Testing).

For real time stability studies, representative samples from a minimum of three batches are stored at the labeled temperature (e.g. room, refrigerated, frozen) for a period at least as long as the proposed expiration date.  The recommended testing schedule is quarterly the first year, semiannually the second year, and yearly thereafter; yearly testing is considered the minimum.

Once a product is approved, firms are expected to maintain a continuing stability program by placing a representative sample from at least one batch a year in a room temperature stability program.

Temperature, humidity, light, air/oxygen may need to be controlled and documented within the specifications set by the stability program protocol. The product is stored in the packaging container(s)/closure(s) intended for marketing. Product containers are stored in such a way so that the product is in contact with as much of the inner surface of the container as possible; for example, a bottle of syrup would be laid on its side or inverted so that the syrup is in contact with both the bottle and the bottle cap. 

For Active Pharmaceutical Ingredients, the retest date is the date after which a sample of the drug substance should be tested to ensure that the material is still potent for use. They are not required to have an expiration date.

  • Visually examine the products in the stability chambers.  Determine if the products are in the containers in which they will be marketed.  Determine if the product is stored properly in the chamber.  For example, liquids should be stored in both the upright and inverted positions.
  • Evaluate the temperature and humidity controls and determine if they will deliver the correct heat and moisture.  Are the controls standardized with NIST traceable temperatures devices and is there sufficient documentation to support this?
  • Review stability written procedures and protocols and determine if the firm is in compliance.
  • Review stability testing records and determine whether stability tests are performed as scheduled using stability indicating methods (See Method Validation section).
  • For sterile products, an evaluation of final product stability at the specified expiration date should also be performed.

Personnel 

  • Obtain a roster of laboratory employees.
  • Examine training records for compliance with written procedures.
  • Examine employees experience and training records and evaluate whether employees are qualified to perform job duties.
  • Ensure employees' workloads are doable.

Documentation

  • Ensure analytical test, equipment calibrations, and sample accountability are thoroughly documented.
  • Ensure analytical records are reviewed and signed off on by authorized personnel.
  • Ensure all mark outs, crossovers, and errors are properly explained, initialed and dated in accordance with SOPs and GMPs.

Standard Operating Procedures (SOPs)/Written Procedures

  • Examine laboratory SOPs and written procedures and evaluate for thoroughness and compliance with GMPs.
  • Determine if SOPs are easily found by employees.
  • Review implementation dates and sign-off dates.
  • Determine if the SOP or written procedure that is to be used is actually being used by the laboratory analysts.

Method Validation, Stability Indicating Methods, and Preservative Effectiveness Testing

There are several guidance documents written about chemistry method validation.  Note that some of these documents are still in draft form. Review of these documents is critical in order to gain an understanding of what method validation means and what the Agency's thinking is. The United States Pharmacopoeia also describes and defines the concept of method validation. 

There are seven common threads throughout these documents: accuracy, linearity, range, precision, detection limit, quantitation limit, specificity, and ruggedness/robustness.  Other factors listed in one or more of the documents include recovery, stability of solutions, and system suitability.  Each factor is clearly explained in the references listed. Remember that all of these validation factors are not needed for each and every method. The amount of validation will depend on the type of method being validated and its intended use.

Table 1    Recommended Validation Characteristics of the Various Types of Tests

Type of Tests / Characteristics ID Testing for Impurities Assay Dissolution (Measurement Only), Content/Potency Specific Tests
    Quantitative Limit    
Accuracy - + - + +4
Precision-Repeatability - + - + +4
Precision-Intermediate Precision - +1 - +1 +4
Specificity +2 + + +5 +4
Detection Limit - -3 + - -
Quantitation Limit - + - - -
Linearity - + - + -
Range - + - + -
Robustness - + -3 + +4

NOTE:

-  Signifies that this characteristic is not normally evaluated.
+  Signifies that this characteristic is normally evaluated.
1  In cases where reproducibility has been performed, intermediate precision is not needed.
2  Lack of specificity for an analytical procedure may be compensated for by the addition of a second analytical procedure.
3  May be needed in some cases.
4  May not be needed in some cases.
5  Lack of specificity for an assay for release may be compensated for by impurities testing.

For microbiology methods, it would be virtually impossible to completely validate test procedures for every organism that may be objectionable, and methods need to be tailored to different types of products. It is essential to inactivate preservatives or inhibitory substances present in order to provide a better medium for damaged or slow growing cells. Other growth parameters include lowering the temperature and increasing the incubation time, which may provide a better survival condition for damaged or slow-growing cells.

The selection of the correct neutralizing agents is largely dependent upon the preservative, inhibitory substance, and formulation of the product under evaluation. If there is growth in the enrichment broth, transfer to more selective agar media or an enrichment agar may be needed for subsequent identification. The method should optimize the recovery of all potential pathogens. There are instances when the product may enhance microbial growth and this may need to be determined for various assays.

Stability Indicating Methods and Preservative Effectiveness Testing

A stability-indicating assay accurately measures the active ingredients, without interference from degradation products, process impurities, excipients, or other potential impurities. This may be demonstrated by performing stress studies, also called forced degradation.  Stress studies expose the product and/or drug substance to acid and base hydrolysis, thermal degradation, photolysis, oxidation etc.  The stress studies should demonstrate that impurities and degradants from the active ingredient and drug product excipients do not interfere with the quantitation of the active ingredient.  Drug product stress testing (forced degradation) may not be needed when the routes of degradation and the suitability of the analytical procedures can be determined through use of the following:

  • Data from stress testing of drug substance.
  • Reference materials for process impurities and degradants.
  • Data from accelerated and long-term studies on drug substance. 
  • Data from accelerated and long-term studies on drug product.
  • Additional supportive information on the specificity of the analytical methods and on degradation pathways of the drug substance are found in literature sources.

Method Validation is also addressed in parts 211.160(a), 211.165 (e), 211.166 (a)(3) and 211.194(a)(2) of the GMPs. It is also addressed in various ICH (The International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use) documents.

There are four basic microbial tests for finished product and stability samples.

  • Sterility Tests: For products such as sterile intravenous solutions and intrathecal injections.
  • Bacterial Endotoxtin Tests (Limulus Amebocyte Lysate): Tests for the presence of endotoxins in injectable drug products and implanted medical devices. 
  • Antimicrobial Effectiveness Tests: For products containing preservatives to inhibit the growth of microorganisms such as nasal and ophthalmic products. 
  • Microbial Limits Tests: For products that must be free of certain microorganisms which may be Salmonella, E. coli, Pseudomonas, or S. aureus such as topically applied preparations. 

For sterile products, microbial preservative effectiveness testing needs to be performed during stability studies. This test is usually later replaced by chemical testing. It is important to note that for sterile products, container/closure integrity needs to be assessed not only at the beginning of the study but also at the end to demonstrate that the product remains sterile. 

Method Transfer

Method transfer occurs when a validated method is transferred from one group, site, or company to another. There needs to be a Method Transfer protocol/plan/procedure in place. This protocol outlines the testing to occur, the roles of the two laboratories, and defines the acceptable values for the transfer to be accepted.  During the inspection, the data from the transfer should be reviewed and compared to the protocol to determine if the data meets the acceptance criteria. Typical instances when method transfer occurs are from the Research and Development (R&D) laboratory to the Quality Control (QC) laboratory, Site A to Site B when a product line is moved, and Company X to Company Y when a product is purchased by another company.

Out of Specification (OOS) Results

During the course of analytical testing, there will be times when results are generated that do NOT meet product specifications.  It is imperative that the firm has a procedure in place to handle these occurrences. During the inspection, an analyst needs to obtain a list of all batches which had an OOS result as well as a list of all failure investigations performed. It is important that when reviewing data, raw data should be reviewed. Often passing results are obtained by averaging a passing result with an OOS result. 

FDA regulations require that an investigation be conducted whenever an OOS test result is obtained. The purpose of the investigation is to determine the cause of the OOS. Even if a batch is rejected based on an OOS result, the investigation is needed to determine if the result is associated with other batches of the same drug product or other products. Batch rejection does not negate the need to perform the investigation. The regulations require that a written record of the investigation be made including the conclusions of the investigation and follow-up (21 CFR 211.192).

The FDA has published a draft guidance document to assist industry and FDA investigators and analysts when reviewing OOS investigations. The concepts mentioned in the guidance document apply to laboratory testing during the manufacture of active pharmaceutical ingredients, excipients, and other components and the testing of finished products to the extent that current good manufacturing practice (CGMP) regulations apply (21 CFR parts 210 and 211). The guidance discusses how to investigate suspect or OOS test results, including responsibilities of laboratory personnel, laboratory phase of the investigation, additional testing that may be needed, when to expand the investigation outside the laboratory, and the final evaluation of all test results.

5.3.3 Team Inspections versus Solo Contract Laboratory Inspections

For many of the inspections performed by analysts, most often the analyst will be part of a team. The team may consist of one investigator and one analyst or it may be comprised of two or more investigators and/or analysts.  It may be a two person team or it may include multiple people such as investigators, chemists, microbiologists, and compliance officers.  It all depends on the firm and the focus of the inspection.  

As a team member, the analyst will be expected to follow the Investigations Operations Manual (IOM) and communicate with the lead investigator.  The IOM can be found at http://www.fda.gov/ora/inspect_ref/iom/iomtc.html .  The electronic copy is the official version and should be referenced prior to performing an inspection.  

The lead investigator is ultimately responsible for the inspection. However, an analyst needs to make sure that he or she is also aware of the objectionable conditions that the other team members are finding. Communication between team members is important. Manufacturing cGMP violations may also result in laboratory violations.   The analyst will be responsible for writing their part of the  FDA-483 Observations, but the lead investigator may change the wording to follow his/her style.  In many instances, it is a give and take situation.  When the investigator is rewording the analyst's observations, focus on whether the intent and meaning of the observation is the same.  If the rewording changes the meaning or intent, clearly explain this to the investigator and work together to write the observation so all team members understand the objectionable condition.  

If the analyst is performing an inspection of a contract laboratory, he or she may not have an investigator present.  In this case, the analyst is acting as the lead investigator.  This is a good reason for the analyst to familiarize him or herself with the IOM. The analyst will need to issue the FDA-482 Notice of Inspection upon entering the inspection site.  Issuance of the FDA-483 is clearly explained in the IOM.  While performing a "solo" inspection, extremely violative conditions may be found.  Consult with the local district office management to determine the course of action.  Documentary samples and/or physical samples may need to be collected and an affidavit and/or receipt for the sample may need to be issued.  Some districts may choose to send an investigator out for this part of the inspection or the analyst may be asked to do this on their own.  If the analyst is to proceed alone, he or she should consult the IOM for the proper procedure and paperwork, as well as, with the local investigation staff, determine the best course of action. 

If an analyst is conducting a solo inspection, the following sections in the IOM are extremely helpful:

CHAPTER 5 - ESTABLISHMENT INSPECTION

Subchapter 5.1 Inspection Information
Subchapter 5.2 Inspection Procedures
Subchapter 5.3 Evidence Development
Subchapter 5.4 Food
Subchapter 5.5 Drugs
Subchapter 5.6 Devices
Subchapter 5.7 Biologics
Subchapter 5.8 Pesticides
Subchapter 5.9 Veterinary Medicine
Subchapter 5.10 Reporting

These subchapters identify the basics of an inspection.  In the case of Drug Inspections, the IOM outlines the general areas covered during inspections.

 

Page Top Previous Page| Document TOC| Next Page