HPE Drug stability: What do we need to know? | Page 30

biological peptide/protein drugs are beyond the scope of this article, but suffice to say, that establishing the conservation of the active site and the secondary and tertiary structure of biologicals requires an array of sophisticated and specialised techniques including LC-MS, circular dichroism, electrophoresis and ELISA immunoassays. Results The results for chemical and physical testing at each time-point must be clearly reported and any gaps in the data or outlier points identified and justified. It is also helpful if stability reports include methodology validation data and details of any statistical methods used. within the manufacturer’s expiry date and must be produced according to the principles of EU GMP. The initial drug concentration must be within 10% of the target value and the batch numbers and expiry dates of all drugs, containers and diluents must be recorded. Storage incubators and refrigerators used for the study must be subject to continual temperature monitoring and any variances must be recorded. Similarly, any deviation in sample times from the study protocol should have been recorded and justified. All analytical methods must have been fully validated. For HPLC, the validation will include linearity of analytical response over the concentration range expected for diluted samples, inter- and intra-day precision, and the assay must be shown to be ‘stability-indicating’. This means that the assay will be sensitive to any drug degradation and can separate degradation product peaks from the main drug analyte peak that is to be quantified. This aspect is often validated using forced degradation conditions to break down the drug deliberately (acid, alkali, oxidising agents, light) and ensure that degradation products are well resolved chromatographically from the drug analyte. Modern HPLC systems record the peak-purity of the analyte peak to ensure this condition is met. pH meters and sub-visual particulate measuring systems should have been calibrated according to Pharmacopoeia recommendations, and visual inspections should follow Pharmacopoeial principles using light and dark backgrounds. The methodologies for stability studies on 30 | 2019 | hospitalpharmacyeurope.com References 1 Manufacturers Summary of Product Characteristics. www. emc.org.uk (accessed August 2019) 2 Stabilis stability data base. www.stabilis.org (accessed August 2019). 3 Bardin C et al. Guidelines for practical stability studies of anticancer drugs: A European Consensus Conference. Ann Pharm Fr 2011;69(4):221–31. 4 NHS Pharmaceutical Quality Control Committee. A Standard Protocol for Deriving and Assessment of Stability Part 1: Aseptic Preparations (Small Molecules), Edition 4, April 2017 (yellow cover). 5 NHS Pharmaceutical Quality Control Committee. A Standard Protocol for Deriving and Assessment of Stability Part 2: Aseptic Preparations (Biopharmaceuticals), Edition 3, April 2017 (yellow cover). 6 Kaestner S, Sewell G. A sequential temperature cycling study for the investigation of carboplatin infusion stability to facilitate dose-banding. J Oncol Pharm Pract 2007;13(2):119–26. Interpretation Clear limits or acceptance criteria for each parameter measured (for example, drug assay, infusion pH) should have been clearly defined before the study commenced. Any trends need to be accounted for and the point at which infusion stability falls outside the pre-determined acceptance criteria must be clearly identified. If a single container falls outside the acceptance criteria for a particular test, this cannot be ignored and should have been investigated. When applying stability data to assign a shelf- life, it is crucial that the conditions used in the study match those to be used in practice (see appropriateness, above). It might be acceptable to interpolate stability study data, for example applying the study finding to a different concentration range that fits within the range used in the study. However, extrapolation of stability data to, for example, higher drug concentrations, different container materials or different storage temperatures must never be attempted without consultation with a recognised expert and a document rationale for the variance. The acceptance criteria used in stability studies is a source of constant debate. For example, should the active drug concentration remain within ±5% of the original concentration, or is ±10% acceptable? In practice this depends on the reproducibility of the drug assay, the toxicity of the degradation products and the therapeutic index of the drug. For cytotoxic drugs, where the identity and pharmacology of most degradation products is unknown, 5% limits are normally appropriate. Conclusions Aseptic infusions and injections are usually required for very sick patients, and any significant instability of the infusion or active components may compromise treatment efficacy and could even cause patient harm if decomposition products are toxic. Stability studies reported in the literature continue to become more complex and there is a critical requirement to ensure that infusions stored or used over extended periods remain effective and safe. This brief article summarises the clinical and pharmaceutical need for shelf-life extension, pharmacist responsibilities and practical guidance on how to assess and interpret stability literature when assigning product shelf-lives. It is important to recognise, in addition to any chemical and physical stability issues, that infusions and injections must remain sterile over the assigned shelf-life. Evidence that microorganisms are excluded and integrity of containers and closures is maintained over the duration of the shelf-life must be included in the assignment of product shelf-lives.