EP3937928A1 - Feste formulierung eines kapsidanordnungmodulators - Google Patents

Feste formulierung eines kapsidanordnungmodulators

Info

Publication number
EP3937928A1
EP3937928A1 EP20709618.1A EP20709618A EP3937928A1 EP 3937928 A1 EP3937928 A1 EP 3937928A1 EP 20709618 A EP20709618 A EP 20709618A EP 3937928 A1 EP3937928 A1 EP 3937928A1
Authority
EP
European Patent Office
Prior art keywords
compound
oral dosage
dosage form
hpmc
solid dispersion
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP20709618.1A
Other languages
English (en)
French (fr)
Inventor
Michaël Bertil S ANNÉ
Claire Elisabeth BALMAIN
Maria JANSENS
Oliver Lenz
Abhishek Singh
Jan SNOEYS
Joris Jozef VANDENBOSSCHE
Frederic Anne R VAN DYCKE
Dominique Josiane W VERSTRAETE
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Sciences Ireland ULC
Original Assignee
Janssen Sciences Ireland ULC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/IB2019/000231 external-priority patent/WO2019175657A1/en
Application filed by Janssen Sciences Ireland ULC filed Critical Janssen Sciences Ireland ULC
Publication of EP3937928A1 publication Critical patent/EP3937928A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1652Polysaccharides, e.g. alginate, cellulose derivatives; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/146Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses

Definitions

  • the present disclosure is directed to solid oral dosage forms of a capsid assembly inhibitor for the treatment of hepatitis B virus infection.
  • Chronic hepatitis B virus (HBV) infection is a persistent, potentially progressive necroinflammatory liver disease associated with chronic HBV infection.
  • HBV infection is a major global cause of severe liver morbidity and liver-related mortality (Hepatitis B Factsheet, World Health Organization, 2013; Hoofnagle JH, et al. , Management of Hepatitis B: Summary of a Clinical Research Workshop, Hepatology, 2007, 45(4):1056- 1075; EASL Clinical Practice Guidelines: Management of chronic hepatitis B virus infection, J. Hepatology, 2012, 57:167-185 (EASL 2012); Lesmana LA, et al.
  • Hepatitis B overview of the burden of disease in the Asia-Pacific region, Liver International, 2006, 26:3-10; Lok ASF and McMahon BJ, Chronic Hepatitis B: Update 2009, Hepatology, September 2009:1-36 (Lok 2009)).
  • HBV antiviral therapies that can achieve sustained viral response during and after treatment.
  • pill burden The number and/or volume of dosage forms that need to be administered are commonly referred to as the "pill burden”.
  • a high pill burden is undesirable for many reasons, such as the number of dosage forms to intake, often combined with the inconvenience of having to swallow large dosage forms, as well as the need to store and transport a large number or volume of pills.
  • a high pill burden increases the risk of patients not taking their entire dose, thereby failing to comply with the prescribed dosage regimen. As well as reducing the effectiveness of the treatment, this also leads to the emergence of viral resistance. The problems associated with this pill burden are increased where a patient must take a combination of different antiviral agents or agents in combination.
  • WO 2014/184350 discloses compound A, and synthesis thereof, as a compound having HBV inhibitory activity:
  • a tablet containing 100 mg of compound A as active ingredient and having a total weight of 1200 mg per tablet is disclosed in PCT/IB2019/000231.
  • the oral dosage forms are formed by spray drying of the compound with a stabiliser, dry granulation and compression of the ingredients. In order to achieve a total dosing of 250-300 mg of compound A, intake of at least three dosage forms of acceptable size are required when using that tablet composition.
  • HBV inhibitory therapy that involves the administration of dosage forms of a practical size while reducing the number of dosage forms.
  • the present invention is based on the unexpected finding that a higher weight% load of compound A per dosage form is feasible while maintaining an acceptable stability of product upon storage and an acceptable bioavailability.
  • the present invention thus provides an anti-HBV therapy involving the
  • dosage forms of compound A having acceptable size and/or number of dosage forms are beneficial in terms of pill burden and drug compliance of the patient.
  • the invention relates to a solid dispersion comprising compound A:
  • Hypromellose HPMC
  • HPMC-AS Hypromellose acetate succinate
  • Said solid dispersion can be further incorporated into an oral dosage form, in particular a solid oral dosage form, more in particular, a tablet.
  • the invention also relates to a solid oral dosage form, in particular a tablet, comprising a solid dispersion comprising compound A
  • a stabiliser selected from at least one of Hypromellose (HPMC) and Hypromellose acetate succinate (HPMC-AS) at a ratio of 1 :1 to less than 1 :3 by weight, and additional pharmaceutical excipients, with the proviso that the ratio compound A: stabiliser is not 1 :2 by weight.
  • the invention relates to a process for preparing an oral dosage form according to the invention, comprising the steps of:
  • the invention relates to a process for preparing an oral dosage form according to the invention, comprising the steps of:
  • a spray-dried powder by preparing a solution of the compound with the stabiliser and spray drying the solution to form a spray dried powder; mixing the spray-dried powder with tablet excipients; and
  • the present invention relates to an oral dosage form according to the invention for use in medicine, more specifically for use in the treatment of HBV infection.
  • the invention relates to a method for the treatment of an HBV infection in a subject which comprises administering to the subject a therapeutically effective amount of an oral dosage form according to the invention.
  • Fig. 1 depicts the geometric mean concentration (cone) (ng/mL) as a function of time (hours) (initial 24 hours) in two pharmacokinetic studies in healthy volunteers evaluating different formulations.
  • the present invention provides a solid dispersion comprising compound A or a pharmaceutically acceptable salt or solvate thereof, and a stabiliser selected from at least one of Hypromellose (HPMC) and Hypromellose acetate succinate (HPMC-AS) at a ratio of 1 :1 to less than 1 :3 by weight, with the proviso that the ratio is not 1 :2 by weight.
  • a stabiliser selected from at least one of Hypromellose (HPMC) and Hypromellose acetate succinate (HPMC-AS) at a ratio of 1 :1 to less than 1 :2 by weight.
  • the invention relates to a solid dispersion as described herein, comprising compound A
  • Hypromellose HPMC
  • HPMC-AS Hypromellose acetate succinate
  • compound A is present in the solid dispersion in amorphous form, more in particular, as an amorphous, base form.
  • the solid dispersion of compound A and stabilizer as described herein is a solid solution.
  • the present invention also provides a solid oral dosage form, in particular a tablet, of compound A, that is manufactured by first providing a solid dispersion as described herein, comprising compound A or a pharmaceutically acceptable salt or a solvate thereof, and a stabiliser selected from at least one of Hypromellose (HPMC) and
  • Hypromellose acetate succinate at a ratio of 1 : 1 to less than 1 :3 by weight, with the proviso that the ratio is not 1 :2 by weight, more in particular, at a ratio of 1 : 1 to less than 1 :2.
  • the invention also relates to a solid oral dosage form, in particular a tablet, comprising a solid dispersion comprising compound A
  • a stabiliser selected from at least one of Hypromellose (HPMC) and Hypromellose acetate succinate (HPMC-AS) at a ratio of 1 :1.5 by weight and additional pharmaceutical excipients.
  • Hypromellose may e.g. be Hypromellose E5 (HPMC E5) (i.e., HPMC with a viscosity of 5 mPa.s).
  • compound A is present in the solid dispersion in amorphous form, more in particular, as an amorphous, base form.
  • the solid dispersion of compound A and stabilizer as described herein is a solid solution.
  • the oral dosage form in particular the tablet, according to the invention, may further comprise one or several agents selected from fillers, disintegrants, glidants, and lubricants.
  • a tablet of the invention may further comprise at least one filler selected from microcrystalline cellulose, silicified microcrystalline cellulose, lactose monohydrate and pre-gelatinized maize starch, at least one disintegrant such as croscarmellose sodium, at least one glidant such as colloidal anhydrous silica, hydrophobic colloidal silica, and at least one lubricant such as magnesium stearate.
  • the tablet comprises 50-300 mg of compound A, in particular 50-250 mg of compound A (more particularly at the above-mentioned doses or daily doses) and 75-450 mg of stabilizer, in particular, 75-375 mg of stabiliser. In a further embodiment, the tablet comprises 250 mg of compound A.
  • the weight percentage of compound A compared to the 100 mg tablet described in PCT/IB2019/000231 can be increased per dosage form by about 60%, thus generating oral dosage forms with a higher drug load of compound A (e.g. tablet having 160 mg compound A and having a total weight of 1200 mg per tablet).
  • the size and weight of existing dosage forms e.g. tablets having 100 mg compound A and having a total weight of 750 mg per tablet may be reduced by about 38%.
  • the size of the dosage forms of the invention i.e. the total weight of the dosage forms, in particular a tablet, should be below a limit of convenience which is below the size at which a number of patients start having difficulty taking in the dosage form.
  • the term“comprising” can include the embodiments“consisting of” and“consisting essentially of.”
  • the terms“comprise(s),” “include(s),”“having,”“has,”“can,”“contain(s),” and variants thereof, as used herein, are intended to be open-ended transitional phrases, terms, or words that require the presence of the named ingredients/steps and permit the presence of other ingredients/steps.
  • compositions or processes as“consisting of” and“consisting essentially of” the enumerated components, which allows the presence of only the named components, along with any
  • approximating language can be applied to modify any quantitative representation that can vary without resulting in a change in the basic function to which it is related. Accordingly, a value modified by a term or terms, such as“about” and “substantially,” can not be limited to the precise value specified, in some cases. In at least some instances, the approximating language can correspond to the precision of an instrument for measuring the value.
  • the modifier“about” should also be considered as disclosing the range defined by the absolute values of the two endpoints. For example, the expression“from about 50 to about 250” also discloses the range“from 50 to 250.”
  • the term“about” can refer to plus or minus 10% of the indicated number.
  • “about 10%” can indicate a range of 9% to 11 %, and“about 1” can mean from 0.9 to 1.1.
  • Other meanings of“about” can be apparent from the context, such as rounding off, so, for example“about 1” can also mean from 0.5 to 1.4.
  • treatment is defined as the application or administration of a therapeutic agent, i.e., a compound of the invention (alone or in combination with another pharmaceutical agent), to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g., for diagnosis or ex vivo applications), who has HBV infection, chronic HBV infection, a symptom of HBV infection or the potential to develop HBV infection, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect HBV infection, the symptoms of HBV infection or the potential to develop HBV infection.
  • Such treatments can be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics.
  • prevent comprises the prevention of at least one symptom associated with or caused by the state, disease or disorder being prevented.
  • the term“patient,”“individual” or“subject” refers to a human or a non-human mammal.
  • Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline and murine mammals.
  • the patient, subject or individual is human.
  • the term“pharmaceutically acceptable” refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material can be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • the term‘compound A’ is meant to comprise the base form, any pharmaceutically acceptable acid addition salt thereof, as well as any pharmaceutically acceptable solvate thereof.
  • the pharmaceutically acceptable addition salts as mentioned hereinabove are the therapeutically active non-toxic acid addition salt forms, which compound A is able to form.
  • the term‘compound A’ is meant to comprise the base form, as well as any pharmaceutically acceptable solvate thereof.
  • pharmaceutically acceptable solvate comprises the hydrates and the solvent addition forms that compound A can form. Examples of such forms are e.g.
  • composition or“pharmaceutical composition” refers to a mixture of at least one compound useful within the invention with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration.
  • Oral dosage forms according to the invention will preferably comprise
  • Such inactive ingredients are added to help hold the tablet together and give it strength, and are selected among binders, fillers, disintegrants, glidants and lubricants.
  • the term“pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabiliser, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the invention within or to the patient such that it can perform its intended function. Typically, such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be“acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound useful within the invention, and not injurious to the patient.
  • “pharmaceutically acceptable carrier” also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound useful within the invention, and are physiologically acceptable to the patient.
  • lubricant means a pharmaceutically acceptable material which avoids manufacturing probles such as tablet sticking when the drug product blend is compressed into tablets.
  • a tablet formulation may also contain a disintegrant to aid disintegration and dissolution of the formulation upon administration to the patients.
  • compositions used in the practice of the invention are known in the art and described, for example in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.
  • stabiliser refers to polymers capable of chemically inhibiting or preventing degradation or conversion to crystalline state of compound A.
  • Stabilisers are added to formulations of compounds to improve chemical and physical stability of the compound.
  • a tablet formulation can be used to make tablet cores in a conventional manner for example by initially dry blending the ingredients, that preferably having been sieved or by dry granulation, in order to improve blend flow. Subsequently, the lubricant is added to the dry-blended mixture for final dry-blending of the total tablet core blend, which is then compressed into tablets having the desired size and weight.
  • the tablet cores according to the invention are generally provided with a film coating. Different coloring agents may be used in the film coating in order to differentiate between tablet strengths.
  • the coating can be applied to the core in coating suspension for example in purified water, followed by drying of the coated cores.
  • “combination,”“therapeutic combination,”“pharmaceutical combination,” or“combination product” as used herein refer to a non-fixed combination or a kit of parts for the combined administration where two or more therapeutic agents can be
  • treatment naive refers to a patient not having previously received treatment with a drug, investigational or approved, for HBV infection, in particular a nucleos(t)ide drug. “Treatment naive” also refers to a patient not having been on treatment with HBV antiviral medicines within six months of entering a clinical study.
  • patients treated according to the methods of the disclosure can be “treatment experienced.”
  • “treatment experienced” refers to a patient who has had at least one previous course of an HBV antiviral therapy, in particular a nucleos(t)ide drug.
  • the last dose in this previous course occurred at least three months prior to implementing a method according to the present disclosure.
  • HBV infections that may be treated according to the disclosed methods include HBV genotype A, B, C, and/or D infections.
  • the methods disclosed may treat any HBV genotype (“pan-genotypic treatment”).
  • HBV genotyping may be performed using methods known in the art, for example, IN NO-LI PA® HBV Genotyping, Innogenetics N.V., Ghent, Belgium).
  • the term“synergistic effect” refers to the action of two agents, such as, for example, a capsid assembly modulator and a nucleos(t)ide analogue, producing an effect, for example, slowing the symptomatic progression of HBV-infection or symptoms thereof, which is greater than the simple addition of the effects of each drug administered alone.
  • a synergistic effect can be calculated, for example, using suitable methods such as the Sigmoid-Emax equation (Holford, N. H. G. and Scheiner, L. B., Clin. Pharmacokinet. 6: 429-453 (1981)), the equation of Loewe additivity (Loewe, S. and Muischnek, H., Arch. Exp. Pathol Pharmacol. 114: 313-326 (1926)) and the median-effect equation (Chou, T.
  • each equation referred to above can be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination.
  • the corresponding graphs associated with the equations referred to above are the concentration-effect curve, isobologram curve and combination index curve, respectively.
  • the combination of compounds exhibits a synergistic effect (/.e., greater than additive effect) in the treatment of HBV infection.
  • Synergy volumes of ⁇ -100, -100 to -50, -50 to -25, -25 to 25, 25 to 50, 50 to 100, and >100 indicate strong antagonism, moderate antagonism, slight antagonism, insignificant synergism/antagonism (additivity), slight synergism, moderate synergism, and strong synergism respectively.
  • Synergy can be defined as an improvement in any beneficial effect of compound A or a nucleos(t)ide analogue, alone or in combination.
  • the improvement may exceed an additive effect of the combination or may only occur as a result of the combination.
  • the effect is complete or sustained reduction of viral load, HBsAg and/or anti-HBsAb during and/or after treatment.
  • the effect is sustained virological response (SVR) and/or sustained viral clearance.
  • the estimated daily dose of compound A is 250 mg compound A once per day.
  • a stabiliser selected from Hypromellose acetate succinate (HPMC-AS) and Hypromellose (HPMC) at a ratio of 1 :3 by weight in the form of a spray dried powder composition ingestion of two tablets of 1200 mg nominal weight, containing 100 mg of compound A and one or two smaller tablets containing 50 or 25 mg of compound A, respectively, is required.
  • Such tablets have a drug load of 0.083 mg compound A per mg tablet.
  • One option in order to reduce the tablet size and/or number of tablets required to achieve a particular dose could be to increase the drug load, and/or decrease the amount of excipients. Decreasing the amount of stabiliser in a given formulation however, could have a detrimental impact in the physical stability of the active pharmaceutical ingredient (API), as well as decrease the amount of API dissolved in the gastrointestinal tract, and therefore bioavailability of the active pharmaceutical ingredient.
  • API active pharmaceutical ingredient
  • By providing a solid dispersion obtained by spray drying compound A with the stabilizer compound A is present in amorphous form, which in turn improves the solubility and bioavailability of compound A.
  • the stabilizer must be present in a sufficient amount in order to prevent compound A from converting to a crystalline form, but also to prevent it from precipitating in the gastrointestinal tract, which in turn might affect the bioavailability of compound A. It has now been unexpectedly discovered that the amount of stabiliser can be reduced and, by using the composition of the invention, tablets with a drug load of 0.133 mg compound A per mg tablet can be made available. In order to ensure appropriate exposure levels, dose adjustment may therefore be necessary.
  • the present invention provides a solid oral dosage form according to the invention for use in medicine, more specifically for use in the treatment of HBV infection.
  • the invention relates to a method for the treatment of an HBV infection in a subject which comprises administering the subject an effective amount of an oral dosage form according to the invention.
  • the invention relates to a method of preventing or treating HBV infection in a subject, said method comprising administering to said subject compound A at a daily dose of 50-300 mg, in particular 50-250 mg, wherein compound A is administered in at least one dosage form comprising a solid dispersion comprising compound A or a pharmaceutically acceptable salt or a solvate thereof, and a stabiliser selected from at least one of Hypromellose (HPMC) and Hypromellose acetate succinate (HPMC-AS) at a ratio of 1 : 1 to less than 1 :3 by weight with the proviso that the ratio is not 1 :2 by weight; more in particular at a ratio of 1 : 1 to less than 1 :2 by weight, yet more in particular, at a ratio of 1 :1.5 by weight.
  • a stabiliser selected from at least one of Hypromellose (HPMC) and Hypromellose acetate succinate (HPMC-AS) at a ratio of 1 : 1 to less than 1 :3 by weight with the
  • the treatment is curative and the patient does not have to continue treatment after the specified treatment time.
  • the treatment is finite.
  • the use or method further comprises administering, in particular co-administering, a transcription inhibitor to the subject.
  • the transcription inhibitor is a nucleoside analog.
  • the nucleoside analog is administered at the approved daily dosing regimen for the nucleoside analogue.
  • the nucleoside analog is tenofovir, or a pharmaceutically acceptable salt or prodrug thereof (e.g. tenofovir disoproxil fumarate or tenofovir alafenamide), or entecavir, or a
  • the nucleoside analog is tenofovir disoproxil fumarate.
  • the nucleoside analog is tenofovir alafenamide.
  • the nucleoside analog is entecavir monohydrate.
  • the tenofovir disoproxil fumarate is administered in an amount of 60-600 mg.
  • the tenofovir disoproxil fumarate is administered in an amount of 300 mg.
  • the entecavir monohydrate is administered in an amount of 0.1-1 g.
  • the entecavir monohydrate is administered in an amount of 0.5 mg.
  • the use or method further comprises administering an immune modulator.
  • the immune modulator is interferon, for example interferon alpha or pegylated interferon alpha.
  • the subject is treatment naive.
  • the use or method further comprises administering at least one Nucleic Acid Polymer (NAP), more particularly at least one NAP which inhibits the release of subviral particles from hepatocytes.
  • NAP Nucleic Acid Polymer
  • the use or method further comprises administering at least one short interfering RNA (siRNA) or antisense oligonucleotide (ASO), more particularly at least one siRNA or ASO sleeted from the group of siRNAs and ASOs which inhibit the expression of one or more genes that are necessary for replication or pathogenesis of HBV.
  • siRNA short interfering RNA
  • ASO antisense oligonucleotide
  • the administration of compound A is performed for an administration period of about 24 weeks. In another embodiment, the administration of compound A is performed for an administration period of longer than 24 weeks. In yet another embodiment, the administration of compound A is performed for an administration period shorter than 24 weeks (e.g., 10, 12, 14, 16, 18, 20, or 22 weeks). In embodiments, compound A is administered for a duration of 28 days. In embodiments, compound A is administered for a duration of about 48 weeks. In embodiments, compound A is administered for a duration of longer than 48 weeks.
  • the co-administration of compound A and the transcription inhibitor is performed for an administration period of about 24 weeks. In another embodiment, the administration of compound A and the transcription inhibitor is performed for an administration period of longer than 24 weeks. In yet another embodiment, the administration of compound A and the transcription inhibitor is performed for an administration period shorter than 24 weeks (e.g., 10, 12, 14, 16, 18, 20, or 22 weeks). In embodiments, compound A and the transcription inhibitor is
  • transcription inhibitor is administered for a duration of about 48 weeks. In embodiments, compound A and the transcription inhibitor is administered for a duration of longer than 48 weeks.
  • compound A is administered to reach a maximal concentration (Cmax) of at least 3,000 ng/mL (e.g., at steady state), in the plasma of the patient and/or an AUC of at least 50,000 ng.h/mL (e.g., at steady state), in the plasma of the patient.
  • Compound A is administered to reach a maximal concentration (Cmax) of at least 3,000 ng/mL, in the plasma of the patient and/or an AUC of at least 50,000 ng.h/mL, in the plasma of the patient.
  • the daily doses described herein are calculated for an average body weight of about 60 to about 70 kg and should be recalculated in case of paediatric applications, or when used with patients with a substantially diverting body weight.
  • kits of parts for treating HBV infections comprising a pharmaceutical composition comprising compound A, or a pharmaceutically acceptable salt or a solvate thereof, as described herein.
  • the kit of parts further comprises packaging and instructions.
  • the kit of parts comprises a pharmaceutical composition comprising compound A, or a pharmaceutically acceptable salt or solvate thereof, as described herein; an additional HBV antiviral agent; and a pharmaceutically acceptable carrier or diluent.
  • the additional HBV antiviral agent can e.g., be a nucleos(t)ide analogue (such as tenofovir or a pharmaceutically acceptable salt or prodrug thereof, or entecavir or a pharmaceutically acceptable salt thereof); an immune modulator (such as interferon), at least one Nucleic Acid Polymer (more particularly at least one NAP which inhibits the release of subviral particles from hepatocytes), or at least one small interfering RNA (siRNA) or antisense oligonucleotide (more particularly at least one siRNA or ASO sleeted from the group of siRNAs and ASOs which inhibit the expression of one or more genes that are necessary for replication or pathogenesis of HBV).
  • an immune modulator such as interferon
  • at least one Nucleic Acid Polymer more particularly at least one NAP which inhibits the release of subviral particles from hepatocytes
  • siRNA small interfering RNA
  • antisense oligonucleotide more
  • kits are provided.
  • the kit includes a sealed container approved for the storage of pharmaceutical compositions, the container containing one of the above-described pharmaceutical compositions.
  • the sealed container minimizes the contact of air with the ingredients, e.g. an airless bottle.
  • the sealed container is a sealed tube.
  • An instruction for the use of the composition and the information about the composition are to be included in the kit.
  • the qualitative and quantitative composition of the Compound A 100-mg (G009) 25- mg (G008) and 5-mg (G007) oral tablets is provided in Table 2 below.
  • the tablets contain compound A and a stabiliser at a ratio of 1 :3 by weight.
  • the quantitative and qualitative composition of the Compound A 250 mg/g spray dried powder (G021) is provided in Table 3 below.
  • the qualitative and quantitative composition of the Compound A 100-mg (G022) oral tablets, Compound A 100-mg (G024) oral tablets and Compound A 25-mg (G-25) oral tablets are provided in Table 4 below.
  • the tablets contain compound A and a stabiliser at a ratio of 1 :3 by weight.
  • Table 4 Qualitative and Quantitative Composition of Compound A 100-mg oral tablets (G022), 100-mg oral tablets (G024) and 25-mg oral tablets (G025)
  • Example 1 New formulations according to the invention
  • the qualitative and quantitative composition of the Compound A 100-mg (G030) oral tablets and 100-mg (G031) oral tablets is provided in Table 7 below.
  • the tablets contain compound A and a stabiliser at a ratio of 1 : 1.5 by weight.
  • Table 7 Qualitative and Quantitative Composition of Compound A 100-mg oral tablets (G030) and (G031)
  • PK pharmacokinetics
  • Study 1 evaluated the exposure of G020, a 10 mg/mL aqueous suspension of Compound A in a crystalline form, as well as the exposure of G009, a 100-mg tablet containing an amorphous solid dispersion of compound A and HPMC as a stabiliser at a ratio of 1 :3 by weight.
  • Study 2 evaluated the exposure of G024, a 100-mg tablet containing an
  • PK data is summarized in Fig. 1 and table 8.
  • Tablet formulations containing an amorphous solid dispersion of compound A and HPMC or HPMC-AS as a stabiliser at a ratio of 1 :3 by weight result in exposures in healthy subjects that can result in treatment regimen in cHBV patients with acceptable pill burden.
  • Tablet formulations containing an amorphous solid dispersion of compound A and HPMC or HPMC-AS as a stabiliser at a ratio of 1 :1.5 by weight result in exposures that can result in treatment regimen in cHBV patients with comparable exposures as formulations with the higher stabilizer amount and allow for formulation of a tablet with higher concentration of Compound A and thus reduced pill burden.
  • Exposure of the tablet formulations containing an amorphous solid dispersion of compound A and HPMC or HPMC-AS as a stabiliser at a ratio of 1 :1.5 or 1 :3 by weight is significantly higher compared to exposure of a crystalline aqueous suspension of Compound A.
  • Exposure of the crystalline suspension formulation was approximately 16 fold lower compared to the tablet formulation G024 after dosing in fed state.
  • the dose of the crystalline suspension would need to be approximately 16 fold higher to reach the same exposure as the oral tablet G024; based on this, and without wishing to be bound by theory, a tablet formulation with the compound in a crystalline form could therefore result in treatment regimen in cHBV patients with a high pill burden.
  • High pill burden can affect patient adherence as it is not convenient for the patient and certainly not for a treatment regimen that has to be administered for a prolonged period of time.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
EP20709618.1A 2019-03-13 2020-03-13 Feste formulierung eines kapsidanordnungmodulators Withdrawn EP3937928A1 (de)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
PCT/IB2019/000231 WO2019175657A1 (en) 2018-03-14 2019-03-13 Capsid assembly modulator dosing regimen
US16/352,754 US10973801B2 (en) 2018-03-14 2019-03-13 Capsid assembly modulator dosing regimen
EP2019056348 2019-03-13
EP19197566 2019-09-16
PCT/EP2020/056991 WO2020183020A1 (en) 2019-03-13 2020-03-13 Capsid assembly modulator solid formulation

Publications (1)

Publication Number Publication Date
EP3937928A1 true EP3937928A1 (de) 2022-01-19

Family

ID=72427202

Family Applications (1)

Application Number Title Priority Date Filing Date
EP20709618.1A Withdrawn EP3937928A1 (de) 2019-03-13 2020-03-13 Feste formulierung eines kapsidanordnungmodulators

Country Status (16)

Country Link
EP (1) EP3937928A1 (de)
JP (1) JP2022524819A (de)
KR (1) KR20210137484A (de)
CN (1) CN113557016A (de)
AU (1) AU2020235442A1 (de)
BR (1) BR112021017525A2 (de)
CA (1) CA3132095A1 (de)
CR (1) CR20210481A (de)
EC (1) ECSP21067052A (de)
IL (1) IL286209A (de)
JO (1) JOP20210250A1 (de)
MA (1) MA55280A (de)
MX (1) MX2021011030A (de)
PE (1) PE20212107A1 (de)
SG (1) SG11202109710QA (de)
WO (1) WO2020183020A1 (de)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2017248828A1 (en) 2016-04-15 2018-11-01 Janssen Sciences Ireland Uc Combinations and methods comprising a capsid assembly inhibitor
CN111867582A (zh) * 2018-03-14 2020-10-30 爱尔兰詹森科学公司 衣壳组装调节剂给药方案

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JO3603B1 (ar) 2013-05-17 2020-07-05 Janssen Sciences Ireland Uc مشتقات سلفامويل بيرولاميد واستخدامها كادوية لمعالجة التهاب الكبد نوع بي
CN111867582A (zh) * 2018-03-14 2020-10-30 爱尔兰詹森科学公司 衣壳组装调节剂给药方案

Also Published As

Publication number Publication date
MA55280A (fr) 2022-01-19
AU2020235442A1 (en) 2021-08-12
KR20210137484A (ko) 2021-11-17
JP2022524819A (ja) 2022-05-10
CR20210481A (es) 2021-10-25
IL286209A (en) 2021-10-31
CN113557016A (zh) 2021-10-26
SG11202109710QA (en) 2021-10-28
CA3132095A1 (en) 2020-09-17
PE20212107A1 (es) 2021-11-04
ECSP21067052A (es) 2021-11-18
MX2021011030A (es) 2021-10-13
WO2020183020A1 (en) 2020-09-17
JOP20210250A1 (ar) 2023-01-30
BR112021017525A2 (pt) 2021-11-23

Similar Documents

Publication Publication Date Title
JP6491174B2 (ja) ニタゾキサニドの制御放出医薬配合物
TWI287988B (en) Low dose entecavir formulation and use
TW389696B (en) Accelerated release composition containing bromocriptine
US20180042944A1 (en) Pharmaceutical compositions
US10973801B2 (en) Capsid assembly modulator dosing regimen
TW200412934A (en) Pharmaceutical formulations of modafinil
ES2865278T3 (es) Amantadina para mejorar la marcha en esclerosis múltiple
CN111918646B (zh) 延迟释放去铁酮片剂及其使用方法
CA2961528A1 (en) Long acting pharmaceutical compositions
JP5514123B2 (ja) 卵巣癌を治療するための、パクリタキセルを含む配合剤
EP3937928A1 (de) Feste formulierung eines kapsidanordnungmodulators
NZ541187A (en) Use of a combination containing a non-nucleoside reverse transcriptase inhibitor (NNRTI) with an inhibitor of cytochrome P450, such as protease inhibitors
JP2004525143A5 (de)
JP2006524190A5 (de)
WO2010022581A1 (zh) 用于治疗高尿酸血症的药物组合物及其用途
CZ20021315A3 (cs) Jednotka dávkování
WO1996011682A1 (fr) Composition preventive et remede pour les maladies allergiques de type i
JP6420923B1 (ja) 医薬
CN107213138A (zh) 定时释放药物治疗高血压的方法和药物组合物
TW202245772A (zh) 一種衣殼蛋白抑制劑的醫藥組成物及其製備方法
EP4262805A1 (de) Minitablettendosierungsform eines viralen terminasehemmers und verwendungen davon
WO2015016694A1 (es) Composición sólida estable de inmunosupresores
CN105434364B (zh) 一种托匹司他缓释颗粒及其制备方法
CN111643506A (zh) 一种奥氮平氟西汀复方胶囊制剂及其制备方法
WO2008106738A1 (en) Compositions for the treatment of sexual dysfunction

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20211013

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20220503