Fact-checked by Grok 2 weeks ago

Sterility assurance level

Sterility assurance level (SAL) is defined as the probability of a single viable occurring on an item after it has been subjected to a sterilization process. This metric quantifies the effectiveness of sterilization in achieving sterility, expressed mathematically as $10^{-n}, where n represents the logarithm of the reduction in microbial population. In practice, SAL values are selected based on the intended use of the sterilized product, with $10^{-6} (a 1 in 1,000,000 probability of non-sterility) serving as the standard for medical devices that contact sterile body tissues or fluids, such as implants and surgical instruments. Lower assurance levels, like $10^{-3} (1 in 1,000), may apply to devices intended for contact with intact skin or mucous membranes where lower risk is acceptable. These levels ensure by minimizing infection risks while balancing process feasibility and product integrity. SAL is validated through rigorous testing and monitoring rather than routine , relying on assessments, microbial resistance data, and process parameters like time, , or dose. standards, including ISO 11139:2018 for and ISO 11137 for sterilization, guide the establishment and verification of SAL, emphasizing statistical confidence over absolute sterility. Alternative SALs (e.g., $10^{-4} or $10^{-5}) are permitted under standards like ANSI/ ST67:2019 for cases where achieving $10^{-6} is impractical, such as in , provided equivalent safety is demonstrated.

Fundamentals

Definition

The sterility assurance level (SAL) is defined as the probability of a single viable microorganism occurring on an item after it has been subjected to a sterilization process, typically expressed as $10^{-n}, where n is the desired level of assurance (corresponding to the number of logs of microbial reduction achieved). This probabilistic measure quantifies the effectiveness of sterilization by estimating the likelihood of microbial survival, acknowledging that log reduction corresponds to a logarithmic decrease in viable microorganisms. Absolute sterility, defined as the complete absence of all viable microorganisms, cannot be practically proven without destructively testing every unit, making a key tool for providing probabilistic assurance of sterility instead. In practice, offers a statistically validated level of in the sterilization outcome, balancing the impossibility of guaranteeing zero survivors with the need for reliable microbial control. Common SAL values include $10^{-3} for applications with lower risk, such as devices intended for intact skin contact, and $10^{-6} for high-risk medical products like those used in invasive procedures, corresponding to a probability of one nonsterile unit per million. These targets ensure patient safety by minimizing the chance of contamination while accounting for process variability. The concept of SAL originated in microbiology as a method to quantify the survival of microorganisms following exposure to sterilizing agents, providing a standardized framework for evaluating process efficacy across various applications.

Historical Context

The development of sterility assurance concepts traces back to late 19th-century advancements in heat-based sterilization techniques. In 1879, French microbiologist Charles Chamberland, a collaborator of , invented the —a pressurized chamber that enabled efficient destruction of microorganisms in medical instruments and supplies. This innovation marked a shift from rudimentary methods to more reliable processes, establishing foundational principles for validating sterilization efficacy that would evolve into probabilistic frameworks. The mid-20th century saw further progress influenced by emerging technologies, particularly the adoption of nuclear for sterilization in the . Commercial gamma using sources began in the late , offering a non-thermal method suitable for heat-sensitive materials and introducing log-reduction models to quantify microbial inactivation based on dose. This approach, initially applied to pharmaceuticals and later to medical devices, highlighted the limitations of direct sterility testing and paved the way for probability-based assurance metrics. Post-World War II, the rapid expansion of disposable manufacturing in the 1950s and created urgent demands for standardized, quantifiable sterility validation to ensure amid increasing production scales. Early guidelines from pharmacopeias in the , including the newly established (1964) and updates to the (USP), began prioritizing probabilistic assurance over sole reliance on direct sterility testing, with authorities like mandating an SAL of 10^{-6} for critical devices. The Sterility Assurance Level (SAL) was formally defined and integrated into regulatory practices during the 1970s and 1980s, as organizations such as the Association for the Advancement of Medical Instrumentation (AAMI) developed consensus standards to specify acceptable non-sterility probabilities for various applications.

Mathematical Foundations

Probability and Log Reduction

The sterility assurance level (SAL) is fundamentally modeled using survivor curves, which depict the decline in microbial over time or exposure to a sterilizing under constant conditions. These curves assume , where the rate of microbial inactivation is proportional to the number of surviving organisms, leading to an pattern. In this model, the probability of P for a single after exposure time t is given by P = 10^{-t/D}, where D is the decimal reduction time (D-value), representing the time required to reduce the by one logarithmic (90%) at a specified condition. This arises because each has an independent probability of inactivation under sterilization stress, such as or , resulting in a semilogarithmic plot of survivors versus time that yields a straight line. The model implies that complete sterility is theoretically unattainable but can be approached to arbitrarily low probabilities by extending exposure. Log reduction quantifies the extent of microbial kill as the number of decimal (logarithmic base-10) decreases in ; for instance, a 6-log reduction means the initial population is reduced by a factor of $10^6. In the context of , a 6-log reduction achieves an SAL of $10^{-6} when the initial is 1 per . While measures the absolute kill extent independent of starting , represents the probability of survival for a , which incorporates the initial ; thus, higher bioburden requires greater to attain the same .

Key Parameters

The key parameters in modeling sterility assurance level (SAL) include , D-value, Z-value, and the choice between and bioburden-based sterilization approaches, which collectively inform the microbial inactivation process without direct computation of SAL. refers to the total number of microorganisms associated with a product, component, or prior to sterilization, typically measured in colony-forming units (CFU) per unit area, volume, or item. In validation, worst-case levels are assumed to ensure process robustness, often set as maximum acceptable limits (e.g., 10-100 CFU per unit) based on routine and environmental controls to minimize initial microbial load. This parameter is critical as higher requires greater inactivation to achieve target SAL. The D-value, or decimal reduction value, is the time or radiation dose required at specified conditions to reduce the population of a specific microorganism by 90% (one logarithmic cycle). It varies significantly by microorganism species (e.g., bacterial spores are more resistant than vegetative cells), sterilization method (e.g., moist heat at 121°C versus gamma radiation), and product factors like moisture content or packaging. For biological indicators used in validation, D-values are standardized, such as D121 ≥ 1.5 minutes for Geobacillus stearothermophilus in steam sterilization. The Z-value quantifies thermal resistance and is defined as the temperature change needed to alter the D-value by a factor of 10. It is primarily applied in sterilization , such as moist , where a typical Z-value for spores is around 10°C, meaning a 10°C increase reduces the D-value by one log cycle, allowing equivalent at lower temperatures over longer times. This enables adjustments for varying load temperatures or non-isothermal conditions. In sterilization validation, two primary approaches utilize these parameters: the method and the -based method. The approach assumes a high initial (e.g., 106 CFU) and applies a fixed, conservative (typically 12 logs) using resistant biological indicators, independent of routine measurement, to ensure SAL regardless of actual load. In contrast, the -based approach measures actual pre-sterilization microbial load and resistance (via D-values), tailoring the process to achieve the required for the verified , often requiring ongoing monitoring but allowing for more efficient cycles. These parameters feed into calculations as the primary output metric for SAL.

Validation Methods

Establishing SAL

Establishing the sterility assurance level () in sterilization processes involves rigorous validation methods to ensure that the probability of a non-sterile unit is acceptably low, typically through controlled challenges to the sterilization system. Biological indicators (BIs), which consist of highly resistant microbial spores, are commonly employed to simulate worst-case microbial loads and verify process efficacy. For steam sterilization, spores are frequently used due to their high resistance to moist heat. These BIs are placed in the most challenging locations within the load, such as areas with potential cold spots, to confirm that the process achieves the targeted microbial inactivation. In validation approaches, a half-cycle —operating at half the exposure time or of the full —is used to demonstrate at least a 6-log reduction in BI populations, providing a conservative margin that ensures the full cycle exceeds the required SAL. This method is particularly suitable for where the natural on products is variable or difficult to control, as it relies on the of highly resistant organisms rather than precise data. Full-cycle validation may follow to confirm overall performance, but the half-cycle serves as the primary evidence of lethality. The bioburden method, in contrast, bases validation on the actual microbial load present on the product prior to sterilization, allowing for tailored process parameters that avoid unnecessary overprocessing. This approach involves characterizing the bioburden through sampling multiple product lots to identify spore-forming organisms and their resistance, followed by dose auditing where exposed samples are tested for sterility to verify the SAL. It requires strict control of manufacturing cleanliness to maintain consistent bioburden levels, enabling more efficient cycles compared to overkill methods. As of the ISO 11137-1:2025 revision (published April 2025), bioburden monitoring requirements have been updated, removing mandatory monthly testing for low-bioburden products and adding guidance on demonstrating stability in bioburden numbers and types. For sterilization, the VDmax establishes a maximum allowable dose by first verifying limits on the product and then applying a dose to confirm partial inactivation, ensuring the full dose achieves the desired without excessive exposure. This involves irradiating representative product units with the verification dose, followed by sterility testing of at least 10 units to demonstrate at a preliminary level. The ISO 11137-1:2025 update introduces more flexible dose audit frequencies and simplified language, while maintaining the core VDmax . Ongoing monitoring supports the method's continued validity. The selection of for these validations often considers their D-value to match the conditions.

Calculation Formulas

The sterility assurance level () is calculated based on the initial microbial and the applied during the sterilization process. The general for SAL in a single unit is given by: \text{SAL} = N_0 \times 10^{-\frac{F}{D}} where N_0 is the initial (number of microorganisms per unit before sterilization), F is the factor (such as exposure time for heat or for ), and D is the D-value (the required to achieve a 1-log in the microbial ). This equation assumes a log-linear survivor curve, where microbial inactivation follows first-order kinetics. To determine the required lethality for a target SAL, the formula is rearranged to calculate the necessary log reduction, defined as: \text{Log reduction} = \log_{10}\left(\frac{N_0}{\text{SAL}}\right) Thus, the required lethality F becomes: F = D \times \left( \log_{10} N_0 - \log_{10} \text{SAL} \right) This approach ensures the process achieves the desired probability of nonsterility by accounting for the initial contamination level and the resistance of the most resistant microorganism. For radiation sterilization, an example illustrates the computation. Consider an initial of N_0 = 10^2 colony-forming units (CFU) per unit, a target of $10^{-6}, and a D-value of 1 kGy for the most resistant . The required is \log_{10}(10^2) - \log_{10}(10^{-6}) = 2 - (-6) = 8. Therefore, the minimum F = 1 \times 8 = 8 kGy. This dose would reduce the microbial population to the target . When considering an entire batch, the probability of nonsterility for the batch (i.e., at least one nonsterile unit) must account for the number of units processed. The formula is: \text{SAL}_{\text{batch}} = 1 - (1 - \text{SAL}_{\text{unit}})^U where \text{SAL}_{\text{unit}} is the SAL per unit and U is the number of units in the batch. For low values of \text{SAL}_{\text{unit}} (e.g., $10^{-6}), this approximates to \text{SAL}_{\text{batch}} \approx U \times \text{SAL}_{\text{unit}}, highlighting the increased risk in large batches if per-unit SAL is not sufficiently stringent.

Standards and Regulations

ISO Standards

The ISO 11137 series establishes requirements for the development, validation, and routine control of radiation sterilization processes for products, including gamma, , and methods. It specifies a sterility assurance level () of $10^{-6} as the standard for achieving sterility in products intended to be sterile, representing a probability of one viable surviving per million units processed. The series outlines dose-setting methods such as VDmax15, which verifies a maximum acceptable dose while substantiating an SAL of $10^{-6}, and bioburden-based approaches that account for the initial microbial load to determine the minimum sterilizing dose. ISO 11135:2014 details the requirements for (EtO) sterilization es, emphasizing validation through either the approach, which assumes a high initial and applies sufficient lethality to achieve the desired , or the approach, which uses product-specific microbial data. It defaults to an SAL of $10^{-6} for devices but requires demonstration of via biological indicators and sterility testing. The standard also addresses routine control, including to remove residuals, to ensure safety and compliance. ISO 17665:2024 focuses on , providing guidance for cycle development, validation, and monitoring to attain the targeted , typically $10^{-6}, through biological indicator () testing that confirms microbial inactivation. It describes and methods for validation, with emphasis on physical parameters like , time, and to ensure across loads. The supports both and biological release criteria for routine processing. The ISO 11138 series specifies requirements for biological indicators used in validating sterilization processes across various modalities, including production, labeling, and performance testing to ensure resistance characteristics suitable for demonstration. These indicators, often based on spores like for moist heat or for EtO and radiation, enable half-cycle and full-cycle validation to confirm process lethality. Recent updates in ISO 11137-1:2025, building on the 2006 edition, emphasize a risk-based approach to SAL selection, allowing levels higher than $10^{-6} (e.g., $10^{-3} or $10^{-4}) for low-risk items where patient safety is not compromised, provided a thorough risk assessment justifies the deviation. This flexibility integrates with broader quality management systems to balance efficacy and product integrity.

Regulatory Frameworks

In the United States, the (FDA) enforces (SAL) requirements under 21 CFR Part 820, the Quality System Regulation, which mandates validation of sterilization processes for sterile medical devices to ensure they meet specifications with a high degree of assurance. The FDA establishes 10^{-6} as the standard SAL benchmark for devices labeled as sterile, representing a probability of one viable in one million units. Guidance on sterilization process controls, originally issued in 1994 and updated in 2023, emphasizes monitoring parameters such as temperature, pressure, and exposure time to verify consistent SAL achievement. In the , the (MDR) (EU) 2017/745 requires SAL compliance for sterile devices through conformity assessment procedures outlined in Annex IX, particularly for high-risk Class III devices, where notified bodies conduct audits of systems and technical documentation to confirm sterility maintenance. These audits ensure that manufacturers demonstrate effective sterilization processes aligned with general safety and performance requirements in Annex I, focusing on preventing microbial during , , and . The (WHO) emphasizes in its prequalification programs for and medical devices, recommending validation to achieve an of 10^{-6} for terminally sterilized products while imposing limits to control microbial load prior to sterilization. Similarly, Japan's () requires validation in approval processes for sterile pharmaceuticals and devices, defining as the probability of a viable surviving sterilization and mandating documentation of process efficacy under the Act on Securing Quality, Efficacy and Safety of Products Including Pharmaceuticals and Medical Devices. Global harmonization efforts, led by the Global Harmonization Task Force (GHTF) until 2011 and continued by the International Medical Device Regulators Forum (IMDRF), promote consistent SAL application across regions through guidelines on essential principles of safety and performance, including documentation of attained SAL in technical files since the 2010s. Routine regulatory inspections worldwide focus on verifying through review of process records, such as F_0 values for moist cycles or dose mappings, to confirm ongoing and detect deviations in sterilization parameters. These frameworks generally ISO standards for foundational guidance on SAL validation.

Applications

Medical Devices

Terminal sterilization is the preferred method for many medical devices, such as implants and syringes, as it allows the device to be sterilized after packaging, achieving a sterility assurance level (SAL) of $10^{-6}, which represents a probability of one non-sterile unit per million. This approach minimizes risks during handling and ensures the device remains sterile until use. Radiation sterilization, particularly using gamma rays or electron beams, is commonly employed for heat-sensitive medical devices due to its penetrating power and lack of chemical residues. The SAL of $10^{-6} is validated through absorbed dose mapping, which identifies minimum and maximum dose locations within the product to confirm uniform lethality against microorganisms. Ethylene oxide (EtO) sterilization is suitable for devices with complex geometries that require deep penetration, offering an SAL of $10^{-6} while accommodating temperature-sensitive materials. However, EtO residuals must be rigorously monitored and controlled to levels below established safety thresholds, ensuring both sterility and . For devices that cannot undergo sterilization, aseptic processes are used, targeting an SAL ranging from $10^{-3} to $10^{-6} through stringent environmental controls, such as operations and microbial monitoring, rather than direct sterilization of the final product. This method relies on preventing during to maintain overall sterility assurance. A representative case is cardiovascular implants, which, due to their invasive placement in sterile body sites, mandate an SAL of $10^{-6} to mitigate risks, often achieved via or EtO methods.

Pharmaceuticals and Biologics

In the production of pharmaceuticals and biologics, terminal sterilization is the preferred method for achieving a high sterility assurance level (SAL) when the product can withstand the process, particularly for injectables such as aqueous solutions. This involves sterilizing the final packaged product, often through autoclaving at 121°C for 15 minutes, to attain an SAL of 10^{-6}, meaning a probability of no more than one viable in one million units. Sterile , typically using 0.2 μm filters, serves as a critical pre-step to reduce prior to filling and terminal sterilization, enhancing overall process reliability. This approach aligns with pharmacopeial standards, where <71> and EP 2.6.1 outline requirements for sterility testing of terminally sterilized parenterals, implicitly supporting an SAL of 10^{-6} through validation of the sterilization cycle. For heat-sensitive biologics like , aseptic processing is employed to maintain product integrity while ensuring sterility, relying on controlled environments rather than terminal sterilization. This method achieves an SAL through stringent cleanroom controls, such as ISO 5 classification in critical zones to minimize airborne contamination, combined with media fills (aseptic process simulations) that challenge the to detect potential failures. Parametric release, based on validated process parameters and real-time monitoring, targets a contamination probability of less than 10^{-3}, verified by zero growth in media fill runs representing the batch scale. USP <71> and EP 2.6.1 further support this by requiring sterility testing to confirm the absence of viable microorganisms in aseptically processed products. Validation of SAL in lyophilization and filling lines for biologics emphasizes barrier integrity and to prevent microbial ingress during the aseptic fill-finish operations. Restricted access barrier systems (RABS) or isolators maintain ISO 5 conditions, with integrity tested via pressure holds or leak rate measurements, while continuous viable and non-viable particle monitoring ensures compliance with limits of fewer than 1 per cubic meter in Grade A zones. These elements collectively contribute to the targeted SAL by simulating worst-case scenarios in media fills that include hold times for lyophilization cycles. An illustrative example is the production of mRNA vaccines, where aseptic fill-finish into vials or pre-filled syringes relies on to assure SAL through validated media fills and environmental controls, enabling rapid scale-up without compromising sterility. In terminal processes, values, such as a 6-log reduction for resistant spores, underpin the SAL calculation by quantifying microbial inactivation.

Monitoring and Limitations

Process Monitoring

Process monitoring in sterilization operations involves ongoing and measures to ensure that the validated consistently achieves the required sterility assurance level (SAL), typically 10^{-6}, after initial validation. These controls rely on a combination of physical, chemical, and biological indicators to confirm that critical parameters are met in each cycle, thereby maintaining product sterility without relying solely on end-product sterility testing. Physical monitors track essential process parameters to verify the delivery of lethality. For heat-based methods like or dry heat, sensors continuously record , , and exposure time to ensure conditions exceed those required for microbial inactivation. In (EtO) sterilization, humidity and gas concentration are similarly monitored alongside time and . For radiation sterilization, dosimeters measure to confirm sufficient energy delivery for the targeted . These monitors provide , enabling immediate detection of deviations and cycle aborts if necessary. Chemical indicators, particularly Classes 4 through 6, serve as adjunct tools for cycle verification by responding to specific sterilization conditions equivalent to achievement. Class 4 indicators react to at least two parameters, such as time and , changing color to confirm in multi-parameter cycles. Class 5 integrating indicators mimic biological responses by accounting for multiple variables including sterilant penetration, providing higher assurance of lethality. Class 6 emulating indicators are calibrated for exact cycle parameters, offering the most precise verification of -equivalent conditions like a 6-log reduction in resistant spores. These indicators are placed inside and outside product packs to validate process attainment. Biological indicators (BIs) are employed routinely in qualified loads to directly assess microbial kill efficacy. Containing highly resistant spores such as Geobacillus stearothermophilus for steam or Bacillus atrophaeus for EtO, BIs are incubated post-cycle to confirm no growth, verifying periodic log reductions of 1-3 logs in routine monitoring or higher in validation contexts. They are used at least weekly for routine steam sterilizers, or daily for high-volume operations, and mandatorily for loads containing implants, with negative results required before release. Parametric release enables batch approval based on confirmed process parameters rather than sterility testing, enhancing efficiency while upholding . This approach, applicable to terminally sterilized products like those using moist heat, relies on validated controls of critical variables—such as temperature-time profiles—to demonstrate a non-sterile unit probability below 10^{-6}, supported by load monitors like or dosimeters. It bypasses traditional testing when process robustness is proven, as outlined in regulatory guidance. Data trending supports long-term process oversight through bioburden surveillance and cycle efficacy reviews. levels on pre-sterilization products are routinely sampled and trended to detect shifts, informing adjustments to maintain low microbial loads and compliance. Cycle efficacy is reviewed quarterly via analysis of monitoring records, including results and parameter logs, to identify trends in performance and ensure ongoing validation.

Challenges and Constraints

The sterility assurance level (SAL) is inherently probabilistic, defining the probability of a single viable surviving on an item after sterilization, such as SAL 10^{-6} indicating approximately one nonsterile unit per million processed. This framework cannot guarantee absolute sterility, as it relies on statistical modeling rather than empirical proof of zero across an entire batch. Practical of such low probabilities is impossible due to experimental constraints, where rates below 10^{-2} cannot be directly measured and must be extrapolated, introducing uncertainties. Bioburden variability poses a significant challenge, as unpredictable initial microbial loads on products can compromise the targeted if not conservatively estimated during validation. Real-world often deviates from controlled test conditions, where artificially elevated levels are used, potentially leading to under- or over-estimation of sterilization efficacy and undermining process reliability. Sterility testing under USP <71> is limited by its small sample sizes, typically involving only 10-20 units per batch, which are statistically insufficient to confirm an SAL of 10^{-6} and increase the risk of false negatives. These tests detect probabilistically but cannot reliably extrapolate to the full production scale, as microbial distribution is uneven and low-level may evade sampling. Method-specific issues further constrain SAL achievement; for instance, (EtO) sterilization requires rigorous aeration to reduce toxic residuals, which can persist and pose risks if not adequately controlled, complicating validation. Similarly, sterilization may induce product degradation, such as breakdown of polymers or bioactive components like growth factors, potentially affecting device integrity and functionality without compromising microbial kill. As of 2025, ongoing challenges include regulatory pressures on EtO facilities, stemming from EPA emissions rules finalized in 2024 requiring up to 92% reductions (with compliance delays extended to 2028), and continued supply constraints for sources in , driven by production limitations and increasing demand that exceeds domestic supply. These issues have prompted exploration of alternatives and delayed some process qualifications, underscoring vulnerabilities in sterilization infrastructure.

References

  1. [1]
  2. [2]
    Sterility Assurance Levels (SALs): Irradiation - steris ast
    An SAL is normally expressed as 10-n with historically, a 10-3 or a 10-6 value being used most frequently for sterilization.
  3. [3]
    Sterility Assurance Level - an overview | ScienceDirect Topics
    Sterility Assurance Level (SAL) is defined as a measure of the effectiveness of a sterilization process, indicating the probability of a sterile product ...
  4. [4]
    Sterility Assurance Levels and Aseptic Processing - AAMI Array
    Mar 9, 2023 · SAL generally is defined as “probability of a single viable microorganism occurring on an item after sterilization” (ISO 11139:2018). It is ...
  5. [5]
    [PDF] STERILITY - Pacific BioLabs
    Sterility is defined as the state of being free from viable microorganisms . A sterility assurance level (SAL) is defined as the probability of an item ...
  6. [6]
    Sterilization of Objects, Products, and Packaging Surfaces and Their ...
    Mar 2, 2021 · To call a product “sterile” does not mean absolute sterility as it is impossible to prove the absence of all microorganisms.
  7. [7]
    <1211> STERILIZATION AND STERILITY ASSURANCE OF ...
    Absolute sterility cannot be practically demonstrated without complete destruction of every finished article. The sterility of a lot purported to be sterile ...Missing: impossible | Show results with:impossible
  8. [8]
    The limits of sterility assurance - PMC - NIH
    A sterility assurance level (SAL) of 10–6 is currently required for sterilization procedures, i.e., a probability of not more than one viable microorganism in ...
  9. [9]
    Which parameters must be validated during a steam sterilization ...
    Dec 7, 2021 · For example, an SAL of ≤ 10-6 means that there is a probability of one in a million devices being non-sterile after exposure to a sterilization ...
  10. [10]
    Sterility Assurance The Fundamentals - Pharmaceutical Online
    Oct 8, 2024 · Sterility assurance is a critical aspect of the production of pharmaceuticals, biopharmaceuticals, medical devices, and other sterile products.
  11. [11]
  12. [12]
    Autoclave Machine: Uses, Guidelines & Cost | Knowledge Center
    Mar 24, 2022 · It wasn't until 1879 that the French microbiologist Charles Chamberland created a new version called the autoclave to be used in medical ...
  13. [13]
    Radiation Sterilization - Stanford University
    Mar 13, 2018 · Commercial radiation sterilization has existed since the late 1950s and has grown tremendously in popularity over the last 60 years.
  14. [14]
    [PDF] Trends in Radiation Sterilization of Health Care Products
    In Eastern Europe, the history of radiation processing in Croatia dates back to the 1950s with the foundation of the Ruder Bokovi Institute as a nuclear ...
  15. [15]
    History - European Directorate for the Quality of Medicines ...
    The Council Decision of 16 June 1994 (94/358/EC) accepts, on behalf of the European Community, the Convention on the elaboration of a European Pharmacopoeia.
  16. [16]
    USP <1211>: The Compendial Informational Chapter on Sterility ...
    The original chapter on sterilization appeared in USP IX, which became official in September 1916 (2). Although this chapter was sparse on technical detail, it ...
  17. [17]
    [PDF] ANSI/AAMI ST67:2003 Sterilization of healthcare products
    Also in the 1960s, the Swedish public health authorities required a stated SAL of 10–6 (or an SAL providing a greater assurance of sterility) for medical ...<|separator|>
  18. [18]
    Microbial Inactivation Kinetics and Heat Disinfection
    May 9, 2021 · THE SEMILOGARITHMIC SURVIVOR CURVE MODEL. As can be seen in ... STERILITY ASSURANCE LEVEL. Thus far, we have presented an analysis of ...
  19. [19]
    Understanding Overkill Sterilization: An End to the Confusion
    May 1, 2007 · ... curve is logarithmically linear. This is a reasonable assumption because microbial death follows essentially first-order kinetics. Figure 3.
  20. [20]
    [PDF] This guidance was written prior to the February 27, 1997 ... - FDA
    Sterility Assurance Level (SAL): A value indicating the probability of a survivor after a sterilization process. For example, an SAL of is the probability ...
  21. [21]
    [PDF] guideline-sterilisation-medicinal-product-active-substance-excipient ...
    Mar 6, 2019 · The SAL should always be established in relation to a D-value that is higher than that of the normal bioburden at routine production. Time and ...
  22. [22]
    [PDF] Guidance for Industry - FDA
    purpose of achieving a predetermined sterility assurance level (SAL) of usually less than 10-6. (i.e., a probability of a nonsterile unit of greater than one ...
  23. [23]
    [PDF] Question-based Review (QbR) for Sterility Assurance of Terminally ...
    Sep 29, 2011 · Like the Chemistry QbR-QOS, we believe the QBR approach can be used to address sterility assurance aspects of an application, ...Missing: seminal | Show results with:seminal<|control11|><|separator|>
  24. [24]
    [PDF] Validation and Management of Heat Sterilization | DCVMN
    A Dvalue of 2.0 means more resistant while a Dvalue of 1min means less resistant. ▫ What is a Z value? ▫ Refers to the temperature change required to produce a ...<|control11|><|separator|>
  25. [25]
  26. [26]
    [PDF] 3 Methods to Establish the Sterilization Dose of a Medical Device
    Oct 1, 2021 · In the medical device industry, sterilization processing is performed to obtain a Sterility Assurance Level. (SAL) as required by regulatory ...
  27. [27]
    Sterility Assurance Level (SAL) and Log Reduction (D Value) in ...
    Dec 19, 2013 · It is 90% reduction in a specified microbial population. The time (in minutes) required to achieve 1 log reduction (at a specified temperature) ...<|control11|><|separator|>
  28. [28]
    Pharmacopeial sterility test: The statistical limitations of sampling
    Aug 2, 2023 · q = 1. From this it follows that: The probability of rejection = 1 - (1 – p)n. For example, the pharmacopoeia test for sterility passes batches ...
  29. [29]
  30. [30]
    Recognized Consensus Standards: Medical Devices - FDA
    Dec 19, 2022 · ISO 11137-2:2013 also specifies methods of sterilization dose audit used to demonstrate the continued effectiveness of the sterilization dose.
  31. [31]
  32. [32]
    ISO 11135: Overview of Medical Device Sterilization [Guide]
    Nov 16, 2021 · ISO 11135 is the international standard that details the development and validation of a process for sterilizing medical devices using ethylene oxide.
  33. [33]
    Overview of an Ethylene Oxide Validation | TechTip | STERIS AST
    This TechTip provides a step-by-step overview of an ethylene oxide validation process that complies with the guidelines established in 11135 using a Half ...
  34. [34]
  35. [35]
    ISO 11138-1:2017 - Sterilization of health care products
    In stock 2–5 day deliveryISO 11138-1:2017 specifies general requirements for production, labelling, test methods and performance characteristics of biological indicators.
  36. [36]
  37. [37]
    Recognized Consensus Standards: Medical Devices - FDA
    ISO 11137-1 specifies requirements for radiation sterilization of medical devices, using 60Co/137Cs, electron, or X-ray beams. It does not cover virus ...Missing: SAL | Show results with:SAL
  38. [38]
    [PDF] Submission and Review of Sterility Information in Premarket ... - FDA
    Jan 21, 2016 · The sponsor should state the sterility assurance level (SAL) of 10-6 for devices labeled as sterile unless the device is intended only for ...
  39. [39]
    Sterilization Process Controls - FDA
    Mar 28, 2023 · For sterilization processes, the primary device specification is the desired Sterility Assurance Level (SAL). Other specifications may include ...Missing: 21 CFR 820
  40. [40]
  41. [41]
    [PDF] Annex 6 WHO good manufacturing practices for sterile ...
    4.12 Components and most products should be prepared in at least a. Grade D environment to ensure low microbial bioburden and particulate counts prior to ...Missing: Z- | Show results with:Z-
  42. [42]
    [PDF] Guidance on the Manufacture of Sterile Pharmaceutical Products ...
    2.50 Sterility assurance level (SAL): Probability of viable microorganisms being present on a product unit after exposure to the proper sterilization process, ...
  43. [43]
    [PDF] ghtf-sg1-n011r20-essential-principles-safety-performance-medical ...
    Mar 5, 2007 · The purpose of such guidance is to harmonize ... sterility assurance level attained, standards applied, the sterilisation protocol developed.
  44. [44]
    Comparing Terminal Sterilization and Aseptic Processing of ...
    The process is validated to provide a Sterility Assurance Level (SAL) lower than 10-6, which means a probability of less than one unsterile product on a one ...
  45. [45]
    Process validation, sterilization by ionizing radiation - Canada.ca
    Nov 12, 2024 · Gamma radiation delivers a specified dose relatively slowly. Both electron beam generators and X-rays deliver the same dose much more quickly.
  46. [46]
    [PDF] Guidelines for the Development, Validation and Routine Control of ...
    Risk management is mentioned in ISO 11137 but is required by many quality management references (GMP, ISO 13485 [8.4] and ISO 22000 [8.5]). Because of the ...Missing: 11137-1:2015 | Show results with:11137-1:2015
  47. [47]
    Dose mapping for documentation of radiation sterilization
    Dose mapping is a part of two elements of the validation programme for the radiation sterilization of a medical device: Installation Qualification and ...
  48. [48]
    Advancing the Sustainable Use of Ethylene Oxide through Process ...
    EO sterilization of medical devices is validated in accordance with AAMI/ANSI/ISO 11135:2014 via a manner in which a sterility assurance level (SAL) of 10−6 is ...
  49. [49]
    Sterilization for Medical Devices - FDA
    The FDA announced, in May 2022, a Sterility Change Master File Pilot Program for sterilization changes to 510(k) cleared medical devices for sterilization ...Sterilization For Medical... · Fda's Actions To Advance... · 510(k) Eto Sterility Change...
  50. [50]
    ISO 10993-7 Primer: Testing for Ethylene Oxide Sterilization Residuals
    Apr 4, 2025 · The standard requires you test for ethylene oxide sterilization residues for both single-use and reusable devices that use EO sterilization.Missing: monitoring | Show results with:monitoring
  51. [51]
    ISO 13408-1:2023(en), Aseptic processing of health care products
    SAL is a mathematical extrapolation applicable only to a validated and controlled terminal sterilization process of known microbial lethality and which is ...
  52. [52]
    Sterilization Assurance & SAL selection for Medical Devices
    Critical devices (e.g., implants, surgical instruments) require an SAL of 10-6 due to direct contact with sterile body sites or blood. Semi-critical devices ( ...
  53. [53]
    [PDF] 71 STERILITY TESTS Change to read - US Pharmacopeia (USP)
    Nov 21, 2016 · The test for sterility is carried out under aseptic conditions. In order to achieve such conditions, the test environment has to be adapted ...Missing: 1960s | Show results with:1960s
  54. [54]
  55. [55]
    [PDF] 20220825_gmp-an1_en_0.pdf
    Aug 22, 2022 · Integrity testing of the product filling lines and filtration systems (as relevant). v. Duration of the batch or filling campaign. vi ...
  56. [56]
    Development of mRNA manufacturing for vaccines and therapeutics
    The fourth and final step in the downstream process is a 0.2 µm filtration and then closed bulk drug substance fill into sterile fill containers. It should ...Missing: SAL | Show results with:SAL
  57. [57]
    [PDF] Guideline for Disinfection and Sterilization in Healthcare Facilities ...
    In short, a SAL is an estimate of lethality of the entire sterilization process and is a conservative calculation. Dual SALs (e.g., 10-3 SAL for blood ...
  58. [58]
    Sterilizing Practices | Infection Control - CDC
    Nov 28, 2023 · Biological indicators are the only process indicators that directly monitor the lethality of a given sterilization process. Spores used to ...
  59. [59]
    [PDF] Guidance for Industry - FDA
    Parametric release is defined as a sterility assurance release program where demonstrated control of the sterilization process enables a firm to use defined ...
  60. [60]
    Container and Closure System Integrity Testing in Lieu of Sterility ...
    Feb 22, 2008 · Because of the limitations of sterility tests described below, sterility tests are not recommended as a component of a stability program for ...
  61. [61]
    [PDF] FDA Innovation Challenges: Identify Sterilization Alternatives and ...
    Aug 13, 2019 · Since microbial activation is expected to be exponential we express how thorough a cycle is through probability in what we call the sterility.Missing: probabilistic | Show results with:probabilistic
  62. [62]
    Risks of Using Sterilization by Gamma Radiation - NIH
    Jan 18, 2018 · Gamma rays induced degradation of growth factors which were crucially involved in osteoinduction (bone morphogenetic protein and transforming ...
  63. [63]
    Sterility Assurance Across-Sectors—New Paradigms and Tools - PMC
    Aug 9, 2021 · ... sterility assurance level, SAL. The term is defined by ISO in their vocabulary document: sterility assurance level, SAL—ISO 11139.