[go: up one dir, main page]

HK40005874A - Depot systems comprising glatiramer acetate - Google Patents

Depot systems comprising glatiramer acetate Download PDF

Info

Publication number
HK40005874A
HK40005874A HK19129347.1A HK19129347A HK40005874A HK 40005874 A HK40005874 A HK 40005874A HK 19129347 A HK19129347 A HK 19129347A HK 40005874 A HK40005874 A HK 40005874A
Authority
HK
Hong Kong
Prior art keywords
patient
depot formulation
glatiramer
injection
certain embodiments
Prior art date
Application number
HK19129347.1A
Other languages
Chinese (zh)
Inventor
埃胡德·马罗姆
纳达夫·布莱希基默尔曼
尤里·达农
塞·鲁比诺夫
Original Assignee
Mapi医药公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mapi医药公司 filed Critical Mapi医药公司
Publication of HK40005874A publication Critical patent/HK40005874A/en

Links

Description

Reservoir system comprising glatiramer acetate
Technical Field
The present invention relates to long acting dosage forms of glatiramer acetate and other pharmaceutically acceptable salts of glatiramer. In particular, the present invention relates to depot formulations and other implantable systems for the extended release of glatiramer acetate.
Background
Multiple Sclerosis (MS) is a chronic, debilitating autoimmune disease of the Central Nervous System (CNS). MS disease activity can be monitored by brain Magnetic Resonance Imaging (MRI), accumulation of disability, and the rate and severity of relapse. There are several forms of multiple sclerosis, including relapsing-remitting multiple sclerosis (RRMS). Patients with RRMS experience sporadic exacerbations or relapses and remissions.
Various therapies have been approved for relapsing remitting form of Multiple Sclerosis (MS), including(glatiramer acetate),(natalizumab)),(PEG-interferon β -1a) and(daclizumab)). Various measures of efficacy have been reported for these therapies. An important therapeutic goal, no signs of disease activity (NEDA), has become a new measure of outcome (Rotstein et al, JAMA neurol, 2015, Vol.72(2):152- & 158).
Glatiramer acetate
Under the trade name ofMarketed copolymer-1, also known as Glatiramer Acetate (GA), contains acetate salts of a mixture of synthetic polypeptides containing L-glutamic acid, L-alanine, L-tyrosine and L-lysine. The average molar fractions of the amino acids were 0.141, 0.427, 0.095 and 0.338, respectively, and the average molecular weight of copolymer-1 was between 5,000 and 9,000 daltons. Chemically, glatiramer acetate is designated as the acetate salt of a polymer of L-glutamic acid with L-alanine, L-lysine and L-tyrosine. The structural formula is as follows: (Glu, Ala, Lys, Tyr)x·xCH3COOH;(C5H9NO4·C3H7NO2·C6H14N2O2·C9H11NO3)x·xC2H4O2[CAS-147245-92-9]。Is a clear, colorless to yellowish, sterile, pyrogen-free solution for subcutaneous injection. Contains glatiramer acetate 20mg or 40mg and mannitol 40mg per ml. The pH of the solution is in the range of about 5.5 to 7.0(Full prescription information (Full prescriptbindinformation), 2009).Is indicated for reducing the frequency of relapse in patients with RRMS, including patients who have experienced a first clinical episode and who have MRI characteristics consistent with multiple sclerosis ((s))Glatiramer acetate injection, complete prescription information, 2016).
Mechanism of action
The mechanism of action of glatiramer acetate is not known, although such copolymers have shown some important immunological properties. Administration of copolymer-1 transferred the T cell population from pro-inflammatory Th1 cells to regulatory Th2 cells (FDA) that suppressed the inflammatory responseA label). Given its similarity to myelin basic protein, copolymer-1 can also act as a bait, diverting the autoimmune response to myelin. However, at least in the early stages of treatment, the integrity of the blood-brain barrier is not significantly affected by copolymer-1.
Copolymer-1 is a non-autoantigen that has been shown to inhibit Experimental Allergic Encephalomyelitis (EAE) induced by various encephalitogenic agents including Mouse Spinal Cord Homogenate (MSCH) including all myelin antigens such as Myelin Basic Protein (MBP) in many species (Sela et al, Bull inst. pasteur (1990) 88303-. EAE is a well-established model for multiple sclerosis.
In a phase III clinical trial, daily subcutaneous injections of copolymer-1 were found to slow the progression of disability and reduce the relapse rate of exacerbation-remitting multiple sclerosis (exarbenting-remitting multiple sclerosis) (Johnson KP, Neurology (1995),1, 65-70). Copolymer-1 therapy is currently limited to daily subcutaneous administration or three times weekly subcutaneous administration. US 6,214,791 discloses treatment by ingestion or inhalation of copolymer-1, but these routes of administration have not been shown to achieve clinical efficacy in human patients.
Approved dosage
Since the time of 1996,administration of a dose of 20mg by daily subcutaneous injection has been approved for the treatment of relapsing-remitting multiple sclerosis (RRMS). Since the time of 2014, the technology has been,a dose of 40mg has also been approved, administered by three injections per week with a spacing of at least 48 hours between each injection. And administering a dose of 20mg per dayThe latter dose and regimen, in contrast, reduced the number of injections per year by about 200, while maintaining the same efficacy.
Side effects
Frequently observed injection site problems include irritation, hypersensitivity, inflammation, pain, and even necrosis (in the case of interferon 1 β treatment) and consequent patient compliance issues.
Side effects typically include lumps at the injection site (injection site reactions), pain, fever, and chills. These side effects are generally mild in nature. Occasionally, the response occurs within minutes after injection, with flushing, tachypnea, anxiety and tachycardias. These side effects subside within thirty minutes. Over time, a visible depression called lipoatrophy may form at the injection site due to local destruction of adipose tissue. Thus, an alternative method of administration is desired.
According to FDA prescription labeling, several serious side effects of glatiramer acetate have been reported, including serious side effects on the cardiovascular system, the digestive system (including liver), the blood and lymphatic systems, the musculoskeletal system, the nervous system, the respiratory system, special senses (particularly the eyes), the urogenital system of the body; metabolic and nutritional disorders have also been reported; however, the link between glatiramer acetate and these adverse effects has not been established explicitly (FDA)A label).
Storage system
The parenteral route by Intravenous (IV), Intramuscular (IM) or Subcutaneous (SC) injection is the most common and effective delivery form for small and large molecular weight drugs. However, this mode of drug delivery is least popular with patients due to the pain, discomfort and inconvenience associated with needle sticks. Therefore, any drug delivery technology that can minimize (at a minimum) the total number of injections is preferred. Such a reduction in the frequency of drug administration in practice can be achieved by using injectable depot formulations capable of releasing the drug in a slow but predictable manner and thus improving compliance. For many small molecular weight drugs, depending on the dose, it may be possible to reduce the frequency of injections from daily to once or twice a month or even longer (6 months) by using a depot system. For large molecular weight drugs, and in particular for protein or polypeptide drugs, there has been little success in the development of depot formulations.
Microparticles, implants and gels are the most common forms of biodegradable polymeric devices used in practice to prolong the release of drugs in vivo. The microparticles are suspended in the aqueous medium just prior to injection and the skilled person can load up to 40% solids in the suspension. The implant/rod formulation is delivered to SC/IM tissue in a dry state with the aid of a specialized needle, without the need for an aqueous medium. This feature of the rod/implant allows for higher quality formulations and drug content to be delivered. Furthermore, in the rod/implant, the initial burst problem is minimized since the implant area is much smaller compared to the microparticles. In addition to biodegradable systems, there are non-biodegradable implants and infusion pumps that can be worn outside the body. Non-biodegradable implants require not only the physician to implant the device into SC/IM tissue, but also the physician to remove them after the drug release period.
Injectable compositions containing particulate preparations are particularly prone to problems. Particulate suspensions may contain up to 40% solids compared to 0.5% -5% solids in other types of injectable suspensions. Furthermore, the size range of microparticles used in injectable depot products is up to about 250 μm (60 μm to 100 μm on average) compared to the particle size of less than 5 μm recommended for IM or SC administration. Higher solids concentrations and larger solid particle sizes require larger size needles (about 18-21 gauge) for injection. In general, despite the infrequent use of large and uncomfortable needles, patients still prefer dosage forms with greatly reduced frequency of administration over daily injections of medication using smaller needles.
Polyesters of biodegradable polylactic acid (PLA) and copolymers of lactide and glycolide known as poly (lactic-co-glycolic acid) or poly (lactide-co-glycolide) (PLGA) are the most common polymers used in biodegradable dosage forms. PLA is a hydrophobic molecule, and PLGA degrades faster than PLA because more hydrophilic glycolide groups are present. These biocompatible polymers undergo random, non-enzymatic, hydrolytic cleavage of ester linkages to form lactic and glycolic acids, which are normal metabolic compounds in the body. Resorbable sutures, clips and implants were the earliest applications of these polymers. Southern Research Institute developed the first synthetic resorbable suture in 1970The first patent describing the use of PLGA polymers in sustained release dosage forms appeared in 1973 (U.S. Pat. No. 3,773,919)). According to some embodiments, the biodegradable polymer is selected from PLGA, PLA, PGA, and any combination thereof. Each possibility represents a separate embodiment of the invention.
PLGA polymers are commercially available today from a variety of suppliers. In addition to PLGA and PLA, natural cellulosic polymers such as starch, starch derivatives, dextran, and non-PLGA synthetic polymers are also being explored as biodegradable polymers in such systems.
WO 2011/080733, which is some of the present people, describes long-acting parenteral pharmaceutical compositions comprising glatiramer. Khan et al (ann. neuron.2013, vol.73, p. 705-713) describe a study evaluating the efficacy and safety of administration of 40mg glatiramer acetate 3 times per week in patients with relapsing-remitting multiple sclerosis (RRMS). US patents 8,232,250, 8,399,413, 8,969,302, US 9,155,776 and 9,402,874 describe methods of alleviating the symptoms of relapsing-remitting multiple sclerosis by three injections of 40mg GA over a period of seven days.
To date, long acting dosage forms of glatiramer acetate have not been tested for safety and efficacy in humans with RRMS, and are therefore not commercially available for treatment of humans. This is an unmet medical need as these formulations would be extremely beneficial to many patients.
Summary of The Invention
The present invention provides compositions and methods for treating relapsing-remitting form of multiple sclerosis comprising a single parenteral administration or implantation of a composition in a depot formulation that provides a monthly dose of 40mg of a pharmaceutically acceptable salt of Glatiramer Acetate (GA).
It is now disclosed for the first time that long acting pharmaceutical compositions and depot formulations according to the principles of the present invention provide injectable dosage forms that are compatible with commercially available three times daily or weekly, i.e.Equal or better curative effect. It was further surprisingly found that a single administration of a depot of 40mg glatiramer acetate according to the principles of the present invention every 4 weeks was at least as effective therapeutically as a 20mg glatiramer acetate injection per day or a 40mg glatiramer acetate administration three times per week, thus reducing the need for GA administration (injection) by 28-fold and 12-fold, respectively. Importantly, despite the fact that the total dose of GA administered was reduced 14-fold and 12-fold, respectively, the therapeutic efficiency was comparable. Treatment of MS patients with the methods disclosed herein requires fewer injections and lower GA doses than previously available therapies using glatiramer acetate.
It is also disclosed herein for the first time, for other MS drugs, such as(natalizumab),(Pegylated interferon β -1a) and(dallizumab) the one-year No Evidence of Disease Activity (NEDA) results reported have significantly better results in achieving NEDA after one year of treatment according to the method of the invention than the one-year No Evidence of Disease Activity (NEDA) results. Thus, treatment with a composition according to the principles of the present invention increases the likelihood of no evidence of disease activity compared to known therapies.
In one aspect, the invention provides a method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS) in a human patient suffering from RRMS or a human patient who has experienced a first clinical attack and is determined to be at high risk of developing clinically determined multiple sclerosis,
the method comprises administering to the patient a single intramuscular injection of a therapeutically effective regimen comprising a 40mg dose of a depot formulation of Glatiramer Acetate (GA) every 2 to 6 weeks sufficient to alleviate at least one symptom of the patient, wherein the symptom is selected from the group consisting of frequency of relapse of the patient, number of MRI-enhanced lesions or number of new lesion images of the brain, and Extended Disability Status Scale (EDSS) score.
In certain embodiments, the depot formulation is administered once every 3 to 5 weeks. In certain embodiments, the depot formulation is administered once every 3 weeks. In certain embodiments, the depot formulation is administered once every 4 weeks. In certain embodiments, the depot formulation is administered once every 5 weeks.
In certain embodiments, alleviating a symptom comprises reducing the frequency of relapse.
In certain embodiments, alleviating a symptom comprises reducing the number or volume of enhanced lesions or the number of new lesions in the brain. In certain embodiments, alleviating a symptom comprises reducing brain atrophy, reducing the number or volume of Gd-enhancing lesions, reducing T in a patient1Weighted number or volume of enhanced lesions, or reduction of new T2-the number of weighted lesions. In certain embodiments, alleviating a symptom comprises reducing the average cumulative number of Gd-enhancing lesions, reducing the average number of new T2 lesions, reducing the total volume of T2 lesions, or reducing T in the brain of a patient1-a cumulative number of enhanced lesions on the weighted image. In certain embodiments, alleviating a symptom comprises reducing the number of new low-intensity lesions on an enhanced T1 scan in the patient, or reducing the total volume of low-intensity lesions on an enhanced T1 scan in the patient.
In certain embodiments, alleviating a symptom comprises reducing the EDSS score of the patient. In certain embodiments, alleviating a symptom comprises reducing the patient's EDSS score by at least 0.5 points.
In certain embodiments, alleviating a symptom comprises reducing the level of disability in the patient as measured by the work productivity and activity decline-general health (WPAI-GH) questionnaire or by the EuroQoL (EQ-5D) questionnaire. In certain embodiments, alleviating a symptom comprises reducing a change in an ambulatory Index (Ambulation Index) in the patient.
In certain embodiments of the methods described above, the method reduces the symptoms at least as effectively as administering 20mg GA subcutaneously daily or 40mg GA subcutaneously three times a week.
In certain embodiments, the depot formulation is administered intramuscularly by the patient. In certain embodiments, the depot formulation is injected into the deltoid muscle.
In certain embodiments, the patient does not receive GA treatment prior to the start of the regimen. In certain embodiments, the patient has received GA treatment prior to the beginning of the regimen. In certain embodiments, the patient has at least 1 brain lesion detectable by MRI scanning, and wherein the lesion is associated with brain tissue inflammation, myelin sheath damage, or axonal damage. In certain embodiments, the lesion is a demyelinating white matter lesion (demyelinating white matter lesion) visible on MRI of the brain, and wherein the white matter lesion is at least 3mm in diameter. In certain embodiments, the patient has experienced a first clinical episode, and wherein the first clinical episode comprises a clinical episode of optic neuritis, blurred vision, diplopia, involuntary rapid eye movement, blindness, loss of balance, tremor, ataxia, vertigo, clumsiness of limbs, lack of coordination, weakness in one or more limbs, altered muscle tone, muscle stiffness, spasticity, stinging, paresthesia, burning sensation, muscle pain, facial pain, trigeminal neuralgia, stabbing megaly (stabbing sharp pain), burning stabbing pain, speech retardation, slurred mouth, altered rhythm, dysphagia, fatigue, bladder problems (including urinary urgency, urinary frequency, incomplete emptying, and incontinence), bowel problems (including constipation and bowel loss), impotence, decreased libido, loss of sensation, sensitivity to heat, short-term memory loss, attention loss, or loss of judgment or reasoning.
In certain embodiments, the frequency of immediate post-injection reactions, the severity of immediate post-injection reactions, or the frequency of injection site reactions is reduced relative to daily subcutaneous administration of 20mg GA or three subcutaneous injections per week of 40mg GA.
In certain embodiments, the depot formulation further comprises a pharmaceutically acceptable biodegradable carrier selected from the group consisting of poly (lactic-co-glycolic acid) (PLGA), polylactic acid (PLA), polyglycolic acid (PGA), and any combination thereof. In certain embodiments, the biodegradable carrier is PLGA.
In certain embodiments, the depot formulation is in the form of microparticles comprising an internal aqueous phase comprising GA, a water-immiscible polymer phase comprising a biodegradable polymer or a non-biodegradable polymer, and an external aqueous phase.
In certain embodiments, the depot formulation is in the form of microparticles prepared by a water-in-oil-in-water (w/o/w) double emulsion process. In certain embodiments, the internal aqueous phase comprises GA. In certain embodiments, the water-immiscible polymer phase comprises PLGA.
In certain embodiments, the external aqueous phase comprises a surfactant selected from the group consisting of polyvinyl alcohol (PVA), polysorbates, polyethylene oxide-polypropylene oxide block copolymers, and cellulose esters. In certain embodiments, the surfactant is PVA.
In certain embodiments, the depot formulation is administered in 2mL of water for injection (WFI). In certain embodiments, the depot formulation comprises from 10% to 40% solids. In certain embodiments, the depot formulation comprises 20% to 30% solids. In certain embodiments, the depot formulation comprises 25% solids.
In certain embodiments, the weight ratio between GA and the pharmaceutically acceptable biodegradable carrier is between 1:1 and 1: 100. In certain embodiments, the weight ratio between GA and the pharmaceutically acceptable biodegradable carrier is between 1:5 and 1: 25.
In certain embodiments, the depot formulation provides an extended release or prolonged action of glatiramer in a subject as compared to an immediate release formulation of a substantially similar dose of glatiramer. In certain embodiments, about 80% of glatiramer is released from the depot formulation in PBS over a 22 day period with continuous stirring at 37 ℃. In certain embodiments, about 20% of glatiramer is released from the depot formulation in PBS over a 5 day period at 37 ℃ with continuous stirring.
In another aspect, the invention also provides a method of increasing GA therapy tolerance in a human patient suffering from relapsing-remitting form of multiple sclerosis, the method comprising reducing the frequency of subcutaneous injections from a daily subcutaneous injection of a 20mg dose of GA or three subcutaneous injections of a 40mg dose of GA spaced apart at least one day between each injection over a period of seven days to a therapeutically effective regime of a depot formulation with a single intramuscular injection of a 40mg dose of GA every 2 to 6 weeks, to thereby increase GA therapy tolerance in the patient.
In certain embodiments, the depot formulation is administered once every 3 to 5 weeks. In certain embodiments, the depot formulation is administered once every 4 weeks.
In certain embodiments, increasing tolerance to GA treatment in a patient with relapsing-remitting multiple sclerosis comprises decreasing injection frequency. In certain embodiments, increasing tolerance to GA therapy in a patient with relapsing-remitting multiple sclerosis comprises decreasing the frequency of injection site reactions.
In another aspect, the invention also provides a method of reducing the frequency of relapse in a human patient suffering from relapsing-remitting multiple sclerosis or a human patient who has experienced a first clinical episode and is determined to be at high risk of developing clinically determined multiple sclerosis, the method comprising administering to the patient a therapeutically effective regimen of a single injection of a depot formulation of a 40mg dose of GA every 2 to 6 weeks, which is sufficient to reduce the frequency of relapse in the patient.
In another aspect, the invention also provides a method of preventing or delaying the progression of relapsing-remitting multiple sclerosis (RRMS) in a human patient suffering from RRMS, said method comprising administering to the human patient a therapeutically effective regime of a single intramuscular injection of a depot formulation comprising a 40mg dose of Glatiramer Acetate (GA) every 2 to 6 weeks.
In certain embodiments, the regimen is sufficient to improve (a) the frequency of relapse, (b) the number or volume of enhanced lesions, (c) the number of new lesions, or (d) the Expanded Disability Status Scale (EDSS) score in the patient. In certain embodiments, the regimen is sufficient to improve the patient's frequency of relapse, number or volume of enhanced lesions, number of new lesions, and Expanded Disability Status Scale (EDSS) score.
In another aspect, the invention also provides a method of preventing disease activity in a human patient with relapsing-remitting multiple sclerosis, said method comprising administering to the patient a therapeutically effective regimen of a single injection of a depot formulation of 40mg dose of GA every 2 to 6 weeks, which is sufficient to prevent relapse, 12 weeks of established disability progression (CDP), new lesion formation and enhancement of existing lesions in the patient.
In another aspect, the present invention also provides a kit comprising a first container containing GA encapsulated by poly (lactic-co-glycolic acid) (PLGA) and a second, separate container containing a pharmaceutically acceptable diluent for injection.
In certain embodiments, the kit comprises 40mg GA and 2 to 10ml WFI. In certain embodiments, mixing the contents of the first container with 2ml of diluent provides 40mg of GA/2ml of suspension. In certain embodiments, the kit is for use in a method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS), increasing GA therapy tolerance, preventing or slowing the progression of RRMS, or preventing RRMS activity.
Further embodiments and full scope of applicability of the present invention will become apparent from the detailed description given hereinafter. However, it should be understood that the detailed description and specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
Brief Description of Drawings
FIG. 1 shows saline controls for the C57BL/6 mouse group,Mean clinical score results (2mg GA, days 0-8) and GA depot (4mg GA, day 0). All treatment groups P compared to untreated control group<0.05, one-way ANOVA followed by a one-tailed T-test assuming unequal variance. N ═ 20/group, +/-standard error.
FIG. 2 shows saline controls for the group of C57BL/6 mice,Average body weight results (2mg GA, days 0-8) and GA reservoir (4mg GA, day 0). All treatment groups compared to untreated control group, P<0.05,**Group compared to untreated control group, P<0.05, one-way ANOVA followed by a one-tailed T-test assuming unequal variance. N ═ 20/group, +/-standard error.
Detailed Description
The invention provides a long-acting depot formulation of glatiramer acetate which provides a daily injection or a three-time-weekly injectionComparable or better therapeutic efficacy and thus lead to improved patient compliance. In addition to providing equivalent or better therapeutic efficacy, the long-acting injection or implant of the present invention reduces the risk of frequent injectionsResulting in adverse side effects.
The present invention is based on the surprising finding that: the current standard treatment for MS patients, i.e. administration of 20mg GA per day by injection, is unnecessarily aggressive in terms of total drug used and frequency of invasive administration. In particular, it has been surprisingly found that 40mg of GA administered once every 28 days is sufficient to achieve a beneficial clinical efficacy, which is at least comparable to the beneficial clinical efficacy of GA administered daily over a 28 day period at a total dose of 560 mg. The present invention thus provides a method of treating MS with 14-fold reduction in the dose administered and 28-fold reduction in the frequency of administration compared to the original and ongoing standard of care.
The present invention is also superior to the more recentA treatment regimen comprising administering 40mgGA three times per week. In this case, treatment of RRMS according to the invention provides a 12-fold reduction in total dose administered and GA administration frequency.
In one aspect, the invention provides a method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS) in a human patient having RRMS or having experienced a first clinical episode or a human patient determined to be at high risk of developing clinically determined multiple sclerosis, the method comprising administering to the patient a therapeutically effective regimen of a single injection of a long acting depot formulation comprising a dose of at least 20mg of glatiramer or any pharmaceutically acceptable salt of glatiramer every 2 to 6 weeks, the regimen being sufficient to alleviate at least one symptom of the patient, wherein the symptom is selected from the group consisting of the patient's frequency of relapses, the number of enhanced or new lesion images in the brain, and the Extended Disability Status Scale (EDSS) score.
In one aspect, the invention provides a method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS) in a human patient having RRMS or having experienced a first clinical episode or a human patient determined to be at high risk of developing clinically determined multiple sclerosis, the method comprising administering to the patient a therapeutically effective regimen of a single injection of a long acting depot formulation comprising a dose of at least 20mg of glatiramer or any pharmaceutically acceptable salt of glatiramer every 2 to 6 weeks, the regimen being sufficient to alleviate at least one symptom of the patient, wherein the symptom is selected from the group consisting of the patient's frequency of relapses, the number of enhanced or new lesion images in the brain, and the Extended Disability Status Scale (EDSS) score.
In certain embodiments, the depot formulation is administered once every 3 to 5 weeks. In certain embodiments, the depot formulation is administered once every 4 weeks.
The term "long acting" as used herein refers to a composition that provides for extended (systemic circulation) or sustained (extended) release of glatiramer to the systemic circulation of a subject or to a site of local action in a subject. The term may also refer to compositions that provide an extended, sustained or prolonged duration of action (pharmacodynamics) of glatiramer salt in a subject. Depending on the desired duration of action expected, each depot or implantable device of the invention will typically contain between about 40 and 520mg of active ingredient, which depot or implantable device is designed to release once over a period of 4 weeks or 13 times every four weeks over a period of one year.
The term "treatment" as used herein refers to the inhibition or alleviation of at least one symptom after the onset of multiple sclerosis. Common symptoms after the onset of multiple sclerosis include, but are not limited to, vision loss or loss, teetering and gait irregularity, unclear mouth and teeth, and urinary frequency and incontinence. In addition, multiple sclerosis can cause mood changes and depression, muscle spasms and severe paralysis.
The phrase "alleviating at least one symptom of RRMS" as used herein refers to any beneficial change in the severity of the symptom or frequency of the symptom of RRMS. The phrase also includes preventing or delaying progression of RRMS symptoms compared to an untreated RRMS patient, or compared to a RRMS patient treated by other therapy, drug, or regimen.
The phrase "a human patient determined to have a high risk of developing clinically established multiple sclerosis" as used herein refers to a patient defined as being in at least one risk group for developing RRMS. The RRMS risk group is long known and reviewed by, for example, McKay et al (biomed.res.int.,2015, vol.2015, Article ID 817238). In certain embodiments, a human patient determined to have a high risk of developing clinically established multiple sclerosis has MRI characteristics consistent with multiple sclerosis. In certain embodiments, a human patient determined to be at high risk of developing clinically established multiple sclerosis has experienced a first clinical episode and has MRI characteristics consistent with multiple sclerosis.
The term "multiple sclerosis" as used herein refers to an autoimmune disease of the central nervous system accompanied by one or more of the symptoms described above.
The term "glatiramer acetate" as used herein refers to what has previously been referred to as copolymer 1, under the trade nameCompounds are sold and include acetate salts of synthetic polypeptides containing four naturally occurring amino acids: l-glutamic acid, L-alanine, L-tyrosine and L-lysine with average molar fractions of 0.141, 0.427, 0.095 and 0.338, respectively.The average molecular weight of glatiramer acetate in (B) is 4,700-11,000 daltons (FDA)Tags) and the number of amino acids ranges between about 15 to about 100 amino acids. The term also refers to chemical derivatives and analogs of the compound. Dian (Chinese character)Formally, the compounds are described in U.S. patent No. 5,981,589; 6,054,430 No; 6,342,476 No; 6,362,161 No; 6,620,847 No; and preparation and characterization as described in any of No. 6,939,539.
According to some embodiments, glatiramer acetate comprises acetate salts of L-alanine, L-glutamic acid, L-lysine, and L-tyrosine in a molar ratio of about 0.14 glutamic acid, about 0.43 alanine, about 0.10 tyrosine, and about 0.33 lysine. According to other embodiments, glatiramer acetate or other pharmaceutically acceptable salts of glatiramer comprise from about 15 to about 100 amino acids. According to certain embodiments, the implantable depot is suitable for subcutaneous or intramuscular implantation. According to an alternative embodiment, the long acting parenteral pharmaceutical composition comprises a pharmaceutically acceptable biodegradable carrier or a non-biodegradable carrier for glatiramer salt. According to some embodiments, the carrier is selected from the group consisting of PLGA, PLA, PGA, polycaprolactone, polyhydroxybutyrate, polyorthoesters, polyalkaneanhydrides, gelatin, collagen, oxidized cellulose, and polyphosphazene. According to some embodiments, the carrier is selected from PLGA, PLA, PGA and any combination thereof. Each possibility represents a separate embodiment of the invention.
In certain embodiments, the at least one symptom that is alleviated is the frequency of relapse and the number of enhanced lesions or the number of images of new lesions on the MRI of the brain.
In certain embodiments, the at least one symptom that is alleviated is the number of enhanced lesions or the number of images of new lesions and the Expanded Disability Status Scale (EDSS) score of the MRI of the brain of the patient.
In certain embodiments, the at least one symptom that is alleviated is the frequency of relapse in the patient and the patient's Expanded Disability Status Scale (EDSS) score.
In certain embodiments, the at least one symptom that is alleviated is the frequency of relapse, the number of enhanced lesions or new lesion images on MRI of the brain, and the Expanded Disability Status Scale (EDSS) score of the patient.
The copolymers may be prepared by any procedure available to those skilled in the art. For example, the copolymers can be prepared under condensation conditions using the desired molar ratio of amino acids in solution or by solid phase synthesis procedures. The condensation conditions include appropriate temperature, pH and solvent conditions for the condensation of the carboxyl group of one amino acid with the amino group of another amino acid to form a peptide bond. Condensing agents, such as dicyclohexylcarbodiimide, may be used to promote the formation of peptide bonds.
In some embodiments, the composition may comprise any other pharmaceutically acceptable salt of glatiramer including, but not limited to, sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogen phosphate, dihydrogen phosphate, metaphosphate, pyrophosphate, hydrochloride, hydrobromide, hydroiodide, acetate, nitrate, propionate, decanoate (decanoate), octanoate, acrylate, formate, isobutyrate, decanoate (caprate), heptanoate, valproate (propiolate), oxalate, malonate, succinate, tocopheryl succinate, suberate, sebacate, fumarate, maleate, butyne-1, 4-dioate, hexyne-1, 6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylenesulfonate, phenylacetate, phenylpropionate, citrate, lactate, β -hydroxybutyrate, mandelate, tartrate, mesylate, tosylate, and like salts of embodiments.
In certain embodiments, the dosage form includes, but is not limited to, a biodegradable injectable depot system, such as a PLGA-based injectable depot system; non-PLGA based injectable depot systems, and injectable biodegradable gels or dispersions. Each possibility represents a separate embodiment of the invention. The term "biodegradable" as used herein refers toIs a component that erodes or degrades at its surface over time, at least in part due to contact with substances found in the surrounding tissue fluid or through cellular action. In particular, the biodegradable component is a polymer such as, but not limited to, a lactic acid based polymer, e.g. a polylactide, e.g. poly (D, L-lactide), i.e. PLA; glycolic acid based polymers such as polyglycolic acid (polyglycolide) or polyglycolic acid (PGA), for examplePoly (D, L-lactide-co-glycolide), i.e. PLGA, (L-lactide-co-glycolide)RG-504、RG-502、RG-504H、RG-502H、RG-504S、RG-502S、) (ii) a Polycaprolactone, such as poly (e-caprolactone), PCLA polyanhydride; poly (sebacic acid) SA; poly (ricinoleic acid) RA; polyfumaric acid, FA; poly (fatty acid dimer), FAD; poly (terephthalic acid), TA; poly (isophthalic acid), IPA; poly (p-carboxyphenoxy) methane, CPM; poly (p-carboxyphenoxypropane), CPP; poly (p-carboxyphenoxyhexane) CPH; polyamine, polyurethane, polyesteramide, polyortho acidEsters, CHDM cis/trans-cyclohexyldimethanol, HD: 1, 6-hexanediol, DETOU: (3, 9-diethylene-2, 4,8, 10-tetraoxaspiro undecane); polydioxanone; polyhydroxybutyrate; polyalkylene oxalates; a polyamide; a polyester amide; a polyurethane; a polyacetal; polyketal; a polycarbonate; a poly (orthocarbonate); a polysiloxane; polyphosphazene; a succinate salt; hyaluronic acid; poly (malic acid); poly (amino acids); a polyhydroxy valerate; polyalkylene succinates; polyvinylpyrrolidone; polystyrene; synthesizing a cellulose ester; polyacrylic acid; poly (butyric acid); triblock copolymers (PLGA-PEG-PLGA), triblock copolymers (PEG-PLGA-PEG), poly (N-isopropylacrylamide) (PNIPAAm), poly (ethylene oxide) -poly (propylene oxide) -poly (ethylene oxide) triblock copolymers (PEO-PPO-PEO), poly (valeric acid); polyethylene glycol; polyhydroxyalkylcellulose; chitin; chitosan; polyorthoesters and copolymers, terpolymers; lipids such as cholesterol, lecithin; poly (glutamic acid-ethyl glutamate copolymer) and the like, or mixtures thereof.
In some embodiments, the compositions of the present invention comprise a biodegradable polymer selected from, but not limited to: PLGA, PLA, PGA, polycaprolactone, polyhydroxybutyrate, polyorthoesters, polyalkaneanhydrides, gelatin, collagen, oxidized cellulose, polyphosphazene, and the like. Each possibility represents a separate embodiment.
In certain embodiments, the biodegradable polymer is a lactic acid-based polymer, such as polylactide or poly (D, L-lactide-co-glycolide), i.e., PLGA. The biodegradable polymer is present in an amount between about 10% to about 98% w/w of the composition. The lactic acid-based polymers have a monomer ratio of lactic acid to glycolic acid in the range of 100: 0 to about 0: 100, for example 100: 0 to about 10: 90, and have an average molecular weight of from about 1,000 to 200,000 daltons. It will be appreciated, however, that the amount of biodegradable polymer is determined by parameters such as duration of use.
According to a particular embodiment, the long acting pharmaceutical composition of the invention is in the form of microparticles prepared by an aqueous-in-oil-in-aqueous double emulsion process. In certain embodiments, the long acting pharmaceutical composition of the present invention comprises an internal aqueous phase comprising a therapeutically effective amount of a pharmaceutically acceptable salt of glatiramer, a water immiscible polymer phase comprising a carrier selected from the group consisting of biodegradable polymers and non-biodegradable polymers, and an external aqueous phase. In other embodiments, the water-immiscible polymer phase comprises a biodegradable polymer selected from the group consisting of PLGA, PLA, and combinations thereof. Each possibility represents a separate embodiment of the invention. In a further embodiment, the external aqueous phase comprises a surfactant selected from the group consisting of polyvinyl alcohol (PVA), polysorbates, polyethylene oxide-polypropylene oxide block copolymers, and cellulose esters. Each possibility represents a separate embodiment of the invention.
The compositions of the present invention may also comprise one or more pharmaceutically acceptable excipients selected from, but not limited to, co-surfactants, solvents/co-solvents, water-immiscible solvents, water-miscible solvents, oily components, hydrophilic solvents, emulsifiers, preservatives, antioxidants, antifoaming agents, stabilizers, buffers, pH adjusters, osmotic agents, channeling agents, osmotic adjusting agents, or any other excipient known in the art suitable co-surfactants include, but are not limited to, polyethylene glycols, polyethylene oxide-polypropylene oxide block copolymers known as "poloxamers", polyglycerol fatty acid esters such as decaglycerol monolaurate and decaglycerol monomyristate, sorbitan fatty acid esters such as sorbitan monostearate, polyethylene oxide sorbitan fatty acid esters such as polyethylene oxide sorbitan monooleate (Tween), polyethylene oxide fatty acid esters such as polyethylene oxide monostearate, polyethylene oxide alkyl ethers such as polyethylene oxide lauryl ether, polyethylene oxide castor oil, and hardened castor oil such as polyethylene oxide hardened castor oil, and analogs thereof or mixtures thereof each representing a suitable embodiment of the present invention include, but are not limited to, the use of a suitable co-surfactants, such as a suitable co-sorbitol, polyoxyethylene, or sorbitan, polyoxyethylene, or a suitable embodiment of the invention may include, a suitable excipient, such as a suitable excipient, such as a suitable excipient, such as a suitable for the invention, for example, for the invention, for example, for the use of the invention, for the use of a suitable for the invention, for the use of a suitable for the invention, for the.
The particle size of the "water-in-oil-in-water (w/o/w) multiple emulsion" can be determined by various parameters including, but not limited to, the amount of force applied at this step, the mixing speed, the surfactant type and concentration, and the like. Suitable particle sizes range from about 1 to 100 μm.
The reservoir system of the present invention includes any form known to those skilled in the art. Suitable forms include, but are not limited to, biodegradable or non-biodegradable microspheres, implantable rods, implantable capsules, and implantable rings. Each possibility represents a separate embodiment of the invention. Extended release gel reservoirs and erodible matrices are also contemplated. Each possibility represents a separate embodiment of the invention. Suitable implantable systems are described in, for example, US 2008/0063687. Implantable rods can be prepared as known in the art using a suitable micro-extruder.
In some embodiments, depot formulations of the invention include, but are not limited to, suspensions of glatiramer acetate in water, oil, or wax phases; a poorly soluble polyelectrolyte complex of glatiramer acetate; an "in situ" gel-forming matrix based on a water-miscible solvent in combination with glatiramer acetate; and biodegradable polymeric microparticles with incorporated glatiramer acetate. Each possibility represents a separate embodiment of the invention. In particular, the compositions of the present invention are in the form of injectable microparticles in which glatiramer acetate is embedded in a biodegradable carrier or a non-biodegradable carrier. The microparticle composition of the present invention may comprise an aqueous-in-oil-in-aqueous double emulsion. Within the scope of the present invention is a microparticle composition comprising an inner aqueous phase comprising glatiramer or any pharmaceutically acceptable salt thereof, an oily or water-immiscible phase comprising a biodegradable polymer or a non-biodegradable polymer, and an outer aqueous phase. The external aqueous phase may also comprise a surfactant, such as polyvinyl alcohol (PVA), polysorbate, polyethylene oxide-polypropylene oxide block copolymers or cellulose esters. The terms "oil phase" and "water immiscible phase" are used interchangeably herein.
According to some embodiments, glatiramer acetate comprises acetate salts of L-alanine, L-glutamic acid, L-lysine, and L-tyrosine in a molar ratio of about 0.14 glutamic acid, about 0.43 alanine, about 0.10 tyrosine, and about 0.33 lysine.
According to other embodiments, glatiramer acetate or other pharmaceutically acceptable salts of glatiramer comprise from about 15 to about 100 amino acids.
In certain embodiments, the depot formulation is administered once every 2 weeks. In certain embodiments, the depot formulation is administered once every 3 weeks. In certain embodiments, the depot formulation is administered once every 4 weeks. In certain embodiments, the depot formulation is administered once every 5 weeks. In certain embodiments, the depot formulation is administered once every 6 weeks.
According to some embodiments, the long acting pharmaceutical composition is suitable for a dosing regimen ranging from once a week to once every 6 months. According to a particular embodiment, the composition is suitable for administration from once every 2 weeks to once a month. According to some embodiments, the long acting composition comprises a dose of between 20 and 750mg of glatiramer acetate per injection. According to some embodiments, the long acting composition comprises a dose of between 20 and 160mg of glatiramer acetate per injection. According to some embodiments, the long acting composition comprises a dose of between 30 and 50mg of glatiramer acetate per injection.
In certain embodiments, alleviating a symptom comprises reducing the frequency of relapse. In certain embodiments, alleviating a symptom comprises reducing the frequency of relapse in the patient by 30% or more compared to the frequency of relapse measured over a period of 12 months prior to beginning the regimen. In certain embodiments, alleviating a symptom comprises lengthening the time to confirmation of relapse in the patient compared to the time between relapses measured over a period of 12 months prior to beginning the regimen. In certain embodiments, alleviating a symptom comprises reducing the total number of confirmed relapses measured over a 12 month period during the regimen in the patient compared to the 12 month period prior to starting the regimen.
In certain embodiments, alleviating a symptom comprises reducing the number or volume of enhancing lesions in the brain. In certain embodiments, alleviating a symptom comprises reducing the number of new lesions in the brain. In certain embodiments, alleviating a symptom comprises reducing the number of MRI-enhanced lesion images of the brain measured over a 12 month period. In certain embodiments, alleviating a symptom comprises reducing the number of new lesion images of brain MRI measured over a 12 month period. In certain embodiments, alleviating a symptom comprises reducing brain atrophy, reducing the number or volume of Gd-enhancing lesions, reducing T in a patient1Weighting the number or volume of enhanced lesions, or reducing new T2-weighting the number of lesions. In certain embodiments, alleviating a symptom comprises reducing the average cumulative number of Gd-enhancing lesions, reducing the average number of new T2 lesions, reducing the total volume of T2 lesions, or reducing the cumulative number of enhancing lesions on T1-weighted images in the brain of a patient. In certain embodiments, alleviating a symptom comprises reducing the number of new low-intensity lesions on an enhanced T1 scan in the patient or reducing the total volume of low-intensity lesions on an enhanced T1 scan in the patient. Each possibility represents a separate embodiment of the invention. In another embodiment, alleviating a symptom comprises reducing MRI in the patientProgression of monitored disease activity.
The Kurtzke Expanded Disability Status Scale (EDSS) is a method to quantify disability in multiple sclerosis. The EDSS quantifies disability in eight Functional Systems (FS) and allows the neurologist to assign a functional system score (FSs) in each of these functional systems. In certain embodiments, alleviating a symptom comprises reducing the EDSS score of the patient. In certain embodiments, alleviating a symptom comprises reducing the measured EDSS score of the patient over a 12 month period. In certain embodiments, alleviating a symptom comprises reducing the patient's EDSS score by at least 0.5. In certain embodiments, alleviating a symptom comprises reducing the patient's EDSS score by at least 1. In certain embodiments, alleviating a symptom comprises reducing the patient's EDSS score from a score in the range of 5.0-9.5 to a score in the range of 0.0-4.5.
In certain embodiments, alleviating a symptom comprises reducing the level of disability in the patient as measured by the work productivity and activity decline-general health (WPAI-GH) questionnaire, or as measured by the EuroQoL (EQ5D) questionnaire. In certain embodiments, alleviating a symptom comprises reducing a change in Ambulation Index (Ambulation Index) in the patient.
In another embodiment, alleviating a symptom comprises reducing the level of disability in the patient as measured by the EDSS score. In another embodiment, alleviating a symptom comprises reducing a change in EDSS score in the patient.
In certain embodiments of the methods described above, the method reduces symptoms at least as effectively as subcutaneous administration of 20mg GA daily or subcutaneous injection of 40mg GA three times a week. In accordance with the principles of the present invention, the long acting pharmaceutical compositions of the present invention provide equivalent or better therapeutic efficacy to commercially available daily injectable dosage forms, with reduced incidence and reduced severity of side effects at the local and/or systemic level. In some embodiments, the compositions of the present invention provide for extended release or prolonged action of glatiramer in a subject as compared to a substantially similar dose of an immediate release formulation of glatiramer acetate.
In certain embodiments, the depot formulation is self-administered intramuscularly by the patient. In certain embodiments, the depot formulation is injected into the deltoid muscle.
In certain embodiments, the patient is not receiving GA treatment prior to the start of the regimen. In certain embodiments, the patient has received GA treatment prior to the start of the regimen.
The present invention includes the combination therapy of glatiramer acetate or any other pharmaceutically acceptable salt of glatiramer with at least one other active agent within the scope of the present invention including, but not limited to, interferons, such as pegylated or non-pegylated α -interferon, or β -interferon (e.g., interferon β -la or interferon β -lb), or tau-interferon, immunosuppressive agents with optional antiproliferative/antitumor activity, such as mitoxantrone, methotrexate (methotrexate), azathioprine, cyclophosphamide (cyclophosphamide) or steroids, such as methylprednisolone, prednisone or dexamethasone (dexamethasone), or steroid secretases, such as ACTH, adenosine deaminase inhibitors, such as cladribine (cladribine), immunoglobulin IV directed against various T cell surface markers, such as immunoglobulin IV (1998, monoclonal antibody 81, such as monoclonal antibody, 1275, such as monoclonal antibody, monoclonal antibody IV, such as monoclonal antibody IV, monoclonal antibody IVOr alemtuzumab, a TH2 cytokine such as IL-4, IL-10, or a compound that inhibits the expression of a TH1 cytokine such as a phosphodiesterase inhibitor such as pentoxifylline (pentoxifylline), antispasmodics including baclofen (baclofen), diazepam (diazepam), piracetam (piracetam), dantrolene (dantrolene), lamotrigine (lamotrigine), riluzole (trifluzole), tizanidine (tizanidine), clonidine (clonidine), β blockers, cyproheptadine (cyproheptadine), oxypheniramine (orphenadrine) or a chemical derivative of a cannabinoid (cannabinoid), an AMPA glutamate receptor antagonistAnti-agents, for example 2, 3-dihydroxy-6-nitro-7-sulfamoylbenzo (f) quinoxaline, [1,2,3,4, -tetrahydro-7-morpholinyl-2, 3-dioxo-6- (trifluoromethyl) quinoxalin-1-yl) methylphosphonate, 1- (4-aminophenyl) -4-methyl-7, 8-methylene-dioxo-5H-2, 3-benzodiazepine or (-) l- (4-aminophenyl) -4-methyl-7, 8-methylene-dioxo-4, 5-dihydro-3-methylcarbamoyl-2, 3-benzodiazepine, inhibitors of VCAM-1 expression or antagonists of its ligands, for example α 4 β 1 integrin VLA-4 and/or α 4 β 7 integrin antagonists, for example natalizumabAnti-macrophage migration inhibitory factor (anti-MIF); xii) cathepsin S inhibitors; xiii) mTor inhibitors. Each possibility represents a separate embodiment of the invention. An example of one other active agent is FTY720 (2-amino-2- [2- (4-octylphenyl) ethyl) propane-1, 3-diol, which belongs to the class of immunosuppressants; fingolimod).
In another embodiment, prior to administration, the patient has at least 1 brain lesion that can be detected by MRI scanning and is suggestive of multiple sclerosis. In yet another embodiment, the lesion is associated with brain tissue inflammation, myelin sheath damage, or axonal damage. In further embodiments, the lesion is a demyelinating white matter lesion visible on a brain MRI. In another embodiment, the diameter of the white matter lesion is at least 3 mm. In certain embodiments, the patient has at least 1 lesion in the brain detectable by MRI scanning, and wherein the lesion is associated with brain tissue inflammation, myelin sheath damage, or axonal damage. In certain embodiments, the lesion is a demyelinating white matter lesion visible on brain MRI, and the white matter lesion is at least 3mm in diameter. In certain embodiments, the patient has experienced a first clinical episode, and the first clinical episode includes a clinical episode of optic neuritis, blurred vision, diplopia, involuntary rapid eye movement, blindness, loss of balance, tremor, ataxia, vertigo, clumsy limbs, lack of coordination, weakness in one or more limbs, altered muscle tone, muscle stiffness, spasticity, tingling, paresthesia, burning sensation, muscle pain, facial pain, trigeminal neuralgia, stabbing megalgia, burning tingling pain, speech retardation, slurred mouth, speech rhythms, dysphagia, fatigue, bladder problems (including urgency, urinary frequency, incomplete emptying and incontinence), bowel problems (including constipation and bowel loss), impotence, decreased libido, loss of sensation, susceptibility to heat, short-term memory loss, loss of attention or loss of judgment or reasoning. Each possibility represents a separate embodiment of the invention.
In certain embodiments, the frequency of immediate response after injection is reduced relative to subcutaneous administration of 20mg GA per day or subcutaneous injection of 40mg GA three times per week. In certain embodiments, the severity of the immediate response after injection is reduced relative to subcutaneous administration of 20mg GA per day or subcutaneous injection of 40mg GA three times per week. In certain embodiments, the frequency of injection site reactions is reduced relative to subcutaneous administration of 20mg GA per day or three subcutaneous injections of 40mg GA per week.
In certain embodiments, the depot formulation is administered in 2mL of water for injection (WFI) or a buffer containing a suspending agent (e.g., carboxymethylcellulose, CMC), a buffer (e.g., citrate), and a tonicity agent (e.g., NaCl). In certain embodiments, the depot formulation comprises 20% to 30% solids. As used herein, the term "water for injection" refers to sterile purified water that meets regulatory standards for particulates, dissolved solids, organic matter, inorganic matter, microorganisms, and endotoxin contaminants.
In certain embodiments, in PBS under continuous stirring at 37 ℃, (i) up to 14% of the glatiramer is released from the depot formulation over 0 days, and/or (ii) up to 15% of the glatiramer is released from the depot formulation over 1 day, and/or (iii) up to 21% of the glatiramer is released from the depot formulation over 5 days, and/or (iv) up to 25% of the glatiramer is released from the depot formulation over 8 days, and/or (v) up to 34% of the glatiramer is released from the depot formulation over 13 days, and/or (vi) up to 43% of the glatiramer is released from the depot formulation over 15 days, and/or (vii) up to 80% of the glatiramer is released from the depot formulation over 22 days, and/or (viii) up to 96% of the glatiramer is released from the depot formulation over 27 days, and/or (ix) up to 99% of glatiramer is released from the depot formulation over a 32 day period. Each possibility represents a separate embodiment of the invention.
In certain embodiments, in PBS, with continuous stirring at 37 ℃, (i) up to 14% of the glatiramer is released from the depot formulation over 0 days, and (ii) up to 15% of the glatiramer is released from the depot formulation over 1 day, and (iii) up to 21% of the glatiramer is released from the depot formulation over 5 days, and (iv) up to 25% of the glatiramer is released from the depot formulation over 8 days, and (v) up to 34% of the glatiramer is released from the depot formulation over 13 days, and (vi) up to 43% of the glatiramer is released from the depot formulation over 15 days, and vii) up to 80% of the glatiramer is released from the depot formulation over 22 days, and (viii) up to 96% of the glatiramer is released from the depot formulation over 27 days, and (ix) up to 99% of the glatiramer is released from the depot formulation over 32 days.
In certain embodiments, (i) about 14% of the glatiramer is released from the depot formulation over a 0 day period in PBS under continuous stirring at 37 ℃, and/or (ii) about 15% of the glatiramer is released from the depot formulation within 1 day, and/or (iii) about 21% of the glatiramer is released from the depot formulation within 5 days, and/or (iv) about 25% of the glatiramer is released from the depot formulation over a period of 8 days, and/or (v) about 34% of the glatiramer is released from the depot formulation over a 13 day period, and/or (vi) about 43% of the glatiramer is released from the depot formulation over a 15 day period, and/or (vii) about 80% of the glatiramer is released from the depot formulation within 22 days, and/or (viii) about 96% of glatiramer is released from the depot formulation over a 27 day period, and/or (ix) about 99% of the glatiramer is released from the depot formulation over a 32 day period. Each possibility represents a separate embodiment of the invention.
In certain embodiments, in PBS, with continuous stirring at 37 ℃, (i) about 14% of the glatiramer is released from the depot formulation over 0 days, and (ii) about 15% of the glatiramer is released from the depot formulation over 1 day, and (iii) about 21% of the glatiramer is released from the depot formulation over 5 days, and (iv) about 25% of the glatiramer is released from the depot formulation over 8 days, and (v) about 34% of the glatiramer is released from the depot formulation over 13 days, and (vi) about 43% of the glatiramer is released from the depot formulation over 15 days, and (vii) about 80% of the glatiramer is released from the depot formulation over 22 days, and (viii) about 96% of the glatiramer is released from the depot formulation over 27 days, and (ix) about 99% of the glatiramer is released from the depot formulation over 32 days.
In another aspect, the invention also provides a method of increasing GA therapy tolerance in a human patient suffering from relapsing-remitting multiple sclerosis, the method comprising reducing the frequency of subcutaneous injections from a daily subcutaneous injection of a 20mg dose of GA or three subcutaneous injections of a 40mg dose of GA over a seven day period with at least one day interval between each injection to a therapeutically effective regime of a depot formulation of a single intramuscular injection of a 40mg dose of GA or any other pharmaceutically acceptable salt of glatiramer every 2 to 6 weeks, so as to thereby increase GA therapy tolerance in the patient.
In certain embodiments, the depot formulation is administered once every 3 to 5 weeks. In certain embodiments, the depot formulation is administered once every 4 weeks.
In certain embodiments, increasing tolerance to GA treatment in a patient with relapsing-remitting multiple sclerosis comprises decreasing injection frequency. In certain embodiments, increasing tolerance to GA therapy in a patient with relapsing-remitting multiple sclerosis comprises decreasing the frequency of injection site reactions. In another embodiment, the injection site reaction is erythema, hemorrhage, induration, inflammation, lump, pain, pruritus, urticaria or redness occurring immediately surrounding the injection site.
In another embodiment, increasing the tolerance of GA treatment in a patient with relapsing-remitting form of multiple sclerosis comprises decreasing the frequency of immediate reactions after injection. In yet another embodiment, the immediate response after injection is palpitation, fever, flushing, hot flashes, tachycardia, dyspnea, chest discomfort, chest pain, non-cardiac chest pain, weakness, back pain, bacterial infection, chills, cysts, facial edema, fever, flu syndrome, infection, injection site erythema, injection site hemorrhage, injection site induration, injection site inflammation, injection site lumps, injection site pain, injection site pruritus, injection site urticaria, injection site redness, neck pain, migraine, syncope, tachycardia, vasodilation, anorexia, diarrhea, gastroenteritis, gastrointestinal disorders, nausea, vomiting, ecchymosis, peripheral edema, arthralgia, restlessness, anxiety, confusion, foot distress, hypertonia, tension, tremor, speech impairment, tremor, vertigo, bronchitis, diarrhea, cold flashes, cold, Dyspnea, laryngeal spasm, rhinitis, erythema, herpes simplex, pruritus, rash, skin nodules, sweating, urticaria, earache, ocular disorders, dysmenorrhea, urgency, or vaginal candidiasis. In embodiments, the frequency of immediate response after injection or the frequency of injection site response is reduced relative to subcutaneous administration of 20mg glatiramer acetate daily or subcutaneous administration of 40mg glatiramer acetate three times per week.
In another aspect, the invention also provides a method of reducing the frequency of relapses in a human patient with relapsing-remitting multiple sclerosis or a human patient who has experienced a first clinical episode and is determined to be at high risk of developing clinically established multiple sclerosis, comprising administering to the patient a therapeutically effective regimen of a single injection of a depot formulation of 40mg dose of GA or any other pharmaceutically acceptable salt of glatiramer every 2 to 6 weeks, said regimen being sufficient to reduce the frequency of relapses in the patient.
In another aspect, the invention also provides a method of preventing or delaying the progression of relapsing-remitting multiple sclerosis (RRMS) in a human patient suffering from RRMS, said method comprising administering to the human patient a therapeutically effective regime of a single intramuscular injection of a depot formulation comprising a 40mg dose of Glatiramer Acetate (GA) every 2 to 6 weeks.
In certain embodiments, the regimen is sufficient to improve (a) the frequency of relapse, (b) the number or volume of enhanced lesions, (c) the number of new lesions, or (d) the Expanded Disability Status Scale (EDSS) score in the patient.
In certain embodiments, the regimen is sufficient to improve the patient's frequency of recurrence, number or volume of enhanced lesions, number of new lesions, and Expanded Disability Status Scale (EDSS) score.
In another aspect, the invention also provides a method of preventing disease activity in a human patient with relapsing-remitting multiple sclerosis, the method comprising administering to the patient a therapeutically effective regimen of a single injection of a depot formulation of GA at a dose of 40mg every 2 to 6 weeks, the regimen being sufficient to prevent relapse, 12 weeks of established disability progression (CDP), new lesion formation, and enhancement of existing lesions in the patient.
In certain embodiments, preventing RRMS activity comprises achieving evidence of disease free activity (NEDA).
In another aspect, the invention also provides a kit comprising a first container containing GA or any other pharmaceutically acceptable salt of glatiramer encapsulated by poly (lactic-co-glycolic acid) (PLGA) and a second separate container containing a pharmaceutically acceptable diluent for injection.
In certain embodiments, the kit comprises 40mg GA and 2mL WFI. In certain embodiments, the kit further comprises a syringe and a needle. In certain embodiments, mixing the contents of the first and second containers provides 40mg GA/2mL of diluent. In certain embodiments, the kit is for use in a method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS), increasing GA therapy tolerance, arresting or delaying progression of RRMS, or arresting RRMS activity. In certain embodiments, the kit is for use in a method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS), increasing tolerance of GA therapy, arresting or delaying progression of RRMS, or arresting the activity of RRMS, the method comprising a single intramuscular injection of a depot formulation comprising a 40mg dose of Glatiramer Acetate (GA) every 2 to 6 weeks. In certain embodiments of the methods and kits provided above, the GA depot formulation is administered as a 21G or 20G needle.
In another embodiment, the therapeutically effective dose of glatiramer acetate is 40 mg. According to various embodiments of the invention, a therapeutically effective amount of glatiramer corresponds to a range from about 1 mg/day to about 2 mg/day. Alternatively, such therapeutically effective amount of glatiramer corresponds to about 1.5 mg/day. According to certain embodiments, the dosing regimen ranges from once a week, twice a month (about once every two weeks) to once a month. Depending on the desired duration of action, each depot or implantable device of the invention will typically contain between about 20 to 750mg of active ingredient which is designed to be released over a period of time ranging from several weeks to several months.
In one embodiment, the pharmaceutical composition is in the form of a sterile solution. In another embodiment, the pharmaceutical composition further comprises mannitol. In yet another embodiment, the pharmaceutical composition has a pH in the range of 5.5 to 8.5. In one embodiment, the pharmaceutical composition has a pH in the range of 5.5 to 7.0.
In one aspect, the invention provides a depot formulation comprising Glatiramer Acetate (GA) at a dose of 40mg for use in a method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS) in a human patient having or having experienced a first clinical episode and determined to be at high risk of developing clinically-determined multiple sclerosis.
In certain embodiments, the depot formulation is administered to the patient by a therapeutically effective regimen of a single intramuscular injection of the depot formulation every 2 to 6 weeks sufficient to alleviate at least one symptom of the patient, wherein the symptom is selected from the group consisting of frequency of relapse of the patient, number of enhanced or new lesion images on MRI of the brain, and Extended Disability Status Scale (EDSS) score.
According to any of the above embodiments, the depot formulation is administered every 3-5 weeks. According to any of the above embodiments, the depot formulation is administered every 4 weeks.
According to any of the above embodiments, alleviating a symptom comprises reducing the frequency of relapse. According to any of the embodiments above, alleviating a symptom comprises reducing the number or volume of enhanced lesions or the number of new lesions in the brain. According to any of the above embodiments, alleviating a symptom comprises reducing brain atrophy, reducing the number or volume of Gd-enhanced lesions, reducing the number or volume of T1-weighted enhanced lesions, or reducing the number of new T2-weighted lesions in the patient. According to any of the above embodiments, alleviating the symptom comprises reducing the EDSS score of the patient.
According to any one of the above embodiments, the method reduces symptoms at least as effectively as daily subcutaneous administration of 20mg GA or three times a week subcutaneous injection of 40mg GA.
According to any of the above embodiments, the patient has received GA treatment prior to the start of the regimen.
According to any of the above embodiments, the depot formulation further comprises a pharmaceutically acceptable biodegradable carrier selected from the group consisting of poly (lactic-co-glycolic acid) (PLGA), polylactic acid (PLA), polyglycolic acid (PGA), and any combination thereof. According to any one of the above embodiments, the biodegradable carrier is PLGA.
According to any of the above embodiments, the depot formulation is in the form of microparticles comprising an internal aqueous phase comprising GA, a water-immiscible polymer phase comprising a biodegradable polymer or a non-biodegradable polymer, and an external aqueous phase.
According to any of the above embodiments, the depot formulation is in the form of microparticles prepared by a water-in-oil-in-water (w/o/w) double emulsion process.
According to any one of the above embodiments, the internal aqueous phase comprises GA.
According to any one of the above embodiments, the water immiscible polymer phase comprises PLGA.
According to any one of the above embodiments, the external aqueous phase comprises a surfactant selected from polyvinyl alcohol (PVA), polysorbates, polyethylene oxide-polypropylene oxide block copolymers, and cellulose esters.
According to any of the above embodiments, the depot formulation comprises 20% to 30% solids. According to any one of the above embodiments, the weight ratio between GA and the pharmaceutically acceptable biodegradable carrier is between 1:1 and 1: 100. According to any one of the above embodiments, the weight ratio between GA and the pharmaceutically acceptable biodegradable carrier is between 1:5 and 1: 25. According to any one of the above embodiments, the weight ratio between GA and the pharmaceutically acceptable biodegradable carrier is about 11.
According to any of the above embodiments, the depot formulation provides a prolonged release or prolonged action of glatiramer in the subject as compared to an immediate release formulation of a substantially similar dose of glatiramer. According to any of the above embodiments, about 80% of glatiramer is released from the depot formulation in PBS over a 22 day period at 37 ℃ with continuous stirring. According to any of the above embodiments, about 20% of glatiramer is released from the depot formulation in PBS over a 5 day period at 37 ℃ with continuous stirring.
In another aspect, the invention provides a therapeutically effective regimen of a single intramuscular injection of a depot formulation of GA at a dose of 40mg every 2 to 6 weeks for use in a method of increasing tolerance to GA treatment in a human patient suffering from relapsing-remitting multiple sclerosis.
In certain embodiments, the method comprises a therapeutically effective regimen of reducing the frequency of subcutaneous injections from a subcutaneous injection of 20mg dose of GA per day or three subcutaneous injections of 40mg dose of GA at intervals of at least one day between each injection over a 7 day period to a single intramuscular injection of a depot formulation of 40mg dose of GA every 2 to 6 weeks.
In another aspect, the invention provides a depot formulation comprising Glatiramer Acetate (GA) at a dose of 40mg for use in a method of preventing or delaying the progression of relapsing-remitting multiple sclerosis (RRMS) in a human patient having RRMS.
In certain embodiments, the method comprises administering to the human patient a therapeutically effective regime of a single intramuscular injection of the depot formulation every 2 to 6 weeks.
According to any one of the above embodiments, the method is sufficient to improve (a) the frequency of relapse, (b) the number or volume of enhanced lesions, (c) the number of new lesions, or (d) the Expanded Disability Status Scale (EDSS) score in the patient.
According to any one of the above embodiments, the method is sufficient to improve the patient's frequency of relapse, number or volume of enhanced lesions, number of new lesions, and Expanded Disability Status Scale (EDSS) score.
In another aspect, the invention provides a depot formulation of GA at a dose of 40mg for use in a method of preventing disease activity in a human patient suffering from relapsing-remitting multiple sclerosis.
In certain embodiments, the method comprises administering to the patient a therapeutically effective regimen of a single injection of the depot formulation every 2-6 weeks sufficient to prevent relapse, 12 weeks of established disability progression (CDP), new lesion formation, and enhancement of existing lesions in the patient.
In another aspect, the present invention provides a kit comprising a first container comprising GA encapsulated with poly (lactic-co-glycolic acid) (PLGA) and a second separate container comprising a pharmaceutically acceptable diluent for injection.
According to any one of the above embodiments, the kit comprises 40mg GA and 2mL water for injection (WFI).
According to any one of the above embodiments, the contents of the first and second containers provide 40mg GA/2mL of diluent.
According to any one of the above embodiments, the kit is for use in a method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS), increasing GA therapy tolerance, arresting or delaying progression of RRMS, or arresting RRMS activity.
The following examples are presented to more fully illustrate certain embodiments of the invention. They should in no way be construed, however, as limiting the broad scope of the invention. Many variations and modifications of the principles disclosed herein can be readily devised by those skilled in the art without departing from the scope of the invention.
Examples
Example 1: method for the in vitro preparation of 40mg GA depot.
Preparation of external aqueous phase A partially hydrolysed polyvinyl alcohol (PVA) solution at a concentration of 2% w/w in sterile WFI was prepared in a reactor and filtered through a 0.22 μm membrane. (2) A solution of NaCl in sterile WFI was prepared and filtered through a 0.22 μm membrane into a reactor containing PVA. (3) Organic phase preparation an organic phase consisting of methylene chloride and poly (lactide-co-glycolide) was prepared in a reactor and filtered through a 0.22 μm membrane. (4) Internal aqueous phase preparation a solution containing sterile WFI and glatiramer acetate was prepared and filtered through a 0.22 μm membrane. (5) Water-in-oil (w/o) emulsion preparation: the internal aqueous phase was added to the organic phase and treated at 7,200RPM for 10 minutes using an IKA Ultra-Turrax T50 homogenizer equipped with a rotor stator dispersion device (high shear mixing). (6) Water-in-oil-in-water (w/o/w) emulsion preparation: during the continued mixing of the w/o emulsion, the water-in-oil emulsion (w/o) prepared in step 5 is added to half of the external aqueous phase. At the end of the transfer from w/o to the external aqueous phase, the w/o/w multiple emulsion was treated for 3 minutes at 2,900RPM using an IKA Ultra-Turrax UTS80 homogenizer with a rotor stator head. Subsequently, a further 30 liters of external aqueous phase were added to the emulsion (quench). (7) Solvent removal/evaporation: the w/o/w multiple emulsion formed in step (6) was mixed using an IKAUTS80 homogenizer at different speeds for 15-17 hours. Compressed air was bubbled through the emulsion at 0.5Pa for 10-12 hours. A vacuum is applied to a portion of the process. (8) Separation and washing: the suspension was centrifuged at 5,300RPM for 10 minutes. The supernatant was discarded and the pellet (settled particles) was resuspended in 550g of WFI and mixed with a magnetic stirrer for 3 minutes. The resuspended microparticles were centrifuged at 2,900RPM for 10 minutes. (9) And (3) freeze drying: the washed microparticles were resuspended in approximately 750g sterile WFI and kept at-20 ℃ until freeze-dried. Freeze-drying was performed using sterile lyoguard trays as follows: freezing at-40 deg.C for 24 hr. Primary drying at 0.2hPa, -5 ℃ for 48 hours. Drying at 10 deg.C under 0.2hPa for 48 hr. The resulting composition comprised GA and PLGA (50: 50, molecular weight 7,000-17,000) in a weight ratio of 1: 11.5.
Example 2: GA in vitro release profile from PLGA depot at 40mg GA.
The release of incorporated glatiramer acetate was performed in tightly closed 20ml glass bottles using a 37 ℃ incubator equipped with a multi-point magnetic stirrer. Phosphate Buffered Saline (PBS) pH 7.4 was used as release medium. Table 1 summarizes the GA release profile from PLGA depots.
Table 1.
Sky 0 1 5 8 13 15 22 27 32
% GA Release into Medium 14 15 21 25 34 43 80 96 99
Example 3: stock of 4mg GA with commercially available GAThe in vivo test of (1).
Materials and methods
Animal(s) productionWhat is needed isAnimal studies were approved by the Israeli MOH animal Care and Use Committee. C57BL/6 female mice, 7-9 weeks old, were randomly divided into control or treatment groups with similar mean body weights. Throughout the experiment, animals were given food and water ad libitum.
Induction of EAE: to induce EAE, a recognized animal model of multiple sclerosis, an emulsion of MOG 35-55(GL biochem co. ltd, Shanghai, China) in modified Complete Freund's Adjuvant (CFA) (Sigma-Aldrich, st. louis, MO, USA) was prepared as follows: heat killed Mycobacterium tuberculosis (M.tuberculosis) strain H37RA (Sigma) was added to CFA to a final concentration of 4 mg/mL. Subsequently, 2mg/mL MOG 35-55 was emulsified with an equivalent amount of modified CFA. EAE has been induced by subcutaneous injection (SC) of this emulsion at one site of the shaved back of the hair of mice followed by intraperitoneal injection of bordetella pertussis toxin (Sigma) in PBS on day 0 and 48 hours after MOG immunization. A 21G needle was used for injection in mice.
Measuring: body weight was measured every two days from day 0 to day 28. EAE was assessed by once daily clinical scoring of mice from day 0 to day 28 after immunization (table 2). For the analysis, the dead animals received a clinical score of 5 points and the body weight measured the last time before the animals died was recorded.
Table 2: EAE clinical score.
The following calculations were obtained from the clinical scoring raw data: the mean highest score is the mean of the highest scores for each mouse in a particular group up to the indicated number of days analyzed; the mean duration of disease and mean number of episodes were calculated as follows: mean disease duration ═ (sum of days analyzed-days of seizures per mouse)/(number of mice per group); mean number of attack days (sum of days of attack per mouse)/(number of mice per group). The area under the curve (AUC) of clinical scores was calculated using microsoft excel and represents the disease burden.
Glatiramer acetate depot (GA depot): GA depots were suspended in water for injection (WFI) and immediately injected Intramuscularly (IM) at the indicated dose. The dose of GA depot is given according to the amount of active ingredient (i.e. 4mg of GA depot containing 4mg of GA).
GA-binding antibody assay: on day 35 post-disease induction, 5 animals from each treatment group were sacrificed. Blood samples were collected and sera were separated and stored at-80 deg.C (see tables 3 and 5).
ELISA plates were prepared as follows: flat bottom ELISA plates (Nunc) were coated with 100ul of 50. mu.g/ml GA in Borate Buffer (BB)0.17M, pH 8.0 overnight at 4 ℃. The wells were emptied and washed at room temperature with Phosphate Buffered Saline (PBS) containing 0.05% Tween 20. After washing, 1% BSA2 was applied at Room Temperature (RT) for blocking non-specific binding sites. Subsequently, the wells were washed three times with the washing solution.
The ELISA test was performed as follows: mu.l of serum samples were diluted 1:1,000, added to wells, and washed three times at 4 ℃ for 18 hours (serum dilution was performed using PBS containing 1% BSA and 0.05% Tween 20), followed by Phosphate Buffered Saline (PBS) containing 0.05% Tween 20 at room temperature. Subsequently, 100. mu.l of 1:50,000 diluted alkaline phosphatase conjugated affinity purified goat anti-mouse IgG + IgM (H + L) (Jackson Laboratories) was added to the wells and incubated for 2 hours at RT. The wells were washed three more times with wash solution and the chromogenic reaction was performed by adding 100. mu.l of the substrate p-nitrophenyl phosphate (Jackson Laboratories) and incubating at RT for 40-60 minutes. The reaction was stopped with 30. mu.l 3N NaOH. The absorbance at 405nm was then recorded using a micro-ELISA reader (Dynatech). Each assay plate contained a positive anti-GA serum sample and a control of normal mouse serum (N ═ 5).
The results are expressed as Binding Index (BI) according to the following formula: binding index is the mean optical density of the tested sera/the mean optical density of the control sera. The mean value of normal mouse sera was 0.230OD and the cut-off value of the binding index was 2.0 ± 1.0. Thus, values above 3.0 are considered positive.
Design of experiments: the study experimental design is illustrated in table 3.
TABLE 3 experimental design.
Dose was administered using two consecutive days of injection, as there was a maximum injection volume in the mice that could be tolerated by a single injection.
Statistical analysis: data were analyzed using Microsoft Excel. Each data set was analyzed using one-way analysis of variance (ANOVA) followed by a one-tailed student T-test.
Results
GA depot dose switching.
FIG. 1 shows a saline control (black circle marks), 2mgMean clinical score results for the groups (black square markers) and GA reservoir 4mg (grey triangle markers) as these groups represent the range of recommended human doses of 0, 20 and 40mg, respectively (using allogeneic 1:10 ratios subtracted from previous studies). All treatment groups compared to untreated controls, P<0.05, one-way analysis of variance was followed by a one-tailed T-test assuming unequal variance. N ═ 20/group, +/-standard error.
Mean clinical score AUC (area under curve), mean days to onset and mean duration of disease in GA reservoir groups and compared to untreated controlsSignificant reduction in group (Table 4, p)<0.05). No GA stores were found in any of the calculated valuesLibrary andstatistically significant differences between (table 4). On days 11-19, the mean clinical score was significantly higher in the saline group than in all other groups (fig. 1, p)<0.05). Mean clinical scores were significantly higher in the saline group on day 20 than in the saline groupAnd mean clinical score of the GA reservoir group (FIG. 1, p)<0.05). Day 10 to 17 after immunization, GA depot andthe body weight of the treated group was significantly higher than that of the untreated control. On the 21 st day, the day,the body weight of the group was significantly higher than that of the untreated control group (fig. 2, P)<0.05). All treatment groups compared to untreated controls, P<0.05。**Group compared to untreated control, P<0.05. One-way analysis of variance ANOVA was followed by a one-tailed T-test assuming unequal variance. N ═ 20/group, +/-standard error.
Table 4: the values are calculated.
P <0.05 in all treatment groups compared to untreated controls, one-way analysis of variance ANOVA followed by a one-tailed T-test assuming unequal variance. N ═ 20/group, +/-standard error.
Immune responses in mice were measured with antibodies against GA.
On day 35 post-disease induction, sera were isolated from mice in the MOG-EAE study. For miceGA reservoir (4mg each time) or(2 mg/day, day 0-8). Antibody (Ab) titers were assessed using ELISA assays. The results are expressed as the Binding Index (BI). N is 5. The data shown in Table 5 indicate exposure toOr GA reservoir mice produced similar titers of total anti-GA antibodies, whereas saline-treated control mice did not. Antibody titers were similar for all treatment groups, indicating a similar immune response.
Table 5: the binding index.
The effect of a single administration of 4mg GA depot was compared to the effect of a standard daily administration of 2mg GA, and the presented data shows similar efficacy between the two dosing regimens. In particular, GA depots show a clear significant effect in delaying the onset of disease and ameliorating symptoms, at least in combination withThe treatment groups were as effective as those recorded (see figure 1).
Furthermore, this experiment shows that intramuscular administration of GA depot induces a humoral response of anti-GA antibodies at similar levels as standard daily subcutaneous injections of GA (see table 5). Thus, a similar humoral response to the GA depot compared to a standard GA injection is likely to represent a similarity in immune response to the GA depot. This may therefore also indicate an equivalent clinical immunomodulatory therapeutic effect, as can be seen in the present EAE study of whichAnd the AUC between GA depots did not differ in a statistically significant sense (see table 4).Thus, the presence of anti-GA antibodies can serve as a biomarker for the therapeutic bioavailability of the drug when administered using the novel formulations and by the novel route.
Overall, the data support the efficacy of GA depots at 4mg GA doses in MOG-induced EAE at least as good as that of the MOG-induced EAEThe efficacy of (a) was comparable and the immune response to both treatments was similar. Based on the results obtained with the 4mg GA depot in the EAE animal model, it is predicted that a dose of 80mg would be better than 40mg in humans.
Example 4: 40mg and 80mg GA depots in humans diagnosed with RRMS.
Brief summary:a prospective, 1-year, open label, two-arm, multicenter, phase IIa study in which GA reservoirs of 40mg or 80mg GA at 4-week intervals were administered as Intramuscular (IM) injections to subjects with RRMS for 52 weeks of treatment. The objective of this study was to evaluate the safety, tolerability and efficacy of monthly long-acting IM injections of 40mg or 80mg GA depot in subjects with RRMS. The study included administration of 20mg or 40mg, respectively, from three times daily or weeklySubject to whom injection was transferred.
Detailed Description: subjects diagnosed with relapsing-remitting multiple sclerosis (RRMS) were subcutaneously injected with 20mg or 40mg GA three times daily or weekly, respectively, over the first 12 monthsAnd (6) carrying out treatment. The product of the study was a long-acting injection of GA (GA depot) which was a combination of injectable extended-release microspheres and a diluent for use in the gastrointestinal tract (water for injection). GA depot was administered Intramuscularly (IM). Study duration for individual subjects was 60 weeks from 4-week screeningAssessment (week-4 to week 0), followed by an open label treatment period of 52 weeks, and a follow-up period of 4 weeks consisting of: a total of 17 accesses. The body, vital signs and safety line were evaluated at each visit. MRI was performed at visit 1 (screening), week 24 and week 52 (end of treatment visit). Neurological and safety laboratory tests were performed in screening visits and visits at weeks 4, 12, 24, 36 and 52 (end of treatment). GA depots were administered in 21G or 20G needles.
Population and sample size: 25 RRMS patients were used before study enrollmentThe treatment lasts for at least 12 months. Average age when signing informed consent: both groups were 52.7 years old. Average duration of MS: the 40mg group and the 80mg group were 16.5 years and 14.6 years, respectively. Efficacy groups: 80mg was administered to 12 patients and 40mg was administered to 13 patients.
Extension of experiments outside of core research: the 13 patients who completed the core study (13 injections) required continued treatment with 40mg GA depot (the patient with the longest treatment time received his 34 th injection and was therefore in the second half of the third year of his continued treatment).
Inclusion criteria: a male or female subject diagnosed with RRMS; multiple Sclerosis (MS) diagnosed as meeting McDonald criteria (revision 2010); treatment with 20mg or 40mg of GA (COPAXONE) over the previous 12 months, with treatment continuing at the screening visit; normal renal function; normal liver function; normal hemoglobin concentration; without any clinically significant medical, psychiatric or laboratory abnormalities; the ability to provide written informed consent.
Exclusion criteria: any relevant medical, surgical or psychiatric condition, laboratory value or concomitant medication that renders the subject unsuitable for entry into the study or may fail to complete all aspects of the study; severe anemia (hemoglobin)<10 g/dL); renal dysfunction (serum creatinine)>1.5 xULN); pregnant or lactating women; women with fertilityThe woman must have a negative urine pregnancy test at the screening visit and use the appropriate contraceptive method throughout the study. Women with surgical sterilization (hysterectomy or tubal ligation) or women with the last menstruation 12 months or more before the screening visit are considered to be without fertility. Acceptable forms of contraception include oral, implantation or injection of contraceptives, placement of intrauterine devices for at least 3 months, estrogen patches and appropriate barrier methods in combination with spermicides. Abstinence (abstinence) is considered an acceptable contraceptive regimen; any allergic reaction (anapurity reaction) and/or history of severe allergic reactions after vaccination, allergies that have been proven to any component of study drugs, such as GA, poly (lactic-co-glycolic acid) (PLGA), polyvinyl alcohol (PVA); a history of drug or alcohol abuse is known or suspected; positive tests for HIV, hepatitis, venereal disease research laboratory test (VDRL) or tuberculosis; engaging in investigational drug studies within 30 days before the study began; treatment with any kind of steroid during the last 30 days; the excess recurred within the last 30 days.
Measurement of results: the recurrence rate measured during the study was compared to that observed 12 months prior to study initiation; changes in the number of enhanced lesions and the number of new lesion images on brain MRI from baseline to the end of treatment visit; change in the Expanded Disability Status Scale (EDSS) score from baseline to end of treatment visit.
Adverse Events (AEs) included mainly mild Injection Site Reactions (ISRs) and no unexpected AEs were reported. The 40mg dose was reported to be statistically significantly less ISR than the 80mg dose. No detection as in GAThe recorded injection immediately followed the reaction.
Results
Endpoint # 1: the recurrence rate was compared to that observed 12 months prior to study entry. During the study: 1 recurrence was detected for the entire study population (which did not include EDSS)Change). During the year before the test, the test is carried outThe same subjects treated: relapse occurs for 2 times.
Endpoint # 2: brain MRI scans were compared to baseline and changes were defined as new lesions (by T2-FLAIR), changes in enhanced lesions (by SPGR T1GdE), or both.
Table 6: MRI scanning.
1 patient with 80mg showed 7 newly enhanced lesions at 6 months MRI scan; 1 x 40mg patient showed a reduction in enhanced lesions from 3 at baseline to 0 at 6 months; considered "no change"; 1 patient with 40mg showed 1 new intensive lesion at early termination of MRI scan;1 80mg patient showed 2 newly enhanced lesions at 12 months MRI scan;1 80mg patient diagnosed with breast cancer was not MRI scanned at the early termination.
Endpoint # 3: and (4) EDSS scoring.
Table 7: and (4) EDSS scoring.
3 of 12 patients in the 40mg group had baseline EDSS scores of 5-6; patient 1 had no EDSS data at premature termination.
Notably, the data demonstrate that a high proportion of patients treated with the depot formulations of the invention for one year achieve NEDA, defined as no evidence of disease activity; defined as the absence of all of the following: relapse, 12-week-Confirmed Disability Progression (CDP), new/enlarged T2 lesions, and T1 gadolinium-enhanced lesions. This proportion of patients who reach NEDA is high, particularly compared to NEDA values for other drugs approved for the treatment of MS.
Table 8: NEDA.
40mg group 80mg group All are
Each protocol (PP) group 8(100%) 3(60%) 11(84.6%)
mlTT*(LOCF**) 11(91.7%) 8(72.7%) 19(82.6%)
Modified intent-to-treat (mITT) population with data for 3 NEDA parameters at week 52 or premature termination or week 24. 1 patient was excluded because there was no MRI assessment other than baseline. Last observation of metastasis (LOCF).
And (4) conclusion:
treatment with a single intramuscular injection of 40mg GA depot was superior to the corresponding 80mg GA treatment in preventing new lesion formation and lesion enhancement (table 6).
Treatment with a single intramuscular injection of 40mg GA depot was comparable to the corresponding 80mg GA treatment in preventing an increase in EDSS score (table 7).
Treatment with depot compositions of GA provides for one year's co-treatment(47%)、(34%) and(39%) (as reported by Rotstein et al, JAMA neurol, 2015, Vol.72(2):152- & 158) compared to better results for NEDA.
Although the present invention has been described in detail, those skilled in the art will appreciate that many variations and modifications may be made. Accordingly, the invention is not to be considered as limited to the specifically described embodiments, and the scope and concept of the invention will be more readily understood by reference to the claims that follow.

Claims (30)

1. A method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS) in a human patient having RRMS or having experienced a first clinical attack and determined to be at high risk of developing clinically determined multiple sclerosis, the method comprising administering to the patient a single intramuscular injection of a therapeutically effective regimen comprising a 40mg dose of Glatiramer Acetate (GA) of a depot formulation every 2 to 6 weeks, the regimen being sufficient to alleviate at least one symptom of the patient, wherein the symptom is selected from the group consisting of frequency of relapses in the patient, number of enhanced or new lesion images in an MRI brain, and an Extended Disability Status Scale (EDSS) score.
2. The method of claim 1, wherein the depot formulation is administered once every 4 weeks.
3. The method of claim 1 or claim 2, wherein alleviating a symptom comprises reducing the frequency of relapse.
4. The method of any one of the preceding claims, wherein alleviating a symptom comprises reducing the number or volume of enhanced lesions or the number of new lesions in the brain.
5. The method of claim 4, wherein alleviating a symptom comprises reducing brain atrophy, reducing the number or volume of Gd-enhancing lesions, reducing T in the patient1Weighted number or volume of enhanced lesions or reduction of new T2-the number of weighted lesions.
6. The method of any one of claims 1-4, wherein alleviating a symptom comprises reducing the patient's EDSS score.
7. The method of any one of the preceding claims, wherein the method reduces the symptoms at least as effectively as subcutaneous administration of 20mg GA daily or subcutaneous injection of 40mg GA three times a week.
8. The method of claim 1 or claim 7, wherein the patient has received GA treatment prior to the beginning of the regimen.
9. The method according to any one of the preceding claims, wherein the depot formulation further comprises a pharmaceutically acceptable biodegradable carrier selected from the group consisting of poly (lactic-co-glycolic acid) (PLGA), polylactic acid (PLA), polyglycolic acid (PGA), and any combination thereof.
10. The method of claim 9, wherein the biodegradable carrier is PLGA.
11. The method of any one of the preceding claims, wherein the depot formulation is in the form of microparticles comprising an internal aqueous phase comprising GA, a water-immiscible polymer phase comprising a biodegradable polymer or a non-biodegradable polymer, and an external aqueous phase.
12. The method of any one of the preceding claims, wherein the depot formulation is in the form of microparticles prepared by a water-in-oil-in-water (w/o/w) double emulsion process.
13. The process of claim 12, wherein the internal aqueous phase comprises the GA.
14. The method of any one of claims 11 to 13, wherein the water-immiscible polymer phase comprises PLGA.
15. The method of any one of claims 11 to 14, wherein the external aqueous phase comprises a surfactant selected from polyvinyl alcohol (PVA), polysorbates, polyethylene oxide-polypropylene oxide block copolymers, and cellulose esters.
16. The method of any one of the preceding claims, wherein the depot formulation comprises 20% to 30% solids.
17. The process according to any one of claims 9 to 16, wherein the weight ratio between the GA and the pharmaceutically acceptable biodegradable carrier is between 1:1 and 1: 100.
18. The method of claim 17, wherein the weight ratio between the GA and the pharmaceutically acceptable biodegradable carrier is between 1:5 and 1: 25.
19. The method of any one of the preceding claims, wherein the depot formulation provides a prolonged release or prolonged effect of glatiramer in the subject as compared to an immediate release formulation of a substantially similar dose of glatiramer.
20. The method of claim 19, wherein about 80% of the glatiramer is released from the depot formulation in PBS over a 22 day period with continuous stirring at 37 ℃.
21. The method of claim 19 or claim 20, wherein about 20% of glatiramer is released from the depot formulation in PBS over a 5 day period at 37 ℃ with continuous stirring.
22. A method of increasing GA therapy tolerance in a human patient suffering from relapsing-remitting form of multiple sclerosis, the method comprising reducing the frequency of subcutaneous injections from a daily subcutaneous injection of a 20mg dose of GA or three subcutaneous injections of a 40mg dose of GA spaced at least one day between each injection over a seven day period to a therapeutically effective regime of a depot formulation with a single intramuscular injection of a 40mg dose of GA every 2 to 6 weeks, to thereby increase GA therapy tolerance in the patient.
23. A method of preventing or delaying the progression of relapsing-remitting multiple sclerosis (RRMS) in a human patient having RRMS, the method comprising administering to the human patient a therapeutically effective regimen of a single intramuscular injection of a depot formulation comprising a 40mg dose of Glatiramer Acetate (GA) every 2 to 6 weeks.
24. The method of claim 23, wherein the regimen is sufficient to improve (a) frequency of relapse, (b) number or volume of enhanced lesions, (c) number of new lesions, or (d) Expanded Disability Status Scale (EDSS) score in the patient.
25. The method of claim 24, wherein the regimen is sufficient to improve the patient's frequency of relapse, number or volume of enhanced lesions, number of new lesions, and Expanded Disability Status Scale (EDSS) score.
26. A method of preventing disease activity in a human patient with relapsing-remitting form of multiple sclerosis, the method comprising administering to the patient a therapeutically effective regimen of a single injection of a depot formulation of 40mg dose of GA every 2 to 6 weeks, the regimen being sufficient to prevent relapse, 12 weeks of established disability progression (CDP), new lesion formation, and enhancement of existing lesions in the patient.
27. A kit comprising a first container comprising GA encapsulated by poly (lactic-co-glycolic acid) (PLGA) and a second separate container comprising a pharmaceutically acceptable diluent for injection.
28. The kit of claim 27, wherein the kit comprises 40mg GA and water for injection (WFI).
29. The kit of claim 27 or claim 28, wherein mixing the contents of the first and second containers provides 40mg GA per 2ml of diluent.
30. The kit of any one of claims 27 to 29, for use in a method of alleviating at least one symptom of relapsing-remitting multiple sclerosis (RRMS), increasing GA therapy tolerance, preventing or delaying progression of RRMS, or preventing RRMS activity.
HK19129347.1A 2016-08-31 2017-08-09 Depot systems comprising glatiramer acetate HK40005874A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US62/381,598 2016-08-31

Publications (1)

Publication Number Publication Date
HK40005874A true HK40005874A (en) 2020-05-15

Family

ID=

Similar Documents

Publication Publication Date Title
JP5916622B2 (en) Depot system containing glatiramer or a pharmacologically acceptable salt thereof
JP7193448B2 (en) Depot system containing glatiramer acetate
US12343371B2 (en) Method for suppressing or alleviating primary or secondary progressive multiple sclerosis (PPMS or SPMS) using a sustained release depot formulation comprising glatiramer acetate
US8377885B2 (en) Depot systems comprising glatiramer or pharmacologically acceptable salt thereof
USRE50301E1 (en) Depot systems comprising glatiramer or pharmacologically acceptable salt thereof
HK40005874A (en) Depot systems comprising glatiramer acetate
HK40010052A (en) Glatiramer depot systems for treating progressive forms of multiple sclerosis