Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TOPICAL COMPOSITIONS COMPRISING CARBOMER FOR THE TREATMENT AND PREVENTION OF VIRAL INFECTIONS AND ALLERGIC CONDITIONS
Document Type and Number:
WIPO Patent Application WO/2017/212422
Kind Code:
A1
Abstract:
Methods and topically administrable compositions for treating or preventing viral infections or allergic conditions or the symptoms thereof in a mammalian subject in need thereof which comprises topically administering to the subject a composition comprising a therapeutically effective amount of at least one pharmaceutically acceptable carbomer dispersed in an aqueous monovalent salt solution buffered to approximately neutral pH. The methods and compositions may be used for the prevention or treatment of influenza and symptoms of the common cold, as well as allergen‐induced conditions such as allergic rhinitis, in a mammalian subject (e.g., human) in need thereof.

Inventors:
CHURCH DENNIS JOSEPH (CH)
NICHOLS ANTHONY (CH)
Application Number:
PCT/IB2017/053367
Publication Date:
December 14, 2017
Filing Date:
June 07, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NOVARTIS CONSUMER HEALTH SA (CH)
International Classes:
A61K31/78; A61K9/00; A61P11/02; A61P27/14; A61P31/16
Foreign References:
CN101590074A2009-12-02
EP2803354A12014-11-19
US20050232868A12005-10-20
CN101590074A2009-12-02
Other References:
LUCZAK: "Antiviral and anineoplastic effect of Carbopol", 1 January 1979 (1979-01-01), pages 815 - 821, XP055396901, Retrieved from the Internet [retrieved on 20170808]
CLERCQ ET AL: "Antiviral activity of carbopol, a cross-linked polycarboxylate", 1 January 1976 (1976-01-01), pages 151 - 158, XP055396905, Retrieved from the Internet [retrieved on 20170808]
BLOEMERS H P J ET AL: "Inhibition of RNA-dependent DNA polymerase of oncorna viruses by Carbopol 934", FEBS LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 52, no. 1, 15 March 1975 (1975-03-15), pages 141 - 144, XP025602906, ISSN: 0014-5793, [retrieved on 19750315], DOI: 10.1016/0014-5793(75)80657-0
K.H. MAIR ET AL: "Carbopol improves the early cellular immune responses induced by the modified-life vaccine Ingelvac PRRS MLV", VETERINARY MICROBIOLOGY, vol. 176, no. 3-4, 1 April 2015 (2015-04-01), NL, pages 352 - 357, XP055308822, ISSN: 0378-1135, DOI: 10.1016/j.vetmic.2015.02.001
"Final report on the safety assessment of carbomers - 934, -910, -934P, -940, -941 and -962", INT. J. TOXICOLOGY, vol. 1, 1982, pages 109 - 141
BARRETT B.: "Medicinal properties of Echinacea: a critical review", PHYTOMEDICINE, vol. 10, 2003, pages 66 - 86, XP004957204, DOI: doi:10.1078/094471103321648692
BERTINO JS.: "Cost burden of viral respiratory infections: issues for formulary decision makers", AM. J. MED., vol. 112, 2002, pages 42S - 49S
"Carbopol. Water soluble resins.", SERVICE BULL. GC-36, 1962
BLOEMERS HPJ; VAN DER HORST A.: "Inhibition of RNA-dependent DNA polymerase of oncorna viruses by Carbapol 934", FEBS LET., vol. 52, no. 1, 1975, pages 141 - 144, XP025602906, DOI: doi:10.1016/0014-5793(75)80657-0
CHOULIS N.: "Timed-release tablets containing quinine sulfate", J. PHORM. SCI., vol. 64, no. 6, 1976, pages 1033 - 1035, XP009014686, DOI: doi:10.1002/jps.2600640634
"Final Report on the Safety Assessment of Carbomers-934, -910, -934P, -940, -941, and - 962", JOURNAL OF THE AMERICAN COLLEGE OF TOXICOLOGY, vol. 1, 1982, pages 109 - 141
DE CLERCQ; LUCZAK M.: "Antiviral activity of carpobol, a cross-linked polycarboxylate", ARCH. VIROL, vol. 52, no. 1, 1976, pages 151 - 158, XP055396905
ELGINDY NA.: "Molecular entrapment of cationic drugs by Carbopol 934", CAN. J. PHARM. SCI., vol. 11, no. 1, 1976, pages 32 - 4
FENDRICK AM; MONTO AS; NIGHTENGALE B; SARNES M.: "The economic burden of non-influenza-related viral respiratory tract infection in the United States", ARCH. INTERN. MED., vol. 163, 2003, pages 487 - 494
GERN JE; MOSSER AG; SWENSON CA; RENNIE PJ; ENGLAND RJ; SHAFFER J; MIZOGUCHI H.: "Inhibition of rhinovirus replication in vitro and in vivo by acid-buffered saline.", J. INF. DIS, vol. 195, 2007, pages 1137 - 1143
GIRANDA VL; HEINZ BA; OLIVIERA MA.: "Acid-induced structural changes in human rinovirus-14 and possible role in uncoating", PROC. NAT'I ACAD. SCI. USA, vol. 89, 1992, pages 10213 - 10217
HAYDEN FG; GWALTNEY JM.: "Intranasal interferon-alpha 2 treatment of experimental rhinoviral colds infection", J. INFECT. DIS, vol. 150, 1984, pages 174 - 180
HAYDEN FG; KAISER DL; ALBRECHT JK.: "Intranasal recombinant alpha-2b interferon treatment of naturally occurring common colds.", ANTIMICROB. AGENTS CHEMOTHER., vol. 32, 1988, pages 224 - 230
HAYDEN FG; ANDRIES K; JANSSEN PA.: "Safety and efficacy of intranasal pirodavir (R77975) in experimental rhinovirus infection.", ANTIMICROB. AGENTS CHEMOTHER, vol. 36, 1992, pages 727 - 732
HAYDEN FG; HIPSKIND GJ; WOERNER DH; EISEN GF; JANSSENS M; JANSSEN PA; ANDRIES K.: "Intranasal pirodavir (R77975) treatment of rhinovirus colds.", ANTIMICROB. AGENTS CHEMOTHER, vol. 39, 1995, pages 290 - 294, XP002474491
HAYDEN FG; HERRINGTON DT; COATS TL; KIM K; COOPER EC; VILLANO SA; LIU S; HUDSON S; PEVEAR DC; COLLETT M: "Efficacy and safety of oral pleconaril for treatment of colds due to picornaviruses in adults: results of two double-blind, randomized, placebo-controlled trials.", CLIN. INFECT. DIS., vol. 36, 2003, pages 1523 - 1532, XP002392816, DOI: doi:10.1086/375069
HAYDEN FG; TURNER RB; GWALTNEY JM; CHI-BURRIS K; GERSTEN M; HSYU P; PATICK AK; SMITH GJ; ZALMAN LS: "Phase II, randomized, double-blind, placebo-controlled studies of ruprintrivir nasal spray two-percent suspension for prevention and treatment of experimentally-induced rhinovirus colds in healthy volunteers.", ANTIMICROB. AGENTS CHEMOTHER, vol. 47, 2003, pages 3907 - 3916
HULL D; RENNIE P; NORONHA A; POORE C; HARRINGTON N; FEARNLEY V; PASSALI D.: "Effects of creating a non-specific, virus hostile environment in the nasopharanx on symptoms and duration of the common cold", ACTA OTORHIN. ITAL., vol. 27, 2007, pages 73 - 77
JACOBS SE; LAMSON DM; ST. GEORGE K; WALSH TJ., CLIN. MICROBIOL. REV., vol. 26, no. 1, 2013, pages 135 - 162
JULLAPHANT T: "Colloidal properties of montelukast sodium nasal spray. Paper presented at: 5th Thailand International Nanotechnology Conference (NanoThailand", NANOTECHNOLOGY ASSOCIATION OF THAILAND., 2016
KENNEDY JL; TURNER RB; BRACIALE, T; HEYMANN, P; BORISH, L: "Pathogenesis of rhinovirus infection", CURRENT OPINION IN VIROLOGY, vol. 2, 2012, pages 287 - 293, XP055311472, DOI: doi:10.1016/j.coviro.2012.03.008
KUMAR BV.: "Inhibition of reverse transcriptase and r-DNA polymerase by Carbapol 934", MICROBIOS. LETT., vol. 2, no. 7, 1976, pages 219 - 223
LAZE L; BARENE I; NESTEROVA Z ET AL.: "Formulation of product for topical delivery of heparin", PAPER PRESENTED AT: INTERNATIONAL STUDENT CONFERENCE OF THE RIGAS STRADINA UNIVERSITY: HEALTH AND SOCIAL SCIENCES., 2017
NICHOL KL; D'HEILLY S; EHLINGER E.: "Colds and influenza-like illnesses in university students: impact on health, academic and work performance, and health care use", CLIN. INFECT. DIS., vol. 40, 2005, pages 1263 - 1270
PALMENBERG AC; SPIRO D; KUZMICKAS R; WANG S; DJIKENG A; RATHE JA; FRASER-LIGGETT CM; LIGGETT SB.: "Sequencing and analyses of all known human rhinovirus genomes reveal structure and evolution.", SCIENCE, vol. 324, 2009, pages 55 - 59, XP055118758, DOI: doi:10.1126/science.1165557
PALMENBERG AC; RATHE JA; LIGGETT SB.: "Analysis of the complete genome sequences of human rhinovirus", J. ALLERGY CLIN. IMMUNOL, vol. 125, 2010, pages 1190 - 1201
PATICK AK; BROTHERS MA; MALDONADO F; BINFORD S; MALDONADO O; FUHRMAN S; PETERSEN A; SMITH GJ; ZALMAN LS.: "In vitro antiviral activity and single-dose pharmacokinetics in humans of a novel, orally bioavailable inhibitor of human rhinovirus 3C protease", ANTIMICROB. AGENTS CHEMOTHER, vol. 49, 2005, pages 2267 - 2275
PEVEAR DC; HAYDEN FG; DEMENCZUK TM; BARONE LR; MCKINLAY MA; COLLETT MS.: "Relationship of pleconaril susceptibility and clinical outcomes in treatment of common colds caused by rhinoviruses", ANTIMICROB. AGENTS CHEMOTHER, vol. 49, 2005, pages 4492 - 4499
RAJNIK M; TOLAN RW., RHINOVIRUS INFECTION, 2014, Retrieved from the Internet
RENNIE P; BOWTELL P; HULL D; CHARBONNEAU D; LAMBKIN-WILLIAMS R; OXFORD J.: "Low pH gel intranasal sprays inactivate influenza viruses in vitro and protect ferrets against influenza infection.", RESPIR RES., vol. 8, no. 1, 2007, pages 38, XP021027435, DOI: doi:10.1186/1465-9921-8-38
ROELEN CAM; KOOPMANS PC; NOTENBOMER A; GROOTHOFF JW.: "Job satisfaction and short sickness absence due to the common cold", WORK, vol. 39, 2011, pages 305 - 313
SIMASEK M; BLANDINO DA.: "Treatment of the common cold.", AMER. FAM. PHYSICIAN, vol. 75, no. 4, 2007, pages 515 - 20
WINE TM; ALPER CM.: "Cytokine responses in the common cold and otitis media.", CURR ALLERGY ASTHMA REP., vol. 12, no. 6, 2012, pages 574 - 581, XP035135439, DOI: doi:10.1007/s11882-012-0306-z
WINDHOLZ M: "The Merck Index, 9th ed.", 1976, MERCK & CO
ZUCKERMAN AJ: "Principles and Practice of Clinical Virology, 6th Edition,", 2007, WILEY, pages: 496
Download PDF:
Claims:
What is claimed is:

1. A method for treating or preventing viral infections or allergenic conditions or the symptoms thereof in a mammalian subject in need thereof which comprises topically administering to the subject an aqueous composition buffered to approximately neutral pH comprising a therapeutically effective amount of at least one pharmaceutically acceptable carbomer.

2. A method according to claim 1 for treating or preventing viral infections wherein the viral infection is caused by a virus selected from one or more of the group consisting of human rhinoviruses (HRVs), coronaviruses influenza viruses, adenoviruses, parainfluenza viruses, respiratory syncytial viruses (RSV), metapneumoviruses, enteroviruses other than HRV, coxsackievirus, Severe Acute Respiratory Syndrome (SARS), avian influenza virus, swine influenza virus, filoviridae, paramyxovirus, orthomyxovirus, and Middle East Respiratory Syndrome (MERS).

3. A method according to claim 2 for treating or preventing viral infections wherein the viral infection is caused by a virus selected from one or more of the group consisting of human rhinoviruses (HRVs), coronaviruses, influenza viruses, adenoviruses, parainfluenza viruses, respiratory syncytial viruses (RSV), metapneumoviruses, and enteroviruses other than HRV.

4. A method according to claim 3 for treating or preventing viral infections wherein the viral infection is caused by one or more of human rhinoviruses (HRV) and the subject is human.

5. A method according to any of the preceding claims 1, 2, 3 and 4 wherein the at least one pharmaceutically acceptable carbomer is selected from Carbomer Homopolymer, Copolymer and Interpolymer, and combinations thereof.

6. A method according to any of the preceding claims 1, 2, 3 and 4 wherein the at least one pharmaceutically acceptable carbomer is selected from Carbomer Homopolymer and Carbomer Copolymer, and combinations thereof.

7. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer is a Carbomer Homopolymer.

8. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer is selected from Carbomer Homopolymer of Types A, B and C, and combinations thereof.

9. A method according to claim 8 wherein the at least one pharmaceutically acceptable carbomer is a Carbomer Homopolymer of Type C.

10. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer comprises a Homopolymer of acrylic acid or Copolymer of acrylic acid with a Cio- 3oalkyl acrylate, in either case crosslinked with an allyl ether of pentaerythritol, an allyl ether of sucrose, or an allyl ether of propylene.

11. A method according to claim 10 wherein the at least one pharmaceutically acceptable carbomer comprises a Homopolymer of acrylic acid crosslinked with an allyl ether of pentaerythritol, an allyl ether of sucrose, or an allyl ether of propylene.

12. A method according to claim 11 wherein the at least one pharmaceutically acceptable carbomer comprises a Homopolymer of acrylic acid crosslinked with allyl pentaerythritol.

13. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer comprises a Homopolymer of acrylic acid crosslinked with allyl pentaerythritol and having a viscosity of 13,000 to 30,000 cP, with reference to a 0.2 wt.% mucilage (spindle 6), and a viscosity of 40,000 to 60,000 cP, with reference to a 0.5 wt.% mucilage (spindle 7), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield VT, 20 rpm.

14. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer is Carbopol® 980 NF.

15. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer comprises a Homopolymer of acrylic acid crosslinked with allyl pentaerythritol and having a viscosity of 1,000 to 6,000 cp, with reference to a 0.2 wt.% mucilage (spindle 4), and a viscosity of 4,000 to 10,000, with reference to a 0.5 wt.% mucilage (spindle 5), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield RVT, 20 rpm.

16. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer is a Carbomer Copolymer.

17. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer is selected from Carbomer Copolymer of Types A, B and C, and combinations thereof.

18. A method according to claim 17 wherein the at least one pharmaceutically acceptable carbomer is a Copolymer of acrylic acid and C 10-30 alkyl acrylate crosslinked with an allyl ether of pentaerythritol, an allyl ether of sucrose, or an allyl ether of propylene.

19. A method according to claim 18 wherein the at least one pharmaceutically acceptable carbomer is a Copolymer of acrylic acid and C 10-30 alkyl acrylate crosslinked with an allyl pentaerythritol.

20. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer comprises a Copolymer of acrylic acid and Cio-30 alkyl acrylate crosslinked with an allyl pentaerythritol and having an emulsion viscosity of 6,500 to 15,500 cP, with reference to a 0.2 wt. % emulsion (spindle #5), and a mucilage viscosity of 10,000 to 26,500 cP, with reference to a 1.0 wt.% mucilage (spindle #6), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield VT, 20 rpm.

21. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer comprises a Copolymer of acrylic acid and Cio so alkyl acrylate crosslinked with an allyl pentaerythritol and having an emulsion viscosity of 1,700 to 4,500 cP, with reference to a 0.2 wt.% emulsion (spindle #4), and a mucilage viscosity of 4,000 to 13,500 cP, with reference to a 1.0 wt.% mucilage (spindle #5), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield RVT, 20 rpm.

22. A method according to claim 6 wherein the at least one pharmaceutically acceptable carbomer is Pemulen® TR-1 or Pemulen® TR-2.

23. A method according to any of the preceding claims 1-3 and 5-22 wherein the subject is human.

24. A method according to claim 23 for treating or preventing viral infections of the symptoms of the upper respiratory tract.

25. A method according to claim 24 wherein the treatment is administered as a nasal spray.

26. A method according to claim 23 for treating or preventing viral infections of the eye.

27. A method according to claim 26 wherein the treatment is administered as an ocular drop.

28. A method according to any of the preceding claims 1-27 wherein the composition is free of other active pharmaceutical ingredients.

29. A topically administrable pharmaceutical composition for treating or preventing viral infections or allergenic conditions or the symptoms thereof in a mammalian subject in need thereof which comprises an aqueous composition buffered to approximately neutral pH comprising a therapeutically effective amount of at least one pharmaceutically acceptable carbomer.

30. A topically administrable pharmaceutical composition according to claim 29 wherein the at least one pharmaceutically acceptable carbomer is a Carbomer Homopolymer.

31. A topically administrable pharmaceutical composition according to claim 29 wherein the at least one pharmaceutically acceptable carbomer is selected from Carbomer

Homopolymer of Types A, B and C, and combinations thereof.

32. A topically administrable pharmaceutical composition according to claim 31 wherein the at least one pharmaceutically acceptable carbomer is a Carbomer Homopolymer of Type C.

33. A topically administrable pharmaceutical composition according to claim 29 wherein the at least one pharmaceutically acceptable carbomer is a Homopolymer or Copolymer of acrylic acid and Cio so alkyl acrylate in either case crosslinked with an allyl ether of pentaerythritol, an allyl ether of sucrose, or an allyl ether of propylene.

34. A topically administrable pharmaceutical composition according to claim 33 wherein the at least one pharmaceutically acceptable carbomer is a Homopolymer of acrylic acid crosslinked with an allyl ether of pentaerythritol, an allyl ether of sucrose, or an allyl ether of propylene.

35. A topically administrable pharmaceutical composition method according to claim 34 wherein the at least one pharmaceutically acceptable carbomer is a Homopolymer of acrylic acid crosslinked with allyl pentaerythritol.

36. A topically administrable pharmaceutical composition method according to claim 29 wherein the at least one pharmaceutically acceptable carbomer comprises a Homopolymer comprising acrylic acid crosslinked with allyl pentaerythritol and having a viscosity of 13,000 to 30,000 cP, with reference to a 0.2 wt.% mucilage (spindle 6), and a viscosity of 40,000 to 60,000 cP, with reference to a 0.5 wt.% mucilage (spindle 7), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield VT, 20 rpm.

37. A topically administrable pharmaceutical composition according to claim 29 wherein the at least one pharmaceutically acceptable carbomer is Carbopol® 980 NF.

38. A method according to claim 28 wherein the at least one pharmaceutically acceptable carbomer comprises a Homopolymer comprising acrylic acid crosslinked with allyl pentaerythritol and having a viscosity of 1,000 to 6,000 cp, with reference to a 0.2 wt.% mucilage (spindle 4), and a viscosity of 4,000 to 10,000, with reference to a 0.5 wt.% mucilage (spindle 5), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield RVT, 20 rpm.

39. A topically administrable pharmaceutical composition according to claim 29 wherein the at least one pharmaceutically acceptable carbomer is a Carbomer Copolymer.

40. A topically administrable pharmaceutical composition according to claim 29 wherein the at least one pharmaceutically acceptable carbomer is selected from Carbomer

Copolymer of Types A, B and C, and combinations thereof.

41. A topically administrable pharmaceutical composition according to claim 40 wherein the at least one pharmaceutically acceptable carbomer is a Copolymer of acrylic acid and Cio so alkyl acrylate crosslinked with an allyl ether of pentaerythritol, an allyl ether of sucrose, or an allyl ether of propylene.

42. A topically administrable pharmaceutical composition according to claim 41 wherein the at least one pharmaceutically acceptable carbomer is a Copolymer of acrylic acid and Cio so alkyl acrylate crosslinked with an allyl pentaerythritol.

43. A topically administrable pharmaceutical composition according to claim 29 wherein the at least one pharmaceutically acceptable carbomer comprises a Copolymer of acrylic acid and Cio so alkyl acrylate crosslinked with an allyl pentaerythritol and having an emulsion viscosity of 6,500 to 15,500 cP, with reference to a 0.2 wt. % emulsion (spindle #5), and a mucilage viscosity of 10,000 to 26,500 cP, with reference to a 1.0 wt.% mucilage (spindle #6), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield RVT, 20 rpm.

44. A topically administrable pharmaceutical composition according to claim 29 wherein the at least one pharmaceutically acceptable carbomer comprises a Copolymer of acrylic acid and Cio so alkyl acrylate crosslinked with an allyl pentaerythritol and having an emulsion viscosity of 1,700 to 4,500 cP, with reference to a 0.2 wt.% emulsion (spindle #4), and a mucilage viscosity of 4,000 to 13,500 cP, with reference to a 1.0 wt.% mucilage (spindle #5), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield RVT, 20 rpm.

45. A topically administrable pharmaceutical composition according to claim 29 wherein the at least one pharmaceutically acceptable carbomer is Pemulen® TR-1 or Pemulen® TR-2.

46. A topically administrable pharmaceutical composition according to any of the preceding claims 29-45 which is administered in the form of a nasal spray.

47. A topically administrable pharmaceutical composition according to any of preceding claims 29-45 which is administered in the form of an ocular drop.

48. A topically administrable pharmaceutical composition according to any of preceding claims 29-47 which is devoid of other active pharmaceutical ingredients.

49. A topically administrable pharmaceutical composition according to any of preceding claims 29-48 for use in the treatment or prevention of viral infections or allergenic conditions or the symptoms thereof in a mammalian subject in need thereof.

50. A topically administrable pharmaceutical composition according to any of preceding claims 29-48 for use in the treatment or prevention of viral infections of the upper respiratory tract or the symptoms thereof in a mammalian subject in need thereof.

51. A topically administrable pharmaceutical composition according to claim 49 for use in the form of a nasal spray.

52. A topically administrable pharmaceutical composition according to claim 49 for use in the form of an ocular drop.

53. A topically administrable pharmaceutical composition according to claim 29, wherein the composition:

(a) comprises less than 2% Carbomer 980, carbomer 981, pemulen T -1 or pemulen TR- 2, preferably between 0.1 and 1.5 % w/w of any of the above, more preferably between 0.2 and 1.2% w/w, and still more preferentially between 0.4 and 0.8% w/w; and/or

(b) comprises a quantity of NaCI between 0.5 and 1.5% w/w, more preferably between 0.7 and 1.3% w/w, and still more preferentially between 0.8 and 1.2% w/w; and/or

(c) is buffered at pH 7.2-7.5 with 10-50 mM phosphate, bicarbonate, glyoxaline or citrate/phosphate buffer; and/or

(d) optionally comprises 0.5% phenylethanol or parabens; and/or

(e) has a viscosity between 100-400 mPa.s at 20°C; and/or

(f) is administered with bottle, dropper, pipette, spray, or applicator into the nasal cavity; and/or (g) preferably is compliant with EN ISO 10993 (2009) for use as a topical medical device;

for use of the formulation in the prevention and/or treatment of upper respiratory infections and/or allergic rhinitis.

Description:
TOPICAL COMPOSITIONS COMPRISING CARBOMER FOR THE

TREATMENT AND PREVENTION OF VIRAL INFECTIONS AND ALLERGIC CONDITIONS Field of the Invention

The present invention concerns pharmaceutical compositions for topical, and especially intranasal, application that have rapid, persistent and broad-spectrum antiviral and anti-allergenic activity, and methods of using said compositions for the prevention and treatment of acute viral infections such as the common cold and allergen-induced ailments such as seasonal and/or allergic rhinitis, and the symptoms thereof.

Summary of the Invention

We have discovered that certain polymers of acrylic acid commonly known as carbomers, when prepared in buffered salt solutions suitable for topical administration, exert marked antiviral and/or anti-allergenic activity in mammalian cell culture in the substantial absence of cytotoxicity, and as such, can provide an important therapeutic benefit in the prevention or treatment of acute viral and/or allergenic infections or conditions and the symptoms thereof. In particular, we have found that pH-neutral compositions of carbomers which are cross-linked polyacrylates demonstrate antiviral or anti-allergenic activity in mammalian epithelial cells in vitro, indicating a therapeutic utility for the topical, and especially intranasal, treatment of viral infection and/or allergenic infections or conditions to obtain relief therefrom. Such compositions can reduce both the duration of infection and the severity of symptoms.

The antiviral/anti-allergenic effect of such compositions is found to be dose- dependent, and thus the compositions can provide effective clinical regimens for treatment and/or prevention of viral infections such as influenza, and/or allergen induced conditions, in humans, as well as in other mammals. Accordingly, the invention comprises methods for treating or preventing viral infections or allergenic conditions or the symptoms thereof in a mammalian subject in need thereof which comprise topically administering to the subject an aqueous composition buffered to approximately neutral pH comprising a therapeutically effective amount of at least one pharmaceutically acceptable carbomer; and compositions therefor.

Brief Description of the Figures

FIG. 1 (Panel A) Plot of of inhibition of H V-B14 mRNA (copy number, per well x lO 3 ) in human airway epithelial A459 cells in preventive mode where cells are pre-treated with increasing concentrations of Carbomer 980; Carbomer 980 + HPMC (where the carbomers are in a 1:1 weight ratio) (final concentration of Carbomer 980 only is plotted); and HPMC (expressed as Log[Polymer] in % weight (% w/w)). (Panel B) Bar graph showing cellular levels of HRV-B14 GADPH mRNA (copy number, per well x 10 ~3 ) following treatment of cells with 1% Poloxamer 407 (a non-ionic polymer), 1% pentosan polysulfate, or vehicle control (p<0.05 versus Control) .

FIG. 2 (Panel A) Plot of HRV-14 mRNA copy number (per well x 10 "3 ) in human airway epithelial A459 cells in preventive mode where cells were pre-treated with Carbomer 980 of increasing carbomer concentration (expressed as Log[Carbomer 980] in % w/w). (Panel B) Plot of cellular B14 mRNA copy number (as % untreated Control) following incubation with Carbomer 980 of (A) (expressed as Log [Carbomer 980] in % w/w).

FIG. 3 (Panels A and B) Lytic plaque formation at Day 3 following incubation of confluent monolayers of human cervical epithelial HeLa cells in curative mode where cells were pre- incubated with HRV-B14 and one of the following applied to the agar overlay: 20 μg/ml ribavirin; 0.0015% Carbomer 980; and 0.0015% Pemulen ® TR-2, compared to Control. The resulting number of lytic plaques are indicated in the lower left hand corner of each picture. FIG. 4 HRV-B14 and HRV-29-induced lytic plaque inhibition (expressed in % control) in human cervical epithelial HeLa cells at Day 3 following application of 0.0015 % Carbomer 980 or Carbomer 981 to agar overlays (*= p< 0.05 versus untreated infected control). Fig. 5 Viral load (expressed as viral copy number per ml x 10 "5 ) in human airway epithelia infected with Influenza H1N1 (Panel A) or Coronavirus OC643 (Panel B) after 24 hours following treatment with Carbomer 980 (with 0.5% phenylethanol preservative) compositions of increasing carbomer concentration (0.003, 0.01, 0.03, 0.1, 0.3 and 0.5 % w/w). In Panel A,* = p < 0.05 versus buffer control ("Ctrl"); in Panel B, for the 0.5 % w/w composition, p=0.092 versus buffer control. As additional controls, the antiviral, oseltamivir, 1 μΜ, is tested against H1N1 (Panel A); and chloroquine is tested against Coronavirus (Panel B).

Fig. 6 (Panels A and B) Lytic plaque formation in human cervical epithelial HeLa cells at Day 3 following application of a 10,000-fold (Panel A) or 100,000-fold (Panel B) dilution of HRV- B14 (MOI=0.4) pre-treated for one hour with one of the following : buffer ("Control"); 0.015% Carbomer 980; and 0.015% Pemulen TR-2. No carbomer is added to the agar overlays. Resulting number of lytic plaques are indicated in the lower left hand corner of each picture.

Fig. 7 (Panel A) Viscosity (in mPa's) of 1% Carbomer 980 or 981 as a function of pH between 5 and 7; or as a function of pH between 4 and 7 and salt content (Carbomer 980 composition comprises 150 mM NaCI; and Carbomer 981 compositions comprise 3 concentrations of NaCI: 1.7 mM, 150 mM, or 450 mM). (Panel B) Photograph of plexiglass model of the human nose showing deposition of composition of the invention throughout the nasal cavity, including in the area of the posterior turbinates and adenoid tissue (white arrows), following administration (via two sequential 150μΙ sprays) of 0.5% w/w Carbomer 980 additionally including a red dye (Panel B). Fig. 8 Plot of HRV-B14 viral mRNA copy number (per well x 10 "3 ) by carbomer concentration of Carbomer 980 composition (expressed as log [Carbomer 980] in % w/w), in human airway epithelial A459 cells treated with 0.01% or 01% Carbomer 980, either in the absence of preservative ("Carbomer 980"); or in the presence of phenoxyethanol ("Carbomer 980 + 0.1% phenoxyethanol"); phenylethanol ("Carbomer 980 + 0.5% phenylethanol"); or paraben ("Carbomer 980 + 0.5% paraben") (*= p < 0.05 versus viral mRNA copy number at the lower concentration).

Fig. 9 Plots of P. aeruginosa (Panel A) or S. aureus (Panel B) titer, expressed as Log

[Microorganism] in colony forming units/ml (CFU/ml), on indicated days (Days) following treatment with the 4 compositions as described with reference to Fig. 8.

Fig. 10 Plots of C. albicans (Panel A) or A. brasillensis (Panel B) titer, expressed as Log

[Microorganism] in colony forming units/ml (CFU/ml), on indicated days (Days) following inoculation of the 4 compositions as described with reference to Fig. 8. Fig. 11 Transepithelial resistance (Ohm, cm 2 ' (Panel A); LDH release (% Triton-induced cytotoxicity) (Panel B); cilial beating frequency (Hz) (Panel C); and mucus production (pg/ml) (Panel D), of human airway epithelia treated on the apical, air-facing side, for 24 hours, with Carbomer 980 composition (including 0.5% phenylethanol preservative) of increasing carbomer concentration (Log [Carbomer 980] in % w/w), against Triton and isoproterenol controls (data not shown).

Fig. 12 Plot of body weight (bw) (in g) (Panel A) or relative food consumption (g/kg/bw/day) in male and female rats over 14 day Test Period of intranasal administration of Carbomer 980 composition (including 0.5% phenylethanol preservative) as compared with untreated controls, followed by a Recovery Period of 7 days.

Fig. 13 Plot of HRV-14 mRNA copy number (per well x 10 "3 ) (Panel A) and cell viability (OD at 450 nm) (Panel B) against carbomer concentration (expressed as Log [Carbomer 980] in % w/w) in human airway epithelial A459 cells treated for one hour with Carbomer 980 compositions of increasing carbomer concentration, and subsequently infected with HRV- Fig. 14 Plot of histamine release (ng/ml) (Panel A) or cell viability (OD at 450 nm) (Panel B) against carbomer concentration (expressed as Log [Carbomer 980] in % w/w) in cultured human basophil KU812 cells incubated with 100 μΜ polymixin B or 100 μΜ of compound 48/80 in the presence of Carbomer 980 compositions with increasing carbomer concentration.

Fig. 15 Mouse L292 fibroblast cell viability graded by microscopic examination at the indicated times in agar-coated monolayers treated with 5% w/w Carbomer 980, as well as various marketed nasal sprays (Vicks First Defence ® , Anefrin Nasal Spray, and Nostrilla ® Fast Relief), and 0.9% saline, 0.1% Triton and 0.5% phenylethanol controls, and stained with neutral red. (* indicates cytotoxicity)

Fig. 16 Effect on weight (Panel A) and any histopathological observations (Panel B) of kidneys, larynx, liver, lungs with bronchi, nose (levels 1-5), pituitary glands and trachea, as determined after necropsy, in male and female rats administered 10 μΙ of 0.5% Carbomer 980 (including 0.5% phenylethanol preservative) or 0.9% saline control in each nares twice per day for a period of 14 days ("INFL" = inflammation; "LH" = lymphoid hyperplasia).

Background of the Invention

The common cold, also known as nasopharyngitis, rhinopharyngitis or coryza, is an acute viral respiratory tract infection affecting the nose, throat and sinuses, and also occasionally involving the eyes in the form of conjunctivitis. It is among the most common infectious diseases in man, being endemic to all human populations (Zuckerman, 2007). Adults are typically subject to 2-4 infections annually, with children having as many as 6-10 colds per year (Simasek and Blandino, 2007). While generally self-limiting in non-immuno- compromised patients, colds represent a major economic burden to society in terms of medical visits, work absenteeism and missed school in children (Nichol et al., 2005; Roelen et al., 2011; Bertino, 2002 and Fendrick et al., 2003), and further constitute a substantial health risk in the elderly, transplant recipients, and subjects presenting hematological malignancies, asthma, chronic obstructive pulmonary disease and cystic fibrosis (Jacobs et al, 2013). Cold viruses have also been linked to otitis media, bronchiolitis and pneumonia, most notably in children and in the elderly (Wine and Alper, 2012). The number of viruses that cause the common cold is very large, with 66-75% of cases being due to 200 antigenically distinct viruses from 8 different genera ( ajnik and Tolan, 2014). Among these, human rhinoviruses (HRVs) account for the majority (i.e. 25 up to about 80%, subject to annual fluctuations) of infections, followed by coronaviruses (15%), influenza viruses (e.g., influenza A and B), (10-15%), and adenoviruses (5%), as well as parainfluenza viruses, respiratory syncytial viruses (RSV), metapneumovirus, and enteroviruses other than HRV, with co-infections occurring as well.

HRVs are a group of picornaviridae enteroviruses with approximately one hundred known serotypes that are subdivided into three subgroups termed HRV-A, HRV-B and HRV- C, with the first two subgroups comprising 74 and 25 serotypes respectively (Palmenberg et al., 2009 and 2010). Among these, HRV-C is noted for causing severe infections in children.

HRVs are non-enveloped, positive sense, single-stranded RNA viruses with genomes of approximately 7.2-8.5 Kb in length that consist of a single gene that codes for 11 proteins which are transcribed as a single polypeptide that is subsequently cleaved into structural and non-structural proteins. Four of these proteins, termed VP1, VP2, VP3 and VP4, make up the capsid that encases the viral RNA, while the remaining non-structural proteins are involved in genome replication and assembly. Each virion contains 60 copies of each of the four capsid proteins, giving it an icosahedral structure, with each VP1 protein serving as the site of attachment of the virus to cell surface receptors.

Approximately 85% of known HRV serotypes, termed "major group HRVs", infect cells by binding to the human cell surface inter-cellular adhesion molecule 1 (ICAM-1), also known as CD45, while the remaining "minor group HRVs" infect cells via the cadherin- related family member 3 (CDHR3) protein. Historically, HRVs have also been reported to bind to the low-density lipoprotein receptor (LDL , 15%) or a cell surface sialoprotein to gain entry into cells.

The binding of major group HRVs to cell-surface ICAM-1 receptors triggers the transfer of viral RNA into the host cell cytoplasm, where the viral genome is translated to generate proteins that are essential for viral replication, which in turn leads to the production of new viral particles. The viral particles are released into the extracellular space, where they infect naive as well as previously-infected epithelial cells, which furthers their proliferation. Infections most typically initially occur in the adenoid area of the respiratory tract, with co-infections by multiple serotypes being common. Viral replication cycle times are estimated at 8-12 hours, with hundreds of new viruses being released for every infecting particle.

Evidence of the role of the immune response to symptomatology has been demonstrated for rhinoviruses. Once activated the humoral immune response continues irrespective of viral presence (Kennedy 2012), further contributing to the symptomology of the common cold.

HRVs are typically transmitted from person to person either via airborne droplets (aerosols), direct contact with infected nasal secretions, or through fomites (i.e.

contaminated objects). Of these, hand-to-hand and surface-to-hand contact appear to be the most important routes of transmission, with HRV infection being particularly well initiated by intranasal and conjunctival inoculation when compared to exposure via the oral route alone. Furthermore, HRVs are able to survive in indoor environments for hours to days at ambient temperature, and for approximately 2h on undisturbed skin, which leads to the high transmission and infection rates observed in child day care, preschool and family environments.

Despite decades of research in the field, there is as of yet no recognized cure for the common cold. For example, (a) oral pleconaril has been shown to reduce cold duration by 24h when compared to placebo (Hayden et al., 2003A and Pevear et al., 2005), but its development was discontinued due to safety concerns; b) intranasal pirodavir was abandoned due to a lack of effect on the severity of cold symptoms (Hayden et al., 1992; Hayden at al., 1995); c) rupintrivir was abandoned due to lack of efficacy in naturally- aquired respiratory infections (Hayden et al., 2003B; Patrick et al., 2005); d) IFN-a2 only showed a preventive effect while possibly prolonging the duration of cold symptoms by promoting more severe sore throat, nasal congestion and mucosal bleeding (Hayden and Gwaltney, 1984; Hayden et al., 1988; Jacobs et al, 2013); and e) human trials of Echinacea extracts in H V-induced colds have led to conflicting results (Barret, 2003). Allergic rhinitis, also termed seasonal rhinitis, perennial rhinitis, polinosis or hay fever, is inflammation of the nose that occurs when the immune system overreacts to airborne allergens. Signs and symptoms include sneezing, rhinorrhea and/or nasal congestion that can be accompanied by conjunctival injection and eyelid edema with swelling under the eyes. The fluid from the nose is usually clear, with sinus obstruction sometimes causing frontal headaches. Symptom onset is often within minutes following exposure to allergens, and can affect sleep, the ability to work, and the ability to concentrate.

Seasonal rhinitis is caused by tree pollens in the spring, grass and weed pollens in the summer, and by other weed pollens in the fall, with seasonal rhinitis also being occasionally caused by airborne fungal spores. Perennial rhinitis, on the other hand, is caused by year- round exposure to indoor environmental allergens, such as to dust mite or cockroach excretions, animal dander or mold. The underlying mechanism leading to rhinitis involves the binding of IgE antibodies to allergens which subsequently causes the release of histamine from mast cells. While the symptoms of allergic rhinitis resemble those of the common cold, they often last for more than two weeks, do not usually include a fever, and can most often be diagnosed by history and symptoms alone.

Treatment of seasonal and perennial rhinitis or other allergen-induced inflammatory conditions of the upper respiratory tract, is generally the same, with its goal being the prevention or reduction of symptoms caused by the inflammation of infected tissues. Measures that are effective include avoiding contact with allergen, with front-line therapies being oral antihistamines with oral decongestants, or nasal corticosteroids with or without oral antihistamines. Alternative treatments include nasal mast cell stabilizers such as cromolyn sodium or nedocromil, nasal antihistamines, nasal imatropium to relieve rhinnorhea, nasal saline, or an oral leukotriene receptor antagonist such as monteleukast. Medications, however, are generally insufficient and/or can be associated with side effects such as fatigue and drowsiness.

It has long been an object in the art to identify new pharmaceutical active ingredients for the treatment of acute viral infections and/or allergen-induced inflammatory conditions of the upper respiratory tract such as allergic rhinitis and the symptoms thereof.

Carbomers are widely considered to be non-toxic and well tolerated excipients, as they are unable to permeate the skin or mucosa, and have little-to-no oral bioavailability (CI , 1982). When applied topically, carbomers display little-to-no skin irritation, sensitization, phototoxicity or allergenic effects in mammals.

Carbomers, which are which are white, hygroscopic powders, that typically swell in water when neutralized, are widely used in both pharmaceutical and cosmetic formulations as thickening, suspending, dispersing and emulsifying agents (Windholz M, 1976; ACT, 1982), and are currently classified by the US Food and Drug Administration as inactive pharmaceutical ingredients approved for use in tablets, capsules, solutions, suspensions, emulsions, lotions, ointments, creams and gels at concentrations of up to 3.5% w/w depending on the application (FDA, 2016). Typical carbomer use in cosmetics is at concentrations of 0.1-1% w/w in lotion, eyeshadow, lipstick, hair conditioner, hair dye, dentifrice, bath soap, shaving cream and aftershave preparations, and at up to 5% w/w in blushes, face cream, hand cream, fragrance, after shave and suntan gel formulations. Their physical properties and relative inertness towards most biological systems further makes them suitable for the controlled the release of drugs from time-release tablets or entrapped systems (Choulis N, 1976 and Elgindy NA, 1976), or, e.g., as bulking agents (Goodrich, 1962). Marketed topical formulations containing carbomers include but are not limited to transdermal products within the Voltaren ® brand, such as diclofenac diethylamine 2.32% gel and diclofenac sodium 1% gel (Novartis); ophthalmic formulations such as Viscotears ® Liquid Gel (Bausch& Lomb); and nasal products such as Ocean Nasal Moisturizer Gel ® (Valeant Pharmaceuticals Int'l.), Extra Moisturizing Anefrin Nasal Spray (Walgreen's), and Nostrilla ® Nasal Spray (Insight Pharmaceutical). The physicochemical properties of montelukast sodium nasal spray formulation prepared as a colloid solution using hydroxypropyl cellulose and Carbomer 934 as a mucoadhesive agent were determined and found to meet the requirements for optimum nasal drug delivery (Jullaphant T, 2016).

Carbopol ® was demonstrated to be a suitable gelling agent in heparin gel; and in concentrations of 1.15 %, 1.20 %, and 1.25 %, used with preservatives, the gelling system showed acceptable microbiological and active ingredient stability (Laze L et al, 2017).

There has been only limited investigation of the use of carbomers as antivirals, and the results have been inconclusive. One study reports an in vitro inhibitory effect of

Carbomer 934 on murine leukemia and avian myoblastosis virus reverse transcriptases at concentrations of 5-30 mg/ml (Bloemers and Van der Horst, 1975; Kumar, 1976). Another study reports a protective effect exerted by Carbomer 934 against intravenous vaccinia and intranasal herpes simplex challenge in the mouse when administered by intraperitoneal injection (De Clercq and Luczak, 1976), which appears related to activation of peritoneal macrophages which in turn stimulates interferon production, rather than by direct viral inhibition.

A hallmark of H V is that the viruses are deactivated in acidic environments (Gern et al. 2007). The effect is believed to be due to conformational changes in the viral capsid proteins at pH below about 6.2 which render the virus noninfectious (Giranda et al. 1992). Thus Hull et al., 2007 attempted to treat HRV infections with 1% Carbomer 980 NF in a succinate buffered composition formulated at pH 4.5. See also Rennie P., 2007. However, to applicants' knowledge, no carbomer composition has previously been shown to be clinically effective in the treatment or prevention of acute viral infections such as the common cold. Additionally, CN 101590074 describes a neutralized gel composition of carbomer (e.g., Carbomer 934P), pH 6.0 to 7.0, that is applied intranasally and around the mouth 4-5 times day to prevent and treat allergic rhinitis.

5

Detailed Description of the Invention

We have discovered that certain carbomers are capable of inhibiting H V and influenza infection of human airway epithelial cells, and thus can be particularly useful for the treatment or prevention of virus-induced respiratory infections as well as allergenic) induced conditions such as allergic rhinitis and symptoms thereof in mammals by topical intranasal administration.

In particular, we have found that certain categories of carbomer, including USP/NF- monographed "Homopolymer" which includes species otherwise familiar to workers in the

15 art as "Carbomer 980" or "Carbomer 981" (available as Carbopols ® 980 NF and 981 NF), as well as USP/NF- monographed "Copolymer," which includes species such as Pemulens ® TR-1 and TR-2 NF are capable of inhibiting viral proliferation in human airway epithelial cells. In in vitro studies, the inhibition was found to be dose-dependent, with maximal inhibition occurring at concentrations consistent with the induction of a barrier effect. Additionally,

20 Carbomer 980 has been found to exhibit anti-plaque activity not only against HRV-B14 but also HRV-29, and thus potentially against a broad range of HRV serotypes. Critically, the anti-viral activity is seen when pre-treating HRV with carbomer, rather than when the cells are incubated with carbomer prior to viral inoculation, indicating that the carbomer acts directly on the HRV.

25

Without wishing to be bound thereby, we hypothesize that such anionic polymer compositions not only can provide a viscous physical barrier to the binding of pathogens to cell surface receptors such as epithelial cell ICAM-1 receptors , but may also exert a potent anti-viral, anti-allergenic effect through the binding of negatively-charged groups of the 30 polymer existing in the pH neutral compositions of the invention to positively-charged viral antigens, thereby inhibiting viral infection of the cell as well as the dissemination and further expansion of progeny virions.

Similarly, where the anionic polymer is used for the treatment or prevention of allergic rhinitis, it is hypothesized that the negatively-charged strands are able to bind to positively-charged domains of various allergens, thus preventing their ability to induce local histamine release when mucosal and/or epithelial surfaces are exposed to topical preparations designed for use in the prevention and/or treatment of allergic disorders.

Carbomers, also known as carboxyvinylpolymers, are synthetic, high molecular weight non-linear polymers of 2-propenoic acid (i.e. acrylic acid).

Physically, carbomers are flocculated powders of particles having a median diameter of about 2 to 7 microns. Each particle can be viewed as a network structure of polymer chains interconnected by crosslinks. In the absence of the crosslinks, the primary particle would be a collection of linear polymer chains, intertwined but not chemically bonded. Such linear polymers are soluble in a polar solvent, such as water.

The cross-linked carbomers, however, are not soluble due to their crosslinked nature and high molecular weight. In aqueous dispersions, these polymers swell up to 1000 times their original volume (and ten times their original diameter) to form a viscous gel (e.g., colloidal dispersion) when exposed to a pH environment above 4-6.

Since the pKa of the polymers is slightly acidic, i.e. 6 ± 0.5, the majority of the carboxylate groups on the polymer backbone are ionized at neutral pH and above, with the different variants of Homopolymers, for example, containing between 98.7% and 99.9% acrylic acid and between 56-68% free carboxylic acid groups (USP monograph).

The ionization results in repulsion between the negative particles, which adds to the swelling of the polymer. Whereas unneutralized suspensions or dispersions have relatively low viscosities, in order to achieve maximum viscosity, it is necessary to neutralize the carbomer, typically with a common base such as sodium hydroxide (NaOH) or

triethanolamine (TEA).

Although high viscosities can be achieved in pH ranges of 5.0-9.0, optimal viscosity for the purposes of the invention is achieved in a pH range of 6.5-7.5, which is referred to herein as "approximately neutral pH." Other pH ranges considered to be "approximately neutral pH" are: from about 6.5 to about 7.8 (e.g., from about 6.5 to about 7.3 or from about 7.3 to about 7.8, e.g., 7.5-7.8). The U.S. Pharmacopeia/National Formulary (USP/NF) umbrella monographs define three categories of carbomer: Homopolymer, Copolymer and Interpolymer.

Carbomer Homopolymer is a high molecular weight polymer of acrylic acid cross- linked with allyl ethers of polyalcohols. Carbomer Homopolymer, previously dried at 80° for one hour, contains not less than 56.0 percent and not more than 68.0 percent of carboxylic acid (-COOH) groups, per USP monograph. Homopolymer at 1% concentration in water has a pH of about 3. When neutralized with alkali hydroxides or amines, the polymer swells giving the appearance of dissolving in water; when neutralized with lower amines and alkanolamines, it swells giving the appearance of dissolving in methanol or glycerin; when neutralized with ethoxylated long-chain (C 14 -C is) amines, it swells giving the appearance of dissolving in ethanol. Typical cross-linkers are allyl ether of pentaerythritol, allyl ethers of sucrose, or allyl ethers of propylene.

Carbomer Copolymer is a high molecular weight copolymer of acrylic acid and a long chain alkyl methacrylate cross-linked with allyl ethers of polyalcohols. Carbomer copolymer swells in water when a suspension or dispersion of the copolymer is neutralized with sodium hydroxide to a pH within the range of 7.3 to 7.8 (USP monograph). The typical cross-linker is allyl ether of pentaerythritol. Carbomer Interpolymer is a Carbomer Homopolymer or Copolymer that contains a block copolymer of polyethylene glycol and a long-chain alkyl acid ester. Carbomer Interpolymer swells in water when a suspension or dispersion of it is neutralized with sodium hydroxide to a pH within the range of 5.5 to 9 (USP monograph).

The USP monograph further identifies Carbomer Homo-, Co- and Inter-polymers by three sub-category Types (A, B and C) which differ by viscosity range, as set forth in Table 1:

As will be understood by the skilled worker in the art, the above monographs were implemented to identify Carbomers manufactured without use of benzene, e.g., in ethyl acetate or ethyl acetate/cyclohexane co-solvent, see USP 29-NF 20, 24 and 25. The prior

USP compendial name (i.e. "Carbomer X" where X is a numeric), has now been reserved for the traditional products manufactured with use of benzene, examples of which consist of

Carbomers 941, 934P, 934, 940, and 1342, as shown on Table 2 (which are commercially available, e.g., Carbopols ® 941 NF, 934 P NF, 934 NF, 940 NF and 1342 NF, respectively).

However, usage of a numeric compendial name to identify benzene-free pharmaceutical grade products has persisted in tradenames as well as in informal usage by workers of skill in the art (e.g., "Carbomer 980" or "Carbomer 980 NF"). Thus the terms, "Carbomer 980" or

"Carbomer 980 NF", as used herein, if not otherwise specified, shall be understood to refer to pharmaceutically acceptable, essentially benzene-free, carbomer meeting the viscosity specifications set forth on Table 1 for Carbomer Homopolymer Type C; and similarly, "Carbomer 981" or "Carbomer 981 NF" shall refer to pharmaceutically acceptable essentially benzene-free Carbomer meeting the viscosity specifications set forth on Table 1 for Carbomer Homopolymer Type A.

A description and viscosity ranges for certain grades of carbomers that continue to be manufactured with the use of benzene are set forth in Table 2:

Additionally, for example, carbomer 910 is a white, fluffy hygroscopic powder. The pH of a 1 in 100 dispersion is about 3. When neutralized with alkali hydroxides or with amines, it dissolves in water, in alcohol, and in glycerin.

With respect to the monographed benzene-free carbomers, examples of commercially available Carbomer Homopolymer include Carbopols ® 71G NF, 971P NF, and 981 NF (Type A); 974 P NF and 5984 EP (Type B); and 980 NF (Type C), (all available from Lubrizol Advanced Materials, Inc., Cleveland, Ohio, hereinafter "Lubrizol"). (Of these, Carbopol ® 980 NF is understood to be a benzene-free alternative to Carbomer 940;

Carbopol ® 981 NF to USP 941; Carbopols ® 71G and 971 P NF to Carbomer 941; and Carbopol ® 5984 EP to Carbomer 934.) (See, e.g., "Compendial Specifications Applicable to Carbopol ® and Pemulen ® Pharmaceutical-Grade Polymers Based on Individual

Monographs," (Lubrizol product literature, Oct 2013)

Examples of commercially available Carbomer Copolymer are available, e.g., as Pemulens ® TR-1 NF and TR-2 NF (Lubrizol), which are Type B and Type A, respectively, copolymers of acrylic acid and C10-C30 alkyl acrylate crosslinked with allyl pentaerythritol that contain both hydrophobic and hydrophilic portions within the molecule.

Still other examples of carbomers are polymers of acrylic acid crosslinked with divinyl glycol (e.g., Noveon ® polycarbophil, Lubrizol).

Examples of commercially available Carbomer Interpolymer is available from Lubrizol under the Carbopol ® tradename (e.g., Carbopol® Ultrez® 10 NF; Carbopol® ETF 2020 NF).

As indicated above and shown on Table 3, Carbopol ® s 980 NF amd 981 NF and Pemulen ® TR-1 and TR-2 are polymerized without benzene, and thus are preferred for use in the methods and compositions of the invention:

The manufacturer's viscosity specifications for Carbopols ® 980/980 NF; 981

NF, and Pemulen ® TR-1 and TR-2 are provided in Tables 4 and 5, respectively: Table 4.

Minimum Maximum

Carbopol ® 980 and Carbopol ® 980 NF (2010)

Viscosity, cP, 25°C 13,000 30,000

Brookfield RVT, 20 rpm, neutralized to pH 7.3 - 7.8

0.2 wt% mucilage, spindle #6

Viscosity, cP, 25°C 40,000 60,000

Brookfield RVT, 20 rpm, neutralized to pH 7.3 - 7.8

0.5 wt% mucilage, spindle #7

Carbopol ® 981 (2008)

Brookfield RVF or RVT Viscosity, (mPa»s): 700 3,000

(20 rpm at 25°C, 0.05 % neutralized

Brookfield RVF or RVT Viscosity, mPa-s): 2,000 7,000

(20 rpm at 25°C, 0.2 % neutralized solutions)

Brookfield RVF or RVT Viscosity, mPa-s): 4,000 11,000

(20 rpm at 25°C, 0.5 % neutralized solutions)

Carbopol ® 981 NF (2010)

Viscosity, cP, 25°C 1,000 6,000

Brookfield RVT, 20 rpm, neutralized to pH 7.3 - 7.8

0.2 wt% mucilage, spindle #4

Viscosity, cP, 25°C 4,000 10,000

Brookfield RVT, 20 rpm, neutralized to pH 7.3 - 7.8

0.5 wt% mucilage, spindle #5

Test procedure 430-1

Lubrizol product literature (Issue Date: June 14, 2007; Edition: August 9, 2010)

The manufacturer's viscosity specifications for certain other grades of Carbopol carbomer used for topical applications are provided in Table 6 as follows: Table 6.

Carbopol ® Type Polymerization Neutralized % Viscosity (cP)

Solvent w/w aqueous 25°C dispersion Brookfield RVT, 20 rpm, neutralized to pH 7.3 - 7.8

ULTREZ ® F Interpolymer Type A ethyl acetate/ 0.5 wt% mucilage, 45,000-65,000 cyclohexane spindle #7

974P NF Homopolymer Type ethyl acetate 0.5 wt% mucilage, 29,400 -39,400

B spindle #6

934 NF carboxypolymethylen benzene 0.5 wt% mucilage, 2,050 - 5,450 e spindle #4

0.5 wt% mucilage, 30,500 - 39,400 spindle #6

934P NF carboxypolymethylen benzene 0.5 wt% mucilage, 2,050 - 5,450 e spindle #4

0.5 wt% mucilage, 29,400 - 39,400 spindle #6

1342 NF acrylates/ClO-30 benzene 1.0 wt% mucilage, 9,500-26,500 alkyl acrylate spindle #64 Crosspolymer

1.0 wt% mucilage 5,500-15,000 with 1.0 wt% salt,

spindle #5

5984EP Homopolymer Type ethyl acetate/ 1.0 wt% mucilage, 30,500 - 39,400 (same spec, as 934) B cyclohexane spindle #7

ETD2020NF Interpolymer Type B ethyl acetate/ 1.0 wt% mucilage, 47,000 - 77,000 cyclohexane spindle #7 (Test SA-003)

Source: product literature

Compositions of the invention comprise at least one pharmaceutically acceptable carbomer in a topically administrable vehicle.

The term, "pharmaceutically acceptable carbomer" as employed herein shall be understood to include both the carbomer in its acid form and also the partially or fully neutralized (e.g., alkali metal or ammonium salt) forms thereof.

More particularly, the invention is directed to aqueous compositions of approximately neutral pH comprising, or consisting essentially of, or consisting of: (a) a therapeutically effective amount of one or more pharmaceutically acceptable carbomers,

(b) a tonicity agent; and

(c) a buffer or buffer system.

The term, "therapeutically effective," as used in connection with this invention, refers to an amount effective for the prevention or treatment of viral or allergen-induced conditions or diseases, including the reduction or alleviation of symptoms associated with said conditions or diseases.

Viral infections treatable or preventable by the compositions of the invention comprise infections arising from one or more of human rhinoviruses (HRVs), coronaviruses influenza viruses (e.g., influenza A and B), adenoviruses, as well as parainfluenza viruses, respiratory syncytial viruses (RSV), metapneumovirus, enteroviruses other than HRV, coxsackievirus, Severe Acute Respiratory Syndrome (SARS), avian and/or swine influenza virus, filoviridae, paramyxovirus, orthomyxovirus, Middle East Respiratory Syndrome (M ERS), and co-infections thereof.

Preferred carbomers (a) for use in the methods and compositions of the invention are selected from: Homopolymer of Type A, B or C (allyl pentaerythritol crosslinked); Homopolymer of Type B (allyl sucrose crosslinked); and Copolymer of Types A or B (allyl pentaerythritol crosslinked), and combinations thereof.

Most preferably, the carbomer is selected from Homopolymer of Type C

(allylpentaerythritol crosslinked) having a viscosity of 13,000 to 30,000 cP, with reference to a 0.2 wt.% mucilage (spindle 6), and a viscosity of 40,000 to 60,000 cP, with reference to a 0.5 wt.% mucilage (spindle 7), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield RVT, 20 rpm. Alternatively, the carbomer is selected from Homopolymer of Type A (allyl pentaerythritol crosslinked) having a viscosity of 1,000 to 6,000 cp, with reference to a 0.2 wt.% mucilage (spindle 4), and a viscosity of 4,000 to 10,000, with reference to a 0.5 wt.% mucilage (spindle 5), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield VT, 20 rpm.

Alternatively, the carbomer is selected from Copolymer of Type B, allyl

pentaerythritol crosslinked, having an emulsion viscosity of 6,500 to 15,500 cP, with reference to a 0.2 wt. % emulsion (spindle #5), and a mucilage viscosity of 10,000 to 26,500 cP, with reference to a 1.0 wt.% mucilage (spindle #6), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield RVT, 20 rpm.

Still further, the carbomer may be selected from Copolymer of Type A, allyl pentaerythritol crosslinked, having an emulsion viscosity of 1,700 to 4,500 cP, with reference to a 0.2 wt.% emulsion (spindle #4), and a mucilage viscosity of 4,000 to 13,500 cP, with reference to a 1.0 wt.% mucilage (spindle #5), where viscosity is determined with reference to an aqueous dispersion of the carbomer neuturalized to pH 7.3-7.8 at 25°C, using a Brookfield RVT, 20 rpm.

As previously indicated, the carbomer (a), after being dispersed in water, is preferably neutralized. It is preferred that the carboxylic acid groups of the carbomer be maximally deprotenated since these groups are required for the polymer's binding to HRV. Common neutralizers, and the appropriate ratio to use (as compared to one part of carbomer) to achieve exact neutralization at a pH of 7.0, are set out in Table 7: Table 7.

Neutralizer Chemical name Neutralization

Ratio, Base/

Carbopol ®

Polymer

NaOH (18%) Sodium hydroxide 2.3/1.0

Ammonia (28%) Ammonium hydroxide 0.7/1.0

KOH Potassium hydroxide 2.7/1.0

TEA (99%) Triethanolamine 1.5/1.0

Tris Amino ® (40%) Tromethamine 3.3/1.0

Source: Lubrizol product information

The concentration of carbomer in the composition may vary widely depending on the application and the desired viscosity.

The concentration of pharmaceutically acceptable carbomer (a) in the compositions is generally about 0.01 to about 20 wt.%, more preferably about 0.05 to about 10 wt.%, even more preferably from about 0.1 to about 5 wt.%, even more preferably about 0.5 to about 3 wt.%, for example 0.5 wt.%.

For compositions to be administered as a nasal spray, the viscosity of the formulation would need to be in excess of about 100 mPa's at 25°C. (as measured with a Brookfield Viscometer, spindle 4, 25 °C.) in order to assure sufficient intranasal residency time for clinical efficacy, while not surpassing about 400 mPa.s in order for the preparation to be sprayable using a commonly-employed nasal spray device.

Preferably, the tonicity agent, component (b), should be selected to provide an osmolarity to the compositions of between about 200 and about 400 mOsm/L, preferably between about 220 and about 380 mOsm/L, and more preferably between about 250 and about 340 mOsm/L.

It is preferred that (b) is an ionic agent selected from one or more sources of monovalent cations, such as sodium chloride, potassium chloride, or a balanced salt solution. The ionic tonicity agents are typically present in an amount of about 0. 5 to about 0. 9%, preferably about 0. 6 to about 0. 9% by weight based on the total composition.

Where sodium chloride is the source of monovalent cation, an appropriate concentration in a nasal spray composition of the invention may be about 100 to about 200 milimolar (mM), e.g., about 130 to about 150 mM, depending on the buffer.

The tonicity agent can also be non-ionic. Non-ionic tonicity agents include diols, such as glycerol, mannitol, erythritol; and sugars such as sucrose and dextrose. Other non- ionic tonicity agents include glycerol, polyethylene glycol, and propylene glycol. Yet other non-ionic agents are mannitol, sucrose, and dextrose. The non-ionic agents may be present in an amount of about 1% to about 5% by weight based on the total composition.

Component (c), the buffer or buffering system, preferably comprises any

pharmaceutically-acceptable acid/anion pair that is used to maintain the formulation at a given pH, while maintaining the preferred viscosity, antiviral activity and/or allergen-binding capacity of the anionic polymer solution. Potentially suitable buffer systems include acetic acid/acetate, adipic acid/adipate, ascorbic acid/ascorbate, aspartic acid/aspartate, benzoic acid/benzoate, bicine/bicinate, boric acid/borate, camphoric acid/camphorate, carbonic acid/carbonate, citric acid/citrate, formic acid/formate, fumaric acid/fumarate, gluconic acid/gluconate, glutamic acid/glutamate, glutaric acid/glutarate, glycerophosphoric acid/glycero-phosphorate, glycolic acid/glycolate, glycine/glycinate, hippuric acid/hippurate, isobutyric acid/isobutyrate, lactic acid/lactate, maleic acid/maleate, malic acid/malate, malonic acid/malonate, N-Tris (hydroxymethyl)methyl-3-aminopropane sulfonic acid/sulfonate, oxalicacid/oxalate, oleic acid/oleate, 2-oxoglutaric acid/2-oxoglutarate, phosphoric acid/phosphate, proprionic acid/ proprionate, pyroglutamic acid/pyroglutamate, salicylic acid/salicylate, succinic acid/succinate, tartric acid/tartrate, and combinations(s) thereof.

Of the above, phosphoric acid/phosphate; citric acid/citrate; succinic acid/succinate; and ascorbic acid/ascorbate, are preferred, of which phosphoric acid/phosphate is most preferred. Sufficient buffer is used to maintain a pH that is preferably in the range of from about 6.5 to about 7.5. Other pH ranges considered to be "approximately neutral pH" are: from about 6.5 to about 7.8, e.g., from about 6.5 to about 7.3 and from about 7.3 to about 7.8.

A suitable concentration of buffer may be 1-100 mM, preferably 5-50 mM, more preferably 5-25 mM, and most preferably 10-20 mM.

In the preparation of nasal sprays, the preferred buffer is phosphate-buffered saline (PBS), which is an aqueous solution comprising sodium hydrogen phosphate, sodium chloride and, in some formulations, potassium chloride and/or potassium dihydrogen phosphate to make up physiological solutions. The osmolarity and ion concentrations of the solutions preferably match those of the human body (i.e. are isotonic).

As used herein, the term "isotonic PBS" shall refer to a 20 mM phosphate buffer solution ("PBS") (comprising 0. 89g/L Disodium phosphate and 2.26 g/L Monosodium phosphate) in 150 mM sodium chloride.

In some applications, salt or salt solution functioning as the tonicity agent (b) may be comprised of the anion of the acid employed in buffering system (c). To the extent a source of monovalent cations is provided to the composition as an ingredient of PBS or other buffer system (c), or otherwise provided to the composition (e.g., as an ingredient contained within an optional component), it shall be considered to fulfill the requirement for component (b) and shall be included in the determination of total monovalent cation concentration for purposes of calculating osmolarity of the composition.

An illustrative embodiment of the compositions of the invention comprises 0.5% w/w Carbomer 980 NF or Carbomer 981 NF in isotonic PBS, pH 7.2. It will be within the skill of a worker in the art to prepare compositions of appropriate viscosity and tonicity within the pH range of from about 6.5 to about 7.5, or from about 6.5 to about 7.8, e.g., from about 6.5 to about 7.3 or from about 7.3 to about 7.8, which are suitable for the intended therapeutic application.

For example, compositions of the invention to be administered to the nasal cavity as a nasal spray are preferably isotonic, non-irritating, and sprayable, yet sufficiently viscous to remain in contact with the surface of the posterior nasal and nasopharyngeal cavities for a prolonged period of time after administration.

Similiarly, compositions of the invention intended for ocular administration in the form of an eyedrop are preferably isotonic, non-irritating, and dispensable by dropper, yet sufficiently viscous to remain in contact with the ocular (e.g., corneal) surface. In the compositions of the invention, an optional additional ingredient comprises a viscosity-modulating agent. A viscosity-modulating agent as used herein, refers to a compound that when added to a solution increases (or decreases) the viscosity of the solution. For example, a viscosity-enhancing agent may be used in a nasal spray to provide sustained mucosal contact time, and reduce the possibility of the formulation to 'drip back' from the nasal cavity to the back of the throat ("post-nasal drip"). Viscosity-modulating agents typically comprise uncrosslinked polymers such as hydroxypropylmethylcellulose (i.e. hypromellose), methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl- cellulose, polyvinyl alcohol, and polyvinyl pyrrolidone. A preferred viscosity-enhancing agent is hydroxypropylmethylcellulose. The concentration of a viscosity-enhancing agent in the aqueous formulation is in general from about 0. 001 to about 5% (w/v), preferably from about 0. 05 to about 0. 5%, and more preferably from about 0.1 to about 0.3% (w/v).

Health regulations in various countries require that multi-dose ophthalmic and nasal preparations include a preservative. The pharmaceutical formulation of the present invention optionally comprises an anti-bacterial preservative, preferably a non-cationic preservative. Suitable preservatives for the present invention may be selected, e.g., from phenylethanol, phenoxyethanol, and parabens, methyl parabens, propyl parabens, and mixtures thereof. Typically, preservatives are employed at a level of about 0. 001 to about 1%, preferably, from about 0. 005 to about 0. 25%, and most preferably from about 0. 05 to about 0. 2% (w/v).

Other optional ingredients comprise taste-masking agents (e.g., sweeteners, flavoring agents, or other agents).

It is preferred that the compositions are essentially free of divalent or trivalent cations such as Ca 2+ , Zn 2+ Fe 2+ , Fe 3+ and La 3+ , which may cause the carbomer to precipitate due to the due to the high affinity of the carbomer anionic groups for these ions.

By comparison, the 1% Carbopof 980 formulation of Hull et al. 2007 comprised Zn 2+ in the presence of succinate buffer at pH 4.5, which is incompatible both with maintaining carbomer in solution and maintaining a maximal number of free deprotonated carboxylic acid groups that are required for the polymer's binding to H V.

Similarly, it is preferred that the compositions are free of certain cationic preservatives, such as benzalkonium chloride, bromododecinium bromide, benzethonium chloride, methylbenzethonium chloride, cetylkonium chloride, cetylpyridinium chloride, cetrimonium chloride, dofanium chloride, tetraethylammonium bromide, didecyldimethyl- ammonium chloride, domiphen bromide and the like, since the carbomers can react with amine groups to form thick emulsions of oils in water (Windholz M, 1976).

The pharmaceutical formulations of the present invention are preferably stable at room temperature for at least 12 months, preferably 24 months, and more preferably 36 months. Stable, as used herein, means that carbomer maintains at least 80%, preferably

85%, 90%, or 95% of its initial anti-viral or anti-allergenic activity. Compositions of 0.5% w/w Carbomer ® 980 formulated in isotonic PBS containing either of 0.1% phenoxyethanol, 0.5% phenylethanol, or a mixture of parabens as preservative, have been found to inhibit viral replication by 85%, 82% and 92% respectively.

Thus the compositions of the invention preferably comprise viscous aqueous solutions, dispersions (e.g., colloidal gels) or suspensions of carbomer in isotonic salt solution buffered to approximately neutral pH, for topical administration.

For purposes of administration by the nasal route, the compositions are applied directly to the nasal cavity by conventional means, for example by use of a dropper, pipette or spray, with said formulation being provided in either single or multidose form. In the case of administration via dropper or pipette, this may be achieved by the user

administering an appropriate, predetermined volume of the formulation, while in the case of a spray this may be achieved, for example, by means of a metered atomizing spray pump, both employed by the user as per the manufacturer's instructions. Said formulation may also comprise additional ingredients which have been selected to further: alleviate the symptoms or duration of the common cold, such as in the case of a nasal decongestant, an antihistamine, an anti-inflammatory agent, an antiviral, or a salt of Li + or Zn 2+ in a concentration sufficiently low to avoid precipitating the carbomer ; reduce the occurrence of secondary bacterial infections, such as in the case of an inducer of endogenous antimicrobial peptide production or an antiseptic for use in rhinosinusitis; promote the healing of the nasal mucosa, such as in the case of a cytoprotective moisturizer, and/or increase the retention of the formulation by the nasal mucosa, such as in the case of an additional mucoadhesive and/or thickening agent, and this without limitation and in any combination(s) thereof, with all resulting formulations being available for use in the prevention and/or treatment of upper respiratory infections and/or allergic rhinitis.

The compositions of the invention are topically administered for the prevention or treatment of viral infection or allergen-induced conditions such as allergic rhinitis to a mammalian (e.g., human or veterinary) subject in need thereof. By way of example, an adult human subject may be administered by the nasal route a total dose of from about 17 to about 34 mg of carbomer per day, more preferably about 5 to about 20 mg/day.

The composition may be administered once a day or in divided doses several times a day. For example, the administration protocol may consist of 2-3 metered dose sprays of a 0.5% carbomer composition of the invention per nostril, 3-4 times per day, which provides a maximum number of sprays per day of 24. Each spray may consist of about 0.10 to about 0.20 ml (e.g., 0.14 ml) of composition of the invention. A typical treatment period may consist of about 1 to 7 days, or shorter or longer as needed. A dosing regimen consisting of 3 actuations per nostril (i.e. x 2 nostrils), given up to 4 times per day (approximately 4 hours apart), over a period of 7 days will thus consist of a maximum of 168 nasal spray actuations. The rationale for the number of applications and regimen is to maximize the coverage of the nasal epithelium commensurate with consumer acceptability.

The combination of (a), (b) and (c) of the compositions of the invention, as previously defined, forms a barrier that reduces the ability of viral pathogens to infect human epithelial cells due to its ability to block the binding of viruses to cell surface receptors, thus providing a useful means for the prevention and treatment of a range of infections including but not limited to upper respiratory, labial, gingival, oropharyngeal, ocular, dermal and genital viral infections.

Furthermore, as the anionic polymers are formulated at approximately neutral pH and preferably in the absence of divalent cations and cationic preservatives, negatively- charged strands are able to bind to positively-charged domains of various allergens, thus preventing their ability to induce local histamine release when mucosal and/or epithelial surfaces are exposed to topical preparations designed for use in the prevention and/or treatment of allergic disorders. In one aspect, the compositions of the invention comprise carbomer as the sole active pharmaceutical ingredient, and are devoid of any active pharmaceutical ingredients other than the carbomer.

Carbomers are unable to permeate the skin or mucosa, and have limited to no oral bioavailability.

In a further aspect of the invention, it is preferred that none of the components of the compositions of the invention are systemically available, nor have any expected pharmacologic action other than the anti-viral and/or anti-allergenic activity of the carbomer. Alternatively, the compositions of the invention may comprise one or more active pharmaceutical ingredients in addition to the carbomer.

In another aspect, the compositions of the invention are essentially free of oil formation, and thus are not emulsions.

Other embodiments of the compositions of the invention include carbomer preparations for the prevention, treatment and/or use in, as applicable: (A) buccal disorders such as labial herpes, cold sores, gingivostomatisis, ulcerative lesions of the oropharyngeal structures and/or oral warts; (B) conjunctivitis, idiopathic anterior uveitis and/or ophthalmic herpes zoster infections; (C) dermal ailments including molluscum contagiosum, common, flat, mosaic, periungual or plantar warts, and/or herpes zoster-induced dermal lesions; (C) sexually-transmitted diseases such as genital herpes and human papilloma virus infections; (D) vaginal candidiasis; (E) reducing the hand-to-hand and surface-to-hand transmission of viruses, and (F) the disinfection of contaminated surfaces.

Still other embodiments are carbomer compositions for use in a topical labial or intrabuccal application for use in the prevention and/or treatment of herpes simplex labialis, coxsackie virus-induced gingivostomatitis or herpangina (i.e. cold sores, painful gums and mouth, and/or ulcerative lesions of the posterior oropharyngeal structures), and/or human papilloma virus-induced veruciform proliferations (i.e. oral warts). Anionic polymer (a) preferably consists of 4% or less (w/w) of a cross-linked 2- propenoic acid-derived carbomer listed in Table 1, while buffering agent (c) consists of any buffer that is commonly used in medicinal preparations (ex: phosphate, citrate, succinate, ascorbate, or other), and which also serves to assure a preferred viscosity and antiviral activity of the anionic polymer (a) solution.

In these embodiments, the resulting formulations are applied directly to the oral cavity by conventional means, for example by application of a liquid (i.e. mouthwash), gel, cream, film, eluting strip, dissolving tablet, losange or any similar means of delivery to the surface of the affected labial or buccal mucosa. In this instance, said formulations may also include, for example and without limitation: (d) a local anesthetic such as benzocaine, procaine, tetracaine, lidocaine, prilocaine or other; (e) an antiviral agent such as acyclovir, valacyclovir, penciclovir, famciclovir, docosanol or other; (f) an antiseptic, (g) an essential oil or fragrance such as menthol or eugenol; and (h) an additional mucoadhesive and/or thickening agent, and this without limitation and in any combination(s) thereof for use in the topical treatment of labial, intrabuccal or oral infections.

The compositions may also be used in the topical treatment of acute, recurrent and/or chronic ocular infections such as adenovirus, herpes simplex virus, enterovirus, Coxsackie virus, poxvirus and human immunodeficiency virus-induced ocular infections (i.e. "viral conjunctivitis" or "pinkeye"), viral anterior uveitis (i.e. "idiopathic anterior uveitis") and/or ophthalmic herpes zoster infections.

In variations of these embodiments, the compositions may be liquids or gels that are applied for example by means of a dropper, squeeze bottle or applicator tube. In this instance, said formulations may also include, for example and without limitation: an antiviral drug including but not limited to trifluridine, idoxuridine, vidarabine, ganciclovir or similar; an anti-inflammatory agent such as a glucocorticoid or a non-steroidal antiinflammatory drug; an antibiotic or similar antimicrobial agent for extending the use of formulation to the treatment of infections induced by Staphylococcus aureus, Haemophilius influenza, Streptococcus pneumoniae and/or Pseudomonas aeruginosa (i.e. "bacterial conjunctivitis"); a vasoactive constrictor of ocular arterioles, such as but not limited to the alpha adrenergic agonists tetrahydrozoline and naphazoline to reduces eye redness and congestion; an antihistamine; an antiseptic; an additional moisturizer and/or lubricant, and this without limitation and in any combination(s) thereof for use in the topical treatment of ocular infections.

In another embodiment, the compositions may be used in the prevention or treatment of dermal ailments such as pox virus-induced molluscum contagiosum, common, flat, genital, mosaic, periungual or plantar warts, and/or herpes zoster-induced dermal lesions.

In variations of these embodiments, the formulations of (a) anionic polymer, (b) tonicity agent, and (c) buffer or buffer system may be a solid, liquid, gel, ointment, cream or paste that is applied for example by means of an adhesive pad, patch, bottle, applicator tube or spray.

In this instance, said formulations may also include, for example and without limitation: an antiviral drug such as acyclovir, valacyclovir or other; an antimetabolite or inhibitor of DNA synthesis such as 5-fluorouracil or other; a keratolytic such as salicylic acid, allantoin, hydantoin, urea, glycolic acid or other; a retinoid such as tretinoin; an immunomodulator such as imiquimod, an interferon or other; a blistering agent such as cantharidin; an Hi or H 2 histamine receptor antagonist; an anesthetic such as benzocaine, procaine, tetracaine, lidocaine, prilocaine or other; an antiseptic; and an antibiotic to either kill infected tissue or to limit secondary bacterial infections, and this without limitation and in any combination(s) thereof for use in the topical treatment of dermal lesions and infections.

In still another embodiment compositions of the invention are used in the prevention of sexually-transmitted diseases, pregnancy and/or in the treatment of intravaginal infections.

In variations of these embodiments, the resulting formulations are applied directly to the vaginal cavity by conventional means, for example by application of a suppository, dissolving tablet, liquid, gel or cream with or without use of an applicator, either as such or combined with the use of a barrier device such as a diaphragm, condom, cervical cap or medicated sponge. In these instances, said formulations may also include, for example and without limitation: an antifungal agent; a spermicide such as nonoxynol-9, octoxynol-9, benzalkonium chloride, sodium cholate, lactic acid, neem oil or other; an antiseptic, a lubricant, a cytoprotective moisturizer, an additional mucoadhesive or thickening agent, and/or an essential oil or fragrance, and this in any combination(s) thereof.

Said compositions may also include an antiviral agent such as but not limited to compounds used for the prevention or treatment of genital herpes or human papilloma virus infections.

In another embodiment, the compositions are used in the prevention of hand-to- hand and surface-to-hand transmission of H V and other respiratory pathogens.

In these embodiments, the resulting formulations are applied directly to the skin of the hands by conventional means, for example by using a rigid or squeeze bottle, metered dispenser, presoaked wipes or a dispensing tube that are employed by the user as per the manufacturer's instructions. In this instance, said formulations may be gels, foams, creams or liquids that may also include, for example and without limitation: an organic solvent such as ethanol, n-propanol or isopropanol; an antiseptic; an oxidizing agent; a humectant such as glycerol, glycerin, propylene glycol or an organic acid; and an essential oil or fragrance, and this in any combination(s) thereof. Among cited alcohols, ethanol is favored both for its antiviral properties and for its propensity to evaporate prior to dermal permeation, resulting in the deposition of a thin film of antiviral anionic polymer on the surface of the skin.

A further embodiment comprises the compositions for use for the disinfection of contaminated surfaces, including but not limited to its use in home, school, restroom, hospital and industrial settings. Bicarbonate may be favored as the buffer for both its antiviral and antimicrobial properties.

In these embodiments, the resulting preparations may be liquids, gels, powders or tablets that are diluted in a volume of water prior to their application to said contaminated surfaces by means of a damp cloth, mop, hose, jet, sprayer or similar device by the user as per the manufacturer's instructions. In these instances, the formulation may also include, for example and without limitation: an organic solvent, including but not limited to ethanol, n-propanol, isopropanol and mixtures thereof; a wetting agent; an oxidizing agent including various acids; a phenolic disinfectant such as phenol, o-phenylphenol, chloroxylenol, thymol or other ; one or more quarternary ammonium compounds; an antiseptic; a chelate or salt of a metal that exhibits antimicrobial properties such as Ag + , Cu + , Cu 2+ , Li + or Zn 2+ (the divalent ions being in a concentration sufficiently low to avoid precipitating the carbomer) and/or an essential oil or fragrance, and this in any combination(s) thereof. In specific variations of these embodiments, the diluted preparation is applied to the surface in question, allowed to dry, and either left or subsequently removed from the surface by rinsing with water as per the manufacturer's instructions.

Thus the invention in its various aspects includes: A method for treating or preventing viral infections or allergenic conditions or the symptoms thereof in a mammalian subject in need thereof which comprises topically administering to the subject a composition comprising a therapeutically effective amount of at least one pharmaceutically acceptable carbomer in an aqueous solution or suspension or dispersion buffered to approximately neutral pH, wherein the composition also comprises a steroidal or non-steriodal anti-inflammatory agent; an antihistamine; a decongestant; an inducer of antimicrobial peptide production; an antibiotic; an antifungal; an antiseptic; and each of these either alone or in any combination thereof for use in the prevention and/or treatment of upper respiratory viral infections, allergic rhinitis or viral, bacterial and/or fungal sinusitis; and/or

the composition is a liquid, dispersion, suspension, emulsion, gel, cream, ointment, paste or film comprising an antiviral drug, an anesthetic, an antiseptic, a mucoadhesive or a humectant, and each of these either alone or in any combinations thereof for topical use in the prevention and/or treatment of infections of the oral cavity, lips and perilabial surface, including the prevention and/or the treatment of cold sores, gingivostomatitis, painful gums and mouth, herpangina, oral warts, herpes simplex labialis and/or any other ulcerative lesions of the labial, buccal or posterior oropharyngeal structures;

a steroidal or non-steroidal antiinflammatory agent, an antibiotic, a decongestant, an antihistamine or a humectant, and each of these either alone or in any combinations thereof for topical use in the prevention and/or treatment of acute, recurrent and/or chronic infections or allergic diseases of the eye, including viral conjunctivitis or pinkeye, allergic conjunctivitis, viral anterior uveitis, idiopathic anterior uveitis and ophthalmic herpes zoster infections; an antiviral drug, keratolytic, a dermal blistering agent, an antiseptic or a humectant, and each of these either alone or in any combinations thereof for topical use in the prevention and/or treatment of viral infections of the skin, including molluscum contagiosum, common, flat, mosaic, periungual or planar warts, and herpes zoster-induced dermal lesions; an antiviral drug, a spermicide, an antiseptic, a lubricant, a mucoadhesive or a humectant, and each of these either alone or in any combinations thereof for topical use in the prevention and/or treatment of sexually-transmitted diseases such as genital herpes and/or human papilloma virus infections;.

wherein the composition is a liquid, dispersion, suspension, emulsion or gel used for prevention of the hand-to-hand and surface-to-hand transmission of viral infections comprising an organic solvent, an antiseptic or humectant, and each of these either alone or in any combinations thereof.

wherein the composition is used for the disinfection of contaminated surfaces and comprises an organic solvent, an oxidizing agent, a phenoloic disinfectant, an antiseptic or humectant, and each of these either alone or in any combinations thereof.

The invention also contemplates a method of preparing a pharmaceutically- acceptable formulation with antiviral and anti-allergenic activity consisting of the combination of a salt or salt solution (a), an anionic polymer with antiviral and/or anti- allergenic action (b), and a buffering system (c), wherein:

(a) the salt or salt solution is, or is respectively made with, any pharmaceutically- acceptable salt of a monovalent cation, including but not limited to any halide, sulfate or carboxylate salt of Na + , K + , Li + , NhV, an organic amine, or an amino acid, and in any combination(s) thereof;

(b) the anionic polymer is any pharmaceutically-acceptable, non-linear, cross-linked, Type A, B or C Homo- or Copolymer derived from 2-propenoic acid (i.e. polyacrylic acid) produced in a ethyl acetate/cyclohexane co-solvent system;

(c) the buffering system consists of any pharmaceutically-acceptable acid/anion pair that is used to maintain the formulation at a given pH, including but not limited to acetic acid/acetate, adipic acid/adipate, ascorbic acid/ascorbate, aspartic acid/aspartate, benzoic acid/benzoate, bicine/bicinate, boric acid/borate, camphoric acid/camphorate, carbonic acid/carbonate, citric acid/citrate, formic acid/formate, fumaric acid/fumarate, gluconic acid/gluconate, glutamic acid/glutamate, glutaric acid/glutarate, glycerophosphoric acid/glycero-phosphorate, glycolic acid/glycolate, glycine/glycinate, hippuric acid/hippurate, isobutyric acid/isobutyrate, lactic acid/lactate, maleic acid/maleate, malic acid/malate, malonic acid/malonate, N-Tris (hydroxymethyl)methyl-3-aminopropanesulfonicacid/ sulfonate, oxalicacid/oxalate, oleic acid/oleate, 2-oxoglutaric acid/2-oxoglutarate, phosphoric acid/phosphate, proprionic acid/ proprionate, pyroglutamic acid/pyroglutamate, salicylic acid/salicylate, succinic acid/succinate, tartric acid/tartrate, and this in any combinations(s) thereof;

The invention further includes the above method, wherein the resulting formulation: (a) is a liquid, dispersion, suspension, emulsion, gel, cream, ointment, paste, film, wax, or solid;

(b) is administered by means of a bottle, dropper, pipette, spray, tube, sponge, tissue, wipe, eluting strip, suppository, granulated preparation, applicator, tablet, losange, patch, or by any other similar pharmaceutically-acceptable means;

( c ) is isotonic compared to tissues, mucosal surfaces and biological fluids, with an osmolarity between 200-400 mOsm/L at 20°C;

(d) has a pH between 6.0 and 8.0, and more preferably between 7.2 and 7.5.

does not contain significant amounts of divalent metal cations, cationic surfactants, cationic antiseptics or cationic preservatives.

The invention even further contemplates the use of the above formulation topically for the prevention and/or treatment the following ailments as applicable, including the prevention and/or treatment of their symptoms, severity, duration and/or sequelae:

(a) the common cold and related upper respiratory infections;

(b) seasonal and allergic rhinitis;

(c) viral, bacterial and/or fungal sinusitis;

(d) infections of the oral cavity, lips and perilabial surface, including the prevention and/or the treatment of cold sores, gingivostomatitis, painful gums and mouth, herpangina, oral warts, herpes simplex labialis and/or any other ulcerative lesions of the labial, buccal or posterior oropharyngeal structures;

(e) acute, recurrent and/or chronic infections or allergic diseases of the eye, including viral conjunctivitis or pinkeye, allergic conjunctivitis, viral anterior uveitis, idiopathic anterior uveitis and ophthalmic herpes zoster infections;

(f) viral infections of the skin, including molluscum contagiosum, common, flat, mosaic, periungual or planar warts, and herpes zoster-induced dermal lesions; (g) sexually transmitted diseases such as genital herpes and/or human papilloma virus infections.

The invention also includes the above methods wherein the resulting formulation contains one or more of:

(a) the local anesthetics articaine, benzocaine, bupivacaine, chloroprocaine, cyclomethycaine, dibucaine, dimethocaine, etidocaine, levobupivacaine, lidocaine, lignocaine, mepivacaine, piperocaine, prilocaine, propoxycaine, procaine, proparacaine, ropivacaine, tetracaine and/or trimecaine, as well as any antihistamines and anticholinergics with local anesthetic properties;

(b) the topical antibiotics bacitracin, ciprofloxacin, chloramphenicol, Chloromycetin, erythromycin, neomycin, mafenide, mupirocin, polymyxin B, retapamulin, silver sulfadiazine and/or sulfacetamide;

(c) the antifungal agents benzoic acid, abafungin, albaconazole, amorolifin,

amphotericin B, anidulafungin, bifonazole, butenafine, butoconazole, candicidin, caspofungin, cicloprox, clometrizole, crystal violet, ecoconazole, efinaconazole,

epoxiconazole, fenticonazole, filipin, flucocytosine, fluconazole, griseofulvin, haloprogin, hamycin, isoconazole, isovuconazole, itraconazole, ketoconazole, Miconazole, micafungin, miconazole, naftifine, natamycin, nystatin, omoconazole, oxiconazole, posaconazole, propriconazole, ravuconazole, rimocidin, sertaconazole, sulconazole, terbinafine, terconazole, tioconazole, tolnaftate, undecylenic acid, and/or voriconazole;

(d) the steroidal antiinflammatory agents alclometasone, amcinonide, betamethasone, clobetasol, desonide, desoximetasone, diflorasone, fluandrenolide, fluocinonide, fluocinolone, fluticasone, halobetasol, halcinonide, halometasone, hydrocortisone, mometasone and/or triamcinolone, and/or any of their various derivatives;

(e) the non-steroidal anti-inflammatory agents acetylsalicylic acid, aceclofenac, celecoxib, clonoxin, dexibuprofen, dexketoprofen, diclofenac, diflunisal, droxicam, etodolac, fenoprofen, flufenamic acid, flurbiprofen, harparagide, ibuprofen, indomethacin, ketoprofen, ketorolac, licofelone, lomoxicam, loxaprofen, meclofenamic acid, mefenamic acid, meloxicam, nabumetone, naproxen, nimesulide, oxaprozin, paracetamol, phenylbutazone, piroxicam, salicylic acid, salsalate, sulindac, tolfenamic acid, tenoxicam, tolmetin and vitamin D;

(f) the antihistamines acrivastine, astemisole, azatadine, azelastine, bilastine, brompheniramine, buclizine, bromodiphenhydramine, carbinoxamine, cetririzine, chlorodiphenhydramine, chlorpheniramine, chlorpromazine, cromolyn sodium clemastine, cyclizine, cyproheptadine, desloratidine, dexbrompheniramine, dexchlorpheniramine, dimenhydrinate, dimetindene, diphenhydramine, doxcepin, doxylamine, ebastine, embramine, fexofenadine, hydroxyzine, levocetirizine, loratidine, nedocromil, meclozine, mepyramine, mirtazapine, olpatidine, orphenadrine, phenindamine, pheniramine, phenyltoloxamine, promethazine, pyrilamine, quetiapine, rupatadine, tripelennamine, terfenadine, trimeprazine and/or triprolidine;

(g) the antiseptics benzalkonium chloride, cetyl trimethylammonium bromide, cetylpyridinium chloride, benzethonium chloride, benzododecinium bromide, chlorhexidine, octenidine, boric acid, brilliant green, hydrogen peroxide, acetic acid, peracetic acid, povidone-iodine, phenol, thymol, hexachlorophene, triclosan, polyhexamethylene biguanide, sodium hypochlorite, calcium hypochlorite and/or sodium bicarbonate;

(h) the antiviral drugs acyclovir, docosanol, famciclovir, idoxuridine, penciclovir, trifluridine, valcyclovir and/or vindarabine;

(i) the cytoprotective moisturizers, humectants and/or wetting agents butylene glycol, ceramide, cetyl alcohol, cyclomethicone, dexpanthenol, dimethicone, glyceryl triacetate, glycerin, glycerol, heparin sulfates, hexylene glycol, hyaluronic acids, maltitol,

maltodextrose, mineral oil, petrolatum, poloxamers, propylene glycol, sorbitol and/or xylitol;

(j) the decongestants cafaminol, cyclopentamine, epinephrine, ephedrine,

fenoxazoline, levomethamphetamine, levonordefrin, mephentermine, naphazoline, norepinephrine, oxymetazoline, phenylephrine, phenylpropanolamine, propylhexidine, pseudoephedrine, synephrine, tetrahydrozolidine, tramazoline, tuaminoheptane, tymazoline and xylometazoline, with preference being given to naphazoline, oxymetazoline, phenylephrine, pseudoephedrine and/or xylometazoline;

(k) the essential oils or fragrances including anethole, pinenes, camphor, 1,8-cineole, citral, eugenol, geranial, limonene, linalool, menthol, menthone, and/or thymol; (I) the inducers of endogenous antimicrobial peptide production including (l-3)(l-6) beta glucans, vitamin D, isoleucine, butyrate, litocholic acid, lactose, lactulose, quinine, niacinamide, resveratrol, pterostilbene, polydatin, sulforophane, maitake extract, shiitake extract, Brewer's yeast extract, natto and/or hyaluronan;

(m) the keratolytics salicylic acid, lactic acid, benzoic acid, urea, hydantoin, allantion, sulfur, resorcinol and/or podophyllum derivatives;

(n) the lubricants cellulose ether (multiple), glycerin and/or carrageenan;

(o) the mucoadhesives alginate, carboxymethylcellulose, hyaluronic acid (multiple), hypromellose and/or polycarbophil calcium;

(p) the non-cationic preservatives phenoxyethanol, phenylethanol and/or parabens (multiple);

(q) the spermicides nonoxynol-9, octoxynol-9, benzalkonium chloride, sodium cholate, lactic acid and/or neem oil;

(r) the dermal blistering agent cantharidin;

(s) the salts of the metal cations Li + , Cu + , Cu 2+ and/or Zn 2+ ;

and each of these either alone, or in any combination(s) thereof.

The invention is further illustrated through the following non-limiting examples. Materials and Methods

Carbomer compositions. The above-described, commercially available carbomers, Carbopol ® 980 NF and Carbopol ® 981 NF (alternatively referred to herein as"Carbomer 980 [or 981]" or "Carbomer 980 [or 981] NF"), as well as Pemulens ® T -1 and TR-2, were used. Unless otherwise specified, in Examples 1-13 and the Figures, the terms "Carbomer 980" or "Carbomer 980 composition" (and analogously for the other carbomers tested) shall be understood to refer to neutralized dispersions of the commercially sourced USP/NF carbomer in isotonic, phosphate buffered solution. Percentages recited in connection with the above-described carbomer compositions (e.g., "0.0015% Carbomer 980") refer to the weight percent (w/w %) of carbomer present in the composition. The "vehicle" or "Control" or "Ctrl" referred to in the Examples and Figures consisted of isotonic PBS without carbomer, unless otherwise indicated.

The carbomer compositions were prepared from carbomer stock suspensions in distilled water at pH 3, that were neutralized and added to 20 mM phosphate-buffered isotonic saline, pH 7.2, containing (unless otherwise indicated) 8.8 g/L NaCI [=150 mM] and 0.4 g/L KCI, and diluted in the same medium to provide 0.0015 to 1.5% (w/w) carbomer dispersions of pH 6.9 -7.5. (A) HRV-B14 Infectivity Studies in Human Airway Epithelial A459 Cells. Ten microliter aliquots of each carbomer composition were added to 90 μΙ supernatants of pre-washed confluent monolayers of human airway A549 epithelial cells cultured in 96 well plates with MEM medium containing 10% fetal bovine serum and 20 μg/ml gentamycin, and the cells were incubated at 37°C for lh prior to inoculation with HRV-B14 at a multiplicity of infection (MOI) of 0.1 for 6 h. After the second incubation, the cells were harvested, and total RNA was isolated from infected cells and reverse transcribed into cDNA. Quantitative PCR (qPCR) of an HRV-specific non-coding viral genome sequence was performed. Cellular glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used both as an indicator of cell viability and as a quantitative control for normalizing HRV-specific qPCR results, with GAPDH qPCR being performed in parallel to HRV qPCR using the cDNA obtained from the same cells. For purposes of cytoxicity assays, confluent monolayers of human airway epithelial A459 cells were incubated at 37°C for 6h or 7h with each concentration of carbomer in the absence of HRV-B14, and assays were conducted by adding water-soluble tetrazolium (WST- 1) to the media followed by the measurement of formazan at 450 nm as an additional control. In certain experiments, the protocol above was repeated using carbomer compositions containing 0.5% phenylethanol as preservative.

(B) HRV-B14 Infectivity Studies in Human Cervical Epithelial HeLa Cells. Aliquots of the carbomer compositions were supplemented with 10-fold concentrated MEM cell culture medium at a ratio of 9:1, and then diluted with normal MEM and adjusted to pH 7.4 prior to application to cells. Viral plaque reduction assays were conducted in human cervical epithelial HeLa cells by incubating confluent cell monolayers with H V-B14 at a multiplicity of infection (MOI) of 0.0004 for lh, after which carbomer dispersion was applied directly to the infected cell monolayers in a semi-solid agarose overlay. The number of lytic plaques was determined in quadruplicate determinations three days post infection using an ELISpot reader after fixation and staining of monolayers with cristal violet. Non-infected cells were used as baseline controls; virus infected cells were used to define 100% infection; and the antiviral agent ribavirin was used as a positive control in all experiments.

(C) Influenza A and Coronavirus OC430C43 Infectivity Studies in Human Airway Epithelia. Studies of the effect of six concentrations (0.003, 0.01, 0.03, 0.1, 0.3 and 0.5% w/w) of carbomer compositions additionally containing 0.5% phenylethanol were conducted in mucilair human airway epithelia preparations by first dispensing Influenza A H1N1 or coronavirus OC43 in the carbomer composition and then applying the mixture directly to the apical surface of the cells for 3h, after which the supernatants were removed and renewed with the same six concentrations of carbomer composition alone at 3.5 and 6h. The supernatants were removed at the end of the 24h incubation period and assayed for viral load by qPCR. Oseltamivir was used as a control in H1N1 viral load studies, and chloroquine in Coronavirus OC43 studies. Cytotoxicity studies were conducted by measuring transepithelial resistance in Ohm. cm 2 , LDH release as a percentage of Triton- induced cytotoxicity, cilial beating frequency in Hz, and epithelial mucus production in pg/ml.

(D) Histamine Release Assays. Histamine release assays were performed in KU812 cells (ATCC No. CRL-2256) propagated in RPMI1640 containing 10% FCS and 20 μΜ gentamycin. The cultured KU812 cells were gently washed in Tyrode buffer, and samples of 1 x 10 5 cells were incubated with increasing concentrations of carbomer composition for 15 min prior to a lh incubation with 100 μΜ polymixin B or compound 48/80. The cells were placed on ice, gently centrifuged at 190 g at 4°C for 8 min, and the supernatants collected and centrifuged at high speed to remove any remaining cell fragments. Histamine release was determined by incubating samples of the resulting supernatants with o-phthalaldehyde (OPT) at alkaline pH, which results in the formation of a fluorescent histamine-OPT condensation product (I ex = 360 nm; I em = 450 nm). Cytotoxicity of carbomer was assessed in the same cells using the cell permeable tetrazolium salt WST-1 followed by the measurement of formazan at 450 nm as a control. (E) Viscosity Measurements. Carbomer 980 composition was added to 50 mM succinate buffer containing various concentrations of sodium chloride followed by adjustment of the pH and solubilization of the material using a polytron. Viscosity of the buffered suspension was measured after lh equilibration at 20°C with a Brookfield viscosimeter at 50 rev./min by calculating mPa.s = (K/N)(Sx-So) using the appropriate K constant.

(F) Microbial Challenge Studies. Microbial challenge studies were performed as per European Pharmacopea 5.3.1 using Pseudomonas aeriginosa C.I. P. 82.118, Staphylococcus aureus C.I. P. 4.83, Candida albicans LP. 48.72 and Aspergillus brasiliensis LP. 1431.83, with inocula being prepared as per procedure ET694.

(G) Cytotoxicity studies, (i) Studies in human airway epithelia were conducted by incubating the apical sides of cultures with carbomer composition for 24h, and measuring transepithelial resistance, LDH release, ciliary beating frequency and mucus production versus appropriate detergent and isoproterenol controls. In mouse L292 fibroblasts the assays were performed in compliance with EN ISO 10993-5 (2009) by propagating mycoplasma-free cells in F-12K Nutrient mixture containing 10% newborn calf serum in a humidified 5% CO2 and 95% air atmosphere at 37°C, after which they were grown to confluency in 90mm tissue culture plates. The monolayers were coated with culture medium containing less than 2% agar, which was allowed to harden for 30 min after which neutral red was added to the plates (0.01% in PBS). Thirty minutes after coloration, control and test items were applied to the cells by means of circular filters of 5 mm in diameter that were deposited on the solidified agar surfaces, and the cultures were placed in a cell culture incubator for 6, 14 or 24h after which cell viability was assessed by microscopy. Test items were evaluated in quadruplicate and controls in duplicate determinations. (H) Rat nasal tolerability studies. The studies were conducted in groups of 20 rats (10 males and 10 females) by administering ΙΟμΙ of carbomer composition intranasally at 6h intervals twice per day for a period of 2 weeks followed by a 1 week recovery period to assess the reversibility of any effects observed at Day 14. Clinical observations were made daily after each dosing. Body weight, food and water consumption were recorded at time of allocation and/or weekly thereafter throughout the experimental period as applicable. At necropsy, all superficial tissues were examined visually and by palpation, and key organs and subcutaneous tissues were inspected visually, then the kidneys, larynx, liver, lungs, trachea, pituitary and samples of the nasal turbinates were collected for histopathological examination. All tissues were stained with hematoxylin and eosin for all animals, with samples of kidney and liver tissue being further processed for Oil Red-0 staining, and subsequently evaluated.

Example 1. Comparison of viral inhibition by anionic v. nonionic polymer.

Using the methods of (A) above, Carbomer 980 compositions, certain of which included 0.05% w/w hypromellose (HPMC), were tested for HRV-B14 viral inhibition alongside HPMC, poloxamer 407 and pentosan polysulfate.

As shown in Figure 1, both Carbomer 980 compositions alone and with HPMC were found to dose-dependently inhibit HRV-B14 replication in human airway epithelial A459 cell cultures, whereas HPMC alone is shown to be without effect (p <0.05 versus lowest concentration of same polymer) (Panel A).

By comparison, treatment of cells with 1% poloxamer 407 (a non-anionic polymer) failed to inhibit HRV-B14 replication while 1% pentosan polysulfate (an anionic polymer) has a modest inhibitory effect (Panel B), indicating that the observed antiviral activity was associated with the anionic polymers (Carbomer 980 and pentosan polysulfate) and not non-anionic polymers (HPMC and poloxamer 407) at the concentrations tested. Therefore, in one aspect, the compositions of the invention are preferably free of non-anionic polymers.

Example 2 Activity against HRV-B14, Using the methods of (A) above, human airway epithelial A459 cells were treated for lh with 0.5% w/w Carbomer 980, Carbomer 981, and Pemulen * TR-l and TR-2 compositions, followed by incubation with HRV-B14 for an additional 6h, resulting in decreases in viral mRNA copy number. The viral mRNA copy number for the two experiments ("Exps. 1 and 2") is shown as follows (% inhibition compared to vehicle controls):

Similarly, a study of inhibition of HRV-B14 infectivity in human airway epithelial A459 cells was carried out using increasing concentrations of Carbomer 980 composition with 0.5% phenylethanol, a preservative, at pH 7.2. As depicted in Figure 13, Carbomer 980 NF dose-dependently inhibits HRV-B14 viral infectivity in the presence of the non-cationic preservative (Panel A), exhibiting an IC50 of Carbomer 980 of 0.029% w/w in this assay, with inhibition of viral infectivity occurring at non-cytotoxic concentrations (Panel B), as determined using the formazan/mitochondrial function-based read-out.

Example 3 Investigation of carbomer cvtoxicitv.

Using the methods described in (A) above, cultured human airway epithelial A459 cells were pre-treated with Carbomer 980 composition for lh prior to exposure to HRV-B14 for 6h. As shown in Figure 2, Carbomer 980 NF dose-dependently reduced HRV viral copy number, with an IC50 of 0.051% w/w in this assay (Panel A), but without affecting cellular GAPDH mRNA levels (Panel B), indicating that the inhibitory effect occurs in a non-cytotoxic manner without inhibiting cellular DNA transcription. Since Carbomer 980 does not penetrate cell membranes, it follows that the pre-incubation of human airway epithelial cells with a Carbomer 980 preparation formulated at pH 7.4 reduces their susceptibility to infection by HRV. This result was supported by repeated cytoxicity testing in the WST- 1/formazan and neutral red membrane integrity assays. The above Examples 1-3 demonstrate that carbomer protects cells from infection when applied topically to human airway epithelial A459 cells prior to the exposure of the cell culture to HRV-B14.

Example 4 Investigation of carbomer HRV-14 viral inhibition post-infection.

As shown in Figure 3, incubation of confluent human epithelial cervical HeLa cell cultures with HRV-B14 according to Materials and Methods (B), lead to the formation of lytic plaques over a period of three days, with the number of plaques representing the multiplication of HRV-B14 in said cultures over time (Panels A, well 129 and Panel B, well 135 v. well 0).

Application of 20 μg/ml ribavirin to the cell monolayers lh after infection reduced the number of HRV-B14-induced lytic plaques to less than 90% of corresponding HRV-B14 controls (Panel A, well 18; Panel B, well 10). Following the methods described in (B) above, application of either 0.0015% w/w Carbomer 980 composition (Panel A, well 55) or analogous suspensions of Pemulen ® TR-2 (Panel B, well 73) one hour after infection reduced the number of lytic plaques by 47.5 + 6.25 and 50.1% + 5.52% respectively at p< 0.05 and n = 3.

Accordingly, the above experiments support the use of carbomers for the prevention or treatment of viral infections.

Example 5 Carbomer Inhibition of HRV-B14 and HRV-29-infected cell culture.

Following the methods described in (A) above, 0.0015% w/w Carbomer 980 and 981 compositions were tested in a viral plaque assay conducted in human cervical epithelial HeLa cells infected with HRV-29 as well as HRV-B14. The inhibitory effect of the carbomer compositions on HRV-B14 and HRV-29 viral plaques at 24 hours in the supernatants of Mucilair human airway epithelia preparations, relative to an untreated control, is shown in Figure 4. The reductions in plaque number were comparable in both cases, with effects being significantly different from the corresponding

100% viral controls at p < 0.05 and n = 3 (Panel A). Example 6 Inhibition of Influenza A HlNl and Coronavirus OC63 Virus in Human Airway Epithelia.

Using the methods described in (A) above, 0.003% to 0.5% Carbomer 980 compositions containing 0.5% phenylethanol as a preservative were found to dose dependently reduce HlNl viral load in infected human airway epithelia preparations within 24h of application as depicted in Figure 5. (Panel A: versus buffer control and oseltamivir at 1 μΜ) (* = p < 0.05 versus buffer control).

Using the same methods, the Carbomer 980 compositions were tested for inhibition of coronavirus OC43 viral load in infected human airway epithelia preparations (Panel B, versus buffer control and chloroquine).

In both cases, Carbomer 980 dose dependently inhibited HlNl and OC43 copy numbers in the supernatants, exhibiting an IC50 value of 0.006% w/w for the inhibition of HlNl replication in this assay. The effect on coronavirus replication, while relatively modest, is noteworthy as there are currently no known, medically-acceptable inhibitors of coronavirus replication.

Example 7 Pre-treatment of virus with carbomer prior to infection of human cervical epithelial HeLa cells.

Using the methods described in (B) above, H V-B14 was pretreated at an MOI of 0.4 with 0.05% w/w Carbomer 980 or 0.015% Pemulen ® TR-2 at pH 7.4 for lh followed by a 10'000-fold (Figure 6, Panel A) or 100'000-fold (Figure 6, Panel B) dilution of the samples prior to their application to confluent monolayers of human cervical epithelial HeLa cells.

As shown in Panels A and B, carbomer pretreatment (Panel A, wells 251 and 252; Panel B, wells 77 and 51) reduced the number of HRV-B14-induced lytic plaques at Day 3 compared to saline controls incubated with virus alone (Panel A, well 376; Panel B, well 113). This effect is attributed to the binding of the anionic polymers to the viral capsid of HRV, which limits its ability to bind to cell surface receptors to infect cells. Example 8 Viscosity studies of carbomer preparations.

Carbomer compositions were prepared with the objective of achieving compositions of suitable viscosity for intranasal administration. The tested hypo- and isotonic compositions comprised 0.5% w/w Carbomer 980 in 20 mM phosphate-buffered saline, pH 7.2, with 50 mM NaCI; or 0.5% w/w Carbomer 981 in 20 mM phosphate-buffered saline with 150, 450 or 1.7 mM mM NaCI. All of the compositions additionally comprised 0.1-0.5% of a non-cationic preservative.

As reflected in Figure 7, Panel A, the viscosity of an aqueous dispersion of 1% w/w Carbomer 981 was dependent on both the pH and the concentration of salt that was contained in the formulation, with increases in pH augmenting the formulation's viscosity while increases in salt concentration reduce it. The same was true for Carbomer 980, with dispersions of the latter being more viscous than the former at a same concentration of anionic polymer at neutral pH. The viscosity of carbomer preparations is also dependent on the presence of amines, cationic preservatives and divalent cations, as carbomers form viscous emulsions with amines and cationic preservatives, yet precipitate out in presence of high concentrations of divalent cations.

Panel A shows that hypo- and isotonic solutions at neutral pH have higher viscosity than solutions containing a high amount of salt at low pH.

Panel B depicts the compositions of the invention as being able to penetrate deep into the nasal cavity, up to and beyond the area of the nasopharyngeal tonsil (i.e. the adenoid) which is a primary site for H V infections.

Example 9 Effect on antiviral activity of non-cationic preservatives.

0.1% or 0.05% w/w Carbomer 980 compositions, of which three additionally comprise a non-cationic preservative, i.e. phenoxyethanol (0.1%), phenylethanol (0.5%) or paraben (0.5%), were tested for inhibition of H V-B14 infectivity. As shown in Figure 8, each of these formulations inhibited viral replication in human airway epithelial A459 cells, demonstrating that the presence of non-cationic preservatives is not deleterious to the polymer's antiviral action.

Example 10 Microbial Challenge with P. aeruginosa or S. aureus.

Following the methods described in (D) above, 40g samples of 0.5% w/w Carbomer

980 composition, to which has been added 0.1% phenoxyethanol, 0.5% phenylethanol, or 0.5% paraben, were inoculated with a standardized suspension of P. aeruginosa, S. aureus, C. albicans and A. brasiliensis, mixed and left to incubate for 7, 14 and 28 days, after which times the number of colony units forming per ml was determined for each microorganism. As shown in Figure 9, the formulations containing 0.5% phenylethanol or 0.5% parabens were found to be particularly effective against P. aeruginosa (Panel A), and S. aureus (Panel B), demonstrating that the addition of 0.5% phenylethanol or parabens was preferred over 0.1% phenoxyethanol or Carbomer 980 NF alone for purposes of preserving the formulation for medicinal use. As shown in Figure 10, all of the formulations, even carbomer alone, were effective against C. albicans, with the paraben-containing formulation being the most effective (Panel A); while the formulation containing 0.5% parabens was more effective over the others against A. brasiliensis (Panel B).

Example 11 Cytotoxicity.

Following the methods described in (G)(i) above, apical, air-facing and mucin- expressing sides of Mucilair human airway epithelial cell cultures were incubated for 24 hrs with carbomer compositions comprising 0.5% w/w Carbomer 980 containing 0.5% phenylethanol, while the cultures received cell culture medium from their basal sides. Figure 11 shows that no significant effects were seen on transepithelial resistance

(Panel A), LDH release (Panel B), ciliary beating frequency (Panel C) or epithelial mucus production (Panel D) compared to Triton controls. Following the methods described in (G)(ii) above, a cytotoxicity study in mouse L929 fibroblasts using the agar overlay method yielded similar results, with the carbomer composition inducing no significant cytotoxic effects for up to 14h of exposure, but moderate cytotoxicity after 24h exposure (see Figure 15). While significant, the cytotoxic effect was significantly less than those observed for three marketed nasal sprays, one of which displayed marked cytotoxicity after only 6h of exposure.

Example 12 Nasal tolerabilitv in the rat.

Following the methods described in (H) above, 10 μΙ of 0.5% w/w Carbomer 980 NF composition additionally containing 0.5% phenylethanol was administered to each of 20 rats (10 male/10 female),

As shown in Figure 12, the carbomer composition has no effect on either body weight (Panel A) or food consumption (Panel B), versus as untreated control, in the animals tested. Figure 16 supports the lack of adverse effects of 0.5% w/w Carbomer 980 on the weight of kidneys, liver, lungs with bronchi and pituitary glands in both male and female treated animals. No treatment induced effects were apparent in either male or female animals on Day 14. The results indicate that the formulation of 0.5% w/w Carbomer 980 is compliant with ISO 10993 (2009). Example 13 Carbomer anti-allergenic activity.

Figure 14 shows increasing concentrations of Carbomer 980 compositions

(additionally comprising 0.5% phenylethanol) dose-dependently inhibited both polymixin B and compound 48/80-induced histamine release in human basophil-derived KU812 cells at non-cytotoxic concentrations (Panel A), with the corresponding IC50S being observed in the 0.001% w/w range.

As shown in Panel A, increasing concentrations of carbomer dose-dependently inhibited both polymixin B and compound 48/80-induced histamine release in human basophil-derived KU812 cells at non-cytotoxic concentrations, with the corresponding IC50S being observed in the 0.001% w/w range.

Figure 14 also shows the lack of a cytotoxic effect of Carbomer 980 composition at the same concentrations in the same cells as determined using the formazan/mitochondrial function-based read-out (Panel B).

Example 14

An exemplary carbomer composition of the invention, pH 7.2, comprises th following:

To a solution of sodium chloride in purified water was slowly added, under constant stirring, disodium phosphate, monobasic potassium phosphate and phenylethanol.

Carbomer was then added and dispersed by using a homogenizer followed by the addition of sodium hydroxide or hydrochloric acid to adjust the pH to 7.2.

The finished product is a cloudy viscous liquid with a pH of 7.2, osmality of 250-300 mOsm/Kg and a viscosity of 150-300 mPa.s

The product is for use as a nasal spray to provide a viscous coating of carbomer into the anterior and posterior nasal cavities. The following tests may be performed as part of release and stability testing:

Appearance, identification, preservative content assay, pH, osmolarity, viscosity, spray content uniformity (by weight), droplet size distribution and microbial limit tests. Example 15 Clinical evaluation of Carbomer 980 NF gel delivered as a nasal spray relative to human nasal safety and tolerability (in healthy volunteers) and clinical efficacy and safety in partients with the common cold.

Study A: Safety and Tolerability. Study A was a randomized, parallel-group, open label, placebo-controlled, assessment of safety and tolerability of a 0.5 % wt/wt Carbomer 980 NF formulated as described in Example 14, for delivery as a topical nasal spray to healthy human volunteers. The composition was filled into 10-mL glass vials fitted with a pump capable of delivering a spray upon actuation. Matching placebo consisted of the composition without carbomer.

Thirty (30) healthy male and female adult subjects (50% of either gender), were randomized in a 2:1 ratio to either receive treatment (n=20) or placebo (n=10),

administered via a nasal spray. One dose of the nasal spray consisted of three actuations per nostril. Each actuation was 140μί (equivalent to 140 mg, and a calculated dose of 0.7 mg of carbomer).

Administration was four times a day dosing, approximately four hours apart (i.e. 8:00, 12:00, 16:00, and 20:00), for a period of seven days (Days 1-7).

During the seven days of treatment, each subject received in total 28 doses, which was equivalent to 168 actuations.

Two cohorts were treated, each containing 15 subjects. The second cohort started (Day -1) following completion of the first cohort. Subject randomization was stratified by gender. Assessment of local nasal tolerability was by nasal examination performed at screening visits, on Day 1 prior to first dosing (before 8:00 dose), on Days 1 to 7 following the 12:00 dose, and at 12:00 during the end of study visit (Day 8).

Safety was assessed by (1) reported adverse events (AE's); and (2) clinically significant changes in vital signs or laboratory parameters.

Local nasal tolerability was assessed through nasal examinations and defined by incidences of nasal mucosal assessments of > Grade IB, or moderate or severe mucosal bleeding or moderate or severe crusting of mucosa.

Conclusion: Multiple administrations of intranasally administered Carbomer 980 gel (0.5 wt.%) were well tolerated with regard to local nasal tolerability criteria with no mucosa grading higher than Grade IB (superficial nasal mucosal erosion), and very low incidences of mild mucosal bleeding or crusting. Intranasally administered Carbomer 980 gel (0.5 wt.%) was also safe and well tolerated in healthy adult subjects after multiple administrations, with no major side effects and no meaningful differences in safety profiles observed when compared to placebo.

Study B: Treatment of patients diagnosed with common cold. Study B is a multi- center, randomized, parallel group, double-blind, 2-arm, placebo-controlled, multiple-dose study in adults with early symptoms of a common cold (<72 hours duration) to assess efficacy, and safety of the investigational formulation used in Study A.

Eligibility criteria include the following symptomatic common cold symptoms:

1. A confirmed common cold diagnosis with symptoms < 72 hours;

2. Total symptom score (TSS) > 9 (baseline sum of the 8 common cold symptoms) where the symptoms of common cold and their severity grading are indicated on Table 10; 3. Score >1 for at least one of the following symptoms: sore throat, runny nose, or blocked nose.

Table 10.

Nasal symptoms: runny nose; blocked nose; sneezing; headache;

Other symptoms: Headache; muscle ache; chills; sore throat; cough

0 = absent symptoms (no sign/symptom evident)

1 = mild symptoms (sign/symptom clearly present, but minimal awareness; easily tolerated)

2 = moderate symptoms (definite awareness of sign/symptom that is bothersome but tolerable)

3 = severe symptoms (sign/symptom that is hard to tolerate; causes interference with activities of daily living and/or sleeping)

Approximately 170 eligible subjects (approximately equally divided between males and females) are randomized to treatment with carbomer nasal spray or placebo (vehicle only, nasal spray) in a 1:1 ratio at multiple sites. Subjects perform a baseline self-assessment of common cold symptoms in an e- diary. Subjects are instructed to dose for the full seven days irrespective of symptom resolution to avoid inappropriate discontinuation due to misinterpretation of reduced symptomatology as a result of the fluctuation of symptoms that occurs with a common cold. Subjects are also instructed to change to a new study treatment device every 2 days and to prime the new device prior to the morning dose.

The dosing regimen is one dose (consisting of 3 actuations per nostril, where each actuation is 140 μί, equivalent to 140 mg and a calculated dose of 0.7 mg carbomer), administered four times a day (at approximately 8:00, 12:00, 16:00, and 20:00 hours), for 7 days (except for Day 1 where subjects in the 2nd strata receive 3 doses). Subjects are instructed to blow their noses prior to dosing. Subjects are also instructed to alternate nostrils after each actuation. First dosing can occur any time prior to 13:00 on Day 1.

The study treatment is administered via a nasal spray device containing 12 mL, yielding 50 actuations after an initial one time priming. Each subject requires 168 actuations or 4 vials. Each subject is assigned a kit containing 4 vials for 7 days of dosing, with instructions to use 1 vial for 2 days (48 actuations), and then to start a new vial.

Subject randomization is stratified by dosing time on Day 1 by those study subjects receiving the first dose of study treatment between the hours of: 8:00-11:00 and those receiving the first dose between the hours 11:01-13:00. On Day 1, subjects dosing between 8:00-11:00 are to dose every 4 hours for 4 doses and subjects dosing between 11:01-13:00 are to dose every 4 hours for 3 doses. On Days 2-7 all subjects administer their initial morning dose immediately following recording of their common cold symptom assessment at 07:00 hours and the remaining 3 doses every 4 hours.

Dosing times and number of actuations are recorded in subject's e-diary. Subjects are instructed not to take any additional cough/cold medications, including but not limited to, prescription, OTC, non-drug/nutritional supplement, or procedures throughout the study. Subjects are instructed to use acetaminophen/paracetamol over other OTC medications, but to avoid use if possible. Subjects self-evaluate the severity of the following common cold signs/symptoms before each dose and record their assessment in the e-diary: headache, muscle ache, chills, sore throat, blocked nose, runny nose, cough, and sneezing. Subjects assess symptom severity on a 4-point scale (0=none, l=mild, 2=moderate, 3=severe) at baseline and 4 times a day (except for Day 1 where subjects in the 2nd strata are to receive 3 doses) through Day 7 immediately prior to each dose of nasal spray and record the scores in the e-diary. Safety is assessed by occurrence of adverse events.

Upon awakening on the morning of Day 8, subjects complete a self-assessment of common cold symptoms in the e-diary. Subjects are to return to the study sites on Day 8 for the End of study/Visit 2). Adverse events and the use of concomitant medications are recorded by the subjects in e-diaries and monitored by the site personnel throughout the study.

Efficacy is assessed by subject's evaluation of common cold signs/symptoms. While common cold symptoms manifest for 7-10 days, the most severe symptoms typically present witin the first 4 days of symptom onset. Thus the primary study objective is to assess the efficacy of the carbomer composition in reducing the severity of nasal symptoms on days 1-4, with the endpoint being average nasal symptom score (ANSS1-4), compared to placebo, in adult subjects with the common cold.

Secondary objectives are:

(1) To assess the efficacy of the carbomer composition in reducing the severity of nasal symptoms on days 1-7, using the average nasal symptom score (ANSS), compared to placebo in adult subjects with the common cold

(2) To assess the efficacy of carbomer composition in reducing the severity of

symptoms on days 1-4 and days 1-7, using the total symptom score (ATSS), compared to placebo in adult subjects with the common cold.

(3) To assess the efficacy of carbomer composition in reducing the severity of

individual symptoms, using the individual symptom score, compared to placebo in adult subjects with the common cold, where the endpoint is individual and composite total symptom scores (NSS and TSS), Days 1-7.

Safety is assessed by reported adverse events (including spontaneous and solicited adverse events and serious events).

References

American College of Toxicology (ACT). Final report on the safety assessment of carbomers - 934, -910, -934P, -940, -941 and -962. Int. J. Toxicology 1: 109-141, doi: 10.3109/10915818 209013151 (1982).

Barrett B. Medicinal properties of Echinacea: a critical review. Phytomedicine 10: 66-86 (2003).

Bertino JS. Cost burden of viral respiratory infections: issues for formulary decision makers. Am. J. Med. 112: 42S- 49S (2002).

B.F. Goodrich Chemical Co. (Goodrich). Carbopol. Water soluble resins. Service Bull. GC-36 (1962).

Bloemers HPJ and Van der Horst A. Inhibition of RNA-dependent DNA polymerase of oncorna viruses by Carbapol 934. FEBS Let. 52(1): 141-144 (1975).

Choulis N. Timed-release tablets containing quinine sulfate. J. Pharm. Sci. 64(6): 1033-1035 (1976).

[CIR] Final Report on the Safety Assessment of Carbomers-934, -910, -934P, -940, -941, and - 962. Journal of the American College of Toxicology, 1982; 1: 109-141.

De Clercq and Luczak M. Antiviral activity of carpobol, a cross-linked polycarboxylate. Arch. Virol. 52(1):151-158 (1976).

Elgindy NA. Molecular entrapment of cationic drugs by Carbopol 934, CAN. J. PHARM. SCI. 11(1), 32-4 (1976).

FDA. Inactive ingredients database http://www.fda.gov/Drugs/lnformationOnDrugs (2016).

Fendrick AM, Monto AS, Nightengale B and Sarnes M. The economic burden of non- influenza-related viral respiratory tract infection in the United States. Arch. Intern. Med. 163: 487-494 (2003).

Gern JE, Mosser AG, Swenson CA, Rennie PJ, England RJ, Shaffer J and Mizoguchi H.

Inhibition of rhinovirus replication in vitro and in vivo by acid-buffered saline. J. Inf. Dis. 195: 1137-1143 (2007).

Giranda VL, Heinz BA, and Oliviera MA. Acid-induced structural changes in human rinovirus- 14 and possible role in uncoating. Proc. Nat'l Acad. Sci. USA 89: 10213-10217 (1992).

Hayden FG and Gwaltney JM. Intranasal interferon-alpha 2 treatment of experimental rhinoviral colds infection. 7. Infect. Dis. 150: 174 -180. (1984). Hayden FG, Kaiser DL and Albrecht JK. Intranasal recombinant alpha-2b interferon treatment of naturally occurring common colds. Antimicrob. Agents Chemother. 32: 224-230 (1988).

Hayden FG, Andries K and Janssen PA. Safety and efficacy of intranasal pirodavir ( 77975) in experimental rhinovirus infection. Antimicrob. Agents Chemother. 36: 727-732 (1992).

Hayden FG, Hipskind GJ, Woerner DH, Eisen GF, Janssens M, Janssen PA and Andries K. Intranasal pirodavir (R77975) treatment of rhinovirus colds. Antimicrob. Agents Chemother. 39: 290-294 (1995).

Hayden FG, Herrington DT, Coats TL, Kim K, Cooper EC, Villano SA, Liu S, Hudson S, Pevear DC, Collett M and McKinlay M. Efficacy and safety of oral pleconaril for treatment of colds due to picornaviruses in adults: results of two double-blind, randomized, placebo-controlled trials. Clin. Infect. Dis. 36: 1523-1532 (2003A).

Hayden FG, Turner RB, Gwaltney JM, Chi-Burris K, Gersten M, Hsyu P, Patick AK, Smith GJ, and Zaiman LS. Phase II, randomized, double-blind, placebo-controlled studies of ruprintrivir nasal spray two-percent suspension for prevention and treatment of experimentally- induced rhinovirus colds in healthy volunteers. Antimicrob. Agents Chemother. 47:3907- 3916 (2003B).

Hull D, Rennie P, Noronha A, Poore C, Harrington N, Fearnley V and Passali D. Effects of creating a non-specific, virus hostile environment in the nasopharanx on symptoms and duration of the common cold. Acta Otorhin. Ital. 27: 73-77 (2007).

Jacobs SE, Lamson DM, St. George K and Walsh TJ. Clin. Microbiol. Rev. 26(1): 135-162 (2013).

Jullaphant T, Colloidal properties of montelukast sodium nasal spray. Paper presented at: 5th Thailand International Nanotechnology Conference (NanoThailand) - Nanotechnology Association of Thailand. 2016; Nakhon Ratchasima, Thailand.

Kennedy JL, Turner RB, Braciale, T, Heymann, P and Borish, L, "Pathogenesis of rhinovirus infection," Current Opinion in Virology 2:287-293 (2012). Kumar BV. Inhibition of reverse transcriptase and r-DNA polymerase by Carbapol 934.

Microbios. Lett. 2(7): 219-223 (1976).

Laze L, Barene I, Nesterova Z et al., Formulation of product for topical delivery of heparin. Paper presented at: International Student Conference of the Rigas Stradina University: Health and Social Sciences. 2017; Riga, Latvia.

Nichol KL, D'Heilly S and Ehlinger E. Colds and influenza-like illnesses in university students: impact on health, academic and work performance, and health care use. Clin. Infect. Dis. 40: 1263-1270 (2005). Palmenberg AC, Spiro D, Kuzmickas R, Wang S, Djikeng A, Rathe JA, Fraser-Liggett CM and Liggett SB. Sequencing and analyses of all known human rhinovirus genomes reveal structure and evolution. Science 324: 55- 59 (2009).

Palmenberg AC, Rathe JA and Liggett SB. Analysis of the complete genome sequences of human rhinovirus. J. Allergy Clin. Immunol. 125: 1190-1201 (2010).

Patick AK, Brothers MA, Maldonado F, Binford S, Maldonado O, Fuhrman S, Petersen A, Smith GJ and Zalman LS. In vitro antiviral activity and single-dose pharmacokinetics in humans of a novel, orally bioavailable inhibitor of human rhinovirus 3C protease.

Antimicrob. Agents Chemother. 49: 2267-2275 (2005).

Pevear DC, Hayden FG, Demenczuk TM, Barone LR, Mckinlay MA and Collett MS.

Relationship of pleconaril susceptibility and clinical outcomes in treatment of common colds caused by rhinoviruses. Antimicrob. Agents Chemother. 49: 4492-4499 (2005).

Rajnik M and Tolan RW. Rhinovirus infection. See Medscape at

http://emedicine.medscape.com/ article/ 227820-overview#a0101 (2014).

Rennie P, Bowtell P, Hull D, Charbonneau D, Lambkin-Williams R and Oxford J. Low pH gel intranasal sprays inactivate influenza viruses in vitro and protect ferrets against influenza infection. Respir Res. 8(1): 38 (2007).

Roelen CAM, Koopmans PC, Notenbomer A and Groothoff JW. Job satisfaction and short sickness absence due to the common cold. Work 39: 305-313 (2011).

Simasek M and Blandino DA. Treatment of the common cold. Amer. Fam. Physician 75 (4): 515-20 (2007).

Wine TM and Alper CM. Cytokine responses in the common cold and otitis media. Curr Allergy Asthma Rep. 12(6): 574-581. (2012).

Windholz M (ed). The Merck Index, 9 th ed. Rahway NJ, Merck & Co (1976).

Zuckerman AJ (ed). In Principles and Practice of Clinical Virology, 6 th Edition, p. 496. Wiley, Hoboken, NJ, USA (2007).




 
Previous Patent: SKIN TREATMENT DEVICE

Next Patent: CHEMCICAL COMPOUNDS