Questions and Answers on cGMP about production & process control part

Questions and Answers on cGMP about production & process control part
Questions and Answers on cGMP about production & process control part

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 with questions about Process Analytical Technology?

9. How do I contact CBER with questions about Process Analytical Technology?

10. What is the acceptable media fill frequency in relation to the number of shifts? Normally, media fills

should be repeated twice per shift per line per year. Is the same frequency expected of a process conducted in an isolator?

11. Why is the FDA concerned about human topical antiseptic drug products?

12. What specific current good manufacturing practice (CGMP) regulations might be useful to

manufacturers of topical antiseptic drug products?

13. How can manufacturers assess and address the risk of microbiological contamination of topical

antiseptics?

14. Can Leptospira species penetrate sterilizing-grade filters? If so, what should manufacturers keep in

mind in their ongoing lifecycle risk management efforts to assure microbial control?

15. FDA recently announced the withdrawal of its draft guidance for industry on Powder Blends and

Finished Dosage Units — Stratified In-Process Dosage Unit Sampling and Assessment. What were the Agency?s major concerns with this guidance?

16. Why is FDA concerned about proper sampling of powder blends?

17. What are some recommended innovative approaches to ensuring adequacy of mixing of powder

blends?

18. What are the Agency?s recommendations regarding in-process stratified sampling of finished dosage

units?

19. For a nonsterile compendial drug product that includes an antimicrobial preservative in its

formulation, may I release and market lots of this drug product with initial out-of-specification total

aerobic plate counts if these lots test within specification 2 weeks later?

20. Do pharmaceutical manufacturers need to have written procedures for preventing growth of

objectionable microorganisms in drug products not required to be sterile? What does objectionable mean anyway?

21. For drug products formulated with preservatives to inhibit microbial growth, is it necessary to test for

preservatives as part of batch release and stability testing?

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

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

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 TS

B (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.

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

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

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:

?U.S. Food and Drug Administration website.

?International Organization for Standardization website.

?ISO 14644-1 Part 1: Classification of Air Cleanliness.

?ISO 14644-2 Part 2: Specifications for Testing and Monitoring to Prove Compliance with ISO 14644-

1.

?Guidance for Industry: Sterile Drug Products Produced by Aseptic Processing – Current Good Manufacturing Practice (2004).

?Current Good Manufacturing Practice (cGMP) for Finished Pharmaceuticals (Title 21, Code of Federal Regulations, Part 211).

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 a 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 (see ORA Field Management Directive No.135, available at: https://www.360docs.net/doc/097708975.html,/ICECI/Inspections/FieldManagementDirectives/ucm096042.htm).

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 certified Investigator before implementation. This option should be used, for example, when an end product 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 (see Questions 8 and 9, below).

Date Revised: 9/16/2013

8. How do I contact CDER with questions about Process Analytical Technology?

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

9 . How do I contact CBER with questions about Process Analytical Technology?

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

10. What is the acceptable media fill frequency in relation to the number of shifts? Normally, media fills should be repeated twice per shift per line per year. Is the same frequency expected of a process conducted in an isolator?

A firm's justification for the frequency of media fills in relation to shifts should be risk-based, depending on the type of operations and the media fill study design. For closed, highly automated systems run on multiple shifts, a firm with a rigorous media fill design may be justified to conduct a lower number of total media fill runs. Such a program can be appropriate provided that it still assures performance of media fills for each aseptic processing line at least semi-annually. The 2004 guidance for industry on Sterile Drug Products Produced by Aseptic Processing states that "[A]ctivities and interventions representative of each shift, and shift changeover, should be incorporated into the design of the semi-annual qualification program." In addition, the EU Annex 1, Manufacture of Sterile Medicinal Products, states that "Normally, process simulation tests should be repeated twice a year per shift and process."

Certain modern manufacturing designs (isolators and "closed vial" filling) afford isolation of the aseptic process from microbiological contamination risks (e.g., operators and surrounding room environment) throughout processing. For such closed systems??…, if the design of the processing equipment is robust and the extent of manual manipulation in the manufacturing process is minimized, a firm can consider this information in determining its media fill validation approach. For example, it is expected that a conventional aseptic processing line that operates on two shifts be evaluated twice per year per shift, and culminate in four media fills. However, for aseptic filling conducted in an isolator over two shifts, it may be justified to perform fewer than four media fill runs per year, while still evaluating the line semi-annually to assure a continued state of aseptic process control. This lower total number of media fill runs would be based on sound risk rationale and would be subject to re-evaluation if contamination issues (e.g., product non-sterility, media fill failure, any problematic environmental trends) occur.This does not apply to RABS (Restricted Access Barrier Systems)

References:

1. 21 CFR 211.63, 211.65, and 211.67 address, respectively, "Equipment design, size, and location," "Equipment construction," and "Equipment cleaning and maintenance."

2. 21 CFR 211.84(c)(3) states that "Sterile equipment and aseptic sampling techniques shall be used when necessary."

3. 21 CFR 211.113(b) states that "Appropriate written procedures, designed to prevent microbiological contamination of drug products purporting to be sterile, shall be established and followed. Such procedures shall include validation of all aseptic and any sterilization process."

4. FDA Guidance for Industry Sterile Drug Products Produced by Aseptic Processing (2004)

5. EU Annex 1, Manufacture of Sterile Medicinal Products (2003)

11. Why is the FDA concerned about human topical antiseptic drug products?

FDA has identified several incidents of objectionable microbial contamination of topical antiseptic drug products (e.g., alcohol pads or swabs used to prepare the skin prior to an injection). Microbial contamination may be caused by substandard manufacturing practices and the agency is concerned about safety risks, such as from infection, associated with this contamination.

12. What specific current good manufacturing practice (CGMP) regulations might be useful to manufacturers of topical antiseptic drug products?

Section 501(a) (2) (B) of the Federal Food, Drug, and Cosmetic Act requires all drugs to be manufactured in conformance with CGMP. The CGMP regulations in 21 CFR Parts 210 and 211 for finished pharmaceuticals apply equally to over-the-counter (OTC) and prescription (Rx) drug products.1

The CGMP regulations provide the minimum legal requirements for conducting reliable operations.2 Some relevant CGMP regulations, with a brief description, are given below:

Manufacturing Design and Control – CGMP Requirements and Recommended Guidance for Manufacturers ?Design manufacturing facilities (21 CFR 211.42)and processes (see below) to prevent microbial contamination :

o For non-sterile drug products – establish control procedures to monitor output and validate processes to include bioburden testing (21 CFR 211.110 (a) (6)), 211.111) and

establish and follow written procedures designed to prevent the introduction of objectionable

microorganisms (211.113(a)).

o For sterile drug products3 - establish and follow written procedures designed to prevent microbial contamination (211.113(b)).

?Conduct process validationstudies4 to ensure acceptable output (e.g., with topical antiseptics, particularly product microbiological quality) (211.110(a). Implement and validate needed changes

when deficient manufacturing steps, equipment, or raw materials may be adversely affecting process control.

?Ensure that operating procedures will consistently produce a quality product (211.100). Review and evaluate any deviations or discrepancies documented during manufacturing and testing to

determine if a product lacks assurance of sterility (for sterile antiseptics) or may be contaminated

with objectionable microorganisms (for non-sterile antiseptics). Document and implement any

corrective actions deriving from the evaluation (211.192).

?Ensure that all equipment, including water systems, is clean, sanitary, operates consistently, and is suitable for its intended use (211.63, 211.65, 211.67, and 211.68).

?Establish and follow in-process bioburden testing procedures to help monitor in-process control, including understanding the bioburden challenge to a final sterilization process (211.110(a)(6)). Components, In-process Materials, Container/Closure and Finished Product Testing - CGMP Requirements for Manufacturers

?Establish appropriate written testing standards/specifications and sampling plans for components, in-process materials, containers/closures, and finished products (211.160).

?Establish procedures for testing and approval or rejection of components, drug product containers, and closures (211.80).Testeach lot of a drug product component and container/closure, including those that may be vulnerable to microbiological contamination (211.84)(d)(4-5), including applicator material (e.g., cotton pads) and water used as an ingredient in the product.

?Conduct appropriate microbiological tests before a batch disposition decision is made. Test each batch of a sterile product for sterility (211.167). Test each batch of a non-sterile product to

ensure absence of objectionable microorganisms (211.165(b)).

Management5,6

The CGMPs require that the management of a manufacturing facility maintains a well-functioning quality system, which includes an effective quality unit vested with the responsibilities and authorities required under cGMP (211.22).

13. How can manufacturers assess and address the risk of microbiological contamination of topical antiseptics?

Since there are potentially many different root causes of product contamination by microorganisms, it is imperative that manufacturers perform a manufacturing risk assessment to understand manufacturing failure modes and implement prevention measures.

In addition, any risk assessment approach should be informed by an understanding of the microbial contamination vulnerabilities of the concerned product. For example, some product considerations for manufacturers include, but are not limited to:

?Determine the types of microbes that might survive or thrive in your products. Provide additional controls and testing based on the output of the risk assessment to ensure product quality.

?Ensure that your microbial recovery methods are capable of detecting the types of microbes that may affect product quality.

?Evaluate risk of contamination from components, including during component production, storage, or due to the intrinsic risk from source materials. Consider all possible sources of microbial

contamination, including the following:

o Components or products stored in open bins can be at risk for contamination by spore-forming microbes, such as Bacillus cereus,7,8as well as by Serratia species and other

worrisome airborne microbes. Manufacturing areas exposed to windy or poor HVAC

conditions may increase the potential for this environmental contamination risk.

o Some materials, especially from natural sources, may have high or objectionable intrinsic bioburden.

o Water quality can pose a significant risk, as most antiseptics include water as a key ingredient. Contaminated purified water has been the root cause of multiple recalls of

antiseptics, including instances of antiseptics contaminated with Burkholderia (previously

Pseudomonas) cepacia, an opportunistic pathogen.

o Unsanitary practices or sources

o When manufacturing in areas with high humidity, molds can be of special concern. References:

1. Compliance Policy Guide Sec. 450.100 CGMP Enforcement Policy - OTC vs. Rx Drugs

(https://www.360docs.net/doc/097708975.html,/ICECI/ComplianceManuals/CompliancePolicyGuidanceManual/ucm074387.htm)

2. Code of Federal Regulations (CFR), Title 21, Part 211 - CURRENT GOOD MANUFACTURING

PRACTICE FOR FINISHED PHARMACEUTICALS

(https://www.360docs.net/doc/097708975.html,/scripts/cdrh/cfdocs/cfCFR/CFRSearch.cfm?CFRPart=211)

3. FDA Guidance for Industry on Sterile Drug Products Produced by Aseptic Processing — Current

Good Manufacturing Practice

(https://www.360docs.net/doc/097708975.html,/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM0 70342.pdf)

4. FDA Guidance for Industry on Process

Validation https://www.360docs.net/doc/097708975.html,/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidanc es/UCM070336.pdf

5. FDA Guidance for Industry on ICH Q9 – Quality Risk Management

(https://www.360docs.net/doc/097708975.html,/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM0 73511.pdf)

6. FDA Guidance for Industry on ICHQ10 – Pharmaceutical Quality System

https://www.360docs.net/doc/097708975.html,/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM0 73517.pdf

7. FDA News Release - FDA reminds health care professionals about safe use of non-sterile alcohol

prep pads (2/1/2011)

(https://www.360docs.net/doc/097708975.html,/NewsEvents/Newsroom/PressAnnouncements/ucm241750.htm)

8. CDC Morbidity and Mortality Weekly Report (MMWR) - Contamination of Alcohol Prep Pads with

Bacillus cereus Group and Bacillus Species

(https://www.360docs.net/doc/097708975.html,/mmwr/preview/mmwrhtml/mm6011a5.htm)

14. Can Leptospira species penetrate sterilizing-grade filters? If so, what should manufacturers keep in mind in their ongoing lifecycle risk management efforts to assure microbial control?

FDA is aware of a recent report [ref. 2, below] of Leptospira licerasiae contamination in cell cultures. There is no indication that this bacterium ultimately contaminated either the finished drug substance or drug product. This bacterium has been found to pass through 0.1 μm pore size rated sterilizing-grade membrane filters. While this specific species was the identified contaminant in this case, other Leptospira species also are capable of passing through 0.1 μm pore size rated filters [ref. 3, below]. Compendial microbiological test methods typically used in association with upstream biotechnology and pharmaceutical production are not capable of detecting this type of bacteria. Whether this apparently rare contamination risk may be more widespread is unknown, and we are sharing this information so that manufacturers can consider whether this hazard may be relevant to their operations.

Leptospira are Gram-negative aerobic spirochetes that are flexible, highly motile, and spiral-shaped with internal flagella. The bacteria measure 1μm in diameter and 10-20 μm in length.Leptospira are obligate aerobes that use oxygen as the electron receptor and long-chain fatty acids as a major source of

energy. While some of the Leptospira are harmless fresh-water saprophytes, other species are pathogenic and can cause leptosporosis, a significant disease in humans and animals [ref. 4-6, below).

Based on current information, Leptospira contamination does not appear to occur frequently, and purification steps that follow cell culture in a typical biotechnology operation would be expected to prevent carryover to the finished drug substance. Testing of bulk drug substances produced in the reported cases did not detect Leptospira spp., and no evidence of deleterious effects on in-process product were observed in the known case study. However, we are providing this communication to alert manufacturers that these types of bacteria can potentially:

o penetrate sterilizing-grade membrane filters

o be present in the manufacturing site environment

o impact in-process production (e.g., production yields, impurity levels, process performance)

o go undetected due to the limitations of current compendial bioburden tests in detecting this microbial genus

As a general principle, manufacturers should use sound risk management and be aware of unusual microbiota reported in the literature that may impact their manufacturing processes (e.g., cell culture biotechnology, conventional sterile drug manufacturing).

Manufacturers should assess their operations, be aware of potential risks, and apply appropriate risk management based on an understanding of possible or emerging contamination risks [ref. 1 (see Section 18.3)]. As appropriate, preventive measures should be implemented during the product and process lifecycle.

To illustrate, if leptospiral contamination is considered possible, or has occurred, risk mitigation procedures and practices for this microorganism should include at least the following:

1. Review of available published articles from the scientific literature and technical reports by related

industry organizations that may provide further understanding on how to mitigate this contamination hazard.

2. Use of molecular or nonconventional microbial monitoring methods at appropriate intervals to detect

microbial flora that may exist in processing steps or in the immediate environment, but are not

readily detected by current routine methods. Such expanded testing should be used to modify the strategy (e.g., timing, frequency, types of tests) of detection and control in the event of newly-

identified risk posed by the viable, but not easily cultured, microorganism.

Examples include:

a. Use of specialized media such as EMJH [ref. 7, below] or other suitable media [ref. 8, below]. It should be noted that these bacteria typically grow very slowly.

b. Use of validated PCR methods (e.g., as an investigative tool) for rapid screening and detection of spirochete bacteria.

c. Consideration of special stain techniques or other means to identify the presence of Leptospira [ref. 9, below].

3. Use of conventional approaches. Firms should continue to properly employ basic, standard microbiology laboratory practices to detect contamination. For example, the laboratory should ensure that microscopic examination is part of its routine cell culture process control program, as it provides an important means of detecting microbial contaminants that may not readily grow on conventional media.

4. Implementing such quality risk-management measures into the initial design (i.e., preventive actions), and promptly implementing an appropriate corrective action plan in response to newly-identified contamination sources, throughout the lifecycle of the product.

References:

1. Guidance for Industry ICH Q7 – Good Manufacturing Practices for Active Pharmaceutical Ingredients. 2001

2. Chen, J.; Bergenvin, J.; Kiss, R.; Walker, G.; Battistoni, T.; Lufburrow, P.; Lam, H.; Vinther, A. Case study – A novel bacterial contamination in cell culture production –Leptospira licerasiae. PDA J. Pharm. Sci. Technol. November-December 2012.

3. WHO – Leptospira. Faine, S. (Ed.). Guidelines for the Control of Leptospirosis. World Health Organization, Geneva. 1982.

4. Ricaldi, JN; Fouts, DE; Selengut, JD; Harkins, DM; Patra, KP; et al. Whole Genome Analysis of Leptospira licerasiae Provides Insight into Leptospiral Evolution and Pathogenicity. PLoS Negl Trop Dis 2012 October, 6(10): e1853.

5. Matthias, MA; Ricaldi, JN; Cespedes, M; Diaz, MM; Galloway, RL; et al., Human Leptospirosis Caused by a New Antigenically Unique Leptopspira Associated with a Rattus Species Reservoir in the Peruvian Amazon. PLoS Negl Trop Dis 2008 (2(4): e213.

6. Bharti, AR; Nally, JE; Ricaldi, JN; Matthias, MA; Diaz, MM; et al. Leptospirosis: A zoonotic disease of global importance. Lancet Infect Dis. 2003:3:757-771.

7. Ellinghausen, H. C. and McCullough W. G. Nutrition of Leptospira Pomona and growth of 13 other serotypes: fractionation of oleic albumin complex (OAC) and a medium of bovine albumin and polysorbate 80. Am. J. Vet. 1965, 26: pp 45-51.

8. Rule Pl, Alexander AD. J. Clin Microbiol, 1986, 23(3):500-504.

9. Frank S. Kohn. J. Amer. Med Technology, July-Aug, 1973.

15. FDA recently announced the withdrawal of its draft guidance for industry on Powder Blends and Finished Dosage Units — Stratified In-Process Dosage Unit Sampling and Assessment. What were the Agency’s major concerns wi th this guidance?

FDA?s major concern was that Sections V and VII of the withdrawn draft guidance no longer represented the Agency?s current thinking, as explained below.

Section V (Exhibit/Validation Batch Powder Mix Homogeneity) recommended that at least three replicate samples be taken from at least ten locations in the powder blender, but that only one of the three replicates be evaluated to assess powder blend uniformity. The Agency currently recommends that all replicate

samples taken from various locations in the blender be evaluated to perform a statistically valid

analysis. This analysis can demonstrate that variability attributable to sample location is not significant and that the powder blend is homogenous. Statistical tools are available to ascertain both the number of replicates and the number of sampling locations across the blender that should be analyzed to conduct a valid analysis.

Section VII (Routine Manufacturing Batch Testing Methods) acceptance criteria designated to the Standard Criteria Method and the Marginal Criteria Method were based upon the limits published in the United States Pharmacopeia (USP) General Chapter <905>Uniformity of Dosage Units. However, the procedures and acceptance criteria in USP <905> are not a statistical sampling plan and so the results of the procedures should not be extrapolated to larger populations. Therefore, because the procedure and acceptance criteria prescribed in section VII provided only limited statistical assurance that batches of drug products met appropriate specifications and statistical quality control criteria, FDA no longer supports their use for batch release. Currently, there are several standard statistical practices (see references) that, if used correctly, can help to ensure compliance with the current good manufacturing practice (CGMP) regulations, including 21 CFR 211.110 Sampling and testing of in-process materials and drug products, 21 CFR 211.160 General Requirements [Subpart I, Laboratory Controls],and 21 CFR 211.165 Testing and release [of the finished drug product] for distribution.1

References:

1. FDA CGMP regulations: 21 CFR 211.110; 211.160; 211.165. Available at:

https://www.360docs.net/doc/097708975.html,/scripts/cdrh/cfdocs/cfcfr/CFRsearch.cfm?CFRPart=211

16.Why is FDA concerned about proper sampling of powder blends?

The CGMPs require that all sampling plans be scientifically sound and representative of the batch under test (see 211.160(b)). Further, in-process testing of powder blends to demonstrate adequacy of mixing is a CGMP requirement (21 CFR 211.110). Between- and within-location variability in the powder blend is a critical component of finished product quality and therefore should be evaluated. Drug product manufacturers need to use a science- and risk-based sampling approach to assure (a) adequacy of blend mixing and (b) that sampling of the blend is done at a suitable juncture in the manufacturing process. The sampling and analysis needs to ensure that no differences exist between locations in a blend that could adversely affect finished product quality. Traditional sampling using a powder-thief may have drawbacks and limitations, such as causing disturbance to the powder bed, powder segregation, or other sampling errors. However, powder-thief sampling remains widely used and provides reliable results in many cases. The Agency encourages firms to adopt more innovative approaches to ensuring adequacy of mixing (see, e.g., the PAT guidance). If a manufacturer proposes to use a thief sampling method, the reliability of the method should be evaluated as part of analytical methods development.

17. What are some recommended innovative approaches to ensuring adequacy of mixing of powder blends?

Innovative approaches to consider include, but are not limited to: (a) Process Analytical Technology (PAT) real-time monitoring and feedforward controlling of the powder blending process1 and (b) use of Statistical Process Control (SPC) tools to monitor the powder blending process and to maintain a state of control. When a manufacturer decides to implement PAT or other process-monitoring and control techniques for powder blend homogeneity assessment, its decision should be supported with appropriate data and rationale using a science- and risk-based approach. For example, the effective sample size of powder examined by PAT probes has to be estimated, such that the scale of scrutiny of the PAT powder blending monitoring can be justified.2 The number of PAT probes and their locations also have to be justified. If a scientifically sound PAT monitoring and control strategy is established, it can facilitate the assessment of: (a) variability across locations within the powder bed,3 (b) the variability over time of one location, and (c) the potential correlation between the powder sample and the unit dosage form.

References:

1. FDA Guidance for Industry: PAT - A Framework for Innovative Pharmaceutical Development,

Manufacturing, and Quality Assurance. September

2004. Available at: https://www.360docs.net/doc/097708975.html,/downloads/Drugs/GuidanceComplianceRegulatoryInformatio n/Guidances/UCM070305.pdf

2. Wu, H.; Tawakkul, M.; White, M.; Khan, M. Quality-by-Design (QbD): An Integrated Multivariate

Approach for the Component Quantification in Powder Blends. International Journal of

Pharmaceutics, vol. 372 issue 1-2 May 8, 2009. p. 39-48.

3. El-Hagrasy, A.; Morris, H.; D?Amico, F; et al. Near-infrared Spectroscopy and Imaging for the

Monitoring of Powder Blend Homogeneity.Journal of Pharmaceutical Sciences, vol. 90 issue 9,

September 2001. p. 1298 – 1307.

18. What are the Agency’s recommendations regarding in-process stratified sampling of finished dosage units?

Stratified sampling is recommended to be used when the population is known to have several subdivisions (i.e., locations) which may give different results for the quality characteristics measured. The Agency expects that no significant differences should exist between in-process locations that could affect finished product quality. Between- and within- location variability is a critical component of finished product quality and therefore should be evaluated. Please refer to ASTM E27096 and ASTM E28107 for further guidance on establishing acceptance criteria for a stratified sampling plan.

References

1. ASTM E2709: Standard Practice for Demonstrating Capability to Comply with an Acceptance Procedure.

2. ASTM E2810: Standard Practice for Demonstrating Capability to Comply with the Test for Uniformity

of Dosage Units.

19. For a nonsterile compendial drug product that includes an antimicrobial preservative in its formulation, may I release and market lots of this drug product with initial out-of-specification total aerobic plate counts if these lots test within specification 2 weeks later?

No. 21 CFR 211.113(a) requires appropriate written procedures to be established and followed during manufacturing to prevent objectionable microorganisms in drug products not required to be sterile.

Additionally, the second paragraph of USP General Chapter <51> Antimicrobial Effectiveness Testing reads:

Antimicrobial preservatives should not be used as a substitute for good manufacturing practices, solely to reduce the viable microbial population of a nonsterile product, or control the presterilization bioburden of a multidose formulation during manufacturing.

Drug manufacturers should not rely on antimicrobial preservatives to reduce initial out-of-specification plate counts to within-specification levels and then market the product. Section 211.165(f) mandates that drug products failing to meet established standards or specifications be rejected. The initial test results exhibiting out-of specification levels of microbes are not disqualified even if subsequent test results are within specifications. In such cases, FDA still expects the manufacturer to reject the drug product based on the initial results.

It is also not acceptable for manufacturers to allow an inappropriately long time (e.g., weeks) to pass before testing the product, which might permit the preservative to reduce levels of microbes possibly introduced during manufacture and thus avoid out-of-specification test results.

Finally, drug manufacturers should review their manufacturing process to determine procedures or equipment that might introduce contaminating microorganisms into the process or product.

References:

?21 CFR 211.113: Control of microbiological contamination

?21 CFR 211.165: Testing and release for distribution

?USP 38–National Formulary (NF) 33 (2015) General Chapter <51> Antimicrobial Effectiveness Testing

?USP 38–NF 33 (2015) General Chapter <61> Microbiological Examination of Nonsterile Products: Microbial Enumeration Tests

?USP 38–NF 33 (2015) General Chapter <62> Microbiological Examination of Nonsterile Products: Tests for Specified Microorganisms

20. Do pharmaceutical manufacturers need to have written procedures for preventing growth of objectionable microorganisms in drug products not required to be sterile? What does objectionable mean anyway?

Yes, CGMP regulations do require these written procedures. 21 CFR 211.113(a) specifies that appropriate written procedures be established and followed to prevent growth of objectionable microorganisms in drug products not required to be sterile. Even though a drug product is not sterile, a firm must follow written procedures that proactively prevent introduction and proliferation of objectionable microorganisms. 21 CFR 211.165(b) states that “[t]here shall be appropriate laboratory testing, as necessary, of each batch of drug product required to be free of objectionable microorganisms” before it is released for distribution.

The meaning of the term objectionable needs to be evaluated on a case-by-case basis by each drug manufacturer. The primary meaning relates to microbial contaminants that, based on microbial species, numbers of organisms, dosage form, intended use, patient population, and route of administration, would adversely affect product safety. Microorganisms may be objectionable for several reasons; for example, they: ?Are a known human pathogen

?Adversely affect product stability

?React with, or potentially damage the integrity of, the container closure system (for example, fermentation that creates gaseous pressures sufficient to rupture a product container/closure) ?Interfere with analytical methods or active ingredient bioavailability

Establishing production time limits is an example of a control to prevent growth of objectionable microorganisms. Per 21 CFR 211.111, time limits for the completion of each phase of production, when appropriate, must be established and followed. For example, if a firm finds it necessary to hold a bulk topical or liquid product for several months until it is filled, the firm might establish a holding time limit to help prevent objectionable microbial buildup. Validation and control over microbial content of purified water systems used in certain topical products are also examples of such procedures (see FDA guidance, referenced below). References:

?21 CFR 211.113: Control of microbiological contamination

?21 CFR 211.165: Testing and release for distribution

?21 CFR 211.111: Time limitations on production

?FDA Guidance for Industry, 2011, Process Validation: General Principles and Practices

21. For drug products formulated with preservatives to inhibit microbial growth, is it necessary to test for preservatives as part of batch release and stability testing?

Yes. Two types of tests are generally used. Initially, firms perform antimicrobial preservative effectiveness testing to determine a minimally effective level of preservative. Once that level has been determined, firms may establish appropriate corresponding analytical test specifications. Firms may then apply the analytical tests for preservative content at batch release and throughout the shelf life of lots on stability. References:

?21 CFR 211.165: Testing and release for distribution

?21 CFR 211.166: Stability testing

?USP 38–NF 33 (2015) General Chapter <51> Antimicrobial Effectiveness Testing

中英文对照版

中英文对照版 自检互检,确保产品零缺点! Self and mutual inspection, make sure zero defection! 杜绝不良思想,发扬优质精神! Prevent unhealthy thinking; develop high quality inspirit. 持续的质量改进,是我们永恒的目标。 Quality continuous improvement is our eternal objective. 积累点滴改进,迈向完美品质。 Accumulate every improvement is led to Perfect quality 培养优质素养,提高团队力量。 Quality-cultivation,Strengthens our team 不为失败找理由,要为成功找方法。

Do not make excuses for failure, but find a way to solve it. 客户满意,是检验我们工作的唯一标准。 Customer satisfaction is the only benchmark to inspect our works. 没有执行力,就没有竞争力。 No implementation power; no competitiveness advantage. 执行力文化,是从负责任开始的。 Implementation starts from taking responsibility 消除一切浪费,追求精益求精和不断改善。 Stop waste to pursue perfection and constant improvement 。 节约是美德,珍惜能源点滴。 Saving is our virtue, treasure every bit of resource 。 努力是将工作做完,用心是将工作做好。

GMP(2010年修订版)中的中英文术语

ICH(International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use)人用药物注册技术要求国际协调会。 Q10:药品质量管理体系(PQS),基于QMS(质量管理体系)而来,最合适的指南应该是ICH的Q10。 Q9:质量风险管理。 Q8:药物开发。 自动化生产规范GAMP5(Good Automated Manufacturing Practice)由 ISPE(International Society For Pharmaceutical Engineering国际制药工程协会。创立于1980年,是致力于培训制药领域专家并提升制药行业水准的世界最大的非盈利性组织之一)的GAMP论坛(GAMP Forum)组织编写发布的关于制药企业计算机化系统的综合性指南。 纠正和预防措施CAPA(Corrective Action & Preventive Action)

超出标准的可疑结果OOS(out of specification) 质量管理 QM(Quality Management) 质量保证 QA(Quality Assurance) 质量控制 QC(Quality Control) 生产管理 PM(Production Maanagement) 工程设备管理 EM (Enginccr Equipment Managemant) 销售管理 MS (Maragement Of Sales) 行政人员管理AP (Administration and Personael Managemeht) 工艺验证 PV (Process Validation) 设备验证 EV (Equipment Validation) 工艺规程 PP (Process Procedure) 质量标准 QS (Quality Management) 物料管理 MM (Materid Management) 原料管理 RM (Raw Material Management) 辅料管理 EM (Excipient Management) 质量标准 QS (Quality Standard) 增补和修订A.A.A Addition and Amendments 空调器AC Air Conditioner 药物不良反应ADR Adverse Drug Reaction 食品与药品官员协会(美国)AFDO Association of Food and Drug Officials 接受ACC Accept 合格质量标准AQL Acceptable Quality Level 简化的新药申请ADNA Abbreviated New Drug Application 物料清单BOM Bill of Material 原料药BPC Bulk pharmaceutical Chemiclls 生物制品评价与研究中心CBER Center for Biologics Evaluation Research 菌落形成单位CFU Colony Forming Unet 药品管理档案DMF Drug Master File 药物评价与研究中心CDER Cemter for Drug Evaluation amd Research

《论语》中英文对照版

论语 CONFUCIAN ANALECTS

学而第一 『⒈1』子曰:“学而时习之,不亦说乎?有朋自远方来,不亦乐乎?人不知而不愠,不亦君子乎?” The Master said: "Is it not pleasant to learn with a constant perseverance and application? "Is it not delightful to have friends coming from distant quarters? "Is he not a man of complete virtue, who feels no discomposure though men may take no note of him?" 『⒈2』有子曰:“其为人也孝弟,而好犯上者,鲜矣;不好犯上,而好作乱者,未之有也。君子务本,本立而道生。孝弟也者,其为仁之本与!” The philosopher Yu said, "They are few who, being filial and fraternal, are fond of offending against their superiors. There have been none, who, not liking to offend against their superiors, have been fond of stirring up confusion. "The superior man bends his attention to what is radical. That being established, all practical courses naturally grow up. Filial piety and fraternal submission,-are they not the root of all benevolent actions?" 『⒈3』子曰:“巧言令色,鲜矣仁!” The Master said, "Fine words and an insinuating appearance are seldom associated with true virtue." 『⒈4』曾子曰:“吾日三省吾身——为人谋而不忠乎?于朋友交而不信乎?传不习乎?” The philosopher Tsang said, "I daily examine myself on three points:-whether, in transacting business for others, I may have been not faithful;-whether, in intercourse with friends, I may have been not sincere;-whether I may have not mastered and practiced the instructions of my teacher." 『⒈5』子曰:“道千乘之国,敬事而信,节用而爱人,使民以时。” The Master said, "To rule a country of a thousand chariots, there must be reverent attention to business, and sincerity; economy in expenditure, and love for men; and the employment of the people at the proper seasons." 『⒈6』子曰:“弟子,入则孝,出则弟,谨而信,凡爱众,而亲仁。行有余力,则以学文。” The Master said, "A youth, when at home, should be filial, and, abroad, respectful to his elders. He should be earnest and truthful. He should overflow in love to all, and cultivate the friendship of the good. When he has time and opportunity, after the performance of these things, he should employ them in polite studies." 『⒈7』子夏曰:“贤贤易色;事父母,能竭其力;事君,能致其身;于朋友交,言而有信。虽曰未学,吾必谓之学矣。”

最新澳大利亚GMP(中文版)

澳大利亚TGA 药物良好生产操作规范

导言 (4) 解释 (4) 第一章质量管理 (5) 原则 (5) 质量保证 (5) 良好的生产操作规范 (5) 质量控制 (6) 第二章人员 (7) 原则 (7) 一般原理 (7) 关键人员 (7) 培训 (8) 个人卫生 (8) 第三章厂房与设备 (9) 原则 (9) 厂房 (9) 原则 (9) 生产区 (9) 贮存区 (10) 质量控制区 (10) 设备 (10) 第四章文件 (12) 原则 (12) 一般原理 (12) 必备文件 (12) 制造手册和生产指令 (13) 包装指令 (14) 批生产记录 (14) 批包装记录 (14) 程序和记录 (15) 第五章生产过程 (17) 原则 (17) 一般原理 (17) 生产过程中交叉污染的预防 (17) 验证 (18) 起始物料 (18) 生产操作中间体和未包装产品 (19) 包装材料 (19) 包装 (19) 成品 (20) 不合格品、返工与回收产品 (20) 第六章质量控制 (21) 原则 (21) 一般原理 (21)

优良的质控化验室 (21) 文件 (21) 取样 (22) 检测 (22) 第七章合同加工与分析 (24) 原则 (24) 一般原理 (24) 合同的提供者(甲方) (24) 合同的接受者(乙方) (24) 合同 (24) 第八章投诉与产品召回 (26) 原则 (26) 投诉 (26) 召回 (26) 第九章自检 (27) 原则 (27) 附录 (28) 附录八:起始物料与包装材料的取样 (28) 原则 (28) 人员 (28) 起始物料 (28) 包装材料 (29) 附录十五:确认与验证 (29) 原则 (29) 验证计划 (29) 文件 (29) 确认 (30) 过程确认 (30) 清洁验证 (32) 变更控制 (32) 再验证 (32) 术语 (32) 附录十六:资质人员和批放行 (33) 附录十七:参数的放行 (33) 1、原则 (33) 2、参数放行 (33) 灭菌产品的参数让步 (33) 术语 (33) 附录十八:原料药的GMP指南 (33) 术语 (34)

浅谈中国GMP与美国cGMP 之差别

浅谈中国GMP与美国cGMP 之差别 《药品生产质量管理规范》(Good Manufacturing Practice GMP)是世界各国对药品生产全过程监督管理普遍采用的法定技术规范,也是世界卫生组织向各国推荐采用的技术规范。实施GMP是实行药品质量保证制度的需要----因为药品生产企业若未通过GMP认证,就会被拒之于国际贸易的技术壁垒之外。 GMP的内容包括:机构、人员、厂房、设施设备、卫生、验证、文件、生产管理、质量管理、产品销售与回收、投诉与不良反应报告、自检等各方面的要求。在硬件方面要有符合要求的环境、厂房、设备;在软件方面要有可靠的生产工艺、严格的制度、完善的验证管理,藉以达到一个共同的目的:防止不同药物或其成份之间发生混淆;防止由其它药物或其它物质带来的污染和交叉污染;防止差错与计量传递和信息传递失真;防止遗漏任何生产和检验步骤的事故发生;防止任意操作及不执行标准与低限投料等违章违法事故发生;以及保证药品生产和销售全过程的可追溯性。 ? ???我国20世纪80年代初引进了GMP概念,并于1999年6月发布了《药品生产质量管理规范》(1998年修订),自1999年7月1日开始全面的、强制性的实施。 美国在20世纪90年代中期就提出了GMP的概念,美国现行药品生产管理规范(cGMP)是英文Current Good Manufacture Practices的简称,在《联邦法典》(CFR)的210和2ll部分。 从总体上看,中国GMP和美国CGMP的精神、原则、主要内容和要求方面是一致的,但在许多方面确实存在着不同,下面就几个方面谈谈中国GMP与美国cGMP的差别。 一、认证方式不同 中国GMP认证只是药品生产许可的GMP认证,不包括产品注册认证,产品注册认证在GMP 认证之前进行,企业只有在取得产品注册的批准文号之后才能申请GMP认证。而且不管是产品注册还是GMP认证,都要求上报三批产品生产数据,至少六个月稳定性考察数据。 美国CGMP认证包括产品研发(Product Development)和生产(Chemical Manufacture Control??CMC)两部分,即将产品注册和生产认证同时进行。而且美国将产品注册分为新药注册(NDA)和仿制药注册(ANDA)两种,新药注册要求上报三批产品生产数据,六个月稳定性考察数据;仿制药注册要求上报一批产品生产数据,三个月稳定性考察数据即可,继续考察和验证的数据由企业留存,在以后给FDA的年报中进行说明。 二、指导文件的详细程度不同 在美国,GMP的原则性条款都包含在联邦法规中的CFR210和211部分中,这些条款一般很难变更和增补。因此,FDA以行业指南的形式发布了各种不同类型医药产品的GMP规范和具体GMP操作的行业规范,如一系列的“Guidance for Industry”文件,这些不断增补和修

新版美国GMP工业指导

新版美国GMP工业指导 该指导描述了FDA目前对该问题的规定。它不是给任何人制造和给予了权益,也不是要与FDA和公众实施。假如有其他途径能满足实施的法规和规章制度,能够进行改变。 I.简介(1) A.目的(1.1) 这份文件试图为在质量治理的合适的系统下生产APIs的GMP提供相关指导。它也试图关心去保证生产出来的APIs能满足对它提出的质量和纯度的标准。 在这份指导中,“生产”那个词能够定义为包含所有APIs原料的购入,生产,包装,重包装,标记,重标记,质量操纵,发放,贮存,和分发以及相关的操纵;“应该”那个词定义为保证与CGMP相符。假如有其他途径能满足实施的法规和规章制度,能够进行改变。在那个目的的指导下,CGMP与GMP是相同的, 这份指导作为一个整体没有包含从事生产的人员的安全条款,也没有与环保相关的条款。这些方面的操纵是生产者固有的责任,也由国家法律来约束。 这份指导并不是试图定义登记和档案治理需求或者修改药典的规定,也不是要阻碍负责规章制度的机构在市场/生产授权或者药物申请下建立与APIs有关的特定的登记/档案治理需求。登记/档案治理文件中的所有条款必须被履行。 B.调整适应性(1.2) 在世界那个范畴内,原料像APIs一样依照法律的分类也有所不同。当一个原料在一个地区或者国家被归类于APIs,被生产并用于药物产品,那么它就必须按照该指导进行生产。 C.范畴(1.3) 这份指导用于人类药物(医疗)产品中APIs的生产,也用于由赶忙优先于使APIs无菌的无菌APIs的生产中。这种无菌APIs的无菌操作没有包括在该指导当中,但应该依照当地官方定义的药物(医疗)产品的GMP指导去执行。 这份指导包含了APIs通过化学合成,提取,细胞培养/发酵,天然药物中的获得,或者与这些相关操作的生产。专门的APIs细胞培养/发酵法生产在XVIII(18)中详细描述。 这份指导包含了所有的疫苗,整个细胞,整个血液和原生质,血液和原生质衍生物(原生质片段),和基因治疗的APIs。然而,它并没有包含用血液或原生质作为原料来生产APIs。有些细胞基质(哺乳动物,植物,昆虫或者微生物细胞,组织或动物来源包括转基因动物)和早期生产步骤包含在GMP中但没有包含在这份指导中。另外,这份指导没有包括医疗用气体,大包装药物(医疗)产品(如在大包装中的药片,胶囊),或者放射性药物。 第XIX(19)章中包含了特定用于临床治疗途径(研究性医疗产品)的药物(医疗)产品APIs的生产指导。 API的“起始原料”是初始原料,中间体,或者用于API产品的API,同时与API结合作为重要的结构框架;开始原料是贸易项目,在合同或者贸易协议下,或者自己生产,从一个或者更多的供货者手中购买原料;它总的来说组成了化学物质和结构。 一个公司应该指出和列出API开始生产的点的差不多原理。在合成时期,它指出了API 开始原料该在什么时候加入该进程中;在其他时期(如:发酵,提取,纯化),那个原理应该建立在一件件的具体情形的基础上。表1给出了API开始原料该在什么时候加入该进程中。 从这点开始,如这份指导中定义的合适的GMP应该被应用于这些中间体和API生产步

美国cGMP 中英文对照版

美国药品生产质量管理规范 210部分和211部分 PART 210 CURRENT GOOD MANUFACTURING PRACTICE IN MANUFACTURING, PROCESSING, PACKING, OR HOLDING OF DRUGS; GENERAL 210部分人用及兽用药品的生产、加工、包装或贮存的CGMP 编者说明:英文版2010-05-23取自FDA官方网站 中文版取自中国GMP网友关大鹏2009-12-18的发的2003-12译本 两个版本可能稍有不同,不同之处红字标示。

目录 210.1 cGMP法规的地位 (2) 210.2 cGMP法规的适用性 (2) 210.3 定义 (2) 211-A- 总则 (4) 211-B- 组织与人员 (4) 211-C- 厂房和设施 (5) 211-D- 设备 (7) 211-E- 成份、药品容器和密封件的控制 (8) 211-F- 生产和加工控制 (10) 211-G- 包装和标签控制 (11) 211-H- 贮存和销售 (13) 211-I- 实验室控制 (14) 211-J- 记录和报告 (16) 211-K- 退回的药品和回收处理 (20) Sec. 210.1 Status of current good manufacturing practice regulations. 210.1 cGMP法规的地位 (a) The regulations set forth in this part and in parts 211 through 226 of this chapter contain the minimum current good manufacturing practice for methods to be used in, and the facilities or controls to be used for, the manufacture, processing, packing, or holding of a drug to assure that such drug meets the requirements of the act as to safety, and has the identity and strength and meets the quality and purity characteristics that it purports or is represented to possess. (a) 在本部分及21CFR 211—226部分中陈述的法规是在药品生产、加工、包装或贮存中使用的现行生产质量管理规范及使用的设施或控制的最低标准,以保证该药品符合联邦食品、药品及化妆品法对安全性的要求,具有均一性和效价(或含量)并符合或代表其生产过程的质量及纯度等特征。 (b) The failure to comply with any regulation set forth in this part and in parts 211 through 226 of this chapter in the manufacture, processing, packing, or holding of a drug shall render such drug to be adulterated under section 501(a)(2)(B) of the act and such drug, as well as the person who is responsible for the failure to comply, shall be subject to regulatory action. (b) 凡是在药品生产、加工、包装或贮存过程中存在任何不符合本部分及21CFR 211—226部分中陈述的法规的药品,依据联邦食品、药品及化妆品法501 (a)(2)-(B),该药应被视为劣药,同时

条款中英文对照版

Fédération Internationale Des Ingénieurs-Conseils 通用条件 General Conditions 1.一般规定 General Provisions 1.1 定义 Definitions 在包括专用条件和本通用条件的合同条件(“本合同条件”)中,以下措辞和用语的含义如下所述。除非上下文中另有要求,指当事人和当事各方的词包括公司和其它法律实体。 In the Conditions of Contract ("these Conditions"), which include Particular Conditions and these General Conditions, the following words and expressions shall have the meanings stated. Words indicating persons or parties include corporations and other legal entities, except where the context requires otherwise. 1.1.1 合同 1.1.1 The Contract 1.1.1.1“合同(Contract)”指合同协议书、中标函、投标函、本合同条件、规范、图纸、资料表、以及在合同协议书或中标函中列明的其它进一步的文件(如有时)。 1.1.1.1 "Contract" means the Contract Agreement, the Letter of Acceptance, the Letter of Tender, these Conditions, the Specification, the Drawings, the Schedules, and the further documents (if any) which are listed in the Contract Agreement or in the Letter of Acceptance. 1.1.1.2“合同协议书(Contract Agreement)”指第1.6款【合同协议】中所说明的合同协议(如有时)。 1.1.1.2 "Contract Agreement" means the contract agreement (if any) referred to in Sub-Clause 1.6 [Contract Agreement]. 1.1.1.3“中标函(Letter of Acceptance)”指雇主对投标文件签署的正式接受函,包括其后所附的备忘录(由合同各方达成并签定的协议构成)。在没有此中标函的情况下,“中标函”一词就指合同协议书,颁发或接收中标函的日期就指双方签订合同协议书的日期。 1.1.1.3 "Letter of Acceptance" means the letter of formal acceptance, signed by the Employer, of the Letter of Tender, including any annexed memoranda comprising agreements between and signed by both Parties. If there is no such letter of acceptance, the expression "Letter of Acceptance" means the Contract Agreement and the date of issuing or receiving the Letter of Acceptance means the date of signing the Contract Agreement. 1.1.1.4“投标函(Letter of Tender)”指名称为投标函的文件,由承包商填写,包括已签字的对雇主的工程报价。 1.1.1.4 "Letter of Tender" means the document entitled letter of tender, which was completed by the Contractor and includes the signed offer to the Employer for the Works. 1.1.1.5“规范(Specification)”指合同中名称为规范的文件,及根据合同规定对规范的增加和修改。此文件具体描述了工程。 1.1.1.5 "Specification" means the document entitled specification, as included in the Contract, and any additions and modifications to the specification in accordance with the Contract. Such document specifies the Works. 1.1.1.6“图纸(Drawings)”指合同中规定的工程图纸,及由雇主(或代表)根据合同颁发的对

中国、美国、欧盟GMP中英文版

中华人民共和国卫生部令 第79 号 《药品生产质量管理规范(2010年修订)》已于2010年10月19日经卫生部部务会议审议通过,现予以发布,自2011年3月1日起施行。 部长陈竺 二○一一年一月十七日 第一章总则 第一条为规范药品生产质量管理,根据《中华人民共和国药品管理法》、《中华人民共和国药品管理法实施条例》,制定本规范。 第二条企业应当建立药品质量管理体系。该体系应当涵盖影响药品质量的所有因素,包括确保药品质量符合预定用途的有组织、有计划的全部活动。 第三条本规范作为质量管理体系的一部分,是药品生产管理和质量控制的基本要求,旨在最大限度地降低药品生产过程中污染、交叉污染以及混淆、差错等风险,确保持续稳定地生产出符合预定用途和注册要求的药品。 第四条企业应当严格执行本规范,坚持诚实守信,禁止任何虚假、欺骗行为。 第二章质量管理 第一节原则 第五条企业应当建立符合药品质量管理要求的质量目标,将药品注册的有关安全、有效和质量可控的所有要求,系统地贯彻到药品生产、控制及产品放行、贮存、发运的全过程中,确保所生产的药品符合预定用途和注册要求。 第六条企业高层管理人员应当确保实现既定的质量目标,不同层次的人员以及供应商、经销商应当共同参与并承担各自的责任。 第七条企业应当配备足够的、符合要求的人员、厂房、设施和设备,为实现质量目标提供必要的条件。 第二节质量保证 第八条质量保证是质量管理体系的一部分。企业必须建立质量保证系统,同时建立完整的文件体系,以保证系统有效运行。 第九条质量保证系统应当确保: (一)药品的设计与研发体现本规范的要求; (二)生产管理和质量控制活动符合本规范的要求; (三)管理职责明确;

中国新版GMP与欧盟、美国GMP比较概要

New Chinese, EU and US GMPs comparative overview 中国新版,欧盟和美国GMP 比较概要 Date: 24 – February – 2011 日期:2011年2月24日 Authors作者:: Víctor Samitier (TDV) / Octavi Colomina (TDV)

1.Quality Management 质量管理 SIMILARITIES 相似点: EU and new Chinese GMPs set forth the requirement of establish a quality system, with their own quality objectives, applying all of requirements on safety, efficacy and quality control related with drug registration to the whole process of drug production. EU and new Chinese GMPs introduce the concepts of Quality Assurance in the same way. US GMPs does not establish the requirement of having a quality system, but it can be understood with the whole reading of 21 CFR Parts 210 &211. 欧盟和新版中国GMP都建立了质量系统要求,包括了确立相应质量目标,应用从药品注册到整个药品生产工艺的安全,成效和质量控制。欧盟和新版中国GMP引入了相同的质量保证概念。美国GMP并没有建立质量系统的需求,但是通过对21CFR Parts 210和211的解读能够体现质量系统概念。 Again EU and new Chinese GMPs introduce clearly the Risk Management Approach according to ICH Q9 concepts. Conversely, 21 CFR does not mention these concepts which are much newer than last revision of US GMPs. 欧盟和中国新版GMP又再次重申了了由ICH Q9引出的质量管理方法。相对陈旧的美国GMP 21CFR 没有涉及到这些最新概念。 DIFFERENCES 差异点: New Chinese GMPs identifies the responsibility of top management personnel for the implementation of quality objectives (ICH Q10 concept). 新版中国GMP确立了实施质量目标最高管理层人员的责任,这一点来源于ICH Q10概念。 https://www.360docs.net/doc/097708975.html,anization and personnel 组织机构和人员 SIMILARITIES 相似点: People should be trained in their duties and in cGMPs, previously to perform their duties and continuously, by qualified personnel. A training programme should be in place to assure training is performed periodically, although US GMP does not specify it. 人员需要根据其GMP中的职责,上岗前进行培训,培训需要有连续性,并且由具有资质的人员实施。培训计划应到位以保证培训能够定期进行,但美国GMP没有涉及培训计划。 DIFFERENCES 差异点: Main responsibilities of Head of Production, Quality Unit and Qualified person are described in EU and new Chinese GMPs. Nevertheless, new Chinese GMPs also identify the minimum training of the person in charge of production (degree in pharmacy), quality unit (degree in pharmacy or related), and qualified person (Bachelor degree in pharmacy or related). US GMP does not include any consideration about it. 在欧盟和中国新版GMP中明确了生产,质量主管和质量授权人的主要责任。然而,新版中国GMP 同时明确了人员的最低资质要求:生产负责人(药学或相关专业),质量单元(药学或相关专业)和质量授权人(药学或相关专业本科学位)。美国GMP没有上述要求。 EU GMPs and new Chinese GMPs are much more specific with Personnel hygienic, requiring an specific training programme. They also request periodical medical check to employees in direct contact with the product (at least one per year in new Chinese GMPs) 欧盟GMPs和中国新版GMP都特别关注了人员卫生,需要特别的培训内容。都要求进行直接接触产品的员工进行周期性体检(在中国新版GMP中规定至少每年1次)

各国国旗 (中英文对照版)

各国国旗 Afghanistan Albania Algeria Andorra Angola Antigua Argentina Armenia 阿富汗阿尔巴尼亚阿尔及利亚安道尔安哥拉安提瓜阿根廷亚美尼亚 Australia Austria Azerbaijan Bahamas Bahrain Bangladesh Barbados Belgium 澳大利亚奥地利阿塞拜疆巴哈马巴林孟加拉巴巴多斯比利时 Belize Benin Bhutan Bolivia Botswana Brazil Brunei Bulgaria 伯里兹贝宁不丹玻利维亚博茨瓦纳巴西文莱保加利亚 BurkinaFaso Burundi Cameroon Canada CapeVerde C.A.R Chad Chile 布基纳法索布隆迪喀麦隆加拿大佛得角中非乍得智利 China Colombia Comoros Congo Costa Rica Cuba Cyprus Czech 中国哥伦比亚科摩罗刚果哥斯达黎加古巴塞浦路斯捷克 El Equatorial Estonia Denmark Dominica Dominican Ecuador Egypt Salvador 丹麦多米尼克多米尼加厄瓜多尔埃及萨尔瓦多赤道几内亚爱沙尼亚

Fiji Finland France Gabon Gambia Georgia Germany Ghana 斐济芬兰法国加蓬冈比亚乔治亚德国加纳 Greece Grenada Guatemala GuineaBissau Guinea Guyana Haiti Holy See 希腊格林纳达危地马拉几内亚比绍几内亚圭亚那海地梵蒂冈 Honduras Hungary Iceland India Idonesia Iran Iraq Ireland 洪都拉斯匈牙利冰岛印度印度尼西亚伊朗伊拉克爱尔兰 Israel Italy Jamaica Japan Jordan Kenya Kiribati Korea(N) 以色列意大利牙买加日本约旦肯尼亚基里巴斯朝鲜 Korea(S) Kuwait Laos Latvia LLebanon Lesotho Liberia Libya 韩国科威特老挝拉托维亚黎巴嫩莱索托利比里亚利比亚 Lithuania Madagascar Malawi Malaysia Maldives Mali Malta Man,Isle. 立陶宛马达加斯加马拉维马来西亚马尔代夫马里马耳他马恩岛 Mauritania Mauritius Mexico Monaco Mongolia Morocco Mozambique Namibia 毛里塔尼亚毛里求斯墨西哥摩纳哥蒙古摩洛哥莫桑比克纳米比亚

中英文对照版财务报表

一、企业财务会计报表封面 FINANCIAL REPORT COVER 报表所属期间之期末时间点 Period Ended 所属月份 Reporting Period 报出日期 Submit Date 记账本位币币种 Local Reporting Currency 审核人 Verifier 填表人 Preparer 二、资产负债表 Balance Sheet 资产 Assets 流动资产 Current Assets 货币资金 Bank and Cash 短期投资 Current Investment 一年内到期委托贷款 Entrusted loan receivable due within one year 减:一年内到期委托贷款减值准备 Less: Impairment for Entrusted loan receivable due within one year 减:短期投资跌价准备 Less: Impairment for current investment 短期投资净额 Net bal of current investment 应收票据 Notes receivable 应收股利 Dividend receivable 应收利息 Interest receivable 应收账款 Account receivable 减:应收账款坏账准备 Less: Bad debt provision for Account receivable 应收账款净额 Net bal of Account receivable 其他应收款 Other receivable 减:其他应收款坏账准备 Less: Bad debt provision for Other receivable 其他应收款净额 Net bal of Other receivable 预付账款 Prepayment 应收补贴款 Subsidy receivable 存货 Inventory 减:存货跌价准备 Less: Provision for Inventory 存货净额 Net bal of Inventory 已完工尚未结算款 Amount due from customer for contract work 待摊费用 Deferred Expense 一年内到期的长期债权投资 Long-term debt investment due within one year 一年内到期的应收融资租赁款 Finance lease receivables due within one year 其他流动资产 Other current assets 流动资产合计 Total current assets 长期投资 Long-term investment 长期股权投资 Long-term equity investment 委托贷款 Entrusted loan receivable 长期债权投资 Long-term debt investment

美国GMP中文版

GOOD MANUFACTURE PRACTICE 美国药品生产质量管理规范 (CGMP) 二○○三年十二月

目录 210.1 cGMP法规的地位 (2) 210.2 cGMP法规的适用性 (2) 210.3 定义 (2) 211-A- 总则 (4) 211-B- 组织与人员 (4) 211-C- 厂房和设施 (5) 211-D- 设备 (7) 211-E- 成份、药品容器和密封件的控制 (8) 211-F- 生产和加工控制 (10) 211-G- 包装和标签控制 (11) 211-H- 贮存和销售 (13) 211-I- 实验室控制 (14) 211-J- 记录和报告 (16) 211-K- 退回的药品和回收处理 (20)

210部分—人用及兽用药品 的生产、加工、包装或贮存的CGMP 210.1 cGMP法规的地位 (a) 在本部分及21CFR 211—226部分中陈述的法规是在药品生产、加工、包装或贮存中使用的现行生产质量管理规范及使用的设施或控制的最低标准,以保证该药品符合联邦食品、药品及化妆品法对安全性的要求,具有均一性和效价(或含量)并符合或代表其生产过程的质量及纯度等特征。 (b) 凡是在药品生产、加工、包装或贮存过程中存在任何不符合本部分及21CFR 211—226部分中陈述的法规的药品,依据联邦食品、药品及化妆品法501 (a)(2)-(B),该药应被视为劣药,同时导致该事故发生的负责人应受相应的法规的制裁。 210.2 cGMP法规的适用性 (a) 本部分及21CFR 211—226适用于普通药品,21CFR 600—680适用于人用生物制品,除非另有明确规定,否则上述两者之间应该是相互补充而不是相互取代。如有上述两部分的法规不适用的药品,则可用特定的具体法规来替代。 210.3 定义 (a) 在联邦食品、药品及化妆品法201部分中包含的定义和解释、说明适用于21CFR 211—226部分中的术语。 (b) 下面定义的术语适用于本部分及21CFR 211—226。 (1) 法(Act) 指联邦食品、药品及化妆品法,修订版(21 U.S.C 301 et seq.)。 (2) 批(Batch) 指在规定限度内,按照某一生产指令在同一生产周期内生产出来的,具有同一性质和质量的一定数量的药品或其它物料。 (3) 组分(Component) 指用于药品生产的所有成份,包括那些未在药品中出现的成份。 (4) 药品(Drug Product) 指成品制剂(如:片剂、胶囊剂、口服液等),通常含有一种活性成份并伴有非活性成份(但不是必需的)。本术语也包括不含有活性成份但作为安慰剂使用的成品制剂。 (5) 纤维(Fiber) 指长度大于其宽度的3倍的任何微粒状污染物。 (6) 无纤维脱落的过滤器(Non-fiber-releasing filter) 指任何经过适当的预处理(如清洗或冲洗)后,不会将纤维脱落到已过滤的组分或药品中的所有过滤器。所有含石棉过滤器均被认为是有纤维脱落的过滤器。 (7) 活性成份(Active Ingredient)

相关文档
最新文档