WO2019089734A1 - Méthodes et compositions pour le traitement de la grippe - Google Patents

Méthodes et compositions pour le traitement de la grippe Download PDF

Info

Publication number
WO2019089734A1
WO2019089734A1 PCT/US2018/058417 US2018058417W WO2019089734A1 WO 2019089734 A1 WO2019089734 A1 WO 2019089734A1 US 2018058417 W US2018058417 W US 2018058417W WO 2019089734 A1 WO2019089734 A1 WO 2019089734A1
Authority
WO
WIPO (PCT)
Prior art keywords
influenza
compound
viruses
virus
subject
Prior art date
Application number
PCT/US2018/058417
Other languages
English (en)
Inventor
Jun Wang
Chunlong MA
Christopher Hulme
Original Assignee
Arizona Board Of Regents On Behalf Of The University Of Arizona
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 Arizona Board Of Regents On Behalf Of The University Of Arizona filed Critical Arizona Board Of Regents On Behalf Of The University Of Arizona
Publication of WO2019089734A1 publication Critical patent/WO2019089734A1/fr

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7012Compounds having a free or esterified carboxyl group attached, directly or through a carbon chain, to a carbon atom of the saccharide radical, e.g. glucuronic acid, neuraminic acid
    • 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

Definitions

  • This invention is in the field of medicinal chemistry.
  • the invention relates to ng a tetrazolyl-piperidinyl-benzoimidazole structure
  • influenza polymerase activity which function as inhibitors influenza polymerase activity, and their use as therapeutics for the treatment of influenza.
  • Influenza virus infection is responsible for both seasonal influenza as well as sporadic influenza pandemics (see, e.g., Palese, P.; Shaw, M. L. Orthomyxoviridae: The Viruses and Their Replication. In: Knipe DM, Howley PM, eds. Fields Virology. 5th ed. Philadelphia:
  • Influenza vaccines remain the mainstay for the prophylaxis of influenza infection. They are generally effective in preventing seasonal influenza virus infection with an overall effectiveness of -60% (see, e.g., Jackson, M. L.; et al, N Engl J Med 2017, 377, 534-543). However, there is generally a six-month delay from strain identification to batch production, which impedes its use at the beginning of an influenza outbreak (see, e.g., Koszalka, P.; et al, Influenza Other Respir Viruses 2017, 11, 240-246). Thus, small molecule antivirals are highly desired. They are not alternatives, but essential complements of influenza vaccines.
  • M2 channel blockers such as amantadine and rimantadine that inhibit the early stage of viral uncoating (see, e.g., Wang, I; Li, F.; Ma, C. Biopolymers 2015, 104, 291-309), and
  • neuraminidase inhibitors such as oseltamivir, peramivir, and zanamivir that inhibit the last stage of viral egress (see, e.g., Loregian, A.; et al, Cell Mol Life Sci 2014, 71, 3659-83).
  • the increasing incidences of drug-resistant viruses now call for the development of the next generation of influenza antivirals (see, e.g., Webster, R. G.; Govorkova, E. A. Ann N Y Acad Sci 2014, 1323, 115-39).
  • amantadine and rimantadine are no longer recommended due to the widespread M2-S31N mutant (see, e.g., Li, F.; et al, J Med Chem 2017, 60, 1580-1590; Li, F.; et al, ACS Infect Dis 2016, 2, 726-733).
  • the 2008-2009 seasonal HlNl strain circulating in the United States and Japan is completely resistant to the only orally bioavailable drug, oseltamivir, due to the H275Y mutation in the neuraminidase (see, e.g., Hurt, A. C. Curr Opin Virol 2014, 8, 22-9; Matsuzaki, Y.; et al, Virol J 2010, 7, 53).
  • the emergence of drug-resistant viruses with acquired fitness of transmission is a timely reminder of the urgent need for the next generation of antivirals with a novel mechanism of action and a high genetic barrier to drug resistance.
  • the current invention addresses this need.
  • MCR multi-component reaction
  • the influenza virus RNA-dependent RNA polymerase composed of PBl, PB2 and PA subunits, is responsible for viral RNA transcription and replication (FIG. 1 A) (see, e.g., Pflug, A.; et al, Virus Res 2017; Stevaert, A.; et al, Med Res Rev 2016, 36, 1127-1173).
  • X-ray crystal structures show that the N-terminal tail of PBl (PBIN) interacts extensively with the C-terminal domain of PA (PAc) (FIG.
  • PAc domain is highly conserved among different types and subtypes of influenza viruses, and compounds that inhibit PAc-PB IN interactions have shown to have broad-spectrum antiviral activity (see, e.g., Yuan, S.; et al., Antiviral Res 2016, 125, 34-42; Massari, S.; et al., J Med Chem 2015, 58, 3830-42; Muratore, G; et al, Proc Natl Acad Sci U S A 2012, 109, 6247-52).
  • the in-house library comprises a diverse set of compounds prepared by multicomponent reaction methodologies (see, e.g., Hulme, C; Ayaz, M.; Martinez- Ariza, G; Medda, F.; Shaw, A. Recent Advances in Multicomponent Reaction Chemistry. In Small Molecule Medicinal Chemistry, John Wiley & Sons, Inc: 2015; pp 145- 187). Top hits prioritized by in silico docking were tested in a PAC-PBIN ELISA assay.
  • Compound 5 ( 5 ) was found to inhibit PAC-PBIN interaction in a dose- dependent manner with an IC50 of 4.3 ⁇ 0.1 ⁇ .
  • the antiviral activity of compound 5 was confirmed by the plaque assay and it had single to submicromolar EC50 values against several influenza A and B viruses, including both oseltamivir-sen ant strains.
  • the invention rel small-molecules having a tetrazolyl-
  • piperidinyl-benzoimidazole structure which function as inhibitors influenza polymerase activity, and their use as therapeutics for the treatment of influenza.
  • the present invention generally relates to small-molecules having a tetrazolyl- piperidinyl-benzoimidazole structure or pharmaceutically acceptable salts thereof, and to uses of such small molecules for inhibiting the replication of influenza viruses, for reducing the amount of influenza viruses, for inhibiting influenza polymerase activity (e.g., through inhibiting interaction between the PA and PBl submits of influenza polymerase), for preventing influenza, and for treating influenza.
  • small-molecules having a tetrazolyl having a tetrazolyl
  • the resulting compound is able to inhibit influenza polymerase activity. In some embodiments, the resulting compound is able to inhibit interaction between PA and PB1 (PAC-PB IN) of an influenza polymerase. In some embodiments, the resulting compound is able to inhibit and/or prevent influenza virus activity. In some embodiments, the resulting compound is able to inhibit the replication of drug-resistant influenza viruses. In some embodiments, the following compounds are contemplated for Formula I:
  • Tables 1 , 2, 3, 4 and 5 show inhibition against influenza strains for various compounds encompassed within Formula I.
  • the invention is directed to a method of reducing the amount of influenza viruses in a biological in vitro sample or in a subject (e.g., a mammalian subject) (e.g., a human subject), comprising administering to the sample or subject an effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein (e.g., one or more of the compounds described herein).
  • a subject e.g., a mammalian subject
  • a human subject e.g., a human subject
  • the invention is directed to a method of inhibiting the replication of influenza viruses in a biological in vitro sample or in a subject (e.g., a mammalian subject) (e.g., a human subject), comprising administering to the sample or subject an effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein (e.g., one or more of the compounds described herein).
  • a subject e.g., a mammalian subject
  • a human subject e.g., a human subject
  • the invention is directed to a method of inhibiting influenza polymerase activity in a biological in vitro sample or in a subject (e.g., a mammalian subject) (e.g., a human subject), comprising administering to the sample or subject an effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein (e.g., one or more of the compounds described herein).
  • inhibiting influenza polymerase activity comprises inhibiting interaction between the PA and PB1 submits (e.g., PAC-PBIN) of the influenza polymerase.
  • the invention is directed to a method of preventing or treating influenza in a subject (e.g., a mammalian subject) (e.g., a human subject), comprising administering to the subject a therapeutically effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein (e.g., one or more of the compounds described herein).
  • a subject e.g., a mammalian subject
  • a human subject e.g., a human subject
  • a therapeutically effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein e.g., one or more of the compounds described herein.
  • the method further comprises coadministration of one or more small molecule influenza antivirals (e.g., M2 channel blockers such as amantadine and rimantadine) (e.g., neuraminidase inhibitors such as oseltamivir, peramivir, and zanamivir).
  • M2 channel blockers such as amantadine and rimantadine
  • neuraminidase inhibitors such as oseltamivir, peramivir, and zanamivir.
  • FIG. 1 Structures of the influenza polymerase.
  • A X-ray crystal structures of the viral polymerase complexes from the bat influenza A/H17N10 virus (PDB: 4WSB) (see, e.g., Reich, S.; et al., Nature 2014, 516, 361). PA: green; PB1 : yellow; PB2: magenta.
  • B X-ray crystal structure of PAC-PBIN (PDB: 3CM8) (see, e.g., He, X.; et al, Nature 2008, 454, 1123-1126). PA: green; PB1 : yellow.
  • FIG. 2 Inhibition of PA-PB1 by compound 5 in ELISA assay.
  • FIG. 3 Structure-activity relationship studies of compound 5. Oseltamivir carboxylate and nucleozin were tested at 200 nM and 1 ⁇ , respectively. All other compounds were tested at 5 ⁇ . The results are the mean ⁇ S.D. from two independent experiments.
  • FIG. 4 Compound 12a has a high in vitro genetic barrier to drug resistance as shown by the serial viral pasage experiment.
  • A Cartoon representation of the serial drug passage experiment.
  • B Comparsion of the in vitro genetic barrier of drug resistance between oseltamivir carboxylate and compound 12a.
  • FIG. 5 Docking model of compound 12a in the PB1 -binding pocket in PA.
  • A Surface view of the docking model of compound 12a in PAc.
  • B Ligand interaction diagram of compound 12a with residues in the binding site.
  • Influenza viruses are RNA viruses of the family Orthomyxoviridae, which comprises five genera: Influenza virus A, Influenza virus B, Influenza virus C, ISA virus and Thogoto virus.
  • influenza A virus has one species, influenza A virus. Wild aquatic birds are the natural hosts for a large variety of influenza A. Occasionally, viruses are transmitted to other species and may then cause devastating outbreaks in domestic poultry or give rise to human influenza pandemics.
  • the type A viruses are the most virulent human pathogens among the three influenza types and cause the most severe disease.
  • the influenza A virus can be subdivided into different serotypes based on the antibody response to these viruses.
  • H1N1 which caused Spanish influenza in 1918
  • H2N2 which caused Asian Influenza in 1957
  • H3N2 which caused Hong Kong Flu in 1968
  • H5N1 a pandemic threat in the 2007-08 influenza season
  • H7N7 which has unusual zoonotic potential
  • H1N2 endemic in humans and pigs
  • Influenza virus B genus has one species, influenza B virus. Influenza B almost exclusively infects humans and is less common than influenza A. The only other animal known to be susceptible to influenza B infection is the seal. This type of influenza mutates at a rate 2-3 times slower than type A and consequently is less genetically diverse, with only two lineages,
  • influenza B viruses do not occur. Nevertheless, influenza B viruses are found to be the predominant circulating strains in certain influenza seasons and human infection with influenza B viruses have been shown to led to morbidity and mortality.
  • influenza C The Influenza virus C genus has one species, influenza C virus, which infects humans and pigs and can cause severe illness and local epidemics. However, influenza C is less common than the other types and usually seems to cause mild disease in children.
  • Influenza A, B and C viruses are very similar in structure.
  • the virus particle is 80-120 nanometers in diameter and usually roughly spherical, although filamentous forms can occur.
  • the Influenza A genome encodes 1 1 proteins: hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), Ml , M2, NS 1, NS2(NEP), PA,
  • HA and NA are large glycoproteins on the outside of the viral particles.
  • HA is a lectin that mediates binding of the virus to target cells and entry of the viral genome into the target cell, while NA is involved in the release of progeny virus from infected cells, by cleaving sugars that bind the mature viral particles.
  • these proteins have been targets for antiviral drugs.
  • Influenza A viruses are classified into subtypes based on antibody responses to HA and NA, forming the basis of the H and N distinctions (vide supra) in, for example, H5N1.
  • influenza Current treatment options for influenza include vaccination, and chemotherapy or chemoprophylaxis with antiviral medications.
  • Vaccination against influenza with an influenza vaccine is often recommended for anyone aged 6 months old and older, especially for high-risk groups, such as children and the elderly, or in people that have asthma, diabetes, or heart disease.
  • influenza vaccinated and still get influenza.
  • the vaccine is reformulated each season for a few specific influenza strains but cannot possibly include all the strains actively infecting people in the world for that season. It may take six months for the manufacturers to formulate and produce the millions of doses required to deal with the seasonal epidemics;
  • influenza vaccines Due to the high mutation rate of the virus, a particular influenza vaccine usually confers protection for no more than a few years. A vaccine formulated for one year may be ineffective in the following year, since the influenza virus changes rapidly over time, and different strains become dominant.
  • Antiviral drugs can also be used to treat influenza, with neuraminidase inhibitors being particularly effective, but viruses can develop resistance to the standard antiviral drugs.
  • compound 12a had broad-spectrum antiviral activity against a panel of human clinical isolates of influenza A and B viruses, including both oseltamivir-sensitive and oseltamivir-resistant influenza viruses. Moreover, compound 12a had a higher in vitro genetic barrier to drug resistance than oseltamivir, and no resistant virus emerged under drug selection pressure. Overall, the discovery of compound 12a as a broad-spectrum influenza antiviral with a high in vitro genetic barrier to drug resistance is significant, as it offers a second line of defense to combat the next influenza epidemics and pandemics if vaccines and oseltamivir fail to confine the disease outbreak.
  • the invention relates to a new class of small-molecules having a tetrazolyl-
  • piperidinyl-benzoimidazole structure ( ) which function as inhibitors influenza polymerase activity, and their use as therapeutics for the treatment of influenza.
  • the present invention generally relates to small-molecules having a tetrazolyl- piperidinyl-benzoimidazole structure or pharmaceutically acceptable salts thereof, and to uses of such small molecules for inhibiting the replication of influenza viruses, for reducing the amount of influenza viruses, for inhibiting influenza polymerase activity (e.g., through inhibiting interaction between the PA and PB1 submits of influenza polymerase), for preventing influenza, and for treating influenza.
  • benzoic acid compounds encompassed within Formula I are
  • the resulting compound is able to inhibit influenza polymerase activity. In some embodiments, the resulting compound is able to inhibit interaction between PA and PBl (PAC-PBIN) of an influenza polymerase. In some embodiments, the resulting compound is able to inhibit and/or prevent influenza virus activity. In some embodiments, the resulting compound is able to inhibit drug resistant influenza virus activity.
  • Tables 1, 2, 3, 4 and 5 show inhibition activity against influenza strains for small-molecules having a tetrazolyl-piperidinyl-benzoimidazole structure encompassed within Formula I.
  • Such small-molecules having a tetrazolyl-piperidinyl-benzoimidazole structure encompassed within Formula I can exist in or form different polymorphic forms.
  • polymorphism is an ability of a compound to crystallize as more than one distinct crystalline or "polymorphic" species.
  • a polymorph is a solid crystalline phase of a compound with at least two different arrangements or polymorphic forms of that compound molecule in the solid state.
  • Polymorphic forms of any given compound are defined by the same chemical formula or composition and are as distinct in chemical structure as crystalline structures of two different chemical compounds.
  • polymorphic form includes solvates and neat polymorphic form that does not have any solvates.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, cis-trans, conformational, and rotational) forms of the structure.
  • isomeric e.g., enantiomeric, diastereomeric, cis-trans, conformational, and rotational
  • the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers are included in this invention, unless only one of the isomers is drawn specifically.
  • a substituent can freely rotate around any rotatable bonds. Therefore, single
  • One aspect of the present invention is generally related to the use of the small-molecules having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein (e.g., one or more of the compounds described herein) for inhibiting the replication of influenza viruses in a biological sample or in a patient, for reducing the amount of influenza viruses (reducing viral titer) in a biological sample or in a patient, and for treating influenza in a patient.
  • a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein e.g., one or more of the compounds described herein
  • the invention is directed to a method of reducing the amount of influenza viruses in a biological in vitro sample or in a subject (e.g., a mammalian subject) (e.g., a human subject), comprising administering to the sample or subject an effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein.
  • a subject e.g., a mammalian subject
  • a human subject e.g., a human subject
  • the invention is directed to a method of inhibiting the replication of influenza viruses in a biological in vitro sample or in a subject (e.g., a mammalian subject) (e.g., a human subject), comprising administering to the sample or subject an effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein (e.g., one or more of the compounds described herein).
  • a subject e.g., a mammalian subject
  • a human subject e.g., a human subject
  • the invention is directed to a method of inhibiting influenza polymerase activity in a biological in vitro sample or in a subject (e.g., a mammalian subject) (e.g., a human subject), comprising administering to the sample or subject an effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein (e.g., one or more of the compounds described herein).
  • inhibiting influenza polymerase activity comprises inhibiting interaction between the PA and PB1 submits (e.g., PAC-PBIN) of the influenza polymerase.
  • the invention is directed to a method of preventing or treating influenza in a subject (e.g., a mammalian subject) (e.g., a human subject), comprising administering to the subject a therapeutically effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein (e.g., one or more of the compounds described herein).
  • a subject e.g., a mammalian subject
  • a human subject e.g., a human subject
  • a therapeutically effective amount of a small-molecule having a tetrazolyl-piperidinyl-benzoimidazole structure disclosed herein e.g., one or more of the compounds described herein.
  • the present invention is generally related to the use of the compounds disclosed herein (e.g., in pharmaceutically acceptable compositions) for any of the uses specified above.
  • the compounds disclosed herein can be used to reduce viral titre in a biological sample (e.g. an infected cell culture) or in humans (e.g. lung viral titre in a patient).
  • influenza virus mediated condition is used interchangeably to mean the disease caused by an infection with an influenza virus.
  • Influenza is an infectious disease that affects birds and mammals caused by influenza viruses.
  • Influenza viruses are RNA viruses of the family Orthomyxoviridae, which comprises five genera: Influenza virus A, Influenza virus B, Influenza virus C, ISA virus and Thogoto virus.
  • Influenza virus A genus has one species, influenza A virus which can be subdivided into different serotypes based on the antibody response to these viruses: H1N1 , H2N2, H3N2, H5N1, H7N7, H1N2, H9N2, H7N2, H7N3 and H10N7. Additional examples of influenza A virus include H3N8 and H7N9.
  • Influenza virus B genus has one species, influenza B virus. Influenza B almost exclusively infects humans and is less common than influenza A.
  • Influenza virus C genus has one species, Influenza virus C virus, which infects humans and pigs and can cause severe illness and local epidemics. However, Influenza virus C is less common than the other types and usually seems to cause mild disease in children.
  • influenza or influenza viruses are associated with Influenza virus A or B. In some embodiments of the invention, influenza or influenza viruses are associated with Influenza virus A. In some specific embodiments of the invention, Influenza virus A is H1N1 , H2N2, H3N2 or H5N1. In some specific embodiments of the invention,
  • Influenza virus A is H1N1, H3N2, H3N8, H5N1 , and H7N9. In some specific embodiments of the invention, Influenza virus A is H1N1, H3N2, H3N8, and H5N1.
  • influenza causes pneumonia, which can be fatal, particularly in young children and the elderly. Although it is often confused with the common cold, influenza is a much more severe disease and is caused by a different type of virus. Influenza can produce nausea and vomiting, especially in children, but these symptoms are more characteristic of the unrelated gastroenteritis, which is sometimes called "stomach flu" or "24-hour flu”.
  • Symptoms of influenza can start quite suddenly one to two days after infection. Usually the first symptoms are chills or a chilly sensation, but fever is also common early in the infection, with body temperatures ranging from 38° C. to 39° C. (approximately 100° F. to 103° F.). Many people are so ill that they are confined to bed for several days, with aches and pains throughout their bodies, which are worse in their backs and legs. Symptoms of influenza may include: body aches, especially joints and throat, extreme coldness and fever, fatigue, headache, irritated watering eyes, reddened eyes, skin (especially face), mouth, throat and nose, abdominal pain (in children with influenza B).
  • influenza Symptoms of influenza are non-specific, overlapping with many pathogens ("influenza-like illness"). Usually, laboratory data is needed in order to confirm the diagnosis.
  • the terms, “disease”, “disorder”, and “condition” may be used interchangeably here to refer to an influenza virus mediated medical or pathological condition.
  • the terms “subj ect” and “patient” are used interchangeably.
  • the terms “subject” and “patient” refer to an animal (e.g., a bird such as a chicken, quail or turkey, or a mammal), specifically a “mammal” including a non-primate (e.g., a cow, pig, horse, sheep, rabbit, guinea pig, rat, cat, dog, and mouse) and a primate (e.g., a monkey, chimpanzee and a human), and more specifically a human.
  • a non-primate e.g., a cow, pig, horse, sheep, rabbit, guinea pig, rat, cat, dog, and mouse
  • a primate e.g., a monkey, chimpanzee and a human
  • the subject is a non-human animal such as a farm animal (e.g., a horse, cow, pig or sheep), or a pet (e.g., a dog, cat, guinea pig or rabbit).
  • a farm animal e.g., a horse, cow, pig or sheep
  • a pet e.g., a dog, cat, guinea pig or rabbit
  • the subject is a "human”.
  • biological sample includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • therapeutic treatments includes the reduction or amelioration of the progression, severity and/or duration of influenza viruses mediated conditions, or the amelioration of one or more symptoms (specifically, one or more discernible symptoms) of influenza viruses mediated conditions, resulting from the administration of one or more therapies (e.g., one or more therapeutic agents such as a compound or composition of the invention).
  • the therapeutic treatment includes the amelioration of at least one measurable physical parameter of an influenza virus mediated condition.
  • the therapeutic treatment includes the inhibition of the progression of an influenza virus mediated condition, either physically by, e.g., stabilization of a discernible symptom, physiologically by, e.g., stabilization of a physical parameter, or both.
  • the therapeutic treatment includes the reduction or stabilization of influenza viruses mediated infections.
  • Antiviral drugs can be used in the community setting to treat people who already have influenza to reduce the severity of symptoms and reduce the number of days that they are sick.
  • chemotherapy refers to the use of medications, e.g. small molecule drugs (rather than “vaccines”) for treating a disorder or disease.
  • medications e.g. small molecule drugs (rather than “vaccines”
  • vaccines small molecule drugs
  • prophylaxis or “prophylactic use” and “prophylactic treatment” as used herein, refer to any medical or public health procedure whose purpose is to prevent, rather than treat or cure a disease.
  • the terms “prevent”, “prevention” and “preventing” refer to the reduction in the risk of acquiring or developing a given condition, or the reduction or inhibition of the recurrence or said condition in a subject who is not ill, but who has been or may be near a person with the disease.
  • chemoprophylaxis refers to the use of medications, e.g., small molecule drugs (rather than "vaccines”) for the prevention of a disorder or disease.
  • prophylactic use includes the use in situations in which an outbreak has been detected, to prevent contagion or spread of the infection in places where a lot of people that are at high risk of serious influenza complications live in close contact with each other (e.g. in a hospital ward, daycare center, prison, nursing home, or the like). It also includes the use among populations who require protection from the influenza but who either do not get protection after vaccination (e.g., due to weak immune system), or when the vaccine is unavailable to them, or when they cannot get the vaccine because of side effects. It also includes use during the two weeks following vaccination, since during that time the vaccine is still ineffective.
  • Prophylactic use may also include treating a person who is not ill with the influenza or not considered at high risk for complications, in order to reduce the chances of getting infected with the influenza and passing it on to a high-risk person in close contact with him (for instance, healthcare workers, nursing home workers, or the like).
  • an influenza "outbreak” is defined as a sudden increase of acute febrile respiratory illness (AFRI) occurring within a 48 to 72 hour period, in a group of people who are in close proximity to each other (e.g. in the same area of an assisted living facility, in the same household, etc.) over the normal background rate or when any subject in the population being analyzed tests positive for influenza.
  • AFRI acute febrile respiratory illness
  • a “cluster” is defined as a group of three or more cases of AFRI occurring within a 48 to 72 hour period, in a group of people who are in close proximity to each other (e.g. in the same area of an assisted living facility, in the same household, etc.).
  • index case is the initial patient in the population sample of an epidemiological investigation.
  • primary case or “patient zero” is the initial patient in the population sample of an epidemiological investigation.
  • term is not capitalized.
  • term is used to refer to a specific person in place of that person's name within a report on a specific
  • index case is the first patient that indicates the existence of an outbreak. Earlier cases may be found and are labeled primary, secondary, tertiary, and the like.
  • the methods of the invention are a preventative or "pre-emptive" measure to a patient, specifically a human, having a predisposition to complications resulting from infection by an influenza virus.
  • pre-emptive or "pre-emptively” as used herein, for example, in 'pre-emptive' use, is the prophylactic use in situations in which an "index case” or an "outbreak" has been confirmed, in order to prevent the spread of infection in the rest of the community or population group.
  • the methods of the invention are applied as a "pre-emptive" measure to members of a community or population group, specifically humans, in order to prevent the spread of infection.
  • an "effective amount” refers to an amount sufficient to elicit the desired biological response.
  • the desired biological response is to inhibit the replication of influenza virus, to reduce the amount of influenza viruses or to reduce or ameliorate the severity, duration, progression, or onset of an influenza virus infection, prevent the advancement of an influenza viruses infection, prevent the recurrence, development, onset or progression of a symptom associated with an influenza virus infection, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy used against influenza infections.
  • the precise amount of compound administered to a subject will depend on the mode of
  • an "effective amount" of the second agent will depend on the type of drug used. Suitable dosages are known for approved agents and can be adjusted by the skilled artisan according to the condition of the subject, the type of condition(s) being treated and the amount of a compound described herein being used. In cases where no amount is expressly noted, an effective amount should be assumed.
  • the compounds disclosed herein can be administered to a subject in a dosage range from between approximately 0.01 to 100 mg/kg body weight/day for therapeutic or prophylactic treatment.
  • dosage regimens can be selected in accordance with a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the renal and hepatic function of the subject; and the particular compound or salt thereof employed, the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • the skilled artisan can readily determine and prescribe the effective amount of the compounds described herein required to treat, to prevent, inhibit (fully or partially) or arrest the progress of the disease.
  • Dosages of the compounds described herein can range from 0.01 to 100 mg/kg body weight/day, 0.01 to 50 mg/kg body weight/day, 0.1 to 50 mg/kg body weight/day, or 1 to 25 mg/kg body weight/day. It is understood that the total amount per day can be administered in a single dose or can be administered in multiple dosing, such as twice a day (e.g., every 12 hours), three times a day (e.g., every 8 hours), or four times a day (e.g., every 6 hours).
  • dosages of the small-molecules having a tetrazolyl-piperidinyl- benzoimidazole structure encompassed within Formula I and pharmaceutically acceptable salts thereof are in a range of 100 mg to 1,600 mg, such as 400 mg to 1 ,600 mg or 400 mg to 1 ,200 mg.
  • Each dose can be taken once a day (QD), twice per day (e.g., every 12 hours (BID)), or three times per day (e.g., q8h (TID)). It is noted that any combinations of QD, BID, and TID can be employed, as desired, such as BID on day 1, followed by QD thereafter.
  • dosages of the small-molecules having a tetrazolyl-piperidinyl- benzoimidazole structure disclosed herein and pharmaceutically acceptable salts thereof are in a range of 100 mg to 1,600 mg, such as 400 mg to 1,600 mg or 400 mg to 1 ,200 mg.
  • Each dose can be taken once a day (QD), twice per day (e.g., every 12 hours (BID)), or three times per day (e.g., q8h (TID)).
  • QD twice per day
  • TID three times per day
  • any combinations of QD, BID, and TID can be employed, as desired, such as BID on day 1, followed by QD thereafter, or, when a loading dosage is employed on day 1 , BID on day 2, followed by QD thereafter.
  • dosages of the compounds described herein are 400 mg to 1,600 mg, 400 mg to 1,200 mg, or 600 mg to 1,200 mg once a day. In another specific embodiment, dosages of the compounds described herein are 400 mg to 1 ,600 mg, 400 mg to 1 ,200 mg, or 300 mg to 900 mg twice a day. In yet another specific embodiment, dosages of the compounds described herein are 400 mg to 1 ,000 mg once a day. In yet another specific embodiment, dosages of the compounds described herein are 600 mg to 1 ,000 mg once a day. In yet another specific embodiment, dosages of the compounds described herein are 600 mg to 800 mg once a day.
  • dosages of the compounds described herein are 400 mg to 800 mg twice a day (e.g., 400 mg to 800 mg every 12 hours). In yet another specific embodiment, dosages of the compounds described herein are 400 mg to 600 mg twice a day.
  • a loading dosage regimen is employed.
  • a loading dose of 400 mg to 1 ,600 mg is employed on day 1 of treatment.
  • a loading dose of 600 mg to 1 ,600 mg is employed on day 1 of treatment.
  • a loading dose of 800 mg to 1 ,600 mg is employed on day 1 of treatment.
  • a loading dose of 900 mg to 1,600 mg is employed on day 1 of treatment.
  • a loading dose of 900 mg to 1 ,200 mg is employed on day 1 of treatment.
  • a loading dose of 900 mg is employed on day 1 of treatment.
  • a loading dose of 1,000 mg is employed on day 1 of treatment.
  • a loading dose of 1 ,200 mg is employed on day 1 of treatment.
  • the dosage regimen of the compounds described herein employs a loading dosage of 600 mg to 1,600 mg on day 1 and with a regular dosage of 300 mg to 1 ,200 mg for the rest of the treatment duration. Each regular dose can be taken once a day, twice a day, or three times a day, or any combination thereof.
  • a loading dosage of 900 mg to 1 ,600 mg, such as 900 mg, 1 ,200 mg, or 1,600 mg is employed.
  • a loading dosage of 900 mg to 1 ,200 mg, such as 900 mg or 1 ,200 mg is employed.
  • a regular dosage of 400 mg to 1 ,200 mg such as 400 mg, 600 mg, or 800 mg, is employed for the rest of the treatment duration.
  • a regular dosage of 400 mg to 800 mg is employed for the rest of the treatment duration.
  • a regular dosage of 300 mg to 900 mg twice a day is employed.
  • a regular dosage of 600 mg to 1 ,200 mg once a day is employed.
  • the compounds described herein can be administered to a patient within, for example, 48 hours (or within 40 hours, or less than 2 days, or less than 1.5 days, or within 24 hours) of onset of symptoms (e.g., nasal congestion, sore throat, cough, aches, fatigue, headaches, and chills/sweats).
  • onset of symptoms e.g., nasal congestion, sore throat, cough, aches, fatigue, headaches, and chills/sweats.
  • the compounds described herein can be administered to a patient within, for example, 96 hours of onset of symptoms.
  • the therapeutic treatment can last for any suitable duration, for example, for 3 days, 4 days, 5 days, 7 days, 10 days, 14 days, etc.
  • the compounds described herein can be administered to a patient within, for example, 2 days of onset of symptoms in the index case, and can be continued for any suitable duration, for example, for 7 days, 10 days, 14 days, 20 days, 28 days, 35 days, 42 days, etc., up to the entire flu season.
  • a flu season is an annually -recurring time period characterized by the prevalence of outbreaks of influenza. Influenza activity can sometimes be predicted and even tracked geographically. While the beginning of major flu activity in each season varies by location, in any specific location these minor epidemics usually take 3-4 weeks to peak and another 3-4 weeks to significantly diminish.
  • Centers for Disease Control CDC collects, compiles and analyzes information on influenza activity year round in the United States and produces a weekly report from October through mid- May.
  • the therapeutic treatment lasts for 1 day to an entire flu season. In one specific embodiment, the therapeutic treatment lasts for 3 days to 14 days. In another specific embodiment, the therapeutic treatment lasts for 5 days to 14 days. In another specific embodiment, the therapeutic treatment lasts for 3 days to 10 days. In yet another specific embodiment, the therapeutic treatment lasts for 4 days to 10 days. In yet another specific embodiment, the therapeutic treatment lasts for 5 days to 10 days. In yet another specific embodiment, the therapeutic treatment lasts for 4 days to 7 days (e.g., 4 days, 5 days, 6 days, or 7 days). In yet another specific embodiment, the therapeutic treatment lasts for 5 days to 7 days (e.g., 5 days, 6 days, or 7 days). In one specific embodiment, the prophylactic treatment lasts up to the entire flu season.
  • the compounds described herein are administered to a patient for 3 days to 14 days (e.g., 5 days to 14 days) with a loading dosage of 900 mg to 1,600 mg on day 1 and with a regular dosage of 300 mg to 1 ,200 mg for the rest of the treatment duration.
  • the compounds described herein are administered to a patient for 3 days to 14 days (e.g., 5 days to 14 days) with a loading dosage of 900 mg to 1,200 mg on day 1 and with a regular dosage of 400 mg to 1 ,000 mg for the rest of the treatment duration.
  • the compounds described herein are administered to a patient for 3 days to 14 days (e.g., 5 days to 14 days) with a loading dosage of 900 mg to 1,200 mg on day 1 and with a regular dosage of 400 mg to 800 mg for the rest of the treatment duration.
  • the compounds described herein are administered to a patient for 3 days to 14 days (e.g., 5 days to 14 days) with a loading dosage of 900 mg to 1,200 mg on day 1 and with a regular dosage of 400 mg to 800 mg for the rest of the treatment duration.
  • Each dose can be taken once a day, twice a day, or three times a day, or any combination thereof.
  • the compounds described herein are administered to a patient for 3 days to 14 days with a loading dosage of 900 mg to 1 ,600 mg on day 1 and with a regular dosage of 600 mg to 1,000 mg once a day for the rest of the treatment duration.
  • the compounds described herein are administered to a patient for 3 days to 14 days with a loading dosage of 900 mg to 1,200 mg on day 1 and with a regular dosage of 600 mg to 800 mg (e.g., 600 mg, 650 mg, 700 mg, 750 mg, or 800 mg) once a day for the rest of the treatment duration.
  • the treatment duration is for 4 days to 10 days, 5 days to 10 days, or 5 days to 7 days.
  • the compounds described herein are administered to a patient for 3 days to 14 days with a loading dosage of 900 mg to 1 ,600 mg on day 1 and with a regular dosage of 400 mg to 800 mg twice a day for the rest of the treatment duration.
  • the compounds described herein are administered to a patient for 3 days to 14 days with a loading dosage of 900 mg to 1 ,200 mg on day 1 and with a regular dosage of 400 mg to 600 mg (e.g., 400 mg, 450 mg, 500 mg, 550 mg, or 600 mg) twice a day for the rest of the treatment duration.
  • the duration is for 4 days to 10 days, 5 days to 10 days, or 5 days to 7 days.
  • the compounds described herein are administered to a patient for 4 days or 5 days with a loading dosage of 900 mg to 1 ,200 mg (e.g., 900 mg or 1 ,200 mg) on day 1 and with a regular dosage of 400 mg to 600 mg (e.g., 400 mg or 600 mg) twice a day for the rest of the treatment duration (e.g., days 2 through 4, or days 2 through 5).
  • a loading dosage of 900 mg to 1 ,200 mg e.g., 900 mg or 1 ,200 mg
  • a regular dosage of 400 mg to 600 mg e.g., 400 mg or 600 mg
  • twice a day for the rest of the treatment duration e.g., days 2 through 4, or days 2 through 5.
  • the compounds described herein are administered to a patient for 4 days or 5 days with a loading dosage of 900 mg to 1,200 mg (e.g., 900 mg or 1,200 mg) on day 1 and with a regular dosage of 600 mg to 800 mg (e.g., 600 mg or 800 mg) once a day for the rest of the treatment duration.
  • a loading dosage of 900 mg to 1,200 mg e.g., 900 mg or 1,200 mg
  • a regular dosage of 600 mg to 800 mg e.g., 600 mg or 800 mg
  • an effective amount can be achieved in the method or pharmaceutical composition of the invention employing a compound of the invention (including a pharmaceutically acceptable salt or solvate (e.g., hydrate)) alone or in combination with an additional suitable therapeutic agent, for example, an antiviral agent or a vaccine.
  • an additional suitable therapeutic agent for example, an antiviral agent or a vaccine.
  • an effective amount can be achieved using a first amount of a compound of the invention and a second amount of an additional suitable therapeutic agent (e.g. an antiviral agent or vaccine).
  • a compound of the invention and the additional therapeutic agent are each administered in an effective amount (i.e., each in an amount which would be therapeutically effective if administered alone).
  • a compound of the invention and the additional therapeutic agent are each administered in an amount which alone does not provide a therapeutic effect (a sub-therapeutic dose).
  • a compound of the invention can be administered in an effective amount, while the additional therapeutic agent is administered in a sub-therapeutic dose.
  • a compound of the invention can be administered in a sub-therapeutic dose, while the additional therapeutic agent, for example, a suitable cancer-therapeutic agent is administered in an effective amount.
  • the terms “in combination” or “co-administration” can be used interchangeably to refer to the use of more than one therapy (e.g., one or more prophylactic and/or therapeutic agents).
  • the use of the terms does not restrict the order in which therapies (e.g., prophylactic and/or therapeutic agents) are administered to a subject.
  • Co-administration encompasses administration of the first and second amounts of the compounds of the coadministration in an essentially simultaneous manner, such as in a single pharmaceutical composition, for example, capsule or tablet having a fixed ratio of first and second amounts, or in multiple, separate capsules or tablets for each.
  • coadministration also encompasses use of each compound in a sequential manner in either order.
  • the present invention is directed to methods of combination therapy for inhibiting Influenza viruses replication in biological samples or patients, or for treating or preventing Influenza virus infections in patients using the compounds described herein.
  • compositions of the invention also include those comprising an inhibitor of Influenza virus replication of this invention in combination with an anti-viral compound exhibiting anti-Influenza virus activity.
  • Methods of use of the compounds described herein and compositions of the invention also include combination of chemotherapy with a compound or composition of the invention, or with a combination of a compound or composition of this invention with another anti-viral agent and vaccination with an Influenza vaccine.
  • the compounds are administered sufficiently close in time to have the desired therapeutic effect.
  • the period of time between each administration which can result in the desired therapeutic effect can range from minutes to hours and can be determined taking into account the properties of each compound such as potency, solubility, bioavailability, plasma half-life and kinetic profile.
  • a compound of the invention and the second therapeutic agent can be administered in any order within 24 hours of each other, within 16 hours of each other, within 8 hours of each other, within 4 hours of each other, within 1 hour of each other or within 30 minutes of each other.
  • a first therapy e.g., a prophylactic or therapeutic agent such as a compound of the invention
  • a first therapy can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy (e.g., a prophylactic or therapeutic agent such as an anti-cancer agent) to a subject.
  • a second therapy e.g., a prophylactic or therapeutic agent such as an anti-cancer agent
  • the method of co-administration of a first amount of a compound of the invention and a second amount of an additional therapeutic agent can result in an enhanced or synergistic therapeutic effect, wherein the combined effect is greater than the additive effect that would result from separate administration of the first amount of a compound of the invention and the second amount of an additional therapeutic agent.
  • the term "synergistic” refers to a combination of a compound of the invention and another therapy (e.g., a prophylactic or therapeutic agent), which is more effective than the additive effects of the therapies.
  • a synergistic effect of a combination of therapies can permit the use of lower dosages of one or more of the therapies and/or less frequent administration of said therapies to a subject.
  • the ability to utilize lower dosages of a therapy (e.g., a prophylactic or therapeutic agent) and/or to administer said therapy less frequently can reduce the toxicity associated with the administration of said therapy to a subject without reducing the efficacy of said therapy in the prevention, management or treatment of a disorder.
  • a synergistic effect can result in improved efficacy of agents in the prevention, management or treatment of a disorder.
  • a synergistic effect of a combination of therapies e.g., a combination of prophylactic or therapeutic agents
  • both therapeutic agents can be administered so that the period of time between each administration can be longer (e.g. days, weeks, or months).
  • Suitable methods include, for example, 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)).
  • Each equation referred to above can be applied with 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.
  • neuraminidase inhibitors such as oseltamivir (Tamiflu®) and Zanamivir (Rlenza®)
  • viral ion channel (M2 protein) blockers such as amantadine (Symmetrel®) and rimantadine
  • T-705 (flavipiravir) and related compounds: Novel broad-spectrum inhibitors of RNA viral infections”
  • the compounds described herein can be co- administered with a traditional influenza vaccine.
  • the compounds described herein can be co-administered with zanamivir. In some embodiments, the compounds described herein can be co-administered with flavipiravir (T-705). In some embodiments, the compounds described herein can be coadministered with oseltamivir. In some embodiments, the compounds described herein can be co-administered with amantadine or rimantadine. Oseltamivir can be administered in a dosage regimen specified in its label. In some specific embodiments, it is administered 75 mg twice a day, or 150 mg once a day.
  • the compounds described herein can be formulated into pharmaceutical compositions that further comprise a pharmaceutically acceptable carrier, diluent, adjuvant or vehicle.
  • the present invention relates to a pharmaceutical composition comprising a compound of the invention described above, and a pharmaceutically acceptable carrier, diluent, adjuvant or vehicle.
  • the present invention is a pharmaceutical composition comprising an effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier, diluent, adjuvant or vehicle.
  • Pharmaceutically acceptable carriers include, for example, pharmaceutical diluents, excipients or carriers suitably selected with respect to the intended form of administration, and consistent with conventional pharmaceutical practices.
  • an “effective amount” includes a “therapeutically effective amount” and a
  • prophylactically effective amount refers to an amount effective in treating and/or ameliorating an influenza virus infection in a patient infected with influenza.
  • prophylactically effective amount refers to an amount effective in preventing and/or substantially lessening the chances or the size of influenza virus infection outbreak.
  • a pharmaceutically acceptable carrier may contain inert ingredients which do not unduly inhibit the biological activity of the compounds.
  • the pharmaceutically acceptable carriers should be biocompatible, e.g., non-toxic, non-inflammatory, non-immunogenic or devoid of other undesired reactions or side-effects upon the administration to a subject. Standard pharmaceutical formulation techniques can be employed.
  • the pharmaceutically acceptable carrier, adjuvant, or vehicle includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • An adverse effect from a therapy might be harmful or uncomfortable or risky.
  • Side effects include, but are not limited to fever, chills, lethargy, gastrointestinal toxicities (including gastric and intestinal ulcerations and erosions), nausea, vomiting, neurotoxicities, nephrotoxicities, renal toxicities (including such conditions as papillary necrosis and chronic interstitial nephritis), hepatic toxicities (including elevated serum liver enzyme levels), myelotoxicities (including leukopenia, myelosuppression, thrombocytopenia and anemia), dry mouth, metallic taste, prolongation of gestation, weakness, somnolence, pain (including muscle pain, bone pain and headache), hair loss, asthenia, dizziness, extra-pyramidal symptoms, akathisia, cardiovascular disturbances and sexual dysfunction.
  • materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as twin 80, phosphates, glycine, sorbic acid, or potassium sorbate), partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, or zinc salts), colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, methylcellulose, hydroxypropyl methylcellulose, wool fat, 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;
  • glycols such a propylene glycol or polyethylene glycol
  • esters such as ethyl oleate and ethyl laurate
  • agar buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
  • compositions described above can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated.
  • Liquid dosage forms for oral administration include, but are not limited to,
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweet
  • sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile inj ectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U. S. P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • the rate of compound release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly (anhydrides).
  • Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are specifically suppositories which can be prepared by mixing the compounds described herein with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin,
  • the dosage form may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active compounds can also be in microencapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • buffering agents include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound described herein include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention.
  • transdermal patches which have the added advantage of providing controlled delivery of a compound to the body.
  • dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • compositions described herein may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes, but is not limited to, subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrastemal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions described herein may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • a long-chain alcohol diluent or dispersant such as carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions described herein may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include, but are not limited to, lactose and com starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • the pharmaceutical compositions described herein may be administered in the form of suppositories for rectal administration.
  • Suitable non-irritating excipient which is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • compositions described herein may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically -transdermal patches may also be used.
  • compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol,
  • the pharmaceutical compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2 octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, specifically, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutical compositions may be formulated in an ointment such as petrolatum.
  • compositions may also be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • the compounds for use in the methods of the invention can be formulated in unit dosage form.
  • unit dosage form refers to physically discrete units suitable as unitary dosage for subjects undergoing treatment, with each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, optionally in association with a suitable pharmaceutical carrier.
  • the unit dosage form can be for a single daily dose or one of multiple daily doses (e.g., 1 to 4 or more times per day). When multiple daily doses are used, the unit dosage form can be the same or different for each dose.
  • compositions, and methods of the present invention are compositions, and methods of the present invention.
  • Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered in clinical therapy and which are obvious to those skilled in the art are within the spirit and scope of the invention.
  • This example describes PAC-PBIN inhibition and antiviral activity of the initial hit compound 5.
  • the initial hit compound 5 was confirmed as a potent inhibitor of the PAC-PB IN polymerase subunit interactions in the ELISA assay with an IC50 of 4.3 ⁇ 0.1 ⁇ (FIG. 2).
  • the antiviral activity of compound 5 was tested in the antiviral plaque assay. It was found that compound 5 inhibits multiple strains of influenza A and B viruses with single to submicromolar EC50 values (Table 1).
  • the cellular cytotoxicity of compound 5 in MDCK cells with a 48 h incubation time was 17.4 ⁇ 1.2 ⁇ ; therefore, the antiviral activity of compound 5 was not due to its cellular cytotoxicity.
  • R2 substitution was benzene
  • a methoxyl group was tolerated at the ortho-, meta-, and /3 ⁇ 4zra-positions (9k and 91).
  • a small alkyl group such as ethyl (9g) was also tolerated at the para- position; however, branched alkyl group such as isopropyl (9j) and a bulky substitution such as benzene (9f) were not tolerated.
  • Compound 9u had antiviral activity similar to compound 5 (22.7% vs 16.5% plaque formation at 5 ⁇ ) as it combines the favorable substitutions from both the aldehyde component ((o- trifluoromethoxy)phenyl) and the isocyanide component (benzyl).
  • Ugi-azide 4-CR produces a new chiral center during the reaction, and hence the product is a mixture of enantiomers.
  • the enantiomers could be separated by chiral HPLC or other methods, chiral separation is generally time consuming and expensive, which presents a challenge for future development. Indeed, this somewhat compromises the advantages of exploring MCR products in the drug discovery arena. With grams of material required for possible downstream pharmacokinetic and in vivo animal studies, experiments therefore sought to develop a convenient synthesis and separation strategy to bypass chiral separation.
  • compound 12a had no cross-resistance with the FDA- approved influenza antivirals amantadine and oseltamivir, as shown by the results that compound 12a had potent antiviral activity against viruses that are resistant to amantadine, oseltamivir, or both.
  • Drug-induced resistance is one of the major obstacles facing antiviral drugs (see, e.g., Loregian, A.; et al, Cell Mol Life Sci 2014, 71, 3659-83).
  • the A/WSN/33 (H1N1) virus was amplified in the presence of increasing concentrations of compound 12a and the drug sensitivity of the resulting viruses at different passages was assayed against compound 12a using a plaque assay.
  • Oseltamivir carboxylate was included as a control. Gratifyingly, viruses at passage 10 remained sensitive to compound 12a, and no increase in EC50 value was observed (FIG. 4B and Table 4). In contrast, the EC50 for oseltamivir carboxylate increased 10-fold at passage six and onwards, which is consistent with previous reports (see, e.g., Ehrhardt, C; et al, Cell Microbiol 2013, 15, 1198-211; Shih, S. R.; et al, J Antimicrob Chemother 2010, 65, 63-71). These results indicate that compound 12a targets a vital viral replication component such as the viral polymerase that is less prone to mutate. Overall, compound 12a demonstrated a high in vitro genetic barrier to drug resistance, rendering it a desired drug candidate for further development.
  • Passage was performed using the A/WSN/33 (HlNl) virus by following our reported procedure (see, e.g., Ma, C; et al., Antiviral Res 2016, 133, 62-72; Ma, C; et al, Mol Pharmacol 2016, 90, 188-98; Hu, Y.; et al., Antiviral Res 2017, 140, 45-54).
  • Oseltamivir carboxylate was included as a control.
  • This example shows the % plaque formation, EC50, and CC50 values for specific compounds of the present invention against the A/California/07/2009 (HlNl) virus (see, Table 5).
  • This example describes a docking model of compound 12a in PAc.
  • the carbonyl from the benzimidazol-2-one in 12a forms a hydrogen bond with the E623 backbone amide NH, while the benzene ring from benzimidazol-2-one fits in the hydrophobic pocket formed by F41 1 and 1621.
  • Plaque assay The plaque reduction assay was performed as previously reported (see, e.g., Wang, J.; et al, PNAS 2013, 110, 1315-1320), except MDCK cells expressing ST6Gal I were used instead of regular MDCK cells (see, e.g., Hatakeyama, S.; et al, Journal of Clinical Microbiology 2005, 43, 4139-4146). Briefly, the confluent monolayers of ST6Gal MDCK cells were incubated with -100 pfu virus samples in DMEM with 0.5% BSA for 1 h at 4 °C, then 37 °C for 1 h. The inoculums were removed, and the cells were washed with phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the cells were then overlaid with DMEM containing 1.2% Avicel microcrystalline cellulose (FMC BioPolymer, Philadelphia, PA) and NAT (2.0 ⁇ g/mL).
  • DMEM fetal calf serum
  • NAT 2.0 ⁇ g/mL
  • the overlay media was supplemented with compounds at testing concentrations.
  • the monolayers were fixed and stained with crystal violet dye solution (0.2% crystal violet, 20% methanol).
  • Influenza A virus A/WSN/33 H1N1 was obtained from Dr. Robert Lamb at the Northwestern University.
  • the influenza viruses A/Texas/04/2009 (H1N1), B/Wisconsin/1/2010, and B/Brisbane/60/2008 were obtained from Dr. James Arthur at the Southern Research Institute. Influenza A and B viruses
  • H3N2 A/Switzerland/9715293/2013 X-247 (H3N2), FR-1366; A/Washington/29/2009 (H1N1), FR- 460; A/California/07/2009 (H1N1), FR-201; A/Washington/29/2009 (H1N1), FR-460;
  • B/Memphis/20/1996, FR-486; B/Utah/9/2014, FR-1372; and B/Phuket 3073/2013, FR-1364; were obtained through the Influenza Reagent Resource, Influenza Division, WHO Collaborating Center for Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention, Atlanta, GA, USA.
  • the influenza viruses A/Denmark/524/2009 (H1N1) and A/Denmark/528/2009 (H1N1) was obtained from Dr. Maria Govorkova at St. Jude Children's Research Hospital.
  • Cytotoxicity assay Evaluation of the cytotoxicity of compounds was carried out using the neutral red uptake assay (see, e.g., Repetto, G; del Peso, A.; Zurita, J. L. Neutral red uptake assay for the estimation of cell viability/cytotoxicity. Nat Protoc 2008, 3, 1125-31). Briefly, 80,000 cells/mL of MDCK or A549 cells in DMEM medium supplemented with 10% FBS and 100 U/mL Penicillin-Streptomycin were dispensed into 96-well cell culture plates at 100 ⁇ .
  • ELISA assay To test the inhibitory activity of compound on PAC-PB IN interaction, ELISA was performed (see, e.g., Yuan, S.; et al, Antiviral Res 2016, 125, 34-42). Briefly, microliter plates were coated with 400 ng of His-tagged PA239-716 (PAc ) for 3 h at 37 °C, followed by blocking with 2% (wt/vol) BSA in phosphate buffer saline (PBS) for 1 h. After washing with PBS containing 0.3% Tween 20, plates were incubated with 200 ng of GST- tagged PB11-25 (PB IN ) protein and compounds overnight at room temperature.
  • PAc His-tagged PA239-716
  • PBS phosphate buffer saline
  • HRP horseradish peroxidase
  • Serial drug passage experiments were performed accordingly to previously published protocol (see, e.g., Ma, C; et al., Antiviral Res 2016, 133, 62-72; Ma, C; et al, Mol Pharmacol 2016, 90, 188-98; Hu, Y.; et al., Antiviral Res 2017, 145, 103-113). Briefly, MDCK cells were infected with the A/WSN/33 (H1N1) virus at MOI 0.001 for 1 h. Then the inoculum was removed and MDCK cells were incubated with 1 ⁇ compound 12a in the first passage and the concentration of 12a was gradually increased 2-fold in passages 2-7 and kept constant at 64 ⁇ in passages 7-10.
  • the viruses were harvested when a significant cytopathic effect was observed, which usually takes 2-3 days after virus infection.
  • the titers of harvested viruses were determined by plaque assay.
  • the drug sensitivity after passages 3, 6, and 10 was determined via plaque assay as described previously (see, e.g., Hu, Y.; et al., Eur J Med Chem 2017, 135, 70-76).
  • Oseltamivir carboxylate was included as a control and similar fold of drug selection pressure was applied.
  • the drug sensitivity of oseltamivir at passages 3, 6, and 10 was determined via plaque assay.

Abstract

La présente invention concerne le domaine de la chimie pharmaceutique. En particulier, l'invention concerne une nouvelle classe de petites molécules qui présentent une structure tétrazolyl-pipéridinyl-benzoimidazole : Structure (I) qui fonctionnent comme des inhibiteurs de l'activité polymérase du virus influenza, et leur utilisation à titre d'agents thérapeutiques pour le traitement de la grippe.
PCT/US2018/058417 2017-10-31 2018-10-31 Méthodes et compositions pour le traitement de la grippe WO2019089734A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762579590P 2017-10-31 2017-10-31
US62/579,590 2017-10-31

Publications (1)

Publication Number Publication Date
WO2019089734A1 true WO2019089734A1 (fr) 2019-05-09

Family

ID=66332333

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/058417 WO2019089734A1 (fr) 2017-10-31 2018-10-31 Méthodes et compositions pour le traitement de la grippe

Country Status (1)

Country Link
WO (1) WO2019089734A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002072576A1 (fr) * 2001-03-09 2002-09-19 Pfizer Products Inc. Composes de benzimidazole anti-inflammatoires
US20140005197A1 (en) * 2010-12-16 2014-01-02 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002072576A1 (fr) * 2001-03-09 2002-09-19 Pfizer Products Inc. Composes de benzimidazole anti-inflammatoires
US20140005197A1 (en) * 2010-12-16 2014-01-02 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE Pubchem 18 May 2011 (2011-05-18), XP55613253, Database accession no. 51360314 *
ZHANG ET AL.: "Exploring Ugi-Azide Four-Component Reaction Products for Broad-Spectrum Influenza Antivirals with a High Genetic Barrier to Drug Resistance", SCIENTIFIC REPORTS, vol. 8, 15 March 2018 (2018-03-15), pages 1 - 14, XP55613235 *

Similar Documents

Publication Publication Date Title
KR102607599B1 (ko) 인플루엔자의 치료 방법
JP6615755B2 (ja) インフルエンザウイルスの複製の阻害剤
JP6618901B2 (ja) インフルエンザウイルスの複製の阻害剤を調製する方法
JP2021191796A (ja) アザインドール化合物の製剤
JP6704416B2 (ja) インフルエンザウイルスの複製の阻害剤を調製する方法
EA037529B1 (ru) Способ получения соединения для ингибирования репликации вирусов гриппа
EA037372B1 (ru) Ингибиторы репликации вирусов гриппа
JP6953308B2 (ja) インフルエンザa型ウイルスバリアント
JP6953307B2 (ja) インフルエンザa型ウイルスバリアント
US10273233B2 (en) Inhibitors of influenza viruses replication
JP2020516634A (ja) インフルエンザウイルス感染の処置方法および併用療法
WO2019089734A1 (fr) Méthodes et compositions pour le traitement de la grippe
TW201144296A (en) Tetrazolones as inhibitors of fatty acid synthase

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: 18874776

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: 18874776

Country of ref document: EP

Kind code of ref document: A1