NZ619924B2 - Combination als therapy - Google Patents

Combination als therapy Download PDF

Info

Publication number
NZ619924B2
NZ619924B2 NZ619924A NZ61992412A NZ619924B2 NZ 619924 B2 NZ619924 B2 NZ 619924B2 NZ 619924 A NZ619924 A NZ 619924A NZ 61992412 A NZ61992412 A NZ 61992412A NZ 619924 B2 NZ619924 B2 NZ 619924B2
Authority
NZ
New Zealand
Prior art keywords
riluzole
subject
effective amount
dose
therapeutically effective
Prior art date
Application number
NZ619924A
Other versions
NZ619924A (en
Inventor
Jesse Cedarbaum
Fady Malik
John Mao
Andrew A Wolff
Original Assignee
Cytokinetics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cytokinetics Inc filed Critical Cytokinetics Inc
Priority claimed from PCT/US2012/046523 external-priority patent/WO2013010015A2/en
Publication of NZ619924A publication Critical patent/NZ619924A/en
Publication of NZ619924B2 publication Critical patent/NZ619924B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/428Thiazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

Provided is a method for treating ALS in a subject, comprising administering to the subject a therapeutically effective amount of riluzole and a therapeutically effective amount of CK-2017357 (Tirasemtiv). Also provided are methods of reducing the variability of riluzole exposure (e.g., Cmax and/or AUC24h) in a subject, methods of reducing the variability of riluzole exposure (e.g., Cmax and/or AUC24h) between two or more subjects, methods of decreasing the total daily dose of riluzole in a subject, methods of increasing the half-life of riluzole in a subject, methods for decreasing the frequency of riluzole dosing in the subject, and methods for reducing the incidence and/or severity of adverse events in a subject treated with riluzole. AUC24h) in a subject, methods of reducing the variability of riluzole exposure (e.g., Cmax and/or AUC24h) between two or more subjects, methods of decreasing the total daily dose of riluzole in a subject, methods of increasing the half-life of riluzole in a subject, methods for decreasing the frequency of riluzole dosing in the subject, and methods for reducing the incidence and/or severity of adverse events in a subject treated with riluzole.

Description

Combination ALS Therapy This application claims the benefit of priority to US Appln. Nos. 61/507,381, filed July 13, 2011, 61/637,770, filed April 24, 2012, 61/544,533, filed October 7, 2011; 61/646,699, filed May 14, 2012, and 61/637,759, filed April 24, 2012, each of which is incorporated by reference for all purposes.
Amyotrophic lateral sclerosis (ALS) is a degenerative and progressive disorder of the nervous system. ALS is characterized by progressive loss of motor neurons in the lateral column of the spinal cord and/or motor cortex. With ssive loss of motor neurons, the innervation to skeletal muscle is lost which results in an inability to ambulate, conduct daily ties and affects swallowing and breathing. ALS is a rare and usually fatal disease; its progression can be variable but on average patients end up dying within 3 years of diagnosis. Most patients succumb to respiratory failure with broncho-pneumonia and pneumonia as the main causes of death. Treatment is tive because no curative treatment exists. le currently is the only drug ed to treat ALS. While the exact mechanism of action of riluzole is unknown, it is believed to act by inhibiting the deleterious effects of an overload of ic acid and other ransmitters in the central nervous system (see, e.g., ier et al., NeuroReport (1994), 5(8):1012-14; Estevez et al., Eur. J. Pharmacol. (1995), 280(1): 47-53; Rothstein et al., J. Neorochem. (1995), 65(2): 643-51). Studies have shown that the serum and plasma concentrations of riluzole vary y from person to person (see, e.g., Groeneveld et al., J. Neurol. Sci. , 191: 310-13). Such variability can lead to difficulties in determining and delivering a eutically effective dose to a patient and may affect the incidence and severity of adverse events (see, e.g., Groeneveld et al., Neurology (2003), 61: 1141-43). 6-Ethynyl(pentanyl)—1H-imidazo[4,5-b]pyrazin-2(3H)—one (also known as CK-2017357 or ) is a selective activator of the fast skeletal muscle troponin complex, which sensitizes fast skeletal muscle to calcium and results in an amplification of the response to neuromuscular input, an increase in muscle power, and a decrease in the fatigability of muscle (see, e.g., US. Patent No. 248). The use of CK-2017357 is being studied for use in the treatment of patients with ALS. By increasing the contractile force developed by fast skeletal muscle fibers in response to any level of neuronal input, treatment with CK-2017357 may improve activities of daily living (such as tion, ing and feeding), reduce hospitalizations, and possibly prolong the survival of patients with ALS.
CK-2017357 has been shown to be a mechanism-based inhibitor of human CYP1A2 in vitro, with a KI ivation constant) of 1.9 uM and kinact (max rate constant for vation) of 0.031 min'1. In humans, riluzole is primarily metabolized by CYP1A2 (see, e.g., Sanderink et al., J. Pharmacol. & Exp. Ther. (1997), 282(3): 1465-72). A number of other drugs have been assessed for their ability to inhibit the metabolism of riluzole by human hepatic microsomes in vitro; although inhibition has been observed in some cases (e.g., amitriptyline, diclofenac, diazepam, nicergoline, clomipramine, imipramine), the high concentrations required for inhibition make it unlikely that these drugs would alter riluzole trations if used in combination with riluzole in the al setting (see Bensimon et al., Expert Opin. Drug Saf. (2004), 3(6): 525-34).
Provided is a method for treating ALS in a subject, comprising administering to the t a therapeutically effective amount of riluzole and a therapeutically effective amount of CK-2017357.
Also provided is a method for reducing the variability of riluzole exposure (e.g., Cmax and/or ) in a subject, comprising administering to the subject a therapeutically effective amount of 7357.
Also provided is a method for ng the variability of riluzole exposure (e.g., Cmax and/or AUC24h) between two or more subjects, comprising administering to the subjects a therapeutically effective amount of CK-2017357.
Also provided is a method of increasing the exposure of riluzole (e.g., Cmax and/or AUC24h) in a subject, comprising administering to the subject a therapeutically effective amount of 7357.
Also provided is a method of increasing the half-life of riluzole in a subject, comprising administering to the subject a therapeutically effective amount of CK-2017357.
Also provided is a method for decreasing the frequency of riluzole dosing in the subject, comprising administering to the subject a eutically effective amount of CK-2017357.
Also provided is a method for decreasing the total daily dose of riluzole in a t, comprising administering to the subject a therapeutically effective amount of CK-2017357.
Also provided is a method for reducing the incidence and/or ty of adverse events in a subject treated with riluzole, comprising administering to the subject treated with le a therapeutically effective amount of CK-2017357.
Also provided is a pharmaceutical composition comprising a therapeutically effective amount of riluzole and a therapeutically effective amount of CK-2017357.
Also provided is a method for treating ALS in a subject, comprising administering to the subject at least two doses daily of CK-2017357.
DESCRIPTION OF FIGURES Figure 1 provides mean riluzole plasma concentration as a function of time for riluzole alone (50 mg single dose) and le (50 mg single dose) and CK-2017357 (250 mg daily x 11 days in healthy subjects.
Figure 2 rates the effect of steady-state CK-2017357 on riluzole pharmacokinetics in healthy subjects. Panel A provides Cmax for riluzole alone (50 mg single dose) and riluzole (50 mg single dose) + CK-2017357 (250 mg daily for 11 days). Panel B provides AUCime for riluzole alone (50 mg single dose) and riluzole (50 mg single dose) + CK-2017357 (250 mg daily for 11 days). The figure shows that in healthy subjects, the inter-subject variability of Cmax of riluzole has been reduced through co-administration of CK-2017357.
Figure 3 provides mean riluzole plasma concentration as a function of time for 50 mg riluzole administered BID and CK-2017357 administered at either 250 or 500 mg single dose in ALS ts.
Figure 4 shows a study flow diagram for a clinical study investigating a daily dose titration regimen of CK-2017357 in patients with ALS.
As used herein, the following abbreviations, words and phrases are generally intended to have the gs as set forth below, except to the extent that the context in which they are used tes othenNise.
As used herein, AUCz4h is the area under the plasma concentrationtime curve from hour 0 to the last measurable plasma concentration (e.g., 24 , calculated by the linear trapezoidal rule.
As used herein, ALS or amyotrophic lateral sclerosis refers to the motor neuron disease commonly known as Lou Gehrig's Disease, and in some embodiments, ophic lateral sclerosis with early bulbar involvement or the bulbar form of the disease, and in some ments, "limb-onset" ALS.
As used herein, QD refers to once a day.
As used herein, BID refers to twice a day.
As used herein, Cmax refers to maximum plasma concentration As used herein, fm refers to fraction metabolized.
Provided is a method for treating ALS in a subject, comprising administering to the subject a therapeutically ive amount of riluzole and a therapeutically effective amount of 7357.
In some embodiments, administering the riluzole in combination with CK-2017357 extends survival and/or time to tracheostomy.
In some embodiments, the eutically ive amount of riluzole administered in combination with CK-2017357 is a smaller dose than the therapeutically effective amount of riluzole when administered singly (i.e., without CK-2017357 treatment). In some embodiments, both the therapeutically effective amount of riluzole and the therapeutically effective amount of CK-2017357 when administered in combination are smaller doses than the therapeutically effective amount of riluzole and the therapeutically effective amount of CK-2017357 when each is administered singly.
In some embodiments, the stration of riluzole is BID. In some embodiments, the administration of le is OD.
In some embodiments, the administration of CK-2017357 is BID. In some embodiments, the administration is OD.
In some embodiments, the administration of riluzole is OD and the administration of CK-2017357 is BID. In some embodiments, the administration of riluzole is BID and the administration of CK-2017357 is BID.
In some embodiments, the administration of riluzole is OD and the stration of CK-2017357 is QD. In some embodiments, the administration of riluzole is BID and the administration of CK-2017357 is OD.
In some embodiments, the CK-2017357 is administered in two or more doses at different times (e.g., once in the morning and once in the evening). In some embodiments, the CK-2017357 is administered in two or more equal doses.
In some embodiments, the CK-2017357 is administered in two or more different doses. In some embodiments, the dose of CK-2107357 is titrated over time to a different (e.g., higher) daily dose level.
In some embodiments, single daily dose of 50 mg riluzole is administered in combination with a eutically effective amount of CK- 2017357. In some embodiments, a single daily dose of 50 mg riluzole is administered in combination with a total daily dose of between about 125 mg and 2000 mg of CK-2017357. In some embodiments, a single daily dose of 50 mg riluzole is administered in combination with a total daily dose of 125, 250, 375, or 500 mg of CK-2017357.
In some embodiments, a twice daily dose of 25 mg riluzole is administered in combination with a therapeutically effective amount of CK- 2017357. In some embodiments, a twice daily dose of 25 mg riluzole is administered in combination a total daily dose of between about 125 mg and 2000 mg of CK-2017357. In some embodiments, a twice daily dose of 25 mg riluzole is administered in ation with a total daily dose of 125, 250, 375, or 500 mg of CK-2017357.
In some ments, a single daily dose of 25 mg le is administered in combination with a therapeutically effective amount of CK- 2017357. In some embodiments, a single daily dose of 25 mg riluzole is administered in ation with a total daily dose of between about 125 mg and 2000 mg of 7357. In some embodiments, a single daily dose of 25 mg riluzole is administered in combination with a total daily dose of 125, 250, 375, or 500 mg of CK-2017357.
In some embodiments, a twice daily dose of 12.5 mg riluzole is stered in combination with a therapeutically effective amount of CK- 2017357. In some embodiments, a twice daily dose of 12.5 mg riluzole is administered in combination with a total daily dose of between about 125 mg and 2000 mg of CK-2017357. In some embodiments, a twice daily dose of 12.5 mg riluzole is administered in combination with 125, 250, 375, or 500 mg of CK- Also disclosed is a method for increasing the half-life of riluzole in a t, comprising administering to the subject a therapeutically effective amount of CK-2017357.
Also disclosed is a method for sing the frequency of riluzole dosing in a subject (e.g., from twice-daily to once-daily) or ng the dose of riluzole stered to the subject (e.g., from 200 mg daily to 100 mg daily, or from 100 mg daily to 50 mg daily, or from 50 mg daily to 25 mg daily).
Also disclosed is a method for reducing the inter-subject variability of le exposure (e.g., Cmax and/or AUC24h) between two or more subjects, comprising administering to the subjects a therapeutically effective amount of CK- 2017357.
Also disclosed is a method for reducing the intra-subject ility of riluzole exposure (e.g., Cmax and/or AUC24h), comprising administering to the subject a therapeutically effective amount of CK-2017357.
Also disclosed is a method for reducing the incidence and/or severity of adverse events in a subject treated with riluzole, comprising administering to the subject a therapeutically effective amount of CK-2017357. In some embodiments, the adverse event is a CNS-related adverse event (see, e.g., o et al., Anesthesia & Analgesia (2007), 104:1415-21).
In some embodiments, CK-2017357 is administered to the subject concurrently with riluzole administration, i.e., CK-2017357 and le are stered simultaneously, essentially simultaneously or within the same treatment protocol. In some instances of concurrent administration, administration of CK-2017357 and riluzole begin and end at the same time (i.e., on the same day or within the same treatment protocol). In other instances of concurrent administration, only one of CK-2017357 and riluzole is stered for a first period of time, followed by co-administration of the 7357 and riluzole for a second period of time. For example, the subject may receive riluzole for a first period of time, then receive both CK-2017357 and riluzole for a second period of time. Administration of either CK-2017357 or riluzole may then continue for a third period of time. In another example, the subject may receive 7357 for a first period of time, then receive both CK-2017357 and riluzole for a second period of time. Administration of either CK-2017357 or riluzole may then ue for a third period of time. In other instances of concurrent administration, CK-2017357 and riluzole are co-administered for a first period of time, followed by stration of only one of CK-2017357 and riluzole for a second period of time. For example, the subject may receive both 7357 and riluzole for a first period of time, then receive CK-2017357 for a second period of time. In another example, the subject may receive both 7357 and riluzole for a first period of time, then receive riluzole for a second period of time. In all instances, alternate administration may be repeated during a single treatment protocol. The ination of the order of administration and the number of repetitions of administration of each therapy during a treatment ol is within the knowledge of the skilled physician after evaluation of the condition of the patient.
In some embodiments, riluzole and CK-2017357 are administered sequentially. In some instances of sequential administration, CK-2017357 is administered to the subject after riluzole administration has ended. The administration of CK-2017357 may begin immediately following ation of riluzole administration, or there may be a time interval (e.g., one day, one week, one month, six months, one year, etc.) between the end of riluzole administration and the beginning of CK-2017357 administration. In other instances of sequential administration, riluzole is administered to the subject after CK-2017357 administration has ended. The administration of riluzole may begin immediately following termination of CK-2017357 administration, or there may be a time al (e.g., one day, one week, one month, six months, one year, etc.) between the end of CK-2017357 administration and the ing of riluzole administration. In each instance, alternate administration may be repeated during a single ent protocol. The determination of the order of stration and the number of repetitions of administration of each therapy during a treatment protocol is within the knowledge of the d physician after evaluation of the condition of the patient.
In some embodiments, CK-2017357 and riluzole are administered in a single pharmaceutical composition. The single pharmaceutical composition may be administered via any of the accepted modes of administration for therapeutic agents including, but not d to, , sublingually, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarily, vaginally, rectally, or intraocularly. In some embodiments, the single ceutical composition is administered . In some embodiments, the single pharmaceutical composition is formulated to administer both CK-2017357 and riluzole at the same time or rate. In some embodiments, the single pharmaceutical composition is formulated to administer both CK-2017357 and riluzole at different times or rates. For example, the single pharmaceutical composition may deliver riluzole at a slower rate the CK- 2017357, or CK-2017357 at a slower rate than le. In another example, the single pharmaceutical composition may deliver CK-2017357 first followed by riluzole (i.e., delayed release of riluzole), or riluzole first followed by CK-2017357 (i.e., delayed release of CK-2017357).
In some embodiments, 7357 and riluzole are stered in separate pharmaceutical compositions. Each agent may, because of different physical and chemical characteristics, be stered by different . For example, one agent can be administered orally, while the other is administered intravenously. Alternatively, each agent may be administered by the same route.
For example, both CK-2017357 and riluzole may be administered orally (i.e., in the form of two separate pills or capsules). The determination of the mode of stration and the advisability of administration, in the same pharmaceutical composition (if le) is within the dge of the skilled clinician. The initial administration can be made according to established ols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician.
Also provided is a method for treating ALS by administering to a patient at least two doses daily of CK-2017357. In some embodiments, two doses of CK-2017357 are administered at different times (e.g., once in the morning and once in the evening). In some embodiments, the total daily dose is at least about 250 mg, or at least about 300 mg, or at least about 350 mg, or at least about 400 mg, or at least about 450 mg, or at least about 500 mg. In some embodiments, at least one of the doses is equal to or greater than about 125 mg, or equal to or r than about 150 mg, or equal to or greater than about 200 mg, or equal to or r than about 250 mg. In some embodiments, at least two of the doses are equal to or greater than about 125 mg, or equal to or greater than about 150 mg, or equal to or greater than about 200 mg, or equal to or greater than about 250 mg. In some embodiments, the CK-2017357 is administered in two or more equal doses (e.g., two equal doses of 125 mg or two equal doses of 250 mg). In some embodiments, the CK-2017357 is administered in two or more different doses (e.g., 125mg/250mg or 250mg/125mg). In some embodiments, the dose of CK-2107357 is titrated over time to a different (e.g., higher) daily dose level.
Administration of CK-2017357 can be via any of the accepted modes of administration for therapeutic agents including, but not limited to, orally, gually, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarily, lly, rectally, or intraocularly. In some embodiments, CK-2017357 is administered orally. In other embodiments, CK-2017357 is administered intravenously. In still other embodiments, CK-2017357 is administered into the lungs by inhalation or spraying of a dry powder, sion, solution or aerosol sing CK- 2017357.
Pharmaceutically acceptable compositions include solid, semi-solid, liquid and aerosol dosage forms, such as, e.g., tablets, capsules, powders, liquids, suspensions, itories, aerosols or the like. CK-2017357 can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills, transdermal (including electrotransport) patches, and the like, for ged and/or timed, pulsed administration at a predetermined rate. In certain ments, the compositions are provided in unit dosage forms suitable for single administration of a precise dose. 7357 can be administered either alone or in combination with a conventional pharmaceutical carrier, excipient or the like (e.g., mannitol, lactose, , magnesium stearate, sodium saccharine, talcum, cellulose, sodium crosscarmellose, glucose, gelatin, sucrose, magnesium carbonate, and the like).
If d, the pharmaceutical composition can also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like (e.g., sodium acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, anolamine acetate, anolamine oleate, and the like). Generally, depending on the intended mode of administration, the pharmaceutical composition will contain about 0.005% to 95% or, in certain ments, about 0.5% to 50% by weight of CK-2017357. Actual methods of preparing such dosage forms are known, or will be nt, to those skilled in this art; for example, see Remington’s Pharmaceutical Sciences, Mack Publishing Company, Easton, Pennsylvania.
In n embodiments, the itions will take the form of a pill or tablet and thus the composition may contain, along with the active ingredient, a diluent such as lactose, sucrose, ium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, ose, cellulose derivatives or the like. In certain embodiments of a solid dosage form, a powder, marume, solution or suspension (e.g., in propylene carbonate, vegetable oils or triglycerides) is encapsulated in a gelatin capsule.
Liquid pharmaceutically administrable itions can, for example, be prepared by dissolving, dispersing, etc. the active ient and optional pharmaceutical adjuvants in a carrier (e.g., water, saline, aqueous se, glycerol, glycols, ethanol or the like) to form a solution or suspension. lnjectables can be prepared in conventional forms, either as liquid solutions or suspensions, as emulsions, or in solid forms suitable for dissolution or suspension in liquid prior to ion. The percentage of CK-2017357 contained in such parenteral compositions is dependent on the specific nature of the compound, as well as the activity of CK-2017357 and the needs of the subject. In some embodiments, percentages of active ingredient of 0.01% to 10% in solution are employable, and may be higher if the composition is a solid which will be subsequently diluted. ceutical compositions of CK-2017357 may also be administered to the respiratory tract as an aerosol or solution for a zer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose. In some embodiments, the particles of the pharmaceutical composition have diameters of less than 50 microns, in certain embodiments, less than 10 microns.
Example 1: Effect of multiple daily doses of CK-2017357 on the PK of a single dose of riluzole in healthy subjects The primary ive of this study was to evaluate the effect of multiple daily doses of CK-2017357 on the pharmacokinetics (PK) of a single dose of riluzole in healthy subjects. On Day 1, 12 subjects (7 males and 5 females) received a single 50 mg oral dose of riluzole. On Day 6, all subjects began 11 days of oral dosing with 250 mg CK-2017357 QD (through Day 16). On Day 13, 11 of the 12 subjects received another single 50 mg oral dose of riluzole.
Following each dose of riluzole, plasma samples were collected at pre-dose, 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, 48, 72, and 96 hours post-dose. Plasma concentrations of riluzole were determined using a validated HPLC/MS/MS method with a quantitation range of 5.00 to 2,000 ng/mL. Samples were analyzed using a 50.0 uL aliquot volume and a n-precipitation extraction procedure ed by HPLC/MS/MS. Riluzole concentrations were calculated with a 1/x2 linear regression over a concentration range of 5.00 to 2,000 using riluzole-130,15N2 as an al standard. Separately, another set of plasma samples were collected at pre-dose of CK-2017357 on days 11, 12, and 13.
These plasma samples were ed using a ted LC/MS/MS method for trough CK-2017357 levels to determine the attainment of steady-state pharmacokinetics of CK-2017357.
Plasma concentration data of riluzole were analyzed by non- compartmental methods to ine pharmacokinetic parameters of riluzole.
Descriptive pharmacokinetic parameters such as Cmax, Tmax, AUC, t1/2, Cl/F, and V/F were ated using Phoenix WinNonlin 6.1 ight, in View, CA). All concentrations < LLOQ were set to zero for the purpose of calculating descriptive statistics and noncompartmental analysis. Summary PK parameters of riluzole are presented in Tables 1A and 18 below.
Table 1A: Summary cokinetic parameters of riluzole in healthy subjects Tmax AUClast AUCinf t1/2 CL/F hr hr*no/ml hr*no/ml hr ml/hr N 12 12 12 Mean 190 1.1 845 8.0 86258 so 113 0.5 416 440 2.5 75198 CV% 59.6 46.9 87.2 Table 13: Summary pharmacokinetic parameters of riluzole + CK2017357 (250 mg QD) in healthy subjects AU Clast AU Cinf t1/2 hr*no/ml hr*no/ml hr N 11 11 11 11 11 Min 191 0.5 1309 1412 9.6 Median 324 1.0 2443 2559 14.9 Max 482 2.0 6613 7019 23.2 CV% 27.0 46.0 55.6 54.7 32.2 The extent of drug-drug interaction was assessed by comparing AUCinf values from Period 1 (riluzole alone) to values from Period 2 (riluzole + CK- 2017357). Results of this study are ted in Table 2 below. Mean plasma concentration-time es are presented graphically in Figure 1, and Cmax and AUCinf data for Periods 1 and 2 are represented in Figure 2.
Table 2: Individual and Summary of AUCinf-fold increase of riluzole in the presence of steady-state CK-2015375 (250mg) in y ts AUCinf AUCinf Fold increase Period 1 Period 2 (hr*ng/ml) (hr*ng/ml) 1 1335.38 2 813.03 1622.66 3 619.77 1931.23 4 162.75 1951.67 883.12 2608.46 6 1189.32 9 7 1674.53 3577.29 8 1263.88 7018.50 9 630.27 3 573.21 2559.42 11 378.92 1412.02 12 612.18 2540.58 N 12 11 844.70 2929.00 440.32 1600.93 162.75 1412.02 721.65 2559.42 1674.53 7018.50 52.1 54.7 This study showed that -state CK-2017357 ) raised the mean Cmax of riluzole approximately 1.7-fold, and its mean AUCinf approximately 4.4-fold compared with riluzole alone. The mean tug of riluzole increased from 8.0 hours to 15.5 hours in the presence of steady-state CK-2017357. As illustrated in Figure 2, co—administration of CK-2017357 generally reduced the inter-subject variability of riluzole pharmacokinetics in these healthy subjects.
Example 2: Drug-drug interaction (DDI) between riluzole and CK-2017357 in ALS patients This was a placebo controlled, three-period crossover study. Each t received 50 mg riluzole BID and single doses of placebo, 250 mg CK- 2017357, and 500 mg CK-2017357 in random order, separated by 6-10 days.
Duplicate PK samples were collected from each t in the clinical study. One set was analyzed for CK-2017357, and the other analyzed for riluzole using a validated LC/MS/MS . Riluzole exposure (AUC24h) was calculated for each patient and each treatment of CK-2015357 (placebo, 250mg, and 500 mg). le AUC-fold increase over CK-2017357 placebo period was used to assess the extent of drug-drug interaction.
Plasma concentrations of riluzole were determined using a validated HPLC/MS/MS method with a quantitation range of 5.00 to 2,000 ng/mL. Samples were analyzed using a 50.0 uL aliquot volume and a protein-precipitation extraction procedure followed by HPLC/MS/MS. Riluzole concentrations were calculated with a 1/x2 linear regression over a concentration range of 5.00 to 2,000 using riluzole-13C,15N2 as an internal standard. ptive pharmacokinetic parameters such as Cmax, Tmax, AUC, t1/2, Cl/F, and VIP were calculated using Phoenix WinNonlin 6.1 (Pharsight, Mountain View, CA). All concentrations < LLOQ were set to zero for the purpose of calculating descriptive statistics and noncompartmental analysis. Since the time of dosing for riluzole was not ed, time elapsed was ated based on nominal time points of 7357, and used to calculate riluzole .
The results of this experiment are summarized in Table 3 below and mean plasma concentration-time profiles are presented in Figure 3.
Table 3: Individual and summary riluzole AUC24h and AUC24hr-fold se following oral le at 50mg b.i.d.
CK357 = CK357 = CK357 = Auc24hr-fold AUC24hr-fold 0mg 250mg 500mg Increase Increase (CK'357 = (CK357 = (hr*ng/ml) (hr*ng/ml) (hr*ng/ml) 1 2831.49 7 4062.97 1.57 1.43 2 1093.68 7 2346.72 2.25 2.15 3 4778.27 4 2492.82 3767.26 4082.12 1.51 1.64 307.49 6 829.77 9 4510.3 6.07 5.44 7 747.85 1828.87 2623.22 2.45 3.51 8 2540.48 1841.12 5652.57 0.72 2.23 9 945.95 1761.84 4024.18 1.86 4.25 1400.63 2509.68 7921.64 1.79 5.66 11 1263.74 4464.6 3.53 12 992.23 2194.64 3132.45 2.21 3.16 13 2172.04 4805.42 5410.5 2.21 2.49 14 2220.49 3939.67 5655.82 1.77 2.55 2647.84 1850.43 2055.75 0.70 0.78 16 1125.41 2947.04 3972.68 2.62 3.53 17 4 1564.05 5600.95 0.85 3.05 18 3047.02 7371.82 7084.32 2.42 2.33 19 1155.09 4366.21 3438.4 3.78 2.98 2699.12 3939.62 4753.02 1.46 1.76 21 683.16 4034.31 4295.43 5.91 6.29 22 1511.98 2 2986.23 3.18 1.98 23 2087.11 3828.63 2197.83 1.83 1.05 24 1384.24 2003.85 2644.38 2773.55 1.32 1.38 26 1477.45 2971.36 5 2.01 2.67 27 4116.82 4620.8 5 1.12 1.28 28 1460.67 2136.77 3992.31 1.46 2.73 29 2344.66 2437.78 7 1.04 1.66 2083.63 1598.71 5707.86 0.77 2.74 31 996.99 1250.21 830.83 1.25 0.83 32 2569.22 3265.26 4360.36 1.27 1.70 33 4171.45 3685.49 34 4841.83 5862.48 9116.2 1.21 1.88 1163.34 4568.86 3138.61 3.93 2.70 36 838.09 2295.37 7 2.74 4.30 37 2812.13 4350.55 4292.15 1.55 1.53 38 1438.58 1 4386.6 2.59 3.05 39 3547.69 40 1158.27 1575.61 1817.17 1.36 1.57 41 1387.18 887.4 3869.44 0.64 2.79 42 536.67 1448.56 849.78 2.70 1.58 43 1379.36 N 39 38 40 Mean 1775.54 3252.18 4025.65 SD 969.09 1462.15 1721.25 Min 307.49 887.4 830.83 Median 7 3118.31 3982.49 Max 4841.83 7371.82 9116.2 CV% 54.6 45 42.8 Analysis of riluzole AUC24h following 50 mg BID showed a general trend of increasing riluzole trations in CK-2017357 active periods (250 and 500 mg vs. 0 mg). Compared to the CK-2017357 placebo period, mean riluzole AUC24h increased approximately 2.1- and 2.6-fold for the 250- and 500-mg CK- 7 dose, respectively. As illustrated in Figure 3, the mean riluzole Cmax roughly doubled in the presence of steady-state concentrations of CK-2017357. .
Example 3: Pharmacokinetics and Interactive Effects of the Fast Skeletal Muscle Activator CK-2017357 and Riluzole The ive of this study was to determine the cokinetics of repeated doses of CK-2017357 both in the presence and absence of riluzole, and to determine the effects of CK-2017357 at g doses on plasma le serum levels.
In this study, 49 patients with ALS were treated; 24 patients were not taking riluzole and the remainder took a stable but reduced dose of le (50 mg daily). Patients (n = off/on riluzole) ed single daily doses of placebo (n = 6/7), 125, 250, or 375 mg of CK-357 (n = 6/6 for all 3 CK-2017357 groups) for 14 days. CK-2017357 and riluzole levels were ed on Days 1, 2, 8 and 15.
Plasma levels of CK-2017357 achieved steady state by Day 8; levels four hours after dosing on Day 8 were approximately 70% higher than four hours after the first dose on Day 1. CK-2017357 Cmax increased proportionally by dose with no apparent effect of riluzole, as shown in Table 4.
Table 4: CK-2017357 Cmax levels with and without riluzole —CK-2017357 Cmax mc/mL CK-2017357 Daily Dose No Riluzole On Riluzole ———m_ CK-2017357 approximately doubled riluzole levels across all dose groups as shown in Table 5 below Adverse event frequencies were not altered by the presence of riluzole at any dose of CK-2017357.
Table 5: Riluzole Cmax levels with various doses of CK-2017357 Dose Group Cmax (ng/mL) AUCIast (hr*pg/mL) Median 1 SD Median 1 SD 125 mg once daily 243 i 89 42.6 1r 14.8 250 mg once daily 143 1r 125 30.0 1r 21.7 375 mg once daily 228 1r 169 46.3 1r 18.2 CK-2017357 had predictable linear kinetics at the repeated doses used in the current study, reaching steady state within 1 week. CK- 2017357 plasma levels were not affected by the presence of le. Riluzole levels were increased by CK-2017357 with plasma levels increasing approximately 2-fold across all dose levels of CK-2017357. r, no adverse events were reported during this study attributable to higher riluzole levels with the daily riluzole dose reduced to 50 mg daily. These results suggest that 7357 and riluzole may be given safely in combination.
Example 4: Investigation of Twice-daily Dose Titration Regimen of CK-2017357 in Patients with ALS In previous clinical ments, 1-(ethy|propyl)ethynylimidazo[4,5- b]pyrazinol (CK-2017357) has been administered once a day in the morning.
While improvements in patient and investigator global assessments, muscle fatigability and pulmonary on have been observed in these studies, dizziness has been a dose-limiting adverse event in both healthy volunteers and in patients with ALS. This e examines whether the maximum tolerable total daily dose of 7357 can be increased by dividing the daily dose into two portions (morning and evening), and beginning treatment with a low dose and titrating upward to a target of 250 mg twice daily. ts enrolled in this clinical study were randomized, double-blind, and placebo-controlled. There was a 7-day stabilization period for le at a reduced dose of 50 mg OD. The patients were then randomized 3:1 to CK- 2017357 or placebo for 14 days.
The CK-2017357 titration regimen was as follows: dosing ted at 125 mg twice daily for 7 days (250 mg total daily dose); on Day 8, up titration to 125 mg in the morning and 250 mg in the evening (375 mg total daily dose); on Day 15, up titration to 250 mg twice daily (500 mg total daily dose) continued through the morning dose on Day 22. Patients who did not tolerate a dose escalation returned to the previous ted dose level and remained at that dose level to complete the study. Placebo patients underwent a similar dummy dose titration to maintain the blind. Figure 4 illustrates the study design.
Twenty-seven patients were treated in this study. All six patients randomized to placebo completed three weeks of dosing. Of the 21 patients randomized to treatment with CK-2017357, 14 were escalated to the t total daily dose of 500 mg and completed three weeks of . The most commonly reported treatment-emergent adverse event was dizziness, which was mild in 10 of the 12 patients in whom it occurred and only te in the other two. ess was self-limited in 6 of 12 ts in whom it occurred. Encouraging trends toward increases in the ALSFRS-R score and MVV were observed on CK- 2017357 relative to placebo. Thus, this study suggests that CK-2017357 administered in a twice-daily, dose titration regimen is safe and well tolerated.
While the present invention has been described with reference to the specific embodiments described herein, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, modifications may be made to adapt a particular situation, material, composition of matter and/or process to the objective, spirit and scope of the present invention.
All such modifications are intended to be within the scope of the claims appended The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information d from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.
Throughout this ication and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the ion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.

Claims (6)

The claims ng the invention are as follows:
1. Use of a therapeutically effective amount of riluzole and a eutically effective amount of 6-Ethynyl(pentanyl)-1H-imidazo[4,5-b]pyrazine-2(3H)- one, in the manufacture of a medicament for treating amyotrophic l sclerosis in a subject.
2. Use according to claim 1, wherein the therapeutically effective amount of riluzole to administered in combination with 6-Ethynyl(pentanyl)-1H- imidazo[4,5-b]pyrazine-2(3H)-one is a smaller dose than the therapeutically effective amount of riluzole when intended to be administered singly.
3. Use of a therapeutically effective amount of 6-Ethynyl(pentanyl)-1H- imidazo[4,5-b]pyrazine-2(3H)-one, in the manufacture of a medicament for reducing the variability of riluzole exposure in a subject with amyotrophic lateral sclerosis.
4. Use of a therapeutically effective amount of 6-Ethynyl(pentanyl)-1H- imidazo[4,5-b]pyrazine-2(3H)-one, in the manufacture of a medicament for reducing the total daily dose of riluzole in a subject with amyotrophic lateral sclerosis.
5. Use of a therapeutically effective amount of 6-Ethynyl(pentanyl)-1H- imidazo[4,5-b]pyrazine-2(3H)-one, in the manufacture of a medicament for sing the half-life of le in a subject with ophic lateral sclerosis.
6. Use of a therapeutically effective amount of 6-Ethynyl(pentanyl)-1H- imidazo[4,5-b]pyrazine-2(3H)-one, in the manufacture of a medicament for decreasing the frequency of riluzole dosing in a subject with amyotrophic l sclerosis. H:\ACG\Interwoven\NRPortbl\DCC\ACG\9411033_1.docx-
NZ619924A 2011-07-13 2012-07-12 Combination als therapy NZ619924B2 (en)

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US201161507381P 2011-07-13 2011-07-13
US61/507,381 2011-07-13
US201161544533P 2011-10-07 2011-10-07
US61/544,533 2011-10-07
US201261637759P 2012-04-24 2012-04-24
US201261637770P 2012-04-24 2012-04-24
US61/637,770 2012-04-24
US61/637,759 2012-04-24
US201261646699P 2012-05-14 2012-05-14
US61/646,699 2012-05-14
PCT/US2012/046523 WO2013010015A2 (en) 2011-07-13 2012-07-12 Combination als therapy

Publications (2)

Publication Number Publication Date
NZ619924A NZ619924A (en) 2016-02-26
NZ619924B2 true NZ619924B2 (en) 2016-05-27

Family

ID=

Similar Documents

Publication Publication Date Title
AU2012281042B2 (en) Combination ALS therapy
US10912744B2 (en) Therapeutic formulation for reduced drug side effects
JP4938905B2 (en) Administration method of selective S1P1 receptor agonist
EA020193B1 (en) Combinations comprising methotrexate and dhodh inhibitors
EP2722045B1 (en) Compositions for treating centrally mediated nausea and vomiting
JP2023171776A (en) Synthetic transdermal cannabidiol for treatment of focal epilepsy in adults
US20170105983A1 (en) Compositions for reduction of side effects
US9066949B2 (en) Compositions and methods for the treatment of catatonia
EA009935B1 (en) New synergistic combination comprising roflumilast and formoterol
NZ619924B2 (en) Combination als therapy
JP6420923B1 (en) Medicine
US10258617B2 (en) Dosing regimens for fast onset of antidepressant effect
JP6860984B2 (en) Administration regimen for ataxia in spinocerebellar degeneration
MX2014000455A (en) Combination als therapy.
US20230172895A1 (en) Methods of treating or preventing organophosphorus poisoning
AU2013205648B2 (en) Combination treatment
EP2482658B1 (en) Methods and compositions for treatment of raynaud&#39;s phenomenon
WO2016208045A1 (en) Dosing regimen of therapeutic agent for ataxia associated with spinocerebellar degeneration
Cole American Academy of Neurology 64th Annual Meeting, New Orleans, Louisiana, USA-April 21-28, 2012