US20110060279A1 - Reduced Volume Formulation of Glatiramer Acetate and Methods of Administration - Google Patents

Reduced Volume Formulation of Glatiramer Acetate and Methods of Administration Download PDF

Info

Publication number
US20110060279A1
US20110060279A1 US12/761,367 US76136710A US2011060279A1 US 20110060279 A1 US20110060279 A1 US 20110060279A1 US 76136710 A US76136710 A US 76136710A US 2011060279 A1 US2011060279 A1 US 2011060279A1
Authority
US
United States
Prior art keywords
injection
study
assisting device
syringe
glatiramer acetate
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/761,367
Other languages
English (en)
Inventor
Ayelet Altman
Doris Saltkill
Tomer El-Gad
Dalton L. Tomlinson
Paul Greenbalch
David George Robinson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Team Consulting Ltd
Teva Pharmaceutical Industries Ltd
Original Assignee
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=42283140&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110060279(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US12/761,367 priority Critical patent/US20110060279A1/en
Priority to US12/785,125 priority patent/US7855176B1/en
Assigned to TEVA PHARMACEUTICAL INDUSTRIES, LTD. reassignment TEVA PHARMACEUTICAL INDUSTRIES, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EL-GAD, TOMER, ALTMAN, AYELET, SALTKILL, DORIS, TOMLINSON, DALTON L.
Priority to PCT/US2010/001972 priority patent/WO2011008274A2/en
Priority to JP2012520598A priority patent/JP2012533540A/ja
Priority to AU2010273234A priority patent/AU2010273234A1/en
Priority to US13/384,021 priority patent/US8920373B2/en
Priority to EP10800156A priority patent/EP2453907A2/en
Priority to BR112012000878A priority patent/BR112012000878A2/pt
Priority to MX2012000687A priority patent/MX2012000687A/es
Priority to EA201270167A priority patent/EA201270167A1/ru
Priority to ARP100102598A priority patent/AR077484A1/es
Priority to US12/948,611 priority patent/US9018170B2/en
Assigned to TEAM CONSULTING LIMITED reassignment TEAM CONSULTING LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GREENHALGH, PAUL, ROBINSON, DAVID GEORGE
Assigned to TEVA PHARMACEUTICAL INDUSTRIES, LTD. reassignment TEVA PHARMACEUTICAL INDUSTRIES, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TEAM CONSULTING LIMITED
Priority to TW100103482A priority patent/TWI435721B/zh
Publication of US20110060279A1 publication Critical patent/US20110060279A1/en
Priority to IL217240A priority patent/IL217240A0/en
Priority to ZA2012/00586A priority patent/ZA201200586B/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/02Peptides of undefined number of amino acids; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Definitions

  • MS Multiple sclerosis
  • CNS central nervous system
  • RRMS relapsing-remitting
  • RRMS progressive course leading to neurologic deterioration and disability.
  • RRMS is the most common form of the disease (1) which is characterized by unpredictable acute episodes of neurological dysfunction (relapses), followed by variable recovery and periods of clinical stability.
  • SP secondary progressive
  • SP secondary progressive
  • PP primary progressive
  • MS axonal damage and associated inflammation
  • a confluence of elements must be present for MS to occur: genetic predisposition, immune dysregulation and one or more environmental factors.
  • prevalence varies considerably around the world, MS is the most common cause of chronic neurological disability in young adults (3,4).
  • Anderson et al. estimated that there were about 350,000 physician-diagnosed patients with MS in the United States in 1990 (approx. 140 per 100,000 population) (5). It is estimated that about 2.5 million individuals are affected worldwide (6). In general, there has been a trend toward an increasing prevalence and incidence of MS worldwide, but the reasons for this trend are not fully understood (5).
  • Glatiramer acetate is the active substance in Copaxone®, a marketed product indicated for reduction of the frequency of relapses in patients with RRMS.
  • Glatiramer acetate consists of the acetate salts of synthetic polypeptides containing four naturally occurring amino acids: L-glutamic acid, L-alanine, L-tyrosine and L-lysine. The average molecular weight of glatiramer acetate is between 5,000 and 9,000 Daltons.
  • the marketed medicinal product, Copaxone® contains 20 mg glatiramer acetate and 40 mg mannitol in 1.0 ml water for injection.
  • GA mechanism of action addresses the main pathological mechanisms driving MS, i.e. anti-inflammation, remyelination and neuroprotection (prevention of axonal loss) (11).
  • GA binds HLA class II (DR) on antigen-presenting cells in lymph nodes.
  • DR HLA class II
  • GA can block the activation of myelin-reactive T cells or render these cells anergic.
  • GA induces GA-specific Th2 cells that cross the blood-brain barrier (BBB) and produce bystander suppression as a result of cross-recognition of myelin antigens.
  • BBB blood-brain barrier
  • These cells secrete both anti-inflammatory cytokines as well as neurotrophic factors and therefore induce both anti-inflammatory and neuroprotective functions (12).
  • Clinical experience with GA consists of information obtained from completed and ongoing clinical trials and from post-marketing experience.
  • the clinical program includes three double-blind, placebo-controlled studies in RRMS subjects treated with GA 20 mg/day (13,14,15).
  • the relapse rate was reduced by 32% from 1.98 under placebo to 1.34 under GA 20 mg.
  • GA 20 mg has also demonstrated beneficial effects over placebo on MRI parameters relevant to RRMS.
  • a significant effect in median cumulative number of Gd-enhancing lesions over 9 months of treatment 11 lesions in the 20 mg group compared to 17 lesions under placebo) was demonstrated.
  • the clinical program with GA also includes one double-blind study in chronic-progressive MS subjects (16), one double-blind placebo-controlled study in primary progressive patients (17), one double-blind placebo-controlled study in CIS patients (20,21) and numerous open-label and compassionate use studies, mostly in RRMS.
  • the clinical use of GA has been extensively reviewed and published in the current literature (18,19,22,23).
  • IPIR Immediate Post-Injection Reaction
  • injection-site reactions were seen to be the most frequent adverse reactions and were reported by the majority of patients receiving GA. In controlled studies, the proportion of patients reporting these reactions, at least once, was higher following treatment with GA (70%) than placebo injections (37%). The most commonly reported injection-site reactions, which were more frequently reported in GA vs. placebo-treated patients, were erythema, pain, mass, pruritus, edema, inflammation and hypersensitivity.
  • Such formulations suffer from poor shelf-life, unacceptable turbidity, changes in pH, chemical degradation including hydrolysis and aggregation (both reversible and irreversible) and increases in viscosity; all of which potentially reduce shelf-life and bioavailability (25).
  • Drug administration by subcutaneous injection results in delivery of the drug to the interstitial area underneath the skin.
  • the fluid environment of the interstitial space is essentially that of plasma although the constituent proteins are at a lower concentration. This physiological medium may conflict with the solubility characteristics of the concentrated peptide drug (26).
  • the interaction of the delivered drug with the interstitial environment dictates the pattern of absorption of the peptide.
  • Formulation characteristics particularly concentration, injection volume and pH, influence the rate of diffusion and absorption by the patient. Because the interstitium also comprises a fibrous matrix of collagen and glycosaminoglycans, it acts as a barrier to the diffusion and permeability of the drug.
  • drugs delivered in a concentrated form to the interstitial space may be susceptible to enzymatic degradation at the injection site, precipitation and/or aggregation in the interstitial fluid, and endocytic/phagocytic mechanisms (26).
  • a peptide drug product such as glatiramer acetate
  • clinical testing is therefore required to determine whether any modification can effectively reduce the number and severity of injection-site reactions while still substantially maintaining therapeutic efficacy.
  • This invention provides a method for reducing frequency of relapses in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS) comprising administering to the patient by subcutaneous injection 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • RRMS relapsing-remitting multiple sclerosis
  • the invention also provides a method for reducing the frequency of relapse in a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least one lesion consistent with multiple sclerosis comprising administering to the patient by subcutaneous injection 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • This invention also provides an injection, assisting device comprising:
  • This invention provides a syringe pre-filled with 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • This invention also provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing frequency of relapses in a human patient afflicted with relapsing, remitting multiple sclerosis (RRMS).
  • aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing frequency of relapses in a human patient afflicted with relapsing, remitting multiple sclerosis (RRMS).
  • This invention additionally provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing the frequency of relapse in a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least one lesion consistent with multiple sclerosis.
  • This invention also provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing the frequency of relapse in a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has a high risk of developing clinically definite multiple sclerosis (CDMS).
  • CDMS clinically definite multiple sclerosis
  • This invention provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing the frequency of relapse in a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least two clinically silent MRI lesions characteristic of multiple sclerosis.
  • This invention further provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol in the manufacture of a medicament for treating a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol in the manufacture of a medicament for treating a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • This invention yet further provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol in the manufacture of a medicament for treating a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least one lesion consistent with multiple sclerosis.
  • This invention also provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in treating a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has been determined to be at high risk of developing clinically definite multiple sclerosis (CDMS).
  • CDMS clinically definite multiple sclerosis
  • This invention provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol in the manufacture of a medicament for treating a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least two clinically silent MRI lesions characteristic of multiple sclerosis.
  • This invention provides a pharmaceutical composition for use in treating a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS) comprising a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • RRMS relapsing-remitting multiple sclerosis
  • This invention also provides a pharmaceutical composition for use in treating a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least one lesion consistent with multiple sclerosis comprising a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • This invention provides a method of treating a patient suffering from a relapsing form of multiple sclerosis which comprises periodically administering to the patient by subcutaneous injection a 20 mg dose of a pharmaceutical composition, wherein the subcutaneous injection is delivered by automatic injection and wherein the pharmaceutical composition comprises 20 mg of glatiramer acetate in 0.5 ml of solution so as to thereby treat the patient.
  • This invention provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in decreasing the frequency of clinical exacerbations or reducing the number and volume of active MRI brain lesions in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in decreasing the frequency of clinical exacerbations or reducing the number and volume of active MRI brain lesions in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • This invention also provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in delaying the onset of Clinically Definite Multiple Sclerosis or decreasing the number and volume of active MRI brain lesions in a human patient who experienced a single demyelinating event and who is considered to be at risk of developing Clinically Definite Multiple Sclerosis.
  • This invention provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for decreasing the frequency of clinical exacerbations or reducing the number and volume of active MRI brain lesions in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for decreasing the frequency of clinical exacerbations or reducing the number and volume of active MRI brain lesions in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • This invention also provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for delaying the onset of clinically definite multiple sclerosis or decreasing the number and volume of active MRI brain lesions in a human patient who experienced a single demyelinating event and who is considered to be at risk of developing clinically definite multiple sclerosis.
  • This invention provides a pharmaceutical composition for use in treating a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS) comprising a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • RRMS relapsing-remitting multiple sclerosis
  • This invention provides a pharmaceutical composition for use in treating a human patient who experienced a single demyelinating event and who is considered to be at risk of developing clinically definite multiple sclerosis comprising a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • FIG. 1 is a diagrammatic representation of FIG. 1 .
  • FIG. 2 is a diagrammatic representation of FIG. 1 .
  • FIG. 3 is a diagrammatic representation of FIG. 3 .
  • FIG. 4 (A-D).
  • FIG. 5 (A-D).
  • FIG. 6 is a diagrammatic representation of FIG. 6 .
  • FIG. 7 (A-C).
  • the “autoject2 for glass syringe” is an automatic injection device for use with the 1 ml long glass syringe for the subcutaneous injection of approved drugs.
  • the insert contains the instructions for use of the “autoject2 for glass syringe”.
  • “autoject2 for glass syringe” For instructions regarding the use of the drug, refer to the drug Patient Information Booklet provided with the drug product.
  • FIG. 7A represents the parts of the auto-injection device.
  • FIG. 7B is a continuation of the instructions to use the auto-injection device.
  • Step 2 Loading and Using the “Autoject2 for Glass Syringe”
  • FIG. 7C is a further continuation of the instructions to use the autoinjection device.
  • the Needle Cap has two parts: a grey/dark rubber inner part covered by a transparent plastic outer part. Make sure both parts of the Needle Cap are intact when the cap falls out of the Cap Remover. If both parts of the Needle Cap have not been removed, unscrew the Syringe Housing and Injector Body, remove the syringe and start over from instruction step 4 with a new syringe. Save the Cap Remover for future use. Dispose of the Needle Cap.
  • the external components and the inside of the Syringe Housing of the “autoject2 for glass syringe” should be cleaned by wiping with a clean damp doth or on alcohol wipe. Do not immerse in water.
  • This invention provides a method for reducing frequency of relapses in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS) comprising administering to the patient by subcutaneous injection 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • RRMS relapsing-remitting multiple sclerosis
  • the invention also provides a method for reducing the frequency of relapse in a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least one lesion consistent with multiple sclerosis comprising administering to the patient by subcutaneous injection 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • the pH of the aqueous pharmaceutical solution is 5.5 to 7.0.
  • the 20 mg of glatiramer acetate does not form polypeptide aggregates in the 0.5 ml of aqueous pharmaceutical solution.
  • the 20 mg of glatiramer acetate does not precipitate in the subcutaneous environment after injection.
  • the 20 mg of glatiramer acetate is absorbed by the patient after the subcutaneous injection.
  • the 20 mg of glatiramer acetate in 0.5 ml of solution is absorbed by the patient at least as readily as 20 mg of glatiramer acetate in 1 ml of solution.
  • the 20 mg of glatiramer acetate in 0.5 ml of solution is co-injected with a vasodilator.
  • the 20 mg of glatiramer acetate in 0.5 ml of solution is co-injected with a vasoconstrictor.
  • 20 mg of glatiramer acetate in 0.5 ml of solution is co-injected with an extracellular matrix-modifying enzyme.
  • the subcutaneous injection is administered to the upper back portion of the arm, to the stomach area outside of a 2 inch area around the navel, to the upper outer-rear quadrant of the buttocks, or to the front and outer area of the thigh 2 inches above the knee and 2 inches below the groin.
  • the pain associated with the subcutaneous injection is reduced relative to pain associated with subcutaneous injection of 1.0 ml of an aqueous pharmaceutical solution of 20 mg glatiramer acetate and 40 mg mannitol.
  • the pain is the patient-reported total injection pain rating on a visual analogue scale (VAS) occurring immediately after injection.
  • VAS visual analogue scale
  • the patient-reported total injection pain rating is reduced by about 27%.
  • the pain is the patient-reported total injection pain rating on a visual analogue scale (VAS) experienced five minutes following subcutaneous injection.
  • VAS visual analogue scale
  • the patient-reported total injection pain rating experienced five minutes following subcutaneous injection is reduced by about 31%.
  • the pain is the immediate pain presence following the subcutaneous injection.
  • the immediate pain presence is reduced by about 19%.
  • the pain is pain presence five minutes following the subcutaneous injection.
  • the pain presence five minutes following the subcutaneous injection is reduced by about 19%.
  • the total number or total severity of Local Injection Site Reactions is reduced relative to the total number or total severity of LISRs associated with subcutaneous injection of 1.0 ml of an aqueous pharmaceutical solution of 20 mg glatiramer acetate and 40 mg mannitol.
  • the total number or total severity of Local Injection Site Reactions (LISRs) five minutes after subcutaneous injection is reduced.
  • the total number of Local Injection Site Reactions (LISRs) five minutes after the subcutaneous injection is reduced by about 24%.
  • the total severity of Local Injection Site Reactions (LISRs) five minutes after the subcutaneous injection is reduced by about 29%.
  • the total number or total severity of Local Injection Site Reactions (LISRs) 24 hours after subcutaneous injection of glatiramer is reduced.
  • the total number of Local Injection Site Reactions (LISRs) 24 hours after the subcutaneous injection is reduced by about 23%.
  • the total severity of Local Injection Site Reactions (LISRs) 24 hours after the subcutaneous injection is reduced by about 25%.
  • the daily five-minute Local Injection Site Reaction (LISR) score is reduced relative to the daily 5-minute LISR score associated with subcutaneous injection of 1.0 ml of an aqueous pharmaceutical solution of 20 mg glatiramer acetate and 40 mg mannitol.
  • LISR Local Injection Site Reaction
  • the daily 24-hour Local Injection Site Reaction (LISR) score is reduced relative to the daily 24-hour LISR score associated with subcutaneous injection of 1.0 ml of an aqueous pharmaceutical solution of 20 mg glatiramer acetate and 40 mg mannitol.
  • LISR Local Injection Site Reaction
  • the Local Injection Site Reactions comprise redness, itching and formation of a lump.
  • the percent of patients who report no Local Injection Site Reactions (LISRs) 5-minutes after injection is increased relative to the percent of patients who report no LISRs 5-minutes after subcutaneous injection of 1.0 ml of an aqueous pharmaceutical solution of 20 mg glatiramer acetate and 40 mg mannitol.
  • LISRs Local Injection Site Reactions
  • the percent of patients who report no Local Injection Site Reactions is increased by 3 fold.
  • the percent of patients who report no Local Injection Site Reactions (LISRs) 24-hours after injection is increased relative to the percent of patients who report no LISRs 24-hours after subcutaneous injection of 1.0 ml of an aqueous pharmaceutical solution of 20 mg glatiramer acetate and 40 mg mannitol.
  • LISRs Local Injection Site Reactions
  • the percent of patients who report no Local Injection Site Reactions is increased by about 50%.
  • the 0.5 ml aqueous pharmaceutical solution is in a pre-filled syringe.
  • the administration is by an automated subcutaneous injection device containing the prefilled syringe and a means for initiating the subcutaneous injection, completing the subcutaneous injection and indicating to the user that the subcutaneous injection of the 0.5 ml aqueous pharmaceutical solution is complete.
  • the 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol is at least as effective as 1.0 ml of an aqueous pharmaceutical solution of 20 mg glatiramer acetate and 40 mg mannitol in reducing the frequency of relapses in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • RRMS relapsing-remitting multiple sclerosis
  • This invention also provides an injection assisting device comprising:
  • the injection assisting device further comprising:
  • the injection assisting device further comprising:
  • a color of the injection lock indicator is configured to substantially contrast with a color of at least one of the first outer shell and the second outer shell, and
  • a color of the attention director is configured to substantially contrast with a color of proximally located components
  • This invention provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • the 20 mg of glatiramer acetate does not form polypeptide aggregates in the 0.5 ml of aqueous pharmaceutical solution.
  • the aqueous pharmaceutical solution has a pH of 5.5-7.0.
  • the aqueous pharmaceutical solution is in a prefilled syringe.
  • This invention also provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing frequency of relapses in a human patient afflicted with relapsing, remitting multiple sclerosis (RRMS).
  • aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing frequency of relapses in a human patient afflicted with relapsing, remitting multiple sclerosis (RRMS).
  • This invention additionally provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing the frequency of relapse in a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least one lesion consistent with multiple sclerosis.
  • This invention also provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing the frequency of relapse in a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has been determined to be at high risk of developing clinically definite multiple sclerosis (CDMS).
  • CDMS clinically definite multiple sclerosis
  • This invention provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in reducing the frequency of relapse in a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least two clinically silent MRI lesions characteristic of multiple sclerosis.
  • This invention further provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol in the manufacture of a medicament for treating a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol in the manufacture of a medicament for treating a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • This invention yet further provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol in the manufacture of a medicament for treating a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least one lesion consistent with multiple sclerosis.
  • This invention also provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in treating a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has been determined to be at high risk of developing clinically definite multiple sclerosis (CDMS).
  • CDMS clinically definite multiple sclerosis
  • This invention provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol in the manufacture of a medicament for treating a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least two clinically silent MRI lesions characteristic of multiple sclerosis.
  • This invention provides a pharmaceutical composition for use in treating a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS) comprising a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • RRMS relapsing-remitting multiple sclerosis
  • This invention also provides a pharmaceutical composition for use in treating a human patient who experienced a first clinical episode consistent with multiple sclerosis and who has at least one lesion consistent with multiple sclerosis comprising a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • This invention provides a method of treating a patient suffering from a relapsing form of multiple sclerosis which comprises periodically administering to the patient by subcutaneous injection a 20 mg dose of a pharmaceutical composition, wherein the subcutaneous injection is delivered by automatic injection and wherein the pharmaceutical composition comprises 20 mg of glatiramer acetate in 0.5 ml of solution so as to thereby treat the patient.
  • the injection of 20 mg glatiramer acetate in 0.5 ml of solution is as effective as injection of 20 mg of glatiramer acetate in 1 ml of solution.
  • the 20 mg of glatiramer acetate in 0.5 ml of solution has a pH equivalent to that of 20 mg of glatiramer acetate in 1 ml of solution.
  • the 20 mg of glatiramer acetate is completely soluble in 0.5 ml of solution.
  • This invention provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in decreasing the frequency of clinical exacerbations or reducing the number and volume of active MRI brain lesions in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in decreasing the frequency of clinical exacerbations or reducing the number and volume of active MRI brain lesions in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • This invention also provides a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for use in delaying the onset of clinically definite multiple sclerosis or decreasing the number and volume of active MRI brain lesions in a human patient who experienced a single demyelinating event and who is considered to be at risk of developing clinically definite multiple sclerosis.
  • This invention provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for decreasing the frequency of clinical exacerbations or reducing the number and volume of active MRI brain lesions in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for decreasing the frequency of clinical exacerbations or reducing the number and volume of active MRI brain lesions in a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS).
  • This invention also provides a use of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol for delaying the onset of clinically definite multiple sclerosis or decreasing the number and volume of active MRI brain lesions in a human patient who experienced a single demyelinating event and who is considered to be at risk of developing clinically definite multiple sclerosis.
  • This invention provides a pharmaceutical composition for use in treating a human patient afflicted with relapsing-remitting multiple sclerosis (RRMS) comprising a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • RRMS relapsing-remitting multiple sclerosis
  • This invention provides a pharmaceutical composition for use in treating a human patient who experienced a single demyelinating event and who is considered to be at risk of developing clinically definite multiple sclerosis comprising a unit dose of 0.5 ml of an aqueous pharmaceutical solution which contains in solution 20 mg glatiramer acetate and 20 mg mannitol.
  • unit dose or pharmaceutical composition disclosed herein adapted for subcutaneous injection to the upper back portion of the arm, to the stomach area outside of a 2 inch area around the navel, to the upper outer-rear quadrant of the buttocks, or to the front and outer area of the thigh 2 inches above the knee and 2 inches below the groin.
  • a patient at risk of developing MS is a patient presenting any of the known risk factors for MS.
  • the known risk factors for MS include anyone of a clinically isolated syndrome (CIS), a single attack suggestive of MS without a lesion, the presence of a lesion (in any of the CNS, PNS, or myelin sheath) without a clinical attack, environmental factors (geographical location, climate, diet, toxins, sunlight) (31, 32, 33), genetics (variation of genes encoding HLA-DRB1, IL7R-alpha and IL2R-alpha) (34, 35), and immunological components (viral infection such as by Epstein-Barr virus, high avidity CD4+ T cells, CD8+ T cells, anti-NF-L, antiCSF114(Glc)) (36, 37, 38).
  • clinically isolated syndrome refers to 1) a single clinical attack (used interchangeably herein with “first clinical event” and “first demyelinating event”) suggestive of MS, which, for example, presents as an episode of optic neuritis, blurring of vision, diplopia, involuntary rapid eye movement, blindness, loss of balance, tremors, ataxia, vertigo, clumsiness of a limb, lack of co-ordination, weakness of one or more extremity, altered muscle tone, muscle stiffness, spasms, tingling, paraesthesia, burning sensations, muscle pains, facial pain, trigeminal neuralgia, stabbing sharp pains, burning tingling pain, slowing of speech, slurring of words, changes in rhythm of speech, dysphagia, fatigue, bladder problems (including urgency, frequency, incomplete emptying and incontinence), bowel problems (including constipation and loss of bowel control), impotence, diminished sexual arousal, loss of sensation, sensitivity to heat,
  • VAS Visual Analogue Scale
  • the VAS is a measurement instrument that tries to measure a characteristic that is believed to range across a continuum of values and cannot easily be directly measured.
  • the amount of pain that a patient feels ranges across a continuum from “no pain” to “worst possible pain”. From the patient's perspective this spectrum appears continuous; their pain does not take discrete jumps, as a categorization of none, mild, moderate and severe would suggest.
  • the VAS is a horizontal line, 100 mm in length, anchored by the above word descriptors at each end. The patient marks on the line the point that they feel represents their perception of their current state. The VAS score is determined by measuring in millimeters from the left hand end of the line to the point that the patient marks.
  • patient-reported total injection pain refers to pain occurring after the injection as recorded on a 100 mm VAS, where 0 mm represents “no pain” and 100 mm represents “worst possible pain.”
  • subcutaneous injection refers to delivery of a bolus to the interstitial area underlying the dermis of the skin.
  • Relapses are characterized by the occurrence of neurological dysfunction symptoms, appearing after a 30-day period of stability or improvement and lasting for more than 24 hours (no infection, no fever). The number of relapses are analyzed using a logistic regression model controlling for treatment and age.
  • Relapse Rate is the number of confirmed relapses per unit time. “Annualized relapse rate” is the mean value of the number of confirmed relapses per each patient multiplied by 365 and divided by the number of days on study drug per each patient.
  • MS MS
  • Benign multiple sclerosis is a retrospective diagnosis which is characterized by 1-2 exacerbations with complete recovery, no lasting disability and no disease progression for 10-15 years after the initial onset. Benign multiple sclerosis may, however, progress into other forms of multiple sclerosis.
  • SPMS may evolve from RRMS. Patients afflicted with SPMS have relapses, a diminishing degree of recovery during remissions, less frequent remissions and more pronounced neurological deficits than RRMS patients. Enlarged ventricles, which are markers for atrophy of the corpus callosum, midline center and spinal cord, are visible on MRI of patients with SPMS.
  • PPMS is characterized by a steady progression of increasing neurological deficits without distinct attacks or remissions. Cerebral lesions, diffuse spinal cord damage and evidence of axonal loss are evident on the MRI of patients with PPMS. PPMS has periods of acute exacerbations while proceeding along a course of increasing neurological deficits without remissions. Lesions are evident on MRI of patients suffering from PRMS (28).
  • CIS clinically isolated syndrome
  • MS such as optic neuritis, brain stem symptoms, and partial myelitis.
  • CDMS clinically definite multiple sclerosis
  • Multiple sclerosis may present with optic neuritis, blurring of vision, diplopia, involuntary rapid eye movement, blindness, loss of balance, tremors, ataxia, vertigo, clumsiness of a limb, lack of co-ordination, weakness of one or more extremity, altered muscle tone, muscle stiffness, spasms, tingling, paraesthesia, burning sensations, muscle pains, facial pain, trigeminal neuralgia, stabbing sharp pains, burning tingling pain, slowing of speech, slurring of words, changes in rhythm of speech, dysphagia, fatigue, bladder problems (including urgency, frequency, incomplete emptying and incontinence), bowel problems (including constipation and loss of bowel control), impotence, diminished sexual arousal, loss of sensation, sensitivity to heat, loss of short term memory, loss of concentration, or loss of judgment or reasoning.
  • relapsing MS includes:
  • relapsing forms of multiple sclerosis include:
  • RRMS Relapsing-remitting multiple sclerosis
  • SPMS Secondary Progressive MS
  • PPM Primary progressive-relapsing multiple sclerosis
  • PRMS progressive-relapsing multiple sclerosis
  • active MRI brain lesions refers to active brain lesions identified on Magnetic Resonance Imaging (MRI) scans.
  • MRI Magnetic Resonance Imaging
  • the unit dose disclosed herein can be administered daily, every other day, weekly, twice weekly, three times weekly, four times weekly, five times weekly or six times weekly.
  • a multicenter, randomized, two arm, single crossover study was undertaken to compare the subject-reported pain of GA 20 mg/1.0 ml (F1) versus GA 20 mg/0.5 ml (F2) administered subcutaneously in subjects with RRMS. Safety and tolerability of the F2 formulation were also assessed. Subjects received both doses once daily in a cross over fashion, for a total treatment duration of five (5) weeks. Subject-reported injection pain was recorded in a daily diary. The primary endpoint was the difference in daily subject-reported injection pain occurring immediately after the injection, for the two GA formulations (F1 versus F2), as recorded on a 100 mm VAS. Secondary objectives included:
  • the purpose of the study was to provide comparative data between a new 20 mg/0.5 ml formulation (F2) of GA and the known profile of the currently marketed 20 mg/1.0 ml formulation (F1). This study was performed in subjects with RRMS who have been on the currently marketed formulation of GA (F1) for a minimum of 90 days.
  • This study was designed to compare subject-reported pain associated with injection of the 20 mg/1.0 ml formulation (F1) of GA versus a new 20 mg/0.5 ml formulation of GA (F2) immediately after and at five minutes following injections. Additionally, the study compared the presence of local injection site reactions (LISRs), severity of LISRs present and adverse events. Evidence was gathered to assess the safety of the 20 mg/0.5 ml formulation.
  • F1 20 mg/1.0 ml formulation
  • F2 new 20 mg/0.5 ml formulation of GA
  • the crossover study design was chosen to allow each eligible subject to use the 20 mg/1.0 ml and the 20 mg/0.5 ml formulation for comparable lengths of time.
  • the two week duration of each cross over period was of sufficient length to determine if differences between the two formulations, as defined by the study outcomes, exist.
  • Each subject served as his/her own control, thereby, reducing subject variability and increasing statistical power in comparison of the two formulations.
  • Blinding in this study was not possible due to the subjects' ability to detect differences in the volume of each formulation. The lack of blinding in this study was a known limitation.
  • Table 2 summarizes subject demographics for the safety population. Overall, the population was predominantly female (81.0%) and white (90.5%). The mean age of the population was 46.0 years with the range between 22 to 71 years. The subject groups receiving Sequence F1/F2 and Sequence F2/F1 were comparable in demographic characteristics.
  • GA injection is a clear, colorless to slightly yellow, sterile, non-pyrogenic solution for subcutaneous injection. It is supplied as a single-use PFS.
  • the marketed medicinal product, Copaxone® contains 20 mg GA and 40 mg mannitol in 1.0 ml of water for injection.
  • the tested, investigational medicinal product (IMP) contains 20 mg GA and 20 mg mannitol in 0.5 ml water for injection.
  • GA is the name used to designate the active ingredient of Copaxone®.
  • study drug was packed and shipped in appropriate storage boxes. Study drug was examined immediately upon arrival at the study center. If the drug supplies appeared to be damaged, the sponsor was contacted immediately. Individual subject's kit should not be open. The tamper proof seal should remain intact until the kit was open when providing the subject with study drug and instructions.
  • Each shipment of drug supplies for the study contained a shipment form to assist in maintaining current and accurate inventory records.
  • the investigator/coordinator/pharmacist acknowledged receipt.
  • Drug supplies must be kept in a secure, limited-access, refrigerated (2° C.-8° C.) and temperature-controlled storage area. Only authorized personnel had access to the study drug at the study centers.
  • the study drugs were dispensed to the subject at the study center by a person authorized by the study investigator at each scheduled visit. Instructions regarding study drug storage were provided to the subject. The subject returned all unused study drug at each visit. The investigator or designee was responsible for performing study drug accountability.
  • the subjects were randomly assigned in a 1:1 assignment ratio to one of two drug sequences; study drugs were distributed to the patient in the order of the sequence (F1/F2 or F2/F1). Block randomization stratified across study was done according to a computer generated schedule to ensure that subjects were distributed equally between the drug sequences.
  • Prior medications for the safety population are summarized in the Table 3 below. All randomized subjects had received prior medications. By design, all subjects were on glatiramer acetate (GA) prior to study, entry. Patient 11/02 inadvertently had the end date of GA listed as the same day as the start of study drug resulting in GA not being listed as a prior medication but as a concomitant medication.
  • GA glatiramer acetate
  • Concomitant medications referred to any medication taken after the first injection of study medication, including those medications that started prior to entry into the study and continued into the study. All medications were allowed except for those medications listed below.
  • Table 4 summarizes the concomitant medications used during the study by >5% of subjects in the safety population.
  • the most frequently used drug classes were multivitamins (27.8% during use of each formulation), selective serotonin reuptake inhibitors (21.5% during 20 mg/1.0 ml GA and 22.9% during 20 mg/0.5 ml GA), other antidepressants (26.4% during 20 mg/1.0 ml GA and 25.7% during 20 mg/0.5 ml GA), benzodiazepine derivatives (21.2% during 20 mg/1.0 ml GA and 22.9% during 20 mg/0.5 ml GA), and propionic acid derivatives (21.5% during use of each formulation).
  • Study drug accountability records were maintained at the site at all times. The identification number of the subject, the date, batch code, expiry date and quantity of study drug dispensed and the date and quantity of study drug returned by the subject were recorded. The returned study drug was noted on the appropriate inventory forms.
  • the subject was requested to return all unused study drug syringes in the original box to the study site at every visit. Compliance with the dosing regimen was determined by performing accountability of returned study drug. The number of returned syringes was counted and recorded by site personnel. The subject number, randomization number, quantity of study drug returned by the subject and visit date were recorded by the site personnel.
  • Compliance with the dosing regimen for each period was determined by performing accountability of returned unused study drug syringes. Compliance was computed as the actual number of used injections (dispensed minus returned) divided by the number of expected number of injections (number of days in the period) times 100 percent. Subjects with compliance ⁇ 75% were considered compliant.
  • Medication compliance was quite high in this trial with the percentage of subjects showing 100% compliance equal to 94.4% following for 20 mg/0.5 ml dose and 91.7% following the 20 mg/1.0 ml dose of GA.
  • Visit 1 screening and first day of dosing for the Run-in Period could occur on the same day. If the first day of dosing could not be done during the screening visit, the first day of dosing must be done within two days of screening. The first day of dosing was captured in the CRF (run-in period).
  • the run-in period was seven (+two) days. Subjects were treated with F1 for the run-in period. The first day of the run-in period was the first day of dosing with study drug and continued for seven days. Run-in Period procedures for the subject consist of:
  • Visit 2 occurred seven (+two) days after the date of first day of dosing in the run-in period.
  • Visit 3 occurred 14 (+three) days after Visit 2/Period 1 starts.
  • Visit 4 occurred 14 (+three) days after the start of Period 2 or at time of early discontinuation.
  • Visit 4/end of study or early discontinuation procedures consisted of:
  • the primary outcome measure was the difference in daily subject-reported injection pain occurring immediately after the injection for the two GA formulations as recorded on a 100 mm VAS, where 0 mm represents “no pain” and 100 mm represents “worst possible pain.”
  • the VAS was scored (i.e., measured) by four central raters. Inter-rater consistency was confirmed as follows; each of the four raters independently measured and scored each of the diary responses and recorded findings on a separate scoring sheet. Scorers provided responses in succession and submitted their scoring sheets to a designated team member in order to blind scores between scorers. Each score was subsequently listed and sent to a statistician for analysis.
  • Adverse events were recorded as soon as the subject signed the Informed Consent Form and throughout the study. Adverse events were reviewed and updated at each subsequent visit and during any phone contact with the subject.
  • AE adverse event
  • any event occurring after the clinical study subject has signed the study Informed Consent was recorded and reported as an AE.
  • An adverse event could therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of an investigational medicinal product, whether or not considered related to the investigational medicinal product.
  • a new condition or the worsening of a pre-existing condition was considered an AE.
  • Stable chronic conditions such as arthritis that were present prior to study entry and do not worsen during the study were not considered AEs.
  • Worsening of the disease under study was measured by clinical impression of the Investigator and was only recorded as an AE if the outcome was more serious than would normally be expected from the normal course of the disease in a particular subject.
  • AEs were recorded as soon as the subject signed the ICF and throughout the study. AES were to be reviewed and updated at each subsequent visit and during any phone contact with the subject.
  • SAE Serious Adverse Event
  • Important medical events were those which may not have been immediately life-threatening, but may have jeopardized the subject and may have required intervention to prevent one of the other serious outcomes listed above. Events such as intensive treatment in an emergency room or at home for allergic bronchospasm; blood dyscrasias or convulsions that do not result in hospitalization, resulting in an adverse event are normally considered serious by this criterion.
  • Inpatient hospitalization or prolongation of existing hospitalization means that hospital inpatient admission and/or prolongation of hospital stay were required for treatment of AE, or that they occurred as a consequence of the event. It does not refer to pre-planned elective hospital admission for treatment of a preexisting condition that has not significantly worsened, or to diagnostic procedure. It does not refer either to hospitalization for I.V. steroid treatment of a relapse, unless it was a worsening of condition beyond expected disease progression.
  • pregnancies including normal pregnancies without an AE, were to be reported to the CRO for inclusions to the safety database. Pregnancies were to be followed up to determine outcome, including spontaneous or voluntary termination, details of birth, presence, or absence of any birth defect, congenital abnormalities, or maternal and newborn complications.
  • Pregnancy report forms and pregnancy follow-up forms were to be provided by the CRO.
  • the pregnancies reporting procedure was the same as SAE reporting procedure.
  • Visit 1 Screening
  • Visit 4 End of study
  • Examinations included mental status, pupil and fundi, cranial nerves, motor examination, gait (if not wheelchair bound), coordination, reflexes and sensory function.
  • Descriptive statistics for continuous variables consisted of N, mean, median, standard deviation (SD), minimum, and maximum values.
  • SD standard deviation
  • minimum minimum
  • maximum values the number and percent (%) of each category are displayed.
  • Baseline was defined as the last assessment prior to the first injection of study medication. For clinical outcomes analyses, baseline was any measurement just prior to starting the Period 1 study treatment. For safety, baseline was defined as the measurements assessed just prior to starting the run-in period treatment (non-missing screening visit).
  • “Screened subjects” refers to those subjects that signed an informed consent and had screening assessments.
  • Demographics and background variables included age, gender, race, height, and weight and were summarized using descriptive statistics. Age was calculated as the integer portion of the date of the screening visit minus the date of birth, divided by 365.25 days per year. Height and weight are presented in cm and kg.
  • Protocol violations/deviations identified by the monitors and data management were discussed prior to data freeze. A review of the data including, but not limited to, inclusion/exclusion criteria, drug and diary compliance, and prohibited medications to exclude subjects from the PP population was made by the sponsor prior to data freeze. Unless a documented decision of protocol violations/deviations inclusion was made, no subjects were allowed to be removed from statistical summaries.
  • Exposure was defined as duration of treatment and was calculated for the run-in period as well as from Day 1 (the day of first dose for each period) to the last dose of each period. Total exposure was the summation of the exposure from both periods, summarized descriptively and by drug formulation.
  • the primary clinical outcomes variable was the total injection pain rating occurring immediately after injection (i.e., Immediate Total Pain score).
  • the null hypothesis was that there was no difference between the 2 study drug formulations.
  • the primary analysis for testing the Immediate Total Pain score was based on the analysis of variance (ANOVA) model for a 2-treatment crossover study with treatment, sequence, and period as fixed effect terms, and a random effect term of subject within sequence. The corresponding 95% confidence interval for the treatment difference in scores is presented.
  • ANOVA analysis of variance
  • Tolerability was based on between group comparisons in subjects prematurely discontinuing the study due to AEs, injection site pain or reactions, and laboratory abnormalities.
  • the Kaplan-Meier product-limit algorithm was applied to compute the time to discontinuation curves, the median event time, and the 95% confidence interval for the median for each study drug formulation.
  • AEs were classified by system organ class (SOC) and preferred term using the Medical Dictionary for Regulatory Affairs (MedDRA) dictionary version 11.1 and the incidence summarized by study drug formulation.
  • SOC system organ class
  • MedDRA Medical Dictionary for Regulatory Affairs
  • a treatment-related AE was defined as an AE considered possibly or probably related to the study drug by an investigator. The most severe occurrence of each preferred term and the most related occurrence of each preferred term were selected for each subject.
  • Clinical Laboratory Tests Clinical laboratory assessments, including tests from hematology and chemistry were summarized using descriptive statistics, by study drug formulation at baseline (Screening) and at each assessment time point, including change from baseline, during the study.
  • Vital Signs Vital signs, including blood pressure, pulse rate, and temperature were summarized using descriptive statistics, by study drug formulation at baseline and at each assessment time point, including change from baseline, during the study
  • Neurological Examinations Neurological examination changes from baseline were summarized in a shift table for: mental status, pupil and fundi, cranial nerves, motor examination, gait (if not wheelchair bound), coordination reflexes and sensory function.
  • a subject After a subject meets the eligibility criteria, he/she was allocated to a treatment sequence, based on a randomization procedure employing a 1:1 assignment ratio, with blocks stratified by center.
  • the randomization scheme was prepared by the CRO.
  • a subject ID number (subject number) was assigned.
  • Table 7 presents the analysis of average immediate VAS total pain scores for the ITT population.
  • the mean immediate VAS total pain score was 11.89 after administration of 20 mg/1.0 ml dose of GA and was 8.64 after administration of 20 mg/0.5 ml dose of GA. This indicates that less pain was experienced after 20 mg/0.5 ml injection of GA compared with 20 mg/1.0 ml injection of GA.
  • the ranked VAS scores differed by 21.1 (95% CI: 13.4, 28.8) observations; this difference in the ranks was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose of GA, although neither group had a high level of pain on average as the VAS scale ranges from 0 to 100.
  • FIG. 1 presents the plot of daily total immediate VAS total scores for the ITT population.
  • the analysis of average immediate VAS total scores for the PP population was also performed.
  • the mean immediate VAS total score for the PP population was 11.44 after administration of 20 mg/1.0 ml dose of GA and was 8.31 after administration of 20 mg/0.5 ml dose of GA.
  • the ranked VAS scores differed by 20.4 (95% CI: 12.7, 28.0) observations; this difference in the ranks was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose of GA
  • the plot of daily total immediate VAS total scores for the PP population is presented in FIG. 2 .
  • similar results were observed in the PP population with statistically significant difference (p ⁇ 0.0001) in the 2 formulations of GA in favor of 20 mg/0.5 ml dose of GA.
  • Table 8 presents the analysis of average 5-minute VAS total scores for the ITT population.
  • the mean VAS total pain score was 17.19 at 5 minutes after administration of 20 mg/1.0 ml dose of GA and was 11.85 at 5 minutes after administration of 20 mg/0.5 ml dose of GA.
  • the ranked VAS scores differed by 27.2 (95% CI: 20.2, 34.3) observations; this difference in the ranks was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose of GA.
  • FIG. 3 presents the plot of daily total 5-minute VAS total scores for the ITT population.
  • Table 9 presents the analysis of average 5-minute LISR total presence scores for the ITT population.
  • LISR total presence scores could range from 0 to 4 for an individual subject depending on how many of the following symptoms were experienced—redness, itching, swelling, and lump.
  • the mean LISR total presence score was 1.85 at 5 minutes after administration of 20 mg/1.0 ml dose of GA and was 1.41 at 5 minutes after administration of 20 mg/0.5 ml dose of GA.
  • the ranked VAS scores differed by 35.0 (95% CI: 25.4, 44.6) observations; this difference in the ranks was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose of GA.
  • Table 10 presents the analysis of average 5-minute LISR total severity scores for the ITT population.
  • LISR total severity scores could range from 0 to 12 for an individual subject depending on the severity (rated 0 to 3) of each of the following symptoms experienced—redness, itching, swelling, and lump.
  • the mean LISR total severity score was 2.30 at 5 minutes after administration of 20 mg/1.0 ml dose of GA and was 1.64 at 5 minutes after administration of 20 mg/0.5 ml dose of GA.
  • the ranked VAS scores differed by 36.9 (95% CI: 27.3, 46.5) observations; this difference in the ranks was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose of GA.
  • Table 11 presents the analysis of average 24-hour LISR total presence scores for the ITT population.
  • the mean LISR total presence score was 1.19 at 24 hours after administration of 20 mg/1.0 ml dose of GA and was 0.92 at 24 hours after administration of 20 mg/0.5 ml dose of GA.
  • the ranked VAS scores differed by 23.8 (95% CI: 14.9, 32.6) observations; this difference in the ranks was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose of GA.
  • Table 12 presents the analysis of average 24-hour LISR total severity scores for the ITT population.
  • the mean LISR total severity score was 1.47 at 24 hours after administration of 20 mg/1.0 ml dose of GA and was 1.10 at 24 hours after administration of 20 mg/0.5 ml dose of GA.
  • the ranked VAS scores differed by 23.8 (95% CI: 15.0, 32.7) observations; this difference in the ranks was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose of GA.
  • Injections of F2 are at least as effective as injections of F1 in treating RRMS.
  • the mean immediate VAS pain presence total score was 0.53 after administration of 20 mg/1.0 ml dose of GA and was 0.43 after administration of 20 mg/0.5 ml dose of GA. This suggests that the pain following injection of 20 mg/1.0 ml dose of GA was greater than the pain following injection of 20 mg/0.5 ml dose of GA.
  • the ranked VAS pain presence scores differed by 20.3 (95% CI: 12.0, 28.6) observations; this difference in the ranks was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose of GA.
  • the mean VAS pain presence total score was 0.67 at 5 minutes after administration of 20 mg/1.0 ml dose of GA and was 0.54 at 5 minutes after administration of 20 mg/0.5 ml dose of GA.
  • the ranked 5-minute VAS pain presence total scores differed by 27.0 (95% CI: 19.0, 34.9) observations; this difference in the ranks was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose of GA.
  • Table 15 presents the average daily 5-minute LISR scores for the ITT population. Plot of average daily 5-minute LISR scores is presented for the ITT population in FIG. 4(A-D) . Swelling, redness, itching and lump were the 4 symptoms considered as local injection site reactions during the study. The mean LISR scores for each of these symptoms were lower for subjects receiving 20 mg/0.5 ml dose of GA than for subjects receiving 20 mg/1.0 ml dose of GA at 5 minutes after administration of injections at each visit.
  • Table 16 presents the average daily 24-hour LISR scores for the ITT population.
  • the plot of average daily 24-hour LISR scores is presented for the ITT population in FIG. 5(A-D) .
  • the mean LISR scores for each of these symptoms were lower for subjects receiving 20 mg/0.5 ml dose of GA than for subjects receiving 20 mg/1.0 ml dose of GA at 24 hours after administration of injections at each visit.
  • Table 17 presents the number and percent of subjects reporting no symptoms at 5 minutes following injection for the ITT population. Most subjects reported some symptoms 5 minutes following injection of either formulation. However, the number and percentage of subjects reporting no symptoms at 5 minutes after 20 mg/0.5 ml GA injection was two to three times the number and percentage of subjects reporting no symptoms at 5 minutes after 20 mg/1.0 ml GA injection for each day interval.
  • Table 18 presents the number and percent of subjects reporting no symptoms 24 hours following injection for the ITT population. The majority of subjects reported symptoms 24 hours following injection of either formulation; however there were fewer subjects reporting symptoms 24 hours following injection than those reporting at 5 minutes following injections. The number and percentage of subjects reporting no symptoms after 24 hours following injection of 20 mg/0.5 ml GA was approximately 25%-50% higher than the number and percentage of subjects reporting no symptoms after 24 hours following injection of 20 mg/1.0 ml GA for most day intervals.
  • Table 19 presents the number and percent of subjects reporting symptoms at 5-minutes following injection by injection site for the ITT population. Overall, the number and percentage of subjects reporting symptoms at 5 minutes following injection of 20 mg/0.5 ml GA was less than the number and percentage of subjects reporting symptoms at 5 minutes following injection of 20 mg/1.0 ml GA with the exception of redness in the left arm and itching in the right arm.
  • Table 20 presents the number and percent of subjects reporting symptoms at 24-hours following injection by injection site for the ITT population. Overall, the number and percentage of subjects reporting symptoms at 24 hours following injection of 20 mg/0.5 ml GA was less than the number and percentage of subjects reporting symptoms at 24 hours following injection of 20 mg/1.0 ml GA with the exception of redness when the left arm was the injection site.
  • the 20 mg/0.5 ml formulation exhibits surprising properties in clinical testing in view of the 20 mg/ml and 40 mg/ml formulations.
  • the 40 mg/ml solution of glatiramer acetate is no more effective than administering a 20 mg/ml dose 42 .
  • the doubling of the drug dosage together with a doubling of drug concentration did not lead to a doubling in the efficacy to the patient 40-42 .
  • doubling both the amount of active drug and the drug concentration does not double the efficacy of the unit dosage form.
  • the prior art indicates that doubling drug concentration may have inhibited the effectiveness of the drug.
  • IPIRs Injection increase over reactions
  • Injection site burning mass, 5% higher reactions pain, and incidence of urticartia are reactions observed relative to 20 mg/ml 40 patient-reported VAS scores taken Reduced by about total injection as baseline 27% relative to pain rating on a those present for visual analogue 20 mg/ml (see page scale (VAS) 19, lines 26-27 of occurring the subject immediately after application) injection the patient- VAS scores taken Reduced by about reported total as baseline 31% relative to injection pain those present for rating on a visual 20 mg/ml (see page analogue scale 20, lines 5-7 of (VAS) experienced the subject five minutes application) following the subcutaneous injection Immediate pain Dichotomized
  • the 20 mg/0.5 ml form of glatiramer acetate represents a unique and novel formulation that differs from prior art formulations of glatiramer acetate. Specifically:
  • the primary clinical outcome variable was the total injection pain rating occurring immediately after injection (i.e., immediate total pain score). Pain immediately following injection and at later timepoints was relatively low for both formulations. However, the mean immediate VAS total pain score was less after administration of 20 mg/0.5 ml GA injection compared with 20 mg/1.0 ml GA injection. The difference in ranked scores between the 2 formulations of GA was statistically significant (p ⁇ 0.0001) in favor of 20 mg/0.5 ml dose.
  • the most frequently reported TEAEs after administration of 20 mg/0.5 ml GA injection were urinary tract infection (2.8%), and viral upper respiratory tract infection, arthralgia, and headache (1.4% each).
  • the 20 mg/0.5 ml GA formulation may be advantageous to patients with RRMS who require daily injections, as subjects in this study reported less pain and fewer injection site reactions following use of this formulation in comparison with the 20 mg/1.0 ml GA formulation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Dermatology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Infusion, Injection, And Reservoir Apparatuses (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US12/761,367 2009-07-15 2010-04-15 Reduced Volume Formulation of Glatiramer Acetate and Methods of Administration Abandoned US20110060279A1 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
US12/761,367 US20110060279A1 (en) 2009-07-15 2010-04-15 Reduced Volume Formulation of Glatiramer Acetate and Methods of Administration
US12/785,125 US7855176B1 (en) 2009-07-15 2010-05-21 Reduced volume formulation of glatiramer acetate and methods of administration
MX2012000687A MX2012000687A (es) 2009-07-15 2010-07-14 Formulación de volumen reducido de acetato de glatiramer y métodos de administración.
EA201270167A EA201270167A1 (ru) 2009-07-15 2010-07-14 Лекарственная форма глатирамера ацетата с уменьшенным объемом и способы введения
BR112012000878A BR112012000878A2 (pt) 2009-07-15 2010-07-14 formulação de volume reduzido de acetato de glatiramer e métodos de administração
JP2012520598A JP2012533540A (ja) 2009-07-15 2010-07-14 グラチラマーアセテートの減量製剤および投与法
AU2010273234A AU2010273234A1 (en) 2009-07-15 2010-07-14 Reduced volume formulation of glatiramer acetate and methods of administration
US13/384,021 US8920373B2 (en) 2009-07-15 2010-07-14 Reduced volume formulation of glatiramer acetate and methods of administration
EP10800156A EP2453907A2 (en) 2009-07-15 2010-07-14 Reduced volume formulation of glatiramer acetate and methods of administration
PCT/US2010/001972 WO2011008274A2 (en) 2009-07-15 2010-07-14 Reduced volume formulation of glatiramer acetate and methods of administration
ARP100102598A AR077484A1 (es) 2009-07-15 2010-07-15 Formulacion de acetato de glatiramer de volumen reducido y metodos para su administracion
US12/948,611 US9018170B2 (en) 2009-07-15 2010-11-17 Reduced volume formulation of glatiramer acetate and methods of administration
TW100103482A TWI435721B (zh) 2010-01-29 2011-01-28 格拉替雷(glatiramer)醋酸鹽之降低體積調配物及施用方法
IL217240A IL217240A0 (en) 2009-07-15 2011-12-27 Reduced volume formulation of glatiramer acetate and methods of administration
ZA2012/00586A ZA201200586B (en) 2009-07-15 2012-01-24 Reduced volume formulation of glatiramer acetate and methods of administration

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US27100909P 2009-07-15 2009-07-15
US27134009P 2009-07-20 2009-07-20
US33701110P 2010-01-29 2010-01-29
US12/761,367 US20110060279A1 (en) 2009-07-15 2010-04-15 Reduced Volume Formulation of Glatiramer Acetate and Methods of Administration

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/785,125 Continuation US7855176B1 (en) 2009-07-15 2010-05-21 Reduced volume formulation of glatiramer acetate and methods of administration
US13/384,021 Continuation-In-Part US8920373B2 (en) 2009-07-15 2010-07-14 Reduced volume formulation of glatiramer acetate and methods of administration

Publications (1)

Publication Number Publication Date
US20110060279A1 true US20110060279A1 (en) 2011-03-10

Family

ID=42283140

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/761,367 Abandoned US20110060279A1 (en) 2009-07-15 2010-04-15 Reduced Volume Formulation of Glatiramer Acetate and Methods of Administration
US12/785,125 Expired - Fee Related US7855176B1 (en) 2009-07-15 2010-05-21 Reduced volume formulation of glatiramer acetate and methods of administration
US12/948,611 Expired - Fee Related US9018170B2 (en) 2009-07-15 2010-11-17 Reduced volume formulation of glatiramer acetate and methods of administration

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/785,125 Expired - Fee Related US7855176B1 (en) 2009-07-15 2010-05-21 Reduced volume formulation of glatiramer acetate and methods of administration
US12/948,611 Expired - Fee Related US9018170B2 (en) 2009-07-15 2010-11-17 Reduced volume formulation of glatiramer acetate and methods of administration

Country Status (20)

Country Link
US (3) US20110060279A1 (ja)
EP (2) EP2275086B1 (ja)
JP (1) JP2012533540A (ja)
AR (1) AR077484A1 (ja)
AT (1) ATE549013T1 (ja)
AU (1) AU2010273234A1 (ja)
BR (1) BR112012000878A2 (ja)
CA (1) CA2697570C (ja)
DK (1) DK2275086T3 (ja)
EA (1) EA201270167A1 (ja)
ES (1) ES2383347T3 (ja)
HK (1) HK1152249A1 (ja)
HR (1) HRP20120349T1 (ja)
IL (1) IL217240A0 (ja)
MX (1) MX2012000687A (ja)
PL (1) PL2275086T3 (ja)
PT (1) PT2275086E (ja)
RS (1) RS52367B (ja)
WO (1) WO2011008274A2 (ja)
ZA (1) ZA201200586B (ja)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100298227A1 (en) * 1998-07-23 2010-11-25 President and Fellows of Harvard College and Yeda Research Treatment of autoimmune conditions with Copolymer 1 and related copolymers
US20110230413A1 (en) * 2010-03-16 2011-09-22 Suhayl Dhib-Jalbut Predictive biomarkers of clinical response to glatiramer acetate therapy in multiple sclerosis
US8709433B2 (en) 2010-10-11 2014-04-29 Teva Pharmaceutical Industries Ltd. Cytokine biomarkers as predictive biomarkers of clinical response for Glatiramer acetate
US8815511B2 (en) 2011-10-10 2014-08-26 Teva Pharmaceutical Industries, Ltd. Determination of single nucleotide polymorphisms useful to predict response for glatiramer acetate
US9155775B1 (en) 2015-01-28 2015-10-13 Teva Pharmaceutical Industries, Ltd. Process for manufacturing glatiramer acetate product
US9155776B2 (en) 2009-08-20 2015-10-13 Yeda Research & Development Co., Ltd. Low frequency glatiramer acetate therapy
US9415176B1 (en) 2015-01-22 2016-08-16 West Pharmaceutical Services, Inc. Autoinjector having an end-of-dose visual indicator
US9617596B2 (en) 2012-10-10 2017-04-11 Teva Pharmaceutical Industries, Ltd. Biomarkers predictive for clinical response for glatiramer acetate
US9702007B2 (en) 2013-10-21 2017-07-11 Teva Pharmaceuticals Industries, Ltd. Genetic markers predictive of response to glatiramer acetate
US10780218B2 (en) 2014-02-26 2020-09-22 Allergan, Inc. Intraocular implant delivery apparatus and methods of use thereof
US11167003B2 (en) 2017-03-26 2021-11-09 Mapi Pharma Ltd. Methods for suppressing or alleviating primary or secondary progressive multiple sclerosis (PPMS or SPMS) using sustained release glatiramer depot systems
USRE49251E1 (en) 2010-01-04 2022-10-18 Mapi Pharma Ltd. Depot systems comprising glatiramer or pharmacologically acceptable salt thereof

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2275086T3 (pl) 2009-07-15 2012-09-28 Teva Pharma Formulacja octanu glatirameru o zredukowanej objętości oraz sposoby podawania
US8920373B2 (en) 2009-07-15 2014-12-30 Teva Pharmaceutical Industries, Ltd. Reduced volume formulation of glatiramer acetate and methods of administration
CN104684603B (zh) * 2012-08-20 2017-11-24 卡贝欧洲有限公司 自动注射装置
WO2014128079A1 (en) * 2013-02-19 2014-08-28 Synthon B.V. Glatiramer acetate multidose formulation
KR101780191B1 (ko) * 2013-03-08 2017-09-21 테바 파마슈티컬 인더스트리즈 리미티드 주사기용 재사용 가능한 주입기 장치
US8591463B1 (en) 2013-03-08 2013-11-26 Teva Pharmaceutical Industries Ltd. Re-useable injector device for syringe
AU2014224340B2 (en) * 2013-03-08 2016-12-22 Teva Pharmaceutical Industries Limited Re-useable injector device for syringe
EP3534893A1 (en) * 2016-11-02 2019-09-11 To Pharmaceuticals LLC Combination therapy of cbd and copaxone
EP4252629A3 (en) 2016-12-07 2023-12-27 Biora Therapeutics, Inc. Gastrointestinal tract detection methods, devices and systems
US10323289B2 (en) 2017-06-26 2019-06-18 Institut Pasteur Treatments to eliminate HIV reservoirs and reduce viral load
US20200306516A1 (en) 2017-08-14 2020-10-01 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with glatiramer or a pharmaceutically acceptable salt thereof

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4822340A (en) * 1985-10-11 1989-04-18 Duphar International Research B.V. Automatic injector
US4950246A (en) * 1987-05-08 1990-08-21 Spruyt-Hillen B.V. Injection pen
US5137516A (en) * 1989-11-28 1992-08-11 Glaxo Group Limited Triggered application device for medicament to be more descriptive of the invention
US5981589A (en) * 1994-05-24 1999-11-09 Yeda Research And Development Co., Ltd. Copolymer-1 improvements in compositions of copolymers
US20020077278A1 (en) * 2000-06-05 2002-06-20 Yong V. Wee Use of glatiramer acetate (copolymer 1) in the treatment of central nervous system disorders
US6448225B2 (en) * 1988-04-15 2002-09-10 Genentech, Inc. Human growth hormone aqueous formulation
US6454746B1 (en) * 1997-06-04 2002-09-24 Eli Lilly And Company Medication delivery apparatus
US20040039336A1 (en) * 2000-12-14 2004-02-26 Mikael Amark Auto-injector
US20050277885A1 (en) * 2002-11-25 2005-12-15 Benjamin Scherer Auto-injector comprising a resettable releasing safety device
US20060154862A1 (en) * 2004-10-29 2006-07-13 Ray Anup K Processes for preparing a polypeptide
US20070161566A1 (en) * 2006-01-11 2007-07-12 Teva Pharmaceutical Industries, Ltd. Method of treating multiple sclerosis
US20070161960A1 (en) * 2006-01-12 2007-07-12 Fu-Yuan Li Lancet device
US20080118553A1 (en) * 2006-06-12 2008-05-22 Anton Frenkel Tannate salt form of polypeptide mixtures, their preparation and use
US20090048181A1 (en) * 2004-09-02 2009-02-19 Schipper Hyman M Combination therapy with glatiramer acetate and n-acetylcysteine for the treatment of multiple sclerosis
US20090149541A1 (en) * 2007-11-28 2009-06-11 Yafit Stark Method of delaying the onset of clinically definite multiple sclerosis
US7560100B2 (en) * 2004-09-09 2009-07-14 Yeda Research And Development Co., Ltd. Mixtures of polypeptides, compositions containing and processes for preparing same, for treating neurodegenerative diseases
US7585843B2 (en) * 2003-10-03 2009-09-08 Bayhill Therapeutics, Inc. Treatment of demyelinating autoimmune disease with modified ordered peptides
USD607558S1 (en) * 2008-09-19 2010-01-05 Becton Dickinson France S.A.S. Medicine injector
US7655221B2 (en) * 2004-05-07 2010-02-02 Peptimmune, Inc. Methods of treating disease with random copolymers
US20100160894A1 (en) * 2006-06-30 2010-06-24 Julian Joseph F Automatic injection device
USD622374S1 (en) * 2006-09-06 2010-08-24 Abbott Biotechnology Ltd. Automatic injection device
US20110066112A1 (en) * 2009-07-15 2011-03-17 Teva Pharmaceutical Industries, Ltd. Reduced volume formulation of glatiramer acetate and methods of administration

Patent Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4822340A (en) * 1985-10-11 1989-04-18 Duphar International Research B.V. Automatic injector
US4950246A (en) * 1987-05-08 1990-08-21 Spruyt-Hillen B.V. Injection pen
US6448225B2 (en) * 1988-04-15 2002-09-10 Genentech, Inc. Human growth hormone aqueous formulation
US5137516A (en) * 1989-11-28 1992-08-11 Glaxo Group Limited Triggered application device for medicament to be more descriptive of the invention
US5981589A (en) * 1994-05-24 1999-11-09 Yeda Research And Development Co., Ltd. Copolymer-1 improvements in compositions of copolymers
US6054430A (en) * 1994-05-24 2000-04-25 Yeda Research And Development Co., Ltd. Copolymer-1 improvements in compositions of copolymers
US6342476B1 (en) * 1994-05-24 2002-01-29 Yeda Research & Development Company Limited Copolymer-1 improvements in compositions of copolymers
US6362161B1 (en) * 1994-05-24 2002-03-26 Yeda Research & Development Company Limited Copolymer-1 improvements on compositions of copolymers
US7199098B2 (en) * 1994-05-24 2007-04-03 Yeda Research And Development Co., Ltd. Copolymer-1 improvements in compositions of copolymers
US6048898A (en) * 1994-05-24 2000-04-11 Yeda Research And Development Co., Ltd. Copolymer-1 improvements in compositions of copolymers
US6620847B2 (en) * 1994-05-24 2003-09-16 Yeda Research And Development Co., Ltd. Copolymer-1 improvements in compositions of copolymers
US6939539B2 (en) * 1994-05-24 2005-09-06 Yeda Research & Development Copolymer-1 improvements in compositions of copolymers
US20040106554A1 (en) * 1994-05-24 2004-06-03 Eliezer Konfino Copolymer-1 improvements in compositions of copolymers
US6454746B1 (en) * 1997-06-04 2002-09-24 Eli Lilly And Company Medication delivery apparatus
US7033582B2 (en) * 2000-06-05 2006-04-25 Teva Pharmaceutical Industries, Ltd. Use of glatiramer acetate (copolymer 1) in the treatment of central nervous system disorders
US20020077278A1 (en) * 2000-06-05 2002-06-20 Yong V. Wee Use of glatiramer acetate (copolymer 1) in the treatment of central nervous system disorders
US20040039336A1 (en) * 2000-12-14 2004-02-26 Mikael Amark Auto-injector
US7442185B2 (en) * 2000-12-14 2008-10-28 Shl Group Ab Auto-injector
US20050277885A1 (en) * 2002-11-25 2005-12-15 Benjamin Scherer Auto-injector comprising a resettable releasing safety device
US7585843B2 (en) * 2003-10-03 2009-09-08 Bayhill Therapeutics, Inc. Treatment of demyelinating autoimmune disease with modified ordered peptides
US7655221B2 (en) * 2004-05-07 2010-02-02 Peptimmune, Inc. Methods of treating disease with random copolymers
US20090048181A1 (en) * 2004-09-02 2009-02-19 Schipper Hyman M Combination therapy with glatiramer acetate and n-acetylcysteine for the treatment of multiple sclerosis
US7560100B2 (en) * 2004-09-09 2009-07-14 Yeda Research And Development Co., Ltd. Mixtures of polypeptides, compositions containing and processes for preparing same, for treating neurodegenerative diseases
US20060154862A1 (en) * 2004-10-29 2006-07-13 Ray Anup K Processes for preparing a polypeptide
US20070161566A1 (en) * 2006-01-11 2007-07-12 Teva Pharmaceutical Industries, Ltd. Method of treating multiple sclerosis
US20070161960A1 (en) * 2006-01-12 2007-07-12 Fu-Yuan Li Lancet device
US20080118553A1 (en) * 2006-06-12 2008-05-22 Anton Frenkel Tannate salt form of polypeptide mixtures, their preparation and use
US20100160894A1 (en) * 2006-06-30 2010-06-24 Julian Joseph F Automatic injection device
USD622374S1 (en) * 2006-09-06 2010-08-24 Abbott Biotechnology Ltd. Automatic injection device
US20090149541A1 (en) * 2007-11-28 2009-06-11 Yafit Stark Method of delaying the onset of clinically definite multiple sclerosis
USD607558S1 (en) * 2008-09-19 2010-01-05 Becton Dickinson France S.A.S. Medicine injector
US20110066112A1 (en) * 2009-07-15 2011-03-17 Teva Pharmaceutical Industries, Ltd. Reduced volume formulation of glatiramer acetate and methods of administration

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100298227A1 (en) * 1998-07-23 2010-11-25 President and Fellows of Harvard College and Yeda Research Treatment of autoimmune conditions with Copolymer 1 and related copolymers
US9155776B2 (en) 2009-08-20 2015-10-13 Yeda Research & Development Co., Ltd. Low frequency glatiramer acetate therapy
US9402874B2 (en) 2009-08-20 2016-08-02 Yeda Research & Development Co., Ltd. Low frequency glatiramer acetate therapy
USRE49251E1 (en) 2010-01-04 2022-10-18 Mapi Pharma Ltd. Depot systems comprising glatiramer or pharmacologically acceptable salt thereof
US20110230413A1 (en) * 2010-03-16 2011-09-22 Suhayl Dhib-Jalbut Predictive biomarkers of clinical response to glatiramer acetate therapy in multiple sclerosis
US8759302B2 (en) 2010-03-16 2014-06-24 Teva Pharmaceutical Industries, Ltd. Methods of treating a subject afflicted with an autoimmune disease using predictive biomarkers of clinical response to glatiramer acetate therapy in multiple sclerosis
US9687522B2 (en) 2010-03-16 2017-06-27 Teva Pharmaceutical Industries, Ltd. Methods of treating a subject afflicted with an autoimmune disease using predictive biomarkers of clinical response to glatiramer acetate therapy in multiple sclerosis
US9625473B2 (en) 2010-10-11 2017-04-18 Teva Pharmaceutical Industries Ltd. Cytokine biomarkers as predictive biomarkers of clinical response for glatiramer acetate
US9063153B2 (en) 2010-10-11 2015-06-23 Teva Pharmaceuticals Industries Ltd. Cytokine biomarkers as predictive biomarkers of clinical response for glatiramer acetate
US8709433B2 (en) 2010-10-11 2014-04-29 Teva Pharmaceutical Industries Ltd. Cytokine biomarkers as predictive biomarkers of clinical response for Glatiramer acetate
US9499868B2 (en) 2011-10-10 2016-11-22 Teva Pharmaceutical Industries, Ltd. Determination of single nucleotide polymorphisms useful to predict response for glatiramer acetate
US8815511B2 (en) 2011-10-10 2014-08-26 Teva Pharmaceutical Industries, Ltd. Determination of single nucleotide polymorphisms useful to predict response for glatiramer acetate
US9617596B2 (en) 2012-10-10 2017-04-11 Teva Pharmaceutical Industries, Ltd. Biomarkers predictive for clinical response for glatiramer acetate
US9702007B2 (en) 2013-10-21 2017-07-11 Teva Pharmaceuticals Industries, Ltd. Genetic markers predictive of response to glatiramer acetate
US10780218B2 (en) 2014-02-26 2020-09-22 Allergan, Inc. Intraocular implant delivery apparatus and methods of use thereof
US9415176B1 (en) 2015-01-22 2016-08-16 West Pharmaceutical Services, Inc. Autoinjector having an end-of-dose visual indicator
US9155775B1 (en) 2015-01-28 2015-10-13 Teva Pharmaceutical Industries, Ltd. Process for manufacturing glatiramer acetate product
US9763993B2 (en) 2015-01-28 2017-09-19 Teva Pharmaceutical Industries Ltd. Process for manufacturing glatiramer acetate product
US11167003B2 (en) 2017-03-26 2021-11-09 Mapi Pharma Ltd. Methods for suppressing or alleviating primary or secondary progressive multiple sclerosis (PPMS or SPMS) using sustained release glatiramer depot systems

Also Published As

Publication number Publication date
EP2275086B1 (en) 2012-03-14
JP2012533540A (ja) 2012-12-27
RS52367B (en) 2012-12-31
EP2275086A1 (en) 2011-01-19
DK2275086T3 (da) 2012-07-09
ZA201200586B (en) 2013-10-30
WO2011008274A4 (en) 2011-11-10
AR077484A1 (es) 2011-08-31
HRP20120349T1 (hr) 2012-07-31
WO2011008274A3 (en) 2011-09-22
ATE549013T1 (de) 2012-03-15
CA2697570C (en) 2011-11-01
ES2383347T3 (es) 2012-06-20
US20110066112A1 (en) 2011-03-17
PL2275086T3 (pl) 2012-09-28
WO2011008274A2 (en) 2011-01-20
EA201270167A1 (ru) 2012-08-30
HK1152249A1 (en) 2012-02-24
AU2010273234A1 (en) 2012-02-23
CA2697570A1 (en) 2010-06-22
US7855176B1 (en) 2010-12-21
PT2275086E (pt) 2012-05-18
US9018170B2 (en) 2015-04-28
EP2453907A2 (en) 2012-05-23
IL217240A0 (en) 2012-02-29
BR112012000878A2 (pt) 2019-09-24
MX2012000687A (es) 2012-09-07

Similar Documents

Publication Publication Date Title
CA2697570C (en) Reduced volume formulation of glatiramer acetate and methods of administration
US8920373B2 (en) Reduced volume formulation of glatiramer acetate and methods of administration
RU2564104C2 (ru) Композиции инсулинов длительного действия
TWI643614B (zh) 低頻率格拉替雷(glatiramer)醋酸鹽之治療
RU2733464C2 (ru) Препараты tpp1 и способы лечения заболевания cln2
EP3909609A1 (en) Methods for treatment of refractory generalized myasthenia gravis
CN109069595A (zh) 标靶医疗性溶酶体酵素融合蛋白质、其相关调配物与用途
KR20170102550A (ko) 릭시세나티드를 이용한 소아 제2형 당뇨병 환자의 치료
US20080038252A1 (en) Histamine-Containing Composition for the Treatment of Allergic Diseases
TWI435721B (zh) 格拉替雷(glatiramer)醋酸鹽之降低體積調配物及施用方法
Agent et al. BASAGLAR™
CARE Approach to the Identification and Differentiation of Migraine
Shuster Anaphylaxis Seen with a Special Dye;• Cholestasis Caused by Cetirizine;• Acarbose and Lymphocytic Colitis;• Seizures Caused by Intravenous Alatrofloxacin;• Pedal Edema Caused by Olanzapine;• Disturbances in Electrolyte Homeostasis as an Adverse Effect of Gentamicin Therapy
TW201318636A (zh) 用於治療膽道狹窄或/及阻塞之glp-1促效劑

Legal Events

Date Code Title Description
AS Assignment

Owner name: TEVA PHARMACEUTICAL INDUSTRIES, LTD., ISRAEL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ALTMAN, AYELET;SALTKILL, DORIS;EL-GAD, TOMER;AND OTHERS;SIGNING DATES FROM 20100419 TO 20100516;REEL/FRAME:024425/0618

AS Assignment

Owner name: TEAM CONSULTING LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GREENHALGH, PAUL;ROBINSON, DAVID GEORGE;REEL/FRAME:025371/0549

Effective date: 20101108

Owner name: TEVA PHARMACEUTICAL INDUSTRIES, LTD., ISRAEL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TEAM CONSULTING LIMITED;REEL/FRAME:025371/0530

Effective date: 20101108

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION