Skip to main content

Selectivity Assessment of Bioanalytical Assay

Selectivity studies, also known as specificity studies, are an integral part of bioanalytical assay validation. These studies determine whether the assay can accurately measure the analyte of interest in the presence of potentially interfering substances, ensuring that the assay is specific to the analyte being measured. Here's how selectivity studies are typically performed:

Selectivity Experiment Design: The study is designed to evaluate potential interference from endogenous substances, metabolites, degradation products, or other analytes that might be present in the sample matrix. Known interfering substances, if identified, are included in the study.

Sample Preparation: A blank matrix (typically the same type of matrix as the study samples, such as plasma or urine) is prepared. The blank matrix is spiked with the analyte of interest at a low concentration (LLOQ) to mimic the lower limit of quantification.

Interfering Substances: If known interfering substances are identified, they are spiked into the blank matrix either individually or in combination with the analyte of interest. Interfering substances might include endogenous compounds, metabolites, or concomitant medications.

Analysis: The spiked samples are then analyzed using the bioanalytical assay. The goal is to determine whether the presence of interfering substances affects the accurate measurement of the analyte.

Data Analysis: The data is analyzed to determine whether the presence of interfering substances results in significant changes in the measured analyte concentration. Statistical analysis may be used to assess the impact of interference.

Acceptance Criteria: Acceptance criteria are established based on regulatory guidelines or predefined criteria. The criteria define the extent to which interference can be tolerated without compromising the accuracy of the assay.

Interpretation: If interference is observed, the laboratory must assess the potential impact on the assay's reliability. Depending on the degree of interference, corrective actions may be taken. These actions could include adjusting the assay conditions, matrix purification, or applying mathematical corrections.

Documentation: The results of the selectivity study, including any observed interferences and the laboratory's response, are thoroughly documented. This documentation serves as a record of how the assay responds to potential interferences.

Example: Selectivity Assessment for Drug Y Assay

Background:

  • Drug Y is a new investigational compound intended for treating a specific medical condition.
  • The bioanalytical assay measures the concentration of Drug Y in human plasma samples.
  • The assay must demonstrate selectivity by accurately measuring Drug Y in the presence of potentially interfering substances commonly found in plasma.

Selectivity Assessment:


Sample Preparation:

  • Prepare a set of human plasma samples as blank matrix samples (no Drug Y or interfering substances).
  • Prepare spiked samples by adding Drug Y at the lower limit of quantification (LLOQ) concentration (e.g., 5 ng/mL) to human plasma.

Interfering Substances:

  • Identify known interfering substances that could be present in plasma (e.g., endogenous compounds, metabolites, concomitant medications).

Spiking Interfering Substances:

  • Prepare spiked samples by adding the interfering substances to blank plasma samples at relevant concentrations.

Analysis:

  • Analyze the spiked samples containing Drug Y alone, spiked samples containing both Drug Y and interfering substances, and blank plasma samples using the bioanalytical assay.

Calculation of Selectivity:

  • Calculate the response (peak area or signal) of Drug Y in the presence of interfering substances and compare it to the response of Drug Y alone.

Data Interpretation:

  • If the presence of interfering substances does not significantly affect the accuracy and precision of Drug Y measurement, the assay is considered selective.
  • Calculate the percent change in response due to interference and assess whether it falls within acceptable limits (e.g., within ±20% of the response without interference).

Threshold for Selectivity:

  • Define a threshold for acceptable interference. If the response change is within this threshold, the interference is considered negligible.

Reporting:

  • Summarize the results of the selectivity assessment in the bioanalytical validation report.
  • Document the interfering substances tested, the concentrations used, and the impact on Drug Y measurement.

Conclusion:

The selectivity assessment demonstrates whether the bioanalytical assay can accurately measure Drug Y in the presence of potential interfering substances commonly found in plasma. If the assay's accuracy and precision remain within acceptable limits in the presence of these interferences, it confirms that the assay is selective for measuring Drug Y.

Popular posts from this blog

Human Genome Editing: FDA Draft Guidance Summary

Consideration for Developing Gene Editing Product  1. Genome Editing Methods: Genome editing can be achieved through nuclease-dependent or nuclease-independent methods. Nuclease-dependent methods involve introducing site-specific breaks in DNA using technologies like zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), modified-homing endonucleases, and CRISPR-associated (Cas) nucleases. These breaks can lead to modification of the DNA sequence at the cleavage site. Nuclease-independent methods can change DNA sequences without cleaving the DNA and include techniques like base editing and synthetic triplex-forming peptide nucleic acids. The choice of GE technology should consider factors such as the mechanism of action, the ability to target specific DNA sequences, and the potential to optimize components for efficiency, specificity, or stability. 2. Type and Degree of Genomic Modification: Different GE approaches rely on DNA repair pathways such as ho

Human Gene Therapy for Neurodegenerative Diseases: FDA Guidance Summary

  Neurodegenerative diseases are a diverse group of disorders characterized by the progressive degeneration of the central or peripheral nervous system, and they can have various causes and clinical characteristics. This guidance document is a resource for sponsors on different aspects of product development, preclinical testing, and clinical trial design. It acknowledges the unique challenges and considerations associated with developing GT products for such complex and varied diseases. Below are the key summaries from the guidance. CONSIDERATIONS FOR CHEMISTRY, MANUFACTURING AND CONTROLS (CMC) The considerations for Chemistry, Manufacturing, and Controls (CMC) when developing gene therapy (GT) products for the treatment of neurodegenerative diseases are crucial for ensuring the safety and efficacy of these advanced therapies. Here, we will elaborate on the specific CMC considerations outlined in your text: Route of Administration and Product Volume: Neurodegenerative diseases often r

Cellular & Gene Therapy Guidances (Food and Drug Administration)

  Studying Multiple Versions of a Cellular or Gene Therapy Product in an Early-Phase Clinical Trial; Draft Guidance for Industry 11/2022 Human Gene Therapy for Neurodegenerative Diseases; Guidance for Industry 10/2022 Considerations for the Development of Chimeric Antigen Receptor (CAR) T Cell Products; Draft Guidance for Industry 3/2022 Human Gene Therapy Products Incorporating Human Genome Editing; Draft Guidance for Industry 3/2022 Interpreting Sameness of Gene Therapy Products Under the Orphan Drug Regulations; Guidance for Industry 9/2021 Manufacturing Considerations for Licensed and Investigational Cellular and Gene Therapy Products During COVID-19 Public Health Emergency; Guidance for Industry 1/2021 Chemistry, Manufacturing, and Control (CMC) Information for Human Gene Therapy Investigational New Drug Applications (INDs); Guidance for Industry 1/2020 Long Term Follow-up After Administration of Human Gene Therapy Products; Guidance for Industry 1/2020 Testing of Retroviral Vecto

Stem loop RT-PCR for Detection of siRNA in Animal Tissues

Step Loop RT-PCR for Detection of Small Interfering RNA (siRNA) The recent publications described a novel used the novel method for the detection of siRNAs using a TaqMan®-based approach. This approach utilizes similar strategy that has been used for microRNA detection. The approach is illustrated in below.  In brief, the RT step occurs in the presence of a stem-loop RT primer that is complementary to the last 6–10 bases of the 3′ end of the antisense strand of the target siRNA. The stem-loop primer contains an additional universal sequence at the 5′ end that facilitates a TaqMan-based detection strategy in the subsequent qPCR step. As in the case of microRNA, the forward primer for qPCR is sequence-specific for the target siRNA. For sequence compositions that yield a low predicted melting temperature (Tm), the forward primer is designed as a tailed primer to help increase Tm. Stem Loop PCR for SiRNA Detection Step 1: Preparation of liver and plasma samples for the quantification of si

Engineering AAV vectors for enhanced safety profiles

 Engineering AAV vectors for enhanced safety profiles involves multiple strategies at both the vector genome and capsid levels. Here is a breakdown of these strategies: Vector Genome Level: Modifying Vector Genome Sequences: Scientists modify AAV vector genomes by adding, mutating, or deleting specific sequences. For example, self-complementary AAV (scAAV) vectors are designed by deleting key signals from the second inverted terminal repeat (ITR), allowing for more efficient genome replication. Codon Optimization: Optimizing the codon usage of the transgene can enhance its expression efficiency. Promoter and PolyA Sequence Selection: Careful selection and manipulation of promoter and polyadenylation (polyA) sequences can influence transgene expression and tissue specificity. Capsid Engineering: Capsid engineering strategies can be categorized into four main categories: Directed Evolution: This approach involves creating capsid mutant libraries using error-prone PCR or introducing pepti