Drug Release Rate Validation in Mucoadhesive Formulations Manufacturing

Drug Release Rate Validation in Mucoadhesive Formulations Manufacturing

Comprehensive Guide to Drug Release Rate Validation in Mucoadhesive Formulations Manufacturing

All equipment used in this process validation must be duly qualified and validated for its intended use and performance specifications. Equipment qualification (IQ/OQ/PQ) is assumed to be completed prior to this process validation.

Introduction to Drug Release Rate Validation in Mucoadhesive Formulations

The validation of drug release rate is a critical component in the manufacturing of mucoadhesive formulations. These dosage forms are designed to adhere to mucosal membranes, releasing the active pharmaceutical ingredient (API) in a controlled manner over time. Ensuring consistent and predictable drug release profiles is essential to achieve therapeutic efficacy and patient safety. This validation process is a formal documented approach to confirm that the manufacturing process consistently yields mucoadhesive products that meet predefined release criteria.

Drug release rate validation, within the context of mucoadhesive dosage forms, integrates formulation science, process control, and analytical testing. This approach establishes confidence in product performance and compliance with regulatory requirements aligned with current Good Manufacturing Practices (cGMP).

Role of Drug Release Rate Validation in cGMP and Product Consistency

Step 1: Understand the regulatory framework—cGMP requires manufacturers to demonstrate that critical quality attributes, such as drug release rate, are controlled within established limits to guarantee batch-to-batch consistency.

Step 2: Define the scope of validation—drug release validation ensures that in-process controls and final product testing confirm the release profile meets the target product profile. This safeguards patients by delivering the API at the proper rate and duration, critical to the safety and efficacy of mucoadhesive systems.

Step 3: Integrate quality risk management to identify potential sources of variability in release rates, such as raw material variability, process parameters, and equipment performance. Address these risks through controls and monitoring strategies within the validation protocol.

Step 4: Establish documented procedures to demonstrate process reproducibility and robustness over multiple batches. This includes frequent review of validation data to detect trends or deviations indicating process drift.

Quality Target Product Profile (QTPP) for Mucoadhesive Formulations

Step 1: Define the QTPP focused on drug release attributes relevant to mucoadhesive dosage forms. Key elements include:

  • Desired release kinetics (e.g., immediate, sustained, or controlled release).
  • Target duration of adhesion to mucosal tissue.
  • Drug release rate range (amount released per hour or percentage released over defined times).
  • Physicochemical characteristics impacting release and adhesion, such as viscosity, polymer type, and swelling behavior.

Step 2: Cohesively align the QTPP with patient-centric criteria such as ease of administration, minimal irritation, and compliance with stability requirements. These parameters influence drug release consistency and must be considered during validation.

Desired Attributes for Drug Release Rate in Mucoadhesive Products

Step 1: Establish the critical physical and chemical attributes of the formulation affecting the drug release rate, including polymer composition, molecular weight, and crosslinking density that govern the mucoadhesive strength and API diffusion.

Step 2: Define target ranges for viscosity and gel formation properties that influence release kinetics. The release profile must be reproducible within set limits across batches.

Step 3: Identify dissolution medium parameters that reflect physiological mucosal fluid conditions, including pH and ionic strength, since these critically impact drug release and adhesive performance.

Step 4: Consider the mechanical retention time on mucosa, which affects cumulative drug release—this informs acceptance criteria for release rate validation tests.

Impact of Drug Release Rate on Quality Target Product Profile

Step 1: Correlate drug release rate to clinical performance endpoints defined in the QTPP, ensuring that the release rate supports the intended therapeutic outcome, such as prolonged analgesia or localized drug delivery.

Step 2: Recognize that deviations in drug release behavior can lead to sub-therapeutic exposure or toxicity. Therefore, precise control of the release rate is integral to maintaining the QTPP consistency.

Step 3: Understand that drug release affects other CQAs like content uniformity, mucoadhesive strength, and stability. Altered release rates may signal underlying formulation or process deficiencies needing corrective action.

Critical Quality Attributes (CQAs) Related to Drug Release Rate

Step 1: Identify CQAs directly affecting drug release rate including:

  • API particle size distribution impacting dissolution.
  • Polymer concentration and characteristics influencing matrix swelling and erosion.
  • Mucoadhesive strength correlating with retention time and release profile stability.
  • Dose uniformity ensuring consistent API availability.
  • Moisture content affecting polymer hydration and drug diffusion.

Step 2: Establish analytical methods validated for sensitivity and specificity to monitor these CQAs throughout development and production.

Step 3: Define acceptance criteria for each CQA that align with desired drug release performance and QTPP objectives.

Key Properties Influencing Drug Release Rate in Mucoadhesive Formulations

Step 1: Evaluate polymer selection and matrix architecture, as these largely determine the swelling, erosion, and drug diffusion mechanisms governing release.

Step 2: Assess formulation parameters such as drug load, excipient ratios, and plasticizer content, which modulate the matrix integrity and release kinetics.

Step 3: Control manufacturing parameters including mixing time, temperature, and drying conditions that impact the microstructure and homogeneity, influencing release consistency.

Step 4: Employ in vitro release testing methods standardized to simulate mucosal conditions, enabling reliable and reproducible assessment of drug release profiles.

Step 5: Monitor environmental factors during storage, such as humidity and temperature, which may alter polymer properties and consequently, drug release rate.

Drug Release Rate Validation in Mucoadhesive Formulations Manufacturing

Drug Release Rate Validation in Mucoadhesive Formulations Manufacturing

All equipment used in this process validation must be duly qualified and validated for its intended use and performance specifications. Equipment qualification (IQ/OQ/PQ) is assumed to be completed prior to this process validation.

Quality Target Product Profile (QTPP) and Desired Attributes

Step 1: Define the Quality Target Product Profile (QTPP) that establishes the desired clinical and quality attributes of the mucoadhesive formulation, emphasizing the drug release characteristics such as onset, duration, and consistency.

Step 2: Determine the critical desired attributes impacting drug release, including mucoadhesion strength, polymer composition, viscosity, and gelation behavior, all of which influence the release kinetics.

See also  Mechanical Integrity Validation in Implants (Subdermal or Intraocular) Manufacturing

Step 3: Specify acceptable ranges for drug release profiles that align with therapeutic objectives, ensuring patient compliance and minimizing variability.

Impact of Drug Release Rate on QTPP

Step 1: Correlate drug release rate with therapeutic efficacy—ensure release profiles meet absorption and bioavailability targets outlined in the QTPP.

Step 2: Identify how changes in release rate affect mucoadhesion time, dosing frequency, and overall patient experience.

Step 3: Monitor the influence of formulation excipients and manufacturing parameters on the drug release rate to maintain consistency within the QTPP framework.

Critical Quality Attributes (CQAs) Relevant to Drug Release Rate

Step 1: Identify key CQAs such as API particle size, polymer molecular weight, cross-linking density, pH of formulation, and in vitro mucoadhesion time that affect drug release behavior.

Step 2: Establish acceptance criteria for these CQAs based on historical data, formulation development studies, and regulatory guidelines.

Step 3: Implement routine testing of CQAs during manufacturing to ensure ongoing control over the drug release profile.

Key Properties of Mucoadhesive Formulations Influencing Release Rate

Step 1: Evaluate the polymer type and concentration as primary determinants of matrix integrity and drug diffusion rates.

Step 2: Assess viscosity and rheological properties that affect adhesion and drug diffusion from the formulation.

Step 3: Measure surface pH and swelling behavior, which can alter the microenvironment and impact drug solubility and release kinetics.

Step 4: Consider formulation homogeneity and content uniformity, ensuring consistent API distribution for reproducible release.

Introduction to Drug Release Rate Validation in Mucoadhesive Formulations

Validating the drug release rate in mucoadhesive formulations is crucial to ensure consistent therapeutic efficacy and patient safety. This process validation must encompass a thorough understanding of the formulation characteristics, manufacturing variables, and analytical methods to reliably demonstrate that the release profile meets predetermined specifications throughout commercial production.

Define Critical Quality Attributes and Critical Process Parameters

Begin by establishing the Critical Quality Attributes (CQAs) related to drug release rate, including dissolution profile, adhesion time, and amount of drug released at specific time points. Subsequently, identify Critical Process Parameters (CPPs) that influence these CQAs, such as polymer concentration, mixing time, granule moisture content, compression force, and coating thickness. Use prior knowledge, historical data, and laboratory studies to develop a risk-based understanding of CPP impact.

Conduct Risk Assessment Using FMEA

Perform a comprehensive Failure Mode and Effects Analysis (FMEA) focused on drug release variability in mucoadhesive dosage forms. Follow these steps:

  • Identify potential failure modes: e.g., incomplete polymer swelling, drug-polymer interaction variability, improper adhesion leading to premature detachment, inconsistent drug loading.
  • Assess severity: rate the impact of each failure on patient safety and product efficacy (e.g., 1=minimal, 10=critical).
  • Evaluate occurrence: estimate how often each failure might happen during manufacturing.
  • Determine detectability: analyze likelihood of detection through in-process controls or final product testing.
  • Calculate Risk Priority Number (RPN): multiply severity, occurrence, and detectability scores to prioritize risks.

Prioritize high RPN factors to address them in the validation strategy.

Design of Experiments (DoE) to Optimize CPPs

Develop a structured DoE plan targeting high-risk CPPs identified via FMEA. Steps include:

  • Select CPPs for study, such as polymer type/concentration, drying temperature, mixing speed, or compression force.
  • Choose a suitable experimental design model (e.g., factorial design, central composite design) to explore parameter effects and interactions.
  • Define response variables linked to drug release rate, including percentage release at predetermined timepoints and mucoadhesive strength.
  • Execute experiments in a controlled laboratory setting, collecting data on formulation and process variations.
  • Analyze results statistically to establish design space boundaries where CQAs are consistently met.

Selection and Justification of Critical Process Parameters

Based on DoE findings, finalize the list of CPPs to tightly control during commercial production. Justify selections with data showing their direct effect on drug release behavior and product consistency. Establish acceptable ranges for each CPP within the validated design space to minimize variability.

Develop a Control Strategy for Drug Release Rate Consistency

Create a control strategy integrating in-process controls and release testing targeted to maintain CQAs, including:

  • In-process controls: monitor and record CPPs such as polymer blend uniformity, granule moisture content, compression parameters, and coating thickness.
  • Sampling plans: define sampling frequency and location to capture batch representativeness and reduce variability risks.
  • Analytical method validation: ensure drug release testing methods are robust, precise, and accurate for detecting release rate deviations.
  • Specification limits: define acceptance criteria for drug release profile, guided by clinical relevance and regulatory standards.

Process Flow and Stepwise Workflow for Validation Execution

Establish a detailed process flow covering all manufacturing steps critical to drug release including:

  1. Raw Material Qualification: verify polymer, drug substance, and excipient quality per specifications.
  2. Material Preparation: blend and granulate ingredients with controlled parameters to ensure reproducibility.
  3. Formulation and Dosage Form Production: undergo tableting or film casting with tightly monitored CPPs.
  4. Coating/Application of Mucoadhesive Layer: control solvent evaporation rate, coating thickness, and drying parameters.
  5. Packaging: maintain environmental controls to prevent moisture or heat impact on formulation.

This workflow ensures systematic control over all variables impacting drug release rate.

Sampling and Decision Points During Validation Batches

Define sampling strategy aligned with process steps where variability can affect drug release:

  • Collect samples post-blending to verify uniformity.
  • Sample intermediate granules to assess moisture and particle size.
  • Test finished dosage forms from multiple lot positions for release rate consistency.

Implement real-time monitoring where feasible. Return to prior step or reject batch if drug release deviations are detected beyond acceptable limits.

Performance Qualification (PPQ) Batch Execution

Execute a minimum of three consecutive PPQ batches under defined conditions with full adherence to control strategy and sampling plan:

  • Document all process parameters and deviations in batch records.
  • Perform drug release testing using validated analytical procedures.
  • Analyze batch data statistically to confirm reproducibility, uniformity, and compliance with release criteria.

Success criteria include demonstration that all CQAs are controlled within predefined limits and no trends suggesting process drift.

Evaluation and Reporting of Validation Results

Compile comprehensive validation report containing:

  • Summary of FMEA and risk assessment results.
  • DoE findings and justification of CPP ranges.
  • Control strategy details and sampling rationale.
  • PPQ batch data, drug release results, trend analysis, and deviation handling.
  • Conclusions affirming process capability to consistently produce mucoadhesive formulations meeting drug release specifications.
  • Recommendations for ongoing monitoring and routine quality assurance.
See also  Filling Accuracy Validation in Powder for Injections Manufacturing

Submit documentation to regulatory authorities as part of process validation package.

Ongoing Monitoring and Continuous Improvement

Post-validation, implement robust process monitoring through routine batch analysis and trending of drug release data. Should release rate variability increase or approach specification limits, initiate root cause investigations and preventive actions. Integrate continuous improvement feedback into manufacturing protocols and training to sustain control over drug release characteristics.

Establish Control Strategy and Acceptable Ranges

Based on DoE results and FMEA prioritization, establish a robust control strategy to maintain CPPs within validated ranges that ensure consistent drug release characteristics. This includes:

  • Setting upper and lower acceptable limits for each CPP, e.g., polymer concentration (%) between 2.0 and 2.5%
  • Implementing in-process controls such as moisture content checks, mixing time verification, and compression force monitoring
  • Defining sampling frequency during manufacturing to detect deviations early
  • Including specifications for key intermediate and final product attributes related to release rate

Process Flow and Stepwise Workflow Definition

Outline the complete manufacturing process with emphasis on steps critical to drug release rate, including:

  1. Raw material receipt and characterization
  2. Polymer blending and granulation to achieve homogeneity
  3. Drying and moisture content adjustment
  4. Compression with controlled force settings
  5. Coating application with specified thickness and uniformity
  6. In-process testing and adjustments as per CPP limits
  7. Packaging under controlled environmental conditions

Define clear decision points for sampling and corrective actions if CPPs or CQAs deviate from established criteria.

Sampling Strategy and Decision Points

Develop a detailed sampling plan for process validation batches:

  • Collect samples at predetermined stages (e.g., post-blend, post-compression, post-coating)
  • Perform drug release testing at multiple time points according to dissolution protocol
  • Monitor adhesion time and other mucoadhesive properties alongside
  • Use statistical sampling techniques to ensure representative batch assessment
  • Establish criteria for acceptance, reprocessing, or batch rejection based on results

Process Performance Qualification (PPQ) Batch Execution and Evaluation

Execute at least three consecutive PPQ batches under defined manufacturing conditions to demonstrate process consistency. Key aspects include:

  • Strict adherence to validated CPP ranges and control strategy
  • Comprehensive documentation of batch records, deviations, and corrective actions
  • Perform complete drug release profile assessments for all batches to confirm reproducibility
  • Evaluate adhesion and other mucoadhesive performance parameters in parallel
  • Analyze data statistically to confirm batch-to-batch consistency within predefined limits

The evaluation results should confirm that the process robustly produces mucoadhesive formulations matching drug release rate specifications.

Protocol Design and Documentation

Develop a detailed process validation protocol encompassing all stages of the drug release rate validation, including:

  • Scope, objectives, and responsibilities
  • Comprehensive list of CPPs and CQAs with acceptance criteria
  • Sampling plans and analytical methods description
  • Batch manufacturing instructions highlighting control points and parameter limits
  • Defined investigation and contingency measures for failures
  • Data analysis plan for assessing process capability and validation success

This protocol should be approved by quality assurance and manufacturing management prior to execution.

Ongoing Monitoring and Continuous Verification

Following successful validation, implement a continuous monitoring program to ensure ongoing control of drug release rate, including:

  • Routine in-process sampling and release testing
  • Trend analysis of CPPs and CQAs to detect drift or shifts
  • Periodic review and revalidation triggers based on significant process or formulation changes
  • Utilization of statistical process control tools to maintain quality within validated limits

This ongoing verification ensures sustained product performance and compliance over the lifecycle of the mucoadhesive formulation.

Introduction to Drug Release Rate Validation in Mucoadhesive Formulations

Drug release rate validation in mucoadhesive formulations is a critical step to ensure consistent therapeutic performance and product quality. This validation confirms that the manufacturing process consistently produces mucoadhesive dosage forms that release the active pharmaceutical ingredient (API) within the predetermined specifications. This section assumes that all manufacturing equipment used in the drug release testing has undergone and passed its installation, operational, and performance qualification (IQ/OQ/PQ).

Define Validation Protocol and Acceptance Criteria

Begin by drafting a detailed process validation protocol specific to the drug release rate testing of mucoadhesive formulations. This protocol must include:

  • Objective of the validation
  • Scope describing the dosage form and manufacturing scale
  • Sampling plan, including the number of batches and sample units per batch
  • Analytical methods and equipment to be used for drug release measurement, including calibration details
  • Acceptance criteria for drug release rate, typically expressed as a range of release percentages over specified time points based on pharmacopoeial or in-house specifications
  • Risk assessment addressing potential sources of variability from formulation, equipment, and environment
  • Documentation responsibilities and timelines

Conduct Three Consecutive Batch Validation (CPV Phase)

Process performance qualification (PPQ) involves executing the manufacturing process for three consecutive commercial-scale batches under predefined conditions, followed by drug release testing. Follow these instructions precisely:

  1. Manufacture the first batch under routine conditions, collecting representative mucoadhesive dosage units as per the sampling plan.
  2. Perform drug release testing using a validated dissolution apparatus appropriate for mucoadhesive systems (e.g., paddle-over-disk or flow-through cell) and validated analytical methods.
  3. Document drug release metrics at specified intervals (for example, 1, 2, 4, and 8 hours), recording mean release percentage and standard deviation.
  4. Repeat steps 1–3 identically for two additional batches, ensuring environmental and process parameters remain controlled and consistent.

Compile Validation Results – Tabulation and Initial Data Analysis

After completing release testing for all three batches, compile the results into a Drug Release Rate Validation Result Table, as illustrated below:

Batch No. Sampling Time (hr) Mean % Release Standard Deviation (SD) Relative Standard Deviation (RSD, %) Acceptance Criteria Met (Yes/No)
Batch 1 1 xx.x x.x x.x Yes
2 xx.x x.x x.x Yes
4 xx.x x.x x.x Yes
8 xx.x x.x x.x Yes
Batch 2 1 xx.x x.x x.x Yes
2 xx.x x.x x.x Yes
4 xx.x x.x x.x Yes
8 xx.x x.x x.x Yes
Batch 3 1 xx.x x.x x.x Yes
2 xx.x x.x x.x Yes
4 xx.x x.x x.x Yes
8 xx.x x.x x.x Yes
See also  Emulsification Speed Validation in Emulsions Manufacturing

Ensure all RSD values are within acceptable limits, usually < 10% depending on regulatory expectations and product complexity. Confirm all values fall within the defined acceptance criteria.

Perform Comparative Summary and Statistical Analysis

Create a comparative summary table to consolidate key performance indicators (KPIs) across all three batches:

Parameter Batch 1 Batch 2 Batch 3 Mean Overall RSD (%) Compliance Status
% Drug Released at 1 hr xx.x xx.x xx.x xx.x x.x Pass
% Drug Released at 2 hr xx.x xx.x xx.x xx.x x.x Pass
% Drug Released at 4 hr xx.x xx.x xx.x xx.x x.x Pass
% Drug Released at 8 hr xx.x xx.x xx.x xx.x x.x Pass

Interpretation guidelines:

  • Mean: Average of all batches for each sampling time point, representing the central tendency.
  • RSD: Calculated as (SD/Mean) × 100, reflecting batch-to-batch variability.
  • Compliance Status: Indicate “Pass” if the mean and individual batch results meet acceptance criteria and RSD is within limits.

Use this analysis to confirm process consistency and identify any trends or deviations requiring further investigation.

Documentation and Trending in APQR

Validation results, including raw data, summary tables, and statistical analysis, must be documented comprehensively in the validation report. The report should include:

  • Introduction and objective
  • Test methods and equipment description
  • Batch manufacturing records
  • Drug release data and analysis
  • Compliance statements
  • Conclusion and recommendations

Following approval of the initial process validation, incorporate drug release rate monitoring into routine quality control and Annual Product Quality Review (APQR) procedures. Trending data over time should be analyzed to detect shifts or trends that could indicate potential process drift or quality risks.

Routine Monitoring and Revalidation Triggers

Routine monitoring of drug release rate as a Critical Quality Attribute (CQA) must be established as part of the in-process and finished product quality control protocol. Steps include:

  1. Periodic sampling from routine batches according to established sampling plans.
  2. Comparative analysis against validated batch data.
  3. Trend analysis using control charts or statistical tools to ensure process stability.

Revalidation triggers include but are not limited to:

  • Significant changes to formulation or raw material suppliers
  • Equipment modifications or relocation
  • Changes in manufacturing scale or site
  • Failure trends identified during routine monitoring or APQR reviews
  • Regulatory requirements or corrective actions

Annexures (Templates)

The following annexures must be prepared and appended to the validation documentation. Use standardized templates to ensure consistency and compliance:

Annexure I: Validation Protocol Template

  • Objective and scope of validation
  • Equipment details and method outline for drug release testing
  • Sampling plan and acceptance criteria
  • Responsibilities and timelines

Annexure II: Batch Manufacturing Records Checklist

  • Batch identification
  • Process parameters
  • Sampling records
  • Raw material traceability

Annexure III: Drug Release Test Result Sheet

  • List batch number and sampling times
  • Data entry section for mean, SD, and RSD values
  • Acceptance criteria confirmation

Annexure IV: Validation Summary and Comparative Table

  • Field to summarize validation outcomes and statistical analysis
  • Batch-to-batch comparison table
  • Compliance declaration

Annexure V: Change Control/Revalidation Form

  • Template to document any changes impacting drug release
  • Risk assessment section
  • Decision on revalidation necessity

Tabulation of Validation Results

Compile drug release data from the three consecutive validation batches into a structured tabulation table for clarity and ease of analysis. Include the following data points for each batch and time interval:

  • Batch identification number
  • Mean percent drug release
  • Standard deviation (SD)
  • Relative standard deviation (RSD)
  • Compliance status against acceptance criteria


Validation Result Tabulation Table
Batch No. Time (hrs) Mean % Drug Release Standard Deviation (SD) Relative Standard Deviation (RSD %) Compliance (Y/N)
Batch 1 1 XX.X X.X X.X Y
Batch 1 2 XX.X X.X X.X Y
Batch 1 4 XX.X X.X X.X Y
Batch 1 8 XX.X X.X X.X Y

Comparative Summary and Statistical Analysis

Conduct a comparative summary analysis across the three batches to verify consistency and compliance. Follow the guidelines below:

  1. Calculate batch-to-batch variability and mean values at each time point.
  2. Determine the overall RSD and assess if it meets predetermined acceptance thresholds, typically ≤ 10% for drug release assays.
  3. Prepare a Comparative Summary Table highlighting means, SDs, and RSDs for all batches at each time interval.


Comparative Summary Table of Drug Release Rate
Time (hrs) Batch 1 Mean ± SD Batch 2 Mean ± SD Batch 3 Mean ± SD Overall Mean ± SD RSD (%) Compliance Status
1 XX.X ± X.X XX.X ± X.X XX.X ± X.X XX.X ± X.X X.X Compliant / Non-compliant
2 XX.X ± X.X XX.X ± X.X XX.X ± X.X XX.X ± X.X X.X Compliant / Non-compliant

Analyze results considering pharmacopoeial guidelines and process capability. Confirm that the drug release profiles meet the predetermined specifications and demonstrate process robustness.

Continued Process Verification (CPV) and Routine Monitoring

To maintain validated status and product consistency, establish ongoing monitoring protocols as follows:

  1. Implement a routine in-process and finished product testing schedule for drug release rate with predefined sampling frequencies.
  2. Use control charts and trending analysis to monitor drug release data for drifts or shifts beyond control limits.
  3. Perform periodic risk assessments based on trending data to identify potential process deviations or formulation stability issues.
  4. Adapt corrective and preventive actions (CAPA) promptly upon detection of out-of-specification (OOS) or non-compliance trends.

Annual Product Quality Review (APQR) and Trending Analysis

Incorporate drug release rate validation data into the APQR as part of a comprehensive quality monitoring strategy:

  • Review all drug release test results from the past year, including CPV and routine releases.
  • Assess data trends via statistical tools—averages, RSD, and control limits.
  • Document any deviations, investigations, CAPA implementation, and their effectiveness.
  • Recommend opportunities for process improvements or further validations if required.

Annexures – Templates and Documentation

To ensure standardization and traceability, attach the following annexures when submitting validation documentation:

  • Annexure I: Drug Release Rate Validation Protocol Template
  • Annexure II: Batch Manufacturing Record Extract – Sampling and Testing
  • Annexure III: Drug Release Analytical Method Validation Report
  • Annexure IV: Validation Result Tabulation Template (blank)
  • Annexure V: Trending and CPV Monitoring Chart Template

Ensure all annexures are duly signed, dated, and approved by the quality and manufacturing teams responsible.