Drug Content Uniformity Validation in Nanostructured Lipid Carriers (NLCs) Manufacturing

Drug Content Uniformity Validation in Nanostructured Lipid Carriers Manufacturing

Validating Drug Content Uniformity During Nanostructured Lipid Carrier (NLC) 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 Content Uniformity in NLCs

Drug content uniformity is a critical quality attribute (CQA) for nanostructured lipid carrier (NLC) dosage forms. Achieving uniform drug distribution within NLCs directly influences therapeutic efficacy, safety, and patient compliance. Due to the nanoscale structure and lipid-based matrix, validating content uniformity in NLC manufacturing requires tailored approaches that address their unique physicochemical properties and manufacturing complexities. This section introduces the foundational concepts and outlines why drug content uniformity validation is pivotal for consistent product quality under current Good Manufacturing Practices (cGMP).

The Role of Drug Content Uniformity Validation in cGMP and Consistency

Under cGMP guidelines, pharmaceutical manufacturers must ensure every batch of the drug product exhibits consistent quality attributes, including drug content uniformity. Validation demonstrates control over variability during the NLC production process and confirms that all doses contain the intended drug amount within established specification limits. This minimizes risks related to under- or overdosing and assures regulatory compliance.

Specifically, content uniformity validation in NLCs helps to:

  1. Confirm robust mixing and homogenization of the active pharmaceutical ingredient (API) within the lipid matrix.
  2. Establish reproducible process parameters that maintain homogeneity batch-to-batch.
  3. Validate analytical methods sensitive enough to quantify drug content at nanoscale level.
  4. Support product stability and shelf-life by ensuring uniform drug distribution that prevents dose segregation during storage.

Defining the Quality Target Product Profile (QTPP) for NLCs

The QTPP sets the desired quality benchmarks from a patient and regulatory perspective and guides content uniformity validation. When defining the QTPP for nanostructured lipid carriers, focus on these attributes:

  • Dosage form: NLCs characterized by nano-sized lipid particles carrying the API.
  • Route of administration: Often topical, oral, or parenteral, influencing drug release and absorption.
  • Drug loading capacity: The maximum drug amount stably incorporated in NLC.
  • Drug content uniformity: API amount per dose unit with tight specification limits established.
  • Particle size distribution: Influences drug release and content uniformity.
  • Stability criteria: Maintaining uniformity during shelf life, avoiding drug migration or crystallization.

Use the QTPP to identify CQAs relevant for drug content uniformity and guide manufacturing controls.

Desired Attributes and Their Impact on Content Uniformity

Achieving uniform content in NLCs depends on several critical attributes related to formulation and process:

  1. Homogeneous API distribution: The API must be evenly dispersed within the lipid phase and throughout the nanoscale particles.
  2. Particle size and polydispersity: Narrow particle size distribution supports uniform dosing and reproducible quantification.
  3. Stability of lipid matrix: Preventing phase separation or drug crystallization maintains drug uniformity.
  4. Controlled mixing and emulsification conditions: Optimized homogenization power and duration promote consistent encapsulation and API dispersion.

Improper control of these attributes can lead to variation in API distribution, impacting drug release profiles and batch-to-batch consistency.

Identification of Critical Quality Attributes (CQAs) Related to Drug Content Uniformity

Validation requires careful selection of CQAs that directly influence and demonstrate drug content uniformity. For NLCs, the main CQAs include:

  • Drug concentration per batch: Ensures targeted loading level is achieved in manufacturing.
  • Drug content uniformity within the batch: Measured in multiple samples to confirm even API distribution.
  • Particle size and polydispersity index (PDI): Impacts drug encapsulation efficiency and uniform dosing.
  • Zeta potential and colloidal stability: Affect API retention within the lipid carriers.
  • Encapsulation efficiency: Percentage of drug successfully incorporated versus free drug, affecting dose uniformity.

These CQAs form the basis of acceptance criteria and are tightly monitored during and after manufacturing steps critical to content uniformity.

Key Properties and Parameters for Validation of Drug Content Uniformity

To effectively validate content uniformity in NLC manufacturing, focus on key properties and process parameters that influence uniform drug distribution:

  1. Sampling strategy: Develop a statistically justified sampling plan across batches, including samples at different production stages and from multiple container units.
  2. Analytical method development and validation: Employ sensitive, accurate, and precise assay methods specific for nanoscale formulations, such as HPLC, UV-Vis spectroscopy, or LC-MS, to quantify the drug in NLC suspensions or lyophilized powders.
  3. Process parameters affecting homogeneity: Optimize emulsification speed, temperature control, lipid phase composition, and mixing duration to minimize batch variability.
  4. Validation of mixing and homogenization equipment: Confirm that equipment used consistently produces uniform dispersions through performance qualification.
  5. Drug release characteristics: Monitor in vitro release profiles to detect any heterogeneity in API availability that may reflect content uniformity issues.

Collectively, these properties support robust validation protocols ensuring documented control over drug content uniformity.

Summary of Stepwise Approach for Content Uniformity Validation in NLCs

  1. Define QTPP and select CQAs related to drug content uniformity.
  2. Develop and validate analytical methods with sensitivity for nanoscale formulations.
  3. Design sampling plan covering multiple points within batches and container units.
  4. Establish manufacturing process parameters that promote homogeneous API distribution.
  5. Trace and document equipment qualification status ensuring consistent performance.
  6. Execute validation runs verifying consistent content uniformity within specification limits.
  7. Analyze data statistically to confirm process robustness and batch-to-batch consistency.
  8. Maintain ongoing monitoring via in-process checks and stability testing.

Following this logical framework ensures thorough validation of drug content uniformity in nanostructured lipid carrier manufacturing under cGMP compliance.

Comprehensive Drug Content Uniformity Validation in Nanostructured Lipid Carriers Manufacturing

Comprehensive Drug Content Uniformity Validation in Nanostructured Lipid Carriers 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.

Critical Quality Attributes (CQAs) Relevant to Drug Content Uniformity in NLCs

Identifying the CQAs that impact drug content uniformity is essential for designing and executing an effective validation strategy. Key CQAs include:

  • Particle size distribution: Influences drug encapsulation efficiency and uniformity within lipid matrices.
  • Polydispersity index (PDI): Reflects homogeneity of particle size, affecting dose consistency.
  • Drug loading and encapsulation efficiency: Indicate the actual drug content per particle versus the theoretical amount.
  • Zeta potential: Determines particle stability and aggregation tendencies, directly influencing content uniformity.
  • Lipid matrix composition and crystallinity: Affect drug distribution and release profiles.
See also  Moisture Content Validation in Powder for Injections Manufacturing

Monitoring these CQAs throughout the manufacturing process ensures detection of deviations that may compromise drug uniformity.

Key Physicochemical Properties Affecting Content Uniformity in NLCs

The inherent physicochemical properties of NLCs demand careful consideration during validation:

  1. Lipid polymorphism and phase behavior: Lipid rearrangements during storage or processing can displace drug molecules, altering uniformity.
  2. Drug solubility in lipid matrices: Highly soluble drugs tend to distribute more evenly, while poorly soluble drugs risk dose variability.
  3. Surface properties and adsorption: Affect interaction with excipients and potential drug migration within the formulation.
  4. Stability under processing conditions: Thermal and mechanical stresses can induce drug degradation or migration.
  5. Nano-distribution homogeneity: Ensuring even distribution of the drug at nanoscale is critical for batch uniformity.

Impact of Drug Content Uniformity on Quality Target Product Profile (QTPP)

Drug content uniformity directly influences multiple parameters outlined in the QTPP:

  • Safety and efficacy: Uniform drug dosing prevents sub-therapeutic or toxic effects.
  • Stability and shelf-life: Consistent drug distribution minimizes degradation and ensures predictable release profiles.
  • Bioavailability: Homogeneous drug presence improves absorption predictability.
  • Patient compliance: Assurance of consistent dosing fosters trust and adherence.
  • Regulatory compliance: Adherence to pharmacopeial content uniformity criteria ensures market approval.

Thus, content uniformity validation is integral to confirming that the NLC product meets intended QTPP standards throughout its lifecycle.

Introduction to Drug Content Uniformity Validation in NLCs Manufacturing

Validating drug content uniformity in Nanostructured Lipid Carriers (NLCs) manufacturing is critical to ensure consistent therapeutic efficacy and safety. This validation confirms that each dosage unit contains the intended drug amount within predefined limits, complying with pharmacopeial standards and regulatory requirements. The following stepwise approach provides a comprehensive framework for the process validation of drug content uniformity in NLCs.

Risk Assessment and Failure Modes Effects Analysis (FMEA)

Begin by conducting a thorough Risk Assessment and FMEA focused on factors impacting drug content uniformity in NLCs. This identifies critical failure modes that could lead to variability in active pharmaceutical ingredient (API) distribution.

  1. List all process parameters potentially affecting drug loading and homogeneity in NLCs, e.g., lipid concentration, mixing speed, temperature, homogenization pressure, and drug solubilization conditions.
  2. Identify potential failure points such as incomplete drug dispersion, nanoparticle aggregation, batch-to-batch variability, and sampling errors.
  3. Assign severity (S), occurrence (O), and detectability (D) scores to each failure mode using a 1-to-10 scale.
  4. Calculate Risk Priority Numbers (RPN = S × O × D) to prioritize risks affecting content uniformity.
  5. Focus on failure modes with high RPN for further process optimization and control strategy development.

Design of Experiments (DoE) for Critical Process Parameter (CPP) Selection

Use a systematic DoE approach to identify and optimize CPPs that significantly impact drug content uniformity in NLCs manufacturing. This approach supports robust process design and control.

  1. Select primary factors from FMEA results (e.g., lipid-to-drug ratio, homogenization pressure, temperature, mixing time) and specify realistic ranges.
  2. Design an appropriate experimental matrix, such as factorial or response surface methodology (RSM), to evaluate the effects and interactions of selected CPPs on drug loading efficiency and content uniformity.
  3. Execute experiments according to the DoE matrix using qualified equipment under controlled conditions.
  4. Analyze results statistically to determine significant CPPs and their optimal operating ranges ensuring uniform drug distribution.
  5. Establish acceptable critical quality attribute (CQA) ranges for drug content uniformity based on DoE outputs and regulatory guidelines.

Establishing the Control Strategy

Develop a comprehensive control strategy integrating process controls, monitoring, and acceptance criteria to maintain drug content uniformity during commercial-scale manufacturing of NLCs.

  1. Define in-process controls (IPCs) such as monitoring lipid phase temperature, homogenization parameters, and API solubilization time.
  2. Implement online or at-line analytical techniques, such as near-infrared spectroscopy (NIR) or high-performance liquid chromatography (HPLC), for real-time drug content monitoring where feasible.
  3. Set control limits for CPPs and CQAs based on DoE optimization results and validation batch data.
  4. Specify corrective actions and deviations procedures for excursions beyond control limits to prevent compromised content uniformity.

Process Flow and Sampling Points Definition

Map out the complete process flow and specify critical sampling points where drug content sampling will be performed to assess uniformity.

  1. Outline process steps from raw material dispersion, lipid phase melting, drug incorporation, homogenization, cooling, and final product collection.
  2. Identify key sampling points, such as after drug incorporation, post-homogenization, and final bulk NLC formulation.
  3. Define representative sampling methods ensuring statistically valid samples for content uniformity testing, adhering to pharmacopeial sampling guidelines.

Protocol Design for Process Performance Qualification (PPQ)

Design the PPQ protocol to confirm that the manufacturing process consistently produces nanostructured lipid carriers meeting drug content uniformity specifications.

  1. Define the number of consecutive batches (typically three) to be produced under routine commercial conditions.
  2. Specify detailed manufacturing steps, equipment settings, batch sizes, and sampling plans with exact time points and quantities.
  3. Include comprehensive acceptance criteria based on pharmacopeial standards (e.g., USP ) and internal quality requirements, generally requiring drug content to be within 85-115% of the label claim with low variability.
  4. Outline analytical methods for drug content determination, validated for accuracy, precision, specificity, and sensitivity for the NLC matrix.
  5. Incorporate failure mode contingencies, re-sampling protocols, and batch disposition criteria if uniformity is not achieved.

Batch Execution and Evaluation

Execute the PPQ batches according to the protocol and evaluate the results for drug content uniformity compliance and process robustness.

  1. Manufacture each batch under defined CPP settings, strictly adhering to the control strategy.
  2. Collect samples at predetermined points for drug content testing in accordance with the sampling plan.
  3. Analyze drug content data and calculate uniformity statistics such as mean, standard deviation, and coefficient of variation.
  4. Compare drug content values to the acceptable range; assess batch compliance and variability trends.
  5. Identify any out-of-specification (OOS) results and initiate investigation and corrective action procedures.
  6. Perform batch-to-batch comparability assessments to confirm process consistency and capability.
See also  Device Functionality Validation in Drug-Device Combinations Manufacturing

Documentation and Continuous Monitoring

Complete detailed validation reports and establish ongoing monitoring to maintain drug content uniformity in routine manufacturing.

  1. Compile comprehensive documentation including FMEA outcomes, DoE data, control strategy justification, PPQ batch records, and analytical test results.
  2. Document any deviations, investigations, and risk mitigation actions taken during validation.
  3. Establish a continuous process verification (CPV) plan incorporating periodic content uniformity testing of production batches.
  4. Implement trend analysis and capability indices monitoring to detect process drifts early.
  5. Update control strategies and validation documentation based on process improvements, formulation changes, or regulatory updates.

Summary

Following this structured approach to drug content uniformity validation in Nanostructured Lipid Carriers manufacturing ensures a scientifically sound, risk-managed process. By integrating risk assessment, DoE optimization, comprehensive control strategies, rigorous sampling, PPQ execution, and continuous monitoring, manufacturers can achieve and maintain drug uniformity compliance, guaranteeing patient safety and product efficacy.

Control Strategy Development and Establishing Acceptable Ranges

Develop a comprehensive control strategy to maintain drug content uniformity within the identified acceptable ranges during manufacturing.

  • Define Critical Quality Attributes (CQAs) related to drug loading and content uniformity according to pharmacopeial standards.
  • Establish control limits for each CPP based on DoE results, ensuring process parameters remain within ranges that guarantee uniformity.
  • Implement real-time monitoring methods (e.g., near-infrared spectroscopy, particle size analysis) for key parameters to detect deviations promptly.
  • Develop Standard Operating Procedures (SOPs) detailing corrective actions when parameters deviate from control limits.
  • Integrate in-process controls reflecting critical sampling points for batch assessment.

Sampling Plan and Decision Points

Define a rigorous sampling plan to verify drug content uniformity throughout the manufacturing process and at final product stage.

  • Specify sampling locations, including intermediate product streams and final NLC batches, considering homogeneity and scale of manufacturing.
  • Choose representative sample sizes and frequency per batch in compliance with pharmacopeial uniformity criteria (e.g., USP or Ph. Eur. 2.9.6).
  • Establish decision rules for acceptance or rejection based on drug content results and statistical confidence intervals.
  • Document sampling methodology clearly to ensure repeatability and compliance during routine and validation manufacturing runs.

Process Performance Qualification (PPQ) and Protocol Design

Design a robust PPQ protocol to demonstrate consistent drug content uniformity under commercial manufacturing conditions.

  • Outline objectives, scope, process descriptions, CPPs and CQAs, sampling plans, and acceptance criteria.
  • Include detailed stepwise workflow of NLC manufacturing—from raw material handling through homogenization to final packaging.
  • Incorporate risk mitigation plans for identified failure modes and predefined corrective actions.
  • Define responsibilities, data collection format, and statistical methods for analysis.
  • Schedule multiple consecutive commercial-scale batches for qualification to capture process variability.

Batch Execution and Evaluation

Execute PPQ batches following the approved protocol while rigorously documenting all process parameters and quality results.

  • Monitor CPPs in real time and record any deviations or out-of-specification findings immediately.
  • Perform drug content uniformity testing on sampled units as per sampling plan using validated analytical methods.
  • Analyze data statistically to ensure all batches meet predefined acceptance criteria and show consistent uniformity.
  • Investigate and document root cause analysis for any anomalies, implementing corrective and preventive actions (CAPA) as needed.
  • Compile a comprehensive validation report summarizing results, deviations, conclusions, and recommendations for routine manufacturing.

Introduction to Drug Content Uniformity Validation in NLCs Manufacturing

Drug content uniformity validation in Nanostructured Lipid Carriers (NLCs) manufacturing is crucial to ensure consistent therapeutic efficacy, patient safety, and regulatory compliance. This validation confirms that the active pharmaceutical ingredient (API) is uniformly distributed within each batch and across individual units of the dosage form. This process demands a systematic and rigorous approach using validated analytical methods, qualified equipment, and well-documented procedures.

Preparation Phase

Prior to initiating drug content uniformity validation, ensure the following preparatory tasks are completed:

  • Confirm that all manufacturing and analytical equipment have undergone installation qualification (IQ), operational qualification (OQ), and performance qualification (PQ).
  • Verify the availability of validated analytical methods specific for detecting and quantifying the API in NLCs formulation.
  • Define acceptance criteria based on pharmacopeial standards (e.g., USP ) or internal specifications, typically requiring individual unit content to be within 85% to 115% of the label claim, with relative standard deviation (RSD) ≤ 6%.
  • Establish sampling plans and batch size in line with regulatory guidance for content uniformity testing.

Sampling Procedure

Sampling plays a pivotal role in content uniformity validation:

  1. Select three consecutive pilot-scale or commercial-scale batches for evaluation to generate representative data.
  2. For each batch, randomly sample the minimum required number of individual units (typically 10 to 30 units) to cover variability within the batch.
  3. Ensure sample handling prevents degradation or contamination, maintaining integrity.

Analytical Testing

Perform content uniformity assay on sampled units using validated analytical techniques, such as High-Performance Liquid Chromatography (HPLC) or Ultra Performance Liquid Chromatography (UPLC), adapted for the lipid matrix:

  1. Calibrate instruments with appropriate standards before each run.
  2. Prepare samples using extraction protocols optimized to release API from NLC matrices without degradation.
  3. Run individual unit assays, following procedural controls and system suitability tests.
  4. Document assay results meticulously with raw data and chromatograms for traceability.

Data Compilation – Validation Result Tabulation Table

Compile assay results for each batch in a structured Validation Result Tabulation Table, including:


Batch No. Unit No. Assay Result (% Label Claim)
Batch 1 1 102.3
Batch 1 2 97.5
Batch 1
Batch 2 1 99.0

Note: This table should be fully populated with all unit assay results for each batch tested.

Statistical Analysis and Comparative Summary Table

Calculate the following for each batch:

  • Mean assay value of all units
  • Relative Standard Deviation (RSD) (%)
  • Minimum and maximum assay values
  • Compliance with established acceptance criteria

Use these results to create a Comparative Summary Table:

Batch No. Mean Assay (%) RSD (%) Min Assay (%) Max Assay (%) Compliance Status
Batch 1 99.4 3.5 95.0 105.0 Compliant
Batch 2 98.7 4.1 94.8 103.8 Compliant
Batch 3 100.2 2.9 96.1 104.1 Compliant

Optimum Analysis and Conclusion

Evaluate compliance based on pharmacopeial acceptance limits and statistical reproducibility:

  • Confirm that the RSD for each batch is within the target ≤ 6%, indicating minimal variability in drug content within the batch.
  • Ensure individual unit assay values fall within the 85%-115% range of labeled content.
  • Analyze trends across batches to detect systematic deviations or drifts using mean values and RSD comparisons.
  • If all batches meet criteria, conclude that the process consistently produces content uniform NLC formulations, validating the drug loading process.
  • If non-compliance is detected, investigate root causes such as process inconsistencies or analytical issues and implement corrective actions followed by re-validation.

Documentation and Reporting

Accurate documentation is a regulatory and quality imperative. Include the following in the validation report:

  • Complete Validation Result Tabulation Table and Comparative Summary Table.
  • Description of sampling plans and analytical procedures.
  • Statistical analysis method description including formulae for mean and RSD calculations.
  • Interpretation of results with conclusions on process capability and compliance.
  • Signatures of authorized personnel validating completeness and accuracy.

Change Control and Continued Process Verification (CPV)

Following initial validation, ongoing monitoring ensures sustained content uniformity:

  1. Establish a CPV protocol with predefined batch sampling frequency (e.g., first three commercial batches, then periodic sampling).
  2. Monitor assay variability over time using control charts to track mean and RSD values for drug content.
  3. Trigger investigations and corrective actions if trends indicate deviation beyond control limits.
  4. Document all CPV data and trends in Annual Product Quality Reviews (APQRs) for compliance and continuous improvement.

Annexure Templates for Validation Documentation

Include the following annexures in the validation dossier to standardize data capture and facilitate regulatory review:

Annexure I: Equipment Qualification Summary

Document IQ/OQ/PQ status of critical equipment used in manufacturing and analysis.

Annexure II: Sampling Plan Template

Defines batch sizes, number of units sampled, sampling locations, and handling procedures.

Annexure III: Analytical Method Validation Summary

Details validation parameters (specificity, accuracy, precision, linearity) for the assay method.

Annexure IV: Raw Data Capture Sheets

Provide forms or spreadsheets templates for recording unit assay results with batch and unit numbers.

Annexure V: Statistical Analysis Worksheets

Include formulas and example calculations for mean, standard deviation, RSD, and tabulation formats.

Summary

By following this stepwise approach to drug content uniformity validation for NLCs, pharmaceutical manufacturers can confidently demonstrate that the API is homogeneously distributed within the lipid carrier matrix and individual dosage units. Maintaining rigorous documentation, applying robust statistical analysis, and implementing ongoing monitoring through CPV are critical for ensuring consistent product quality and regulatory compliance.

Data Analysis and Compliance Assessment

Analyze the assay data to determine content uniformity compliance per batch:

  • Calculate mean drug content and relative standard deviation (RSD) for each batch.
  • Assess individual unit content against acceptance limits (typically 85-115% of label claim).
  • Verify that RSD does not exceed predefined thresholds (usually ≤6%).
  • Identify any outliers or units outside specifications and evaluate potential root causes.
Validation Result Tabulation Table: Content Uniformity Assay (Three Batches)
Batch No. Number of Units Tested Mean Drug Content (% Label Claim) Standard Deviation Relative Standard Deviation (RSD %) Units Outside Limits Compliance Status
Batch 1 30 98.5 2.5 2.54 0 Pass
Batch 2 30 99.2 3.0 3.02 1 Pass
Batch 3 30 97.8 3.2 3.27 0 Pass

Comparative Summary and Optimum Process Analysis

Compile and compare data across validation batches to confirm process consistency:

  • Create a comparative summary table highlighting key statistics for each batch.
  • Evaluate trends in mean content, variability, and out-of-specification occurrences.
  • Perform Root Mean Square Deviation (RMSD) or similar statistical techniques to identify batch-to-batch variability.
  • Determine optimum manufacturing conditions that minimize variability while maintaining compliance.
Comparative Summary Table: Drug Content Uniformity Across Batches
Parameter Batch 1 Batch 2 Batch 3 Overall Mean ± SD
Mean Drug Content (%) 98.5 99.2 97.8 98.5 ± 0.7
RSD (%) 2.54 3.02 3.27 2.94 ± 0.36
Units Outside Limits 0 1 0 1/90 units (1.1%)

Continued Process Verification (CPV) and Routine Monitoring

To sustain validated uniformity, implement ongoing monitoring and verification activities as part of the pharmaceutical quality system:

  • Establish routine sampling and content uniformity testing for commercial batches at pre-defined intervals.
  • Monitor key process parameters influencing uniformity, such as lipid-to-drug ratio, mixing speed, and homogenization cycles.
  • Use control charts (e.g., X-bar and R charts) to identify trends or deviations early.
  • Document all findings in CPV reports and trigger investigations if process drift is observed.

Annual Product Quality Review (APQR) and Trending

Include content uniformity data in periodic quality reviews and trending evaluations:

  • Compile comprehensive datasets including batch assays, process parameters, deviations, and out-of-specification (OOS) investigations.
  • Perform statistical trending to identify subtle shifts or patterns affecting drug content uniformity.
  • Review effectiveness of corrective and preventive actions (CAPA) related to uniformity failures.
  • Update control strategies and process parameters based on review outcomes for continual improvement.

Annexures

The following annexures are recommended for documentation and quality assurance:

  • Annexure I: Sample Collection Log Template
  • Annexure II: Analytical Test Report Template
  • Annexure III: Equipment Calibration and Qualification Records
  • Annexure IV: Validation Summary and Data Analysis Template
  • Annexure V: CPV and APQR Documentation Template
See also  Seal Integrity Validation in Capsules (Soft Gelatin) Manufacturing