RegisterSign in
ViewPDF
- Access throughyour institution
Article preview
- Abstract
- Introduction
- Section snippets
- References (66)
- Cited by (17)
- Recommended articles (6)
Regulatory Toxicology and Pharmacology
Volume 79, Supplement 1,
15 August 2016
, Pages S3-S10
Author links open overlay panel
https://doi.org/10.1016/j.yrtph.2016.05.029Get rights and content
Abstract
Cross-contamination in multi-product pharmaceutical manufacturing facilities can impact both product safety and quality. This issue has been recognized by regulators and industry for some time, leading to publication of a number of continually evolving guidelines. This manuscript provides a historical overview of the regulatory framework for managing cross-contamination in multi-product facilities to provide context for current approaches. Early guidelines focused on the types of pharmaceuticals for which dedicated facilities and control systems were needed, and stated the requirements for cleaning validation. More recent guidelines have promoted the idea of using Acceptable Daily Exposures (ADEs) to establish cleaning limits for actives and other potentially hazardous substances. The ADE approach is considered superior to previous methods for setting cleaning limits such as using a predetermined general limit (e.g., 10ppm or a fraction of the median lethal dose (LD50) or therapeutic dose). The ADEs can be used to drive the cleaning process and as part of the overall assessment of whether dedicated production facilities are required. While great strides have been made in using the ADE approach, work remains to update good manufacturing practices (GMPs) to ensure that the approaches are clear, consistent with the state-of-the-science, and broadly applicable yet flexible enough for adaptation to unique products and situations.
Introduction
Pharmaceutical companies, regulators, and other stakeholders such as public health advocacy groups maintain a partnership in their concern for product quality and occupational safety during the manufacture of pharmaceutical products. While principles of risk management are considered integral to all aspects of pharmaceutical business, risk management principles and quality systems approaches have not been consistently utilized or applied in pharmaceutical manufacturing (ISPE, 2010). In recent years, cross-contamination of medicinal products in shared facilities has come under increased regulatory scrutiny. Global regulations and guidelines for preventing cross-contamination have been published and have increasingly taken a risk-based approach. To evaluate and discuss a consistent approach for assessing health hazards of drug substances (DS) and intermediates (IM) and derivation of health-based exposure limits, representatives of pharmaceutical companies and consultants joined efforts in an expert workshop (Weideman etal., 2015). The purpose of this manuscript is to review how Good Manufacturing Practice (GMP) regulations have evolved with respect to cross-contamination issues in multi-product facilities, the influences that led to this evolution, and finally identification of areas of future dialogue between private partners and regulatory authorities. This dialogue would ensure that the interests of all parties in achieving practical, consistent, science-based, and health-protective strategies for preventing cross-contamination can be achieved. Table1 lists a summary of available guidelines for use in pharmaceutical manufacturing and safety. It also lists additional resources for using risk-based approaches to set safe exposure limits for environmental and workplace chemical exposures that can also provide useful information for pharmaceutical risk assessments.
Section snippets
Historical review of regulatory approaches for GMPs as they pertain to cross-contamination in multi-product facilities
In 1978, the US Food and Drug Administration (US FDA) issued regulations pertaining to “minimum current GMP” for preparation of drug products for administration to humans or animals (US FDA, 1978). Subpart C of 21 CFR 211.42 broadly outlined requirements for the prevention of cross-contamination. It states “Operations shall be performed within specifically defined areas of adequate size. There shall be separate or defined areas or such other control systems for the firm's operations as are
Historical review of approaches for establishing cleaning limits
Cleaning pharmaceutical manufacturing equipment is a fundamental means of preventing cross-contamination in multi-product facilities (Walsh, 2011a). WHO, PIC/S, Heath Canada, and ANVISA (Brazil) guidelines all focus principally on systems rather than cleaning and health limit determinations. Risk-MaPP, US FDA, and EMA have provided additional guidance on the process for determining health-based limits for cleaning. The US FDA briefly mentions acceptance criteria (see quote below) but does not
Approaches to further harmonization of GMPs
To achieve global consistency in GMPs for pharmaceutical products further harmonization is necessary. GMP practices in some countries may require updated language specifically to eliminate undefined terms such as “certain hormones”, “certain steroids”, “cytotoxics”, and “highly active”. Lack of definition allows for individual interpretation and potentially impacts the safe and efficient manufacture of pharmaceuticals (Olson etal., 2016, Sussman etal., 2016a, this issue). Similarly, with
Summary and conclusions
In multi-product pharmaceutical facilities, the possibility of cross-contamination exists, particularly when there is change-over from one product line to another. Since pharmaceuticals by nature have biological activity, including some at very low doses, steps are necessary to ensure that cross-contamination is well controlled. The importance of controlling cross-contamination has been recognized by the pharmaceutical industry and regulators alike for decades. Early approaches, as described in
Acknowledgements
The statements and conclusions in this paper reflect the opinions of the authors and do not necessarily represent official policies of the organizations as listed on the title page. The authors would like to acknowledge Patricia Weideman, Andrew Maier, and Alison Pecquet for organizing and facilitating the workshop that served as the basis for developing this manuscript. The authors would also like to thank all of the participants of the workshop for their contributions at the workshop and
References (66)
- I.C. Munro et al.The threshold of toxicological concern (TTC) in risk assessment
Toxicol. Lett.
(2008)
- R. Kroes et al.Structure-based thresholds of toxicological concern (TTC): guidance for application to substances present at low levels in the diet
Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc.
(2004)
- M.L. Dourson et al.Regulatory history and experimental support of uncertainty (safety) factors
Regul. Toxicol. Pharmacol. RTP
(1983)
- D.G. Dolan et al.Application of the threshold of toxicological concern concept to pharmaceutical manufacturing operations
Regul. Toxicol. Pharmacol.
(2005)
- J.P. Bercu et al.Application of the threshold of toxicological concern concept when applied to pharmaceutical manufacturing operations intended for short-term clinical trials
Regul. Toxicol. Pharmacol.
(2013)
- ANVISA
Resolution – RDC No 17, of 16.04.10, Provisions for the Good Practices of Medicament Manufacturing
(2010)
- ATSDR
Public Health Assessment Guidance Manual
(2015)
- J.P. Bercu et al.
Point of departure (PoD) selection for the derivation of acceptable daily exposure (ADE) values for active pharmaceutical ingredients (APIS)
Regul. Toxicol. Pharmacol.
(2016)
- CEFIC
APIC Guidance on Aspects of Cleaning Validation in Active Pharmaceutical Ingredient Plants
(2014)
- D.L. Conine et al.
Setting health-based residue limits for contaminants in pharmaceuticals and medical devices
Qual. Assur. San. Diego Calif.
(1992)
IEH Reports and Publications
(2015)
European Centre for Ecotoxicology and Toxicology of Chemicals (ECETOC) Publications
(2015)
Guidance on Information Requirements and Chemical Safety Assessment
(2015)
Part 1- chapter 3: premises and equipment
Part 1- chapter 5: production
Guideline on Setting Health Based Exposure Limits for Use in Risk Identification in the Manufacture of Different Medicinal Products in Shared Facilities
(2014)
Guideline on Setting Specifications for Related Impurities in Antibiotics
(2012)
Guideline on the Approach to Establish a Pharmacological ADI
(2012)
Questions and Answers on the Guideline on the Limits of Genotoxic Impurities; EMA/CHMP/SWP/431994/2007
(2010)
Update on Revision of Chapters 3 and 5 of the GMP Guide: “Dedicated facilities.” EMA/INS/GMP/809387/2009
(2009)
Guideline on the Specification Limits for Residues of Metal Catalysts or Metal Reagents. Committee for Medicinal Products for Human Use (CHMP). Doc. Ref.EMEA/CHMP/Swp/4446/2000
(2008)
Guideline on Excipients in the Dossier for Application for Marketing Authorisation of a Medicinal Product. Doc. Ref.EMEA/CHMP/QWP/396951/2006
(2007)
Guideline on the Limits of Genotoxic Impurities. Committee for Medicinal Products for Human Use (CHMP)
(2006)
Q6B Specifications: Test Procedures and Acceptance Criteria for Biotechnological/Biological Products
(1999)
The Rules Governing Medicinal Products in the European Union
(2008)
Using default methodology to derive the acceptable daily exposure (ADE)
Regul. Toxicol. Pharmacol.
(2016)
Harmonization effort for acceptable exposure methodology applied to pharmaceutical cleaning validation- operational aspects
Regul. Toxicol. Pharmacol.
(2016)
Good Manufacturing Practices (GMP) Guidelines: 2009 Edition, Version 2 (GUI-0001)
(2009)
Cleaning Validation Guidelines (GUIDE-0028)
(2008)
ICH Guidelines
(2015)
ICH Guideline M7 on Assessment and Control of DNA Reactive (Mutagenic) Impurities in Pharmaceuticals to Limit Potential Carcinogenic Risk
(2014)
ICH Q3D Technical Document Q3D: Guideline for Elemental Impurities
(2014)
Impurities: Guideline for Residual Solvents Q3C(R5) Step 4 Version
(2011)
Cited by (17)
- Setting impurity limits for endogenous substances: Recommendations for a harmonized procedure and an example using fatty acids
2022, Regulatory Toxicology and Pharmacology
Endogenous substances, such as fatty, amino, and nucleic acids, are often purposefully used in parenterally pharmaceuticals, but may be present as impurities. Currently, no consensus guidance exists on setting impurity limits for these substances. Specific procedures are needed, as the amount and types of toxicity data available for endogenous substances are typically far less than those for other chemical impurities. Additionally, the parenteral route of administration of these substances is inherently non-physiological, resulting in potentially different or increased severity of toxicity. Risk Assessment Process Maps (RAPMAPs) are proposed as a model to facilitate the development of health-based exposure limits (HBELs) for endogenous substances. This yielded a framework that was applied to derive HBELs for several fatty acids commonly used in parenteral pharmaceuticals. This approach was used to derive HBELs with further vetting based on anticipated perturbations in physiological serum levels, impacts of dose-rate, and consideration of intermittent dosing. Parenteral HBELs of 100–500mg/day were generated for several fatty acids, and a proposed class-based limit of 50mg/day to be used in the absence of chemical-specific data. This default limit is consistent with the low toxicity of this chemical class and ICH Q3C value for Class 3 solvents.
- Green chemistry and sustainability metrics in the pharmaceutical manufacturing sector
2022, Current Opinion in Green and Sustainable Chemistry
This review article summarizes recent developments of green chemistry and sustainability metrics with a focus on the pharmaceutical industry. We discuss synergies and discrepancies of the green chemistry principles with regulatory guidelines in the field of pharmaceutical process development and manufacturing under Good Manufacturing Practices (GMP). This review captures selected views and publications on green chemistry and sustainability metrics. It discusses two recent process development highlights from the pharmaceutical industry which use different approaches to optimize the process mass intensity (PMI)of small-molecule active pharmaceutical ingredient (API) manufacturing processes. The first example is the development of green process conditions by switching reaction media from organic solvents to micellar catalysis in water in the Takeda process of the API TAK-954. The second example features process intensifications accomplished by the application of innovative technologies in pharmaceutical processing in the Merck process of the API MK-7264.
(Video) Pharmaceutical Quality Symposium 2021 Part 7 - Health-based exposure limits and toxicology in the pharmaceutical industry
2021, Toxicological Risk Assessment and Multi-System Health Impacts from Exposure
Cross-contamination is inevitable when it comes to the production of different products in the same manufacturing facility. For decades, many different approaches have been suggested and applied by regulators and institutions to control the risk of cross-contamination in shared facilities. Empirical methods have been replaced by the determination of health-based exposure limits in recent years. This innovation has resulted in toxicological risk assessment being included in manufacturing processes. With the determination of the permitted daily exposure (PDE) value, which includes a very comprehensive scientific evaluation, it is expected that no health risks will occur at this level. Harmonization and standardization in this process will provide significant benefits to increase the quality of the reports. The aim of this chapter is to provide a basic understanding for the PDE determination strategy.
- Considerations when deriving compound-specific limits for extractables and leachables from pharmaceutical products: Four case studies
2020, Regulatory Toxicology and Pharmacology
Leachables from pharmaceutical container closure systems are a subset of impurities that present in drug products and may pose a risk to patients or compromise product quality. Extractable studies can identify potential leachables, and extractables and leachables (E&Ls) should be evaluated during development of the impurity control strategy. Currently, there is a lack of specific regulatory guidance on how to risk assess E&Ls; this may lead to inconsistency across the industry. This manuscript is a cross-industry Extractables and Leachables Safety Information Exchange (ELSIE) consortium collaboration and follow-up to Broschard et al. (2016), which aims to provide further clarity and detail on the conduct of E&L risk assessments. Where sufficient data are available, a health-based exposure limit termed Permitted Daily Exposure (PDE) may be calculated and to exemplify this, case studies of four common E&Ls are described herein, namely bisphenol-A, butylated hydroxytoluene, Irgafos® 168, and Irganox® 1010. Relevant discussion points are further explored, including the value of extractable data, how to perform route-to-route extrapolations and considerations around degradation products. By presenting PDEs for common E&L substances, the aim is to encourage consistency and harmony in approaches for deriving compound-specific limits.
- Harmonization efforts for deriving health-based exposure limits in the pharmaceutical industry – Advancing the current science and practice
2016, Regulatory Toxicology and Pharmacology
- Identifying and assessing highly hazardous drugs within quality risk management programs
2016, Regulatory Toxicology and Pharmacology
Citation Excerpt :
However, control of contamination as well as occupational exposures must also be considered. In past years, regulatory authorities have identified “categories of concern” for potentially hazardous active pharmaceutical ingredients (APIs) to implement this mission (Sargent et al., 2016, this issue). This method allowed manufacturers to provide appropriate segregation, where necessary, according to current Good Manufacturing Practices (cGMP).
Historically, pharmaceutical industry regulatory guidelines have assigned certain active pharmaceutical ingredients (APIs) to various categories of concern, such as “cytotoxic”, “hormones”, and “steroids”. These categories have been used to identify APIs requiring segregation or dedication in order to prevent cross-contamination and protect the quality and safety of drug products. Since these terms were never defined by regulatory authorities, and many novel pharmacological mechanisms challenge these categories, there is a recognized need to modify the historical use of these terms. The application of a risk-based approach using a health-based limit, such as an acceptable daily exposure (ADE), is more appropriate for the development of a Quality Risk Management Program (QRMP) than the use of categories of concern. The toxicological and pharmacological characteristics of these categories are discussed to help identify and prioritize compounds requiring special attention. Controlling airborne concentrations and the contamination of product contact surfaces in accordance with values derived from quantitative risk assessments can prevent adverse effects in workers and patients, regardless of specific categorical designations to which these APIs have been assigned. The authors acknowledge the movement away from placing compounds into categories and, while not yet universal, the importance of basing QRMPs on compound-specific ADEs and risk assessments. Based on the results of a risk assessment, segregation and dedication may also be required for some compounds to prevent cross contamination during manufacture of APIs.
Recommended articles (6)
Research article
Regulatory framework of pharmaceutical compounding and actual developments of legislation in EuropeHealth Policy, Volume 117, Issue 3, 2014, pp. 328-333
Pharmaceutical preparations are medicines that the pharmacist makes for the special needs of the patients that the pharmaceutical industry cannot comply for economic and logistic reasons. Pharmacy compounding is still an important component of pharmacy practice and a valuable therapeutical service that is an integrant part of the modern health care system, but its legislation is not harmonized among European and US countries.
In 2011 the Committee of Ministers of the Council of Europe has adopted a Resolution on quality and safety assurance requirements for medicinal products prepared in pharmacies for the special needs of patients. Aim of this resolution is to harmonize quality assurance and standards for pharmacy-made medicinal products among European countries and to pass the gap in quality assurance and standards between preparation in pharmacies and medicines prepared by the pharmaceutical industry. This article will analyze the actual rules and technical norms that regulate compounding activity and the expectations resultants from the new European and US laws.
Research article
Cleaning Validation and Its Regulatory Aspects in the Pharmaceutical IndustryDevelopments in Surface Contamination and Cleaning, 2015, pp. 129-186
(Video) Sterility Validation 101: Ensuring a robust sterilization validation program from start to finishPharmaceutical products and active pharmaceutical ingredients (APIs) can be contaminated by other pharmaceutical products or APIs, by cleaning agents, by microorganisms, or by other materials (e.g., airborne particles, dust, lubricants, raw materials, intermediates, auxiliaries). To avoid contamination of the product, adequate cleaning procedures are essential. Cleaning validation describes validation of cleaning procedures for the removal of contaminants associated with the previous products, residues of cleaning agents, and the control of potential microbial contaminants and significantly reduces the amount of actives, excipient(s), and cleaning agent(s) to a concentration within defined acceptance limits.
Research article
Use of low-dose clinical pharmacodynamic and pharmacokinetic data to establish an occupational exposure limit for dapagliflozin, a potent inhibitor of the renal sodium glucose co-transporter 2Regulatory Toxicology and Pharmacology, Volume 67, Issue 1, 2013, pp. 89-97
Classical risk assessment models for setting safe occupational exposure limits (OEL) have used multiple uncertainty factors (UF) applied to a point of departure (POD), e.g., a No Observed Effect Level (NOEL), which in some cases is the pharmacological effect. Dapagliflozin promotes glucosuria by inhibiting the renal sodium–glucose cotransporter-2 transporter. The initial OEL for dapagliflozin (0.002mg/m3) was calculated when low dose clinical data was not available to identify a NOEL resulting in the need to use excessive UFs. To reduce the UFs from the OEL, a clinical pharmacodynamic [glucosuria and urinary glucose dipstick (UGD)] and pharmacokinetic study was conducted with single oral doses of 0.001, 0.01, 0.1, 0.3, 1.0 or 2.5mg administered to 36 healthy subjects. Dose-related dapagliflozin systemic exposures were observed at doses ⩾0.1mg and glucosuria was observed at doses ⩾0.3mg and corroborated by UGD. The NOEL was therefore 0.1mg for glucosuria. For setting the new OEL, no UFs were required. Dividing the POD by 10m3 (the volume of air an adult inhales in a workday), the resulting OEL was 0.01mg/m3. In conclusion, low-dose clinical pharmacodynamic and pharmacokinetic data can allow the OEL to be adjusted to the highest safe level.
Research article
Bioavailability of protein therapeutics in rats following inhalation exposure: Relevance to occupational exposure limit calculationsRegulatory Toxicology and Pharmacology, Volume 100, 2018, pp. 35-44
Protein therapeutics represent a rapidly growing proportion of new medicines being developed by the pharmaceutical industry. As with any new drug, an Occupational Exposure Limit (OEL) should be developed to ensure worker safety. Part of the OEL determination addresses bioavailability (BA) after inhalation, which is poorly understood for protein therapeutics. To explore this, male Sprague-Dawley rats were exposed intravenously or by nose-only inhalation to one of five test proteins of varying molecular size (10–150 kDa), including a polyethylene glycol-conjugated protein. Blood, lung tissue and bronchoalveolar lavage (BAL) fluid were collected over various time-points depending on the expected test protein clearance (8 minutes-56 days), and analyzed to determine the pharmacokinetic profiles. Since the BAL half-life of the test proteins was observed to be > 4.5 h after an inhalation exposure, accumulation and direct lung effects should be considered in the hazard assessment for protein therapeutics with lung-specific targets. The key finding was the low systemic bioavailability after inhalation exposure for all test proteins (∼≤1%) which did not appear molecular weight-dependent. Given that this study examined the inhalation of typical protein therapeutics in a manner mimicking worker exposure, a default 1% BA assumption is reasonable to utilize when calculating OELs for protein therapeutics.
Research article
Cleaning verification: Exploring the effect of the cleanliness of stainless steel surface on sample recoveryJournal of Pharmaceutical and Biomedical Analysis, Volume 134, 2017, pp. 108-115
The parameters affecting the recovery of pharmaceutical residues from the surface of stainless steel coupons for quantitative cleaning verification method development have been studied, including active pharmaceutical ingredient (API) level, spiking procedure, API/excipient ratio, analyst-to-analyst variability, inter-day variability, and cleaning procedure of the coupons. The lack of a well-defined procedure that consistently cleaned coupon surface was identified as the major contributor to low and variable recoveries. Assessment of acid, base, and oxidant washes, as well as the order of treatment, showed that a base-water-acid-water-oxidizer-water wash procedure resulted in consistent, accurate spiked recovery (>90%) and reproducible results (Srel≤4%). By applying this cleaning procedure to the previously used coupons that failed the cleaning acceptance criteria, multiple analysts were able to obtain consistent recoveries from day-to-day for different APIs, and API/excipient ratios at various spike levels. We successfully applied our approach for cleaning verification of small molecules (MW<1000Da) as well as large biomolecules (MW up to 50,000Da). Method robustness was greatly influenced by the sample preparation procedure, especially for analyses using total organic carbon (TOC) determination.
Research article
Modern science for better quality control of medicinal products “Towards global harmonization of 3Rs in biologicals”: The report of an EPAA workshopBiologicals, Volume 48, 2017, pp. 55-65
This article summarizes the outcome of an international workshop organized by the European Partnership for Alternative Approaches to Animal Testing (EPAA) on Modern science for better quality control of medicinal products: Towards global harmonization of 3Rs in biologicals. As regards the safety testing of biologicals, the workshop participants agreed to actively encourage the deletion of abnormal toxicity tests and target animal batch safety tests from all relevant legal requirements and guidance documents (country-specific guidelines, pharmacopoeia monographs, WHO recommendations). To facilitate the global regulatory acceptance of non-animal methods for the potency testing of, e.g., human diphtheria and tetanus vaccines and veterinary swine erysipelas vaccines, international convergence on the scientific principles of the use of appropriately validated invitro assays for replacing invivo methods was identified as an overarching goal. The establishment of scientific requirements for new assays was recognized as a further means to unify regulatory approaches in different jurisdictions. It was recommended to include key regulators and manufacturers early in the corresponding discussions. Manufacturers and responsible expert groups, e.g. at the European Directorate for the Quality of Medicines and Health Care of the Council of Europe or the European Medicines Agency, were invited to consider leadership for international collaboration.
© 2016 Elsevier Inc. All rights reserved.
FAQs
How can cross contamination be prevented in pharmaceutical industry? ›
Disinfect: Regular cleaning and sanitization is the core of preventing pharmaceutical contamination. In addition to thoroughly cleaning and disinfecting the facility, cleanliness, and hygiene of workwear is also important.
What is cross contamination in pharmaceutical industry? ›Pharmaceutical product cross-contamination refers to the. process by which foreign chemical, microbial, or physical. substances are unintentionally transferred from one. substance or object to medicines with harmful effects that. might affect the purity and quality of the pharmaceutical.
What is cross contamination in GMP? ›Cross-Contamination – Contamination of a starting material, intermediate product, finished product with another starting material or a product.
What is contamination and cross contamination in pharma industry? ›Contamination and Cross Contamination are two different things needs to be controlled in pharmaceuticals . Contamination is Spoilage of product with unknown Contaminant. Cross contamination/Mix up means the product is contaminated with known contaminant like paracetamol is contaminated with aspirin .
What are 4 methods for preventing cross contamination? ›- use different utensils, plates and chopping boards for raw and cooked food.
- wash utensils, plates and chopping boards for raw and cooked food thoroughly between tasks.
- make sure you do not wash raw meat.
- wash your hands after touching raw food and before you handle ready-to-eat food.
To prevent this: Wash hands with soap and hot water before and after handling food, and after using the bathroom, changing diapers; or handling pets. Use hot, soapy water and paper towels or clean cloths to wipe up kitchen surfaces or spills. Wash cloths often in the hot cycle of your washing machine.
What are the 4 types of cross contamination? ›There are four main types of contamination: chemical, microbial, physical, and allergenic. All food is at risk of contamination from these four types. This is why food handlers have a legal responsibility to ensure that the food they prepare is free from these contaminants and safe for the consumer.
What are the 3 types of cross contamination? ›There are three main types of cross contamination: food-to-food, equipment-to-food, and people-to-food.
What are 5 examples of cross contamination? ›Some examples are: Touching raw meats then handling vegetables or other ready-to-eat foods without washing hands between tasks. Using a food soiled apron or towel to wipe your hands between handling different foods. Failing to change gloves between handling different foods.
What are the 6 ways to prevent cross contamination? ›- Mind Your Cutting Boards. Every kitchen should have at least two cutting boards (at least!) ...
- Wash Your Hands! The signs in restaurants say this for a reason. ...
- Watch the Juices. ...
- Don't Rinse Meats. ...
- Properly Rinse Produce. ...
- Clean All Surfaces.
What do OSHA standards require in order to prevent cross contamination? ›
Cross contamination of the workplace can be prevented by removing or decontaminating PPE and washing hands before exiting the work zone. To prevent cross contamination at home, workers should wash their hands and faces at the end of a work shift and change into clean clothes and shoes.
What are the three elements of GMP? ›- Primary Materials and Products.
- Premises.
- People.
- Procedures.
- Processes.
- using the same knife or chopping board to cut both raw and ready-to-eat foods.
- using the handwash basin for defrosting food or placing dirty utensils and equipment.
- storing food uncovered or on the floor of the fridge or freezer.
- storing raw food above ready-to-eat food.
There are three main types of contamination to consider in pharmaceutical manufacturing and transportation. When you think about types of contaminants, consider which category they may fall into and how dangerous they may actually be.
What are the consequences of contamination in pharmaceutical setting? ›Because different drugs interact differently, cross contamination will negatively impact the drug's efficacy, cause other health problems, or trigger an allergic reaction in the patient. Even contamination via particle buildup from a single substance can cause problems by altering the intended dosage of the product.
What are the 5 steps that must be taken in order to prevent cross contact? ›- Read labels and be aware of allergens. ...
- Wash hands with soap and water and change gloves before prepping food. ...
- Wash, dry, and sanitize food prep areas. ...
- Use separate cooking utensils and a separate cutting board when preparing allergen-safe foods.
Hand hygiene. Use of personal protective equipment (e.g., gloves, masks, eyewear). Respiratory hygiene / cough etiquette. Sharps safety (engineering and work practice controls).
How do you prevent cross contamination provide 5 ways or guidelines on how we can prevent cross contamination in preparing food? ›- Check the delivery temperature. ...
- Keep Toxic Chemicals away from Food. ...
- Floor Level storage is a no-go. ...
- A well looked after Storage Space goes a long way. ...
- Use the FIFO system. ...
- Stick it with a Label.
While there are many food safety hazards that can cause food contamination, most fall into one of three categories: biological, physical or chemical contamination. In many cases, a single hazard can introduce more than one type of contamination to food.
What tool do you need to use to prevent cross contamination? ›Use Separate Equipment to Prevent Cross-Contamination
Use separate plates and utensils for cooked and raw foods. Use more than one cutting board – one for fresh produce and one for raw meat, poultry or seafood. Be sure to replace these cutting boards once they get cracks or excessive cuts from your knives.
What are the control measures for contamination? ›
To prevent airborne contamination, high-efficiency particulate air (HEPA) filters, airlocks and cleanroom suits are used.
What is the most common factor of cross contamination? ›It is most likely to happen when raw food touches or drips onto ready-to-eat food, equipment or surfaces.
What is the most common source of cross contamination? ›- Clothing: Dirty clothes can transport bacteria from one place to another. ...
- Utensils: Different utensils should be used to prepare different types of foods. ...
- Food Handlers: Coughing, sneezing or even touching your face or hair before handling food can cause cross-contamination.
- Microbiological hazards. Microbiological hazards include bacteria, yeasts, moulds and viruses.
- Chemical hazards. ...
- Physical hazards. ...
- Allergens.
Preventing cross contamination
Cross contamination is when harmful bacteria are accidentally transferred from raw food to ready-to-eat food. Most cases of food poisoning are caused by bacteria from raw food coming into contact with food that is ready to eat.
Wipe Up Spills Immediately. In addition to helping reduce the growth of the Listeria bacteria (which can grow at refrigerated temperatures), getting rid of spills — especially drips from thawing meats — will help prevent “cross-contamination,” where bacteria from one food spread to another.
What are five ways of preventing chemical contamination? ›- always label and store chemicals separately from food.
- use the appropriate chemical for the job you're doing.
- always follow the chemical manufacturer's instructions with regards to dilution, contact time and water temperature.
There are four groups of OSHA standards: General Industry, Construction, Maritime, and Agriculture. (General Industry is the set that applies to the largest number of workers and worksites). These standards are designed to protect workers from a wide range of hazards.
What are OSHA's 5 priorities? ›- Imminent Danger has top priority. ...
- Fatalities and Catastrophes are next in priority. ...
- Complaints and Referrals are OSHA's third priority. ...
- Programmed Inspections are the fourth priority.
With an estimated 7 million worksites to cover, OSHA prioritizes its inspections based on immediate danger situations, severe injuries, worker complaints, referrals, targeted inspections, and follow-up inspections.
What are the 5 P's of GMP? ›
Good manufacturing practices (GMPs) help to ensure the consistent quality and safety of products by focusing attention on five key elements, which are often referred to as the 5 P's of GMP—people, premises, processes, products and procedures (or paperwork).
What are the 10 basic principles of GMP? ›- Create Standard Operating Procedures (SOPs)
- Enforce / Implement SOPs and work instructions.
- Document procedures and processes.
- Validate the effectiveness of SOPs.
- Design and use working systems.
- Maintain systems, facilities, and equipment.
- Develop job competence of workers.
The CGMP regulations for drugs contain minimum requirements for the methods, facilities, and controls used in manufacturing, processing, and packing of a drug product. The regulations make sure that a product is safe for use, and that it has the ingredients and strength it claims to have.
Which factors increase the risk of cross contamination? ›- Food from unsafe sources.
- Inadequate cooking.
- Improper holding temperatures.
- Contaminated equipment.
- Poor personal hygiene.
Microscopical analysis is particularly well suited to the analysis of particulate contamination because the particles are usually too small to be analyzed using conventional methods. Proper methods of sample isolation and preparation are also critical to the successful pharmacuetical contamination identification.
What are the 3 main contaminants? ›The three types of contamination are biological, physical, and chemical.
What principles will help to Minimise the risk of contamination in pharmaceutical production? ›Orderly placing and logical positioning of equipment and materials. Entry of materials through separate airlocks. - should permit easy and effective cleaning. Pipework, ventilation and light points and other services should be designed to avoid creation of recesses which are difficult to clean.
What is contamination in pharmaceutical industry? ›Contamination is defined as the undesired introduction of impurities of a chemical or microbiological nature, or of foreign matter, into or onto a raw material, intermediate, or API (Active Pharmaceutical Ingredient) during production, sampling, packaging or repackaging, storage or transport.
What are seven ways you can prevent cross contamination? ›- Mind Your Cutting Boards. Every kitchen should have at least two cutting boards (at least!) ...
- Wash Your Hands! The signs in restaurants say this for a reason. ...
- Watch the Juices. ...
- Don't Rinse Meats. ...
- Properly Rinse Produce. ...
- Clean All Surfaces.
Coating and Lining
Coatings or linings that are non-metallics should be used to combat metal corrosion. Electrical cladding, organic coatings, and electroplating can also be employed. By coating the anodes and cathodes with barrier coatings or by isolating them both, galvanic corrosion can be controlled.
How is cross contamination prevented in dispensing and production areas? ›
Use of bristles, brushes, fiber, shedding clothes to be avoided. The sequence of washing, cleaning and drying operation should be designed so as to prevent cross-contamination. Clean common equipment according to validated cleaning procedure during the production of different products.
What are 3 ways to reduce the risk of contamination in the lab? ›- Automate the process with lab automation.
- Wear proper protective equipment.
- Sterilize equipment.
- Check your water source.
- Clean surfaces regularly.
- Reduce the number of touches.
- Use an air filter and laminar flow hood.
- Stay organized.
Cross-contamination is the accidental transfer of contaminants into the food from a surface, object, or person. Four common sources of cross-contamination include clothing, utensils, food handlers, and pests.
What are the safety methods in pharmaceutical industry? ›- Quickly clean up all spills.
- Maintain a tidy work area.
- Frequently wash hands.
- Wear a laboratory coat.
- Never smoke inside the laboratory.
- Label containers correctly.
- Wear eye protection and other personal protective equipment (PPE)
- evaluating starting material purity.
- minimizing impurity levels in synthesis and manufacturing processes.
- identifying impurity structures.
- isolating and synthesizing impurities for qualification in toxicity studies.
- monitoring the stability of APIs and DPs to detect degradation products.
- Materials: The quality, handling and control of raw materials can also cause pharmaceutical contamination:
- Improper handling and storage.
- Poor labelling, sampling and testing of raw materials.
- Degradation of materials due to environmental conditions.
- Using materials that do not meet acceptance guidelines.
There are three main types of cross contamination: food-to-food, equipment-to-food, and people-to-food. In each type, bacteria are transferred from a contaminated source to uncontaminated food.
How can we prevent or reduce contamination? ›...
You can also buy pre-aliquoted reagents.
- Use sterile labware. ...
- Use filter tips and change them often. ...
- Check your cells often. ...
- Bleach your contaminated samples. ...
- Use good labeling practice.
Hand washing is the single most effective way to break disease transmission from one patient to another.