Skip to main content

Posts

Ago2 Immunoprecipitation for RISC-siRNA Quantitation

Recent posts

Stem-Loop PCR for siRNA

 Stem-loop PCR is a method often used for detecting and quantifying small RNAs, such as siRNA or miRNA, which are typically difficult to amplify directly due to their short lengths. The method involves the design of a stem-loop reverse transcription (RT) primer, which enhances specificity and stability of the short RNA during the RT-PCR process, allowing for sensitive detection and quantification of the siRNA. Here’s a detailed guide to how stem-loop PCR can be applied to siRNA detection: Key Steps in Stem-Loop PCR for siRNA Designing the Stem-Loop RT Primer : Structure : The stem-loop RT primer consists of a loop region flanked by complementary sequences on either side (the "stem"), which will fold back on itself to form a hairpin structure. Specific Binding Region : A short sequence complementary to the 3’ end of the siRNA is added at the end of the stem-loop primer to ensure specific binding to the siRNA target. Stabilization : The loop structure helps prevent primer-dimer

EMSA for siRNA Plasma Protein Binding

Electrophoretic Mobility Shift Assay (EMSA) can be an effective technique for assessing plasma protein binding (PPB) of siRNA. EMSA measures shifts in the migration of siRNA on a gel in the presence of plasma proteins, which form complexes with siRNA and slow its movement through the gel matrix. This method, while less common for quantifying PPB, can provide qualitative or semi-quantitative insights into the interactions between siRNA and specific plasma proteins. Key Steps in EMSA for siRNA Plasma Protein Binding Preparation of siRNA-Plasma Mixture siRNA Labeling : For visual detection, label the siRNA with a fluorescent dye (e.g., Cy5 or fluorescein) or a radioactive isotope. Fluorescent labeling is often preferred for safety and ease of detection. Protein Mixture : Prepare a plasma sample or specific plasma protein (e.g., albumin or α-1 acid glycoprotein) in physiological buffer. It’s essential to use a concentration range to observe changes in siRNA binding at different protein lev

Ultrafiltration Method for siRNA PPB Evaluation

The ultrafiltration method is a widely used technique for assessing plasma protein binding (PPB) of siRNA molecules, including those conjugated with GalNAc. This method separates free (unbound) siRNA from protein-bound siRNA, enabling quantification of the unbound fraction. Here’s a detailed look at the ultrafiltration method, particularly for siRNA molecules, which can pose unique challenges due to their size, charge, and potential for non-specific binding. Steps in Ultrafiltration Method for siRNA PPB Evaluation Preparation of Plasma-siRNA Mixture : Dilute siRNA Sample : The test siRNA is diluted in plasma to achieve a physiologically relevant concentration. Equilibration : The plasma-siRNA mixture is incubated, typically at 37°C, to allow binding equilibrium between the siRNA and plasma proteins (e.g., albumin, α-1 acid glycoprotein). Ultrafiltration Device and Membrane Selection : Device : Specialized ultrafiltration devices (e.g., Amicon, Centrifree) are used to separate free siRN

Plasma Protein Binding (PPB) evaluation for GalNAc-conjugated small interfering RNA (siRNA)

The plasma protein binding (PPB) evaluation for GalNAc-conjugated small interfering RNA (siRNA) is an important parameter in understanding its pharmacokinetics (PK), distribution, and clearance from the body. GalNAc-siRNAs are designed for targeted delivery to hepatocytes via the asialoglycoprotein receptor (ASGPR), but their interaction with plasma proteins can impact their bioavailability and efficacy. Key Considerations in PPB Evaluation of GalNAc-siRNA Purpose of Plasma Protein Binding (PPB) Evaluation Distribution : Determines the extent to which the siRNA is free in plasma vs. bound to proteins, impacting its availability for hepatocyte targeting. Clearance : High protein binding often reduces renal clearance, prolonging circulation time, while low protein binding may lead to rapid clearance. Efficacy and Safety : Protein binding influences the siRNA’s pharmacodynamic (PD) effect by modulating its free fraction available for receptor binding. Methodology for PPB Evaluation Equili

Preclinical Studies for AAV Gene Therapy

 Preclinical studies for AAV gene therapy are crucial to assess the safety, efficacy, biodistribution, and immunogenicity of the therapy before progressing to human trials. These studies help in understanding the potential risks and therapeutic effects in animal models, which is essential for regulatory approval to proceed to first-in-human studies. Here’s a breakdown of key preclinical study types and their objectives: 1. Efficacy Studies Objective : Determine whether the gene therapy delivers a therapeutic benefit in relevant disease models, such as improvement in phenotypic markers or functional outcomes. Study Design : Use disease-specific animal models that reflect the condition the therapy intends to treat (e.g., knockout models for genetic disorders). Evaluate therapeutic endpoints, such as protein expression, functional assays, or phenotypic changes. Example : For a neurological condition, measure motor function or cognitive outcomes in treated versus control groups. 2. Biodist

Companion Diagnostic Assay: Introduction and Regulatory Perspective

 A companion diagnostic assay, as defined by the FDA, is a test that provides essential information for the safe and effective use of a corresponding therapeutic product. For gene therapies that use adeno-associated virus (AAV) vectors, an FDA-approved companion diagnostic can help identify patients likely to benefit from therapy by screening for factors such as pre-existing anti-AAV antibodies. Such a companion assay is especially valuable in gene therapy, where pre-existing immunity can significantly influence efficacy and safety. Key FDA Requirements for Companion Diagnostics Definition and Regulatory Oversight The FDA defines a companion diagnostic as a test essential for the therapeutic’s safe and effective use. This could mean identifying patients who may benefit from the therapy, avoiding patients likely to experience adverse reactions, or helping to tailor treatment plans. Companion diagnostics typically require FDA premarket approval (PMA) or clearance under a specific pathway

Companion assay to screen for anti-AAV antibodies (AAV Abs)

Developing a companion assay to screen for anti-AAV antibodies (AAV Abs) is a critical step in patient selection for AAV-based gene therapy. Since anti-AAV antibodies, particularly neutralizing antibodies (NAbs), can prevent effective transduction, an assay that reliably detects their presence and quantifies their activity is essential for determining patient eligibility and adjusting treatment protocols. Here’s a breakdown of key elements in designing a companion assay for AAV antibody screening: 1. Assay Type Selection ELISA (Enzyme-Linked Immunosorbent Assay) : Overview : Detects total anti-AAV antibodies, including both binding and non-neutralizing antibodies. Plates are coated with AAV capsid proteins, allowing antibodies in patient serum to bind and be detected via a secondary antibody. Advantages : Easy to standardize, high throughput, and cost-effective. Suitable for initial screening to determine if antibodies are present. Limitations : Cannot differentiate between neutralizin

Predicting the first-in-human (FIH) dose for AAV gene therapy

 Predicting the first-in-human (FIH) dose for AAV gene therapy from non-human primates, such as cynomolgus monkeys (cynos), involves careful extrapolation to ensure safety while aiming for therapeutic efficacy. Cynos are commonly used for AAV studies because of their physiological and immunological similarities to humans, making them an effective model for dose prediction. Here’s a structured approach to predicting the FIH dose for AAV gene therapy based on cyno data: 1. Determining the Relevant Cyno Dose Therapeutic Efficacy Threshold : In cyno studies, a dose that achieves therapeutic efficacy is identified first. For AAV gene therapy, this could mean achieving a target transgene expression level or a specific biomarker change in the target tissue. No Observed Adverse Effect Level (NOAEL) : The NOAEL is the highest dose in cynos that does not produce observable adverse effects. It’s crucial to identify this dose to balance efficacy with safety in humans. Maximum Tolerated Dose (MTD)

Allometric scaling in AAV gene therapy dose estimation

 Allometric scaling in AAV gene therapy dose estimation is crucial for translating effective and safe doses from animal models to humans. Since AAV dosing often involves high viral vector concentrations, proper dose scaling is essential to minimize adverse effects and optimize therapeutic outcomes. Here’s a breakdown of how allometric scaling is applied in AAV gene therapy dosing and the considerations involved: 1. Concept of Allometric Scaling Allometric scaling is a method of adjusting drug doses across species based on body size, physiology, and metabolism. It is especially useful in biologics and gene therapies where the pharmacokinetics and pharmacodynamics are more complex than small-molecule drugs. For AAV vectors, dosing is commonly scaled by body weight (e.g., vector genomes [vg] per kilogram) or body surface area (BSA), as these parameters can approximate dose distribution and vector exposure across different species. 2. Standard Scaling Approaches Body Weight Scaling (mg/kg