FDA Logo links to FDA home page
Center for Drug Evaluation and Research, U.S. Food and Drug Administration U.S. Food and Drug Administration Center for Drug Evaluation and Research
  HHS Logo links to Department of Health and Human Services website

FDA Home Page | CDER Home Page | CDER Site Info | Contact CDER | What's New @ CDER

horizonal rule

Questions and Answers on Current Good Manufacturing Practices,
Good Guidance Practices, Level 2 Guidance

Production and Process Controls

  1. Do the CGMPs require a firm to retain the equipment status identification labels with the batch record or other file?  Assuming each major piece of equipment has a unique "Cleaning and Use Log" that is adequately retained, is it acceptable to discard these 'quick reference' equipment labels?

  2. Can containers, closures, and packaging materials be sampled for receipt examination in the warehouse?

  3. A firm has multiple media fill failures. They conducted their media fills using TSB (tryptic soy broth) prepared by filtration through 0.2 micron sterilizing filter.  Investigation did not show any obvious causes.  What could be the source of contamination?

  4. Some products, such as transdermal patches, are made using manufacturing processes with higher in-process material reject rates than for other products and processes.  Is this okay?

  5. Do CGMPs require three successful process validation batches before a new active pharmaceutical ingredient (API) or a finished drug product is released for distribution?

  6. Is it generally acceptable from a cGMP perspective for a manufacturer of sterile drug products produced by aseptic processing to rely solely on ISO 14644-1 and ISO 14644-2 when qualifying their facility?

  7. In 2004, FDA issued a guidance entitled “PAT - A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality Assurance” that encouraged industry to modernize manufacturing through enhancements in process control.   How can I implement PAT (Process Analytical Technology)?

  8. How do I contact CDER's Process Analytical Technology Team?

  9. How do I contact CBER's Process Analytical Technology Team?

1. Do the CGMPs require a firm to retain the equipment status identification labels with the batch record or other file?  Assuming each major piece of equipment has a unique "Cleaning and Use Log" that is adequately retained, is it acceptable to discard these 'quick reference' equipment labels?

The CGMP regulations for finished pharmaceuticals require the retention of cleaning and use logs for non-dedicated equipment, but no similar requirement exists for retaining what are intended to be "quick reference" or temporary status labels.  Examples of these kinds of status labels include "mixing lot ###"; "clean, ready for use as of d/M/y"; "not clean."  We see no value in the retention of such labels in addition to the required equipment log or batch record documentation.  The labels serve a valuable, temporary purpose of positively identifying the current status of equipment and the material under process.  Any status label should be correct, legible, readily visible, and associated with the correct piece of equipment.  The information on the temporary status label should correspond with the information recorded in the equipment cleaning and use log, or the previous batch record for non-dedicated equipment.

Labels are merely one way to display temporary status information about a piece of equipment.  It is considered acceptable practice to display temporary equipment status information on dry-erase boards or chalkboards.  And it would be appropriate for an FDA investigator to verify that the information on a temporary status label is consistent with the log.

References:

  • 21 CFR 211.182: Equipment cleaning and use log

  • 21 CFR 211.105:  Equipment identification

Contact for further information:

Brian Hasselbalch, CDER
brian.hasselbalch@fda.hhs.gov

Back to Top


2. Can containers, closures, and packaging materials be sampled for receipt examination in the warehouse?

Yes.  Generally, we believe that sampling in a typical drug manufacturing facility warehouse would not represent a risk to the container/closure or affect the integrity of the sample results. But whether the act of collecting a sample in the warehouse violates the CGMPs requirement that containers "be opened, sampled, and sealed in a manner designed to prevent contamination of their contents..." will depend on the purported quality characteristics of the material under sample and the warehouse environment. For container/closures purporting to be sterile or depyrogenated, sampling should be under conditions equivalent to the purported quality of the material: a warehouse environment would not suffice (see 211.94 and 211.113(b)).  This is to preserve the fitness for use of the remaining container/closures as well as ensure sample integrity, if they are to be examined for microbial contamination.  At a minimum, any sampling should be performed in a manner to limit exposure to the environment during and after the time samples are removed (i.e., wiping outside surfaces, limiting time that the original package is open, and properly resealing original package). Well-written and followed procedures are the critical elements.

Note that the CGMPs at 211.84 permit a manufacturer to release for use a shipment of containers/closures based on the supplier's certificate of analysis and a visual identification of the containers/closures.  Once a supplier's reliability has been established by validation of their test results, a manufacturer could perform the visual examination entirely in the warehouse.

References:

  • 21 CFR 211.84: Testing and approval or rejection of components, drug product containers, and closures

  • 21 CFR 211.94: Drug product containers and closures

  • 21 CFR 211.113(b): Control of microbiological contamination

  • 21 CFR 211.122: Materials examination and usage criteria

Contact for further information:

Anthony Charity, CDER
anthrony.charity@fda.hhs.gov


3. A firm has multiple media fill failures. They conducted their media fills using TSB (tryptic soy broth) prepared by filtration through 0.2 micron sterilizing filter.  Investigation did not show any obvious causes.  What could be the source of contamination?

A firm recently had multiple media fill failures.  The media fill runs, simulating the filling process during production, were conducted inside an isolator.  The firm used TSB (non-sterile bulk powder) from a commercial source, and prepared the sterile solution by filtering through a 0.2 micron sterilizing filter.  An investigation was launched to trace the source of contamination.  The investigation was not successful in isolating or recovering the contaminating organism using conventional microbiological techniques, including the use of selective (e.g., blood agar) and nonselective (e.g., TSB and tryptic soy agar) media, and examination under a microscope.  The contaminant was eventually identified to be Acholeplasma laidlawii by using 16S rRNA gene sequence.  The firm subsequently conducted studies to confirm the presence of Acholeplasma laidlawii in the lot of TSB used.  Therefore, it was not a contaminant from the process, but from the media source.

Acholeplasma laidlawii belongs to an order of mycoplasma. Mycoplasma contain only a cell membrane and have no cell wall.  They are not susceptible to beta-lactams and do not take up Gram stain.  Individual organisms are pleomorphic (assume various shape from cocci to rods to filaments), varying in size from 0.2 to 0.3 microns or smaller.  It has been shown that Acholeplasma laidlawii is capable of penetrating a 0.2 micron filter, but is retained by a 0.1 micron filter (see Sundaram, et al.). Acholeplasma laidlawii is known to be associated with animal-derived material, and microbiological media is often from animal sources.  Environmental monitoring of mycoplasma requires selective media (PPLO broth or agar).

Resolution:

For now, this firm has decided to filter prepared TSB, for use in media fills, through a 0.1 micron filter (note: we do not expect or require firms to routinely use 0.1 micron filters for media preparation).  In the future, the firm will use sterile, irradiated TSB when it becomes available from a commercial supplier.  (Firm's autoclave is too small to permit processing of TSB for media fills, so this was not a viable option.)  The firm will continue monitoring for mycoplasma and has revalidated their cleaning procedure to verify its removal.  In this case, a thorough investigation by the firm led to a determination of the cause of the failure and an appropriate corrective action.

References:

  • 21 CFR 211.113: Control of microbiological contamination

  • 21 CFR 211.72: Filters

  • 21 CFR 211.84(d)(6): Testing and approval or rejection of components, drug product container, and closures

  • Sundaram, S., Eisenhuth, J., Howard, G., Brandwein, H. Application of membrane filtration for removal of diminutive bioburden organisms in pharmaceutical products and processes. PDA J. Pharm. Sci. Technol. 1999 Jul-Aug; 53(4): 186-201.

  • Kong, F., James, G., Gordon, S., Zekynski, A., Gilbert, G.L. Species-specific PCR for identification of common contaminant mollicutes in cell culture. Appl. Environ. Microbiol. 2001 Jul; 67(7): 3195-200.

  • Murray, P., Baron, E., Pfaller, M., Tenover, F., Yolken, R. Manual of Clinical Microbiology ASM Press, Sixth Edition.

Contact for further information:

Brenda Uratani, CDER
brenda.uratani@fda.hhs.gov

Back to Top


4. Some products, such as transdermal patches, are made using manufacturing processes with higher in-process material reject rates than for other products and processes.  Is this okay?

Maybe.  It depends on the cause and consistency of the reject rate.  Many transdermal patch manufacturing processes produce more waste (i.e., lower yield from theoretical) than other pharmaceutical processes.  This should not of itself be a concern.  The waste is usually due to the cumulative effect of roll splicing, line start-ups and stoppages, roll-stock changes, and perhaps higher rates of in-process sampling.  This is most pronounced for processes involving lamination of rolls of various component layers.  Roll-stock defects detected during adhesive coating of the roll, for example, can often only be rejected from the roll after final fabrication/lamination of the entire patch, which contributes to the final process waste stream.

We expect that validated and well-controlled processes will achieve fairly consistent waste amounts batch-to-batch.  Waste in excess of the normal operating rates may need (see 211.192) to be evaluated to determine cause (e.g., due to increase in sampling or higher than normal component defects... or both) and the consequences on product quality assessed.  We've seen a small number of cases where unusually high intra-batch rejects/losses were due to excessive component quality variability and poorly developed processes.

References:

  • 21 CFR 211.100: Written procedures; deviations

  • 21 CFR 211.103: Calculation of yield

  • 21 CFR 211.110: Sampling and testing of in-process materials and drug products

  • 21 CFR 211.192: Production record review

Contact for further information:

Brian Hasselbalch, CDER
brian.hasselbalch@fda.hhs.gov

Back to Top


5.  Do CGMPs require three successful process validation batches before a new active pharmaceutical ingredient (API) or a finished drug product is released for distribution?

No.  Neither the CGMP regulations nor FDA policy specifies a minimum number of batches to validate a manufacturing process. The current industry guidance on APIs (see ICH Q7A for APIs) also does not specify a specific number of batches for process validation. 

 FDA recognizes that validating a manufacturing process, or a change to a process, cannot be reduced to so simplistic a formula as the completion of three successful full scale batches.  The agency acknowledges that the idea of three validation batches has become prevalent, in part due to language in its own guidance documents.  However, FDA is now clarifying current expectations on process validation.  The 1987 Guideline of General Principles of Process Validation is currently being revised to address this issue. The emphasis for demonstrating validated processes is placed on the manufacturer’s process design and development studies in addition to its demonstration of  reproducibility at scale, a goal that has always been expected. 

 However, a minimum number of conformance (a.k.a. validation) batches necessary  to validate the manufacturing processes is not specified.  The manufacturer is expected to have a sound rationale for its choices in this regard.  The agency encourages the use of science based approaches to process validation.

 In March 2004, FDA revised the Compliance Policy Guide (CPG) (Sec. 490.100) on Process Validation Requirements for Drug Products and Active Pharmaceutical Ingredients Subject to Pre-Market Approval.  The CPG describes the concept that, after having identified and establishing control of all critical sources of variability, conformance batches are prepared to demonstrate that under normal conditions and operating parameters, the process results in the production of acceptable product.  Successful completion of the initial conformance batches would normally be expected before commercial distribution begins, but some possible exceptions are described in the CPG.  For example, although the CPG does not specifically mention concurrent validation for an API in short supply, the agency would consider the use of concurrent validation when it is necessary to address a true short-supply situation, and if the concurrent validation study conforms to the conditions identified in the CPG (See paragraph 4. a-c).

 The conditions outlined in the CPG include expanded testing for each batch intended to address a short-supply situation.  Expanded testing, conducted according to an established validation protocol could provide added assurance that the batch meets all established and appropriate criteria before the API is used in the finished drug product.  Additionally, confidence in the API manufacturing process may be gained by enhanced sampling (larger sample size representative of batch) and perhaps the testing of additional attributes.  Validated analytical methods are needed for testing every batch, including validation batches.  The agency would also expect the manufacturer to use a validation protocol which includes a review and final report after multiple batches are completed, even though the earlier batches may have been distributed or used in the finished drug product.

 References:

  • 21 CFR 211.100: Written procedures; deviations

  • 21 CFR 211.110: Sampling and testing of in-process materials and drug products

  • CPG 490.100 Process Validation Requirements for Drug Products and Active Pharmaceutical Ingredients Subject to Pre-Market Approval. 

  • ICH Q7A Good Manufacturing Practice Guidance for Active Pharmaceutical Ingredients

Contact for further information:

Grace McNally, grace.mcnally@fda.hhs.gov

Back to Top


6. Is it generally acceptable from a cGMP perspective for a manufacturer of sterile drug products produced by aseptic processing to rely solely on ISO 14644-1 and ISO 14644-2 when qualifying their facility?

No.  It is generally not acceptable from a current good manufacturing practice (“cGMP”) perspective for a manufacturer of sterile drug products produced by aseptic processing to rely solely on ISO 14644-1 Part 1: Classification of Air Cleanliness (“14644-1”) and ISO 14644-2 Part 2: Specifications for Testing and Monitoring to Prove Compliance with ISO 14644-1 (“14644-2”) when qualifying their facility. Rather, a manufacturer of sterile drug products produced by aseptic processing should use these ISO standards in combination with applicable FDA regulations, guidance and other relevant references to ensure a pharmaceutical facility is under an appropriate state of control. Consequently, appropriate measures augmenting ISO’s recommendations (e.g., with microbiological data) would likely be expected for a firm to meet or exceed CGMP in a pharmaceutical facility.

Please understand that 14644-1 and 14644-2 have superseded Federal Standard 209E, Airborne Particulate Cleanliness Classes in Cleanrooms and Clean Zones (“Federal Standard 209E”).  In November 2001, the U.S. General Services Administration canceled Federal Standard 209E.

While not FDA regulations or FDA guidance, the Agency believes 14644-1 and 14644-2 are useful in facilitating the international harmonization of industrial air classification for non-viable particle cleanliness in multiple industries (e.g., computer, aerospace, pharmaceutical).  As such, FDA adopted these particle cleanliness ratings in the 2004 guidance for industry Sterile Drug Products Produced by Aseptic Processing – Current Good Manufacturing Practice.  However, due to the unique aspects of producing sterile drug products by aseptic processing (e.g., microbiological issues) an aseptic processing manufacturer should not rely solely on 14644-1 and 14644-2 when qualifying their facility. 

References:

Author:
Stephen Mahoney (HFD-326)

Contact Information:
Division of Manufacturing and Product Quality (HFD-320): CGMP Subject Contacts.

Back to Top


7. In 2004, FDA issued a guidance entitled “PAT - A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality Assurance” that encouraged industry to modernize manufacturing through enhancements in process control.   How can I implement PAT (Process Analytical Technology)?

The objective of FDA's PAT program is to facilitate adoption of PAT.  In our 2004 guidance, we discuss FDA's collaborative approach to promote industry uptake of new and beneficial technologies that modernize manufacturing operations and enhance process control.  FDA recognizes that firms should be encouraged to promptly implement new systems that improve assurance of quality and process efficiency.  Accordingly, our approach to PAT implementation is risk based, and includes multiple options:

1. PAT can be implemented under the facility's own quality system. CGMP inspections by the PAT Team or PAT certified Investigator can precede or follow PAT implementation.

2. As another quality system implementation option, FDA invites manufacturers to request a preoperational review of their PAT manufacturing facility and process by the PAT Team (see ORA Field Management Directive No.135).

3. A supplement (CBE, CBE-30 or PAS) can be submitted to the Agency prior to implementation, and, if necessary, an inspection can be performed by a PAT Team or PAT certified Investigator before implementation.  This option should be used, for example, when an endproduct testing specification established in the application will be changed. 

4. A comparability protocol can be submitted to the Agency outlining PAT research, validation and implementation strategies, and time lines. Following collaborative review of the general strategy outlined in the comparability protocol, the regulatory pathway can include implementation under the facility's own quality system, a pre-operational review, CGMP inspections (either before or after PAT implementation), a combination of these, or another flexible approach.

Manufacturers should evaluate and discuss with the Agency the most appropriate option for PAT implementation.  For products regulated by the CDER, contact the Process Analytical Technology Team with any questions.

Back to Top


8. How do I contact CDER's Process Analytical Technology Team? 

Manufacturers are encouraged to contact the team via email regarding any PAT questions at: PAT@cder.fda.gov
 
To contact our PAT Team via mail, please see the PAT Web page (under the section “Contact Us”) for our new mailing address at White Oak. 

All correspondence should be identified clearly as "Process Analytical Technology" or "PAT."

Please also refer to the Web page to keep abreast of the latest information on PAT.

Back to Top


9 . How do I contact CBER's Process Analytical Technology Team? 

Manufacturers should contact the appropriate review division in CBER to discuss applicability of PAT to CBER-regulated products. 

References:

PAT - A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality Assurance

Contact Information:

 

Back to Top     Back to cGMP

Date created: August 4, 2004, updated November 12, 2008

horizonal rule