Application of Micro-Flow Imaging (MFI TM ) to The Analysis of Particles in Parenteral Fluids. October 2006 Ottawa, Canada

Similar documents
Flow Microscopy: Dynamic Image Analysis for Particle Counting

Micro-Flow Imaging: Flow Microscopy Applied to Sub-visible Particulate Analysis in Protein Formulations

Testing for particles in injectable products

Technical Note. How to make Concentration Measurements using NanoSight LM Series Instruments

Characterization of protein aggregates in suspension and on a filter membrane by Morphologically-Directed Raman Spectroscopy

The Coulter Principle for Outlier Detection in Highly Concentrated Solutions

Fundamentals of Particle Counting

429 LIGHT DIFFRACTION MEASUREMENT OF PARTICLE SIZE

Standardization of Optical Particle Counters

The Ultimate Guide to Dynamic Imaging Particle Analysis for Protein Therapeutics:

Is NIST SRM2806b Responsible for the Sudden Increase in Particle Counts you have Seen on your Oil Analysis Reports?

Introducing the Morphologi G3 ID The future of particle characterization

Zeta Potential Analysis using Z-NTA

Development of an Automatic Sampling Module to Monitor Concentration of Liquid-Borne Nanoparticles

Technical Paper 3 Verifying Particle Counter Calibration: Size Verification

The Elzone II Particle Count and Size Analyzer

Holographic Characterization of Agglomerates in CMP Slurries

Particle Size Analysis with Differential Centrifugal Sedimentation. Marc Steinmetz Sales & Support Manager CPS Instruments Europe

Particle size analysis -Chapter 3

PARTICLE MEASUREMENT IN CLEAN ROOM TECHNOLOGY

CRITICAL EVALUATION OF THE EMERGING ANALYTICAL METHODS FOR CHARACTERIZATION OF SUB-VISIBLE PARTICLES

ORTHOGONAL PARTICLE CHARACTERIZATION TECHNIQUES FOR BIO-APPLICATIONS: AN INTRODUCTION TO DLS (DYNAMIC LIGHT SCATTERING), NTA (NANOPARTICLE

White Paper. Perform Conductivity Measurements In Compliance with USP <645>

Analytical methods to characterize aggregates and subvisible particles

Introduction to Differential Sedimentation

Model 2300XP PSL & Process-Particle Wafer Deposition System

Seeing the Nano-scale: Nanoparticle Tracking Analysis HVM MNT Expo 2006 Oxford. Jeremy Warren CEO, NanoSight Ltd

CD stage. ISO/CD Hydraulic fluid power Calibration of automatic particle counters for liquids ISO TC 131/SC 6 ISO TC 131/SC 6/WG 1 N313

The Characterization of Nanoparticle Element Oxide Slurries Used in Chemical-Mechanical Planarization by Single Particle ICP-MS

BECKMAN COULTER QbD1200 Total Organic Carbon Analyzer Critical Measurements Made Simple

MicroCell User manual

Analysis of Subvisible Particles. Linda O. Narhi Formulation and Analytical Resources, R&D Amgen, Inc.

Determination of particle size distribution Single particle light interaction methods. Part 4:

Latest Technology and Standardization Trends for Liquid-borne Particle Counters

Visualize and Measure Nanoparticle Size and Concentration

The solution for all of your

ITRS, SEMI and ASTM Guidelines for Semiconductor Ultrapure Water (UPW) Production and the Consequences for UPW Particle Metrology

Particle Size and Shape Analysis in PHARMACEUTICAL INDUSTRY.

Measuring Lysozyme Monomer at 0.1 mg/ml Concentration. Equipment used : Sample Preparation and Measurement :

REX Evaluation Guide. American Micro Detection Systems Inc March Lane, Suite 200 Stockton, CA 95219

Automated determination of the uniformity of dosage in quinine sulfate tablets using a fiber optics autosampler

OPTICAL PARTICLE SIZER MASS CALIBRATION METHOD

An introduction to particle size characterisation by DCS:

PARTICLE SIZE ANALYTICAL RANGES AND APPLICABILITY. m mm (10-6 m) nm (10-9 m)

Draft PS 18 APPENDIX A Dynamic Spiking Procedure ( ) 1.1 This appendix to Performance Specification 18

Test Method Development and Validation as Pertaining to Microtrac Particle Size Measuring Instruments

Introduction to Fourier Transform Infrared Spectroscopy

ActiPix SDI300 Surface Dissolution Imaging System

S2 PICOFOX. Innovation with Integrity. Spectrometry Solutions TXRF

Accurate Measurement of Transmittance and Reflectance for Engineering Applications

Holographic Characterization of Protein Aggregates

Pharmaceutical Particulate Matter

Biofuels Series. Application Note 301LS RAPID MEASUREMENT OF XYLOSE AND GLUCOSE MONITORING CORN STOVER FERMENTATION IN BIOETHANOL PRODUCTION

Advances in particle concentration measurements

The Better Way to Characterize Nanoparticles MANTA s ViewSizer 3000

MORPHOLOGI 4 RANGE. Automated imaging for advanced particle characterization

CALIBRATION CERTIFICATE FAILURE EXPLANATION FOR AEROTRAK PARTICLE COUNTERS

ISO INTERNATIONAL STANDARD

DI 3500 Discrete Aoalvzer

An Automated Application Template for Dissolution Studies

Introduction to Fourier Transform Infrared Spectroscopy

Developments & Limitations in GSR Analysis

Automated, intelligent sample preparation: Integration of the ESI prepfast Auto-dilution System with the Thermo Scientific icap 7400 ICP-OES

Method Development. Creating the Perfect Standard Operating Procedure (SOP) 2007 HORIBA, Ltd. All rights reserved.

Application Note PB 401. Improve HPLC Sample Preparation in an Analytical Laboratory A New Automated Sample Preparation Process

Algorithm User Guide:

High-Speed Environmental Analysis Using the Agilent 7500cx with Integrated Sample Introduction System Discrete Sampling (ISIS DS)

Multi-residue analysis of pesticides by GC-HRMS

Your partner in Science!

Measuring Colocalization within Fluorescence Microscopy Images

by Ray Garvey, Mong Ching Lin, and John Mountain Computational Systems Inc. 835 Innovation Drive, Knoxville, TN (423)

DDefense. MMetals. Marie C. Vicéns

Particle Characterization of Pharmaceutical Products by Dynamic Image Analysis

Particle Analysis at the Touch of a Button. Litesizer series

Using runt Disposable Cuvettes

Quantitative Analysis of Caffeine in Energy Drinks by High Performance Liquid Chromatography

BIOO FOOD AND FEED SAFETY. Histamine Enzymatic Assay Kit Manual. Catalog #: Reference #:

ANALYSIS OF LOW DENSITY PARTICLES USING DIFFERENTIAL CENTRIFUGAL SEDIMENTATION

BIO & PHARMA ANALYTICAL TECHNIQUES. Chapter 5 Particle Size Analysis

Standard Operating Procedure. Immunology

Mag-Bind Soil DNA Kit. M preps M preps M preps

Mixing in Colliding, Ultrasonically Levitated Drops

Method Development for a Simple and Reliable Determination of PCBs in Mineral Insulating Oil by SPME-GC-ECD

Simplifying the Process: Automated USP 643 / EP

Approval Block. Prepared by: Signature Date Evan Parnell 08 NOV Reviewed by: Signature Date. Approved by: Signature Date

An ultra-sensitive, versatile, and affordable single tube luminometer for Life Science Research.

Figure 2. Size distribution and concentration of a cocktail of PSL particles measured by three different sizing instruments.

Technical Procedure for Concentration Determination of Methamphetamine in Liquids via HPLC

NanoParticle Tracking Analysis The NANOSIGHT system.

Standard Operating Procedure for the Analysis of Dissolved Inorganic Carbon CCAL 21A.1

Confocal Microscopy Imaging of Single Emitter Fluorescence and Hanbury Brown and Twiss Photon Antibunching Setup

ITS. One of the highest resolutions for particle sizing and counting. Multisizer 3 COULTER COUNTER SCIENCE

Detection of Protein Aggregation by Enzyme Immunoassay

CEINT/NIST PROTOCOL REPORTING GUIDELINES FOR THE PREPARATION OF AQUEOUS NANOPARTICLE DISPERSIONS FROM DRY MATERIALS. Ver. 2.0

Fine Tuning Coriolis Flow Meter Calibrations Utilizing Piece- Wise Linearization

Quantos GraviPrep. Bye Bye, ml Hello, Quantos

Automated multi-vapor gravimetric sorption analyzer for advanced research applications

Peptide Isolation Using the Prep 150 LC System

Measuring NP Aggregation Propensities

MicroBrook Eyetech. Particle Size, Shape, and Concentration Analyzer. Particle Sizing. Particle Concentration. Particle Shape

Transcription:

Application of Micro-Flow Imaging (MFI TM ) to The Analysis of Particles in Parenteral Fluids October 26 Ottawa, Canada

Summary The introduction of a growing number of targeted protein-based drug formulations poses a significant challenge to the instrumentation technologies traditionally used for sub-visible and visible particle population analysis in parenteral fluids. US Pharmacopeia Chapter 788 (USP788) specifies maximum concentrations of sub-visible particles having equivalent a circular diameter greater than or equal to 1µm and 25µm as determined by light obscuration or, when applicable, by microscopic analysis following filtration. Since proteinaceous particles are highly transparent when compared to the polystyrene (PS) beads used for obscuration instrument calibration, they may be grossly undersized or missed entirely using this technique. The result is that a formulation may be measured as meeting USP788 requirements when, in fact, it does not. The limitations of light obscuration instruments have led to an interest in alternative technologies which are less dependent on particle properties and can provide additional information on particle parameters. This paper will describe the application of Micro-Flow Imaging (MFI TM ) technology to the analysis of parenteral particle populations in both the sub-visible and visible range. The instrument s performance with respect to its range of measurement parameters which complement this application will be outlined. A direct comparison will show that MFI can detect and measure particle concentrations in proteinaceous samples which are one or more orders of magnitude higher than the standard method, and that this effect is most pronounced for the larger particles of interest in USP788. The capability of the instrument to provide and analyze images of each particle will be shown. The value of the additional information provided by particle morphology data in assessing clinical outcome and in assisting to identify particle origin for further process control, quality control, diagnostics and troubleshooting will be discussed. 1. Introduction USP788 (Particulate Matter in Injections) describes physical tests to be performed for the purpose of enumerating sub-visible particles within specific size ranges contained within parenteral fluids. USP788 identifies light obscuration and microscopy as the methods for determining particle counts within the specified size ranges. Light obscuration is an indirect measurement technique. When a particle transits the measurement zone an optical beam is obscured with a resulting change in signal strength at the detector. This signal change is then equated to a particle s equivalent circular diameter (ECD) based on a calibration curve created using polystyrene (PS) spheres of a known size. To the extent that particles in intravenous solutions are composed of different materials and are often far from spherical, errors in sizing and counting are inevitable. Particles which are composed of highly transparent materials can be grossly undersized and, as a result, the concentration of larger particles is underestimated. A 23 Workshop on Particle Size Analysis was co-sponsored by the American Association of Pharmaceutical Scientists (AAPS), the Food and Drug Administration (FDA) and the United States Pharmacopeia (USP). The subsequent report 1 states: The measurement and expression of particle size is intimately bound with the shape and morphology of the constituent units that make up the ensemble of particles. The difficulties encountered when relating empirical information derived using different methods would not exist if the component particles were spherical. In the real world of pharmaceutics, particles are rarely (if ever) spherical, and consequently it is important to understand the importance of particle shape and morphology. 1 AAPS PharmaSci 24; 6 (3) Article 2, Page 3 Brightwell Technologies Inc. October 26 Page 2 of 11

This paper explores the application of MFI to the analysis of subvisible and visible particles in parenteral fluids. The following measurement parameters are considered: Section 3.1 Sensitivity for highly transparent particles Section 3.2 Dependence on particle material type Section 3.3 Sizing accuracy and repeatability Section 3.4 Concentration accuracy and repeatability Section 3.5 Visible/dense particle introduction Section 3.6 Very low concentrations of larger particles Section 3.7 Shape Analysis and Particle Morphology 2. Micro-Flow Imaging (MFI) MFI is a flow microscopic technology which operates by capturing images of suspended particles in a flowing stream. Different magnification set-points are available to suit the desired particle size range and image quality. The images are used to produce a particle database including count, size, transparency and shape parameters. The database can be interrogated to produce particle size distributions and isolate sub-populations using any measured parameter. Particle images are available for verification, further investigation and analysis. The sample volume in each frame is precisely defined by the flow cell geometry so particle concentrations are determined absolutely. No calibration by the user is required and all critical system parameters are automatically pre-set to eliminate run-torun and user dependent variations. For parenteral fluids, the primary interest is in detecting and measuring relatively large particles (>2µm). Therefore the low magnification (x5) option was selected. Very large (>7µm) particles can pose particular problems for sample introduction by aspiration due to particle densities and settling velocities. MFI includes a top-down gravity-assisted sample introduction method to address this problem. The sample introduction module permits the MFI instrument to analyze large particles which cannot be reliably suspended and aspirated. This sample introduction methodology is equally effective for small and large particles. The equipment setup is shown in Figure 1. Figure 1 - Isometric View of Sampling System Brightwell Technologies Inc. October 26 Page 3 of 11

A volume sampling speed of 1ml per 5 minutes is typically used. Clean baselines are achieved by flushing for 2 minutes between sample runs. A typical image frame (shown after thresholding has been applied) from a proteinaceous sample is shown Figure 2. The system software provides measurement of area, equivalent circular diameter, perimeter, intensity, circularity, and maximum Feret s diameter for each particle. Figure 2 - Typical Image Frame 3. Performance Parameters of MFI 3.1 Sensitivity For Highly Transparent Particles The results in Figure 3 are averaged from separate studies conducted by pharmaceutical laboratories on parenteral formulations containing proteinaceous particles. The solutions were analyzed using both traditional obscuration particle counters and MFI. MFI vs. Obscuration - Detection Sensitivity 1, 1, Particles/ml 1, 1 1 1 >5µm >1µm >25µm >4µm Size Range Micro-Flow Imaging Light Obscuration Figure 3 - Comparative Studies of MFI and Obscuration Brightwell Technologies Inc. October 26 Page 4 of 11

For particle size ranges relating to USP788, the measured concentrations for these sample types differ by one or more orders of magnitude and this difference becomes even greater for larger particles. The dramatically improved results provided by MFI were manually verified by direct observation of the images stored by the MFI system. Comparable experiments on transparent mineral particles have also demonstrated the higher sensitivity of the MFI technique. It can be hypothesized that the highly transparent particles are undersized and/or undetected by the obscuration instrument (due to a less sensitive threshold) resulting in these observed differences in measured concentration. To illustrate the differences in transparency between PS beads and real proteinaceous particles, an experiment was performed to compare their respective measured intensities. MFI measures the transparency of a particle by reading the intensity outputs of the pixels from the particle s image on the digital camera. A higher pixel intensity value equates to a higher degree of transparency. The results in Figure 4 illustrate a clear difference between the observed MFI mean intensity for proteinaceous particles versus PS beads which are similar in size (1µm) and shape (as measured by circularity). Transparency: Proteinaceous Particles vs. PS Beads Normalized Particle Count 8 7 6 5 4 3 2 1 1µm Polystyrene Spheres Proteinaceous Particles 5 55 6 65 7 75 8 85 Mean Intensity (1-124 units) Figure 4 - Transparency of Proteinaceous Particles vs. PS Beads 3.2 Dependence on Particle Material Type The direct, pixel-based imaging technique employed in MFI makes no assumptions of particle material type. Provided the presence of a particle results in sufficient contrast relative to the surrounding suspension fluid, the particle will be accurately sized. No calibration by the user is required. In order to explore the material dependence of parameter measurements, MFI has been evaluated with unstained and stained PS beads and beads of borosilicate glass. The results in Figure 5 compare measurements of PS beads which were stained red and nearly transparent borosilicate glass beads (both nominally sized at 1µm). Despite the Brightwell Technologies Inc. October 26 Page 5 of 11

widely different optical properties of the two types of beads, the sizing results are almost identical (note that these samples were not NIST-traceable). This relative material-insensitivity demonstrates that MFI is well suited for the heterogeneous populations commonly found in intravenous solutions. Material Insensitivity Concentration (#/ml) 5, 45, 4, 35, 3, 25, 2, 15, 1, 5, 5 6 7 8 9 1 11 12 13 14 15 Particle Size (µm) Borosilicate Glass Red Polystyrene Beads 3.3 Sizing Accuracy and Repeatability Figure 5 - Colored and Transparent Particle Size Distributions MFI contains design features which maximize sensitivity, accuracy and repeatability. These include an automatic, high sensitivity threshold setting, 1-bit threshold resolution and automatic background compensation for noise suppression. Experimental results with NIST-traceable PS spheres demonstrate the accuracy of MFI for sizing particles across its entire performance range of.75 to 4µm (see Figure 6). PS Bead Sizing Accuracy Measured Size (µm) 35 3 25 2 15 1 5 R 2 =.9999 5 1 15 2 25 3 35 NIST Certified Mean Diameter (µm) 5% 4% 3% 2% 1% % -1% -2% -3% -4% -5% Percent Error MFI Sizing Percent Error Linear (MFI Sizing) Figure 6 - PS Bead Sizing Accuracy The results in Figure 7 illustrate MFI sizing repeatability. In this experiment three (3) consecutive runs were performed on 15µm PS bead samples. The instrument was re-initialized, and the experiment repeated twice more for a total of 9 runs. Brightwell Technologies Inc. October 26 Page 6 of 11

15um PSL Bead Sizing Repeatibility (3 calibrations, 3 tests per calibration) 5 45 Concentration (/ml) 4 35 3 25 2 15 1 Cal1-R1 Cal1-R2 Cal1-R3 Cal2-R1 Cal2-R2 Cal2-R3 Cal3-R3 Cal3-R2 Cal3-R3 5 13. 13.25 13.5 13.75 14. 14.25 14.5 14.75 15. 15.25 15.5 15.75 16. 16.25 16.5 16.75 17. Size_ECD (um) 3.4 Concentration Accuracy and Repeatability Figure 7-15µm PS Bead Sizing Repeatability MFI instruments provide sensitive detection, counting, and sizing of individual particles in each image frame. Since a frame represents a known sample volume, particle concentration can be directly measured. Figure 8 and Figure 9 highlight experimental results with NIST-traceable PS bead concentration standards (Duke Scientific Count-Cal) demonstrating the concentration accuracy of MFI. The manufacturer guarantees a specification of 3 particles/ml with an accuracy of ±1% for both standards. The instrument was re-initialized between samples for a total of 6 runs on the 7µm standard and 3 runs on the 1µm standard. 1um PSL Concentration Standard Accuracy (CC1-PK Count-Cal) Concentration >7.5um (/ml) 3,3 3, 2,7 2,4 2,1 1,8 1,5 1,2 9 6 3 R1 R2 R3 Test Number CC1-PK Figure 8-1µm PSL Concentration Standard Accuracy Brightwell Technologies Inc. October 26 Page 7 of 11

7um PSL Concentration Standard Accuracy (CC7-PK Count-Cal) Concentration >5um (#/ml) 33 3 27 24 21 18 15 12 9 6 3 R1-Test1 R2-Test1 R3-Test1 R1-Test2 R2-Test2 R3-Test3 Test Number CC7-PK Figure 9-7µm PSL Concentration Standard Accuracy An additional benefit of MFI for concentration measurement is a higher upper limit compared to obscuration instruments (~275, particles/ml vs. ~12, particles/ml respectively). This provides a correspondingly higher threshold before coincidence errors occur and reduces the requirement for sample dilution to obtain accurate results. Real-time images provided by MFI permit the user to immediately verify the likelihood of coincidence errors in each sample as well as determine the presence of potential sources of contamination. Figure 1 shows measurements of a 1-fold dilution series carried out with 1µm PS bead size standards. Concentration Linearity 1, R 2 =.9999 Measured Concentration 1, 1, 1 1 1 1 1 1 1, 1, 1, Expected Concentration Figure 1 - MFI Dilution Series Brightwell Technologies Inc. October 26 Page 8 of 11

3.5 Visible/Dense Particle Introduction The stirring mechanism in the top-down gravity-assisted sample introduction system is designed to keep particles in a uniform suspension during testing. Results from a time study of a 7µm PS bead sample introduced with a 1ml syringe are shown in Figure 11. The data demonstrates that the concentration of the sample measured over a 15 minute period remains stable and settling is not a significant factor. Suspension Stability of 7µm PSDVB Spheres 6, Concentration (#/ml) 5, 4, 3, 2, 1, 3 6 9 12 15 Elapsed Test Time (min) Figure 11-7um Concentration vs. Time 3.6 Very Low Concentrations of Large Particles An emerging requirement for parenteral drug analysis is to detect and measure very low concentrations of large (visible) particles in the presence of high concentrations of smaller particles. The source of these large particles can include contamination and formulation instability. Table 1 and Table 2 are the results of experiments for the measurement of low concentration suspensions of NISTtraceable, 2µm PS beads. The first test used a concentration of ~2 particles per ml created by manually counting and suspending 11 particles into 5ml of filtered water. The second test used a concentration of 1 particle per ml created by mixing 5 particles into 5ml of filtered water. Parameter/Count per 5ml R1 Sample 11 MFI Count (particles >4µm) 92 Glassware Count (did not enter the system for analysis) 16 Image Verification Count (manual verification of stored images 79 % Recovery - based upon Image Verification 72% Table 1 - Low Concentration Measurement (2 Particles/ml) Brightwell Technologies Inc. October 26 Page 9 of 11

Parameter/Count per 5ml R1 R2 R3 R4 Sample 5 5 5 5 Count (particles >4µm) 7 26 5 5 Image Verification Count 5 5 3 5 % Recovery (Image Verification) 1% 1% 6% 1% Table 2 - Low Concentration Measurement (1 Particle/ml) Note 1: R1 and R2 contained additional particles which were shown by image analysis to result from contamination during sample preparation and handling. Particles may be lost either by lodging in the glassware and tubing or by having passed through the flow cell outside the field of view where the frame is captured. These initial results demonstrate that MFI is capable of reliably detecting very low concentrations of large particles. They also demonstrate the importance of fluidics design, development of optimum protocols for sample introduction and glassware preparation. The value of stored image analysis in providing a method of verifying the analysis and diagnosing unexpected results is also demonstrated. Additional development is expected to further improve on current performance in rare event particle detection. 3.7 Shape Analysis and Particle Morphology Because the light obscuration technique can only compare the signals received from real particles with those from PS spheres, particles are perceived as uniform spheres and particle size expressed in equivalent circular diameter. As was seen in Figure 2 and the images shown below, this assumption is misleading and particles vary widely in shape and uniformity. In contrast to obscuration, MFI provides an image of each particle detected. These images are observed by the user and analyzed by the system software to provide quantitative information on particle morphology. Measurement parameters, which include Feret s Diameter, area, perimeter, transparency and circularity, may be employed to create graphs and scatter plots which characterize the observed particle population. As an example, MFI images shown below demonstrate the differences between ECD (equivalent circular diameter) and maximum Feret Diameter for various proteinaceous particles found in a sample of parenteral fluid. In this case, Feret s Diameter is an effective parameter for distinguishing the particles based on their maximum dimension. Images on the left are the greyscale images as seen in the instrument while the images on the right are binary representations of the particles after thresholding. ECD = 12.13µm Max. Feret Diameter = 113.88µm ECD = 12.88µm Max. Feret Diameter = 237.88µm Brightwell Technologies Inc. October 26 Page 1 of 11

ECD = 113.13µm, Maximum Feret Diameter = 339.63µm 4. Conclusions Micro-Flow Imaging has distinct advantages over traditional obscuration counting in the analysis of sub-visible and visible particles in parenteral solutions. These include: Improved sensitivity and accuracy for near-transparent particle detection Results confirmation by direct image observation Insensitivity to particle material and shape Extended concentration range Ability to measure extremely low concentrations of outlier particles Additional information on particle morphology obtained by shape analysis The potential to isolate material types using morphological parameters The combined image and data information provided by this new technology may be employed in a wide variety of applications in the parenteral development and manufacturing process stages. Examples include formulation development, stability assessment, contamination analysis, process control, quality control, diagnostics and troubleshooting. Brightwell Technologies Inc. October 26 Page 11 of 11