Interview with MPI Research - Cornerstones for Successful Bioanalysis of Macromolecules

What should a company expect from their CRO when outsourcing macromolecule bioanalysis?

When a company outsources macromolecule bioanalysis, the expectation of their CRO largely depends on the company’s setting. If the outsourcing request is from a virtual company without laboratory space or hands-on expertise in bioanalytical development, the CRO is expected to provide both consulting and mentoring with suggestions and guidance on study design, critical reagent procurement or creation, bioanalytical method development, and spearheading the progress towards method validation and sample analysis. It is crucial that the CRO provides clear, concise updates on progress through timely communication. Alternatively, the sponsor may already have the bioanalytical methods developed and validated and needs a cost-effective method transfer for their preclinical or clinical programs. It is important to understand that not all “validated methods” can be validated, due to a myriad of reasons. Effective communication should be initiated with cost savings in mind, and without compromising method integrity or regulatory compliance. Often, an abbreviated method assessment/development will successfully bring it to validation. In cases of big pharmaceutical/biotechnology companies that have developed a vast amount of knowledge of the assay, they may want the CRO to act as a seamless extension of their own laboratory. This can be challenging, since the CRO maintains and trains on its own standard operating procedures (SOPs), which could vary in subtle interpretation. Mutual expectations of how a process will be implemented are paramount for a fruitful collaboration.

How does life cycle management of critical reagents factor in?

Critical reagents for ligand binding assays (LBA) are monoclonal or polyclonal antibodies, receptors, target ligands, or therapeutic proteins, used for capture and detection. They directly impact the sensitivity, specificity, and robustness of bioanalytical methods that either support pharmacokinetic (PK) measurements or determine an anti-drug antibody (ADA) response. In instances where critical reagents are not commercially available, the sponsors should initiate the generation of such reagents. Alternatively, the CRO may guide this process either by conducting the work internally (especially for CROs with animal facilities), or by subcontracting the work to companies that specialize in generating monoclonal and polyclonal antibodies (or other types of recombinant proteins). It is worthwhile to consider both the time required for the completion of the reagent creation and its impact on the bioanalytical work timeline. Monitoring the quality control of the reagents over the lifetime of the assay is paramount to the success of preclinical and clinical programs. Reagent lotto- lot differences may lead to significant variability in assay performance, which in turn may require method modification to return assay performance to established acceptable limits. In preclinical settings, where the life span of the assay is short and predictable, securing sufficient quantity of the reagents can be established through efficient communication and relationship building with reagent vendors. In prolonged and large scale clinical settings, implementing more stringent physicochemical and biophysical characterization methods for critical reagents is essential to ensuring the integrity of the bioanalytical method and data quality.

What are the important factors regarding assay formats and platforms?

The selection of assay formats and platforms is influenced by many factors. For instance, preclinical assay formats are largely dependent on the availability of critical reagents, while dose and sensitivity may dictate the platforms used. Conversely, clinical programs are impacted by specific human protein-recognizing reagents capable of selectively detecting human therapeutics. Additionally, concerns about method transferability across different facilities may also dictate assay platform.

How does analyst technique relate to assay development and validation?

The course of assay development, validation, and sample analysis requires well-trained bioanalytical scientists. Troubleshooting and out-of-specification investigations are based on the confidence of solid bioanalytical techniques. If assay anomalies are discovered during any of the analytical stages, misinterpretation of their root cause, due to uncertainty of the analyst's technique, will significantly impact working progress, efficiency, study cost, and timeline management. In addition to individual training, harmonizing the techniques and streamlining bioanalytical processes across laboratories are critical measures for success. Furthermore, the use of a dependable liquid handling automation system is recommended for a 21st century laboratory, and is expected to dramatically reduce human error while increasing productivity.

Should I be concerned with Out-Of- Specification (OOS) investigations?

Out-of-specification (OOS) investigations may be conducted during validation, sample analysis, and incurred sample reanalysis (ISR). The laboratory investigation process should be controlled and driven by SOPs, which may be established according to the FDA guidance for industry “Investigation Out-of-Specification (OOS) Test Results for Pharmaceutical Production.” While such processes may vary among CROs, they should all be driven by the same final goal: to identify a root cause for the anomalous event. The investigation SOP should outline three general procedural areas: assessment of root cause, impact on study interpretation, and corrective actions for moving forward.

What should I expect from my CRO’s final report?

Both FDA and EMA (European Medicine Agency) guidelines outline the requirements of validation as well as bioanalytical reports. Validation (or sample analysis) reports should include a summary of the assay procedures, details of the immunoassay method, information about the stock standard (certificate of analysis, stability, and storage conditions), descriptive information regarding the matrix, preparation of calibration standards and quality control samples, and the run acceptance criteria. Data analyses are displayed in tabular format. The report should include all of the analytical runs, their dates, pass or fail status, and the reason for failure, if failed. Additionally, both PK and ADA immunoassay validation reports must include unexpected results obtained during validation, and the findings of any OOS investigations. For sample analysis, the sample tracking information should refer to study sample results. Re-assay values must be specifically identified with a justifiable reason. All reports must document deviations from the method, SOPs or protocol. With this level of required report detail, the use of a Laboratory Information Management Systems (e.g., Thermo Watson LIMS) for sample tracking, data analysis, and generation of report tables is highly recommended.

References

  1. US Department of Health and Human Services. US FDA, Center for Drug Evaluation and Research. Guideline for industry: bioanalytical method validation. Rockville, MD, USA (2001).
  2. European Medicine Agency. Guideline on bioanalytical method validation. Committee for Medicinal Products for Human use (CHMP), London, UK (2011).
  3. Ray, C.A. and Ahene, A.B. "Ligand binding assays in the 21st century laboratory-a call for change." Aaps J. 14(3): p. 377-9 (2012).
  4. US Department of Health and Human Services. US FDA, Center for Drug Evaluation and Research. Guideline for industry: Investigating Out-of-Specification (OOS) Test Results for Pharmaceutical Production. (2006).
  • <<
  • >>

Join the Discussion