WO2012016042A2 - Glatiramer acetate molecular weight markers - Google Patents

Glatiramer acetate molecular weight markers Download PDF

Info

Publication number
WO2012016042A2
WO2012016042A2 PCT/US2011/045724 US2011045724W WO2012016042A2 WO 2012016042 A2 WO2012016042 A2 WO 2012016042A2 US 2011045724 W US2011045724 W US 2011045724W WO 2012016042 A2 WO2012016042 A2 WO 2012016042A2
Authority
WO
WIPO (PCT)
Prior art keywords
molecular weight
daltons
glatiramer acetate
markers
kda
Prior art date
Application number
PCT/US2011/045724
Other languages
French (fr)
Other versions
WO2012016042A3 (en
Inventor
Santhanakrishnan Srinivasan
Karthik Ramasamy
Chakravarthula Kalyan Narasimham Nallam
Yagna Kiran Kumar Komaravolu
Sudheer Reddy Kallam
Bala Harsha Vardhan Ganji
Ravindra Chary Bathoju
Basanthi Devi
Original Assignee
Dr. Reddy's Laboratories Ltd.
Dr. Reddy's Laboratories, Inc.
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
Priority to CA2806997A priority Critical patent/CA2806997A1/en
Priority to RU2013108828/15A priority patent/RU2013108828A/en
Priority to NZ606723A priority patent/NZ606723A/en
Priority to MX2013001150A priority patent/MX2013001150A/en
Priority to US13/812,684 priority patent/US9109006B2/en
Priority to CN201180041836.0A priority patent/CN103080747B/en
Priority to AU2011282635A priority patent/AU2011282635B2/en
Priority to EP11813184.6A priority patent/EP2598889A4/en
Application filed by Dr. Reddy's Laboratories Ltd., Dr. Reddy's Laboratories, Inc. filed Critical Dr. Reddy's Laboratories Ltd.
Priority to KR1020137003967A priority patent/KR20130043196A/en
Priority to BR112013002230A priority patent/BR112013002230A2/en
Priority to JP2013521976A priority patent/JP2013535474A/en
Publication of WO2012016042A2 publication Critical patent/WO2012016042A2/en
Publication of WO2012016042A3 publication Critical patent/WO2012016042A3/en
Priority to ZA2013/00980A priority patent/ZA201300980B/en

Links

Classifications

    • 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
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/34Size selective separation, e.g. size exclusion chromatography, gel filtration, permeation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2/00Peptides of undefined number of amino acids; Derivatives thereof
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/62Detectors specially adapted therefor
    • G01N30/72Mass spectrometers
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/10Composition for standardization, calibration, simulation, stabilization, preparation or preservation; processes of use in preparation for chemical testing
    • Y10T436/105831Protein or peptide standard or control [e.g., hemoglobin, etc.]
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/25Chemistry: analytical and immunological testing including sample preparation
    • Y10T436/25375Liberation or purification of sample or separation of material from a sample [e.g., filtering, centrifuging, etc.]
    • Y10T436/255Liberation or purification of sample or separation of material from a sample [e.g., filtering, centrifuging, etc.] including use of a solid sorbent, semipermeable membrane, or liquid extraction

Definitions

  • aspects of the present application relate to polypeptides having no predetermined amino acid sequence for use as molecular weight markers of glatiramer acetate and for the accurate determination of the average molecular weight of glatiramer acetate.
  • Glatiramer acetate (formerly known as copolymer-1 ) is chemically designated L-glutamic acid polymer with L-alanine, L-lysine, and L-tyrosine, acetate salt. It has the structural formula of Formula (I).
  • Glatiramer acetate is the acetate salt of a synthetic mixture of polypeptides, each of which consists essentially of the four naturally occurring amino acids: L- glutamic acid, L-alanine, L-tyrosine, and L-lysine with an average molar fraction of 0.141 , 0.427, 0.095, and 0.338, respectively.
  • the average molecular weight of glatiramer acetate is 5,000-9,000 Daltons.
  • Glatiramer acetate is sold in USA as Copaxone®, a registered trademark of Teva Pharmaceutical Industries Ltd.
  • Glatiramer acetate is indicated for reduction of the frequency of relapses in patients with Relapsing-Remitting Multiple Sclerosis (RRMS).
  • U.S. Patent No. 6,514,938 discloses seven polypeptides having an amino acid sequences which are identified in the patent.
  • U.S. Patent No. 7,074,580 discloses a process for obtaining a pharmaceutical product containing a mixture of polypeptides.
  • Each of the polypeptides consists essentially of alanine, glutamic acid, tyrosine, and lysine.
  • the mixture has an average molecular weight from 4000 to 13,000 Daltons and in the mixture the molar fraction of alanine is 0.427, of glutamic acid is 0.141 , of lysine is 0.337, and of tyrosine is 0.093.
  • the patent teaches determining the molecular weight distribution of a batch of an aqueous mixture of polypeptides, each of which consists essentially of alanine, glutamic acid, tyrosine, and lysine.
  • the molecular weight distribution determination uses a gel permeation chromatography column to determine whether the mixture has an average molecular weight from 4000 to 13,000 Daltons for inclusion in a pharmaceutical product.
  • the determination method comprises calibrating the molecular weight obtained using the gel permeation chromatography column by subjecting a plurality of molecular weight markers, each of which is a polypeptide consisting essentially of alanine, glutamic acid, tyrosine and lysine and having a predetermined amino acid sequence, to chromatography on the column to establish a relationship between retention time on the column and molecular weight.
  • molecular weight markers each of which is a polypeptide consisting essentially of alanine, glutamic acid, tyrosine and lysine and having a predetermined amino acid sequence
  • U.S. Patent No. 7,163,802 discloses in a process for obtaining a pharmaceutical product containing a mixture of polypeptides, each of which consists essentially of alanine, glutamic acid, tyrosine, and lysine.
  • the mixture of polypeptides has an average molecular weight between 2000 and 40,000 Daltons and in the mixture the molar fraction of alanine is 0.38 to 0.5, of glutamic acid is 0.13 to 0.15, of tyrosine is 0.08 to 0.10 and of lysine is 0.3 to 0.4.
  • a batch of a mixture of polypeptides each of which consists essentially of alanine, glutamic acid, tyrosine, and lysine, is tested using a gel permeation chromatography column to determine whether the mixture has an average molecular weight between 2000 and 40,000 Daltons for inclusion in the pharmaceutical product.
  • the testing method comprises calibrating the gel permeation chromatography column by subjecting a plurality of molecular weight markers, each of which is a polypeptide consisting essentially of alanine, glutamic acid, tyrosine and lysine and having a predetermined amino acid sequence, to chromatography on the column to establish a relationship between retention time on the column and molecular weight said relationship being used to determine average molecular weight of the mixture of polypeptides.
  • molecular weight markers each of which is a polypeptide consisting essentially of alanine, glutamic acid, tyrosine and lysine and having a predetermined amino acid sequence
  • the present application provides polypeptides having no predetermined amino acid sequence for use as molecular weight markers of glatiramer acetate , which are useful as standards for determining the peak average molecular weight of glatiramer acetate a polypeptide.
  • Fig. 1 is a plot of the retention time (RT) of the present molecular weight markers versus the log molecular weight of those markers, using the RT-based algorithm.
  • Fig.2 is an illustration of MALDI spectrum of molecular weight marker 1 prepared according to Example 1.
  • Fig.3 is an illustration of MALDI spectrum of molecular weight marker 2 prepared according to Example 1.
  • Fig.4 is an illustration of MALDI spectrum of molecular weight marker 3 prepared according to Example 1.
  • Fig.5 is an illustration of MALDI spectrum of molecular weight marker 4 prepared according to Example 1.
  • Fig.6 is an illustration of circular dichroism spectrum of Copaxone®.
  • Fig. 7 is an illustration of circular dichroism spectrum of molecular weight marker 2 prepared according to Example .
  • Fig. 8 is an illustration of circular dichroism spectrum of molecular weight marker 3 prepared according to Example 1.
  • Fig. 9 is an illustration of circular dichroism spectrum of molecular weight marker 4 prepared according to Example 1.
  • the present application provides polypeptides having no predetermined amino acid sequence for use as molecular weight markers of glatiramer acetate , which are useful as standards for determining the peak average molecular weight of glatiramer acetate a polypeptide.
  • the present application provides polypeptides for use as molecular weight markers of glatiramer acetate for determining the peak average molecular weight of glatiramer acetate a polypeptide.
  • the present application provides polypeptides having an amino acid composition corresponding to glatiramer acetate polypeptide and an identified molecular weight which is between about 2,000 Daltons and about 40,000 Daltons or between about 2,000 Daltons and about 20,000 Daltons or between about 2,000 Daltons and about 15,000 Daltons or between about 2,000 Daltons and about 10,000 Daltons; for use as molecular weight markers for determining the average molecular weight of a polypeptide.
  • the present application provides polypeptides having an amino acid composition corresponding to glatiramer acetate and an identified molecular weight which is between about 2,000 Daltons and about 40,000 Daltons or between about 2,000 Daltons and about 20,000 Daltons or between about 2,000 Daltons and about 15,000 Daltons or between about 2,000 Daltons and about 10,000 Daltons; for use as molecular weight markers for determining the average molecular weight of glatiramer acetate.
  • the present application provides polypeptides consisting essentially of amino acids alanine, glutamic acid, tyrosine, and lysine in molar fractions of from about 0.38 to about 0.50 alanine, from about 0.13 to about 0.155 glutamic acid, from about 0.08 to about 0.12 tyrosine, and from about 0.28 to about 0.4 lysine, for use as molecular weight markers for determining the average molecular weight of a polypeptide.
  • the present application provides polypeptides consisting essentially of amino acids alanine, glutamic acid, tyrosine, and lysine in molar fractions of from about 0.38 alanine to about 0.50 alanine, from about 0.13 glutamic acid to about 0.155 glutamic acid, from about 0.08 tyrosine to about 0.12 tyrosine, and from about 0.28 lysine to about 0.4 lysine, for use as molecular weight markers for determining the average molecular weight of glatiramer acetate.
  • the present application provides processes for preparing molecular weight markers of the present application, which includes one or more of the following steps, individually or in the sequence recited:
  • step (b) passing the fractions collected in step (a) through molecular weight cut-off membranes;
  • step (c) isolating the molecular weight markers by lyophilizing the fractions collected from step (b).
  • the present application provides processes for preparing molecular weight markers of the present application, which includes one or more of the following steps, individually or in the sequence recited:
  • step (b) passing the fractions collected in step (a) through molecular weight cut-off membranes;
  • step (c) isolating the molecular weight markers by lyophilizing the fractions collected from step (b).
  • Step (a) involves fractionating a sample of glatiramer acetate in a gel permeation chromatography (GPC) column.
  • Suitable chromatography media for gel filtration that may be used in step (a) include, but are not limited to, SuperdexTM, SephacrylTM, SuperoseTM and SephadexTM , and the like.
  • SuperdexTM is a composite medium based on highly cross-linked porous agarose particles to which dextran have been covalently bonded.
  • SuperdexTM is a trademark of GE Healthcare companies.
  • SephacrylTM is an allyl dextran cross- linked with N, N'-methylene bisacrylamide. SephacrylTM is a trademark of GE Healthcare.
  • SuperoseTM is a medium with high physical and chemical stability based on highly cross-linked porous agarose particles. SuperoseTM is a trademark of GE Healthcare Ltd, a General Electric Company. SephadexTM is dextran cross-linked with epichlorohydrin. SephadexTM is a trademark of GE Healthcare companies.
  • a sample of glatiramer acetate may be prepared by dissolving in a suitable solvent and then the resultant solution is filtered to remove any unwanted particulate matter through syringe filters.
  • Suitable solvent that may be used for the preparation of a sample of glatiramer acetate include, but are not limited to, Milli Q® water, and the like. Milli-Q® is a registered trademark of the Millipore Corporation, Billerica, MA
  • Suitable buffers that may be used in step (a) include, but are not limited to, acetate buffers, phosphate buffers, citrate buffers, and sodium chloride, or mixtures thereof.
  • Step (b) involves passing of the fractions collected in step (a) through molecular cut-off membranes for obtaining the markers with a narrow molecular weight distribution.
  • Suitable molecular cut-off membranes may range from 1 KDa to 10 KDa, or 1 KDa to 20 KDa, or 1 KDa to 30 KDa.
  • Step (b) may be preceded by an optional lyophilization of the individual fractions collected in step (a), dissolving the lyophilizates in a solvent, and then passing through molecular cut-off membranes.
  • Step (c) involves isolation of molecular weight markers by lyophilizing the fractions collected from step (b).
  • the molecular weights of the markers of the present application may be determined by any method including the methods described in the art.
  • molecular weight of markers of present application may be determined by Matrix-Assisted Laser Desorption/lonization (MALDI) or Low Angle Laser Light Scattering Detection (LALLS) or Multi Angle Light Scattering Detection (MALLS).
  • MALDI Matrix-Assisted Laser Desorption/lonization
  • LALLS Low Angle Laser Light Scattering Detection
  • MALLS Multi Angle Light Scattering Detection
  • Glatiramer acetate that is used as the input for the process of the present application may be obtained by any process including the processes described in the art.
  • glatiramer acetate may be prepared by the processes described in International Application No. PCT/US1 1/34102.
  • the present application provides a method for determining the peak average molecular weight of a polypeptide or a pharmaceutically acceptable salt thereof, which comprises subjecting the polypeptide or a pharmaceutically acceptable salt thereof to chromatography on a chromatographic column so as to determine the average molecular weight of the polypeptide or a pharmaceutically acceptable salt thereof, wherein the chromatographic column is calibrated by using a plurality of molecular weight markers of the present application, wherein each of the molecular weight marker is a polypeptide having no predetermined amino acid sequence.
  • the present application provides a method for determining the peak average molecular weight of glatiramer acetate or a pharmaceutically acceptable salt thereof, which comprises subjecting the glatiramer acetate or a pharmaceutically acceptable salt thereof to chromatography on a chromatographic column so as to determine the peak average molecular weight of the glatiramer acetate or a pharmaceutically acceptable salt thereof, wherein the chromatographic column is calibrated by using a plurality of molecular weight markers of the present application, wherein each of the molecular weight markers is a polypeptide having no predetermined amino acid sequence.
  • the present application provides a process of calibration of gel permeation chromatography column by using molecular weight markers of the present application.
  • the present application provides a plurality of molecular weight markers for determining the average molecular weight of a polypeptide on a molecular weight fractionation column.
  • the present application provides a plurality of molecular weight markers for determining the average molecular weight of glatiramer acetate on a molecular weight fractionation column.
  • the present application provides a linear relationship between the log molecular weight of the glatiramer acetate molecular weight markers of the present application and the retention time of the molecular weight markers on a molecular weight fractionation column.
  • polypeptide e.g., copolymer 1 glatiramer acetate preparation for use in a pharmaceutical product
  • Molecular weight markers that have chemical and physical characteristics similar to polypeptide provide an accurate and robust calibration set for determination of molecular weights of production batches.
  • the present application provides derivatives of polypeptide e.g., glatiramer acetate useful as molecular weight markers for determining the average molecular weight of polypeptide e.g., glatiramer acetate preparations.
  • Molecular weight markers of the present application include polypeptides having an amino acid composition approximately corresponding to polypeptide e.g., glatiramer acetate , and an identified molecular weight which is between about 2,000 Daltons and about 40,000 Daltons, between about 2,000 Daltons and about 20,000 Daltons, between about 2,000 Daltons and about 15,000 Daltons, or between about 2,000 Daltons and about 10,000 Daltons; and are useful for accurately determining the average molecular weight of polypeptide e.g., glatiramer acetate. It follows from the requirement for an identified molecular weight that a molecular weight marker should not be highly polydispersed and should have a narrow molecular weight distribution. The molecular weight markers of the present application are not fixed sequence markers.
  • the present application provides molecular weight markers consisting essentially of amino acids alanine, glutamic acid, tyrosine, and lysine in defined molar ratios.
  • the molar ratio of amino acids of molecular weight markers of present application is same as that found in a polypeptide e.g., glatiramer acetate.
  • Such a correspondence in molar ratios provides the best molecular weight markers because those markers will have a charge and a molecular shape which is similar to that of a polypeptide e.g., glatiramer acetate.
  • the markers may migrate or elute somewhat differently from polypeptide e.g., glatiramer acetate copolymer preparations, though these preparations have the same molecular weight as that of markers.
  • the plurality of molecular weight markers are polypeptides.
  • the plurality of markers can be two to about ten or more.
  • Each of these polypeptides has an identified peak average molecular weight which is between about 2,000 Daltons and about 40,000 Daltons, between about 2,000 Daltons and about 20,000 Daltons, between about 2,000 Daltons and about 15,000 Daltons, or between about 2,000 Daltons and about 10,000 Daltons; and an amino acid composition which corresponds approximately to that of polypeptide e.g., glatiramer acetate.
  • the molecular weight markers of the present application may have therapeutic activity which is similar to glatiramer acetate polypeptide. These markers may be used in any molecular size discrimination system using any available molecular weight determination procedure or apparatus. For example, the present markers may be used for calibration of any chromatographic procedure or apparatus which is used for molecular weight determinations of polypeptides.
  • a chromatographic apparatus may be a molecular weight sizing column which separates polypeptides on the basis of their molecular size. Examples of molecular weight sizing columns include TSK-GEL® columns, SephadexTM columns, Sepharose® columns, SephacrylTM columns, and SuperoseTM columns.
  • Sephacryl® is a covalently cross linked dextran/bisacrylamide copolymer gel formed into beads. It is used in gel filtration columns for separating molecules in the size range 5 kD to 1.5 million Dalton. Sephacryl® S-100 has a 1 kD - lOOkD molecular weight range and has a particle size of 25-75 ⁇ . Sephacryl® is a registered trademark of GE Healthcare. SuperoseTM 12 is a cross-linked, agarose-based medium optimized for high performance gel filtration of biomolecules.
  • EXAMPLE 1 Preparation of molecular weight markers of glatiramer acetate.
  • glatiramer acetate 350 mg is dissolved in Milli-Q® water (10 mL) and filtered through 0.45 ⁇ syringe filter.
  • the column is stabilized with the above prepared mobile phase.
  • the flow rate of the mobile phase is kept constant and stabilization is continued until base line reaches constant value.
  • Sample is loaded on to the column at the same flow rate through pump B assembly.
  • the set mark option is used to indicate injection marking.
  • Absorbencies are recorded at dual wavelengths such as 215 nm & 280 nm. At nearly 111 mL of elution volume, the peak at 280 nm starts rising up.
  • the fraction collector is activated to deliver 9 mL of each fraction. At 180 mL of elution volume the peak reaches its maximum value. Mobile phase flow still continues. At around 300 mL of elution volume, the peak almost reaches its baseline.
  • the four selected fractions, at about RT 23.0 minutes, at about RT 26 minutes, at about RT 29 minutes and at about RT 30 minutes are then concentrated through a 10 KDa molecular weight cutoff membrane, a 3 KDa molecular weight cutoff membrane, a 3 KDa molecular weight cutoff membrane and a 3 KDa molecular weight cut off membrane respectively.
  • the concentrated solutions are lyophilized individually to afford 15 mg, 30 mg, 35 mg, and 12 mg of the title compound.
  • EXAMPLE 2 Preparation of glatiramer acetate molecular weight markers.
  • glatiramer acetate (1500 mg) is dissolved in Milli-Q® water (10 mL) and filtered through 0.45 ⁇ syringe filter.
  • the column is stabilized with the above prepared mobile phase.
  • the flow rate of the mobile phase is kept constant and stabilization is continued until base line at absorbance @ 215 nm reaches constant value.
  • Sample is loaded on to the column at the same flow rate through pump B assembly.
  • the set mark option is used to indicate injection loading.
  • Absorbencies are recorded at dual wavelengths such as 2 5 nm & 280 nm.
  • the peak at 215 nm starts rising up.
  • the fraction collector is activated to deliver 10 mL of each fraction.
  • At nearly 640 mL of elution volume the peak reaches its maximum value.
  • Mobile phase flow still continues.
  • the peak almost reaches its baseline.
  • fractionation is stopped. Two consecutive fractions from the fraction collector are mixed together to get the required fraction volume of 20 mL each. After fractionation, the column is stabilized with mobile phase for the next cycle. The same procedure was repeated for numerous batches. Pooled fractions from the each batch were chromatographed on SuperoseTM 12 column and the peak retention time for each of the fractions recorded. Each of these fractions elutes at a distinct retention time as given in tables 1 , 2, & 3 respectively. Fractions with similar retention time from different batches are pooled together for further processing.
  • Molecular weight markers that are prepared according to the procedure given in Example 1 above and a glatiramer acetate preparation are expected to demonstrate a similar correlation between retention time (RT) and log molecular weight.
  • the molecular weight markers of the present application are chromatographed on SuperoseTM 12 column. The peak retention time for each of the markers is recorded.
  • the linear correlation between Log Molecular Weight (MW) and the Retention Time (RT) is calculated as follows:
  • MW is the molecular weight
  • RT is the retention time
  • a and B are the intercept and the slope of the calculated regression function.
  • molecular weight markers that are prepared according to the procedure given in Example 2 above and a glatiramer acetate preparation are expected to demonstrate a similar correlation between retention time (RT) and log molecular weight.
  • the molecular weight markers of the present application are chromatographed on SuperoseTM 12 column. The peak retention time for each of the markers is recorded.
  • the linear correlation between Log Molecular Weight (MW) and the Retention Time (RT) is calculated as follows:
  • MW is the molecular weight
  • RT is the retention time
  • a and B are the intercept and the slope of the calculated regression function.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Analytical Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Molecular Biology (AREA)
  • General Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Food Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Pyrane Compounds (AREA)
  • Investigating Or Analyzing Non-Biological Materials By The Use Of Chemical Means (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Polyesters Or Polycarbonates (AREA)
  • Heterocyclic Compounds That Contain Two Or More Ring Oxygen Atoms (AREA)

Abstract

Aspects of the present application relate to molecular weight markers of glatiramer acetate for accurate determination of the average molecular weight of glatiramer acetate.

Description

GLATIRAMER ACETATE MOLECULAR WEIGHT MARKERS
INTRODUCTION
Aspects of the present application relate to polypeptides having no predetermined amino acid sequence for use as molecular weight markers of glatiramer acetate and for the accurate determination of the average molecular weight of glatiramer acetate.
Glatiramer acetate (formerly known as copolymer-1 ) is chemically designated L-glutamic acid polymer with L-alanine, L-lysine, and L-tyrosine, acetate salt. It has the structural formula of Formula (I).
(Glu, Ala, Lys, Tyr)x. XCH3COOH
(C5H9NO4.C3H7NO2.C6Hu 2O2.C9H! ! NO3)x.XC2H4O2
Formula (I)
Glatiramer acetate is the acetate salt of a synthetic mixture of polypeptides, each of which consists essentially of the four naturally occurring amino acids: L- glutamic acid, L-alanine, L-tyrosine, and L-lysine with an average molar fraction of 0.141 , 0.427, 0.095, and 0.338, respectively. The average molecular weight of glatiramer acetate is 5,000-9,000 Daltons. Glatiramer acetate is sold in USA as Copaxone®, a registered trademark of Teva Pharmaceutical Industries Ltd. Glatiramer acetate is indicated for reduction of the frequency of relapses in patients with Relapsing-Remitting Multiple Sclerosis (RRMS).
U.S. Patent No. 6,514,938 discloses seven polypeptides having an amino acid sequences which are identified in the patent. U.S. Patent No. 7,074,580 discloses a process for obtaining a pharmaceutical product containing a mixture of polypeptides. Each of the polypeptides consists essentially of alanine, glutamic acid, tyrosine, and lysine. The mixture has an average molecular weight from 4000 to 13,000 Daltons and in the mixture the molar fraction of alanine is 0.427, of glutamic acid is 0.141 , of lysine is 0.337, and of tyrosine is 0.093. The patent teaches determining the molecular weight distribution of a batch of an aqueous mixture of polypeptides, each of which consists essentially of alanine, glutamic acid, tyrosine, and lysine. The molecular weight distribution determination uses a gel permeation chromatography column to determine whether the mixture has an average molecular weight from 4000 to 13,000 Daltons for inclusion in a pharmaceutical product. The determination method comprises calibrating the molecular weight obtained using the gel permeation chromatography column by subjecting a plurality of molecular weight markers, each of which is a polypeptide consisting essentially of alanine, glutamic acid, tyrosine and lysine and having a predetermined amino acid sequence, to chromatography on the column to establish a relationship between retention time on the column and molecular weight.
U.S. Patent No. 7,163,802 discloses in a process for obtaining a pharmaceutical product containing a mixture of polypeptides, each of which consists essentially of alanine, glutamic acid, tyrosine, and lysine. The mixture of polypeptides has an average molecular weight between 2000 and 40,000 Daltons and in the mixture the molar fraction of alanine is 0.38 to 0.5, of glutamic acid is 0.13 to 0.15, of tyrosine is 0.08 to 0.10 and of lysine is 0.3 to 0.4. During the process, a batch of a mixture of polypeptides, each of which consists essentially of alanine, glutamic acid, tyrosine, and lysine, is tested using a gel permeation chromatography column to determine whether the mixture has an average molecular weight between 2000 and 40,000 Daltons for inclusion in the pharmaceutical product. The testing method comprises calibrating the gel permeation chromatography column by subjecting a plurality of molecular weight markers, each of which is a polypeptide consisting essentially of alanine, glutamic acid, tyrosine and lysine and having a predetermined amino acid sequence, to chromatography on the column to establish a relationship between retention time on the column and molecular weight said relationship being used to determine average molecular weight of the mixture of polypeptides.
There remains a need to provide molecular weight markers of glatiramer acetate useful as standards for determining the peak average molecular weight of glatiramer acetate polypeptide compositions.
SUMMARY
The present application provides polypeptides having no predetermined amino acid sequence for use as molecular weight markers of glatiramer acetate , which are useful as standards for determining the peak average molecular weight of glatiramer acetate a polypeptide. BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 is a plot of the retention time (RT) of the present molecular weight markers versus the log molecular weight of those markers, using the RT-based algorithm.
Fig.2 is an illustration of MALDI spectrum of molecular weight marker 1 prepared according to Example 1.
Fig.3 is an illustration of MALDI spectrum of molecular weight marker 2 prepared according to Example 1.
Fig.4 is an illustration of MALDI spectrum of molecular weight marker 3 prepared according to Example 1.
Fig.5 is an illustration of MALDI spectrum of molecular weight marker 4 prepared according to Example 1.
Fig.6 is an illustration of circular dichroism spectrum of Copaxone®.
Fig. 7 is an illustration of circular dichroism spectrum of molecular weight marker 2 prepared according to Example .
Fig. 8 is an illustration of circular dichroism spectrum of molecular weight marker 3 prepared according to Example 1.
Fig. 9 is an illustration of circular dichroism spectrum of molecular weight marker 4 prepared according to Example 1.
DETAILED DESCRIPTION
The present application provides polypeptides having no predetermined amino acid sequence for use as molecular weight markers of glatiramer acetate , which are useful as standards for determining the peak average molecular weight of glatiramer acetate a polypeptide.
The present application provides polypeptides for use as molecular weight markers of glatiramer acetate for determining the peak average molecular weight of glatiramer acetate a polypeptide.
In an aspect, the present application provides polypeptides having an amino acid composition corresponding to glatiramer acetate polypeptide and an identified molecular weight which is between about 2,000 Daltons and about 40,000 Daltons or between about 2,000 Daltons and about 20,000 Daltons or between about 2,000 Daltons and about 15,000 Daltons or between about 2,000 Daltons and about 10,000 Daltons; for use as molecular weight markers for determining the average molecular weight of a polypeptide.
In an aspect, the present application provides polypeptides having an amino acid composition corresponding to glatiramer acetate and an identified molecular weight which is between about 2,000 Daltons and about 40,000 Daltons or between about 2,000 Daltons and about 20,000 Daltons or between about 2,000 Daltons and about 15,000 Daltons or between about 2,000 Daltons and about 10,000 Daltons; for use as molecular weight markers for determining the average molecular weight of glatiramer acetate.
In an aspect, the present application provides polypeptides consisting essentially of amino acids alanine, glutamic acid, tyrosine, and lysine in molar fractions of from about 0.38 to about 0.50 alanine, from about 0.13 to about 0.155 glutamic acid, from about 0.08 to about 0.12 tyrosine, and from about 0.28 to about 0.4 lysine, for use as molecular weight markers for determining the average molecular weight of a polypeptide.
In an aspect, the present application provides polypeptides consisting essentially of amino acids alanine, glutamic acid, tyrosine, and lysine in molar fractions of from about 0.38 alanine to about 0.50 alanine, from about 0.13 glutamic acid to about 0.155 glutamic acid, from about 0.08 tyrosine to about 0.12 tyrosine, and from about 0.28 lysine to about 0.4 lysine, for use as molecular weight markers for determining the average molecular weight of glatiramer acetate.
In an aspect, the present application provides processes for preparing molecular weight markers of the present application, which includes one or more of the following steps, individually or in the sequence recited:
(a) fractionating the glatiramer acetate polypeptide in a gel permeation chromatography (GPC) column;
(b) passing the fractions collected in step (a) through molecular weight cut-off membranes; and
(c) isolating the molecular weight markers by lyophilizing the fractions collected from step (b). In an aspect, the present application provides processes for preparing molecular weight markers of the present application, which includes one or more of the following steps, individually or in the sequence recited:
(a) fractionating the glatiramer acetate in a gel permeation chromatography (GPC) column;
(b) passing the fractions collected in step (a) through molecular weight cut-off membranes; and
(c) isolating the molecular weight markers by lyophilizing the fractions collected from step (b).
Step (a) involves fractionating a sample of glatiramer acetate in a gel permeation chromatography (GPC) column. Suitable chromatography media for gel filtration that may be used in step (a) include, but are not limited to, Superdex™, Sephacryl™, Superose™ and Sephadex™ , and the like. Superdex™ is a composite medium based on highly cross-linked porous agarose particles to which dextran have been covalently bonded. Superdex™ is a trademark of GE Healthcare companies. Sephacryl™ is an allyl dextran cross- linked with N, N'-methylene bisacrylamide. Sephacryl™ is a trademark of GE Healthcare. Superose™ is a medium with high physical and chemical stability based on highly cross-linked porous agarose particles. Superose™ is a trademark of GE Healthcare Ltd, a General Electric Company. Sephadex™ is dextran cross-linked with epichlorohydrin. Sephadex™ is a trademark of GE Healthcare companies.
A sample of glatiramer acetate may be prepared by dissolving in a suitable solvent and then the resultant solution is filtered to remove any unwanted particulate matter through syringe filters. Suitable solvent that may be used for the preparation of a sample of glatiramer acetate include, but are not limited to, Milli Q® water, and the like. Milli-Q® is a registered trademark of the Millipore Corporation, Billerica, MA
Suitable buffers that may be used in step (a) include, but are not limited to, acetate buffers, phosphate buffers, citrate buffers, and sodium chloride, or mixtures thereof. Step (b) involves passing of the fractions collected in step (a) through molecular cut-off membranes for obtaining the markers with a narrow molecular weight distribution. Suitable molecular cut-off membranes that may be used in step (b) may range from 1 KDa to 10 KDa, or 1 KDa to 20 KDa, or 1 KDa to 30 KDa.
Step (b) may be preceded by an optional lyophilization of the individual fractions collected in step (a), dissolving the lyophilizates in a solvent, and then passing through molecular cut-off membranes.
Step (c) involves isolation of molecular weight markers by lyophilizing the fractions collected from step (b).
The molecular weights of the markers of the present application may be determined by any method including the methods described in the art. For example molecular weight of markers of present application may be determined by Matrix-Assisted Laser Desorption/lonization (MALDI) or Low Angle Laser Light Scattering Detection (LALLS) or Multi Angle Light Scattering Detection (MALLS).
Glatiramer acetate that is used as the input for the process of the present application may be obtained by any process including the processes described in the art. For example glatiramer acetate may be prepared by the processes described in International Application No. PCT/US1 1/34102.
In an aspect, the present application provides a method for determining the peak average molecular weight of a polypeptide or a pharmaceutically acceptable salt thereof, which comprises subjecting the polypeptide or a pharmaceutically acceptable salt thereof to chromatography on a chromatographic column so as to determine the average molecular weight of the polypeptide or a pharmaceutically acceptable salt thereof, wherein the chromatographic column is calibrated by using a plurality of molecular weight markers of the present application, wherein each of the molecular weight marker is a polypeptide having no predetermined amino acid sequence.
In an aspect, the present application provides a method for determining the peak average molecular weight of glatiramer acetate or a pharmaceutically acceptable salt thereof, which comprises subjecting the glatiramer acetate or a pharmaceutically acceptable salt thereof to chromatography on a chromatographic column so as to determine the peak average molecular weight of the glatiramer acetate or a pharmaceutically acceptable salt thereof, wherein the chromatographic column is calibrated by using a plurality of molecular weight markers of the present application, wherein each of the molecular weight markers is a polypeptide having no predetermined amino acid sequence.
In another aspect, the present application provides a process of calibration of gel permeation chromatography column by using molecular weight markers of the present application.
In an aspect, the present application provides a plurality of molecular weight markers for determining the average molecular weight of a polypeptide on a molecular weight fractionation column.
In an aspect, the present application provides a plurality of molecular weight markers for determining the average molecular weight of glatiramer acetate on a molecular weight fractionation column.
In another aspect, the present application provides a linear relationship between the log molecular weight of the glatiramer acetate molecular weight markers of the present application and the retention time of the molecular weight markers on a molecular weight fractionation column.
To certify a polypeptide e.g., copolymer 1 (glatiramer acetate preparation for use in a pharmaceutical product, it is necessary to accurately determine the average molecular weight of the polypeptides in the preparation. Molecular weight markers that have chemical and physical characteristics similar to polypeptide provide an accurate and robust calibration set for determination of molecular weights of production batches. The present application provides derivatives of polypeptide e.g., glatiramer acetate useful as molecular weight markers for determining the average molecular weight of polypeptide e.g., glatiramer acetate preparations.
Molecular weight markers of the present application include polypeptides having an amino acid composition approximately corresponding to polypeptide e.g., glatiramer acetate , and an identified molecular weight which is between about 2,000 Daltons and about 40,000 Daltons, between about 2,000 Daltons and about 20,000 Daltons, between about 2,000 Daltons and about 15,000 Daltons, or between about 2,000 Daltons and about 10,000 Daltons; and are useful for accurately determining the average molecular weight of polypeptide e.g., glatiramer acetate. It follows from the requirement for an identified molecular weight that a molecular weight marker should not be highly polydispersed and should have a narrow molecular weight distribution. The molecular weight markers of the present application are not fixed sequence markers.
In an aspect, the present application provides molecular weight markers consisting essentially of amino acids alanine, glutamic acid, tyrosine, and lysine in defined molar ratios. The molar ratio of amino acids of molecular weight markers of present application is same as that found in a polypeptide e.g., glatiramer acetate.
Such a correspondence in molar ratios provides the best molecular weight markers because those markers will have a charge and a molecular shape which is similar to that of a polypeptide e.g., glatiramer acetate. When structurally dissimilar markers are used, the markers may migrate or elute somewhat differently from polypeptide e.g., glatiramer acetate copolymer preparations, though these preparations have the same molecular weight as that of markers.
The plurality of molecular weight markers are polypeptides. The plurality of markers can be two to about ten or more. Each of these polypeptides has an identified peak average molecular weight which is between about 2,000 Daltons and about 40,000 Daltons, between about 2,000 Daltons and about 20,000 Daltons, between about 2,000 Daltons and about 15,000 Daltons, or between about 2,000 Daltons and about 10,000 Daltons; and an amino acid composition which corresponds approximately to that of polypeptide e.g., glatiramer acetate.
When such a plurality of molecular weight markers of the present application are used as standards for determining the average molecular weight of a polypeptide e.g., glatiramer acetate , a relationship which is approximately linear exists between the retention time of the molecular weight markers of the present application on the chromatographic column and the log of the molecular weight of the markers. A plurality of markers is used which is sufficient to establish approximately the linear relationship between the retention time and the molecular weight.
The molecular weight markers of the present application may have therapeutic activity which is similar to glatiramer acetate polypeptide. These markers may be used in any molecular size discrimination system using any available molecular weight determination procedure or apparatus. For example, the present markers may be used for calibration of any chromatographic procedure or apparatus which is used for molecular weight determinations of polypeptides. Such a chromatographic apparatus may be a molecular weight sizing column which separates polypeptides on the basis of their molecular size. Examples of molecular weight sizing columns include TSK-GEL® columns, Sephadex™ columns, Sepharose® columns, Sephacryl™ columns, and Superose™ columns.
DEFINITIONS
The following definitions are used in connection with the present invention unless the context indicates otherwise. Sephacryl® is a covalently cross linked dextran/bisacrylamide copolymer gel formed into beads. It is used in gel filtration columns for separating molecules in the size range 5 kD to 1.5 million Dalton. Sephacryl® S-100 has a 1 kD - lOOkD molecular weight range and has a particle size of 25-75 μητι. Sephacryl® is a registered trademark of GE Healthcare. Superose™ 12 is a cross-linked, agarose-based medium optimized for high performance gel filtration of biomolecules. It has an exclusion limit of -2x106 Mr and is a composite of cross-linked agarose with a bead diameter of 8-12 μηη. Superose™ is a trademark of GE Healthcare Ltd. TSK-GEL® is a registered trademark of Tosoh Corporation. Sepharose® is a registered trademark of GE Healthcare.
Certain specific aspects and embodiments of the present application will be explained in greater detail with reference to the following examples, which are provided only for purposes of illustration and should not be construed as limiting the scope of the application in any manner. Reasonable variations of the described procedures are intended to be within the scope of the present invention. While particular aspects of the present invention have been illustrated and described, it would be obvious to those skilled in the art that various other changes and modifications can be made without departing from the spirit and scope of the invention. It is therefore intended to cover in the appended claims all such changes and modifications that are within the scope of this invention. EXAMPLES
EXAMPLE 1 : Preparation of molecular weight markers of glatiramer acetate.
Matrix : Sephacryl™ S-100 HR (high resolution)
Bed volume : 320 mL
Elution mode : Isocratic
Flow rate : 1.8 mL/min
Wave length : 280 nm & 215 nm
Mobile phase: Phosphate buffer (mM), pH 7.2, 7.72 g of NaH2PO4> 20.45 g of Na2HPO4 with 35.05 g of NaCI (150 mM)/4 L water
Sample Preparation: glatiramer acetate (350 mg) is dissolved in Milli-Q® water (10 mL) and filtered through 0.45 μητι syringe filter.
The column is stabilized with the above prepared mobile phase. The flow rate of the mobile phase is kept constant and stabilization is continued until base line reaches constant value. Sample is loaded on to the column at the same flow rate through pump B assembly. The set mark option is used to indicate injection marking. Absorbencies are recorded at dual wavelengths such as 215 nm & 280 nm. At nearly 111 mL of elution volume, the peak at 280 nm starts rising up. The fraction collector is activated to deliver 9 mL of each fraction. At 180 mL of elution volume the peak reaches its maximum value. Mobile phase flow still continues. At around 300 mL of elution volume, the peak almost reaches its baseline. Around 15 fractions are collected (9 mL of each volume), the fractionation is stopped. Of the collected fractions, one fraction from the rising peak, two near the peak apex and one at the tailing peak are chromatographed on Superose™ 12 column and the peak retention time for each of the fractions recorded. Each of these fractions elutes at a distinct retention time.
The same procedure was repeated for numerous batches and the selected fractions eluted at the same retention time in the Superose™-12 column when their elution time and elution volume during fractionation were constant. The collected fractions are lyophilized individually to give 80-90 mg each.
The four selected fractions, at about RT 23.0 minutes, at about RT 26 minutes, at about RT 29 minutes and at about RT 30 minutes are then concentrated through a 10 KDa molecular weight cutoff membrane, a 3 KDa molecular weight cutoff membrane, a 3 KDa molecular weight cutoff membrane and a 3 KDa molecular weight cut off membrane respectively. The concentrated solutions are lyophilized individually to afford 15 mg, 30 mg, 35 mg, and 12 mg of the title compound.
Confirmation of molecular weight of the markers of the present application by MALDI (Matrix-Assisted Laser Desorption/lonization) is given below:
Figure imgf000012_0001
EXAMPLE 2: Preparation of glatiramer acetate molecular weight markers.
Matrix : Sephacryl™ S-100 HR (high resolution)
Bed volume : 900 mL
Bed height : 460 mm
Elution mode : Isocratic
Flow rate : 4.8 mL/min
Wave length : 280 nm & 215 nm
Mobile phase: Phosphate buffer (mM), pH 7.2, 7.72 g of NaH2P04, 20.45 g of Na2HPO4 with 1 1.688 g of NaCI/4 L water
Sample Preparation: glatiramer acetate (1500 mg) is dissolved in Milli-Q® water (10 mL) and filtered through 0.45 μιη syringe filter.
The column is stabilized with the above prepared mobile phase. The flow rate of the mobile phase is kept constant and stabilization is continued until base line at absorbance @ 215 nm reaches constant value. Sample is loaded on to the column at the same flow rate through pump B assembly. The set mark option is used to indicate injection loading. Absorbencies are recorded at dual wavelengths such as 2 5 nm & 280 nm. At nearly 420 mL of elution volume, the peak at 215 nm starts rising up. The fraction collector is activated to deliver 10 mL of each fraction. At nearly 640 mL of elution volume the peak reaches its maximum value. Mobile phase flow still continues. At around 1025 mL of elution volume, the peak almost reaches its baseline. The fractionation is stopped. Two consecutive fractions from the fraction collector are mixed together to get the required fraction volume of 20 mL each. After fractionation, the column is stabilized with mobile phase for the next cycle. The same procedure was repeated for numerous batches. Pooled fractions from the each batch were chromatographed on Superose™ 12 column and the peak retention time for each of the fractions recorded. Each of these fractions elutes at a distinct retention time as given in tables 1 , 2, & 3 respectively. Fractions with similar retention time from different batches are pooled together for further processing.
Table 1
Figure imgf000013_0001
Table 2
Bate h No. 2
Fraction No. Retention time (minutes)
Fraction No. 1 & 2 Low response
Fraction No. 3& 4 24.465
Fraction No. 5&6 25.362
Fraction No. 7&8 26.185
Fraction No. 1 1 &12 27.890
Fraction No.15&16 29.582
Fraction No. 19 & 20 31 .095
Fraction No. 23 & 24 33.054
Fraction No. 27 &28 36.271
Table 3
Figure imgf000014_0001
The solution (60 mL) from each pooled fractions (Fractions 3 & 4 to Fractions 19 & 20) are poured in to the Centrifugal CutOff membrane (CutOff membrane of 5KDa from GE Amersham Life science) and centrifuged at 25°C and 3000 RPM of speed for 60 minutes till the volume concentrated to 20 mL. Milli Q water (20 mL) is added to the retentate side of the tube for further washing and again centrifuged until 20 mL removal in permeate side. The retentate solution from each of the membrane tube was collected separately. The concentrated solutions are lyophilized individually to afford 150 mg, 187 mg, 292 mg, 537 mg, 580 mg, and 277 mg of the title compound. Confirmation of molecular weight of the markers of the present application by MALDl (Matrix-Assisted Laser Desorption/lonization) is given below:
Table 4
Molecular
No. Fraction No weight Molar fraction
(Daltons)
Glutamic
Alanine Lysine Tyrosine acid
Fraction No.
1 23.731 14600 - - - - 3 & 4
Fraction No.
2 24.723 12000 0.147 0.465 0.083 0.305
5 & 6
Fraction No.
3 25.764 9600 0.153 0.476 0.084 0.287
7 & 8
Fraction No.
4 27.564 6614 0.153 0.472 0.090 0.284
1 1 &12
Fraction
5 29.268 4460 0.130 0.444 0.094 0.332 No.15 &16
Fraction No.
6 30.833 3600 0.147 0.437 0.1 16 0.300
19 & 20 EXAMPLE 3: Superose™ 12 Column Calibration with molecular weight markers of the present application.
Molecular weight markers that are prepared according to the procedure given in Example 1 above and a glatiramer acetate preparation are expected to demonstrate a similar correlation between retention time (RT) and log molecular weight. The molecular weight markers of the present application are chromatographed on Superose™ 12 column. The peak retention time for each of the markers is recorded. The linear correlation between Log Molecular Weight (MW) and the Retention Time (RT) is calculated as follows:
Log MW=A+B X RT
where MW is the molecular weight, RT is the retention time, A and B, respectively, are the intercept and the slope of the calculated regression function.
Figure imgf000015_0002
In the similar manner, molecular weight markers that are prepared according to the procedure given in Example 2 above and a glatiramer acetate preparation are expected to demonstrate a similar correlation between retention time (RT) and log molecular weight. The molecular weight markers of the present application are chromatographed on Superose™ 12 column. The peak retention time for each of the markers is recorded. The linear correlation between Log Molecular Weight (MW) and the Retention Time (RT) is calculated as follows:
Log MW=A+B X RT
where MW is the molecular weight, RT is the retention time, A and B, respectively, are the intercept and the slope of the calculated regression function.
Figure imgf000015_0001
Figure imgf000016_0001
Before utilizing these markers for determining the molecular weight of Copaxone® or any other pharmaceutical composition consisting of glatiramer acetate , it is essential to establish a structural correlation between the markers and glatiramer acetate. The CD (Circular Dichroism) profile of the molecular weight markers that are prepared according to the procedure given in Example 1 above were compared against Copaxone® and the two profiles were found to be similar, indicating their structural homogeneity.
The Superose™-12 column pre-calibrated in the above manner was then used to determine the molecular weight of Copaxone® lots of innovator and different batch samples glatiramer acetate. The data in the table below shows that the results are well within the specified limits of 5 to 9KDa proposed for Copaxone®.
Figure imgf000016_0002
Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art as known to those skilled therein as of the date of the invention described and claimed herein.
While particular embodiments of the present invention have been illustrated and described, it would be obvious to those skilled in the art that various other changes and modifications can be made without departing from the spirit and scope of the invention. It is therefore intended to cover in the appended claims all such changes and modifications that are within the scope of this invention.

Claims

CLAIMS:
1. A molecular weight marker having an amino acid composition corresponding to glatiramer acetate and an identified molecular weight which is between about 2,000 Daltons and about 40,000 Daltons.
2. The molecular weight marker of claim 1 , wherein the identified molecular weight is between about 2,000 Daltons and about 20,000 Daltons.
3. The molecular weight marker of claim 1 , wherein the identified molecular weight is between about 2,000 Daltons and about 15,000 Daltons.
4. The molecular weight marker of claim 1 , wherein the identified molecular weight is between about 2,000 Daltons and about 10,000 Daltons.
5. The molecular weight marker of claim 1 , consisting essentially of the amino acids alanine, glutamic acid, tyrosine, and lysine in molar fractions of from about 0.38 to about 0.50 alanine, from about 0.13 to about 0.15 glutamic acid, from about 0.08 to about 0.10 tyrosine, and from about 0.3 to about 0.4 lysine.
6. The molecular weight marker of claim 1 , which is molecular weight marker 1 with a peak average molecular weight of 16833 Daltons, molecular weight marker 2 with a peak average molecular weight of 8908 Daltons, molecular weight marker 3 with a peak average molecular weight of 5006 Daltons, or molecular weight marker 4 with a peak average molecular weight of 3709 Daltons.
7. A processes for preparing a molecular weight marker of claim 1 comprising:
(a) fractionating the glatiramer acetate polypeptide in a gel permeation chromatography (GPC) column;
(b) passing the fractions collected in step (a) through molecular weight cut-off membranes; and
(c) isolating the molecular weight markers by lyophilizing the fractions collected from step (b).
8. The process of claim 7, wherein the gel permeation chromatography (GPC) column of step (a) is Superdex™, a composite medium based on highly cross-linked porous agarose particles to which dextran have been covalently bonded, Sephacryl™, an allyl dextran cross-linked with N, N'-methylene bisacrylamide, Superose™, a medium with high physical and chemical stability based on highly cross-linked porous agarose particles, or Sephadex™, a dextran cross-linked with epichlorohydrin.
9. The process of claim 7, wherein the molecular cut-off membranes used in step (b) range from 1 KDa to 10 KDa, 1 KDa to 20 KDa, or 1 KDa to 30 KDa.
10. A method for determining the peak average molecular weight of glatiramer acetate or another pharmaceutically acceptable salt thereof, which comprises subjecting the glatiramer acetate or another pharmaceutically acceptable salt thereof to chromatography on a chromatographic column so as to determine the average molecular weight of the glatiramer acetate or a pharmaceutically acceptable salt thereof, wherein the chromatographic column is calibrated by using a plurality of molecular weight markers of claim 1.
1 1. A process of calibration of a gel permeation chromatography column by using a molecular weight marker of claim 1.
PCT/US2011/045724 2010-07-29 2011-07-28 Glatiramer acetate molecular weight markers WO2012016042A2 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
AU2011282635A AU2011282635B2 (en) 2010-07-29 2011-07-28 Glatiramer acetate molecular weight markers
NZ606723A NZ606723A (en) 2010-07-29 2011-07-28 Glatiramer acetate molecular weight markers
MX2013001150A MX2013001150A (en) 2010-07-29 2011-07-28 Glatiramer acetate molecular weight markers.
US13/812,684 US9109006B2 (en) 2010-07-29 2011-07-28 Glatiramer acetate molecular weight markers
CN201180041836.0A CN103080747B (en) 2010-07-29 2011-07-28 glatiramer acetate molecular weight marker
CA2806997A CA2806997A1 (en) 2010-07-29 2011-07-28 Glatiramer acetate molecular weight markers
EP11813184.6A EP2598889A4 (en) 2010-07-29 2011-07-28 Glatiramer acetate molecular weight markers
RU2013108828/15A RU2013108828A (en) 2010-07-29 2011-07-28 MARKERS OF MOLECULAR MASS OF GLATIAMETER ACETATE
KR1020137003967A KR20130043196A (en) 2010-07-29 2011-07-28 Glatiramer acetate moleuclar weight markers
BR112013002230A BR112013002230A2 (en) 2010-07-29 2011-07-28 glatiramer acetate molecular weight markers
JP2013521976A JP2013535474A (en) 2010-07-29 2011-07-28 Molecular weight marker for glatiramer acetate
ZA2013/00980A ZA201300980B (en) 2010-07-29 2013-02-06 Glatiramer acetate molecular weight markers

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
IN2146CH2010 2010-07-29
IN2146/CHE/2010 2010-07-29
US38936010P 2010-10-04 2010-10-04
US61/389,360 2010-10-04

Publications (2)

Publication Number Publication Date
WO2012016042A2 true WO2012016042A2 (en) 2012-02-02
WO2012016042A3 WO2012016042A3 (en) 2012-05-31

Family

ID=45530721

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/045724 WO2012016042A2 (en) 2010-07-29 2011-07-28 Glatiramer acetate molecular weight markers

Country Status (13)

Country Link
US (1) US9109006B2 (en)
EP (1) EP2598889A4 (en)
JP (1) JP2013535474A (en)
KR (1) KR20130043196A (en)
CN (1) CN103080747B (en)
AU (1) AU2011282635B2 (en)
BR (1) BR112013002230A2 (en)
CA (1) CA2806997A1 (en)
MX (1) MX2013001150A (en)
NZ (1) NZ606723A (en)
RU (1) RU2013108828A (en)
WO (1) WO2012016042A2 (en)
ZA (1) ZA201300980B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104098655A (en) * 2013-04-09 2014-10-15 深圳翰宇药业股份有限公司 Polypeptides of mass spectrography internal standard used for synthesis of glatiramer acetate
US9029507B2 (en) 2011-02-14 2015-05-12 Usv Limited Copolymer-1, process for preparation and analytical methods thereof
EP3147667A1 (en) * 2015-09-24 2017-03-29 CHEMI S.p.A. Analysis of the molecular weight distribution of complex polypeptide mixtures

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3290432B1 (en) * 2015-04-28 2022-03-30 Hybio Pharmaceutical Co., Ltd. High performance liquid chromatography method for polypeptide mixtures
CA3050086A1 (en) 2017-03-26 2018-10-04 Mapi Pharma Ltd. Glatiramer depot systems for treating progressive forms of multiple sclerosis
KR102438261B1 (en) * 2017-12-14 2022-08-30 엘지디스플레이 주식회사 Display Device
JP7273996B2 (en) * 2019-12-20 2023-05-15 株式会社日立ハイテク Molecular weight marker for electrophoresis, nucleic acid fractionation method, and nucleic acid size analysis method

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL113812A (en) 1994-05-24 2000-06-29 Yeda Res & Dev Copolymer-1 pharmaceutical compositions containing it and its use
US6800287B2 (en) * 1998-09-25 2004-10-05 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US6514938B1 (en) * 1998-09-25 2003-02-04 Yeda Research And Development Co. Ltd. At The Weizmann Institute Of Science Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US20030157720A1 (en) 2002-02-06 2003-08-21 Expression Technologies Inc. Protein standard for estimating size and mass
SI1797109T1 (en) * 2004-09-09 2016-07-29 Yeda Research And Development Co., Ltd. Mixtures of polypeptides, compositions containing and processes for preparing same, and uses thereof
WO2006050122A1 (en) * 2004-10-29 2006-05-11 Sandoz Ag Processes for preparing glatiramer
AU2006211510B8 (en) * 2005-02-02 2011-04-21 Teva Pharmaceutical Industries, Ltd. Process for producing polypeptide mixtures using hydrogenolysis
WO2007030573A2 (en) * 2005-09-09 2007-03-15 Yeda Research And Development Co. Ltd. Polypeptides useful for molecular weight determinations
US20090111133A1 (en) 2007-10-31 2009-04-30 Abbott Laboratories Gel Filtration Standard
EP2215460A4 (en) * 2007-11-26 2010-12-29 Waters Technologies Corp Internal standards and methods for use in quantitatively measuring analytes in a sample
BR112012027753A2 (en) * 2010-04-27 2017-01-10 Reddys Lab Inc Dr preparation of polypeptides and salts thereof
ES2654291T3 (en) * 2011-02-14 2018-02-13 Usv Private Limited Copolymer-1, preparation process and its methods of analysis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2598889A4 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9029507B2 (en) 2011-02-14 2015-05-12 Usv Limited Copolymer-1, process for preparation and analytical methods thereof
CN104098655A (en) * 2013-04-09 2014-10-15 深圳翰宇药业股份有限公司 Polypeptides of mass spectrography internal standard used for synthesis of glatiramer acetate
CN104098655B (en) * 2013-04-09 2018-01-30 深圳翰宇药业股份有限公司 Target polypeptide in mass spectrum for synthesizing acetic acid copaxone
EP3147667A1 (en) * 2015-09-24 2017-03-29 CHEMI S.p.A. Analysis of the molecular weight distribution of complex polypeptide mixtures
US10234461B2 (en) 2015-09-24 2019-03-19 Chemi S.P.A. Analysis of the molecular weight distribution of complex polypeptide mixtures

Also Published As

Publication number Publication date
BR112013002230A2 (en) 2016-05-24
US9109006B2 (en) 2015-08-18
NZ606723A (en) 2015-11-27
CA2806997A1 (en) 2012-02-02
EP2598889A4 (en) 2014-01-22
KR20130043196A (en) 2013-04-29
ZA201300980B (en) 2014-03-26
AU2011282635B2 (en) 2015-05-28
US20130205877A1 (en) 2013-08-15
RU2013108828A (en) 2014-09-10
AU2011282635A1 (en) 2013-02-28
MX2013001150A (en) 2013-05-30
CN103080747A (en) 2013-05-01
CN103080747B (en) 2016-04-27
JP2013535474A (en) 2013-09-12
WO2012016042A3 (en) 2012-05-31
EP2598889A2 (en) 2013-06-05

Similar Documents

Publication Publication Date Title
US9109006B2 (en) Glatiramer acetate molecular weight markers
EP2117685B1 (en) Method for making cross-linked cellulose membranes
US10457705B2 (en) Carrier for ligand immobilization
EP3290432B1 (en) High performance liquid chromatography method for polypeptide mixtures
KR20120124412A (en) Specific sorbent for binding proteins and peptides, and separation method using the same
EP2675824A2 (en) Copolymer-1, process for preparation and analytical methods thereof
JP6764464B2 (en) Porous crosslinked cellulose gel and its manufacturing method
Bund et al. Chromatographic purification and characterization of whey protein–dextran glycation products
EP3512866A1 (en) The use of a polymeric mesh for the purification of macromolecules
KR20100070994A (en) Packing material for liquid chromatography and process for separation and purification of biopolymer by means of the packing material
JP2022184990A (en) Composite material for bioseparation
CN110791491A (en) Method for extracting defibrase from snake venom
JP2010145240A (en) Liquid chromatography filler for immobilizing hydrophobic amino acid or aminomethyl benzoic acid, and method for separating, refining, collecting and recovering biological polymer using the same
JP2008069073A (en) Lactoferrin conjugate and its manufacturing method
WO2015044876A1 (en) Molecular weight markers for protected polypeptides
JP7344232B2 (en) Composite materials for bioseparation
KR102020995B1 (en) A method of preparing gcsf and polyol_conjugated conjugates with high yield
CN113720955A (en) Method for detecting ganirelix acetate high-molecular polymer
WO2019192877A1 (en) Cex chromatography media and low salt elution of target proteins from biopharmaceutical feeds
Khabarov et al. HPLC study of the physicochemical characteristics of chitosan preparations during their manufacture and storage
JPS60161923A (en) Preparation of human urogastrone
JPH0118775B2 (en)

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180041836.0

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11813184

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2013521976

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2806997

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 224474

Country of ref document: IL

Ref document number: MX/A/2013/001150

Country of ref document: MX

REEP Request for entry into the european phase

Ref document number: 2011813184

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011813184

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20137003967

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A201302457

Country of ref document: UA

ENP Entry into the national phase

Ref document number: 2011282635

Country of ref document: AU

Date of ref document: 20110728

Kind code of ref document: A

Ref document number: 2013108828

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13812684

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013002230

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013002230

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20130129