WO2025147598A1 - Device system and use for hpv infection and cervical intraepithelial neoplasia - Google Patents
Device system and use for hpv infection and cervical intraepithelial neoplasia Download PDFInfo
- Publication number
- WO2025147598A1 WO2025147598A1 PCT/US2025/010220 US2025010220W WO2025147598A1 WO 2025147598 A1 WO2025147598 A1 WO 2025147598A1 US 2025010220 W US2025010220 W US 2025010220W WO 2025147598 A1 WO2025147598 A1 WO 2025147598A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- certain embodiments
- retaining device
- monofumarate
- vaginal
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/66—Phosphorus compounds
- A61K31/675—Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0034—Urogenital system, e.g. vagina, uterus, cervix, penis, scrotum, urethra, bladder; Personal lubricants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/08—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
- A61K47/10—Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/08—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
- A61K47/12—Carboxylic acids; Salts or anhydrides thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/30—Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
- A61K47/32—Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/30—Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
- A61K47/36—Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
- A61K47/38—Cellulose; Derivatives thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
Definitions
- the present disclosure provides a device and therapeutic system for the topical delivery of the PMEG prodrug Compound I or its pharmaceutically acceptable salt such as monofumarate and hemifumarate salts thereof, Compound II, Compound III, or one or more morphic forms of the foregoing to treat a female in need thereof with a human papilloma virus (HPV) infection or a related disorder such as HPV-induced cervical or vaginal intraepithelial neoplasia, in a manner that reduces toxicity to nearby cells and tissues.
- HPV human papilloma virus
- HPV HPV is so common that most sexually active people have been infected at some point in their lives.
- Papillomaviruses are a group of non-enveloped DNA viruses, which in humans infect keratinocytes of skin and mucous membranes including in the cervical area. HPV infections can cause cellular transformations in the human patient that have not yet progressed to cancer but have reached the stage of neoplasia.
- HPV-induced neoplasia forms include cervical intraepithelial neoplasia (“CIN”), anal intraepithelial neoplasia (“AIN”), perianal intraepithelial neoplasia (“PAIN”), vulvar intraepithelial neoplasia (“VIN”), penile intraepithelial neoplasia (“PIN”) and vaginal intraepithelial neoplasia (“VAIN”).
- Cancers caused by HPV include cervical, anal, perianal, penile, vaginal, vulvar, and oropharyngeal cancer.
- HPV can cause viral infection, neoplasia and cancer.
- Most of the cancer- causing HPV types are from the alpha-7 and alpha-9 species and include types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73, and 82.
- the most common cancer-causing HPV types are 16 and 18.
- the causal role of HPV in cancer of the cervix has been firmly established biologically and epidemiologically. Persistent infection with high-risk HPV is necessary to promote progression of pre-malignant stages to invasive cancer.
- Oncogenic HPV types are detected in almost all cervical cancer specimens, with types 16 and 18 accounting for about 70% of cervical cancers and about 50% of high-grade lesions. The majority of venereal warts are caused by the low-risk HPV types 6 and 11.
- Commonly used drug therapies include trichloroacetic acid, 5-fluorouracil, imiquimod and podofilox. Imiquimod (Aldara TM , Zyclara TM ) stimulates the immune system to clear the infection through toll-like receptor signaling and causes redness and swelling.
- SUMMARY OF THE DISCLOSURE It has been discovered that an effective composition for the treatment of cervical or vaginal HPV infection, and related conditions such as cervical or vaginal intraepithelial neoplasia, requires the combination of the selection of a number of aspects that work together to achieve the desired results.
- the present disclosure in certain aspects provides Compound II and in particular the isolated morphic form Compound II Pattern 1, pharmaceutical compositions containing such compound, methods of treating an HPV infection or intraepithelial neoplasia related to HPV infection using the selected morphic form described herein, and methods of preparing such compound and morphic form which is used in the therapeutic delivery with a retaining device as disclosed herein.
- the monofumarate salt of ethyl ((R)-((2-(2-amino-6-methoxy-9H-purin-9-yl)ethoxy)methyl)(benzyloxy)phosphoryl)-L- alaninate (Compound II) has very high tissue penetration when administered topically to the target tissue. Topical administration avoids toxicity associated with systemic administration of the drug.
- Attorney Docket No.01394-0006-01PCT [0053] Because precancerous and/or cancerous cells that are infected with HPV are several layers into the epithelium, the compound must have high penetration into the tissue to reach and treat these affected cells.
- Compound I monofumarate has superior tissue permeation and penetration in both porcine and human vaginal tissues over ABI-1968, which failed in clinical trials despite also being a phosphonate ester of an acyclic purine nucleoside.
- Compound I monofumarate can reach concentrations of about 40-85 ng/mL in vaginal tissue at a 0.1% dose.
- ABI-1968 even when used in a 3% dose does not reach 15 ng/mL concentration (see FIG.2). The significant improvement in tissue penetration, especially considering the decrease in dose, could not have been predicted in advance.
- Compound I monofumarate is surprisingly stable (Example 22) in comparison to other salt forms of the compound.
- Nonlimiting advantageous solid dosage forms include a vaginal tablet, which can be inserted into the affected area.
- a formulation to use in the vaginal tablet (or acceptable gel, cream or other semisolid formulation with sufficient integrity) for the treatment of HPV or an Attorney Docket No.01394-0006-01PCT intraepithelial neoplasia, such as CIN is a dosage form containing from about 0.01 milligram to about 0.03 milligram, from about 0.03 milligrams to about 0.25 milligrams, from about 0.20 milligrams to about 0.5 milligrams, from about 0.4 milligrams to about 1 milligram, from about 0.75 milligram to about 3 milligrams, from about 1 milligram to about 10 milligrams, from about or 5 milligrams to about 20 milligrams of the active ingredient (e.g., Compound I).
- the active ingredient e.g., Compound I
- containing when used in relation to a composition or dosage form, for example, means “comprising” (i.e., consisting at least in part of).
- containing when used in relation to a composition or dosage form, for example, means “comprising” (i.e., consisting at least in part of).
- a formulation for the treatment of HPV or an intraepithelial neoplasia, such as CIN is a dosage form containing about or at least 0.03, 0.05, 0.1 mg, 0.3 mg.0.5 mg, 0.7 mg, 1 mg, 1.5 mg, 2 mg, 2.5 mg, 3 mg, 4 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45mg or 50 mg of Compound I monofumarate, Compound II or Compound III.
- a formulation for the treatment of HPV or an intraepithelial neoplasia, such as CIN is a dosage form containing about 0.05 mg, 0.1 mg, 0.3 mg, 1 mg, 1.5 mg, or 2 mg of Compound I monofumarate, Compound II or Compound III.
- the dose strengths in mg used herein refer to the mass of the active acyclic phosphonamidate (the free base) and do not include the salt in the molecular weight.
- the 0.1 mg dose is referring to the amount of the corresponding free base.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 0.1 mg of the active agent (e.g., Compound I) 3 times a week for one week (0.3 mg/week).
- the active agent e.g., Compound I
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 0.3 mg of the active agent (e.g., Compound I) 3 times a week for one week (0.9 mg/week).
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 0.3 mg of the active agent (e.g., Compound I) 3 times a week for two weeks (1.8 mg/2 weeks).
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 0.3 mg of the active agent (e.g., Compound I) 3 times a week for one week, followed by 0.1 mg of the active agent 3 times a week for one week (1.2 mg/2 weeks).
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 1.0 mg of the active agent (e.g., Compound I) 1 time per week for three weeks.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with a total of 3.0 mg of the active agent (e.g., Compound I) in about 2 weeks (e.g., 15 days).
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 1.0 mg of the active agent (e.g., Compound I) 1 time per week for three weeks, broken into 2 cycles.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with a total of 6.0 mg of the active agent (e.g., Compound I) in about 6 weeks (e.g, Attorney Docket No.01394-0006-01PCT about 43 or 44 days), broken into 2 cycles.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 1.0 mg of the active agent (e.g., Compound I) 4 times in two weeks, broken into 2 cycles.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with a total of 8.0 mg of the active agent (e.g., Compound I) in about 6 weeks (e.g., about 43 or 44 days), broken into 2 cycles.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 1.0 mg of the active agent (e.g., Compound I) 1 time every two weeks, broken into four cycles.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with a total of 4.0 mg of the active agent (e.g., Compound I) in about 6 weeks (e.g., about 43 or 44 days), broken into four cycles).
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 1.0 mg of the active agent (e.g., Compound I) 1 time every two weeks, broken into three cycles.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with a total of 3.0 mg of the active agent (e.g., Compound I) in about 6 weeks (e.g., about 43 or 44 days), broken into four cycles).
- the patient with an HPV infection or an intraepithelial neoplasia, such as CIN administers a tablet comprising a monofumarate salt of the active agent (e.g., Compound I monofumarate salt, Compound II, or Compound III).
- a monofumarate salt of the active agent e.g., Compound I monofumarate salt, Compound II, or Compound III.
- the patient administers the tablet by means of a retaining device, which is inserted to, for example, collect post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG.
- the patient administers the tablet by means of an applicator that is removed before insertion of a retaining device.
- Dosage forms which do not adhere well to the target site may be dislodged, interfering with treatment.
- the present disclosure provides dosage forms that adhere to the target site and dissolve rapidly in low fluid volumes. Adhesion to the target site also minimizes exposure to non-target tissues, which may limit toxicity, unwanted systemic Attorney Docket No.01394-0006-01PCT exposure, and side effects, especially when used in combination with the retaining device. Dosage forms which soften, break down, and/or disintegrate quickly in low fluid volumes are advantageous to cause a rapid release of the active compound to the target tissue.
- the dosage form is a vaginal tablet.
- the dosage form is a semisolid such as a gel or cream.
- the dosage form disintegrates in about one to about ten seconds. In certain embodiments, the dosage form disintegrates in about ten seconds to about one minute.
- the dosage form disintegrates in about one minute to about one hour. In certain embodiments, the dosage form disintegrates in about one hour to about six hours.
- the physical dimensions of the dosage form can impact the effectiveness of the dosage form. A tablet that is thinner provides a greater surface area to volume ratio and may degrade quicker and cover the target area better. In certain embodiments, the dosage form is less than 3 millimeters thick in its smallest dimension. [0075] The formulation of the dosage form may be important for adequate administration of the active agent into the intraepithelial tissue.
- Tablet formulations should display the properties of mucoadhesion and substantivity and include excipients that have solubilizing, erosion-generating (for disintegration), porosity (for water uptake) and viscosity enhancing (to keep the drug at the target site) properties.
- excipients that will cause rapid disintegration of a solid dosage form to cover the cervix or vaginal areas include, but are not limited to mannitol, microcrystalline cellulose, lactose, sucrose, calcium phosphate, sodium phosphate, sodium bicarbonate, citric acid, maleic acid, adipic acid or fumaric acid.
- excipients that can enhance disintegration and coverage of the affected area include but are not limited to sodium starch glycollate, pregelatinized starch, crospovidone and croscarmellose sodium.
- Mucoadhesive excipients that are useful in the present disclosure include but are not limited to microcrystalline cellulose, polycarbophil, hydroxymethyl cellulose, hypromellose, hydroxypropyl cellulose, and PVP.
- a nonlimiting example of a tablet formulation includes, but is not limited to, microcrystalline cellulose, crospovidone, magnesium stearate, silicon dioxide, polyethylene Attorney Docket No.01394-0006-01PCT oxide and mannitol.
- Semi solid dosage forms may include, for example, a mucoadhesive polymer, a solubility/penetration enhancer, a lipophilic solubilizer and a penetration enhancer.
- the mucoadhesive polymer may be, but is not limited to, a carbomer, polyethylene glycol, crospovidone, hypromellose, polycarbophil and/or hydroxyethyl cellulose.
- Penetration enhancers include but are not limited to propylene glycol, transcutol, oleic acid, isopropyl myristate, propylene glycol glycerol monooleate, propylene glycol monocaprylate, PEG-8 Bees wax, cetyl alcohol, stearic acid, cetyl palmitate and/or cetostearyl alcohol.
- a non-limiting example of a semi-solid formulation comprises, for example, one or more of a carbomer, propylene glycol, sorbic acid, EDTA and water.
- a pessary (vaginal suppository) can be formulated with, for example but not limited to, hard fat (such as Ovucire, Witepsol, Supposi-Base), polyethylene glycol, macrogols, cocoa butter and glycerol.
- hard fat such as Ovucire, Witepsol, Supposi-Base
- polyethylene glycol such as polyethylene glycol
- macrogols such as polyethylene glycol
- cocoa butter and glycerol glycerol
- a non-limiting example of a pessary can be made from Witepsol H 15 or Ovucire WL 3264.
- the present disclosure includes at least the following features: (i) A method to treat a cervical or vaginal HPV-induced infection or an associated condition in a female in need thereof, including but not limited to cervical or vaginal intraepithelial neoplasia, comprising administering to a host in need thereof an effective amount of Compound I or a pharmaceutically acceptable salt thereof such as the monofumarate or hemifumarate, or other solid form, or Compound II or Compound III, in a pharmaceutical Attorney Docket No.01394-0006-01PCT composition, in a vaginal tablet or semisolid formulation with a vaginal applicator in combination with the use of a retaining device for sufficient time to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue.
- a method to treat a cervical or vaginal HPV-induced infection, intraepithelial neoplasia, or an associated condition in a human in need thereof comprising (i) administering an effective amount of the anti-HPV therapeutic agent Compound I or a pharmaceutically acceptable salt thereof, such as the monofumarate or hemifumarate, or other solid form or Compound II or Compound III typically in tablet form, to the cervix using a vaginal applicator (which is optionally coated with a lubricant) and then (ii) inserting a retaining device (which is optionally coated with a lubricant) that is maintained at the base of the cervix or in the vaginal canal for sufficient time to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue.
- a vaginal applicator which is optionally coated with a lubricant
- FIG.1A and FIG 1B depict the molecular structure of Compound I monofumarate pattern 1 as determined by the single crystal X-ray diffraction analysis of Example 21. There exists an intermolecular interaction between protonated N5-atom of free base and O7-atom of fumaric acid anion (N(5)–H(5) ⁇ O(7)) in the single-crystal form of Compound II.
- FIG.2 is an in vitro tissue permeation test in vaginal tissue comparing Compound I monofumarate salt to ABI-1968.
- Attorney Docket No.01394-0006-01PCT salts form solids with favorable properties for solid dosage forms including morphic forms for administration in combination with a retaining device to a host such as a human with an HPV infection or an HPV-related disease such as cervical or vaginal intraepithelial neoplasia.
- the monofumarate optionally in morphic form displays superior properties to the hemifumarate and monosuccinate.
- the monofumarate salt remains a desired salt form of Compound I for administration in combination with a retaining device.
- the compound used in combination with the retaining device is an isolated morphic form of the compound of the formula: wherein the isolated comprising peaks independently selected from at least 3, 4, 5, or 6 of the following 2theta values 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
- the compound used in combination with the retaining device is an isolated morphic form of the compound of the formula:
- the compound used in combination with the retaining device is an isolated morphic form of the compound of the formula: wherein the isolated comprising peaks independently or 2theta values 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 20.38+0.2°, 22.94+0.2°, 25.09+0.2°, 26.54+0.2°, 26.90+0.2°, 27.38+0.2°, 28.28+0.2°, 28.95+0.2°, 29.64+0.2°, and 38.07+0.2°.
- the compound used in combination with the retaining device is an isolated morphic form of the compound of the formula: wherein the isolated comprising peaks independently selected from at least 3, 4, 5, or 6 of the following 2theta values 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
- Compound I monofumarate pattern 1 characterized by an XRPD pattern in or substantially similar to that in FIG.9 is used in combination with the retaining device.
- Compound I monofumarate pattern 1 can be produced, for example, by crystallization from isopropyl alcohol and heptane, as described in Example 23.
- Compound I (free base) and about 1.0 equivalents of fumaric acid can for example be dissolved in Attorney Docket No.01394-0006-01PCT isopropanol at a concentration from about 25% to about 40% w/v and stirred at an elevated temperature, for example about 45°C, about 50°C, about 55°C. The solution is stirred at this temperature until some solids form and is then optionally seeded with Compound I monofumarate pattern 1 crystalline solids.
- the mixture is stirred and cooled to a lower temperature to facilitate crystallization, for example less than about 40°C, less than about 30°C, less than about 25°C, less than about 20°C, or less than about 15°C.
- the mixture is then stirred while heptane is added in an amount ranging from about 1mL per mL of isopropanol to about 5 mL per mL of isopropanol, such as about 4 mL per mL isopropanol.
- the resulting suspension is stirred while the product crystallizes, for example for at least about 24 hours at 25°C.
- the suspension is cooled to facilitate crystallization further.
- the solution can be cooled to less than about 10°C, less than about 5°C, less than about 0°C, or less than about -5°C.
- the solution is stirred at the decreased temperature to allow time for additional product to crystallize, such as for at least about one day, and then the solids are collected by filtration.
- the collected solids are dried under reduced pressure and optionally at elevated temperature to provide Compound I monofumarate pattern 1.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least about 3, 4, or 52theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least twelve of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least eleven of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least ten of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least eight of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least seven of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least six of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least five of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least four of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least three of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- XRPD pattern comprising at least three of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
- Attorney Docket No.01394-0006-01PCT [00201]
- Compound II Pattern 1 can be produced, for example, by recrystallizing Compound II (Example 28, Table 40), equilibration of Compound II in a suitable solvent, or crystallization by slow evaporation of solvent (Example 28, Table 41).
- the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least three, four or five 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
- the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least seven 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
- the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least six 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
- the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least five 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
- the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least four 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
- the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least three 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
- the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least one 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
- the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least three, four, or five 2theta values selected from 3.08+0.2°, 9.30+0.2°, 10.66+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 17.45+0.2°, 18.13+0.2°, 19.78+0.2°, 20.14+0.2°, 22.91+0.2°, 23.34+0.2°, 25.14+0.2°, 25.33+0.2°, 25.86+0.2°, 26.78+0.2°, 27.99+0.2°, and 28.82+0.2°.
- Compound III pattern 1 characterized by an XRPD pattern in or substantially similar to that in FIG.59 is used in combination with the retaining device.
- Compound III pattern 1 can be produced, for example, by precipitation from isopropanol and heptanes (Example 30, Table 48).
- Compound III pattern 1 was prepared by dissolving SP-Compound I free base in isopropanol, for example about 100 mg of SP-Compound I in about 0.25 mL to about 0.5 mL of isopropanol. To this solution was added about 1.0 eq of fumaric acid, and the mixture stirred at ambient or elevated temperature, for example about 25°C to about 60°C.
- the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising independently at least 2, 3, 4, 5, or 6 of the 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°.
- the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least ten 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°.
- the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least nine 2theta Attorney Docket No.01394-0006-01PCT values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°.
- the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least eight 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°.
- the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least seven 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°.
- the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least six 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°.
- the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least five 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°.
- the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least ten of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°.
- the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least nine of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°.
- the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least eight of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°.
- the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least seven of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°.
- the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least six of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°.
- the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least five of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°.
- the present disclosure includes but is not limited to compounds, pharmaceutical compositions, and the use of any of the compounds described herein in combination with the retaining device, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as fumarate, Compound II or Compound III, with desired isotopic substitutions of atoms at amounts above the natural abundance of the isotope, i.e., enriched.
- Isotopes are atoms having the same atomic number but different mass numbers, i.e., the same number of protons but a different number of neutrons.
- isotopes of hydrogen for example, deuterium ( 2 H) and tritium ( 3 H) may be used anywhere in described structures.
- isotopes of carbon e.g., 13 C and 14 C
- isotopes of carbon e.g., 13 C and 14 C
- a preferred isotopic substitution is Attorney Docket No.01394-0006-01PCT deuterium for hydrogen at one or more locations on the molecule to improve the performance of the drug.
- the deuterium can be bound in a location of bond breakage during metabolism (an ⁇ -deuterium kinetic isotope effect) or next to or near the site of bond breakage (a ⁇ - deuterium kinetic isotope effect).
- Achillion Pharmaceuticals, Inc. (WO/2014/169278 and WO/2014/169280) describes deuteration of nucleotides to improve their pharmacokinetic or pharmacodynamic, including at the 5-position of the molecule.
- substitution with isotopes such as deuterium can afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements.
- Substitution of deuterium for hydrogen at a site of metabolic break-down can reduce the rate of or eliminate the metabolism at that bond.
- the hydrogen atom can be any isotope of hydrogen, including protium ( 1 H), deuterium ( 2 H) and tritium ( 3 H).
- isotopically-labeled refers to an analog that is a "deuterated analog", a " 13 C-labeled analog,” or a “deuterated/ 13 C-labeled analog.”
- deuterated analog means a compound described herein, whereby a H, i.e., hydrogen/protium ( 1 H), is substituted by a H-isotope, i.e., deuterium ( 2 H).
- Deuterium substitution can be partial or complete. Partial deuterium substitution means that at least one hydrogen is substituted by at least one deuterium.
- the isotope is 90, 95 or 99% or more enriched in an isotope at any location of interest.
- the patient can be instructed to remain supine for several hours, for example, 3, 4, 5, 6, 7 or 8 hours with minimal activities and may resume normal activities after that time-period.
- the patient should repeat this procedure once daily for a total of 2, 3 or 4 applications in a week as instructed. It is preferred to carry out the two step administration process in the evening.
- Patients may administer three doses on alternate days over a 1-week period. Patients can optionally be instructed to use the following preventative measures to protect against leakage of drug or metabolite-containing fluid to healthy tissue: • Minimize amount of water-based lubricant (e.g.,KY gel) used for dosing. • Do not bathe the genital area until after the instructed administration dosage time period.
- water-based lubricant e.g.,KY gel
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with the active agent (e.g., Compound I) once per week.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with the active agent (e.g., Compound I) once every two weeks.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with the active agent (e.g., Compound I) once per 10 days.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 0.1 mg of the active agent (e.g., Compound I), followed by up to three doses of 0.1 within one week.
- the active agent e.g., Compound I
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with 0.1 mg of the active agent (e.g., Compound I) 3 times a week.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with up to 0.3 mg of the active agent (e.g., Compound I) for up to 3 times over one week.
- the active agent e.g., Compound I
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with up to 0.3 mg of the active agent (e.g., Compound I) for six doses over two weeks.
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with up to 1.0 mg of the active agent (e.g., Compound I) for up to six doses over up to two weeks.
- the active agent e.g., Compound I
- a patient with an HPV infection or an intraepithelial neoplasia, such as CIN is dosed with up to 1.0 mg of the active agent (e.g., Compound I) for up to six doses over up to four weeks.
- Dosage forms that disintegrate in, for example, less than about 50 ⁇ L, less than about 100 ⁇ L, less than about 125 ⁇ L, less than about 150 ⁇ L, less than about 175 ⁇ L, less than about 200 ⁇ L, or less than about 250 ⁇ L fluid particularly facilitate drug penetration into the target site.
- Attorney Docket No.01394-0006-01PCT [00265]
- the dosage form is a gel.
- the dosage form is a cream.
- the dosage form is a tablet.
- the dosage form disintegrates in about one to about ten seconds. In certain embodiments, the dosage form disintegrates in about ten seconds to one minute.
- the dosage form disintegrates in about one minute to about one hour. In certain embodiments, the dosage form disintegrates in about one to six hours. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 5 minutes. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 4 minutes. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 3 minutes. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 2 minutes. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 1 minutes. Disintegration can be tested by, for example, Ph.
- the physical dimensions of the dosage form can impact the effectiveness of the dosage form.
- a tablet that is thinner provides a greater surface area to volume ratio and may degrade quicker and cover the target area better.
- the dosage form is less than about 6, 5, 4, 3, or 2 millimeters thick in its smallest dimension.
- the formulation of the dosage form is important for adequate administration of the active agent into the intraepithelial tissue.
- the formulation for example, can be prepared for use as a tablet, a reconstituted powder, a dry powder, a semi solid dosage form, a film or a pessary (i.e., a vaginal suppository).
- Some embodiments disclosed herein include the use of an effective amount of Compound I monofumarate, Compound II or Compound III, in the manufacture of a medicament for ameliorating or treating a human papillomavirus infection, wherein the infection can be ameliorated or treated by inhibiting viral replication by inhibiting the synthesis of viral DNA.
- Other embodiments disclosed herein include the use of an effective amount of any of the compounds described herein including but not limited to Compound I monofumarate, Compound II or Compound III, for ameliorating or treating a human papillomavirus infection, wherein the human papillomavirus infection can be ameliorated or treated by inhibiting viral replication by inhibiting the synthesis of viral DNA.
- Certain nonlimiting embodiments disclosed herein include a method for ameliorating or treating a human papillomavirus infection that comprises contacting a cell infected with the human papillomavirus in a subject with an effective amount of any of the compounds described herein including but not limited to Compound I monofumarate, Attorney Docket No.01394-0006-01PCT Compound I hemifumarate, or other solid form of Compound I, Compound II or Compound III, wherein the infection is ameliorated or treated by inhibiting the synthesis of viral DNA.
- Yet still other embodiments disclosed herein include a method for ameliorating or treating a human papillomavirus infection that comprise administering to a subject infected with the human papillomavirus an effective amount of Compound I monofumarate, Compound II or Compound III, wherein the human papillomavirus infection can be ameliorated or treated by inhibiting viral replication by inhibiting the synthesis of viral DNA.
- Some embodiments disclosed herein relate to Compound I monofumarate, Compound II or Compound III, for use in ameliorating or treating a human papillomavirus infection, wherein the human papillomavirus infection can be ameliorated or treated by inhibiting viral replication by inhibiting the synthesis of viral DNA.
- the human papillomavirus can be a high-risk human papillomavirus (hrHPV), such as those described herein.
- the high-risk human papillomavirus can be selected from HPV-16, HPV-18, HPV-31, HPV-33, HPV-35, HPV-39, HPV-45, HPV-51, HPV-52, HPV-56, HPV- 58, HPV-59, HPV-68, HPV-73, and HPV-82.
- the human papillomavirus can be HPV-16.
- the human papillomavirus can be HPV-11.
- the human papillomavirus can be HPV-18. In some embodiments, the human papillomavirus can be one or more of the following high-risk types: HPV-31, HPV-33, HPV-35, HPV-39, HPV-45, HPV-51, HPV-52, HPV-56, HPV-58, HPV- 59, HPV-68, HPV-73, and HPV-82.
- HPV-31, HPV-33, HPV-35, HPV-39, HPV-45, HPV-51, HPV-52, HPV-56, HPV-58, HPV- 59, HPV-68, HPV-73, and HPV-82 can be detected using the Papanicolaou test (Pap smear) and/or DNA probe testing (for example, HPV DNA probe testing for one or more high-risk HPV types).
- an effective amount of Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II or Compound III
- a subject diagnosed with an HPV infection for example a high-risk HPV infection
- an effective amount of Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II or Compound III
- an effective amount of Compound I monofumarate, Compound II, or Compound III may be provided to a subject with a Papanicolaou test result that does not indicate the disease has progressed to cervical cancer.
- the Bethesda system is a Attorney Docket No.01394-0006-01PCT standardized scoring system for reporting pap smear test results and assigns a grade of 1-3 based on severity. Grade 1 CIN (CIN 1) indicates mild dysplasia. Grades 2 and 3 CIN (CIN 2, CIN 3) are more serious and typically require intervention.
- a pharmaceutical composition comprising Compound I monofumarate, Compound II or Compound III, is used in the manufacture of a system for the treatment of CIN 1 (Grade 1 cervical intraepithelial neoplasia) that comprises the use of a retaining device, as described further herein.
- a pharmaceutical composition comprising Compound I monofumarate, Compound II or Compound III, is used in the manufacture of a system for the treatment of CIN 2 (Grade 2 cervical intraepithelial neoplasia) that comprises the use of a retaining device as described further herein.
- a pharmaceutical composition comprising Compound I monofumarate, Compound II or Compound III, is used in the manufacture of a medicament for the treatment of CIN 3 (Grade 3 cervical intraepithelial neoplasia).
- Compound I monofumarate, Compound II or Compound III optionally in a pharmaceutically acceptable carrier, is used to treat a condition selected from the group consisting of atypical squamous cells of undetermined significance (ASC-US), atypical glandular cells (AGC), low-grade squamous intraepithelial lesions (LSIL), atypical squamous cells (cannot exclude high grade squamous intraepithelial lesion) (ASC-H), high grade squamous intraepithelial lesions (HSIL), adenocarcinoma in situ (AIS), and cervical cancer (e.g.
- ASC-US atypical squamous cells of undetermined significance
- ASC atypical glandular cells
- LSIL low
- Compound II optionally in a pharmaceutically acceptable carrier, is used in the therapeutic use and retaining device system to treat a condition selected from the group consisting of atypical squamous cells of undetermined significance (ASC-US), atypical glandular cells (AGC), low-grade squamous intraepithelial lesions (LSIL), atypical squamous cells (cannot exclude high grade squamous intraepithelial lesion) (ASC-H), high grade squamous intraepithelial lesions (HSIL), adenocarcinoma in situ (AIS), and cervical cancer (e.g.
- ASC-US atypical squamous cells of undetermined significance
- ASC atypical glandular cells
- LSIL low-grade squamous intraepithelial lesions
- ASC-H atypical squamous cells (cannot exclude high grade squamous intraepithelial lesion)
- HSIL high grade squamous intraepit
- Compound I monofumarate, Compound II or Compound III can be used in the manufacture of a system that comprises a retaining device for use in ameliorating and/or treating an infection of HPV-16 and/or HPV-18.
- Compound I monofumarate, Compound II or Compound III can be used in the manufacture of a system that comprises a retaining device for use in treating a high-risk HPV infection.
- Compound I monofumarate, Compound II or Compound III can be used in the manufacture of a system that comprises a retaining device for use in treating a related disease or condition occurring as a result of a high-risk HPV infection.
- Compound I monofumarate, Compound II or Compound III can be used in the manufacture of a system that comprises a retaining device for use in ameliorating and/or treating an infection comprising both high-risk and low-risk HPV.
- a pharmaceutical composition comprising Compound II is used in the therapeutic system that comprises a retaining device to treat conditions related to or occurring as a result of exposure to or an infection of HPV.
- a pharmaceutical composition comprising Compound II is used to treat precancerous cervical lesions.
- a pharmaceutical composition comprising Compound II is used to treat cervical intraepithelial neoplasia.
- a pharmaceutical composition comprising Compound II is used to treat vaginal intraepithelial neoplasia. In certain embodiments, a pharmaceutical composition comprising Compound II is used to treat cervical cancer. In certain embodiments, a pharmaceutical composition comprising Compound II is used to treat vaginal cancer. [00280] In certain embodiments, a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of conditions related to or occurring as a result of exposure to or an infection of HPV. In certain embodiments, a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of precancerous cervical lesions.
- a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the Attorney Docket No.01394-0006-01PCT treatment of cervical intraepithelial neoplasia.
- a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of vaginal intraepithelial neoplasia.
- a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of cervical cancer.
- a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of vaginal cancer.
- a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of oropharyngeal cancer.
- the dosage form may be easily applied to the target site. Direct application to the target site prevents systemic exposure and toxicity.
- the dosage form may be applied with an applicator.
- the dosage form is applied with a vaginal applicator.
- the dosage form is applied with a retaining device.
- additional fluid (such as a lubricant) is delivered along with the dosage form, applied to the dosage form, or applied to the target site or surrounding tissues.
- a lubricating fluid is administered in combination with the dosage form to enhance the coverage of the cervix, vagina or vulva.
- water is used as the fluid administered with the dosage form.
- a lubricating glycerol- or hydroxyethylcellulose-based, water soluble fluid is used in combination with the dosage form.
- the dosage form is administered without additional fluid.
- the dosage form will soften, disintegrate, and/or dissolve in less than about 5 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 4 milliliters of fluid.
- the dosage form will soften, disintegrate, and/or dissolve in less than about 3 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 2 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 1 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.75 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.5 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.25 milliliter of fluid.
- the dosage form will soften, disintegrate, and/or dissolve in less than about 0.2 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.15 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.125 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.1 milliliter of fluid. [00284] In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 10 microliters to about 100 microliters of fluid.
- the dosage form will soften, disintegrate, and/or dissolve in from about 75 microliters to about 250 microliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 200 microliters to about 500 microliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 400 microliters to about 750 microliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 700 microliters to about 1,000 microliters of fluid. [00285] In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 1 milliliter to about 2 milliliters of fluid.
- Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week. In certain embodiments, Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week for up to 12 weeks. In certain embodiments, Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week for up to 10 weeks. In certain embodiments Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week for up to 8 weeks.
- the dosage form is from about 4 mm to about 5 mm thick. In certain embodiments, the tablet is from about 5 mm to about 15 mm thick. In certain embodiments, the dosage form is less than 5 grams. In certain embodiments, the dosage form is from about 0.05 gram to about 0.15 gram. In certain embodiments, the dosage form is from about 0.1 to about 1 gram. In certain embodiments, the dosage from about 0.75 grams to about 2 grams. In certain embodiments, the dosage form is from about 1 gram to about 5 grams. In certain embodiments, the dosage form has a mass of from about 50 mg to about 500 mg. In certain embodiments, the dosage form has a mass of from about 100 mg to about 250 mg.
- the dosage form is not easily removed, dislodged, or moved from the target site. These desirable properties may be achieved by inclusion of a mucoadhesive polymer into the pharmaceutical composition.
- the pharmaceutical composition comprises a mucoadhesive polymer or mucoadhesive excipient.
- mucoadhesive polymers and excipients include: Hypromellose, lectin, thiolated polymers (e.g.
- chitosan–iminothiolane poly(acrylic acid)–cysteine, poly(acrylic acid)–homocysteine, chitosan–thioglycolic acid, chitosan–thioethylamidine, alginate– cysteine, poly(methacrylic acid)–cysteine and sodium carboxymethylcellulose– cysteine), polyethylene glycol, polyvinyl alcohol, polyvinyl pyrrolidinone, polyacrylic acid Attorney Docket No.01394-0006-01PCT (Carbopol®), polyheroxyethyl methacrylate, chitosan, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, methylcellulose, sodium carboxymethyl cellulose, aminated corn starch, cellulose derivatives, poly (acrylic acid) polymers, poly (hydroxyethyl methylacrylate), poly (ethylene oxide), poly (vinyl pyrrolidone), poly (
- the pharmaceutical composition comprises propylene glycol.
- the pharmaceutical composition comprises carboxypolymethylene.
- the pharmaceutical composition comprises ethylenediaminetetraacetic acid (EDTA).
- the pharmaceutical composition comprises sorbic acid.
- the pharmaceutical composition comprises carbomer.
- the pharmaceutical composition comprises hydroxyethyl cellulose.
- the pharmaceutical composition comprises polyethylene glycol.
- the pharmaceutical composition comprises two diluents and a glidant.
- the pharmaceutical composition comprises mannitol.
- Mannitol is a Generally Recognized As Safe (GRAS) listed naturally occurring sugar alcohol, widely used in pharmaceutical dosage forms and food products. It has been extensively studied including by vaginal route, and is currently in oral, parenteral, nasal and topical products.
- GRAS Generally Recognized As Safe
- the pharmaceutical composition comprises magnesium stearate.
- the pharmaceutical composition comprises microcrystalline cellulose.
- the pharmaceutical composition comprises polycarbophil.
- the pharmaceutical composition comprises polyethylene oxide.
- the pharmaceutical composition comprises colloidal silicon dioxide.
- the pharmaceutical composition comprises povidone. In certain embodiments, the pharmaceutical composition comprises isopropyl alcohol. In certain embodiments, the pharmaceutical composition comprises sodium starch glycolate. In certain embodiments, the pharmaceutical composition comprises croscarmellose sodium. In certain embodiments, the pharmaceutical composition comprises crospovidone. In certain embodiments, the pharmaceutical composition comprises hydroxypropylmethylcellulose. In certain embodiments, the pharmaceutical composition comprises lactose.
- a powder pharmaceutical composition comprises one or more excipient from the group consisting of xanthan gum, microcrystalline cellulose, polyethylene oxide, hydroxypropylmethylcellulose, hydroxypropylcellulose, carboxymethylcellulose sodium, povidone, mannitol, colloidal silicon dioxide, sodium benzoate, sodium starch glycolate, sodium lauryl sulfate, poloxamer 407, polyoxypropylene- polyoxyethylene copolymers, and the like.
- the pharmaceutical composition comprising an effective amount of a fumarate salt of any of the active compounds described herein including but not limited to Compound I, further comprises a pharmaceutically acceptable Attorney Docket No.01394-0006-01PCT excipient selected from the list consisting of Acacia, agar, alginic acid, ascorbyl palmitate, bentonite, benzoic acid, butylated hydroxyanisole, butylated hydroxytoluene, butylene glycol, calcium acetate, calcium hydroxide, canola oil, carob bean gum, carrageenan, castor oil, cellulose, corn starch, disodium edetate, erythorbic acid, ethyl lactate, ethylcellulose, glycerin, glyceryl behenate, glyceryl monooleate, glyceryl monostearate, hydroxyethylmethyl cellulose, hydroxypropyl cellulose, hypromel
- the pharmaceutical composition comprises pharmaceutically acceptable excipients for use as a pessary.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II or Compound III, further comprises up to 99.9% pessary excipient selected from the group consisting of hard fat, PEG, macrogols, cocoa butter, and glycerol.
- Non-limiting examples of hard fat include Ovucire® (mono-, di- and triglyceride esters of fatty acids (C 10 to C 18 ), the triester fraction being predominant and ethoxylated fatty alcohols), Witepsol® (glycerol esters of vegetable saturated fatty acids, such as lauric acid), and Supposi-baseTM (a blend of saturated polyglycolysed glycerides).
- Ovucire® mono-, di- and triglyceride esters of fatty acids (C 10 to C 18 ), the triester fraction being predominant and ethoxylated fatty alcohols
- Witepsol® glycerol esters of vegetable saturated fatty acids, such as lauric acid
- Supposi-baseTM a blend of saturated polyglycolysed glycerides
- the pharmaceutical composition comprising an effective amount of any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II or Compound III, further comprises a pharmaceutically acceptable excipient that enhances the penetration, disintegration, film forming and/or controlled release properties of the composition.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II or Attorney Docket No.01394-0006-01PCT Compound III, further comprises a penetration enhancing excipient.
- the penetration enhancing excipient is selected from the group consisting of oleic acid, eucalyptol, Caprylol, Labrafil, Labrasol, Lauroglycol, diethylene glycol monomethyl ether (Transcutol), propylene glycol, sodium laurate, sodium lauryl sulfate, cetyltrimethylammonium bromide, poloxamer (231, 182, 184), Tween 20, 40, 60, 80, fatty acids and fatty acid esters, isostearic acid, glycerin, and chitosan.
- the pharmaceutical composition comprising Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III contains from 0% to about 20% penetration enhancing excipients selected from the group consisting of cetyl alcohol, propylene glycol, transcutol P, oleic acid, isopropyl myristate, propylene glycol dicaprylate, glyceryl monooleate, propylene glycol monocaprylate, PEG-8 bees wax, cetyl alcohol, stearic acid, cetyl palmitate, and cetosteryl alcohol.
- penetration enhancing excipients selected from the group consisting of cetyl alcohol, propylene glycol, transcutol P, oleic acid, isopropyl myristate, propylene glycol dicaprylate, glyceryl monooleate, propylene glycol monocaprylate, PEG-8 bees wax, cetyl alcohol, stearic acid, cetyl palmitate, and
- the pharmaceutical composition comprises from about 0 to about 25% penetration enhancing excipients selected from the list consisting of stearyl alcohol, polysorbate 80, sodium lauryl sulfate, mono and diglycerides, sorbitan monostearate, glyceryl isostearate, polyoxyl 15 hydroxystearate, polyoxyl 40 hydrogenated castor oil, octyl dodecanol, and soybean lecithin.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III further comprises a film forming excipient.
- the pharmaceutical composition comprising any of the active compounds described herein including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III contains from 0% to about 99% film forming excipients selected from the group consisting of hypromellose, polyethylene glycol, polymethacrylates, microcrystalline cellulose, guar gum, xanthan gum, and polyvinylpyrrolidone.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises an excipient that allows for controlled release of the active compound.
- the controlled release pharmaceutical composition comprises ethylcellulose, hypromellose, microcrystalline wax, polycarbophil, and/or beeswax.
- Attorney Docket No.01394-0006-01PCT [00331] Percentage ranges of excipients and other components of the pharmaceutical composition are given as a percent by weight, unless otherwise specified.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises a disintegration enhancing excipient.
- the disintegration enhancing excipient is selected from the group consisting of cellulose, guar gum, crospovidone, polyplasdone, soy polysaccharides, calcium silicate, gelatin, cation exchange resins, bentonite, citrus pulp, alginic acid, calcium alginate, methylcellulose, microcrystalline cellulose, sodium carboxymethylcellulose, croscarmellose, solka floc, corn starch, sodium starch glycolate (Explotab, Primojel), glycine, hydroxypropyl starch, and starch 1500.
- the pharmaceutical composition comprises up to about 99% of a disintegration enhancing excipient(s) such as mannitol and/or microcrystalline cellulose.
- the pharmaceutical composition comprises from about 0 to about 70% of a disintegration enhancing excipient(s) selected from the list consisting of lactose, sucrose, and calcium phosphate. In certain embodiments, the pharmaceutical composition comprises from about 0 to about 50% of a disintegration enhancing excipient(s) selected from the list consisting of sodium bicarbonate, citric acid, maleic acid, adipic acid, and fumaric acid. In certain embodiments, the pharmaceutical composition comprises from about 0 to about 20% of a disintegration enhancing excipient(s) selected from the list consisting of sodium starch glycollate, pregelatinized starch, crospovidone, and croscarmellose sodium.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 70% mannitol, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 20%, about 30% about 40%, about 50%, about 60% or about 70%.
- the pharmaceutical composition comprising Compound I or a pharmaceutically acceptable salt thereof, such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 70% lactose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60% or about 70%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from about 0 to about 70% sucrose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60% or about 70%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from about 0 to about 70% microcrystalline cellulose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60% or about 70%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 20% sodium starch glycolate, including but not limited to any amount that achieves the desired results, for example up to about 1%, about 2%, about 3%, about 5%, about 7%, about 10%, about 12%, about 15% or about 20%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from about 0 to about 20% pregelatinized starch, including but not limited to any amount that achieves the desired results, for example up to about 1%, about 2%, about 3%, about 5%, about 7%, about 10%, about 12%, about 15% or about 20%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from about 0 to about 20% crospovidone, including but not limited to any amount that achieves the desired results, for example up to about 1%, about 2%, about 3%, about 5%, about 7%, about 10%, about 12%, about 15% or about 20%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Attorney Docket No.01394-0006-01PCT Compound III, further comprises from about 0 to about 20% croscarmellose sodium, including but not limited to any amount that achieves the desired results, for example up to about 1%, about 2%, about 3%, about 5%, about 7%, about 10%, about 12%, about 15% or about 20%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% xanthan gum, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20% about 25%, about 30%, about 35%, about 40%, about 45% or about 50%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% polycarbophil, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% polyethylene oxide, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% hydroxyethylmethyl cellulose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a Attorney Docket No.01394-0006-01PCT pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% hydroxyethyl cellulose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%.
- the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% hypromellose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%.
- a tablet used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV further comprises about 155 mg microcrystalline cellulose, about 1.75 mg of magnesium stearate, and about 17.5 mg of mannitol.
- a semi-solid formulation used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises about 15 mg of carbomer, about 50 mg of propylene glycol, about 10 mg of sorbic acid, about 5 mg of EDTA, and about 920 mg of water.
- a semi-solid formulation used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises about 20 mg of carbomer; about 70 mg of mineral oil; about 80 mg of a mixture of polyoxyl 6 stearate Type I, ethylene glycol stearates and polyoxyl 32 stearate type 1; about 5 mg parabens; about 60 mg propylene glycol; about 5 mg EDTA; and about 760 mg water.
- a dry powder for reconstitution is used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises about 15.5 mg xanthan gum, about 19.8 mg mannitol, about 5 mg silicon dioxide, and about 0.5 mg sodium benzoate.
- the treatments described herein in combination with the retaining device are used to lessen, ameliorate or substitute the use of these conventional practices.
- the treatments described herein can be used in combination with a surgical technique.
- a patient in need thereof can receive surgery before, during and/or after administration of an effective amount of a compound described herein.
- the compound is administered in combination with the retaining device about or less than about 8 months, 6 months, 4 months, 3 months, 2 months, or 1 month prior to surgery.
- the compound is administered in combination with the retaining device about or at least about 1 month, 2 months, 3 months, 4 months, 6 months, or 8 months after surgery.
- the surgical procedure can be an excision of the target and/or diseased tissue, including but not limited to loop electrosurgical excision procedure (LEEP), large loop excision of the transformation zone (LLETZ), knife conization, cold knife conization, knife cone biopsy, or laser conization.
- the surgical procedure can be ablation, including but not limited to laser ablation or cryoablation.
- the pharmacokinetic, biodistribution, half-life, or other parameters of the drug can be altered by such combination therapy (which may include alternation therapy if considered concerted).
- combination therapy with anticancer therapeutics can provide better outcomes for patients.
- the disclosed Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III are DNA polymerase inhibitors, it may be Attorney Docket No.01394-0006-01PCT useful to administer the compound to a host in need thereof in combination with, for example: a) a protease inhibitor; b) another DNA polymerase inhibitor; c) an inhibitor of E6 or E6AP such as MEDI0457, luteolin, CAF-24 or gossypetin; d) an inhibitor of E7; e) an inhibitor of E1 or E2, including inhibitors of the E1-E2 protein interaction; f) L2 lipopeptides; g) an inhibitor or degrader of L1 or L2; h) an HDAC inhibitor such as vorinostat; i) degraders of tetraspanins such as CD9, CD63 or CD151; j) immunotherapeutics such as T-cell therapies (including adoptive T-cell therapies) and check
- a salt is an example of a multicomponent solid, and a crystalline salt is a multicomponent crystalline form. Salts typically exhibit proton transfer between an acid and a base.
- Other multicomponent crystalline forms include cocrystals.
- a cocrystal is typically a crystalline solid of two or more neutral species. Because charge balance is not necessary for neutral species, the stoichiometry of cocrystals cannot be determined based on charge balance.
- one component is typically an active drug or prodrug and the other (or others) is usually termed a coformer, and it interacts nonionically with the active drug or prodrug in the crystal lattice.
- the solid form may be both a salt and a cocrystal.
- the active drug or prodrug may be in the form of a salt with a counterion and interact nonionically with a coformer.
- Such a solid form would be a multicomponent solid and, when crystalline, a multicomponent crystalline form. Examples of such cocrystals when the coformer is volatile includes solvates.
- a solid form may exist in a single crystalline solid form or be polymorphic where more than one crystalline form of the same solid form exists. Polymorphism is typically determined by use of x-ray powder diffraction.
- Other examples of multicomponent solids are hydrates and solvates.
- a hydrate is a solid form that includes water in the solid form.
- the water may form part of the unit cell of the crystal in which case it is often a stoichiometric hydrate.
- a solvate is a solid form that includes one or more solvent molecules and, when crystalline, may form part of the unit cell of the crystal in which case it is often a stoichiometric solvate.
- a hydrate or a solvate can also be used in the therapy described generally herein.
- the solid form may be partially crystalline, disordered, or amorphous.
- any of the solid forms described above that comprise Compound I can be used in an effective amount in the treatment of HPV infection or cervical or vaginal intraepithelial neoplasia in a patient in need thereof, as generally taught herein.
- a multicomponent crystal comprising Compound I can include a salt form of Compound I, a cocrystal form of Compound I, and/or a mixture of a salt and cocrystal form of Compound I.
- the solid form formed can be a function of the synthesis and crystallization or solidification conditions used, among other factors.
- an effective amount of a multicomponent crystal comprising Compound I and a pharmaceutically acceptable coformer is used in the treatment of cervical or vaginal intraepithelial neoplasia in a patient in need thereof.
- an effective amount of a multicomponent crystal comprising Compound I and a pharmaceutically acceptable coformer is used in combination with a retaining device to treat cervical or vaginal intraepithelial neoplasia in a patient in need thereof.
- the crystalline form formed by addition of about 1 equivalent of fumaric acid to a solution of Compound I optionally in isopropyl alcohol and heptane, optionally in a pharmaceutically acceptable carrier, is used in the treatment of cervical or vaginal intraepithelial neoplasia, optionally in combination with a retaining device.
- Any solid form comprising Compound I or a salt thereof can be used in an effective amount with or without a retaining device to deliver therapy to the HPV-infected patient in need thereof.
- any solid form comprising Compound I or a salt thereof can be used in an effective amount with or without a retaining device to deliver therapy to a patient infected with a virus chosen from adenovirus, human cytomegalovirus, BK virus, and hepatitis B virus.
- a virus chosen from adenovirus, human cytomegalovirus, BK virus, and hepatitis B virus.
- the present disclosure that includes the combined use of the therapeutic compounds as described herein with a retaining device includes the use of one of the following compounds below which may optionally be used in a morphic form as described herein, and further embodiments that include such compounds. 1.
- a compound of the formula: Attorney Docket No.01394-0006-01PCT or or A compound of the formula: Attorney Docket No.01394-0006-01PCT or . Attorney Docket No.01394-0006-01PCT 5.
- the therapeutic use and/or retaining device system of embodiment 6, wherein the pharmaceutical composition is in a solid dosage form.
- the therapeutic use and/or retaining device system of embodiment 6, wherein the pharmaceutical composition is in a semi-solid dosage form. 9.
- the therapeutic use and/or retaining device system of embodiment 6, wherein the pharmaceutical composition is in the form of a film. 10. The therapeutic use and/or retaining device system of embodiment 6, wherein the pharmaceutical composition is in the form of a pessary. 11. The therapeutic use and/or retaining device system of embodiment 7, wherein the pharmaceutical composition is in the form of a tablet. Attorney Docket No.01394-0006-01PCT 12. The therapeutic use and/or retaining device system of embodiment 8, wherein the pharmaceutical composition is in the form of a cream. 13. The therapeutic use and/or retaining device system of embodiment 8, wherein the pharmaceutical composition is in the form of a gel. 14. The therapeutic use and/or retaining device system of any one of embodiments 6-13, wherein the pharmaceutical composition is formulated for topical administration. 15.
- a method to treat a cervical or vaginal HPV-induced infection or a condition caused by a human papillomavirus in a human in need thereof, including but not limited to cervical or vaginal intraepithelial neoplasia comprising administering to a host in need thereof an effective amount of the compound or pharmaceutical composition for use as described in any one of embodiments 1-43, with a vaginal applicator in combination with a retaining device for a sufficient time to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue. 45.
- a method to treat a cervical or vaginal HPV-induced infection or a condition caused by a human papillomavirus, including by not limited to intraepithelial neoplasia, in a human in need thereof comprising: (i) administering an effective amount of the compound or pharmaceutical composition as described in any one of embodiments 1-43 to the cervix using a vaginal applicator (which is optionally coated with a lubricant); and then (ii) inserting a retaining device (which is optionally coated with a lubricant) that is maintained at the base of the cervix or in the vaginal canal for sufficient time to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its Attorney Docket No.01394-0006-01PCT metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non- diseased tissue.
- the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap, and a sponge.
- the retaining device is used as both the applicator and a retaining means to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue.
- the retaining device is a menstrual cup.
- the retaining device is a menstrual disc. 50.
- the method of embodiment 44 or 45, wherein the retaining device is a tampon. 51.
- the method of embodiment 44 or 45, wherein the retaining device is a cervical cap.
- the method of embodiment 44 or 45, wherein the retaining device is a sponge.
- the method of embodiment 47, wherein the retaining device used as both an applicator and a retaining means is a diaphragm.
- the method of any one of embodiments 44-54, wherein the condition caused by a human papillomavirus is atypical squamous cells of undetermined significance (ASC- US). 56.
- ASC atypical glandular cells
- ASC-H high grade squamous intraepithelial lesion
- AIS adenocarcinoma in situ
- any one of embodiments 44-54, wherein the intraepithelial neoplasia is cervical intraepithelial neoplasia.
- the method of embodiment 59, wherein the cervical intraepithelial neoplasia is Grade 1 cervical intraepithelial neoplasia.
- the method of embodiment 59, wherein the cervical intraepithelial neoplasia is Grade 2 cervical intraepithelial neoplasia.
- 62. The method of embodiment 59, wherein the cervical intraepithelial neoplasia is Grade 3 cervical intraepithelial neoplasia.
- any one of embodiments 44-54 wherein the intraepithelial neoplasia is vaginal intraepithelial neoplasia.
- 64. The method of any one of embodiments 44-63, wherein from about 0.05 milligrams to about 40 milligrams of active compound is administered.
- 65. The method of any one of embodiments 44-63, wherein from about 0.1 milligrams to about 30 milligrams of active compound is administered. 66.
- any one of embodiments 44-63 wherein from about 0.001 to about 20 mg, from about 0.005 to about 10 mg, from about 0.01 mg to about 5 mg, from about 0.03 mg to about 1 mg, from about 0.05 mg to about 0.3 mg, from about 0.03 mg to about 0.07 mg, from about 0.05 mg to about 0.15 mg, or from about 0.15 mg to about 0.45 mg of the active compound is administered.
- 67 The method of any one of embodiments 44-63, wherein from about 0.05 mg to about 0.3 mg of the active compound is administered.
- 68. The method of any one of embodiments 44-67, further comprising applying a lubrication means to the epithelial tissue before administering the dosage form to the affected area. 69.
- any one of embodiments 44-67 further comprising applying a lubrication means to the dosage form before administering the dosage form to the affected area.
- the lubrication means is selected from water, a glycerol-based lubricant, and a hydroxyethylcellulose-based lubricant.
- 71. The method of any one of embodiments 44-70, wherein the compound or pharmaceutical composition is administered once per day.
- 72. The method of any one of embodiments 44-70, wherein the compound or pharmaceutical composition is administered twice per day.
- 73 The method of any one of embodiments 44-70, wherein the compound or pharmaceutical composition is administered twice per week.
- a method of treating a cervical or vaginal HPV-induced infection or an associated condition in a female in need thereof, including cervical or vaginal intraepithelial neoplasia comprising (i) administering to the base of the cervix an effective amount of dosage form of a therapeutic compound selected from: Attorney Docket No.01394-0006-01PCT (ii) and then inserting a retaining device. 2.
- a method of treating a cervical or vaginal HPV-induced infection or an associated condition in a female in need thereof, including cervical or vaginal intraepithelial neoplasia comprising (i) administering an effective amount of dosage form of a therapeutic compound selected from: or Attorney Docket No.01394-0006-01PCT and then (ii) inserting a retaining device.
- a method of treating a cervical or vaginal HPV-induced infection or an associated condition in a female in need thereof, including cervical or vaginal intraepithelial neoplasia comprising (i) administering a dosage form with an effective amount of a therapeutic compound selected from: or a pharmaceutically acceptable salt thereof; (ii) inserting a retaining device.
- the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap and a sponge. 12. The method of any one of embodiments 1-11, wherein the retaining device is used as both the applicator and a retaining means. 13. The method of embodiment 11, wherein the retaining device is a menstrual cup. 14. The method of embodiment 11, wherein the retaining device is a menstrual disc. 15. The method of embodiment 11, wherein the retaining device is a tampon. 16. The method of embodiment 11, wherein the retaining device is a diaphragm. Attorney Docket No.01394-0006-01PCT 17.
- the retaining device is a cervical cap. 18. The method of embodiment 11, wherein the retaining device is a sponge. 19. The method of any one of embodiments 1-18, wherein the compound is administered in a morphic form. 20. The method of any one of embodiments 1-19, wherein the dosage form is administered to the base of the cervix. 21. The method of any one of embodiments 1-20, wherein the retaining device is placed in the vaginal area, for example, below the cervical base and, for example, for a sufficient time to collect the post-treatment leakage of the therapeutic compound or its PMEG metabolite. 22.
- any one of embodiments 4 or 6-20 wherein the compound is an isolated morphic form of: wherein the isolated comprising peaks independently selected from at least 3, 4, 5, or 6 of the following 2theta values 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. 23.
- any one of embodiments 5-20 wherein the compound is an isolated morphic form of: wherein the isolated comprising peaks independently selected from at least 3, 4, 5, 6, 7, 8 or 9 of the following 2theta values Attorney Docket No.01394-0006-01PCT 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. 24.
- any one of embodiments 5-20 wherein the compound is an isolated morphic form of: wherein the isolated comprising peaks independently or 2theta values 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 20.38+0.2°, 22.94+0.2°, 25.09+0.2°, 26.54+0.2°, 26.90+0.2°, 27.38+0.2°, 28.28+0.2°, 28.95+0.2°, 29.64+0.2°, and 38.07+0.2°. 25.
- any one of embodiments 1 or 6-20 wherein the compound is an isolated morphic form of the compound of the formula: wherein the isolated comprising peaks independently selected from at least 3, 4, 5, or 6 of the following 2theta values 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. 26.
- the table disintegrated in less than about 150 ⁇ L of fluid. 28.
- the method of any one of embodiments 1-27 wherein about 0.05 to 1 mg of therapeutic compound or pharmaceutically acceptable salt thereof is administered.
- the pharmaceutical composition comprises a polymer. 35. The method of embodiment 6, wherein the pharmaceutical composition comprises a disintegration enhancing excipient. 36. The method of embodiment 6, wherein the pharmaceutical composition comprises a penetration enhancing excipient. 37. The method of embodiment 6, wherein the pharmaceutical composition comprises an excipient that allows for controlled release. 38. The method of embodiment 6, wherein the pharmaceutical composition comprises at least one of a light mineral oil, propylparaben, Tefose 63, water, EDTA, methylparaben, and Carbopol 974P. 39. The method of embodiment 6, wherein the pharmaceutical composition comprises at least one of EDTA, methyl paraben, Carbopol 974P, propylene glycol, and sorbic acid.
- any one of embodiments 1-40 wherein the condition associated with a human papillomavirus is atypical squamous cells, cannot exclude high grade squamous intraepithelial lesion (ASC-H). 44. The method of any one of embodiments 1-40, wherein the condition associated with a human papillomavirus is adenocarcinoma in situ (AIS). 45. The method of any one of embodiments 1-40, wherein the condition is cervical intraepithelial neoplasia. 46. The method of any one of embodiments 1-40, wherein the condition is cervical intraepithelial neoplasia Grade 1. 47.
- the lubrication means is selected from water, a glycerol-based lubricant, and a hydroxyethylcellulose-based lubricant.
- Attorney Docket No.01394-0006-01PCT 56 The method of any one of embodiments 1-55, wherein the dosage form is administered for about one week. 57. The method of any one of embodiments 1-55, wherein the dosage form is administered for about two weeks.
- the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap, and a sponge. 70.
- the kit of embodiment 70, wherein the retaining device is a menstrual cup.
- the kit of embodiment 70, wherein the retaining device is a menstrual disc. Attorney Docket No.01394-0006-01PCT 73.
- the kit of embodiment 70, wherein the retaining device is a tampon.
- the kit of embodiment 70, wherein the retaining device is a diaphragm.
- the kit of embodiment 70, wherein the retaining device is a cervical cap.
- 76. The kit of embodiment 70, wherein the retaining device is a sponge. 77.
- the hemi-fumarate seed crystals discussed immediately above were prepared by adding about 50 mg of Compound I (free base) to EtOH.0.5 molar amount of fumaric acid was added under stirring at 50°C for 2 hours and then at 25°C for at least 12 hours. Seeds crystals (prepared from a 30 mg batch as described immediately below) were added, and heptanes (0.4 mL) was added as antisolvent. Obtained mixture was stirred at 5°C for about 3 days. Then the suspension was taken out and centrifuged. Solids obtained were dried in oven at 50°C for about 1 hour under vacuum, and analyzed by XRPD to confirm the crystals were the Hemifumarate Pattern 1.
- the monofumarate seed crystals discussed immediately above were prepared by adding about 30 mg of Compound I (free base) to 0.1 mL of IPA. An equimolar amount of fumaric acid was added to the mixture under stirring at 50°C for 2 hours, a yellow clear solution was obtained, and 0.3 mL of heptane was added as antisolvent. Some seeds (prepared from a 50 mg batch as described immediately below) were added. Suspension was obtained and it was stirred at 25°C for at least 12 hours.
- the monofumarate seed crystals discussed immediately above were prepared by adding about 50 mg of Compound I (free base) to 0.1 mL of IPA.1.5 molar amount of fumaric acid was added to the mixture and the mixture was stirred at 25°C. After about 3 hours, the sample was too sticky, 0.1 mL additional IPA was added and kept stirring at 25°C for about 22 hours. Sticky sample was obtained and dried in a vacuum oven at 50°C for about 2 hours, and reslurried in 1.0 mL of MTBE at 25°C for about 5 days.
- the mixture was stirred at 50°C for about 1.5 hours and cooled to 25°C, then it was stirred at 25°C for about 10 min. Then, 40 mL of heptanes was added as antisolvent. Obtained suspension was stirred at 25°C for about 24 hours, then cooled to 5°C at a rate of 0.1°C/min and stirred at 5°C for about 1 day. The solids were collected by filtration and dried in the oven at 40°C for about 2 hours under vacuum. About 4.1 g of light pink solids were obtained in a yield of 81.2%.
- Example 3 Synthesis of Mixture of (R, S) and (S, S) ethyl-2-((((2-(2-amino-6-methoxy- 9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate (Compound I) Attorney Docket No.01394-0006-01PCT Step 1: Preparation of diethyl-((2-(2-amino-6-chloro-9H-purin-9-yl)-ethoxy)-methyl)- phosphonate (3) 1 (50 g, 0.296 mol, 1 equiv.), Cs 2 CO 3 (96.37 g, 0.296 mol, 1 equiv.) and DMF (250 mL) under N 2 atmosphere at room temperature.
- diethyl 2-chloroethoxymethyl phosphonate 2 (74.85 g, 0.325 mol, 1.1 equiv.) in a drop-wise manner.
- the reaction was stirred at 40-50°C for 0.5 to 1.5 hours, heated to 60-70°C and stirred for 0.5-1.5 hours, and then stirred at 75 to 85°C for 18-24 h.
- the reaction mixture was filtered and the resulting cake was washed with DMF (100 mL x 2).
- the combined filtrate was concentrated to a half volume below 70°C, diluted with n-heptane (250 mL) and again concentrated to a half volume below 75°C.
- the resulting crude product 3 was then purified by silica gel column chromatography using DCM to 1% MeOH in DCM. The fractions containing products were combined and the solvent was evaporated below 40°C.
- the solid product 3 was treated with the repeated dilution with MTBE and concentration (up to 1/3 rd volume). The resulting slurry was then diluted with MTBE (400-500 mL) and agitated at 40-50°C for 4-6 h and at 15-25°C for 8-15 h.
- TMSBr 167.47 g, 1.102 mol, 4.0 equiv.
- a drop-wise addition of 1144 g aqueous 1N NaOH was performed.
- the aqueous alkaline layer was separated and repeatedly washed with MTBE.
- Step 3 Preparation of ((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)- phosphonic acid (5) Attorney Docket No.01394-0006-01PCT [00383] To a flask containing MeOH (350 mL) was charged ((2-(2-amino-6-chloro- 9H-purin-9-yl)-ethoxy)-methyl)-phosphonic acid 4 (50 g, 0.162 mol) at 20-30°C, and stirred for 10-30 min.
- Step 4a Preparation of a mixture of (R,S)- and (S,S)-ethyl-2-((((2-(2-amino-6-methoxy- 9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate (8)
- ((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)- phosphonic acid 5 40 g, 0.132 mol, 1 equiv.) in DCM (560 mL) at 20-30°C under stirring was charged (S)-ethyl 2-aminopropionate hydrogen chloride salt 6 (20.19 g, 0.132 mol, 1 equiv.), benzyl alcohol 7 (71.28 g, 0.66 mol, 5 equiv.) and TEA (159.98 g, 1.58 mol, 12 equiv.),
- the remaining aqueous phase was then extracted with DCM (2 x 400 mL) and upon concentration of DCM under vacuum below 40°C, the crude product was purified by silica gel column chromatography with DCM to 2% MeOH in DCM as a mobile phase.
- Example 4 Preparation of (+)-Compound I Monofumarate ((+)-(2S)-Ethyl-2-((((2-(2- amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)- propionate monofumarate) (9) ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate (Compound I) 8 (29.72 g, 0.06 mol, 1 equiv.) in IPA was added a solution of fumaric acid (7.66 mol, 1.1 equiv.) in IPA through a filter at 45-55°C and stirring was continued for 1-2 h.
- the seeds of compound 13 were added to the reaction mixture and stirring was continued for 1-2 h at 45-55°C. After allowing the reaction mixture to settle at 20-30°C for 4-6 h, a drop-wise addition of n-heptane ( ⁇ 30 mL) was performed and stirring was continued for another 8-15h at 20-30°C and 0-5°C for 8-15h. The solid observed was filtered and the wet cake was washed with a mixture of IPA/n- heptane (1/3, v/v, ⁇ 5 mL).
- Step 2c Preparation of (R,R)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate and (S,R)-ethyl-2- ((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)- amino)-propionate monofumarate - - - methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate and (S,R)-ethyl-2-((((2- (2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate can be
- a topical gel formulation can be prepared, for example, by mixing an aqueous gel carrier with an active pharmaceutical ingredient or salt thereof, for example, Compound I, Compound I monofumarate, Compound II, or Compound III.
- an active pharmaceutical ingredient or salt thereof for example, Compound I, Compound I monofumarate, Compound II, or Compound III.
- the aqueous phase of the topical gel was prepared by mixing water, EDTA, methylparaben (or sorbic acid) and Carbopol®974P.
- the active pharmaceutical ingredient or salt thereof and Attorney Docket No.01394-0006-01PCT propylene glycol was added to this solution, mixed and pH adjusted, then filled into tubes.
- from about 0.001% w/w to about 10% w/w active pharmaceutical ingredient or salt thereof is added to the semisolid formulation.
- from about 0.0025% w/w to about 2.5% w/w such as 0.003%, 0.01%, 0.03%, 0.1%, 0.3% or 1%.
- Example 7 Preparation of vaginal tablets [00402] A nonlimiting example of the preparation of vaginal tablets of Compound I monofumarate is provided below (see FIG.5 for a flow diagram). Two or more of the excipients are combined, blended, and screened to make the excipient blend.
- the monofumarate salt of the active pharmaceutical ingredient (such as Compound I monofumarate) is screened and added to a portion of the excipient blend.
- the resulting mixture is then blended and then more excipient blend is added.
- the mixture is thus gradually diluted with the excipient blend, with thorough mixing after each addition of excipient blend.
- the magnesium stearate is added and the mixture blended once more. The mixture is then compressed into tablets and packaged. Table 2.
- Compression Compress the blend on a rotary tablet press using appropriate tooling (punches and die), to target weight. Check friability and disintegration at the beginning of the compression run, and check periodically for individual tablet weights, thickness and hardness.
- Packaging 1. Package bulk tablets into double lined re-closable clear PE bags with desiccants between the bags and further into an aluminum foil pouch with desiccant and heat sealed.
- Illustrative excipients for a vaginal tablet formulation Attorney Docket No.01394-0006-01PCT [00403] Tablet formulations are optionally selected to display the properties of mucoadhesion and substantivity and include excipients that have solubilizing, erosion- generating (for disintegration), porosity (for water uptake) and viscosity enhancing (to keep the drug at the target site) properties.
- excipients that will cause rapid disintegration to cover the cervix or vaginal areas include, but are not limited to mannitol, microcrystalline cellulose, lactose, sucrose, calcium phosphate, sodium phosphate, sodium bicarbonate, citric acid, maleic acid, adipic acid or fumaric acid.
- excipients that can enhance disintegration and coverage of the affected area include but are not limited to sodium starch glycollate, pregelatinized starch, crospovidone and croscarmellose sodium.
- Mucoadhesive excipients that are useful in the present disclosure include but are not limited to microcrystalline cellulose, polycarbophil, hydroxymethyl cellulose, hypromellose, hydroxypropyl cellulose, and PVP.
- a tablet formulation may for example comprise the active pharmaceutical ingredient, microcrystalline cellulose and may contain mannitol.
- the tablet formulation comprises one or more excipients selected from the rapid disintegrant category (left column of Table 3).
- the tablet formulation comprises one or more excipients selected from the disintegration enhancement category (middle column of Table 3).
- the tablet formulation comprises one or more excipients selected from the mucoadhesive excipient category (right column of Table 3).
- Table 3 Excipients for Tablets Rapidly disintegrating To Enhance Mucoadhesive polymers Attorney Docket No.01394-0006-01PCT Sucrose (0 to 70%) Pregelatinised starch (0 to Polyethylene Oxide (0 to 20%) 50%) e o Example 8.
- Illustrative excipients for a reconstitution powder or dry powder formulation [00405] A reconstitution powder or dry powder formulation may improve the shelf stability of a pharmaceutical agent or formulation.
- the dry powder formulation may be mixed with saline, propylene glycol or other aqueous carrier shortly before it is administered, minimizing the time for degradation.
- the dry powder formulation is mixed with an oil, cream, or other nonaqueous carrier shortly before it is administered.
- the reconstitution powder or dry powder formulation rapidly covers the infected or diseased tissue in the cervix, vulva or vagina. Excipients which enhance the rapid coverage of the cervix, vulva or vagina, include but are not limited to mannitol, lactose, sucrose, calcium phosphate, and microcrystalline cellulose.
- the excipient for rapid coverage of the cervix, vulva or vagina is mannitol.
- the reconstitution powder or dry powder formulation has good coverage of the cervix, vulva or vagina.
- excipients which enhance the coverage of the cervix, vulva or vagina include sodium starch glycollate, pregelatinized starch, crospovidone, and croscarmellose sodium.
- Attorney Docket No.01394-0006-01PCT [00408]
- the reconstitution powder or dry powder formulation contains mucoadhesive properties once it has been reconstituted.
- Excipients which improve the mucoadhesive properties of the reconstituted powder or dry powder formulation include but are not limited to xanthan gum, polycarbophil, polyethylene oxide, hydroxyethylmethyl cellulose, hydroxyethyl cellulose, Hypromellose, hydroxypropyl cellulose, PVP, and microcrystalline cellulose.
- the excipient which improves mucoadhesion is xanthan gum.
- a dry powder or reconstitution powder formulation comprises the active pharmaceutical ingredient and may contain mannitol and/or xanthan gum.
- the dry powder or reconstitution powder formulation comprises one or more excipients selected from the rapid coverage category (left column of Table 4).
- the dry powder or reconstitution powder formulation comprises one or more excipients selected from the coverage enhancement category (middle column of Table 4).
- the dry powder or reconstitution powder formulation comprises one or more excipients selected from the mucoadhesive excipient category (right column of Table 4). Table 4.
- Illustrative excipients for a semisolid formulation [00410] Semisolid formulations are selected to display the properties of mucoadhesion and assist in the drug penetration into the tissue.
- Semisolid formulations may include excipients that have solubilizing, lipophilic (to assist in solubilizing lipophilic compound), penetration enhancing (for higher activity) and mucoadhesive (to keep the drug at the target site) properties.
- the semisolid formulation is mucoadhesive. Excipients which contribute to the mucoadhesive properties include but are not limited to carbomer, polyethylene glycol, crospovidone, polycarbophil, hypromellose, and hyroxyethyl cellulose.
- the semisolid formulation enhances the penetration and/or solubility of the active pharmaceutical ingredient.
- Excipients which enhance the penetration and/or solubility of the active pharmaceutical ingredient include but are not limited to polyoxyl 6 stearate type I, ethylene glycol stearate, polyoxyl 32 stearate type I, and propylene glycol.
- a semisolid formulation comprises the active pharmaceutical ingredient and one or more excipients from each column of Table 5.
- the semisolid formulation comprises one or more excipients selected from the mucoadhesive polymer category (left column of Table 5).
- the tablet formulation comprises one or more excipients selected from the solubility and penetration enhancers category (second column of Table 5).
- the semisolid formulation comprises one or more excipients selected from the lipophilic solubilizer category (third column of Table 5). In certain non-limiting embodiments, the semisolid formulation comprises one or more excipients selected from the penetration enhancer category (right column of Table 5). Table 5. Excipients for semisolid dosage forms Mucoadhesive Solubility and penetration Lipophilic Penetration polymer enhancers solubilizers Enhancer 0 o Attorney Docket No.01394-0006-01PCT Polyoxyl 40 hydrogenated Stearic acid (0 to castor oil (0 to 25%) 20%) ol Example 10.
- Pessary and film forming formulations are selected to be solid at room temperature but soften to release the active pharmaceutical ingredient at body temperature. This allows for easy handling and storage of the formulation as well as achieving desired tissue coverage at the cervix.
- one or more excipients from the left column of Table 6 provide the desired properties.
- one or more excipients from the right column of Table 6 provide the desired properties. Table 6.
- the formulation for a vaginal tablet dosage form comprises the ingredients in Table 7.
- the formulation for a tablet dosage form comprises the ingredients in Table 7.
- An illustrative process for combining these ingredients into a tablet dosage form can be found in Example 7. Table 7.
- Porcine vaginal tissue on Franz diffusion cells [00425] The circular portion of tissue was sandwiched between two chambers of a Franz diffusion cell with an active diffusion area of 1 cm 2 , and the mucosal layer was exposed to the donor chambers. The resistance across porcine vaginal tissue was measured using a wave form generator to ensure the integrity of the tissue segment used for the permeation study. Porcine vaginal tissue with resistance of ⁇ 3 K ⁇ .cm 2 was used for study. The receiver chamber was filled with 8 ml of 5% solutol PBS 7.4 pH, which was stirred at 600 rpm with a 3 mm magnetic stir bar and the temperature was maintained at 37 °C with a circulating water bath.
- Compound I monofumarate vaginal tablets are formulated for topical administration at the site of action with the goal of producing minimal systemic exposure.
- dose-dependent increases in systemic exposure Cmax and AUC
- Cmax and AUC dose-dependent increases in systemic exposure
- No accumulation was observed after repeated (i.e., 3x/week for 2 weeks) administration.
- the extent of systemic exposure in humans is expected to be lower than observed in rabbits due to the differences in body mass.
- AUC Systemic exposures at a dose of 3 mg/kg after IV dosing were more than 20-fold higher than those observed in intravaginal toxicology studies at maximally tolerated dose strengths and are, on that basis, also expected to be much higher than potential human exposure by intravaginal application.
- Toxicity Studies in Dogs [00440] A similarly designed single-dose IV toxicity study was also completed in female dogs given doses of 0, 0.02, 0.06 and 0.3 mg/kg, again with 7- and 23-day post- treatment observation periods. As in rabbits, Compound I was well tolerated with no clinical signs and all animals survived to their scheduled necropsy. There were no changes in chemistry, hematology, urinalysis or coagulation parameters.
- Compound I monofumarate salt was investigated in a single and multiple- dose, ascending, randomized, placebo-controlled study in healthy human volunteers. The study was conducted in 2 parts. Part A consisted of single ascending dose cohorts administering Compound I monofumarate Vaginal Gel 0.01% or placebo and Compound I monofumarate vaginal tablet up to 0.3 mg or placebo. Part B consisted of 2 multiple ascending dose cohorts administering vaginal tablets with a dose of 0.1 mg or 0.3 mg Compound I or placebo (3 doses over 1-week period). The study completed in June 2023.
- Compound I hemifumarate Pattern 1 is an anhydrate. The stoichiometry of free form: fumaric acid is about 1:0.5 based on 1 H NMR result. It has a melting peak at Tonset of 85.2°C with an enthalpy of about 37 J/g. It about 1.0% weight loss at about 85°C. KF shows it contains about 1.7% of water. About 0.7% of EtOH and 0.7% of heptanes (by weight residual) was detected by 1 H NMR. [00466] Compound I monofumarate Pattern 1 is an anhydrate.
- the stoichiometry of free form: fumaric acid is about 1:1.0 based on 1 H NMR result. It has a melting peak at T onset of 107.2°C with split peaks and with an of about 78 J/g. The two thermal events cannot be resolved with 2 K/min and 0.5 K/min heating rate by DSC as well. It shows about 0.3% weight loss at about 107°C. KF shows it contains about 1.2% of water. About 0.7% of IPA and 2.2% of heptanes (by weight) residue was detected by 1 H NMR. Example 24.
- Compound I hemifumarate Pattern 1 tends to degrade more than Compound I monofumarate Pattern 1 in the two conditions mentioned above.
- Table 19 Bulk stability of Compound I free base Pattern 1, Compound I monofumarate Pattern 1 and Compound I hemifumarate Pattern 1 Compound I Compound I Compound I Storage Conditions free base Hemifumarate Monofumarate P tt rn 1 P tt rn 1 P tt rn 1 Example 25.
- Solubility of Compound I free base Pattern 1, Compound I Monofumarate Pattern 1 and Compound I Hemifumarate Pattern 1 [00470] About 4 mg of Compound I Pattern 1 was added into 2 mL of buffer solution. For Compound I hemifumarate Pattern 1 and Compound I monofumarate Pattern 1, about 4 mg was added into 1.8 mL and 1.6 mL buffer solution, respectively. pH value was adjusted in simulated vaginal fluid with 0.2N NaOH. After stirring at 37°C for 0.5 hour and 2 hours, clear solution was obtained for all the three candidates. Table 20.
- Example 26 Hygroscopicity of Compound I free base Pattern 1, Compound I Hemifumarate Pattern 1, and Compound I Monofumarate Pattern 1
- Hygroscopicity of Compound I free base Pattern 1, Compound I hemifumarate Pattern 1, and Compound I monofumarate Pattern 1 Compound I free base Compound I Compound I Pattern 1 Hemifumarate Pattern 1 Monofumarate Pattern 1 le 9 8 7 7 7 N [00475] Hygroscopicity was investigated by DVS at 25°C. All three Patterns are moderately hygroscopic. Compound I Pattern 1 shows about 4.3% water uptake up to 90% RH but absorbs about 14.3% water in 95% RH. For Compound I monofumarate Pattern 1 and Attorney Docket No.01394-0006-01PCT Compound I hemifumarate Pattern 1, they show about 2.9% and 6.9% water uptake in up to 95% RH.
- Example 27 Large Scale Preparation of Mono-Fumarate Pattern 1
- Compound I free base was dissolved in 11 mL of IPA.
- 1.0 eq. of fumaric acid was added to the yellow clear solution with stirring at 50°C. After about 1 hour, some solids precipitated out.
- 10 mg seeds of mono-fumarate (from the above ‘Small scale preparation’) were added.
- the mixture was stirred at 50°C for about 1.5 hours and cooled to 25°C, then it was stirred at 25°C for about 10 min.
- 40 mL of heptanes was added as antisolvent.
- Compound I monofumarate Pattern 1 (Example 27) was weighed to a 2 mL glass vial and aliquot of 20 ⁇ L of each solvent (as shown in Table 23 below) was added to determine solubility at 50°C. Maximum volume of each solvent added was 1 mL. Approximate solubility was determined by visual observation. Table 23.
- Cycle 2 0°C to 130°C at 10°C /min; 130°C to 0°C at 10°C /min; reheat from 0°C to 250°C at 10°C /min.
- Cycle 2 0°C to 130°C at 10°C /min; 130°C to 0°C at 10°C /min; reheat from 0°C to 250°C at 10°C /min.
- the results are presented in Table 44 and in FIG.54 and FIG.55. Table 44. Behavior of Compound I monofumarate Pattern 1 under heating and cooling Exp. Heating rate Thermal events 1 Cycle 1: 0°C to 106°C at 10°C/min; 106°C Heating: Behavior under compression [00507] About 10 mg of Compound I monofumarate Pattern 1 was compressed for 5 minutes at 10 MPa with a hydraulic press.
- Pattern B, Pattern C and Pattern E are hemifumarate salts, and Pattern D is a degradation product.
- Pattern B is an anhydrate of hemifumarate with HPLC purity of about 99.6%.
- MEK, acetone, acetone/heptanes, MEK/heptanes and EtOH/MTBE as solvents it can be obtained from equilibration experiment, addition of antisolvent, slow cooling and fast Attorney Docket No.01394-0006-01PCT cooling experiments.
- the ratio of free base form to fumaric acid is about 1: 0.52 by 1H- NMR.
- Pattern C is not a monofumarate. It has two thermal events with T onset of about 74.0°C with an enthalpy of about 89 J/g and Tonset of about 90.6°C with an enthalpy of about 15 J/g. It shows about 0.4% weight loss at about 73°C and 2.1% weight loss from 73°C to 144°C. About 2.0% (by weight) ACN residue was detected by 1 H NMR. [00514] Pattern D is a degradation product with HPLC purity of about 0.2%. It was obtained in water by equilibration experiment experiments.
- Pattern E is an anhydrate of hemifumarate with HPLC purity of about 98.9%. It was obtained in acetone/toluene by equilibration experiment. The ratio of free base form to fumaric acid of the mixture is about 1: 0.69 by 1 H NMR.
- EQ indicates the form can be prepared through equilibration in the listed solvent.
- SC indicates the form can be prepared by slowly cooling a solution of Compound I monofumarate in the listed solvent.
- FC indicates t the form can be prepared by quickly cooling a solution of Compound I monofumarate in the listed solvent.
- Attorney Docket No.01394-0006-01PCT Table 45 Summary of identified Compound I fumarate salt Patterns 1, B, C, and E Melting Polymorph Salt Temperatures Salt ratio Preparation (Enthalpy) Example 29: Preparation of Compound II Pattern 1 Experiment 1.
- Compound II Pattern 1 is an anhydrate.
- the stoichiometry of free base:fumaric acid is about 1:1.0 based on 1 H NMR result. It has a melting peak at Tonset of 141.5°C combined with TGA shows about 0.3% weight loss at about 130°C. No residual solvent was detected by 1 H NMR.
- Example 30 Preparation of III Pattern 1 [00520] S P Compound I free base (100 mg) and 0.3 mL of IPA were added into a glass vial. To it was added 1.0 equiv.
- the sample was stirred at 50°C for 1 h, then cooled to 3°C with 0.1°C/min. After stirring at 3°C for about 20 h, the sample was heated from 3°C to 50°C within 20 mins, then 0.2 equiv. of fumaric acid and 1.5 ml of heptanes were added into the mixture. The resulting mixture was stirred at 50°C for about 2 h then cooled to 3°C with 0.1°C/min. After stirring at 3°C for about 13 h, it was reheated to 50°C within 20 mins and kept stirred at 50°C for about 6 h. It was cooled to 3°C with 0.1°C/min and stirred at 3°C for about 2 days.
- Part B may include up to 3 proof of concept (POC) cohorts, and Part B enrollment may proceed when a dose regimen tested in Part A is deemed safe and tolerated and demonstrates some level of efficacy.
- POC proof of concept
- Each Part B POC cohort (up to 3 cohorts will be expanded; Expanded Cohorts could be from A3 through A11, based on the data from Part A) will serve as an expansion of the corresponding Part A cohort as follows: Expanded Cohort A3: up to 0.3 mg for up to 3 doses over 1 week Expanded Cohort A4: up to 0.3 mg for 6 doses over 2 weeks Attorney Docket No.01394-0006-01PCT Expanded Cohort A5: up to 1.0 mg for up to 6 doses over up to 2 weeks Expanded Cohort A6: up to 1.0 mg for up to 6 doses over up to 4 weeks Expanded Cohort A7: up to 1.0 mg for up to 6 doses over up to 2 weeks (defined as a single treatment cycle); up to 2 treatment cycles Expanded Cohort A8: up to 1.0 mg, 2 intermittent weekly dosing (1 week of treatment, followed by 1 week treatment break, followed by 1 week of treatment over 3 total weeks) for up to 6 doses (defined as a single treatment cycle
- cHSIL will be confirmed by positive p16 status. Histology diagnosis must be obtained from the biopsy within 60 days prior to the first dose administration. The lesion must be fully visible and evaluable and involve no more than 3 quadrants based on colposcopic evaluation at the time of enrollment. [00550] Participants who will be using a vaginal applicator will be instructed in insertion of the vaginal tablet to the cervix with the applicator. Participants may be asked to insert a tampon (as a retaining device) after the vaginal tablet is administered with the applicator (e.g., immediately after administration or after some period of time after administration).
- a tampon as a retaining device
- HPV status will be evaluated with the Roche cobas® HPV Test collected using a cervical swab.
- HPV testing will occur at Baseline, Days 15, 29, 42, and prior to the LLETZ procedure.
- 2-week extended (dosing 3x/week) treatment be administered in Cohorts A4 and A5 or Part B POC cohort(s)
- HPV testing will be performed at Baseline, Days 19, 29, 42, and prior to the LLETZ procedure.
- Vaginal tablets were prepared from Compound I monofumarate as described above in Example 38.
- eligible participants will be randomized to receive Compound I monofumarate vaginal tablets vs. placebo in a 3:1 ratio, stratified (in the expansion cohorts) based on age at randomization (25 to ⁇ 30 years of age vs. ⁇ 30 years of age) and hrHPV genotype (HPV-16 and/or -18 vs. any other hrHPV genotype).
- Expanded Proof of Concept (POC) cohorts may include up to 4 POC cohorts, and randomization may proceed when a dose regimen tested in a sentinel cohort is deemed safe and tolerated by the SMC and demonstrates some level of efficacy.
- POC Expanded Proof of Concept
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Gynecology & Obstetrics (AREA)
- Reproductive Health (AREA)
- Urology & Nephrology (AREA)
- Virology (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Medicinal Preparation (AREA)
Abstract
The disclosure relates to a pharmaceutically acceptable salt of an acyclic nucleotide phosphonamidate to treat HPV and related conditions including neoplasia, as well as pharmaceutical compositions, morphic forms and dosage forms thereof.
Description
Attorney Docket No.01394-0006-01PCT DEVICE SYSTEM AND USE FOR HPV INFECTION AND CERVICAL INTRAEPITHELIAL NEOPLASIA CROSS REFERENCE [0001] This application claims the benefit of U.S. Provisional Application No.63/618,174, filed January 5, 2024, and international application No. PCT/2024/10538, filed January 5, 2024, the entire contents of both of which are incorporated by reference herein. FIELD OF THE DISCLOSURE [0002] The present disclosure provides a device and therapeutic system for the topical delivery of the PMEG prodrug Compound I or its pharmaceutically acceptable salt such as monofumarate and hemifumarate salts thereof, Compound II, Compound III, or one or more morphic forms of the foregoing to treat a female in need thereof with a human papilloma virus (HPV) infection or a related disorder such as HPV-induced cervical or vaginal intraepithelial neoplasia, in a manner that reduces toxicity to nearby cells and tissues. BACKGROUND [0003] According to the U.S. Center for Disease Control, there is no direct cure for human papilloma virus. HPV is so common that most sexually active people have been infected at some point in their lives. [0004] Papillomaviruses are a group of non-enveloped DNA viruses, which in humans infect keratinocytes of skin and mucous membranes including in the cervical area. HPV infections can cause cellular transformations in the human patient that have not yet progressed to cancer but have reached the stage of neoplasia. Forms of HPV-induced neoplasia include cervical intraepithelial neoplasia (“CIN”), anal intraepithelial neoplasia (“AIN”), perianal intraepithelial neoplasia (“PAIN”), vulvar intraepithelial neoplasia (“VIN”), penile intraepithelial neoplasia (“PIN”) and vaginal intraepithelial neoplasia (“VAIN”). Cancers caused by HPV include cervical, anal, perianal, penile, vaginal, vulvar, and oropharyngeal cancer.
Attorney Docket No.01394-0006-01PCT [0005] Thus, HPV can cause viral infection, neoplasia and cancer. Most of the cancer- causing HPV types are from the alpha-7 and alpha-9 species and include types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73, and 82. The most common cancer-causing HPV types are 16 and 18. The causal role of HPV in cancer of the cervix has been firmly established biologically and epidemiologically. Persistent infection with high-risk HPV is necessary to promote progression of pre-malignant stages to invasive cancer. Oncogenic HPV types are detected in almost all cervical cancer specimens, with types 16 and 18 accounting for about 70% of cervical cancers and about 50% of high-grade lesions. The majority of venereal warts are caused by the low-risk HPV types 6 and 11. [0006] The current therapeutic options for HPV and cervical intraepithelial neoplasia are adjunctive only. Commonly used drug therapies include trichloroacetic acid, 5-fluorouracil, imiquimod and podofilox. Imiquimod (AldaraTM, ZyclaraTM) stimulates the immune system to clear the infection through toll-like receptor signaling and causes redness and swelling. Podofilox (CondyloxTM) destroys tissues by destabilizing microtubules which prevents host cell replication. [0007] The cervical epithelium is composed of several layers of tissue and is referred to as stratified squamous epithelium. The layers are the superficial cell layer, the intermediate cell layer, the parabasal cell layer and the basal cell layer. It is essential that a topical drug for the treatment of cervical intraepithelial neoplasia is able to penetrate these multiple layers of tissue to adequately reach and treat the transformed cells. This is a formidable task because the cells are tightly bound and without blood vessels. [0008] Cervical intraepithelial neoplasia is most often treated by observation (the wait and see approach) or by excision or ablation of the cervical transformation zone. Techniques include cryotherapy, laser therapy, loop electrosurgical procedure (LEEP) and cone biopsy. All of these surgical procedures damage the affected areas and can lead to scarring. The most common intervention, LEEP, is effective in 60-90% of cases, however, it can be a painful procedure and may be associated with a significantly increased risk of miscarriage, ectopic pregnancies, and negative psychological outcomes. Despite extensive research, no drug has been approved to replace or combine with these surgical methods. [0009] Cervical high-grade squamous intraepithelial lesions (cHSIL), sometimes referred to as CIN2 and CIN3, is a disease caused by the abnormal hyperproliferation (dysplasia) of squamous cells in the cervical epithelium (Waxman, A. G., et. al 2012. “Revised terminology for cervical histopathology and its implications for management of high-grade squamous intraepithelial lesions of the cervix”. Obstet Gynecol, 120, 1465-71). Hyperproliferation
Attorney Docket No.01394-0006-01PCT usually occurs where the simple columnar, endometrial-type epithelium of the endocervix transitions to the stratified squamous epithelium of the ectocervix; this region is referred to as the “transformation zone” (Sellors, J. W. & Sankaranarayanan, R.2003. An introduction to the anatomy of the uterine cervix. Colposcopy and Treatment of Cervical Intraepithelial Neoplasia: A Beginners' Manual). Cervical HSIL is classified as a pre-cancerous condition because apoptosis is impaired in these hyperproliferating cells, which can lead to the accumulation of genetic alterations that transform the cells into cancer. The probability that the patient’s immune system will be sufficiently activated to clear lesions diminishes over time and is contingent upon a variety of factors including the patient’s immunocompetency, lifestyle (e.g., smoking), and nutritional status (Schiffman, M., et al 2011. “Human papillomavirus testing in the prevention of cervical cancer”. J Natl Cancer Inst, 103, 368-83). [0010] For women already infected with HPV, there is no approved drug therapy to treat HPV infections or cHSIL and thereby, to prevent the progression of HSIL to cervical cancer. [0011] Frantz Viral Therapies, LLC is conducting clinical trials to assess the use of artesunate vaginal inserts for the treatment of women with cervical high grade intraepithelial neoplasia (CIN2/3). Artesunate is an artemisinin derivative with cytotoxic activity. Artesunate is a known WHO-approved anti-malarial agent. The cytotoxic agent is delivered in the trial at a dosage, for example, of 50 to 200 mg for a 5-day cycle on weeks 0, 2 and 4. The artesunate vaginal inserts are self-administered at bedtime with a vaginal applicator, followed by use of a tampon, which is removed in the morning. Artesunate has a cytotoxic effect but is not an anti-viral agent, so does not directly stop the HPV replication. Therefore, artesunate cannot be used to treat patients who have an HPV infection that has not progressed to cervical intraepithelial neoplasia. Also, artesunate does have some systemic exposure under these conditions. See generally Trimble, et al., “A first-in-human proof-of-concept trial of intravaginal artesunate to treat cervical intraepithelial neoplasia 2/3 (CIN 2/3)”, Gynecologic Oncology 157 (2020)188-194 as well as U.S. Patents 6,586,464; 8,394,849; 8,940,787 and 7,989,491. [0012] Cidofovir, a pyrimidine based acyclic phosphonate nucleoside, which has broad spectrum activity against DNA viruses, is recognized as one of the effective treatments for HPV lesions that have not become cancerous. It is a DNA terminator and causes cell death via apoptosis of HPV transformed cells and regression of HPV-induced tumors. Cidofovir has been tested as a topical treatment of CIN2 and CIN3. See Van Pachterbeke, et al., “Topical treatment of CIN 2+ by cidofovir: Results of a phase II, double-blind, prospective, placebo-controlled study”, Gynecologic Oncology 115 (2009) 69-74. Researchers found that
Attorney Docket No.01394-0006-01PCT cidofovir cannot replace conization but may be used for topical chemotherapy. See also Snoeck, et al, “Cidofovir, a New Approach for the Treatment of Cervix Intraepithelial Neoplasia Grade III (CIN III)” Journal of Medical Virology 60:205-209 (2000). According to Snoeck, et al., except for two patients, patients had at least partial responses and half had a total response. In the partial responses, the transformed cells persisted in the deep tissues that can lead to neoplasia. Therefore, because of imperfect bioavailability, modifications were needed. The clinical trial thus had mixed results and was not progressed through completed clinical trials to an approved product. [0013] The Regents of the University of California, with Karl Hostetler, et. al, as named inventors, has filed a series of patents on various acyclic nucleotide derivatives to treat papilloma infections, including (i) U.S. Patent Nos.8,835,603; 9,629,860; 9,156,867; 10,449,207; 10,195,222; 10,076,533; 10,076,532; 9,775,852; 9,387,217 with a priority date of March 15, 2013; (ii) U.S. Patent Nos.10,702,532; 10,213,430; 9,493,493; and 9,801,884, with a priority date of September 15, 2014 (PCT/US2015/050202; published as WO 2016/044281); and (iii) U.S. Patent No.11,014,950 and 10,377,782 with a priority date of September 15, 2015. [0014] Antiva Biosciences carried out human clinical trials with the phosphonate ABI-1968 to assess its ability to adequately penetrate the various layers of cervical epithelium and release the antiviral agent PMEG (9-(2-phosphonomethyoxy)ethyl)guanine). PMEG is phosphorylated to PMEGpp (PMEG polyphosphate). It was determined that ABI-1968, when used even up to a 3% dose, does not reach 15 ng/mg of tissue concentration for ABI-1968 (see Bar F and G in FIG.2) and thus is not suitable as a topical drug to treat cervical intraepithelial neoplasia. The clinical trials were terminated as not successful. It was administered with or without a diaphragm or cervical cap retaining device.
[0015] It is thus a formidable challenge to topically dose HPV-infected epithelial stratified tissue in an effective manner that destroys the neoplasia cells in the multiple epithelial layers.
Attorney Docket No.01394-0006-01PCT The drug must be lipophilic enough to pass through the tissue layers and be metabolized if necessary to the active agent in a sufficient concentration to kill the pathogenic cells. [0016] It is an object of the present disclosure to provide an improved method for the treatment of cervical or vaginal HPV infection and related conditions such as HPV-induced cervical or vaginal intraepithelial neoplasia in a female in need thereof, that is effective against the infection or related conditions and also minimize toxicity to nearby tissues and cells. SUMMARY OF THE DISCLOSURE [0017] It has been discovered that an effective composition for the treatment of cervical or vaginal HPV infection, and related conditions such as cervical or vaginal intraepithelial neoplasia, requires the combination of the selection of a number of aspects that work together to achieve the desired results. It was essential to select the right compound with advantageous lipophilic and tissue penetrating properties combined with a selected pharmaceutically acceptable salt or other solid form optionally in an advantageous morphic form to achieve the long-sought ability to penetrate the deep epithelial stratified tissues in an effective amount to deliver the active agent. It required years of research to solve this problem, after many failures, to the benefit of patients globally suffering from interepithelial neoplasia that may become cancerous. In addition, Applicant has found that the use of a retaining means (such as a retaining device) is advantageous to minimize potential adverse effects on surrounding tissue that can be uncomfortable or even painful for the patient and possibly damaging to normal tissue. Thus, the disclosure includes a device system and therapeutic use to effectively treat HPV cervical or vaginal HPV infection or cervical or vaginal intraepithelial neoplasia. [0018] Specifically, it has been discovered that a particularly advantageous treatment for cervical or vaginal HPV infection or cervical or vaginal intraepithelial neoplasia is achieved by the administration of an effective amount of the PMEG prodrug Compound I or a pharmaceutically acceptable salt thereof such as the monofumarate or hemifumarate salt, or other solid form including in particular Compound II or Compound III. For example, such treatment may be performed by (i) administering an effective amount of the anti-HPV therapeutic agent Compound I (or a pharmaceutically acceptable salt thereof) or Compound II or III, typically in solid tablet form (although semi-solid formulations may also be acceptable), to the cervix using a vaginal applicator and then (ii) inserting a retaining device
Attorney Docket No.01394-0006-01PCT that is maintained at the base of the cervix or in the vaginal canal for sufficient time to collect the post-treatment leakage of vaginal fluids which may contain, for example, remaining therapeutic agent or its metabolite such as PMEG, in a manner that minimizes toxicity- causing damage to non-diseased tissue. In certain embodiments, the retaining device is separate from the applicator. In certain embodiments, the retaining device is both the applicator and the retaining means. For example, in some embodiments, the retaining device is used as the applicator. [0019] In some embodiments, the retaining device is inserted immediately after administration of the solid or semi-solid formulation. In some embodiments, the retaining device is inserted from about zero hours to about one hour after administration of the solid or semi-solid formulation. In some embodiments, the retaining device is inserted one hour or more after administration of the solid or semi-solid formulation. In some embodiments, the retaining device is inserted about 1-10 hours, about 1-9 hours, or about 1-8 hours after administration of the solid or semi-solid formulation. In some embodiments, the retaining device is inserted about 2-10 hours, about 2-9 hours, or about 2-8 hours after administration of the solid or semi-solid formulation. In some embodiments, the retaining device is inserted about 3-10 hours, about 3-9 hours, or about 3-8 hours after administration of the solid or semi-solid formulation. In some embodiments, the retaining device is inserted about 4-10 hours, about 4-9 hours, or about 4-8 hours after administration of the solid or semi-solid formulation. In some embodiments, the retaining device is inserted about 5-10 hours, about 5-9 hours, or about 5-8 hours after administration of the solid or semi-solid formulation. In some embodiments, the retaining device is inserted about 6-10 hours, about 6-9 hours, or about 6-8 hours after administration of the solid or semi-solid formulation. [0020] In some embodiments, the retaining device is held in place for 3-10 hours, for example 4-8 hours or 5-7 hours or about or at least around 6 hours. In some embodiments, the retaining device is held in place for 6-8 hours. In some embodiments, the retaining device is left in place overnight. The retaining device is then removed. In some embodiments, the retaining device is used after every topical application of the anti-HPV therapeutic agent to prevent undesired PMEG toxicity to surrounding tissue or cells. The retaining device can be, in non-limiting embodiments, a retaining period device, a birth control device, or other type of device that adequately separates the cervix from the vagina, or part of the vagina. Alternatively, it can be any device that forms an adequate barrier between the inserted tablet and surrounding vaginal tissue for a time period of at least 6 hours, and more generally, 7, 8, 9, 10, 11 or 12 hours, for example for 6-8 hours.
Attorney Docket No.01394-0006-01PCT [0021] For treatment of vaginal HPV infection or vaginal intraepithelial neoplasia potentially without cervical involvement, the patient can, in non-limiting embodiments, insert the retaining device lower in the vaginal canal. In some non-limiting embodiments, a semisolid dosage form such as a gel or cream may be used for the treatment of vaginal HPV infection or vaginal intraepithelial neoplasia, in combination with a retaining device placed such that the infected tissue is exposed to the active PMEG metabolite but uninfected tissues are not. Alternatively, the vaginal tablet can be inserted below the cervix or just above the infected parts of the vaginal canal to treat an infection in the vaginal canal. [0022] In certain embodiments, the retaining device is both the applicator and the retaining means. In this embodiment, the vaginal applicator and retaining device are combined into one device that can be used to both insert a vaginal tablet and act as a retaining device to protect surrounding tissue. In principal embodiments, the combined device and applicator is similar in structure to a diaphragm with a contoured surface that the vaginal tablet can be placed in for administration. [0023] In an alternative embodiment, instead of using a vaginal tablet, a gel or cream formulation with the active therapeutic, as described further below, can be administered and then used in combination with the retaining device. While the vaginal tablet (which can also be called a vaginal insert) is preferred for its solid structure and physical properties, a gel or cream with sufficient rigidity to deliver the active therapeutic and not unduly escape to surrounding tissue is also appropriate. A lubricant or other suitable material can be applied to the retaining device to hold the vaginal tablet in place. [0024] In certain embodiments, the vaginal applicator and/or retaining device is first lubricated with a water-based gel such as for example, a glycerol based lubricant, hydroxyethylcellulose-based lubricant, KY jelly or Surgilube to help maintain the vaginal tablet in place during application and for patient comfort. The lubricant may help the vaginal tablet remain in place for effective disintegration. Alternatively or in addition, a water-based gel can also be applied to the vaginal area optionally up to the cervix prior to vaginal tablet administration. In addition or alternatively, the lubricant can be coated on the retaining device for ease of placement as well as to help maintain the device in position. [0025] Applicant is now in clinical development of a pharmaceutical salt of Compound I below, which is a prodrug of PMEG, for topical, intravaginal treatment of HPV as well as several precancerous and potentially precancerous conditions related to human papillomavirus (HPV) infection, including high-grade squamous intraepithelial lesions (HSIL) of the cervix and high-risk HPV (hrHPV) infection in women.
Attorney Docket No.01394-0006-01PCT [0026] Specifically, it was discovered that the key compound is a specific salt of: .
[0027] Compound I is (ethyl(((2-(2-amino-6-methoxy-9H-purin-9- yl)ethoxy)methyl)(benzyloxy)-phosphoryl)-L-alaninate). U.S. Patent Nos.9,801,884 and 11,344,555 assigned to the Regents of the University of California claim Compound I and pharmaceutically acceptable salts generally, as well as methods of using the same for treating a papillomavirus infection. Compound I is an acyclic nucleotide phosphonate that metabolizes to a known potent antiviral compound (PMEG; ((9-[2- phosphonomethyoxy)ethyl)guanine])), but PMEG has poor cellular permeability and use- limiting systemic toxicity. Applicant has discovered how to improve the prodrug to be delivered topically in a manner that it is rapidly taken up into epithelial cells, a challenging task to date and one that ABI-1968 failed. Advantageous salt forms of Compound I include
[0028] At high local exposures, inhibition of DNA replication by Compound I or its
Attorney Docket No.01394-0006-01PCT pharmaceutically acceptable salt (for example in the form of Compound II) in healthy epithelial tissues may lead to immune cell infiltration, edema, epithelial thinning, and inflammation at sites of administration. Such effects have been observed in rabbits in toxicology studies evaluating repeated intravaginal administration over 2 weeks given a maximal dose volume of gel containing relatively high concentrations of Compound I monofumarate. These findings were dose level-dependent, with evidence of recovery observed 4 weeks following cessation of treatment. In a clinical trial in healthy participants, adverse events (AEs) that were observed to be related to vaginal tablets comprising Compound I monofumarate have mostly involved the lower genitourinary tract that were graded as mild or moderate in severity. Genitourinary AEs appeared approximately 1 to 2 weeks following the first dose, were persistent for 1 to 2 weeks after onset, and were reversible. In the multiple dose escalation portion of a clinical trial in healthy participants, multiple doses (3 doses over a 1-week period) of Compound I monofumarate vaginal tablets (0.1 mg) were safe and well tolerated, however, it is important to further minimize these negative side effects of the very potent therapy using a retaining device, as described in detail herein. [0029] As a non-limiting example, the female patient optionally coats the vagina with a lubricant and then inserts a vaginal tablet into the base of the cervical area with a vaginal applicator (optionally that has been coated with a lubricant), and then removes the vaginal applicator. The female then inserts the retaining device (also which has been optionally coated with a lubricant) and leaves it in place for at least 4-12 hours, such as 6-10 hours or 6- 8 hours and typically at least 6 hours. [0030] In some embodiments, the retaining device is inserted immediately after the tablet has been administered. In some embodiments, the retaining device is inserted from about zero hours to about one hour after administration of the tablet. In some embodiments, the retaining device is inserted one hour or more after the tablet has been administered. In some embodiments, the retaining device is inserted about 1-10 hours, about 1-9 hours, or about 1-8 hours after administration of the tablet. In some embodiments, the retaining device is inserted about 2-10 hours, about 2-9 hours, or about 2-8 hours after administration of the tablet. In some embodiments, the retaining device is inserted about 3-10 hours, about 3-9 hours, or about 3-8 hours after administration of the tablet. In some embodiments, the retaining device is inserted about 4-10 hours, about 4-9 hours, or about 4-8 hours after administration of the tablet. In some embodiments, the retaining device is inserted about 5-10 hours, about 5-9 hours, or about 5-8 hours after administration of the tablet. In some embodiments, the
Attorney Docket No.01394-0006-01PCT retaining device is inserted about 6-10 hours, about 6-9 hours, or about 6-8 hours after administration of the tablet [0031] As a non-limiting example, the female patient optionally coats the vagina with a lubricant and then inserts a vaginal tablet into the base of the cervical area with a retaining device (optionally that has been coated with a lubricant), which remains in place for at least 4-12 hours, such as 6-10 hours or 6-8 hours and typically at least 6 hours. [0032] In some embodiments, it is advantageous to insert the vaginal tablet and then place the retaining device in in the evening and leave it in overnight, for at least 5 or 6 hours or more (such as 6-8 hours). In the morning, the retaining device is removed and cleaned for the next use if reusable or disposed of if not. By administering before evening sleep, the topical medication is allowed to penetrate the intraepithelial tissue while the patient is in a supine position with little movement, and the retaining device protects surrounding healthy tissue from the PMEG prodrug toxicity. Nonlimiting examples of retaining devices are those that are conventionally used for period protection or those normally used for birth control. [0033] Nonlimiting examples of retaining devices normally used for period protection are: a menstrual cup, a menstrual disc, and a tampon. These devices are commercially available without a prescription. [0034] A menstrual cup is a cup that is placed in the vagina below the cervix that stays in place and normally holds blood flow. A variety of menstrual cups are commercially available with different shapes and sizes, and are typically bell-shaped, V-shaped, round or asymmetrical. A menstrual cup is typically made from medical grade silicone or latex rubber. Menstrual cups can typically be used for up to 8 to 12 hours, which is well within the time needed for the Compound I, II, or III or PMEG fluid retainment. It should be thoroughly cleaned between uses, with an antibiotic soap or in boiling water. Some menstrual cups have a tapered shape that can make insertion and removal easy. Nonlimiting examples of current commercially available products include Flex™, Cora™, Saalt™, Lena™, Lumma™, Tampax™, Shordy™ and Diva™. [0035] A menstrual disc is a product that is worn inside the vaginal canal that is flat and disc- shaped. It is placed differently in the vaginal canal than a menstrual cup. It is designed to fit at the base of the cervix, where it creates a seal to prevent leaks. The menstrual disc comes is a range of sizes, shapes and materials to fit the comfort of the patient. Menstrual cups sit lower in the vaginal canal than menstrual discs and are usually designed to create a suction. Nonlimiting examples of current commercially available menstrual disc products include, but
Attorney Docket No.01394-0006-01PCT are not limited to, those made by nixit™, Cora™, Saalt™, Lena™, Lumma™, Shordy™ and Diva™. [0036] A tampon is a well-known device that is a pocket-sized absorbent material that can optionally have a cardboard or plastic applicator that is inserted into the body normally to collect period fluid. It is normally removed every few hours up to once a day, for example every 4-6 hours or 6-8 hours. It is sold in many different varieties by many different vendors. [0037] Non-limiting birth control devices include, but are not limited to a diaphragm, a cervical cap, and a sponge. The diaphragm and cervical cap devices often need a doctor’s prescription as part of gynecological healthcare for birth control, while sponges do not. [0038] A diaphragm is a shallow bendable (e.g., flexible) cup that is placed in the vagina typically as a means for contraception. It covers the cervix during sex to prevent pregnancy. It is typically made of a soft silicone and is used in combination with a spermicide. Nonlimiting examples of current commercially available diaphragm products include, but are not limited to, those made by Caya™, Milex™ and Cooper Surgical™. In an alternative embodiment, the separate vaginal applicator and diaphragm can be combined into one device by including a contoured surface in the diaphragm that the vaginal tablet can be placed in. [0039] A cervical cap is smaller than a diaphragm and can be left in place longer. It covers the cervix and should be covered with a spermicide. A nonlimiting example of a current commercially available cervical cap product is the Femcap™. [0040] Sponges can also be used according to this disclosure. They are typically made of polyurethane or natural sea sponges. It is soft, spongy and usually round. It is placed against the cervix and typically contains applied spermicide (when used as a birth control device but not in the present disclosure). Often it has a loop attached for easy displacement and removal. Nonlimiting examples of products include, but are not limited to, those made by Safe-T™ and Today Sponge™. [0041] In certain embodiments, a female diagnosed with HPV or cervical or vaginal squamous intraepithelial lesions (SIL, either low-grade and high-grade lesions) is treated with a PMEG prodrug, for example, a Compound I monofumarate vaginal tablet in a dose strength of 0.1 mg to 2 mg, for example 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0 mg, once daily on a schedule of 1, 2, 3 or 4 times a week, or once every 10 days, optionally for 1, 2, 3, 4, 5 or 6 weeks with the use of a retaining device to adequately protect surrounding tissue. The dose strengths in mg used herein refer to the mass of the active acyclic phosphonamidate (e.g., Compound I) and do not include the salt in the molecular weight. In other words, when referring to a dosage
Attorney Docket No.01394-0006-01PCT form comprising 0.1 mg of Compound I monofumarate, for example, the 0.1 mg dose is referring to the amount of the corresponding free base. Thus, when referring to a 0.1 mg tablet of Compound I monofumarate, for example, it should be understood that the tablet contains 0.124 mg of the monofumarate as this amount is equivalent to a dose of 0.1 mg of Compound I free base. Similarly, when referring to a 0.3 mg tablet of Compound I monofumarate, it should be understood that the tablet contains 0.372 mg of the monofumarate as this amount is equivalent to a dose of 0.3 mg of Compound I free base. In addition, when referring to a 1 mg tablet of Compound I monofumarate, for example, it should be understood that the tablet contains 1.241 mg of the monofumarate as this amount is equivalent to a dose of 1 mg of Compound I free base. [0042] Short-term treatment with the vaginal tablet that comprises Compound I (or a pharmaceutically acceptable salt thereof), Compound II or Compound III is intended to exert two effects on cervical or vaginal lesions caused by HPV. First, the selected therapeutic has an anti-viral effect in HPV-infected cells by inhibiting viral genome replication. Second, it selectively and effectively induces apoptosis in hyperproliferating lesional cells, while the retaining device protects the surrounding normal tissue. The therapeutic effects of Compound I (or a pharmaceutically acceptable salt thereof), Compound II or Compound III (and other compounds of the same class) are mediated by inhibition of cellular DNA polymerases. Inhibition of HPV DNA replication in productively infected cells results in elimination of the replicating episomal viral DNA accompanied by arrest of supra basal cell DNA synthesis and attendant cellular apoptosis. Cells in high-grade lesions that are rapidly proliferating and deficient in normal cell cycle regulatory mechanisms should be highly sensitive to the pro-apoptotic effects of these therapeutic compounds and salts thereof that result from DNA synthesis blockage. Normal cells can typically withstand this temporary insult with well characterized checkpoints and DNA regulatory mechanisms, and the use of the retaining device further assures protection. Compound I (or a pharmaceutically acceptable salt thereof), Compound II or Compound III is therefore expected to eliminate HPV infection in productively infected cells typical of low-grade lesions and to induce apoptosis in advanced lesions, which often have integrated HPV genomes and express elevated levels of HPV oncoproteins. [0043] Compound I (ethyl (((2-(2-amino-6-methoxy-9H-purin-9-yl)ethoxy)methyl)- (benzyloxy)phosphoryl)-L-alaninate) has two chiral centers, one at the phosphorus atom and one in the amino acid moiety, either of which can be in the R or S stereoconfiguration. Therefore, Compound I exists as four stereoisomers, or two diastereomeric pairs: (RP, SC)/(SP,
Attorney Docket No.01394-0006-01PCT SC) and (RP, RC)/(SP, RC). While U.S. Patent Nos.9,801,884 and 11,344,555 describe Compound I generally, the patents do not address the potential stereochemistry of the phosphorus atom. It has been discovered that the stereoisomer of Compound I with R- stereochemistry at the phosphorus and S-stereochemistry at the amino acid carbon has advantageous properties over the other three stereoisomers.
[0044] In a non-limiting embodiment, the advantageous salt (for example fumarate) of Compound I is used as a mixture of (R,S) and (S,S) diastereomers, wherein the first R/S designates the stereochemistry at the phosphorus atom and the second S is the stereochemistry of the carbon in the amino acid moiety (corresponding to the L-alanine residue having S-configuration). While any ratio of the diastereomers can be used that provides the desired results, the (R,S) diastereomer stands out. In other embodiments, the ratio is approximately 1:1 of the R to S enantiomer at the phosphorus atom. In aspects, the compound is enantiomerically enriched with the R chirality at the phosphorus atom, wherein the amount of R by weight is for example, greater than about 50%, or equal to or greater than about 60%, 70%, 75%, 80%, or even 85% or more. [0045] The S-stereoconfiguration at the chiral carbon corresponding to the natural amino acid configuration is advantageous in the present disclosure. In certain aspects, the amount of S by weight is for example, greater than about 50%, or equal to or greater than about 60%, 70%,75%, 80%, or even 85% or more. In alternative embodiments, the compound is used with R-stereoconfiguration at the chiral carbon and wherein the R-stereoconfiguration is greater than about 50%, or equal to or greater than about 60%, 70%, 75%, 80%, or even 85% or more. [0046] A pharmaceutically acceptable salt of the enantiomerically pure (Rp,Sc, or simply “R,S”) version of Compound I is a principal embodiment in the disclosed therapeutic use with
Attorney Docket No.01394-0006-01PCT retaining device therapy as disclosed herein. An enantiomerically pure Compound II is at least 90% free of the opposite enantiomer. Surprisingly, the free base parent compound is an oil, not a solid, and thus would not have been selected as the active ingredient for the topical formulation. This is especially true because the racemic mixture or enantiomerically enriched R,S with S,S as a free base is a solid. However, when formed as the fumarate salt, the R,S enantiomerically pure Compound I becomes a highly crystalline material and the most advantageous for intraepithelial topical administration. Thus, the monofumarate salt of Compound I exhibits unexpected stability, processability, and thus has therapeutic advantages over the free base Compound I. [0047] Compound II as referred to herein and illustrated below is the enantiomerically enriched or pure embodiment that has predominately R-stereochemistry at the phosphorus atom and S-stereochemistry at the amino acid carbon atom. In an enantiomerically pure form, Compound II exhibits superior stability properties over its stereoisomer, ethyl ((S)-((2-(2- amino-6-methoxy-9H-purin-9-yl)ethoxy)methyl)(benzyloxy) phosphoryl)-L-alaninate monofumarate (Compound III). This is important for the success of the topical application to the cervix or vagina.
[0048] Other advantageous salts of Compound I that have been discovered include the hemifumarate salts ethyl ((R)-((2-(2-amino-6-methoxy-9H-purin-9- yl)ethoxy)methyl)(benzyloxy)phosphoryl)-L-alaninate hemifumarate (Compound IV) and
Attorney Docket No.01394-0006-01PCT ethyl ((S)-((2-(2-amino-6-methoxy-9H-purin-9-yl)ethoxy)methyl)(benzyloxy)phosphoryl)-L- alaninate hemifumarate (Compound V).
[0049] In or acceptable salts thereof are used in a morphic form described in detail below. [0050] There are many strains of HPV, some of which are strongly associated with the development of cancer and are known as high-risk strains. In some embodiments, Compound I fumarate or Compound II can be used to treat any strain of HPV including the high-risk types of HPV, such as HPV-16 and HPV-18. Other exemplary strains include HPV-31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73, and 82. [0051] The present disclosure in certain aspects provides Compound II and in particular the isolated morphic form Compound II Pattern 1, pharmaceutical compositions containing such compound, methods of treating an HPV infection or intraepithelial neoplasia related to HPV infection using the selected morphic form described herein, and methods of preparing such compound and morphic form which is used in the therapeutic delivery with a retaining device as disclosed herein. [0052] In particular, it has been surprisingly discovered that the monofumarate salt of ethyl ((R)-((2-(2-amino-6-methoxy-9H-purin-9-yl)ethoxy)methyl)(benzyloxy)phosphoryl)-L- alaninate (Compound II) has very high tissue penetration when administered topically to the target tissue. Topical administration avoids toxicity associated with systemic administration of the drug.
Attorney Docket No.01394-0006-01PCT [0053] Because precancerous and/or cancerous cells that are infected with HPV are several layers into the epithelium, the compound must have high penetration into the tissue to reach and treat these affected cells. [0054] Compound I monofumarate has superior tissue permeation and penetration in both porcine and human vaginal tissues over ABI-1968, which failed in clinical trials despite also being a phosphonate ester of an acyclic purine nucleoside. Compound I monofumarate can reach concentrations of about 40-85 ng/mL in vaginal tissue at a 0.1% dose. ABI-1968, even when used in a 3% dose does not reach 15 ng/mL concentration (see FIG.2). The significant improvement in tissue penetration, especially considering the decrease in dose, could not have been predicted in advance. [0055] Compound I monofumarate is surprisingly stable (Example 22) in comparison to other salt forms of the compound. As shown in Table 17, Compound I monofumarate melts at 100°C, whereas the next highest melting salt, the sulfate, has a 15% lower melting point of 85°C. A higher melting point generally correlates with higher stability. [0056] Careful selection of each aspect of the disclosure in combination with the use of a retaining device to protect surrounding tissue was key to achieving the desired results. One important aspect of the disclosure is the selection of the formulation, with detailed descriptions provided herein. Nonlimiting advantageous solid dosage forms include a vaginal tablet, which can be inserted into the affected area. Alternative topical formulations as used herein include semisolid dosage forms such as gels, creams or ointments which are stable enough to have integrity at body temperature to deliver the active agent in the target area in a time period that allows deep tissue penetration. [0057] It has been discovered that Compound I or its pharmaceutically acceptable salt, or other solid form or Compound II or Compound III can be prepared in a solid dosage form for topical administration. In some embodiments, the tablet formulation provides similar tissue penetration of the gel formulation (55-85 ng/mg for gel and 44-79 ng/mg for the tablet, FIG. 6). [0058] In exemplary non-limiting embodiments, a method for the treatment of HPV infection or HPV-induced intraepithelial neoplasia is provided that comprises administering an effective amount of one or a combination of the active compounds as described herein in a topical formulation that is sufficient to treat the neoplasia in combination with the use of a retaining device as described further herein. [0059] In certain embodiments, a formulation to use in the vaginal tablet (or acceptable gel, cream or other semisolid formulation with sufficient integrity) for the treatment of HPV or an
Attorney Docket No.01394-0006-01PCT intraepithelial neoplasia, such as CIN, is a dosage form containing from about 0.01 milligram to about 0.03 milligram, from about 0.03 milligrams to about 0.25 milligrams, from about 0.20 milligrams to about 0.5 milligrams, from about 0.4 milligrams to about 1 milligram, from about 0.75 milligram to about 3 milligrams, from about 1 milligram to about 10 milligrams, from about or 5 milligrams to about 20 milligrams of the active ingredient (e.g., Compound I). [0060] The term “containing” when used in relation to a composition or dosage form, for example, means “comprising” (i.e., consisting at least in part of). When interpreting statements in this specification and claims which include the term “containing” or “comprising”, it is to be understood that other features that are additional to the features prefaced by this term in each statement or claim may also be present. Related terms such as “comprise” and “comprised” are to be interpreted in a similar manner. [0061] In certain embodiments, a formulation for the treatment of HPV or an intraepithelial neoplasia, such as CIN, is a dosage form containing about or at least 0.03, 0.05, 0.1 mg, 0.3 mg.0.5 mg, 0.7 mg, 1 mg, 1.5 mg, 2 mg, 2.5 mg, 3 mg, 4 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45mg or 50 mg of Compound I monofumarate, Compound II or Compound III. In certain embodiments, a formulation for the treatment of HPV or an intraepithelial neoplasia, such as CIN, is a dosage form containing about 0.05 mg, 0.1 mg, 0.3 mg, 1 mg, 1.5 mg, or 2 mg of Compound I monofumarate, Compound II or Compound III. As discussed above, the dose strengths in mg used herein refer to the mass of the active acyclic phosphonamidate (the free base) and do not include the salt in the molecular weight. In other words, when referring to a dosage form comprising 0.1 mg of Compound I monofumarate, for example, the 0.1 mg dose is referring to the amount of the corresponding free base. Thus, as explained earlier, when referring to a 0.1 mg dosage form of Compound I monofumarate, for example, it should be understood that the dosage form contains 0.124 mg of the monofumarate as this amount is equivalent to a dose of 0.1 mg of Compound I free base; when referring to a 0.3 mg dosage form of Compound I monofumarate, it should be understood that the dosage form contains 0.372 mg of the monofumarate as this amount is equivalent to a dose of 0.3 mg of Compound I free base; when referring to a 1 mg dosage form of Compound I monofumarate, for example, it should be understood that the dosage form contains 1.241 mg of the monofumarate as this amount is equivalent to a dose of 1 mg of Compound I free base; and so on. [0062] In certain embodiments, a particular dosage is 0.05 mg, 0.1 mg, 0.2 mg, 0.3 mg, or 1.0 mg of Compound I monofumarate, Compound II or Compound III. In certain embodiments,
Attorney Docket No.01394-0006-01PCT the 0.05 mg, 0.1 mg, 0.2 mg, 0.3 mg, or 1.0 mg dosage of Compound I monofumarate, Compound II or Compound III is administered once, twice, or three times per week as needed. In certain embodiments, the 0.1-1.0 mg dosage, such as 0.1 mg to 0.3 mg dosage of Compound I monofumarate, Compound II or Compound III is administered for the prescribed time instructed by the healthcare provider, including daily dosing, in combination with the retaining device. [0063] In certain embodiments, the compound can be administered in one or more therapeutic cycles comprising a treatment cycle and a rest cycle (also known as a treatment break), wherein the treatment cycle comprises administering the compound as described herein, followed by a rest cycle (comprising a period of no treatment) before the next treatment cycle. In certain embodiments, the rest cycle is from about one day to about six months. In certain embodiments, the rest cycle is one, two, three, four, five, six, seven, eight, or more weeks before the next treatment cycle. In certain embodiments, multiple therapeutic cycles are administered, for example one, two, three, four, five, or six therapeutic cycles. [0064] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.1 mg of the active agent (e.g., Compound I) 3 times a week for one week (0.3 mg/week). [0065] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.3 mg of the active agent (e.g., Compound I) 3 times a week for one week (0.9 mg/week). In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.3 mg of the active agent (e.g., Compound I) 3 times a week for two weeks (1.8 mg/2 weeks). In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.3 mg of the active agent (e.g., Compound I) 3 times a week for one week, followed by 0.1 mg of the active agent 3 times a week for one week (1.2 mg/2 weeks). [0066] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 1.0 mg of the active agent (e.g., Compound I) 1 time per week for three weeks. In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with a total of 3.0 mg of the active agent (e.g., Compound I) in about 2 weeks (e.g., 15 days). In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 1.0 mg of the active agent (e.g., Compound I) 1 time per week for three weeks, broken into 2 cycles. In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with a total of 6.0 mg of the active agent (e.g., Compound I) in about 6 weeks (e.g,
Attorney Docket No.01394-0006-01PCT about 43 or 44 days), broken into 2 cycles. In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 1.0 mg of the active agent (e.g., Compound I) 4 times in two weeks, broken into 2 cycles. In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with a total of 8.0 mg of the active agent (e.g., Compound I) in about 6 weeks (e.g., about 43 or 44 days), broken into 2 cycles. [0067] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.05 mg of the active agent (e.g., Compound I) 3 times a week for two weeks (0.3 mg/2 weeks). [0068] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.1 mg of the active agent (e.g., Compound I) 3 times a week for two weeks (0.6 mg/2 weeks). [0069] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 1.0 mg of the active agent (e.g., Compound I) 1 time every two weeks, broken into four cycles. In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with a total of 4.0 mg of the active agent (e.g., Compound I) in about 6 weeks (e.g., about 43 or 44 days), broken into four cycles). In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 1.0 mg of the active agent (e.g., Compound I) 1 time every two weeks, broken into three cycles. In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with a total of 3.0 mg of the active agent (e.g., Compound I) in about 6 weeks (e.g., about 43 or 44 days), broken into four cycles). [0070] In certain embodiments, the patient with an HPV infection or an intraepithelial neoplasia, such as CIN, administers a tablet comprising a monofumarate salt of the active agent (e.g., Compound I monofumarate salt, Compound II, or Compound III). In certain embodiments, the patient administers the tablet by means of a retaining device, which is inserted to, for example, collect post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG. In certain embodiments, the patient administers the tablet by means of an applicator that is removed before insertion of a retaining device. [0071] Dosage forms which do not adhere well to the target site may be dislodged, interfering with treatment. The present disclosure provides dosage forms that adhere to the target site and dissolve rapidly in low fluid volumes. Adhesion to the target site also minimizes exposure to non-target tissues, which may limit toxicity, unwanted systemic
Attorney Docket No.01394-0006-01PCT exposure, and side effects, especially when used in combination with the retaining device. Dosage forms which soften, break down, and/or disintegrate quickly in low fluid volumes are advantageous to cause a rapid release of the active compound to the target tissue. Dosage forms that disintegrate in, for example, less than about 50 µL, less than about 100 µL, less than about 125 µL, less than about 150 µL, less than about 175 µL, less than about 200 µL, less than about 250 µL, less than about 500 μL, less than about 1 mL, or less than about 2 mL fluid facilitate drug penetration into the target site. [0072] In certain embodiments, the dosage form is a vaginal tablet. In certain embodiments, the dosage form is a semisolid such as a gel or cream. [0073] In certain embodiments, the dosage form disintegrates in about one to about ten seconds. In certain embodiments, the dosage form disintegrates in about ten seconds to about one minute. In certain embodiments, the dosage form disintegrates in about one minute to about one hour. In certain embodiments, the dosage form disintegrates in about one hour to about six hours. [0074] The physical dimensions of the dosage form can impact the effectiveness of the dosage form. A tablet that is thinner provides a greater surface area to volume ratio and may degrade quicker and cover the target area better. In certain embodiments, the dosage form is less than 3 millimeters thick in its smallest dimension. [0075] The formulation of the dosage form may be important for adequate administration of the active agent into the intraepithelial tissue. Tablet formulations should display the properties of mucoadhesion and substantivity and include excipients that have solubilizing, erosion-generating (for disintegration), porosity (for water uptake) and viscosity enhancing (to keep the drug at the target site) properties. [0076] Examples of excipients that will cause rapid disintegration of a solid dosage form to cover the cervix or vaginal areas include, but are not limited to mannitol, microcrystalline cellulose, lactose, sucrose, calcium phosphate, sodium phosphate, sodium bicarbonate, citric acid, maleic acid, adipic acid or fumaric acid. Examples of excipients that can enhance disintegration and coverage of the affected area include but are not limited to sodium starch glycollate, pregelatinized starch, crospovidone and croscarmellose sodium. Mucoadhesive excipients that are useful in the present disclosure include but are not limited to microcrystalline cellulose, polycarbophil, hydroxymethyl cellulose, hypromellose, hydroxypropyl cellulose, and PVP. [0077] A nonlimiting example of a tablet formulation includes, but is not limited to, microcrystalline cellulose, crospovidone, magnesium stearate, silicon dioxide, polyethylene
Attorney Docket No.01394-0006-01PCT oxide and mannitol. Another non-limiting example of a tablet formulation has microcrystalline cellulose, magnesium stearate and mannitol. [0078] Semi solid dosage forms may include, for example, a mucoadhesive polymer, a solubility/penetration enhancer, a lipophilic solubilizer and a penetration enhancer. The mucoadhesive polymer, for example, may be, but is not limited to, a carbomer, polyethylene glycol, crospovidone, hypromellose, polycarbophil and/or hydroxyethyl cellulose. The solubility/penetration enhancer can be, for example, but not limited to, a mixture of polyoxyl 6 stearate Type I, ethylene glycol stearate and polyoxy 32 stearate Type I, cetyl alcohol, stearyl alcohol, polysorbate 80, sodium lauryl sulphate, mono and di-glycerides, sorbitan monostearate, glyceryl isostearate, polyoxy 15 hydroxystearate, poly15 hydroxystearate, polyoxy 40 hydrogenated castor oil, octyl dodecanol, and/or soybean lecithin. Lipophilic solubilizers include, but are not limited to light mineral oil, mineral oil, white wax and silicone fluid. Penetration enhancers include but are not limited to propylene glycol, transcutol, oleic acid, isopropyl myristate, propylene glycol glycerol monooleate, propylene glycol monocaprylate, PEG-8 Bees wax, cetyl alcohol, stearic acid, cetyl palmitate and/or cetostearyl alcohol. [0079] A non-limiting example of a semi-solid formulation comprises, for example, one or more of a carbomer, propylene glycol, sorbic acid, EDTA and water. Another non-limiting example of a semi solid formulation comprises one or more of a carbomer, mineral oil, a mixture of polyoxy 6 stearate Type I, ethylene glycol stearate, polyoxy 32 stearate Type I, parabens, propylene glycol, EDTA and/or water. [0080] Films can be produced, for example, with, but not limited to, hypromellose, polyethylene glycol, polymethacrylates, microcrystalline cellulose, xanthan gum, guar gum and/or polyvinylpyrrolidone. [0081] A pessary (vaginal suppository) can be formulated with, for example but not limited to, hard fat (such as Ovucire, Witepsol, Supposi-Base), polyethylene glycol, macrogols, cocoa butter and glycerol. A non-limiting example of a pessary can be made from Witepsol H 15 or Ovucire WL 3264. [0082] Therefore, the present disclosure includes at least the following features: (i) A method to treat a cervical or vaginal HPV-induced infection or an associated condition in a female in need thereof, including but not limited to cervical or vaginal intraepithelial neoplasia, comprising administering to a host in need thereof an effective amount of Compound I or a pharmaceutically acceptable salt thereof such as the monofumarate or hemifumarate, or other solid form, or Compound II or Compound III, in a pharmaceutical
Attorney Docket No.01394-0006-01PCT composition, in a vaginal tablet or semisolid formulation with a vaginal applicator in combination with the use of a retaining device for sufficient time to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue. (ii) A method to treat a cervical or vaginal HPV-induced infection, intraepithelial neoplasia, or an associated condition in a human in need thereof, the method comprising (i) administering an effective amount of the anti-HPV therapeutic agent Compound I or a pharmaceutically acceptable salt thereof, such as the monofumarate or hemifumarate, or other solid form or Compound II or Compound III typically in tablet form, to the cervix using a vaginal applicator (which is optionally coated with a lubricant) and then (ii) inserting a retaining device (which is optionally coated with a lubricant) that is maintained at the base of the cervix or in the vaginal canal for sufficient time to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue. (iii) The method of (i) or (ii), wherein the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap and a sponge. (iv) The method of (i) or (ii) wherein the retaining device is used as both the applicator and a retaining means in a manner that minimizes toxicity-causing damage to non- diseased tissue. (v) The method of (i) or (ii) wherein the retaining device is a menstrual cup. (vi) The method of (i) or (ii) wherein the retaining device is a menstrual disc. (vii) The method of (i) or (ii) wherein the retaining device is a tampon. (viii) The method of (i), (ii), or (iv) wherein the retaining device is a diaphragm. (ix) The method of (i) or (ii) wherein the retaining device is a cervical cap. (x) The method of (i) or (ii) wherein the retaining device is a sponge. (xi) The method of (v)-(x) wherein the retaining device is used as both an applicator and a retaining means. (xii) The method of (i)-(xi) wherein the tablet or semisolid formulation contains of from 0.1 mg to 30 mg, from 0.05 to 0.3 mg, from 0.5 mg to 20 mg, from 1 mg to 20 mg, from 1 mg to 15 mg, from 1 mg to 10 mg of the active agent and in certain embodiments, about or at least 0.03, 0.05, 0.1 mg, 0.3 mg.0.5 mg, 0.7 mg, 1 mg, 1.5 mg, 2 mg, 2.5 mg, 3 mg, 4 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45mg or 50 mg of the active agent.
Attorney Docket No.01394-0006-01PCT (xiii) The method of (i)-(xi) wherein the tablet or semisolid formulation contains from about 0.05 mg to about 1.0 mg, such as 0.1 to 0.5 mg, for example 0.2, 0.3 or 0.4 mg of the active agent. (xiv) The method of (i)-(xiii) wherein the retaining device is held in place for between 4-12 hours, 5-10 hours, 6-8 hours, or 6-9 hours and then removed. (xv) The method of (i)-(xiv) wherein the tablet or semisolid formulation with retaining device is administered once a day for one, two, three or four days a week for as long as necessary to achieve the desired results. (xvi) The method of (i)-(xiv) wherein the tablet or semisolid formulation with retaining device is administered three time a week for as long as necessary to achieve the desired results. (xvii) The method of (xv)-(xvi) wherein the tablet or semisolid formulation with retaining device is administered for 2, 3, 4, 5, or 6 weeks. (xviii) The method of (i)-(xvii), wherein the retaining device and/or vaginal applicator is first lubricated with a water-based gel such as for example, a glycerol-based lubricant, hydroxyethylcellulose-based lubricant, KY jelly or Surgilube. (xix) The method of (i)-(xviii) wherein a water-based gel is inserted into the vaginal area up to the cervix prior to vaginal tablet administration. (xx) The method of (i)-(xix) wherein a lubricant is coated on the retaining device and/or vaginal applicator for ease of placement as well as to help maintain the device in position before inserting. (xxi) The method of (i)-(xx) wherein the vaginal applicator and the retaining device are combined into one device for both administration of the vaginal tablet and retaining the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue (xxii) The method of (xx), wherein the lubricant is selected from water, a glycerol- based lubricant and a hydroxyethylcellulose-based lubricant. (xxiii) The methods of any of the above, wherein the active compound is delivered in a morphic form as described herein. (xxiv) A kit that comprises a dosage form with a therapeutic compound as described herein and a retaining device. (xxv) The kit of (xxiv) that further comprises a vaginal applicator. (xxvi) The kit of (xxiv) or (xxv) wherein the dosage form is a vaginal tablet.
Attorney Docket No.01394-0006-01PCT (xxvii) The kit of (xxiv)-(xxvi) wherein the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap and a sponge. (xxviii) The kit of (xxv), wherein the retaining device is used as both the vaginal applicator and a retaining means. BRIEF DESCRIPTION OF THE FIGURES [0083] FIG.1A and FIG 1B depict the molecular structure of Compound I monofumarate pattern 1 as determined by the single crystal X-ray diffraction analysis of Example 21. There exists an intermolecular interaction between protonated N5-atom of free base and O7-atom of fumaric acid anion (N(5)–H(5)···O(7)) in the single-crystal form of Compound II. [0084] FIG.2 is an in vitro tissue permeation test in vaginal tissue comparing Compound I monofumarate salt to ABI-1968. Bar A shows the tissue penetration of a 0.1% Compound I monofumarate gel in porcine vaginal tissue. Bars B and C show the tissue penetration of a 0.1% Compound I monofumarate gel in human cervical tissue. Bar D shows the tissue penetration of a 1% formulation of ABI-1968 in 6% NMP into porcine vaginal tissue. Bar E shows the tissue penetration of a 1% nanosuspension of ABI-1968 in porcine vaginal tissue. Bar F shows the tissue penetration of a 3% formulation of ABI-1968 in 6% NMP into porcine vaginal tissue. Bar G shows the tissue penetration of a 3% formulation of ABI-1968 in 20% NMP into porcine vaginal tissue. ABI-1968 penetrates the tissue to a substantially smaller degree, which hinders the ability of the compound to reach the cells which are infected with HPV. This may be a contributing factor to the performance of ABI-1968 in clinical studies. Surprisingly, Compound I and/or its monofumarate salt displays high tissue penetration in both porcine and human tissues. High tissue penetration may lead to high activity against HPV. This is described in Example 15. [0085] FIG.3 shows a flow diagram for the process of preparing a topical cream formulation described in Example 6. [0086] FIG.4 shows a flow diagram for the process of preparing a topical gel formulation described in Example 6. [0087] FIG.5 shows a flow diagram for the process of preparing a tablet formulation described in Example 7. [0088] FIG.6 is a bar graph comparing the tissue penetration of a topical gel and a topical tablet dosage form as described in Example 15. The tablet dosage form produces similar tissue penetration to the topical gel, with an average of 58 ng/mg of compound in the tissue.
Attorney Docket No.01394-0006-01PCT [0089] FIGS.7A, 7B, 7C, 7D, 7E, 7F, 7G, 7H, 7I, and 7J depict representative administration procedures to insert a vaginal tablet as described in Example 20. [0090] FIG.8 depicts the structure of Compound I monofumarate. The synthesis of Compound I and stereoisomers and pharmaceutically acceptable salts thereof is provided in Examples 1-5. [0091] FIG.9 is a comparison of XRPD diffractograms of Compound I hemifumarate Pattern 1 and Compound I monofumarate Pattern 1 (small scale preparation) obtained in Example 23. [0092] FIG.10 is a DSC thermogram of Compound I hemifumarate Pattern 1 obtained in Example 23. [0093] FIG.11 is a TGA thermogram of Compound I hemifumarate Pattern 1 obtained in Example 23. [0094] FIG.12A is a DSC thermogram of Compound I monofumarate Pattern 1 (small scale preparation, Example 23), recorder at heating rate of 10°C/min. [0095] FIG.12B is a DSC thermogram of Compound I mono-fumarate Pattern 1 (small scale preparation, Example 23), recorder at heating rate of 2°C/min. [0096] FIG.12C is a DSC cycle of Compound I mono-fumarate Pattern 1 (DSC cycle, 0- 150ºC, 150-0ºC, 0-250ºC, 10ºC/min) for the small-scale preparation sample (Example 23). [0097] FIG.13 is TGA thermogram of Compound I mono-fumarate Pattern 1 for the small- scale preparation sample (Example 23). [0098] FIG.14 is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 obtained in stability test experiments at 25°C/84%RH (2 days, open container), 25°C/92%RH (1 week, open container), 40°C/75%RH (1 week, open container), and 60°C (1 week, tight container) as described in Example 24 with original sample of Compound I monofumarate Pattern 1 before the test. [0099] FIG.15 is a Dynamic Vapor Sorption (DVS) plot and DVS change in mass plot for Compound I Pattern 1(Example 26). [00100] FIG.16 is a comparison of XRPD diffractograms of Compound I Pattern 1 before and after DVS study (Example 26). [00101] FIG.17 is a Dynamic Vapor Sorption (DVS) plot and DVS change in mass plot for Compound I hemifumarate Pattern 1 (Example 26). [00102] FIG.18 is a comparison of XRPD diffractograms of Compound I hemifumarate Pattern 1 before and after DVS study (Example 26). [00103] FIG.19 is a Dynamic Vapor Sorption (DVS) plot and DVS change in mass plot for Compound I monofumarate Pattern 1 (Example 26).
Attorney Docket No.01394-0006-01PCT [00104] FIG.20 is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 before and after DVS study (Example 26). [00105] FIG.21A is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 obtained in Example 27 (large scale) before and after heating to 106°C. [00106] FIG.21B is a DSC thermogram of Compound I monofumarate Pattern 1 (Example 27), recorded at heating rate of 10°C/min. [00107] FIG.21C is a DSC thermogram of Compound I monofumarate Pattern 1 (large scale preparation, Example 27), recorded at heating rate of 2°C/min. [00108] FIG.21D is a DSC thermogram of Compound I monofumarate Pattern 1 (obtained in Example 27). [00109] FIG.22 is TGA thermogram of Compound I monofumarate Pattern 1 (obtained in Example 27). [00110] FIG.23 is a comparison of XRPD diffractograms of Compound I mono- fumarate Pattern 1, hemi-fumarate Pattern 2, hemi-fumarate Pattern 3, Pattern 4 obtained from equilibration experiment in water at 25°C for 2 weeks and fumaric acid pattern (obtained in Example 28). [00111] FIG.24 is a comparison of XRPD diffractograms of Compound I Pattern 4 obtained from equilibration experiment in water at 25°C for 2 and 3 weeks (obtained in Example 28). [00112] FIG.25 is a DSC thermogram of Compound I Pattern 4 obtained from equilibration experiment in water at 25°C for 2 weeks (obtained in Example 28). [00113] FIG.26 is a DSC thermogram of Compound I Pattern 4 obtained from equilibration experiment in water at 25°C for 3 weeks (obtained in Example 28). [00114] FIG.27 is a TGA thermogram of Compound I Pattern 4 obtained from equilibration experiment in water at 25°C for 2 weeks (obtained in Example 28). [00115] FIG.28 is a TGA thermogram of Compound I Pattern 4 obtained from equilibration experiment in water at 25°C for 3 weeks (obtained in Example 28). [00116] FIG.29 is a comparison of XRPD diffractograms of fumaric acid pattern and Compound I hemifumarate Pattern C obtained in equilibration experiment EQ2 (in acetonitrile) and EQ15 (in 2.9:97.1 v/v water/ acetonitrile) at 25°C for 3 weeks (obtained in Example 28).
Attorney Docket No.01394-0006-01PCT [00117] FIG.30 is a DSC thermogram of Compound I hemi-fumarate Pattern C obtained from equilibration experiment in acetonitrile at 25°C for 2 weeks (obtained in Example 28). [00118] FIG.31 is a TGA thermogram of Compound I hemi-fumarate Pattern C obtained from equilibration experiment in acetonitrile at 25°C for 2 weeks (obtained in Example 28). [00119] FIG.32 is a comparison of XRPD diffractograms of Compound I hemi- fumarate Pattern 2 obtained in equilibration experiments EQ3 (in methyl ethyl ketone), EQ4 (in acetone), and EQ7 (in 1:1 v/v acetone/heptane) at 25°C for 3 weeks (obtained in Example 28). [00120] FIG.33 is a DSC thermogram of Compound I hemi-fumarate Pattern 2 obtained in equilibration experiment in methyl ethyl ketone at 25°C for 2 weeks (obtained in Example 28). [00121] FIG.34 is a TGA thermogram of Compound I hemi-fumarate Pattern 3 obtained in equilibration experiment in methyl ethyl ketone at 25°C for 2 weeks (obtained in Example 28). [00122] FIG.35 is a comparison of XRPD diffractograms of fumaric acid and a mixture of Compound I monofumarate Pattern 1 and unknown pattern obtained in equilibration experiments EQ5 (in isopropanol), EQ8 (in 1:1 v/v isopropanol /heptane), EQ9 (in 1:1 v/v isopropanol/toluene), EQ10 (in 1:3 v/v isopropanol/methyl tertbutyl ether), and EQ12 (in 1:3 v/v ethanol/heptanes) at 25°C for 3 weeks (obtained in Example 28). [00123] FIG.36 is a DSC thermogram of a mixture of Compound I mono-fumarate Pattern 1 and unknown pattern obtained in equilibration experiment EQ5 in isopropanol at 25°C for 2 weeks (obtained in Example 28). [00124] FIG.37 is a TGA thermogram of a mixture of Compound I mono-fumarate Pattern 1 and unknown pattern obtained in equilibration experiment EQ5 in isopropanol at 25°C for 2 weeks (obtained in Example 28). [00125] FIG.38 is a comparison of XRPD diffractograms of Compound I hemi- fumarate Pattern 5 obtained in equilibration experiment EQ6 (in 1:1 v/v acetone/toluene) at 25°C for 2 and 3 weeks (obtained in Example 28). [00126] FIG.39 is a DSC thermogram of Compound I hemi-fumarate Pattern 5 obtained in equilibration experiment EQ6 (in 1:1 v/v acetone/toluene) at 25°C for 2 weeks (obtained in Example 28).
Attorney Docket No.01394-0006-01PCT [00127] FIG.40 is a DSC thermogram of Compound I hemi-fumarate Pattern 5 obtained in equilibration experiment EQ6 (in 1:1 v/v acetone/toluene) at 25°C for 3 weeks (obtained in Example 28). [00128] FIG.41 is a TGA thermogram of Compound I hemi-fumarate Pattern 5 obtained in equilibration experiment EQ6 (in 1:1 v/v acetone/toluene) at 25°C for 2 weeks (obtained in Example 28). [00129] FIG.42 is a TGA thermogram of Compound I hemi-fumarate Pattern 5 obtained in equilibration experiment EQ6 (in 1:1 v/v acetone/toluene) at 25°C for 3 weeks (obtained in Example 28). [00130] FIG.43 is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 obtained in equilibration experiments (procedure in Example 28) EQ11 (in 1:3 v/v tetrahydrofuran/heptane), EQ13 (in 1:3 v/v ethyl acetate/toluene), EQ14 (in 1:3 v/v ethanol/toluene) at 25°C for 2 weeks and Compound I monofumarate Pattern 1 (material obtained in Example 23). [00131] FIG.44 is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 obtained in equilibration experiments EQ11 (in 1:3 v/v tetrahydrofuran/heptane), EQ13 (in 1:3 v/v EA/toluene), EQ14 (in 1:3 v/v ethanol/toluene) at 25°C for 3 weeks (obtained in Example 28). [00132] FIG.45 is a comparison of XRPD diffractograms of a mixture of Compound I monofumarate Pattern 1 and unknown pattern obtained in equilibration experiments EQ16 (in 1:4 v/v isopropanol/heptane), EQ5 (in isopropanol) (obtained in Example 28) at 25°C for 2 weeks and Compound I monofumarate Pattern 1 obtained in Example 23. [00133] FIG.46 is a comparison of XRPD diffractograms of reference fumaric acid pattern, Compound I monofumarate Pattern 1 (Example 23), Compound I monofumarate Pattern 2 (obtained by precipitation from acetone solution with heptane antisolvent in Experiment AS1, Example 28), Compound I monofumarate Pattern 2 (obtained by precipitation from methyl ethyl ketone solution with heptane antisolvent in Experiment AS4, Example 28), and fumaric acid pattern obtained in Experiment AS2, Example 28. [00134] FIG.47 is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 (Example 23), Compound I monofumarate Pattern 2 (obtained by precipitation from ethanol solution with heptanes antisolvent in Experiment AS6, Example 28) and Compound I monofumarate Pattern 2 (obtained by precipitation from tetrahydrofuran solution with heptane antisolvent in Experiment AS7, Example 28).
Attorney Docket No.01394-0006-01PCT [00135] FIG.48 is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 (Example 23) and Compound I monofumarate Pattern 1 obtained by crystallization at room temperature by slow evaporation from acetone, methylethylketone and ethyl acetate as described in Example 28, Table 41. [00136] FIG.49 is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 (Example 23) and Compound I monofumarate Pattern 1 obtained by crystallization at room temperature by slow evaporation from methanol, ethanol, isopropanol and tetrahydrofuran as described in Example 28, Table 41. [00137] FIG. 50 is a comparison of XRPD diffractograms of reference fumaric acid pattern, Compound I monofumarate Pattern 1 (Example 23) and Compound I monofumarate Pattern 2 obtained by crystallization from hot methyl ethyl ketone saturated solution by slow cooling, Compound I monofumarate Pattern 2 obtained by crystallization from acetone hot saturated solution by slow cooling and Compound I monofumarate Pattern 3 obtained by crystallization from acetonitrile hot saturated solution by slow cooling. [00138] FIG.51 is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 (Example 23), Compound I monofumarate Pattern 1 obtained by crystallization from hot water saturated solution by slow cooling, and Compound I monofumarate Pattern 1 obtained by crystallization from hot saturated solution in ethanol/toluene (1/1 v/v) by slow cooling (Example 28, Table 42). [00139] FIG.52 is a comparison of XRPD diffractograms of Compound I monofumarate Pattern 1 (Example 23), Compound I monofumarate Pattern 2 obtained by crystallization from hot acetone saturated solution by fast cooling, and Compound I monofumarate Pattern 2 obtained by crystallization from hot saturated solution in methyl ethyl ketone by fast cooling (Example 28, Table 43). [00140] FIG. 53 is a comparison of XRPD diffractograms of reference fumaric acid pattern, Compound I monofumarate Pattern 1 (Example 23), Compound I monofumarate Pattern 1 obtained by crystallization from hot water saturated solution by fast cooling, Compound I monofumarate Pattern 3 obtained by crystallization from hot saturated solution in acetonitrile by fast cooling, and Compound I monofumarate Pattern 1 obtained by crystallization from hot saturated solution in ethanol/toluene (1/1 v/v) by fast cooling (Example 28). [00141] FIG. 54 is a heat-cool-heat DSC thermogram of Compound I monofumarate Pattern 1 (heating to 106°C) (Example 28, Table 44).
Attorney Docket No.01394-0006-01PCT [00142] FIG. 55 is a heat-cool-heat DSC thermogram of Compound I monofumarate Pattern 1 (heating to 130°C) (Example 28, Table 44). [00143] FIG.56 is an XRPD diffractogram of Compound II Pattern 1 (Example 29). [00144] FIG.57 is a DSC thermogram of Compound II Pattern 1 (Example 29). [00145] FIG.58 is a TGA thermogram of Compound II Pattern 1 (Example 29). [00146] FIG.59 is an XRPD diffractogram of Compound III Pattern 1 (Example 30). [00147] FIG.60 is a DSC thermogram of Compound III Pattern 1 (Example 30). [00148] FIG.61 is a TGA thermogram of Compound III Pattern 1 (Example 30). [00149] FIG.62 is an XRPD diffractogram of Compound III Pattern 2 (Example 31). [00150] FIG.63 is a DSC thermogram of Compound III Pattern 2 (Example 31). [00151] FIG.64 is a TGA thermogram of Compound III Pattern 2 (Example 31). DETAILED DESCRIPTION [00152] It has been discovered that an effective composition for the treatment of cervical or vaginal HPV infection and cervical or vaginal intraepithelial neoplasia, requires the combination of the selection of a number of aspects that work together to achieve the desired results. It was essential to select the right compound with advantageous lipophilic and tissue penetrating properties combined with a selected pharmaceutically acceptable salt optionally in an advantageous morphic form to achieve the long-sought ability to penetrate the deep epithelial stratified tissues in an effective amount to deliver the active agent. It required years of research to solve this problem, after many failures, to the benefit of patients globally suffering from interepithelial neoplasia that may become cancerous. In addition, an aspect of successful treatment is the use of a retaining device to minimize potential adverse effects on surrounding tissue that can be uncomfortable or even painful for the patient and possibly damaging to the tissue. Thus, the disclosure includes a device system and advantageous therapeutic compound administration to effectively treat HPV cervical or vaginal HPV infection or cervical or vaginal intraepithelial neoplasia. [00153] It has also been discovered that a particularly advantageous treatment for cervical or vaginal HPV infection or cervical or vaginal intraepithelial neoplasia is achieved by the administration of an effective amount of the PMEG prodrug Compound I or a pharmaceutically acceptable salt thereof, such as the hemifumarate or monofumarate salts, including in particular Compound II or Compound III, which may be in a morphic form as described further herein, by steps comprising (i) administering an effective amount of the
Attorney Docket No.01394-0006-01PCT anti-HPV therapeutic agent Compound I (or a pharmaceutically acceptable salt thereof) or Compound II or III, typically in solid tablet form, to the cervix using a vaginal applicator and then (ii) inserting a retaining device that is maintained at the base of the cervix or in the vaginal canal for sufficient time to collect the post-treatment leakage of vaginal fluids and the remaining therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue. In certain embodiments, the retaining device is separate from the applicator. In certain embodiments, the retaining device is both the applicator and the retaining means. [00154] In some embodiments, the retaining device is inserted immediately after administration of the anti-HPV therapeutic agent. In some embodiments, the retaining device is inserted from about zero hours to about one hour after administration of the anti-HPV therapeutic agent. In some embodiments, the retaining device is inserted one hour or more after administration of the anti-HPV therapeutic agent. In some embodiments, the retaining device is inserted about 1-10 hours, about 1-9 hours, or about 1-8 hours after administration of the anti-HPV therapeutic agent. In some embodiments, the retaining device is inserted about 2-10 hours, about 2-9 hours, or about 2-8 hours after administration of the anti-HPV therapeutic agent. In some embodiments, the retaining device is inserted about 3-10 hours, about 3-9 hours, or about 3-8 hours after administration of the anti-HPV therapeutic agent. In some embodiments, the retaining device is inserted about 4-10 hours, about 4-9 hours, or about 4-8 hours after administration of the anti-HPV therapeutic agent. In some embodiments, the retaining device is inserted about 5-10 hours, about 5-9 hours, or about 5-8 hours after administration of the anti-HPV therapeutic agent. In some embodiments, the retaining device is inserted about 6-10 hours, about 6-9 hours, or about 6-8 hours after administration of the anti-HPV therapeutic agent. [00155] In some embodiments, the retaining device is held in place for 3-10 hours, for example 4-8 hours or 5-7 hours or about or at least around 6 hours. In some embodiments, the retaining device is held in place for 6-8 hours. In some embodiments, the retaining device is left in place overnight. The retaining device is then removed. In some embodiments, the retaining device is used after every topical application of the anti-HPV therapeutic agent to prevent undesired PMEG toxicity to surrounding tissue or cells. The retaining device can be, in non-limiting embodiments, a retaining period device, a birth control device, or other type of device that adequately separates the cervix from the vagina, or part of the vagina. Alternatively, it can be any device that forms an adequate barrier between the inserted tablet
Attorney Docket No.01394-0006-01PCT and surrounding vaginal tissue for a time period of at least 6 hours, and more typically, 7, 8, 9, 10, 11 or 12 hours, for example for 6-8 hours. [00156] In certain embodiments, the retaining device is both the applicator and the retaining means. In this embodiment, the vaginal applicator and retaining device are combined into one device that can be used to both insert the vaginal tablet (which can also be called a vaginal insert) and act as a retaining device to protect surrounding tissue. In principal embodiments, the combined device and applicator is similar in structure to a diaphragm with a contoured surface that the vaginal tablet can be placed in for administration, and the tablet can be held in place with a lubricating fluid or other appropriate means. [00157] In certain embodiments, the retaining device and/or the vaginal applicator is intended for use of vaginal contact for about 24 hours or less. [00158] In certain embodiments, the retaining device and/or vaginal applicator has a barrel length of from about 3 to about 6 inches, such as from about 3.5 to about 5.5 inches or from about 4 to about 5 inches. In certain embodiments, the barrel length of the applicator or retaining device is from about 4.50 to about 4.65, such as from 4.52 to about 4.60 inches. In certain embodiments, the barrel length of the applicator or retaining device is 4.6 ± 0.2 inches. [00159] In certain embodiments, the retaining device and/or vaginal applicator comprises a plunger and barrel. In certain embodiments, the separation force between the plunger and barrel is greater than or equal to 0.2 N. In certain embodiments, the separation force between the plunger and barrel is from about 0.2 N to about 10.0 N, such as from about 0.2 N to about 9.0 N, from about 0.2 N to about 8.5 N, from about 0.2 N to 8.0 N, from about 0.2 N to about 7.0 N, from about 0.2 N to about 6.0 N, from about 0.2 N to about 5.0 N, from about 0.2 N to about 4.0 N, from about 0.2 N to about 3.0 N, from about 0.2 N to about 2.0 N, or from about 0.2 N to about 1.0 N. [00160] In certain embodiments, when the retaining device and/or vaginal applicator comprises a plunger and barrel, the force to load the vaginal tablet into the barrel such that the tablet is retained when the applicator or retaining device is held vertically, tablet side down, is less than or equal to 12.0 N. In certain embodiments, the force to load the tablet is from about 1.0 N to about 12.0 N, from about 2.0 N to about 12.0 N, from about 3.0 N to about 12.0 N, from about 4.0 N to about 12.0 N, from about 5.0 N to about 12.0 N, from about 6.0 N to about 12.0 N, from about 7.0 N to about 12.0 N, from about 8.0 N to about 12.0 N, from about 9.0 N to about 12.0 N, from about 10.0 N to about 12.0 N, or from about 11.0 N to about 12.0 N.
Attorney Docket No.01394-0006-01PCT [00161] In certain embodiments, when the retaining device and/or vaginal applicator comprises a plunger and barrel, the force on the plunger to deliver the tablet must be less than or equal to 12 N. In certain embodiments, the force on the plunger to deliver the tablet is from about 1.0 N to about 12.0 N, from about 2.0 N to about 12.0 N, from about 3.0 N to about 12.0 N, from about 4.0 N to about 12.0 N, from about 5.0 N to about 12.0 N, from about 6.0 N to about 12.0 N, from about 7.0 N to about 12.0 N, from about 8.0 N to about 12.0 N, from about 9.0 N to about 12.0 N, from about 10.0 N to about 12.0 N, or from about 11.0 N to about 12.0 N. [00162] In an alternative embodiment, instead of using a vaginal tablet, a gel or cream formulation with the active therapeutic, as described further below, is administered and then used in combination with the retaining device. While the vaginal tablet is preferred for its solid structure and physical properties, a gel or cream with enough rigidity to deliver the active therapeutic and not unduly escape to surrounding tissue would work. [00163] In certain embodiments, the vaginal applicator and/or retaining device is first lubricated with a water-based gel such as for example, a glycerol-based lubricant, hydroxyethylcellulose-based lubricant, KY jelly or Surgilube to help maintain the vaginal tablet in place during application and for patient comfort. The lubricant may help the vaginal tablet remain in place for effective disintegration. Alternatively or in addition, a water-based gel can also be inserted into the vaginal area up to the cervix prior to vaginal tablet administration. In addition or alternatively, the lubricant can be coated on the retaining device for ease of placement as well as to help maintain the device in position. [00164] As noted earlier, it was discovered that the key compound for delivery of the active agent is a specific salt of:
. [00165] Compound I is (ethyl(((2-(2-amino-6-methoxy-9H-purin-9- yl)ethoxy)methyl)(benzyloxy)-phosphoryl)-L-alaninate). Compound I is an acyclic nucleotide phosphonate that metabolizes to a known potent antiviral compound (PMEG; ((9-
Attorney Docket No.01394-0006-01PCT [2-phosphonomethyoxy)ethyl)guanine])) but has poor cellular permeability and use-limiting systemic toxicity. Applicant has discovered how to improve the prodrug to be delivered topically in a manner that it is rapidly taken up into epithelial cells, a challenging task to date and one that ABI-1968 failed. [00166] Compound I (ethyl (((2-(2-amino-6-methoxy-9H-purin-9-yl)ethoxy)methyl)- (benzyloxy)phosphoryl)-L-alaninate) has two chiral centers, one at the phosphorus atom and one in the amino acid moiety, either of which can be in the R or S stereoconfiguration. Therefore, Compound I has four stereoisomers. While U.S. Patent Nos.9,801,884 and 11,344,555 describe Compound I generally, the patents do not address the potential stereochemistry of the phosphorus atom. It has been discovered that the stereoisomer of Compound I with R-stereochemistry at the phosphorus and S-stereochemistry at the amino acid carbon has advantageous properties over the other three stereoisomers, as discussed further herein. [00167] In a non-limiting embodiment, the advantageous salt (for example fumarate) of Compound I is used as a mixture of (R,S) and (S,S) diastereomers, wherein the first R/S designates the stereochemistry at the phosphorus atom and the second S is the stereochemistry of the carbon in the amino acid moiety (corresponds to the L-alanine residue having S-configuration). While any ratio of the diastereomers can be used that provides the desired results, the (R,S) diastereomer stands out. In other embodiments, the ratio is approximately 1:1 of the R to S enantiomer at the phosphorus atom. In certain aspects, the compound is enantiomerically enriched with the R enantiomer at the phosphorus atom, wherein the amount of R by weight is for example, greater than about 50%, or equal to or greater than about 60%, 70%, 75%, 80%, or even 85% or more. [00168] The S-stereoconfiguration at the chiral carbon corresponding to the natural amino acid configuration is advantageous in the present disclosure. In some aspects, the amount of S by weight is for example, greater than about 50%, or equal to or greater than about 60%, 70%, 75%, 80%, or even 85% or more. In alternative embodiments, the R- stereoconfiguration of the chiral carbon is predominant and is greater than about 50%, or equal to or greater than about 60%, 70%, 75%, 80%, or even 85% or more, [00169] In principal aspects, therefore, the disclosure includes the administration of an effective amount of the fumarate salt of (RP, SC) ethyl (((2-(2-amino-6-methoxy-9H-purin-9- yl)ethoxy)methyl)(benzyloxy)-phosphoryl)-L-alaninate (Compound II) as described herein, optionally in a pharmaceutically acceptable carrier. The present disclosure provides a
Attorney Docket No.01394-0006-01PCT pharmaceutical salt of an acyclic nucleotide, methods, compositions, and dosage forms for the treatment of diseases associated with human papilloma virus (HPV).
[00170] The may used in combination with a retaining device to treat conditions related to or occurring as a result of an HPV viral exposure or infection. For example, the active compound can be used to treat precancerous cervical lesions, cervical intraepithelial neoplasia, vaginal intraepithelial neoplasia, cervical cancer and vaginal cancer. [00171] The active compounds and compositions with a retaining device can be used to treat an infection caused by any strain of HPV. Most of the cancer-causing HPV types are from the alpha-7 and alpha-9 species including types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73, and 82. The most common cancer-causing HPV types are 16 and 18. HPV-16 and HPV-18 are reported to be the cause of 50% of cervical cancers. And 90% of venereal warts are caused by HPV-6 and HPV-11 (World Health Organization, “Cervical Cancer” https://www.who.int/news-room/fact-sheets/detail/cervical-cancer). Infection with one type of genotype does not preclude a later infection with a different genotype. [00172] In some embodiments, Compound I monofumarate, Compound II or Compound III is used to treat HPV in combination with the retaining device. In some embodiments, Compound I monofumarate, Compound II or Compound III is used to treat HPV-16. In one embodiment, Compound I monofumarate, Compound II or Compound III is used to treat HPV-18. In one embodiment, Compound I monofumarate, Compound II or Compound III is used to treat a high risk HPV infection. In one embodiment, Compound I monofumarate, Compound II or Compound III is used to treat HPV type 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73, or 82. [00173] In some embodiments, the compound, formulations, or solid dosage forms that comprise an active compound described herein can also be used prophylactically to prevent
Attorney Docket No.01394-0006-01PCT or retard the progression of clinical illness in individuals who are HPV positive or who have been exposed to HPV. [00174] In particular, it has been discovered that Compound II, optionally in a morphic form as described herein, exhibits superior drug-like and pharmacological properties for use in this therapeutic use with retaining device combination. [00175] Compound II has R-stereochemistry at the phosphorus atom which has been confirmed with X-ray crystallography (Example 21, see FIG.1A and FIG.1B). In alternative embodiments, Compound II can be used in the form of any desired ratio of phosphorus R- and S-enantiomers, including up to pure enantiomers. In some embodiments, Compound II is used in a form that is at least 90% free of the opposite enantiomer, and can be at least 98%, 99%, or even 100% free of the opposite enantiomer. Unless described otherwise, an enantiomerically pure Compound II is at least 90% free of the opposite enantiomer. In certain embodiments Compound II is used as a racemic mixture of isomers. In addition, in an alternative embodiment, the amino acid of the phosphonamidate can be in the D- or L- configuration, or a mixture thereof, including a racemic mixture. [00176] Where a phosphonamidate exhibits chirality, it can be provided as an R or S chiral phosphorus derivative or a mixture thereof, including an enantiomerically enriched form including a racemic mixture. All of the combinations of these stereoconfigurations are alternative embodiments in the disclosure described herein. In another embodiment, at least one of the hydrogen atoms of Compound I, Compound II, or Compound III can be replaced with deuterium. [00177] In certain embodiments, Compound I may be:
Attorney Docket No.01394-0006-01PCT a pharmaceutically acceptable salt thereof. und I or salt thereof may be:
II may be ,
Attorney Docket No.01394-0006-01PCT . III may be
.
of the disclosure is Compound II, which can be provided in a pharmaceutically acceptable composition or solid dosage form thereof. In an embodiment, Compound II is an amorphous solid. In yet a further embodiment, Compound II is a crystalline solid.
Attorney Docket No.01394-0006-01PCT [00182] In certain embodiments, a compound of the disclosure is Compound I, RP Compound I, or SP Compound I as a monofumarate salt. In certain embodiments, a compound of the disclosure is Compound I, RP Compound I, or SP Compound I as a hemifumarate salt. In certain embodiments, a compound of the disclosure is Compound I, RP Compound I, or SP Compound I as a sesquifumarate salt. In certain embodiments, a compound of the disclosure is RP Compound I or SP Compound I as a sulfate salt. In certain embodiments, a compound of the disclosure is RP Compound I or SP Compound I as a hydrochloride salt. In certain embodiments, a compound of the disclosure is Compound I, RP Compound I, or SP Compound I as a benzenesulfonate salt. In certain embodiments, a compound of the disclosure is RP Compound I or SP Compound I as a tosylate salt. In certain embodiments, a compound of the disclosure is RP Compound I or SP Compound I as a succinate salt. or
Attorney Docket No.01394-0006-01PCT
Attorney Docket No.01394-0006-01PCT RP Salts of Compound I
Attorney Docket No.01394-0006-01PCT
Attorney Docket No.01394-0006-01PCT
Attorney Docket No.01394-0006-01PCT
salts form solids with favorable properties for solid dosage forms including morphic forms for administration in combination with a retaining device to a host such as a human with an HPV infection or an HPV-related disease such as cervical or vaginal intraepithelial neoplasia. However, the monofumarate optionally in morphic form displays superior properties to the hemifumarate and monosuccinate. Thus, the monofumarate salt remains a desired salt form of Compound I for administration in combination with a retaining device. [00184] In certain embodiments, the compound used in combination with the retaining device is an isolated morphic form of the compound of the formula: wherein the isolated
comprising peaks independently selected from at least 3, 4, 5, or 6 of the following 2theta values 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. [00185] In certain embodiments, the compound used in combination with the retaining device is an isolated morphic form of the compound of the formula:
Attorney Docket No.01394-0006-01PCT wherein the isolated morphic form is characterized by an XRPD pattern comprising peaks independently selected from at least 3, 4, 5, 6, 7, 8 or 9 of the following 2theta values 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. [00186] In certain embodiments, the compound used in combination with the retaining device is an isolated morphic form of the compound of the formula: wherein the isolated comprising peaks
independently or 2theta values 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 20.38+0.2°, 22.94+0.2°, 25.09+0.2°, 26.54+0.2°, 26.90+0.2°, 27.38+0.2°, 28.28+0.2°, 28.95+0.2°, 29.64+0.2°, and 38.07+0.2°. [00187] In certain embodiments, the compound used in combination with the retaining device is an isolated morphic form of the compound of the formula: wherein the isolated
comprising peaks independently selected from at least 3, 4, 5, or 6 of the following 2theta values 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. [00188] In certain embodiments, Compound I monofumarate pattern 1 characterized by an XRPD pattern in or substantially similar to that in FIG.9 is used in combination with the retaining device. [00189] Compound I monofumarate pattern 1 can be produced, for example, by crystallization from isopropyl alcohol and heptane, as described in Example 23. Compound I (free base) and about 1.0 equivalents of fumaric acid can for example be dissolved in
Attorney Docket No.01394-0006-01PCT isopropanol at a concentration from about 25% to about 40% w/v and stirred at an elevated temperature, for example about 45°C, about 50°C, about 55°C. The solution is stirred at this temperature until some solids form and is then optionally seeded with Compound I monofumarate pattern 1 crystalline solids. The mixture is stirred and cooled to a lower temperature to facilitate crystallization, for example less than about 40°C, less than about 30°C, less than about 25°C, less than about 20°C, or less than about 15°C. The mixture is then stirred while heptane is added in an amount ranging from about 1mL per mL of isopropanol to about 5 mL per mL of isopropanol, such as about 4 mL per mL isopropanol. The resulting suspension is stirred while the product crystallizes, for example for at least about 24 hours at 25°C. Next, the suspension is cooled to facilitate crystallization further. The solution can be cooled to less than about 10°C, less than about 5°C, less than about 0°C, or less than about -5°C. The solution is stirred at the decreased temperature to allow time for additional product to crystallize, such as for at least about one day, and then the solids are collected by filtration. The collected solids are dried under reduced pressure and optionally at elevated temperature to provide Compound I monofumarate pattern 1. [00190] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least about 3, 4, or 52theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°. [00191] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least twelve of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°. [00192] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least eleven of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°. [00193] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least ten of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
Attorney Docket No.01394-0006-01PCT [00194] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least nine of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°. [00195] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least eight of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°. [00196] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least seven of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°. [00197] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least six of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°. [00198] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least five of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°. [00199] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least four of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°. [00200] In certain embodiments, the Compound I monofumarate pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least three of the 2theta values selected from 6.0+0.2°, 8.9+0.2°, 9.3+0.2°, 9.7+0.2°, 11.9+0.2°, 14.8+0.2°, 18.0+0.2°, 20.0+0.2°, 23.4+0.2°, 25.2+0.2°, 25.9+0.2°, 26.8+0.2°, and 28.0+0.2°.
Attorney Docket No.01394-0006-01PCT [00201] In certain embodiments, Compound II Pattern 1 characterized by an XRPD pattern in or substantially similar to that in FIG.56 is used in combination with the retaining device. [00202] Compound II Pattern 1 can be produced, for example, by recrystallizing Compound II (Example 28, Table 40), equilibration of Compound II in a suitable solvent, or crystallization by slow evaporation of solvent (Example 28, Table 41). [00203] In certain embodiments, the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least three, four or five 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. [00204] In certain embodiments, the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least seven 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. [00205] In certain embodiments, the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least six 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. [00206] In certain embodiments, the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least five 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. [00207] In certain embodiments, the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least four 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. [00208] In certain embodiments, the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least three 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. [00209] In certain embodiments, the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least two 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°.
Attorney Docket No.01394-0006-01PCT [00210] In certain embodiments, the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least one 2theta values selected from 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. [00211] In certain embodiments, the Compound II pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least three, four, or five 2theta values selected from 3.08+0.2°, 9.30+0.2°, 10.66+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 17.45+0.2°, 18.13+0.2°, 19.78+0.2°, 20.14+0.2°, 22.91+0.2°, 23.34+0.2°, 25.14+0.2°, 25.33+0.2°, 25.86+0.2°, 26.78+0.2°, 27.99+0.2°, and 28.82+0.2°. [00212] In certain embodiments, Compound III pattern 1 characterized by an XRPD pattern in or substantially similar to that in FIG.59 is used in combination with the retaining device. [00213] Compound III pattern 1 can be produced, for example, by precipitation from isopropanol and heptanes (Example 30, Table 48). In certain nonlimiting embodiments, Compound III pattern 1 was prepared by dissolving SP-Compound I free base in isopropanol, for example about 100 mg of SP-Compound I in about 0.25 mL to about 0.5 mL of isopropanol. To this solution was added about 1.0 eq of fumaric acid, and the mixture stirred at ambient or elevated temperature, for example about 25°C to about 60°C. Next, about two to about five times as much heptanes was added as isopropanol. The resulting mixture was stirred at ambient or elevated temperature, for example about 25°C to about 60°C, and then cooled gradually, for example about 0.01°C/min to 1°C/min the solids isolated by filtration and then dried at ambient or reduced pressure. [00214] In certain embodiments, the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising independently at least 2, 3, 4, 5, or 6 of the 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. [00215] In certain embodiments, the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least ten 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. [00216] In certain embodiments, the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least nine 2theta
Attorney Docket No.01394-0006-01PCT values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. [00217] In certain embodiments, the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least eight 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. [00218] In certain embodiments, the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least seven 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. [00219] In certain embodiments, the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least six 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. [00220] In certain embodiments, the Compound III pattern 1 used in combination with the retaining device is characterized by an XRPD pattern comprising at least five 2theta values selected from 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. [00221] In certain embodiments, the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising independently at least 3, 4, 5, or 6 of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 20.38+0.2°, 22.94+0.2°, 25.09+0.2°, 26.54+0.2°, 26.90+0.2°, 27.38+0.2°, 28.28+0.2°, 28.95+0.2°, 29.64+0.2°, and 38.07+0.2°. [00222] In certain embodiments, the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising independently at least 3, 4, 5, or 6 of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°.
Attorney Docket No.01394-0006-01PCT [00223] In certain embodiments, the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least ten of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°. [00224] In certain embodiments, the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least nine of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°. [00225] In certain embodiments, the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least eight of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°. [00226] In certain embodiments, the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least seven of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°. [00227] In certain embodiments, the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least six of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°. [00228] In certain embodiments, the Compound III pattern 2 used in combination with the retaining device is characterized by an XRPD pattern comprising at least five of the 2theta values selected from 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 25.09+0.2°, 29.64+0.2°, and 38.07+0.2°. [00229] The present disclosure includes but is not limited to compounds, pharmaceutical compositions, and the use of any of the compounds described herein in combination with the retaining device, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as fumarate, Compound II or Compound III, with desired isotopic substitutions of atoms at amounts above the natural abundance of the isotope, i.e., enriched. Isotopes are atoms having the same atomic number but different mass numbers, i.e., the same number of protons but a different number of neutrons. By way of general example and without limitation, isotopes of hydrogen, for example, deuterium (2H) and tritium (3H) may be used anywhere in described structures. Alternatively, or in addition, isotopes of carbon, e.g., 13C and 14C, may be used. A preferred isotopic substitution is
Attorney Docket No.01394-0006-01PCT deuterium for hydrogen at one or more locations on the molecule to improve the performance of the drug. The deuterium can be bound in a location of bond breakage during metabolism (an α-deuterium kinetic isotope effect) or next to or near the site of bond breakage (a β- deuterium kinetic isotope effect). Achillion Pharmaceuticals, Inc. (WO/2014/169278 and WO/2014/169280) describes deuteration of nucleotides to improve their pharmacokinetic or pharmacodynamic, including at the 5-position of the molecule. [00230] Substitution with isotopes such as deuterium can afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. Substitution of deuterium for hydrogen at a site of metabolic break-down can reduce the rate of or eliminate the metabolism at that bond. At any position of the compound that a hydrogen atom may be present, the hydrogen atom can be any isotope of hydrogen, including protium (1H), deuterium (2H) and tritium (3H). Thus, reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise. [00231] The term "isotopically-labeled" analog refers to an analog that is a "deuterated analog", a "13C-labeled analog," or a "deuterated/13C-labeled analog." The term "deuterated analog" means a compound described herein, whereby a H, i.e., hydrogen/protium (1H), is substituted by a H-isotope, i.e., deuterium (2H). Deuterium substitution can be partial or complete. Partial deuterium substitution means that at least one hydrogen is substituted by at least one deuterium. In certain embodiments, the isotope is 90, 95 or 99% or more enriched in an isotope at any location of interest. In some embodiments, it is deuterium that is 90, 95 or 99% enriched at a desired location. Unless indicated to the contrary, the deuteration is at least 80% at the selected location. Deuteration of the nucleoside can occur at any replaceable hydrogen that provides the desired results. [00232] In exemplary non-limiting embodiments, a method for the treatment of HPV infection or a condition associated with HPV is provided that comprises (i) administering an effective amount of the anti-HPV therapeutic agent to a human in need thereof and (ii) inserting a retaining device. In some embodiments, the anti-HPV therapeutic agent is administered to the cervix. In some embodiments, the retaining device is maintained at the base of the cervix or in the vaginal canal. [00233] In exemplary non-limiting embodiments, a method for the treatment of HPV infection or HPV-induced intraepithelial neoplasia is provided that comprises (i) administering an effective amount of the anti-HPV therapeutic agent such as Compound I or a pharmaceutically acceptable salt thereof, or other compound or its salt described herein,
Attorney Docket No.01394-0006-01PCT optionally in morphic form, and optionally in tablet form, to the cervix using a vaginal applicator and then (ii) inserting a retaining device that is maintained at the base of the cervix or in the vaginal canal for sufficient time to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue as described in detail above. Types of HPV- induced intraepithelial neoplasia which can be treated include but are not limited to cervical and vaginal intraepithelial neoplasia. [00234] Patients can be instructed in the procedure for self-administration. The females can optionally administer the first dose of one of the compounds disclosed herein in a vaginal tablet to the cervix using the vaginal applicator followed by the retaining device in the clinic or alternatively, administer the first dose using a retaining device. The single-use tablet applicator or retaining device can be pre-moistened with a water-based lubricating gel. The patient or healthcare provider can then place the retaining device in its proper location. Following placement of the retaining device, the patient can be instructed to remain supine for several hours, for example, 3, 4, 5, 6, 7 or 8 hours with minimal activities and may resume normal activities after that time-period. The patient should repeat this procedure once daily for a total of 2, 3 or 4 applications in a week as instructed. It is preferred to carry out the two step administration process in the evening. [00235] Patients may administer three doses on alternate days over a 1-week period. Patients can optionally be instructed to use the following preventative measures to protect against leakage of drug or metabolite-containing fluid to healthy tissue: • Minimize amount of water-based lubricant (e.g.,KY gel) used for dosing. • Do not bathe the genital area until after the instructed administration dosage time period. An example is to bathe in the morning after dosing the night before. • After bathing, a non-water based barrier gel (for example zinc oxide) can be applied to the external genitalia and perineum for additional protection. • Peripads can be used and can be changed frequently to avoid continued exposure to any leaked drug or metabolite containing fluid. • It is preferred to avoid dosing during the patient’s period. Dosings are preferably initiated and completed prior to anticipated starting date of the period. • Examples of suitable schedules include dosing on days 1, 3, and 5 followed by dosing on days 8, 10, and 12, followed by dosing on days 15, 17, and 19.
Attorney Docket No.01394-0006-01PCT [00236] Patients who have screening biopsy results of HSIL (CIN2 or CIN3) can combine this protocol with other standard care procedures. For example, 10-14 weeks after the first administration of the vaginal tablet with retainer device, the patient may opt to undergo a LLETZ procedure or another procedure described herein. In some embodiments, the patient may be taking a non-systemic corticosteroid. [00237] In certain embodiments, the Compound I, or a pharmaceutically acceptable salt thereof such as Compound II or Compound III, vaginal tablet is self-administered. In certain embodiments, one or more doses of the vaginal tablets are administered by a physician. In certain embodiments, the first dose is administered by a physician or medical provider and the remaining doses, for example doses on days 3, 5, 8, and 10, are administered at home. [00238] In certain embodiments, a formulation for the treatment of HPV and associated conditions, such as intraepithelial neoplasia, is a dosage form that contains from about 0.01 mg to about 10 mg, from about 0.05 to about 5 mg, from about 0.05 to about 0.15 mg, from about 0.15 mg to about 0.45 mg, or from about 0.5 to about 1.5 mg of any of the active compounds described herein including but not limited to Compound I or a pharmaceutically acceptable sale such as a monofumarate, hemifumarate, or other solid form, or Compound II or Compound III. In certain embodiments, a formulation for the treatment of HPV and associated conditions, such as intraepithelial neoplasia, is a dosage form that contains about or at least 0.01, 0.03, 0.05, 0.1 mg, 0.3 mg.0.5 mg, 0.7 mg, 1 mg, 1.5 mg, 2 mg, 2.5 mg, 3 mg, 4 mg, 5 mg, 10 mg, 15 mg or 20 mg, of Compound I monofumarate, Compound II or Compound III, optionally in a selected morphic form. As explained earlier, the dose strengths in mg used herein refer to the mass of the active acyclic phosphonamidate (e.g., Compound I) and do not include the salt in the molecular weight. In other words, when referring to a dosage form comprising 0.1 mg of Compound I monofumarate, for example, the 0.1 mg dose is referring to the amount of the corresponding free base. Thus, when referring to a 0.1 mg dosage form of Compound I monofumarate, for example, it should be understood that the dosage form contains 0.124 mg of the monofumarate as this amount is equivalent to a dose of 0.1 mg of Compound I free base; when referring to a 0.3 mg dosage form of Compound I monofumarate, it should be understood that the dosage form contains 0.372 mg of the monofumarate as this amount is equivalent to a dose of 0.3 mg of Compound I free base; when referring to a 1 mg dosage form of Compound I monofumarate, for example, it should be understood that the dosage form contains 1.241 mg of the monofumarate as this amount is equivalent to a dose of 1 mg
Attorney Docket No.01394-0006-01PCT of Compound I free base; and so on. [00239] In certain embodiments, the topical formulation is administered once a day, or several days a week (such as 2 or 3 days a week) in combination with the retaining device, as long as necessary to achieve the desired results. In certain embodiments, the topical formulation is administered on a weekly schedule for one, two, three, four, five, six, eight, or more weeks. In certain aspects, the topical formulation is administered on a schedule of three dosages a week for two, three, four, five, six, or eight weeks. [00240] In certain embodiments, the formulation can be administered in one or more therapeutic cycles comprising a treatment cycle and a rest cycle, wherein the treatment cycle comprises administering a formulation as described herein in combination with the retaining device, followed by a rest cycle (comprising a period of no treatment) before the next treatment cycle. In certain embodiments, the rest cycle (also known as a treatment break) is from about one day to about six months. In certain embodiments, the rest cycle is one, two, three, four, five, six, seven, eight or more weeks before the next treatment cycle. In certain embodiments, the rest cycle is one week before the next treatment cycle. In certain embodiments, multiple therapeutic cycles are administered, for example one, two, three, four, five, or six therapeutic cycles. [00241] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with the active agent (e.g., Compound I) once per week. In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with the active agent (e.g., Compound I) once every two weeks. In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with the active agent (e.g., Compound I) once per 10 days. [00242] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with a single dose of the active agent (e.g., Compound I) as 0.01% vaginal gel. In some embodiments, one gram of the 0.01% vaginal gel is dosed. [00243] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.3 mg of the active agent (e.g., Compound I) 3 times over one week. [00244] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.1 mg of the active agent (e.g., Compound I). [00245] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.3 mg of the active agent (e.g., Compound I).
Attorney Docket No.01394-0006-01PCT [00246] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.05 mg of the active agent (e.g., Compound I) 3 times a week for two weeks. [00247] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.1 mg of the active agent (e.g., Compound I) 3 times a week for two weeks. [00248] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 0.3 mg of the active agent (e.g., Compound I) up to three times a week over up to two weeks. [00249] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed up to 0.1 mg of the active agent (e.g., Compound I) up to three times a week over up to two weeks, for up to two treatment cycles. [00250] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 0.3 mg of the active agent (e.g., Compound I) up to three times a week over up to two weeks, for up to two treatment cycles. [00251] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 1.0 mg of the active agent (e.g., Compound I) up to three times a week over up to two weeks. [00252] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 1.0 mg of the active agent (e.g., Compound I) up to three times a week over up to two weeks, for up to two treatment cycles. [00253] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 1.0 mg of the active agent (e.g., Compound I) for up to six times per treatment cycle. [00254] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.1 mg of the active agent (e.g., Compound I), followed by up to three doses of 0.1 within one week. [00255] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with 0.1 mg of the active agent (e.g., Compound I) 3 times a week. [00256] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 0.3 mg of the active agent (e.g., Compound I) for up to 3 times over one week.
Attorney Docket No.01394-0006-01PCT [00257] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 0.3 mg of the active agent (e.g., Compound I) for six doses over two weeks. [00258] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 1.0 mg of the active agent (e.g., Compound I) for up to six doses over up to two weeks. [00259] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 1.0 mg of the active agent (e.g., Compound I) for up to six doses over up to four weeks. [00260] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 1.0 mg of the active agent (e.g., Compound I) for up to six doses over up to two weeks for up to 2 treatment cycles. [00261] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 1.0 mg of the active agent (e.g., Compound I), two intermittent weekly dosing (1 week of treatment, followed by 1 week treatment break, followed by 1 week of treatment over 3 total weeks) for up to 6 doses (defined as a single treatment cycle) for up to 2 treatment cycles. [00262] In certain embodiments, a patient with an HPV infection or an intraepithelial neoplasia, such as CIN, is dosed with up to 1.0 mg of the active agent (e.g., Compound I) for up to six doses over up to two weeks. [00263] In some embodiments, the dosage is administered with a retaining device such as a diaphragm. In some embodiments, the dosage is administered with a vaginal applicator, which is subsequently removed. In some embodiments, the dosage is administered with a vaginal applicator, which is subsequently removed, and then a retaining device is inserted. [00264] Dosage forms which do not adhere well to the target site may be dislodged, interfering with treatment. Dosage forms have been discovered that adhere to the target site and dissolve rapidly in low fluid volumes. Adhesion to the target site also prevents exposure to healthy tissues, which may limit toxicity and side effects. Dosage forms which soften, break down, and/or disintegrate quickly in low fluid volumes are advantageous to cause a rapid release of the active compound to the target tissue. Dosage forms that disintegrate in, for example, less than about 50 µL, less than about 100 µL, less than about 125 µL, less than about 150 µL, less than about 175 µL, less than about 200 µL, or less than about 250 µL fluid particularly facilitate drug penetration into the target site.
Attorney Docket No.01394-0006-01PCT [00265] In certain embodiments, the dosage form is a gel. In certain embodiments, the dosage form is a cream. In certain embodiments, the dosage form is a tablet. [00266] In certain embodiments, the dosage form disintegrates in about one to about ten seconds. In certain embodiments, the dosage form disintegrates in about ten seconds to one minute. In certain embodiments, the dosage form disintegrates in about one minute to about one hour. In certain embodiments, the dosage form disintegrates in about one to six hours. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 5 minutes. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 4 minutes. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 3 minutes. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 2 minutes. In certain embodiments, the dosage form (such as a tablet) disintegrates in not more than 1 minutes. Disintegration can be tested by, for example, Ph. Eur <2.9.1> or USP<701> using water at 37°C as a media. [00267] The physical dimensions of the dosage form can impact the effectiveness of the dosage form. A tablet that is thinner provides a greater surface area to volume ratio and may degrade quicker and cover the target area better. In certain embodiments, the dosage form is less than about 6, 5, 4, 3, or 2 millimeters thick in its smallest dimension. [00268] The formulation of the dosage form is important for adequate administration of the active agent into the intraepithelial tissue. The formulation, for example, can be prepared for use as a tablet, a reconstituted powder, a dry powder, a semi solid dosage form, a film or a pessary (i.e., a vaginal suppository). [00269] Some embodiments disclosed herein include the use of an effective amount of Compound I monofumarate, Compound II or Compound III, in the manufacture of a medicament for ameliorating or treating a human papillomavirus infection, wherein the infection can be ameliorated or treated by inhibiting viral replication by inhibiting the synthesis of viral DNA. Other embodiments disclosed herein include the use of an effective amount of any of the compounds described herein including but not limited to Compound I monofumarate, Compound II or Compound III, for ameliorating or treating a human papillomavirus infection, wherein the human papillomavirus infection can be ameliorated or treated by inhibiting viral replication by inhibiting the synthesis of viral DNA. [00270] Certain nonlimiting embodiments disclosed herein include a method for ameliorating or treating a human papillomavirus infection that comprises contacting a cell infected with the human papillomavirus in a subject with an effective amount of any of the compounds described herein including but not limited to Compound I monofumarate,
Attorney Docket No.01394-0006-01PCT Compound I hemifumarate, or other solid form of Compound I, Compound II or Compound III, wherein the infection is ameliorated or treated by inhibiting the synthesis of viral DNA. Yet still other embodiments disclosed herein include a method for ameliorating or treating a human papillomavirus infection that comprise administering to a subject infected with the human papillomavirus an effective amount of Compound I monofumarate, Compound II or Compound III, wherein the human papillomavirus infection can be ameliorated or treated by inhibiting viral replication by inhibiting the synthesis of viral DNA. Some embodiments disclosed herein relate to Compound I monofumarate, Compound II or Compound III, for use in ameliorating or treating a human papillomavirus infection, wherein the human papillomavirus infection can be ameliorated or treated by inhibiting viral replication by inhibiting the synthesis of viral DNA. [00271] In some embodiments, in any of the methods disclosed herein, the human papillomavirus can be a high-risk human papillomavirus (hrHPV), such as those described herein. For example, the high-risk human papillomavirus can be selected from HPV-16, HPV-18, HPV-31, HPV-33, HPV-35, HPV-39, HPV-45, HPV-51, HPV-52, HPV-56, HPV- 58, HPV-59, HPV-68, HPV-73, and HPV-82. In some embodiments, the human papillomavirus can be HPV-16. In some embodiments, the human papillomavirus can be HPV-11. In some embodiments, the human papillomavirus can be HPV-18. In some embodiments, the human papillomavirus can be one or more of the following high-risk types: HPV-31, HPV-33, HPV-35, HPV-39, HPV-45, HPV-51, HPV-52, HPV-56, HPV-58, HPV- 59, HPV-68, HPV-73, and HPV-82. [00272] As described herein, the presence of an HPV infection can be detected using the Papanicolaou test (Pap smear) and/or DNA probe testing (for example, HPV DNA probe testing for one or more high-risk HPV types). Therefore, in some embodiments, an effective amount of Compound I or a pharmaceutically acceptable salt thereof, such as a monofumarate, Compound II or Compound III, can be provided in combination with the retaining device to a subject diagnosed with an HPV infection (for example a high-risk HPV infection) by a DNA test. In some embodiments, an effective amount of Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II or Compound III can be provided to a subject diagnosed with an HPV infection, or a disease associated with HPV infection, as identified by a Papanicolaou test. [00273] In certain embodiments, an effective amount of Compound I monofumarate, Compound II, or Compound III may be provided to a subject with a Papanicolaou test result that does not indicate the disease has progressed to cervical cancer. The Bethesda system is a
Attorney Docket No.01394-0006-01PCT standardized scoring system for reporting pap smear test results and assigns a grade of 1-3 based on severity. Grade 1 CIN (CIN 1) indicates mild dysplasia. Grades 2 and 3 CIN (CIN 2, CIN 3) are more serious and typically require intervention. In certain embodiments, Compound I monofumarate, Compound II or Compound III is used to treat CIN 1 (Grade 1 cervical intraepithelial neoplasia). In certain embodiments, Compound I monofumarate, Compound II or Compound III is used to treat CIN 2 (Grade 2 cervical intraepithelial neoplasia) in combination with the retaining device. In certain embodiments, Compound I monofumarate, Compound II or Compound III is used to treat CIN 3 (Grade 3 cervical intraepithelial neoplasia) in combination with the retaining device. [00274] In certain embodiments, a pharmaceutical composition comprising Compound I monofumarate, Compound II or Compound III, is used in the manufacture of a system for the treatment of CIN 1 (Grade 1 cervical intraepithelial neoplasia) that comprises the use of a retaining device, as described further herein. In certain embodiments, a pharmaceutical composition comprising Compound I monofumarate, Compound II or Compound III, is used in the manufacture of a system for the treatment of CIN 2 (Grade 2 cervical intraepithelial neoplasia) that comprises the use of a retaining device as described further herein. In certain embodiments, a pharmaceutical composition comprising Compound I monofumarate, Compound II or Compound III, is used in the manufacture of a medicament for the treatment of CIN 3 (Grade 3 cervical intraepithelial neoplasia). [00275] In certain embodiments, Compound I monofumarate, Compound II or Compound III, optionally in a pharmaceutically acceptable carrier, is used to treat a condition selected from the group consisting of atypical squamous cells of undetermined significance (ASC-US), atypical glandular cells (AGC), low-grade squamous intraepithelial lesions (LSIL), atypical squamous cells (cannot exclude high grade squamous intraepithelial lesion) (ASC-H), high grade squamous intraepithelial lesions (HSIL), adenocarcinoma in situ (AIS), and cervical cancer (e.g. squamous cell carcinoma or adenocarcinoma). [00276] In certain embodiments, Compound II optionally in a pharmaceutically acceptable carrier, is used in the therapeutic use and retaining device system to treat a condition selected from the group consisting of atypical squamous cells of undetermined significance (ASC-US), atypical glandular cells (AGC), low-grade squamous intraepithelial lesions (LSIL), atypical squamous cells (cannot exclude high grade squamous intraepithelial lesion) (ASC-H), high grade squamous intraepithelial lesions (HSIL), adenocarcinoma in situ (AIS), and cervical cancer (e.g. squamous cell carcinoma or adenocarcinoma).
Attorney Docket No.01394-0006-01PCT [00277] In some embodiments, Compound I monofumarate, Compound II or Compound III, can be used in the manufacture of a system that comprises a retaining device for use in ameliorating and/or treating an infection caused by one or more types of human papillomaviruses. In some embodiments, the human papillomavirus is a low-risk human papillomavirus, including those described herein. In some embodiments, the human papillomavirus is HPV-6. In some embodiments, the human papillomavirus is HPV-11. [00278] In some embodiments, Compound I monofumarate, Compound II or Compound III, can be used in the manufacture of a system that comprises a retaining device for use in ameliorating and/or treating an infection of HPV-16 and/or HPV-18. In certain embodiments, Compound I monofumarate, Compound II or Compound III can be used in the manufacture of a system that comprises a retaining device for use in treating a high-risk HPV infection. In certain embodiments, Compound I monofumarate, Compound II or Compound III can be used in the manufacture of a system that comprises a retaining device for use in treating a related disease or condition occurring as a result of a high-risk HPV infection. In some embodiments, Compound I monofumarate, Compound II or Compound III can be used in the manufacture of a system that comprises a retaining device for use in ameliorating and/or treating an infection comprising both high-risk and low-risk HPV. [00279] In certain embodiments, a pharmaceutical composition comprising Compound II is used in the therapeutic system that comprises a retaining device to treat conditions related to or occurring as a result of exposure to or an infection of HPV. In certain embodiments, a pharmaceutical composition comprising Compound II is used to treat precancerous cervical lesions. In certain embodiments, a pharmaceutical composition comprising Compound II is used to treat cervical intraepithelial neoplasia. In certain embodiments, a pharmaceutical composition comprising Compound II is used to treat vaginal intraepithelial neoplasia. In certain embodiments, a pharmaceutical composition comprising Compound II is used to treat cervical cancer. In certain embodiments, a pharmaceutical composition comprising Compound II is used to treat vaginal cancer. [00280] In certain embodiments, a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of conditions related to or occurring as a result of exposure to or an infection of HPV. In certain embodiments, a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of precancerous cervical lesions. In certain embodiments, a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the
Attorney Docket No.01394-0006-01PCT treatment of cervical intraepithelial neoplasia. In certain embodiments, a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of vaginal intraepithelial neoplasia. In certain embodiments, a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of cervical cancer. In certain embodiments, a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of vaginal cancer. In certain embodiments, a pharmaceutical composition comprising Compound II is used in the manufacture of a system that comprises a retaining device for the treatment of oropharyngeal cancer. [00281] It is advantageous for the dosage form to be easily applied to the target site. Direct application to the target site prevents systemic exposure and toxicity. To place the dosage form on the target site, the dosage form may be applied with an applicator. In certain embodiments, the dosage form is applied with a vaginal applicator. In certain embodiments, the dosage form is applied with a retaining device. In certain embodiments, additional fluid (such as a lubricant) is delivered along with the dosage form, applied to the dosage form, or applied to the target site or surrounding tissues. [00282] In certain embodiments, a lubricating fluid is administered in combination with the dosage form to enhance the coverage of the cervix, vagina or vulva. In certain embodiments, water is used as the fluid administered with the dosage form. In certain embodiments, a lubricating glycerol- or hydroxyethylcellulose-based, water soluble fluid is used in combination with the dosage form. In certain embodiments, the dosage form is administered without additional fluid. [00283] In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 5 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 4 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 3 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 2 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 1 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.75 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.5 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.25 milliliter of fluid. In
Attorney Docket No.01394-0006-01PCT certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.2 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.15 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.125 milliliter of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in less than about 0.1 milliliter of fluid. [00284] In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 10 microliters to about 100 microliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 75 microliters to about 250 microliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 200 microliters to about 500 microliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 400 microliters to about 750 microliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 700 microliters to about 1,000 microliters of fluid. [00285] In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 1 milliliter to about 2 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 2 milliliters to about 3 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 3 milliliters to about 4 milliliters of fluid. In certain embodiments, the dosage form will soften, disintegrate, and/or dissolve in from about 4 milliliters to about 5 milliliters of fluid. [00286] In certain embodiments, Compound II is administered in combination with the use of a retaining device for at least 1, 2, 3, 4, 5, or 6 consecutive or nonconsecutive days. [00287] In certain embodiments, Compound II is administered once a week in combination with the use of a retaining device. In certain embodiments, Compound II is administered once a week in combination with the use of a retaining device for up to 12 weeks. In certain embodiments, Compound II is administered once a week in combination with the use of a retaining device for up to 10 weeks. In certain embodiments, Compound II is administered once a week in combination with the use of a retaining device for up to 8 weeks. In certain embodiments, Compound II is administered once a week in combination with the use of a retaining device for up to 6 weeks. In certain embodiments, Compound II is administered once a week in combination with the use of a retaining device for up to 4 weeks. In certain embodiments, Compound II is administered once a week in combination with the
Attorney Docket No.01394-0006-01PCT use of a retaining device for up to 2 weeks. In certain embodiments, Compound II is administered once a week in combination with the use of a retaining device for up to 1 week. [00288] In certain embodiments, Compound II is administered in combination with the use of a retaining device twice a week. In certain embodiments, Compound II is administered in combination with the use of a retaining device twice a week for up to 12 weeks. In certain embodiments, Compound II is administered in combination with the use of a retaining device twice a week for up to 10 weeks. In certain embodiments, Compound II is administered in combination with the use of a retaining device twice a week for up to 8 weeks. In certain embodiments, Compound II is administered in combination with the use of a retaining device twice a week for up to 6 weeks. In certain embodiments, Compound II is administered in combination with the use of a retaining device twice a week for up to 4 weeks. In certain embodiments, Compound II is administered in combination with the use of a retaining device twice a week for up to 2 weeks. In certain embodiments, Compound II is administered in combination with the use of a retaining device twice a week for up to 1 week. [00289] In certain embodiments, Compound II is administered in combination with the use of a retaining device three times a week. In certain embodiments, Compound II is administered in combination with the use of a retaining device three times a week for up to 12 weeks. In certain embodiments, Compound II is administered in combination with the use of a retaining device three times a week for up to 10 weeks. In certain embodiments, Compound II is administered in combination with a retaining device three times a week for up to 8 weeks. In certain embodiments, Compound II is administered in combination with a retaining device three times a week for up to 6 weeks. In certain embodiments, Compound II is administered in combination with a retaining device three times a week for up to 4 weeks. In certain embodiments, Compound II is administered in combination with a retaining device three times a week for up to 2 weeks. In certain embodiments, Compound II is administered in combination with a retaining device three times a week for up to 1 week. [00290] In certain embodiments, Compound II is administered in combination with a retaining device daily. In certain embodiments, Compound II is administered in combination with a retaining device daily for up to 12 weeks or indefinitely as instructed by a healthcare provider. In certain embodiments, Compound II is administered in combination with a retaining device daily for up to 10 weeks. In certain embodiments, Compound II is administered in combination with a retaining device daily for up to 8 weeks. In certain embodiments, Compound II is administered in combination with a retaining device daily for up to 6 weeks. In certain embodiments, Compound II is administered in combination with a
Attorney Docket No.01394-0006-01PCT retaining device daily for up to 4 weeks. In certain embodiments, Compound II is administered in combination with a retaining device daily for up to 2 weeks. In certain embodiments, Compound II is administered in combination with a retaining device daily for up to 1 week. [00291] In certain embodiments, from about 0.05 mg to about 1.0 mg, of Compound II is administered in combination with a retaining device daily for one, two, three, four, five, six, or more weeks, as instructed by a healthcare provider. In certain embodiments, from about 0.05 mg to about 0.3 mg, of Compound II is administered in combination with a retaining device daily for one, two, three, four, five, six, or more weeks, as instructed by a healthcare provider. In certain embodiments, from about 0.1 mg to about 0.3 mg, of Compound II is administered in combination with a retaining device daily for one, two, three, four, five, six, or more weeks, as instructed by a healthcare provider. As explained earlier, the dose strengths in mg used herein refer to the mass of the active acyclic phosphonamidate (e.g., Compound II free base) and do not include the salt in the molecular weight. In other words, when referring to a dosage form comprising 0.1 mg of Compound II, for example, the 0.1 mg dose is referring to the amount of the corresponding free base. Thus, when referring to the administration of 0.1 mg of Compound II, for example, it should be understood that the administration is of 0.124 mg of the monofumarate, as this amount is equivalent to a dose of 0.1 mg of the free base of Compound II; when referring to the administration of 0.3 mg of Compound II, it should be understood that the administration is of 0.372 mg of the monofumarate, as this amount is equivalent to a dose of 0.3 mg of the free base of Compound II; when referring to the administration of 1 mg of Compound II, for example, it should be understood that the administration is of 1.241 mg of the monofumarate, as this amount is equivalent to 1 mg of the free base of Compound II; and so on. [00292] In certain embodiments, Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week. In certain embodiments, Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week for up to 12 weeks. In certain embodiments, Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week for up to 10 weeks. In certain embodiments Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week for up to 8 weeks. In certain embodiments, Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week for up to 6 weeks. In
Attorney Docket No.01394-0006-01PCT certain embodiments, Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week for up to 4 weeks. In certain embodiments, Compound I monofumarate or hemifumarate or other solid form is administered in combination with a retaining device three times a week for up to 2 weeks. In certain embodiments, Compound I monofumarate is administered in combination with a retaining device three times a week for up to 1 week. [00293] In certain embodiments, Compound I monofumarate is administered in combination with a retaining device daily. In certain embodiments, Compound I monofumarate is administered in combination with a retaining device daily for up to 12 weeks or indefinitely as instructed by a healthcare provider. In certain embodiments, Compound I monofumarate is administered in combination with a retaining device daily for up to 10 weeks. In certain embodiments, Compound I monofumarate is administered in combination with a retaining device daily for up to 8 weeks. In certain embodiments, Compound I monofumarate is administered in combination with a retaining device daily for up to 6 weeks. In certain embodiments, Compound I monofumarate is administered in combination with a retaining device daily for up to 4 weeks. In certain embodiments, Compound I monofumarate is administered in combination with a retaining device daily for up to 2 weeks. In certain embodiments, Compound I monofumarate is administered in combination with a retaining device daily for up to 1 week. [00294] In certain embodiments, Compound I monofumarate, or other solid form such as Compound II or Compound III, may be administered in combination with a retaining device three, four, five or six times a week. In certain embodiments, Compound I monofumarate, Compound II or Compound III may be administered in combination with a retaining device once per day. In certain embodiments, Compound I monofumarate, Compound II or Compound III may be administered in combination with a retaining device twice per day. In certain embodiments, Compound I monofumarate, Compound II or Compound III may be administered in combination with a retaining device three, four, or more times per day. In certain embodiments, Compound I monofumarate, Compound II or Compound III may be administered in combination with a retaining device daily. [00295] In certain embodiments, Compound I monofumarate, Compound II or Compound III can be administered in combination with a retaining device in one or more therapeutic cycles comprising a treatment cycle and a rest cycle, wherein the treatment cycle comprises administering the compound as described herein, followed by a rest cycle (comprising a period of no treatment) before the next treatment cycle. In certain
Attorney Docket No.01394-0006-01PCT embodiments, the rest cycle is from about one day to about six months. In certain embodiments, the rest cycle is one, two, three, four, five, six, seven, eight or more weeks before the next treatment cycle. In certain embodiments, multiple therapeutic cycles are administered, for example, one, two, three, four, five, or six therapeutic cycles. [00296] As described above, a number of compounds have been investigated for the treatment of HPV-induced neoplasia, however none has been approved yet. For non-limiting examples of investigated approaches, see: Ahn W.S., et al. Protective effects of green tea extracts (polyphenon E and EGCG) on human cervical lesions. Eur. J. Cancer Prev. 2003;12:383–390; Ashrafian L,et al. Double-blind randomized placebo-controlled multicenter clinical trial (phase IIa) on diindolymethane’s efficacy and safety in the treatment of CIN: implications for cervical cancer prevention. EPMA J.2015;6:doi: 10.1186/s13167- 13015-10048-13169; Bossens M., et al. Safety and tolerance of cidofovir as a 2% gel for local application in high-grade cervical intraepithelial neoplasia: A phase 1 investigation. Int. J. Clin. Pharmacol.2018;56:134–141; Chen F.P. Efficacy of imiquimod 5% cream for persistent human papillomavirus in genital intraepithelial neoplasm. Taiwanese J. Obstetrics Gynecol.2013;52(4):475–478; Choo Y., et al. Intravaginal application of leukocyte interferon gel in the treatment of cervical intraepithelial neoplasia (CIN) Arch Gynecol.1985;237:51– 54; de Witte C.J et al. Imiquimod in cervical, vaginal and vulvar intraepithelial neoplasia: a review. Gynecol. Oncol.2015;139:377–384; Desravines N, et al. Low dose 5-fluorouracil intravaginal therapy for the treatment of cervical intraepithelial neoplasia 2/3: A case series. J. Gynecol. Surg.2020;36; DiSilvestro P.A., et al. Treatment of cervical intraepithelial neoplasia levels 2 and 3 with adapalene, a retinoid-related molecule. J. Low Genit Tract. Dis. 2001;5:33–37; Graham V., et al. Phase II trial of beta-all-transretinoic acid for cervical intraepithelial neoplasia via a collagen sponge and cervical cap. West. J. Med.1986;145:192– 195; Grimm C., et al. Treatment of cervical intraepithelial neoplasia with topical imiquimod: a randomized controlled trial. Obstet. Gynecol.2012;120(1):152–159; Hampson L., et al. A single-arm, proof-of-concept trial of lopimune (lopinavir/ritonavir) as treatment for HPV- related pre-invasive cervical disease. PLoS ONE.2016;11; Helm C.W. et al. Retinoids for preventing the progression of cervical intra-epithelial neoplasia. Cochrane Systematic Review.2013; Hubert P., et al. Local applications of GM-CSF induce the recruitment of immune cells in cervical low-grade squamous intraepithelial lesions. Am. J. Reprod. Immunol.2010;64:126–136; Koeneman MM, et al. Topical Imiquimod treatment of high- grade Cervical intraepithelial neoplasia (TOPIC trial): study protocol for a randomized controlled trial. BMC Cancer.2016:doi: 10.1186/s12885-12016-12187-12883; Krause S., et
Attorney Docket No.01394-0006-01PCT al. Interferon and cervical dysplasia: CIN III treated with local interferon application. Colposcopy Gynecologic Laser Surgery.1987;3:195–198; Krebs H.B., et al. Chronic ulcerations following topical therapy with 5-fluorouracil for vaginal human papillomavirus- associated lesions. Obstet. Gynecol.1991;78(2):205–208; Laccetta G. et al. Effect of the treatment with beta-glucan in women with cervical cytologic report of atypical squamous cells of undetermined significance (ASCUS) and low-grade intraepithelial lesions (L-SIL) Minerva Ginecol.2015;67:113–120; Meyskens F.L., et al. A phase I trial of beta-all- transretinoic acid delivered via a collagen sponge and a cervical cap for mild or moderate intraepithelial cervical neoplasia. J. Natl Cancer Inst.1983;71:921–925; Niwa K., et al. Topical vidarabine of 5-fluoruracil treatment against persistent HPV in genital (pre)cancerous lesions. Oncol Reports.2003;10:1437–1441; Pachman DR, et al. Randomized clinical trial of imiquimod: an adjunct to treating cervical dysplasia. Am. J. Obstet. Gynecol.2012;206(1):42 e41-47; Rahangdale L et al. Topical 5-fluorouracil for treatment of Cervical Intraepithelial Neoplasia 2: a randomized controlled trial. Am. J. Obstet. Gynecol.2014;210:e1–e8; Schneider A., et al. Efficacy trial of topically administered Interferon gamma-1beta gel in comparison to laser treatment in cervical intraepithelial neoplasia. Arch. Gynecol Obste. 1995;256:75–83; Silman F.H., et al.5-fluorouracil/chemosurgery for intraepithelial neoplasia of the lower genital tract. Obstet. Gynecol.1981;58:356–360; Snoeck R., Noel J.C., Muller C., Clercq De, Bossens M. Cidofovir, a new approach for the treatment of cervix intraepithelial neoplasia III (CIN III) J. Med. Virol.2000;60:205–209; Stentella P., Biamonti A., Carraro C. Efficacy of carboxymethyl beta-glucan in cervical intraepithelial neoplasia: a retrospective, case-control study. Minerva Ginecol.2017;69:425–430; Suh-Burgmann E., Sivret J., Duska L.R., Del Carmen M., Seiden M.V. Long-term administration of intravaginal dehydroepiandrosterone on regression of low-grade cervical dysplasia - a pilot study. Gynecol. Obstet. Invest.2003;55:25–31; Valencia M.H., Pacheco A.C., Quijano T.H., Giron A.V., Lopez C.V. Clinical response to glycyrrhizinic acid in genital infection due to human papillomavirus and low-grade squamous intraepithelial lesion. Clin. Pract.20111(e93); van de Sande A., Koeneman M., Gerestein C., Kruse A., van Kemenade F., van Beekhuizen H. Topical Imiquimod treatment of residual or recurrent cervical intraepithelial neoplasia (TOPIC-2 trial): a study protocol for a randomized controlled trial. BMC Cancer. 2018;18:4510–4517; and Van Pachterbeke C., Bucella D., Rozenberg S. Topical treatment of CIN 2+ by cidofovir: Results of a phase II, double-blind, prospective, placebo-controlled study. Gynecol Onc.2009;115:69–74.
Attorney Docket No.01394-0006-01PCT [00297] In an aspect of the disclosure, pharmaceutical compositions according to the present disclosure comprise an anti-HPV effective amount of any of the active compounds described herein, optionally in a morphic form as described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof, including the monofumarate, Compound II or Compound III as described herein, optionally in combination with a pharmaceutically acceptable carrier, additive, or excipient, and/or in combination or alternation with at least one other active compound and used in combination with the retaining device as described herein. In some embodiments, the pharmaceutical composition comprises an anti-HPV effective amount of Compound I monofumarate, Compound II or Compound III in combination with a pharmaceutically acceptable carrier, additive, or excipient and in combination with a retaining device as described herein. In one embodiment, the disclosure includes a solid dosage form comprising Compound II and a pharmaceutically acceptable carrier. In certain embodiments, the pharmaceutical composition is administered directly to the cervix, vagina or vulva. [00298] Table 1 provides a nonlimiting example of a vaginal tablet formulation. Table 1. A nonlimiting example of a formulation for vaginal tablet Ingredient Function Amount per 175 mg Tablet
a sa o e.g., o o u a a e sa s use o p epa e e a e , e a ou g e a jus e suc at the amount in the tablet is equivalent to 0.1 mg to 1 mg of the free base. For example, if the API is Compound I monofumarate and the dose is 1 mg per tablet, then the tablet will contain 1.241 mg of the fumarate salt (as this amount is equivalent to 1 mg of the free base). [00299] In certain embodiments, vaginal tablet products are formulated to deliver drug to the cervical epithelium following topical administration onto the cervix. In certain embodiments, the tablet has good adherence to mucosal tissue. The excipients in the tablet products should be preferably compendial (USP/NF), included in the US FDA’s List of Inactive Ingredients, or appear on the US FDA’s Food Additive
Attorney Docket No.01394-0006-01PCT [00300] In certain embodiments, the vaginal tablets can be packaged for example in bottles with a desiccant. Nonlimiting examples are high density polyethylene bottles with a silica gel desiccant canister and rayon, induction sealed and capped with a child-resistant closure. The vaginal tablet can be administered using a single-use vaginal tablet applicator or a retaining device. [00301] It is preferred that the tablets are stored refrigerated (2-8℃) in bottles with a desiccant. Prior to administration, the bottle should be brought to room temperature for at least 30 minutes before opening and removing the tablet. [00302] Tablets are typically stored refrigerated (2-8℃) in bottles with a desiccant. Prior to clinical use, the tablet should be removed from refrigeration (2-8℃) and brought to room temperature for at least 30 minutes before opening and removing the tablet. The tablets have been designed to deliver the required dose with a single tablet at each application using a tablet applicator or retaining device. [00303] In an aspect of the disclosure, pharmaceutical compositions used in the present disclosure comprise an anti-HPV effective amount of Compound II described herein, optionally in a morphic form, in combination with a pharmaceutically acceptable carrier, additive, or excipient, further optionally in combination with at least one other antineoplastic agent or antiviral agent, such as an anti-HPV agent, in combination with the retaining device. In certain embodiments, the pharmaceutical composition comprises Compound II in combination with a second antiviral drug in combination with the retaining device. In certain embodiments, the pharmaceutical composition comprises Compound II in combination with an anticancer drug in combination with the retaining device. [00304] One of ordinary skill in the art will recognize that a therapeutically effective amount will vary with the infection or condition to be treated, its severity, the treatment regimen to be employed, the pharmacokinetic of the agent used, as well as the patient or subject (animal or human) to be treated, and such therapeutic amount can be determined by the attending physician or specialist. [00305] In certain pharmaceutical dosage forms, the prodrug form of the compounds, especially including acylated (acetylated or other) and ether (alkyl and related) derivatives, phosphate esters, thiophosphonamidates, phosphonamidates, and various salt forms of the present compounds, may be used to achieve the desired effect. One of ordinary skill in the art will recognize how to readily modify the present compounds to prodrug forms to facilitate delivery of active compounds to a targeted site within the host organism or patient. The person of ordinary skill in the art also will take advantage of favorable pharmacokinetic
Attorney Docket No.01394-0006-01PCT parameters of the prodrug forms, where applicable, in delivering the present compounds to a targeted site within the host organism or patient to maximize the intended effect of the compound. [00306] The amount of any of the active compounds described herein (including but not limited to Compound I monofumarate, Compound II, or Compound III) included within the therapeutically active formulation according to the present disclosure is an effective amount to achieve the desired outcome according to the present disclosure in combination with the retaining device, for example, for treating the HPV infection, reducing the likelihood of a HPV infection or the inhibition, reduction, and/or abolition of HPV or its secondary effects, including disease states, conditions, and/or complications which occur secondary to HPV infection. [00307] In certain embodiments, Compound I monofumarate, Compound II, or Compound III may be administered in a gel in combination with the use of a retaining device. In certain embodiments, the gel contains from about 0.001% to about 10%, from about 0.01% to about 10%, from about 0.05% to about 5%, from about 0.1 to about 3% from about 0.1 to about 2% Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II, or Compound III (weight/weight). In certain embodiments, the gel contains from about 0.001% to about 0.05% Compound I monofumarate, Compound II, or Compound III. In certain embodiments, the gel contains from about 0.01% to about 0.5% Compound I monofumarate, Compound II, or Compound III. In certain embodiments, the gel contains from about 0.1% to about 5% Compound I monofumarate, Compound II, or Compound III. In certain embodiments, one dose of a gel comprising Compound I monofumarate, Compound II, or Compound III provides about 0.1 mg to about 10 mg of the corresponding free base. In certain embodiments, one dose of the gel provides about 0.1 mg of the corresponding free base. In certain embodiments, one dose of the gel provides about 1 mg of the corresponding free base. In certain embodiments, one dose of the gel provides about 10 mg of the corresponding free base. [00308] In certain non-limiting embodiments, any of the active compounds described herein (including but not limited to Compound I monofumarate, Compound II, or Compound III) are administered topically via a tablet, capsule, suspension, liquid, emulsion, implant, particle, sphere, cream, ointment, suppository, pessary, transdermal form, gel, mucosal, and the like in combination with the retaining device. The dosage form may also be a bilayer tablet, in which the full dose of active compound is released one direction (for example towards the target tissue).
Attorney Docket No.01394-0006-01PCT [00309] In certain embodiments, the dosage form can soften, disintegrate, and/or release the drug in low fluid volumes. In certain embodiments, the dosage form softens and begins to release the drug immediately. In certain embodiments, the dosage form softens and begins to release the drug gradually. In certain embodiments, the dosage form softens and begins to release the drug within one hour. In certain embodiments, the dosage form softens and begins to release the drug within two hours. In certain embodiments, the dosage form is an immediate release vaginal tablet. [00310] The dosage form may be prepared to maximize surface area, facilitating disintegration. In certain embodiments, the dosage form is a round tablet. In certain embodiments, the dosage form is an oval tablet. In certain embodiments, the dosage form is a caplet. [00311] In certain embodiments, the dosage form is a tablet with a width that is the largest dimension and a tablet thickness that is the smaller dimension. In certain embodiments, the dosage form is twice as wide as it is thick. In certain embodiments, the dosage form is three times as wide as it is thick. In certain embodiments, the dosage form is four or more times as wide as it is thick. In certain embodiments, the dosage form is from about 0.1 mm thick to about 5 mm thick. In certain embodiments, the dosage form is from about 1 mm to about 2 mm thick. In certain embodiments, the dosage form is from about 2 mm to about 3 mm thick. In certain embodiments, the dosage form is from about 3 mm to about 4 mm thick. In certain embodiments, the dosage form is from about 4 mm to about 5 mm thick. In certain embodiments, the tablet is from about 5 mm to about 15 mm thick. In certain embodiments, the dosage form is less than 5 grams. In certain embodiments, the dosage form is from about 0.05 gram to about 0.15 gram. In certain embodiments, the dosage form is from about 0.1 to about 1 gram. In certain embodiments, the dosage from about 0.75 grams to about 2 grams. In certain embodiments, the dosage form is from about 1 gram to about 5 grams. In certain embodiments, the dosage form has a mass of from about 50 mg to about 500 mg. In certain embodiments, the dosage form has a mass of from about 100 mg to about 250 mg. In certain embodiments, the dosage form has a mass of from about 150 mg to about 200 mg. In certain embodiments, the dosage form has a mass of about 175 mg. [00312] In some embodiments, the dosage form is a tablet with a hardness of greater than about 1 Kp. In some embodiments, the dosage form is a tablet with a hardness of greater than about 2 Kp. In some embodiments, the dosage form is a tablet with a hardness of greater than about 3 Kp. In some embodiments, the dosage form is a tablet with a hardness of
Attorney Docket No.01394-0006-01PCT greater than about 4 Kp. In some embodiments, the dosage form is a tablet with a hardness of greater than about 5 Kp. [00313] In some embodiments, the dosage form is a tablet with a hardness of from about 1 Kp to about 10 Kp. In some embodiments, the dosage form is a tablet with a hardness of from about 2 Kp to about 10 Kp. In some embodiments, the dosage form is a tablet with a hardness of from about 3 Kp to about 10 Kp. In some embodiments, the dosage form is a tablet with a hardness of from about 4 Kp to about 10 Kp. In some embodiments, the dosage form is a tablet with a hardness of from about 5 Kp to about 10 Kp. [00314] In some embodiments, the dosage form is a tablet with a hardness of about 1 Kp. In some embodiments, the dosage form is a tablet with a hardness of about 2 Kp. In some embodiments, the dosage form is a tablet with a hardness of about 3 Kp. In some embodiments, the dosage form is a tablet with a hardness of about 4 Kp. In some embodiments, the dosage form is a tablet with a hardness of about 5 Kp. In some embodiments, the dosage form is a tablet with a hardness of about 6 Kp. In some embodiments, the dosage form is a tablet with a hardness of about 7 Kp. In some embodiments, the dosage form is a tablet with a hardness of about 8 Kp. In some embodiments, the dosage form is a tablet with a hardness of about 9 Kp. In some embodiments, the dosage form is a tablet with a hardness of about 10 Kp. [00315] In some embodiments, the dosage form is a tablet with a friability of not more than about 5%. In some embodiments, the dosage form is a tablet with a friability of not more than about 4%. In some embodiments, the dosage form is a tablet with a friability of not more than about 3%. In some embodiments, the dosage form is a tablet with a friability of not more than about 2%. In some embodiments, the dosage form is a tablet with a friability of not more than about 1%. Friability can be tested by, for example, Ph. Eur <2.9.7> or USP<1216>. [00316] In certain embodiments, the dosage form is not easily removed, dislodged, or moved from the target site. These desirable properties may be achieved by inclusion of a mucoadhesive polymer into the pharmaceutical composition. In certain embodiments, the pharmaceutical composition comprises a mucoadhesive polymer or mucoadhesive excipient. Nonlimiting examples of mucoadhesive polymers and excipients include: Hypromellose, lectin, thiolated polymers (e.g. chitosan–iminothiolane, poly(acrylic acid)–cysteine, poly(acrylic acid)–homocysteine, chitosan–thioglycolic acid, chitosan–thioethylamidine, alginate– cysteine, poly(methacrylic acid)–cysteine and sodium carboxymethylcellulose– cysteine), polyethylene glycol, polyvinyl alcohol, polyvinyl pyrrolidinone, polyacrylic acid
Attorney Docket No.01394-0006-01PCT (Carbopol®), polyheroxyethyl methacrylate, chitosan, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, methylcellulose, sodium carboxymethyl cellulose, aminated corn starch, cellulose derivatives, poly (acrylic acid) polymers, poly (hydroxyethyl methylacrylate), poly (ethylene oxide), poly (vinyl pyrrolidone), poly (vinyl alcohol), tragacanth, sodium alginate, karaya gum, guar gum, xanthan gum, soluble starch, gelatin, pectin, chitosan, methyl cellulose, hyaluronic acid, hydroxy propyl methylcellulose, hydroxy propyl cellulose, gellan gum, carrageenan, cationic hydroxyethyl celluloses, hydrogel, dihydroxyphenylalanine, and alginate-polyethylene glycol acrylate. In certain embodiments, the pharmaceutical composition comprises from about 0 to about 10% mucoadhesive polymer excipients selected from the list consisting of carbomer, polyethylene glycol, crospovidone, polycarbophil, hypromellose, and hydroxyethyl cellulose. [00317] In certain embodiments, the pharmaceutical composition comprises from at least from about 0.1% to about 90, from about 0.1% to about 92%, from about 0.1% to about 93%, from about 0.1% to about 95%, from about 0.1% to about 98%, from about 0.1% to about 97%, from about 0.1% to about 98%, or from about 0.1% to about 99% mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises from about 0.1% to about 1% mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises from about 0.5% to about 5% mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises from about 1% to about 10% mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises from about 5% to about 20% mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises from about 10% to about 50% mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises from about 20% to about 75% mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises from about 50% to about 90% mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises from about 75% to about 99% mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises about or at least about 0.1, 0.25, 0.5, 0.75, 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95 percent mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises no more than about 0.1, 0.25, 0.5, 0.75, 1, 2, 3, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95 percent mucoadhesive polymer. In certain embodiments, the pharmaceutical composition comprises 0% mucoadhesive polymer. In this instance, the adhesion to the target site is achieved by use of other pharmaceutically acceptable excipients.
Attorney Docket No.01394-0006-01PCT [00318] To prepare the pharmaceutical compositions used in the present disclosure, a therapeutically effective amount of any of the active compounds described herein (including but not limited to Compound I monofumarate, Compound II, or Compound III, optionally in morphic form) may be admixed with a pharmaceutically acceptable carrier, as described for example in Example 7, to produce a dosage form. [00319] For example, in certain embodiments, the present disclosure relates to a method of preparing a pharmaceutical composition, the method comprising mixing Compound I monofumarate, Compound II or Compound III to at least one pharmaceutically acceptable carrier, additive, and/or excipient. In certain embodiments, the Compound I monofumarate, Compound II or Compound III is screened prior to mixing with the at least one pharmaceutically acceptable carrier, additive, and/or excipient. In certain embodiments, the at least one pharmaceutically acceptable carrier, additive, and/or excipient is screened and/or blended prior to mixing with the Compound I monofumarate, Compound II or Compound III. In certain embodiments, after the Compound I monofumarate, Compound II or Compound III is mixed with the at least one pharmaceutically acceptable carrier, additive, and/or excipient, this first mixture is optionally screened and then mixed with at least one additional pharmaceutically acceptable carrier, additive, and/or excipient, which may be the same type of pharmaceutically acceptable carrier, additive, and/or excipient that was previously added. In some embodiments, this process of optionally screening and mixing with at least one additional pharmaceutically acceptable carrier, additive, and/or excipient is repeated two or more times. [00320] In preparing pharmaceutical compositions in topical dosage form, any of the usual pharmaceutical media may be used. Thus, for liquid or semi-solid topical preparations such as gels, creams, ointments, suspensions, elixirs, and solutions, suitable carriers and additives including water, glycols, oils, alcohols, preservatives, and the like may be used. In certain embodiments, the pharmaceutical composition comprises propylene glycol. In certain embodiments, the pharmaceutical composition comprises carboxypolymethylene. In certain embodiments, the pharmaceutical composition comprises ethylenediaminetetraacetic acid (EDTA). In certain embodiments, the pharmaceutical composition comprises sorbic acid. In certain embodiments, the pharmaceutical composition comprises carbomer. In certain embodiments, the pharmaceutical composition comprises hydroxyethyl cellulose. In certain embodiments, the pharmaceutical composition comprises polyethylene glycol. [00321] For solid topical preparations such as powders, tablets, capsules, and for solid preparations such as suppositories, suitable carriers and additives including starches, sugar
Attorney Docket No.01394-0006-01PCT carriers, such as dextrose, mannitol, lactose, and related carriers, diluents, granulating agents, lubricants, binders, mucoadhesive polymer, disintegrating agents, and the like may be used. If desired, the tablets or capsules may be coated or sustained release by standard techniques. The use of these dosage forms may significantly enhance the bioavailability of the compounds in the patient. In certain embodiments, the pharmaceutical composition comprises a diluent and a glidant. In some embodiments, the pharmaceutical composition comprises two diluents and a glidant. [00322] In certain embodiments, the pharmaceutical composition comprises mannitol. Mannitol is a Generally Recognized As Safe (GRAS) listed naturally occurring sugar alcohol, widely used in pharmaceutical dosage forms and food products. It has been extensively studied including by vaginal route, and is currently in oral, parenteral, nasal and topical products. [00323] In certain embodiments, the pharmaceutical composition comprises magnesium stearate. In certain embodiments, the pharmaceutical composition comprises microcrystalline cellulose. In certain embodiments, the pharmaceutical composition comprises polycarbophil. In certain embodiments, the pharmaceutical composition comprises polyethylene oxide. In certain embodiments, the pharmaceutical composition comprises colloidal silicon dioxide. In certain embodiments, the pharmaceutical composition comprises povidone. In certain embodiments, the pharmaceutical composition comprises isopropyl alcohol. In certain embodiments, the pharmaceutical composition comprises sodium starch glycolate. In certain embodiments, the pharmaceutical composition comprises croscarmellose sodium. In certain embodiments, the pharmaceutical composition comprises crospovidone. In certain embodiments, the pharmaceutical composition comprises hydroxypropylmethylcellulose. In certain embodiments, the pharmaceutical composition comprises lactose. [00324] In certain embodiments, a powder pharmaceutical composition comprises one or more excipient from the group consisting of xanthan gum, microcrystalline cellulose, polyethylene oxide, hydroxypropylmethylcellulose, hydroxypropylcellulose, carboxymethylcellulose sodium, povidone, mannitol, colloidal silicon dioxide, sodium benzoate, sodium starch glycolate, sodium lauryl sulfate, poloxamer 407, polyoxypropylene- polyoxyethylene copolymers, and the like. [00325] In certain embodiments, the pharmaceutical composition comprising an effective amount of a fumarate salt of any of the active compounds described herein including but not limited to Compound I, further comprises a pharmaceutically acceptable
Attorney Docket No.01394-0006-01PCT excipient selected from the list consisting of Acacia, agar, alginic acid, ascorbyl palmitate, bentonite, benzoic acid, butylated hydroxyanisole, butylated hydroxytoluene, butylene glycol, calcium acetate, calcium hydroxide, canola oil, carob bean gum, carrageenan, castor oil, cellulose, corn starch, disodium edetate, erythorbic acid, ethyl lactate, ethylcellulose, glycerin, glyceryl behenate, glyceryl monooleate, glyceryl monostearate, hydroxyethylmethyl cellulose, hydroxypropyl cellulose, hypromellose, lactic acid, lauric acid, lecithin, linoleic acid, medium chain triglycerides, methyl paraben, methylcellulose, microcrystalline cellulose, microcrystalline wax, myristic acid, oleic acid, palmitic acid, peanut oil, pectin, phosphoric acid, polycarbophil, potassium alginate, propionic acid, propyl gallate, propyl paraben, propylene glycol, propylene glycol alginate, silicon dioxide, simethicone, sodium alginate, sodium benzoate, sodium bicarbonate, sodium carboxymethylcellulose, sodium chloride, sodium citrate, sodium lactate, sodium lauryl sulfate, sodium metabisulfate, sodium phosphate, sodium sulfite, sodium thiosulfate, sorbic acid, stearic acid, talc, tapioca starch, tartaric acid, thymol, urea, vitamin E polyethylene succinate, beeswax, xanthan gum, and zinc acetate. [00326] In certain embodiments, the pharmaceutical composition comprises pharmaceutically acceptable excipients for use as a pessary. In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II or Compound III, further comprises up to 99.9% pessary excipient selected from the group consisting of hard fat, PEG, macrogols, cocoa butter, and glycerol. Non-limiting examples of hard fat include Ovucire® (mono-, di- and triglyceride esters of fatty acids (C10 to C18), the triester fraction being predominant and ethoxylated fatty alcohols), Witepsol® (glycerol esters of vegetable saturated fatty acids, such as lauric acid), and Supposi-base™ (a blend of saturated polyglycolysed glycerides). [00327] In certain embodiments, the pharmaceutical composition comprising an effective amount of any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II or Compound III, further comprises a pharmaceutically acceptable excipient that enhances the penetration, disintegration, film forming and/or controlled release properties of the composition. [00328] In certain embodiment, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate, Compound II or
Attorney Docket No.01394-0006-01PCT Compound III, further comprises a penetration enhancing excipient. In certain embodiments, the penetration enhancing excipient is selected from the group consisting of oleic acid, eucalyptol, Caprylol, Labrafil, Labrasol, Lauroglycol, diethylene glycol monomethyl ether (Transcutol), propylene glycol, sodium laurate, sodium lauryl sulfate, cetyltrimethylammonium bromide, poloxamer (231, 182, 184), Tween 20, 40, 60, 80, fatty acids and fatty acid esters, isostearic acid, glycerin, and chitosan. In certain embodiments, the pharmaceutical composition comprising Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III contains from 0% to about 20% penetration enhancing excipients selected from the group consisting of cetyl alcohol, propylene glycol, transcutol P, oleic acid, isopropyl myristate, propylene glycol dicaprylate, glyceryl monooleate, propylene glycol monocaprylate, PEG-8 bees wax, cetyl alcohol, stearic acid, cetyl palmitate, and cetosteryl alcohol. In certain embodiments, the pharmaceutical composition comprises from about 0 to about 25% penetration enhancing excipients selected from the list consisting of stearyl alcohol, polysorbate 80, sodium lauryl sulfate, mono and diglycerides, sorbitan monostearate, glyceryl isostearate, polyoxyl 15 hydroxystearate, polyoxyl 40 hydrogenated castor oil, octyl dodecanol, and soybean lecithin. [00329] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III further comprises a film forming excipient. In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III contains from 0% to about 99% film forming excipients selected from the group consisting of hypromellose, polyethylene glycol, polymethacrylates, microcrystalline cellulose, guar gum, xanthan gum, and polyvinylpyrrolidone. [00330] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises an excipient that allows for controlled release of the active compound. In certain embodiments, the controlled release pharmaceutical composition comprises ethylcellulose, hypromellose, microcrystalline wax, polycarbophil, and/or beeswax.
Attorney Docket No.01394-0006-01PCT [00331] Percentage ranges of excipients and other components of the pharmaceutical composition are given as a percent by weight, unless otherwise specified. [00332] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises a disintegration enhancing excipient. In certain embodiments, the disintegration enhancing excipient is selected from the group consisting of cellulose, guar gum, crospovidone, polyplasdone, soy polysaccharides, calcium silicate, gelatin, cation exchange resins, bentonite, citrus pulp, alginic acid, calcium alginate, methylcellulose, microcrystalline cellulose, sodium carboxymethylcellulose, croscarmellose, solka floc, corn starch, sodium starch glycolate (Explotab, Primojel), glycine, hydroxypropyl starch, and starch 1500. In certain embodiments, the pharmaceutical composition comprises up to about 99% of a disintegration enhancing excipient(s) such as mannitol and/or microcrystalline cellulose. In certain embodiments, the pharmaceutical composition comprises from about 0 to about 70% of a disintegration enhancing excipient(s) selected from the list consisting of lactose, sucrose, and calcium phosphate. In certain embodiments, the pharmaceutical composition comprises from about 0 to about 50% of a disintegration enhancing excipient(s) selected from the list consisting of sodium bicarbonate, citric acid, maleic acid, adipic acid, and fumaric acid. In certain embodiments, the pharmaceutical composition comprises from about 0 to about 20% of a disintegration enhancing excipient(s) selected from the list consisting of sodium starch glycollate, pregelatinized starch, crospovidone, and croscarmellose sodium. [00333] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 70% mannitol, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 20%, about 30% about 40%, about 50%, about 60% or about 70%. [00334] In certain embodiments, the pharmaceutical composition comprising Compound I or a pharmaceutically acceptable salt thereof, such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 70% lactose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60% or about 70%.
Attorney Docket No.01394-0006-01PCT [00335] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from about 0 to about 70% sucrose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60% or about 70%. [00336] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from about 0 to about 70% microcrystalline cellulose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60% or about 70%. [00337] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 20% sodium starch glycolate, including but not limited to any amount that achieves the desired results, for example up to about 1%, about 2%, about 3%, about 5%, about 7%, about 10%, about 12%, about 15% or about 20%. [00338] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from about 0 to about 20% pregelatinized starch, including but not limited to any amount that achieves the desired results, for example up to about 1%, about 2%, about 3%, about 5%, about 7%, about 10%, about 12%, about 15% or about 20%. [00339] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from about 0 to about 20% crospovidone, including but not limited to any amount that achieves the desired results, for example up to about 1%, about 2%, about 3%, about 5%, about 7%, about 10%, about 12%, about 15% or about 20%. [00340] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or
Attorney Docket No.01394-0006-01PCT Compound III, further comprises from about 0 to about 20% croscarmellose sodium, including but not limited to any amount that achieves the desired results, for example up to about 1%, about 2%, about 3%, about 5%, about 7%, about 10%, about 12%, about 15% or about 20%. [00341] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% xanthan gum, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20% about 25%, about 30%, about 35%, about 40%, about 45% or about 50%. [00342] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% polycarbophil, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%. [00343] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% polyethylene oxide, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%. [00344] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% hydroxyethylmethyl cellulose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%. [00345] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a
Attorney Docket No.01394-0006-01PCT pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% hydroxyethyl cellulose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%. [00346] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% hypromellose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%. [00347] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% hydroxypropyl cellulose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%. [00348] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% PVP, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%. [00349] In certain embodiments, the pharmaceutical composition comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises from 0 to about 50% microcrystalline cellulose, including but not limited to any amount that achieves the desired results, for example up to about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45% or about 50%. [00350] In typical embodiments according to the present disclosure, any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically
Attorney Docket No.01394-0006-01PCT acceptable salt thereof such as a monofumarate or Compound II or Compound III, and the compositions described are used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV. [00351] In certain embodiments, a tablet used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises about 250 mg of microcrystalline cellulose, about 20 mg of crospovidone, about 5 mg of magnesium stearate, about 5 mg of silicon dioxide, about 5 mg of polyethylene oxide, and about 100 mg of mannitol. In certain embodiments, a tablet used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV further comprises about 155 mg microcrystalline cellulose, about 1.75 mg of magnesium stearate, and about 17.5 mg of mannitol. [00352] In certain embodiments, a semi-solid formulation used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises about 15 mg of carbomer, about 50 mg of propylene glycol, about 10 mg of sorbic acid, about 5 mg of EDTA, and about 920 mg of water. In certain embodiments, a semi-solid formulation used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises about 20 mg of carbomer; about 70 mg of mineral oil; about 80 mg of a mixture of polyoxyl 6 stearate Type I, ethylene glycol stearates and polyoxyl 32 stearate type 1; about 5 mg parabens; about 60 mg propylene glycol; about 5 mg EDTA; and about 760 mg water. [00353] In certain embodiments, a dry powder for reconstitution is used to treat, prevent, or delay an HPV infection or a secondary disease state, condition, or complication of HPV comprising any of the active compounds described herein, including but not limited to Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III, further comprises about 15.5 mg xanthan gum, about 19.8 mg mannitol, about 5 mg silicon dioxide, and about 0.5 mg sodium benzoate.
Attorney Docket No.01394-0006-01PCT [00354] The treatments described herein for cervical or vaginal intraepithelial neoplasia can be combined with conventional approaches such as, but not limited to, excision or ablation of the transformed zone. Techniques include cryotherapy, laser therapy, loop electrosurgical procedure (LEEP), and cone biopsy. All of these surgical procedures damage the affected areas and can lead to scarring. The most common intervention for cervical intraepithelial neoplasia, LEEP, is effective in 60-90% of cases, however, it has been associated with a significantly increased risk of miscarriage, ectopic pregnancies, and negative psychological outcomes. In certain embodiments, the treatments described herein in combination with the retaining device are used to lessen, ameliorate or substitute the use of these conventional practices. [00355] In certain embodiments, the treatments described herein can be used in combination with a surgical technique. In certain embodiments, a patient in need thereof can receive surgery before, during and/or after administration of an effective amount of a compound described herein. In certain embodiments, the compound is administered in combination with the retaining device about or less than about 8 months, 6 months, 4 months, 3 months, 2 months, or 1 month prior to surgery. In certain embodiments, the compound is administered in combination with the retaining device about or at least about 1 month, 2 months, 3 months, 4 months, 6 months, or 8 months after surgery. In certain embodiments, the surgical procedure can be an excision of the target and/or diseased tissue, including but not limited to loop electrosurgical excision procedure (LEEP), large loop excision of the transformation zone (LLETZ), knife conization, cold knife conization, knife cone biopsy, or laser conization. In certain embodiments, the surgical procedure can be ablation, including but not limited to laser ablation or cryoablation. [00356] The efficacy of a drug against an HPV infection or neoplasia may be prolonged, augmented, or restored by administering the compound in combination or alternation with another, and perhaps even two or three other, antiviral compounds that induce a different mutation or act through a different pathway, from that of the principal drug. Alternatively, the pharmacokinetic, biodistribution, half-life, or other parameters of the drug can be altered by such combination therapy (which may include alternation therapy if considered concerted). Furthermore, since HPV is associated with several types of cancer, combination therapy with anticancer therapeutics can provide better outcomes for patients. Since the disclosed Compound I or a pharmaceutically acceptable salt thereof such as a monofumarate or Compound II or Compound III are DNA polymerase inhibitors, it may be
Attorney Docket No.01394-0006-01PCT useful to administer the compound to a host in need thereof in combination with, for example: a) a protease inhibitor; b) another DNA polymerase inhibitor; c) an inhibitor of E6 or E6AP such as MEDI0457, luteolin, CAF-24 or gossypetin; d) an inhibitor of E7; e) an inhibitor of E1 or E2, including inhibitors of the E1-E2 protein interaction; f) L2 lipopeptides; g) an inhibitor or degrader of L1 or L2; h) an HDAC inhibitor such as vorinostat; i) degraders of tetraspanins such as CD9, CD63 or CD151; j) immunotherapeutics such as T-cell therapies (including adoptive T-cell therapies) and checkpoint inhibitors; k) anti-proliferative drugs; l) a therapeutic vaccine; m) a prophylactic vaccine; n) trichloroacetic acid; o) salicylic acid; p) imiquimod; q) podofilox; r) Gardasil® 9; s) Gardasil® 4; t) Cervarix; u) VGX-3100; v) GGX-188E; and/or w) ADXS11-001. [00357] Any of the materials described herein that include Compound I in any stereochemical designation, or mixtures thereof, along with one or more additional compounds in any selected ratio, such as but not limited to an acid as described herein or water (or other solvent) in a solid form, can be used for HPV anti-viral or intraepithelial neoplasia therapy. [00358] Such solid forms, when they contain Compound I so described and one or more additional compounds, may be considered multicomponent solids and, when crystalline,
Attorney Docket No.01394-0006-01PCT multicomponent crystalline forms. For example, “crystalline A B” would be considered a multicomponent crystalline form of compounds A and B. [00359] A salt is an example of a multicomponent solid, and a crystalline salt is a multicomponent crystalline form. Salts typically exhibit proton transfer between an acid and a base. [00360] Other multicomponent crystalline forms include cocrystals. A cocrystal is typically a crystalline solid of two or more neutral species. Because charge balance is not necessary for neutral species, the stoichiometry of cocrystals cannot be determined based on charge balance. In pharmaceutically relevant cocrystals, one component is typically an active drug or prodrug and the other (or others) is usually termed a coformer, and it interacts nonionically with the active drug or prodrug in the crystal lattice. [00361] In some embodiments, the solid form may be both a salt and a cocrystal. For example, the active drug or prodrug may be in the form of a salt with a counterion and interact nonionically with a coformer. Such a solid form would be a multicomponent solid and, when crystalline, a multicomponent crystalline form. Examples of such cocrystals when the coformer is volatile includes solvates. [00362] A solid form may exist in a single crystalline solid form or be polymorphic where more than one crystalline form of the same solid form exists. Polymorphism is typically determined by use of x-ray powder diffraction. [00363] Other examples of multicomponent solids are hydrates and solvates. A hydrate is a solid form that includes water in the solid form. In crystalline hydrates, the water may form part of the unit cell of the crystal in which case it is often a stoichiometric hydrate. A solvate is a solid form that includes one or more solvent molecules and, when crystalline, may form part of the unit cell of the crystal in which case it is often a stoichiometric solvate. A hydrate or a solvate can also be used in the therapy described generally herein. [00364] In some embodiments, the solid form may be partially crystalline, disordered, or amorphous. [00365] In certain embodiments, any of the solid forms described above that comprise Compound I can be used in an effective amount in the treatment of HPV infection or cervical or vaginal intraepithelial neoplasia in a patient in need thereof, as generally taught herein. A multicomponent crystal comprising Compound I can include a salt form of Compound I, a cocrystal form of Compound I, and/or a mixture of a salt and cocrystal form of Compound I. The solid form formed can be a function of the synthesis and crystallization or solidification conditions used, among other factors.
Attorney Docket No.01394-0006-01PCT [00366] In certain embodiments, an effective amount of a multicomponent crystal comprising Compound I and a pharmaceutically acceptable coformer is used in the treatment of cervical or vaginal intraepithelial neoplasia in a patient in need thereof. In certain embodiments, an effective amount of a multicomponent crystal comprising Compound I and a pharmaceutically acceptable coformer is used in combination with a retaining device to treat cervical or vaginal intraepithelial neoplasia in a patient in need thereof. [00367] In certain embodiments, the crystalline form formed by addition of about 1 equivalent of fumaric acid to a solution of Compound I optionally in isopropyl alcohol and heptane, optionally in a pharmaceutically acceptable carrier, is used in the treatment of cervical or vaginal intraepithelial neoplasia, optionally in combination with a retaining device. [00368] Any solid form comprising Compound I or a salt thereof can be used in an effective amount with or without a retaining device to deliver therapy to the HPV-infected patient in need thereof. [00369] Any solid form comprising Compound I or a salt thereof can be used in an effective amount with or without a retaining device to deliver therapy to a patient infected with a virus chosen from adenovirus, human cytomegalovirus, BK virus, and hepatitis B virus. Additional Embodiments [00370] In certain embodiments, the present disclosure that includes the combined use of the therapeutic compounds as described herein with a retaining device includes the use of one of the following compounds below which may optionally be used in a morphic form as described herein, and further embodiments that include such compounds. 1. A compound of the formula: ,
Attorney Docket No.01394-0006-01PCT or
or
A compound of the formula:
Attorney Docket No.01394-0006-01PCT or
.
Attorney Docket No.01394-0006-01PCT 5. The compound of embodiment 3 of the formula: or .
neoplasia) and/or retaining device system as described herein that includes a pharmaceutical composition comprising the compound of any one of embodiments 1-5, and a pharmaceutically acceptable carrier. 7. The therapeutic use and/or retaining device system of embodiment 6, wherein the pharmaceutical composition is in a solid dosage form. 8. The therapeutic use and/or retaining device system of embodiment 6, wherein the pharmaceutical composition is in a semi-solid dosage form. 9. The therapeutic use and/or retaining device system of embodiment 6, wherein the pharmaceutical composition is in the form of a film. 10. The therapeutic use and/or retaining device system of embodiment 6, wherein the pharmaceutical composition is in the form of a pessary. 11. The therapeutic use and/or retaining device system of embodiment 7, wherein the pharmaceutical composition is in the form of a tablet.
Attorney Docket No.01394-0006-01PCT 12. The therapeutic use and/or retaining device system of embodiment 8, wherein the pharmaceutical composition is in the form of a cream. 13. The therapeutic use and/or retaining device system of embodiment 8, wherein the pharmaceutical composition is in the form of a gel. 14. The therapeutic use and/or retaining device system of any one of embodiments 6-13, wherein the pharmaceutical composition is formulated for topical administration. 15. The therapeutic use and/or retaining device system of any one of embodiments 6-14, wherein the pharmaceutical composition is formulated for delivery to the cervix. 16. The therapeutic use and/or retaining device system of any one of embodiments 6-14, wherein the pharmaceutical composition is formulated for delivery to the vagina. 17. The therapeutic use and/or retaining device system of any one of embodiments 6-14, wherein the pharmaceutical composition is formulated for delivery to the vulva. 18. The therapeutic use and/or retaining device system of embodiment 11, wherein the tablet is a bilayer tablet. 19. The therapeutic use and/or retaining device system of embodiment 11 or 18, wherein the tablet disintegrates in less than about 250 μL of fluid. 20. The therapeutic use and/or retaining device system of embodiment 19, wherein the tablet disintegrated in less than about 150 μL of fluid. 21. The therapeutic use and/or retaining device system of any one of embodiments 6-20, wherein the pharmaceutical composition comprises from about 0.005 to about 10 mg, from about 0.01 mg to about 5 mg, from about 0.03 mg to about 1 mg, or from about 0.05 mg to about 0.3 mg of the active compound. 22. The therapeutic use and/or retaining device system of any one of embodiments 6-20, wherein the pharmaceutical composition comprises from about 0.05 mg to about 5 mg of the active compound. 23. The therapeutic use and/or retaining device system of embodiment 22, wherein the pharmaceutical composition comprises from about 0.05 mg to about 0.5 mg of the active compound. 24. The therapeutic use and/or retaining device system of embodiment 23, wherein the pharmaceutical composition comprises from about 0.1 mg to about 0.3 mg of the active compound. 25. The therapeutic use and/or retaining device system of embodiment 22, wherein the pharmaceutical composition comprises at least about 0.05 mg of the active compound.
Attorney Docket No.01394-0006-01PCT 26. The therapeutic use and/or retaining device system of embodiment 22, wherein the pharmaceutical composition comprises at least about 0.1 mg of the active compound. 27. The therapeutic use and/or retaining device system of embodiment 22, wherein the pharmaceutical composition comprises at least about 0.3 mg of the active compound. 28. The therapeutic use and/or retaining device system of embodiment 22, wherein the pharmaceutical composition comprises at least about 1 mg of the active compound. 29. The therapeutic use and/or retaining device system of embodiment 22, wherein the pharmaceutical composition comprises at least 1.5 mg of the active compound. 30. The therapeutic use and/or retaining device system of embodiment 22, wherein the pharmaceutical composition comprises at least about 2 mg of the active compound. 31. The therapeutic use and/or retaining device system of embodiments 12-13, wherein the pharmaceutical composition comprises from about 0.01% to about 10% of the active compound. 32. The therapeutic use and/or retaining device system of embodiments 12-13, wherein the pharmaceutical composition comprises from about 0.01% to 0.5% of the active compound. 33. The therapeutic use and/or retaining device system of embodiments 12-13, wherein the pharmaceutical composition comprises from about 0.1% to 5% of the active compound. 34. The therapeutic use and/or retaining device system of any one of embodiments 6-33, wherein the pharmaceutical composition comprises a mucoadhesive polymer. 35. The therapeutic use and/or retaining device system of any one of embodiments 6-33, wherein the pharmaceutical composition comprises from about 5% to about 20% of a mucoadhesive polymer. 36. The therapeutic use and/or retaining device system of any one of embodiments 6-33, wherein the pharmaceutical composition comprises from about 10% to about 50% of a mucoadhesive polymer. 37. The therapeutic use and/or retaining device system of any one of embodiments 6-33, wherein the pharmaceutical composition comprises from about 50% to about 90% of a mucoadhesive polymer.
Attorney Docket No.01394-0006-01PCT 38. The therapeutic use and/or retaining device system of any one of embodiments 6-37, wherein the pharmaceutical composition comprises a disintegration enhancing excipient. 39. The therapeutic use and/or retaining device system of any one of embodiments 6-38, wherein the pharmaceutical composition comprises a penetration enhancing excipient. 40. The therapeutic use and/or retaining device system of any one of embodiments 6-39, wherein the pharmaceutical composition comprises an excipient which allows for the controlled release of the active compound. 41. The therapeutic use and/or retaining device system of embodiment 12, wherein the pharmaceutically acceptable carrier is selected from at least one of light mineral oil, propylparaben, Tefose 63, water, EDTA, methylparaben, and Carbopol 974P. 42. The therapeutic use and/or retaining device system of embodiment 13, wherein the pharmaceutically acceptable carrier is selected from at least one of EDTA, methyl paraben, Carbopol 974P, propylene glycol, and sorbic acid. 43. The therapeutic use and/or retaining device system of embodiment 11, wherein the tablet comprises mannitol, a polycrystalline cellulose (such as microcrystalline cellulose), and magnesium stearate. 44. A method to treat a cervical or vaginal HPV-induced infection or a condition caused by a human papillomavirus in a human in need thereof, including but not limited to cervical or vaginal intraepithelial neoplasia, the method comprising administering to a host in need thereof an effective amount of the compound or pharmaceutical composition for use as described in any one of embodiments 1-43, with a vaginal applicator in combination with a retaining device for a sufficient time to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue. 45. A method to treat a cervical or vaginal HPV-induced infection or a condition caused by a human papillomavirus, including by not limited to intraepithelial neoplasia, in a human in need thereof, the method comprising: (i) administering an effective amount of the compound or pharmaceutical composition as described in any one of embodiments 1-43 to the cervix using a vaginal applicator (which is optionally coated with a lubricant); and then (ii) inserting a retaining device (which is optionally coated with a lubricant) that is maintained at the base of the cervix or in the vaginal canal for sufficient time to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its
Attorney Docket No.01394-0006-01PCT metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non- diseased tissue. 46. The method of embodiment 44 or 45, wherein the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap, and a sponge. 47. The method of embodiment 44 or 45, wherein the retaining device is used as both the applicator and a retaining means to collect the post-treatment leakage of vaginal fluids and the therapeutic agent or its metabolite such as PMEG in a manner that minimizes toxicity-causing damage to non-diseased tissue. 48. The method of any one of embodiment 44-47, wherein the retaining device is a menstrual cup. 49. The method of any one of embodiment 44-47, wherein the retaining device is a menstrual disc. 50. The method of embodiment 44 or 45, wherein the retaining device is a tampon. 51. The method of any one of embodiments 44, 45, or 47, wherein the retaining device is a diaphragm. 52. The method of embodiment 44 or 45, wherein the retaining device is a cervical cap. 53. The method of embodiment 44 or 45, wherein the retaining device is a sponge. 54. The method of embodiment 47, wherein the retaining device used as both an applicator and a retaining means is a diaphragm. 55. The method of any one of embodiments 44-54, wherein the condition caused by a human papillomavirus is atypical squamous cells of undetermined significance (ASC- US). 56. The method of any one of embodiments 44-54, wherein the condition caused by a human papillomavirus is atypical glandular cells (AGC). 57. The method of any one of embodiments 44-54, wherein the condition caused by a human papillomavirus is atypical squamous cells, cannot exclude high grade squamous intraepithelial lesion (ASC-H). 58. The method of any one of embodiments 44-54, wherein the condition caused by a human papillomavirus is adenocarcinoma in situ (AIS). 59. The method of any one of embodiments 44-54, wherein the intraepithelial neoplasia is cervical intraepithelial neoplasia.
Attorney Docket No.01394-0006-01PCT 60. The method of embodiment 59, wherein the cervical intraepithelial neoplasia is Grade 1 cervical intraepithelial neoplasia. 61. The method of embodiment 59, wherein the cervical intraepithelial neoplasia is Grade 2 cervical intraepithelial neoplasia. 62. The method of embodiment 59, wherein the cervical intraepithelial neoplasia is Grade 3 cervical intraepithelial neoplasia. 63. The method of any one of embodiments 44-54, wherein the intraepithelial neoplasia is vaginal intraepithelial neoplasia. 64. The method of any one of embodiments 44-63, wherein from about 0.05 milligrams to about 40 milligrams of active compound is administered. 65. The method of any one of embodiments 44-63, wherein from about 0.1 milligrams to about 30 milligrams of active compound is administered. 66. The method of any one of embodiments 44-63, wherein from about 0.001 to about 20 mg, from about 0.005 to about 10 mg, from about 0.01 mg to about 5 mg, from about 0.03 mg to about 1 mg, from about 0.05 mg to about 0.3 mg, from about 0.03 mg to about 0.07 mg, from about 0.05 mg to about 0.15 mg, or from about 0.15 mg to about 0.45 mg of the active compound is administered. 67. The method of any one of embodiments 44-63, wherein from about 0.05 mg to about 0.3 mg of the active compound is administered. 68. The method of any one of embodiments 44-67, further comprising applying a lubrication means to the epithelial tissue before administering the dosage form to the affected area. 69. The method of any one of embodiments 44-67, further comprising applying a lubrication means to the dosage form before administering the dosage form to the affected area. 70. The method of embodiment 68 or 69, wherein the lubrication means is selected from water, a glycerol-based lubricant, and a hydroxyethylcellulose-based lubricant. 71. The method of any one of embodiments 44-70, wherein the compound or pharmaceutical composition is administered once per day. 72. The method of any one of embodiments 44-70, wherein the compound or pharmaceutical composition is administered twice per day. 73. The method of any one of embodiments 44-70, wherein the compound or pharmaceutical composition is administered twice per week.
Attorney Docket No.01394-0006-01PCT 74. The method of any one of embodiments 44-70, wherein the compound or pharmaceutical composition is administered three times or more per week. 75. The method of any one of embodiments 44-74, wherein the compound or pharmaceutical composition is administered for about one week. 76. The method of any one of embodiments 44-74, wherein the compound or pharmaceutical composition is administered for about two weeks. 77. The method of any one of embodiments 44-74, wherein the compound or pharmaceutical composition is administered for about three weeks. 78. The method of any one of embodiments 44-74, wherein the compound or pharmaceutical composition is administered for about four weeks. 79. The method of any one of embodiments 44-74, wherein the compound or pharmaceutical composition is administered for about five weeks. 80. The method of any one of embodiments 44-74, wherein the compound or pharmaceutical composition is administered for about six weeks or about eight weeks. 81. The method of any one of embodiments 44-80, wherein the compound or pharmaceutical composition is wherein the compound is administered in a therapeutic cycle comprising: a. a treatment cycle comprising administering the compound or pharmaceutical composition, and b. a rest cycle comprising a period of no treatment. 82. The method of embodiment 81, wherein the rest cycle is about one week. 83. The method of embodiment 81, wherein the rest cycle is about two weeks. 84. The method of embodiment 81, wherein the rest cycle is about three weeks. Further Embodiments [00371] In certain embodiments, the present disclosure relates to the following further embodiments: 1. A method of treating a cervical or vaginal HPV-induced infection or an associated condition in a female in need thereof, including cervical or vaginal intraepithelial neoplasia, comprising (i) administering to the base of the cervix an effective amount of dosage form of a therapeutic compound selected from:
Attorney Docket No.01394-0006-01PCT
(ii) and then inserting a retaining device. 2. A method of treating a cervical or vaginal HPV-induced infection or an associated condition in a female in need thereof, including cervical or vaginal intraepithelial neoplasia, comprising (i) administering an effective amount of dosage form of a therapeutic compound selected from: or
Attorney Docket No.01394-0006-01PCT and then (ii) inserting a retaining device. 3. A method of treating a cervical or vaginal HPV-induced infection or an associated condition in a female in need thereof, including cervical or vaginal intraepithelial neoplasia, comprising (i) administering a dosage form with an effective amount of a therapeutic compound selected from: or
a pharmaceutically acceptable salt thereof;
(ii) inserting a retaining device.
Attorney Docket No.01394-0006-01PCT 4. The method of embodiment 2, wherein the pharmaceutically acceptable salt is selected from:
acceptable salt is selected from:
Attorney Docket No.01394-0006-01PCT .
wherein the compound or salt thereof is in a pharmaceutically acceptable composition. 7. The method of embodiment 6, wherein the pharmaceutical composition is a solid dosage form. 8. The method of embodiment 6, wherein the pharmaceutical composition is a semi- solid dosage form. 9. The method of embodiment 7, wherein the solid pharmaceutical composition is in the form of a vaginal tablet. 10. The method of any one of embodiments 1-9, wherein the pharmaceutical composition is administered with a vaginal applicator. 11. The method of any one of embodiments 1-10, wherein the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap and a sponge. 12. The method of any one of embodiments 1-11, wherein the retaining device is used as both the applicator and a retaining means. 13. The method of embodiment 11, wherein the retaining device is a menstrual cup. 14. The method of embodiment 11, wherein the retaining device is a menstrual disc. 15. The method of embodiment 11, wherein the retaining device is a tampon. 16. The method of embodiment 11, wherein the retaining device is a diaphragm.
Attorney Docket No.01394-0006-01PCT 17. The method of embodiment 11, wherein the retaining device is a cervical cap. 18. The method of embodiment 11, wherein the retaining device is a sponge. 19. The method of any one of embodiments 1-18, wherein the compound is administered in a morphic form. 20. The method of any one of embodiments 1-19, wherein the dosage form is administered to the base of the cervix. 21. The method of any one of embodiments 1-20, wherein the retaining device is placed in the vaginal area, for example, below the cervical base and, for example, for a sufficient time to collect the post-treatment leakage of the therapeutic compound or its PMEG metabolite. 22. The method of any one of embodiments 4 or 6-20, wherein the compound is an isolated morphic form of: wherein the isolated
comprising peaks independently selected from at least 3, 4, 5, or 6 of the following 2theta values 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. 23. The method of any one of embodiments 5-20, wherein the compound is an isolated morphic form of: wherein the isolated
comprising peaks independently selected from at least 3, 4, 5, 6, 7, 8 or 9 of the following 2theta values
Attorney Docket No.01394-0006-01PCT 9.53+0.2°, 10.04+0.2°, 11.60+0.2°, 14.57+0.2°, 17.22+0.2°, 17.50+0.2°, 20.04+0.2°, 20.36+0.2°, 22.34+0.2°, 23.73+0.2°, 25.48+0.2°, 26.06+0.2°, 27.38+0.2°, and 32.20+0.2°. 24. The method of any one of embodiments 5-20, wherein the compound is an isolated morphic form of: wherein the isolated comprising peaks
independently or 2theta values 8.94+0.2°, 9.89+0.2°, 9.91+0.2°, 11.66+0.2°, 12.11+0.2°, 15.13+0.2°, 17.85+0.2°, 18.15+0.2°, 19.90+0.2°, 20.38+0.2°, 22.94+0.2°, 25.09+0.2°, 26.54+0.2°, 26.90+0.2°, 27.38+0.2°, 28.28+0.2°, 28.95+0.2°, 29.64+0.2°, and 38.07+0.2°. 25. The method of any one of embodiments 1 or 6-20, wherein the compound is an isolated morphic form of the compound of the formula: wherein the isolated
comprising peaks independently selected from at least 3, 4, 5, or 6 of the following 2theta values 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. 26. The method of embodiment 9, wherein the tablet disintegrates in less than about 250 μL of fluid. 27. The method of embodiment 9, wherein the table disintegrated in less than about 150 μL of fluid. 28. The method of any one of embodiments 1-27, wherein about 0.05 to 1 mg of therapeutic compound or pharmaceutically acceptable salt thereof is administered.
Attorney Docket No.01394-0006-01PCT 29. The method of any one of embodiments 1-27, wherein from about 0.1 to 1 mg of therapeutic compound or pharmaceutically acceptable salt thereof is administered. 30. The method of any one of embodiments 1-27, wherein from about 0.1 to 0.5 mg of therapeutic compound or pharmaceutically acceptable salt thereof is administered. 31. The method of any one of embodiments 1-27, wherein from about 0.2 to 0.3 mg of therapeutic compound or pharmaceutically acceptable salt thereof is administered. 32. The method of any one of embodiments 1-27, wherein about 0.3 mg of therapeutic compound or pharmaceutically acceptable salt thereof is administered. 33. The method of embodiment 6, wherein the pharmaceutical composition comprises from about 0.01% to 10% of the therapeutic compound or pharmaceutically acceptable salt thereof. 34. The method of embodiment 6, wherein the pharmaceutical composition comprises a polymer. 35. The method of embodiment 6, wherein the pharmaceutical composition comprises a disintegration enhancing excipient. 36. The method of embodiment 6, wherein the pharmaceutical composition comprises a penetration enhancing excipient. 37. The method of embodiment 6, wherein the pharmaceutical composition comprises an excipient that allows for controlled release. 38. The method of embodiment 6, wherein the pharmaceutical composition comprises at least one of a light mineral oil, propylparaben, Tefose 63, water, EDTA, methylparaben, and Carbopol 974P. 39. The method of embodiment 6, wherein the pharmaceutical composition comprises at least one of EDTA, methyl paraben, Carbopol 974P, propylene glycol, and sorbic acid. 40. The method of embodiment 6, wherein the pharmaceutical composition comprises mannitol, a polycrystalline cellulose (such as microcrystalline cellulose), and magnesium stearate. 41. The method of any one of embodiments 1-40, wherein the condition associated with a human papillomavirus is atypical squamous cells of undetermined significance (ASC-US).
Attorney Docket No.01394-0006-01PCT 42. The method of any one of embodiments 1-40, wherein the condition associated with a human papillomavirus is atypical glandular cells (AGC). 43. The method of any one of embodiments 1-40, wherein the condition associated with a human papillomavirus is atypical squamous cells, cannot exclude high grade squamous intraepithelial lesion (ASC-H). 44. The method of any one of embodiments 1-40, wherein the condition associated with a human papillomavirus is adenocarcinoma in situ (AIS). 45. The method of any one of embodiments 1-40, wherein the condition is cervical intraepithelial neoplasia. 46. The method of any one of embodiments 1-40, wherein the condition is cervical intraepithelial neoplasia Grade 1. 47. The method of any one of embodiments 1-40, wherein the condition is cervical intraepithelial neoplasia Grade 2. 48. The method of any one of embodiments 1-40, wherein the condition is cervical intraepithelial neoplasia Grade 3. 49. The method of any one of embodiments 1-40, wherein the condition is vaginal intraepithelial neoplasia. 50. The method of any one of embodiments 1-49, further comprising applying a lubrication means to the epithelial tissue before inserting the dosage form in the affected area. 51. The method of any one of embodiments 1-50, further comprising applying a lubrication means to the dosage form before administering the dosage form to the affected area. 52. The method of embodiment 50 or 51, wherein the lubrication means is selected from water, a glycerol-based lubricant, and a hydroxyethylcellulose-based lubricant. 53. The method of any one of embodiments 1-52, wherein the dosage form is administered once per day. 54. The method of any one of embodiment 1-53, wherein the dosage form is administered twice per week. 55. The method of any one of embodiment 1-53, wherein the dosage form is administered three times or more per week.
Attorney Docket No.01394-0006-01PCT 56. The method of any one of embodiments 1-55, wherein the dosage form is administered for about one week. 57. The method of any one of embodiments 1-55, wherein the dosage form is administered for about two weeks. 58. The method of any one of embodiments 1-55, wherein the dosage form is administered for about three weeks. 59. The method of any one of embodiments 1-55, wherein the dosage form is administered for about four weeks. 60. The method of any one of embodiments 1-55, wherein the dosage form is administered for about five weeks. 61. The method of any one of embodiments 1-55, wherein the dosage form is administered for about six weeks or for about eight weeks. 62. The method of any one of embodiments 1-55, wherein the dosage form is administered in a therapeutic cycle comprising: a treatment cycle comprising administering the compound, and a rest cycle comprising a period of no treatment. 63. The method of embodiment 62, wherein the rest cycle is about one week. 64. The method of embodiment 62, wherein the rest cycle is about two weeks. 65. The method of embodiment 62, wherein the rest cycle is about three weeks. 66. A kit that comprises a dosage form with a therapeutic compound or salt thereof of any one of embodiments 1-5 and a retaining device. 67. The kit of embodiment 66, that further comprises a vaginal applicator. 68. The kit of embodiment 66 or 67, wherein the dosage form is a vaginal tablet. 69. The kit of any one of embodiments 66-68, wherein the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap, and a sponge. 70. The kit of any one of embodiments 66-69, wherein the retaining device is used as both the applicator and a retaining means. 71. The kit of embodiment 70, wherein the retaining device is a menstrual cup. 72. The kit of embodiment 70, wherein the retaining device is a menstrual disc.
Attorney Docket No.01394-0006-01PCT 73. The kit of embodiment 70, wherein the retaining device is a tampon. 74. The kit of embodiment 70, wherein the retaining device is a diaphragm. 75. The kit of embodiment 70, wherein the retaining device is a cervical cap. 76. The kit of embodiment 70, wherein the retaining device is a sponge. 77. The kit of any one of embodiments 66-76, wherein the therapeutic compound or salt thereof is administered in a morphic form. EXAMPLES [00372] All vaginal gels and tablets used in these examples were prepared using the monofumarate salt of Compound I. The dose strengths in mg used herein refer to the mass of the active acyclic phosphonamidate (Compound I) and do not include the salt in the molecular weight unless specifically noted. Example 1: Preparation of Fumarate Salts Preparation of Compound I Hemifumarate [00373] About 200 mg of Compound I free base was added to 0.5 mL of EtOH. While stirring, 0.5 molar amount of fumaric acid was added at 50°C and the mixture was stirred for 2 hours. A clear solution was obtained. The solution was then cooled to 25°C within 1 hour. Hemi-fumarate seeds (prepared from a 50 mg batch as described immediately below) were added, followed by addition of 2.5 mL of heptanes to induce precipitation. An oil was obtained and stirred at 25°C for about 4 days. After 4 days, the resulting suspension was cooled to 5°C. After stirred at 5°C for about 4 days, the precipitated solids were collected by filtration and dried at 40°C under vacuum for about 3 hours. As a result, 116 mg of light- orange hemi-fumarate were obtained in yield of 52%. [00374] The hemi-fumarate seed crystals discussed immediately above were prepared by adding about 50 mg of Compound I (free base) to EtOH.0.5 molar amount of fumaric acid was added under stirring at 50°C for 2 hours and then at 25°C for at least 12 hours. Seeds crystals (prepared from a 30 mg batch as described immediately below) were added, and heptanes (0.4 mL) was added as antisolvent. Obtained mixture was stirred at 5°C for about 3 days. Then the suspension was taken out and centrifuged. Solids obtained were dried in oven at 50°C for about 1 hour under vacuum, and analyzed by XRPD to confirm the crystals were the Hemifumarate Pattern 1.
Attorney Docket No.01394-0006-01PCT [00375] The monofumarate seed crystals discussed immediately above were prepared by adding about 30 mg [00376] Compound I (free base) into an ethanol-heptane solvent system, followed by addition of 0.5 molar equivalents fumaric acid under stirring at 50°C. The mixture was stirred for 2 hours at 50°C and then at 25°C for at least 12 hours. Half the volume was evaporated in a fume hood and the obtained suspensions were taken out and centrifuged. Solids obtained were analyzed and found to be Hemifumarate Pattern 1. Preparation of Compound I Monofumarate Pattern 1 (small scale preparation) [00377] About 244 mg of Compound I free base was added into 0.8 mL of IPA. Then, 1.0 eq. of fumaric acid was added with stirring at 50°C for about 1.5 hours. The yellow clear solution obtained was cooled to 25°C and stirred for about 5 minutes. Monofumarate seeds(prepared from a 30 mg batch as described immediately below) were added to the mixture, followed by addition of 4 mL of heptanes as an antisolvent. The mixture was stirred at 25°C for 4 days. The precipitated material was collected by filtration and dried at 40°C under vacuum for about 2 hours. As a result, 208 mg of mono-fumarate solids were obtained in yield of 69%. [00378] The monofumarate seed crystals discussed immediately above were prepared by adding about 30 mg of Compound I (free base) to 0.1 mL of IPA. An equimolar amount of fumaric acid was added to the mixture under stirring at 50°C for 2 hours, a yellow clear solution was obtained, and 0.3 mL of heptane was added as antisolvent. Some seeds (prepared from a 50 mg batch as described immediately below) were added. Suspension was obtained and it was stirred at 25°C for at least 12 hours. Obtained suspension was taken out and centrifuged and analyzed by XRPD to confirm the crystals were the Monofumarate Pattern 1. [00379] The monofumarate seed crystals discussed immediately above were prepared by adding about 50 mg of Compound I (free base) to 0.1 mL of IPA.1.5 molar amount of fumaric acid was added to the mixture and the mixture was stirred at 25°C. After about 3 hours, the sample was too sticky, 0.1 mL additional IPA was added and kept stirring at 25°C for about 22 hours. Sticky sample was obtained and dried in a vacuum oven at 50°C for about 2 hours, and reslurried in 1.0 mL of MTBE at 25°C for about 5 days. Obtained suspension was taken out and centrifuged. Solids obtained were dried in oven at 40°C under vacuum for about 1 hour, and analyzed by XRPD to confirm the crystals and found to be Monofumarate Pattern 1.
Attorney Docket No.01394-0006-01PCT Example 2: Large Scale Preparation of Mono-Fumarate [00380] About 4.16 g of Compound I free base was dissolved in 11 mL of IPA. Then, 1.0 eq. of fumaric acid was added to the yellow clear solution under stirring at 50°C. After about 1 hour, some solids precipitated out. Then, 10 mg seeds of mono-fumarate (from the above ‘Small scale preparation’) were added. The mixture was stirred at 50°C for about 1.5 hours and cooled to 25°C, then it was stirred at 25°C for about 10 min. Then, 40 mL of heptanes was added as antisolvent. Obtained suspension was stirred at 25°C for about 24 hours, then cooled to 5°C at a rate of 0.1°C/min and stirred at 5°C for about 1 day. The solids were collected by filtration and dried in the oven at 40°C for about 2 hours under vacuum. About 4.1 g of light pink solids were obtained in a yield of 81.2%. Example 3: Synthesis of Mixture of (R, S) and (S, S) ethyl-2-((((2-(2-amino-6-methoxy- 9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate (Compound I)
Attorney Docket No.01394-0006-01PCT Step 1: Preparation of diethyl-((2-(2-amino-6-chloro-9H-purin-9-yl)-ethoxy)-methyl)- phosphonate (3)
1 (50 g, 0.296 mol, 1 equiv.), Cs2CO3 (96.37 g, 0.296 mol, 1 equiv.) and DMF (250 mL) under N2 atmosphere at room temperature. To this at room temperature and under stirring was added diethyl 2-chloroethoxymethyl phosphonate 2 (74.85 g, 0.325 mol, 1.1 equiv.) in a drop-wise manner. The reaction was stirred at 40-50°C for 0.5 to 1.5 hours, heated to 60-70°C and stirred for 0.5-1.5 hours, and then stirred at 75 to 85°C for 18-24 h. After bringing the reaction temperature to 20-30°C, the reaction mixture was filtered and the resulting cake was washed with DMF (100 mL x 2). The combined filtrate was concentrated to a half volume below 70°C, diluted with n-heptane (250 mL) and again concentrated to a half volume below 75°C. The resulting solution was poured into DCM (1 L), stirred at 20 to 30°C for 20-40 min., then aqueous 10% Na2SO4 solution (~100 mL) was added. The resulting bi-phasic solution was stirred for 20-40 minutes then filtered through diatomite and the wet cake was washed with DCM (~100 mL). From the filtrate, the aqueous phase was separated and the organic phase was again washed with aqueous 10% Na2SO4 solution (~100 mL). The combined aqueous phases upon washing (back extraction) with DCM (200-300 mL), the organic phases were combined and concentrated. The resulting crude product 3 was then purified by silica gel column chromatography using DCM to 1% MeOH in DCM. The fractions containing products were combined and the solvent was evaporated below 40°C. The solid product 3 was treated with the repeated dilution with MTBE and concentration (up to 1/3rd volume). The resulting slurry was then diluted with MTBE (400-500 mL) and agitated at 40-50°C for 4-6 h and at 15-25°C for 8-15 h. The suspension was filtered and washed with MTBE and dried at 35-40°C for 15-20 h to afford the desired product, diethyl- ((2-(2-amino-6-chloro-9H-purin-9-yl)-ethoxy)-methyl)-phosphonate 3 in 43.4% (48.66 g) isolated yield with 91.8 % purity by HPLC.1H NMR (400 MHz, DMSO-d6), δppm: 8.08 (s, 1H), 6.91 (s, 2H), 4.24 (d, 2H, J= 8 Hz), 3.92 (m, 4H), 3.86 (q, 4H, J= 8Hz), 1.14 (t, 6H, J= 8 Hz). LCMS (m/z): 364.2 (MH+) and 366.2 (MH+).
Attorney Docket No.01394-0006-01PCT Step 2: Preparation of ((2-(2-amino-6-chloro-9H-purin-9-yl)-ethoxy)-methyl)- phosphonic acid (4)
was charged with diethyl-((2-(2-amino-6-chloro-9H-purin-9-yl)-ethoxy)-methyl)-phosphonate 3 (100 g, 0.275 mol) followed by 2,6-lutidine (147.33 g, 1.375 mol, 5 equiv.) and the temperature was adjusted to 0-5°C. To this was added TMSBr (167.47 g, 1.102 mol, 4.0 equiv.) in a drop-wise manner and stirred further for 0.5-1 h at 0-5°C and 15-20 h at 20-25°C. After adjusting the reaction temperature at 0-5°C, a drop-wise addition of 1144 g aqueous 1N NaOH was performed. After maintaining the temperature at 20-30°C for 1-2 h, the aqueous alkaline layer was separated and repeatedly washed with MTBE. An aqueous solution was acidified with a drop-wise addition of aqueous 2N HCl to pH= 6-7 at 15-25°C and charged with MeOH (10 Vol.). This resulting methanolic solution was further acidified with a drop- wise addition of aqueous 2N HCl to pH= 3-4 at 35-45°C After addition of seeds of product 4, the methanolic acidic solution was stirred at 35-45°C for 3-5 h and acidified furthermore using a drop-wise addition of aqueous 2N HCl to pH= 1.5-2.5 and stirred for 11-20 h at 15- 20°C. The resulting solid was isolated by filtration, washing with MeOH (2 x 100 mL) and drying at 45-55°C for 20-30 h to yield the desired product, ((2-(2-amino-6-chloro-9H-purin- 9-yl)-ethoxy)-methyl)-phosphonic acid 4 in 96.5 % (84.4 g) isolated yield with 99.8 % purity by HPLC.1H NMR (400 MHz, DMSO-d6), δ ppm: 8.1 (s, 1H), 6.92 (bs, 2H), 4.5-5.5 (bs, 2H), 4.22 (dd= t, 2H, J= 8 Hz), 3.84 (t, 2H, J= 8 Hz), 3.58 (t, 2H, J= 8 Hz). LCMS (m/z): 308 (MH+) and 310 (MH+). Step 3: Preparation of ((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)- phosphonic acid (5)
Attorney Docket No.01394-0006-01PCT [00383] To a flask containing MeOH (350 mL) was charged ((2-(2-amino-6-chloro- 9H-purin-9-yl)-ethoxy)-methyl)-phosphonic acid 4 (50 g, 0.162 mol) at 20-30°C, and stirred for 10-30 min. To this solution was added a 30 wt% NaOMe solution in MeOH (1.62 mol, 10 equiv.) in a drop-wise manner and then stirred at 50-60°C for 15-24 h. The reaction was maintained at 20-30°C for 20-40 min and then filtered. The filtrate was then acidified at 20- 30°C by a drop-wise addition of conc. HCl to adjust pH= 6-7 and concentrated below 40°C to one third of the volume. The temperature of the concentrated solution was raised to 35-45°C and acidified to adjust the pH= 3-4 by means of a drop-wise addition of conc. HCl. The resulting acidic solution was charged with the seeds of product 5, and stirred at 35-45°C for 1.5-2.5 h. At this temperature, the addition of conc. HCl to adjust the pH= 2-3 was done in a drop-wise rate, stirred for 3-5 h, cooled to -3°C to 3°C range and stirred for 8-15 h. The resulting solid was filtered, washed with MeOH (~100 mL) and n-heptane (~100 mL). The resulting cake was dried at 50-60°C for 16-24 h under vacuum to afford the desired product, ((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-phosphonic acid 5 in 89.3 % (48.22 g) isolated yield with 99.5 % purity by HPLC.1H NMR (400 MHz, DMSO-d6), δ, ppm: 7.88 (s, 1H), 6.47 (bs, 4H), 4.18 (t, 2H, J= 8 Hz), 3.96 (t, 2H, J= 8 Hz), 3.60 (d, 2H, J= 12 Hz). LCMS (m/z): 304.20 (MH+). Step 4a: Preparation of a mixture of (R,S)- and (S,S)-ethyl-2-((((2-(2-amino-6-methoxy- 9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate (8)
[00384] To a solution of ((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)- phosphonic acid 5 (40 g, 0.132 mol, 1 equiv.) in DCM (560 mL) at 20-30°C under stirring was charged (S)-ethyl 2-aminopropionate hydrogen chloride salt 6 (20.19 g, 0.132 mol, 1 equiv.), benzyl alcohol 7 (71.28 g, 0.66 mol, 5 equiv.) and TEA (159.98 g, 1.58 mol, 12 equiv.), and the solution was stirred for 10-30 min. To this was added a solution prepared from Ph3P (207.5 g, 0.792 mol, 6 equiv.) and 2,2’-dithiopyridine (Aldrithiol-2) (174.24 g,
Attorney Docket No.01394-0006-01PCT 0.792 mol, 6 equiv.) in DCM (320 mL) at 20-30°C over 60 min. The resulting reaction mixture was stirred at 35-45°C for 15-20 h and concentrated to remove 3/4th of solvent under vacuum below 40°C. To the resulting residue were added MeOH (~120 mL), distilled water (~400 mL), toluene (~400 mL) and n-heptane (~400 mL) and stirred at 20-30°C for 0.5-1 h. After allowing the reaction mixture to stand for 0.5 to 1 h at 20-30°C, the organic phase was separated and the aqueous phase was extracted few more times with a mixture of toluene (~400 mL) and n-heptane (~400 ml) to remove maximum amount of remaining reagents and by-products. The remaining aqueous phase was then extracted with DCM (2 x 400 mL) and upon concentration of DCM under vacuum below 40°C, the crude product was purified by silica gel column chromatography with DCM to 2% MeOH in DCM as a mobile phase. The eluting fractions containing product were combined and solvent was removed under vacuum below 40°C to give the desired product as a mixture of diastereoisomers namely, (R, S) and (S, S); (+) (2S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)- (benzyloxy)-phosphoryl)-amino)-propionate 8 (Compound I) in 45.8 % (29.74 g) isolated yield with 98.8 % purity by HPLC.1H NMR (400 MHz, DMSO-d6), δ, ppm: 7.85 (s, 1H), 7.34 (m, 5H), 6.44 (s, 2H), 5.36 (m, 1H), 4.90 (m, 2H), 4.17 (m, 2H), 4.07 (m, 2H), 3.95 (s, 3H), 3.82 (m, 5H), 1.18-1.24 (m, 6H). LCMS (m/z): 493.3 (MH+). Step 4b: Preparation of a mixture of (R,R)- and (S,R)-ethyl-2-((((2-(2-amino-6-methoxy- 9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate
[00385] To synthesize the (R,R) and (S,R) mixture, the procedure of Step 4a can be performed substituting D-alanine ethyl ester ((R)-ethyl 2-aminopropionate hydrogen chloride salt) for the L-alanine ethyl ester ((S)-ethyl 2-aminopropionate hydrogen chloride salt).
Attorney Docket No.01394-0006-01PCT Step 4c: Preparation of ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate
(ethyl 2-aminopropionate hydrogen chloride salt) for the L-alanine ethyl ester ((S)-ethyl 2- aminopropionate hydrogen chloride salt). Example 4: Preparation of (+)-Compound I Monofumarate ((+)-(2S)-Ethyl-2-((((2-(2- amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)- propionate monofumarate) (9)
ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate (Compound I) 8 (29.72 g, 0.06 mol, 1 equiv.) in IPA was added a solution of fumaric acid (7.66 mol, 1.1 equiv.) in IPA through a filter at 45-55°C and stirring was continued for 1-2 h. The seeds of compound 9 were added to the reaction mixture and stirring was continued for 1-2 h at 45-55°C. After allowing the reaction mixture to settle at 20-30°C for 4-6 h, a drop-wise addition of n-heptane (~300 mL) was performed and stirring was continued for another 8-15 h at 20-30°C and 0- 5°C for 8-15 h. The solid observed was filtered and the wet cake was washed with a mixture of IPA/n-heptane (1/3, v/v, ~50-60 mL). The solid cake was dried at 35-45°C for 16-24 h under vacuum to afford the desired product, (+)-(2S)-ethyl-2-((((2-(2-amino-6-methoxy-9H- purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate 9 (Compound I monofumarate) in 87.9 % (32.74 g) isolated yield with 99.1 % purity by HPLC. 1H NMR (DMSO-d6), δ, ppm: 1.14 (t, 3H, J= 7.2 Hz), 1.22 (d, 3H, J= 7.2 Hz), 3.82 (m, 2H; dd, 1H, J= 4.0 Hz; bs, 2H), 3.95 (s, 3H), 4.06 (m, 2H), 4.17 (m, 2H), 4.87 (m, 2H), 5.38 (q,
Attorney Docket No.01394-0006-01PCT 1H, J= 4 Hz), 6.44 (s, 2H), 6.64 (s, 2H), 7.33 (m, 5H), 7.82 (s, 1H), 13.18 (bs, 2H). LCMS (m/z): 493.20 (MH+). Example 5: Chiral Separation of (R,S)- and (S,S)-Isomers of Compound I and preparation of their monofumarate salts, Compound II and Compound III
Step 1a: Chiral Separation of (R,S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)- ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate and (S,S)-ethyl-2-((((2-(2- amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)- propionate [00388] Compound I (a diastereoisomeric mixture of Isomers I and II) (22.50 g) was subjected to a chiral chromatography separation under SFC separation conditions as shown below to separate and obtain the 11.7 g of (R,S)-Isomer I (10) with 98.6% purity by HPLC and 9.1 g of (S,S)-Isomer II (11) with 95.6% purity by HPLC. SFC Conditions: Column: ChiralPak AD, 250×30mm I.D., 10µm; Mobile phases: A: CO2 and B: Ethanol (0.1% NH3H2O); Gradient: B 45% isocratic; Flow rate: 200 mL /min;
Attorney Docket No.01394-0006-01PCT Wavelength: 310 nm; Cycle time: ~6 min; Back pressure: 100 bar; Injection amount: ~1g. [00389] Characterization of (R,S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9- yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate (Isomer I) as a free base: Purity by HPLC: 98.6%; 1H NMR (DMSO-d6), δ, ppm: 7.82 (s, 1H), 7.30 (m, 5H), 6.38 (s, 2H), 5.30 (t, 1H), 4.83 (d, 2H), 4.18 (t, 2H), 4.05 (m, 2H), 3.95 (s, 3H), 3.84 (m, 2H), 3.60 (m, 5H), 1.20 (d, 3H), 1.15 (t, 3H); LCMS (m/z): 493 (MH+). [00390] Characterization of (S,S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9- yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate (Isomer II) as a free base: Purity by HPLC: 95.6%; 1H NMR (DMSO-d6): δppm 7.82 (s, 1H), 7.35 (m, 5H), 6.45 (s, 2H), 5.30 (t, 1H), 4.80 (d, 2H), 4.18 (t, 2H), 4.05 (m, 2H), 3.95 (s, 3H), 3.80 (m, 3H), 3.70 (m, 2H), 1.20 (d, 3H), 1.15 (t, 3H); LCMS (m/z): 493 (MH+). [00391] In certain nonlimiting embodiments, the stereoisomers are separated using HPLC or SFC with achiral or chiral stationary phases. Non limiting examples of chiral stationary phases which may be used include Chiralpak AD, Chiralpak AS, Chiralcel OG, and Chiralcel OJ. [00392] In alternative non limiting embodiments, the individual isomers can be synthesized asymmetrically. For nonlimiting examples of asymmetric synthesis of phosphonamidates see Numan, A et al. “Asymmetric Synthesis of Stereogenic Phosphorus P(V) Centers Using Chiral Nucleophilic Catalysis”, Molecules 2021, 26, 3661 and Ambrosi, A. et al. “Synthesis of Rovafovir Etalafenamide (Part III): Evolution of the Synthetic Process to the Phosphonamidate Fragment” 2021, Org. Process Res. Dev.25, 5, 1247-1262.
Attorney Docket No.01394-0006-01PCT Step 1b: Chiral Separation of (R,S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)- ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate and (S,S)-ethyl-2-((((2-(2- amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)- propionate OCH3
ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate and (S,R)-ethyl-2-((((2-(2- amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)- propionate (synthesis described in Step 4b of Example 3) can be performed using same techniques for the (R,S) and (S,S) mixture as described above.
Attorney Docket No.01394-0006-01PCT Step 1c: Chiral Separation of (RP)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)- ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate and (SP)-ethyl-2-((((2-(2- amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)- propionate
[00394] Separation of (RP)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate and (SP)-ethyl-2-((((2-(2-amino-6- methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate (synthesis described in Step 4c of Example 3) can be performed using same techniques for the (R,S) and (S,S) mixture as described above. Step 2a: Preparation of (R,S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate (Compound II)
- - ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate 10 (3 g, 6 mmol, 1 equiv.) in IPA was added a solution of fumaric acid ( 0.765 g, 6.6 mmol, 1.1 equiv.) in IPA through a filter at 45-55°C and stirring was continued for 1-2. The seeds of compound 12 were added to
Attorney Docket No.01394-0006-01PCT the reaction mixture and stirring was continued for 1-2 h at 45-55°C. After allowing the reaction mixture to settle at 20-30°C for 4-6 h, a drop-wise addition of n-heptane (~30 mL) was performed and stirring was continued for another 8-15h at 20-30°C and 0-5°C for 8-15h. The solid observed was filtered and the wet cake was washed with a mixture of IPA/n- heptane (1/3, v/v, ~5 mL). The solid cake was dried at 35-45°C for 16-24 h under vacuum to afford the desired product, (R,S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate 12 (Isomer I monofumarate or Compound II) in 85 % (3.1 g) isolated yield with 98.6 % purity by HPLC. 1H NMR (DMSO-d6), δ, ppm:δ7.80 (s, 1H), 7.35 (m, 5H), 6.63 (s, 2H), 6.40 (s, 2H), 5.53 (t, 1H), 4.84 (d, 2H), 4.15 (t, 2H), 4.00 (m, 2H), 3.92 (s, 3H), 3.80 (m, 3H), 3.75 (m, 2H), 1.20 (d, 3H), 1.13 (t, 3H); Base (10): Fumaric acid ratio: 1: 1.00 (by 1H NMR). Step 2b: Preparation of (S,S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate (13), also referred as Compound III
[00396] To a solution of (R,S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)- ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)-propionate 11 (3 g, 6 mmol, 1 equiv.) in IPA was added a solution of fumaric acid ( 0.765 g, 6.6 mmol, 1.1 equiv.) in IPA through a filter at 45-55°C and stirring was continued for 1-2. The seeds of compound 13 were added to the reaction mixture and stirring was continued for 1-2 h at 45-55°C. After allowing the reaction mixture to settle at 20-30°C for 4-6 h, a drop-wise addition of n-heptane (~30 mL) was performed and stirring was continued for another 8-15h at 20-30°C and 0-5°C for 8-15h. The solid observed was filtered and the wet cake was washed with a mixture of IPA/n- heptane (1/3, v/v, ~5 mL). The solid cake was dried at 35-45°C for 16-24 h under vacuum to afford the desired product, (S,S)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate 13 (Isomer II monofumarate or Compound III) in 80 % (2.9 g) isolated yield with 95.6 % purity by HPLC.
Attorney Docket No.01394-0006-01PCT 1H NMR (DMSO-d6), δ, ppm: δ7.82 (s, 1H), 7.35 (m, 5H), 6.62 (s, 2H), 6.35 (s, 2H), 5.30 (t, 1H), 4.90 (d, 2H), 4.15 (t, 2H), 4.05 (m, 2H), 3.95 (s, 3H), 3.80 (m, 3H), 3.70 (m, 2H), 1.20 (d, 3H), 1.15 (t, 3H); Base (11): Fumaric acid ratio: 1: 1.2 (by 1H NMR). Step 2c: Preparation of (R,R)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate and (S,R)-ethyl-2- ((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)- amino)-propionate monofumarate
- - - methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate and (S,R)-ethyl-2-((((2- (2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)- propionate monofumarate can be carried out as in Steps 2b and 2c for the (R,S) and (S,S) stereosiomers, substituting the starting materials in Steps 2b and 2c with the product of the chiral separation in Step 1b.
Attorney Docket No.01394-0006-01PCT Step 2d: Preparation of (RP)-ethyl-2-((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate and (SP)-ethyl-2- ((((2-(2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)- amino)-propionate monofumarate
- methyl)-(benzyloxy)-phosphoryl)-amino)-propionate monofumarate and (SP)-ethyl-2-((((2- (2-amino-6-methoxy-9H-purin-9-yl)-ethoxy)-methyl)-(benzyloxy)-phosphoryl)-amino)- propionate monofumarate can be carried out as in Steps 2b and 2c for the (R,S) and (S,S) stereosiomers, substituting the starting materials in Steps 2b and 2c with the product of the chiral separation in Step 1c. Example 6. Nonlimiting examples of preparation of semisolid formulations [00399] A topical cream formulation can be prepared, for example, by emulsifying an oil phase and an aqueous phase along with an active pharmaceutical ingredient. In a non- limiting embodiment, the oil phase of the cream was prepared by mixing light mineral oil, propylparaben and Tefose® 63. Next, the aqueous phase of the cream was prepared by mixing water, EDTA, methylparaben, and Carbopol®974P. The oil and water phases were then emulsified. To the emulsified mixture was added the active pharmaceutical ingredient and propylene glycol. The mixture was pH adjusted and then filled into tubes. [00400] A topical gel formulation can be prepared, for example, by mixing an aqueous gel carrier with an active pharmaceutical ingredient or salt thereof, for example, Compound I, Compound I monofumarate, Compound II, or Compound III. In a non-limiting embodiment, the aqueous phase of the topical gel was prepared by mixing water, EDTA, methylparaben (or sorbic acid) and Carbopol®974P. The active pharmaceutical ingredient or salt thereof and
Attorney Docket No.01394-0006-01PCT propylene glycol was added to this solution, mixed and pH adjusted, then filled into tubes. [00401] In certain nonlimiting embodiments, from about 0.001% w/w to about 10% w/w active pharmaceutical ingredient or salt thereof is added to the semisolid formulation. For example, from about 0.0025% w/w to about 2.5% w/w, such as 0.003%, 0.01%, 0.03%, 0.1%, 0.3% or 1%. Example 7. Preparation of vaginal tablets [00402] A nonlimiting example of the preparation of vaginal tablets of Compound I monofumarate is provided below (see FIG.5 for a flow diagram). Two or more of the excipients are combined, blended, and screened to make the excipient blend. Then the monofumarate salt of the active pharmaceutical ingredient (such as Compound I monofumarate) is screened and added to a portion of the excipient blend. The resulting mixture is then blended and then more excipient blend is added. The mixture is thus gradually diluted with the excipient blend, with thorough mixing after each addition of excipient blend. Once the Excipient blend has been used up, the magnesium stearate is added and the mixture blended once more. The mixture is then compressed into tablets and packaged. Table 2. Batch Formula for vaginal tablets, 0.3 mg free base dose for batch size of 1.0 Kg Ingredient % w/w Qty/batch (g)
Compound I monofumarate vaginal tablets, 0.3 mg A non-limiting example of a process to prepare the vaginal tablet of Compound I monofumarate is provided below. Dispensing
Attorney Docket No.01394-0006-01PCT 1. Weigh the materials as per the batch manufacturing formula and dispense in separate poly bags. Screening 1. Sieve all excipients through a screen. Blending of Active and Screening 1. Blend the screened excipients: microcrystalline cellulose and mannitol in a diffusion blender. 2. Take 49.5 grams of the excipients blend and add 2.12 grams of Compound I monofumarate 3. Blend the Compound I monofumarate and the excipients, and screen to remove chunks 4. To this blend add an additional 148.5 grams of the excipients blend 5. Blend the Compound I monofumarate blend and the excipients and screen to remove chunks 6. To this blend add an additional 247.5 grams of the excipients blend 7. Blend the Compound I monofumarate blend and the excipients and screen to remove chunks 8. To this blend add the remaining 495 grams of the excipients blend 9. Blend the Compound I monofumarate blend and the excipients and screen to remove chunks Final Blending 10. Add magnesium stearate to the diffusion blender and mix the contents. 11. Discharge and reconcile blend. Compression 1. Compress the blend on a rotary tablet press using appropriate tooling (punches and die), to target weight. Check friability and disintegration at the beginning of the compression run, and check periodically for individual tablet weights, thickness and hardness. Packaging 1. Package bulk tablets into double lined re-closable clear PE bags with desiccants between the bags and further into an aluminum foil pouch with desiccant and heat sealed. Illustrative excipients for a vaginal tablet formulation
Attorney Docket No.01394-0006-01PCT [00403] Tablet formulations are optionally selected to display the properties of mucoadhesion and substantivity and include excipients that have solubilizing, erosion- generating (for disintegration), porosity (for water uptake) and viscosity enhancing (to keep the drug at the target site) properties. Examples of excipients that will cause rapid disintegration to cover the cervix or vaginal areas include, but are not limited to mannitol, microcrystalline cellulose, lactose, sucrose, calcium phosphate, sodium phosphate, sodium bicarbonate, citric acid, maleic acid, adipic acid or fumaric acid. Examples of excipients that can enhance disintegration and coverage of the affected area include but are not limited to sodium starch glycollate, pregelatinized starch, crospovidone and croscarmellose sodium. Mucoadhesive excipients that are useful in the present disclosure include but are not limited to microcrystalline cellulose, polycarbophil, hydroxymethyl cellulose, hypromellose, hydroxypropyl cellulose, and PVP. [00404] The table below lists non-limiting examples of combinations of excipients which have the desired properties for a tablet formulation. A tablet formulation may for example comprise the active pharmaceutical ingredient, microcrystalline cellulose and may contain mannitol. In certain non-limiting embodiments, the tablet formulation comprises one or more excipients selected from the rapid disintegrant category (left column of Table 3). In certain non-limiting embodiments, the tablet formulation comprises one or more excipients selected from the disintegration enhancement category (middle column of Table 3). In certain non-limiting embodiments, the tablet formulation comprises one or more excipients selected from the mucoadhesive excipient category (right column of Table 3). Table 3. Excipients for Tablets Rapidly disintegrating To Enhance Mucoadhesive polymers
Attorney Docket No.01394-0006-01PCT Sucrose (0 to 70%) Pregelatinised starch (0 to Polyethylene Oxide (0 to 20%) 50%) e o
Example 8. Illustrative excipients for a reconstitution powder or dry powder formulation [00405] A reconstitution powder or dry powder formulation may improve the shelf stability of a pharmaceutical agent or formulation. In certain nonlimiting embodiments, the dry powder formulation may be mixed with saline, propylene glycol or other aqueous carrier shortly before it is administered, minimizing the time for degradation. In certain nonlimiting embodiments, the dry powder formulation is mixed with an oil, cream, or other nonaqueous carrier shortly before it is administered. [00406] In certain embodiments, the reconstitution powder or dry powder formulation rapidly covers the infected or diseased tissue in the cervix, vulva or vagina. Excipients which enhance the rapid coverage of the cervix, vulva or vagina, include but are not limited to mannitol, lactose, sucrose, calcium phosphate, and microcrystalline cellulose. In certain embodiments, the excipient for rapid coverage of the cervix, vulva or vagina is mannitol. [00407] In certain embodiments, the reconstitution powder or dry powder formulation has good coverage of the cervix, vulva or vagina. Nonlimiting examples of excipients which enhance the coverage of the cervix, vulva or vagina, include sodium starch glycollate, pregelatinized starch, crospovidone, and croscarmellose sodium.
Attorney Docket No.01394-0006-01PCT [00408] In certain embodiments, the reconstitution powder or dry powder formulation contains mucoadhesive properties once it has been reconstituted. This prevents smearing of the dosage form or otherwise exposing healthy tissues to the active pharmaceutical ingredient. Excipients which improve the mucoadhesive properties of the reconstituted powder or dry powder formulation include but are not limited to xanthan gum, polycarbophil, polyethylene oxide, hydroxyethylmethyl cellulose, hydroxyethyl cellulose, Hypromellose, hydroxypropyl cellulose, PVP, and microcrystalline cellulose. In certain embodiments, the excipient which improves mucoadhesion is xanthan gum. [00409] The table below lists combinations of excipients which have the desired properties for a reconstitution powder or dry powder formulation. A dry powder or reconstitution powder formulation comprises the active pharmaceutical ingredient and may contain mannitol and/or xanthan gum. In certain non-limiting embodiments, the dry powder or reconstitution powder formulation comprises one or more excipients selected from the rapid coverage category (left column of Table 4). In certain non-limiting embodiments, the dry powder or reconstitution powder formulation comprises one or more excipients selected from the coverage enhancement category (middle column of Table 4). In certain non-limiting embodiments, the dry powder or reconstitution powder formulation comprises one or more excipients selected from the mucoadhesive excipient category (right column of Table 4). Table 4. Excipients for Reconstitution Powders of Dry Powder Dosage Forms Rapidly covering To Enhance coverage Mucoadhesive polymers t r t ti with f t r t ti
Attorney Docket No.01394-0006-01PCT Microcrystalline Croscarmellose Na (0 Hydroxyethylcellulose cellulose (0 to to 20%) (0 to 50%) e
Example 9. Illustrative excipients for a semisolid formulation [00410] Semisolid formulations are selected to display the properties of mucoadhesion and assist in the drug penetration into the tissue. Semisolid formulations may include excipients that have solubilizing, lipophilic (to assist in solubilizing lipophilic compound), penetration enhancing (for higher activity) and mucoadhesive (to keep the drug at the target site) properties. [00411] In certain embodiments, the semisolid formulation is mucoadhesive. Excipients which contribute to the mucoadhesive properties include but are not limited to carbomer, polyethylene glycol, crospovidone, polycarbophil, hypromellose, and hyroxyethyl cellulose. [00412] In certain embodiments, the semisolid formulation enhances the penetration and/or solubility of the active pharmaceutical ingredient. Excipients which enhance the penetration and/or solubility of the active pharmaceutical ingredient include but are not limited to polyoxyl 6 stearate type I, ethylene glycol stearate, polyoxyl 32 stearate type I, and propylene glycol. [00413] The table below lists combinations of excipients which have the desired properties for a semisolid formulation. A semisolid formulation comprises the active pharmaceutical ingredient and one or more excipients from each column of Table 5. In certain non-limiting embodiments, the semisolid formulation comprises one or more excipients selected from the mucoadhesive polymer category (left column of Table 5). In certain non-limiting embodiments, the tablet formulation comprises one or more excipients selected from the solubility and penetration enhancers category (second column of Table 5).
Attorney Docket No.01394-0006-01PCT In certain non-limiting embodiments, the semisolid formulation comprises one or more excipients selected from the lipophilic solubilizer category (third column of Table 5). In certain non-limiting embodiments, the semisolid formulation comprises one or more excipients selected from the penetration enhancer category (right column of Table 5). Table 5. Excipients for semisolid dosage forms Mucoadhesive Solubility and penetration Lipophilic Penetration polymer enhancers solubilizers Enhancer 0 o
Attorney Docket No.01394-0006-01PCT Polyoxyl 40 hydrogenated Stearic acid (0 to castor oil (0 to 25%) 20%) ol
Example 10. Illustrative excipients for a semisolid formulation [00414] Pessary and film forming formulations are selected to be solid at room temperature but soften to release the active pharmaceutical ingredient at body temperature. This allows for easy handling and storage of the formulation as well as achieving desired tissue coverage at the cervix. In a non-limiting embodiment of a film forming formulation, one or more excipients from the left column of Table 6 provide the desired properties. In a non-limiting embodiment of a pessary formulation, one or more excipients from the right column of Table 6 provide the desired properties. Table 6. Excipients for films and pessaries Films (Film Formers) Pessaries (Vaginal Suppositories)
Attorney Docket No.01394-0006-01PCT Example 11. Illustrative vaginal tablet formulations [00415] In certain non-limiting embodiments, the formulation for a vaginal tablet dosage form comprises the ingredients in Table 7. In certain non-limiting embodiments, the formulation for a tablet dosage form comprises the ingredients in Table 7. An illustrative process for combining these ingredients into a tablet dosage form can be found in Example 7. Table 7. Example Tablet Formulation Material Amount in tablet d such that
the amount in the tablet is equivalent to 0.05 mg to 5 mg of the free base. Table 8. Example Tablet Formulation Material Amount in tablet
Attorney Docket No.01394-0006-01PCT Magnesium stearate 1.75 mg d such that
Example 12. Illustrative semisolid formulations [00416] In certain non-limiting embodiments, the formulation for a gel semisolid dosage form comprises the ingredients in Table 9. In certain non-limiting embodiments, the formulation for a cream semisolid dosage form comprises the ingredients in Table 10. An illustrative process for combining these ingredients into a cream or gel semisolid dosage form can be found in Example 6 above. Table 9. Example semisolid formulation (gel) Material Amount per Gram * d such that
the amount in the tablet is equivalent to 0.025 mg to 25 mg of the free base. Table 10. Example semisolid formulation (cream) Material Amount per Gram
Attorney Docket No.01394-0006-01PCT Active pharmaceutical ingredient 0.025 mg to 25 mg (for example 0.03 mg to 10 mg, such as 0.03, 0.1 mg, 0.3 mg, 1.0 mg, 3 ed such
. . Example 13. Illustrative pessary formulation [00417] In certain non-limiting embodiments, the pessary formulation comprises the ingredients listed in Table 11 or Table 12. The pessary dosage form can be prepared, for example, by mixing the active pharmaceutical ingredient with the excipient. In one non- limiting embodiment, the excipient is heated in a mixing apparatus while stirring until it has softened or melted, then the active pharmaceutical ingredient is added portionwise. The temperature, stirring speed, and rate of addition are controlled to ensure an even distribution of active pharmaceutical ingredient. The mixture is then mixed until homogeneous and placed into pessary or suppository molds to solidify.
Attorney Docket No.01394-0006-01PCT Table 11. Example formulation for a pessary Pessary Excipients Amount per Gram * d such
Table 12. Example formulation for a pessary Pessary Excipients Amount per Gram * d such
q . g g . Example 14. In-vitro cytotoxicity testing Compounds: [00418] Three compounds (Compound I, Compound II and Compound III) were solubilized at 40 mM in DMSO and stored at -20°C. The test compounds were evaluated using a high test concentration of 50 µM. Serial half-logarithmic dilutions were performed for the in vitro cytotoxicity assays. Tamoxifen citrate was purchased from Sigma-Aldrich (St. Louis, MO). Tamoxifen citrate was solubilized in DMSO at 40 mM and used as a positive control compound at a high test concentration of 100 µM for the cytotoxicity assays. In Vitro Cytotoxicity Evaluations: [00419] Cells listed in Table 13 were enumerated by Trypan Blue Dye exclusion method and seeded in the interior wells of a 96 well flat bottom microtiter plate at 100 µL/well. Proliferating cells were incubated overnight at 37°C/5% CO2 to allow the cells to adhere to the plates at approximately 70% confluency. Tissue culture medium was
Attorney Docket No.01394-0006-01PCT removed and replaced with 100 µL/well of fresh medium. One-hundred microliters (100 µL) of each compound at six concentrations was transferred to the 96-well plate containing the cells in triplicate. Table 13 lists the cell line, type of cell, source of cell stock, base tissue culture medium supplemented with 10% fetal bovine serum, 2 mM L- glutamine, 100 U/mL penicillin and 100 µg/mL streptomycin, and microtiter plate seeding density. Table 13: Cell Culture for in vitro Cytotoxicity Cell Seeding Cell Line Cell Type Cell Source Culture Density 4
y o ox c y : [00420] Following incubation at 37°C in a 5% CO2 incubator for five days, the test plates were stained with the tetrazolium dye XTT (2,3-bis(2-methoxy-4-nitro-5- sulfopheny1)-5-[(phenylamino)carbonyl]-2H-tetrazolium hydroxide). XTT-tetrazolium was metabolized by the mitochondrial enzymes of metabolically active cells to a soluble formazan product. XTT solution was prepared daily as a stock of 1 mg/ml in RPMl1640. Phenazine methosulfate (PMS) solution was prepared at 0.15 mg/ml in PBS and stored in the dark at -20°C. XTT/PMS stock was prepared immediately before use by adding 40 µL of PMS per ml of XTT solution. Fifty microliters of XTT/PMS were added to each well of the plate and the plate was reincubated for 4 hours at 37°C. Plates were sealed with adhesive plate sealers and shaken gently or inverted several times to mix the soluble formazan product and the plate was read spectrophotometrically at 450/650 nm with a Molecular Devices Vmax plate reader.
Attorney Docket No.01394-0006-01PCT Data Analysis and Evaluation: [00421] Microsoft Excel 2010 was used to analyze and graph the raw data. CCso (50% reduction in cell viability) values are tabulated and provided. Raw data for cytotoxicity with a graphic representation of the data are provided in a printout summarizing the compound effect on cell viability. In Vitro Cytotoxicity Evaluations: [00422] Compounds I, II, and III were evaluated for cytotoxicity to proliferating Hs27, HeLa, C33A and HEK293 cells by measuring cell viability using XTI tetrazolium dye following five days in culture (Table 14). The CC50 values calculated from these assays are summarized in the below tables. [00423] Tamoxifen citrate was evaluated in parallel as a control compound. The CC50 value for tamoxifen citrate in proliferating C33A, HeLa, Hs27, and HEK293 cells was 17.2, 19.9, 21.2 and 21.3 µM, respectively. Compound I and its two isomers were similarly cytotoxic when evaluated in parallel against each of the four cell lines. CC50 values for the three test compounds ranged from approximately 0.1 to 0.28 µM in C33A cells. In HeLa cells, CC50 values for the three test compounds ranged from 15.1 to 18.6 µM. The CC50 values for the three test compounds ranged from approximately 7.62 to 23.2 µM in Hs27 cells. In HEK293 cells, CC50 values for the three test compounds was approximately 0.1 µM. Table 14: In vitro Cytotoxicity Data Compound C33A HeLa Hs27 HEK293 )
Example 15. Ex Vivo permeation and penetration of antiviral drugs across porcine vaginal tissue Preparation of Porcine Vaginal Tissue [00424] Freshly harvested porcine vaginal tissue was procured from local slaughterhouse in an ice box. The vaginal tissue was cut open to expose the mucosal surface
Attorney Docket No.01394-0006-01PCT and tissue was cleaned by gentle flow of PBS pH 7.4. The porcine vaginal tissue was cut into circular portions (approximately 2 cm2) with the help of a telemetric punch. Mounting Porcine vaginal tissue on Franz diffusion cells [00425] The circular portion of tissue was sandwiched between two chambers of a Franz diffusion cell with an active diffusion area of 1 cm2, and the mucosal layer was exposed to the donor chambers. The resistance across porcine vaginal tissue was measured using a wave form generator to ensure the integrity of the tissue segment used for the permeation study. Porcine vaginal tissue with resistance of ≥3 KΩ.cm2 was used for study. The receiver chamber was filled with 8 ml of 5% solutol PBS 7.4 pH, which was stirred at 600 rpm with a 3 mm magnetic stir bar and the temperature was maintained at 37 °C with a circulating water bath. Loading of formulation in the donor chamber [00426] The ~200mg of gel was filled into a tared 1mL syringe and gel was dispensed to donor chamber. The gel was spread on to a mucosal surface with a pre-weighed applicator. After loading and spreading of the gel onto surface of mucosal surface, the weight of 1mL syringe and applicator was noted to determine the exact amount of gel loaded into donor chamber. Permeation and penetration study [00427] Time course (2h, 4h and 8h; Table 15) porcine vaginal permeation studies were performed. After loading gel, 500 μL samples were withdrawn from the receiver compartment at different time intervals and each time an equal volume of fresh receiver media was used to replace withdrawn samples. The sample withdrawn at each time interval were stored immediately in -20 ̊C until analysis. After 2, 4 and 8h, the formulation was removed from the donor chamber with the help of a syringe and cleaned with cotton swab. The tissue was removed and washed gently with wash solution (50% methanol in water) 5 times and alternatively cleaning with cotton swab. Table 15: Time course IVPT study design Study Receptor fluid sampling time points Study Period
Attorney Docket No.01394-0006-01PCT Mincing the Porcine vaginal tissue (active diffusion area) after IVPT [00428] An 8 mm punch of active diffusion area of washed porcine vaginal tissue was removed, weighed and transferred into a tube. This tube was immediately placed into dry ice for ~15 min. After specified time, tissue was removed and placed in precooled dish. The tissue was minced into smaller pieces on a dish with a precooled surgical blade. The minced tissue was transferred to sample tubes and dish was rinsed with 1 ml of 5% solutol in PBS 7.4 pH and transferred to same tissue sample tube. These tubes were stored in -70̊C until analysis. Preparation of receptor fluid for analysis [00429] The samples stored at -20 °C were removed and thawed at room temperature for 30 min. The drug from receptor fluid was centrifuged at 13000 rpm for 5 min and to 200μL of supernatant equal volume of extraction solvent (acetonitrile) was added. These samples were centrifuged at 13000 rpm for 5 min and supernatant was transferred into vials for analysis. Extraction of Drug from Porcine Vaginal tissue [00430] The minced tissue samples stored at -70̊C were removed and thawed at room temperature for ~90 min. The samples were kept for shaking at room temperature for 4 h in BioShaker. After 4 h, samples were centrifuged at 13000 rpm for 5 min. To 100 μL of supernatant, 400 μL of extraction solvent was added and vortexed for 2 min. These samples were centrifuged at 13000 rpm for 5 min and supernatant was transferred into vials for analysis. Example 16. Nonclinical Studies in Non-human Animals [00431] Two in vivo studies evaluated the local toxicity and toxicokinetics of Compound I monofumarate vaginal tablets following repeated intravaginal administration to rabbits for 2 weeks (6 doses, 3x/week) at doses ranging from 0.003 mg to 2.9 mg Compound I per tablet. Rabbits (approximately 3 kg in size) were administered tablets with the same excipient profile as those proposed for use in clinical studies, but smaller in size (50 mg rabbit tablets vs.175 mg clinical tablets). Tablets were administered through a lubricated cannula inserted into the vagina, followed by a small amount of normal saline to aid in tablet disintegration. Rabbits were dosed 6 times over 2 weeks (dose administration on Study Days 1, 3, 5, 8, 10, and 12) and necropsied on Day 15. In general, Compound I monofumarate
Attorney Docket No.01394-0006-01PCT vaginal tablet administration was well tolerated. Findings from the two rabbit studies in the locally exposed tissues of vulva, vagina and cervix are summarized below. Vulva [00432] The most significant findings in both tablet studies were in the vulvar tissues, with dose-dependent erythema and edema observed after about the third dose (i.e., the second week when administered 3x/week) that was dose-strength related. While some transient increases in erythema and edema were seen across all groups (including placebo control tablets and sham controls in which the cannula was inserted into the vagina) which was attributed to the dosing procedure, in both studies there were individual erythema and edema scores of ≥ 3 (scored out of 4) seen in the 2.9 mg Compound I animals (high dose group) starting at Day 8. [00433] In the first of the two studies, there were a few instances of more substantial vulvar irritation seen at necropsy in the 2.9 mg dose group (abrasion, scab and/or thickened vulva) and microscopic findings of moderate vulvar ulceration and inflammation; these vulvar findings were not seen in the second study where animals were also dosed at 2.9 mg and may be due in part to more familiarity with the dosing procedure by the technical staff. The vulvar findings are presumed to be related to direct contact with Compound I and/or its salt, possibly due to leakage given the position of the urethra in the vagina of rabbits, resulting in increased vulvar exposure to Compound I and/or its salt during urination. These results support the need for the use of a retaining device in combination with the therapeutic administration. Vagina and Cervix [00434] Many animals, including sham and placebo controls, had microscopic infiltrates of a mixed cell population noted within the lamina propria of sections of vulva and vagina after repeated administration of the Compound I monofumarate vaginal tablets. Within the vagina, microscopic findings in the two studies included minimal to mild epithelial degeneration, mixed cell inflammation and luminal exudate in animals administered tablets ≥ 0.03 mg Compound I. In the first study, two cases of minimal arterial degeneration/necrosis were seen at the 2.9 mg dose, which was characterized by disruption of the arterial wall with karyorrhectic (nuclear) debris and/or fibrinous change. In addition, one animal treated at 2.9 mg had moderate ulceration in the middle portion of the vagina.
Attorney Docket No.01394-0006-01PCT [00435] These findings were not observed in the second, larger GLP study. In general, the lower portion of the vagina near the vaginal opening was less affected than the upper, or cervical/cranial, vagina nearer the site of administration. Systemic Exposure [00436] Compound I monofumarate vaginal tablets are formulated for topical administration at the site of action with the goal of producing minimal systemic exposure. In rabbits, dose-dependent increases in systemic exposure (Cmax and AUC) were observed following intravaginal administration of the Compound I monofumarate vaginal tablets at doses of 0.03, 0.29 and 2.9 mg Compound I. No accumulation was observed after repeated (i.e., 3x/week for 2 weeks) administration. The extent of systemic exposure in humans is expected to be lower than observed in rabbits due to the differences in body mass. In addition, physiologic/anatomic differences between rabbits and humans are expected to result in decreased bioavailability in humans compared with rabbits due to a smaller relative surface area and increased thickness of vaginal and cervical epithelial tissue. Toxicity Studies in Rabbits [00437] In addition, three in vivo studies of up to 2-week duration were conducted in rabbits using Compound I monofumarate Vaginal Gel. Those studies were designed to examine both the local and systemic toxicity associated with intravaginal administration of Compound I at dose strengths ranging from 0.01% to 1% (in a 1 mL dose, this correlates to administering 0.1 mg to 10 mg of Compound I). In general, the local findings were similar in nature to those described following administration of the vaginal tablet, although more vulvar irritation was seen, presumably due to the greater extent of direct tissue exposure with gel leakage and urination. One of the 2-week studies included 4-week recovery groups to understand the reversibility of any toxicity findings. By Day 45 (Recovery), there was partial recovery of Compound I-related microscopic findings in the vulva and lower vagina, and complete recovery of the findings seen in the middle and upper vagina. Systemically, the Compound I monofumarate Vaginal Gel was well tolerated, with no significant findings. [00438] Although systemic exposure in patients is intended to be low, the safety evaluation of Compound I included studies with parenteral administration to ensure 100% systemic exposure to the dose, to characterize the potential systemic toxicity. Compound I administered by the IV route in safety pharmacology studies evaluated the potential for
Attorney Docket No.01394-0006-01PCT adverse effects on the respiratory, cardiovascular, and central nervous systems. No adverse effects were observed. [00439] Single-dose IV toxicity studies were also conducted to describe potential systemic toxicity. In rabbits, single doses of 0 (vehicle control), 0.03, 0.3 and 3.0 mg/kg Compound I monofumarate were given intravenously and animals were monitored for 7 or 23 days before sacrifice for necropsy and microscopic examination of tissues. Compound I was well tolerated with no signs of toxicity at any dose and all animals survived to the scheduled termination. There were no changes in chemistry or coagulation parameters. Decreased bone marrow cellularity, likely due to inhibition of DNA polymerase, was observed at the high dose only (3 mg/kg) in animals sacrificed on Day 7. Although there were no cytologic abnormalities observed in bone marrow smears, there was evidence in bone marrow smears of increased myeloid and erythroid precursors. The bone marrow changes completely reversed by Day 23. Hence, the NOAEL was 3 mg/kg. Systemic exposures (AUC) at a dose of 3 mg/kg after IV dosing were more than 20-fold higher than those observed in intravaginal toxicology studies at maximally tolerated dose strengths and are, on that basis, also expected to be much higher than potential human exposure by intravaginal application. Toxicity Studies in Dogs [00440] A similarly designed single-dose IV toxicity study was also completed in female dogs given doses of 0, 0.02, 0.06 and 0.3 mg/kg, again with 7- and 23-day post- treatment observation periods. As in rabbits, Compound I was well tolerated with no clinical signs and all animals survived to their scheduled necropsy. There were no changes in chemistry, hematology, urinalysis or coagulation parameters. The only treatment-related finding was microscopic renal tubular karyocytomegaly at the high dose (0.3 mg/kg) on both Days 7 and 23. Although the change was not considered adverse, it was considered to be related to treatment with Compound I. Karyomegaly is an enlargement of the cell nucleus that has been proposed to be a response to chemical/drug exposure or insult (Hard, 2018). As indicated, the NOAEL for this study was 0.3 mg/kg which is approximately 12-fold greater exposure (by AUC) than the plasma exposure in rabbits dosed intravaginally at the 0.29 mg dose strength. Genotoxicity [00441] A standard battery of genotoxicity studies to assess the potential mutagenicity of Compound I was completed. A bacterial reverse mutation (Ames) study was negative. A
Attorney Docket No.01394-0006-01PCT mammalian chromosomal aberrations study was positive in the presence of metabolic activation at the highest concentration tested; however, there was no evidence of mutagenicity in an in vivo micronucleus assay. These data suggest the risk of genotoxicity following administration of Compound I is low. [00442] Taken together, data from 5 intravaginal safety studies (2 with tablet, 3 with gel) suggest that if adverse effects following administration of Compound I vaginal tablet were to occur, they would be expected to be minor and limited to the site of application. Example 17. Clinical Studies in Healthy Human Volunteers [00443] Compound I monofumarate salt was investigated in a single and multiple- dose, ascending, randomized, placebo-controlled study in healthy human volunteers. The study was conducted in 2 parts. Part A consisted of single ascending dose cohorts administering Compound I monofumarate Vaginal Gel 0.01% or placebo and Compound I monofumarate vaginal tablet up to 0.3 mg or placebo. Part B consisted of 2 multiple ascending dose cohorts administering vaginal tablets with a dose of 0.1 mg or 0.3 mg Compound I or placebo (3 doses over 1-week period). The study completed in June 2023. A total of 35 subjects were enrolled in the study: 19 subjects in Part A (13 received Compound I monofumarate; 6 received placebo) and 16 subjects in Part B (12 received Compound I monofumarate; 4 received placebo). [00444] In Part A and the 0.1 mg dose cohort of Part B, the vaginal tablets were safe and well tolerated. No dose limiting toxicities (DTLs) were reached or serious adverse events (SAEs) reported. All AEs were mild or moderate in severity. [00445] After review of safety data, it was determined that DLT at the 0.3 mg dose in Part B was reached. Although no SAEs were reported in this cohort, 3 out of 6 subjects receiving the Compound I monofumarate vaginal tablets experienced 2 or more moderate genitourinary AEs (i.e. vaginal discomfort, vulvovaginal erythema, vulvovaginal pain, vulvovaginal swelling, urinary tract infection, and bacterial vaginosis), thereby meeting the protocol definition of DLT. As such, the SMC confirmed the maximum tolerated dose (MTD) at 0.1 mg administered 3 times over 1-week period. There was no detectable Compound I in plasma from any subjects in any cohort at any time-points (up to Day 8; LLOQ = 0.1 ng/mL). This study supports the use of a retaining device as an integral part of the therapeutic protocol.
Attorney Docket No.01394-0006-01PCT Example 18. Human Patient Self-Administration [00446] Patients should keep the bottles of vaginal tablets from extreme high temperatures and preferably store the tablets in the refrigerator. Prior to administration, the vaginal tablet is removed from the refrigerator and allowed to come to room temperature for at least 30 minutes. Compound I monofumarate vaginal tablets can be administered with a vaginal applicator, followed by application (immediately or after some period of time, such as 6-8 hours) of a retaining device such as a tampon. In certain embodiments, Compound I monofumarate vaginal tablets can be administered with a diaphragm or menstrual cup. Example 19. Exemplary Instructions For Self-Administration Of Compound I monofumarate vaginal tablets With Vaginal Applicator [00447] Tablet administration is preferably carried out at bedtime to reduce displacing medication that may occur when standing or walking. 1. Place all supplies on a clean flat surface 2. Optionally, gloves for administration, vaginal tablet applicator, water-based lubricant (such as KY Jelly, Surgilube or similar), and bottle of Compound I monofumarate vaginal tablets are assembled. 3. Wash and dry hands, and put on gloves. 4. Remove the applicator from its packaging. 5. Remove the Compound I monofumarate vaginal tablet from the bottle and place it into the end of the applicator. 6. To load the tablet into the applicator, retract the plunger slightly and insert the tablet into the barrel of the applicator until it is securely seated. Illustrations of the tablet placed in the barrel of the applicator are shown in FIG 7A. If loaded properly, the tablet will be firmly in the barrel and will not drop even when the applicator is held with tablet facing down. In this position the tablet can be released by pushing in the plunger. 7. Prior to self-administration, add a small amount (about a teaspoon, which should be enough to cover/coat the tablet and applicator) of water-based lubricant onto the tablet and applicator. The lubricant may be provided in individual packets if available; in that case, open the packet and dispense the entire amount of lubricant onto the Compound I monofumarate vaginal tablet. 8. For insertion, the female can position her body in one of two ways: a. Stand upright (standing with her feet apart and her knees bent). It may also be helpful to raise one leg up on a chair or the bed.
Attorney Docket No.01394-0006-01PCT b. Lie on her back with her knees bent and legs slightly apart. 9. When in a comfortable position, spread the lips at the entrance to the vagina and gently insert the applicator into vagina (as she would for inserting a tampon). Gently advance the applicator until resistance is met. When in position, deliver the Compound I monofumarate tablet onto the cervix by pushing the plunger until it stops. 10. Remove the applicator from the vagina and place in the disposal bag. 11. It is preferred to not engage in any vigorous activities such as jogging, working out, or swimming for 24 hours after applying the vaginal tablet. Example 20. Participant Instructions for Self-Administration of Compound I monofumarate vaginal tablets with Vaginal Retaining Device 1. The diaphragm should be a flexible device that is created to be inserted through the vagina and placed to cover the cervix. The retaining device is used to help prevent leakage from the dissolved Compound I monofumarate vaginal tablet. The device will form a seal against the cervix. The diaphragm with the vaginal tablet is inserted, and then removed approximately 5-10 hours after insertion, and more typically 5-8 hours, for example around or at least 6 hours. 2. Vaginal tablet administration should be preferably done at bedtime to reduce displacing medication that may occur when standing or walking. 3. Remove the diaphragm from its packaging. 4. Prior to self-administration, remove the Compound I monofumarate vaginal tablet from the bottle and place it inside the diaphragm first (FIG.7B) 5. Place a small amount (about a teaspoon or 5 mL) of water-based lubricant into the diaphragm to cover the Compound I monofumarate vaginal tablet. The lubricant may be provided in individual packets if available; in that case, open the packet and dispense the amount of lubricant onto the Compound I monofumarate vaginal tablet (FIG.7C). 6. Hold the diaphragm with one hand by placing the thumb and index finger on the grip dimples along the rim. The arrow should point towards her body. See FIG.7D. 7. Proceed to fold the diaphragm. The vaginal tablet will be located on either the left or right side of the fold (See FIG.7E). 8. Alternatively, if the lubricant and vaginal tablet rise up too close to the rim when folding, and appear to be coming out, the patient can push down on the diaphragm dome while folding to allow for one big fold (See FIG.7F).
Attorney Docket No.01394-0006-01PCT 9. For insertion of the diaphragm the patient can position her body in one of three ways: kneeling down in a sitting position; lying down with legs bent; or standing with one leg elevated. 10. Using her free hand, spread the labia. Push the diaphragm with her other hand into the vagina, and push down gently towards the back, advancing the diaphragm until the cervix rests inside the diaphragm (FIG.7G). 11. Optionally use a finger to verify that the cervix is covered by the diaphragm. If you can feel the cervix through the diaphragm (it should feel like the tip of the nose), then it is positioned correctly (See FIGS.7H, 7I, and 7J). 12. The edge of the removal dome should be located securely and safely behind the upper part. 13. It is preferred not to engage in any vigorous activities such as jogging, working out, or swimming for 24 hours after applying the vaginal tablet. Example 21: Single-Crystal X-Ray Diffraction (SC-XRD) study of Compound II Pattern 1 [00448] Single crystals of Compound II Pattern 1 suitable for SC-XRD study were obtained in a temperature cycling experiment in MeOH. X-ray diffraction data were collected on a D8 Venture diffractometer equipped with a CMOS area detector at 170(2) K using Cu- Kα radiation (λ = 1.5418 Å); X-ray generator power: 50 kV, 1.4 mA; Distance from sample to area detector: 40 mm; Exposure time 150 seconds; Resolution: 0.81. Structure refinement: on F2. Hydrogen site location: mixed. H atoms were treated by a mixture of independent and constrained refinement. X-ray diffraction data and crystal data are presented in Table 16. Table 16. Crystallographic parameters and x-ray diffraction data Chemical Formula C21H30N6O6P·C4H3O4 M l l i ht 4
Attorney Docket No.01394-0006-01PCT Dcalc, g/cm3 1.403 µ/mm-1 0.16 of
[ ] rys a ne orm o ompoun a ern crys a ze n monoc n c sys em, P21 space group with Rint = 5.7% and the final R1 [I>2σ(I)] = 7.4%. The crystalline form did not contain solvent molecule. This crystalline form of Compound II Pattern 1 was found to have a free base to fumaric acid ratio of 1:1 and corresponds to monofumarate salt of Isomer I with Flack parameter (absolute structure parameter) of 0.16(10). As shown in FIG.1A and 1B, protonated free base and fumaric acid anion are linked through a N+(5)-H(5)···O(7) ionic bond in the single crystal structure. Proton transfer was observed from fumaric acid to the N(5)-nitrogen atom of purine. Example 22: Synthesis of Salts of Compound I Compound I Sulfate [00450] Two methods were used to synthesize the sulfate salt of Compound I.
Attorney Docket No.01394-0006-01PCT Method A [00451] To a solution of Compound I (0.049 g, 0.1 mmol) in dry THF (1 mL) at 0- 10°C was added a solution of sulfuric acid (1N in THF) slowly. During the addition of sulfuric acid solution, the pink colored, clear THF solution of Compound I free base was changed to an off-white semisolid. The reaction mixture was brought to room temperature over 20-30 minutes and shaken. After allowing the solid material to settle, the supernatant was decanted carefully. The resulting semi-solids were washed with an additional amount of dry THF (2 x 2 mL), and the resulting solids were dried under high vacuum to yield 0.053 g of Compound I Sulfate Salt-1 as an off-white solid. Method B [00452] To a solution of Compound I (0.024 g, 0.05 mmol) in dry ethyl acetate (EtOAc, 0.5 mL) at 0-10°C was added a solution of sulfuric acid (1N in EtOAc) slowly. During the addition of sulfuric acid solution, the pink colored EtOAc clear solution of Compound I free base was changed to an off-white colored solid. The reaction mixture was brought to room temperature over 20-30 min and shaken well. After allowing the solid material to settle, the supernatant was decanted carefully. The off-white solids were washed with an additional amount of EtOAc (2 x 2 mL). The resulting off-white solids were dried under high vacuum to yield 0.023 g of Compound I Sulfate Salt-2. Compound I Methanesulfonate
used to synthesize the methanesulfonate salt of Compound I. Method A [00454] To a solution of Compound I (0.049 g, 0.1 mmol) in dry EtOAc (1mL) at 0- 10°C was added neat methanesulfonic acid (MSA; MW=96.11; d= 1.47; 0.007 mL; 0.11 mmol) dropwise. During the addition of MSA solution, the pink colored EtOAc clear solution of Compound I free base was changed in to an off-white semi-solid (glue like). The heterogeneous mixture was brought to room temperature in 20-30 min and shaken well. After allowing semi-solid material to settle down, the supernatant was decanted carefully. The resulting semi-solid (glue like) was washed with methyl tert-butyl ether (MTBE; 2 x 2 mL) and was dried under high vacuum to yield 0.054 g of Compound I Methanesulfonate Salt-1.
Attorney Docket No.01394-0006-01PCT Method B [00455] To a solution of Compound I (0.049 g, 0.1 mmol) in dry IPA (1mL) at 0-10°C was added methanesulfonic acid solution (1N in THF; 0.11 mmol; 0.110 mL) dropwise. During the addition of MSA solution, the pink colored EtOAc clear solution of Compound I free base was changed in to an off-white solid. The heterogeneous mixture was brought to room temperature in 20-30 min and shaken well. After allowing solid material to settle down, the supernatant was decanted carefully. The resulting solid was washed with methyl tert-butyl ether (MTBE; 2 x 2 mL) and was dried under high vacuum to yield 0.049 g of Compound I Methanesulfonate Salt-2. Compound I Hydrochloride Salt Compound I (MW=492; 0.049 g, 0.1 mmol) in dry EtOAc
solution (4N in dioxane; 0.11mmole; 0.027 mL) dropwise. During the addition of HCl solution, the pink colored EtOAc clear solution of Compound I free base was changed in to an off-white solid. This heterogeneous mixture was brought to room temperature in 20-30 min and shaken well. After allowing solid material to settle down, the supernatant was decanted carefully. The resulting solid was washed with methyl tert- butyl ether (MTBE; 2 x 2 mL) and was dried under high vacuum to yield 0.045 g of Compound I HCl Salt. Compound I monofumarate
of Compound I (0.035 g, 0.071 mmol) in dry iso-propanol (0.1 mL) at 0-10°C was added a fumaric acid (MW= 116; 12.3 mg; 0.106 mmol; 1.5 equivalents). The reaction mixture was brought to room temperature, then heated at 60°C for 30 min and stirred at room temperature for 12 h. The reaction mixture was filtered, and the filtrate was concentrated under a reduced pressure at 40°C. The mixture was diluted with MTBE (2 mL), shaken well and the MTBE was decanted carefully. The colorless solid was washed with an additional amount of MTBE (2 mL) and dried under high vacuum to yield 0.022 g of Compound I sesquifumarate salt. Compound I monofumarate salt can be synthesized by washing Compound I sesquifumarate salt with additional MTBE and drying under high vacuum.
Attorney Docket No.01394-0006-01PCT Compound I Benzenesulfonic Acid Salt I (0.057 g, 0.115 mmol) in dry EtOAc (1.0 mL) at
acid (BsOH, MW=158; 0.020 g; 0.127 mmol in 0.2 mL of EtOAc) dropwise. During the addition of BsOH solution, the pink colored EtOAc clear solution of Compound I free base precipitated as a colorless solid. The reaction mixture was brought to room temperature in 20-30 min, shaken well. After allowing solid material to settle, the supernatant was decanted carefully. The colorless solid was washed with an additional amount of MTBE (2 x 2 mL). The resulting solid was dried under high vacuum to yield 0.064 g of Compound I Besylate Salt. Compound I Tosylate Salt of Compound I (0.024 g, 0.05 mmol) in dry EtOAc (0.5 mL) at
of p-toluenesulfonic acid (p-TsOH, 0.055 mL; 0.055 mmol; 1N in EtOAc) dropwise. During the addition of p-TsOH solution, the pink colored EtOAc clear solution of Compound I free base precipitated as a colorless solid. The reaction mixture was brought to room temperature in 20-30 min and shaken well. After allowing solid material to settle, the supernatant was decanted carefully. The colorless solid was washed with an additional amount of MTBE (2 x 2 mL). The resulting solid was dried under high vacuum to yield 0.027 g of Compound I Tosylate Salt. [00460] Melting points of salts of Compound I obtained in this example were determined by differential scanning calorimetry. The results are presented in Table 4. The monofumarate salt, produced by washing of the sesquifumarate salt has the highest melting point of the salts tested. Table 17. Melting points of salts of Compound I Compound I salt Onset of melting (°C) S lf t -1 85
Attorney Docket No.01394-0006-01PCT Example 23: Preparation of Fumarate Salts Preparation of Compound I Hemifumarate Pattern 1 [00461] About 200 mg of Compound I free base was added to 0.5 mL of EtOH. While stirring, 0.5 molar amount of fumaric acid was added at 50°C and the mixture was stirred for 2 hours. A clear solution was obtained. The solution was then cooled to 25°C within 1 hour. Hemi-fumarate seeds of Sample RC13 were added, followed by addition of 2.5 mL of heptanes to induce precipitation. An oil was obtained and stirred at 25°C for about 4 days. After 4 days, the resulting suspension was cooled to 5°C. After stirring at 5°C for about 4 days, the precipitated solids were collected by filtration and dried at 40°C under vacuum for about 3 hours. As a result, 116 mg of light-orange hemi-fumarate Pattern 1 were obtained in yield of 52%. XRPD is shown in FIG.9; DSC is shown in FIG.10; and TGA is shown in FIG.11. Preparation of Compound I Monofumarate Pattern 1 (small scale preparation) [00462] About 244 mg of Compound I free base was added into 0.8 mL of IPA. Then, 1.0 eq. of fumaric acid was added with stirring at 50°C for about 1.5 hours. The yellow clear solution obtained was cooled to 25°C and stirred for about 5 minutes. Mono-fumarate seeds of Sample RC 18 were added to the mixture, followed by addition of 4 mL of heptanes as an antisolvent. The mixture was stirred at 25°C for 4 days. The precipitated material was collected by filtration and dried at 40°C under vacuum for about 2 hours. As a result, 208 mg of mono-fumarate Pattern 1 solids were obtained in yield of 69%. XRPD is shown in FIG.9. DSC at 10°C/min is shown in FIG.12A, DSC at 2°C/min is shown in FIG.12B. DSC cycle results are shown in FIG.12C. TGA is shown in FIG.13. Table 18. Characterization of Compound I free base pattern and its mono- and hemifumarate patterns Compound I Compound I Compound I Monofumarate Pattern %
Attorney Docket No.01394-0006-01PCT Crystallinity (XRPD) High Medium Medium Melting point (DSC), °C 75.0 85.2 107.2
[00 63] Compound a ern s n g crys a n y. e em umara e and monofumarate Patterns are in medium crystallinity. [00464] Compound I Pattern 1 is an anhydrate and has a melting peak at Tonset of 75.0°C with an enthalpy of about 64 J/g. It shows about 0.3% weight loss at about 70°C. KF shows it contains about 1.7% of water. About 0.7% of MTBE (by weight) residue was detected by 1H NMR. [00465] Compound I hemifumarate Pattern 1 is an anhydrate. The stoichiometry of free form: fumaric acid is about 1:0.5 based on 1H NMR result. It has a melting peak at Tonset of 85.2°C with an enthalpy of about 37 J/g. It about 1.0% weight loss at about 85°C.
KF shows it contains about 1.7% of water. About 0.7% of EtOH and 0.7% of heptanes (by weight residual) was detected by 1H NMR. [00466] Compound I monofumarate Pattern 1 is an anhydrate. The stoichiometry of free form: fumaric acid is about 1:1.0 based on 1H NMR result. It has a melting peak at Tonset of 107.2°C with split peaks and with an
of about 78 J/g. The two thermal events cannot be resolved with 2 K/min and 0.5 K/min heating rate by DSC as well. It shows about 0.3% weight loss at about 107°C. KF shows it contains about 1.2% of water. About 0.7% of IPA and 2.2% of heptanes (by weight) residue was detected by 1H NMR. Example 24. Stability of Compound I free base Pattern
I Monofumarate Pattern 1 and Compound I hemifumarate Pattern 1 [00467] Initial chemical purity: Initial purities of Compound I free base Pattern 1, Compound I monofumarate Pattern 1 and Compound I hemifumarate Pattern 1 are 98.7%, 98.6% and 97.8%, respectively. [00468] Bulk stability: Accelerated stability experiments were conducted at 25°C/92%RH and 40°C/75%RH in an open container, 60°C in a tight container for one week. Results are presented in Table 19. Results for Compound I monofumarate Pattern 1 are also presented in FIG.14. [00469] All the three candidates showed good physical stability after exposure to the three conditions. They all show some degradation after exposure to 40°C/75% RH for one
Attorney Docket No.01394-0006-01PCT week, and two are not stable at high temperature (60°C for one week). Compound I hemifumarate Pattern 1 tends to degrade more than Compound I monofumarate Pattern 1 in the two conditions mentioned above. Table 19. Bulk stability of Compound I free base Pattern 1, Compound I monofumarate Pattern 1 and Compound I hemifumarate Pattern 1 Compound I Compound I Compound I Storage Conditions free base Hemifumarate Monofumarate P tt rn 1 P tt rn 1 P tt rn 1
Example 25. Solubility of Compound I free base Pattern 1, Compound I Monofumarate Pattern 1 and Compound I Hemifumarate Pattern 1 [00470] About 4 mg of Compound I Pattern 1 was added into 2 mL of buffer solution. For Compound I hemifumarate Pattern 1 and Compound I monofumarate Pattern 1, about 4 mg was added into 1.8 mL and 1.6 mL buffer solution, respectively. pH value was adjusted in simulated vaginal fluid with 0.2N NaOH. After stirring at 37℃ for 0.5 hour and 2 hours, clear solution was obtained for all the three candidates. Table 20. Solubility of Compound I free base Pattern 1, Compound I monofumarate Pattern 1 and Compound I hemifumarate Pattern 1 Compound I free base Compound I Compound I Hemifumarate Monofumarate
Attorney Docket No.01394-0006-01PCT Simulated vaginal fluid (pH adjusted with 0.2N >2 >2 >2 >2 (5.53 >2 >2 NaOH) ) (5.30) (5.39)
o u y as es e e e a, p . c a e u e , p . ace a e u e , p 6.8 phosphate buffer, water, simulated vaginal fluid (pH 4.2) at 37°C for 0.5 h and 2 h. All the three candidates are highly soluble in the media (>2 mg/mL). Example 26: Hygroscopicity of Compound I free base Pattern 1, Compound I Hemifumarate Pattern 1, and Compound I Monofumarate Pattern 1 [00472] Hygroscopicity was investigated by DVS at 25°C, using the following method: [00473] Method: 40-0-95-0-40%RH, dm/dt=0.002 [00474] The results are presented in Table 21 and in FIGS.15 to 20. Table 21. Hygroscopicity of Compound I free base Pattern 1, Compound I hemifumarate Pattern 1, and Compound I monofumarate Pattern 1 Compound I free base Compound I Compound I Pattern 1 Hemifumarate Pattern 1 Monofumarate Pattern 1 le 9 8 7 7 7 N
[00475] Hygroscopicity was investigated by DVS at 25°C. All three Patterns are moderately hygroscopic. Compound I Pattern 1 shows about 4.3% water uptake up to 90% RH but absorbs about 14.3% water in 95% RH. For Compound I monofumarate Pattern 1 and
Attorney Docket No.01394-0006-01PCT Compound I hemifumarate Pattern 1, they show about 2.9% and 6.9% water uptake in up to 95% RH. There is no form change after the DVS tests. Example 27: Large Scale Preparation of Mono-Fumarate Pattern 1 [00476] About 4.16 g of Compound I free base was dissolved in 11 mL of IPA. Then, 1.0 eq. of fumaric acid was added to the yellow clear solution with stirring at 50°C. After about 1 hour, some solids precipitated out. Then, 10 mg seeds of mono-fumarate (from the above ‘Small scale preparation’) were added. The mixture was stirred at 50°C for about 1.5 hours and cooled to 25°C, then it was stirred at 25°C for about 10 min. Then, 40 mL of heptanes was added as antisolvent. Obtained suspension was stirred at 25°C for about 24 hours, then cooled to 5°C at a rate of 0.1°C/min and stirred at 5°C for about 1 day. The solids were collected by filtration and dried in the oven at 40°C for about 2 hours under vacuum. About 4.1 g of light pink solids were obtained in a yield of 81.2%. XRPD is shown in FIG. 21A; DSC at 10°C/min rate is shown in FIG.21B; DSC at 2°C/min rate is shown in FIG. 21C; mDSC thermogram is shown in FIG.21D; TGA is shown in FIG.22. Table 22. Properties of Compound I Monofumarate Pattern 1 Parameter Method Result Purity HPLC 99.3%
Attorney Docket No.01394-0006-01PCT Example 28: Polymorph Screening Study of Compound I fumarate salts [00477] The polymorph screening study was performed for fumarate salts of Compound I (mixture of diastereomers). Their polymorphic behaviors were investigated by equilibration, precipitation by addition of antisolvent, slow cooling, fast cooling and slow evaporation experiments. Approximate solubility of the Compound I Monofumarate Pattern 1 at 25°C and 50°C glass
solubility at 25°C. About 10 mg of Compound I monofumarate Pattern 1 (Example 27) was weighed to a 2 mL glass vial and aliquot of 20 μL of each solvent (as shown in Table 23 below) was added to determine solubility at 50°C. Maximum volume of each solvent added was 1 mL. Approximate solubility was determined by visual observation. Table 23. Approximate solubility of Compound I monofumarate Pattern 1 at 25°C and 50°C Experiment Solvent Solubility (mg/mL) 25°C 50°C
Equilibration with solvents at 25°C for 2 weeks and 3 weeks [00479] About 30 mg of Compound I monofumarate Pattern 1 (obtained in Example 27) was equilibrated in suitable amount of solvent at 25°C for 2 weeks and 3 weeks with a stirring plate. Obtained suspension was filtered. The solid part (wet cake) was investigated by XRPD. When differences were observed, additional studies were performed (e.g., NMR, DSC, TGA, HPLC, and PLM). The results are presented in Tables 24-39 and in FIGS.23-45.
Attorney Docket No.01394-0006-01PCT Table 24. Results of equilibration with water for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks) EQ1 Pattern D DSC onset (enthalpy): Pattern D DSC onset (enthalpy): e
Table 25. Results of equilibration with acetonitrile for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks)
, . is shown in FIG.30; and TGA thermogram is shown in FIG.31. Table 26. Results of equilibration with MEK for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks)
Attorney Docket No.01394-0006-01PCT [00481] For Pattern B, XRPD diffractogram is shown in FIG.32; DSC thermogram is shown in FIG.33; and TGA thermogram is shown in FIG.34. Table 27. Results of equilibration with acetone for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks) EQ4 Pattern B n/a Pattern B n/a
[00482] XRPD diffractogram is shown in FIG.32 (3 weeks). Table 28. Results of equilibration with isopropanol for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks)
, . s shown in FIG.36; and TGA thermogram is shown in FIG.37. Table 29. Results of equilibration with acetone/toluene (1:1 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 test (2 weeks) (3 weeks) test (3 weeks)
Attorney Docket No.01394-0006-01PCT [00484] For Pattern E, XRPD diffractogram is shown in FIG.38; DSC thermograms are shown in FIG.39 and FIG.40; and TGA thermograms are shown in FIG.41 and FIG.42. Table 30. Results of equilibration with acetone/heptane (1:1 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks) EQ7 Simil r t S lt r ti =1: 056 Simil r t S lt r ti =1: 062
[00485] XRPD diffractogram is shown in FIG.32 (3 weeks). Table 31. Results of equilibration with IPA/heptane (1:1 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks)
[00486] XRPD diffractogram is shown in FIG.35. Table 32. Results of equilibration with IPA/toluene (1:1 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks)
Attorney Docket No.01394-0006-01PCT Note: Salt ratio is free base form:fumaric acid ratio [00487] XRPD diffractogram is shown in FIG.35. Table 33. Results of equilibration with IPA/MTBE (1:3 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks) EQ10 Mixture of Salt ratio =1: 065 Mixture of n/a
Table 34. Results of equilibration with THF/heptanes (1:3 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks)
. . . Table 35. Results of equilibration with EtOH/heptanes (1:3 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks) by
Attorney Docket No.01394-0006-01PCT Note: Salt ratio is free base form:fumaric acid ratio [00490] XRPD diffractogram is shown in FIG.35. Table 36. Results of equilibration with EA/toluene(1:3 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks) EQ13 Pattern 1 Salt ratio =1: 095 Pattern 1 n/a
[00491] XRPD is shown in FIG.43 and FIG.44 Table 37. Results of equilibration with EtOH/toluene (1:3 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks)
Table 38. Results of equilibration with water/ACN (2.9:97.1 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional (Solvent) (2 weeks) test (2 weeks) (3 weeks) test (3 weeks)
[00 93] d rac ogram s s own n G. 9. Table 39. Results of equilibration with IPA/heptanes (1:4 v/v) for Compound I monofumarate Pattern 1 at 25°C for 2 weeks and 3 weeks Exp. XRPD Additional XRPD Additional l t 2 k t t 2 k k t t k
Attorney Docket No.01394-0006-01PCT heptane, unknown 1.5% (by weight) heptane and 1:4 v/v) pattern 0.7% (by weight) IPA DSC, onset (enthalpy):
[00494] XRPD diffractogram is shown in FIG.45. Precipitation by addition of antisolvent [00495] About 30 mg of Compound I monofumarate Pattern 1 (Example 7) was dissolved in a good solvent. Antisolvent was added into the obtained solutions slowly. Precipitates were collected by filtration. The solid part (wet cake) was investigated by XRPD. When differences were observed, additional investigations were performed (e.g., NMR, DSC, TGA). If no precipitation was obtained, the solutions were cooled to 5°C for crystallization. After stirred at 5°C for about 23 days, no precipitation was obtained, the solutions were put in a -20°C freezer for crystallization. [00496] Results are presented in Table 40. XRPD diffractograms are shown in FIG.46 and FIG.47. Table 40. Precipitation of Compound I fumarate salts by addition of antisolvent Exp. Solvent Antisolvent XRPD Comments AS1 Acetone Heptanes Pattern B + Salt ratio =1:0.89 s ); at he nd ); at a
Attorney Docket No.01394-0006-01PCT AS7 THF Heptanes Pattern 1, Salt ratio =1:0.87 medium Residual solvent: 1.6% (by weight) heptanes crystallinity + solvent C at
Crystallization at room temperature by slow evaporation [00497] Combined with approximate solubility experiment, solubility samples were filtered through a 0.45 μm nylon filter. Obtained solutions were slow evaporated at ambient condition. Solid residues were examined for their polymorphic form. [00498] Results are presented in Table 41. XRPD diffractograms are shown in FIG.48 and FIG.49. Table 41. Crystallization at room temperature by slow evaporation Exp. Solvent XRPD 1 Acetone Pattern 1 with medium crystallinity
Crystallization from hot saturated solutions by slow cooling [00499] About 30 mg of Compound I monofumarate Pattern 1 was dissolved in the minimal amount of selected solvents at 50°C. Obtained solutions were cooled to 5°C at the rate of 0.1°C/min. Precipitates were collected by filtration. The solid part (wet cake) was investigated by XRPD. When differences were observed, additional investigations were performed (e.g., NMR, DSC, TGA). If no precipitation was obtained, the solutions were put in a -20°C freezer for crystallization. After storing in -20°C freezer for about 5 days, no precipitation was obtained, heptane was added as antisolvent. Precipitates were collected by filtration. The solid part (wet cake) was investigated by XRPD.
Attorney Docket No.01394-0006-01PCT [00500] Results are presented in Table 42. XRPD diffractograms are shown in FIG.50 and FIG.51. Table 42. Crystallization from hot saturated solutions by slow cooling Exp. Solvent XRPD Comments 1 Acetone Pattern B with medium Salt ratio =1:0.66; cr stallinit Residual solvent: 15% (b wei ht) acetone N % N s: s:
Crystallization from hot saturated solutions by fast cooling [00501] About 30 mg of Compound I monofumarate Pattern 1 was dissolved in the minimal amount of selected solvents at 50°C. Obtained solutions were put into an ice bath and agitated. Precipitates were collected by filtration. The solid part (wet cake) was investigated by XRPD. When differences were observed, additional investigations were performed (e.g., NMR, DSC, TGA). If no precipitation was obtained, the solutions were put in -20°C freezer for crystallization. After storing in -20°C freezer for about 7 days, no precipitation was obtained, heptanes was added as antisolvent. Precipitates were collected by filtration. The solid part (wet cake) was investigated by XRPD. [00502] Results are presented in Table 43. XRPD diffractograms are shown in FIG.52 and FIG.53. Table 43. Crystallization from hot saturated solutions by fast cooling Exp. Solvent XRPD Comments
Attorney Docket No.01394-0006-01PCT 1 Acetone Pattern B with low Free base form:fumaric acid=1:0.66; crystallinity 1.0% (by weight) acetone residue 2 IPA n/a Clear solution s:
Behavior of Compound I Monofumarate Pattern 1 under heating and cooling
by [00504] Cycle 1: 0°C to 106°C at 10°C/min; 106°C to 0°C at 10°C /min; reheat from 0°C to 250°C at 10°C/min. [00505] Cycle 2: 0°C to 130°C at 10°C /min; 130°C to 0°C at 10°C /min; reheat from 0°C to 250°C at 10°C /min. [00506] The results are presented in Table 44 and in FIG.54 and FIG.55. Table 44. Behavior of Compound I monofumarate Pattern 1 under heating and cooling Exp. Heating rate Thermal events 1 Cycle 1: 0°C to 106°C at 10°C/min; 106°C Heating:
Behavior under compression [00507] About 10 mg of Compound I monofumarate Pattern 1 was compressed for 5 minutes at 10 MPa with a hydraulic press. XRPD characterization was performed to
Attorney Docket No.01394-0006-01PCT investigate the polymorphic behavior under compression. According to the XRPD, no form change was observed. Grinding simulation experiments [00508] About 10 mg of Compound I monofumarate Pattern 1 was ground manually with a mortar and pestle for 5 min. Form transformation and degree of crystallinity was evaluated by XRPD. According to the XRPD, no form change was observed; crystallinity slightly decreased. Granulation simulation experiments [00509] Water or ethanol was added drop wise to about 10 mg of Compound I monofumarate Pattern 1 until solids were wetted sufficiently. The samples were ground manually with a mortar and pestle for 3 minutes. Samples are dried under ambient condition for 10 min. Form transformation and degree of crystallinity were evaluated by XRPD. According to the XRPD, neither in ethanol, nor in water form change was observed. Summary of identified mono- and hemifumarate polymorphs [00510] Compound I monofumarate Pattern 1 used in the study was prepared from Compound I free base according to Example 7. The initial form of monofumarate used in the polymorph study described below (Pattern 1) is an anhydrate of monofumarate with HPLC purity of about 99.3%. The ratio of free form and fumaric acid is about 1: 0.96 by 1H NMR. It has two melting peaks with Tonset of about 98.5°C with an enthalpy of about 14 J/g and 109.6°C with an enthalpy of about 25 J/g, as measured by differential scanning calorimetry (DSC). By thermogravimetric analysis (TGA), Pattern 1 shows about 0.5% weight loss at about 98°C and 0.6 % weight loss from 98°C to 140°C. About 1.0% (by weight) heptanes and 0.2% (by weight) IPA residue were detected by 1HNMR. Karl Fischer titration shows it contains about 1.3% water. [00511] During the polymorph study, four new patterns were identified. Although monofumarate was used as initial physical form obtained new patterns showed different stoichiometric ratio. Pattern B, Pattern C and Pattern E are hemifumarate salts, and Pattern D is a degradation product. [00512] Pattern B is an anhydrate of hemifumarate with HPLC purity of about 99.6%. With MEK, acetone, acetone/heptanes, MEK/heptanes and EtOH/MTBE as solvents, it can be obtained from equilibration experiment, addition of antisolvent, slow cooling and fast
Attorney Docket No.01394-0006-01PCT cooling experiments. The ratio of free base form to fumaric acid is about 1: 0.52 by 1H- NMR. It has two thermal events with Tonset of about 77.4°C with an enthalpy of about 71 J/g and 88.4°C with an enthalpy of about 18 J/g. It shows about 0.7% weight loss at about 77°C and 4.2% weight loss from 77°C to 130°C. About 4.6% (by weight) MEK residue was detected by 1H NMR. [00513] With ACN and ACN/water as solvents, a mixture of Pattern C and fumaric acid was obtained by equilibration, slow cooling and fast cooling experiments. The ratio of free base form to fumaric acid of the mixture is about 1: 0.95 by 1H NMR. After washing by water, the ratio decreased to about 1:0.76. This indicates that Pattern C is not a monofumarate. It has two thermal events with Tonset of about 74.0°C with an enthalpy of about 89 J/g and Tonset of about 90.6°C with an enthalpy of about 15 J/g. It shows about 0.4% weight loss at about 73°C and 2.1% weight loss from 73°C to 144°C. About 2.0% (by weight) ACN residue was detected by 1H NMR. [00514] Pattern D is a degradation product with HPLC purity of about 0.2%. It was obtained in water by equilibration experiment experiments. It has two thermal events with Tonset of about 41.4°C with an enthalpy of about 67 J/g and Tonset of about 72.1°C with an enthalpy of about 29 J/g. It shows about 0.6% weight loss at about 41°C and 8.5% weight loss from 41°C to 178°C. [00515] Pattern E is an anhydrate of hemifumarate with HPLC purity of about 98.9%. It was obtained in acetone/toluene by equilibration experiment. The ratio of free base form to fumaric acid of the mixture is about 1: 0.69 by 1H NMR. It has two thermal events with Tonset of about 53.1°C with an enthalpy of about 33 J/g and Tonset of about 96.5°C with an enthalpy of about 34 J/g. It shows about 1.0% weight loss at about 53°C and 3.6% weight loss from 53°C to 96°C. About 0.6% (by weight) acetone residue was detected by 1H NMR. [00516] Results of the study on Compound I mono- and hemifumarate polymorphs are summarized in Table 45 below. The salt ratio is the ratio between Compound I free base and the salt counterion. “AS” indicates the form can be prepared by antisolvent addition, using the solvent/antisolvent pairs listed in the table. “EQ” indicates the form can be prepared through equilibration in the listed solvent. “SC” indicates the form can be prepared by slowly cooling a solution of Compound I monofumarate in the listed solvent. “FC” indicates t the form can be prepared by quickly cooling a solution of Compound I monofumarate in the listed solvent.
Attorney Docket No.01394-0006-01PCT Table 45. Summary of identified Compound I fumarate salt Patterns 1, B, C, and E Melting Polymorph Salt Temperatures Salt ratio Preparation (Enthalpy)
Example 29: Preparation of Compound II Pattern 1 Experiment 1. Small-scale synthesis and preparation of seeds [00517] One hundred milligrams of RP Compound I free base and 0.3 mL of IPA were added into a glass vial. To it was added 1.0 equiv. of fumaric acid and the resulting mixture was stirred at 50°C for 2 min, most of the material was precipitated out. To it 1.0 mL of heptanes was added. The sample was stirred at 50°C for 1 h, then cooled to 3°C with 0.1°C/min. After stirring at 3°C for about 8 h, 0.4 mL of heptanes was added. Solids obtained were isolated by filtration and drying in vacuum oven at 40°C for about 2 h to obtain Compound II Pattern 1. The characterization results are reported in Table 46. XRPD diffractogram of Compound II Pattern 1 is shown in FIG.56. Table 46. Properties of Compound II Pattern 1 (small scale preparation) Parameter Method Result ° :
Attorney Docket No.01394-0006-01PCT Experiment 2. Large-scale preparation [00518] Three grams of RP Compound I free base and 9 mL of IPA were added into a glass vial. To it was added 1.0 equiv. of fumaric acid and the resulting mixture was stirred at 50°C for 5 min. About 21 mg of Compound II Pattern 1 seeds from Experiment 1 were added into the mixture.30 mL of heptanes was added into the mixture. The sample was stirred at 50°C for 1 h then cooled to 3°C with 0.1°C/min. After stirring at 3°C for about 16 h, the solid obtained was isolated by filtration and drying in vacuum oven at 40°C for about 4 h and at 50°C for about 3 h. This resulted in 2.9 g of white solid Compound II Pattern 1 in a yield of 78.3%. XRPD diffractogram of Compound II Pattern 1 is shown in FIG.56. DSC thermogram of Compound II Pattern 1 is shown in FIG.57. TGA thermogram of Compound II Pattern 1 is shown in FIG.58. Table 47. Properties of Compound II Pattern 1 (large scale preparation) Parameter Method Result Yield 78.3% of :
[00519] Compound II Pattern 1 is an anhydrate. The stoichiometry of free base:fumaric acid is about 1:1.0 based on 1H NMR result. It has a melting peak at Tonset of 141.5°C combined with
TGA shows about 0.3% weight loss at about 130°C. No residual solvent was detected by 1H NMR. Example 30: Preparation of
III Pattern 1 [00520] SP Compound I free base (100 mg) and 0.3 mL of IPA were added into a glass vial. To it was added 1.0 equiv. of fumaric acid and the mixture was stirred at 50°C for 15 min, most of the material was precipitated out. After addition of 1.0 mL heptanes, the sample was stirred at 50°C for 1 h, then cooled to 3°C with 0.1°C/min. After stirring at 3°C for about 8 h, 0.4 mL of heptanes was added into the mixture to get better suspension. White solid was isolated by filtration and drying in vacuum oven at 40°C for about 2 h to yield Compound III
Attorney Docket No.01394-0006-01PCT Pattern 1. The characterization results are reported in Table 48. XRPD diffractogram of Compound III Pattern 1 is shown in FIG.59. DSC thermogram of Compound III Pattern 1 is shown in FIG.60. TGA thermogram of Compound III Pattern 1 is shown in FIG.61. Table 48. Properties of Compound III Pattern 1 Parameter Method Result X-ray diffraction 3-40° (2 theta) Compound III Pattern 1 ° ° :
Example 31: Preparation of Compound III Pattern 2 [00521] SP Compound I free base (3 g) and 9 mL of IPA were added into a glass vial. After the addition of 1.0 equiv. of fumaric acid, a lot of solid precipitated out immediately.30 mL of heptanes was added into the mixture, then about 20 mg of Compound III Pattern 1 seeds were added into the mixture. The sample was stirred at 50°C for 1 h, then cooled to 3°C with 0.1°C/min. After stirring at 3°C for about 20 h, the sample was heated from 3°C to 50°C within 20 mins, then 0.2 equiv. of fumaric acid and 1.5 ml of heptanes were added into the mixture. The resulting mixture was stirred at 50°C for about 2 h then cooled to 3°C with 0.1°C/min. After stirring at 3°C for about 13 h, it was reheated to 50°C within 20 mins and kept stirred at 50°C for about 6 h. It was cooled to 3°C with 0.1°C/min and stirred at 3°C for about 2 days. The resulting solid was isolated by filtration and drying in vacuum oven at 50°C for about 3 h to obtain 3.1 g of white solid in a yield of 83.6%. The results are reported in Table 49. XRPD diffractogram of Compound III Pattern 2 is shown in FIG.62. DSC thermogram of Compound III Pattern 2 is shown in FIG.63. TGA thermogram of Compound III Pattern 2 is shown in FIG.64. Table 49. Properties of Compound III Pattern 2 Parameter Method Result
Attorney Docket No.01394-0006-01PCT Stoichiometry and 1H NMR Free base : fumaric acid=1:1.2; residual solvent No residual solvent M r h l PLM Irr l r rti l
o pou a e s a a y a e. e s oc o e y o ee ase: fumaric acid is about 1:1.2 based on 1H NMR result. It has two melting peaks at Tonset of 106.4°C with an enthalpy of about 47 J/g and Tonset of 118.8°C with an enthalpy of about 63 J/g. It shows about 0.3% weight loss at about 105°C. No residual solvent was detected by 1H NMR. Example 32: Bulk stability of Compound II Pattern 1 and Compound III Pattern 2 [00523] Compound II Pattern 1 and Compound III Pattern 2 were placed in an open container at 25°C/92% RH, in an open container at 40°C/75% RH, and in a closed container at 60°C for 1 week. Samples were characterized by XRPD and HPLC and inspected for color change. The results are presented in Table 50. Table 50. Stability: purity and appearance Parameter Physical form Compound II Pattern 1 Compound III Pattern 2 ge ge ge
Initial chemical and chiral purity [00524] Initial chemical purity of Compound II Pattern 1 and Compound III Pattern 2 is 99.7% and 98.8%, respectively. Chiral purity (%de) of Compound II Pattern 1 is 98.4%. Bulk stability
Attorney Docket No.01394-0006-01PCT [00525] Accelerated stability experiments were conducted at 25°C/92% RH in an open container, at 40°C/75% RH in an open container, and at 60°C in a tight container for one week. Compound II Pattern 1 showed good physical and chemical stability after exposure to the three conditions. Compound III Pattern 2 showed good physical stability at the above mentioned three conditions. Degradation product increased by 1.6% and 1.5% after exposure to 40°C/75% RH in an open container and 60°C in a tight container, respectively. Example 33: Solubility study of Compound II Pattern 1 and Compound III Pattern 2 [00526] Accurately 12 mg of Compound II Pattern 1, 12 mg of Compound III Pattern 2 were weighed into an 8 mL glass vial and to it 5 mL of solubility medium was added. The salt amount used is equivalent to 10 mg anhydrous free base. All the samples were clear solution in the media after 0.5 and 2h at 37°C. The obtained clear solutions were analyzed by pH meter for pH, and solubility was determined by observation. Table 51. Solubility at 37°C, target concentration 10 mg/5 mL, equilibration for 0.5 h and 2 h Compound II Pattern 1 Compound III Pattern 2 E S lv nt m di )
[00527] Solubility was tested in five media, pH 3.0 citrate buffer, pH 4.5 acetate buffer, pH 6.8 phosphate buffer, water, simulated vaginal fluid (pH 4.2) at 37°C for 0.5 h and 2 h. The two candidates, Compound II Pattern 1 and Compound III Pattern 2, are highly soluble in the media (>2 mg/mL).
Attorney Docket No.01394-0006-01PCT Example 34: Hygroscopicity of Compound II Pattern 1 and Compound III Pattern 2 [00528] Hygroscopicity was investigated by DVS at 25°C, using the following method: [00529] Method: 40-0-95-0-40%RH, dm/dt=0.002 [00530] Both Compound II Pattern 1 and Compound III Pattern 2 are slightly hygroscopic. Compound II Pattern 1 shows about 0.2% water uptake up to 95% RH. No form change after the DVS test was observed. Compound III Pattern 2 shows about 1.0% water uptake up to 95% RH. No form change after the DVS test was observed. Results are presented in Table 52. Table 52. Compound II Pattern 1 Compound III Pattern 2 Desorp Sorp Desorp Sorp Desorp Sorp Desorp Sorp 2
Example 35: Polymorph screening of Compound II Pattern 1 and Compound III Pattern 2 [00531] Monofumarate (50 mg) was equilibrated in suitable amount of solvent or solvent mixture. Obtained suspensions were equilibrated for 1 week. Solids were isolated by centrifugation filtration. Wet cakes obtained after equilibration were analyzed by XRPD to determine crystal form change.
Attorney Docket No.01394-0006-01PCT Table 53. Mini-polymorph screening: Crystal modification after 1 week equilibration at 25°C (salt ratio is defined as free base : fumaric acid ratio Exp. Parameter Compound II Pattern 1 Compound III Pattern 2 Slvnt Chr triztin r lt Chr triztin r lt by C, y: y: by C, C, y: K II ue
Attorney Docket No.01394-0006-01PCT PS8 water/ACN Compound II Pattern 2 Compound III Pattern 4 (2.9/97.1, v/v) C m nd II P tt rn 1 C m nd III P tt rn 6 g; g;
Polymorphism evaluation of Compound II Pattern 1 during equilibration experiments
and 2 II Pattern 2 and Compound II Pattern 3) of Isomer I were obtained from acetonitrile and MEK, respectively. These both showed lower melting temperature than for Compound II Pattern 1. Both polymorphs of isomer 1 show unchanged high chiral purity. Polymorphism evaluation of Compound III Pattern 2 [00533] In this study, dissociation was observed during equilibration experiments, a hemifumarate (hemifumarate Pattern 2) of Isomer II was obtained. In addition, 4 new monofumarate salts (Compound III Pattern 3, Compound III Pattern 4, Compound III Pattern 5 and Compound III Pattern 6) of Isomer II were obtained. Example 36: X-ray Powder Diffraction (XRPD) [00534] The XRPD analysis was carried out on a Bruker D8 Advance diffractometer. [00535] Table 54 below provides the results of the XRPD performed on Compound II Pattern 1. The XRPD exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Compound II Pattern 1 at about 3.1±0.2°, about 9.3±0.2°, about 12.1±0.2°, about 14.9±0.2°, about 15.1±0.2°, about 18.1±0.2°, about 19.8±0.2°, about 20.1±0.2°, about 25.1±0.2°, about 25.9±0.2°, and about 28.8±0.2°. Table 54. XRPD Peak List For Compound II Pattern 1 Pos. [°2θ] d-spacing d-spacing Å Rel. Int. [%] Pos. [°2θ] Å Rel. Int. [%]
Attorney Docket No.01394-0006-01PCT 9.300 9.50181 33.0 27.587 3.23082 3.2 10.660 8.29244 9.9 27.994 3.18474 10.6 12084 731818 846 28824 309496 639
[00536] Table 55 below provides the results of the XRPD performed on Compound IV Pattern 1. Significant peaks were observed in the XRPD on Compound IV Pattern 1 at about 6.5±0.2°, about 12.1±0.2°, about 17.5±0.2°, about 18.1±0.2°, about 18.5±0.2°, about 19.6±0.2°, about 19.8±0.2°, about 20.2±0.2°, about 20.6±0.2°, and about 21.3±0.2. Table 55. XRPD Peak List For Compound IV Pattern 1 Pos. [°2θ] d-spacing Rel. I d-spacing [Å] nt. [%] Pos. [°2θ] [Å] Rel. Int. [%]
Attorney Docket No.01394-0006-01PCT 17.522 5.05725 17.9 32.193 2.77826 2.8 18.141 4.88628 28.5 32.628 2.74228 3.7 18526 478544 304 35573 252168 32
[0053 ] ab e 56 be ow prov des e resu s o e per ormed on Compound III Pattern 1. Significant peaks were observed in the XRPD on Compound III Pattern 1 at about 9.5±0.2°, about 11.7±0.2°, about 14.6±0.2°, about 17.5±0.2°, about 18.0±0.2°, about 20.0±0.2°, about 20.4±0.2°, about 22.3±0.2°, about 23.7±0.2°, and about 25.5±0.2. Table 56. XRPD Peak List For Compound III Pattern 1 Pos. [°2θ] d-spacing d-s [Å] Rel. Int. [%] Pos. [°2θ] pacing [Å] Rel. Int. [%]
[00538] Table 57 below provides the results of the XRPD performed on Compound III Pattern 2. The XRPD exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Compound III Pattern 2 at about 8.9±0.2°, about 9.9±0.2°, about 11.7±0.2°, about 12.1±0.2°, about 15.1±0.2°, about 17.9±0.2°, about 18.2±0.2°, about 19.9±0.2°, about 25.1±0.2°, about 29.6±0.2°, and about 38.1±0.2°.
Attorney Docket No.01394-0006-01PCT Table 57. XRPD Peak List For Compound III Pattern 2 Pos. [°2θ] d-spacing Rel. d-spacing [Å] Int. [%] Pos. [°2θ] [Å] Rel. Int. [%] 6078 1453063 86 22362 397249 80
[00539] Table 58 below provides the results of the XRPD performed on Compound V Pattern 1. Significant peaks were observed in the XRPD on Compound V Pattern 1 at about 5.0±0.2°, about 7.2±0.2°, about 10.1±0.2°, about 12.1±0.2°, about 17.5±0.2°, about 17.9±0.2°, about 19.3±0.2°, about 22.0±0.2°, about 24.3±0.2°, about 25.1±0.2°, and about 26.3±0.2. Table 58. XRPD Peak List For Compound V Pattern 1 Pos. [°2θ] d-spacing Re d-spacing [Å] l. Int. [%] Pos. [°2θ] [Å] Rel. Int. [%]
Attorney Docket No.01394-0006-01PCT 15.674 5.64928 17.0 28.741 3.10368 12.8 17.625 5.02802 32.8 28.955 3.08118 25.5 17457 507608 696 33035 270936 144
[ ] a e e ow prov es e resu s o e per orme on ompoun V Pattern 2. Significant peaks were observed in the XRPD on Compound V Pattern 2 at about 5.1±0.2°, about 6.9±0.2°, about 7.6±0.2°, about 10.2±0.2°, about 11.6±0.2°, about 12.1±0.2°, about 15.1±0.2°, about 17.6±0.2°, about 18.1±0.2°, about 18.7±0.2°, about 19.5±0.2°, about 19.8±0.2°, and about 25.1±0.2°. Table 59. XRPD Peak List For Compound V Pattern 2 Pos. [°2θ] d-spacing Rel d-spacing [Å] . Int. [%] Pos. [°2θ] [Å] Rel. Int. [%]
Example 37. Tablet Stability Study [00541] Tablets were prepared using the excipients and excipient ratios shown in the first column of the following tables. The tablets were then stored under the conditions indicated and periodically sampled and tested for purity by HPLC. The tablets produced from Compound I monofumarate degraded less than tablets produced from Compound I across a range of different formulations.
Attorney Docket No.01394-0006-01PCT Table 60. Tablets produced with Compound I Excipients 0 months 1 month 25C/60% RH 1 month 40C/75% RH PC (49%) + Anhydrous 99.40 95.98 41.3 Lactose (49%)
Excipients 0 months 1 month 25C/60% RH 1 month 40C/75% RH PEO-15 (25%) + MCC 99.03 97.76 83.13
Example 38. Clinical Studies in Patients with Cervical Squamous Intraepithelial Lesions [00542] A single and multiple-dose study was undertaken to evaluate the safety, tolerability, and efficacy of Compound I monofumarate vaginal tablets (also known as vaginal inserts) in participants with cervical squamous intraepithelial lesions. A primary objective was to assess the safety and tolerability of the vaginal tablets when administered intravaginally to the cervix of participants with cervical intraepithelial neoplasia (CIN), also referred to as Squamous Intraepithelial Lesions (SIL). [00543] Vaginal tablets were prepared from Compound I monofumarate in sufficient amount to the required dose of Compound I. The compositions of the vaginal tablets of different doses (weighing 175 mg total) are shown below in Tables 62 and 63. Table 62 Ingredient 0.05 mg dose tablet 0.1 mg dose tablet
Attorney Docket No.01394-0006-01PCT Microcrystalline cellulose QS to 175 mg QS to 175 mg Total Tablet Weight 175 mg 175 mg
Table 63 Ingredient 0.3 mg dose tablet 1 mg dose tablet
. [00544] In this study, vaginal tablets are introduced into the cervix using an applicator or a retaining device. In cohorts using the applicator, a retaining device (such as a tampon) is optionally used after dosing in order to minimize leakage of vaginal secretions. When self- administered at home, the vaginal tablets are preferably inserted at bedtime. In some cohorts, the vaginal tablets are pre-moistened with a water-based lubricant. [00545] The study consisted of two parts – Part A (including an optional cohort) and Part B, as described below. [00546] Part A includes up to11 cohorts and an optional cohort: Cohort A1: single dose of 0.1 mg followed by up to 3 doses of 0.1 mg within 1 week Cohort A2: 0.1 mg for up to 3 doses over 1 week Cohort A3: up to 0.3 mg for up to 3 doses over 1 week Cohort A4: up to 0.3 mg for 6 doses over 2 weeks Cohort A5: up to 1.0 mg for up to 6 doses over up to 2 weeks Cohort A6: up to 1.0 mg for up to 6 doses over up to 4 weeks (dose regimen to be determined based on safety data from all previous cohorts in Part A and available pharmacokinetic (PK) and efficacy data; in the event of menstrual cycles occurring during the 3- or 4-week treatment period, dosing may be held for up to a week and resume after completion of the menstrual cycle)
Attorney Docket No.01394-0006-01PCT Cohort A7: up to 1.0 mg for up to 6 doses over up to 2 weeks (defined as a single treatment cycle); up to 2 treatment cycles (the second treatment cycle may be administered after a week break at a minimum, and in the event of menstrual cycles occurring between the two cycles, dosing may be held for another week and retreatment resumed after completion of the menstrual cycle) Cohort A8: up to 1.0 mg, 2 intermittent weekly dosing (1 week of treatment, followed by 1 week treatment break, followed by 1 week of treatment over 3 total weeks) for up to 6 doses (defined as a single treatment cycle); up to 2 treatment cycles (the second treatment cycle may be initiated after about a week break at a minimum, allowing for the menstrual cycle to complete between the two cycles.) Cohorts A9, A10, and A11: doses and regimens with or without retreatment to be determined based on safety data from all previous cohorts through 7 days following last dose of study drug, and available PK and efficacy/pharmacodynamic (PD) data Optional Cohort: up to 1.0 mg for up to 6 doses over up to 2 weeks. For cohorts A7-A11, the maximum dose will not exceed 24 mg. [00547] The proposed dosing regime will be guided using the following parameters: Dosage: up to 1.0 mg Number of doses per treatment cycle: up to 6 doses (cumulative dose not to exceed 3- fold that tested in any prior cohort) Number of treatment cycles: up to 2 cycles Treatment breaks (time between 2 cycles when no drug is given): minimum of 1 week - Repeat cycles may only be initiated if Grade ≥ 2 lower genitourinary AEs from previous cycles are resolving or stabilizing (i.e., returning to normal), per the Investigator assessment. Total treatment duration: up to 8 weeks (i.e., inclusive of all treatment cycles; no treatment within 4 weeks of LLETZ) Method of administration: vaginal applicator (with or without tampon) or retaining device (e.g., menstrual cup/disc or diaphragm) [00548] Part B may include up to 3 proof of concept (POC) cohorts, and Part B enrollment may proceed when a dose regimen tested in Part A is deemed safe and tolerated and demonstrates some level of efficacy. Each Part B POC cohort (up to 3 cohorts will be expanded; Expanded Cohorts could be from A3 through A11, based on the data from Part A) will serve as an expansion of the corresponding Part A cohort as follows: Expanded Cohort A3: up to 0.3 mg for up to 3 doses over 1 week Expanded Cohort A4: up to 0.3 mg for 6 doses over 2 weeks
Attorney Docket No.01394-0006-01PCT Expanded Cohort A5: up to 1.0 mg for up to 6 doses over up to 2 weeks Expanded Cohort A6: up to 1.0 mg for up to 6 doses over up to 4 weeks Expanded Cohort A7: up to 1.0 mg for up to 6 doses over up to 2 weeks (defined as a single treatment cycle); up to 2 treatment cycles Expanded Cohort A8: up to 1.0 mg, 2 intermittent weekly dosing (1 week of treatment, followed by 1 week treatment break, followed by 1 week of treatment over 3 total weeks) for up to 6 doses (defined as a single treatment cycle); up to 2 treatment cycles Expanded Cohorts A9, A10, and A11: doses and regimens with or without retreatment to be determined based on safety data from all previous cohorts through 7 days following last dose of study drug, and available PK and efficacy/PD data [00549] The presence of SIL lesions will be determined based on historical pathology. In Part A cohorts A3 through A11 and Part B POC cohorts, cHSIL will be confirmed by positive p16 status. Histology diagnosis must be obtained from the biopsy within 60 days prior to the first dose administration. The lesion must be fully visible and evaluable and involve no more than 3 quadrants based on colposcopic evaluation at the time of enrollment. [00550] Participants who will be using a vaginal applicator will be instructed in insertion of the vaginal tablet to the cervix with the applicator. Participants may be asked to insert a tampon (as a retaining device) after the vaginal tablet is administered with the applicator (e.g., immediately after administration or after some period of time after administration). In some embodiments, the participants may be asked to insert the tampon about 6 to about 8 hours after the vaginal tablet is administered. Participants may be instructed to remove tampon within about 8 hours. [00551] Participants using the retaining device will be instructed in the procedure for placing the vaginal tablet in the retaining device (menstrual cup/disc or diaphragm) with water-based lubricating gel and inserting the device over the cervix. [00552] If administration is in a clinic, participants will be instructed to remain supine whenever possible/feasible for 6 hours (± 1 hour) with minimal activities, and normal activities may resume 6 hours post-dose. [00553] All participants in each of the cohorts will receive wellness checks during the treatment period to ensure compliance and monitoring for AEs. [00554] In addition to assessing the safety, tolerability and the plasma concentrations of Compound I, the efficacy of treatment will be evaluated by analyzing changes in lesions detected by colposcopy and histologically by LLETZ procedure. All participants in both Parts A and B will undergo colposcopy with documentation of any lesions present and visual
Attorney Docket No.01394-0006-01PCT assessment of lesion size on Day 42, and all participants with screening biopsy results of HSIL (CIN 2 or CIN 3) will undergo LLETZ procedure within 12 weeks of administration of the first vaginal tablet dose. Participants with screening biopsy results of LSIL (CIN 1) will be referred for follow-up according to the standard of care. [00555] Efficacy will also be evaluated by the detection of HPV as a viral biomarker. HPV status will be evaluated with the Roche cobas® HPV Test collected using a cervical swab. For Parts A and B for 1-week dose regimen (dosing 3x/week), HPV testing will occur at Baseline, Days 15, 29, 42, and prior to the LLETZ procedure. Should the 2-week extended (dosing 3x/week) treatment be administered in Cohorts A4 and A5 or Part B POC cohort(s), HPV testing will be performed at Baseline, Days 19, 29, 42, and prior to the LLETZ procedure. For Cohorts A6 through A11 and if expanded in Part B, HPV testing will be performed at Baseline, Days 15 or 19, 29 or last day of dose, 42, and prior to the LLETZ procedure. Cytology will also be evaluated using the same cervical swab. Example 39. Clinical Studies in Patients diagnosed with persistent cervical hrHPV infection [00556] A randomized, double-blind, placebo-controlled Phase 1b/2 study in women diagnosed with persistent cervical hrHPV infection was undertaken to assess safety, tolerability, and efficacy following the use of Compound I monofumarate vaginal tablets (also known as vaginal inserts) delivered intravaginally. In addition, the study was undertaken to evaluate persistence of type-specific hrHPV DNA test negativity following treatment with various doses and dosing regimens of the Compound I monofumarate vaginal tablets vs. placebo. [00557] Vaginal tablets were prepared from Compound I monofumarate as described above in Example 38. [00558] Following screening, eligible participants will be randomized to receive Compound I monofumarate vaginal tablets vs. placebo in a 3:1 ratio, stratified (in the expansion cohorts) based on age at randomization (25 to < 30 years of age vs. ≥ 30 years of age) and hrHPV genotype (HPV-16 and/or -18 vs. any other hrHPV genotype). All participants will end the study at 24 weeks after randomization. A post-study hrHPV test will be performed at Week 52 (± 4 weeks) in the clinic or at home, as feasible. [00559] Participants will administer the study drug using a vaginal applicator or, depending on participant experience and emerging safety/tolerability/efficacy data during the study, study drug may be administered using a retaining device (in countries where available) instead. Participants assigned to vaginal applicator use may be directed to use a tampon as a
Attorney Docket No.01394-0006-01PCT hygienic measure; unless otherwise specified, the tampon should be placed 6 to 8 hours following dosing. [00560] The proposed doses/regimens of the Compound I monofumarate vaginal tablets or matching placebo for the sentinel cohorts are as follows: Cohort 1: 0.05 mg, 3×/week over 2 weeks Cohort 2: 0.1 mg, 3×/week over 2 weeks Cohort 3: up to 0.3 mg, up to 3×/week over up to 2 weeks Cohort 4: up to 0.1 mg, up to 3×/week over up to 2 weeks; up to 2 treatment cycles (Optional) Cohort 5: up to 0.3 mg, up to 3×/week over up to 2 weeks; up to 2 treatment cycles Cohort 6: up to 1.0 mg, up to 3×/week over up to 2 weeks Cohort 7: up to 1.0 mg, up to 3×/week over up to 2 weeks; up to 2 treatment cycles (Optional) Cohorts 8 through 11: up to 1.0 mg, up to 6 doses/treatment cycle [00561] All participants in sentinel cohorts with ≥ 2 treatment cycles will receive the same treatment as assigned for the first treatment cycle regardless of their HPV DNA test results at Week 4. [00562] The parameters for selecting doses and dosing regimens for Cohorts 8 through 11 are as follows: Maximum individual dose: up to 1.0 mg Dose escalation increment: Cumulative dose will be no more than 3-fold higher than a cumulative dose tested in a prior cohort in this study. Maximum total treatment duration: up to 8 weeks (inclusive of treatment breaks) Minimum treatment breaks (i.e., time between 2 cycles when no drug is given): 1 week Method of administration: vaginal applicator or alternate method as specified [00563] Repeat cycles will only be initiated if Grade ≥ 2 lower genitourinary AEs from the previous cycle are resolving or stabilizing (ie, returning to normal). The maximum cumulative dose of Compound I to be tested will not exceed 24 mg. [00564] Expanded Proof of Concept (POC) cohorts: These may include up to 4 POC cohorts, and randomization may proceed when a dose regimen tested in a sentinel cohort is deemed safe and tolerated by the SMC and demonstrates some level of efficacy. [00565] All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were
Attorney Docket No.01394-0006-01PCT specifically and individually indicated to be incorporated by reference. [00566] Although the foregoing disclosure has been described in some detail by way of illustration and example for the purposes of clarity of understanding, it will be readily apparent to one of ordinary skill in the art in light of the teaching of this disclosure that certain changes and modifications may be made thereto without departing from the spirit or scope of the disclosure as defined in the embodiments and/or claims.
Claims
Attorney Docket No.01394-0006-01PCT CLAIMS: 1. A method of treating a cervical or vaginal HPV infection or an associated condition in a female in need thereof, comprising (i) administering a pharmaceutically acceptable composition comprising an effective amount of dosage form of a therapeutic compound selected from:
(ii) and inserting a retaining device.
Attorney Docket No.01394-0006-01PCT 2. The method of claim 1, wherein the therapeutic compound is: . acceptable composition
is a 4. The method of claim 3, wherein the solid dosage form is in the form of a vaginal tablet. 5. The method of any one of claims 1-4, wherein the pharmaceutically acceptable composition is administered with a vaginal applicator. 6. The method of any one of claims 1-5, wherein the pharmaceutically acceptable composition is administered to the base of the cervix. 7. The method of any one of claims 1-6, wherein the retaining device is inserted in the vaginal area below the cervical base. 8. The method of any one of claims 1-7, wherein the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap and a sponge. 9. The method of any one of claims 1-8, wherein the retaining device is used as both the applicator and a retaining means. 10. The method of any one of claims 1-9, wherein the retaining device is inserted below the cervical base for a sufficient time to collect the post-treatment leakage of the therapeutic compound or its PMEG metabolite. 11. The method of any one of claims 1-10, wherein the compound is an isolated morphic form of: wherein the
pattern comprising peaks independently selected from at least 3, 4, 5, or 6 of the following 2theta values
Attorney Docket No.01394-0006-01PCT 3.08+0.2°, 9.30+0.2°, 12.08+0.2°, 14.92+0.2°, 15.10+0.2°, 20.14+0.2°, 25.14+0.2°, and 28.82+0.2°. 12. The method of any one of claims 4-11, wherein the tablet disintegrates in less than about 250 μL of fluid or less than about 150 μL of fluid. 13. The method of any one of claims 1-12, wherein the amount of therapeutic compound administered is equivalent to from about 0.05 to 1 mg, or from about 0.1 to 1 mg, or from about 0.1 to 0.5 mg, or from about 0.2 to 0.3 mg, or about 0.3 mg of the free base. 14. The method of any one of claim 1-13, wherein the therapeutic compound comprises from about 0.01% to 10% of the pharmaceutical composition. 15. The method of any one of claim 1-14, wherein the pharmaceutically acceptable composition comprises an excipient chosen from a polymer, a disintegration enhancing excipient, a penetration enhancing excipient, and an excipient that allows for controlled release. 16. The method of any one of claim 1-15, wherein the pharmaceutically acceptable composition comprises at least one of a light mineral oil, propylparaben, Tefose 63, water, EDTA, methylparaben and Carbopol 974P. 17. The method of any one of claim 1-16, wherein the pharmaceutically acceptable composition comprises at least one of EDTA, methyl paraben, Carbopol 974P, propylene glycol and sorbic acid. 18. The method of any one of claim 1-17, wherein the pharmaceutically acceptable composition comprises mannitol, a polycrystalline cellulose (such as microcrystalline cellulose), and magnesium stearate. 19. The method of any one of claims 1-18, wherein the condition is atypical squamous cells of undetermined significance (ASC-US), atypical glandular cells (AGC), low- grade squamous intraepithelial lesions (LSIL), high grade squamous intraepithelial lesions (HSIL), or adenocarcinoma in situ (AIS). 20. The method of any one of claims 1-18, wherein the condition is atypical squamous cells, cannot exclude high grade squamous intraepithelial lesion (ASC-H). 21. The method of any one of claims 1-18, wherein the condition is cervical intraepithelial neoplasia. 22. The method of any one of claims 1-18, wherein the condition is cervical intraepithelial neoplasia Grade 1, Grade 2, or Grade 3.
Attorney Docket No.01394-0006-01PCT 23. The method of any one of claims 1-22, further comprising applying a lubrication means to the epithelial tissue and/or to the dosage form before inserting the dosage form in the affected area. 24. The method of claim 23, wherein the lubrication means is selected from water, a glycerol-based lubricant, and a hydroxyethylcellulose-based lubricant. 25. The method of any one of claims 1-24, wherein the pharmaceutical composition is administered once per day, twice per week, or three times or more per week. 26. The method of any one of claims 1-25, wherein the pharmaceutical composition is administered for about one week, for about two weeks, for about three weeks, for about four weeks, for about five weeks, for about six weeks, or for about eight weeks. 27. The method of any one of claims 1-26, wherein the pharmaceutical composition is administered in a therapeutic cycle comprising: a. a treatment cycle comprising administering the pharmaceutical composition, and b. a rest cycle comprising a period of no treatment. 28. The method of claim 27, wherein the rest cycle is about one week, about two weeks, or about three weeks. 29. A kit that comprises: (i) a dosage form comprising a therapeutic compound selected from: ,
Attorney Docket No.01394-0006-01PCT and
30. a 31. The kit of claim 28 or 29, wherein the dosage form is a vaginal tablet. 32. The kit of any one of claims 28-31, wherein the retaining device is selected from a menstrual cup, a menstrual disc, a tampon, diaphragm, cervical cap and a sponge. 33. The kit of any one of claims 28-32, wherein the retaining device is used as both the applicator and a retaining means.
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202463618174P | 2024-01-05 | 2024-01-05 | |
| USPCT/US2024/010538 | 2024-01-05 | ||
| PCT/US2024/010538 WO2025147262A1 (en) | 2024-01-05 | 2024-01-05 | Device system and use for hpv infection and cervical intraepithelial neoplasia |
| US63/618,174 | 2024-01-05 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025147598A1 true WO2025147598A1 (en) | 2025-07-10 |
Family
ID=96300711
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2025/010220 Pending WO2025147598A1 (en) | 2024-01-05 | 2025-01-03 | Device system and use for hpv infection and cervical intraepithelial neoplasia |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025147598A1 (en) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20080268022A1 (en) * | 2007-02-23 | 2008-10-30 | Mccabe R Tyler | Mehtods for treating and preventing ailments caused by human papillomavirus |
| US8859756B2 (en) * | 2010-03-31 | 2014-10-14 | Gilead Pharmasset Llc | Stereoselective synthesis of phosphorus containing actives |
| US20220395510A1 (en) * | 2014-09-15 | 2022-12-15 | The Regents Of The University Of California | Nucleotide Analogs |
| US11814406B2 (en) * | 2020-08-27 | 2023-11-14 | Gilead Sciences, Inc. | Compounds and methods for treatment of viral infections |
-
2025
- 2025-01-03 WO PCT/US2025/010220 patent/WO2025147598A1/en active Pending
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20080268022A1 (en) * | 2007-02-23 | 2008-10-30 | Mccabe R Tyler | Mehtods for treating and preventing ailments caused by human papillomavirus |
| US8859756B2 (en) * | 2010-03-31 | 2014-10-14 | Gilead Pharmasset Llc | Stereoselective synthesis of phosphorus containing actives |
| US20220395510A1 (en) * | 2014-09-15 | 2022-12-15 | The Regents Of The University Of California | Nucleotide Analogs |
| US11814406B2 (en) * | 2020-08-27 | 2023-11-14 | Gilead Sciences, Inc. | Compounds and methods for treatment of viral infections |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| KR101940824B1 (en) | Treatment of pain associated with dislocation of basal endometrium | |
| US20240148729A1 (en) | Lopinavir and ritonavir for the treatment of cervix disorders | |
| TWI737861B (en) | N-[5-(aminosulfonyl)-4-methyl-1,3-thiazol-2-yl]-n-methyl-2-[4-(2-pyridinyl)-phenyl]-acetamide free base hemihydrate, methods of manufacture and uses thereof | |
| MX2011001410A (en) | Lower dosage strength imiquimod formulations and short dosing regimens for treating genital and perianal warts. | |
| WO2019224777A1 (en) | Pharmaceutical compositions | |
| US20250295679A1 (en) | Compositions and Dosage Forms for Treatment of HPV Infection and HPV-Induced Neoplasia | |
| US20230285287A1 (en) | Pharmaceutical compositions | |
| WO2019224776A1 (en) | Pharmaceutical compositions | |
| WO2025147598A1 (en) | Device system and use for hpv infection and cervical intraepithelial neoplasia | |
| WO2025147262A1 (en) | Device system and use for hpv infection and cervical intraepithelial neoplasia | |
| CN117751129A (en) | Compositions and dosage forms for treating HPV infection and HPV-induced neoplasia | |
| WO2005007191A1 (en) | Medicinal composition | |
| WO2025160227A1 (en) | Advantageous compounds for treatment of hpv infection and hpv-induced neoplasia | |
| HK40083240A (en) | Pharmaceutical composition for preventing or treating viral perivaginal disease | |
| TW202216142A (en) | Pharmaceutical composition for preventing or treating viral perivaginal disease | |
| JP2007022980A (en) | Treatment for endometriosis | |
| WO2015125082A1 (en) | Improved process for the preparation of ritonavir |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 25736519 Country of ref document: EP Kind code of ref document: A1 |