WO1997046860A2 - Improvements in detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form - Google Patents

Improvements in detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form Download PDF

Info

Publication number
WO1997046860A2
WO1997046860A2 PCT/US1997/011791 US9711791W WO9746860A2 WO 1997046860 A2 WO1997046860 A2 WO 1997046860A2 US 9711791 W US9711791 W US 9711791W WO 9746860 A2 WO9746860 A2 WO 9746860A2
Authority
WO
WIPO (PCT)
Prior art keywords
dissolution
dosage form
active agent
drug
release
Prior art date
Application number
PCT/US1997/011791
Other languages
French (fr)
Other versions
WO1997046860A3 (en
WO1997046860A9 (en
Inventor
Kurt Roinestad
Frank S. Cheng
Philip J. Palermo
Kevin Bynum
Original Assignee
Euro-Celtique, S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Euro-Celtique, S.A. filed Critical Euro-Celtique, S.A.
Priority to AU43262/97A priority Critical patent/AU729814B2/en
Priority to CA002257106A priority patent/CA2257106C/en
Priority to EP97941329A priority patent/EP0938657A4/en
Priority to JP1998500947A priority patent/JP3380864B6/en
Priority to US08/915,785 priority patent/US6174497B1/en
Publication of WO1997046860A2 publication Critical patent/WO1997046860A2/en
Publication of WO1997046860A3 publication Critical patent/WO1997046860A3/en
Publication of WO1997046860A9 publication Critical patent/WO1997046860A9/en
Priority to US09/707,498 priority patent/US6558957B1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N13/00Investigating surface or boundary effects, e.g. wetting power; Investigating diffusion effects; Analysing materials by determining surface, boundary, or diffusion effects
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/15Medicinal preparations ; Physical properties thereof, e.g. dissolubility
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N13/00Investigating surface or boundary effects, e.g. wetting power; Investigating diffusion effects; Analysing materials by determining surface, boundary, or diffusion effects
    • G01N2013/006Dissolution of tablets or the like

Definitions

  • Dissolution testing is required for all solid oral pharmaceutical solid dosage form forms in which absorption of the drug is necessary for the product to exert the desired therapeutic effect
  • the U.S. Pharmacopeia (USP) is one well-known standard source of information which provides for dissolution and drug release testing in the majority of monographs for such dosage forms Exceptions are for tablets meeting a requirement for completeness of solution or for rapid ( 10 to 15 minutes) disintegration for soluble or radiolabled drugs.
  • the apparatus and procedure conform to the requirements and specificaitons given, e.g., USP 23rd edition Chapter 711 (Dissolution) pages 1791-1793 Dissolution testing serves as a measure of quality control, stability and uniformity as well as a means by which to correlate in-vitro with in-vivo drug release characteristics
  • a small aliquot of sample is taken from each vessel, usually by a multi channeled pumping system, and transported to either a cuvette or a sample vial for subsequent spectrophotometric or high pressure liquid chromatography (HPLC) analysis, respectively. Plotting percentage dissolution of a solid dosage form through time results in a dissolution profile.
  • HPLC high-density lipoprotein
  • spectrophotometric method the HPLC method is usually favored over the spectrophotometric method.
  • HPLC dissolution offers the advantage of specificity, acceptable accuracy, precision and sensitivity
  • the disadvantage of the status quo rather lies with the inherent burden of creating, manipulating, and storing voluminous numbers of sequence and data files.
  • the cost of HPLC, columns, mobile phases, and the waste solvent disposal, etc. is substantial and the limited number of data points that can be determined may result in a less than ideal representation of the release profile of a solid dosage form over time.
  • HPLC dissolution offers the advantage of specificity, acceptable accuracy, precision and sensitivity
  • the disadvantage of the status quo rather lies with the inherent burden of creating, manipulating, and storing voluminous numbers of sequence and data files.
  • the cost of HPLC, columns, mobile phases, and the waste solvent disposal, etc. is substantial and the limited number of data points that can be determined may result in a less than ideal representation of the release profile of a solid dosage form
  • HPLC analysis is a sequential time consuming process. In general, a typical 24 hour dissolution requires up to 60 hours to generate a dissolution profile.
  • the present invention relates to an improvement in a detection system used for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a singular dissolution vessel or multiple dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft and a probe placed within the mixing shaft or outside the individual dissolution vr ⁇ sels, the probe capable of measuring the dissolution characteristics using UV " • , aear- IR, fluorescence, electrochemical, nuclear magnetic resonance (NMR), and Raman spectroscopy techniques.
  • the present invention also relates to a method for predicting the dissolution curve provided by a controlled release pharmaceutical dosage form comprising taking continuous measurements of the amount of drug released from a dosage form for a portion of the time over which the drug is expected to be released and predicting the remainder of the dissolution curve based on the values obtained.
  • the present invention relates to a in-situ dissolution methods to evaluate and study the dissolution characteristics of drug formulations
  • Such methods utilize systems that include fiber optics, ultraviolet spectroscopy, fluorescence spectroscopy, NMR and the like.
  • the present invention specifically relates to detection systems for measuring dissolution characteristics of pharmaceutical dosage forms using ultraviolet, LR, near-LR, and Raman spectroscopy techniques as well as electrochemical techniques such as polarography, and NMR.
  • Figure 1 shows the UV-vis spectra of tramadol standard solutions at four different concentrations
  • Figure 2 the linearity plot of tramadol HCl solutions of Example 1 ;
  • Figure 3 is a graphical representation of repeated UV-vis scans at 30 minute intervals over 25 hours for a tramadol HCl 200mg once-a-day tablet of Example 2,
  • Figure 4 shows the plot of the average dissolution of three tramadol HCl once-a-day tablets of Example 2 and the results from the HPLC method
  • Figure 5 displays the plot of the dissolution of a tramadol tablet of Example 3 over 45 minutes
  • Figure 6 is a graph of the dissolution profile of a tramadol controlled release tablet, using the average dissolution results from table 1, by using TableCurve 2D program, using the best fit equation (as described in Example 4);
  • Figure 7 is a graph showing the dissolution profile of a tramadol controlled release tablet as described in Example 4 obtained from 12 hour sampling data, at 1 hour intervals, using the to the best fit equation (as described in Example 4),
  • Figure 8 is a graph showing the dissolution profile of a tramadol controlled release tablet obtained from 16 hour data, taken at 1 hour interval,using the best fitted equation (as described in Example 4),
  • Figure 9 is a graph of a dissolution profile of a tramadol controlled release tablet, when 16 hour data, generated at every half hour, are used to find the best fit curve (as described in Example 4);
  • Figure 10 shows a plot comparison of dissolution data obtained from both a fiber optics v. HPLC methods (as described in Example 6);
  • Figure 1 1 depicts a preferred configuration of the present invention
  • Figure 12 depicts a closed-vessel embodiment of the invention
  • Figure 13 depicts a UV probe in shaft embodiment of the invention.
  • One aspect of the present invention is related to an improvement in a detection system for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a dissolution vessel containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft having a probe contained within, the probe capable of measuring the release of the drug using fluorescence, ultraviolet (UV), Infrared (IR), near-Infrared (IR), electrochemical, and Raman spectroscopy techniques.
  • UV ultraviolet
  • IR Infrared
  • IR near-Infrared
  • electrochemical and Raman spectroscopy techniques.
  • the present invention further provides an improvement wherein the probe utilizes ultraviolet spectroscopy techniques, electrochemical techniques, Infrared
  • Another aspect of the present invention provides a method for predicting the dissolution curve provided by a controlled release pharmaceutical dosage form comprising taking continuous measurements of the amount of drug released from a dosage form for a portion of the time over which the drug is expected to be released and predicting the remainder of the dissolution curve based on the values obtained.
  • a method according to the present invention utilizes a detection system comprising a singular dissolution vessel or multiple dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement in the detection system comprising a mixing shaft and a probe placed within the mixing shaft or outside the individual dissolution vessels, the probe capable of measuring the dissolution characteristics using UV, IR, near-IR, fluorescence, electrochemical, and Raman spectroscopy techniques.
  • Yet another aspect of the present invention relates to an improvement in a detection system for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a plurality of dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft having a probe contained within, the probe capable of measuring the release of the drug using fluorescence, ultraviolet, Infrared, near-Infrared, electrochemical, and Raman spectroscopy techniques. It is further provided that this aspect of the present invention may utilize at least two vessels to optionally hold a dissolution medium or a placebo formulation for baseline correction.
  • the present invention also provides an improvement in a detection system for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a singular dissolution vessel or multiple dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft and a probe placed outside the individual dissolution vessels, the probe capable of measuring the dissolution characteristics using UV, IR, near-LR, fluorescence, electrochemical, and Raman spectroscopy techniques.
  • At least two vessels in the inventive system optionally hold a dissolution medium or a placebo formulation for baseline correction.
  • the present invention particularly relates to detection systems for measuring dissolution characteristics of pharmaceutical dosage forms using ultraviolet, LR, near-LR, and Raman spectroscopy techniques as well as electrochemical techniques such as polarography.
  • the present invention also relates to a dissolution apparatus for determining a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, the apparatus including a detector for quantifying one or more physical and/or chemical properties of the therapeutically active agent, the detector operatively associated with the dissolution medium for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent; and a data processor for continually processing the generated data for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form.
  • the present invention also relates to a method for determining a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, including the steps of continually generating physical and/or chemical data characteristic of the therapeutically active agent by operatively associating a detector with the dissolution medium for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent; and continually processing the generated data with a data processor for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form.
  • Another preferred embodiment of the invention relates to a dissolution arrangement for measuring in- vitro release of an active agent from a dosage form containing the active agent, including a plurality of vessels, each of the vessels containing a dissolution media and a dosage form containing an active agent to be measured, a fiberoptic probe associated with each of the vessels, each of the fiberoptic probes including a detector which simultaneously and continuously measures the concentration of active agent in the dissolution media, and a data processor connected to the fiberoptic probes, the data processor continually processing information received from the probes concerning the concentration of the drug to obtain a dissolution profile of the dosage form.
  • the dissolution arrangement further includes utilizing the data processor to predict future concentrations of the active agent.
  • the dissolution arrangement further comprises utilizing the data processor to predict a 24 hour dissolution profile of the active agent after 16 hours of dissolution time has elapsed.
  • the term releasable quantity is defined, for purposes of the present invention, as the maximum amount of therapeutically active agent that can be released from a pharmaceutical dosage form during the dissolution testing time period.
  • the releasable amount may be less than 100% of the total amount of agent contained in the pharmaceutical dosage form.
  • the dissolution testing time period is preferably at least one hour, and in certain embodiments is 8-24 hours or longer, e.g., 48, 72 or 96 hours.
  • the term physical and/or chemical properties is a physical and or chemical property that is characteristic of a particular therapeutically active agent.
  • a non-limiting list of physical and/or chemical properties include ultraviolet abso ⁇ tion or radiation spectra; infrared abso ⁇ tion or radiation spectra; , ⁇ or gamma radiation; electron states; polarity; magnetic resonance; concentration electro-chemical properties and the like.
  • the physical and/or chemical properties of a agent are any property characteristics of a agent or group of agents that can be used to detect, e.g. the presence, absence, quantity, physical state or chemical state of that agent.
  • agent is defined as any chemical or physical entity or combination of entities, particles or organisms either that are detectable by a detector.
  • agents include chemicals, therapeutically active agents, radiation particles (e.g., ⁇ -particles); microbes such as bacteria, viruses, individual cells from a multi-cellular organism (e.g., blood cells); and the like.
  • a detector is defined for pu ⁇ oses of the present invention as any device that detects a physical and/or chemical property of a agent and generate data regarding about the physio-chemical property.
  • detectors include UV- spectrophotometers, Geiger counters, fluorscopic devices and the like.
  • the physical and/or chemical property detected by the detector and the type of data generated by the detector are not critical to the present invention.
  • operatively associated is defined for pu ⁇ oses of the present invention as positioning the detector in proximity to the vessel containing the subject agent such that the detector can quantify the desired physical and/or chemical data characteristic of the agent, and transmit the data to a data processor.
  • dissolution apparatus of the present invention is particularly useful for determin .g a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, the apparatus including a detector for generating physical and/or chemical data characteristic of the therapeutically active agent, said detector operatively associated with the dissolution medium for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent, and a data processor for continually processing the generated data for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form.
  • the detector may be any detector known in the art that generates physical and/or chemical data of the test agent, e.g. a uv spectrophotometer.
  • the detector has a probe communicably attached thereto.
  • there is a corresponding detector capable of continuously generating physical and/or chemical data characteristic of the agent to be analyzed.
  • the data processor may be any device capable of continuously processing the data generated by the detector.
  • the data processor is a computer.
  • the data generated by the detector is preferably stored and/or analyzed by the computer.
  • the data collector is a computer that has data processing software, e.g. Microsoft Excel 5.0 or Tablecurve.
  • the data generated by the detector is processed by the software and reorganized into a preferred form, e.g. as a graph or a table.
  • the data is preferably continuously processed by the software as it is received from the detector.
  • the apparatus further comprises a shaft.
  • the shaft has at least one aperature therein, which aperature allows the detector to detect the necessary physical and or chemical properties of the subject agent and generate the required physical and/or chemical data.
  • the size and position of the opening along the shaft will depend ona variety of factors, including, but not limited to, the type of detector used and the physical and/or chemical property to be detected.
  • the shaft has an orifice therein for receiving the detector
  • the detector is attached to the shaft
  • the detector is attached to the shaft by any attachment means known in the art, including, but not limited to, welds, adhesives, soldering, screws, friction, and the like
  • the detector is permanently attached to the shaft by, for example, soldering the detector to the shaft
  • the detector is rotatably attached to the shaft in a manner such that when the detector is receivedin the shaft, the shaft can freely rotate about the detector, allowing the shaft to perform other functions independent of the detector
  • a paddle or basket may then be affixed to at least one end of the shaft such when the shaft is rotated, the paddle or basket also rotates to provide, e g , agitation when the paddle or basket is contacted with an external environment, e g dissolution media
  • the detector measures the concentration of the agent, e g , therapeutically active agent, in the media surrounding the dosage form, e g , simulated gastic fluid, or simulated intestinal fluid
  • concentration of the agent in the surrounding media e g , therapeutically active agent, in the media surrounding the dosage form, e g , simulated gastic fluid, or simulated intestinal fluid
  • the present invention also relates to a method for determining a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, including the steps of continually generating physical and/or chemical data characteristic of the therapeutically active agent by oneratively associating a detector with the dissolution medium for at least the time per IOJ required for the dosage form to release the maximum releasable quantity of therapeutically active agent, and continually processing the generated data with a data processor for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form
  • the invention includes three components, a conventional dissolution apparatus, a UV detection unit and a Pentium computer running Windows 95 and Excel 5 0 software
  • the conventional dissolution apparatus, a Distek 5100 bathless unit (or equivalent unit), is interfaced to a UV radiation source with fiber optic transmission dip probes, and
  • the agent is dissolved in the solvent, however, for pu ⁇ oses of the present invention, the agent may be dispersed or suspended throughout the solvent in a solid or semi-solid media
  • the agent need not be dissolved in the solvent, but may, instead, provide a dispersion or suspension medium for the agent
  • the device comprises a detector for monitoring chemical and/or physical properties of an agent, wherein the detector is mounted to a shaft having a hollow portion capable of receiving said detection means, said shaft having an aperature therein that allows said detection means to communicate with said external environment when said detection means is received by said hollow portion
  • the detection means may be permanently mounted to the shaft, or preferably removably mounted to the shaft to as allow a near infinate combination of shafts and detection means
  • the mount is preferably a universal mount which will allow an almost infinite combination of detection means and shafts
  • the detection means is capable of aquiring data characteristic of a particular agent by a method selected from the group consisting of ultraviolet radiation, infrared radiation, nuclear magnetic resonance, Raman sprctroscopy, electrochemical, and mixtures thereof, with ultraviolet radiation detection being particularly preferred
  • the shaft is rotatably attached to said detection means, such that the shaft is freely rotatabie around the peripheral edges
  • the device includes a data collecting means, e g a computer
  • the computer is capable of operating data collection software which facilitates analysis or collection of the data generated by the detection means
  • the software may serve to merely store the data, or it may provide compariitive analysis to rfrence standards, produce graphic representations of the data (e g , dissolution vs time curves), or other assorted functions known in the art
  • the software will preferably be capable of continuously receiving said data from said detection means, providing near-instantaneous access to the data derived from a given test
  • the present invention is also directed to a method for continuously monitoring a agent in an external environment, e.g dissolution media, including the steps of collecting data characteristic to a particular agent in an external environment by positioning at an effective distance to the external environment a device for continually monitoring the agent in the external environment comprising a detection means for detecting a agent in an external environment mounted to a shaft having a hollow portion capable of receiving the detection means; the shaft having an
  • electrochemical techniques used in the present invention optionally include biosensors, in which a transducer is coupled to a biological element, to quantitate a change in concentration of target analyte(s)
  • the new dissolution system has been applied to study the dissolution characteristics of pharmaceutical dosage forms, for example analgesic products, such as Tramadol HCI QD Tablets and Hydromo ⁇ hone Capsules
  • the Ocean Optics Inc. PC Plug-In Fiber Optic Miniature Spectrometer is used with an ultraviolet probe as the method of detection.
  • the probe is coupled to a LS-1 deuterium light source and detection is conducted using a SI 000 spectrometer. Data is processed using SpectraScope and Microsoft Excel 5.0 software The detector is capable of scanning the entire UV and visible spectrum in under 2 seconds. Comparison with the current method for dissolution analysis of solid dosage forms was conducted.
  • a more powerful deuterium light source from Oriel Co ⁇ oration, Stratford, CT can also be used to replace the LS-1 deuterium lamp when higher light throughput is required.
  • This light source also has the advantage of using a condensing lens to manipulate the quality of light hitting the fiber optic interface.
  • a xenon arc lamp source from Oriel Co ⁇ oration may also be used for applications requiring increased sensitivity, such as Hydromorphone and Hydrocodone Controlled Release Products.
  • a variable path length dip probe from CIC Photonics, Inc. of Albuquerque NM can be used for method development pu ⁇ oses to determine optimal flow cell pathlength for a given drug product
  • the dissolution bath was a Hansen Research model SR5 with type II (paddle) agitation.
  • the bath temperature was maintained at 37 +/-0.5 degrees and solution was agitated at 100 rpm.
  • Figure 1 shows the UV-vis spectra of Tramadol standard solutions at four different concentrations. Inspection of the spectra in Figure 1 reveals relatively noise free data with well defined spectral features The absorbance of Tramadol vs concentration at the maximum absorbtivity (272 nm) are shown in Table 1 below The correlation coefficient of the regression line is 0 999825 indicating a linear relationship between concentration and absorption The linearity plot is shown in Figure 2
  • Three Tramadol 200 mg QD tablets were placed in the dissolution medium to check its release rate over three different days by the in-situ system.
  • the repeated UV-vis scans at 30 minute intervals over 25 hours for one of the tablets is shown in Figure 3.
  • the dissolution data of these tablets are shown in Table 2.
  • Table 2 also shows the average of the three and the dissolution results obtained from an existing, validated, HPLC method for comparison.
  • a Tramadol 200 mg QD tablet was placed in the in-situ dissolution system and the amount of Tramadol released monitored in real time. This was obtained by a process called history channel evaluation in which the UV-vis scans of the analyte are acquired in about every 2.5 seconds. The abso ⁇ tion at a pre-selected wavelength is plotted against time to generate a dissolution profile.
  • Figure 5 displays the plot of the dissolution of Tramadol tablet over 45 minutes. This example illustrates the feasibility of applying the in-situ system to generate the dissolution profile in real time. This is one of the most important applications of the proposed system for immediate release products, because FDA is increasingly requiring for such information.
  • the present invention can also be practiced using techniques and equipment generally described below
  • Electrochemical Detection System Quantitative electrochemical techniques as described by Cooper, J.C and Hall, E.A., Journal of Biomedical Engineering, (1988) 10, 210-219, including Differential Pulse Voltametry, Current Polarography and Osteryoung Square Wave Voltametry can be applied to monitor analytes dissolved in dissolution media
  • Electrochemical Detection System Quantitative electrochemical techniques as described by Cooper, J.C and Hall, E.A., Journal of Biomedical Engineering, (1988) 10, 210-219, including Differential Pulse Voltametry, Current Polarography and Osteryoung Square Wave Voltametry can be applied to monitor analytes dissolved in dissolution media
  • a biosensor in which a transducer is coupled to a biological element, can be used to quantitate a change in concentration of target analyte as described by Buerk, D.G in Biosensors Theory and Applications, Technomic Publishing, (1993), inco ⁇ orated herein by reference
  • the biological element can be an enzyme or enzyme system, antigen/antibody, lectin, protein, organelle, cell, or tissue, though enzymes and antigen/antibodies predominate as biological elements of choice, as described by Lowe et al in Journal of Chromatography (1990) 510,
  • the biological element is generally immobilized on a support as described by Coulet et al in Journal of Pharmaceutical and Biomedical Engineering (1988) 10, 210-219, inco ⁇ orated herein by reference
  • the transducer may be optic or fiber optic (measuring most commonly changes in abso ⁇ tion or luminescence), or electrochemical Superior specificity is one of the advantages of biosensors Such sensors will be used in our systems.
  • the dissolution vessel temperature in the in-situ system can be controlled either by a water bath in which vessels are submerged to maintain appropriate temperature or a bathless configuration, in which each vessel is surrounded by a heating element.
  • the latter configuration reduces the size of the equipment and consequently the bench space, as well as minimizing maintenance. It also allows temperature control of each vessel and also helps to minimize vibration associated with thermocirculation. This is commercially available from Distek, Inc.
  • FIG 11 shows seven vessels in which six can be the samples and one for the dissolution medium itself, or a placebo formula for baseline correction.
  • the actual system can be any number of vessels in the system.
  • This system design may be inco ⁇ orated with a "closed vessel” design as shown in Figure 12. The major advantage of this closed design is to minimize loss of dissolution media.
  • Figure 13 shows a UV probe in shaft embodiment of the present invention.

Abstract

The present invention relates to an improvement in a detection system used for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a singular dissolution vessel or multiple dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft and a probe placed within the mixing shaft or outside the individual dissolution vessels, the probe capable of measuring the dissolution characteristics using UV, IR, near-IR, fluorescence, electrochemical, and Raman spectroscopy techniques. The present invention also relates to a method for predicting the dissolution curve provided by a controlled release pharmaceutical dosage form comprising taking continuous measurements of the amount of drug released from a dosage form for a portion of the time over which the drug is expected to be released and predicting the remainder of the dissolution curve based on the values obtained.

Description

IMPROVEMENTS IN DETECTION SYSTEMS AND METHODS FOR
PREDICTING THE DISSOLUTION CURVE OF A DRUG FROM A
PHARMACEUTICAL DOSAGE FORM
BACKGROUND OF THE INVENTION
Dissolution testing is required for all solid oral pharmaceutical solid dosage form forms in which absorption of the drug is necessary for the product to exert the desired therapeutic effect The U.S. Pharmacopeia (USP) is one well-known standard source of information which provides for dissolution and drug release testing in the majority of monographs for such dosage forms Exceptions are for tablets meeting a requirement for completeness of solution or for rapid ( 10 to 15 minutes) disintegration for soluble or radiolabled drugs. The apparatus and procedure conform to the requirements and specificaitons given, e.g., USP 23rd edition Chapter 711 (Dissolution) pages 1791-1793 Dissolution testing serves as a measure of quality control, stability and uniformity as well as a means by which to correlate in-vitro with in-vivo drug release characteristics
Current USP dissolution methods most commonly employ a temperature programmable water bath, maintained at about 37 °C, in which sample vessels are submerged These vessels contain a predetermined volume of a dissolution media and a means to agitate the contents of the vessel. This may be accomplished by means of a rotating basket attached to a shaft or with a paddle which is also attached to a shaft, both means generally described in USP 23 rd edition Chapter 71 1 (Dissolution) pages 1791-1793 The solid dosage form is placed into the media filled vessel at time zero and specific vessel temperature and mixing speeds are maintained. At fixed time intervals (e.g. 2, 4, 8 hours, etc.) a small aliquot of sample is taken from each vessel, usually by a multi channeled pumping system, and transported to either a cuvette or a sample vial for subsequent spectrophotometric or high pressure liquid chromatography (HPLC) analysis, respectively. Plotting percentage dissolution of a solid dosage form through time results in a dissolution profile.
Of the two methods discussed above, the HPLC method is usually favored over the spectrophotometric method. While HPLC dissolution offers the advantage of specificity, acceptable accuracy, precision and sensitivity, the disadvantage of the status quo rather lies with the inherent burden of creating, manipulating, and storing voluminous numbers of sequence and data files. The cost of HPLC, columns, mobile phases, and the waste solvent disposal, etc., is substantial and the limited number of data points that can be determined may result in a less than ideal representation of the release profile of a solid dosage form over time. Furthermore,
HPLC analysis is a sequential time consuming process. In general, a typical 24 hour dissolution requires up to 60 hours to generate a dissolution profile.
Because of the aforementioned disadvantages of currently available systems, an in-situ dissolution method is desirable.
SUMMARY OF THE INVENTION The present invention relates to an improvement in a detection system used for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a singular dissolution vessel or multiple dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft and a probe placed within the mixing shaft or outside the individual dissolution vr^sels, the probe capable of measuring the dissolution characteristics using UV "• , aear- IR, fluorescence, electrochemical, nuclear magnetic resonance (NMR), and Raman spectroscopy techniques.
The present invention also relates to a method for predicting the dissolution curve provided by a controlled release pharmaceutical dosage form comprising taking continuous measurements of the amount of drug released from a dosage form for a portion of the time over which the drug is expected to be released and predicting the remainder of the dissolution curve based on the values obtained.
The present invention relates to a in-situ dissolution methods to evaluate and study the dissolution characteristics of drug formulations Such methods utilize systems that include fiber optics, ultraviolet spectroscopy, fluorescence spectroscopy, NMR and the like.
The present invention specifically relates to detection systems for measuring dissolution characteristics of pharmaceutical dosage forms using ultraviolet, LR, near-LR, and Raman spectroscopy techniques as well as electrochemical techniques such as polarography, and NMR. These and other aspects of the present invention can be followed by one skilled in the art by reading the detailed description and the methods provided by the instant invention.
BRIEF DESCRIPTION OF THE DRAWINGS The following drawings are illustrative of the invention and are not meant to limit the scope of the claims.
Figure 1 shows the UV-vis spectra of tramadol standard solutions at four different concentrations;
Figure 2 the linearity plot of tramadol HCl solutions of Example 1 ; Figure 3 is a graphical representation of repeated UV-vis scans at 30 minute intervals over 25 hours for a tramadol HCl 200mg once-a-day tablet of Example 2,
Figure 4 shows the plot of the average dissolution of three tramadol HCl once-a-day tablets of Example 2 and the results from the HPLC method;
Figure 5 displays the plot of the dissolution of a tramadol tablet of Example 3 over 45 minutes;
Figure 6 is a graph of the dissolution profile of a tramadol controlled release tablet, using the average dissolution results from table 1, by using TableCurve 2D program, using the best fit equation (as described in Example 4);
Figure 7 is a graph showing the dissolution profile of a tramadol controlled release tablet as described in Example 4 obtained from 12 hour sampling data, at 1 hour intervals, using the to the best fit equation (as described in Example 4),
Figure 8 is a graph showing the dissolution profile of a tramadol controlled release tablet obtained from 16 hour data, taken at 1 hour interval,using the best fitted equation (as described in Example 4),
Figure 9 is a graph of a dissolution profile of a tramadol controlled release tablet, when 16 hour data, generated at every half hour, are used to find the best fit curve (as described in Example 4);
Figure 10 shows a plot comparison of dissolution data obtained from both a fiber optics v. HPLC methods (as described in Example 6);
Figure 1 1 depicts a preferred configuration of the present invention, Figure 12 depicts a closed-vessel embodiment of the invention; and Figure 13 depicts a UV probe in shaft embodiment of the invention.
DETAILED DESCRIPTION
One aspect of the present invention is related to an improvement in a detection system for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a dissolution vessel containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft having a probe contained within, the probe capable of measuring the release of the drug using fluorescence, ultraviolet (UV), Infrared (IR), near-Infrared (IR), electrochemical, and Raman spectroscopy techniques.
The present invention further provides an improvement wherein the probe utilizes ultraviolet spectroscopy techniques, electrochemical techniques, Infrared
(LR), near-Infrared (IR) or Raman spectroscopy techniques
Another aspect of the present invention provides a method for predicting the dissolution curve provided by a controlled release pharmaceutical dosage form comprising taking continuous measurements of the amount of drug released from a dosage form for a portion of the time over which the drug is expected to be released and predicting the remainder of the dissolution curve based on the values obtained.
A method according to the present invention utilizes a detection system comprising a singular dissolution vessel or multiple dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement in the detection system comprising a mixing shaft and a probe placed within the mixing shaft or outside the individual dissolution vessels, the probe capable of measuring the dissolution characteristics using UV, IR, near-IR, fluorescence, electrochemical, and Raman spectroscopy techniques.
Yet another aspect of the present invention relates to an improvement in a detection system for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a plurality of dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft having a probe contained within, the probe capable of measuring the release of the drug using fluorescence, ultraviolet, Infrared, near-Infrared, electrochemical, and Raman spectroscopy techniques. It is further provided that this aspect of the present invention may utilize at least two vessels to optionally hold a dissolution medium or a placebo formulation for baseline correction.
The present invention also provides an improvement in a detection system for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a singular dissolution vessel or multiple dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft and a probe placed outside the individual dissolution vessels, the probe capable of measuring the dissolution characteristics using UV, IR, near-LR, fluorescence, electrochemical, and Raman spectroscopy techniques.
It is further provided that at least two vessels in the inventive system optionally hold a dissolution medium or a placebo formulation for baseline correction. The present invention particularly relates to detection systems for measuring dissolution characteristics of pharmaceutical dosage forms using ultraviolet, LR, near-LR, and Raman spectroscopy techniques as well as electrochemical techniques such as polarography.
The present invention also relates to a dissolution apparatus for determining a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, the apparatus including a detector for quantifying one or more physical and/or chemical properties of the therapeutically active agent, the detector operatively associated with the dissolution medium for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent; and a data processor for continually processing the generated data for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form. The present invention also relates to a method for determining a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, including the steps of continually generating physical and/or chemical data characteristic of the therapeutically active agent by operatively associating a detector with the dissolution medium for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent; and continually processing the generated data with a data processor for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form.
Another preferred embodiment of the invention relates to a dissolution arrangement for measuring in- vitro release of an active agent from a dosage form containing the active agent, including a plurality of vessels, each of the vessels containing a dissolution media and a dosage form containing an active agent to be measured, a fiberoptic probe associated with each of the vessels, each of the fiberoptic probes including a detector which simultaneously and continuously measures the concentration of active agent in the dissolution media, and a data processor connected to the fiberoptic probes, the data processor continually processing information received from the probes concerning the concentration of the drug to obtain a dissolution profile of the dosage form.
In a more preferred embodiment, the dissolution arrangement further includes utilizing the data processor to predict future concentrations of the active agent. In other preferred embodiments, further include utilizing the data processor to predict the entire dissolution profile of the active agent after at least 50 percent of the entire desired dissolution time frame has elapsed. For example, the dissolution arrangement further comprises utilizing the data processor to predict a 24 hour dissolution profile of the active agent after 16 hours of dissolution time has elapsed. The term releasable quantity is defined, for purposes of the present invention, as the maximum amount of therapeutically active agent that can be released from a pharmaceutical dosage form during the dissolution testing time period. It will be understood by the skilled artisan that the releasable amount may be less than 100% of the total amount of agent contained in the pharmaceutical dosage form. The dissolution testing time period is preferably at least one hour, and in certain embodiments is 8-24 hours or longer, e.g., 48, 72 or 96 hours.
The term physical and/or chemical properties, for purposes of the present invention, is a physical and or chemical property that is characteristic of a particular therapeutically active agent. A non-limiting list of physical and/or chemical properties include ultraviolet absoφtion or radiation spectra; infrared absoφtion or radiation spectra; , β or gamma radiation; electron states; polarity; magnetic resonance; concentration electro-chemical properties and the like. The physical and/or chemical properties of a agent are any property characteristics of a agent or group of agents that can be used to detect, e.g. the presence, absence, quantity, physical state or chemical state of that agent.
For puφoses of the present invention, the term agent is defined as any chemical or physical entity or combination of entities, particles or organisms either that are detectable by a detector. An exemplary list of agents include chemicals, therapeutically active agents, radiation particles (e.g., β-particles); microbes such as bacteria, viruses, individual cells from a multi-cellular organism (e.g., blood cells); and the like.
A detector is defined for puφoses of the present invention as any device that detects a physical and/or chemical property of a agent and generate data regarding about the physio-chemical property. Examples of detectors include UV- spectrophotometers, Geiger counters, fluorscopic devices and the like. The physical and/or chemical property detected by the detector and the type of data generated by the detector are not critical to the present invention.
The term "operatively associated" is defined for puφoses of the present invention as positioning the detector in proximity to the vessel containing the subject agent such that the detector can quantify the desired physical and/or chemical data characteristic of the agent, and transmit the data to a data processor.
DETAiILED DESCRIPTION OF THE INVENTION Th? dissolution apparatus of the present invention is particularly useful for determin .g a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, the apparatus including a detector for generating physical and/or chemical data characteristic of the therapeutically active agent, said detector operatively associated with the dissolution medium for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent, and a data processor for continually processing the generated data for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form.
The detector may be any detector known in the art that generates physical and/or chemical data of the test agent, e.g. a uv spectrophotometer. Preferably, the detector has a probe communicably attached thereto. In preferred embodiments, there is at least one detector per sample vessel; i.e. the ratio of detectors to sample vessels is at least 1 : 1. In other words, for each sample to be analyzed, there is a corresponding detector capable of continuously generating physical and/or chemical data characteristic of the agent to be analyzed.
The data processor may be any device capable of continuously processing the data generated by the detector. In preferred embodiments, the data processor is a computer. The data generated by the detector is preferably stored and/or analyzed by the computer. In a particularly preferred embodiment, the data collector is a computer that has data processing software, e.g. Microsoft Excel 5.0 or Tablecurve. The data generated by the detector is processed by the software and reorganized into a preferred form, e.g. as a graph or a table. The data is preferably continuously processed by the software as it is received from the detector.
In an alternative embodiment, the apparatus further comprises a shaft. The shaft has at least one aperature therein, which aperature allows the detector to detect the necessary physical and or chemical properties of the subject agent and generate the required physical and/or chemical data. The size and position of the opening along the shaft will depend ona variety of factors, including, but not limited to, the type of detector used and the physical and/or chemical property to be detected.
In preferred embodiments, the shaft has an orifice therein for receiving the detector When the shaft is received by the connector, it is preferable that the detector is attached to the shaft In preferred embodiments, the detector is attached to the shaft by any attachment means known in the art, including, but not limited to, welds, adhesives, soldering, screws, friction, and the like In a preferred embodiment, the detector is permanently attached to the shaft by, for example, soldering the detector to the shaft In other preferred embodiments, the detector is rotatably attached to the shaft in a manner such that when the detector is receivedin the shaft, the shaft can freely rotate about the detector, allowing the shaft to perform other functions independent of the detector For example, a paddle or basket may then be affixed to at least one end of the shaft such when the shaft is rotated, the paddle or basket also rotates to provide, e g , agitation when the paddle or basket is contacted with an external environment, e g dissolution media
In certain preferred embodiments of the present invention, the detector measures the concentration of the agent, e g , therapeutically active agent, in the media surrounding the dosage form, e g , simulated gastic fluid, or simulated intestinal fluid By measuring the concentration of the agent in the surrounding media, the amount of agent released from the dosage form can be calculated
The present invention also relates to a method for determining a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, including the steps of continually generating physical and/or chemical data characteristic of the therapeutically active agent by oneratively associating a detector with the dissolution medium for at least the time per IOJ required for the dosage form to release the maximum releasable quantity of therapeutically active agent, and continually processing the generated data with a data processor for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form In a preferred embodiment, the invention includes three components, a conventional dissolution apparatus, a UV detection unit and a Pentium computer running Windows 95 and Excel 5 0 software The conventional dissolution apparatus, a Distek 5100 bathless unit (or equivalent unit), is interfaced to a UV radiation source with fiber optic transmission dip probes, and a series of charge coupled detector (CCD) spectrometers which are internalized in the Pentium computer The computer is configured with Windows 95 and Excel 5 0 for operation of the system and connected to a Novell file server for data storage Within the Excel software is a template used to run the system A Dissolution Apparatus is used where vessels are rapidly heated with a thin sheath of electrically resistant material (Distek Premiere 5100 Bathless Unit) A thermocouple present in the shaft of each paddle constantly monitors the temperature of each vessel The unit uses vessel covers which have been tooled to tightly hold fiber optic probes at specified heights Alternatively, a dissolution apparatus utilizing a water bath may be used in place of the bathless unit A fiber optic dip probe, used for transmission, is interfaced via a sheathed fiber to a deuterium lamp to provide the UV radiation source for the analysis The dip probe is connected to a CCD spectrometer Radiation returns from the probe to the CCD spectrometer where it is analyzed and quantitated Preferably, the internal core of the fiber consists of fused silica, which allows UV radiation to be efficiently propagated UV radiation is transmitted from the source lamp through the fiber (which extends into the probe) and through a quartz lens seated directly above the flow cell UV radiation travels through the flow cell and is reflected off a mirror positioned at the terminal end of the probe The radiation then travels back through the flow cell and quartz lens It is directed into a second fiber where it travels to the spectrometer for analysis Quantitation of the drug substance is accomplished by determining the change in intensity ofUV radiation as it is transmitted through the flow cell The spectrometer itself is comprised of a closed optics bench mounted on a printed circuit board which is situated in the computer system Upon entering the spectrometer, UV radiation is propagated through an optical slit and onto a grating via a mirror The radiation is then reflected off a second mirror and onto a charge coupled detector Each fiber optic probe is interfaced to its own spectrometer using universal SMA fittings.The CCD spectrometer is calibrated for both wavelength accuracy and for quantitative accuracy and precision A second order polynomial equation is used to determine wavelength accuracy This equation matches each wavelength of light hitting the CCD with a discrete pixel on the array The control unit is comprised of a Pentium class computer interfaced to the Novell network and fitted with several CCD spectrometers The spectrometers are entirely controlled through a Microsoft Excel 5 0 template consisting of multiple sheets Excel communicates with the spectrometers via a device driver library The system parameters can be adjusted by accessing the data acquisition parameters within the Excel worksheet The parameters for spectrometer control can be set by using either the mouse or keystrokes The applicable information such as lot numbers and package types are manually entered into the spreadsheet before the test begins A worksheet presenting real-time data can then be accessed throughout the dissolution. As the data is collected it is stored on the network
Generally, the agent is dissolved in the solvent, however, for puφoses of the present invention, the agent may be dispersed or suspended throughout the solvent in a solid or semi-solid media Thus, for purposes of the present invention, the agent need not be dissolved in the solvent, but may, instead, provide a dispersion or suspension medium for the agent
In a preferred embodiment of the invention the device comprises a detector for monitoring chemical and/or physical properties of an agent, wherein the detector is mounted to a shaft having a hollow portion capable of receiving said detection means, said shaft having an aperature therein that allows said detection means to communicate with said external environment when said detection means is received by said hollow portion The detection means may be permanently mounted to the shaft, or preferably removably mounted to the shaft to as allow a near infinate combination of shafts and detection means To facilitate the interchangeabiliy, the mount is preferably a universal mount which will allow an almost infinite combination of detection means and shafts In a preferred embodiment, the detection means is capable of aquiring data characteristic of a particular agent by a method selected from the group consisting of ultraviolet radiation, infrared radiation, nuclear magnetic resonance, Raman sprctroscopy, electrochemical, and mixtures thereof, with ultraviolet radiation detection being particularly preferred In a particularly preferred embodiment, the shaft is rotatably attached to said detection means, such that the shaft is freely rotatabie around the peripheral edges of the detection means when the detection means is situated in the hollow portion of the shaft In this embodiment, the detection means may or may not be attached in a manner to allow the detection means to independently rotate about an axis within the hollow portion of the shaft, as desired
In other preferred embodiments of the invention, the device includes a data collecting means, e g a computer In particularly preferred embodiments, the computer is capable of operating data collection software which facilitates analysis or collection of the data generated by the detection means For example, the software may serve to merely store the data, or it may provide compariitive analysis to rfrence standards, produce graphic representations of the data (e g , dissolution vs time curves), or other assorted functions known in the art The software will preferably be capable of continuously receiving said data from said detection means, providing near-instantaneous access to the data derived from a given test The present invention is also directed to a method for continuously monitoring a agent in an external environment, e.g dissolution media, including the steps of collecting data characteristic to a particular agent in an external environment by positioning at an effective distance to the external environment a device for continually monitoring the agent in the external environment comprising a detection means for detecting a agent in an external environment mounted to a shaft having a hollow portion capable of receiving the detection means; the shaft having an aperature that allows the detection means to communicate with the external environment; and continuously retreiving data obtained from the detection means during the time interval that the device is exposed to the external environment.
It should be understood that the electrochemical techniques used in the present invention optionally include biosensors, in which a transducer is coupled to a biological element, to quantitate a change in concentration of target analyte(s)
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
The following examples illustrate various aspects of the present invention They are not to be construed to limit the claims in any manner whatsoever.
I METHODS
I 1 Material
The new dissolution system has been applied to study the dissolution characteristics of pharmaceutical dosage forms, for example analgesic products, such as Tramadol HCI QD Tablets and Hydromoφhone Capsules
1.2 Apparatus
1.2.1 Fiber Optics Ultraviolet Spectroscopy
The Ocean Optics Inc. PC Plug-In Fiber Optic Miniature Spectrometer is used with an ultraviolet probe as the method of detection. The probe is coupled to a LS-1 deuterium light source and detection is conducted using a SI 000 spectrometer. Data is processed using SpectraScope and Microsoft Excel 5.0 software The detector is capable of scanning the entire UV and visible spectrum in under 2 seconds. Comparison with the current method for dissolution analysis of solid dosage forms was conducted.
A more powerful deuterium light source from Oriel Coφoration, Stratford, CT can also be used to replace the LS-1 deuterium lamp when higher light throughput is required. This light source also has the advantage of using a condensing lens to manipulate the quality of light hitting the fiber optic interface. A xenon arc lamp source from Oriel Coφoration may also be used for applications requiring increased sensitivity, such as Hydromorphone and Hydrocodone Controlled Release Products. In addition, a variable path length dip probe from CIC Photonics, Inc. of Albuquerque NM can be used for method development puφoses to determine optimal flow cell pathlength for a given drug product
1.2.2 Fluorescence Spectroscopy
Fluorescence studies were conducted on a Perkin Elmer model LS5 Luminescence Spectrometer. The excitation spectrum was obtained from 220 nm to 500 nm and the emission spectrum was taken from 300 to 800 nm.
1.2.3 Dissolution Apparatus
The dissolution bath was a Hansen Research model SR5 with type II (paddle) agitation. The bath temperature was maintained at 37 +/-0.5 degrees and solution was agitated at 100 rpm.
1.2.4 Software Modules
SpectraScope software from Ocean Optics and Microsoft Excel 5.0 was used for data collection. TableCurve 2D and Table curve 3D from Jandel Scientific Software was used to mathematically model tabulated data and predict experimental results. Disclosure generally describing the above software from Jandel Scientific Software is included under Annexure I, and the same is incoφorated herein by reference. EXAMPLES
Example 1
In-situ system using an Ultraviolet-visible spectrometer: Figure 1 shows the UV-vis spectra of Tramadol standard solutions at four different concentrations. Inspection of the spectra in Figure 1 reveals relatively noise free data with well defined spectral features The absorbance of Tramadol vs concentration at the maximum absorbtivity (272 nm) are shown in Table 1 below The correlation coefficient of the regression line is 0 999825 indicating a linear relationship between concentration and absorption The linearity plot is shown in Figure 2
Table 1. Linearity of Tramadol HCl
Tramadol HCI (mg/mL) Absorption at 272 nm
0 0112 0 058
0.0224 0.119
0.0447 0.246
0 112 0.625
0.224 1.21 1
Correlation Coefficient(r): 0.999825
Slope. 5.4292 y-intercept: 0.001935
This demonstrates the feasibility of using the fiber optical probe as a
spectrometer. Example 2
Dissolution of Tramadol 200 mg QD tablets:
Three Tramadol 200 mg QD tablets were placed in the dissolution medium to check its release rate over three different days by the in-situ system. The repeated UV-vis scans at 30 minute intervals over 25 hours for one of the tablets is shown in Figure 3. The dissolution data of these tablets are shown in Table 2. Table 2 also shows the average of the three and the dissolution results obtained from an existing, validated, HPLC method for comparison.
The table clearly demonstrates that the in-situ dissolution system gives results which are precise and correlate well to the current HPLC method. Figure 4 shows the plot of the average dissolution of three tablets and the results from the HPLC method.
Table 2 Dissolution of Tramadol QD 200 mg
UV Probe vs. HPLC
Figure imgf000020_0001
EXAMPLE 3
Dissolution profiles generated in real time:
A Tramadol 200 mg QD tablet was placed in the in-situ dissolution system and the amount of Tramadol released monitored in real time. This was obtained by a process called history channel evaluation in which the UV-vis scans of the analyte are acquired in about every 2.5 seconds. The absoφtion at a pre-selected wavelength is plotted against time to generate a dissolution profile. Figure 5 displays the plot of the dissolution of Tramadol tablet over 45 minutes. This example illustrates the feasibility of applying the in-situ system to generate the dissolution profile in real time. This is one of the most important applications of the proposed system for immediate release products, because FDA is increasingly requiring for such information.
Example 4 Prediction of the dissolution profile by curve fitting:
Controlled-release pharmaceutical dosage forms in general follow certain release patterns controlled by physical and chemical properties of the matrix. These release patterns can be predicted mathematically.
For example, using the average dissolution results from table 1, by using TableCurve 2D program, one can fit the dissolution data into the equation as shown in Figure 6. On top of figure 6, it displays the best-fitted equation for the data
If one uses the 12 hour data, at 1 hour interval, and fits them to the best fit equation the dissolution profile generate would be as the one shown in Figure 7. When 16 hour data, taken at 1 hour interval, are taken to find the best fitted equation, it produced the curve ofFigure 8. Note that this curve is closer so that actual experimental data than that generated in Figure 7.
Finally, when 16 hour data, generated at every half hour, are used to find the best fit curve, the equation produced, as shown in Figure 9, is exactly what one would get in 24 hour experiment, as shown in Figure 6. This example clearly demonstrates that it is possible to shorten the experiment by taking more frequent data in a short time and predict the remaining results with mathematical modeling The in-situ system can work to this puφose because it generates instant data in real time It is therefore able to give a predicted result to formulators in shorter time
EXAMPLE 5
Detection of drug products with low dosage strength Dissolution testing by conventional spectroscopic methods for low dosage strength products, such as hydromoφhone HCl Controlled Release 12 mg capsules, may be difficult due to low concentrations of active drug in the dissolution vessel Using the combination of a high intensity lamp and a probe tip with a relatively long pathlength (20mm), a 24 hour dissolution profile has been generated for hydromoφhone HCl 12 mg which is comparable to that obtained from a validated HPLC method Results are displayed in Table 3 and Figure 10
Table 3 Data of Hydromorphone HCl 12 mg Capsule Dissolution
Fiber Optics v. HPLC
D
% Hydromorphone HCI Dissolved
Hour Fiber HPLC Hour Fiber HPLC Hour Fiber HPLC Hour Fiber HPLC
0 1 7 0
0 17 276 6 17 389 122 62 18 17 79
0.33 55 633 392 123 625 1833 797
0.5 7 18 65 40 125 632 185 798
067 867 667 406 127 637 1867 302
083 10 683 41 5 128 644 1883 805
1 11 4 11.3 7 422 13 649 19 808
1 17 125 7 17 43 132 655 19 17 81 2
1 33 137 733 435 133 66 1933 81 6
1 5 148 75 442 135 664 195 82
1 67 16 767 449 137 67 1967 82 1
1 83 17 2 783 457 138 675 1983 826
2 18 1 18 2 8 46 3 56 14 68 20 829
2 17 193 8 17 468 142 68 5 20 17 829
233 20 1 833 472 143 69 2033 835
25 21 2 85 484 145 696 205 836
267 ?1 8 867 492 147 70r2 2067 84 283 228 883 496 148 707 2083 841
3 23 8 9 503 15 71 3 21 844
3 17 247 9 17 51 1 152 71 5 21 17 846
333 254 933 51 8 153 72 1 21 33 849
35 26 1 95 523 155 723 21 5 852
367 269 9 67 533 157 729 21 67 854
383 28 9 83 537 158 734 21 83 855
4 287 31 8 10 544 16 739 22 857
4 17 297 102 55 1 162 74.3 22 17 862
433 305 103 559 163 748 2233 863
45 31 4 105 562 165 753 225 866
467 322 107 57 1 167 756 2267 87 1
483 328 108 573 168 758 2283 87 1
5 337 11 58 17 76 5 23 87 1
5 17 344 11 2 586 172 769 23 17 873
5 33 35 3 11 3 59 1 173 774 2333 875
55 36 11 5 597 175 779 23 5 877
567 36 7 11 7 602 177 779 2367 879
5 B3 37 5 11 8 609 178 784 2383 879
6 38 3 12 61 4 738 18 789 90 3 24 883 99 8
The present invention can also be practiced using techniques and equipment generally described below
II 1 Detection Systems
II 1 1 Other Fiber Optic Systems
Fluorescence as described in the publication by Glazier, S A et al , Analytical Letters (1995) 28, 2607-24, Infrared techniques as described by Krska, R et al in Appl Phys Lett (1993) 63, 1868-70, Near IR and Raman techniques as described by Cram, D J and Hammond, G S , Organic Chemistry, McGraw-Hill (1959), all of which are hereby incoφorated by reference, either with a grating or interferometer system, are potentially powerful techniques useful for dissolution testing The techniques can be used with fiber optic spectrometers similar to that of UV These technologies will be incoφorated into the in-situ system similar to that for UV detection
II 1 2 Electrochemical Detection System Quantitative electrochemical techniques as described by Cooper, J.C and Hall, E.A., Journal of Biomedical Engineering, (1988) 10, 210-219, including Differential Pulse Voltametry, Current Polarography and Osteryoung Square Wave Voltametry can be applied to monitor analytes dissolved in dissolution media These techniques will be used in the in-situ system with different electrode designs, such as platinum or glassy carbon electrodes, for evaluation of different products
II.1.3. Biosensor
A biosensor, in which a transducer is coupled to a biological element, can be used to quantitate a change in concentration of target analyte as described by Buerk, D.G in Biosensors Theory and Applications, Technomic Publishing, (1993), incoφorated herein by reference The biological element can be an enzyme or enzyme system, antigen/antibody, lectin, protein, organelle, cell, or tissue, though enzymes and antigen/antibodies predominate as biological elements of choice, as described by Lowe et al in Journal of Chromatography (1990) 510,
347-354, incoφorated herein by reference The biological element is generally immobilized on a support as described by Coulet et al in Journal of Pharmaceutical and Biomedical Engineering (1988) 10, 210-219, incoφorated herein by reference The transducer may be optic or fiber optic (measuring most commonly changes in absoφtion or luminescence), or electrochemical Superior specificity is one of the advantages of biosensors Such sensors will be used in our systems.
11.2 Probe Design
II.2.1 Probe in Shaft
A distinct configuration, not having been explored in the present art, is the design of a probe which is totally contained in the mixing shaft of each dissolution vessel. The advantage of this system is the absence of flow aberration, since an additional probe need not be submerged in the dissolution media II.2.2. Probe Outside of Vessel
An alternate design to the above places probes outside the dissolution vessels. Near IR which has limited interference from the container, such as glass vessel, is good candidate of such detection probe.
III. Temperature Control
The dissolution vessel temperature in the in-situ system can be controlled either by a water bath in which vessels are submerged to maintain appropriate temperature or a bathless configuration, in which each vessel is surrounded by a heating element. The latter configuration reduces the size of the equipment and consequently the bench space, as well as minimizing maintenance. It also allows temperature control of each vessel and also helps to minimize vibration associated with thermocirculation. This is commercially available from Distek, Inc.
IV. System Design
With the above configurations, a system is therefore designed as in Figure 11. The figure shows seven vessels in which six can be the samples and one for the dissolution medium itself, or a placebo formula for baseline correction. The actual system, of course, can be any number of vessels in the system. This system design may be incoφorated with a "closed vessel" design as shown in Figure 12. The major advantage of this closed design is to minimize loss of dissolution media. Figure 13 shows a UV probe in shaft embodiment of the present invention.
All of the above-identified references are hereby incoφorated by reference The examples provided above are not meant to be exclusive. Many other variations of the present invention will be readily apparent to those skilled in the art, and are contemplated to be encompassed within the appended claims.

Claims

We claim:
1. In a detection system for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a dissolution vessel containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising utilizing a mixing shaft having a probe contained within, the probe capable of measuring the release of the drug using fluorescence, ultraviolet, Infrared, near-Infrared, electrochemical, NMR and Raman spectroscopy techniques.
2. A method for predicting the dissolution curve provided by a controlled release pharmaceutical dosage form comprising taking continuous measurements of the amount of drug released from a dosage form for a time period over which the drug is expected to be released and predicting the remainder of the dissolution curve based on the values obtained.
3. In a detection system for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a plurality of dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising a mixing shaft having a probe contained within, the probe capable of measuring the release of the drug using fluorescence, ultraviolet, Infrared, near-Infrared, electrochemical, and Raman spectroscopy techniques.
4. The system of claim 3 further comprising at least two vessels containing dissolution medium or a placebo formulation for baseline correction.
5. In a detection system arrangement for continuously measuring the release of a drug from a pharmaceutical dosage form comprising a singular dissolution vessel or multiple dissolution vessels containing a dissolution medium and a measuring device for detecting the amount of drug released at a given time, the improvement comprising arranging a mixing shaft and a probe outside the individual dissolution vessels, the probe capable of measuring the dissolution characteristics using UV, IR, near-IR, fluorescence, electrochemical, and Raman spectroscopy techniques.
6. A dissolution arrangement for measuring in-vitro release of an active agent from a dosage form containing the active agent, comprising a plurality of vessels, each of said vessels containing a dissolution media and a dosage form containing an active agent to be measured, a fiberoptic probe associated with each of said vessels, each of said fiberoptic probes including a detector which simultaneously and continuously measures the concentration of active agent in the dissolution media, and a data processor connected to said fiberoptic probes, said data processor continually processing information received from said probes concerning the concentration of said drug to obtain a dissolution profile of the dosage form.
7. The dissolution arrangement of claim 6, further comprising utilizing said data processor to predict future concentrations of said active agent.
8. The dissolution arrangement of claim 6, further comprising utilizing said data processor to predict the entire dissolution profile of said active agent after at least 50 percent of the entire desired dissolution time frame has elapsed.
9. The dissolution arrangement of claim 6, further comprising utilizing said data processor to predict a 24 hour dissolution profile of said active agent after 16 hours of dissolution time has elapsed.
10. A dissolution apparatus for determining a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, comprising: a detector for generating physical and/or chemical data characteristic of the therapeutically active agent, said detector operatively associated with the dissolution medium for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent; and a data processor for continually processing said generated data for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form.
11. A method for determining a dissolution profile of a pharmaceutical dosage form containing a releasable quantity of a therapeutically active agent wherein the dosage form is immersed in a dissolution medium contained in a vessel, comprising: continually generating physical and/or chemical data characteristic of the therapeutically active agent by operatively associating a detector with the dissolution medium for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent; and continually processing said generated data with a data processor for at least the time period required for the dosage form to release the maximum releasable quantity of therapeutically active agent to obtain a dissolution profile of the dosage form.
PCT/US1997/011791 1996-06-04 1997-06-04 Improvements in detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form WO1997046860A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
AU43262/97A AU729814B2 (en) 1996-06-04 1997-06-04 Improvements in detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form
CA002257106A CA2257106C (en) 1996-06-04 1997-06-04 Improvements in detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form
EP97941329A EP0938657A4 (en) 1996-06-04 1997-06-04 Improvements in detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form
JP1998500947A JP3380864B6 (en) 1996-06-04 1997-06-04 Improvements in detection systems and methods for predicting dissolution curves of drugs from pharmaceutical formulations
US08/915,785 US6174497B1 (en) 1997-06-04 1997-08-21 Detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form
US09/707,498 US6558957B1 (en) 1996-06-04 2000-11-07 Detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US1894496P 1996-06-04 1996-06-04
US60/018,944 1996-06-04

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US08/915,785 Continuation US6174497B1 (en) 1996-06-04 1997-08-21 Detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form

Publications (3)

Publication Number Publication Date
WO1997046860A2 true WO1997046860A2 (en) 1997-12-11
WO1997046860A3 WO1997046860A3 (en) 1998-02-19
WO1997046860A9 WO1997046860A9 (en) 1998-04-23

Family

ID=21790557

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/011791 WO1997046860A2 (en) 1996-06-04 1997-06-04 Improvements in detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form

Country Status (4)

Country Link
EP (1) EP0938657A4 (en)
AU (1) AU729814B2 (en)
CA (1) CA2257106C (en)
WO (1) WO1997046860A2 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1210583A1 (en) * 1999-08-30 2002-06-05 Euro-Celtique S.A. In situ methods for measuring the release of a substance from a dosage form
EP1308724A2 (en) * 2001-11-05 2003-05-07 Rohm And Haas Company Buccal dissolution test for pharmaceutically active substrates
US6595678B2 (en) * 2000-02-17 2003-07-22 Astrazeneca Ab Mixing apparatus
WO2016030583A1 (en) * 2014-08-29 2016-03-03 Svanbäck Sami Method and system for determining dissolution properties of matter
US9884025B2 (en) 2000-11-15 2018-02-06 Aptalis Pharma S.R.L. Microspheres of pancreatic enzymes with high stability and production method thereof
US9976171B2 (en) 2011-08-08 2018-05-22 Allergan Pharmaceuticals International Limited Method for dissolution testing of solid compositions containing digestive enzymes
US10087493B2 (en) 2008-03-07 2018-10-02 Aptalis Pharma Canada Ulc Method for detecting infectious parvovirus in pharmaceutical preparations
US10184121B2 (en) 2013-06-28 2019-01-22 Allergan Pharmaceuticals International Limited Methods for removing viral contaminants from pancreatic extracts
US10206882B2 (en) 2007-02-20 2019-02-19 Allergan Pharmaceuticals International Limited Stable digestive enzyme compositions
US10993996B2 (en) 2013-08-09 2021-05-04 Allergan Pharmaceuticals International Limited Digestive enzyme composition suitable for enteral administration
US11119041B2 (en) 2016-03-21 2021-09-14 Research Center Pharmaceutical Engineering Gmbh Monitoring dissolution of a dosage form in progress during dissolution by low coherence interferometry
US11364205B2 (en) 2010-10-01 2022-06-21 Societe Des Produits Nestle S.A. Stable low digestive enzyme content formulation
CN117783459A (en) * 2024-02-28 2024-03-29 沈阳科惠生物医药科技有限公司 Drug dissolution curve determination method and system

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4335438A (en) * 1980-04-17 1982-06-15 Smolen Victor F Method and apparatus for automatic dissolution testing of products
US5133892A (en) * 1990-10-17 1992-07-28 Lever Brothers Company, Division Of Conopco, Inc. Machine dishwashing detergent tablets
US5142920A (en) * 1989-12-20 1992-09-01 Prolabo Dissolution cell and apparatus for determining solids-dissolving kinetics
US5589649A (en) * 1994-02-25 1996-12-31 Distek, Inc. Dissolution testing apparatus

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4335438A (en) * 1980-04-17 1982-06-15 Smolen Victor F Method and apparatus for automatic dissolution testing of products
US5142920A (en) * 1989-12-20 1992-09-01 Prolabo Dissolution cell and apparatus for determining solids-dissolving kinetics
US5133892A (en) * 1990-10-17 1992-07-28 Lever Brothers Company, Division Of Conopco, Inc. Machine dishwashing detergent tablets
US5589649A (en) * 1994-02-25 1996-12-31 Distek, Inc. Dissolution testing apparatus

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP0938657A2 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1210583A4 (en) * 1999-08-30 2004-08-11 Delphian Technology Inc In situ methods for measuring the release of a substance from a dosage form
EP1210583A1 (en) * 1999-08-30 2002-06-05 Euro-Celtique S.A. In situ methods for measuring the release of a substance from a dosage form
US6595678B2 (en) * 2000-02-17 2003-07-22 Astrazeneca Ab Mixing apparatus
US7144147B2 (en) * 2000-02-17 2006-12-05 Astrazeneca Ab Mixing apparatus
US9884025B2 (en) 2000-11-15 2018-02-06 Aptalis Pharma S.R.L. Microspheres of pancreatic enzymes with high stability and production method thereof
KR100954393B1 (en) * 2001-11-05 2010-04-26 롬 앤드 하스 캄파니 Buccal Dissolution of Active Substances
EP1308724A2 (en) * 2001-11-05 2003-05-07 Rohm And Haas Company Buccal dissolution test for pharmaceutically active substrates
EP1308724A3 (en) * 2001-11-05 2004-06-23 Rohm And Haas Company Buccal dissolution test for pharmaceutically active substrates
US10206882B2 (en) 2007-02-20 2019-02-19 Allergan Pharmaceuticals International Limited Stable digestive enzyme compositions
US10087493B2 (en) 2008-03-07 2018-10-02 Aptalis Pharma Canada Ulc Method for detecting infectious parvovirus in pharmaceutical preparations
US11364205B2 (en) 2010-10-01 2022-06-21 Societe Des Produits Nestle S.A. Stable low digestive enzyme content formulation
US9976171B2 (en) 2011-08-08 2018-05-22 Allergan Pharmaceuticals International Limited Method for dissolution testing of solid compositions containing digestive enzymes
US10184121B2 (en) 2013-06-28 2019-01-22 Allergan Pharmaceuticals International Limited Methods for removing viral contaminants from pancreatic extracts
US10993996B2 (en) 2013-08-09 2021-05-04 Allergan Pharmaceuticals International Limited Digestive enzyme composition suitable for enteral administration
US10520411B2 (en) 2014-08-29 2019-12-31 The Solubility Company Oy Method and system for determining dissolution properties of matter
WO2016030583A1 (en) * 2014-08-29 2016-03-03 Svanbäck Sami Method and system for determining dissolution properties of matter
US11119041B2 (en) 2016-03-21 2021-09-14 Research Center Pharmaceutical Engineering Gmbh Monitoring dissolution of a dosage form in progress during dissolution by low coherence interferometry
CN117783459A (en) * 2024-02-28 2024-03-29 沈阳科惠生物医药科技有限公司 Drug dissolution curve determination method and system

Also Published As

Publication number Publication date
WO1997046860A3 (en) 1998-02-19
AU729814B2 (en) 2001-02-08
JP3380864B2 (en) 2003-02-24
AU4326297A (en) 1998-01-05
JPH11514088A (en) 1999-11-30
CA2257106C (en) 2002-04-23
EP0938657A4 (en) 1999-10-27
EP0938657A2 (en) 1999-09-01
CA2257106A1 (en) 1997-12-11

Similar Documents

Publication Publication Date Title
US6558957B1 (en) Detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form
Song et al. High-performance fiber-optic pH microsensors for practical physiological measurements using a dual-emission sensitive dye
AU729814B2 (en) Improvements in detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form
WO2001016582A9 (en) In situ methods for measuring the release of a substance from a dosage form
Pimenta et al. Application of sequential injection analysis to pharmaceutical analysis
US6764651B2 (en) Fiber-optic dissolution systems, devices, and methods
Escandar et al. A review of multivariate calibration methods applied to biomedical analysis
Skoulika et al. Rapid quantitative determination of ciprofloxacin in pharmaceuticals by use of solid-state FT-Raman spectroscopy
WO1997046860A9 (en) Improvements in detection systems and methods for predicting the dissolution curve of a drug from a pharmaceutical dosage form
Kassis et al. Attenuated total reflection infrared spectroscopy (ATR-IR) as an in situ technique for dissolution studies
JP3860118B2 (en) System and method for in vitro analysis of therapeutic agents
Song et al. Hyperspectral Raman line mapping as an effective tool to monitor the coating thickness of pharmaceutical tablets
Legnerová et al. Automated sequential injection fluorimetric determination and dissolution studies of ergotamine tartrate in pharmaceuticals
Liyanage et al. Ultraviolet absorption spectroscopy of peptides
Fanget et al. Correction of inner filter effect in mirror coating cells for trace level fluorescence measurements
Li et al. Flow-injection system for automated dissolution testing of isoniazid tablets with chemiluminescence detection
El-Zohry et al. Environmental method to determine dopamine and ascorbic acid simultaneously via derivative spectrophotometry
WO2003098199A1 (en) In situ methods for measuring the release of a substance from a dosage form
Wiberg et al. Multivariate chemometric approach to fiber-optic dissolution testing
JP3380864B6 (en) Improvements in detection systems and methods for predicting dissolution curves of drugs from pharmaceutical formulations
Mirza et al. Comprehensive validation scheme for in situ fiber optics dissolution method for pharmaceutical drug product testing
CN1276250C (en) Optical fiber in situ monitor for dissolution and release of medicine
CN1271401C (en) Optical fiber in situ tester for dissolution and release of medicine
Drennen et al. Pharmaceutical applications of near-infrared spectrometry
Hui et al. Diffusional microtitration of 20 μL samples with optical indication

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH HU IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN YU AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
COP Corrected version of pamphlet

Free format text: PAGES 1/12-12/12, DRAWINGS, REPLACED BY NEW PAGES 1/13-13/13; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

ENP Entry into the national phase in:

Ref country code: CA

Ref document number: 2257106

Kind code of ref document: A

Format of ref document f/p: F

Ref document number: 2257106

Country of ref document: CA

ENP Entry into the national phase in:

Ref country code: JP

Ref document number: 1998 500947

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1997941329

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWP Wipo information: published in national office

Ref document number: 1997941329

Country of ref document: EP