WO2021170741A1 - Treatment of hbv - Google Patents

Treatment of hbv Download PDF

Info

Publication number
WO2021170741A1
WO2021170741A1 PCT/EP2021/054727 EP2021054727W WO2021170741A1 WO 2021170741 A1 WO2021170741 A1 WO 2021170741A1 EP 2021054727 W EP2021054727 W EP 2021054727W WO 2021170741 A1 WO2021170741 A1 WO 2021170741A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
treatment
patient
hbeag
nucleos
Prior art date
Application number
PCT/EP2021/054727
Other languages
French (fr)
Inventor
Oliver Lenz
Willem Jan-Paul Edmond TALLOEN
Joris Jozef VANDENBOSSCHE
Jeysen Zivan YOGARATNAM
Original Assignee
Janssen Sciences Ireland Unlimited Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janssen Sciences Ireland Unlimited Company filed Critical Janssen Sciences Ireland Unlimited Company
Publication of WO2021170741A1 publication Critical patent/WO2021170741A1/en

Links

Classifications

    • 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
    • A61K31/401Proline; Derivatives thereof, e.g. captopril
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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

  • 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)).
  • Compound 1 a potent class N capsid assembly modulator, has been tested in phase 1 clinical trials and has been found well tolerated with dose-dependent PK and efficacy up to 250 mg orally qd for 4 weeks in treatment-naive chronic hepatitis B (CHB) patients (patients).
  • CHB chronic hepatitis B
  • HBV antiviral therapies that can achieve sustained viral response during and after treatment.
  • the present disclosure is directed to methods of using a capsid assembly inhibitor for the treatment of hepatitis B virus infection.
  • provided herein is a method of treating Hepatitis B viral infection in a not treated HBeAg+ patient comprising administering to the patient, a therapeutically effective amount of a combination of a nucleos(t)ide analogue and a compound of Formula 1 or a pharmaceutically acceptable salt thereof.
  • a method of treating Hepatitis B viral infection in a not treated HBeAg+ patient comprising administering to the patient, a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
  • a method of treating Hepatitis B viral infection in a not treated HBeAg+ patient comprising: evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and treating the patient with a therapeutically effective amount of a combination of a nucleos(t)ide analogue and a compound of Formula 1 or a pharmaceutically acceptable salt thereof.
  • a method of treating Hepatitis B viral infection in a not treated HBeAg+ patient comprising: evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and treating the patient with a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
  • nucleos(t)ide analogue in another aspect, provided herein is a combination of a nucleos(t)ide analogue and a compound of Formula 1 or a pharmaceutically acceptable salt thereof for use in cotherapy for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient.
  • nucleos(t)ide analogue in a further aspect, provided herein is a combination of a nucleos(t)ide analogue and a compound of Formula 1 or a pharmaceutically acceptable salt thereof, for use in the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient.
  • a combination of a nucleos(t)ide analogue and a compound Formula 1 or a pharmaceutically acceptable salt thereof for use in the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient, wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering the combination of a nucleos(t)ide analogue and compound 1 or a pharmaceutically acceptable salt thereof.
  • a compound Formula 1 or a pharmaceutically acceptable salt thereof for use in the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient, wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering the compound of Formula 1 or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
  • a combination as described herein wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering a cotherapy by combination of a nucleos(t)ide analogue and compound 1 or a pharmaceutically acceptable salt thereof.
  • a combination of a nucleos(t)ide analogue and a compound Formula 1 or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient.
  • a compound Formula 1 or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient, wherein the patient is also administered a nucleos(t)ide analogue.
  • a use of a combination of a nucleos(t)ide analogue and a compound Formula 1 or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering the combination of a nucleos(t)ide analogue and compound 1 or a pharmaceutically acceptable salt thereof.
  • a use of a compound Formula 1 or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering the compound 1 or a pharmaceutically acceptable salt thereof in combination with a nucleos(t)ide analogue.
  • Hepatitis B viral infection as referred to herein is, in particular, chronic hepatitis B viral infection.
  • Fig. 1 A depicts a schematic representation of the study design.
  • Fig. 1B depics a schematic representation of the study design to week 24.
  • Fig. 2 depicts the mean standard error (SE) change in HBV DNA (assessed using SE).
  • HBV DNA lower limit of quantification (LLOQ): 20 lll/mL.
  • HBV DNA lower limit of quantification (HBeAg: hepatitis B e antigen, NA: nucleos(t)ide analogue; IU: international unit).
  • Fig. 3 depicts the mean (SE) change in HBV RNA over 24 weeks of treatment from baseline in NT and virologically suppressed (VS) patients (HBeAg+ and HBeAg-).
  • HBV RNA LLOQ 4.04 logio copies/mL. (TND: target not detected)
  • Fig. 4 depicts the mean (SE) change in hepatitis B surface antigen (HBsAg) over 24 wks of treatment from baseline by patient population.
  • HBsAg LLOQ 0.05 lU/mL and ULOQ:
  • Fig. 5 depicts the mean (SE) change in HBeAg over 24 weeks of treatment from baseline in HBeAg+ patients.
  • Fig. 6 depicts the individual changes in A) HBsAg and B) HBeAg over 24 weeks of treatment from baseline in HBeAg+ NT patients.
  • the present disclosure is directed to methods of using a capsid assembly inhibitor for the treatment of hepatitis B virus infection. It has now been found that at 24 weeks, administration of compound 1 in combination with a nucleos(t)ide analogue, in not treated patients, increases suppression of HBV DNA, and HBV RNA in all patients, compared to nucleos(t)ide analogue alone. Unexpectedly, in not treated HBeAg+ patients, at 24 weeks, at an oral dose of 250 mg compound 1 daily a mean 0.4 logio HBsAg decline was observed.
  • 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.
  • such description should be construed as also describing compositions or processes as “consisting of” and “consisting essentially of” the enumerated compounds, which allows the presence of only the named compounds, along with any pharmaceutically acceptable carriers, and excludes other compounds.
  • treatment is defined as the application or administration of a therapeutic agent, i.e. , a combination of the invention (alone or in combination with another pharmaceutical agent), to a patient as defined herein, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect HBV infection, or the symptoms of HBV infection.
  • a therapeutic agent i.e. , a combination of the invention (alone or in combination with another pharmaceutical agent)
  • 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 “pharmaceutically acceptable salt” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.
  • 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.
  • the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, 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.
  • a pharmaceutically acceptable material, composition or carrier such as a liquid or solid filler, stabilizer, 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.
  • 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.
  • materials that can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline
  • “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. Supplementary active compounds can also be incorporated into the compositions.
  • the “pharmaceutically acceptable carrier” can further include a pharmaceutically acceptable salt of the compound useful within the invention.
  • Other additional ingredients that can be included in the pharmaceutical 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.
  • stabilizer refers to polymers capable of chemically inhibiting or preventing degradation of a Compound of Formula 1. Stabilizers are added to formulations of compounds to improve chemical and physical stability of the compound.
  • combination refers to a non-fixed combination or a kit of parts for the combined administration where two or more therapeutic agents can be administered independently, at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g., synergistic, effect.
  • 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.
  • 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.
  • Not treated refers to a patient that is not receiving any chronic hepatitis B treatment at screening, ie, has not received within 6 months prior to baseline treatment with HBV antiviral medicines, including nucleot(s)ide analogues (NAs) or interferon (IFN) products, or has not been on treatment with HBV antiviral medicines, including NAs or IFN products.
  • virologically suppressed refers to patients treated with HBV antiviral therapy, in particular a nucleos(t)ide drug.
  • 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, INNO-LIPA® HBV Genotyping, Innogenetics N.V., Ghent, Belgium).
  • 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.
  • 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 each of the compound of Formula 1 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.
  • Nucleos(t)ide analogs can include deoxyadenosine analogues, adenosine analogues, deoxycytidine analogues, guanosine analogues, thymine analogues, and deoxyuridine analogues.
  • the nucleos(t)ide analogs can include Lamivudine (Lamivudine,3TC or LMV), Adefovir (Adefovir, ADV), Entecavir (Entecavir, ETV), Telbivudine (Telbivudine, LdT) and Tenofovir, or prodrugs thereof (e.g.
  • Tenofovir disoproxil fumarate, TDF) and pharmaceutically acceptable salts thereof include tenofovir disoproxil fumarate, tenofovir disoproxil succinate, and tenofovir alafenamide fumarate, in particular tenofovir disoproxil fumarate and tenofovir alafenamide fumarate.
  • the nucleos(t)ide analogue used in the invention can be selected from tenofovir, or a pharmaceutically acceptable salt or prodrug thereof, and entecavir, or a pharmaceutically acceptable salt thereof.
  • tenofovir or a pharmaceutically acceptable salt or prodrug thereof can be selected from tenofovir alafenamide, tenofovir disoproxil succinate and tenofovir disoproxil fumarate, more in particular tenofovir alafenamide and tenofovir disoproxil fumarate.
  • entecavir or a pharmaceutically acceptable salt thereof is entecavir monohydrate.
  • the nucleos(t)ide analogue used in the invention is administered at the approved dose/dosage.
  • tenofovir disoproxil fumarate is administered in an amount of 60-600 mg.
  • tenofovir disoproxil fumarate is administered in an amount of 300 mg.
  • tenofovir alafenamide is administered in an amount of 25 mg.
  • the entecavir monohydrate is administered in an amount of 0.1-1 mg.
  • entecavir monohydrate is administered in an amount of 0.5 mg.
  • the nucleos(ti)de analogue is tenofovir or a produg thereof, in particular, tenofovir alafenamide, or tenofovir disoproxil fumarate.
  • the amount of the compound of Formula 1 in the invention is from about 50 mg per day to about 500 mg per day (e.g. 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500 mg). In some embodiments, the amount of a compound of Formula 1 is from 75 mg per day to 250 mg per day. In some embodiments, the amount of a Compound of Formula 1 is 75 mg per day. In some embodiments, the amount of a Compound of Formula 1 is 150 mg per day. In some embodiments, the amount of a Compound of Formula 1 is 250 mg per day.
  • the compound of Formula 1 is administered at a dose of 75- 300mg, more particularly 80-300mg, more particularly 100-300mg, more particularly 100-250mg once daily. In a further embodiment, the compound of Formula 1 is administered at a dose of 75 mg once daily. In a further embodiment, the compound of Formula 1 is administered at a dose of 150 mg once daily. In a further embodiment, the compound of Formula 1 is administered at a dose of 250 mg once daily.
  • the compound of Formula 1 and the nucleos(t)ide analogue are co-administered.
  • the compound of Formula 1 and the nucleos(t)ide analogue are administered simultaneously, or they may be administered separately.
  • the combination or cotherapy further comprises another HBV antiviral.
  • Typical HBV antivirals include, but are not limited to immune modulators, nucleic acid polymers, short interfering RNAs or antisense oligonucleotides.
  • the method further comprises administering an immune modulator.
  • the immune modulator is interferon, for example interferon alpha or pegylated interferon alpha.
  • the 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 method further comprises administering at least one short interfering RNA (siRNA) or antisense oligonucleotide (ASO), more particularly at least one siRNA or ASO selected 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 present invention aims at reducing serum HBV DNA, serum HBV RNA, and/or quantitative serum HBsAg and HBeAg in patients.
  • the methods of treating HBV infection provided herein, in particular, treat HBV infection by reducing serum HBV DNA in a patient, by reducing serum HBV RNA in a patient and/or by reducing serum HBsAg and HBeAg in a patient and/or by inducing seroconversion (against sAg and/or eAg) in a patient. Therefore, in a particular embodiment, the present invention relates to a method of reducing HBsAg in a patient in need thereof, more particularly in a NT HBeAg+ patient in need thereof.
  • the present invention relates to a method of reducing HBsAg about at least 0.3 logio lll/mL in 24 weeks, more in particular about at least 0.4 logio lll/mL in 24 weeks.
  • the invention relates to a method of reducing HBV RNA, more particularly HBsAg RNA, in a patient in need thereof, more particularly in a NT HBeAg+ patient in need thereof.
  • the treatment is curative and the patient does not have to continue treatment after the specified treatment time.
  • the treatment is finite.
  • the (co-)administration of compound 1 is performed for an administration period of at least 24 weeks. In another embodiment, the (co-) administration of compound 1 is performed for an administration period of longer than 24 weeks. In yet another embodiment, compound 1 is (co-)administered for a duration of about 48 weeks. In embodiments, compound 1 is (co-)administered for a duration of longer than 48 weeks.
  • HBV infections that can be treated according to the disclosed methods include HBV genotype A, B, C, and/or D infections.
  • the methods disclosed can treat any HBV genotype (“pan-genotypic treatment”).
  • HBV genotyping can be performed using methods known in the art, for example, INNO-LIPA® HBV Genotyping, Innogenetics N.V., Ghent, Belgium).
  • Serum HBV DNA quantitation can be performed according to methods known in the art, for example, using the polymerase chain reaction (PCR)-based assay COBAS® TAQMAN® HBV Test v2.0 (Roche Diagnostics), which has been validated to quantify HBV DNA from serum samples for diverse HBV genotypes (A-H) including pre-core mutant HBV strains, with a reported lower limit of detection of 35 lll/mL and a linear dynamic range of quantitation of 1.7 x 10 2 to 8.5 x 10 8 lll/mL lll/mL, using the WHO pooled serum reference standard for quantitation.
  • PCR polymerase chain reaction
  • COBAS® TAQMAN® HBV Test v2.0 Roche Diagnostics
  • Serum HBsAg and HBeAg levels can be measured using for example, the investigational Abbott ARCHITECTTM assays (Abbott Laboratories; Abbott Park, IL, USA).
  • 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.
  • Compound 1 can be administered in a suitable composition, for example, in a tablet, as described in WO2019175657 (or the US counterparts thereof), which is hereby incorporated by reference in its entirety.
  • CHB-infected subject populations (1) subjects who were not being treated for their chronic hepatitis B (CHB) infection (including CHB treatment-naive subjects) and (2) subjects virologically suppressed by current NA treatment (either ETV or TDF as per local practice).
  • CHB chronic hepatitis B
  • NA represents entecavir (ETV) or tenofovir disoproxil fumarate (TDF)
  • HBV hepatitis B virus
  • NA nucleos(t)ide analog
  • ETV entercavir
  • TDF tenofovir disoproxil fumarate
  • Virologically suppressed participants received matching placebo along with NA (ETV or TDF) tablets orally for 24 weeks.
  • NA ETV or TDF
  • the eligible participants entered the extension phase and continue study drugs up to 48 weeks.
  • Virologically suppressed participants received compound 1 along with NA (ETV or TDF) tablets orally for 24 weeks.
  • NA ETV or TDF
  • the eligible participants entered the extension phase and continued study drugs up to 48 weeks.
  • participant 1 received compound 1 (250 mg dose), orally for 24 weeks.
  • the eligible participants entered the extension phase and received compound 1 along with NA (ETV or TDF) from Week 24 to Week 48.
  • NA ETV or TDF
  • Placebo Comparator Part B: Arm 7 (placebo + NA [ETV or TDF]) Participants with HBV currently not being treated received matching placebo along with NA (ETV or TDF) tablets orally for 24 weeks. The eligible participants entered the extension phase and continued study drugs up to 48 weeks.
  • Placebo Comparator Part B: Arm 9 (placebo + NA [ETV or TDF])
  • Virologically suppressed participants received matching placebo along with NA (ETV or TDF) tablets orally for 24 weeks.
  • NA ETV or TDF
  • the eligible participants entered the extension phase and continued study drugs up to 48 weeks.
  • Virologically suppressed participants received compound 1 tablet (250 mg dose) along with NA (ETV or TDF) tablets orally for 24 weeks. The eligible participants entered the extension phase and continued study drugs up to 48 weeks.
  • Part B ie, Treatment Arms 6 to 10
  • Part A Treatment Arms 6 to 10
  • Part B Treatment Arms 6 to 10
  • ⁇ HBsAg level at screening (310,000 versus ⁇ 10,000 lll/mL for HBeAg-positive subjects currently not being treated and 31 ,000 versus ⁇ 1 ,000 lll/mL for all other subjects).
  • the aim was to have approximately 40% HBeAg-positive subjects who were currently not being treated and 30% HBeAg-positive virologically suppressed subjects.
  • enrollment of HBeAg-negative subjects could be capped at 60% in subjects who were currently not being treated and at 70% in virologically suppressed subjects.
  • Part A started first and evaluated a 75-mg once-daily (qd) dose of compound 1.
  • the compound 1 dose to be administered in Part B of the study was 250 mg given once daily. This dose was selected, based on all safety, PK, and antiviral activity data available following completion of the highest compound 1 dose group (who received 250 mg once daily) in the Phase 1 study involving compound 1 in treatment-naive CHB-infected subjects treated for 28 days.
  • Each part of the study consisted of a screening phase (up to 8 weeks), a treatment phase (24 weeks or 48 weeks, depending on treatment response), and a post-treatment follow-up phase (24 weeks or 48 weeks, depending on treatment response).
  • a screening phase up to 8 weeks
  • a treatment phase 24 weeks or 48 weeks, depending on treatment response
  • a post-treatment follow-up phase 24 weeks or 48 weeks, depending on treatment response.
  • NA treatment either ETV or TDF as per local practice
  • was started in case of compound 1 monotherapy
  • continued in case NA treatment was part of the assigned study treatment
  • the duration of individual participation was approximately up to 56 weeks (subjects not eligible to continue treatment in the extension phase), up to 80 weeks (subjects continuing treatment in the extension phase but not meeting the treatment completion criteria), or up to 104 weeks (subjects meeting treatment completion criteria).
  • Subjects must be 18 to 70 years of age, inclusive.
  • Subjects must have not been receiving any CHB treatment at screening, ie, o Have never received treatment with HBV antiviral medicines, including NAs or interferon (IFN) products, OR o Have not been on treatment with HBV antiviral medicines, including NAs or IFN products, within 6 months prior to baseline (first intake of study drugs), AND ⁇ Subjects must have been HBeAg-positive and have HBV DNA 320,000 lll/mL, OR been HBeAg-negative and have HBV DNA 32,000 IU /ml_ at screening, AND
  • Subjects must have been virologically suppressed by current NA treatment (ETV or TDF) as defined by HBV DNA ⁇ 60 lll/mL at screening and at least 6 months prior to screening, AND
  • the current NA treatment could either be a branded product or a locally approved generic alternative (including different salt forms [eg, tenofovir maleate or succinate]).
  • a branded product or a locally approved generic alternative (including different salt forms [eg, tenofovir maleate or succinate]).
  • subjects would receive branded ETV (Baraclude®) or TDF (Viread®) treatment, as applicable.
  • liver disease of non-HBV etiology includes hepatitis virus infections mentioned above, drug- or alcohol-related liver disease, autoimmune hepatitis, hemochromatosis, Wilson’s disease, Gilbert’s syndrome, a-1 antitrypsin deficiency, primary biliary cirrhosis, primary sclerosing cholangitis, or any other non-HBV liver disease considered clinically significant by the investigator.
  • HCC hepatocellular carcinoma
  • DOSAGE AND ADMINISTRATION Compound 1 in this study was administered as 100 g and 25 mg tablets with the composition as described in WO2019175657, page 51, table 9, tablet (G009) and (G008).
  • Study drugs (compound 1/NA/placebo) were administered orally.
  • Compound 1 was provided as 25-mg and/or 100-mg tablets and placebo as matching tablets.
  • Subjects were instructed to take compound 1/placebo in the morning at approximately the same time each day, together with breakfast.
  • the tablets should be swallowed as a whole.
  • the study drug should be taken on site under the supervision of the study staff.
  • ETV Baraclude®
  • TDF Viread®
  • NA ETV or TDF
  • Baraclude® and Viread® were to be taken as per the package insert.
  • dosing interval for ETV or TDF may be adjusted as per the package insert.
  • HBV DNA and HBV RNA were quantified at a central laboratory using in vitro nucleic acid amplification tests for the quantification of HBV DNA and HBV RNA.
  • Samples for the determination of HBV DNA were processed in real-time using a test such as the COBAS® TaqMan® HBV Test, v2.0 (Roche Molecular Systems). Samples for the determination of HBV RNA could be analyzed in batch.
  • PHARMACOKINETIC EVALUATIONS Venous blood samples were collected for the determination of plasma concentrations of total compound 1 and/or NA (ETV or TDF), as applicable.
  • 24-hour urine sampling was performed in approximately 35% of all subjects (pharmacokinetic subgroup, at selected sites only) for the determination of compound 1 or NA (ETV or TDF) concentrations, as applicable, on Day 1 and Day 84 (Week 12) (0-2, 2-12, and 12-24 hours postdose).
  • compound 1 or NA ETV or TDF
  • CHB Chronic Hepatitis B
  • NT virologically suppressed
  • VS virologically suppressed
  • HBeAg+ or HBeAg- patient were randomized to 75mg or 250mg compound 1 qd or placebo (pbo) with a nucleos(t)ide analogue (NA; TDF/ETV) or received compound 1 alone for 24 weeks.
  • NA nucleos(t)ide analogue
  • BL baseline
  • NA nucleos(t)ide analogue
  • SD standard deviation
  • NT not treated at study start
  • VS virologically suppressed
  • ALT alanine aminotransferase
  • ULN upper limit of normal
  • HBeAg hepatitis B e antigen
  • HBV hepatitis B virus
  • IU international unit
  • HBsAg hepatitis B surface antigen
  • ⁇ DNA and RNA values in the VS subpopulation are mostly ‘imputed’ since values were below lower limit of quantification (LLOQ) or target not detected (TND), respectively.
  • LLOQ lower limit of quantification
  • TTD target not detected
  • HBV RNA* at week 24 In NT HBeAg+/- pts, compound 1 75mg and 250mg + NA showed a clear mean (SE) HBV RNA decline from baseline (BL) (2.96 (0.23) and 3.15 (0.33) copies/mL, respectively) compared with placebo + NA (1.33 (0.32) copies/mL), thereby differentiating compound 1 from NAs.
  • HBsAg were 8/23 (35%) vs 1/8 (13%) with NA+placebo
  • HBeAg were 19/23 (83%) vs 4/8 (50%), respectively ( Figures 4 and 5).
  • Compound 1 75 Compound 1 250 from baseline at Placebo + NA mg + NA mg + NA week 24, n (%) >0.3 logio IU/mL ⁇ 4/12 (33) 4/11 (36) 1/8 (13)
  • ALT flares occurred early on-treatment with compound 1 250mg + NA in NT HBeAg+ patients and were associated with HBsAg decline but not bilirubin increase, and resolved quickly with continuous treatment
  • AE adverse event
  • ALT alanine aminotransferase
  • AST aspartate aminotransferase
  • eGFR cr estimated glomerular filtration rate based on serum creatinine adjusted for body surface area (mL/min/1 73m 2 ).
  • Safetyty analyses were conducted with all available data, including VS patients receiving compound 1 75 mg or 250 mg and NT patients receiving compound 1 75 mg who had been treated beyond 24 weeks. **Most frequent Grade 3 or 4 AEs were gastrointestinal disorders (colitis and dyspepsia, placebo + NA) and investigations (ALT and ALT elevations in the compound 1 75 mg + NA and compound 1 250 mg + NA arms).
  • ⁇ Laboratory abnormalities were transient (Large fluctuations in eGFR occurred. Patients with eGFR 60 mL/min/1 73m 2 (lower limit of Grade 2) were included in the study and sometimes dipped to ⁇ 60 mL/min/1 .73 m 2 . Grade 3 creatine kinase increases were isolated and in many cases were linked to strenuous exercise.
  • ⁇ ALT/AST elevations were not associated with bilirubin increases.
  • Compound 1 in combination with NA achieved substantial HBV DNA and RNA reductions at week 24 in CHB patients.
  • Compound 1 + NA had a limited effect on mean HBsAg or HBeAg levels in HBeAg+ NT patients during the 24-week treatment period

Abstract

The present disclosure is directed to methods of using a capsid assembly inhibitor for the treatment of hepatitis B virus infection.

Description

TREATMENT OF HBV
BACKGROUND
Chronic hepatitis B virus (HBV) infection is a persistent, potentially progressive necroinflammatory liver disease associated with chronic HBV infection. Worldwide about 240- 400 million persons are chronically infected with HBV, and chronic 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)).
Compound 1 , a potent class N capsid assembly modulator, has been tested in phase 1 clinical trials and has been found well tolerated with dose-dependent PK and efficacy up to 250 mg orally qd for 4 weeks in treatment-naive chronic hepatitis B (CHB) patients (patients).
With the continued worldwide prevalence of HBV-associated mortality and severe morbidity, there remains a need for improved HBV antiviral therapies that can achieve sustained viral response during and after treatment.
SUMMARY
The present disclosure is directed to methods of using a capsid assembly inhibitor for the treatment of hepatitis B virus infection.
In an aspect, provided herein is a method of treating Hepatitis B viral infection in a not treated HBeAg+ patient comprising administering to the patient, a therapeutically effective amount of a combination of a nucleos(t)ide analogue and a compound of Formula 1
Figure imgf000002_0001
or a pharmaceutically acceptable salt thereof. In an aspect, provided herein is a method of treating Hepatitis B viral infection in a not treated HBeAg+ patient comprising administering to the patient, a therapeutically effective amount of a compound of Formula 1
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
In another aspect, provided herein is a method of treating Hepatitis B viral infection in a not treated HBeAg+ patient comprising: evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and treating the patient with a therapeutically effective amount of a combination of a nucleos(t)ide analogue and a compound of Formula 1
Figure imgf000003_0002
or a pharmaceutically acceptable salt thereof.
In another aspect, provided herein is a method of treating Hepatitis B viral infection in a not treated HBeAg+ patient comprising: evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and treating the patient with a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
In another aspect, provided herein is a combination of a nucleos(t)ide analogue and a compound of Formula 1
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof for use in cotherapy for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient.
In a further aspect, provided herein is a combination of a nucleos(t)ide analogue and a compound of Formula 1
Figure imgf000004_0002
or a pharmaceutically acceptable salt thereof, for use in the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient. In a yet further aspect, provided herein is a combination of a nucleos(t)ide analogue and a compound Formula 1
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof, for use in the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient, wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering the combination of a nucleos(t)ide analogue and compound 1 or a pharmaceutically acceptable salt thereof.
In a further aspect, provided herein is a compound Formula 1
Figure imgf000005_0002
or a pharmaceutically acceptable salt thereof, for use in the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient, wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering the compound of Formula 1 or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
In another aspect, provided herein is a combination as described herein, wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering a cotherapy by combination of a nucleos(t)ide analogue and compound 1 or a pharmaceutically acceptable salt thereof. In a yet further aspect, provided herein is a use of a combination of a nucleos(t)ide analogue and a compound Formula 1
Figure imgf000006_0001
or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient.
In a further aspect, provided herein is a use of a compound Formula 1
Figure imgf000006_0002
or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient, wherein the patient is also administered a nucleos(t)ide analogue.
In an additional aspect, provided herein is a use of a combination of a nucleos(t)ide analogue and a compound Formula 1
Figure imgf000006_0003
or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient, wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering the combination of a nucleos(t)ide analogue and compound 1 or a pharmaceutically acceptable salt thereof.
In a further aspect, provided herein is a use of a compound Formula 1
Figure imgf000007_0001
or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient, wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering the compound 1 or a pharmaceutically acceptable salt thereof in combination with a nucleos(t)ide analogue. Hepatitis B viral infection as referred to herein is, in particular, chronic hepatitis B viral infection.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 A depicts a schematic representation of the study design.
Fig. 1B depics a schematic representation of the study design to week 24. Fig. 2 depicts the mean standard error (SE) change in HBV DNA (assessed using
Roche COBAS HBV DNA assay) over 24 weeks of treatment from baseline in not treated at study start (NT) patients. HBV DNA lower limit of quantification (LLOQ): 20 lll/mL. (HBeAg: hepatitis B e antigen, NA: nucleos(t)ide analogue; IU: international unit). Fig. 3 depicts the mean (SE) change in HBV RNA over 24 weeks of treatment from baseline in NT and virologically suppressed (VS) patients (HBeAg+ and HBeAg-). HBV RNA LLOQ: 4.04 logio copies/mL. (TND: target not detected)
Fig. 4 depicts the mean (SE) change in hepatitis B surface antigen (HBsAg) over 24 wks of treatment from baseline by patient population. HBsAg LLOQ: 0.05 lU/mL and ULOQ:
>125000 ILI/mL.
Fig. 5 depicts the mean (SE) change in HBeAg over 24 weeks of treatment from baseline in HBeAg+ patients. HBeAg LLOQ: 0.11 ILI/mL and ULOQ: 1400 lU/mL.
Fig. 6 depicts the individual changes in A) HBsAg and B) HBeAg over 24 weeks of treatment from baseline in HBeAg+ NT patients.
DETAILED DESCRIPTION
The present disclosure is directed to methods of using a capsid assembly inhibitor for the treatment of hepatitis B virus infection. It has now been found that at 24 weeks, administration of compound 1 in combination with a nucleos(t)ide analogue, in not treated patients, increases suppression of HBV DNA, and HBV RNA in all patients, compared to nucleos(t)ide analogue alone. Unexpectedly, in not treated HBeAg+ patients, at 24 weeks, at an oral dose of 250 mg compound 1 daily a mean 0.4 logio HBsAg decline was observed.
Definitions
As used in the specification and in the claims, 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. However, such description should be construed as also describing compositions or processes as “consisting of” and “consisting essentially of” the enumerated compounds, which allows the presence of only the named compounds, along with any pharmaceutically acceptable carriers, and excludes other compounds.
All ranges disclosed herein are inclusive of the recited endpoint and independently combinable inclusive of the endpoints, and all the intermediate values. The endpoints of the ranges and any values disclosed herein are not limited to the precise range or value; they are sufficiently imprecise to include values approximating these ranges and/or values. As used herein, 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.
As used herein, the term “treatment” or “treating,” is defined as the application or administration of a therapeutic agent, i.e. , a combination of the invention (alone or in combination with another pharmaceutical agent), to a patient as defined herein, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect HBV infection, or the symptoms of HBV infection.
As used herein, 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.
As used herein, the term “pharmaceutically acceptable salt” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.
As used herein, the term “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.
As used herein, the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, 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. Some examples of materials that can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline; Ringer’s solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations. As used herein, “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. Supplementary active compounds can also be incorporated into the compositions. The “pharmaceutically acceptable carrier” can further include a pharmaceutically acceptable salt of the compound useful within the invention. Other additional ingredients that can be included in the pharmaceutical 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.
The term “stabilizer,” as used herein, refers to polymers capable of chemically inhibiting or preventing degradation of a Compound of Formula 1. Stabilizers are added to formulations of compounds to improve chemical and physical stability of the compound.
The term “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 administered independently, at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g., synergistic, effect.
As used herein, 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. Preferably, the patient, subject or individual is human.
As used herein, “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. “Not treated” refers to a patient that is not receiving any chronic hepatitis B treatment at screening, ie, has not received within 6 months prior to baseline treatment with HBV antiviral medicines, including nucleot(s)ide analogues (NAs) or interferon (IFN) products, or has not been on treatment with HBV antiviral medicines, including NAs or IFN products.
As used herein, “virologically suppressed” refers to patients treated with HBV antiviral therapy, in particular a nucleos(t)ide drug.
HBV infections that may be treated according to the disclosed methods include HBV genotype A, B, C, and/or D infections. However, in an embodiment, the methods disclosed may treat any HBV genotype (“pan-genotypic treatment”). HBV genotyping may be performed using methods known in the art, for example, INNO-LIPA® 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. C. and Talalay, P., Adv. Enzyme Regul. 22: 27-55 (1984) and Chou, Pharmacol. Rev. 58: 621-681 (2006). 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. In some embodiments, 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 each of the compound of Formula 1 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. For example, in an embodiment, the effect is complete or sustained reduction of viral load, HBsAg and/or anti-HBsAb during and/or after treatment. For example, in an embodiment, the effect is sustained virological response (SVR) and/or sustained viral clearance.
Nucleos(t)ide analogs can include deoxyadenosine analogues, adenosine analogues, deoxycytidine analogues, guanosine analogues, thymine analogues, and deoxyuridine analogues. In some embodiments, the nucleos(t)ide analogs can include Lamivudine (Lamivudine,3TC or LMV), Adefovir (Adefovir, ADV), Entecavir (Entecavir, ETV), Telbivudine (Telbivudine, LdT) and Tenofovir, or prodrugs thereof (e.g. Tenofovir disoproxil fumarate, TDF) and pharmaceutically acceptable salts thereof. Pharmaceutically acceptable prodrugs of tenofovir, for example, include tenofovir disoproxil fumarate, tenofovir disoproxil succinate, and tenofovir alafenamide fumarate, in particular tenofovir disoproxil fumarate and tenofovir alafenamide fumarate.
In an embodimentof the invention, the nucleos(t)ide analogue used in the invention can be selected from tenofovir, or a pharmaceutically acceptable salt or prodrug thereof, and entecavir, or a pharmaceutically acceptable salt thereof. In a particular embodiment, tenofovir or a pharmaceutically acceptable salt or prodrug thereof can be selected from tenofovir alafenamide, tenofovir disoproxil succinate and tenofovir disoproxil fumarate, more in particular tenofovir alafenamide and tenofovir disoproxil fumarate. In a particular embodiment, entecavir or a pharmaceutically acceptable salt thereof is entecavir monohydrate.
In an embodiment, the nucleos(t)ide analogue used in the invention is administered at the approved dose/dosage. In a particular embodiment, tenofovir disoproxil fumarate is administered in an amount of 60-600 mg. In another particular embodiment, tenofovir disoproxil fumarate is administered in an amount of 300 mg. In another particular embodiment, tenofovir alafenamide is administered in an amount of 25 mg. In yet another particular embodiment of the method, the entecavir monohydrate is administered in an amount of 0.1-1 mg. In still another particular embodiment of the method, entecavir monohydrate is administered in an amount of 0.5 mg. In a particular embodiment, the nucleos(ti)de analogue is tenofovir or a produg thereof, in particular, tenofovir alafenamide, or tenofovir disoproxil fumarate.
In some embodiments, the amount of the compound of Formula 1 in the invention is from about 50 mg per day to about 500 mg per day (e.g. 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500 mg). In some embodiments, the amount of a compound of Formula 1 is from 75 mg per day to 250 mg per day. In some embodiments, the amount of a Compound of Formula 1 is 75 mg per day. In some embodiments, the amount of a Compound of Formula 1 is 150 mg per day. In some embodiments, the amount of a Compound of Formula 1 is 250 mg per day.
In some embodiments, the compound of Formula 1 is administered at a dose of 75- 300mg, more particularly 80-300mg, more particularly 100-300mg, more particularly 100-250mg once daily. In a further embodiment, the compound of Formula 1 is administered at a dose of 75 mg once daily. In a further embodiment, the compound of Formula 1 is administered at a dose of 150 mg once daily. In a further embodiment, the compound of Formula 1 is administered at a dose of 250 mg once daily.
In some embodiments, the compound of Formula 1 and the nucleos(t)ide analogue are co-administered. In particular, the compound of Formula 1 and the nucleos(t)ide analogue are administered simultaneously, or they may be administered separately.
In a particular embodiment of the method, the combination or cotherapy further comprises another HBV antiviral. Typical HBV antivirals include, but are not limited to immune modulators, nucleic acid polymers, short interfering RNAs or antisense oligonucleotides.
In an embodiment of the method, the method further comprises administering an immune modulator. In an embodiment of the method, the immune modulator is interferon, for example interferon alpha or pegylated interferon alpha.
In some embodiments, the 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.
In some embodiments, the method further comprises administering at least one short interfering RNA (siRNA) or antisense oligonucleotide (ASO), more particularly at least one siRNA or ASO selected 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.
The present invention aims at reducing serum HBV DNA, serum HBV RNA, and/or quantitative serum HBsAg and HBeAg in patients. The methods of treating HBV infection provided herein, in particular, treat HBV infection by reducing serum HBV DNA in a patient, by reducing serum HBV RNA in a patient and/or by reducing serum HBsAg and HBeAg in a patient and/or by inducing seroconversion (against sAg and/or eAg) in a patient. Therefore, in a particular embodiment, the present invention relates to a method of reducing HBsAg in a patient in need thereof, more particularly in a NT HBeAg+ patient in need thereof. More in particular, the present invention relates to a method of reducing HBsAg about at least 0.3 logio lll/mL in 24 weeks, more in particular about at least 0.4 logio lll/mL in 24 weeks. Alternatively, or complementarily, the invention relates to a method of reducing HBV RNA, more particularly HBsAg RNA, in a patient in need thereof, more particularly in a NT HBeAg+ patient in need thereof.
In certain embodiments of the methods of treating HBV infection provided herein, the treatment is curative and the patient does not have to continue treatment after the specified treatment time. In a particular embodiment of the method of treating HBV provided herein, the treatment is finite.
In some embodiments, the (co-)administration of compound 1 is performed for an administration period of at least 24 weeks. In another embodiment, the (co-) administration of compound 1 is performed for an administration period of longer than 24 weeks. In yet another embodiment, compound 1 is (co-)administered for a duration of about 48 weeks. In embodiments, compound 1 is (co-)administered for a duration of longer than 48 weeks.
HBV infections that can be treated according to the disclosed methods include HBV genotype A, B, C, and/or D infections. However, in an embodiment, the methods disclosed can treat any HBV genotype (“pan-genotypic treatment”). HBV genotyping can be performed using methods known in the art, for example, INNO-LIPA® HBV Genotyping, Innogenetics N.V., Ghent, Belgium).
Serum HBV DNA quantitation can be performed according to methods known in the art, for example, using the polymerase chain reaction (PCR)-based assay COBAS® TAQMAN® HBV Test v2.0 (Roche Diagnostics), which has been validated to quantify HBV DNA from serum samples for diverse HBV genotypes (A-H) including pre-core mutant HBV strains, with a reported lower limit of detection of 35 lll/mL and a linear dynamic range of quantitation of 1.7 x 102 to 8.5 x 108 lll/mL lll/mL, using the WHO pooled serum reference standard for quantitation.
Serum HBsAg and HBeAg levels can be measured using for example, the investigational Abbott ARCHITECT™ assays (Abbott Laboratories; Abbott Park, IL, USA). 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.
Pharmaceutical Compositions and Kits
Compound 1, including the synthesis thereof, is disclosed in PCT Publication No. WO/2014/184350 (or the US counterparts thereof), which is hereby incorporated by reference in its entirety.
Compound 1 can be administered in a suitable composition, for example, in a tablet, as described in WO2019175657 (or the US counterparts thereof), which is hereby incorporated by reference in its entirety.
The following examples are merely illustrative and are not intended to limit the disclosure to the materials, conditions, or process parameters set forth therein.
EXAMPLES
EXAMPLE 1: OVERVIEW OF STUDY DESIGN
This was an multicenter, interventional, randomized, partially-blind, placebo-controlled, two-part study to assess the efficacy, safety, and pharmacokinetics of treatment with two doses of compound 1 administered as monotherapy and in combination with NA (either entecavir (ETV) or tenofovir disoproxil fumarate (TDF) as per local practice) in HBeAg-positive and -negative CHB-infected subjects. Subjects who completed the initial 24 weeks of treatment with a virologic response by Week 20 (HBV DNA <200 lU/mL of the HBV DNA assay) and without experiencing any safety concerns precluding continued study drug treatment as determined by the investigator, continued study drugs up to 48 weeks in a treatment extension phase.
The study initially consisted of 2 parts, Part A and Part B. For each part, a sample size of 110 subjects was targeted who would receive compound 1 at two dose levels as monotherapy or in combination with an NA. Additional parts could be added through a protocol amendment based on emerging data.
Each part consisted of 2 types of CHB-infected subject populations: (1) subjects who were not being treated for their chronic hepatitis B (CHB) infection (including CHB treatment-naive subjects) and (2) subjects virologically suppressed by current NA treatment (either ETV or TDF as per local practice).
Study Arms (NA represents entecavir (ETV) or tenofovir disoproxil fumarate (TDF)): • Experimental: Part A: Arm 1 (compound 1 or NA) (open label)
Participants with hepatitis B virus (HBV) currently not being treated and receiving compound 1 tablet (75 mg dose) orally for 24 weeks, stopped further dosing with compound and start treatment with nucleos(t)ide analog (NA) (entecavir [ETV] or tenofovir disoproxil fumarate [TDF]), and entered the 24 week post treatment follow-up phase.
• Placebo Comparator: Part A: Arm 2 (Placebo+NA [ETV] or [TDF])
Participants with HBV currently not being treated received matching placebo along with NA (ETV or TDF) tablets orally for 24 weeks. The eligible participants entered the extension phase and continue study drugs up to 48 weeks.
• Experimental: Part A: Arm 3 (Compound 1 + NA [ETV or TDF])
Participants with HBV currently not being treated received compound 1 along with NA (ETV or TDF) tablet orally for 24 weeks. The eligible participants entered the extension phase and continued study drugs up to 48 weeks.
• Placebo Comparator: Part A: Arm 4 (Placebo + NA [ETV or TDF])
Virologically suppressed participants received matching placebo along with NA (ETV or TDF) tablets orally for 24 weeks. The eligible participants entered the extension phase and continue study drugs up to 48 weeks.
• Experimental: Part A: Arm 5 (Compound 1 + NA [ETV or TDF])
Virologically suppressed participants received compound 1 along with NA (ETV or TDF) tablets orally for 24 weeks. The eligible participants entered the extension phase and continued study drugs up to 48 weeks.
• Experimental: Part B: Arm 6 (Compound 1 + NA [ETV or TDF]) (open label)
Participants with HBV currently not being treated received compound 1 (250 mg dose), orally for 24 weeks. The eligible participants entered the extension phase and received compound 1 along with NA (ETV or TDF) from Week 24 to Week 48.
Placebo Comparator: Part B: Arm 7 (placebo + NA [ETV or TDF]) Participants with HBV currently not being treated received matching placebo along with NA (ETV or TDF) tablets orally for 24 weeks. The eligible participants entered the extension phase and continued study drugs up to 48 weeks.
• Experimental: Part B: Arm 8 (compound 1 + NA [ETV or TDF])
Participants with HBV currently not being treated received compound 1 tablet (250 mg dose) along with NA (ETV or TDF) tablets orally for 24 weeks. The eligible participants entered the extension phase and continue study drugs up to 48 weeks.
• Placebo Comparator: Part B: Arm 9 (placebo + NA [ETV or TDF])
Virologically suppressed participants received matching placebo along with NA (ETV or TDF) tablets orally for 24 weeks. The eligible participants entered the extension phase and continued study drugs up to 48 weeks.
Interventions:
• Experimental: Part B: Arm 10 (Compound 1 + NA [ETV or TDF])
Virologically suppressed participants received compound 1 tablet (250 mg dose) along with NA (ETV or TDF) tablets orally for 24 weeks. The eligible participants entered the extension phase and continued study drugs up to 48 weeks.
PART A:
Approximately 70 subjects who were currently not being treated for their CHB infection were randomized in a 3:1 :3 ratio to receive one of the following treatments:
□ Treatment Arm 1: Compound 1 (N=30) (open-label)
After full recruitment, this treatment arm was discontinued early per Protocol Amendment 4. All subjects were to stop further dosing with compound 1 75 mg, start treatment with NA, and enter the post-treatment follow-up phase.
□ Treatment Arm 2: Placebo + NA (ETV or TDF) (N=10)
□ Treatment Arm 3: compound 1 + NA (ETV or TDF) (N=30)
At the same time, approximately 40 subjects who were virologically suppressed by current NA treatment were randomized in a 1:3 ratio to receive one of the following treatments: □ Treatment Arm 4: Placebo + NA (ETV or TDF) (N=10)
□ Treatment Arm 5: compound 1 + NA (ETV or TDF) (N=30)
PART B:
Part B (ie, Treatment Arms 6 to 10) followed the same format as Part A, with a different dose of compound 1.
Randomization in each part was stratified by:
□ HBeAg status at screening (positive versus negative);
□ HBsAg level at screening (³10,000 versus <10,000 lll/mL for HBeAg-positive subjects currently not being treated and ³1 ,000 versus <1 ,000 lll/mL for all other subjects).
The aim was to have approximately 40% HBeAg-positive subjects who were currently not being treated and 30% HBeAg-positive virologically suppressed subjects. To this end, enrollment of HBeAg-negative subjects could be capped at 60% in subjects who were currently not being treated and at 70% in virologically suppressed subjects.
Part A started first and evaluated a 75-mg once-daily (qd) dose of compound 1. The compound 1 dose to be administered in Part B of the study was 250 mg given once daily. This dose was selected, based on all safety, PK, and antiviral activity data available following completion of the highest compound 1 dose group (who received 250 mg once daily) in the Phase 1 study involving compound 1 in treatment-naive CHB-infected subjects treated for 28 days.
Each part of the study consisted of a screening phase (up to 8 weeks), a treatment phase (24 weeks or 48 weeks, depending on treatment response), and a post-treatment follow-up phase (24 weeks or 48 weeks, depending on treatment response). A high-level overview of the study design is provided in Figure 1.
All subjects were planned to be treated for at least 24 weeks. Following this initial treatment period, subjects who completed the 24-week treatment period with virologic response by Week 20 (HBV DNA <200 lll/mL of the HBV DNA assay) and without experiencing any safety concerns precluding continued study drug treatment as determined by the investigator, would continue study drugs up to 48 weeks in a treatment extension phase. Subjects choosing not to participate in the extension phase would complete their assigned 24-week treatment after which they would enter the 24-week follow-up period. Per Protocol Amendment, subjects in the compound 1 250 mg monotherapy arm who were eligible for treatment extension, based on the treatment extension criteria assessed at Week 20, would receive compound 1 in combination with NA treatment from Week 24 to Week 48.
In addition, a futility rule for this 250 mg monotherapy arm was installed: if ³1 subject in the 250 mg monotherapy arm experienced virological breakthrough during the first 24 weeks of treatment, NA treatment would be added to compound 1 treatment as soon as possible for all remaining subjects. These subjects would continue the visit schedule as planned. Subjects with virological breakthrough would discontinue compound 1 treatment and would start NA treatment per protocol.
For subjects who were not eligible to participate in the treatment extension phase:
- Subjects were considered to have completed the study treatment if they had completed 24 weeks of study drug administration, after which they discontinued compound 1 or placebo treatment. NA treatment (either ETV or TDF as per local practice) was started (in case of compound 1 monotherapy) or continued (in case NA treatment was part of the assigned study treatment) as per local treatment guidelines.
- Subjects were considered to have completed the study if they have completed the assessments of the end of study visit (Follow-up [FU] Week 24).
- For subjects who did not continue or start NA treatment at Week 24, an additional 24-week follow-up (as per local treatment guidelines) by their primary care physician, outside of the study, was recommended after study completion.
For subjects who continued treatment in the extension phase:
- Subjects were considered to have completed the study treatment if they had completed 48 weeks of study drug administration, after which they discontinued compound 1 or placebo treatment.
At Week 48: o Subjects who met all of the Individual Subject Treatment Completion Criteria completed all treatment. These subjects were followed-up until 48 weeks after end of trial (EOT). If, during the 48-week follow-up, a subject mets one of the re-treatment criteria, NA treatment (ETV or TDF) was re-started. o Subjects who did not meet the Individual Subject Treatment Completion Criteria would start (in case of compound 1 monotherapy) or continue (in case NA treatment was part of the assigned study treatment) NA treatment as per local treatment guidelines. These subjects were followed-up until 24 weeks after the end of study treatment.
- Subjects were considered to have completed the study if they had completed the assessments of the end of study visit (FU Week 24 or FU Week 48).
- For subjects who did not meet the Individual Subject Treatment Completion Criteria and did not start or continue NA treatment at Week 48, an additional 24-week follow-up (as per local treatment guidelines) by their primary care physician, outside of the study, was recommended after study completion.
The duration of individual participation was approximately up to 56 weeks (subjects not eligible to continue treatment in the extension phase), up to 80 weeks (subjects continuing treatment in the extension phase but not meeting the treatment completion criteria), or up to 104 weeks (subjects meeting treatment completion criteria).
The study was considered completed with the last visit of the last subject participating in the study.
SUBJECT POPULATION
KEY INCLUSION CRITERIA
□ Subjects must be 18 to 70 years of age, inclusive.
□ Subjects must have CHB infection documented by:
□ Serum HBsAg-positive at screening and serum HBsAg- or HBV DNA-positive at least 6 months prior to screening;
□ Serum immunoglobulin M (IgM) anti-HBc antibody negative at screening.
□ In subjects currently not being treated (Treatment Arms 1-2-3 and 6-7-8):
□ Subjects must have not been receiving any CHB treatment at screening, ie, o Have never received treatment with HBV antiviral medicines, including NAs or interferon (IFN) products, OR o Have not been on treatment with HBV antiviral medicines, including NAs or IFN products, within 6 months prior to baseline (first intake of study drugs), AND □ Subjects must have been HBeAg-positive and have HBV DNA ³20,000 lll/mL, OR been HBeAg-negative and have HBV DNA ³2,000 IU /ml_ at screening, AND
□ Subjects must have HBsAg >250 lll/mL at screening, AND
□ Subjects must have ALT > upper limit of normal (ULN) and £5 x ULN at screening, determined in the central laboratory.
Note: If subjects were treated with investigational anti-HBV agents more than 6 months before screening, the sponsor had to be contacted to discuss the case. Subjects who had received treatment with a CAM for more than 4 weeks any time prior to screening were excluded.
In virologically suppressed subjects (Treatment Arms 4-5 and 9-10):
□ Subjects must have been virologically suppressed by current NA treatment (ETV or TDF) as defined by HBV DNA <60 lll/mL at screening and at least 6 months prior to screening, AND
□ Subjects must have been on the same NA treatment (ETV or TDF) and the same dose for ³12 months prior to screening, AND
□ Subjects must have had HBsAg >250 lll/mL at screening, AND
□ Subjects must have had ALT £2x ULN at screening.
Note: If subjects were treated with investigational anti-HBV agents more than 6 months before screening, the sponsor had to be contacted to discuss the case. Subjects who had received treatment with a CAM for more than 4 weeks any time prior to screening were excluded.
Note: The current NA treatment could either be a branded product or a locally approved generic alternative (including different salt forms [eg, tenofovir maleate or succinate]). During the study, subjects would receive branded ETV (Baraclude®) or TDF (Viread®) treatment, as applicable.
□ Subjects must have had:
□ A liver biopsy result classified as Metavir F0-F2 within 1 year prior to screening or at the time of screening, OR
□ FibroScan™ liver stiffness measurement <8.0 kPa within 6 months prior to screening or at the time of screening. KEY EXCLUSION CRITERIA
□ Subjects who tested positive for anti-HBs antibodies.
□ Subjects with current hepatitis A virus infection (confirmed by hepatitis A antibody IgM), hepatitis D virus (HDV) infection (confirmed by HDV antibody), hepatitis E virus infection (confirmed by hepatitis E antibody IgM), or human immunodeficiency virus type 1 (HIV-1) or HIV-2 infection (confirmed by antibodies) at screening; subjects with a history of or current HCV infection (confirmed by HCV antibody). Evidence of other active infection (bacterial, viral, fungal, including acute tuberculosis) deemed clinically relevant by the investigator that would interfere with study conduct or its interpretation would also lead to exclusion.
□ Subjects with any evidence of hepatic decompensation at any time point prior to or at the time of screening:
□ Direct bilirubin >1 2x ULN, or
□ International normalized ratio >1.5x ULN, or
□ Serum albumin < lower limit of normal (LLN), or
□ Documented history or current evidence of variceal bleeding, ascites, or hepatic encephalopathy.
□ Subjects with any evidence of liver disease of non-HBV etiology. This included but was not limited to hepatitis virus infections mentioned above, drug- or alcohol-related liver disease, autoimmune hepatitis, hemochromatosis, Wilson’s disease, Gilbert’s syndrome, a-1 antitrypsin deficiency, primary biliary cirrhosis, primary sclerosing cholangitis, or any other non-HBV liver disease considered clinically significant by the investigator.
□ Subjects who had signs of hepatocellular carcinoma (HCC) on an abdominal ultrasound performed within 2 months prior to screening or at the time of screening. In case of suspicious findings on conventional ultrasound the subject could still be eligible if HCC had been ruled out by a more specific imaging procedure (contrast enhanced ultrasound, computed tomography [CT] or magnetic resonance imaging [MRI]).
□ Subjects who had received an organ transplant.
DOSAGE AND ADMINISTRATION Compound 1 in this study was administered as 100 g and 25 mg tablets with the composition as described in WO2019175657, page 51, table 9, tablet (G009) and (G008).
Study drugs (compound 1/NA/placebo) were administered orally. Compound 1 was provided as 25-mg and/or 100-mg tablets and placebo as matching tablets. Subjects were instructed to take compound 1/placebo in the morning at approximately the same time each day, together with breakfast.
The tablets should be swallowed as a whole. At each study visit, the study drug should be taken on site under the supervision of the study staff.
Subjects received commercially available Baraclude® (ETV) or Viread® (TDF) during the study (provided through this study), as applicable. Subjects who were already being treated with Baraclude® or Viread® at screening, continued this NA treatment. Subjects were switched to these branded NA treatments for the course of the study in case they are on another originator branded or a generic ETV or TDF treatment, respectively, at screening.
NA (ETV or TDF) was to be taken as per the package insert (Baraclude® and Viread®, respectively). In the event of decline in renal function, dosing interval for ETV or TDF may be adjusted as per the package insert.
PRIMARY OUTCOME MEASURE
Change from baseline in Hepatitis B surface antigen (HBsAg) levels at Week 24.
EFFICACY EVALUATIONS
Qualitative and quantitative HBsAg and HBeAg, and quantitative HBcrAg as well as anti-HBs and anti-HBe antibodies were determined using standard serologic assays in a central laboratory. Samples for the determination of HBsAg and HBeAg were processed in real-time using an assay such as the ARCHITECT platform (Abbott Laboratories). Samples for the determination of HBcrAg could be analysed in batch.
HBV DNA and HBV RNA were quantified at a central laboratory using in vitro nucleic acid amplification tests for the quantification of HBV DNA and HBV RNA. Samples for the determination of HBV DNA were processed in real-time using a test such as the COBAS® TaqMan® HBV Test, v2.0 (Roche Molecular Systems). Samples for the determination of HBV RNA could be analyzed in batch.
PHARMACOKINETIC EVALUATIONS Venous blood samples were collected for the determination of plasma concentrations of total compound 1 and/or NA (ETV or TDF), as applicable.
24-hour urine sampling was performed in approximately 35% of all subjects (pharmacokinetic subgroup, at selected sites only) for the determination of compound 1 or NA (ETV or TDF) concentrations, as applicable, on Day 1 and Day 84 (Week 12) (0-2, 2-12, and 12-24 hours postdose).
This 24-hour sampling schedule required an overnight stay on site.
RESULTS - Week 24 antiviral activity, safety and pharmacokinetics results of a phase 2 study of compound 1, a capsid assembly modulator, administered in combination with a nucleos(t)ide analogue in patients with chronic hepatitis B
METHODS: Chronic Hepatitis B (CHB) not treated (NT) and virologically suppressed (VS), HBeAg+ or HBeAg- patient were randomized to 75mg or 250mg compound 1 qd or placebo (pbo) with a nucleos(t)ide analogue (NA; TDF/ETV) or received compound 1 alone for 24 weeks. Primary endpoint was change from baseline (BL) in HBsAg at week 24. Only data from the combination arms are presented here.
RESULTS: Of 172 patients in the combination arms (88 NT; 84 VS; age 18-65 years, mostly male (73%)), 48% were Asian and 34% HBeAg+.
Table 1: Baseline Demographics and Disease Characteristics
Figure imgf000025_0001
NA, nucleos(t)ide analogue; SD, standard deviation; NT, not treated at study start; VS, virologically suppressed; ALT, alanine aminotransferase; ULN, upper limit of normal; HBeAg, hepatitis B e antigen; HBV, hepatitis B virus; IU, international unit; HBsAg, hepatitis B surface antigen
Black or African American, Native Hawaiian or Other Pacific Islander. †Metavir fibrosis stage was assessed by elastography; N=21 , 20, 28, 32, 28 and 28 in the respective subgroups.
†DNA and RNA values in the VS subpopulation are mostly ‘imputed’ since values were below lower limit of quantification (LLOQ) or target not detected (TND), respectively. Mean Change in HBV DNA at week 24
In NT HBeAg+ patients (mean baseline HBV DNA levels 7.65 to 8.24 logio lll/mL), there pronounced declines in mean (SE) HBV DNA from baseline of 5.53 (0.23) and 5.88 (0.34) logio lll/mL for compound 1 75 mg and 250 mg + NA, respectively, and 5.21 (0.42) for placebo + NA (Figure 2). In NT HBeAg- patients (mean baseline HBV DNA levels 4.89 to 5.40 logio lU/mL), interpretation of mean (SE) HBV DNA decline was confounded since many patients (Figure 2) had HBV DNA <LLOQ from Week 4 onwards.
Patients with HBV
Compound 1 75 Compound 1 250
DNA<LLOQ at Placebo + NA mg + NA mg + NA week 24, n (%)
Figure imgf000026_0001
Mean Change in HBV RNA* at week 24 In NT HBeAg+/- pts, compound 1 75mg and 250mg + NA showed a clear mean (SE) HBV RNA decline from baseline (BL) (2.96 (0.23) and 3.15 (0.33) copies/mL, respectively) compared with placebo + NA (1.33 (0.32) copies/mL), thereby differentiating compound 1 from NAs. HBV RNA was target not detected (TND) at week 24 in 19/30 (58%), 23/30 (77%) and 9/21 (43%) patients, respectively in the 75 mg, 250 mg and placebo groups. All compound 1 VS patients with detectable HBV RNA at BL achieved HBV RNA TND at week 24 vs 1/7 (14%) with placebo +
NA patients (Figure 3).
HBV Compd Compd
Compd Compd
RNA 1 250 Placebo HBV RNA 1 250 Placebo
Figure imgf000026_0002
Week 19/33 23/30 Baseline 19/32 21/29 14/21
9/21 (43)
24 (58) (77) (BL (59) (72) (67) Week 24
(when 13/13 8/8
1/7 (14) detectable (100) (100)
_ at BL) _
* assessed using a quantitative reverse transcription polymerase chain reaction assay.
Mean Change in HBsAg* and HBeAg
In NT HBeAg+ patients, compound 1 75 mg and 250 mg + NA resulted in a mean (SE) HBsAg decline of 0.13 (0.10) and 0.40 [0.15] logio lll/mL), respectively, compared with 0.22 (0.11) for NA + placebo at week 24 (Figure 4). Patients with HBsAg declines also had HBeAg declines (Figures 4-6) and frequently early on treatment isolated ALT flares.
In both compound 1 + NA arms:
Maximal individual HBsAg and HBeAg reductions were 1.28 and 1.8 logio lU/mL at week 24, respectively (Figure 6). Proportion of NT HBeAg+ patients with >0.3 logio lU/mL reductions from BL in:
HBsAg were 8/23 (35%) vs 1/8 (13%) with NA+placebo
HBeAg were 19/23 (83%) vs 4/8 (50%), respectively (Figures 4 and 5).
Proportion of VS HBeAg+ patients with >0.3 logio lU/mL reductions in HBeAg was 7/19 (37%) vs 1/5 (20%) NA+placebo (Figure 5).
NT HBeAg+ patients
HBsAg reduction
Compound 1 75 Compound 1 250 from baseline at Placebo + NA mg + NA mg + NA week 24, n (%) >0.3 logio IU/mL 4/12 (33) 4/11 (36) 1/8 (13)
>0.5 logio lU/mL 0/12 4/11 (36) 1/8 (13)
Figure imgf000027_0001
*Assessed using an Abbott Architect™ assay; HBsAg lower limit of quantification: 0.05 lll/mL and upper limit of quantification (ULOQ): >125000 lll/mL. †Proportions of VS HBeAg+ patients with >0.3 logio reductions from baseline at 24 weeks in HBsAg were: 2/9 (22%) (compound 1 75 mg + NA), 0/10 (compound 1 250 mg + NA) and 0/5 (placebo + NA).
No NT or VS HBeAg- patients had >0.3 logio reductions from baseline at 24 weeks in HBsAg. †After retesting of the baseline sample (1/2 dilution), one additional compound 1 250 mg + NA treated patient and one additional Placebo + NA treated patient had >0.3 logio lll/mL reduction from baseline in HBsAg levels.
Figure imgf000029_0001
*Assessed using an Abbott Architect™ assay; HBeAg lower limit of quantification: 0.11 lll/mL and upper limit of quantification (ULOQ): 1400 lll/mL.
After retesting of the baseline sample (1/10 dilution), one additional Placebo + NA treated patient had >0.3 Iog10 lll/mL reduction from baseline in HBeAg levels.
Viral breakthroughs
There were no viral breakthroughs in the compound 1+NA combination arms.
Confirmed viral breakthrough (VBT; confirmed >1 logio lll/mL increase in HBV DNA from nadir) occurred in 5/28 patients on compound 1 75 mg monotherapy These patients had emerging high-level resistant variant T33N (compound 1 fold change (FC) of 85).
One patient on compound 1 250 mg monotherapy with non-response (<1 logio lll/mL decline from baseline at week 4 had subsequent VBT. This patient (genotype E) carrie I105T baseline polymorphism (FC of 2.7) and had no emerging mutations at HBV core protein positions of interest. Safety
Most treatment-emergent adverse events (AEs) were Grade 1 or 2 (Table 2), and no study treatment related serious adverse events occurred.
There were no clinically significant changes in laboratory parameters, including hematologic parameters and renal function (Table 2)
ALT flares occurred early on-treatment with compound 1 250mg + NA in NT HBeAg+ patients and were associated with HBsAg decline but not bilirubin increase, and resolved quickly with continuous treatment
Transient, Grade 3 (<60 ml/min/1.73m2) decreases in eGFRcrwere observed in each treatment group.
Table 2: Summary of Treatment-Emergent Adverse Events and Laboratory Abnormalities*
Figure imgf000030_0001
AE: adverse event; ALT: alanine aminotransferase; AST: aspartate aminotransferase; eGFRcr: estimated glomerular filtration rate based on serum creatinine adjusted for body surface area (mL/min/1 73m2). *Safety analyses were conducted with all available data, including VS patients receiving compound 1 75 mg or 250 mg and NT patients receiving compound 1 75 mg who had been treated beyond 24 weeks. **Most frequent Grade 3 or 4 AEs were gastrointestinal disorders (colitis and dyspepsia, placebo + NA) and investigations (ALT and ALT elevations in the compound 1 75 mg + NA and compound 1 250 mg + NA arms).
†Serious AEs (none related to study drugs) were colitis (n=1 , placebo + NA); streptococcal toxic shock syndrome, acute cardiac failure, myocarditis, muscle necrosis (n=1), post-traumatic neck syndrome (n=1) and lymphadenitis (n=1) (compound 1 75 mg + NA) and appendicitis, ligament rupture, ovarian haemorrhage and intervertebral disc protrusion (each n=1 ; compound 1 250 mg + NA).
†AEs leading to treatment discontinuation were abdominal discomfort/gastrointestinal upset (n=1 , placebo + NA) and streptococcal toxic shock syndrome, acute cardiac failure, myocarditis and muscle necrosis (n=1 ; compound 1 75 mg + NA).
^Laboratory abnormalities were transient (Large fluctuations in eGFR occurred. Patients with eGFR 60 mL/min/1 73m2 (lower limit of Grade 2) were included in the study and sometimes dipped to <60 mL/min/1 .73 m2. Grade 3 creatine kinase increases were isolated and in many cases were linked to strenuous exercise.
§ALT/AST elevations were not associated with bilirubin increases.
Pharmacokinetics (PK)
After repeated once daily (qd) dosing with 75 and 250 g of compound 1 (up to 24 weeks), observed plasma concentrations were dose proportional and within the range predicted by the population PK model.
Comparing plasma concentrations and PK parameters between the compound 1 monotherapy arms and combination arms with an NA (ETV and TDF), a PK drug-drug interaction between ETV and compound 1 was not apparent. A PK-drug drug interaction between TDF (TFV) and compound 1 was apparent.
CONCLUSIONS:
Compound 1 in combination with NA achieved substantial HBV DNA and RNA reductions at week 24 in CHB patients.
Compound 1 + NA had a limited effect on mean HBsAg or HBeAg levels in HBeAg+ NT patients during the 24-week treatment period
- Compound 1 250 mg + NA resulted in a mean 0.4 Iog10 lll/mL HBsAg decline in HBeAg+ NT patients - Patients with HBsAg decline mostly had HBeAg declines and frequently early on-treatment isolated ALT flares.
• Due to cases of viral breakthrough, Compound 1 monotherapy will not be developed further. No viral breakthrough was observed in the Compound 1 + NA combination arms. · Compound 1 either at 75 mg or 250 mg doses was well tolerated and demonstrated target engagement.
The disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated herein by reference in their entirety.
While the invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of this invention can be devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims are intended to be construed to include all such embodiments and equivalent variations.

Claims

1. A method of treating Hepatitis B viral infection in a not treated HBeAg+ patient comprising administering to the patient, a therapeutically effective amount of a compound of Formula 1
Figure imgf000033_0001
or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
2. The method according to claim 1, comprising: evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and treating the patient with a therapeutically effective amount of a compound of Formula 1
Figure imgf000033_0002
or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
The method according to claim 1 or 2, wherein the nucleos(ti)de analogue is selected from the group consisting of tenofovir, or a pharmaceutically acceptable salt or prodrug thereof, and entecavir, or a pharmaceutically acceptable salt thereof.
The method according to claim 3, wherein the nucleos(ti)de analogue is tenofovir or a produg thereof, in particular, tenofovir alafenamide, tenofovir disoproxil fumarate or tenofovir disoproxil succinate.
5. The method according to any one of claims 1 to 4, wherein the treatment is performed for a period of at least 24 weeks.
6. The method according to any one of claims 1 to 5, wherein the treatment is performed for a period of 48 weeks.
7. The method according to any one of claims 1 to 6, wherein the combination further comprises another HBV antiviral.
8. The method according to claim 7, wherein the additional HBV antiviral is selected from the group consisting of an immune modulator, e.g. interferon alpha or pegylated interferon alpha, a nucleic acid polymer, a short interfering RNA or an antisense oligonucleotide.
9. The method according to any one of claims 1 to 8, wherein compound 1 is administered at a dose of 100-300mg once daily.
10. A combination of a nucleos(t)ide analogue and a compound of Formula 1
Figure imgf000034_0001
or a pharmaceutically acceptable salt thereof for use in cotherapy for the treatment of Hepatitis B viral infection in a not treated HBeAg+ patient.
11. The combination of claim 10, wherein the treatment comprises evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering a cotherapy by combination of a nucleos(t)ide analogue and compound 1 or a pharmaceutically acceptable salt thereof.
12. A method of reducing HBsAg in a patient in need thereof, more particularly in a not treated HBeAg+ patient in need thereof, comprising administering to the patient, a therapeutically effective amount of a compound of Formula 1
Figure imgf000034_0002
or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
13. The method according to claim 12 comprising: evaluating a biological sample from the patient for the presence of HBeAg, confirming the patient is HBeAg+, and administering to the patient a therapeutically effective amount of a compound of Formula 1
Figure imgf000035_0001
or a pharmaceutically acceptable salt thereof, in combination with a nucleos(t)ide analogue.
14. The method according to claim 12 or 13, wherein the nucleos(ti)de analogue is selected from the group consisting of tenofovir, or a pharmaceutically acceptable salt or prodrug thereof, and entecavir, or a pharmaceutically acceptable salt thereof.
15. The method according to claim 14, wherein the nucleos(ti)de analogue is tenofovir or a produg thereof, in particular tenofovir alafenamide, tenofovir disoproxil succinate or tenofovir disoproxil fumarate.
16. The method according to any one of claims 12 to 15, wherein the treatment is performed for a period of at least 24 weeks.
17. The method according to any one of claims 12 to 16, wherein the treatment is performed for a period of 48 weeks.
18. The method according to any one of claims 12 to 17, wherein the method further comprises administration of another HBV antiviral.
19. The method according to claim 18, wherein the additional HBV antiviral is selected from the group consisting of an immune modulator, e.g. interferon alpha or pegylated interferon alpha, a nucleic acid polymer, a short interfering RNA or an antisense oligonucleotide.
20. The method according to any one of claims 12 to 19, wherein compound 1 is administered at a dose of 100-300mg once daily.
PCT/EP2021/054727 2020-02-27 2021-02-25 Treatment of hbv WO2021170741A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202062982671P 2020-02-27 2020-02-27
US62/982671 2020-02-27
EP20191427 2020-08-18
EP20191427.2 2020-08-18

Publications (1)

Publication Number Publication Date
WO2021170741A1 true WO2021170741A1 (en) 2021-09-02

Family

ID=74856829

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/054727 WO2021170741A1 (en) 2020-02-27 2021-02-25 Treatment of hbv

Country Status (2)

Country Link
TW (1) TW202146008A (en)
WO (1) WO2021170741A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014184350A1 (en) 2013-05-17 2014-11-20 Janssen R&D Ireland Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
WO2017181141A2 (en) * 2016-04-15 2017-10-19 Novira Therapeutics, Inc. Combinations and methods comprising a capsid assembly inhibitor
WO2019175657A1 (en) 2018-03-14 2019-09-19 Janssen Sciences Ireland Unlimited Company Capsid assembly modulator dosing regimen

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014184350A1 (en) 2013-05-17 2014-11-20 Janssen R&D Ireland Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
WO2017181141A2 (en) * 2016-04-15 2017-10-19 Novira Therapeutics, Inc. Combinations and methods comprising a capsid assembly inhibitor
WO2019175657A1 (en) 2018-03-14 2019-09-19 Janssen Sciences Ireland Unlimited Company Capsid assembly modulator dosing regimen

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
"EASL Clinical Practice Guidelines: Management of chronic hepatitis B virus infection", J. HEPATOLOGY, vol. 57, 2012, pages 167 - 185
"Hepatitis B Factsheet", 2013, WORLD HEALTH ORGANIZATION
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
CHOU, PHARMACOL. REV., vol. 58, 2006, pages 621 - 681
CHOU, T. C.TALALAY, P., ADV. ENZYME REGUL., vol. 22, 1984, pages 27 - 55
HOLFORD, N. H. G.SCHEINER, L. B., CLIN. PHARMACOKINET., vol. 6, 1981, pages 429 - 453
HOOFNAGLE JH ET AL.: "Management of Hepatitis B: Summary of a Clinical Research Workshop", HEPATOLOGY, vol. 45, no. 4, 2007, pages 1056 - 1075
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2
LESMANA LA ET AL.: "Hepatitis B: overview of the burden of disease in the Asia-Pacific region", LIVER INTERNATIONAL, vol. 26, 2006, pages 3 - 10
LOEWE, S.MUISCHNEK, H., ARCH. EXP. PATHOL PHARMACOL., vol. 114, 1926, pages 313 - 326
LOK ASFMCMAHON BJ: "Chronic Hepatitis B: Update 2009", HEPATOLOGY, September 2009 (2009-09-01), pages 1 - 36

Also Published As

Publication number Publication date
TW202146008A (en) 2021-12-16

Similar Documents

Publication Publication Date Title
US10973801B2 (en) Capsid assembly modulator dosing regimen
JP5869469B2 (en) Treatment of liver diseases where iron is involved in the pathogenesis
EP2694087B1 (en) Treatment for infection with hepatitis b virus alone or in combination with hepatitis delta virus and associated liver diseases
JP2005511749A (en) Use of phosphonate nucleotide analogues to treat hepatitis B virus infection
WO2020188448A1 (en) Toll-like receptor agonists for use in the treatment of hepatitis b
KR20220018552A (en) Use of Amlexanox for the manufacture of anti-hepatitis virus drugs
WO2021170741A1 (en) Treatment of hbv
JP2024502673A (en) Synergistic effect of FXR agonist and IFN for treatment of HBV infection
EP2519243B1 (en) Compositions and methods for treating hepatitis b virus infection
US20220313690A1 (en) Synergistic effect of eyp001 and ifn for the treatment of hbv infection
OA19838A (en) Capsid assembly modulator dosing regimen.
EA046230B1 (en) SYNERGIC ACTION OF EYP001 AND IFN IN THE TREATMENT OF HBV INFECTION
CN112891510A (en) Methods of treatment with IAP inhibitors
US20130324497A1 (en) Compositions and methods for treating hepatitis b virus infection

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21709630

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21709630

Country of ref document: EP

Kind code of ref document: A1