Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
2-S RIMANTADINE AND 2-R RIMANTADINE FOR TREATING CANCER AND PRECANCEROUS PAPILLOMA VIRUS LESIONS
Document Type and Number:
WIPO Patent Application WO/2022/177908
Kind Code:
A1
Abstract:
The present invention relates to the use of purified 2-S rimantadine or purified 2-R rimantadine or racemic rimantadine, or a pharmaceutically acceptable salt thereof, to treat cancers and precancer lesions, including cancers and precancer lesions associated with papillomavirus (HPV) in subjects in need of treatment. The present invention also relates to compositions comprising purified 2-S rimantadine or purified 2-R rimantadine or racemic rimantadine, or a pharmaceutically acceptable salt thereof, in combination with one or more immune checkpoint inhibitors.

Inventors:
LUMPKIN RICHARD (US)
Application Number:
PCT/US2022/016471
Publication Date:
August 25, 2022
Filing Date:
February 15, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TORAGEN INC (US)
International Classes:
A61K31/13; A61K39/395; A61P31/16; A61P31/20; A61P35/00; C07K16/28; C12Q1/70
Domestic Patent References:
WO2020118096A12020-06-11
Foreign References:
US20130072553A12013-03-21
Other References:
WETHER1LL, L.F. ET AL.: "Alkyl-imino sugars inhibit the pro-oncogenic ion channel function of human papillomavirus (HPV) E5", ANTIVIRAL RESEARCH, vol. 158, 2018, pages 113 - 121, XP085477631, DOI: 10.1016/j.antiviral.2018.08.005
KASEMSUK THITIMA, KAEOPU RUENRUTHAI, YUBOLPHAN RUEDEEMARS, PHUAGKHAOPONG SUTTINEE, VIVITHANAPORN PORNPUN: "Apoptotic and antiproliferative effects of amantadine and rimantadine in glioblastoma cells", THAI JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 43, no. 3, 30 October 2019 (2019-10-30), pages 119 - 124, XP055964948
MIYAUCHI, S. ET AL.: "HPV16 E5 Mediates Resistance to PD-L1 Blockade and can be targeted with Rimantadine in Head and Neck Cancer", CANCER RESEARCH, vol. 80, no. 4, 2020, pages 732 - 746, XP055875312, DOI: 10.1158/0008-5472.CAN-19-1771
DRAKOPOULOS ANTONIOS, TZITZOGLAKI CHRISTINA, MA CHULONG, FREUDENBERGER KATHRIN, HOFFMANN ANJA, HU YANMEI, GAUGLITZ GÜNTER, SCHMIDT: "Affinity of Rimantadine Enantiomers against Influenza A/M2 Protein Revisited", ACS MEDICINAL CHEMISTRY LETTERS, vol. 8, no. 2, 9 February 2017 (2017-02-09), US , pages 145 - 150, XP055964966, ISSN: 1948-5875, DOI: 10.1021/acsmedchemlett.6b00311
Attorney, Agent or Firm:
PESSIAN, Maysam (US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAIMED IS:

1. A method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of either enantiomerically pure 2- S rimantadine or a pharmaceutically acceptable salt thereof.

2. The method of claim 1, wherein the side effects associated with administration of 2-S rimantadine are reduced as compared to the side effects associated with racemic rimantadine.

3. The method of claims 1 or 2, wherein the subject is administered a pharmaceutically acceptable salt of 2-S rimantadine.

4. The method of any one of claims 1-3, wherein the pharmaceutically acceptable salt is a hydrochloride salt.

5. The method of any one of claims 1 -4, wherein the cancer is selected from one or more of melanoma, head and neck cancer, lung cancer, colon cancer, breast cancer, esophageal cancer, pancreatic cancer, prostate cancer, cervical cancer, and stomach cancer.

6. The method of any one of claims 1 -4, wherein the cancer is a sarcoma, carcinoma, lymphoma, or leukemia.

7. The method of claim 6, wherein the carcinoma is a squamous cell carcinoma.

8. The method of claim 7, wherein the squamous cell carcinoma is head and neck squamous cell carcinoma.

9. The method of claim 5, wherein the cancer is selected from the group consisting of head and neck cancer, breast cancer, and melanoma.

10. The method of any one of claims 1-9, wherein the cancer is an HPV-associated cancer.

11. The method of claim 10, wherein the HPV -associated cancer is associated with the alpha genus of HPV.

12. The method of any one of claims 1-11, wherein one or more cancer cells from the subject express a human papilloma virus (HPV) protein.

13. The method of claim 12, wherein the HPV protein is E5, HPV protein.

14. The method of claim 13, wherein the HPVE5, protein is from one ormoreHPV subtypes selected from the group consisting of HPV 6, HPV 11 , HPV 16, HPV 18, HPV 31 , HPV 33, HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV58, HPV 66, and HPV 69.

15. The method of claim 14, wherein the HPV protein is E5 from HPV 16

16. The method of claim 14, wherein the HPV protein is E5 from HPV 18.

17. A method of treating cancer in a subject, the method comprising: (a) detecting in a sample from the subject a cancer cell that expresses a human papilloma virus (HPV) protein; and

(b) administering to the subject a therapeutically effective amount of 2-S rimantadine or a pharmaceutically acceptable salt thereof.

18. The method of claim 17, wherein the cancer is associated with the alpha genus of HPV.

19. The method of claims 17 or 18, wherein the HPV protein is one or more of an E5, E6, or E7 HPV protein.

20. The method of claim 19, wherein the HPVE5, E6, or E7 protein is from one or more HPV subtypes selected from the group consisting of HPV 6, HPV 11 , HPV 16, HPV 18, HPV 31, HPV 33, HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV 58, HPV 66, and HPV 69.

21. The method of any one of claims 17-18, wherein the cancer is selected from the group consisting of head and neck cancer, mucosal squamous cell carcinomas, cutaneous squamous cell carcinomas, hepatic cancer, cervical cancer, vaginal cancer, vulvar cancer, penile cancer, and anal cancer.

22. The method of any one of claims 1-21, wherein the method further comprises administering an additional anti-cancer agent.

23. The method of claim 22, wherein the additional anti-cancer agent is selected from the group consisting of: carboplatin, cisplatin, gemcitabine, methotrexate, paclitaxel, pemetrexed, lomustine, temozolomide, dacarbazine, and a combination thereof.

24. The method of claim 22, wherein the additional anti-cancer agent is an immunotherapy.

25. The method of claim 24, wherein the additional anti-cancer agentis an immune checkpoint inhibitor.

26. The method of claim 23, wherein the immune checkpoint inhibitor targets one or more of: CTLA-4, PD-1, PD-L1, BTLA, LAG-3, A2AR, TIM-3, B7-H3, VISTA, and IDO.

27. The method of claims 25 or 26, wherein the immune checkpoint inhibitor is selected form the group consisting of: ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, tremelimumab, cemiplimab, and a combination thereof.

28. The method of any one of claims 1 -27, wherein the method further comprises subjecting the subject to radiation therapy, surgery, or a combination thereof.

29. The method of any one of claims 1-18, wherein the subject is a human.

30. A method of treating a precancerous HPV lesion in a subject needing treatment comprising administering a therapeutically effective amount of rimantadine.

31. The method of claim 30, wherein the HPV lesion is associated with the alpha genus of HPV.

32. The method according to claims 30 or 31, wherein the rimantadine is a racemic mixture.

33. The method according to claims 30 or 31, wherein the rimantadine is purified 2-S rimantadine.

34. The method according to claims 30 or 31, wherein the rimantadine is purified 2-R rimantadine.

35. The method according to any one of claims 30-34, wherein the HPV precancerous lesion is a lesion of the cervix, skin, urethra, nasal cavity, paranasal sinus, larynx, tracheobronchial mucosa or oral cavity.

36. The method of any one of claims 30-35, wherein the HPV precancerous lesion expresses one or more HPV proteins selected from one or more of E5, E6, or E7 HPV protein.

37. The method of claim 36, wherein the HPVE5, E6, orE7 protein is from one or more HPV subtypes selected from the group consisting of one or more of HPV 6, HPV 11, HPV 16, HPV 18, HPV 31 , HPV 33, HPV 35, HPV 39, HPV 45, HPV 51 , HPV 52, HPV 56, HPV 58, HPV 66, and HPV 69.

38. The method according to any one of claims 1-37, wherein the rimantadine is administered topically, orally, subcutaneously, or parenterally.

39. A method of treating or preventing avian bird flu in poultry comprising administering a therapeutically effective amount of pure 2-S rimantadine or pure 2-S rimantadine or a pharmaceutically acceptable salt thereof.

40. The method according to claim 39, wherein the avian bird flu is H5N1.

41. The method of any one of claims 3-29, wherein the side effects associated with administration of 2-S rimantadine are reduced as compared to the side effects associated with racemic rimantadine or enantiomerically pure 2-R rimantadine.

42. A method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of enantiomerically pure 2-R rimantadine or a pharmaceutically acceptable salt thereof.

43. The method of claim 42, wherein the side effects associated with administration of pure 2- R rimantadine are reduced as compared to the side effects associated with racemic rimantadine or 2-S rimantadine.

44. The method of claims 42 or 43, wherein the subject is administered a pharmaceutically acceptable salt of pure 2-R rimantadine.

45. The method of any one of claims 42 or 44, wherein the pharmaceutically acceptable salt is a hydrochloride salt.

46. The method of any one of claims 42-45, wherein the cancer is selected from one or more of melanoma, head and neck cancer, lung cancer, colon cancer, breast cancer, esophageal cancer, pancreatic cancer, prostate cancer, cervical cancer, and stomach cancer.

47. The method of any one of claims 42-45, wherein the cancer is a sarcoma, carcinoma, lymphoma, or leukemia.

48. The method of claim 47, wherein the carcinoma is a squamous cell carcinoma.

49. The method of claim 48, wherein the squamous cell carcinoma is head and neck squamous cell carcinoma.

50. The method of claim 46, wherein the cancer is selected from the group consisting of head and neck cancer, breast cancer, and melanoma.

51. The method of any one of claims 42-50, wherein the cancer is an HPV-associated cancer.

52. The method of any one of claims 42-51 , wherein one or more cancer cells from the subject express a human papilloma virus (HPV) protein.

53. The method of claim 51 , wherein the HPV -associated cancer is associated with the alpha genus of HPV.

54. The method of claims 51 or 53, wherein the HPV protein is E5, HPV protein.

55. The method of claim 54, wherein the HPVE5, protein is from one or more HPV subtypes selected from the group consisting of HPV 6, HPV 11 , HPV 16, HPV 18, HPV 31 , HPV 33, HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV58, HPV 66, and HPV 69.

56. The method of claim 55, wherein the HPV protein is E5 from HPV 16.

57. The method of claim 55, wherein the HPV protein is E5 from HPV 18.

58. A method of treating cancer in a subj ect, the method comprising:

(a) detecting in a sample from the subject a cancer cell that expresses a human papilloma virus (HPV) protein; and

(b) administering to the subject a therapeutically effective amount of pure 2-R rimantadine or a pharmaceutically acceptable salt thereof.

59. The method of claim 58, wherein the HPV protein is associated with the alpha genus of HPV.

60. The method of claim 59, wherein the HPV protein is one or more of an E5, E6, or E7 HPV protein.

61. The method of claim 60, wherein the HPVE5, E6, or E7 protein is from one or more HPV subtypes selected from the group consisting of HPV 6, HPV 11 , HPV 16, HPV 18, HPV 31, HPV 33, HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV 58, HPV 66, and HPV 69.

62. The method of any one of claims 58-61, wherein the cancer cell is from a cancer selected from the group consisting of head and neck cancer, mucosal squamous cell carcinomas, cutaneous squamous cell carcinomas, cervical cancer, vaginal cancer, vulvar cancer, penile cancer, and anal cancer.

63. The method of any one of claims 42-62, wherein the method further comprises administering an additional anti-cancer agent.

64. The method of claim 63, wherein the additional anti-cancer agent is selected from the group consisting of: carboplatin, cisplatin, gemcitabine, methotrexate, paclitaxel, pemetrexed, lomustine, temozolomide, dacarbazine, and a combination thereof.

65. The method of claim 63, wherein the additional anti-cancer agent is an immunotherapy.

66. The method of claim 63 , wherein the additional anti-cancer agent is an immune checkpoint inhibitor.

67. The method of claim 66, wherein the immune checkpoint inhibitor targets one or more of: CTLA-4, PD-1, PD-L1, BTLA, LAG-3, A2AR, TIM-3, B7-H3, VISTA, and IDO.

68. The method of claims 66 or 67, wherein the immune checkpoint inhibitor is selected form the group consisting of: ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, tremelimumab, cemiplimab, and a combination thereof.

69. The method of any one of claims 42-68, wherein the method further comprises subjecting the subject to radiation therapy, surgery, or a combination thereof.

70. The method of any one of claims 42-69, wherein the subject is a human.

71. The method of any one of claims 42-70, wherein the side effects associated with administration of pure 2 -A rimantadine are reduced as comparedto the side effects associated with racemic rimantadine or pure 2-S rimantadine.

72. A composition comprising: pure 2-R rimantadine or a pharmaceutically acceptable salt thereof, pure 2-S rimantadine or a pharmaceutically acceptable saltthereof, or racemic rimantadine or a pharmaceutically acceptable salt thereof; and one or more immune checkpoint inhibitors.

73. The composition of claim 72, wherein the one or more immune checkpoint inhibitors comprises CTLA-4, PD-1, PD-L1, BTLA, LAG-3, A2AR, TIM-3, B7-H3, VISTA, IDO, or any combination thereof.

74. The composition of claim 72, wherein the pure 2 -R rimantadine or a pharmaceutically acceptable salt thereof, pure 2-S rimantadine or a pharmaceutically acceptable salt thereof, or racemic rimantadine or a pharmaceutically acceptable salt thereof is formulated for an injection.

Description:
2-S RIMANTADINE AND 2-R RIMANTADINE FOR TREATING CANCER AND PRECANCEROUS PAPILLOMA VIRUS LESIONS

CROSS-REFERENCE

[0001] The present application claims the benefit of U.S. Provisional Application Serial No. 63/150,027, filed on February 16, 2021, which is incorporated herein by reference in its entirety.

INCORPORATION BY REFERENCE

[0002] All publications, patents, and patent applications disclosed herein are incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. In the event of a conflict between a term disclosed herein and a term in an incorporated reference, the term herein controls.

FIELD OF THE INVENTION

[0003] The present disclosure relates to methods of treating or preventing cancer, including cancers caused by papilloma viruses comprising administering enantiomerically pure 2-S enantiomer of rimantadine or enantiomerically pure 2-R rimantadine.

BACKGROUND OF THE DISCLOSURE

[0004] Genital human papillomavirus (HPV) is the most common sexually transmitted infection in the United States. The Centers for Disease Control and Prevention (CDC) states that 90% of HPV infections cause no symptoms and resolve spontaneously within two years. However, in some cases, an HPV infection persists and results in either warts or precancerous lesions. These lesions, depending on the site affected, increase the risk of cancer of the cervix, vulva, vagina, penis, anus, rectum, and oropharynx. HPV types associated with cervical oncogenicity are classified into 15 "high-risk types" (HPV 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73 and 82) and 3 “possibly high-risk types” (HPV 26, 53 and 66). Researchers have also shown an association of HPV 16 and 18 with breast cancer. HPV types (HPV 6, 11, 40, 42, 43, 44, 54, 61, 70, 72 and 81) are classified as "low risk types" and are known to show benign low-grade cervical changes, genital warts and recurrent respiratory papillomatosis. Skin HPV types 5, 8, and 92 are associated with skin cancer.

[0005] Rimantadine hydrochloride (a-methyl-l-adamantane-methalamine hydrochloride) is an oral medication sold under the brand name Flumadine® that is used to treat influenza A. Rimantadine inhibits influenza activity by binding to amino acids in the virus M2 transmembrane channel and blocking proton transport across the M2 channel. Flumadine® contains a racemic mixture of rimantadine. One study found evidence that the R-enantiomer binds the M2 channel pore with greater affinity than the S-enantiomer. However, that finding is in conflict with several earlier findings that found no differences between the enantiomers against M2. The absence of a distinction between the enantiomers against M2 was confirmed in later studies. Rimantadine has also been suggested to have some anti-Parkinsonian activity. However, its use for this indication has not been developed or approved.

[0006] Flumadine® has gastrointestinal and central nervous system adverse effects including nausea, upset stomach, vomiting, anorexia, dry mouth, abdominal pain, asthenia, nervousness, tiredness, lightheadedness, dizziness, headache, trouble sleeping, difficulty concentrating confusion and anxiety. Anxiety and insomnia are the most commonly cited toxicities for discontinuation of treatment.

SUMMARY OF THE DISCLOSURE

[0007] Formula I below shows the chemical structures of the 2-R enantiomer of rimantadine and the 2-S enantiomer of rimantadine.

Formula I

[0008] An aspect of the present disclosure comprises a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically eff ective amount of either enantiomerically pure 2-S rimantadine or a pharmaceutically acceptable salt thereof. In some embodiments, the side effects associated with administration of 2-S rimantadine are reduced as compared to the side effects associated with racemic rimantadine. In some embodiments, the subject is administered a pharmaceutically acceptable salt of 2-S rimantadine. In some embodiments, the pharmaceutically acceptable salt is a hydrochloride salt. In some embodiments, the cancer is selected from one or more of melanoma, head and neck cancer, lung cancer, colon cancer, breast cancer, esophageal cancer, pancreatic cancer, prostate cancer, cervical cancer, and stomach cancer. In some embodiments, the cancer is a sarcoma, carcinoma, lymphoma, or leukemia. In some embodiments, the carcinoma is a squamous cell carcinoma. In some embodiments, the squamous cell carcinoma is head and neck squamous cell carcinoma. In some embodiments, the cancer is selected from the group consisting of head and neck cancer, breast cancer, and melanoma. In some embodiments, the cancer is an HPV-associated cancer. In some embodiments, the HPV-associated cancer is associated with the alpha genus of HPV. In some embodiments, one or more cancer cells from the subject express a human papilloma vims (HPV) protein. In some embodiments, the HPV protein is E5, HPV protein. In some embodiments, the HPV E5, protein is from one or more HPV subtypes selected from the group consisting of HPV 6, HPV 11, HPV 16, HPV 18, HPV 31, HPV 33, HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV58, HPV 66, and HPV69. In some embodiments, the HPV protein is E5 from HPV 16. In some embodiments, the HPV protein is E5 from HPV 18.

[0009] Another aspect of the present disclosure comprises a method of treating cancer in a subject, the method comprising: (a) detecting in a sample from the subject a cancer cell that expresses a human papilloma vims (HPV) protein; and (b) administering to the subject a therapeutically effective amount of 2-S rimantadine or a pharmaceutically acceptable salt thereof. In some embodiments, the cancer is associated with the alpha genus of HPV. In some embodiments, the HPV protein is one or more of an E5, E6, or E7 HPV protein. In some embodiments, the HPV E5, E6, or E7 protein is from one or more HPV subtypes selected from the group consisting of HPV 6, HPV 11 , HPV 16, HPV 18, HPV 31 , HPV 33 , HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV 58, HPV 66, and HPV 69. In some embodiments, the cancer is selected from the group consisting of head and neck cancer, mucosal squamous cell carcinomas, cutaneous squamous cell carcinomas, cervical cancer, vaginal cancer, vulvar cancer, penile cancer, and anal cancer. In some embodiments, the methodfurther comprises administering an additional anti -cancer agent. In some embodiments, the additional anti-cancer agent is selected from the group consisting of: carboplatin, cisplatin, gemcitabine, methotrexate, paclitaxel, pemetrexed, lomustine, temozolomide, dacarbazine, and a combination thereof. In some embodiments, the additional anti-cancer agent is an immunotherapy. In some embodiments, the additional anti-cancer agent is an immune checkpoint inhibitor. In some embodiments, the immune checkpoint inhibitor targets one or more of: CTLA-4, PD-1, PD-L1, BTLA, LAG-3, A2AR, TIM-3, B7-H3, VISTA, and IDO. In some embodiments, the immune checkpoint inhibitor is selected form the group consisting of: ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, tremelimumab, cemiplimab, and a combination thereof. In some embodiments, the method further comprises subjecting the subject to radiation therapy, surgery, or a combination thereof. In some embodiments, the subject is a human. [0010] Another aspect of the present disclosure comprises a method of treating a precancerous HPV lesion in a subject needing treatment comprising administering a therapeutically effective amount of rimantadine. In some embodiments, the HPV lesion is associated with the alpha genus of HPV. In some embodiments, the rimantadine is a racemic mixture. In some embodiments, the rimantadine is purified 2 -S rimantadine. In some embodiments, the rimantadine is purified 2-R rimantadine. In some embodiments, the HPV precancerous lesion is a lesion of the cervix, skin, urethra, nasal cavity, paranasal sinus, larynx, tracheobronchial mucosa or oral cavity. In some embodiments, the HPV precancerous lesion expresses one or more HPV proteins selected from one or more ofE5, E6, orE7 HPV protein. In some embodiments, the HPV E5, E6, or E7 protein is from one or more HPV subtypes selected from the group consisting of one or more of HPV 6, HPV 11 , HPV 16, HPV 18, HPV 31 , HPV 33, HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV 58, HPV 66, and HPV 69. In some embodiments, the rimantadine is administered topically, orally, subcutaneously, or parenterally.

[0011] Another aspect of the present disclosure comprises a method of treating or preventing avian bird flu in poultry comprising administering a therapeutically effective amount of pure 2-S rimantadine or pure 2-S rimantadine or a pharmaceutically acceptable salt thereof. In some embodiments, the avian bird flu is H5N 1. In some embodiments, the side effects associated with administration of 2-S rimantadine are reduced as compared to the side effects associated with racemic rimantadine or enantiomerically pure 2-R rimantadine.

[0012] Another aspect of the present disclosure comprises a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of enantiomerically pure 2-R rimantadine or a pharmaceutically acceptable salt thereof. In some embodiments, the side effects associated with administration of pure 2-R rimantadine are reduced as compared to the side effects associated with racemic rimantadine or 2-S rimantadine. In some embodiments, the subject is administered a pharmaceutically acceptable salt of pure 2-R rimantadine. In some embodiments, the pharmaceutically acceptable salt is a hydrochloride salt. In some embodiments, the cancer is selected from one or more of melanoma, head and neck cancer, lung cancer, colon cancer, breast cancer, esophageal cancer, pancreatic cancer, prostate cancer, cervical cancer, and stomach cancer. In some embodiments, the cancer is a sarcoma, carcinoma, lymphoma, or leukemia. In some embodiments, the carcinoma is a squamous cell carcinoma. In some embodiments, the squamous cell carcinoma is head and neck squamous cell carcinoma. In some embodiments, the cancer is selected from the group consisting of head and neck cancer, breast cancer, and melanoma. In some embodiments, the cancer is an HPV-associated cancer. In some embodiments, one or more cancer cells from the subject express a human papilloma virus (HPV) protein. In some embodiments, the HPV-associated cancer is associated with the alpha genus of HPV. In some embodiments, the HPV protein is E5, HPV protein. In some embodiments, the HPVE5, protein is from one or more HPV subtypes selected from the group consisting of HPV 6, HPV 11, HPV 16, HPV 18, HPV 31, HPV 33 , HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV58, HPV 66, and HPV 69. In some embodiments, the HPV protein is E5 from HPV 16. In some embodiments, the HPV protein is E5 from HPV 18.

[0013] Another aspect of the present disclosure comprises a method of treating cancer in a subject, the method comprising: (a) detecting in a sample from the subject a cancer cell that expresses a human papilloma virus (HPV) protein; and (b) administering to the subject a therapeutically effective amount of pure 2-R rimantadine or a pharmaceutically acceptable salt thereof. In some embodiments, the HPV protein is associated with the alpha genus of HPV. In some embodiments, the HPV protein is one or more of an E5, E6, or E7 HPV protein. In some embodiments, the HPV E5, E6, or E7 protein is from one or more HPV subtypes selected from the group consisting of HPV 6, HPV 11 , HPV 16, HPV 18, HPV 31 , HPV 33 , HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV 58, HPV 66, and HPV 69. In some embodiments, the cancer cell is from a cancer selected from the group consisting of head and neck cancer, mucosal squamous cell carcinomas, cutaneous squamous cell carcinomas, cervical cancer, vaginal cancer, vulvar cancer, penile cancer, and anal cancer. In some emb odiments, the method further comprises administering an additional anti-cancer agent. In some embodiments, the additional anti -cancer agent is selected from the group consisting of: carboplatin, cisplatin, gemcitabine, methotrexate, paclitaxel, pemetrexed, lomustine, temozolomide, dacarbazine, and a combination thereof. In some embodiments, the additional anti-cancer agent is an immunotherapy. In some embodiments, the additional anti-cancer agent is an immune checkpoint inhibitor. In some embodiments, the immune checkpoint inhibitor targets one or more of: CTLA-4, PD-1, PD-L1, BTLA, LAG-3, A2AR, TIM-3, B7-H3, VISTA, and IDO. In some embodiments, the immune checkpoint inhibitor is selected form the group consisting of: ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, tremelimumab, cemiplimab, and a combination thereof. In some embodiments, the method further comprises subjecting the subject to radiation therapy, surgery, or a combination thereof. In some embodiments, the subject is a human. In some embodiments, the side effects associated with administration of pure 2-R rimantadine are reduced as compared to the side effects associated with racemic rimantadine or pure 2-S rimantadine.

[0014] Another aspect of the present disclosure comprises a composition comprising: pure 2-R rimantadine or a pharmaceutically acceptable salt thereof, pure 2-S rimantadine or a pharmaceutically acceptable salt thereof; and one or more immune checkpoint inhibitors. In some embodiments, the one or more immune checkpoint inhibitors comprises CTLA-4, PD-1, PD-L1, BTLA, LAG-3, A2AR, TIM-3, B7-H3, VISTA, IDO, or any combination thereof. In some embodiments, the pure 2-R rimantadine or a pharmaceutically acceptable salt thereof, pure 2-S rimantadine or a pharmaceutically acceptable salt thereof, or racemic rimantadine or a pharmaceutically acceptable salt thereof is formulatedfor an injection.

BRIEF DESCRIPTION OF THE DRAWINGS

[0015] Figures 1A-B shows the peak current amplitude measurements and steady state current measurements of 2-S rimantadine (TGN-S15) and 2-R rimantadine (TGN-S16) for NR2A. Figures 1C-D shows the peak current amplitude measurements and steady state current measurements of 2-S rimantadine (TGN-S15) and 2 -A rimantadine (TGN-S16) forNR2B. [0016] Figure 2 shows the proliferation of CAL-27 cells with varying concentrations of it'-rimantadine (TGN-S11), S-rimantadine (TGN-S15), R-rimantadine (TGN-S16), and memantine (TGN-S13).

DETAILED DESCRIPTION OF THE EMBODIMENTS

[0017] An aspect of the present disclosure is the use of enantiomerically pure 2-S rimantadine or enantiomerically pure 2-R rimantadine for treating cancers. In some embodiments disclosed herein is the use of 2-S rimantadine (also referred to as “S-rimantadine”) or enantiomerically pure 2-R rimantadine for treating cancers associated with papillomaviruses, such as human papilloma viruses (HPV). In some embodiments, the HPV is an HPV from the alpha genus.

[0018] Another aspect of the disclosure is the use of 2-S rimantadine or enantiomerically pure 2-R rimantadine for treating precancerous lesions associated with papilloma viruses, such as human papilloma viruses.

[0019] Racemic rimantadine has side effects at currently prescribed doses. The side effects include central nervous system (CNS) side effects, sleep side effects, gastrointestinal side effects, and atropinic side effects, such as, without limitation light headedness, dizziness, depression, confusion, difficulty concentrating, anxiety (such as nervousness), irritability, hallucinations, and headache, insomnia, excess fatigue, loss of appetite, nausea, vomiting, constipation, dry mouth, blurred vision, difficulty voiding and difficulty swallowing. Anxiety and insomnia are the most commonly cited toxicities of racemic rimantadine for discontinuation of treatment.

[0020] As disclosed herein, 2-S rimantadine inhibits the N-methyl-D-aspartate subtype glutamate receptors (NMD A) subunit NR2B subunits to a lesser degree as compared to 2-R rimantadine and racemic rimantadine (See, Table 2 in Example 2 below).

[0021] In some embodiments, disclosed herein is the use of 2-S rimantadine for treating cancer, cancer associated with HPV, precan cerous lesions associated with HPV, and/or influenza A. Due to its lower ability to inhibit NR2B as compared to racemic rimantadine, 2-S rimantadine can have less side effects as compared to treating these conditions with racemic rimantadine or 2- R rimantadine. Due to its ability to inhibit NR2B to a greater degree than 2-S rimantadine, 2-R rimantadine can have less side effects as compared to treating these conditions with racemic rimantadine or 2-S rimantadine.

[0022] In some embodiments, disclosed herein is the use of 2-R rimantadine for treating cancer, cancer associated with HPV, precan cerous lesions associated with HPV, and/or influenza A. Due to its greater ability to inhibit NR2B as compared to racemic rimantadine, 2 -A rimantadine can have less side effects as compared to treating these conditions with racemic rimantadine or 2- S rimantadine.

[0023] In some embodiments, disclosed herein 2-S rimantadine can have less side effects as compared to treating these conditions with racemic rimantadine or 2-R rimantadine due to 2-S rimantadine’s lower ability to antagonize NMDA receptors and/or inhibit NMDA-mediated biological pathways. In some embodiments, the degree of NMDA receptor inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is about 10 % less to about 100 % less. In some embodiments, the degree of NMD A receptor inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is about 10 % less to about 20 % less, about 10 % less to about 30 % less, about 10 % less to about40 % less, about 10 % less to about 50 % less, about 10 % less to about 60 % less, about 10 % less to about70 % less, about 10 % less to about 80 % less, about 10 % less to about 90 % less, about 10 % less to about 100 % less, about 20 % less to about 30 % less, about 20 % less to about 40 % less, about 20 % less to about 50 % less, about 20 % less to about 60 % less, about 20 % less to about 70 % less, about 20 % less to about 80 % less, about 20 % less to about 90 % less, about 20 % less to about 100 % less, about 30 % less to about 40 % less, about 30 % less to about 50 % less, about 30 % less to about 60 % less, about 30 % less to about 70 % less, about 30 % less to about 80 % less, about 30 % less to about 90 % less, about 30 % less to about 100 % less, about 40 % less to about 50 % less, about 40 % less to about 60 % less, about 40 % less to about 70 % less, about 40 % less to about 80 % less, about 40 % less to about 90 % less, about 40 % less to about 100 % less, about 50 % less to about 60 % less, about 50 % less to about 70 % less, about 50 % less to about 80 % less, about 50 % less to about 90 % less, about 50 % less to about 100 % less, about 60 % less to about 70 % less, about 60 % less to about 80 % less, about 60 % less to about 90 % less, about 60 % less to about 100 % less, about 70 % less to about 80 % less, about 70 % less to about 90 % less, about 70 % less to about 100 % less, about 80 % less to about 90 % less, about 80 % less to about 100 % less, or about 90 % less to about 100 % less. In some embodiments, the degree of NMDA receptor inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is about 10 % less, about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the degree of NMDA receptor inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is at least about 10 % less, about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, or about 90 % less. In some embodiments, the degree of NMDA receptor inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is at most about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the NMDA receptor is NR2 A. In some embodiments, the NMDA receptor is NR2B.

[0024] In some embodiments, disclosed herein 2-R rimantadine can have less side effects as compared to treating these conditions with racemic rimantadine or 2-S rimantadine due to 2-R rimantadine’s lower ability to antagonize NMDA receptors and/or inhibit NMD A-mediated biological pathways. In some embodiments, the degree of NMDA receptor inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is about 10 % less to about 100 % less. In some embodiments, the degree of NMDA receptor inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is about 10 % less to about 20 % less, about 10 % less to about 30 % less, about 10 % less to about40 % less, about 10 % less to about 50 % less, about 10 % less to about 60 % less, about 10 % less to about 70 % less, about 10 % less to about 80 % less, about 10 % less to about 90 % less, about 10 % less to about 100 % less, about 20 % less to about 30 % less, about 20 % less to about 40 % less, about 20 % less to about 50 % less, about 20 % less to about 60 % less, about 20 % less to about 70 % less, about 20 % less to about 80 % less, about 20 % less to about 90 % less, about 20 % less to about 100 % less, about 30 % less to about 40 % less, about 30 % less to about 50 % less, about 30 % less to about 60 % less, about 30 % less to about 70 % less, about 30 % less to about 80 % less, about 30 % less to about 90 % less, about 30 % less to about 100 % less, about 40 % less to about 50 % less, about 40 % less to about 60 % less, about 40 % less to about 70 % less, about 40 % less to about 80 % less, about 40 % less to about 90 % less, about 40 % less to about 100 % less, about 50 % less to about 60 % less, about 50 % less to about 70 % less, about 50 % less to about 80 % less, about 50 % less to about 90 % less, about 50 % less to about 100 % less, about 60 % less to about 70 % less, about 60 % less to about 80 % less, about 60 % less to about 90 % less, about 60 % less to about 100 % less, about 70 % less to about 80 % less, about 70 % less to about 90 % less, about 70 % less to about 100 % less, about 80 % less to about 90 % less, about 80 % less to about 100 % less, or about 90 % less to about 100 % less. In some embodiments, the degree of NMDA receptor inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is about 10 % less, about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the degree of NMDA receptor inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is at least about 10 % less, about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, or about 90 % less. In some embodiments, the degree of NMDA receptor inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is at most about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the NMDA receptor is NR2 A. In some embodiments, the NMDA receptor is NR2B.

[0025] In some embodiments, disclosed herein 2-S rimantadine can have less side effects as compared to treating these conditions with racemic rimantadine or 2-R rimantadine due to 2-S rimantadine’s lower ability to antagonize GABA receptors and/or inhibit GABA-mediated biological pathways. In some embodiments, the degree of GABA receptor and/or GABA- mediated biological pathway inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is about 10 % less to about 100 % less. In some embodiments, the degree of GABA receptor and/or GABA-mediated biological pathway inhibition caused by 2- S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is about 10 % less to about 20 % less, about 10 % less to about 30 % less, about 10 % less to about40 % less, about 10 % less to about 50 % less, about 10 % less to about 60 % less, about 10 % less to about70 % less, about 10 % less to about 80 % less, about 10 % less to about 90 % less, about 10 % less to about 100 % less, about 20 % less to about 30 % less, about 20 % less to about 40 % less, about 20 % less to about 50 % less, about 20 % less to about 60 % less, about 20 % less to about 70 % less, about 20 % less to about 80 % less, about 20 % less to about 90 % less, about 20 % less to about 100 % less, about30 % less to about 40 % less, about 30 % less to about 50 % less, about30 % less to about 60 % less, about 30 % less to about 70 % less, about 30 % less to about 80 % less, about 30 % less to about 90 % less, about 30 % less to about 100 % less, about 40 % less to about 50 % less, about 40 % less to about 60 % less, about 40 % less to about 70 % less, about 40 % less to about 80 % less, about 40 % less to about 90 % less, about 40 % less to about 100 % less, about 50 % less to about 60 % less, about 50 % less to about 70 % less, about 50 % less to about 80 % less, about 50 % less to about 90 % less, about 50 % less to about 100 % less, about 60 % less to about 70 % less, about 60 % less to about 80 % less, about 60 % less to about 90 % less, about 60 % less to about 100 % less, about 70 % less to about 80 % less, about 70 % less to about 90 % less, about 70 % less to about 100 % less, about 80 % less to about 90 % less, about 80 % less to about 100 % less, or about 90 % less to about 100 % less. In some embodiments, the degree of GABA receptor and/or GABA-mediated biological pathway inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is about 10 % less, about 20 % less, about30 % less, about40 % less, about 50 %less, about 60 % less, about70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the degree of GABA receptor and/or GABA-mediated biological pathway inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is at least about 10 % less, about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, or about 90 % less. In some embodiments, the degree of GABA receptor and/or GABA- mediated biological pathway inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is at most about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, about 90 % less, or about 100 % less.

[0026] In some embodiments, disclosed herein 2-R rimantadine can have less side effects as compared to treating these conditions with racemic rimantadine or 2-S rimantadine due to 2-R rimantadine’s lower ability to antagonize GABA receptors and/or inhibit GABA-mediated biological pathways. In some embodiments, the degree of GABA receptor and/or GABA- mediated biological pathway inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is about 10 % less to about 100 % less. In some embodiments, the degree of GABA receptor and/or GABA-mediated biological pathway inhibition caused by 2- R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is about 10 % less to about 20 % less, about 10 % less to about 30 % less, about 10 % less to about 40 % less, about 10 % less to about 50 % less, about 10 % less to about 60 % less, about 10 % less to about70 % less, about 10 % less to about 80 % less, about 10 % less to about 90 % less, about 10 % less to about 100 % less, about 20 % less to about 30 % less, about 20 % less to about 40 % less, about 20 % less to about 50 % less, about 20 % less to about 60 % less, about 20 % less to about 70 % less, about 20 % less to about 80 % less, about 20 % less to about 90 % less, about 20 % less to about 100 % less, about30 % less to about40 % less, about30 % less to about 50 % less, about30 % less to about 60 % less, about 30 % less to about 70 % less, about 30 % less to about 80 % less, about 30 % less to about 90 % less, about 30 % less to about 100 % less, about 40 % less to about 50 % less, about 40 % less to about 60 % less, about 40 % less to about 70 % less, about 40 % less to about 80 % less, about 40 % less to about 90 % less, about 40 % less to about 100 % less, about 50 % less to about 60 % less, about 50 % less to about 70 % less, about 50 % less to about 80 % less, about 50 % less to about 90 % less, about 50 % less to about 100 % less, about 60 % less to about 70 % less, about 60 % less to about 80 % less, about 60 % less to about 90 % less, about 60 % less to about 100 % less, about 70 % less to about 80 % less, about 70 % less to about 90 % less, about 70 % less to about 100 % less, about 80 % less to about 90 % less, about 80 % less to about 100 % less, or about 90 % less to about 100 % less. In some embodiments, the degree of GABA receptor and/or GABA-mediated biological pathway inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is about 10 % less, about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the degree of GABA receptor and/or GABA-mediated biological pathway inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is at least about 10 % less, about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, or about 90 % less. In some embodiments, the degree of GABA receptor and/or GABA- mediated biological pathway inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is at most about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, about 90 % less, or about 100 % less.

[0027] In some embodiments, disclosed herein 2-S rimantadine can have less side effects as compared to treating these conditions with racemic rimantadine or 2-R rimantadine due to 2-S rimantadine’s lower ability to antagonize dopamine receptors and/or inhibit dopamine -mediated biological pathways. In some embodiments, the degree of dopamine receptor and/or dopamine- mediated biological pathway inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is about 10 % less to about 100 % less. In some embodiments, the degree of dopamine receptor and/or dopamine-mediated biological pathway inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is about 10 % less to about 20 % less, about 10 % less to about 30 % less, about 10 % less to about 40 % less, about 10 % less to about 50 % less, about 10 % less to about 60 % less, about 10 % less to about 70 % less, about 10 % less to about 80 % less, about 10 % less to about 90 % less, about 10 % less to about 100 % less, about 20 % less to about 30 % less, about 20 % less to about 40 % less, about 20 % less to about 50 % less, about 20 % less to about 60 % less, about 20 % less to about 70 % less, about 20 % less to about 80 % less, about 20 % less to about 90 % less, about 20 % less to about 100 % less, about30 % less to about40 % less, about30 % less to about 50 % less, about30 % less to about 60 % less, about 30 % less to about 70 % less, about 30 % less to about 80 % less, about 30 % less to about 90 % less, about 30 % less to about 100 % less, about 40 % less to about 50 % less, about 40 % less to about 60 % less, about 40 % less to about 70 % less, about 40 % less to about 80 % less, about 40 % less to about 90 % less, about 40 % less to about 100 % less, about 50 % less to about 60 % less, about 50 % less to about 70 % less, about 50 % less to about 80 % less, about 50 % less to about 90 % less, about 50 % less to about 100 % less, about 60 % less to about 70 % less, about 60 % less to about 80 % less, about 60 % less to about 90 % less, about 60 % less to about 100 % less, about 70 % less to about 80 % less, about 70 % less to about 90 % less, about 70 % less to about 100 % less, about 80 % less to about 90 % less, about 80 % less to about 100 % less, or about 90 % less to about 100 % less. In some embodiments, the degree of dopamine receptor and/or dopamine-mediated biological pathway inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is about 10 % less, about 20 % less, about30 % less, about40 % less, about 50 %less, about 60% less, about70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the degree of dopamine receptor and/or dopamine-mediatedbiological pathway inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is at least about 10 % less, about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, or about90 % less. In some embodiments, the degree of dopaminereceptor and/or dopamine- mediated biological pathway inhibition caused by 2-S rimantadine, with respect to 2-R rimantadine or racemic rimantadine is at most about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the dopamine receptor is the D 2 /3 receptor.

[0028] In some embodiments, disclosed herein 2-R rimantadine can have less side effects as compared to treating these conditions with racemic rimantadine or 2-S rimantadine due to 2-R rimantadine’s lower ability to antagonize dopamine receptors and/or inhibit dopamine-mediated biological pathways. In some embodiments, the degree of dopamine receptor and/or dopamine- mediated biological pathway inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine orracemic rimantadine is about 10% less to about 100% less. In some embodiments, the degree of dopamine receptor and/or dopamine-mediated biological pathway inhibition caused by 2-R rimantadine, with respect to 2 -S rimantadine or racemic rimantadine is about 10 % less to about20 % less, about 10 % less to about 30% less, about 10 % less to about40 % less, about 10 % less to about 50 % less, about 10 % less to about 60 % less, about 10 % less to about 70 % less, about 10 % less to about 80 % less, about 10 % less to about 90 % less, about 10 % less to about 100 % less, about 20 % less to about 30 % less, about 20 % less to about 40 % less, about 20 % less to about 50 % less, about 20 % less to about 60 % less, about 20 % less to about 70 % less, about 20 % less to about 80 % less, about 20 % less to ab out 90 % less, about 20 % less to about 100 % less, about30 % less to about40 % less, about30 % less to about 50 % less, about30 % less to about 60 % less, about 30 % less to about 70 % less, about 30 % less to about 80 % less, about 30 % less to about 90 % less, about 30 % less to about 100% less, about 40 % less to about 50 % less, about 40 % less to about 60 % less, about 40 % less to about 70 % less, about 40 % less to about 80 % less, about 40 % less to about 90 % less, about 40 % less to about 100 % less, about 50 % less to about 60 % less, about 50 % less to about 70 % less, about 50 % less to about 80 % less, about 50 % less to about 90 % less, about 50 % less to about 100 % less, about 60 % less to about 70 % less, about 60 % less to about 80 % less, about 60 % less to about 90 % less, about 60 % less to about 100 % less, about 70 % less to about 80 % less, about 70 % less to about 90 % less, about 70 % less to about 100 % less, about 80 % less to about 90 % less, about 80 % less to about 100 % less, or about 90 % less to about 100 % less. In some embodiments, the degree of dopamine receptor and/or dopamine-mediated biological pathway inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is about 10 % less, about 20 % less, about30 % less, about40 % less, about 50 %less, about 60% less, about70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the degree of dopamine receptor and/or dopamine-mediated biological pathway inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is at least about 10 % less, about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, or about 90 % less. In some embodiments, the degree of dopamine receptor and/or dopamine-mediated biological pathway inhibition caused by 2-R rimantadine, with respect to 2-S rimantadine or racemic rimantadine is at most about 20 % less, about 30 % less, about 40 % less, about 50 % less, about 60 % less, about 70 % less, about 80 % less, about 90 % less, or about 100 % less. In some embodiments, the dopamine receptor is the D 2/3 receptor.

[0029] In another aspect, disclosed herein is the use of 2-S rimantadine for the treatment/prevention of flu in veterinary animals, for example, poultry (e.g., chickens, turkeys, and ducks) and horses. Use of 2 -S rimantadine can have less side effects as compared to treating these animals with racemic rimantadine or 2-R rimantadine.

[0030] In another aspect, disclosed herein is the use of 2-R rimantadine for the treatment/prevention of flu in veterinary animals, for example, poultry (e.g., chickens, turkeys, and ducks) and horses. Use of 2-R rimantadine can have less side effects as compared to treating these animals with racemic rimantadine or 2-S rimantadine.

Definitions

[0031] As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the content clearly dictates otherwise. It should also be noted that the term “or” is generally employed in its sense including “and/or” unless the content clearly dictates otherwise. Further, headings provided herein are for convenience only and do not interpret the scope or meaning of the claimed invention.

[0032] In some embodiments, S-rimantadine, R-rimantadine, racemic rimantadine, or a rimantadine derivative as described herein is PEGylated. As used herein, “PEGylated” or “PEGylation” describes the conjugation of a compound with a polyethylene glycol (PEG) moiety. The PEG moiety can be of any length. For example, the PEG moiety can have from 2 to 500 repeating units. In some embodiments, the PEG moiety can have an average molecular weight of about 300 g/mol to about 10,000,000 g/mol. In some embodiments, thePEGmoiety canbe a high molecular weight PEG or low molecular weight PEG. For example, a high molecular weight PEG has a molecular weight greater than or equal to 5 kDa, and a low molecular weight PEG has a molecular weight of less than 5 kDa. In some embodiments, the PEG is selected from the group consisting of: PEG 200, PEG 300, PEG 400, PEG 600, PEG 800, PEG 1000, PEG 1500, PEG 2000, and PEG 3350. A PEG moiety can be a linear PEG or the PEG moiety can be a branched PEG. For example, branchedPEGs includes any PEG having one or more branches of PEG groups extending from a PEG backbone.

[0033] As used herein the term “pure” when applied to a chiral compound, refers to an enantiomer of the chiral compound substantially free from its opposite enantiomer (i.e., in enantiomeric excess). For example, the pure “R” form of a compound is substantially free from the “S” form of the compound and is, thus, in enantiomeric excess of the “S” form. The term “enantiomerically pure” or “pure enantiomer” denotes that the compound comprises an excess of an enantiomer, e.g. more than 75% by weight, more than 80% by weight, more than 85% by weight, more than 90% by weight, more than 91% by weight, more than 92% by weight, more than 93% by weight, more than 94% by weight, more than 95% by weight, more than 96% by weight, more than 97% by weight, more than 98% by weight, more than 98.5% by weight, more than 99% by weight, more than 99.2% by weight, more than 99.5% by weight, more than 99.6% by weight, more than 99.7% by weight, more than 99.8% by weight or more than 99.9% by weight, of the enantiomer. In certain embodiments, the weights are based upon total weight of the compound, i.e. all enantiomers of the compound. In certain embodiments, one enantiomer can be in excess by 30-80%, or by 30-70%, 30-60%, 30%, 35%, 40%, 45%, 50%, 55% or 60%, or any percentage in between.

[0034] As used herein and unless otherwise indicated, the term “enantiomerically pure “ 2-S rimantadine” refers, e.g., to at least about 80% by weight 2-S rimantadine and at most about 20% by weight 2-R rimantadine, at least about 90% by weight 2-S rimantadine and at most about 10% by weight 2-R rimantadine, at least about 95% by weight 2-S rimantadine and at most about 5% by weight 2-R rimantadine, at least about 99% by weight 2-S rimantadine and at most about 1% by weight 2-R rimantadine or at least about 99.9% by weight 2-S rimantadine and at most about 0.1% by weight 2-R rimantadine. In certain embodiments, the weights are based upon total weight of rimantadine, i.e., both or all of the enantiomers of rimantadine.

[0035] As used herein and unless otherwise indicated, the term “enantiomerically pure “2 -R rimantadine” refers, e.g., to at least about 80% by weight 2-R rimantadine and at most about 20% by weight 2-S rimantadine, at least about 90% by weight 2-R rimantadine and at most about 10% by weight 2-S rimantadine, at least about 95% by weight 2-R rimantadine and at most about 5% by weight 2-S rimantadine, at least about 99% by weight 2-R rimantadine and at most about 1% by weight 2-S rimantadine, at least about 99.9% by weight 2-R rimantadine or at most about 0.1% by weight 2-S rimantadine. In certain embodiments, the weights are based upon total weight of rimantadine, i.e., both or all enantiomers of the rimantadine.

[0036] In the compositions provided herein, enantiomerically pure rimantadine or a pharmaceutically acceptable salt, solvate, hydrate, ester or prodrug thereof can be present with other active or inactive ingredients. For example, a pharmaceutical composition comprising enantiomerically pure 2-S rimantadine can comprise, for example, about 90% excipient and about 10% enantiomerically pure 2-S rimantadine. In certain embodiments, the enantiomerically pure 2- S rimantadine in such compositions can, for example, comprise, at least about 99.9% by weight 2-S rimantadine and at most about 0.1% by weight 2-S rimantadine. In certain embodiments, the active ingredient can be formulated with little or no carrier, excipient or diluent.

[0037] As used herein, the terms “subject,” “individual,” or “patient,” used interchangeably, refer to any animal, including poultry, such as chickens, ducks, turkeys and mammals such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, primates, and humans. In some embodiments, the subject is a human. [0038] As used herein, the terms “treat” or “treatment” refer to therapeutic or palliative measures. Beneficial or desired clinical results include, but are not limited to, alleviation, in whole or in part, of symptoms associated with a disease or disorder or condition, diminishmentof the extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state (e.g., one or more symptoms of the disease), and remission (whether partial ortotal), whether detectable or undetectable. “Treatment’ can also mean prolonging survival as compared to expected survival if not receiving treatment. [0039] The term "preventing” as used herein means the prevention of the onset, recurrence or spread, in whole or in part, of the disease or condition as described herein, or a symptom thereof. [0040] The term “administration” or “administering” refers to a method of giving a dosage of a compound or pharmaceutical composition to a vertebrate or invertebrate, including a mammal, a bird, a fish, or an amphibian. The preferred method of administration can vary depending on various factors, e.g., the components of the pharmaceutical composition, the site of the disease, and the severity of the disease. By “therapeutically effective amount” or “pharmaceutically effective amount” of a compound as provided herein is an amount which is sufficient to achieve the desired effect and can vary according to the nature and severity of the disease condition, and the potency of the compound. A therapeutic effect is the relief, to some extent, of one or more of the symptoms of the disease, and can include curing a disease.

[0041] “Curing” means that the symptoms of active disease are eliminated. However, certain long-term or permanent effects of the disease can exist even after a cure is obtained (such as, e.g, extensive tissue damage).

[0042] The terms “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of an agent or a compound being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate “effective” amount in any individual case may be determinedusingtechniques, such as a dose escalation study. An “effective amount” is an amount sufficient for a compound to accomplish a stated purpose relative to the absence of the compound (e.g., achieve the effect for which it is administered, treat a disease, reduce enzyme activity, increase enzyme activity, reduce a signaling pathway, or reduce one or more symptoms of a disease or condition). An example of an “effective amount” is an amount sufficient to contribute to the treatment, prevention, or reduction of a symptom or symptoms of a disease, which could also be referred to as a “therapeutically effective amount.” A “reduction” of a symptom or symptoms (and grammatical equivalents of this phrase) means decreasing of the severity or frequency of the symptom(s), or elimination of the symptom(s). A “prophylactically effective amount” of a drug is an amount of a drug that, when administered to a subject, will have the intended prophylactic effect, e.g., preventing or delaying the onset (or reoccurrence) of an injury, disease, pathology or condition, or reducing the likelihood of the onset (or reoccurrence) of an injury, disease, pathology, or condition, or their symptoms. The full prophylactic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses. Thus, a prophylactically effective amount may be administered in one or more administrations. An “activity decreasing amount,” as used herein, refers to an amount of antagonist required to decrease the activity of an enzyme relative to the absence of the antagonist. A “function disrupting amount,” as used herein, refers to the amount of antagonist required to disrupt the function of an enzyme or protein relative to the absence of the antagonist. The exact amounts will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols. 1 -3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Pickar, Dosage Calculations (1999); andRemington: The Science and Practice of Pharmacy, 20thEdition, 2003, Gennaro, Ed., Lippincott, Williams & Wilkins).

[0043] The term “immunotherapy” refers to an agent that modulates the immune system. In some embodiments, an immunotherapy can increase the expression and/or activity of a regulator of the immune system. In some embodiments, an immunotherapy can decrease the expression and/or activity of a regulator of the immune system. In some embodiments, an immunotherapy can recruit and/or enhance the activity of an immune cell.

Pharmaceutically Acceptable Salts, Prodrugs, Stereoisomers and Tautomers [0044] The pure R or S enantiomers of rimantadine provided herein can be administered as any salt or prodrug that upon administration to the recipient is capable of providing directly or indirectly the parent compound, or that exhibits activity itself. As used herein, the term "pharmaceutically acceptable salt" refers to a salt that retains the desired biological activity of the subject compound and exhibits minimal undesired toxicological effects. The phrase “pharmaceutically acceptable salt or prodrug” is used throughout the specification to describe any pharmaceutically acceptable form (such as an ester, amide, salt of an ester, salt of an amide or related group) of a compound that, upon administration to a patient, provides an active compound of the present disclosure. Modifications like these can affect the biological activity of the compound, in some cases increasingthe activity over the parent compound. The pharmaceutically acceptable salt may be prepared in situ duringthe final isolation and purification of the compound, or by separately reacting the purified compound in its free acid or free base form with a suitable base or acid, respectively. In some embodiments, a pharmaceutically acceptable salt can be preferred over the respective free base or free acid because such a salt imparts greater stability or solubility to the molecule thereby facilitating formulation into a dosage form. Basic compounds are generally capable of f orming pharmaceutically acceptable acid addition salts by treatment with a suitable acid. Suitable acids include pharmaceutically acceptable inorganic acids and pharmaceutically acceptable organic acids. Non-limiting examples of a pharmaceutically acceptable acid addition salt include hydrochloride, hydrobromide, nitrate, methylnitrate, sulfate, bisulfate, sulfamate, phosphate, acetate, hydroxyacetate, phenylacetate, propionate, butyrate, isobutyrate, valerate, maleate, hydroxymaleate, acrylate, fumarate, malate, tartrate, citrate, salicylate, p-aminosalicy elate, glycollate, lactate, heptanoate, phthalate, oxalate, succinate, benzoate, o-acetoxybenzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxy benzoate, methoxybenzoate, mandelate, tannate, formate, stearate, ascorbate, palmitate, oleate, pyruvate, pamoate, malonate, laurate, glutarate, glutamate, estolate, methanesulfonate (mesylate), ethanesulfonate (esylate), 2 -hydroxy ethanesulfonate, benzenesulfonate (besylate),p- aminobenzenesulfonate, p- toluenesulfonate (tosylate),napthalene-2-sulfonate, ethanedisulfonate, and 2,5- dihydroxy benzoate.

[0045] A pharmaceutically acceptable prodrug refers to a compound that is metabolized (i.e., hydrolyzed or oxidized, for example) in the host to form a compound of the present present disclosure. Typical examples of prodrugs include compounds that have biologically labile protecting groups on a functional moiety of the active compound. Prodrugs include compounds that can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, and/or dephosphorylated to produce the active compound.

[0046] In some embodiments, a method as described herein comprises administering pure 2- S rimantadine, or pure 2-R rimantadine or a pharmaceutically acceptable salt thereof.

Pharmaceutical Compositions

[0047] Also provided herein are pharmaceutical compositions comprising pure 2-S rimantadine, pure 2-R rimantadine or pharmaceutically acceptable salt thereof, as described herein. Any of the pharmaceutical compositions described herein can be administered to a subject to treat a cancer as described herein.

[0048] Administration of 2-S rimantadine, pure 2-R rimantadine or the pharmaceutically acceptable salts thereof, or pharmaceutical compositions thereof, can be via any of the accepted modes of administration, including, but not limited to, orally, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly , intrapulmonarilly, vaginally, rectally, ontologically, neurotologically, intraocularly, sub conjuctiv ally, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, via wound irrigation, intrabuccally, intra- abdominally, intra-articularly, intra-aurally, intrabronchially, intracap sularly, intrameningeally, via inhalation, via endotracheal or endobronchial instillation, via direct instillation into pulmonary cavities, intraspinally, intrasynovially, intrathoracically, via thoracostomy irrigation, epidurally, intratympanically, intracistemally, intravascularly, intraventricularly, intraosseously, via irrigation of infected bone, or via application as part of any admixture with a prosthetic devices. In some embodiments, the administration method includes oral or parenteral administration.

[0049] Pharmaceutically acceptable compositions can include solid, semi -solid, liquid, solutions, colloidal, liposomes, emulsions, suspensions, complexes, coacervates and aerosols. Pharmaceutically acceptable compositions can also include dosage forms, such as, e.g., tablets, capsules, powders, liquids, suspensions, suppositories, aerosols, implants, controlled release or the like. 2-S rimantadine, pure 2-R rimantadine or the pharmaceutically acceptable salts thereof, or pharmaceutical compositions thereof, can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills (tablets and or capsules), transdermal (including electrotransport) patches, implants and the like, for prolonged and/or timed, pulsed administration at a predetermined rate.

[0050] In some embodiments, the pharmaceutical composition is a tablet. In some embodiments, the pharmaceutical composition is a film -coated tablet.

[0051] Pure 2-S rimantadine, pure 2-R rimantadine orthe pharmaceutically acceptable salts thereof, or pharmaceutical compositions thereof, can be administered either alone or in combination with a conventional pharmaceutical carrier, excipient or the like. Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol, polyethylene glycol 1000, succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, poly acrylates, waxes, polyethylene -poly oxypropylene-block polymers, and wool fat. Cyclodextrins can also be used to enhance delivery of compounds described herein.

[0052] In some embodiments, a pharmaceutical composition described herein will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with 2-S rimantadine, pure 2-R rimantadine or the pharmaceutically acceptable salt thereof, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like. In another solid dosage form, a powder, marume, solution or suspension (e.g., in propylene carbonate, vegetable oils, PEG’s, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule). Unit dosage forms in which 2-S rimantadine, pure 2-R rimantadine or the pharmaceutically acceptable salts thereof, provided herein or additional active agents are physically separated are also contemplated; e.g., capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two-compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated.

[0053] In some embodiments, the rimantadine, or pharmaceutically acceptable salt thereof, is PEGylated. In some embodiments, the PEGylated rimantadine, or pharmaceutically acceptable salt thereof, comprises a high molecular weight PEG. In some embodiments, the PEGylated rimantadine, or pharmaceutically acceptable salt thereof, comprises a low molecular weight PEG.In some embodiments, the rimantadine, or a pharmaceutically acceptable salt thereof, is modified. In some embodiments, the modification is PEGylation.

[0054] In some embodiments, the PEGylated rimantadine, or a pharmaceutically acceptable salt thereof, is PEGylated with a high molecular weight PEG. In some embodiments, the PEGylated rimantadine, ora pharmaceutically acceptable salt thereof, is PEGylated with a low molecular weight PEG. Accordingly, also provided herein are methods of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of PEGylated rimantadine, or a pharmaceutically acceptable salt thereof.

[0055] In some embodiments, the pharmaceutical composition includes one or more excipients selected from the group consisting of: hypromellose, magnesium stearate, microcrystalline 5 cellulose, and sodium starch glycolate.

[0056] Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc. a compound provided herein and optional pharmaceutical adjuvants in a carrier (e.g., water, saline, aqueous dextrose, glycerol, glycols, ethanol or the like) to form a solution, colloid, liposome, emulsion, complexes, coacervate or suspension. If desired, the pharmaceutical composition can also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, co -solvents, solubilizing agents, pH buffering agents and the like (e.g., sodium acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine acetate, triethanolamine oleate, and the like).

[0057] Dosage forms or compositions containing 2-S rimantadine, pure 2-R rimantadine or a pharmaceutically acceptable salt thereof, as described herein in the range of 0.005% to 100% with the balance made up from nontoxic carrier may be prepared. The contemplated compositions may contain 0.001%-100% of a compound provided herein, in one embodiment 0.1-95%, in another embodiment 75-85%, in a further embodiment 20-80%. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 22nd Edition (Pharmaceutical Press, London, UK. 2012).

[0058] The pharmaceutical compositions herein can contain, per unit dosage unit, e.g., tablet, capsule, suspension, solution, sachet for reconstitution, powder, injection, I.V., suppository, sublingual/buccal film, teaspoonful and the like, from about 0.1 -1000 mg of 2- S rimantadine, pure 2-R rimantadine or pharmaceutically acceptable salt thereof. Pure 2-S rimantadine, pure 2-R rimantadine or pharmaceutically acceptable salt thereof, can be given at a dosage of from aboutO.01-300 mg/kg/day, or any range therein, preferably from about 0.5-50 mg/kg/day, or any range therein. In some embodiments, the pharmaceutical compositions provided herein contain, per unit dosage unit, about 25 mg to about 500 mg of a compound provided herein (for example, about 25 mg to about 400 mg, about 25 mg to about 300 mg, about 25 mg to about 250 mg, about 25 mg to about 200 mg, about 25 mg to about 150 mg, about 25 mg to about 100 mg, about 25 mg to about 75mg, about 50 mg to about 500 mg, about 100 mg to about 500 mg, about 150 mg to about 500 mg, about 200 mg to about 500 mg, about 250 mg to about 500 mg, about 300 mg to about 500 mg, about 400 mg to about 500 mg, about 50 to about 200 mg, about 100 to about 250 mg, about 50 to about 150 mg). In some embodiments, the pharmaceutical compositions provided herein 5 contain, per unit dosage unit, about 25 mg, about 50 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 400 mg, or about 500 mg of a compound provided herein. The dosages, however, may be varied depending upon the requirement of the patients, the severity of the condition being treated and the compound being employed. In some embodiments, the dosages are administered once daily (QD) or twice daily (BID). [0059] In some embodiments, the present disclosure comprises a composition comprising pure 2-R rimantadine or a pharmaceutically acceptable salt thereof, pure 2-S rimantadine or a pharmaceutically acceptable salt thereof Methods of Treatment

[0060] Also provided herein are methods of treating cancer in a subject. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of one or more of the pharmaceutical compositions described herein. In some embodiments, the pharmaceutical compositions comprise either enantiomerically pure 2-S rimantadine or a pharmaceutically acceptable salt thereof. In some embodiments, the pharmaceutical compositions comprise either enantiomerically pure 2 -R rimantadine or a pharmaceutically acceptable salt thereof. In some embodiments, the pharmaceutically acceptable salt is a hydrochloride salt.

[0061] In some embodiments, the cancer is a sarcoma, carcinoma, melanoma, lymphoma, or leukemia. Non-limiting examples of a sarcoma include: bone sarcoma (e.g., angiosarcoma, fibrosarcoma, liposarcoma, chondrosarcoma, chordoma, Ewing’s sarcoma, giant cell tumor, osteosarcoma, rhabdomyosarcoma, and synovial sarcoma) and soft tissue sarcoma (e.g., fibrosarcoma, 5 gastrointestinal stromal tumor (GIST), Kaposi’s sarcoma, leiomyosarcoma, liposarcoma, rhabdomyosarcoma, and soft tissue Ewing’s sarcoma). Non-limiting examples of a carcinoma include: basal cell carcinoma, squamous cell carcinoma, renal cell carcinoma, invasive ductal carcinoma, hepatocellular carcinoma, and adenocarcinoma. Non-limiting examples of lymphoma include: Non-Hodgkin’s lymphoma (e.g., B-cell lymphoma, T-cell lymphoma, Burkitt’s lymphoma, follicular lymphoma, mantle cell lymphoma, primary mediastinal B-cell lymphoma, small lymphocytic lymphoma, Waldenstrom macroglobulinemia) and Hodgkin’s lymphoma (e.g., lymphocyte-depleted Hodgkin’s disease, lymphocyte-rich Hodgkin’s disease, mixed cellularity Hodgkin’s lymphoma, nodular lymphocyte -predominant Hodgkin’ s disease, and nodular sclerosis Hodgkin’ s lymphoma). Non -limiting examples of leukemia include: acute hairy cell leukemia, acute lymphocytic leukemia, acute myeloid leukemia, acute promyelocytic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, a myeloproliferative neoplasm, and systemic mastocytosis.

[0062] In some embodiments, the cancer is selected from the group consisting of : melanoma, head and neck cancer, lung cancer, colon cancer, anal cancer, breast cancer, esophageal cancer, pancreatic cancer, prostate cancer, cervical cancer, hepatic cancer, and stomach cancer.

[0063] In some embodiments, the cancer is a carcinoma. In some embodiments, the carcinoma is selected from the group consisting of: an adenocarcinoma, a squamous cell carcinoma, a transitional cell carcinoma, a hepatocellular carcinoma, and a clear cell carcinoma. In some embodiments, the cancer is a squamous cell carcinoma. In some embodiments, the squamous cell carcinoma is head and neck squamous cell carcinoma. In some embodiments, the cancer is a hepatocellular carcinoma.

[0064] In some embodiments, the cancer is selected from the group consisting of head and neck cancer, breast cancer, and melanoma.

[0065] In some embodiments, pure 2-S rimantadine or pure 2-R rimantadine, or pharmaceutically acceptable salt thereof, as described herein can be used to treat a hepatitis B virus (HBV)-associated cancer in a subject. An “HBV-associated cancer” as used herein is a cancer in which one or more of the cancerous cells express at least one HB V protein (for example, see , Liu et al., Hepatitis B Virus X Protein Induces RHAMM-Dependent Motility in Hepatocellular Carcinoma Cells via PI3K-Akt-Oct-1 Signaling. Mol Cancer Res. 2020 Mar ; 18(3):375-389. doi: 10.1158/1541-7786.MCR-19-0463. Epub 2019 Dec 2. PMID: 31792079.). For example, one or more cancerous cells can express an HBV oncoprotein. In some embodiments, the HBV-associated cancer is a hepatic cancer (e.g., hepatocellular carcinoma). In some embodiments, the HBV-associated cancer is cervical cancer.

[0066] In some embodiments, pure 2-S rimantadine or pure 2-R rimantadine, or pharmaceutically acceptable salt thereof, as described herein can be used to treat a human papillomavirus (HPV)-associated cancer in a subject. An “HPV-associated cancer” as used herein is a cancer in which one or more of the cancerous cells express at least one HPV protein. For example, one or more of the cancerous cells can express a HPV oncoprotein. Human papillomavirus (HPV) can cause malignant transformation by, for example, targeting the critical tumor suppressors p53 andRb (see, e.g., Conway and Meyers. J Dent Res. 2009 Apr;88(4):307- 17; and Hoppe-Seyler. Trends Microbiol. 2018Feb;26(2):158-168). HPV genes can also help HPV-infected cells evade immune responses (see, e.g., Senba. Oncol Rev. 5 2012 Oct 5;6(2):el7). For example, HPV genes and proteins can target the antigen processing and antigen presentation required for effective adaptive immune responses (see, e.g., Senba. Oncol Rev. 2012 Oct 5;6(2):e17; andO’Brien and Saveria Campo. VirusRes. 2002 Sep;88(l-2):103-17). There are many HPV oncoproteins including, but not limited to, HPV 16 E5, E6, and E7. For example, HPV E5 is protein that has been reported to have multiple functions including regulation of tumor cell differentiation and apoptosis, modulation of H+ ATPase responsible for acidification of late endosomes, and immune modulation including direct binding and downregulation of major histocompatibility complex (MHC) class I and MHC class II (see e.g., Venuti. Mol Cancer. 2011, 10:140), which can affect antigen processing and presentation.

[0067] In some embodiments, one or more cancer cells from the subject express anHPV protein. In some embodiments, theHPV protein is one or more of anHPV E5, E6, orE7 protein. In some embodiments, the HPV E5, E6, or E7 protein is from one or more HPV subtypes selected from the group consisting of: HPV 6, HPV 11 , HPV 16, HPV 18, HPV 31 , HPV 33 , HPV 35, HPV 39, HPV 45, HPV 51, HPV 52, HPV 56, HPV 58, HPV 66, and HPV 69. In some embodiments, the HPV protein isHPV16 E5. In some embodiments, the subject has a cancer selected from the group consisting of : head and neck cancer, a mucosal squamous cell carcinoma, a cutaneous squamous cell carcinoma, cervical cancer, vaginal cancer, vulvar cancer, penile cancer, and anal cancer.

[0068] In some embodiments, the cancer is HPV-associated cancer. In some embodiments, the HPV-associated cancer is HPV-associated head and neck squamous cell carcinoma (HNSCC).

[0069] In some embodiments pure 2-S rimantadine, or pure 2-R rimantadine or pharmaceutically acceptable salt thereof, as described herein can be used to treat a human papillomavirus precancerous lesion such as those associated with, without limitation, proliferative verrucous Leukoplakia (PV1), oral leukoplakia, nicotine palatinus in reverse smokers, oral erythroplakia, laryngeal keratosis, actinic cheilosis, smooth thick leukoplakia, smooth, red tongue of plummer-vinson, smokeless tobacco keratosis, syndrome oral submucous fibrosis, erythroleukoplakia, granular leukoplakia, oral lichen planus (erosive forms), smooth thin leukoplakia, nicotine stomatitis, and tobacco pouch keratosis, cervix (cervical dysplasia); and penile intraepithelial neoplasia (PeIN lesions). In the oral cavity, 24 types of HPV (1, 2, 3, 4, 6, 7, 10, 11, 13, 16, 18, 30, 31, 32, 33, 35,45, 52, 55, 57, 59, 69, 72 and 73) have been associated with benign lesions and 12 types (2, 3, 6, 11, 13, 16, 18, 31, 33, 35, 52 and 57) with malignant lesions. Approximately 40% of invasive penile carcinomas are attributable to HPV 16, 18 and 6/11. HPV types associated with cervical oncogenicity are 15 high risk types (HPV 16, 18, 31, 33, 35, 39,45, 51, 52, 56, 58, 59, 68, 73 and 82) and3 “possibly high-risk types” (HPV 26, 53 and 66).

[0070] Testing for HPV is known in the art for example see Coultlee,F., et. al., 2005, CanJ Infect Dis Med Microbiol 16(2):83-91; care HPV Test Kit (QIAGEN, Redwood City, CA); Tang K.D., 2019, Unlockingthe Potential of Saliva-Based Test to Detect HPV-16-Driven Oropharyngeal Cancer, Cancers (Basel), 11 (4):473 ; HPV probes (BIOCARE MEDICAL, Pacheo, CA)

[0071] In some embodiments, of the methods described herein, pure 2-S rimantidine, or pure 2-R rimantadine or pharmaceutically acceptable salt thereof, is administered in combination with a therapeutically effective amount of at least one additional therapeutic agent selected from one or more additional anti-cancer therapies ortherapeutic agents (e.g., chemotherapeutic agents). Using a combination of different forms of treatment to treat a subject with cancer is a common practice in medical oncology. These other form(s) of conjoint treatment or therapy, in addition to 2-S rimantadine, or pharmaceutically acceptable salt thereof described herein, can include, for example, surgery, radiotherapy, and additional anti -cancer agents, such as kinase inhibitors, signal transduction inhibitors, platinum-based chemotherapy, and/or monoclonal antibodies. In some embodiments, the method further comprises administering an additional anti-cancer agent.

[0072] Non-limiting examples of additional anti-cancer agents include: carboplatin, cisplatin, gemcitabine, methotrexate, paclitaxel, pemetrexed, lomustine, temozolomide, and dacarbazine.

[0073] In some embodiments, the additional anti-cancer agent is an immunotherapy. Many types of immunotherapies can be used in combination with pure 2-S rimantadine, or pure 2 -A rimantadine or pharmaceutically acceptable salts thereof, described herein. Non - limiting examples of an immunotherapy include: immune checkpoint inhibitors, antibody therapy, cellular immunotherapy, antibody -drug conjugates, cytokine therapy, mRNA-based immunotherapy, and cancer vaccines.

[0074] In some embodiments, the immunotherapy is one or more immune checkpoint inhibitors. In some embodiments, the immune checkpoint inhibitor targets one or more of: CTLA-4, PD-1, PD-L1, BTLA, LAG-3, A2AR, TIM-3, B7-H3, VISTA, and IDO. In some embodiments, the checkpoint inhibitor is selected form the group consisting of: ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, cemiplimab, tremelimumab, and a combination thereof.

[0075] In some embodiments, the immune checkpoint inhibitor is a CTLA-4 inhibitor, a PD-1 inhibitor or a PD-L1 inhibitor. In some embodiments, the CTLA-4 inhibitor is ipilimumab (YERVOY®) or tremelimumab (CP-675,206). In some embodiments, the PD-1 inhibitor is pembrolizumab (KEYTRUDA®), cemiplimab (LIBTAYO®), or nivolumab (OPDIVO®). In some embodiments, the PD-L1 inhibitor is atezolizumab (TECENTRIQ®), avelumab (BAVENCIO®) or durvalumab (IMFINZI™). [0076] In some embodiments, the antibody therapy is bevacizumab (MVASTI™, AVASTIN®), trastuzumab (HERCEPTIN®), avelumab (BAVENCIO®), rituximab (MABTHERA™, RITUXAN®), edrecolomab (Panorex), daratumuab (DARZALEX®), olaratumab (LARTRUVO™), ofatumumab (ARZERRA®), alemtuzumab (CAMPATH®), cetuximab (ERBITUX®), oregovomab, pembrolizumab(KEYTRUDA®), dinutiximab (UNITUXIN®), obinutuzumab (GAZYVA®), tremelimumab (CP-675,206), ramucirumab (CYRAMZA®), ublituximab (TG-1101), panitumumab (VECTIBIX®), elotuzumab (EMPLICITI™), avelumab (BAVENCIO®), necitumumab (PORTRAZZA™), cirmtuzumab (UC-961), ibritumomab (ZEVALIN®), isatuximab (SAR650984), nimotuzumab, fresolimumab (GC1008), lirilumab (INN), 5 mogamulizumab (POTELIGEO®), ficlatuzumab (AV-299), denosumab (XGEVA®), ganitumab, urelumab, pidilizumab or amatuximab.

[0077] In some embodiments, the immunotherapy is a cellular immunotherapy (e.g., adoptive T-cell therapy, dendritic cell therapy, natural killer cell therapy).

[0078] In some embodiments, the immunotherapy is an antibody -drug conjugate. In some embodiments, the antibody -drug conjugate is gemtuzumab ozogamicin (MYLOTARG™), inotuzumab ozogamicin (BESPONSA®), brentuximab vedotin (ADCETRIS®), ado-trastuzumab emtansine (TDM-1; KADCYLA®), moxetumomab pasudotox (LUMOXITI®), polatuzumab vedotin-piiq (POLIVY®), mirvetuximab soravtansine (IMGN853), or anetumab ravtansine.

[0079] In some embodiments, the immunotherapy is a cytokine therapy. In some embodiments, the cytokine therapy is an interleukin 2 (IL-2) therapy, an interleukin (IL-15) therapy, an interleukin 7 (IL-7) therapy, an interferon alpha (IFNa) therapy, agranulocyte colony stimulating factor (G-CSF) therapy, an interleukin 12 (IL-12) therapy, or an erythropoietin-alpha (EPO) therapy. In some embodiments, the IL-2 therapy is aldesleukin (Proleukin®). In some embodiments, the IFNa therapy is interferon alfa-2b (e.g.,

IntronA®) or interferon alfa-2a (e.g., Roferon-A®). In some embodiments, the G-CSF therapy is filgrastim (Neupogen®).

[0080] In some embodiments, the immunotherapy is mRNA-based immunotherapy. In some embodiments, the mRNA-based immunotherapy is CV9104 (see, e.g., Rausch et al. (2014) Human Vaccin Immunother 10(11): 3146-52; and Kubler et al. (2015) J. Immunother Cancer 3:26). See also, Pardi et al. Nat Rev Drug Discov . 2018 Apr; 17(4): 261-279, which are incorporated by reference herein in their entirety. [0081] In some embodiments, the method comprises subjecting the subject to radiation therapy, surgery, or a combination thereof. For example, a surgery can be open surgery or minimally invasive surgery.

[0082] In some embodiments, the subject is refractory to standard therapy (e.g., standard of care). In some embodiments, the subject has no standard therapy option. In some embodiments, the subject relapsed or progressed after standard therapy. In some embodiments, the methods provided herein are useful for treating locally advanced or metastatic solid tumors refractory to standard therapies. For example, an HPV associated cancer can be refractory to immune checkpoint inhibitors such as those described herein. [0083] In some embodiments, the subject has a cancer that is 5 refractory or intolerant to standard therapy (e.g., administration of a chemotherapeutic agent, an immunotherapy, or radiation). In some embodiments, the subject has a cancer (e.g., a locally advanced or metastatic tumor) that is refractory or intolerant to prior therapy (e.g., administration of a chemotherapeutic agent, immunotherapy (e.g., an immune checkpoint inhibitor), or radiation). In some embodiments, the cancer that is refractory or intolerant to standard therapy is an HPV-associated cancer. In some embodiments, the subject has a cancer (e.g., a locally advanced or metastatic tumor) that has no standard therapy.

[0084] In some embodiments, the subject has undergone prior therapy. In some embodiments, the subject received treatment with a platinum -based chemotherapy, immune checkpoint inhibitor (e.g., PD-1/PDL1 immunotherapy), radiation therapy, or a combination thereof, prior to treatment with 2-S rimantadine, or pharmaceutically acceptable salt thereof. [0085] Optimal dosages of pure 2-S rimantadine, or pure 2-R rimantadine or pharmaceutically acceptable salt thereof, to be administered to a subject can be determined by those skilled in the art, and will vary with the mode of administration, the strength of the preparation, the mode of administration, and the advancement of the disease condition. In some embodiments, a subject can be administered a dosage of 2-S rimantadine, or pharmaceutically acceptable salt thereof, of about 0.01 to 10,000 mg per 25 adult human per day. For example, a pharmaceutical composition comprising pure 2-S rimantadine, or pure 2- R rimantadine, or racemic rimantadine, or pharmaceutically acceptable salt thereof, can be formulated to provide a dosage of about 0.01, about 0.05, about 0.1, about 0.5, about 1, about 2.5, about 5, about 10, about 15, about 25, about 50, about 100, about 150, about 200, about 250 or about 500 milligrams of rimantadine, or pharmaceutically acceptable salt thereof. In some embodiments, an effective amount of pure 2-S rimantadine, pure 2-R rimantadine or pharmaceutically acceptable salt thereof, can be provided at a dosage level of about 0.1 mg/kg to about 1000 mg/kg of body weight per day, or any range therein. For example, about 0.5 to about 500 mg/kg of body weight per day, about 1.0 to about 250 mg/kg of body weight per day, about 0.1 to about 100 mg/kg of body weight per day, 0.1 to about 50.0 mg/kg of body weight per day, 15.0 mg/kg of body weight per day, or about 0.5 to about 7.5 mg/kg of body weight per day. Pure 2-S rimantadine, or pure 2-R rimantadine or pharmaceutically acceptable salt thereof, can be administered to a subject on a regimen of 1 to 5 times per day or in a single daily dose.

[0086] In one aspect, the compounds disclosed herein are used in the preparation of medicaments for the treatment of diseases or conditions described herein. In addition, a method for treating any of the diseases or conditions described herein in a subject in need of such treatment, involves administration of pharmaceutical compositions that include at least one compound disclosed herein or a pharmaceutically acceptable salt, active metabolite, prodrug, or solvate thereof, in therapeutically effective amounts to said subject.

[0087] In certain embodiments, the compositions containing the compounds disclosed herein are administered for prophylactic and/or therapeutic treatments. In certain therapeutic applications, the compositions are administered to a patient already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest at least one of the symptoms of the disease or condition. Amounts effective for this use depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and the judgment of the treating physician. Therapeutically effective amounts are optionally determined by methods including, but not limited to, a dose escalation clinical trial. [0088] In prophylactic applications, compositions containing the compounds disclosed herein are administered to a patient susceptible to or otherwise at risk of a particular disease, disorder or condition.

[0089] In certain embodiments, the dose of drug being administered may be temporarily reduced or temporarily suspended for a certain length of time (i.e., a “drug holiday”).

Methods of Detection of HPV

[0090] Another aspect of the present disclosure comprises methods of treating cancer in a subject, the method comprising detecting in a sample from the subject a cancer cell that expresses a HPV protein and then administering to the subject a therapeutically effective amount of any one of the pharmaceutical compositions described herein. The detection methods described herein are based on determining the presence or absence of an HPV protein or of a functionally equivalent variant thereof. In some embodiments, wherein the presence of an HPV protein or of a functionally equivalent variant thereof is detected in a sample from the subject, the expression level of the HPV protein is determined. In some embodiments, the HPV protein is HPV16 E5. In some embodiments, the pharmaceutical composition comprises at least one additional therapeutic agent selected from one or more additional anti-cancer therapies or therapeutic agents (e.g., chemotherapeutic agents).

[0091] Thus, in another aspect, the present disclosure relates to an in vitro method for the diagnosis of diseases associated the presence of an HPV protein in a subject or for determining the predisposition of a subject to suffer from said disease associated with the presence of an HPV protein, or for determining the stage or severity of said disease associated with the presence of an HPV protein in a subject, or for monitoring the effect of the therapy administered to a subject with said disease associated with the presence of an HPV protein, which comprises quantifying the expression levels of an HPV protein or of a functionally equivalent variant thereof in a biological sample from said subject, wherein an increase of the expression of the gene encoding an HPV protein or of a functionally equivalent variant thereof, with respect to the expression levels of the gene encoding an HPV protein or of a functionally equivalent variant thereof in a control sample, is indicative of a disease associated with the presence of an HPV protein, or of a greater predisposition of said subject to suffer from a disease associated with the presence of an HPV protein or of the non-response to the therapy administered to said subject. In some embodiments, the HPV protein is HPV16 E5. In some embodiments, the pharmaceutical composition comprises at least one additional therapeutic agent selected from one or more additional anti- cancer therapies ortherapeutic agents (e.g., chemotherapeutic agents).

[0092] Therefore, as it is used herein the term “functionally equivalent variant” also includes any functionally equivalent fragment of said marker proteins. The term “fragment” relates to a peptide comprising a portion of said marker protein. In this case, a functionally equivalent fragment is a peptide or protein comprising a portion said marker protein and having essentially the same functions as said protein. “Marker protein” preferably refers to an HPV protein, without being limited thereto.

[0093] As will be understood by the persons skilled in the art, the detecting normally may not be correct for 100% of the subjects, although it is preferably is. However, the term requires being able to identify a statistically significant part of the subjects as possessing enough quantity of the protein -of-interest such that the subject suffers from a disease associated with the presence of the protein -of-interest or has a predisposition to same. The person skilled in the art can determine if a part is statistically significant by simply using one or several well-known statistical evaluation tools, for example, determination of confidence intervals, determination of the p-value, Student's t-test, Mann-Whitney test, etc. The details are in Dowdy and Wearden, Statistics for Research, John Wiley & Sons, New York 1983. The preferred confidence intervals are at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%. The p-values are preferably 0.2, 0.1, 0.05.

[0094] As used herein, the term “predisposition” means that a subject has still not developed the disease or any of the symptoms of the disease mentioned above or other diagnostic criteria but will, however, develop the disease in the future with a certain probability. Said probability will be significantly different from the statistical probability of onset of a disease associated with the presence of an HPV protein. It is preferably diagnosed that the probability of developing a disease associated with the presence of an HPV protein is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or 100% of a predisposition. The diagnosis of a predisposition can sometimes be referred to as prognosis or prediction of the probability of a subject developing the disease. [0095] In the context of the present disclosure, “control sample” is understood as the reference sample which is used to determine the variation of the expression levels of the genes and proteins used in the present disclosure. In an embodiment, the reference value is obtained from the provided signal using a sample of tissue obtained from a healthy individual. Preferably, samples are taken from the same tissue of several healthy individuals and combined, such that the amount of polypeptides in the sample reflects the mean value of said molecules in the population.

[0096] Thus, in a particular embodiment of the present disclosure, the expression levels of an HPV protein can be quantified. In some embodiments, the HPV protein is HPV16 E5. [0097] As is understood by the person skilled in the art, the expression level of a protein can be quantified by means of any conventional method. By way of non -limiting illustration, the levels of protein can be quantified, for example, by means of the use of antibodies with the capacity to bind to said proteins (or to fragments thereof containing an antigenic determinant) and the subsequent quantification of the complexes formed. The antibodies which are used in these assays may or may not be labeled. Illustrative examples of markers which can be used include radioactive isotopes, enzymes, fluorophores, chemiluminescent reagents, enzyme substrates or cofactors, enzyme inhibitors, particles, dyes, etc. There is a large variety of known assays which can be used in the present disclosure which use non-labeled antibodies (primary antibody) and labeled antibodies (secondary antibody); these techniques include Western -blot, ELISA (enzyme-linked immunosorbent assay), RIA (radioimmunoassay), competitive EIA (competitive enzyme immunoassay), DAS-ELISA (double-antibody sandwich ELISA), immunocytochemical and immunohistochemical techniques, techniques based on the use of biochips or microarrays of proteins which include specific antibodies or assays based on colloidal precipitation in formats such as dipsticks. In another particular embodiment, the quantification of the levels of protein is performed by means of an immoanalytical method, such as Western blot, immunohistochemistry or ELISA. In some embodiments, said immunoanalytical method comprises the antibody specific to HPV16 E5.

[0098] Likewise, the detection method of the present disclosure can be applied to any of the diseases associated with the presence of anHPV protein defined above. In a preferred embodiment, the disease associated with the presence of an HPV protein is a cancer, preferably a cancer having high levels of an HPV protein. In some embodiments, the HPV protein is HPV 16 E5.

[0099] Putting the method of the present disclosure into practice comprises obtaining a biological sample from the subject to be studied. Illustrative non-limiting examples of said samples include different types of biological fluids, such as blood, serum, plasma, cerebrospinal fluid, peritoneal fluid, feces, urine and saliva, as well as samples of tissues. The samples of biological fluids can be obtained by any conventional method like the samples of tissues; by way of illustration said samples of tissues can be samples of biopsies obtained by surgical resection.

[0100] In another aspect, the present disclosure relates to a kit comprising reagents for the quantification of the expression levels of an HPV protein or of a functionally equivalent variant thereof, for the diagnosis of cancer in a subject or for determining the predisposition of a subject to suffer from said cancer, or for determining the stage or severity of said cancer in a subject, or for monitoring the effect of the therapy administered to a subject with said cancer, in which if the reagents detect an increase in the expression of said gene or said protein or functionally equivalent variant thereof with respect to a control sample, then said subject can suffer from a disease associated with the presence of an HPV protein, or present a greater predisposition to suffer from said disease associated with the presence of anHPV protein, or present a greater severity of said disease, or the administered therapy is not being effective. In some embodiments, the HPV protein is HPV16 E5. In some embodiments, the pharmaceutical composition comprises at least one additional therapeutic agent selected from one or more additional anti-cancer therapies or therapeutic agents (e.g., chemotherapeutic agents).

[0101] The present disclosure also relates to the use of said kit. [0102] All the terms and expressions used in the definition of the use of the kit have been described above and explained for other inventive aspects and particular embodiments of the present disclosure, and are also applicable to the use of the kit described herein. Methods for Designing Customized Therapies and for Selecting Patients who can Benefit from Administration of 2-S Rimantadine or 2-R Rimantadine [0103] In another aspect, the present disclosure relates to an in vitro method for designing a customized therapy fora patient suffering from a disease associated with the presence of an HPV protein comprising:

(a) quantifying the expression levels of an HPV protein in said patient, and

(b) comparing said expression levels with control levels, wherein if the expression levels of an HPV protein in said patient are greater than the control values, then a therapeutically effective amount of 2-S rimantadine or a pharmaceutically acceptable salt thereof, or 2 -A rimantadine or pharmaceutically acceptable salt thereof, is administered to said patient.

[0104] In some embodiments, the HPV protein is HPV16 E5. In some embodiments, at least one additional therapeutic agent selected from one or more additional anti -cancer therapies or therapeutic agents (e.g., chemotherapeutic agents) is administered to the patient.

[0105] In another aspect, the present disclosure relates to an in vitro method for selecting patients suffering from a disease associated with the presence of an HPV protein, to be treated with a therapeutically effective amount of 2-S rimantadine or a pharmaceutically acceptable salt thereof, or 2 -A rimantadine or pharmaceutically acceptable salt thereof comprising a) quantifying the expression levels of an HPV protein in said patient, and b) comparing said expression levels with control levels, wherein if the expression levels of an HPV protein in said patient are greater than the control values, then said patient is selected to receive a therapeutically effective amount of 2- S rimantadine or a pharmaceutically acceptable salt thereof, or 2-R rimantadine or pharmaceutically acceptable salt thereof.

[0106] In some embodiments, the HPV protein is HPV16 E5. In some embodiments, at least one additional therapeutic agent selected from one or more additional anti -cancer therapies or therapeutic agents (e.g., chemotherapeutic agents) is administered to the patient. EXAMPLES

EXAMPLE-1. ScreenPatch® Assay on NR1/NR2A and NR1/NR2B

[0107] ScreenPatch

• NR1/NR2A ionotropic receptor encoded by the human GRIN1/GRIN2A genes, expressed in HEK293 cells.

• NR1/NR2B ionotropic receptor encoded by the human GRIN1/GRIN2B genes, expressed in HEK293 cells

Formulation

[0108] 2-S rimantadine, 2 -R rimantadine, racemic rimantadine and amantadine solutions were prepared daily and prepared by diluting stock solutions into an appropriate HEPES- buffered physiological saline (HB-PS) solution. Because 0.6% DMSO does not affectchannel current, all test and control solutions contained 0.6% DMSO. Test article formulations were sonicated (Model 2510/5510, Branson Ultrasonics, Danbury, CT), at room temperature for at least 20 minutes to facilitate dissolution.

[0109] Test article effects were evaluated in 8-point concentration-response format (4 replicate wells/concentration). All test and control solutions contained 0.6% DMSO. The test article formulations were loaded in a 384-well compound plate using an automated liquid handling system (Assist Plus, INTEGRA).

Positive Control Treatment Groups

[0110] Stock solutions of positive control articles were prepared in batches, aliquoted for individual use, stored frozen, and were used within six months. The positive control test solutions were prepared fresh daily. The final DMSO concentration was 0.6% (v/v). The NMD A receptors agonists, L-glutamate and glycine were used as a reference agonist in this study. The NMDA receptors antagonist, amantadine, was used as a reference antagonist in this study.

Testing and Concentrations:

[0111] Test articles were evaluated for functional effects on ion channels. Test concentrations are shown in Table 1 below. Table L Test Articles Concentrations

Cloned Test Systems

[0112] Cells were maintained in tissue culture incubators. Stocks were maintained in cryogenic storage. Cells used for electrophysiology were plated in plastic culture dishes.

HEK293 Culture Procedures

[0113] HEK293 cells were transfected with the appropriate ion channel or receptor cDNA(s) encoding NR1 and NR2A-B. Stable transfectants were selected using the G418 and Zeocin-resistance genes incorporated into the expression plasmid. Selection pressure was maintained with 0418 and Zeocin in the culture medium. Cells were cultured in Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 (D-MEM/F-12) supplemented with 10% fetal bovine serum, 100 Ci/mL penicillin O sodium, 100pg/mL streptomycin sulfate, 100 ug/mL Zeocin, 5 ug/mL blasticidin and 500 pg/mL 0418.

ScreenPatch Test Methods:

[0114] All experiments were performed at ambient temperature. Target-specific test procedures are described below. The following procedures apply to all ScreenPatch assays. [0115] Before testing, cells in culture dishes were washed twice with HBS8 solution. Immediately before use in lonWorks Barracuda™, the ceils were washed in HB-PS containing 6 mM CaCh to improve sealing.

[0116] Test articles were evaluated in 8 -point concentration-response format (4 replicate wells/concentration, see Table 1). Previous results have shown that 0,6% DMSQ does not affect channels currents; thus, unless specified otherwise, all test and control solutions contained 0.6% DMSO. The test article formulations were loaded in a 384 -well compound plate and placed in the lonWorks Barracuda ™ plate well. [0117] Positive control articles were prepared in batches, aliquoted for individual use, stored frozen, and used within six months. The positive control test solutions were prepared fresh daily. The final DMSO concentration was 0.6%.

[0118] 2X concentration of test article (as specified in Table 2) was pre-applied 2 minutes before application of L-glutamate (Sigma- Aldrich)/ glycine (Sigma-Aldrich (5mM L- glutamate and 50 μM glycine) mixed with IX concentration of test article.

[0119] To monitor the sensitivity of the assay, the antagonist positive control, amantadine hydrocholoride (Sigma-Aldrich) w as applied at 8 half log concentrations (range 0.3-1000 μM); n = 4, where n = the number of replicates per concentration. The agonist positive control (L-glutamate) was applied at eight (8) concentrations (0.03 - 100 μM; n = 4, where n = the number of replicates) together with 50 μM glycine,

[0120] Inhibitory effects of compounds on the channels were calculated as: where Base is the response at low concentrations of test article, Max is the maximum response at high concentrations, xhalf the EC 50 , or IC 50 , the concentration of test article producing either half-maximal activation or inhibition, and rate is the Hill coefficient. Nonlinear least squares fits weremadeassumingasimplebindingmodel. If appropriate, fits were weighted by the standard deviation. No assumptions about the fit parameters were made; the fit parameters were determined by the algorithm.

[0121] Nonlinear least, squares fits were solved with the XLf it add-in for Excel 2016 (Boston, MA).

[0122] Effects of the compounds were evaluated with two types of measurements: 1 . Peak current, amplitude (PCA) measurements at current maximum.

2. Steady state current amplitude (SSC) measurements, as mean between 4 and 5 seconds following stimulation of receptors with agonist.

Procedures

[0123] Electrophysiological Procedures: a) Intracellular solution (mM): 50 mM CsCl, 90 mM CsF, 2 mM MgCl 2 , 5 mM

EGTA, 10 mM HEPES. Adjusted to pH 7.2 with CsOH. This solution was prepared in batches and stored refrigerated. In preparation for a recording session, the intracellular solution was loaded into the intracellular compartment of the PPC planar electrode. b) Extracellular solution, HB-PS (composition in mM): NaCl, 137; KCl, 1.0;

CaCl 2 , 5; HEPES, 10; Glucose, 10; pH adjusted to 7.4 with NaOH (refrigerated until use). c) Holding potential: -70 mV, potential during test articles application: -70 mV.

[0124] Recording procedure: a) Extracellular buffer was loaded into the PPC plate wells (11 μL per well). Cell suspension was pipetted into the wells (9 μL, per well) of the PPC planar electrode. b) Whole-cell recording configuration was established via patch perforation with membrane currents recorded by on-board patch clamp amplifiers. c) Two recordings (scans) were performed. First scan, during test articles addition at 2X concentration to detect potential agonist effect and for preincubation of test articles with cells (for 2 minutes). Second, during agonist stimulation of receptors (5 μML, - glutamate and 50 μM glycine) co-applied with IX concentration of test articles to detect antagonist effects of the test articles.

[0125] Test article administration: The application consisted of the addition of 20 μL, of 1X concentrated test article solution and agonist at 10 μL/s (2 second total application time). [0126] Positive control agonist: 0,03 - 100 μM L-glutamate (8 concentration dose- response, half log scale) and 50 μM glycine

[0127] Positive control antagonist: 0.3 - 1000 μM amantadine (8 concentration - response, half log scale) co-applied with 5 mM glutamate and 50 μM glycine.

[0128] Evaluation of effects were based on peak current measurements.

Results

[0129] Agonist and antagonistproperties of four (4) compounds were examinedusingan HTS electrophysiology -based approach, Ion Work Barracuda™ (IWB). A two-application protocol was employed.

[0130] Agonist Format: The potential agonist effect of the test articles and the positive control antagonist, amantadine, were examined during first application. Neither test articles nor amantadine had produced significant activation of NMD A receptors (data not shown)

[0131] AntagonistFormat: The antagonist activity of test articles was examined during second application of compounds after stimulation of receptors with 5 μML-glutamate and 50 μM glycine. It was found that all four test articles produced significant concentration dependent inhibition of NMDA receptors function. To access open channel blocktype of inhibition, peak and steady state current amplitudes (between 4 th and 5 th seconds after agonist application) were measured (PCA and SSC respectively). Table 2 shows an average of compounds’ IC 50 at NR1/NR2A and NR1/NR2B receptors with these two types of measurements.

[0132] Amantadine produced inhibition of NR1/NR2A receptors with IC 50_PCA = 97.8μM and IC 50_ssc = 48.9μM for peak and steady state currents amplitudes respectively. NR1/NR2B receptors were inhibited by amantadine with IC 50 PCA= 22.0μMand IC 50_ssc = 17.9μM for peak and steady state currents amplitudes respectively . Leftward shiftin amantadinepotency for steady state currents measurements suggests, atleastin part, open channel block mechanism of inhibition for NR1/NR2A NMD A receptors.

Table 2. Summary of inhibition IC 50 produced by test articles and reference antagonist, amantadine.

[0133] Amantadine produced inhibition ofNRl/NR2A receptors with IC 50 PCA = 97.8μM and IC 50 ssc = 48.9μM for peak and steady state currents amplitudes respectively. NR1/NR2B receptors were inhibited by amantadine with IC 50_PCA = 22.0μM and IC 50_ssc = 17.9μM for peak and steady state currents amplitudes respectively. Leftward shiftin amantadinepotency for steady state currents measurements suggests, atleastin part, open channel block mechanism of inhibition for NR1/NR2A NMD A receptors.

[0134] The results of these assays are further exemplified in FIG. 1A-D.

EXAMPLE 2. Effect of pure 2-S rimantadine or pure 2-R rimantadine in mouse cancer models.

Methods

[0135] Cell lines

[0136] AT-84-E7 and B16-OVA are grown in RPMI 1640 containing 10% FBS, 1% L- flutamine, 1% penicillin/streptomycin, 1% sodium pyruvate, and 200 μg/ml G418. DC2.4, RAW264.7, B3Z, 4T1, B16 and MC38 are grown in RPMI 1640 20 containing 10% FBS, 1% L-glutamine, 1% penicillin/streptomycin, and 1% sodium pyruvate. HEK293T is grown in DMEM containing 10% FBS, 1% L-glutamine, and 1% penicillin/streptomycin. 4MOSC1 is cultured in collagen-coated dish with KSFM media (Invitrogen, Carlsbad, 20 CA) supplemented 1% penicillin/streptomycin, 5 ng/ml EGF (Invitrogen), and 2 x 10-11 M cholera toxin (Sigma, St. Louis, MO)(27). CAL-27, CAL-33, and SCC-47 are grown in DMEM containing 10% FBS, 22 1% L-glutamine, and 1% penicillin/streptomycin. Routine monitoring for Mycoplasma contamination is performed using the MycoAlert PLUS Detection Kit (Lonza, Basel, Switzerland). All cell lines are used within ten passages after thawing.

[0137] Mouse Studies

[0138] Mice are injected subcutaneously with 1.0 to 5.0 x 10 5 AT-84-E7, 1.5 x 10 5 BIO- OVA, or 5.0 x 10 5 4T1 cells are resuspended in 100 pi of PBS in the right flank. For orthotopic models, 1.0 x 10 5 AT-84-E7 or 1.0 x 10 6 4MOSC1 in 30 μl of PBS are injected into tongue. Tumor diameter is measured every 2 to 3 days with an electronic caliper and reported as volume using the formula; tumor volume (mm 3 ) = (length x width 2 )/2. Once tumors are palpable, mice are treated with 200 μg ofanti-PDL1 antibody (BioXcell, West Lebanon, NH) via IP injection every 3 days for a total of three or four injections per mouse, or 5 mice are treated with 10-20 mg/kg body weight of pure 2-S rimantadine or pure 2-R rimantadine via IP injection daily for 7 days. For adoptive transfer experiments, first, single-cell suspension of spleen from OT-1 mice are cultured in media containing 10 ng/ml OVA SIINFEKL peptide (InvivoGen, San Diego, CA) and 2 ng/ml recombinant IL-2 (PeproTech, Rocky Hill, NJ) for days, and then 4.0 x 10 6 cells are intravenously injected into B 16-0 VA-b earing mice.

[0139] Flow cytometry

[0140] Single-cell suspensions are prepared from, lung, liver, tumor-draining lymph nodes, and tumors by mechanical dissociation and are filtered using a 70 μg filter. AT-84- E7 and MOC2 tumors are incubated in collagenase D (Roche, Basel, Switzerland) at 37°C for 1 hour prior to mechanical dissociation. Density gradient centrifugation on 40%/80% Percoll (GE Healthcare, Chicago, IL) gradient is performed for single-cell suspension from tumors. After obtaining single-cell suspensions, each sample is incubated with an Fc blocking reagent (anti-CD16/32 antibody; BioLegend, San Diego, CA). Following Fc blockade, cells are stained with fluorescent-labeled antibodies [BioLegend, BD Bioscience (San Jose, CA), or eBiosciences (Thermo Fisher Scientific, Waltham, MA)]. LIVE/DEAD Fixable Cell Staining Kit (Invitrogen) is used for viability staining. For intracellular staining, cells were processed with Foxp3 / Transcription Factor Fixation/Permeabilization Concentrate and Diluent (Invitrogen). Cells are analyzed using a BD FACS Aria II or LSR II flow cytometer (BD). Data is analyzed on FlowJo (FlowJo, LLC, Ashland, OR). For each antibody, the following clones are used: CD45.2 (104), CD3e (145- 2C11), CD4 (RM4-5), CD8a (5H10), CD25 (3C7, PC61), CD44 (IM7), CD62L (MEL-14), IFN-g (XMG1.2), Foxp3 (MF23), H- 2Kb (AF6-88.5), H-2Kk (36-7-5), H-2Kd (SFl-1.1), H-2Kb/SIINFEKL (eBio25-D1.16), I- A/I-E (2G9), CD49b (DX5), CD 11b (Ml/70), FLAG (L5), CD31 (MEC13.3), NK-T/NK Cell Antigen (U5A2-13), CD102 (3C4 (MIC2/4)), CD62P (RMP-1), CD105 (MJ7/18), CD106 (429 (MVCAM.A)), and CD 162 (2PH1). H-2Kb/SIINFEKL tetramer was purchased from MBL International (Woburn, MA).

[0141] Cell cycle and proliferation assays

[0142] Cell cycle progression was analyzed on the basis of BrdU incorporation following 1 cell staining with BrdU-APC and 7-AAD using BD Pharmingen BrdU Flow Kit. (BD, Franklin Lakes, NJ) according to the manufacture’s protocol. Cells are analyzed using flow cytometry. Cell proliferation is assessed by using MTT [3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide]. First, cells are seeded in 96-well plate and cultured for 2-3 days. Next, culture media is replaced with freshmedia containing 0.5 mg/ml of MTT (Sigma) and the plates are incubated for 4 hours at 37°C. Then, purple formazan crystals are dissolved in lysis buffer (4 mM HCl and 0.1% NP-40 in isopropanol) and the absorbance is recorded on a TECAN infiniteM200 microplate reader (Tecan, Mannedorf, Switzerland) at a wavelength of 570 nm with absorbance at 650 nm as reference.

[0143] B3Z activation assay

[0144] B 16-OVA cells are seeded into a 96-well plate and treated with 100 μM pure 2 -R rimantadine, pure 2-R rimantadine or racemic rimantadine for24 hours, priorto addition of B3Z cells. After 24 hours of co-culture, medium is removed and 100 pi of lysis buffer [0.155 mM chlorophenol red β-D-galactopyranoside (CPRG) (Roche), 0.125% Nonidet P-40 Alternative (EMDCalbiochem), and 9 mM MgCl 2 (Sigma) in PBS] are added. After incubation for 4 hours at 37°C, the absorbance at 570 nm is determined on a TECAN infinite M200 microplate reader.

[0145] Reverse transcription and quantitative PCR

[0146] Total RNA is extracted using TRIzol Reagent (Invitrogen) and reverse transcribed with qScript cDNA Synthesis Kit (Quanta BioSciences, Beverly, MA) according to the manufacturer’s instructions. Quantitative PCR analysis is conducted by using KAPA SYBR 1 FAST (KAPA Biosystems, Wilmington, MA) on the 7900HT Fast Real-Time PCR System (Applied Biosystems, Foster City, CA). [0147] Results

[0148] Pure 2-S Rimantadine and pure 2-R rimantadine have anti-tumor activity alone and significantly decreased tumor growth. Six mice are inoculated with 5 x 10 5 AT84-E7/E5 tumor cells and treated with intraperitoneal (IP) injections of 10 mg/kg body weight pure 2- S rimantadine or pure 2-R rimantadine once daily fora total of 7 injections starting on day 8. The tumor volumes are measured over the course of the experiment. Mice that receive pure 2- S rimantadine or pure 2-R rimantadine show statistically significant decreases in tumor size compared to control groups. Six mice are inoculated with 1.5 x 10 5 B16-OVA tumor cells and treated with IP injections of 10 mg/kg body weight pure 2-S rimantadine or pure 2-R rimantadine once daily for a total of 7 injections starting on day 10. The tumor volumes are measured over the course of the experiment. Mice that receive pure 2-S rimantadine or pure 2-R rimantadine show statistically significant decreases in tumor sizes compared to control groups. This experiment is repeated three times with similar results. Five mice are inoculated with 5 x 10 5 4T1 tumor cells and treated with IP injections of 10 mg/kg body weight pure 2- S rimantadine or pure 2-R rimantadine once daily fora total of 7 injections starting on day 6. The tumor volumes are measured over the course of the experiment. Mice that receive 2-S rimantadine or pure 2-R rimantadine show statistically significant decreases in tumor sizes compared to control groups. The anti-tumor effect of pure 2-S rimantadine or pure 2-R rimantadine is decreased in AT-84-E7 tumors which do not express E5. Significant increases in surface expression of MHC is observed in multiple cell lines. Cell surface expression of MHC I on E5-positive AT-84-E7 is restored with pure 2-S rimantadine or pure 2-R rimantadine treatment.

[0149] To test the ability of 2-S rimantadine to enhance functional antigen presentation on tumor cells, B16 cells expressing OVA are used as a model tumor antigen and coculture with B3Z cells which respond to OVA SINNFKL peptide. Treatment of B16-OVA cells with pure 2-S rimantadine or pure 2-R rimantadine results in a significant 3 -fold increase in recognition of this model tumor antigen by B3Z cells. Pure 2-S rimantadine or pure 2-R rimantadine with anti-PDLl immunotherapy results in a significant improvement in survival in mice harboring B 16-OVA tumors.

[0150] The ability for pure 2-S rimantadine or pure 2-R rimantadine to increase expression of MHC on antigen presenting cells using the RAW264.7 cell line is tested and significant increases in both MHC class I and MHC class II surface expression is observed. These findings demonstrate that pure 2-S rimantadine or pure 2-R rimantadine has novel anti-tumor activity in multiple pre-clinical tumor models and functions to enhance antigen presentation by upregulating MHC.

[0151] To study the direct cytotoxic activity of pure 2 -S rimantadine or pure 2-R rimantadine, in vitro BrdU incorporation assays are performed to quantify the effects of pure 2-S rimantadine or pure 2-R rimantadine on cell cycling in human HNSCC cell lines. Pure 2- S rimantadine or pure 2-R rimantadine alone result in significant increases in G0/G1 cell cycle arrest and significant decreases in S phase in both AT-84-E7 and B 16-OVA models. Suppression of cell proliferation is also observed. Analysis of the effect of pure 2-S rimantadine or pure 2-R rimantadine on proliferation of T cells is tested, but there are no significant effects.

[0152] Changes in gene expression of cell cycle proteins caused by pure 2-S rimantadine or pure 2-R rimantadine are screened using RTqPCR and significant decreases in microtubule and cell cycle regulatory molecule Stathmin is seen. Decreases in microtubule associated molecule Tau is also observed.

[0153] To confirm pure 2-S rimantadine or pure 2-R rimantadine has activity against human head and neck tumor lines, BrdU incorporation assays and proliferation assays are performed. Significant cell cycle arrest and decreased proliferation with rimantadine alone is observed in the human CAL-27, CAL-33, and SCC-47 squamous cell carcinoma cell lines. Finally, pure 2-S rimantadine or pure 2-R rimantadine induce cell cycle arrest in murine and human cell lines engineered to express HPV16 E5, indicating that pure 2-S rimantadine or pure 2 -A rimantadine is able to functionally reverse effects of HPV E5.

EXAMPLE 3. HPV genotyping

[0154] HPV genotyping is known in the art, for example, see Sichero et al., 2017, Cancer Epidemiol Biomarkers , 26(8):1312-1320. For example, DNA is extracted from exfoliated cervical cells by spin-column chromatography. Mucosal alpha-HPVs are tested using PCR amplification with primers such as MY09/11 and PGMY09/11 (see Table 3) followed by genotyping via hybridization with HPV type-specific oligonucleotide probes and restriction fragment length polymorphism analysis. Negative and positive controls are used to ascertain the quality of template DNA. Table 3- primer sequences

The degenerate base code is as follows: M = A or C, W = A or T, Y = C or T, and R = A or G.

EXAMPLE 4. 2-S RIMANTADINE AND 2-R RIMANTADINE TOXICOLOGY IN VIVO

[0155] A series of in-vivo experiments are performed to determine whether 2-S rimantadine or 2-R rimantadine have higher binding selectivity for any one of glutamate, GABA, dopamine receptors, or any combination thereof. Enhanced selectivity of any one of glutamate, GABA, dopamine receptors, or any combination thereof by 2-S rimantadine or 2-R rimantadine compared to racemic rimantadine results in the absence of central nervous system adverse effects including nausea, upset stomach, vomiting, anorexia, dry mouth, abdominal pain, asthenia, nervousness, tiredness, lightheadedness, dizziness, headache, trouble sleeping, difficulty concentrating, confusion and anxiety, commonly associated with racemic rimantadine.

[0156] To test the above, mice are treated with 10-20 mg/kg body weight of pure 2-S rimantadine, pure 2-R rimantadine, racemic rimantadine (control), or amantadine (control) via IP injection daily for 7 days. Next, a series of SPECT analyses as described in Schramm, N., et al. (2000). Compact high resolution detector for small animal SPECT, are conducted for each of glutamate, GABA, dopamine receptors to assess the binding selectivity of 2-S rimantadine and 2-R rimantadine.

[0157] The SPECT analyses comprise of treatment of the mice with radioligands specific to each receptor. For example, [ 123 I] IBZM has been documented to have a high affinity for the D 2/3 dopamine receptor. Radioligands specific to glutamate and GABA receptors are known to those skilled in the art. The appropriate amount of the respective radioligands for each of glutamate, GABA, dopamine receptors are injected into the lateral tail vein of the mice and SPECT measurements commence 45 mins after radioligand administration. [0158] Surprisingly, 2 -R rimantadine has significantly higher binding selectivity or agonistic behavior to glutamate, GABA, dopamine receptors or pathways, or any combination thereof as compared to 2-S rimantadine. Thus, 2-R rimantadine results in a higher incidence of central nervous system adverse effects including nausea, upset stomach, vomiting, anorexia, dry mouth, abdominal pain, asthenia, nervousness, tiredness, lightheadedness, dizziness, headache, trouble sleeping, difficulty concentrating, confusion and anxiety, as compared to 2-S rimantadine. Taken together with Example 2, 2-S rimantadine is significantly less toxic, while still effective, as a treatment for cancer as compared to 2-R rimantadine.

EXAMPLE 5. 2-S RIMANTADINE AND 2-R RIMANTADINE TOXICOLOGY IN VIVO

[0159] A series of in-vivo experiments are performed to determine whether 2-S rimantadine or 2-R rimantadine have higher binding selectivity for any one of glutamate, GABA, dopamine receptors, or any combination thereof. Enhanced selectivity of any one of glutamate, GABA, dopamine receptors, or any combination thereof by 2-S rimantadine or 2-R rimantadine compared to racemic rimantadine results in the absence of central nervous system adverse effects including nausea, upset stomach, vomiting, anorexia, dry mouth, abdominal pain, asthenia, nervousness, tiredness, lightheadedness, dizziness, headache, trouble sleeping, difficulty concentrating, confusion and anxiety, commonly associated with racemic rimantadine.

[0160] To test the above, mice are treated with 10-20 mg/kg body weight of pure 2-S rimantadine, pure 2-R rimantadine, racemic rimantadine (control), or amantadine (control) via IP injection daily for 7 days. Next, a series of SPECT analyses as described in Schramm, N., et al. (2000). Compact high-resolution detector for small animal SPECT, are conducted for each of glutamate, GABA, dopamine receptors to assess the binding selectivity of 2-S rimantadine and 2-R rimantadine.

[0161] The SPECT analyses comprise of treatment of the mice with radioligands specific to each receptor. For example, [ 123 I] IBZM has been documented to have a high affinity for the D 2/3 dopamine receptor. Radioligands specific to glutamate and GABA receptors are known to those skilled in the art. The appropriate amount of the respective radioligands for each of glutamate, GABA, dopamine receptors are injected into the lateral tail vein of the mice and SPECT measurements commence 45 mins after radioligand administration.

[0162] Surprisingly, 2-S rimantadine has significantly higher binding selectivity or agonistic behavior to glutamate, GABA, dopamine receptors or pathways, or any combination thereof as compared to 2-R rimantadine. Thus, 2-S rimantadine results in a higher incidence of central nervous system adverse effects including nausea, upset stomach, vomiting, anorexia, dry mouth, abdominal pain, asthenia, nervousness, tiredness, lightheadedness, dizziness, headache, trouble sleeping, difficulty concentrating, confusion and anxiety, as compared to 2 -R rimantadine. Taken together with Example 2, 2-S rimantadine is significantly less toxic, while still effective, as a treatment for cancer as compared to 2-R rimantadine.

EXAMPLE 6. 2-S RIMANTADINE AND 2-R RIMANTADINE PROLIFERATION IN VITRO

[0163] Experiments were performed to determine the ability of 2-S rimantadine (also referred to as “S-rimantadine” throughout the application), 2-R rimantadine (also referred to as “R-rimantadine” throughout the application), racemic (RS) rimantadine, and memantine to effect proliferation in CAL-27 cells. S-rimantadine results in enhanced or equivalent cancer cell proliferation as compared to R-rimantadine or racemic rimantadine.

[0164] On day 1, CAL-27 cells were seeded in a 96 -well plate (2 - 4 x 10 3 cells/well, medium 100 μl/well) and left overnight to allow the cells to attach to the plate. On day 2, varying concentrations of rimantadine (0 μM, 100 μM, 250 μM, or 500 μM) were added to the cells and allowed to incubate for 24 hours or 48 hours. On either day 3 or 4, the culture media was aspirated, and 100 μl/well of MTT solution (comprising an MTT concentration of 0.5mg/ml; formed by dilution of thiazolyl blue tetrazolium bromide solution (SIGMA, Cat#

M2 128) with stock solution (5 mg/ml in PBS (-20°C)) was added to the culture media. The cells were incubated in a CO 2 incubator at 37°C for 3 hours, and the MTT solution was aspirated. 100 μl /well of DMSO was then added and the cells were incubated for about 5 minutes. A OD570 nm (Ref 650 nm) was then read. Results of the experiment are shown in FIG. 2.

EXAMPLE 7. IN-VIVO TUMOR MODEL/ANTI-TUMOR ACTIVITY METHODS

[0165] The activity of 2-S rimantadine, 2-R rimantadine, and racemic rimantadine will be tested against HPV associated tumors using in-vivo murine syngeneic tumor models. S- rimantadine will demonstrate equivalent or increased anti-tumor activity as compared to racemic rimantadine and/or R-rimantadine.

Plasmid construction and HPV16 E5-expressing stable cell line [0166] Codon-optimized HPV16 E5 will be amplified. Either C-terminal orN-terminal FLAG-tagged full-length HPV16 E5 and deletion mutants will be cloned into MIP (MSCV- IRES-Puro) or pMSCV-Blasticidin vectors. All the constructs will be confirmed by DNA sequencing. For establishing HPV16 E5 -expressing cell line, HEK293T cells will be cotransfected with MIP-HPV16 E5 and Ecopac (pIK6.1MCV.ecopac.EiTd) using PEI reagent (Sigma-Aldrich). Retroviruses from the culture medium of these cells will then be used to infect AT-84-E7, MOC2, and CAL-27 cells, and the infected cells will be selected by puromycin. pMSCVBlasticidin- HPV16 E5 will be used for MEER cells.

Mouse studies

[0167] Female 6- to 8-week-old mice will be used for experiments. C3H/HeN mice and C57BL/6 and BALB/c will be used. Mice will be injected subcutaneously with 1.0 to 5.0xl0 5 AT-84-E7, 1.5x 10 5 B 16-OVA, 5.0x10 5 4T1 , or 1.0x10 5 MOC2 cells resuspended in 100 mL of PBS in the right flank. For orthotopic models, 1.0 10 5 AT-84-E7 or 1.0 10 6 4MOSC1 in 30 mL of PBS will be injected into tongue. Once tumorsbecome palpable, mice will be treated with 200 mg of anti-PD-Ll antibody (Bio X Cell) via i.p. injection every 3 days for a total of three or four injections per mouse, or mice will be treated with 10 mg/kg body weight of R-rimantadine, S-rimantadine, and/or racemic rimantadine via i.p. injection daily for 7 days. For adoptive transfer experiments, single-cell suspension of spleen from OT-1 mice will be cultured in media containing 10 ng/mL OVA SIINFEKL peptide (InvivoGen) and 2 ng/mL recombinant IL2 (PeproTech) for 5 days, and then 4.0 10 6 cells will be intravenously injected into B16-OVA- bearing mice. Tumor diameter will be measured every 2 to 3 days with an electronic caliper and reported as volume using the formula; tumor volume (mm 3 ) (length width 2 )/2.

[0168] The information and procedures used and disclosed in Miyauchi S., etak, Cancer Res. 2020 Feb 15;80(4):732-746 are hereby incorporated by reference in their entirety. The information and procedures (e.g., protocols) disclosed will be implemented for the study of S- rimantadine, R-rimantadine, and/or racemic rimantadine.

EXAMPLE 8. IN-VITRO ANTI- VIRAL METHODS

[0169] The direct anti-viral activity of enantiomers of rimantadine (e.g., S-rimantadine) will be tested against HPV viral replication using in-vitro HPV viral replication assays. S- rimantadine will demonstrate equivalent or increased direct HPV anti-viral activity compared to racemic rimantadine or R-rimantadine.

Plasmid

[0170] Snls-Cre expression plasmid pCAGGS-nlsCre will be used. pNeo-loxPHPV-18 and pNeo-loxPHPV-18 E6*I plasmids will be used. For both plasmids, the 34-bp loxP sites will flank the linear HPV- 18 sequence upstream of nucleotide 7474 and downstream from nucleotide 7473. The vector will carry the Neomycin resistance marker gene selectable in bacteria and in mammalian cells. In the HPV-18 E6*I mutant, the intron coding sequence (nucleotides 234M15) in the predominant E6* I mRNA will be deleted. For trans- complementation experiments, the empty vector-only retrovirus pLC and pLJ HPV-18 URR-E6 orURR -E6/E7 retro- viruses will be used. Each expresses the Neomycin resistance gene (Cheng et al. 1995. Differentiation-dependent up-regulation of the human papillomavirus E7 gene reactivates cellular DNA replication in suprabasal differentiated keratinocytes. Genes & Dev. 9 : 2335-2349; Chien et al. 2002. Alternative fates of keratinocytes transduced by human papillomavirus type 18 E7 during squamous differentiation. J. Virol. 76: 2964-2972). All plasmids will be purified by banding (e.g., in CsCl-ethidium bromide equilibrium density gradients).

HPV-18 virion recovery and titer determination

[0171] HPV-18 virions will be recovered from day- 14 or day- 16 epithelia as described (Favre, M. 1975. Structural polypeptides of rabbit, bovine, and human papillomaviruses. J.

Virol. 15: 1239-1247). To titer the virus, aliquots of the virus stocks will be digested with DNase I (Invitrogen), which will then be inactivated by heating for 5 min at 100°C. Packaged viral DNA will then be purified by digestion with Proteinase K and phenol/ chloroform extractions. Serial dilutions of viral DNA will be analyzed by real-time quantitative PCR using, for example, SYBR GreenER qPCR SuperMix (Invitrogen) and primers J and K, disclosed in Supplemental Table 1 of WangHK. etal., Genes Dev. 2009 Jan 15; 23(2): 181-194. As standards, purified pNeo-LoxPHPV-18 plasmid DNA will be serially diluted to ~40 to 4 x 10 8 copies per well. Forty cycle PCR amplification reactions in triplicate will be performed (e.g., in 384-well plates usingthe ABI 7900HT). Data will then be processed (e.g., with the use of SDS2.1 software (Applied Biosystems)).

HPV-18 infectivity assays

[0172] Approximately 1 x 10 5 primary human keratinocytes (PHKs) will be inoculated with various amounts of virus stock, corresponding to an MOI of 5200, 1040, 208, 42, 10, 2, 1, or 0 in 1 mL of K-SFM and incubated overnight. The medium will be changed and the cells will be cultured for four more days. Total RNA will then be extracted (e.g., with the use of Trizol (Invitrogen)). Reverse transcription will be conducted in a 50-mL reaction on 10 mg of RNA. One microliter of RT reaction will then then subjected to 30 cycles of PCR or nested PCR amplification (30 cycles each) in a 35 -mL reaction mixture to generate a cDNA fragment of the spliced HPV-18 E6-E7-E1ʌE4, RNA, orthe b- actin mRNA, as described (Meyers et al. 2002. Infectious virions produced from a human papillomavirus type 18/16 genomic DNA chimera. J. Virol. 76: 4723-4733). Fifteen micro- liters of each reaction will be resolvedby electrophoresis in a 2% agarose gel and visualized by ethidium bromide staining. PHKs will also be infected with various MOIs in K-SFM overnight and developed into raft cultures, fixed on day 14, and processed as described.

[0173] The PHKs receiving varying amounts of virus stock will then be exposed to varying concentrations of R-rimantadine, S- rimantadine, and/or racemic rimantadine over a period of time (e.g., 1 day, 2 days, 3 days, 5 days, 7 days, and/or 10 days). The information and procedures (e.g., protocols) disclosed will be implemented forthe study of S -rimantadine, R-rimantadine, and/or racemic rimantadine.

[0174] The information and procedures used and disclosed in Wang HK. et al., Genes Dev. 2009 Jan 15; 23(2): 181-194 are hereby incorporated by reference in their entirety.

The information and procedures (e.g., protocols) disclosed will be implemented forthe study of S-rimantadine, R-rimantadine, and/or racemic rimantadine.

EXAMPLE 9. IN VIVO CENTRAL NERVOUS SYSTEM (“CNS”) ASSAYS [0175] Studies will be conducted to determine the effects of R-rimantadine, S- rimantadine, and racemic rimantadine on the CNS of living animals (e.g., mice and/or rats). Varying doses of R-rimantadine, S-rimantadine, and racemic rimantadine will be studied and the following tests. Animals receiving S-rimantadine will demonstrate less CNS toxicity at similar doses of R-rimantadine and racemic rimantadine. Additionally, animals receiving S-rimantadine will be capable of receiving higher doses of the respective agent as compared to animals receiving R- rimantadine or racemic rimantadine before exhibiting signs and/or symptoms of CNS toxicity. Additionally, mice receiving S-rimantadine will better tolerate signs and symptoms of CNS toxicity as compared to mice receiving similar doses of R-rimantadine and racemic rimantadine. a) Rotarod

[0176] CNS toxicity associated with the use of R-rimantadine, S-rimantadine, and racemic rimantadine will be studied with the use of the rotarod system (e.g., Rotor Rod System, San Diego Instruments). Use of the Rotor Rod system will allow study of the CNS toxicity potentially caused by R-rimantadine, S-rimantadine, and racemic rimantadine by allowing observation of motor coordination in animals (e.g., mice or rats).

[0177] Animals will receive doses (e.g., varying doses) of R-rimantadine, S-rimantadine, or racemic rimantadine. After a period of time (e.g., 1 hour, 2 hours, 3 hours, 5 hours, 10 hours,

1 day, 2 days, 3 days, 5 days, 7 days, and/or 10 days) after receiving a dose, the potential CNS effects will be measured with the use of the rotarod system. Animals receiving S- rimantadine will demonstrate less adverse CNS effects and toxicity when compared to R-rimantadine and racemic rimantadine. In particular, animals (e.g., mice or rats) receiving S-rimantadine will demonstrate less abnormal motor coordination. The information and procedures used and disclosed in Rotor Rod, San Diego Instruments, available at https://sandiegoinstruments.com/product/rotor-rod/; ROTOR-ROD™ System, Biomedical and Obesity Research Core, College of Education and Human Sciences, University ofNebraska- Lincoln, available at https://cehs.unl.edu/borc/rotor-rod%E2%84%A2-system/; Castagne et al., CNS Safety Pharmacology, Reference Module in Biomedical Research , 2014; Dunham NW and Miya TS (1957) A note on a simple apparatus for detecting neurological deficit in rats and mice. Journal of the American Pharmaceutical Association, American Pharmaceutical Association (Baltimore) 46: 208-209; Bohlen et al., Calibration of rotational acceleration for the rotarodtest of rodent motor coordination, Journal of Neuroscience Methods (2009) 178: 10-14; Shiotsuki et al., A rotarod test for evaluation of motor skill learning. JNeurosci Methods. 2010 Jun 15; 189(2): 180-5. doi: 10.1016/j.jneumeth.2010.03.026. Epub 2010 Mar 30. PMID: 20359499; and Rustay NR, Wahlsten D, and Crabbe JC (2003) Influence of task parameters on rotarod performance and sensitivity to ethanol in mice. Behavioural Brain Research 141 : 237-249, are hereby incorporated by reference in their entirety. The information and procedures (e.g., protocols) disclosed will be implemented for the study of S-rimantadine, R-rimantadine, and/or racemic rimantadine. b) Photobeam Activity System-Home Cage

[0178] CNS toxicity associated with the use of R-rimantadine, S-rimantadine, and racemic rimantadine will be studied with the use of a Photobeam Activity System-Home Cage (San Diego Instruments). Use of the photobeam activity system-home cage will allow study of the animal’s locomotive activity. Animals receiving R-rimantadine will demonstrate less CNS toxicity as evidenced by photobeam activity system-home cage testing.

[0179] Animals (e.g., mice or rats) will receive doses (e.g., varying doses) of R- rimantadine, S- rimantadine, or racemic rimantadine. After a period of time (e.g., 1 hour, 2 hours, 3 hours, 5 hours, 10 hours, 1 day, 2 days, 3 days, 5 days, 7 days, and/or 10 days) after receiving a dose, the potential CNS effects will be measured with the use of the photobeam activity system-home cage. Animals receiving S- rimantadine will demonstrate less adverse CNS effects and toxicity when compared to R- rimantadine and racemic rimantadine. In particular, animals (e.g., mice or rats) receiving S- rimantadine will demonstrate less abnormal locomotor activity. The information and procedures used and disclosed in Photobeam Activity System-Home Cage, San Diego Instruments, available at https://sandiegoinstruments.com/product/pas-homecage/, and Tatem et al., Behavioral and locomotor measurements using an open field activity monitoring system for skeletal muscle diseases. J Vis Exp. 2014 Sep 29;(91):51785. doi: 10.3791/51785. PMID: 25286313; PMCID: PMC4672952 are hereby incorporated by reference in its entirety. The information and protocols set forth in these disclosures will be used for the study of R-rimantadine, S-rimantadine, and racemic rimantadine. The study will be used and potentially modified to analyze aspects of the animal’s physiological responses related to the CNS, such as circadian rhythm and anxiety. c) Irwin Test/Functional Observation Battery (FOB)

[0180] CNS toxicity associated with the use of R-rimantadine, S-rimantadine, and racemic rimantadine will be studied with the use of an Irwin Test and FOB. Use of the Irwin test and FOB will allow study of the qualitative effects of R-rimantadine, S- rimantadine, and racemic rimantadine. Animals receiving R-rimantadine will demonstrate less CNS toxicity as evidenced by the Irwin Test/FOB test.

[0181] Animals (e.g., mice or rats) will receive doses (e.g., varying doses (e.g., four different doses)) of R-rimantadine, S-rimantadine, or racemic rimantadine. After a period of time (e.g., 1 hour, 2 hours, 3 hours, 5 hours, 10 hours, 1 day, 2 days, 3 days, 5 days, 7 days, and/or 10 days) after receiving a dose, the behavior and physiological functions of the animals (e.g., mice or rats), will be studied. Animals receiving S-rimantadine will demonstrate less adverse CNS effects and toxicity when compared to R-rimantadine and racemic rimantadine. In particular, animals receiving S-rimantadine are will demonstrate less abnormal behavior and physiological function and similar doses, and animals receiving R-rimantadine will tolerate higher doses before demonstrating either observable effects on behavior and physiological function and/or higher doses before demonstrating clear behavioral toxicity. The information and procedures used and disclosed in Castagne etal., CNS Safety Pharmacology, Reference Module in Biomedical Research , 2014, Irwin S (1968), Comprehensive observational assessment: la. A systematic, quantitative procedure for assessing the behavioral and physiologic state of the mouse, Psychopharmacologia 13: 222-257, Esteve J, Farre AJ, and RoserR (1988) Pharmacological profile of droxicam, General Pharmacology 19: 49-54, Mattson et al., (1996) A performance standardfor clinical and functional observational battery examination of rats. Journal of the American College of Toxicology , 15: 239-250, and Rouxet al., Primary observation (Irwin) test in rodents for assessing acute toxicity of a test agent and its effects on behavior and physiological function. Curr. Protoc. Pharmacol. 2005 Jan 1 ; Chapter lOTJnit 10.10. doi: 10.1002/0471141755. ph1010s27. PMID: 22294127, are hereby incorporated by reference in their entirety. The information and protocols set forth in these disclosures will be used for the study of R-rimantadine, S-rimantadine, and racemic rimantadine. d) Morris Water Maze Test

[0182] CNS toxicity associated with the use of R-rimantadine, S- rimantadine, and racemic rimantadine will be studied with the use of a Morris Water Maze Test. The Morris Water Maze Test will allow the study of potential CNS toxicity experienced by animal (e.g., mouse or rat) by testingthe animal’s spatial learning ability. Animals receiving R-rimantadine will demonstrate less CNS toxicity as evidenced by Morris Water Maze testing.

[0183] Animals (e.g., mice or rats) will receive doses (e.g., varying doses (e.g., four different doses)) of R-rimantadine, S- rimantadine, or racemic rimantadine. After a period of time (e.g., 1 hour, 2 hours, 3 hours, 5 hours, 10 hours, 1 day, 2 days, 3 days, 5 days, 7 days, and/or 10 days) after a dose, animals will be put into the maze. Animals receiving S- rimantadine will demonstrate less adverse CNS effects and toxicity when compared to R- rimantadine and racemic rimantadine. In particular, animals receiving S-rimantadine will demonstrate less inhibition of their spatial learning ability. The information and procedures used and disclosed in Vorhees et al., Morris water maze: procedures for assessing spatial and related forms of learning and memory, NatProtoc 1, 848-858 (2006). https://doi.org/10.1038/nprot.2006.116, andCastagne et al., CNS Safety Pharmacology, Reference Module in Biomedical Research ,2014, Morris RGM (1981) Spatial localization does notrequire the presence of local cues, Learning and Motivation 12: 239-260, are hereby incorporated by reference in their entirety. The information and protocols set forth in these disclosures will be used for the study of R-rimantadine, S-rimantadine, and racemic rimantadine. e) Electroencephalogram (EEG) Scans

[0184] CNS toxicity associated with the use of R-rimantadine, S- rimantadine, and racemic rimantadine will be studied with the use of EEG scans. EEG scans will allow the study of an animal’s (e.g., mouse or rate) electrical activity in the brain. Animals receiving R-rimantadine will demonstrate less CNS toxicity as evidenced by EEG testing.

[0185] Animals (e.g., mice or rats) will receive doses (e.g., varying doses (e.g., four different doses)) of R-rimantadine, S-rimantadine, or racemic rimantadine. After a period of time (e.g., 1 hour, 2 hours, 3 hours, 5 hours, 10 hours, 1 day, 2 days, 3 days, 5 days, 7 days, and/or 10 days) after receiving a dose, EEG signals from the animals will be recorded. Animals receiving S- rimantadine will demonstrate less adverse CNS effects and toxicity when compared to R-rimantadine and racemic rimantadine. In particular, animals receiving S-rimantadine will demonstrate less abnormal EEG signals as compared to animals receiving to R-rimantadine and racemic rimantadine. The information and procedures used and disclosed in Vogler et al., Low CostElectrod Assembly for EEg Recordings in Mice, Front. Neurosci., 14 November 2017, https://doi.org/10.3389/fnins.2017.00629; DanhofM and Visser SA (2002) Pharmaco- electroencephalography and pharmacokinetic-pharmacodynamic modeling in drug development: focus on preclinical steps. Methods & Findings in Experimental & Clinical Pharmacology 24((Suppl D): 127-128; Itil TMand Itil KZ (1995) Quantitative EEG Brain Mapping In Psychotropic Drug Development, Drug Treatment Selection, and Monitoring. American Journal of Therapy 2: 359-367; and Protocol for Rat Sleep EEG, NeuroDetective International, available at https://www.ndineuroscience.com/userfiles/Rat_Sleep_EEG_Meth ods.pdf, are hereby incorporated by reference in their entirety. The information and protocols set forth in these disclosures will be used for the study of R-rimantadine, S-rimantadine, and racemic rimantadine.

EXAMPLE 10. IN VITRO CENTRAL NERVOUS SYSTEM (“CNS”) ASSAYS

[0186] Studies will be conducted to determine the effects of R-rimantadine, S- rimantadine, and racemic rimantadine on the changes in anatomy and/or physiology associated with CNS toxicity. Tissues obtained from animals (e.g., living or deceased) (e.g., mice and/or rats)) receiving S-rimantadine will demonstrate less changes as compared to baseline or normal (e.g., within acceptable limits) tissues when compared against tissues obtained from animals receiving R-rimantadine or racemic rimantadine.

[0187] Animals receiving S-rimantadine will demonstrate less physiological and/or anatomical changes due to CNS toxicity when compared to animals receiving similar doses of R-rimantadine and racemic rimantadine. Animals receiving S-will be capable of receiving higher doses of the S-rimantadine than animals receiving R- rimantadine and racemic rimantadine before exhibiting physiological and/or anatomical changes associated with CNS toxicity. Varying doses of R-rimantadine, S-rimantadine, and racemic rimantadine will be studied and with the use of at least the following tests. The information and protocols set forth in these disclosures will be used for the study of R-rimantadine, S-rimantadine, and racemic rimantadine.

Brain Slice/Whole-Cell Patch-Clamp

[0188] CNS toxicity associated with the use of R-rimantadine, S- rimantadine, and racemic rimantadine will be studied with the use of Brain Slice/Whole -Cell Patch-Clamp studies. Brain Slice/Whole-Cell Patch-Clamp electrophysiology will allow for analysis of the biophysical mechanism (e.g., ionic currents) of neural computation and pathology in neuronal cells. Animals receiving R-rimantadine will demonstrate less CNS toxicity (e.g., less anatomical and/or physiological changes) as evidenced by brain slice/whole-cell patch- clamp tests.

[0189] Animals (e.g., mice or rats) will receive doses (e.g., varying doses (e.g., four different doses)) of R-rimantadine, S- rimantadine, or racemic rimantadine. After a period of time (e.g., 1 hour, 2 hours, 3 hours, 5 hours, 10 hours, 1 day, 2 days, 3 days, 5 days, 7 days, and/or 10 days) after receiving a dose, the animals will be euthanized and brain slices will be obtained and analyzed. Alternatively, whole-cell patch claim may be conducted in vivo. In such a case, after the period of time after receiving a dose, the animal will be analyzed without being euthanized. Animals receiving S-rimantadine will demonstrate less adverse CNS effects and toxicity when compared to animals receiving R-rimantadine or racemic rimantadine. In particular, animals receiving S-rimantadine will demonstrate less abnormal biophysical mechanisms of neural computation and pathology (e.g., ionic currents) than animals receiving either R-rimantadine or racemic rimantadine. The information and procedures used and disclosed in Kodandaramaiah et al., Automated whole -cell patch-clamp electrophysiology of neurons in vivo , Nat Methods. 2012 Jun;9(6):585-7. doi: 10.1038/nmeth.l993. Epub 2012 May 6. PMTD: 22561988; PMCID: PMC3427788, is hereby incorporated by reference in its entirety. The information and protocols set forth in these disclosures will be used for the study of R- rimantadine, S-rimantadine, and racemic rimantadine.

OTHER EMBODIMENTS

[0190] It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention which is defined by the scope of the appended claims. Other aspects, advantages, and modification are within the scope of the following claims.