HK1226664A1 - Combination formulation of two antiviral compounds - Google Patents
Combination formulation of two antiviral compoundsInfo
- Publication number
- HK1226664A1 HK1226664A1 HK17100411.2A HK17100411A HK1226664A1 HK 1226664 A1 HK1226664 A1 HK 1226664A1 HK 17100411 A HK17100411 A HK 17100411A HK 1226664 A1 HK1226664 A1 HK 1226664A1
- Authority
- HK
- Hong Kong
- Prior art keywords
- ledipasvir
- sofosbuvir
- weeks
- solid dispersion
- pharmaceutical composition
- Prior art date
Links
Abstract
Disclosed are pharmaceutical compositions having an effective amount of substantially amorphous ledipasvir and an effective amount of substantially crystalline sofosbuvir.
Description
The application is a divisional application of Chinese patent application with application date of 2014, 1 month and 30 days, application number of 201480000286.1 and invention name of 'a combined preparation of two antiviral compounds'.
Cross Reference to Related Applications
The present application claims the benefit of U.S. provisional application No. 61/759,320 filed on day 31/1/2013, U.S. provisional application No. 61/772,292 filed on day 4/3/2013, U.S. provisional application No. 61/828,899 filed on day 30/5/2013, U.S. provisional application No. 61/870,729 filed on day 27/8/2013, U.S. provisional application No. 61/897,793 filed on day 30/10/2013, and U.S. provisional application No. 61/907,332 filed on day 21/11/2013 under 35U.S. c. § 119 (e). The entire contents of these provisional applications are incorporated herein by reference.
Background
Hepatitis c is considered a chronic viral disease of the liver, characterized by liver disease. Although liver-targeted drugs have been widely used and are also effective, toxicity and other side effects limit the use of these drugs. Hepatitis C Virus (HCV) inhibitors can be used to limit the establishment and progression of HCV infection, as well as diagnostic assays for HCV.
Ledipasvir is a selective inhibitor of nonstructural protein 5A (NS5A) and has been described previously (see, e.g., WO 2010/132601). The chemical name of Ledipasvir is (1- {3- [6- (9, 9-difluoro-70 {2- [5- (2-methoxycarbonylamino-3-methyl-butyryl) -5-aza-spiro [2.4] hept-6-yl ] -3H-imidazol-4-yl } -9H-fluoren-2-yl) -1H-benzimidazol-2-yl ] -2-aza-bicyclo [2.2.1] heptane-2-carbonyl } -2-methyl-propyl) -carbamic acid methyl ester.
Sofosbuvir (sof) is a selective inhibitor of nonstructural protein 5B (NS5B) (see, e.g., WO2010/132601 and U.S. patent application 7,964,580). The chemical name of Sofosbuvir is (S) -isopropyl 2- (((S) - ((((2R, 3R,4R,5R) -5- (2, 4-dioxo-3, 4-dihydropyrimidin-1 (2H) -yl) -4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl) methoxy) (phenoxy) phosphoryl) amino) propionate.
Disclosure of Invention
In some embodiments, the invention provides a pharmaceutical composition comprising ledipasvir and sofosbuvir, wherein ledipasvir is in substantially amorphous form and sofosbuvir is in substantially crystalline form.
The chemical name of Ledipasvir is (1- {3- [6- (9, 9-difluoro-70 {2- [5- (2-methoxycarbonylamino-3-methyl-butyryl) -5-aza-spiro [2.4] hept-6-yl ] -3H-imidazol-4-yl } -9H-fluoren-2-yl) -1H-benzimidazol-2-yl ] -2-aza-bicyclo [2.2.1] heptane-2-carbonyl } -2-methyl-propyl) -carbamic acid methyl ester, the formula is as follows:
sofosbuvir (sof) has the chemical name (S) -2- (((S) - (((((2R, 3R,4R,5R) -5- (2, 4-dioxo-3, 4-dihydropyrimidin-1 (2H) -yl) -4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl) methoxy) (phenoxy) phosphoryl) amino) isopropyl propionate, and has the formula:
in some embodiments, there is provided a pharmaceutical composition comprising: a) an effective amount of ledipasvir, wherein ledipasvir is substantially amorphous; and b) an effective amount of sofosbuvir, wherein the sofosbuvir is substantially crystalline.
Furthermore, embodiments of the present invention relate to pharmaceutical dosage forms and tablets. The invention also provides a method for treating hepatitis C by using the composition.
Brief Description of Drawings
Figure 1 is an XRPD pattern of ledipasvir solid dispersion containing copovidone with a drug to polymer ratio of 1: 1. The solid dispersion was in an amorphous state as shown by XRPD.
Figure 2 is a modified Differential Scanning Calorimetry (DSC) curve for ledipasvir solid dispersion comprising copovidone with a drug to polymer ratio of 1: 1. The glass transition temperature of the solid dispersion is about 140 ℃.
Figure 3 shows the solid state characterization by solid state nuclear magnetic resonance (SS-NMR) of ledipasvir solid dispersion containing copovidone with a drug to polymer ratio of 1: 1.
Figure 4 is a fourier transform raman spectrum of a solid dispersion of ledipasvir containing copovidone with a drug to polymer ratio of 1: 1.
FIG. 5 shows the dissolution rate of sofosbuvir (400mg)/ledipasvir (90mg) in the composition described in example 7.
FIG. 6 shows the dissolution rate of ledipasvir (400mg)/ledipasvir (90mg) in the sofosbuvir (400mg) composition described in example 3.
Figures 7A-D show HCVRNA levels during 12 weeks treatment and during 24 weeks post treatment in patients who were naive (rheomentaive) (figure 7A) or null responder (figure 7B) with sofosbuvir (sof) and Ribavirin (RBV) and naive (figure 7C) or null responder (figure 7D) with sofosbuvir (sof), ledipasvir and Ribavirin (RBV). The data and test methods are further described in example 5.
Figures 8A-B list graphs to show that all three formulations have comparable dissolution profiles, similar to that of the single drug control. Example 7 is more fully described.
FIG. 9 shows the pH-solubility curve of ledipasvir at Room Temperature (RT). The curve is obtained using equation ST=S0[(1+10(pKa1-pH)+10(pKa1+pKa2-2·pH))]And an inherent solubility (S) of 0.04. mu.g/mL0) And weakly basic pKa1 and pKa2 values non-linear least squares regression fits of 5.0 to 4.0. Example 8 is more fully described.
Figure 10 shows the study design for treatment of naive (no cirrhosis) and non-responsive (50% cirrhosis) patients for 8 and 12 weeks with a fixed dose composition of sofosbuvir (sof) and ledipasvir in the presence or absence of Ribavirin (RBV). The data and protocol are described in example 9.
Figure 11 shows the results of treatment of naive (non-cirrhosis) and non-responder (50% cirrhosis) patients at 8 and 12 weeks with a fixed dose composition of sofosbuvir (sof) and ledipasvir in the presence or absence of Ribavirin (RBV). The data and protocol are further described in example 9.
Detailed Description
1. Definition of
As used in this specification, the following words and phrases generally have the meanings as set forth below, unless the context of their use indicates otherwise.
The term "about" as used herein in the context of quantitative detection means the indicated amount ± 10%, alternatively ± 5%, or ± 1%. For example, "about 2: 8" means 1.8-2.2:7.2-8.8 according to the. + -. 10% range.
The term "amorphous" refers to a state in which a substance lacks long-range order at the molecular level and can exhibit physical properties of a solid or liquid depending on temperature. Generally, such materials do not give a unique X-ray diffraction pattern and, even if they exhibit solid properties, are more formally described as liquids. Upon heating, a change of solid to liquid properties occurs, which is mainly characterized by a change of state, usually a second order change (glass transition).
The term "crystal" refers to a solid phase of a substance having a regular ordered internal structure at the molecular level, with a unique X-ray diffraction pattern and specific peaks. The substance will also exhibit the properties of a liquid when heated sufficiently, but the solid to liquid change is characterized by a phase change, usually a first order change (melting point).
The term "substantially amorphous" as used herein refers to compounds in the composition in excess of 70%; or more than 75%; or more than 80%; or more than 85%; or more than 90%; or more than 95%; or more than 99% in amorphous form. "substantially amorphous" may also refer to a material having no more than about 20% crystallinity, or no more than about 10% crystallinity, or no more than about 5% crystallinity, or no more than about 2% crystallinity.
The term "substantially crystalline" as used herein means that the compound in the composition is more than 70%; or more than 75%; or more than 80%; or more than 85%; or more than 90%; or more than 95%; or more than 99% is in crystalline form. "substantially crystalline" may also refer to a material having no more than about 20%, or no more than about 10%, or no more than about 5%, or no more than about 2% amorphous form.
The term "polymer" refers to a chemical compound or mixture of compounds comprised of repeating structural units formed by a polymerization process. Suitable polymers useful in the present invention are fully described.
The term "polymer matrix" as used herein refers to a composition comprising one or more polymers in which the active agent is dispersed or contained within the matrix.
The term "solid dispersion" refers to a solid dispersion of one or more active agents in a polymer matrix made by a variety of methods, including spray drying, melting (fusing), solvent or melt-solvent methods.
The term "amorphous solid dispersion" as used herein refers to a stable solid dispersion containing an amorphous active drug and a polymer. By "amorphous active drug" it is meant that the amorphous solid dispersion contains the active drug in a substantially amorphous solid state form. In certain aspects, as shown by XRPD in fig. 1, the solid dispersion is in an amorphous state, the solid dispersion having a glass transition temperature of about 140 ℃ (see fig. 2).
The term "pharmaceutically acceptable" means that the substance does not have properties that would allow a reasonably prudent physician to avoid administration of the substance to a patient in view of the disease or condition to be treated and the respective route of administration. For example, it is often desirable that the material be substantially sterile, such as when used in an injection.
The term "pharmaceutically acceptable polymer" means a polymer that does not possess properties that would allow a reasonably prudent physician to avoid administration of the substance to a patient in view of the disease or condition to be treated and the respective route of administration.
The term "carrier" refers to a glidant, diluent, adjuvant, excipient, or the like, administered with a compound without limitation. Examples of carriers are described herein and in the "complete raminten pharmacy" of e.w. martin.
The term "diluent" refers to a chemical compound used to dilute a compound of interest prior to shipment. Diluents may also be used to stabilize the compound. Non-limiting examples of diluents include starch, sugars, disaccharides, sucrose, lactose, polysaccharides, cellulose ethers, hydroxypropyl cellulose, sugar alcohols, xylitol, sorbitol, maltitol, microcrystalline cellulose, calcium or sodium carbonate, lactose monohydrate, calcium hydrogen phosphate, cellulose, compressible sugars, dibasic calcium phosphate dehydrate, mannitol, microcrystalline cellulose, and tricalcium phosphate.
The term "binder" as used herein refers to any pharmaceutically acceptable film that can be used to bind the active and inert components of a carrier together to maintain the bound and separated portions together. Non-limiting examples of binders include hydroxypropyl cellulose, hydroxypropyl methylcellulose, povidone, copovidone, and ethylcellulose.
The term "disintegrant" refers to a substance that, when added to a solid formulation, promotes its decomposition or disintegration after administration and allows the active ingredient to be released as efficiently as possible for rapid dissolution. Non-limiting examples of disintegrants include corn starch, sodium carboxymethyl starch, croscarmellose sodium, crospovidone, microcrystalline cellulose, modified corn starch, sodium carboxymethyl starch, povidone, pregelatinized starch, and alginic acid.
The term "lubricant" refers to an excipient added to the powdered mixture to prevent the compacted powder material from sticking to equipment during tableting or packaging. It helps the tablet to be ejected from the die and improves the flowability of the powder. Non-limiting examples of lubricants include magnesium stearate, stearic acid, silicon dioxide, fats, calcium stearate, polyethylene glycol, sodium stearyl fumarate, or talc; and solubilizing agents such as fatty acids including lauric acid, oleic acid and C8/C10A fatty acid.
The term "film coating" refers to a thin, uniform film on the surface of a substrate (e.g., a tablet). Film coatings are particularly useful for protecting active ingredients from light degradation. Non-limiting examples of film coatings include polyvinyl alcohol based, hydroxyethyl cellulose, hydroxypropyl methyl cellulose, sodium carboxymethyl cellulose, polyethylene glycol 4000, and cellulose acetate phthalate film coatings.
The term "glidant" as used herein refers to substances used in tablets and capsules to improve flowability during compression and to produce an anti-caking effect. Non-limiting examples of glidants include colloidal silicon dioxide, talc, fumed silica, starch derivatives, and bentonite.
The term "effective amount" refers to an amount sufficient to be therapeutically effective when administered to a mammal in need of treatment as defined below. The therapeutically effective amount will vary depending on the patient to be treated, the weight and age of the patient, the severity of the disease, the mode of administration, and the like, and can be readily determined by one of ordinary skill in the art.
The terms "treating" or "treating" in reference to a disease or disorder include avoiding the occurrence of the disease or disorder, inhibiting the disease or disorder, eliminating the disease or disorder, and/or alleviating one or more symptoms of the disease or disorder.
The term "sustained viral response" means that no viral RNA (or RNA below the limit of detection) is detectable in the patient sample (i.e., blood sample) for a specified period of time after treatment is discontinued. For example, a 4-week SVR indicates that no RNA was detected or that RNA was below the limit of detection in the patient at 4 weeks after cessation of HCV treatment.
The term "% w/w" as used herein refers to the weight of a component based on the total weight of the composition in which the component is contained. For example, if component A is present in an amount of 50% w/w of 100mg of the composition, the amount of component A is 50 mg.
2. Pharmaceutical compositions
The pharmaceutical composition comprises a combination of an effective amount of ledipasvir and an effective amount of sofosbuvir, wherein the ledipasvir is substantially amorphous and the sofosbuvir is substantially crystalline.
The experimental examples demonstrate that such a combined composition exhibits unexpected properties. Both Sofosbuvir and ledipasvir have previously been shown to be effective anti-HCV drugs. Ledipasvir showed a negative food effect when administered alone in the form of a traditional formulation, as evidenced by an approximately 2-fold reduction in drug exposure when compared to administration on a high fat diet to a fasted state (see, e.g., tables 10 and 11, example 3). This negative food effect did not occur when ledipasvir was administered as a solid dispersion and in combination with sofosbuvir (table 12, example 3).
In the combination composition, ledipasvir is substantially in the amorphous form. Amorphous drugs are expected to be unstable and have non-linear solubility and exposure profiles compared to crystalline drugs. The data presented herein show that ledipasvir in the combination composition is stable under a variety of conditions, both short-term and long-term, and maintains a consistently high solubility and exposure profile (example 6).
Furthermore, according to conventional wisdom, it is not suggested to co-prepare an amorphous drug and a crystalline drug, because the crystals can act as seeds to induce crystallization of the amorphous drug, resulting in instability of the amorphous drug. However, the present data show that ledipasvir remains stable and does not form crystals in the composition, whether co-granulated or co-blended with the same layer or with the sofosbuvir integrated as a separate layer (example 6).
It was also found that no drug-drug interaction occurred at the time of tablet formation of the combination composition, i.e. when sofosbuvir and ledipasvir were co-granulated or blended (example 7).
A.Ledipasvir
Ledipasvir has been described previously (see, e.g., WO2010/132601) and can be prepared by the methods described therein. In one embodiment, the pharmaceutical composition comprises ledipasvir formulated as a solid dispersion dispersed in a polymer matrix, wherein the polymer matrix is formed from a pharmaceutically acceptable polymer. The starting material for the solid dispersion may be ledipasvir in a variety of forms, including crystalline forms, non-crystalline forms, salts, solvates and free radicals. For example, acetone solvate, D-tartrate, anhydrous crystalline free radical, amorphous free radical, solvate or desolvated form of ledipasvir may be used. Solvates of Ledipasvir include, for example, those described in U.S. patent 2013/0324740 (incorporated herein by reference), such as, for example, monoacetone solvate, diacetone solvate, ethyl acetone solvate, isopropyl acetate solvate, methyl acetate solvate, ethyl formate solvate, acetonitrile solvate, tetrahydrofuran solvate, methyl ethyl ketone solvate, and methyl tert-butyl ether solvate. Particular starting materials which are expected to be useful are the monoacetone solvate, the diacetone solvate, the anhydrous crystalline radical, the D-tartrate salt, the anhydrous crystalline radical and the amorphous radical. These forms are characterized and described in U.S. publication No. 2013/0324496.
After dispersion in the polymer, the solid dispersion is in an amorphous form. Figures 1-4 depict the characteristics of amorphous solid dispersions containing ledipasvir. As shown by XRPD in fig. 1, the solid dispersion is in an amorphous state, and the glass transition temperature of the solid dispersion is about 140 ℃.
Solid dispersions can be prepared by a variety of techniques well known in the art, including but not limited to melt extrusion, spray drying, lyophilization, and evaporation of solutions.
Melt extrusion is the process of implanting a compound into a thermoplastic carrier. The mixture is treated at elevated temperature and pressure to disperse the compound at the molecular level into the matrix to form a solid solution. The extruded material can be further processed into a variety of dosage forms including capsules, tablets and transmucosal systems.
For the solution evaporation method, a solid dispersion can be prepared by dissolving the compound in a suitable liquid solvent, then combining the solution directly into a melt of the polymer, and then evaporating until a clear, solvent-free film remains. The film was further dried to constant weight.
For lyophilization techniques, the compound and carrier can be co-dissolved in a common solvent, frozen, and sublimed to obtain a lyophilized molecular dispersion.
For spraying of a dry solid dispersion, the solid dispersion may be prepared by a) mixing the compound and the polymer in a solvent to obtain a feed solution; and b) spray drying the feed solution to obtain a solid dispersion.
The spray dried solid dispersion of ledipasvir may result in improved in vivo and in vitro performance and generating/extending ability compared to other formulation methods, such as wet and dry granulation formulations. Ledipasvir may also be provided as the free base, D-tartrate, crystalline acetone solvate, or other solvate as described herein.
The choice of polymer for the solid dispersion depends on the stability and physical properties of ledipasvir in solution. Both hypromellose and copovidone solid dispersions showed sufficient stability and physical properties. Thus, in one embodiment, the polymer used for the solid dispersion is selected from hypromellose and copovidone. Furthermore, the increase in bioavailability of the copolyvidone-based dispersion when prepared at the 2:1API: polymer ratio was higher than the equivalent hypromellose-based formulation (30% and 22% F, respectively). The bioavailability of the copovidone-based formulation further increased as the polymer component increased to a ratio of 1:1, reaching 35% in famotidine-pretreated dogs.
In one embodiment, the polymer used for the ledipasvir solid dispersion is hydrophilic. Non-limiting examples of hydrophilic polymers include polysaccharides, polypeptides, cellulose derivatives such as methylcellulose, sodium carboxymethylcellulose, hydroxyethylcellulose, ethylcellulose, hydroxypropyl methylcellulose acetate succinate, hydroxypropyl methylcellulose phthalate, cellulose acetate phthalate, hypromellose, povidone, copovidone, hypromellose, cellulose nitrate polyethylene oxide, polyvinyl alcohol, and methacrylic acid copolymers.
In another embodiment, the polymer is non-ionic. Nonionic polymers have shown advantages in solubility screening assays. Non-limiting examples of nonionic polymers include hypromellose, copovidone, povidone, methylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, ethylcellulose, nitrocellulose, polyethylene oxide, polyvinyl alcohol, polyethylene glycol, and polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol.
In another embodiment, the polymer is ionic. Examples of ionic polymers include hydroxypropyl methylcellulose acetate succinate, hydroxypropyl methylcellulose phthalate, cellulose acetate phthalate, and copolymers of methacrylic acid.
In another embodiment, the polymer is selected from the group consisting of hypromellose, copovidone, and povidone. Both hypromellose and copovidone solid dispersions showed sufficient stability and physical properties. When spray dried at a 2:1ledipasvir polymer ratio, the increase in bioavailability of the copolyvidone-based dispersion was higher than that of the equivalent hypromellose-based formulation (30% and 22% F, respectively). Thus, in a particular embodiment, the polymer is copovidone.
In certain embodiments, the weight ratio of ledipasvir to polymer ranges from about 5:1 to about 1: 5. In other embodiments, the weight ratio of ledipasvir to polymer ranges from about 5:1 to about 1:4, or from about 5:1 to about 1:3, or from about 5:1 to about 1:2, or from about 2:1 to about 1: 1. In a particular embodiment, the weight ratio of ledipasvir to polymer is about 1: 1. In another embodiment, the weight ratio of ledipasvir to polymer is about 2: 1. In other embodiments, the weight ratio of ledipasvir to polymer is about 5:1, 1:4, 1:3, or 1: 2. In some instances, increasing the composition of the polymer to a 1:1 ratio may result in increased bioavailability. For example, a 1:1 ratio of ledipasvir to copovidone resulted in increased bioavailability in famotidine-pretreated dogs (F ═ 35%).
A therapeutically effective amount of a solid dispersion comprising ledipasvir may be present in the pharmaceutical composition. In some embodiments, the pharmaceutical composition comprises from about 1% to about 50% w/w solid dispersion of ledipasvir. In other embodiments, the composition comprises from about 5% to about 40% w/w, or from about 5% to about 35% w/w, or from about 5% to about 30% w/w, or from about 10% to about 25% w/w, or from about 15% to about 20% w/w of the ledipasvir of the solid dispersion. In other embodiments, the pharmaceutical composition comprises ledipasvir as a solid dispersion at about 1% w/w, about 5% w/w, about 10% w/w, about 20% w/w, about 25% w/w, about 30% w/w, about 35% w/w, about 40% w/w. In a particular embodiment, the pharmaceutical composition comprises ledipasvir as an approximately 18% w/w solid dispersion.
A therapeutically effective amount of ledipasvir may be present in the pharmaceutical composition. In some embodiments, the pharmaceutical composition comprises from about 1% to about 50% w/w ledipasvir. In other embodiments, the composition comprises from about 1% to about 40% w/w, or from about 1% to about 30% w/w, or from about 1% to about 20% w/w, or from about 5% to about 15% w/w, or from about 7% to about 12% w/w ledipasvir. In other embodiments, the pharmaceutical composition comprises about 1% w/w, about 3% w/w, about 5% w/w, about 7% w/w, about 11% w/w, about 13% w/w, about 15% w/w, about 17% w/w, about 20% w/w, about 23% w/w, about 25% w/w, or about 28% w/w, about 30% w/w ledipasvir. In a particular embodiment, the pharmaceutical composition comprises about 9% w/w ledipasvir.
As described above, after mixing ledipasvir with the polymer, the mixture can be dissolved in a solvent. One skilled in the art can select an appropriate solvent depending on the properties of the drug and/or polymer, such as solubility, glass transition temperature, viscosity, and molecular weight. Acceptable solvents include, but are not limited to, water, acetone, methyl acetate, ethyl acetate, chlorinated solvents, ethanol, methylene chloride, and methanol. In one embodiment, the solvent is selected from the group consisting of ethanol, dichloromethane, and methanol. In another embodiment, the solvent is ethanol or methanol. In a particular embodiment, the solvent is ethanol.
The mixture of compound and polymer is dissolved in a solvent, and then the mixture may be spray-dried. Spray drying is a well known process in which a liquid feedstock is dispersed into droplets into a drying chamber, accompanied by a heated process gas stream to assist in removing the solvent and producing a powder product. Suitable spray drying parameters are well known in the art and one skilled in the art knows how to select suitable spray drying parameters. The target feed concentration is typically about 10% to about 50%, the target is about 20%, and the viscosity is about 15cP to about 300 cP. The inlet temperature of the spray-drying apparatus is typically about 50-190 c and the outlet temperature is about 30-90 c. Two liquid nozzles and a hydraulic nozzle can be used for spray drying ledipasvir. The gas flow rate of the two liquid jets may be about 1-10kg/hr, the flow rate of the hydraulic jets may be about 15-300kg/hr, and the gas flow rate of the cell may be about 25-2500 kg/hr. The spray-dried material generally has a particle size (D)90) Below 80 μm. In some instances, a milling step may be used if further reduction in particle size is desired. Further description of spray drying and other techniques for forming amorphous dispersions is provided in U.S. patent No. 6,763,607 and U.S. patent publication No. 2006-0189633, the entire contents of each of which are incorporated herein by reference.
High yields (88, 90, 92, 95, 97, 98, 99%) were obtained from alcohol spray drying over a wide range of spray drying exit temperatures (30-90 ℃) with no accumulation of starting material in the spray drying chamber, whereas yields obtained from DCM spray drying were 60%, 78% and 44%. Furthermore, ledipasvir has good chemical stability in ethanol feed solution.
B.Sofosbuvir
Sofosbuvir has been previously described in U.S. patent 7,964,580 and U.S. patent publication nos.: 2010/0016251, 2010/0298257, 2011/0251152 and 2012/0107278. The sofosbuvir provided in the pharmaceutical compositions described herein is substantially crystalline. Examples of the preparation of crystalline form sofosbuvir are found in U.S. patent publication nos.: 2010/0298257, and 2100/0251152, both of which are incorporated herein by reference. Crystalline forms, i.e., forms 1-6, of sofosbuvir are disclosed in U.S. patent publication nos.: 2010/0298257 and 2100/0251152, both of which are incorporated herein by reference. The sofosbuvir of forms 1-6 have the following characteristic X-ray powder diffraction (XRPD) pattern 2 θ values measured according to the XRPD method disclosed in the patent:
(1) the 2 θ -reflection (° ± 0.2 θ) lies at about: 7.5, 9.6 and 18.3 (form 1);
(2) the 2 θ -reflection (° ± 0.2 θ) lies at about: 5.0,7.3 and 18.1 (form 1);
(3) the 2 θ -reflection (° ± 0.2 θ) lies at about: 6.9, 24.7 and 25.1 (form 2);
(4) the 2 θ -reflection (° ± 0.2 θ) lies at about: 19.7, 20.6 and 24.6 (form 3);
(5) the 2 θ -reflection (° ± 0.2 θ) lies at about: 5.0, 6.8 and 24.9 (form 4);
(6) the 2 θ -reflection (° ± 0.2 θ) lies at about: 5.2, 6.6 and 19.1 (form 5);
and
(7) the 2 θ -reflection (° ± 0.2 θ) lies at about: 6.1, 20.1 and 20.8 (form 6).
Form 6 as described in the above-mentioned patent may be referred to as form 2, for example by the food and drug administration. According to the teachings disclosed in U.S. patent publication nos.: 2010/0298257 and 2100/0251152, the following characteristic XRDP pattern 2 θ values alternately characterize form 1 and form 6:
(1) the 2 θ -reflection (°) is at about: 5.0 and 7.3 (form 1); and
(2) the 2 θ -reflection (°) is at about: 6.1 and 12.7 (form 6).
In one embodiment, the sofosbuvir crystals have an XRPD2 θ -reflection (° ± 0.2 θ), said XRPD2 θ -reflection (° ± 0.2 θ) is at about:
(1)7.5, 9.6 and 18.3; (form 1A)
(2)5.0, 7.3 and 18.1; (form 1B)
(3)6.9, 24.7 and 25.1; (form 2)
(4)19.7, 20.6 and 24.6; (form 3)
(5)5.0, 6.8 and 24.9; (form 4)
(6)5.2, 6.6 and 19.1; (form 5) or
(7)6.1, 20.1 and 20.8; (form 6).
In certain embodiments, the sofosbuvir crystals have an XRPD2 θ -reflection (° ± 0.2 θ), said XRPD2 θ -reflection (° ± 0.2 θ) is at about:
(1)5.2, 7.5, 9.6, 16.7, 18.3 and 22.2 (form 1);
(2)5.0, 7.3, 9.4 and 18.1 (form 1);
(3)4.9, 6.9, 9.8, 19.8, 20.6, 24.7, 25.1 and 26.1 (form 2);
(4)6.9, 9.8, 19.7, 20.6 and 24.6 (form 3);
(5)5.0, 6.8, 19.9, 20.6, 20.9 and 24.9 (form 4);
(6)5.2, 6.6, 7.1, 15.7, 19.1 and 25.0 (form 5); or
(7)6.1、8.2、10.4、12.7、17.2、17.7、18.0、18.8、19.4、
19.8, 20.1, 20.8, 21.8 and 23.3 (form 6).
In another embodiment, the sofosbuvir crystals are in the range of about: 6.1, 8.2, 10.4, 12.7, 17.2, 17.7, 18.0, 18.8, 19.4, 19.8, 20.1, 20.8, 21.8 and 23.3 have XRPD2 theta-reflections (° + -0.2 theta). In another embodiment, the sofosbuvir crystals are in the range of about: 6.1 and 12.7 had XRPD2 theta-reflections (° + -0.2 theta).
A therapeutically effective amount of sofosbuvir may be present in the pharmaceutical composition. In some embodiments, from about 10% to about 70% w/w of sofosbuvir is included in the pharmaceutical composition. In other embodiments, the composition comprises from about 15% to about 65% w/w, or from about 20% to about 60% w/w, or from about 25% to about 55% w/w, or from about 30% to about 50% w/w, or from about 35% to about 45% w/w of sofosbuvir. In other embodiments, the pharmaceutical composition comprises about 10% w/w, about 15% w/w, about 20% w/w, about 25% w/w, about 30% w/w, about 35% w/w, about 45% w/w, about 50% w/w, about 55% w/w, about 60% w/w, about 65% w/w, or about 70% w/w, or about 75% w/w of sofosbuvir. In a particular embodiment, the pharmaceutical composition comprises about 40% w/w sofosbuvir.
C. Auxiliary materials
The pharmaceutical compositions provided according to the invention are typically administered orally. The present invention thus provides pharmaceutical compositions comprising a solid dispersion comprising ledipasvir as described herein and one or more pharmaceutically acceptable excipients or carriers, including but not limited to inert solid diluents and fillers, including sterile aqueous solutions and various organic solvents, penetration enhancers, solubilizers, disintegrants, lubricants, binders, glidants, adjuvants, and combinations thereof. Such compositions are prepared by methods well known in the pharmaceutical art (see, for example, the entire Reminden pharmaceutical monograph, MacePathishing Co., Philadelphia, PA, 17 th edition (1985); and modern pharmaceuticals, Marcel Dekker, Inc. 3 rd edition (G.S. Bank & C.T. Rhodes, Eds.)).
The pharmaceutical composition may be administered orally in a single dose or in multiple doses. Administration can be by capsule, tablet, and the like. In one embodiment, the ledipasvir is in the form of a tablet. In another embodiment, the tablet is a compressed tablet. In preparing a pharmaceutical composition comprising a solid as described herein, the active ingredient is typically diluted with an excipient and/or enclosed within a carrier, which may be in the form of a capsule, tablet, sachet, paper or other container. When the adjuvant serves as a diluent, it may be in the form of a solid, semi-solid or liquid material (as above) which acts as a vehicle, carrier or medium for the active ingredient.
The pharmaceutical composition can be made into quick release or sustained release. A "sustained release formulation" is one designed to slowly release the therapeutic agent over a longer period of time in vivo, while an "immediate release formulation" is one designed to rapidly release the therapeutic agent over a shorter period of time in vivo. In some cases immediate release formulations may be coated so that the therapeutic agent is only released when the intended target in the body (e.g., the stomach) is reached. In a particular embodiment, the pharmaceutical composition is formulated as an immediate release formulation.
The pharmaceutical composition may further comprise pharmaceutical excipients such as diluents, binders, fillers, glidants, disintegrants, lubricants, solubilizers and combinations thereof. Some examples of suitable excipients are described herein. When the pharmaceutical composition is formulated as a tablet, the tablet may be uncoated or may be coated by known techniques, including microencapsulation, to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained release effect over a longer period of time. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
In one embodiment, the pharmaceutical composition comprises a diluent selected from the group consisting of dibasic calcium phosphate, cellulose, compressible sugar, dibasic calcium phosphate dehydrate, lactose monohydrate, mannitol, microcrystalline cellulose, starch, tribasic calcium phosphate, and combinations thereof.
In another embodiment, the pharmaceutical composition comprises lactose monohydrate in an amount from about 1% to about 50% w/w, or from about 1% to about 45% w/w, or from about 5% to about 40% w/w, or from about 5% to about 35% w/w, or from about 1% to about 25% w/w, or from about 10% to about 20% w/w. In particular embodiments, the lactose monohydrate is about 5% w/w, about 10% w/w, about 15% w/w, about 20% w/w, about 25% w/w, about 30% w/w, about 35% w/w, about 40% w/w, about 45% w/w, or about 50% w/w. In another particular embodiment, the lactose monohydrate is present in an amount of about 16.5% w/w.
In yet another embodiment, the pharmaceutical composition comprises microcrystalline cellulose in an amount from about 1% to about 40% w/w, or from about 1% to about 35% w/w, or from about 1% to about 25% w/w, or from about 5% to about 25% w/w, or from about 10% to about 25% w/w, or from about 15% to about 20% w/w. In particular embodiments, the amount of microcrystalline cellulose is about 5%, or about 10%, or about 15%, or about 20%, or about 25%, or about 30%, or about 35%, or about 40% w/w. In another particular embodiment, the amount of microcrystalline cellulose is 18% w/w.
In other embodiments, the pharmaceutical composition comprises a disintegrant selected from the group consisting of croscarmellose sodium, crospovidone, microcrystalline cellulose, modified corn starch, povidone, pregelatinized starch, sodium starch glycolate, and combinations thereof.
In certain embodiments, the pharmaceutical composition comprises croscarmellose sodium in an amount from about 1% to about 20% w/w, or from about 1% to about 15% w/w, or from about 1% to about 10% w/w, or from about 1% to about 8% w/w, or from about 2% to about 8% w/w. In particular embodiments, the amount of croscarmellose sodium is about 1%, or about 3%, or about 6%, or about 8%, or about 10%, or about 13%, or about 15% w/w. In another particular embodiment, the amount of croscarmellose sodium is about 5% w/w.
In other embodiments, the pharmaceutical composition comprises a glidant selected from the group consisting of colloidal silicon dioxide, talc, starch derivatives, and combinations thereof.
In other embodiments, the pharmaceutical composition comprises colloidal silicon dioxide in an amount from about 0.1% to about 5% w/w, or from about 0.1% to about 4.5% w/w, or from about 0.1% to about 4% w/w, or from about 0.5% to about 5.0% w/w, or from about 0.5% to about 3% w/w, or from about 0.5% to about 2% w/w, or from about 0.5% to about 1.5% w/w. In particular embodiments, the amount of colloidal silica is about 0.1% w/w, 0.5% w/w, 0.75% w/w, 1.25% w/w, 1.5% w/w, or 2% w/w. In another particular embodiment, the amount of colloidal silica is about 1% w/w.
In other embodiments, the pharmaceutical composition comprises a lubricant selected from the group consisting of calcium stearate, magnesium stearate, polyvinyl alcohol, sodium stearyl fumarate, stearic acid, talc, and combinations thereof.
In other embodiments, the pharmaceutical composition comprises magnesium stearate in an amount from about 0.1% to about 3% w/w, or from about 0.1% to about 2.5% w/w, or from about 0.5% to about 3% w/w, or from about 0.5% to about 2.5% w/w, or from about 0.5% to about 2% w/w, or from about 1% to about 3% w/w, or from about 1% to about 2% w/w. In particular embodiments, the amount of magnesium stearate is about 0.1%, or about 0.5%, or about 1%, or about 2%, or about 2.5%, or about 3% w/w. In another particular embodiment, the amount of magnesium stearate is about 1.5% w/w.
In one embodiment, the pharmaceutical composition comprises a) about 30% to about 50% w/w sofosbuvir and b) about 5% to about 35% w/w solid dispersion containing ledipasvir. In a related embodiment, the composition comprises a) about 40% w/w sofosbuvir and b) about 18% w/w ledipasvir containing solid dispersion. In yet another related embodiment, the composition further comprises a) from about 5% to about 25% w/w lactose monohydrate, b) from about 5% to about 25% w/w microcrystalline cellulose, c) from about 1% to about 10% w/w croscarmellose, d) from about 0.5% to about 3% w/w colloidal silicon dioxide, and e) from about 0.1% to about 3% w/w magnesium stearate. In another embodiment, a pharmaceutical composition comprises a) about 40% w/w of ospibuvir, b) about 18% w/w of a solid dispersion containing ledipasvir, c) about 16.5% w/w of lactose monohydrate, d) about 18% w/w of microcrystalline cellulose, e) about 5% w/w of croscarmellose, f) about 1% w/w colloidal silicon dioxide, and g) about 1.5% w/w magnesium stearate.
3. Pharmaceutical dosage form
The present invention provides tablets, pills, and the like, comprising the pharmaceutical compositions or dosage forms described herein. The tablets or pills of the invention may be coated to provide a dosage form with a sustained release effect or which is not affected by the acidic conditions of the stomach. Tablets may also be formulated as immediate release formulations as previously described. In certain embodiments, the tablet comprises a film coating. Film coatings may be used to limit photodegradation. Suitable film coatings can be selected by routine screening of commercially available formulations. In one embodiment, the film coating is a polyvinyl alcohol based coating.
The tablet can be made into single layer or double layer tablet. Generally, a monolayer tablet comprises the active ingredients (i.e., ledipasvir and sofosbuvir) co-mixed with a uniform monolayer. To prepare a single layer tablet, exemplary methods include, but are not limited to, co-blending (or double granulation) and co-dry granulation. Co-blend granulation is a multi-step process consisting of dry granulation of each active ingredient with excipients separately and mixing the two granulations together. The co-dry granulation consists of dry granulation of the two active ingredients together with excipients.
Bilayer tablets containing the active ingredients (i.e. ledipasvir and sofosbuvir) in separate layers can be prepared by making a mixture containing the excipient and one active ingredient (i.e. ledipasvir) and making a separate mixture containing the second active ingredient (i.e. sofosbuvir) and the excipient. One mixture is then pre-compressed and a second mixture may then be added to the top of the first pre-compressed mixture. The resulting tablet comprises two separate layers, each layer containing a different active ingredient.
In one embodiment, the tablet comprises a) from about 30% to about 50% w/w of ospobrevir and b) from about 10% to about 40% w/w of a solid dispersion comprising ledipasvir. In a related embodiment, the tablet comprises a) about 40% w/w of ospibuvir and b) about 18% w/w of a solid dispersion containing ledipasvir. In another embodiment, the tablet comprises a) about 300mg to about 500mg sofosbuvir and b) about 50mg to 130mg ledipasvir. In another embodiment, the tablet comprises a) about 400mg of sofosbuvir and b) about 90mg of ledipasvir. In a related embodiment, the tablet further comprises a) from about 5% to about 25% w/w lactose monohydrate, b) from about 5% to about 25% w/w microcrystalline cellulose, c) from about 1% to about 10% w/w croscarmellose sodium, d) from about 0.5% to about 3% w/w colloidal silicon dioxide, and e) from about 0.1% to about 3% w/w magnesium stearate.
In some embodiments, the pharmaceutical compositions described herein are formulated as a unit dose or pharmaceutical dosage form. The term "unit dosage form" or "pharmaceutical dosage form" refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient (e.g., a tablet or capsule). The compounds are generally administered in a pharmaceutically effective amount. In some embodiments, each dosage unit contains from 3mg to 2 gledipasvir. In other embodiments, the pharmaceutical dosage form contains from about 3mg to about 360mg, or about 10mg to about 200mg, or about 10mg to about 50mg, or about 20mg to about 40mg, or about 25mg to about 35mg, or about 40mg to about 140mg, or about 50mg to about 130mg, or about 60mg to about 120mg, or about 70mg to about 110mg, or about 80mg to about 100 mg. In particular embodiments, the pharmaceutical dosage form contains about 40mg, or about 45mg, or about 50mg, or about 55mg, or about 60mg, or about 70mg, or about 80mg, or about 100mg, or about 120mg, or about 140mg, or about 160mg, or about 180mg, or about 200mg, or about 220mg ledipasvir. In another specific embodiment, the pharmaceutical dosage form contains about 90mg ledipasvir. In yet another specific embodiment, the pharmaceutical dosage form contains about 30mg ledipasvir.
In other embodiments, the pharmaceutical dosage form contains from about 1mg to about 3g of sofosbuvir. In other embodiments, the pharmaceutical dosage form contains from about 1mg to about 800mg, or about 100mg to about 700mg, or about 200mg to about 600mg, or about 300mg to about 500mg, or about 350mg to about 450mg of sofosbuvir. In particular embodiments, the pharmaceutical dosage form contains about 50mg, or about 100mg, or about 150mg, or about 200mg, or about 250mg, or about 300mg, or about 350mg, or about 450mg, or about 500mg, or about 550mg, or about 600mg, or about 650mg, or about 700mg, or about 750mg, or about 800 sofosbuvir. In another specific embodiment, the pharmaceutical dosage form contains about 400mg of sofosbuvir. It will be appreciated that the amount of ledipasvir and/or sofosbuvir actually administered will generally be determined by a physician in view of the relevant circumstances including the condition to be treated, the chosen route of administration, the compound actually administered and its relative activity, the age, weight and response of the individual patient, the severity of the patient's symptoms, and the like.
In a particular embodiment, the pharmaceutical dosage form contains about 400mg of sofosbuvir and about 90mg of ledipasvir.
In one embodiment, the pharmaceutical composition or pharmaceutical dosage form or tablet comprises about 90mg of amorphous ledipasvir (wherein the ratio of polymer: ledipasvir is 1:1), about 400mg of crystalline sofosbuvir, lactose monohydrate in an amount from about 5% to about 25% w/w, microcrystalline cellulose in an amount from about 5% to about 25% w/w, croscarmellose sodium in an amount from about 1% to about 10% w/w, colloidal silicon dioxide in an amount from about 0.5% to about 3% w/w, and magnesium stearate in an amount from about 0.1% to about 3% w/w, in a solid dispersion. In one embodiment, the polymer is copovidone.
In other embodiments, the pharmaceutical composition, pharmaceutical dosage form, or tablet described herein does not have negative drug-drug interactions. In a related embodiment, the pharmaceutical composition, pharmaceutical dosage form, or tablet does not have a negative drug-drug interaction with the acid-suppression therapy. In another embodiment, the pharmaceutical composition, pharmaceutical dosage form, or tablet described herein can be administered without regard to food and with or without regard to whether the patient is undergoing acid suppression therapy.
4. Application method
The solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms and tablets of ledipasvir and sofosbuvir described herein are orally administered to patients suffering from Hepatitis C Virus (HCV) infection at daily doses. In one embodiment, the patient is a human.
Ledipasvir has previously been shown to have a negative food effect when administered alone. Unexpectedly, the combination treatment of ledipasvir and sofosbuvir did not show negative food effects. Thus, the pharmaceutical composition comprising sofosbuvir and ledipasvir is administered without regard to food.
In some embodiments, the combination composition achieves a reduced food impact. In some aspects, the composition achieves an initial exposure when administered to a patient after a meal that is no more than 25%, or no more than 20%, 15%, or 10% less than a second exposure when not administered to a patient after a meal. The exposure can be detected as Cmax、AUClastOr AUCinf. In some aspects, administration is performed within four, three, two, or one hour after a meal.
In one embodiment, the solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms, and tablets of ledipasvir and sofosbuvir described herein are effective in treating one or more of a genotype 1 HCV-infected patient, a genotype 2 HCV-infected patient, a genotype 3 HCV-infected patient, a genotype 4 HCV-infected patient, a genotype 5 HCV-infected patient, and/or a genotype 6 HCV-infected patient. In one embodiment, the solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms, and tablets of ledipasvir and sofosbuvir described herein are effective in treating genotype 1HCV infected patients, including genotype 1a and/or genotype 1 b. In another embodiment, the solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms, and tablets of ledipasvir and sofosbuvir described herein are effective in treating genotype 2HCV infected patients, including genotype 2a, genotype 2b, genotype 2c, and/or genotype 2 d. In another embodiment, the solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms, and tablets of ledipasvir and sofosbuvir described herein are effective in treating genotype 3HCV infected patients, including genotype 3a, genotype 3b, genotype 3c, genotype 3d, genotype 3e, and/or genotype 3 f. In another embodiment, the solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms, and tablets of ledipasvir and sofosbuvir described herein are effective in treating genotype 4 HCV-infected patients, including genotype 4a, genotype 4b, genotype 4c, genotype 4d, genotype 4e, genotype 4f, genotype 4g, genotype 4h, genotype 4i, and/or genotype 4 j. In another embodiment, the solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms, and tablets of ledipasvir and sofosbuvir described herein are effective in treating patients infected with genotype 5HCV, including genotype 5 a. In another embodiment, the solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms, and tablets of ledipasvir and sofosbuvir described herein are effective in treating patients infected with genotype 6HCV, including genotype 6 a. In one embodiment, the compositions are pan-genotypic, meaning that they are effective for all genotypes and their resistant mutants.
In some embodiments, the solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms, and tablets of ledipasvir and sofosbuvir described herein are administered alone or in combination with one or more therapeutic agents for treating HCV (e.g., HCV ns3 protease inhibitor or HCV ns5B polymerase inhibitor) for about 24 weeks, about 16 weeks, or about 12 weeks or less. In other embodiments, the solid dispersions, pharmaceutical compositions, pharmaceutical dosage forms, and tablets of ledipasvir and sofosbuvir are administered alone or in combination with one or more therapeutic agents for treating HCV (e.g., HCV ns3 protease inhibitor or HCV ns5B polymerase inhibitor) for about 24 weeks or less, about 22 weeks or less, about 20 weeks or less, about 18 weeks or less, about 16 weeks or less, about 12 weeks or less, about 10 weeks or less, about 8 weeks or less, about 6 weeks or less, or about 4 weeks or less. The solid dispersion, pharmaceutical composition, pharmaceutical dosage form and tablet may be administered once daily, twice daily, once bidaily, twice weekly, thrice weekly, four times weekly or five times weekly.
In other embodiments, a sustained viral response is achieved at about 4 weeks, 6 weeks, 8 weeks, 12 weeks, or 16 weeks, or at about 20 weeks, or at about 24 weeks, or at about 4 months, or at about 5 months, or at about 6 months, or at about 1 year, or at about 2 years.
In one embodiment, the daily dose is 90mg of ledippsvir and 400mg of sofosbuvir administered in tablet form. In another embodiment, the daily dose is a tablet comprising a) about 30% to about 50% w/w of ospibuvir, b) about 10% to about 40% w/w of a solid dispersion comprising ledipasvir, c) about 5% to about 25% w/w of lactose monohydrate, d) about 5% to about 25% w/w of microcrystalline cellulose, e) about 1% to about 10% w/w of croscarmellose sodium, f) about 0.5% to about 3% w/w colloidal silicon dioxide, and g) about 0.1 to about 3% w/w magnesium stearate.
In other embodiments, the patient also suffers from cirrhosis. In yet another embodiment, the patient does not have cirrhosis.
5. Combination therapy
In the methods described herein, the methods may further comprise the use of another therapeutic agent for the treatment of HCV and other conditions, such as HIV infection. In one embodiment, non-limiting examples of suitable additional therapeutic agents include one or more interferons, ribavirin or its analogs, HCV ns3 protease inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, nucleoside or nucleotide inhibitors of HCV ns5B polymerase, non-nucleoside or inhibitors of HCV ns5B polymerase, HCV ns5A inhibitors, TLR-7 agonists, cyclophilin inhibitors, HCV ires inhibitors, pharmacokinetic enhancers, and other drugs or therapeutic agents for treating HCV.
More specifically, the other therapeutic agent may be selected from the group consisting of:
1) interferons, such as PEGylated rIFN-. alpha.2b (PEG-Intron), PEGylated rIFN-. alpha.2a (Pegasys), rIFN-. alpha.2b (Intron-A), rIFN-. alpha.2a (Roferon-A), interferon-. alpha. (MOR-22, OPC-18, Alfaferone, Alfanative, Multiferon, subalin), consensus interferon (Infergen), interferon-. alpha.n 1(Wellferon), interferon-. alpha.n 3(Alferon), interferon-. beta.Avonex, DL-8234, interferon-. omega. (ω DUROS, Biomed510), binderleton. alpha.2b (Albuferon), IFN-. alpha.2bXL, BLX-883(Locteron), DA-3021, glycosylated interferon-. alpha.2b (AVI-005), PEG-replin (Inferon), PEG-1 (PEG-29), and PEG-foel;
2) ribavirin and its analogs, such as ribavirin (rebeol, Copegus) and taribavirin (viramidine);
3) HCVNS3 protease inhibitors, for example, boceprevir (SCH-503034, SCH-7), telaprevir (VX-950), TMC435350, BI-1335, BI-1230, MK-7009, VBY-376, VX-500, GS-9256, GS-9451, BMS-605339, PHX-1766, AS-101, YH-5258, YH5530, YH5531, ABT-450, ACH-1625, ITMN-191, MK5172, MK6325, and MK 2748.
4) Alpha-glucosidase 1 inhibitors, such as Cigosivir (MX-3253), miglitol and UT-231B.
5) Hepatoprotective agents such as emericasan (IDN-6556), ME-3738, GS-9450(LB-84451), siliblin, and MitoQ;
6) nucleoside or nucleotide inhibitors of HCV NS5B polymerase such as R1626, R7128(R4048), IDX184, IDX-102, BCX-4678, valopicitabine (NM-283), MK-0608 and INX-189 (now known as BMS 986094);
7) non-nucleoside inhibitors of HCV NS5B polymerase, such as PF-868554, VCH-759, VCH-916, JTK-652, MK-3281, GS-9190, VBY-708, VCH-222, A848837, ANA-598, GL60667, GL59728, A-63890, A-48773, A-48547, BC-2329, VCH-796(nesbuvir), GSK625433, BILN-1941, XTL-2125, ABT-072, ABT-333, GS-9669, PSI-7792 and GS-9190;
8) inhibitors of HCVNS5A, such as AZD-2836(A-831), BMS-790052, ACH-3102, ACH-2928, MK8325, MK4882, MK8742, PSI-461, IDX719, ABT-267, and A-689;
9) TLR-7 agonists such as imiquimod, 852A, GS-9524, ANA-773, ANA-975, AZD-8848(DSP-3025) and SM-360320;
10) cyclophilin inhibitors such as DEBIO-025, SCY-635 and NIM 811;
11) HCVIRES inhibitors, such as MCI-067;
12) pharmacokinetic enhancers, such as BAS-100, SPI-452, PF-4194477, TMC-41629, GS-9350, GS-9585 and roxithromycin; and
13) other drugs for HCV treatment, such as thymosin alpha 1 (Zadaxin), nitazoxanide (Alinean, NTZ), BIVN-401(virostat), PYN-17(altirex), KPE02003002, actilon (CPG-10101), GS-9525, KRN-7000, civacir, GI-5005, XTL-6865, BIT225, PTX-111, ITX2865, TT-033i, ANA971, NOV-205, tarvacin, EHC-18, VGX-410C, EMZ-702, AVI4065, BMS-650032, BMS-791325, bavin, MDX-1106(ONO-4538), Oglufanide and VX-497 (merepodii).
More specifically, the additional therapeutic agent may be combined with one or more compounds selected from the group consisting of HCV NS5B protease inhibitors (ABT-072 and ABT-333), HCV NS5A inhibitors (ACH-3102 and ACH-2928), and HCV NS3 protease inhibitors (ABT-450 and ACH-125).
In another embodiment, the therapeutic agent used in combination with the pharmaceutical composition described herein can be any agent that has a therapeutic effect when used in combination with the pharmaceutical composition described herein. For example, therapeutic agents for use in combination with the pharmaceutical compositions described herein can be interferons, virazole analogs, NS3 protease inhibitors, NS5B protease inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, non-nucleoside inhibitors of HCV, and other agents for treating HCV.
In another embodiment, the other therapeutic agent used in combination with the pharmaceutical composition described herein is a cyclophilin inhibitor, including, for example, one disclosed in WO 2013/185093. Non-limiting examples include compounds selected from the group consisting of the following compounds and their stereoisomers and mixtures of stereoisomers:
in another embodiment, the additional therapeutic agent used in combination with the pharmaceutical composition described herein is a non-nucleoside inhibitor of hcv ns5B polymerase. Non-limiting examples include compound E (described below).
Other examples of anti-HCV drugs that can be used in combination with the compositions provided herein include, but are not limited to, the following:
A. interferons, for example PEGylated rIFN-. alpha.2b (PEG-Intron), PEGylated rIFN-. alpha.2a (Pegasys), rIFN-. alpha.2b (Intron-A), rIFN-. alpha.2a (Roferon-A), interferon-. alpha. (MOR-22, OPC-18, alfafenone, Alfanative, Multiferon, subalin), consensus interferon (interferon alfacon-1) (Infergen), interferon-. alpha.n 1(Wellferon), interferon-. alpha.n 3(Alferon), interferon-. beta.Avonex, DL-8234), interferon-. omega.DUROS, Biomed510, interferon-. alpha.2 b (Albuferon), IFN. alpha.XL, BLX-883 (Loctron), DA-3021, glycosylated interferon-. alpha.2 b (I-I), interferon-. alpha.005-2 b (PEG-beta.L), PEG-25, PEG-IFN-. alpha. 2bXL, PEG-IFN-. alpha., rIFN α -2a, consensus interferon α (consensus IFN α), xerophthalmia (benefigen), rebif, PEGylated INF- β, oral interferon a, feron, reaferon, INTERmaxa, r-INF- β, and xerophthalmia (benefigen) + actimmenobrevin and viramidine analogs such as rebeol, copogus, VX-497, and viramidine (taribavirin);
NS5A inhibitors, for example, Compound B (described below), Compound C (described below), ABT-267, Compound D (described below), JNJ-47910382, daclatasvir (BMS-790052), ABT-267, MK-8742, EDP-239, IDX-719, PPI-668, GSK-2336805, ACH-3102, A-831, A-689, AZD-2836(A-831), AZD-7295(A-689), and BMS-790052;
NSB5 polymerase inhibitors, for example Compound E (described below), Compound F (described below), ABT-333, Compound G (described below), ABT-072, Compound H (described below), tegobrevir (GS-9190), GS-9669, TMC647055, setobrevir (ANA-598), non-ribavirin (PF-868554), VX-222, IDX-375, IDX-184, IDX-102, BI-207127, valicitabine (NM-283), PSI-6130(R1656), PSI-7851, BCX-4678, nesbuvir (HCV-796), BILB1941, MK-0608, NM-107, R7128, VCH-759, GSK625433, XTL-2125, VCH-916, JTK-652, MK-3281, VBY-708, A848837, GL59728, A-63890, A-48773, A-48547, BC-2329, BMS-791325 and BILB-1941;
NS3 protease inhibitors, such as Compound I, Compound J, Compound K, ABT-450, Compound L (below), simeprevir (TMC-435), Pospervir (SCH-503034), narraprevir (SCH-900518), vaniprevir (MK-7009), MK-5172, danoprevir (ITMN-191), sovaprevir (ACH-1625), neceprevir (ACH-2684), Telaprevir (VX-950), VX-813, VX-500, faaprevir (BI-335), asunaprevir (BMS-650032), BMS-605339, VBY-376, PHX-1766, 55YH 31, BILN-2065 and BILN-2061;
E. alpha-glucosidase 1 inhibitors, such as Cigosivir (MK-3253), miglitol and UT-231B;
F. hepatoprotective agents such as IDN-6556, ME3738, MitoQ and LB-84451;
non-nucleoside inhibitors of hcv, such as benzimidazole derivatives, benzo-1, 2, 4-thiadiazine derivatives and phenylalanine derivatives; and
H. other anti-HCV drugs, such as Ridaxin, nitazoxanide (alinea), BIVN-401(virostat), DEBIO-025, VGX-410C, EMZ-702, AVI4065, bavaximab, oglufanide, PYN-17, KPE02003002, actilon (CPG-10101), KRN-7000, civacir, GI-5005, ANA-975, XTL-6865, ANA971, NOV-205, tarvacin, EHC-18, and NIM 811.
Compound B is an NS5A inhibitor, represented by the following chemical structure:
compound C is an NS5A inhibitor, represented by the following chemical structure:
compound D is an NS5A inhibitor, represented by the following chemical structure:
see U.S. patent publication No. 2013/0102525 and references cited therein.
Compound E is an NS5 bthumbiii polymerase inhibitor, represented by the following chemical structure:
compound F is a nucleotide inhibitor prodrug designed to inhibit viral RNA replication by hcv ns5B polymerase, represented by the following chemical structure:
compound G is an HCV polymerase inhibitor represented by the following chemical structure:
see U.S. patent publication No. 2013/0102525 and references cited therein.
Compound H is an HCV polymerase inhibitor represented by the following chemical structure:
see U.S. patent publication No. 2013/0102525 and references cited therein.
Compound I is an HCV protease inhibitor represented by the following chemical structure:
see U.S. patent publication No. 2014/0017198 and references cited therein.
Compound J is an HCV protease inhibitor represented by the following chemical structure:
see U.S. patent No. 8,178,491 and references cited therein.
Compound K is an HCV protease inhibitor represented by the following chemical structure:
compound L is an HCV protease inhibitor represented by the following chemical structure:
see U.S. patent publication No. 2013/0102525 and references cited therein.
In one embodiment, the additional therapeutic agent used in combination with the pharmaceutical composition described herein is an hcv ns3 protease inhibitor. Non-limiting examples include, but are not limited to, one or more compounds selected from the group consisting of:
and
in another embodiment, the present application provides a method of treating hepatitis C in a human patient comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition described herein and an additional therapeutic agent selected from the group consisting of pegylated rIFN- α 2b, pegylated rIFN- α 2a, rIFN- α 2b, IFN α -2bXL, rIFN- α 2a, consensus IFN α, Dry Diffjin (Infergen), Libiper, Loctreron, AVI-005, PEG-Dry Difjin, PEG-Pepper, oral Interferon α, feron, Reaferon, Intermax α, r-IFN- β, Dry Difujin + Interferon γ -1b, IFN- ω and DUROS, albuferon, Rebetol, Copegus, levovirin, VX-497, Ribavirin, A-9, and a pharmaceutically acceptable carrier or excipient, NM-283, valopicitabine, R1626, PSI-6130(R1656), HCV-796, BILB1941, MK-0608, NM-107, R7128, VCH-759, PF-868554, GSK625433, XTL-2125, SCH-503034(SCH-7), VX-950(Telaprevir), ITMN-191, and BILN-2065, MX-3253 (Cigosavir), UT-231B, IDN-6556, ME3738, MitoQ, and LB-84451, benzimidazole derivatives, benzo-1, 2, 4-thiadiazine derivatives, and phenylalanine derivatives, DADAXIAN (zadaxin), nitazoxanide (alinea), BIVN-401(virostat), DEBIO-025, VGX-410C, EMZ-702, AVI4065, KRI, valacitrexib, PYLUE-02017, CPK-01, GI-5005, GI-84451, SAG-84451, and its derivatives, ANA-975 (isatoribine), XTL-6865, ANA971, NOV-205, tarvacin, EHC-18, and NIM 811.
In yet another embodiment, the present application provides a pharmaceutical combination comprising:
a) a first pharmaceutical composition comprising an effective amount of ledipasvir that is substantially amorphous as described herein; and an essentially crystalline effective amount of sofosbuvir and
b) a second pharmaceutical composition comprising at least one additional therapeutic agent selected from the group consisting of HIV protease inhibiting compounds, HIV reverse transcriptase non-nucleoside inhibitors, HIV reverse transcriptase nucleotide inhibitors, HIV integrase inhibitors, gp41 inhibitors, CXCR4 inhibitors, gp120 inhibitors, CCR5 inhibitors, interferons, virazole analogs, NS3 protease inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, HCV non-nucleoside inhibitors, and other agents for treating HCV, and combinations thereof.
The other therapeutic agent may be a drug for treating other conditions such as HIV infection. Thus, the additional therapeutic agent may be a compound for the treatment of HIV, such as HIV protease inhibiting compounds, HIV reverse transcriptase non-nucleoside inhibitors, HIV reverse transcriptase nucleotide inhibitors, HIV integrase inhibitors, gp41 inhibitors, CXCR4 inhibitors, gp120 inhibitors, CCR5 inhibitors, interferons, virazole analogs, NS3 protease inhibitors, NS5b polymerase inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, HCV non-nucleoside inhibitors, and other agents for the treatment of HCV.
More specifically, the other therapeutic agent may be selected from the group consisting of:
1) HIV protease inhibitors, such as amprenavir, atazanavir, fosamprenavir, indinavir, lopinavir, ritonavir, lopinavir + ritonavir, nelfinavir, saquinavir, tipranavir, beconavir, darunavir, TMC-126, TMC-114, mozenavir (DMP-450), JE-2147(AG1776), AG1859, DG35, L-756423, RO0334649, KNI-272, DPC-681, DPC-684 and GW640385X, DG17, PPL-100,
2) non-nucleoside inhibitors of HIV reverse transcriptase, such as cappuccinin, emivirin, delavirdine (delavirdine), efavirenz, nevirapine, Calophyllum extract ((+) calanolide A), etravirin, GW5634, DPC-083, DPC-961, DPC-963, MIV-150 and TMC-120, TMC-278 (rilpivirin), efavirenz, BILR355BS, VRX840773, UK-453,061, RDEA806,
3) HIV reverse transcriptase nucleoside inhibitors, for example zidovudine, emtricitabine, didanosine, stavudine, zalcitabine, lamivudine, abacavir, amdoxovir, elvucitabine, alovudine, MIV-210, lacivir (. + -. -FTC), D-D4FC, emtricitabine, triazophos, fozivudine tidoxil, fosvudine tidoxil, aricitabine (AVX754), amdoxovir, KP-1461, abacavir + lamivudine + zidovudine, zidovudine + lamivudine,
4) HIV reverse transcriptase nucleotide inhibitors, for example tenofovir, tenofovir disoproxil fumarate + emtricitabine + efavirenz and adefovir,
5) HIV integrase inhibitors, such as curcumin, curcumin derivatives, chicoric acid derivatives, 3, 5-dicaffeoylquinic acid derivatives, aurintricarboxylic acid derivatives, caffeic acid phenethyl ester derivatives, tyrphostin derivatives, quercetin derivatives, S-1360, zintevir (AR-177), L-870812 and L-870810, MK-0518 (raltegravir), BMS-70707035, MK-2048, BA-011, BMS-538158, GSK3647 364735C,
6) gp41 inhibitors, such as enfuvirdine, cidevir peptide, FB006M, TRI-1144, SPC3, DES6, locus gp41(Locusgp41), CovX and REP9,
7) CXCR4 inhibitors, such as AMD-070,
8) entry inhibitors, such as SP01A, TNX-355,
9) gp120 inhibitors, such as BMS-488043 and BlockAide/CR,
10) g6PD and NADH-oxidase inhibitors, such as an immunotin (immutin), 10) CCR5 inhibitors, such as aplaviroc, vkelinor, INCB9471, PRO-140, INCB15050, PF-232798, CCR5mAb004, and molalvuno,
11) interferons, such as PEGylated rIFN-. alpha.2b, PEGylated rIFN-. alpha.2a, rIFN-. alpha.2b, IFN-. alpha.2bXL, rIFN-. alpha.2a, consensus interferon alpha (consensus IFN. alpha.), siccatin, Libix, lotteron, AVI-005, PEG-siccatin, PEGylated IFN-. beta.IFN-. alpha.feron, referon, interfax. alpha., r-IFN-. beta., siccatin + actimmune, interferon-. omega. with DUROS, and albuferon,
12) azole analogs of viruses, such as rebeol, copegus, levovirin, VX-497 and ribavirin prophase (taribavirin)
13) NS5a inhibitors, such as A-831, A-689, and BMS-790052,
14) NS5b polymerase inhibitors, such as NM-283, valopicitabine, R1626, PSI-6130(R1656), HCV-796, BILB1941, MK-0608, NM-107, R7128, VCH-759, PF-868554, GSK625433 and XTL-2125,
15) NS3 protease inhibitors, such as SCH-503034(SCH-7), VX-950(Telaprevir), ITMN-191 and BILN-2065,
16) alpha-glucosidase 1 inhibitors, such as MX-3253 (Cigosivir) and UT-231B,
17) hepatoprotective agents, such as IDN-6556, ME3738, MitoQ and LB-84451,
18) non-nucleoside inhibitors of HCV, such as benzimidazole derivatives, benzo-1, 2, 4-thiadiazine derivatives and phenylalanine derivatives,
19) other drugs for treating hepatitis C, such as Ridaxin, nitazoxanide (alinea), BIVN-401(virostat), DEBIO-025, VGX-410C, EMZ-702, AVI4065, baveximab, oglufanide, PYN-17, KPE02003002, actilon (CPG-10101), KRN-7000, civacir, GI-5005, ANA-975 (isatoribine), XTL-6865, ANA971, NOV-205, tarvacin, EHC-18 and NIM811,
20) pharmacokinetic enhancers, e.g., BAS-100 and SPI452, 20) RNAseH inhibitors, e.g., ODN-93 and ODN-112, and
21) other anti-HIV drugs, such as VGV-1, PA-457(bevirimat), ampligen, HRG214, cytolin, polymun, VGX-410, KD247, AMZ0026, CYT99007, A-221HIV, BAY50-4798, MDX010(iplimumab), PBS119, ALG889, and PA-1050040.
In one embodiment, the additional therapeutic agent is ribavirin. Accordingly, the methods described herein include a method of treating hepatitis c in a human patient in need thereof comprising administering to the patient a therapeutically effective amount of ribavirin and a therapeutically effective amount of a pharmaceutical composition, pharmaceutical dosage form, or tablet described herein. In another embodiment, ribavirin and the pharmaceutical composition, dosage form or tablet comprising sofosbuvir and ledipasvir are administered for about 12 weeks or less. In other embodiments, ribavirin and the pharmaceutical composition, dosage form, or tablet comprising sofosbuvir and ledipasvir are administered for about 8 weeks or less, about 6 weeks or less, or about 4 weeks or less.
It is contemplated that the other therapeutic agents will be administered in methods well known in the art and the dosage may be selected by one skilled in the art. For example, the additional agent may be administered in a dosage of about 0.01 mg to about 2g per day.
Examples
In the following examples and throughout the present invention, abbreviations used herein have the following meanings, respectively:
example 1: synthesis of amorphous Ledipasvir
Methods for preparing various forms of ledipasvir can be found in U.S. publication nos. 2013/0324740 and 2013/0324496. Both of these applications are incorporated herein by reference. The following is a method for isolating ledipasvir amorphous free base.
The ledipasvir acetone solvate (191.4g) and acetonitrile (1356g) were combined in one reaction vessel and the contents mixed until a solution formed. This solution of ledipasvir in acetonitrile was slowly added to another reaction vessel containing vigorously stirred water (7870 g). The contents were stirred at about 23 ℃ for about 30 minutes. The contents were filtered and dried at about 40-45 ℃ until constant weight was reached to give a non-crystalline solid of ledipasvir (146.4g, 82% yield).
Example 2: preparation and formulation of tablets
A. Dosage selection of tablets
i.Sofosbuvir
Tablets the selected dose of sofosbuvir was 400mg once daily. EmaxThe PK/PD model and early virology and human exposure data support a dose of400 mgsofosbuvir, and other trials also support a dose selection of400 mgsofosbuvir.
Mean AUC of major metabolites of sofosbuvir at a dose of400 mgsofosbuvirtauThis value is the plateau of the exposure-response sigmoid curve for approximately 77% of the maximum change in hcv rna from baseline measured according to this model. In S shape EmaxIn the model, there is a relatively linear exposure-response relationship during the 20% to 80% maximum effect range. Thus, whereas the sofosbuvir exposure of a 200mg tablet is dose proportional to a single dose of up to 1200mg, doses below 400mg are expected to show a considerable reduction in the magnitude of the change in hcv rna from baseline. Also, to improve the 77% efficacy prediction in the exposure-response curve plateau, a large increase in antiviral effect would require a large increase in exposure (and thus dose).
A dose of400mg sofosbuvir per day was associated with a higher SVR rate in genotype 1HCV infected patients compared to a dose of 200mg once per day when combined with other HCV treatments for 24 weeks. Both dosage levels have similar safety and tolerability. In addition, 100% SVR24 was observed when 400mg sofosbuvir were administered once daily with other HCV treatments to HCV infected patients of genotype 2 or 3.
ii.Ledipasvir
In all groups administered at doses of 3mg ledipasvir, the maximum median of HCVRNAlog10 was reduced by 3 or more. EmaxThe PK/PD model showed that the exposure achieved after 30mg dose administration provides for patients with genotype 1aHCV infection>95% of the maximal antiviral response. Based on analysis of NS5A mutants that appeared in response to ledipasvir exposure, it was also observed that ledipasvir of 30mg or more may provide coverage for some drug-related mutations that were not provided by doses below 30 mg. Thus, 30mg and 90mg ledipasvir were selected as the dosages for the formulations described herein.
Other studies have shown that the Breakthrough (BT) rate (number of patients reaching hcv rna > lower limit of quantitation (LLOQ) after vRVR/total number of patients reaching vRVR) at a 30mg dose (BT 33%, 11/33; 30mg ledipasvir) is higher than at a 90mg dose (BT 12%, 9/74; 90mg ledipasvir) when ledipasvir is combined with other therapeutic agents. Thus, a 90mg dose of ledipasvir can result in a greater antiviral range that prevents virus breakthrough.
B. Solid dispersion containing Ledipasvir
To prepare tablets containing the sofosbuvir and ledipasvir compositions described herein, a solid dispersion containing ledipasvir is co-formulated with crystalline sofosbuvir. As described herein, the starting material for the solid dispersion may be ledipasvir in a variety of forms, including crystalline forms, non-crystalline forms, salts, solvates, and free bases. This form was used for amorphous solid dispersions of ledipasvir due to high solubility in organic solvents and excipients and the ability to isolate the ledipasvir free base crystalline acetone solvate.
The method of spray drying the solid dispersion results in the most desirable properties compared to other formulation methods, including improved in vivo and in vitro performance and productivity/expandability.
The spray dried feed solution was prepared by dissolving ledipasvir acetone solvate and polymer in the feed solvent. Vigorous mixing or homogenization is performed to avoid agglomeration of the composition.
To obtain the preferred properties of the solid dispersion, different polymers were tested. Nonionic polymers such as hypromellose and copovidone solid dispersions all exhibit sufficient stability and physical characteristics.
The feed solution was first evaluated for solubility, stability and viscosity to select the appropriate solvent. Ethanol, methanol and Dichloromethane (DCM) all showed good solubility (ledipasvir solubility >500 mg/mL). The ease of preparation of ethanol and DCM-based feed stocks was evaluated and the robustness of the spray-drying process was evaluated by spray-drying at a range of inlet and outlet temperatures. Both of these solvents allow rapid dissolution of ledipasvir and copovidone.
High yields (88, 90, 92, 94, 95, 97, 98, 99%) were obtained from ethanol spray drying over a wide range of spray drying exit temperatures (49-70 ℃) with no material accumulation in the spray drying chamber. Yields of 60%, 78% and 44% were obtained from DCM spray drying. In summary, a solid dispersion of ledipasvir (50% w/w) with a 1:1 ratio of ledipasvir to copovidone has good chemical stability in ethanol feed solution.
A homogenization procedure was used to prepare a 10% ledipasvir acetone solvate and a 10% copovidone in ethanol solution. Ledipasvir, an ethanolic solution of copovidone, has a very low viscosity (-65 cP) as measured by 30% solids content.
Spray drying is carried out using two liquid nozzles or one hydraulic nozzle. Table 1 lists the spray drying process parameters evaluated using an AnhydroMS35 spray dryer for 100g to 4000g total feed solution, and Table 2 shows the spray drying process parameters using a hydraulic nozzle. The particle size data indicate that the particle size is sufficiently large (10-14 μm average PS) and there is substantially no effect using a higher spray velocity or larger diameter spray nozzle. The nozzle gas flow rate was not adjusted to increase the particle size.
TABLE 1Ledipasvir spray drying parameters on an AnhydroMS35 spray dryer using two liquid nozzles
| Parameter(s) | Test 1 | Test 2 | Test 3 | Test 4 |
| Batch size (g) | 100 | 250 | 250 | 4000 |
| Solids% | 20 | 20 | 20 | 20 |
| Feed rate (mL/min) | 30 | 40 | 40 | 40 |
| Spray nozzle (mm) | 1.0 | 1.0 | 1.2 | 1.2 |
| Nozzle air flow (kg/hr) | 6.0 | 6.0 | 6.0 | 6.0 |
| Spray chamber airflow (kg/hr) | 35.0 | 35.0 | 35.0 | 35.0 |
| Inlet temperature (. degree.C.) | 125 | 165 | 165 | 165 |
| Outlet temperature (. degree.C.) | 70 | 73 | 72 | 76 |
| PS d10/d50/d90Average value (μm) | 4/9/18/10 | 5/10/20/12 | 5/10/19/11 | 6/12/22/14 |
| LOD after spraying (%) | 5.56 | 4.86 | 4.29 | 3.42 |
TABLE 2 examples of Ledipasvir spray drying parameters using a hydraulic nozzle
| Parameter(s) | Test 1 |
| Batch size (g) | 200 |
| Solids% | 20 |
| Feed rate (kg/hr) | 178 |
| Feed pressure (bar) | 52 |
| Inlet temperature (. degree.C.) | 158 |
| Outlet temperature (. degree.C.) | 65 |
| PS d10/d50/d90Average value (μm) | 3/14/34 |
| LOD after spraying (%) | 0.6 |
Organic volatile impurities, including spray-dried solvent ethanol and residual acetone from ledipasvir acetone solvate, can be removed rapidly during the second drying at 60 ℃. Smaller scale production can be dried in trays. For larger scale batches, a double cone dryer or a stirred dryer may be used. Loss On Drying (LOD) is proportionately slower due to water and can subsequently be confirmed by karl fischer titration.
By 6 hours drying (or 8 hours on a larger scale), the residual ethanol was reduced to less than 0.5% w/w of the ICH guidelines. The ethanol content at the end of drying was 0.08% w/w and the residual acetone was 0.002%, indicating that the second drying process was sufficient to remove the residual solvent.
C. Preparation of tablets
i. Single layer sheet
Ledipasvir A solid dispersion of copovidone (1:1) is prepared by dissolving Ledipasvir and copovidone in ethanol and then spray-drying the mixture. Spray dried ledipasvir the copovidone solid dispersion was also dried in a second dryer. The amorphous solid dispersion containing ledipasvir was mixed with sofosbuvir and excipients and ground to facilitate uniform mixing. A co-blending or co-drying granulation process may be used. Co-blend granulation is a multi-step process involving dry granulation of each active ingredient with an excipient separately, followed by mixing the two granulation portions together. Co-dry granulation involves drying the granulation active ingredient and excipients together. The co-blending and co-drying processes have comparable physical and chemical tablet properties. Exemplary co-blended and co-dried formulations are shown in tables 3 and 4, listed below.
TABLE 3 representative compositional examples of Sofosbuvir/Ledipasvir co-dried (co-granulated) tablets of varying fill weights
TABLE 4 representative compositional examples of Sofosbuvir/Ledipasvir co-blended (double granulation) tablets
The granules are then mixed with a lubricant prior to tableting. The total weight of the resulting tablet core was 1000 mg.
The tablets were film coated to reduce photodegradation. The tablet weight increased by 3% after coating. The film coating material is polyvinyl alcohol-based coating. See table 5 for exemplary tablets.
TABLE 5 representative examples of compositions for tablets containing solid dispersions of Ledipasvir and Sofosbuvir
| Composition (I) | %w/w | Component weight (mg/tablet) |
| Sofosbuvir | 40.00 | 400 |
| Ledipasvir solid dispersion | 18.00 | 180.0 |
| Lactose monohydrate | 16.50 | 165.0 |
| Microcrystalline cellulose | 18.00 | 180.0 |
| Croscarmellose sodium | 5.00 | 50.0 |
| Colloidal silicon dioxide | 1.00 | 10.0 |
| Magnesium stearate | 1.50 | 15 |
| Total weight of tablet core | 100.0 | 1000.0 |
| Film coating | 3.00 | 30.029 --> |
| Pure water | -- | -- |
| Total weight of coated tablets | 1030.0 |
A bilayer tablet
Tablets containing a co-formulation of a solid dispersion comprising ledipasvir and crystalline sofosbuvir, in one layer for each active ingredient, may also be formulated as bilayer tablets. To prepare the bilayer tablets, ledipasvir copovidone (1:1) solid dispersions were prepared by dissolving ledipasvir and copovidone in ethanol and then spray drying the mixture. Spray dried ledipasvir the copovidone solid dispersion was further dried in a second dryer. Subsequently, the spray dried solid dispersion of ledipasvir copovidone is mixed with excipients. The mixture was milled and then blended with a lubricant prior to dry granulation. The ledipasvir granulate is mixed with an extragranular lubricant. Additionally the sofosbuvir drug substance is separately mixed with excipients and the mixture is then ground and mixed with a lubricant before dry granulation. The sofosbuvir granules were then mixed with the extra-granular lubricant. Finally, the sofosbuvir powder mix and the ledipasvir powder mix were compressed into bilayer tablet cores. The bilayer tablet cores are then coated prior to packaging. An example of a representative composition of a bilayer tablet containing a solid dispersion of ledipasvir and sofosbuvir is shown in table 6. In this table, the solid dispersion comprises ledipasvir to copovidone in a 1:1 ratio.
TABLE 6 representative compositional examples of bilayer tablets containing solid dispersions of ledipasvir and sofosbuvir
Example 3: ledipasvir single drug tablets and Ledipasvir/Sofosbuvir tablets with reduced PK, stability and dissolution properties, food effect and gastric acid inhibitor effect
Bioavailability of Ledipasvir single drug tablets
A series of in vivo experiments were performed to assess the potential benefits of this solid dispersion approach compared to conventional formulations, and the solid dispersion was optimized by identifying the optimal polymer type and relative polymer concentration within the dispersion.
In the pentagastrin-pretreated canine model, the preparation containing the free base amorphous form (4% w/w, 10mg amorphous free base tablet) and the preparation containing ledipasvirD-tartrate (5.85% w/w, 10 mgD-tartrate tablet) had equivalent bioavailability, both of which were conventional formulations, and the results are shown in table 7. The five gastrins are synthetic polypeptides that stimulate the secretion of gastric acid, pepsin and intrinsic factors.
TABLE 7 mean (RSD) pharmacokinetic parameters of Ledipasvir after oral administration of tablets, 25mg in beagle dogs (n ═ 6)
Since these formulations showed similar PK properties, while the individual properties of D-tartrate were superior to the free base amorphous form, the crystalline D-tartrate formulation was chosen for comparison with the amorphous solid dispersion composition. In these studies, 30mg tablets containing ledipasvir crystalline D-tartrate and 30mg or 90mg tablets containing ledipasvir amorphous solid dispersions were used. The results of the canine pharmacokinetics of the immediate release ledipasvir tablets selected to contain the ledipasvir solid dispersion are shown in table 8.
Table 8 mean (RSD) pharmacokinetic parameters of Ledipasvir after oral administration of Ledipasvir tablets in fasted beagle dogs (n ═ 6)
Amorphous solid dispersion tablets showed higher bioavailability and lower variability compared to the crystalline D-tartrate ledipasvir formulation. In pentagastrin-pretreated animals, an approximately 40% increase in exposure and a 2-fold decrease in variability was observed. More importantly, in famotidine-pretreated animals, up to a 3.5-fold increase in bioavailability was observed compared to D-tartrate tablets.
The bioavailability of the copolyvidone-based dispersion increased more than equivalent hypromellose-based formulations when spray dried at a ratio of API to polymer of 2:1 (30% and 22%, respectively). The bioavailability of the copovidone-based formulation was further increased as the polymer component was increased to the 1:1 ratio, resulting in a bioavailability of 35% in famotidine-pretreated dogs.
A 1:1 mixture of ledipasvir copovidone was chosen as the starting material for spray drying due to improved in vivo performance and acceptable stability and physical properties.
Formulations containing amorphous solid dispersions proved to be superior to formulations containing amorphous free base or D-tartrate. The bioavailability of the amorphous free base formulation was observed to be similar to that of the D-tartrate formulation. Additional data indicate a decrease in bioavailability when ledipasvir is administered with a gastric acid inhibitor (famotidine), indicating that the free base amorphous form of ledipasvir and the D-tartrate formulation exhibit adverse drug-drug interactions. Solid dispersions using spray drying and with hydrophilic polymers were identified as having acceptable stability, physical properties and in vivo performance. A rapidly disintegrating tablet was developed using a dry granulation process and common excipients. A bioavailability study comparing D-tartrate containing formulations with formulations containing amorphous solid dispersions shows that improved biopharmaceutical performance is observed in D-tartrate formulations and overcomes most of the negative drug-drug interactions with acid suppression therapy.
Bioavailability of Ledipasvir + Sofosbuvir tablets
The PK results for Sofosbuvir combined with ledipasvir (where ledipasvir and copovidone in the solid dispersion are 1:1) are shown in table 9, which indicates that there is no significant interaction between Sofosbuvir and ledipasvir.
TABLE 9 pharmacokinetic data for Sofosbuvir and Ledipasvir when administered alone or in combination with Sofosbuvir and Ledipasvir
Sofosbuvir plasma exposure was increased by-2.3 fold by ledipasvir. The effect of Ledipasvir on Sofosbuvir, which is known to be a substrate for P-gp, is probably due to the inhibition of P-gp. The increase in Sofosbuvir was considered insignificant because its exposure was low and transient compared to total drug related substance (DRM) exposure (DRM, calculated as the sum of AUCs per analyte, corrected for molecular weight). According to this calculation, the AUC of sofosbuvir and ledipasvir is only 5.7% of DRMAUC. Exposure of the major circulating metabolite of Sofosbuvir, metabolite II, was unaffected by ledipasvir administration, indicating no significant interaction between Sofosbuvir and ledipasvir.
Reduction of food Effect of Ledipasvir solid Dispersion and Ledipasvir/Sofosbuvir tablets
Conventional formulations of ledipasvir alone (not solid dispersions) have been shown to have negative food effects. Table 10 summarizes the PK parameters of ledipasvir following a single dose of 30mg ledipasvir administration under fasting and fed conditions. The PK profile of ledipasvir changes in the presence of food. In particular, a high fat diet may delay the absorption of ledipasvir, prolonging Tmax(TmaxMedian of 8 hours) and reduced plasma exposure of ledipasvir (C)max、AUClastAnd AUCinfThe average value is respectively reduced by about45%)。
Table 10: plasma LedipasvirPK profile following single dose administration of Ledipasvir in a food intake regime
Table 11 shows the GLSMs ratio (30 mgledipasvir under fasted/30 mgledipasvir under fed) for each of the major PK parameters.
Table 11: statistical evaluation of food Effect of LedipasvirPK parameters
Similar median ledipasvir half-life (t under fasting conditions) was observed when administered either in fasting or fed conditions1/239.82 hours, 36.83 hours under fasting conditions), indicating that food can reduce its bioavailability by reducing solubility and/or absorption of ledipasvir.
Since ledipasvir has been shown to have a negative food effect, the food effect was examined for compositions containing sofosbuvir and ledipasvir, which are solid dispersions formed with copovidone (1: 1). See table 12 for these results. The food can slow the absorption rate (T) of sofosbuvirmaxMedian value: 1.00 and 2.00 hours), but only slightly altered bioavailability, which was demonstrated by a 2-fold or less increase in plasma exposure of sofosbuvir and sofosbuvir metabolite I. As for the metabolite II of sofosbuvir, C was observed when sofosbuvir was administered with foodmaxThe decrease was about 20-30% with no change in AUC. Of Sofosbuvir metabolite IIAUCThe% GMR and the associated 90% CI (fed/fasted treatment) lie within the equivalent limits of 70% to 143%. Due to sofosbuvir metabolite IICmaxThe decrease in AUC was slight and the AUC parameters met the equivalence criterion, so the effect of food on sofosbuvir metabolite II was considered insignificant.
Similar plasma exposure (AUC and C) of ledipasvir was achieved with administration of ledipasvir under fasting or fed conditionsmax). The% GMR and the associated 90% CI (fed/fasted treatment) lie within the equivalent limits of 70% to 143%. Ledipasvir has been previously observed to have a "negative" dietary effect when administered alone (as an amorphous free base rather than as a solid dispersion), but the pharmacokinetics of ledipasvir (amorphous solid dispersion; copovidone (1:1)) administered in combination with sofosbuvir is not affected by diet. Thus, the combination of sofosbuvir and ledipasvir can be administered without regard to food.
Table 12: pharmacokinetic data for Sofosbuvir, Sofosbuvir metabolites I and II, and Ledipasvir when administered in fasted or medium-fat or high-calorie/high-fat diets
Reduction of gastric acid inhibitor Effect in Ledipasvir/Sofosbuvir tablets
30mg of ledipasvir alone in conventional formulations (as D-tartrate) and as a solid dispersion have demonstrated reduced bioavailability when administered with certain gastric acid inhibitors; most notably proton pump inhibitors (PPI's, e.g., omeprazole), but also histamine-2 antagonists (H2 RA's, e.g., famotidine, data not shown). Table 12A summarizes the PK parameters of ledipasvir given with 90mg ledipasvir solid dispersion together with conventional single drug tablets of ledipasvir 30mg, solid dispersion tablets of ledipasvir (ledipasvir: copovidone 1:1)30mg, and sofosvir/ledipasvir FDC tablets with or without copovidone 1:1 of omeprazole. The bioavailability of ledipasvir as a single drug tablet when administered with omeprazole is reduced by a factor of about 2; however, when ledipasvir was administered with omeprazole as part of the sofosbuvir/ledipasvir fdc tablets, the exposure to ledipasvir (AUC and Cmax) was not significantly reduced compared to when the sofosbuvir/ledipasvir fdc tablets were not administered with omeprazole.
Table 12A: pharmacokinetic data for Ledipasvir when Ledipasvir single tablets or Sofosbuvir/Ledipasvir tablets are administered with omeprazole or alone
Dissolution rate of Ledipasvir/Sofosbuvir tablets
Dissolution studies were conducted comparing sofosbuvir400mg/ledipasvir90mg tablets (ledipasvir: copovidone (1: 1)). sofosbuvir ledipasvir tablets (LOT1-5) showed that the two tablet formulation had more than 85% of the sofosbuvir (FIG. 5) and ledipasvir (FIG. 6) dissolved in 30 minutes. These results are shown in fig. 5 and 6.
Example 4: stability of Sofosbuvir/Ledipasvir co-formulation
The compatibility of the sofosbuvir anhydrous crystal bulk drug and ledipasvir, namely the copovidone solid dispersion is evaluated. A mixture of sofosbuvir and ledipasvir, copovidone (1:1) was prepared in a ratio representing the final 400mgsofosbuvir/90mgledipasvir tablet. The mixture was pressed into pellets and placed in a stability chamber at 40 ℃/75% RH and 60 ℃/ambient humidity and tested after two and four weeks storage in open glass vials. The results summarized in table 13 indicate that no degradation was observed for either sofosbuvir or ledipasvir, indicating chemical compatibility between sofosbuvir and ledipasvir, a copovidone solid dispersion.
Table 13: specification and impurity content of Sofosbuvir and Ledipasvir solid Dispersion mixture stored at 40 ℃/75% RH and 60 ℃
Example 5: therapeutic efficacy of Sofosbuvir/Ledipasvir/ribavirin treatment in HCV infected patients
The combination of sofosbuvir, ledipasvir and ribavirin or the combination of sofosbuvir and ribavirin is used to treat patients infected with HCV. Patients for study included those who were naive, i.e., not previously treated for HCV, and those who were non-responsive, i.e., not previously treated for HCV, but who did not respond to the treatment. Standard doses of each drug (e.g., 90mg ledipasvir, 400mg sobossbrevir, and 1000mg ribavirin) were administered to the patient for 12 weeks. HCV RNA was tested during treatment and the persistent viral response (SVR) was tested after treatment was stopped. At four weeks of treatment, the HCV RNA test results reached a limit of detection (LOD of 15IU/mL) in almost all patients, and at the end of treatment, 100% of the patients reached HCV RNA levels below LOD (Table 14).
Table 14: patients with HCVRNA below detection limits over time
Surprisingly, 100% of patients receiving the combination therapy of sofosbuvir, ledipasvir and ribavirin achieved a sustained viral response at weeks 4 and 12 after treatment. In contrast, only 88% of patients treated with sofosbuvir and ribavirin received SVR at week 4 post-treatment, and only 84% of patients treated with sofosbuvir and ribavirin received SVR at week 12 post-treatment, and 10% of non-responders (table 15).
Table 15: sustained viral response
These results are graphically depicted in FIGS. 7A-D, indicating that 100% SVR was obtained at weeks 4 and 12 with the addition of ledipasvir to the treatment regimen. Example 9 below shows that a treatment regimen of less than 12 weeks (i.e., a treatment regimen of about 8 weeks or 6 weeks) can also achieve similar results from treatment regimens of sofosbuvir and ledipasvir without the addition of ribavirin.
Example 6 stability of SOF400mg/Ledipasvir90mg fixed dose combination tablets
This example summarizes the physicochemical stability of packaged Sofosbuvir (SOF)400mg/ledipasvir90mg blue film-coated Fixed Dose Combination (FDC) tablets in the presence of a desiccant at 25 ℃/60% Relative Humidity (RH) and 40 ℃/75% RH. The ledipasvir portion of the tablet contains ledipasvir copovidone in a ratio of 1: 1. In addition, the chemical and physical stability of SOF/ledipasvirFDC tablets was evaluated in an open environment at 40 ℃/75% RH for a period of up to 4 weeks.
The physicochemical properties evaluated included the character, potency, formation of degradation products, dissolution rate and moisture content. The physical stability of the tablets in the absence of a desiccant was evaluated after 24 weeks using FT-raman spectroscopy and a modified differential scanning calorimeter (mDSC).
SOF400mg/ledipasvir90mg blue film coated FDC tablets showed good stability at 25 ℃/60% RH and 40 ℃/75% RH in the presence of 0, 1 and 3g of desiccant for up to 24 weeks. Significant changes in potency, impurity levels or dissolution rate were observed. However, a photodegradation product of ledipasvir was present at 0.1% under all conditions. FT-raman analysis of the tablets stored without desiccant showed no detectable crystallization after 24 weeks.
Method and material
Material
Table 16 lists the physicochemical properties of the SOF drug substance and ledipasvir solid dispersions used to produce the tablets. The amounts of SOF drug substance and ledipasvir solid dispersions were adjusted according to their respective drug content coefficients (DCF) with concomitant adjustment of the amount of lactose monohydrate. The DCF for the SOF and ledipasvir solid dispersion powders 50% w/w were 0.997 and 0.497, respectively (0.994 when the amount of copovidone was adjusted).
TABLE 16 physicochemical Properties of SOF drug substance and Ledipasvir solid Dispersion 50% w/w bulk powder for the production of SOF400mg/Ledipasvir90mg film coated FCD tablets
Device
The main equipment used to produce SOF400mg/Ledipasvir90mg film coated FDC tablets included a 12 qt.V-blender, a sizer equipped with a 0.094 grid screen (Comil197S, Quadro, Waterloo, Canada), a roller tablet/granulator equipped with a 1.0mm abrasive screen and smooth/smooth roller mechanism (MiniPador, Gerteis, Jona, Switzerland), a 12-position rotary disk tablet (XM-12, Korsch, Berlin, Germany) and a tablet coater (LabCoat, O' Hara technologies Inc., Ontario, Canada). A diamond tablet die (elizabethcrarbidedie, inc., McKeesport, PA, USA) consists of a diamond, standard concave D-die, measuring 0.7650inx0.4014in (19.43mmx 10.20mm). The cores were coated using a 15 inch perforated pan film coater.
Container seal
Sofosbuvir/Ledipasvir FDC tablets were packaged in a 100mL white High Density Polyethylene (HDPE) bottle. Each bottle contained 30 tablets and 0, 1 or 3g silica gel desiccant cartridges or pouches and polyester packaging. Each bottle was sealed with a white continuous threaded child-resistant screw cap and an electromagnetic induction sealed aluminum-faced liner.
A selected number of bottles were held open and packaged without desiccant to evaluate the physical and chemical stability at 40 ℃/75% RH under accelerated heat and humidity conditions.
General study design
The solid state and chemical stability of the packaged batches were evaluated in the following combinations:
1) at 25 ℃/60% RH and 40 ℃/75% RH with desiccant. The samples were stored under sealed conditions for a minimum of 24 weeks.
2) Open conditions at 40 ℃/75% RH for up to 4 weeks.
Samples were taken at predetermined time points. Chemical stability tests were performed for traits, efficacy, degradation product formation, dissolution rate and moisture content. Other physical stability analyses were also performed to monitor potential crystallization and phase separation.
Evaluation of physical stability
Physical stability testing includes trait and FT-Raman analysis. The compressed film coated tablets were visually inspected to identify changes in tablet color and coating integrity. FT-raman spectroscopy was used to detect potential crystalline ledipasvir (form III) in film coated tablets.
The tablets were visually inspected for changes in properties at all time points and storage conditions. In contrast, FT-Raman analysis was performed only on tablets at 24 weeks (25 ℃/60% RH and 40 ℃/75% RH) under 0g of desiccant.
Traits
The physical integrity (i.e., color, shape, coating integrity and dishing) of the tablets was checked at all time points.
FT-Raman analysis
FT-Raman testing was performed. Formation of crystalline ledipasvir (form III) was detected using FT-Raman spectroscopy to analyze 24 week SOF/ledipasvir film coated FDC tablets stored in sealed containers at 25 ℃/60% RH and 40 ℃/75% RH. Briefly, using XatoTMThe tablet was carefully coated with a knife and then ground using a pestle. The tablet powder was then filled into cups and the spectra collected using a back-scattering geometry.
Evaluation of chemical stability
Chemical stability analyses were performed including water content, efficacy, formation of impurities/degradation products and dissolution rate as measured by karl fischer (kf) method.
Water content of KF
The water content of SOF400mg/ledipasvir film coated FDC tablets was reported according to USP <921 >.
Potency and impurity/degradation product formation by UPLC assay
A sample solution of 10 tablets of the composition was analyzed according to STM-2542[5], from which the efficacy and degradation product formation of SOF400mg/ledipasvir film-coated FDC tablets were evaluated. The concentrations of the control standards for SOF and ledipasvir were 2.0mg/mL and 0.45mg/mL, respectively. The specification and degradation product content of SOF and ledipasvir were determined by UPLC at 262nm and 325nm wavelength, respectively, using external standard and area normalization methods.
Dissolution method
The SOF400mg/ledipasvir film coated FDC tablets were subjected to dissolution testing. A USP2 type dissolution apparatus and 900mL dissolution media were used, with a paddle speed of 75 rpm. The medium was 1.5% polysorbate 80 in 10mM potassium phosphate buffer, pH6.0, and the temperature was maintained at 37 ℃ during the analysis. The extent of SOF and ledipasvir release over time was monitored by area normalization using UPLC and external control standards at a wavelength of 250 nm.
Results
A. Physical stability
A1. Traits
Visual inspection of the samples at all stability conditions and desiccant levels at all time points revealed blue-like, diamond-shaped film coated tablets.
FT-Raman analysis
The tablets were stored without desiccant for 24 weeks, after which the powder was extracted and subjected to FT-raman analysis. The% crystallinity calculated using the PLS model showed no evidence of crystalline ledipasvir (form III) exceeding 3% LOD under all storage conditions. This is consistent with the results for the original sample (t ═ 0), in which ledipasvir (form III) is also lower than LOD. Spectra of selected samples are seen from 1577cm-1To 1514cm-1The baseline is adjusted artificially according to the transparency. This region is one of four spectral regions used by the PLS model to estimate% ledipasvir (form III) in the tablet.
The top two spectra in the figure (used as a standard for the PLS model) are from a tablet of crystalline ledipasvir (form III) spiked at 10% w/w and 3% w/w. Is connected withThe two spectra represent compressed tablets stored at 40 ℃/75% RH and 25 ℃/60% RH for 24 weeks. The last spectrum represents the initial time point (t ═ 0). Ledipasvir (form III) at 1552cm-1There is a distinct peak which is clearly visible in the labeled tablet and the intensity increases from 3% to 10%. The strength of the pressed samples stored for 24 weeks in this area did not increase compared to the t-0 samples, indicating no change in crystallinity. Ledipasvir (form III) in the T-0 and 24 week samples was below the amount present in the spiked 3% form IIIledipasvir tablets, the current detection limit of this analytical technique.
B. Chemical stability
B.1KF Water content
The water content of the pressed samples stored for 4 weeks under open conditions increased from 2.28% to 5.23%. The moisture content of the pressed samples stored without desiccant, 1g desiccant and 3g desiccant at 25 ℃/60% RH dropped to 1.91%, 1.58% and 1.65%, respectively. The moisture content of the pressed samples stored without desiccant, 1g desiccant and 3g desiccant at 40 ℃/75% RH dropped to 2.03%, 1.79% and 1.46%, respectively.
B.2 potency and impurity/degradation product formation
The efficacy and impurity/degradation product content of SOF400mg/ledipasvir90mg film coated FDC tablets were determined at 25 ℃/60% RH and 40 ℃/75% RH. A representative chromatogram of a stable sample stored at 40 ℃/75% RH was obtained. The data show that SOF and ledipasvir are chemically stable in SOF400mg/ledipasvir90mg film coated FDC tablets stored at 25 ℃/60% RH and 40 ℃/75% RH for 24 weeks. The labeled specifications for SOF and ledipasvir remained unchanged at 25 deg.C/60% RH and 40 deg.C/75% RH.
Dissolution rate
The dissolution profiles of SOF and ledipasvir in a SOF400mg/ledipasvir90mg film coated FDC tablet were obtained. At the 24 week time point, the SOF dissolution range in the tablet at 45 minutes was 99% to 100% and ledipasvir was between 99% to 98% at all tested desiccant levels of 25 ℃/60% RH and 40 ℃/75% RH.
Based on the foregoing results, this example shows that SOF400mg/ledipasvir90mg film coated FDC tablets maintain good stability for up to 24 weeks at 25 ℃/60% RH and 40 ℃/75% RH at 0, 1g, and 3g of desiccant. In addition, FT-raman analysis did not detect crystalline ledipasvir (form III) after 24 weeks of storage.
Example 7 formulation development of SOF400mg/Ledipasvir90mg Fixed Dose Combination (FDC) tablets
This example shows the development of SOF400mg/ledipasvir90mg Fixed Dose Combination (FDC) tablets containing ledipasvir copovidone (1: 1). This development is expected to have a number of difficulties based on the existing formulation of each individual drug, one of which is expected to be poor powder flow, and others associated with a heterogeneous mixture.
Three tablets were tested, including (1) a single layer co-granulated tablet, (2) a single layer co-blended tablet, and (3) a bilayer tablet. In all three formulations, SOF was anhydrous crystalline form II and ledipasvir was an amorphous solid dispersion (ledipasvir: copovidone (1: 1)).
Formulation (1) is typically associated with the highest risk of drug-drug interaction, but is most cost-effective during production. Bilayer formulation (3) in contrast, has the lowest risk of drug-drug interaction.
The dissolution capacity of the preparation is tested by using a dissolution medium comprising 10mM phosphate buffer, pH6.0 (1.5%)80). As shown in fig. 8A-B, all three formulations had comparable dissolution capabilities, similar to the single drug control.
Pharmacokinetic (PK) performance was also tested for each formulation. Plasma concentrations of SOF/ledipasvirFDC and control tablets were determined following oral administration to fasted dogs (100mg/22.5mg fixed/dog). The PK results are shown in table 17 below.
TABLE 17 pharmacokinetic Performance of the formulations in famotidine-pretreated dogs
From these results, monolayer co-granulated sheets were selected for further analysis. The composition of this formulation is shown in table 18.
TABLE 18 composition of SOF400mg/Ledipasvir90mgFDC tablets
A clinical study of bioavailability of this formulation was performed in 24 healthy patients in fasted conditions, using single drug tablets as controls. See table 19 for results.
TABLE 19 bioavailability of SOF/Ledipasvir fixed dose combination tablets and single drug tablets
Thus, these results indicate bioequivalence of SOF/ledipasvir fixed dose combination (co-granulated) tablets and single drug tablets.
EXAMPLE 8 solubility study of amorphous Ledipasvir
This example examines the physicochemical properties of the different ledipasvir forms, including amorphous and crystalline free bases, solvates and salts, in terms of solubility.
A. Materials and methods
pH-solubility curve
The aqueous solubility of ledipasvir amorphous free base was determined over a pH range of 1 to 10. Excess solid ledipasvir was added to a series of pH adjusted aqueous solutions (HCl or NaOH titration) and stirred at room temperature for 48 hours. The suspension was then filtered through a regenerated cellulose syringe filter. Measuring the pH of the supernatant with 50:50H2The supernatant was diluted with an appropriate amount of O + 0.1% TFA ACN and analyzed for ledipasvir content by HPLC-UV method.
Simulating solubility in intestinal media
The solubility of ledipasvir amorphous free base was evaluated in three simulated intestinal fluids at ph6.5 or ph5.0, simulated intestinal bile salts, and a lecithin mixture (SIBLM) at ph 6.4. An excess of solid ledipasvir was added to the respective SIFs and stirred at room temperature for 48 hours. The resulting suspension was then filtered through a regenerated cellulose syringe filter. With 50:50H2The supernatant was diluted with an appropriate amount of O + 0.1% TFA ACN and analyzed for ledipasvir content by HPLC-UV method.
Solubility of auxiliary materials
The solubility of ledipasvir amorphous free base and ledipasvir crystalline D-tartrate is determined in a range of pharmaceutically acceptable solvents including co-solvents, surfactants, fatty acids, triglycerides or mixtures thereof. The material was weighed into a scintillation vial and stirred at room temperature for up to 48 hours. In many cases, the solubility is higher than the amount of solid used in the sample, and therefore, if the concentration is not quantitatively determined by HPLC-UV, many results are reported as "higher" or "higher or equal".
In addition, the water solubility was measured as a function of time in the presence of 0.1% w/w surfactant and polymer at pH2(50mM citrate) and pH5(50mM citrate). Ledipasvir crystalline form (acetone solvate form II; amorphous FB form III; D-tartrate) and amorphous form were evaluated to identify differences in dissolution behavior. Adding excess solids to the aqueous buffer; samples were taken at predetermined times (2, 5, 8, 10, 15, 20, 30, 45, 60 minutes and 24 hours), filtered through a regenerated cellulose filter, diluted and tested for concentration by HPLC-UV method.
B. Results
Solubility and dissolution rate
The pH-solubility curves of all existing ledipasvir forms were examined at room temperature and are shown vividly in fig. 9. The smooth part of the solubility curve (pH >5) represents the intrinsic water solubility of the free base. The aqueous solubility of ledipasvir increases significantly when the pH of the solution is below the pKa of the ionizable group. All forms lose crystallinity and revert to amorphous free base in aqueous solution, once exhibiting similar steady state water solubility. However, the dissolution properties are form dependent and will be described in detail below.
Ledipasvir amorphous free base
The intrinsic solubility of Ledipasvir amorphous Free Base (FB) is approximately 0.04 μ g/mL. Under acidic conditions, the solubility increased to 1mg/mL at pH2.3 and peaked at about 7mg/mL at pH1.6, as shown in Table 20 and FIG. 9. The solubility of ledipasvir in simulated intestinal fluid is governed by both the pH of the medium and the presence or absence of bile salts and lecithin. The solubility was 0.025mg/mL in fasted state simulated intestinal fluid (FaSSIF) at pH6.5 and at room temperature, and was about 10-fold increased to 0.232mg/mL in a simulated bile and lecithin mixture (SIBLM, pH6.5) due to increased concentrations of bile salts and lecithin. A similar increase in solubility of 0.230mg/mL was observed in fasted-state simulated intestinal fluid (FeSSIF, pH5) containing a mixture of bile salts and lecithin below SIBLM. The main reason for the increased solubility in this mixture is the ionized state of the molecules at pH5.
TABLE 20 solubility of Ledipasvir amorphous free base as a function of pH at room temperature
| Aqueous medium | Solubility (mg/mL) |
| Aqueous, pH1.6 (HCl) | 6.855 |
| Aqueous, pH2.3 (HCl) | 1.096 |
| Aqueous, pH 3.1(HCl) | 0.0132 |
| Aqueous, pH 4.1(HCl) | 0.00011 |
| Aqueous, pH 5.5(HCl) | 0.00003 |
| Aqueous, pH 6.2 (unchanged) | 0.00003 |
| Aqueous, pH 7.2(NaOH) | 0.00001 |
| FaSSIF1pH=6.5 | 0.025 |
| FeSSIF2pH=5.0 | 0.230 |
| SIBLM3pH=6.4 | 0.232 |
1FaSSIF is an aqueous solution containing 3mM sodium taurocholate and 0.75mM lecithin, the pH being adjusted to 6.5 with phosphate buffer and the ionic strength being adjusted to 0.15M with NaCl.
2FaSSIF is an aqueous solution containing 15mM sodium taurocholate and 3.75mM lecithin, the pH being adjusted to 6.5 with phosphate buffer and the ionic strength being adjusted to 0.15M with NaCl.
3The SIBLM is an aqueous solution containing 30mM sodium glycocholate, 30mM sodium chenodeoxycholate, 15mM sodium glycodeoxycholate (sodium glycodeoxycholate), 10mM sodium taurocholate, 10mM sodium taurochenodeoxycholate (sodium taurochenodeoxycholate), 5mM sodium taurochenodeoxycholate (sodium taurochenodeoxycholate), 50mM sodium chloride and 11mM lecithin, the pH being adjusted to 6.4 with a phosphate buffer, and the ionic strength being adjusted to 0.15M with NaCl.
The dissolution rate of ledipasvir amorphous free base was also tested at pH3 and 6. At pH3, the dissolution rate of the amorphous free base form was faster than the dissolution rates of the crystalline free base and acetone solvate forms. However, at pH6, all free base forms exhibited similar dissolution rate profiles.
As shown in Table 21, ledipasvir amorphous free base is readily soluble in ethanol and other organic solvents such as propylene glycol and PEG400 ((R))>500 mg/mL). Its solubility in surfactant (e.g., polysorbate 80, CremophorEL, glyceryl oleate (Labrasol)) and lipid mixtures is above 200 mg/mL. Its solubility in oleic acid and caprylic acid is higher than 500 mg/mL. The solubility of Ledipasvir in short chain triglycerides (SCTs, tributyrin) is limited to 20mg/mL, decreasing to below 1mg/mL in long chain triglycerides (LCTs, soybean oil). It was in the selected vehicle for toxicological studies: 45% of propylene glycol and 15% of caprylic/capric polyethylene glycol glyceride (SolutolHS)) And a solubility in 40% water (adjusted to pH2.5 by HCl) of 25 mg/mL.
TABLE 21 solubility of Ledipasvir free base form and LedipasvirD-tartrate in organic solvents and adjuvants at room temperature
1.RSSEDDS: 10% ethanol, 10% PG, 40% polyethylene glycol stearate-15 (SolutolHS-15), 40% caprylic/capric polyethylene glycol glyceride
As shown in table 22, the nonionic weak surfactants generally increased solubility of ledipasvir at pH2 and pH5. Similar effects can be observed for non-ionic polymers, but to a lesser extent. Sodium Lauryl Sulfate (SLS) is an anionic surfactant that increases the solubility of ledipasvir at pH5. However, under acidic conditions (pH2), a significant decrease in solubility was observed in the presence of SLS. This result is consistent with the phenomenon that weakly basic compounds with low intrinsic water solubility are presumed to form insoluble propionate lauryl sulfate.
TABLE 22 solubility of Ledipasvir amorphous free base in surfactant or polymer adjuvant diluted in aqueous medium at pH2 and 5 and room temperature
Ledipasvir crystal acetone solvate (Ledipasvir-03)
Ledipasvir acetone solvate (Ledipasvir-03) has a steady state solubility similar to the other forms. Ledipasvir-03 had the slowest dissolution rate of all the forms tested. At pH6, its dissolution rate did not differ from the other forms due to very poor intrinsic solubility (<0.1 μ g/mL).
Ledipasvir-03 is soluble in a number of organic and pharmaceutically acceptable solvents with a solubility comparable to that listed for the Ledipasvir amorphous free base, as also shown in table 21.
Ledipasvir crystalline free base (form III)
Ledipasvir crystalline free base form III has a similar steady state solubility as the other forms (fig. 9). This form dissolves slower than the amorphous free base, but faster than ledipasvir-03. Its dissolution at pH6 did not differ from the other forms due to very poor intrinsic solubility (<0.1 μ g/mL). Their solubility in a wide range of organic vehicles was not explored, but would be expected to be similar to the other free base forms.
Ledipasvir crystal D-tartrate (Ledipasvir-02)
Ledipasvir crystalline tartrate (Ledipasvir-02) exhibited a steady state solubility similar to the other forms (FIG. 9). The dissolution behavior of ledipasivr-02 is improved compared to all free base forms. At pH3, it showed an initial dissolution rate of about 5 to 10 times faster than the free base form, with the amount of ledipasvir in solution after 60 minutes approximately doubling compared to the amorphous form. The increased dissolution rate was also evident at pH6. However, the solubility values resulting from the rapid dissolution of this salt over several minutes at this pH are equivalent to the other forms.
Ledipasvir-02 is insoluble in a variety of organic media as shown in Table 21. The maximum solubility of Ledipasvir-02 in any organic vehicle is 20mg/mL in ethanol; this limits the use of ledipasvir-02 in a formulation or process requiring solubility in organic media.
Ledipasvir has low water solubility and high permeability, and is a BCS2 compound. The data presented in this example show that in water, all forms of ledipasvir: amorphous free base, crystalline free base acetone solvate (ledipasvir-03), crystalline anhydrous free base (form III) and crystalline D-tartrate (ledipasvir-02), are converted to amorphous free base with similar steady state water solubility. The water solubility of Ledipasvir is below 0.1. mu.g/mL in the neutral form (pH >5), but is substantially increased under acidic conditions due to protonation of the two basic moieties. The aqueous dissolution rate of Ledipasvir amorphous free base is faster than the crystalline free base form. However, the dissolution rates of all the free base forms were slower than that of crystalline D-tartrate (ledipasvir-02). Ledipasvir-02 has improved wettability in an aqueous medium. Both crystalline and amorphous ledipasvir free base forms are highly soluble in a range of co-solvents and surfactants. In contrast, ledipasvir-02 is poorly soluble in organic excipients, a property that may limit its application.
The use of ledipasvir amorphous free base in phase I clinical studies has identified bulk drug production as a key limitation to this format. Ledipasvir crystalline D-tartrate (Ledipasvir-02) was then identified as a major salt and form for phase 2 clinical studies, but its poor solubility in organic excipients limited its use in unconventional formulations. The development of spray-dried dispersion formulations using crystalline ledipasvir acetone solvate (ledipasvir-03) supports future clinical studies because it has better solubility in organic solvents and excipients than crystalline ledipasvir-tartrate and improved productivity compared to other free base forms.
Example 9: therapeutic efficacy of Sofosbuvir and Ledipasvir with or without fixed doses of ribavirin for HCV infected patients
Patients with HCV infection were treated with sofosbuvir and ledipasvir with or without fixed doses of ribavirin. Patients used in the study included those naive (no cirrhosis), i.e., patients who had not previously been treated with HCV, and those non-responders (with and without cirrhosis), i.e., patients who had previously been treated with HCV but who did not respond to treatment, in which treatment failed Protease Inhibitor (PI). Treatment was 6, 8 and 12 weeks in naive patients and 12 weeks in non-responders.
Study 1
Cohort 1 of study 1 included treatment naive, genotype 1 patients without cirrhosis. Patients were randomized 1:1:1 into three groups to receive 1) a fixed dose combination of SOF/ledipasvir for 8 weeks, 2) ribavirin for 8 weeks in combination with a fixed dose of SOF/ledipasvir, or 3) a fixed dose combination of SOF/ledipasvir for 12 weeks (figure 10).
Group 2 of study 1 included patients who had been experienced with protease inhibitor therapy, genotype 1 patients (50% with compensatory cirrhosis before failure of protease inhibitor therapy). Patients were randomized to receive 12 weeks: 1) SOF/ledipasvir fixed dose combination or 2) SOF/ledipasvir fixed dose combination with ribavirin (FIG. 10). In cohort 2, patients were unable to stop dosing before treatment due to adverse events.
In study 1, the inclusion criteria were broad, with no upper limit on age or BMI. Platelet not less than 50,000/mm3. The demographic data for study 1 is seen in table 23 below.
TABLE 23 demographic data
Among 100 patients participating in study 1, 97% achieved a sustained viral response. In failed patients, two relapses (one from group 1 (i.e., SOF/Ledipasvirx8 weeks) and one from group 4 (i.e., SOF/Ledipasvirx12 weeks)). However, patients who lost follow-up reached SVR at 8 weeks and declined on subsequent visits.
In study 1, cohort 1 (i.e., naive, cirrhosis-free patients), 58 of 60 patients treated for 8 or 12 weeks achieved SVR. In cohort 2 of study 1 (i.e., the treatment experienced, PI failure patients), 39 of40 patients who were 12 weeks of treatment achieved SVR 12. All 21 patients with cirrhosis reached SVR12 (fig. 11).
In study 1, seven of nine patients with NS5A tolerance-related variants (RAVs) achieved sustained viral responses. In addition, a sustained viral response was achieved in all patients with the NS3/4A tolerance-related variant. Interestingly, the S282T mutation and multiple NS5ARAVs were detected in patients who failed in group 1 when relapsed (table 24). See table 25 and table 26 for safety summary and adverse effect classification, respectively.
TABLE 24 tolerance assay
Number of patients in group 2 who had been exposed to protease inhibitors
TABLE 25 safety overview
Gastric ulcer, spondylotic fracture
Delirium, suicide and souring
TABLE 26 adverse events (. gtoreq.5% of all patients)
Study 2
In study 2, the naive received a fixed dose combination of SOF/ledipasvir and ribavirin, whereas the previous non-responder was randomized to receive twelve weeks: 1) SOF/ledipasvir fixed dose combination or 2) SOF/ledipasvir fixed dose combination with ribavirin, wherein these non-responders all had cirrhosis.
Results
Of the 144 patients treated in study 1 and study 2, 136 (94%) reached SVR at four weeks post-treatment. Of the 85 naive patients in both studies, 3 of 25 failed to achieve SVR after 6 weeks of SOF/ledipasvir fixed dose combination with ribavirin treatment, and 100% (60/60) of the patients achieved SVR after 8 or 12 weeks of SOF/ledipasvir combined with or without ribavirin fixed dose. Of the 59 patients with treatment experience in these two studies, three patients with cirrhosis relapsed after receiving 12 weeks of treatment with SOF/ledipasvir in combination with no fixed dose of ribavirin. In contrast, no viral treatment failure was observed in the SOF/ledipasvir and ribavirin fixed dose combination treatment groups, but two patients in these groups lost follow-up. SOF/ledipasvir treatment with or without fixed doses of ribavirin is well tolerated with little SAEs and adverse events.
Conclusion
SOF/ledipasvir fixed doses combined with +/-ribavirin may be given for a minimum of 8 weeks to treat naive patients without cirrhosis. Patients with treatment experience, even with cirrhosis, can achieve high SVR rates after 12 weeks of treatment with SOF/ledipasvir with or without fixed doses of ribavirin.
Example 10: therapeutic efficacy of multiple anti-HCV combination therapy in patients with chronic hepatitis C virus infection
To evaluate the safety, tolerability and efficacy of SOF alone or in combination with ledipasvir and/or compound J for 4 to 12 weeks in HCV patients, HCV patients were dosed as shown in table 27.
TABLE 27 dosing regimens
The primary analysis set for safety analysis included patients who received a minimum of one study drug administration. The treatment data was analyzed and data collected from the first administration of the study drug to the last administration date of the study drug plus 30 days was defined as treatment data. The patient receiving the study medication, rather than the patient, dispensing the medication, is analyzed based on the study medication received.
The assay setup for antiviral activity assays included patients who participated in the study and received at least one study drug administration.
The pharmacokinetic analysis setup included all patients who participated in and received at least one study drug administration.
The patient began study treatment after confirming a qualified and well understood rest of the study on day 0 and signing up specific informed consent for treatment grouping if not done previously. Blood was drawn for HCV viral load, study drug levels, lipid level studies, and also for immunological studies if no blood was drawn during screening and blood was preserved prior to dosing as part of the screening consent. Pregnancy tests were performed on women with childbearing potential and must be negative on day 0 prior to study drug administration. The patient may be asked to fill out a baseline compliance questionnaire with an electronic vial cap placed over all study vials that records vial opening. Help is provided to fill out the questionnaire if necessary. Patients in groups B and H were provided with diaries to record gastrointestinal side effects on day 0, week 2, and week 4 (group B only).
At the time of arrival at the clinic for the scheduled study visit, the patient's vital signs are measured, the woman is subjected to pregnancy tests (if planned and fertility is likely), clinical laboratory blood draw tests and review of study limits.
At each scheduled study visit (excluding days 1, 3,5, 10, 2, 3,6 (not applicable to group F, G or H) or weeks 2 and 8 after treatment (these two were used only for laboratory blood draws)), the patient will be asked the health status and use of any concomitant medications since the previous study visit. Vital signs, body weight detection and examination were performed according to the study procedure. A complete list of the study steps and laboratory examinations to be performed is seen in the test plans below. In addition, patients may be observed for grade 3 or 4 adverse events or any unexpected adverse event or possible toxicity at the time of the unscheduled visit.
The patient may be asked to fill out a follow-up compliance questionnaire, with an electronic cap recording vial opening on day 7 (group a), week 4 (group a), week 6 (groups B and C), week 8 (group a) and week 12 (group a). Assistance may be provided to fill out the questionnaire if desired.
Patients of groups B and H will be required to carry their side effect diary for visit on weeks 2,4, 6 (group B only).
Some visits may have a small amount of flexibility depending on when they are needed. Visits made during the period of time that the patient receives study medication are very limited in flexibility, as they are very frequent, skipping a visit at this stage may be considered a missing visit. The window phase of the access plan is seen in table 28.
TABLE 28 Window phase of visit plan
At four week visit, hcv rna was available to determine whether the criteria for treatment cessation based on viral response were met. Patients who failed to achieve >2log10HCV RNA reduction at this time (unless >2log reduction would be below LLOQ) should be withheld unless otherwise decided upon by review of PI/LAI/applicant's medical monitor (see 9.3.1).
At the end of the study group-determined treatment period, the patient may stop the administration of SOF and ledipasvir, compound E and/or compound J. In addition, if patient participation has ceased prior to the scheduled study drug administration period, an end-of-treatment assessment may be made at any end-of-treatment visit. An alternative study liver biopsy for study purposes may be performed in up to 10 patients per study group at this time. This additional liver biopsy data will be used to explore liver hcv rna sequence analysis. If the patient is taking an alternative study liver biopsy, they may complete a safety laboratory check as prescribed prior to surgery and imaging. Patients with HCVVL < LLOQ may receive education on how to avoid HCV re-infection.
All patients evaluated for a sustained viral response at 12 weeks after the end of treatment visit. Patients with HCVVL < LLOQ may receive education on how to avoid HCV re-infection.
After the treatment is discontinued, the patient may receive follow-up visits at weeks 2,4, 8, 12, 24, 36 and 48 after the end of the treatment. Serum pregnancy tests may be performed at each visit if necessary. The 2 nd and 8 th weeks after the end of treatment may include only laboratory blood draws.
Subjects (n-18) received a single dose of sofosbuvir alone (400mg) or in combination with compound E (500mgQD) under fasting conditions. See table 29 for preliminary PK results for Sofosbuvir in combination with compound E, showing that there was no clinically significant interaction between Sofosbuvir and compound E.
Table 29: pharmacokinetic data for individual and Co-administration of SOF, Compound E and ledipasvir
It should be understood that although the present invention has been specifically disclosed by preferred embodiments and that optional features, alterations, modifications and variations of the present invention may be resorted to by those skilled in the art, those variations, modifications and variations are considered to be within the scope of this invention. The materials, methods, and examples provided herein are representative of, and are illustrative of, preferred embodiments and are not intended to be limiting on the scope of the invention.
The present invention is broadly and generically described herein. Each of the narrower species and subgeneric groupings that fall within the generic description also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any substance from the genus, regardless of whether or not the excised material is specifically recited herein.
Furthermore, when features or aspects of the invention are described in terms of Markush structures, those skilled in the art will recognize that the invention can also be described in terms of any individual member or subgroup of members of the Markush structure.
All writings, patent applications, patents, and other references mentioned herein are expressly incorporated herein by reference in their entirety to the same extent as if each was individually incorporated by reference. In case of conflict, the present specification, including definitions, will control.
Claims (25)
1. A pharmaceutical composition in the form of a fixed-dose combination tablet comprising:
a) from about 10% w/w to about 25% w/w of a solid dispersion comprising ledipasvir dispersed in a polymer matrix formed from copovidone, wherein the weight ratio of ledipasvir to copovidone in the solid dispersion is about 1:1, and wherein the ledipasvir is substantially amorphous and has the following structural formula:
b) about 35% w/w to about 45% w/w of sofosbuvir that is substantially crystalline, wherein the crystalline sofosbuvir has an XRPD2 θ -reflection (° ± 0.2 θ) at about 6.1 and 12.7, and wherein the sofosbuvir has the following structural formula:
c) about 5.0% w/w to about 25% w/w lactose monohydrate;
d) about 5.0% w/w to about 25% w/w microcrystalline cellulose;
e) from about 1.0% w/w to about 10% w/w croscarmellose sodium;
f) about 0.5% w/w to about 3% w/w colloidal silica; and
g) about 0.1% w/w to about 3% w/w magnesium stearate.
2. The pharmaceutical composition of claim 1, comprising about 40% w/w sofosbuvir.
3. The pharmaceutical composition of claim 1 or 2, comprising about 18% w/w of the solid dispersion.
4. A pharmaceutical composition in the form of a fixed-dose combination tablet comprising:
a) about 18% w/w of a solid dispersion comprising ledipasvir dispersed in a polymer matrix formed from copovidone, wherein the weight ratio of ledipasvir to copovidone in the solid dispersion is about 1:1, and wherein the ledipasvir is substantially amorphous and has the following structural formula:
b) about 40% w/w of sofosbuvir, which is substantially crystalline, wherein said crystalline sofosbuvir has an XRPD2 θ -reflection (° ± 0.2 θ) at about 6.1 and 12.7, and wherein said sofosbuvir has the following structural formula:
c) about 16.5% w/w lactose monohydrate;
d) about 18.0% w/w microcrystalline cellulose;
e) about 5.0% w/w croscarmellose sodium;
f) about 1.0% w/w colloidal silica; and
g) about 1.5% w/w magnesium stearate.
5. The pharmaceutical composition of claim 1, having from about 50mg to about 130mg of ledipasvir, and from about 300mg to about 600mg of sofosbuvir.
6. A pharmaceutical composition in the form of a fixed-dose combination tablet comprising:
a) about 180mg of a solid dispersion comprising ledipasvir dispersed in a polymer matrix formed from copovidone, wherein the weight ratio of ledipasvir to copovidone in the solid dispersion is about 1:1, and wherein the ledipasvir is substantially amorphous and has the following structural formula:
b) about 400mg of sofosbuvir, which is substantially crystalline, wherein said crystalline sofosbuvir has an XRPD2 θ -reflection (° ± 0.2 θ) at about 6.1 and 12.7, and wherein said sofosbuvir has the following structural formula:
c) about 165mg lactose monohydrate;
d) about 180mg of microcrystalline cellulose;
e) about 50mg of croscarmellose sodium;
f) about 10mg of colloidal silica; and
g) about 15mg of magnesium stearate.
7. The pharmaceutical composition of claim 5 or the pharmaceutical composition of claim 6, further comprising a film coating.
8. Use of a pharmaceutical composition according to any one of claims 1 to 7 for the manufacture of a medicament for the treatment of a patient with hepatitis c.
9. The use of claim 8, wherein the medicament is for about 24 weeks or less.
10. The use of claim 8, wherein the medicament is for about 12 weeks or less.
11. The use of claim 8, wherein the medicament is for about 8 weeks or less.
12. The use of claim 8, wherein the medicament is for about 6 weeks or less.
13. The use of claim 8, wherein the medicament is for once daily for about 12 weeks or less, and wherein the hepatitis C is genotype 1,2, 3,4, 5 or 6.
14. The use of claim 8, wherein the medicament is for once daily for about 8 weeks or less, and wherein the hepatitis C is genotype 1,2, 3,4, 5 or 6.
15. The use of claim 8, wherein the medicament is for once daily for about 6 weeks or less, and wherein the hepatitis C is genotype 1,2, 3,4, 5 or 6.
16. The use of claim 13, wherein the hepatitis c is genotype 1a or 1 b.
17. The use of claim 14, wherein the hepatitis c is genotype 1a or 1 b.
18. The use of claim 15, wherein the hepatitis c is genotype 1a or 1 b.
19. The use of claim 8, wherein the medicament is for once daily use for about 12 weeks, and wherein the hepatitis C virus is genotype 1a,1b,2a,2b,2c,2d,3a,3b,3c,3d,3e,3f,4a,4b,4c,4d,4e,4f,4g,4h,4i,5a or 6 a.
20. The use of claim 8, wherein the medicament is for once daily use for about 8 weeks, and wherein the hepatitis C virus is genotype 1a,1b,2a,2b,2c,2d,3a,3b,3c,3d,3e,3f,4a,4b,4c,4d,4e,4f,4g,4h,4i,5a or 6 a.
21. Use according to claim 8, with ribavirin.
22. The use of claim 8, wherein the patient has not received interferon therapy.
23. The use of claim 8, wherein the patient has not received ribavirin treatment.
24. The use of claim 23, wherein the patient has not received interferon therapy.
25. Use according to claim 8, using simeprevir.
Applications Claiming Priority (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US61/759,320 | 2013-01-31 | ||
| US61/772,292 | 2013-03-04 | ||
| US61/828,899 | 2013-05-30 | ||
| US61/870,729 | 2013-08-27 | ||
| US61/897,793 | 2013-10-30 | ||
| US61/907,332 | 2013-11-21 |
Publications (3)
| Publication Number | Publication Date |
|---|---|
| HK1226664A1 true HK1226664A1 (en) | 2017-10-06 |
| HK1226664A HK1226664A (en) | 2017-10-06 |
| HK1226664B HK1226664B (en) | 2019-11-01 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US10039779B2 (en) | Combination formulation of two antiviral compounds | |
| US11338007B2 (en) | Combination formulation of three antiviral compounds | |
| HK1226664A1 (en) | Combination formulation of two antiviral compounds | |
| HK1226664A (en) | Combination formulation of two antiviral compounds | |
| HK40031813A (en) | Combination formulation of two antiviral compounds | |
| HK1226664B (en) | Combination formulation of two antiviral compounds | |
| HK1219869B (en) | Combination formulation of two antiviral compounds | |
| OA17450A (en) | Combination formulation of two antiviral compounds. | |
| HK1220392A1 (en) | Combination formulation of two antiviral compounds |