This invention is in the field of therapeutic and prophylactic drugs to treat adenoviral infections and disease. In particular, the present invention is directed to the use of filociclovir (FCV) in the treatment and/or prevention of adenoviral infections in mammals, and in particular the treatment and/or prevention of adenoviral infections in humans.
Adenoviruses (AdVs) are ubiquitous DNA viruses that are most commonly associated with pediatric illnesses of the respiratory tract, gastrointestinal tract and conjunctiva. Other rare manifestations of AdV infections include hepatitis, as well as genitourinary and neurologic symptoms (Khanal et al., Biomedicines, 6(1) (2018)). Although most adenoviral infections are self-limiting in the immunocompetent host, disseminated forms of the disease and subsequent high fatalities can occur in immunocompromised patients and other vulnerable populations in closed or crowded settings, such as school children and military recruits (Hendrix et al., Emerg. Infect. Dis., 5(6): 798-801 (1999); Munoz et al., Clin. Infect. Dis. 27(5): 1194-1200 (1998); Trei et al., Emerg. Infect. Dis., 16(5): 769-775 (2010)). Infections in the immunocompromised are similar in scope, but more severe, particularly in transplant recipients (Lion, T., Clin. Microbiol. Rev., 27(3): 441-462 (2014)). HIV infection is also a risk factor for AdV infection and recently serotypes were isolated from this population (De Jong et al., J. Clin. Microbiol., 37(12): 3940-3945 (1999)). Transmission of adenoviruses can occur via inhalation of aerosol droplets, fecal-oral transmission, and contaminated fomites (Lynch, J. P and Kajon, A. E., Seminars in Respiratory and Critical Care Medicine, 37(4): 586-602 (2016)). More than 50 serotypes of adenoviruses have been identified and are classified as six distinct groups (A-F) based on their biochemical, immunological and morphological criteria (Robinson et al., Scientific Reports, 3: 1812 (2013)). Approximately one-third of the described serotypes are associated with human disease. Globally, serotypes 1-7, 21, and 41 are most commonly associated with human disease. Different serotypes display different tissue tropisms that correlate with clinical manifestations of infection (Lynch and Kajon 2016, supra). The spectrum of clinical disease differs with age, immune status and population characteristics. Upper respiratory tract infections (URI) are among the most common, while pneumonia and diarrhea are the deadliest in infants. Epidemic keratoconjunctivitis and acute respiratory distress syndrome (ARDS) are the main concerns in young military recruits. Adenoviruses account for at least 5-10% of pediatric and 1-7% of adult respiratory tract infections (Lee et al., Journal of Medical Virology, 82(4): 624-631 (2010)). Pneumonia occurs in up to 20% of newborns and infants (Zhang et al., Infectious diseases, 48(1): 40-47 (2016); Jain et al., The New England Journal of Medicine, 372(9): 835-845 (2015)). Adenoviruses also account for >50% of pneumonia cases among unvaccinated military recruits, not only in the US but also globally (Lynch and Kajon 2016, supra). In immunocompromised individuals, dissemination and/or severe respiratory failure develops in 10-30% of cases and fatality rates for severe adenoviral pneumonia may exceed 50% (Low et al., Respiratory Medicine, 107(11): 1810-1813 (2013); Ison, M. G., Clinical Infectious Diseases, 43(3): 331-339 (2006)). Very recently, an outbreak of AdV3 infection was reported in New Jersey at a nursing and rehabilitation center that resulted in 54 illnesses and 11 deaths. (See, https://nj.gov/health/cd/topics/adenovirus.shtml).
The options for controlling AdV infections are very limited. Live oral vaccines reduce the risk of respiratory illness and are in routine use by the US military, but currently are not available to civilians and only provide protection against types 4 and 7 (Radin et al., Clinical Infectious Diseases, 59(7): 962-968 (2014)). No antiviral drug has been approved for treating adenoviruses. Previous reports have shown specific antiviral activity for a number of nucleoside analogues in cell culture and animal models, including ribavirin, ganciclovir (GCV), cidofovir (CDV) and its prodrug brincidofovir (BCDV) (Hartline et al., The Journal of Infectious Diseases, 191(3): 396-399 (2005); Naesens et al., Antimicrobial Agents and Chemotherapy, 49(3): 1010-1016 (2005); Ying et al., Antimicrobial Agents and Chemotherapy, 58(12): 7171-7181 (2014); Toth et al., Antiviral Research, 153: 1-9 (2018); Toth et al., Proc. Nat. Acad. Sci. USA, 105(20): 7293-7297 (2008)). Ribavirin is teratogenic, causes hemolytic anemia and is active only against group C adenoviruses (Feld et al., Liver International, 37(1): 5-18 (2017); Morfin et al., Antiviral Therapy, 10(2): 225-229 (2005)). Ribavirin treatment of adenovirus infection in immunocompromised hosts yielded mixed results, which could be correlated to its serotype specificity (Ljungman, P., Eur. J. Clin. Microbiol. Infect. Dis., 23(8): 583-588 (2004); Lankester et al., Clinical Infectious Diseases, 38(11): 1521-1525 (2004)). The potency of GCV against AdV5 is very modest (IC50=66±15.1 μM), and no controlled trials exist that indicate clinical efficacy for the use of GCV (Kinchington et al, The Journal of Antimicrobial Chemotherapy, 55(4): 424-429 (2005)). Long-term use of CDV has been associated with nephrotoxicity and CNS side effects (Bhadri et al., Transplant Infectious Disease, 11(4): 373-379 (2009); Vora et al., Journal of the Pediatric Infectious Diseases Society, 6(4): 399-402 (2017)). Brincidofovir (BCDV), a lipid-linked derivative of CDV, is orally bioavailable, highly potent in vitro and is currently undergoing clinical trials (Hartline et al. 2005, supra; Waye, M. M. Y. and Sing, C. W., Pharmaceuticals, 3(10): 3343-3354 (2010)). However, these trials have only shown modest benefit for AdV disease or viremia (Lopez et al., Current opinion in organ transplantation, 23(4): 395-399 (2018)). In a recent randomized placebo-controlled Phase 2 trial, the pre-emptive treatment of AdV infections with BCDV was shown to rapidly reduce viral loads (Grimley et al., Biology of Blood and Marrow Transplantation, 23(3): 512-521 (2017)). However, GI toxicity and acute kidney injury in solid organ transplant (SOT) and graft vs. host disease in hematopoietic cell transplant (HSCT) patients were reported (Detweiler et al., Journal of Pediatric Hematology/Oncology, 40(6): e364-e368 (2018); Faure et al., Medicine, 95(44): e5226 (2016)). Two Phase 3 AdV trials were completed several years ago (NCT01143181, NCT02087306) however, the company has not sought approval. Development of BCDV for HCMV has been discontinued because of the increased frequency of serious adverse events (primarily graft vs. host disease) and 24-week all-cause mortality observed during Phase 3 (Marty et al., Biology of Blood and Marrow Transplantation, 25(20: 369-381 (2018)). In addition, two Phase 3 clinical trials for CDV in kidney transplant patients were recently terminated (NCT02439957, NCT02439970). These safety issues call into question whether the drug can be administered safely in any population.
Currently there is no drug approved for the prevention or treatment of adenovirus infection and the drug furthest in development has safety issues that will likely preclude its advancement. Therefore, there exists an urgent and unmet medical need for a safe and effective anti-adenovirus drug for use in a method for treating and/or preventing adenovirus infections.
The present invention is directed to a method for treating or preventing adenoviral (AdV) infections via administration of a small molecule inhibitor. In particular, the present invention is directed to a method for treating or preventing adenoviral infections by administration of filociclovir (FCV). The method described herein is suitable for the treatment and/or prevention of adenoviral infections in mammals, in particular humans.
In particular, the present method is directed to treating or preventing infection by specific strains of adenovirus. Therefore, in one embodiment, the present invention is directed to a method for treating or preventing infection in a mammal by adenovirus 5 (AdV5) by administration of a composition comprising filociclovir or a pharmaceutically acceptable salt thereof. In a preferred embodiment, the mammal is a human.
In another embodiment, the present invention is directed to a method for treating or preventing infection in a mammal by adenovirus 6 (AdV6) by administration of a composition comprising filociclovir or a pharmaceutically acceptable salt thereof. In a preferred embodiment, the mammal is a human.
In another embodiment, the present invention is directed to a method for treating or preventing infection in a mammal by adenovirus 8 (AdV8) by administration of a composition comprising filociclovir or a pharmaceutically acceptable salt thereof. In a preferred embodiment, the mammal is a human.
In another embodiment, the present invention is directed to the use of filociclovir in the manufacture of a medicament for treating or preventing adenovirus infection in a mammal. In a preferred embodiment, the present invention is directed to the use of filociclovir in the manufacture of a medicament to treat or prevent infection by AdV5, AdV6, and/or AdV8.
Also disclosed are pharmaceutical compositions comprising a therapeutically effective amount of filociclovir, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. The pharmaceutical compositions are suitable for use in the disclosed methods for treating or preventing adenoviral infections in a mammal, particularly humans. The pharmaceutical compositions may be formulated for both parenteral and/or nonparenteral administration to a subject or patient in need thereof.
In another embodiment, filociclovir may be administered to a subject in need thereof optionally in combination with one or more known antiviral agents. The additional antiviral agent or agents may be administered before, simultaneously with, or after administration of the filociclovir.
In preferred embodiments, the filociclovir compound/composition of the present invention exhibits an inhibitory concentration of ≤10 μM against infection by adenovirus and a cytotoxicity (CC50) of ≥100-150 μM.
A composition or method described herein as “comprising” (or “comprises”) one or more named elements or steps is open-ended, meaning that the named elements or steps are essential, but other elements or steps may be added within the scope of the composition or method. To avoid prolixity, it is also understood that any composition or method described as “comprising” one or more named elements or steps also describes the corresponding, more limited, composition or method “consisting essentially of” (or “consists essentially of”) the same named elements or steps, meaning that the composition or method includes the named essential elements and may also include additional elements or steps that do not materially affect the basic and novel characteristic(s) of the composition or method. It is also understood that any composition or method described herein as “comprising” or “consisting essentially of” one or more named elements or steps also describes the corresponding, more limited, and closed-ended composition or method “consisting of” (or “consists of”) the named elements or steps to the exclusion of any other element or step. In any composition or method disclosed herein, known or disclosed equivalents of any named essential element or step may be substituted for that element or step, respectively.
Filociclovir (also referred to as cyclopropavir, 2-amino-9-{(Z)-[2,2-bis(hydroxymethyl)cyclopropylidene]methyl}-3,9-dihydro-6H-purin-6-one), and salts thereof, is a methylenecyclopropane nucleoside analogue represented by the following structure:
As used herein, the term “subject” can be a human, non-human primate, horse, pig, rabbit, dog, sheep, goat, cow, cat, guinea pig or rodent. A “patient” or “subject in need thereof” refers to a mammal afflicted with a disease or disorder. The term “patient” includes human and veterinary subjects.
Terms such as “parenteral”, “parenterally”, and the like, refer to routes or modes of administration of a compound or composition to an individual other than along the alimentary canal. Examples of parenteral routes of administration include, without limitation, subcutaneous (s.c.), intravenous (i.v.), intramuscular (i.m.), intra-arterial (i.a.), intraperitoneal (i.p.), transdermal (absorption through the skin or dermal layers), nasal (“intranasal”; absorption across nasal mucosa), or pulmonary (e.g., inhalation for absorption across the lung tissue), vaginal, direct injections or infusions into body cavities or organs other than those of the alimentary canal, as well as by implantation of any of a variety of devices into the body (e.g., of a composition, depot, or device that permits active or passive release of a compound or composition into the body).
The terms “non-parenteral”, “non-parenterally”, “enteral”, “enterally”, “oral”, “orally”, and the like, refer to administration of a compound or composition to an individual by a route or mode along the alimentary canal. Examples of enteral routes of administration include, without limitation, oral, as in swallowing solid (e.g., tablet) or liquid (e.g., syrup) dosage forms, sublingual (absorption through the mucosal membranes lining the floor of the mouth, e.g., under the tongue), buccal (absorption through the mucosal membranes lining the cheeks), nasojejunal or gastrostomy tubes (delivery into the stomach), intraduodenal administration, as well as rectal administration (e.g., suppositories for release of a drug composition into and absorption by the lower intestinal tract of the alimentary canal).
Human adenoviruses belong to the adenoviridae family, which is distinct from the herpesviridae family. For example, adenoviruses are double-stranded DNA viruses but, unlike herpesviruses, they are non-enveloped with an icosahedral nucleocapsid, and carry elongated fiber proteins on their outer surface. Adenoviruses are classified into seven species designated human adenovirus A to G. These viruses mainly infect the upper or lower respiratory tracts, conjunctiva, or gastrointestinal tract, causing a whole array of clinical manifestations that are distinct from those caused by herpesviruses. Most infections occur in children and immunocompromised individuals. Adenoviruses are not known to encode virus-encoded kinases, which implies a novel mechanism for filociclovir activation.
Other rare manifestations of AdV infections include hepatitis, as well as genitourinary and neurologic symptoms (Khanal et al. 2018, supra). The options for controlling AdV infections are very limited. Live oral vaccines reduce the risk of respiratory illness and are in routine use by the US military, but currently are not available to civilians and only provide protection against types 4 and 7 (Radin et al. 2014, supra). No antiviral drug has been approved for treating adenoviruses.
Herein we describe the discovery of a novel method for treating and/or preventing adenovirus infection in a mammal by administration of the small molecule inhibitor, filociclovir (FCV). Our results demonstrate that filociclovir is unexpectedly effective for use in a method for treating or preventing adenovirus infection and is more effective at treating or preventing adenovirus infection than previously known treatments.
The present method for the use of filociclovir to treat or prevent adenovirus infection is particularly effective against a number of strains of adenovirus including, but not limited to adenovirus 3 (AdV3), adenovirus 4 (AdV4), adenovirus 5 (AdV5), adenovirus 6 (AdV6), adenovirus 7 (AdV7), adenovirus 8 (AdV8), adenovirus 19 (AdV19), and adenovirus 34 (AdV37).
The present method for the use of filociclovir to treat adenovirus infection is particularly suited for the treatment or prevention of adenovirus infections involving the upper respiratory tract and adenoviral infections of the eye, e.g., conjunctivitis, and in particular, keratoconjunctivitis involving the ocular surface of the eye.
Therefore, the present invention provides a novel method directed to anti-adenovirus therapeutics for treatment or prevention of adenoviral infection of mammalian cells. In particular, the present invention is directed to the use of filociclovir to treat or prevent adenovirus infection in a mammalian subject. In a preferred embodiment, the mammal is a human.
The data described herein demonstrate that filociclovir is an effective pan-adenoviral therapeutic; thus, therapeutic administration of filociclovir is effective to treat or prevent infection by multiple strains of adenovirus. As described below, using several assays, it is demonstrated that filociclovir is a potent, safe, and effective inhibitor of a number of strains of adenovirus. For example, as set forth in Example 1, we used a cytopathic effects (CPE) reduction assay to demonstrate that filociclovir is a more potent inhibitor of adenovirus 5 than a number of other known viral inhibitors including cidofovir and ganciclovir (see, Table 1 and
Next we used a neutral red survival assay to demonstrate that filociclovir is effective to reduce the cytotoxic effects of adenovirus 6. The results are shown in
As described in Example 3, an immunofluorescence stain assay was used to demonstrate that filociclovir is effective at inhibiting ‘late’ replication of AdV in infected human A549 cells. In this assay, cells were infected with either AdV5 or AdV6 or mock-infected. After 1 hour, filociclovir was added to a final concentration of 0, 4, 10, or 40 μM. The results are shown in
Using a Syrian hamster model of adenovirus infection as shown in Examples 4-7, it is demonstrated that filociclovir exhibits favorable bioavailability when administered both orally and intravenously (see,
In Example 6, using the same Syrian hamster model described above, we also demonstrate that following AdV infection, even low doses of filociclovir (3 mg/kg and 10 mg/kg) are effective to prevent morbidity and mortality (
In Example 7 it is shown that late administration of filociclovir (10 mg/kg), e.g., up to 4 days post-challenge, is effective to prevent morbidity and mortality (see,
Therefore, in one embodiment, the present invention is directed to the use of filociclovir in a method for treating or preventing infection of a mammal by adenovirus 5 (AdV5).
In another embodiment, the present invention is directed to the use of filociclovir in a method for treating or preventing infection of a mammal by adenovirus 6 (AdV6).
In another embodiment, the present invention is directed to the use of filociclovir in a method for treating or preventing infection of a mammal by adenovirus 7 (AdV7).
In yet another embodiment, the present invention is directed to the use of filociclovir in a method for treating or preventing infection of a mammal by adenovirus 8 (AdV8).
Results also demonstrate that filociclovir is effective to treat or prevent infection of a mammal by adenovirus 3 (AdV3), adenovirus 4 (AdV4), adenovirus 19 (AdV19), and adenovirus 37 (AdV37). (Data not shown).
In a preferred embodiment, the mammal is a human.
The present invention is further directed to the use of filociclovir in a method for the manufacture of a medicament for treating or preventing adenovirus infection in mammals (e.g., humans) comprising combining filociclovir or a composition comprising filociclovir with a pharmaceutically acceptable carrier or diluent. Thus, in one aspect, the invention relates to a method for manufacturing a medicament comprising combining at least one disclosed compound, e.g., filociclovir, according to the present invention or at least one disclosed product with a pharmaceutically acceptable carrier or diluent.
Individual dose requirements for administration to a subject will be determined by the caregiver, for example, a physician, primary care physician, or nurse.
As described herein, it has been demonstrated that filociclovir exhibits a dose-dependent inhibition of adenovirus infection with IC50 values ≤5 μM and a minimal mammalian cytotoxicity (CC50) of preferably ≥100-150 μM.
Filociclovir can be administered as a pharmaceutically acceptable salt. Such pharmaceutically acceptable salts include the gluconate, lactate, acetate, tartarate, citrate, phosphate, maleate, borate, nitrate, sulfate, and hydrochloride salts. The salts of the compounds described herein can be prepared, for example, by reacting the base compound with the desired acid in solution. After the reaction is complete, the salts are crystallized from solution by the addition of an appropriate amount of solvent in which the salt is insoluble. In some embodiments, the hydrochloride salt is made by passing hydrogen chloride gas into an ethanolic solution of the free base.
In another embodiment, the compounds are formulated into a pharmaceutically acceptable carrier or excipient for administration to a subject in need thereof. In another embodiment, the compounds may be formulated into a pharmaceutical formulation and further comprise an additional antiviral compound. In another embodiment, the pharmaceutical formulation may be formulated to be administered orally, parenterally, or topically.
It is preferable to develop an orally active therapeutic, since that is the most convenient and rapid method to administer a drug to a large exposed population in case of pandemic. Currently, live oral adenovirus vaccines for protection against serotypes 4 and 7 are in use by the military but are not available to the civilian population. However, it is also expected that filociclovir as described herein will be suitable for intravenous (i.v.) administration, because it is envisioned that in case of a natural outbreak the infected patients may require i.v. administration. Therefore, the method described herein will provide an effective, safe, and easy therapeutic option for any newly emerged pandemic strain(s) of adenovirus.
The methods of the presently disclosed subject matter are useful for treating these conditions in that they inhibit the onset, growth, or spread of the condition, cause regression of the condition, cure the condition, or otherwise improve the general well-being of a subject afflicted with, or at risk of, contracting the condition. Thus, in accordance with the presently disclosed subject matter, the terms “treat”, “treating”, and grammatical variations thereof, as well as the phrase “method of treating”, are meant to encompass any desired therapeutic intervention, including but not limited to a method for treating an existing adenovirus infection in a subject, and a method for the prophylaxis (i.e., preventing) of an adenovirus infection, such as in a subject that has been exposed to a virus as disclosed herein or that has an expectation of being exposed to a virus as disclosed herein.
Pharmaceutical compositions according to the invention comprise the adenovirus inhibitor, filociclovir, as described herein, or a pharmaceutically acceptable salt thereof, as the ‘active ingredient’ and a pharmaceutically acceptable carrier (or ‘vehicle’), which may be a liquid, solid, amorphous, or semi-solid compound.
In some embodiments, the presently disclosed subject matter is related to a method of treating or preventing an adenovirus infection in a subject in need of treatment thereof wherein the method comprises administering to the subject an effective amount of a composition comprising filociclovir. Filociclovir may be administered alone or optionally in combination with one or more additional antiviral agents.
In another embodiment, filociclovir may be formulated into a pharmaceutically-acceptable carrier and applied/administered to a subject in need thereof by an injection, including, without limitation, intradermal, transdermal, intramuscular, intraperitoneal and intravenous. According to another embodiment of the invention, the administration is oral and the compound may be presented, for example, in the form of a tablet or encased in a gelatin capsule or a microcapsule, which simplifies oral application. The production of these forms of administration is within the general knowledge of a technical expert. Multiple routes of administration are envisioned for the method described herein and highly cost-effective production strategies can be easily achieved.
In another aspect, the invention relates to pharmaceutical compositions comprising filociclovir, or a pharmaceutically acceptable salt, solvate, hydrate, or polymorph thereof, and a pharmaceutically acceptable carrier.
In a further aspect, the pharmaceutical composition is a solid dosage form selected from a capsule, a tablet, a pill, a powder, a granule, an effervescing granule, a gel, a paste, a troche, and a pastille. In a still further aspect, the pharmaceutical composition is a liquid dosage form selected from an emulsion, a solution, a suspension, a syrup, and an elixir.
As used herein, the term “pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids. When the compound of the present invention is acidic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases.
Salts derived from such inorganic bases include aluminum, ammonium, calcium, copper (-ic and -ous), ferric, ferrous, lithium, magnesium, manganese (-ic and -ous), potassium, sodium, zinc and the like salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, as well as cyclic amines and substituted amines such as naturally occurring and synthesized substituted amines. Other pharmaceutically acceptable organic non-toxic bases from which salts can be formed include ion exchange resins such as, for example, arginine, betaine, caffeine, choline, N,N′-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.
As used herein, the term “pharmaceutically acceptable non-toxic acids”, includes inorganic acids, organic acids, and salts prepared therefrom, for example, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid and the like. Preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.
In preparing the compositions for oral dosage form, any convenient pharmaceutical media can be employed. For example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like can be used to form oral liquid preparations such as suspensions, elixirs and solutions; while carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like can be used to form oral solid preparations such as powders, capsules and tablets. Because of their ease of administration, tablets and capsules are the preferred oral dosage units whereby solid pharmaceutical carriers are employed. Optionally, tablets can be coated by standard aqueous or nonaqueous techniques
A tablet containing the composition of this invention can be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants. Compressed tablets can be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
Pharmaceutical compositions of the present invention suitable for parenteral administration can be prepared as solutions or suspensions of the active compounds in water, from, for example, lyophilized samples of the active compounds. A suitable surfactant can be included such as, for example, hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms.
Pharmaceutical compositions of the present invention suitable for injectable use include sterile aqueous solutions, emulsions, or dispersions. Furthermore, the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be effectively fluid for easy syringability. The pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), vegetable oils, and suitable mixtures thereof.
Pharmaceutical compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, mouth washes, gargles, and the like. Further, the compositions can be in a form suitable for use in transdermal devices. These formulations can be prepared, utilizing a compound of the invention, or pharmaceutically acceptable salts thereof, via conventional processing methods. As an example, a cream or ointment is prepared by mixing hydrophilic material and water, together with about 5 wt % to about 10 wt % of the compound, to produce a cream or ointment having a desired consistency.
Pharmaceutical compositions of this invention can be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories can be conveniently formed by first admixing the composition with the softened or melted carrier(s) followed by chilling and shaping in molds.
In addition to the aforementioned carrier ingredients, the pharmaceutical formulations described above can include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti-oxidants) and the like. Furthermore, other adjuvants can be included to render the formulation isotonic with the blood of the intended recipient. Compositions containing a compound of the invention, and/or pharmaceutically acceptable salts thereof, can also be prepared in powder or liquid concentrate form.
In one aspect, the invention relates to a kit comprising at least filociclovir according to the present invention, or a pharmaceutically acceptable salt, solvate, or polymorph thereof; and one or more of:
The kits can also comprise compounds and/or products co-packaged, co-formulated, and/or co-delivered with other components. For example, a drug manufacturer, a drug reseller, a physician, a compounding shop, or a pharmacist can provide a kit comprising a disclosed compound of the present invention and/or product and another component for delivery to a patient.
In a further aspect, the kit further comprises a plurality of dosage forms, the plurality comprising one or more doses; wherein each dose comprises an amount of the compound and the agent known to have antiviral activity. In another aspect, the kit further comprises a plurality of dosage forms, the plurality comprising one or more doses; wherein each dose comprises an effective amount of the compound and the agent known to have antiviral activity.
In a further aspect, an effective amount is a therapeutically effective amount. In a still further aspect, an effective amount is a prophylactically effective amount.
The following Examples have been included to illustrate modes of the presently disclosed subject matter. In light of the present disclosure and the general level of skill in the art, those of skill will appreciate that the following Examples are intended to be exemplary only and that numerous changes, modifications, and alterations can be employed without departing from the invention as presently disclosed.
Human foreskin tissue was obtained from the University of Alabama at Birmingham tissue procurement facility with approval from the Institutional Review Board. The tissue was stored at 4° C. in cell culture medium consisting of minimum essential media (MEM) with Earle's salts supplemented with 10% fetal bovine serum (FBS; HyClone, Inc., Logan, Utah) and standard concentration of L-glutamine, amphotericin B (Fungizone), and vancomycin. The tissue was then placed in a phosphate-buffered saline solution, minced, and rinsed to remove the red blood cells. Tissue fragments were then resuspended in a trypsin-EDTA solution and incubated at 37° C. to disperse the cells, which were then collected by centrifugation. Cell pellets were then resuspended in 4 ml culture medium, placed in a 25-cm2 tissue culture flask, and incubated at 37° C. for 24 h. The culture medium was then replaced with fresh medium and condition of the cells was monitored daily until a confluent cell monolayer was formed. The HFF cells were then expanded through serial passages in standard growth medium of MEM with Earle's salts supplemented with 10% FBS, L-glutamine, penicillin, and gentamicin. Each lot of cells was confirmed to be free of mycoplasma infection and routinely passaged or used for assays at or before passage 10.
Antiviral and cytotoxicity data were obtained in a series of three to five separate experiments for each virus to provide an accurate estimate of antiviral activity and statistical data. Each assay included positive and negative control compounds as well as infected and uninfected controls to ensure the integrity of the experiment. The concurrent assessment of cytotoxicity was performed in each assay plate using the same number of cells and equivalent levels of compound exposure so that accurate selective index (SI) values could be obtained. All liquid handling steps were performed on a BioMek 4000 and significantly increased the efficiency of the assays and reduced hands-on time of analysts.
The CPE reduction assays were performed in monolayers of human foreskin fibroblast (HFF) cells (5000/well) in 384-well plates in assay media consisting of MEM with Earle's salts, 2% FBS and standard concentrations of L-glutamine, penicillin and gentamycin. 5000 cells were seeded in 384-well microtiter plates and incubated at 37° C. in a humidified 5% CO2 incubator for 24 hours to allow for formation of confluent monolayers. Dilutions of antiviral test compounds, including filociclovir, were prepared directly in the plates in a series of 5-fold dilutions in duplicate wells to yield final concentrations that ranged from 0.1 to 300 μM or from 0.003 to 10 μM which allows for a large dynamic range of compound concentrations to facilitate the detection of antiviral activity of unknown compounds with either weak or potent antiviral activity.
Cell monolayers were infected at a multiplicity of infection (MOI) of approximately 0.005 PFU/cell with virus strains including AdV5. Infected cells were incubated at 37° C. until 100% CPE was observed in the virus control wells. Cytopathology was determined by the addition of CellTiter-Glo® reagent (Promega, Madison, Wis.). Concentrations of antiviral test compounds sufficient to reduce CPE by 50% (EC50) were interpolated from the experimental data using standard methods in Microsoft® Excel. Cytotoxicity was also determined with CellTiter-Glo® luminescent cell viability assay (Promega, Madison, Wis.) and concentrations of the test compounds that decreased cell viability by 50% (CC50) were calculated from the data and selective index (SI) values were calculated as the CC50/EC50 as a measure of antiviral activity. The results are shown in Table 1 below and
aValues shown represent the average EC50 and CC50 values (μM) and SD from five independent experiments.
The results shown in Table 1 demonstrate that filociclovir (FCV) exhibits significantly improved inhibitory activity against adenovirus 5 (SI=43) over all the other test compounds and was at least twice as potent as the next most effective antiviral agent, cidofovir (CDV) (SI=24) and significantly better than ganciclovir (GCV) (SI≥1.5).
The percent reduction in AdV5 replication as a function of filociclovir concentration ranging from 0.01 to 100 μM is shown in
Similar experiments were carried out with filociclovir against adenovirus 6, adenovirus 7, and adenovirus 8. The results are shown below in Table 2. Filociclovir exhibited potent inhibition of adenovirus 8 (SI≥61).
The neutral red cell cytotoxicity assay was used to detect cell viability or drug cytotoxicity. The principle of this assay is based on the detection of viable cells via the uptake of the dye neutral red. Neutral red is a eurhodin dye that stains lysosomes in viable cells. Viable cells can take up neutral red via active transport and incorporate the dye into their lysosomes, but non-viable cells are unable not take up this chromophore. Consequently, after washing, viable cells can release the incorporated dye under acidified-extracted conditions. The amount of released dye can be quantified and used to determine the total number of viable cells or drug cytotoxicity. As such, cytotoxicity is expressed as a concentration-dependent reduction of the uptake of neutral red after exposure to the compound under investigation.
The neutral red survival assays were carried out in a 96-well format. Briefly, A549 adenocarcinomic human alveolar basal epithelial cells were plated at a concentration of 6×103 cells per well one day prior to infection. AdV6 infections were carried out at 5×103 PFU per well (an estimated 0.5 PFU/cell at the time of infection). Filociclovir was serially diluted 1:3 on separate 96-well plates with a no drug final row as control. The AdV6 infections were performed as 9 replicate wells for each drug concentration and there were 3 uninfected drug control wells for each drug concentration. Filociclovir dilutions were added to the cell plates immediately before infection with AdV6. At 6 days post-infection (when viral cytopathic effect had reached 70-90% for the virus-infected, no drug wells), neutral red was added for 1 hour, then plates were washed 3 times with PBS to remove unattached cells, and the neutral red was extracted from the remaining cells using 50% ethanol/l % glacial acetic acid. Filociclovir plates were read and the results were graphed using GraphPad Prism graphing and statistical software (GraphPad Software, San Diego, Calif.).
The results are shown in
Human A549 cells on glass coverslips in 6-well plates were infected at an MOI of 5 PFU/cell with either AdV5 or AdV6 or were mock-infected. After 1 hour, filociclovir was added to a final concentration of 0, 4, 10, or 40 μM. At 27 hours post-infection, the A549 cells were fixed in paraformaldehyde (3.7% in PBS) and permeabilized with methanol. Cells were stained for the Adenovirus DNA-binding protein and adenovirus hexon. DBP staining will be antinuclear and uniform during early AdV infection (prior to AdV DNA replication). DBP associates with replication centers as the infection progresses. The replication centers are initially small “dots” (each dot results from one incoming AdV genome). The replication centers will expand and “multiply” as DNA replication takes place. The nuclei become enlarged and misshapen as infection progresses. AdV hexon, the most abundant component of the AdV viral capsid, is not expressed until after DNA replication is taking place and is therefore considered to be a “late” AdV protein. The results are shown in
As seen in
The results show that filociclovir is a potent inhibitor of human adenovirus 5 (AdV5), AdV6, and AdV8. This level of activity is similar to that of cidofovir (CDV) and far superior to that of ganciclovir (GCV), which has an IC50 value of 66 μM against AdV5. (See, Table 1) The range of AdV serotypes tested so far suggest that FCV could potentially be developed as a pan-adenoviral inhibitor.
In order to test the pharmacokinetics of filociclovir, we use a Syrian male hamster model of adenovirus infection. Syrian hamsters were given either an oral (p.o. 50 mg/kg) or intravenous (i.v. 10 mg/kg) dose of filociclovir. The results are shown in
The Syrian male hamster model is particularly advantageous as it mimics the pathology seen with human patients, and it can be used to test the efficacy of antiviral compounds (reviewed in Wold, W. S. M. and Toth, K., Advances in Cancer Research, 115: 69-92 (2012)). Syrian hamsters are one of the two rodent species (the other is the cotton rat) that are permissive to AdV species C infections (types 1, 2, 5, and 6).
Using this model, it is possible to carry out controlled in vivo experiments to test the efficacy of anti-adenoviral compounds (Ying et al., Antimicrobial Agents and Chemotherapy, 58(12): 7171-7181 (2014); Toth et al., Proc. Natl. Acad. Sci. USA, 105(20): 7293-7297 (2008); Tollefson et al. 2014; Toth et al., Viruses, 7(3): 1409-1428 (2015)).
For these experiments, young hamsters were immunosuppressed with cyclophosphamide (CP), an agent often used as part of the conditioning regimen for human transplant recipients. After the desired degree of immunosuppression was achieved, the hamsters were infected intravenously (iv) with AdV5, leading to replication of AdV5 in most organs, most prominently in the liver (Toth et al. 2008 supra).
For p.o. administration, filociclovir was suspended in 0.4% carboxymethyl cellulose (Sigma C5678) at 5 mg/ml and sonicated to visual homogeneity. For i.v. administration, filociclovir was dissolved in DMSO (Sigma D2650) and then diluted with water to a final concentration of 5 mg/ml for filociclovir and 75% DMSO. The dosing solutions were prepared one day before use and stored at 4° C.
Male Syrian hamsters were purchased from Envigo (Hackensack, N.J.) at ˜100 g body weight. All hamsters were immunosuppressed using cyclophosphamide (CP) administered intraperitoneally at a dose of 140 mg/kg, and then twice weekly at a dose of 100 mg/kg. The animals received three injections of CP before the administration of filociclovir. Two groups of animals (12 hamsters/group) were used for the two routes of administrations. In addition, three untreated animals (the same three hamsters for both routes) served as mock controls. The p.o. and i.v. experiments were performed on two consecutive days. As the hamsters weighed approximately 100 g, (+/−10%), they were administered 1 ml and 0.2 ml for the p.o. and i.v. routes, respectively, to achieve the desired dose. For both routes, 3 hamsters were sacrificed at 0.5, 1, 3, and 6 hr post dosing.
Plasma and liver samples were collected from all animals and stored at −80° C. until analyzed by LC MS as follows:
As set forth above, it has been established that filociclovir is an effective inhibitor against several strains of adenovirus replication in vitro and demonstrates good oral bioavailability in hamsters.
Next, we assessed whether filociclovir exhibits anti-adenoviral efficacy in immunosuppressed Syrian hamsters infected intravenously (i.v.) with AdV6. Four dose levels were tested: 10, 30, 60, and 100 mg/kg p.o. q.d. The highest dose was determined based on rat toxicology data.
All hamsters were immunosuppressed using cyclophosphamide (CP). CP was administered intraperitoneally at a dose of 140 mg/kg, and then twice weekly at a dose of 100 mg/kg.
Filociclovir, in powdered form, was suspended in 0.4% carboxymethyl cellulose (Sigma C5678) at 1, 3, 6, and 10 mg/ml and sonicated to visual homogeneity and aliquots stored at 4° C. Aliquots were allowed to equilibrate to room temperature before dosing. Hamsters weighing approximately 100 g were dosed with a 1 ml volume of the appropriate suspension for the 10, 30, 60, and 100 mg/kg dose levels. References to “vehicle” refers to 0.4% carboxymethyl cellulose
The hamsters were divided into 8 groups, 15 hamsters/group, (with the exception of Group 2, which had only 5 hamsters; see Table 3), immunosuppressed, and then injected i.v. with vehicle (Groups 1-2) or 2×1010 PFU/kg of AdV6 (Groups 5-9). Groups 1 and 3 received drug vehicle (p.o. q.d.), Group 4 received 10 mg/kg filociclovir (p.o. q.d.), Group 5 received 30 mg/kg filociclovir (p.o. q.d.), Group 6 received 60 mg/kg filociclovir (p.o. q.d.), Groups 2 and 7 received 100 mg/kg filociclovir (p.o. q.d.), and Group 8 received cidofovir (20 mg/kg, 3 times weekly). For all groups, drug administration started 1 day before challenge, and continued according to the schedule above for the duration of the study.
The body weights and any signs of morbidity of the animals were recorded daily. At 5 days post challenge, 5 hamsters (designated at the start of the experiment) from each group (with the exception of Group 2) were sacrificed, and gross pathological observation was performed. Serum and liver samples were collected and the virus burden in liver was determined by a TCID50 assay, and the serum was analyzed for transaminase levels. The remaining 10 hamsters were sacrificed at 14 days post challenge. Hamsters that became visually moribund before Day 14 were sacrificed as needed. In addition to the hamsters observed to be moribund, all hamsters that lost more than 20% of their original body weight were also sacrificed. For the hamsters sacrificed at the conclusion of the experiment and those sacrificed as moribund, serum and liver tissues were collected and banked for possible determination of serum transaminase levels and virus burden in the liver. Liver tissue was preserved in formalin for possible histopathological examination.
21
61
71
1Removed from study
There were 5 treatment-related deaths in the experiment, all in the AdV6+Vehicle group (
At the day 5 (D5) sacrifice, extensive kidney pathology was observed for animals in the AdV6+60 mg/kg and 100 mg/kg filociclovir groups.
At 5 days post challenge, serum was collected from 5 animals in each group and analyzed for transaminase levels. Two hamsters in the AdV6+Vehicle group had high transaminase levels, while none of the filociclovir- or CDV-treated animals had elevated serum transaminase levels (
At 5 days post challenge, untreated AdV6-infected hamsters had high virus burden in their liver (
Data with the two lower doses of filociclovir (10 mg/kg and 30 mg/kg) are favorable. Treatment with either of these two doses inhibited virus replication (
Next, we assessed whether low doses of filociclovir exhibit anti-adenoviral efficacy in immunosuppressed Syrian hamsters infected intravenously (i.v.) with AdV6. Three dose levels were tested: 1, 3, and 10 mg/kg p.o. q.d.
All hamsters were immunosuppressed using cyclophosphamide (CP) administered intraperitoneally at a dose of 140 mg/kg, and then twice weekly at a dose of 100 mg/kg. Filociclovir in powdered form was suspended in 0.4% carboxymethyl cellulose (Sigma C5678) at 0.1, 0.3, and 1.0 mg/ml and sonicated to visual homogeneity. Filociclovir was made up once weekly, aliquoted into daily portions, and stored at 4° C. Aliquots were allowed to equilibrate to room temperature before dosing.
Hamsters were distributed into 7 groups, 15 hamsters/group (see Table 4), immunosuppressed as described above, and injected i.v. with vehicle (Groups 1-2) or 4×1010 PFU/kg of AdV6 (Groups 3-7). Groups 1 and 3 received drug vehicle (p.o. q.d.), Groups 2 and 6 received 10 mg/kg filociclovir (p.o. q.d.), Group 4 received 3 mg/kg filociclovir (p.o. q.d.), Group 5 received 1 mg/kg filociclovir (p.o. q.d.), and Group 7 received cidofovir (one dose of 37 mg/kg, followed by 20 mg/kg doses 3 times weekly). For all groups, drug administration started 1 day before challenge, and continued according to the schedule above for the duration of the study.
The body weights and any signs of morbidity of the animals were monitored and recorded daily. At 5 days post challenge, 5 hamsters (designated at the start of the experiment) from each group were sacrificed, and gross pathological observation was performed. Serum and liver were collected and virus burden in liver was determined by TCID50 assay and the serum was analyzed for transaminase levels. The remaining 10 hamsters were sacrificed at 14 days post challenge. Hamsters that became moribund before Day 14 were sacrificed as needed. In addition to the animals judged moribund by observation, we sacrificed all hamsters that lost more than 20% of their original body weight. For all animals, liver tissue was preserved in formalin for histopathological examination.
There were 14 treatment-related deaths in this experiment; 9 in the AdV6+Vehicle group and 5 in the AdV6+1 mg/kg filociclovir-treated group (
At the Day 5 sacrifice, 4 hamsters in the AdV6+Vehicle and 3 hamsters in the AdV6+1 mg/kg filociclovir groups had mottled liver and enlarged gall bladder (a sign of adenovirus pathology). Similar findings were noted in animals that were sacrificed as moribund. No other significant findings were noted at the Day 14 sacrifice.
The 10 mg/kg filociclovir q.d. dose prevented AdV6-induced liver damage as determined by serum transaminase levels, while it was largely ineffective at the 1 mg/kg filociclovir q.d. level (
The 10 mg/kg filociclovir q.d. dose inhibited AdV6 replication in the liver, while it was ineffective at the 1 mg/kg q.d. dose (
The data suggest that the 10 mg/kg p.o. q.d. dose of filociclovir is efficacious in preventing the pathology caused by intravenous infection of immunosuppressed hamsters with AdV6 while 3 mg/kg filociclovir had a moderate effect, however 1 mg/kg filociclovir was not efficacious.
Male Syrian hamsters (Envigo, Hackensack, N.J.) at 60 to 80 g were immunosuppressed by administration of cyclophosphamide (CP) intraperitoneally at a dose of 140 mg/kg, and then twice weekly at a dose of 100 mg/kg.
Filociclovir in powdered form was suspended in 0.4% carboxymethyl cellulose (Sigma C5678) at 1 mg/mi and sonicated to visual homogeneity. Filociclovir was made up once weekly, aliquoted into daily portions, and stored at 4° C. Aliquots are allowed to equilibrate to room temperature before dosing.
Hamsters were distributed into 8 groups, 15 hamsters each (Table 5), immunosuppressed as described above, and then injected i.v. with vehicle or 4×1010 plaque forming units (PFU) of AdV6 per kg. Filociclovir was administered p.o. at 10 mg/kg q.d., starting at one day before, or 1, 2, 3, or 4 days after AdV6 injection. An AdV6-infected group that did not receive filociclovir, and groups that received virus vehicle and drug vehicle only or virus vehicle plus drug (started at one day before challenge) were used as controls.
Body weights and any signs of morbidity in the hamsters were recorded daily. At 7 days post challenge, 5 hamsters from each group (designated at the start of the experiment) were sacrificed, and gross pathological observations were performed. Serum and liver were collected and virus burden in liver was determined by TCID50 assay and the serum was analyzed for transaminase levels. The remaining 10 hamsters were sacrificed at 14 days post challenge. Hamsters that became moribund before Day 14 were sacrificed as needed. In addition to the hamsters that were determined to be moribund by observation, we also sacrificed all hamsters that lost more than 20% of their original body weight.
There were 14 treatment-related deaths in this experiment; 9 in the AdV6+Vehicle group and 5 in the AdV6+1 mg/kg filociclovir group (
At the Day 5 sacrifice, 4 hamsters in the AdV6+Vehicle and 3 in the AdV6+1 mg/kg filociclovir groups had mottled liver and enlarged gall bladder (a sign of adenovirus pathology). Similar findings were noted in hamsters that were sacrificed as moribund. No other significant findings were noted. No significant findings were noted at the Day 14 sacrifice.
The 10 mg/kg q.d. filociclovir dose prevented AdV6-induced liver damage as determined by serum transaminase levels, however, filociclovir was largely ineffective at the 1 mg/kg q.d. dose level (
The 10 mg/kg q.d. filociclovir dose inhibited AdV6 replication in the liver, while it was ineffective at 1 mg/kg q.d. (
The data demonstrate that the 10 mg/kg p.o. q.d. dose of filociclovir is safe and effective in preventing the pathology caused by intravenous infection of immunosuppressed hamsters with AdV6 up to 4 days post-infection. The 1 mg/kg filociclovir dose was not efficacious, while the 3 mg/kg filociclovir dose had a moderate effect.
Consideration of the foregoing data showed that filociclovir was an effective antiviral agent for inhibiting a range of adenovirus types, leading to the conclusion that filociclovir is useful as a pan-adenoviral therapeutic.
All publications, patent applications, patents, and other documents cited herein are incorporated by reference in their entirety. The examples set forth above are illustrative only and not intended to be limiting. Obvious variations to the disclosed methods and alternative embodiments of the invention will be apparent to those skilled in the art in view of the foregoing disclosure. All such obvious variants and alternatives are considered to be within the scope of the invention as described herein
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2020/021548 | 3/6/2020 | WO | 00 |
Number | Date | Country | |
---|---|---|---|
62815630 | Mar 2019 | US |