WO2019224776A1 - Pharmaceutical compositions - Google Patents

Pharmaceutical compositions Download PDF

Info

Publication number
WO2019224776A1
WO2019224776A1 PCT/IB2019/054289 IB2019054289W WO2019224776A1 WO 2019224776 A1 WO2019224776 A1 WO 2019224776A1 IB 2019054289 W IB2019054289 W IB 2019054289W WO 2019224776 A1 WO2019224776 A1 WO 2019224776A1
Authority
WO
WIPO (PCT)
Prior art keywords
anhydrous composition
weight
composition according
muco
anhydrous
Prior art date
Application number
PCT/IB2019/054289
Other languages
French (fr)
Inventor
Fergus Cameron Binnie
Marten Geert Vos
Original Assignee
Douglas Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Douglas Pharmaceuticals Ltd filed Critical Douglas Pharmaceuticals Ltd
Publication of WO2019224776A1 publication Critical patent/WO2019224776A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof

Definitions

  • compositions for topical application comprising at least one active pharmaceutical ingredient and a hydrophilic muco-adhesive agent; methods for their manufacture; and the use of said anhydrous compositions as a medicament.
  • compositions are provided comprising lopinavir and ritonavir for use in treating and/or inhibiting the progression of HPV related dysplasia of the cervix.
  • Topical compositions can be defined as compositions that are applied directly to an external body surface, which includes the skin and membranes such as the rectal lining, the lining of the mouth (buccal mucosa), the vaginal wall (vaginal mucosa), the covering of the eye (cornea and conjunctival membranes), the urethral membrane, the lining of the external ear, and the nasal mucous membrane.
  • an external body surface which includes the skin and membranes such as the rectal lining, the lining of the mouth (buccal mucosa), the vaginal wall (vaginal mucosa), the covering of the eye (cornea and conjunctival membranes), the urethral membrane, the lining of the external ear, and the nasal mucous membrane.
  • the composition once applied to an external body surface must remain at the site of application for a sufficient period in order for an efficacious dose of the active pharmaceutical ingredient to be delivered to the site of treatment.
  • the topical composition needs to remain at the site of application long enough for a sufficient proportion of the active pharmaceutical ingredient to be released from the composition and have the desired efficacious effect on the disease and/or disorder being treated.
  • Many topical compositions are unable to reside at the site of application for a sufficiently long period. For example, fluid like systems may readily flow off the applied area (streak).
  • the composition may be rapidly cleared away from the site of application by the body’s natural clearance mechanisms. This can result in variable amounts of the active pharmaceutical ingredient being dosed to the patient as well as under dosing in certain circumstances.
  • Topical compositions may contain excipients which increase the viscosity of the composition and thereby provide some improvement in residence time at the site of application. While this may reduce the risk of under dosing and/or the patient receiving variable amounts of the active pharmaceutical ingredient, it also typically results in the composition being more difficult to handle. For example, transferring the composition between vessels during the manufacturing process and/or during the filing of tubes/syringes suitable for patients and/or healthcare professionals to use. In addition, manipulating the tube/syringe during dispensing may be difficult if the composition is especially viscous.
  • Topical compositions can be presented in a number of formats. Suitable formats include ointments, creams, pastes, and gels. Some of these formats have water present within the composition. Topical compositions which contain water are not ideal for use with an active pharmaceutical ingredient which is prone to degradation by hydrolysis as this can result in a short shelf life of the pharmaceutical product and/or the requirement to store the composition in certain conditions in order to minimise degradation of the active
  • the present invention is based on work carried out by the inventors to formulate a composition comprising at least one active pharmaceutical ingredient for topical application. They have unexpectedly established that the use of a hydrophilic muco-adhesive agent in an anhydrous composition offers particular benefits.
  • the composition is anhydrous, thus the composition is particularly well suited to active pharmaceutical ingredients which are prone to degradation due to hydrolysis.
  • the site of application is a mucosal membrane. In a particular embodiment, the site of application is the vaginal wall.
  • ICC Invasive cervical cancer
  • CIN1 low-grade cervical intraepithelial neoplasia
  • CIN2/3 high-grade cervical intraepithelial neoplasia
  • Lesions can be screened for by cervical cytology testing where they are diagnosed (or graded) as either borderline atypical squamous cells of undetermined significance (ASCUS), low-grade squamous intraepithelial lesions (LSIL) or high-grade squamous intraepithelial lesions (HSIL).
  • ASCUS borderline atypical squamous cells of undetermined significance
  • LSIL low-grade squamous intraepithelial lesions
  • HSIL high-grade squamous intraepithelial lesions
  • cryotherapy has a higher failure rate compared to other treatment modalities.
  • HPV related cervical dysplasia An effective, inexpensive, non-surgical, self-applied treatment for HPV related cervical dysplasia would have great potential particularly in low resource settings. Furthermore, improved compliance with topical treatment would be enhanced, if the side effects are minimised.
  • WO2015/059485 which describes the protease inhibitors, lopinavir and ritonavir (which had previously been used as orally ingested medicaments for the clinical management of retroviral infections such as HIV) as being clinically useful for topical administration to tissues to prevent or treat malignancies caused by human papilloma virus.
  • KALETRA ® which is marketed by Abbott/ Abbvie for the treatment of HIV infections by oral administration
  • can be administered topically e.g. inserted into the vagina for treatment of the cervix
  • the prevention or treatment of cancerous conditions for the prevention or treatment of oncogenic viral infections and for the prevention or treatment of benign proliferative orders.
  • compositions of the invention offer significant benefits when compared to oral soft capsule compositions when topical formulation is required.
  • compositions comprising at least one active pharmaceutical ingredient and a hydrophilic muco-adhesive agent.
  • anhydrous composition for topical application comprising:
  • anhydrous composition transforms into a muco-adhesive composition
  • a method of manufacturing the anhydrous composition of the first aspect comprising the step of incorporating (e.g. by mixing) a hydrophilic muco-adhesive agent into the anhydrous composition.
  • an anhydrous composition according to the first aspect of the invention for use as a medicament.
  • the anhydrous composition is used as a medicament for treating and/or inhibiting the development of diseases and/or disorders.
  • the anhydrous composition is used as a medicament for treating and/or inhibiting the development or progression of cancers and/or benign proliferative disorders.
  • the anhydrous composition comprises an effective amount of the at least one active
  • the anhydrous composition comprises an effective amount of the at least one active pharmaceutical ingredient for treating and/or inhibiting the development or progression of a disease or disorder. In one embodiment, the anhydrous composition comprises an effective amount of the at least one active
  • the anhydrous composition comprises an effective amount of at least one active pharmaceutical ingredient for treating a Human Papilloma Virus (HPV) infection with or without attendant abnormal pathology.
  • HPV Human Papilloma Virus
  • the anhydrous composition is used as a medicament for treating and/or inhibiting the development of early stage neoplasias.
  • the anhydrous composition is used as a medicament for treating or preventing the development of HPV related cervical, vulval, vaginal, penile, anal, oral or laryngeal neoplasias and/or warts.
  • the anhydrous composition is for use as a medicament for treating or preventing the development of cervical neoplasias.
  • a method of treating and/or inhibiting the development or progression of diseases and/or disorders in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention.
  • a method of treating and/or inhibiting the development or progression of cancers and/or benign proliferative disorders in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention.
  • a method of treating or preventing the development of HPV related cervical, vulval, vaginal, penile, anal, oral or laryngeal neoplasias and/or warts in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention.
  • a method of treating or preventing the development of cervical neoplasias in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention.
  • the cancer or benign proliferative disorder is caused by a viral infection, such as by an oncogenic virus and in particular human tumour viruses such as HPV.
  • the invention concerns treating a subject having an HPV related dysplasia of the cervix comprising administering to said subject a therapeutically effective dose of the disclosed anhydrous compositions.
  • Figure 2 Oscillation frequency rheological response of a prototype vaginal ointment containing no HPMC in both the absence and presence of water
  • Figure 3 Oscillation frequency rheological response comparison of low shear and high- pressure homogenizing processes for a prototype vaginal ointment containing 1% w/w HPMC
  • Figure 4 Oscillation frequency rheological response of an HPH processed prototype vaginal ointment containing 1% w/w HPMC in both the absence and presence of water
  • human papilloma virus HPV
  • ASC-US Atypical squamous cells of undetermined significance
  • LSIL Low grade squamous intraepithelial lesion
  • HSIL High grade squamous intraepithelial lesion
  • Cervical intraepithelial neoplasia 1 CIN1
  • Cervical Intraepithelial neoplasia 2 CIN2
  • Cervical intraepithelial neoplasia 3 CIN3
  • Carcinoma in situ CIS
  • ICC Invasive Cervical Carcinoma
  • treating and like terms refer to reducing the severity and/or frequency of symptoms, eliminating symptoms and/or the underlying cause of said symptoms, reducing the frequency or likelihood of symptoms and/or their underlying cause, delaying, preventing and/or slowing the progression of diseases and/or disorders, such as cancers or benign proliferative disorders, and improving or remediating damage caused, directly or indirectly, by the diseases and/or disorders such as cancers or benign proliferative disorders.
  • anhydrous composition for topical application comprising:
  • anhydrous composition transforms into a muco-adhesive composition
  • anhydrous composition transforms into a muco-adhesive composition
  • anhydrous composition transforms into a muco-adhesive composition
  • anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient;
  • anhydrous composition transforms into a muco-adhesive composition
  • an anhydrous composition for topical application comprising:
  • the anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition; and wherein the anhydrous composition is a semi-solid at room temperature.
  • an anhydrous composition for topical application comprising:
  • anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient; and
  • the anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition; wherein the anhydrous composition is a semi-solid at room
  • the muco-adhesive composition is a semi-solid at room
  • an anhydrous composition for topical application comprising:
  • the anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition; wherein the anhydrous composition is an ointment; and wherein the muco-adhesive composition is a gel.
  • an anhydrous composition for topical application comprising:
  • anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient;
  • the anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition and wherein the muco-adhesive composition has an increase in adhesiveness compared to the anhydrous composition.
  • a site of application such as preferably a mucosal membrane
  • an anhydrous composition for topical application comprising:
  • the anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition and wherein the muco-adhesive composition provides prolonged retention of the at least one active pharmaceutical ingredient at the site of application.
  • a site of application such as preferably a mucosal membrane
  • an anhydrous composition for topical application comprising:
  • the anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition and wherein the muco-adhesive composition provides prolonged retention of the at least one active pharmaceutical ingredient at the site of application for at least 15 minutes, preferably at least 30 minutes, preferably at least 1 hour, preferably at least 90 minutes, preferably at least 2 hours, preferably at least 4 hours, preferably at least 6 hours, preferably at least 8 hours, preferably at least 10 hours, preferably at least 12 hours, or preferably at least 24 hours.
  • an anhydrous composition for topical application comprising:
  • the anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition and wherein the muco-adhesive composition has a greater viscosity than the anhydrous composition.
  • a site of application such as preferably a mucosal membrane
  • anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient;
  • anhydrous composition transforms into a muco-adhesive composition
  • an anhydrous composition for topical application comprising:
  • a hydrophilic muco-adhesive agent such as hydroxypropylmethylcellulose
  • an anhydrous composition for topical application comprising:
  • anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
  • a site of application such as preferably a mucosal membrane
  • the anhydrous composition comprises:
  • the anhydrous composition comprises:
  • anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
  • a site of application such as preferably a mucosal membrane
  • the anhydrous composition comprises:
  • anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
  • a site of application such as preferably a mucosal membrane
  • the anhydrous composition comprises:
  • anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
  • a site of application such as preferably a mucosal membrane
  • anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
  • a site of application such as preferably a mucosal membrane
  • the anhydrous composition comprises:
  • the anhydrous composition comprises:
  • anhydrous composition upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
  • a site of application such as preferably a mucosal membrane
  • an anhydrous composition for topical application comprising:
  • anhydrous composition transforms into a muco-adhesive composition
  • anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient;
  • anhydrous composition transforms into a muco-adhesive composition
  • hydrophilic muco-adhesive agent b. about 0.1 to about 10% by weight of hydrophilic muco-adhesive agent
  • hydrophilic muco-adhesive agent b. about 0.1 to about 10% by weight of hydrophilic muco-adhesive agent
  • anhydrous composition transforms into a muco-adhesive composition
  • an anhydrous composition for topical application comprising:
  • anhydrous composition transforms into a muco-adhesive composition
  • the anhydrous composition for topical application comprises: a. at least one active pharmaceutical ingredient;
  • anhydrous composition transforms into a muco-adhesive composition
  • hydroxypropylmethylcellulose e. about 0.1 to about 10% by weight of hydroxypropylmethylcellulose; f. about 0.05 to about 0.5% by weight of butylated hydroxytoluene; g. about 0.1 to about 10% by weight of mono diglyceride;
  • anhydrous composition transforms into a muco-adhesive composition
  • the muco-adhesive composition is a gel.
  • the muco-adhesive composition has an increase in adhesiveness and/or has a different rheological behaviour, such as an increase in viscosity, compared to the anhydrous composition.
  • this increase in adhesiveness and/or different rheological behaviour means that the anhydrous composition is easier to handle during its manufacturing and/or during filing of dosing apparatus, such as a syringe, but can still provide upon application a muco-adhesive composition that is retained at the site of application for a prolonged period.
  • the muco-adhesive composition has a greater viscosity than the anhydrous composition.
  • the muco-adhesive compositions advantageously provide prolonged retention of the at least one active pharmaceutical ingredient at the site of application. This ensures that the composition remains at the site of application long enough to allow a significant proportion of the active pharmaceutical ingredient to be released from the composition and have the desired efficacious effect on the disease and/or disorder being treated.
  • the skilled person would be aware that various in-vitro techniques can be employed to measure the release of the active pharmaceutical ingredient from the composition.
  • the amount of active pharmaceutical ingredient delivered through a membrane can be determined using, for example, Vertical Diffusion Studies (“Topical and Transdermal Drug Products, Pharmacopeial Forum, Vol. 35(3) [May- June 2009]).
  • the in-vitro release rate can be correlated to how the composition will perform in an in-vivo setting.
  • At least 50% by weight of the at least one active pharmaceutical ingredient is released from the composition, such as at least 60% by weight, such as at least 70% by weight, such as at least 80% by weight, such as at least 90% by weight, or such as at least 95% by weight within 2 hours, within 3 hours, within 4 hours, within 6 hours, or within 8 hours.
  • the anhydrous composition remains at the site of application for at least 15 minutes, such as at least 30 minutes, such as at least 1 hour, such as at least 90 minutes, such as at least 2 hours, such as at least 4 hours, such as at least 6 hours, such as at least 8 hours, such as at least 10 hours, such as at least 12 hours, or such as at least 24 hours.
  • the anhydrous composition remains at the site of application for up to 30 minutes, for up to 1 hour, for up to 90 minutes, for up to 2 hours, up to 4 hours, for up to 6 hours, for up to 8 hours, for up to 10 hours, for up to 12 hours, or for up to 24 hours.
  • a semi-solid is any material that can be present in a stationary material state until an external stress is applied resulting in flow of the material.
  • the physical properties of a semi solid are a mixture of a solid and liquid. It will be apparent to the skilled person that the pharmaceutical composition is in a form of a semi-solid, for example by testing the pharmaceutical composition using a rheometer. From rheometric testing of the
  • the yield stress is a direct measure for the initiation of material flow (solid state to fluid state) in response to an applied stress.
  • the storage modulus is a direct measure of a fluid’s solid-like behaviour.
  • the loss modulus is a direct measure of a fluid’s fluid-like behaviour.
  • Tan d is the ratio of the loss modulus to the storage modulus and is a unitless measure.
  • a tan d value of ⁇ 1 defines a material as largely solid like, while a tan d value >1 defines a material as predominantly fluid-like.
  • the rheological behaviour of the semi-solid can be tested according to the methodology described in Example 6
  • the anhydrous composition is a semi-solid at room temperature.
  • the muco-adhesive composition is a semi-solid at room temperature.
  • the anhydrous composition is a semi-solid at room temperature and the muco-adhesive composition is a semi-solid at room temperature.
  • the rheological behaviour of the anhydrous composition is different to the rheological behaviour of the muco-adhesive composition, for example, the compositions differ in respect to complex viscosity and/or storage modulus and/or loss modulus and/or tan delta.
  • Complex viscosity is defined as the frequency-dependent viscosity function determined for a viscoelastic fluid by subjecting it to oscillatory shear stress.
  • Storage modulus is a measure of the stored energy within a material and represents the elastic portion.
  • the loss modulus measures the energy dissipated as heat and represents the viscous portion.
  • Tan delta is the ratio of loss modulus to the storage modulus. Thus, a tan delta value of greater than 1 means that the composition has a greater loss modulus than storage modulus.
  • the complex viscosity of the muco-adhesive composition is greater than the complex viscosity of the anhydrous composition.
  • the muco-adhesive composition has a complex viscosity at least 50% greater than, such as at least 75% greater than, such as at least 100% greater than, or such as at least 110% greater than, the complex viscosity of the anhydrous composition and wherein the complex viscosity is measured at an angular frequency of 0.1 rad/s.
  • the anhydrous composition has a complex viscosity of between 1500 and 2000 cP.s, such as between 1600 and 1900 cP.s, or such as between 1700 and 1800 cP.s, and wherein the complex viscosity is measured at an angular frequency of 0.1 rad/s.
  • the muco-adhesive composition has a complex viscosity of between 3500 and 4000 cP.s, such as between 3600 and 3900 cP.s, or such as between 3800 and 3900 cP.s, and wherein the complex viscosity is measured at an angular frequency of 0.1 rad/s.
  • the anhydrous composition has a complex viscosity of between 1500 and 2000 cP.s, such as between 1600 and 1900 cP.s, or such as between 1700 and 1800 cP.s, and the muco-adhesive composition has a complex viscosity of between 3500 and 4000 cP.s, such as between 3600 and 3900 cP.s, or such as between 3800 and 3900 cP.s, and wherein the complex viscosity is measured at an angular frequency of 0.1 rad/s.
  • the storage modulus of the muco-adhesive composition is greater than the storage modulus of the anhydrous composition.
  • the muco- adhesive composition has a modulus of at least 100% greater than, such as at least 150% greater than, or such as at least 200% greater than, the storage modulus of the anhydrous composition and wherein the storage modulus is measured at an angular frequency of 0.1 rad/s.
  • the anhydrous composition has a storage modulus of between 50 and 150 Pa, such as between 75 and 125 Pa, or such as between 90 and 110 Pa, and wherein the storage modulus is measured at an angular frequency of 0.1 rad/s.
  • the muco-adhesive composition has a storage modulus of between 250 and 450 Pa, such as between 300 and 400 Pa, or such as between 300 and 350 Pa, and wherein the storage modulus is measured at an angular frequency of 0.1 rad/s.
  • the anhydrous composition has a storage modulus of between 50 and 150 Pa, such as between 75 and 125 Pa, or such as between 90 and 110 Pa
  • the muco- adhesive composition has a storage modulus of between 250 and 450 Pa, such as between 300 and 400 Pa, or such as between 300 and 350 Pa, and wherein the storage modulus is measured at an angular frequency of 0.1 rad/s.
  • the loss modulus of the muco-adhesive composition is at least 20% greater than, such as at least 30% greater than, or such as at least 40% greater than, the loss modulus of the anhydrous composition and wherein the loss modulus is measured at an angular frequency of 0.1 rad/s.
  • the anhydrous composition has a loss modulus of between 130 and 170 Pa, such as between 140 and 160 Pa, or such as between 140 and 150 Pa, and the loss modulus is measured at an angular frequency of 0.1 rad/s.
  • the muco-adhesive composition has a loss modulus of between 190 and 230 Pa, such as between 200 and 220 Pa, or such as between 205 and 215 Pa, and the loss modulus is measured at an angular frequency of 0.1 rad/s.
  • the anhydrous composition has a loss modulus of between 130 and 170 Pa, such as between 140 and 160 Pa, or such as between 140 and 150 Pa
  • the muco-adhesive composition has a loss modulus of between 190 and 230 Pa, such as between 200 and 220 Pa, or such as between 205 and 215 Pa, and the loss modulus is measured at an angular frequency of 0.1 rad/s.
  • the tan delta value of the muco-adhesive composition is at least 15% less than, such as at least 20% less than, or such as at least 25% less than, the tan delta value of the anhydrous composition.
  • the anhydrous composition has a tan delta value greater than 1, such as greater than 1.2, such as greater than 1.3, or such as about 1.4, and wherein the loss modulus and the storage modulus are measured at an angular frequency of 0.1 rad/s.
  • the muco-adhesive composition has a tan delta value less than 1 , such as less than 0 8, such as greater than 0.7, or such as about 0.7, and wherein the loss modulus and the storage modulus are measured at an angular frequency of 0.1 rad/s.
  • the anhydrous composition has a tan delta value greater than 1, such as greater than 1.2, such as greater than 1.3, or such as about 1.4, and the muco-adhesive composition has a tan delta value less than I, such as less than 0.8, such as greater than 0.7, or such as about 0.7, and wherein the loss modulus and the storage modulus are measured at an angular frequency of 0.1 rad/s.
  • the anhydrous composition comprises less than 1% by weight of water, such as less than 0.5% by weight, such as less than 0.2% by weight, such as less than 0.1% by weight, or such as less than 0.05% by weight.
  • the anhydrous composition is an ointment or a paste. In one embodiment, the anhydrous composition is an ointment. In one embodiment, the ointment comprises a hydrocarbon base.
  • the anhydrous composition is for intravaginal delivery. In one embodiment, the anhydrous composition is for intravaginal delivery using a syringe.
  • the anhydrous composition is for topical delivery of the at least one active pharmaceutical ingredient to the cervix (e.g. as an ointment or paste) for preventing the development of, or treating, cervical cancer (e.g. caused by high-risk types of HPV such as HPV16).
  • the cervix e.g. as an ointment or paste
  • cervical cancer e.g. caused by high-risk types of HPV such as HPV16.
  • the hydrophilic muco-adhesive agent only becomes active upon topical application of the anhydrous composition to a site of application, such as preferably a mucosal membrane.
  • the site of application is an environment comprising water. Environments comprising water activate the hydrophilic muco-adhesive agent resulting in the anhydrous composition transforming to a muco-adhesive composition and in particular embodiments becoming a gel.
  • the word gel describes the polymeric interaction between the hydrophilic muco-adhesive agent and the water.
  • the resulting gel enables the composition to be retained at the site of application for a prolonged period.
  • the site of application comprises a mucus membrane
  • the mucus membrane is selected from buccal mucosa, nasal mucosa, ocular mucosa, vaginal mucosa and rectal mucosa.
  • the mucus membranes provide an aqueous environment and as such activates the hydrophilic muco-adhesive agent upon application of the anhydrous composition to the site of application, resulting in the prolonged retention of the at least one active pharmaceutical ingredient at the site of application, for example, activation of the hydrophilic muco-adhesive agent results in the anhydrous composition transforming into a muco-adhesive agent and becoming a gel.
  • the hydrophilic muco-adhesive agent is present in the anhydrous composition at about 0.1 to about 20% by weight of the total anhydrous composition weight, such as about 0.1 to about 10% by weight, such as about 0.1 to about 5% by weight, such as about 0.2 to about 4% by weight, such as about 0.5 to about 3% by weight, such as about 0.5 to about 1.5% by weight, such as about 1% by weight, or such as 1% by weight.
  • the hydrophilic muco-adhesive agent is a non-ionic polymer or an ionic polymer.
  • the non-ionic polymer is a cellulose ether.
  • the hydrophilic muco-adhesive agent is a cellulose ether or an ionic polymer.
  • the cellulose ether is selected from methyl cellulose
  • the cellulose ether is hydroxypropylmethylcellulose.
  • the hydroxypropylmethylcellulose has a degree of methoxy substitution of between 19 and 24% by weight and a degree of hydroxypropyl substitution of between 4 and 12% by weight.
  • the ionic polymer is sodium polyacrylate.
  • the hydrophilic muco-adhesive has a viscosity of about 2000 to about 6000 cps, such as about 3000 to about 5000 cps, such as about 3500 to about 4500 cps, such as about 3800 to about 4200 cps, such as about 4000 cps, or such as 4000 cps.
  • the at least one active pharmaceutical ingredient is a solid at room temperature. In one embodiment, the at least one active pharmaceutical ingredient is synthetically prepared. In one embodiment, the at least one active pharmaceutical ingredient is not a fatty acid (free or bound state).
  • the at least one active pharmaceutical ingredient is present in a dissolved state in the anhydrous composition. In another embodiment, the at least one active pharmaceutical ingredient is present in a dispersed state in the anhydrous composition. In another embodiment, an amount of the least one active pharmaceutical ingredient is present in a dispersed state and an amount is present in a dissolved state in the anhydrous composition.
  • the active pharmaceutical ingredient is dissolved or dispersed within the anhydrous composition by the use of techniques such as optical microscopy using polarised light filters, differential scanning calorimetry or micro FTIR.
  • a placebo anhydrous composition i.e., a composition containing no active pharmaceutical ingredients
  • the crystalline active pharmaceutical ingredient will exhibit birefringence.
  • the spiked placebo composition can be used as a comparison standard in order to confirm there is no crystalline active pharmaceutical ingredient in the anhydrous composition, and thereby demonstrating the active pharmaceutical ingredient is dissolved within the anhydrous composition.
  • micro FTIR can be used to confirm the active
  • spectra obtained for an anhydrous composition spiked with the active pharmaceutical ingredient and for the anhydrous composition can be compared and used to demonstrate the active pharmaceutical ingredient is dissolved in the anhydrous composition.
  • the at least one active pharmaceutical ingredient is stable within the anhydrous composition.
  • the compositions of the invention are particularly suitable for active pharmaceutical ingredients used in the composition that are prone to chemical or physical degradation.
  • the active pharmaceutical is prone to degradation due to hydrolysis.
  • the active pharmaceutical ingredient used in the composition is prone to degradation due to oxidation.
  • the active pharmaceutical ingredient used in the composition is prone to degradation which is accelerated by heat.
  • the active pharmaceutical ingredient used in the composition is prone to physical form changes, e.g. solid-state polymorphic transitions.
  • the at least one active pharmaceutical ingredient is stable within the anhydrous composition for at least 3 months, such as at least 6 months, such as at least 9 months, such as at least 12 months, such as at least 18 months, such as at least 24 months, or such as at least 36 months, at a temperature of 5°C, 25°C, 30°C or at 40°C, and/or at a relative humidity of 60%, 65% or 75% RH.
  • the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition. In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed at room temperature. In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed at elevated temperatures, such as >30°C, such as >40°C, such as >50°C, such as >60°C, such as >65°C such as about 70°C.
  • the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed under an inert atmosphere, such as a vacuum, such as a vacuum at about -0.5 bar. In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed under an inert atmosphere, such as a vacuum, such as a vacuum at about -0.5 bar. In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed under an inert atmosphere, such as a vacuum, such as a vacuum at about -0.5 bar. In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed under an inert atmosphere, such as a vacuum, such as a vacuum at about -0.5 bar. In one embodiment, the at least one active pharmaceutical ingredient is stable within
  • pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed in the absence of water, for example wherein the process is performed under an insert atmosphere, such as a vacuum, such as a vacuum at about -0.5 bar.
  • the at least one active pharmaceutical ingredient is present in the anhydrous composition between about 0.001 and about 50% by weight of the total anhydrous composition weight, such as between about 0.01 and about 50% by weight, such as between about 0.001 and about 5% by weight, such as between about 0.1 and about 25% by weight, such as between about 0.5 and about 15% by weight, such as between about 0.5 and about
  • 0.1 and about 2.5% by weight such as between about 0.2 and about 2% by weight, such as between about 0.2 and about 1.5% by weight, such as between about 0.4 and about 1.5% by weight, such as between about 0.4 and about 0.6% by weight, such as about 0.5% by weight, such as 0.5% by weight, such as between about 0.5 and about 0.7% by weight, such as about 0.6% by weight, such as 0.6% by weight, such as between about 0.9 and about 1.1% by weight, such as about 1% by weight, such as 1% by weight, such as 1% by weight, such as between about 1.2 and about 1.4% by weight, such as about 1.3% by weight, such as 1.3% by weight, such as between about 2 and about 20% by weight, such as between about 2 and about 15% by weight, such as between about 4 and about 12% by weight, such as between about 4 and about 6% by weight, such as about 5% by weight, such as 5% by weight, such as between about 5 and about 7% by weight, such as about 6% by weight, such as 6% by weight,
  • the at least one active pharmaceutical ingredient is classified as a Biopharmaceutics Classification System (BCS) Class II or a BCS Class IV active
  • a BCS class II active pharmaceutical ingredient is classed as an active ingredient having a high permeability and a low solubility.
  • a BCS class IV active pharmaceutical ingredient is classed as an active ingredient having a low permeability and a low solubility.
  • the at least one active pharmaceutical ingredient is selected from a protease inhibitor, a retinoid, a vitamin D analog, an antileprosy active pharmaceutical ingredient, a calcineurin inhibitor, a cannabinoid, a 5 alpha-reductase inhibitor, an androgen receptor inhibitor, a peroxisome proliferator activated receptor activator, an antihistamine, a chloride channel activator, a tyrosine kinase inhibitor, a hormone, a protease inhibitor, and a mTOR kinase inhibitor.
  • the at least one active pharmaceutical ingredient is selected from abacavir, efavirenz, enfuvirtide, nevirapine, ritonavir, lopinavir, tenofovir, adefovir, entecavir, ribavirin, acyclovir, famciclovir, penciclovir, valacyclovir, cidofovir, ganciclovir,
  • valganciclovir oseltamivir, zanamivir, amprenavir, bexarotene, calcifediol, calcitriol, clofazimine, cyclosporin A, doxercalciferol, dronabinol, dutasteride, enzalutamide, fenofibrate, isotretinoin, loratadine, lubiprostone, nintedanib, paricalcitol, progesterone, saquinavir, sirolimus, tipranavir, tretinoin, atorvastatin, carvedilol, itraconazole, ketoprofen, and simvastatin.
  • the at least one active pharmaceutical ingredient is selected from abacavir, efavirenz, enfuvirtide, nevirapine, ritonavir, lopinavir, tenofovir, adefovir, entecavir, ribavirin, acyclovir, famciclovir, penciclovir, valacyclovir, cidofovir, ganciclovir,
  • valganciclovir oseltamivir and zanamivir.
  • the anhydrous composition further comprises an active pharmaceutical ingredient synergist.
  • the active pharmaceutical ingredient synergist is a HIV protease enzyme inhibitor.
  • the at least one active pharmaceutical ingredient is a HIV protease enzyme inhibitor.
  • the HIV protease enzyme inhibitor is selected from lopinavir and ritonavir.
  • Lopinavir (CAS# 192725-17-0) is a protease inhibitor chemically designated as [1S- [lR*(R*), 3R*, 4R*]]-N-[4-[(2,6-dimethylphenoxyacetyl]amino]-3-hydroxy-5-phenyl-l- (phenylmethyl)pentyl]tetrahydro-alpha-(l -methylethyl)-2-oxo- 1 (2H)-pyrimidineacetamide. It has the molecular formula C37H48N405 and a molecular weight of 628.80.
  • Ritonavir (CAS# 155214-67-5) is a protease inhibitor chemically designated as 10- Hydroxy-2-methyl-5-(l-methylethl)-l-[2-(l-methylethyl)-4-thiazolyl]-3,6-dioxo-8,
  • the anhydrous composition comprises lopinavir and ritonavir.
  • the molar ratio of lopinavir to ritonavir present in the composition is between about 1 : 10 and about 18: 1, such as between about 1 : 10 and about 15: 1, such as between about 1 :5 and about 15: 1, such as between about 1 : 1 and about 15: 1, such as between about 2: 1 and about 15: 1, such as between about 4: 1 and about 15: 1, such as between about
  • lopinavir has a molecular weight of 628.8 daltons and ritonavir has a molecular weight of 720.95 daltons. Accordingly molar ratios and w/w ratios will not be the same and a factor of 0.872 should be applied when converting molar ratios to w/w.
  • lopinavir is present in the anhydrous composition at about 0.01 to about 50% of the total anhydrous composition weight, such as about 0.1 to about 25% by weight, such as about 2 to about 20% by weight, such as about 2 to about 15% by weight, such as about 4 to about 12% by weight, such as about 4 to about 6% by weight, such as at about 5% by weight, such as 5% by weight, such as about 5 to about 7% by weight, such as about 6% by weight, such as 6% by weight, such as about 9 to about 11% by weight, such as at about 10% by weight, such as 10% by weight, such as about 11 to about 13% by weight, such as about 12% by weight, or such as 12% by weight.
  • ritonavir is present in the anhydrous composition at about 0.001 to about 5% by weight of the total anhydrous composition weight, such as about 0.01 to about 2.5% by weight, such as about 0.1 to about 2.5% by weight, such as about 0.2 to about 2% by weight, such as about 0.2 to about 1.5% by weight, such as about 0.4 to about 1.5% by weight, such as about 0.4 to about 0.6% by weight, such as at about 0.5% by weight, such as at 0.5% by weight, such as about 0.9 to about 1.1% by weight, such as about 1% by weight, such as 1% by weight, such as about 1.2 to 1.4% by weight, such as about 1.3% by weight, such as 1.3% by weight, such as about 0.5 to about 0.7% by weight, such as about 0.6% by weight, or such as 0.6% by weight.
  • the anhydrous composition further comprises a solvent for the at least one active pharmaceutical ingredient.
  • the solvent is selected from an unsaturated free fatty acid, PEG castor oil, diethylene glycol, monoethyl ether, propylene glycol, polyethylene glycol, and a medium chain triglyceride.
  • Fatty acids are usually derived from triglycerides or
  • Glycerol has three hydroxyl functional groups, which can be esterified with one, two, or three fatty acids to form mono-, di-, or triglycerides respectively.
  • Phospholipid molecules consist of two hydrophobic fatty acid“tails” and a hydrophilic“head” consisting of a phosphate group. These two components are joined together by a glycerol molecule.
  • Both triglyceride and phospholipids comprise fatty acids in a bound state.
  • free fatty acids are fatty acids which are not bound, that is they are not esterified.
  • An unsaturated free fatty acid is a free fatty acid wherein there is at least one double bond between carbon atoms in the fatty acid.
  • the solvent is an unsaturated free fatty acid.
  • the unsaturated free fatty acid is selected from oleic acid, linoleic acid, alpha-linoleic acid, palmitoleic acid, gondoic acid, and ricinoleic acid.
  • the unsaturated free fatty acid is oleic acid.
  • the unsaturated fatty acid present within the composition, at least 90% by weight, such as at least 95% by weight, such as at least 98% by weight, such as at least 99% by weight, or such as at least 99.5% by weight, is in the free form, i.e., not esterified or bound to other components such as glycerol.
  • the free fatty acid content can be measured by reacting the free fatty acid with a chromogeneous compound, thus changing the frequency that the chromogeneous compound absorbs electromagnetic radiation.
  • the concentration of the chromogeneous compound reacted can be determined by monitoring the chromogeneous compound using a suitable wavelength which in turn can be used to determine the free fatty acid content in the sample.
  • free fatty acids products that are commercially available may contain small amounts of other free fatty acids.
  • oleic acid typically contains 7-12% saturated free fatty acids, such as stearic and palmitic acid, together with other unsaturated free fatty acids, such as linoleic acid (Handbook of Pharmaceutical Excipients, 2 nd Edition, see entry for Oleic acid).
  • saturated free fatty acid or unsaturated free fatty acid are to be understood as meaning the saturated free fatty acid or the unsaturated free fatty acid are of Pharmacopeia grade, such as the ETS Pharmacopeia and/or the British
  • the saturated free fatty acid or unsaturated free fatty acid may contain small amounts of other free fatty acids.
  • the unsaturated free fatty acid is not in the form of a triglyceride or polysorbate.
  • the at least one active pharmaceutical ingredient has a solubility measured at ambient temperature of at least 1% w/v in the solvent, such as at least 5% w/v in the solvent, such as at least 10% w/v in the solvent, such as at least 12% w/v in the solvent, such as at least 15% w/v in the solvent, or such as at least 18% w/v in the solvent.
  • the solvent is present in the anhydrous composition at a level of at least 25% by weight of the total anhydrous composition weight, such as at least 35% by weight, such as at least 45% by weight, such as about 25 to about 75% by weight, such as about 55 to about 75% by weight, such as about 45 to about 75% by weight, such as about 50 to about 70% by weight, such as about 55 to about 70% by weight, such as about 55 to about 65% by weight, such as about 50 to about 60% by weight, such as about 25 to about 60% by weight, or such as about 50 to about 65% by weight.
  • the solvent is present in the anhydrous composition at a level of about 55% by weight of the total anhydrous composition weight, such as about 57% by weight, such as about 58% by weight, such as about 60% by weight, such as about 62% by weight, such as about 63% by weight, such as about 65% by weight, such as about 67% by weight, or such as about 68% by weight.
  • excipients may be included in the composition according to the present invention providing that inclusion of such excipients does not unacceptably impact the ability of the composition, upon topical administration to a site of application, to transform into a muco-adhesive composition.
  • the anhydrous composition further comprises a thickener.
  • a thickener is an excipient which when added to a mixture increases the viscosity of the mixture and confers the anhydrous composition with greater physical stability and/or control during delivery of the active pharmaceutical ingredient to the site of application.
  • the thickener is selected from mono di glyceride, ceresin wax, and hydrogenated vegetable oil or a combination thereof.
  • the anhydrous composition comprises mono di glyceride, ceresin wax, and hydrogenated vegetable oil.
  • the anhydrous composition further comprises a stiffening agent.
  • the stiffening agent is an excipient used to stiffening the composition so that the anhydrous composition is a semi-solid at room temperature.
  • the stiffening agent is a solid at room temperature.
  • the stiffening agent is a saturated free fatty acid, such as a C10-C38 saturated free fatty acid, such as a C16-C22 saturated free fatty acid.
  • a saturated free fatty acid is a free fatty acid (i.e., the fatty acid is not bound to another molecule, such as glycerol) wherein there are no double bonds between the carbon atoms in the fatty acid.
  • the stiffening agent is selected from capric acid, undecylic acid, lauric acid, tridecylic acid, myristic acid, pentadecylic acid, palmitic acid, margaric acid, stearic acid, nonadecylic acid, arachidic acid, heneicosylic acid, behenic acid, tricosylic acid, lignoceric acid, pentacosylic acid, cerotic acid, heptacosylic acid, montanic acid, nonacosylic acid, melissic acid, henatriacontylic acid, lacceroic acid, psyllic acid, geddic acid, ceroplastic acid, hexatriacontylic acid, heptatriacontanoic acid and octatriacontanoic acid.
  • the stiffening agent is stearic acid.
  • Stearic acid is a straight chained saturated fatty acid with an 18-carbon chain.
  • the total saturated fatty acid (bound and free form saturated fatty acid) present within the composition at least 90% by weight, such as at least 95% by weight such as at least 98% by weight, such as at least 99% by weight, or such as at least 99.5% by weight, is in the free form, i.e., not esterified or bound to other components such as glycerol.
  • the free fatty acid content can be measured by reacting the free fatty acid with a chromogeneous compound, thus changing the frequency that the chromogeneous compound absorbs electromagnetic radiation.
  • the concentration of the chromogeneous compound reacted can be determined by monitoring the chromogeneous compound using a suitable wavelength which in turn can be used to determine the free fatty acid content in the sample.
  • the saturated free fatty acid is not in the form of a triglyceride or polysorbate.
  • the total fatty acid (unsaturated and saturated fatty acids in the bound and free states) present within the composition at least 90% by weight, such as at least 95% by weight, such as at least 98% by weight, such as at least 99% by weight, or such as at least 99.5% by weight, is in the free form, i.e., not esterified or bound to other components such as glycerol.
  • the free fatty acid content can be measured by reacting the free fatty acid with a chromogeneous compound, thus changing the frequency that the chromogeneous compound absorbs electromagnetic radiation.
  • the concentration of the chromogeneous compound reacted can be determined by monitoring the chromogeneous compound using a suitable wavelength which in turn can be used to determine the free fatty acid content in the sample.
  • the saturated free fatty acid and the unsaturated free fatty acid are not in the form of a triglyceride or polysorbate.
  • the anhydrous composition further comprises oleic acid and stearic acid. In one embodiment, the anhydrous composition further comprises oleic acid, stearic acid, and hydroxypropylmethylcellulose.
  • the amount of thickener present in the anhydrous composition is at least about 0.1% of the total anhydrous composition weight, such as at least 1% by weight, such as at least about 3% by weight, such as about 0.1 to about 40% by weight, such as about 3 to about 40% by weight, such as about 3 to about 35% by weight, such as about 5 to about 21% by weight, such as about 4 to about 12% by weight, such as about 4 to about 7% by weight, such as about 0.1 to about 10% by weight, such as about 0.1 to about 20% by weight, such as about 5% by weight, such as 5% by weight, such as about 6% by weight, such as 6% by weight, such as about 8 to about 12% by weight, such as about 10% by weight, such as 10% by weight, such as about 10 to about 35% by weight, such as about 15 to about 35% by weight, such as about 20 to about 30% by weight, such as about 21% by weight, or such as 21% by weight.
  • the ceresin wax is present in the anhydrous composition at about 0.1 to about 30% by weight of the total anhydrous composition weight, such as about 0.1 to about 10% by weight, such as about 1 to about 10% by weight, such as about 4 to about 8% by weight, such as about 5 to about 7% by weight, such as about 5% by weight, such as about 6% by weight, or such as 6% by weight.
  • the hydrogenated vegetable oil is present in the anhydrous composition at about 0.1 to about 30% by weight of the total anhydrous composition weight, such as about 0.1 to about 20% by weight, such as about 5 to about 15% by weight, such as about 8 to about 12% by weight, such as about 9 to about 11% by weight, such as about 10% by weight, or such as 10% by weight.
  • the stiffening agent is present in the anhydrous composition at about 0.1 to about 65% by weight of the total anhydrous composition weight, such as about 3 to about 60% by weight, such as about 3 to about 50% by weight, such as about 3 to about
  • 40% by weight such as about 3 to about 35% by weight, such as about 3 to about 6% by weight, such as about 4 to about 5% by weight, such as about 4.5% by weight, such as 4.5% by weight, such as about 15 to about 25% by weight, such as about 18 to about 22% by weight, such as about 20% by weight, such as about 20 to about 30% by weight, such as about 24 to about 30% by weight, such as about 26% by weight, such as about 25 to about 35% by weight, such as about 28 to about 32% by weight, such as about 30% by weight, such as 30% by weight, such as about 32% by weight, or such as about 34% by weight.
  • the stearic acid is present in the anhydrous composition at about 0.1 to about 65% by weight of the total anhydrous composition weight, such as about 3 to about 40% by weight, such as about 3 to about 35% by weight, such as about 3 to about 6% by weight, such as about 4 to about 5% by weight, such as about 4.5% by weight, such as 4.5% by weight, such as about 15 to about 25% by weight, such as about 18 to about 22% by weight, such as about 20% by weight, such as about 20 to about 30% by weight, such as about 24 to about 30% by weight, such as about 26% by weight, such as about 25 to about 35% by weight, such as about 28 to about 32% by weight, such as about 30% by weight, such as 30% by weight, such as about 32% by weight, or such as about 34% by weight.
  • the anhydrous composition further comprises a blending agent.
  • a blending agent is an agent which promotes uniformity within the anhydrous composition, for example it promotes uniformity for organoleptic attributes within the anhydrous composition.
  • the blending agent is selected from polyoxy 100 stearate and glycerol monooeleate. In one embodiment, the composition comprises polyoxy 100 stearate and glycerol monooeleate.
  • the blending agent is present in the anhydrous composition at at least 0.1% by weight of the total anhydrous composition weight, such as at least 1% by weight, such as at least 1.5% by weight, such as about 0.1 to about 48% by weight, such as about 0.1 to about 38% by weight, such as about 0.1 to about 10% by weight, such as 0.1 to about 3% by weight, such as about 1 to about 10% by weight, such as about 2 to about 5% by weight, such as about 2% by weight, such as 2% by weight, such as about 3% by weight, such as 3% by weight, such as about 5% by weight, or such as 5% by weight.
  • the polyoxy 100 stearate is present in the anhydrous composition at about 0.1 to about 38% by weight of the total anhydrous composition weight, such as at about 0.1 to about 10% by weight, such as about 0.5 to about 5% by weight, such as about 1 to about 3% by weight, such as about 2% by weight, or such as 2% by weight.
  • the glycerol monooleate is present in the anhydrous composition at about 0.1 to about 10% by weight of the total anhydrous composition weight, such as about 0.5 to about 5% by weight, such as about 2 to about 4% by weight, such as about 3% by weight, or such as 3% by weight.
  • the anhydrous composition comprises an antioxidant.
  • the antioxidant is butylated hydroxytoluene.
  • the antioxidant is present in the anhydrous composition at about 0.05 to about 0.5% by weight of the total anhydrous composition weight, such as about 0.1 to about 0.3% by weight, such as about 0.2% by weight, such as 0.2% by weight, such as about 0.1% by weight, or such as 0.1% by weight.
  • the anhydrous composition further comprises oleic acid, stearic acid, mono di glyceride, ceresin wax, hydrogenated vegetable oil, polyoxyl 100 stearate, glycerol monooleate, and butylatedhydroxytoluene.
  • the anhydrous composition is for use as a medicament.
  • the anhydrous composition is for use as a medicament wherein the anhydrous composition is applied topically. In one embodiment, the anhydrous composition is for use as a medicament wherein the anhydrous composition is topically applied to the part of the body in need of the treatment. In one embodiment, the anhydrous composition is applied topically to an environment comprising water. In one embodiment, the anhydrous composition is topically applied to skin. In one embodiment, the anhydrous composition is topically applied to mucosa. In one embodiment, the anhydrous composition is for use as a medicament wherein the anhydrous composition is topically applied to the cervix. This is particularly advantageous as the patient can self-administer the anhydrous composition and/or treatment with the anhydrous composition negates the need of treatment by surgery.
  • the anhydrous compositions are useful in the treatment and/or prevention of diseases and/or disorders. In one embodiment, the anhydrous compositions are useful in the treatment of benign proliferative disorders.
  • the anhydrous compositions are useful in the treatment of cancer and particularly useful for preventing the development of cancers. Accordingly, normal subjects (i.e. subjects with no detectable cancer), subjects with pre-malignant cells or particularly cancer prone subjects may be treated by topical administration of the anhydrous compositions according to the invention with a view to preventing the development of cancer.
  • the anhydrous compositions are pastes or ointments and may be applied directly to the cervix by techniques known to the art.
  • compositions comprising lopinavir and ritonavir for use as a medicament in the treatment of cancer or benign proliferative disorders (e.g. warts) or in the prevention of the development of cancer.
  • benign proliferative disorders e.g. warts
  • the invention may be applied to a wide range of cancers such as ovarian carcinoma, breast carcinoma, lung carcinoma, uterine carcinoma, cervical carcinoma and thyroid carcinoma. It is also applicable to cancer prone conditions.
  • the invention is applicable particularly, but by no means exclusively, to pre-cancerous conditions and cancers caused by oncogenic viruses, e.g. high-risk or even low-risk forms of human papilloma viruses (HPVs).
  • HPVs human papilloma viruses
  • compositions may be administered to treat, and particularly prevent, the development of cervical cancer.
  • the inhibitors are used to treat, or prevent the development of cervical cancers caused by HPV (particularly high-risk types of HPV such as HPV16 or HPV 18).
  • compositions may be used to prevent or treat cancer as a monotherapy (i.e.
  • anhydrous composition comprising two or more active
  • cancer therapy e.g. chemotherapeutic agents, radiotherapy.
  • compositions are used to treat humans. However, it will be appreciated that the compositions may also have some veterinary use.
  • the amount of the at least one active pharmaceutical ingredient required is determined by biological activity and bioavailability, which in turn depends, in part, on the precise mode of administration, the physicochemical properties of the anhydrous composition employed, and whether the anhydrous compositions are being used as a monotherapy or in a combined therapy with other oral or topical medicines.
  • the at least one active pharmaceutical ingredient could be topically applied in addition to oral dosing of the same compounds or other active pharmaceutical ingredient(s).
  • the frequency of administration will also be influenced by the abovementioned factors and particularly the half-life of the active pharmaceutical ingredients within the subject being treated.
  • Daily doses may be given as a single administration. Alternatively, administration may be twice or more times during a day.
  • the anhydrous compositions may be topically administered at least once a day, such as once a day, or such as twice a day.
  • Optimal dosages to be administered may be determined by those skilled in the art, and will vary with the strength of the preparation, the mode of administration, and the
  • Suitable amounts of the at least one active pharmaceutical ingredient to be given as a daily dose are of about 0.01 mg to about 10 g, such as about 0.1 mg to about 10 g, such as about 1 mg to about 5g, such as about 1 mg to about 1 g, such as about 5 mg to about 2 g, such as about 10 mg to about 1 g, such as about 5 mg to about 500 mg, such as about 10 mg to about 500 mg, such as about 10 mg to about 400 mg, such as about 5 mg to about 200 mg, such as about 5 mg to about 50 mg, such as about 10 mg to about 40 mg, such as about 20 mg to about 40 mg, such as about 25 mg to about 35 mg, such as about 27 mg to about 32 mg, such as about 29 mg, such as 29 mg, such as about 28.7 mg, such as 28.7 mg, such as about 15 mg to about 35 mg, such as about 20 mg to about 30 mg, such as about 23 mg to about 27 mg, such as about 25 mg, such as 25 mg, such as about 5 mg to about 25 mg, such
  • about 10 mg to about 400 mg such as about 100 mg to about 400 mg, such as about 200 mg to about 400 mg, such as about 250 mg to about 350 mg, such as about
  • about 300 mg of lopinavir and about 29 mg ritonavir per day may be administered to the cervix of a woman.
  • about 300 mg of lopinavir and about 25 mg ritonavir per day may be administered to the cervix of a woman.
  • about 150 mg of lopinavir and about 14 mg ritonavir per day may be administered to treat the cervix of a woman.
  • about 150 mg of lopinavir and about 13 mg ritonavir per day may be administered to treat the cervix of a woman.
  • anhydrous composition may be administered to a subject per day.
  • dosage forms may comprise about 300 mg of lopinavir and about 29 mg of ritonavir; or about 150 mg of lopinavir and about 14 mg of ritonavir.
  • Such dosage forms may comprise about 300 mg of lopinavir and about 25 mg of ritonavir; or about 150 mg of lopinavir and about 12.5 mg of ritonavir.
  • anhydrous composition disclosed in Table 3 or Table 4 is administered to the cervix by a syringe applicator as a once per day application (preferably in the evening before retiring for the night).
  • the anhydrous composition may be administered to a subject for as long as treatment is required.
  • the length of time for which treatment will be required will depend upon the exact condition being treated or prevented and its severity. A skilled person will appreciate that treatment should be maintained in view of a number of factors which will include any requirement to eradicate the disease or disorder.
  • a course of treatment may be for 2 - 4 weeks, 7-21 days or for about 14 days. After this time a clinician may assess whether the course of treatment has been successful. A decision may then be made whether or not to continue treatment.
  • a treatment regimen may be for about 14 - 21 days and can be administered between menses.
  • a clinician may elect to stop topical treatment of the cervix during menses and recommence a new course of treatment in the next menstrual cycle.
  • a treatment regimen can be: (1) 14 - 21 days of administration; (2) followed by 1 - 14 days without treatment (during which menses may occur if treating the cervix); and (3) a further cycle of 14 -21 days of treatment if this is considered medically necessary.
  • the anhydrous compositions may be used to treat female subjects having an HPV related dysplasia of the cervix.
  • HPV related pre-invasive lesions include high grade squamous intraepithelial lesion (HSIL), atypical squamous cells of undetermined significance (ASCUS), and low grade squamous intraepithelial lesion (LSIL).
  • HPV related cancers include, for example, cervical
  • CIN intraepithelial neoplasia
  • ICC invasive cervical cancer
  • the disclosed methods and treatment regimens can be used to treat HPV related dysplasia.
  • the disclosed methods and treatment regimens can be used to treat HSIL.
  • the disclosed methods and treatment regimens can be used to treat ASCUS.
  • the disclosed methods and treatment regimens can be used to treat LSIL.
  • the disclosed methods and treatment regimens can be used to treat CIN.
  • the disclosed methods and treatment regimens can be used to treat ICC.
  • the disclosed methods and treatment regimens can be used to inhibit the progression of HPV related dysplasia.
  • the disclosed methods and treatment regimens can be used to inhibit the progression of HSIL.
  • the disclosed methods and treatment regimens can be used to inhibit the progression of ASCUS. In other aspects, the disclosed methods and treatment regimens can be used to inhibit the progression of LSIL. In other aspects, the disclosed methods and treatment regimens can be used to inhibit the progression of CIN. In yet other embodiments, the disclosed methods and treatment regimens can be used to inhibit the progression of ICC.
  • a method of treating a patient having an HPV related dysplasia of the cervix comprising administering intravaginally to said patient a therapeutically effective dose of an anhydrous composition according to the first aspect.
  • the anhydrous composition reduces the severity of the HPV related dysplasia. In one embodiment, the severity of the HPV is reduced from CIN3 to CIN2, from CIN3 to CIN1, from CIN3 to HPV negative, from CIN2 to CIN1, from CIN2 to HPV negative, or from CIN1 to HPV negative.
  • the patient has a cervical cytology of high grade squamous intraepithelial lesion (HSIL), atypical squamous cells of undetermined significance (ASCUS), or low grade squamous intraepithelial lesion (LSIL).
  • HSIL high grade squamous intraepithelial lesion
  • ASCUS atypical squamous cells of undetermined significance
  • LSIL low grade squamous intraepithelial lesion
  • the anhydrous composition reduces the cervical cytology from HSIL to a normal cytology, from HSIL to ACSUS, from HSIL to LSIL, from ACSUS to a normal cytology, or from LSIL to a normal cytology.
  • the composition induces apoptosis of HPV infected cells.
  • a method of manufacturing the anhydrous composition of the first aspect comprising the step incorporating (e.g. mixing) a hydrophilic muco-adhesive agent into the anhydrous composition.
  • a method of manufacturing the anhydrous composition of the first aspect comprising the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition.
  • the step of mixing uses a low shear mixing process or a high pressure homogenizing process.
  • using high pressure homogenizing (HPH) in the mixing step results in greater dispersion of the muco-adhesive agent within the anhydrous composition and a decrease in particle size resulting in the HPH generated anhydrous composition having an increase in product surface area and an increase in viscosity relative to the low shear generated anhydrous composition.
  • HPH high pressure homogenizing
  • the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition is performed at room temperature.
  • This has the advantage of the process being particularly well suited to active pharmaceutical ingredients which are unstable when exposed to temperatures above room temperature, for example >30°C, >40°C, >50°C, and >60°C.
  • the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition is performed at a temperature greater than 30°C, such as greater than 40°C, such as greater than 50°C, such as greater than 60°C, such as greater than 65°C, such as about 70°C.
  • the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition is performed at a temperature greater than the melting point of the thickener and/or stiffening agent, such as greater than 50°C, such as greater than 60°C, such as greater than 65°C, such as about 70°C.
  • the method of manufacturing the anhydrous composition further comprises the step of mixing the at least one active pharmaceutical ingredient, a solvent and a stiffening agent together.
  • the step of mixing the at least one active pharmaceutical ingredient, a solvent and a stiffening agent together uses low shear mixing.
  • the step of mixing the at least one active pharmaceutical ingredient, a solvent and a stiffening agent together is performed at a temperature greater than the melting point of the stiffening agent.
  • the step of mixing the at least one active pharmaceutical ingredient, a solvent and a stiffening agent together is performed at a temperature greater than the melting point of the stiffening agent and until a clear and transparent melt is obtained.
  • the method of manufacturing the anhydrous composition further comprises the step of mixing the at least one active pharmaceutical ingredient, a solvent a thickener, and a stiffening agent together.
  • the step of mixing the at least one active pharmaceutical ingredient, a solvent, a thickener, and a stiffening agent together uses low shear mixing. In one embodiment, the step of mixing the at least one active pharmaceutical ingredient, a solvent, a thickener, and a stiffening agent together is performed at a temperature greater than the melting point of the thickener and/or stiffening agent.
  • the step of mixing the at least one active pharmaceutical ingredient, a solvent, a thickener, and a stiffening agent together is performed at a temperature greater than the melting point of the thickener and/or stiffening agent and until a clear and transparent melt is obtained.
  • the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition using a low shear mixing process is performed until the muco- adhesive agent is dispersed within the anhydrous composition.
  • the process further comprises a cooling step.
  • the cooling step comprises cooling the mixture to a temperature less than 70°C, such as less than 60°C, such as less than 50°C, such as about 45°C.
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • step ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing.
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • step iii cooling the mixture from step ii. while maintaining low shear mixing.
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • step iii cooling the mixture from step ii. while maintaining low shear mixing.
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • step iii cooling the mixture from step ii. while maintaining low shear mixing.
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • step ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing.
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • step ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing.
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • step iii cooling the mixture from step ii. while maintaining low shear mixing.
  • the method of manufacturing the anhydrous composition of the first aspect comprises:
  • the method of manufacture is performed under an inert atmosphere.
  • the inert atmosphere is provided by a vacuum.
  • the vacuum is about -0.5 bar. Performing the method of manufacture under an inert atmosphere has the advantage that any components of the anhydrous composition, such as the at least one active pharmaceutical ingredient, that are prone to degradation, such as oxidative or hydrolysis degradation, are less likely to degrade during the manufacturing process.
  • a syringe comprising the anhydrous composition of the first aspect.
  • the syringe is for vaginal dosing of the anhydrous composition.
  • kits of parts comprising instructions for use and a syringe comprising the anhydrous composition of the first aspect.
  • a kit of parts comprising instructions for use and a syringe comprising the anhydrous composition, the syringe for vaginal dosing of the anhydrous composition.
  • a tube comprising the anhydrous composition of the first aspect.
  • the tube is an aluminium tube.
  • kits of parts comprising instructions for use and a tube comprising the anhydrous composition of the first aspect.
  • the tube is an aluminium tube.
  • API active pharmaceutical ingredient
  • Lopinavir/Ritonavir is thus one that excludes heat, water, and/or oxygen.
  • Formulation 3b Binary mixture (oleic acid and stearic acid); placebo
  • Formulation 3c Mixture (oleic acid, stearic acid and thickeners); full strength - molar ratio of lopinavir to ritonavir 12:1
  • vaginal dosage form full strength active batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 3:- i. Add into the mixer the following materials - 3,4,5,6,7,8,9,1,10,11
  • Formulation 3d Mixture (oleic acid, stearic acid and thickeners); half strength - molar ratio of lopinavir to ritonavir 12:1
  • vaginal dosage form half strength active batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 4:- i.
  • Formulation 3e Mixture (oleic acid, stearic acid and thickeners): placebo
  • vaginal dosage form placebo batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 5:- i. Add into the mixer the following materials - 3, 4, 5, 6, 7, 8, 9,1
  • Formulation 3f Mixture (oleic acid, stearic acid, HPMC and thickeners); full strength molar ratio of lopinavir to ritonavir 13.8:1
  • vaginal dosage form placebo batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 6:- i. Add into the mixer the following materials - 3, 4, 5, 6, 7, 8, 9, 1, 10, 11
  • Formulation 3g Mixture (oleic acid, stearic acid. HPMC and thickeners): half strength
  • vaginal dosage form placebo batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 7:- i.
  • vaginal dosage form exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 8:- i.
  • medicine mixer add 1, 9, 10, 11
  • Formulation 3i Binary mixture (oleic acid and stearic acid): full strength- molar ratio of lopinavir to ritonavir 13.8:1
  • Formulation 3i Binary mixture (oleic acid and stearic acid); full strength- molar ratio of lopinavir to ritonavir 9.2:1
  • Formulation 3k Mixture (oleic acid, stearic acid, HPMC and thickeners); full strength molar ratio of lopinavir to ritonavir 13.8:1; cold process
  • vaginal dosage form exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 11 :-
  • Example 3 The formulations described in Example 3 are suitable for topical dosing and syringe dosing. Organoleptic assessments of formulations 3a, 3b, 3c, 3d, 3e, 3f, 3g, 3i, 3j, 3h and 3k were conducted.
  • the opaque ointment presents as easy to dispense from the tube and is controllable on the skin during rubbing. After a short period of rubbing, the ointment clarifies to leave the skin non- tacky and with minimal greasiness. Approximately 10 minutes after dosing, the skin presents as silky, with negligible greasiness. These attributes denote an ointment as particularly suitable for topical application.
  • the opaque ointment presents as easy to dispense from the tube into the syringe applicator, after a short period of rubbing the ointment becomes increasingly more opaque, increasing whiteness, embodied, gelatinous, and cream-like.
  • These attributes denote an ointment that is compatible with a water environment, and as such, particularly suitable for topical application to an area of the body where water is present, for example vaginal application.
  • compositions according to Example 3 have been proven to be stable during the process employed to manufacture them (i.e. processes described in Example 3). Additionally, the compositions according to Example 3 have been found to be stable upon storage.
  • Formulations 3c, 3d, 3f, 3g, 3i, 3j, and 3h were stored under different time and stability conditions, and subsequently analysed.
  • the stability data is presented in Tables 13 and 14.
  • a stable product is defined as one demonstrating 90-110% w/w assay for both Lopinavir and Ritonavir. Based on the data generated, the hot processed product
  • Formulation 3h when stored at 5°C (range 2-8°C) affords at least 10 months stability.
  • Formulation 3h (prepared using a cold process) demonstrates greater stability than that for formulations 3c and 3d when stored for 3 months stored at 25°C. This is thought to be due to the absence of thermal exposure of Ritonavir and Lopinavir during the manufacture. Forward projection of the stability data suggest at least 6 months/25°C is achievable for Formulation 3h. To get stability beyond 6 months for Formulation 3h, cold storage (5°C) may be required.
  • **Formulation 3j was an early formulation. A stability assessment was not taken at the time of manufacture. For stability trending, formulation 3i (a close variant of formulation 3j) was manufactured. The following analytical methodology was used to analyse the formulations. UHPLC:
  • Example 6 Rheological characterisation of the in-situ transition provided by the hydrophilic muco-adhesive when the anhydrous composition is exposed to an
  • the following methodology was used to determine the rheological response of the anhydrous composition upon exposure to an environment comprising water.
  • a Discovery Hybrid Rheometer (Model HR-3, TA instruments) was used. The sample was introduced onto the peltier plate (base) in excess (l-2g), and the spindle lowered to make contact with the sample. The excess sample was cleaned up. The spindle was then rotated in a predetermined fashion to exert a series of shear forces on the sample. The various sample shear force response parameters (complex viscosity, storage and loss moduli, tan 5) were captured and then plotted in order to make conclusions regarding inherent sample attributes.
  • HPMC level investigated was 0 - 10 % w/w, see Formulations 6a, 6b, 6c and 6d.
  • Formulation 6b 1% HPMC - Manufactured using a low shear mixing process The manufacture of a composition comprising 1% HPMC is described below in accordance with Table 16:- i. Into main vessel add 4, 5, 6, 7, 8, 9, 10, 1
  • vii. Lift the main vessel lid and introduce the solvent phase contents into the main vessel. viii. Under vacuum (e.g. -0.5 to -0.9 bar), stir very well to combine the two phases while cooling to ambient. Aim for a very smooth consistency free from lumps. Faster /longer (e.g 90 rpm 120 mins) stirring can help to achieve this. Use homogenizer periodically at 600 rpm to assist.
  • vacuum e.g. -0.5 to -0.9 bar
  • Formulation 6c 1 % HPMC - Manufactured using high pressure homogenizing
  • the muco-adhesive effect has been interpreted via the rheological response of the formulation in the presence of water, as demonstrated in the oscillation frequency plot as shown in Figure 1 for a prototype vaginal dosage form ointment containing 1% w/w HPMC (formulation 6b). From Figure 1, it can be seen that once 2-3 drops of water are introduced to the product environment, a shift in the rheological response occurs, consistent with the forming of an in situ gel dispersion. This drives an increase in both the complex viscosity (h) and the storage modulus (G’).
  • the storage modulus is a direct measure of a fluid’s solid-like behaviour.
  • the ratio of the loss modulus (G”) to the storage modulus (G’) is a unitless measure, Tan 5.
  • a Tan d value of ⁇ 1 defines a material as largely solid-like; >1 classifies the material as predominately fluid-like. In the presence of water, the increase in complex viscosity and storage modulus can be interpreted as defining a product with increased adherence potential and resistance to movement.
  • a prototype vaginal ointment without HMPC does not exhibit the same upward trend for complex viscosity or storage modulus.
  • the complex viscosity and storage modulus both decrease, which, together with the increasing loss modulus, results in raising Tan d.
  • Such a product can be interpreted as having low potential to adhere within the vaginal cavity.
  • High pressure homogenizing is an alternate manufacturing process step, found to achieve greater product dispersion, a decrease in particle size, leading to an increase in product surface area, and potentially an increase in viscosity.
  • a prototype vaginal ointment with 1% w/w HPMC processed via HPH (formulation 6c) demonstrates the ability to achieve an increase in product viscosity when compared to the same formulation processed via traditional low shear (formulation 6b).
  • the product does nonetheless, still display the same transition from fluid-like to solid-like, as per Figure 4.
  • the storage modulus switches from a position of below the loss modulus, to above that of the loss modulus; Tan d moves from >1 to be ⁇ 1, and the product demonstrates solid-like behaviour.
  • Formulation 7 The manufacture of a composition comprising HPMC 2910 (4000 cps) as the mucosa adhesive is described below in accordance with Table 19:- i. Into main vessel add materials except 2
  • the storage modulus is a direct measure of a fluid’s solid-like behaviour, while the ratio of the loss modulus (G”) to the storage modulus (G’) is a unitless measure, Tan d.
  • a Tan d value of ⁇ 1 defines a material as largely solid-like; >1 classifies the material as predominately fluid-like.
  • the increase in complex viscosity and storage modulus, and reduction in Tan d can be interpreted as defining a product with increased adherence potential and resistance to movement.
  • UV LIGHT Example 8 Excipient One Factor At A Time (OF AT) analysis
  • Formulation 8a Full strength ointment
  • Example 9 A Phase 1, single centre, double blind, randomised, parallel group, ascending single and multiple dose, safety and tolerability, pharmacokinetic (PK) and pharmacodynamic (PD) study of Formulations 3c, 3d and 3e in healthy women volunteers.
  • PK pharmacokinetic
  • PD pharmacodynamic
  • compositions according to Example 3 were investigated according to the clinical trial described below.
  • Period 1 Single dose of Formulation 3d or placebo Formulation 3e followed by confinement. PK blood sampling during confinement.
  • Period 2 21 daily doses of Formulation 3d or placebo Formulation 3e followed by PK blood sampling.
  • Period 2 21 daily doses of Formulation 3c or placebo Formulation 3e followed by PK blood sampling.
  • Inclusion Criteria a. Women, 20 to 45 years old, with an intact uterus and vagina.
  • dronedarone Ranolazine, Fusidic Acid, Colchicine, Astemizole, terfenadine, Lurasidone, Pimozide, Quetiapine, Dihydroergotamine, ergonovine, ergotamine, methylergonovine, Cisapride, Lovastatin, simvastatin, Avanafil, Sildenafil, Vardenafil, Oral midazolam, triazolam, St. John's wort.
  • the physical examination consisted of a review of body systems with height and weight (in indoor clothing).
  • Electrolytes sodium and potassium
  • ALT sodium and potassium
  • GGT ALP
  • albumin total protein
  • total bilirubin urea
  • uric acid serum creatinine
  • TFT fasting lipids
  • amylase glucose
  • the measurement at screening serve as a baseline to monitor any abnormalities that may manifest as a result of dosing
  • benzodiazepines and opiates i.e. morphine, heroin and codeine.
  • Vaginal swabs for microbiology gonorrhoea, Chlamydia, bacterial vaginosis, Candida
  • Serum HCG testing was carried out on all participants as part of the screening procedures and within 3 days before the lst dose.
  • Systolic Blood Pressure > 90 or ⁇ 160 mm Hg; Diastolic Blood Pressure > 50 or ⁇ 90 mm Hg Sitting:
  • Systolic Blood Pressure > 90 or ⁇ 160 mm Hg; Diastolic Blood Pressure > 50 or ⁇ 90 mm Hg
  • Dosing began at approximately 8pm on each day dosing was scheduled. Participants were instructed to insert the medication in private. Dosing applicators were returned to study staff and examined to ensure the full dose has been applied and for reconciliation of study drug. Sample collection
  • Vaginal swabs were self-administered by the participants.
  • PK Blood samples blood samples (8 mL) were drawn through venous catheters and transferred into vacutainers containing sodium heparin as the anti-coagulant. The time of collection was recorded as the time the full 8 mL of blood is collected. The venous catheters were kept patent by flushing with 1.5 mL-2.0 mL of heparinized saline following each sample (0-24 hours). The sampling intervals were at: Day 1-2: 0, 1, 2, 4, 8, 12, 24 hours; Day 22-23: 0, 1, 2, 4, 8, 12, 24 hours. Samples were collected at their due time. Any deviation was noted.
  • Plasma was separated by centrifugation at 3500 rpm for 5 minutes at about 4°C. No aids for separation of plasma from red cells was used. The plasma sample was transferred with clean pipettes. The assay was determined using a validated Analytical method.
  • Each plasma sample was placed into a polypropylene storage tube with a screw cap.
  • the plasma was stored frozen at -60°C or colder at the clinical site pending transfer to a
  • each participant was required to provide a blood sample for analysis. Any abnormalities as compared to initial screening were monitored and followed up until they return to normal.
  • AE was classified by the Principal Investigator as serious adverse event (SAE) or non- serious.
  • SAE serious adverse event
  • Non- serious adverse events were assessed as being mild, moderate, or severe to describe the maximum intensity of the AE.
  • the Principal Investigator also provided the possible relationship between the AE and the study medication as highly probable, probable, possible, remotely or not ("no") related to the study medication.
  • the Principal Investigator should have stated if the cause of the AE is related to the concurrent non-investigational medication(s) if any are being taken, an underlying disease, a combination of these factors or is unknown.
  • Formulations 3c and 3d are deemed to be well-tolerated since all AEs were either minor or not related to administration of study medication
  • AUC area under the plasma drug concentration time curve
  • Cmax peak plasma drug concentration
  • Tmax time to maximum drug concentration
  • the plasma drug concentration (C) versus the real sampling time (t) data were analysed by a "noncompartmental" method to obtain the pharmacokinetic parameters. Initially the plasma data in the post distribution phase of the plasma concentration - time plot were fitted using linear regression to: ⁇
  • the mean lopinavir and ritonavir plasma concentration-time data for each sampling time is listed in Tables 25 and 26.
  • the pharmacokinetic parameters for lopinavir and ritonavir are summarised in Tables 27 and 28.
  • Table 25 Mean ( ⁇ SD) Plasma Lopinavir and Ritonavir Concentration Data vs Sampling Times (Formulation 3d) Vaginal Ointment)
  • Table 26 Mean ( ⁇ SD) Plasma Lopinavir and Ritonavir Concentration Data vs Sampling Times (Formulation 3c) Vaginal Ointment)
  • the mean Cmax for lopinavir was 12.3 ⁇ 5.4 pg/mL (SPMC Kaletra) . Adjusting for dose comparison with a 300 mg dose administered topically in ointment form, the mean Cmax would be 9.23 ⁇ 4.1 pg/mL.
  • the AUCO-t for lopinavir was 113.2 ⁇ 60.5 pg h/mL (SPMC Kaletra). Adjusting for dose comparison with a 300 mg dose administered topically in ointment form, the AUCO-t would be 84.9 ⁇ 45.4 pg h/mL.
  • the AUCO-t was 7368.1 ⁇ 4973.1 pg/mL.
  • the ratio of AUC oral/AUC topical is >11,500 indicating that less than 0.009% of the topical dose is available systemically.
  • the combined Cmax and AUC data indicate that systemic absorption of lopinavir from topical administration of the ointment is negligible.
  • Example 10 A Phase lb. Multicentre. Open Label. Study of the Efficacy. Safety and Tolerability of Formulation 3h in Women with Cvtological Abnormalities of the Uterine Cervix.
  • Tolerability Objective Assess the tolerability of Formulation 3h, measured by compliance with dosing schedule of Formulation 3h during 21 consecutive days of treatment for up to 3 treatment cycles.
  • This study is designed as a Phase lb multicentre, open label study investigating the efficacy, safety and tolerability of Formulation 3h ointment in women with cytological abnormalities of the uterine cervix.

Abstract

The present invention relates to anhydrous pharmaceutical compositions for topical application comprising at least one active pharmaceutical ingredient and a hydrophilic muco-adhesive agent; to the use of said compositions as a medicament; and to processes for the preparation of said compositions.

Description

PHARMACEUTICAL COMPOSITIONS
Provided herein are anhydrous compositions for topical application comprising at least one active pharmaceutical ingredient and a hydrophilic muco-adhesive agent; methods for their manufacture; and the use of said anhydrous compositions as a medicament. In particular, compositions are provided comprising lopinavir and ritonavir for use in treating and/or inhibiting the progression of HPV related dysplasia of the cervix.
BACKGROUND OF INVENTION
Topical compositions can be defined as compositions that are applied directly to an external body surface, which includes the skin and membranes such as the rectal lining, the lining of the mouth (buccal mucosa), the vaginal wall (vaginal mucosa), the covering of the eye (cornea and conjunctival membranes), the urethral membrane, the lining of the external ear, and the nasal mucous membrane.
In order for a topical composition containing an active pharmaceutical ingredient to be successfully used as a medicament, the composition once applied to an external body surface must remain at the site of application for a sufficient period in order for an efficacious dose of the active pharmaceutical ingredient to be delivered to the site of treatment. For example, the topical composition needs to remain at the site of application long enough for a sufficient proportion of the active pharmaceutical ingredient to be released from the composition and have the desired efficacious effect on the disease and/or disorder being treated. Many topical compositions are unable to reside at the site of application for a sufficiently long period. For example, fluid like systems may readily flow off the applied area (streak). Alternatively, and/or additionally, depending on the site of application, the composition may be rapidly cleared away from the site of application by the body’s natural clearance mechanisms. This can result in variable amounts of the active pharmaceutical ingredient being dosed to the patient as well as under dosing in certain circumstances.
Topical compositions may contain excipients which increase the viscosity of the composition and thereby provide some improvement in residence time at the site of application. While this may reduce the risk of under dosing and/or the patient receiving variable amounts of the active pharmaceutical ingredient, it also typically results in the composition being more difficult to handle. For example, transferring the composition between vessels during the manufacturing process and/or during the filing of tubes/syringes suitable for patients and/or healthcare professionals to use. In addition, manipulating the tube/syringe during dispensing may be difficult if the composition is especially viscous.
Topical compositions can be presented in a number of formats. Suitable formats include ointments, creams, pastes, and gels. Some of these formats have water present within the composition. Topical compositions which contain water are not ideal for use with an active pharmaceutical ingredient which is prone to degradation by hydrolysis as this can result in a short shelf life of the pharmaceutical product and/or the requirement to store the composition in certain conditions in order to minimise degradation of the active
pharmaceutical ingredient.
The present invention is based on work carried out by the inventors to formulate a composition comprising at least one active pharmaceutical ingredient for topical application. They have unexpectedly established that the use of a hydrophilic muco-adhesive agent in an anhydrous composition offers particular benefits.
Firstly, the composition is anhydrous, thus the composition is particularly well suited to active pharmaceutical ingredients which are prone to degradation due to hydrolysis.
Secondly, the use of a hydrophilic muco-adhesive agent in the anhydrous composition offers particular benefits in respect to the properties and behaviour of the composition upon topical application. The hydrophilic muco-adhesive agent causes the composition to undergo an in- situ transition when the anhydrous composition is applied to a site of application. Specifically, the hydrophilic muco-adhesive agent attracts water/moisture at the site of application and alters the physical properties of the composition. For example, the resulting muco-adhesive composition exhibits greater adhesiveness and/or tackiness. As such, the composition containing the hydrophilic muco-adhesive agent when administered to a site of application provides a prolonged retention time. Conveniently, the in-situ transition that occurs upon administration causes the viscosity of the muco-adhesive composition after dosing to be greater than the viscosity of the anhydrous composition prior to dosing. Advantageously this helps provides a prolonged retention time at the site of application but also allows ease of handling during manufacturing and/or dispensing, i.e. due to the lower viscosity of the anhydrous composition.
The present invention provides an anhydrous composition for topical application comprising at least one active pharmaceutical ingredient and a hydrophilic muco-adhesive agent, wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition. Compositions of the invention are particularly useful in providing topical treatment for the prevention or treatment of diseases and/or disorders such as for the prevention or treatment of cancerous conditions, for the prevention or treatment of oncogenic viral infections and/or for the prevention or treatment of benign proliferative orders, wherein the site of application comprises a mucosal membrane, for example, the vaginal wall.
In one embodiment, the site of application is a mucosal membrane. In a particular embodiment, the site of application is the vaginal wall.
In particular, the compositions are useful for the prevention or treatment of cervical cancer wherein the site of application is the vaginal wall, and wherein the composition comprises lopinavir and ritonavir.
Many different forms of cancer exist, and it is believed that there are many different causes of the disease. The incidence of cancer varies, but it represents the second highest cause of mortality, after heart disease, in most developed countries.
Human tumour viruses are recognised to be a major cause of human cancer, and there is a great deal of evidence which supports the contention that these viruses cause cancer by inducing genetic instability in infected cells. Indeed, both the human T-cell leukemia virus type 1 (HTLV1) Tax and the human papilloma virus type 16 (HPV16) E6 oncoproteins are known to induce genetic instability producing abnormal numbers of centrosomes,
multinucleation and nuclear atypia.
Invasive cervical cancer (ICC) is an example of a cancer associated with viral infection which causes >270,000 deaths per annum with over 85% of these occurring in low resource countries. Infection with high-risk types of HPV has been established as the main aetiological agent for ICC. The development of ICC can take 10-20 years and is preceded by HPV related pre-invasive pathology which is characterised as either low-grade (CIN1) or high-grade cervical intraepithelial neoplasia (CIN2/3). Lesions can be screened for by cervical cytology testing where they are diagnosed (or graded) as either borderline atypical squamous cells of undetermined significance (ASCUS), low-grade squamous intraepithelial lesions (LSIL) or high-grade squamous intraepithelial lesions (HSIL).
The reduction in ICC related mortality in the developed world has been largely dependent on organised cytology screening and similar trends in cervical cancer mortality have been achieved by organised single screen and treatment in the rest of the world. However, in the poorer nations lack of resources and health education means that most pre-invasive cervical disease remains undiagnosed and untreated. Thus, where resources are limited, low-cost screening and treatment options are clearly a high priority. Current treatment options in clinical practice are either by ablative (destructive) or excisional modalities. Systematic reviews have demonstrated that these treatment modalities have similar success rates but have different morbidities. In the developed world, Large Loop Excision of the Transformation Zone LLETZ (aka loop electrosurgical excision procedure - LEEP) is used in the majority of colposcopy clinics. Over 80% of these procedures are performed under local analgesia and the whole of the transformation zone is available for subsequent histological examination. The procedure is associated with a risk of
primary/secondary haemorrhage, prolonged discharge, infection and a risk of preterm delivery in subsequent pregnancies. The former side effects can be problematic particularly in low resource countries. Ablative treatment in the form of cold coagulation and cryotherapy are often advocated for use in low resource countries since these are low cost, require minimal infrastructure and can be carried out by trained non-medical health professionals. However, some studies have suggested that cryotherapy has a higher failure rate compared to other treatment modalities.
There are a variety of locally-applied, non-surgical approaches which have been evaluated for the treatment of cervical dysplasia including; photodynamic therapy (PDT); off- licence use of the anti-cytomegalovirus (CMV) drug cidofovir; local application of the immune activator Imiquimod and direct application of the cytotoxic drug 5 flurouracil (5FU). Although some of these alternative treatment modalities show promise, their treatment outcomes are inferior to the reported 80-95% success rates obtained in quality assured colposcopy units.
An effective, inexpensive, non-surgical, self-applied treatment for HPV related cervical dysplasia would have great potential particularly in low resource settings. Furthermore, improved compliance with topical treatment would be enhanced, if the side effects are minimised.
A recent advance in the treatment of cancers caused by viruses is disclosed in
WO2015/059485 which describes the protease inhibitors, lopinavir and ritonavir (which had previously been used as orally ingested medicaments for the clinical management of retroviral infections such as HIV) as being clinically useful for topical administration to tissues to prevent or treat malignancies caused by human papilloma virus. The authors were particularly surprised to find that soft capsules of KALETRA® (which is marketed by Abbott/ Abbvie for the treatment of HIV infections by oral administration) can be administered topically (e.g. inserted into the vagina for treatment of the cervix) for the prevention or treatment of cancerous conditions, for the prevention or treatment of oncogenic viral infections and for the prevention or treatment of benign proliferative orders. KALETRA® (or its equivalent LOPIMUNE) is available for oral consumption as a solution comprising 80 mg lopinavir and 20 mg ritonavir per millilitre or as a soft capsule for oral administration that comprises 133.3 mg lopinavir and 33.3 mg ritonavir. The solution additionally contains alcohol (42% w/w), high fructose corn syrup, propylene glycol, purified water, glycerol, povidone, flavourings, polyoxyl 40 hydrogenated castor oil, acesulfame potassium, saccharin sodium, sodium chloride, peppermint oil, sodium citrate, citric acid, and menthol. The soft capsule contents contain, along with lopinavir and ritonavir, oleic acid, propylene glycol, polyoxy 35 castor oil, and purified water (KALETRA® Summary of Product Characteristics, EMA).
The compositions of the invention offer significant benefits when compared to oral soft capsule compositions when topical formulation is required.
SUMMARY OF INVENTION
Disclosed herein are anhydrous compositions comprising at least one active pharmaceutical ingredient and a hydrophilic muco-adhesive agent.
According to a first aspect of the invention, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
According to a second aspect of the invention, there is provided a method of manufacturing the anhydrous composition of the first aspect comprising the step of incorporating (e.g. by mixing) a hydrophilic muco-adhesive agent into the anhydrous composition.
According to a third aspect of the invention there is provided an anhydrous composition according to the first aspect of the invention for use as a medicament. In one embodiment, the anhydrous composition is used as a medicament for treating and/or inhibiting the development of diseases and/or disorders. In one embodiment, the anhydrous composition is used as a medicament for treating and/or inhibiting the development or progression of cancers and/or benign proliferative disorders. In one embodiment, the anhydrous composition comprises an effective amount of the at least one active
pharmaceutical ingredient. In one embodiment, the anhydrous composition comprises an effective amount of the at least one active pharmaceutical ingredient for treating and/or inhibiting the development or progression of a disease or disorder. In one embodiment, the anhydrous composition comprises an effective amount of the at least one active
pharmaceutical ingredient for treating and/or inhibiting the development or progression of cancers and/or benign proliferative disorders. In a further embodiment, the anhydrous composition comprises an effective amount of at least one active pharmaceutical ingredient for treating a Human Papilloma Virus (HPV) infection with or without attendant abnormal pathology. In one embodiment, the anhydrous composition is used as a medicament for treating and/or inhibiting the development of early stage neoplasias. In one embodiment, the anhydrous composition is used as a medicament for treating or preventing the development of HPV related cervical, vulval, vaginal, penile, anal, oral or laryngeal neoplasias and/or warts. In one embodiment, the anhydrous composition is for use as a medicament for treating or preventing the development of cervical neoplasias.
According to a fourth aspect of the invention there is provided a method of treating and/or inhibiting the development or progression of diseases and/or disorders in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention. In one embodiment, there is provided a method of treating and/or inhibiting the development or progression of cancers and/or benign proliferative disorders in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention.
In a further embodiment, there is a provided a method of treating a Human Papilloma
Virus (HPV) infection with or without attendant abnormal pathology in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention. In one embodiment, there is provided a method of treating and/or inhibiting the development of early stage neoplasias in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention. In one embodiment, there is provided a method of treating or preventing the development of HPV related cervical, vulval, vaginal, penile, anal, oral or laryngeal neoplasias and/or warts in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention. In one embodiment, there is provided a method of treating or preventing the development of cervical neoplasias in a subject in need of such treatment or inhibition comprising administering a therapeutically effective amount of the anhydrous composition according to the first aspect of the invention.
Conveniently, the cancer or benign proliferative disorder is caused by a viral infection, such as by an oncogenic virus and in particular human tumour viruses such as HPV.
Conveniently, the invention concerns treating a subject having an HPV related dysplasia of the cervix comprising administering to said subject a therapeutically effective dose of the disclosed anhydrous compositions.
BRIEF DESCRIPTION OF DRAWINGS
The summary, as well as the following detailed description, is further understood when read in conjunction with the appended drawings. For the purposes of illustrating the disclosed compositions, processes of manufacture and methods, there are shown in the drawings exemplary embodiments of the compositions, processes of manufacture and methods; however, the compositions, processes of manufacture and methods are not limited to the specific embodiments disclosed. In the drawings:
Figure 1 : Oscillation frequency rheological response of a prototype vaginal ointment containing 1% w/w HPMC in both the absence and presence of water
Figure 2: Oscillation frequency rheological response of a prototype vaginal ointment containing no HPMC in both the absence and presence of water
Figure 3: Oscillation frequency rheological response comparison of low shear and high- pressure homogenizing processes for a prototype vaginal ointment containing 1% w/w HPMC
Figure 4: Oscillation frequency rheological response of an HPH processed prototype vaginal ointment containing 1% w/w HPMC in both the absence and presence of water
Figure 5: Oscillation frequency rheological response of Formulation 7 in both the absence and presence of water
DETAILED DESCRIPTION
The disclosed compositions, processes of manufacture and methods may be understood more readily by reference to the following detailed description taken in
connection with the accompanying figures, which form a part of this disclosure. It is to be understood that the disclosed compositions, processes of manufacture and methods are not limited to the specific compositions, processes of manufacture and methods described and/or shown herein, and that the terminology used herein is for the purpose of describing particular embodiments by way of example only and is not intended to be limiting of the claimed compositions, processes of manufacture and methods.
Reference to a particular numerical value includes at least that particular value, unless the context clearly dictates otherwise. When a range of values is expressed, another embodiment includes from the one particular value and/or to the other particular value.
Further, reference to values stated in ranges include each and every value within that range. All ranges are inclusive and combinable.
When values are expressed as approximations, by use of the antecedent“about,” it will be understood that the particular value forms another embodiment.
It is to be appreciated that certain features of the disclosed compositions, processes of manufacture and methods which are, for clarity, described herein in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the disclosed compositions, processes of manufacture and methods that are, for brevity, described in the context of a single embodiment, may also be provided separately or in any sub-combination.
As used herein, the singular forms“a,”“an,” and“the” include the plural.
The following abbreviations are used herein: human papilloma virus (HPV); Atypical squamous cells of undetermined significance (ASC-US); Low grade squamous intraepithelial lesion (LSIL); High grade squamous intraepithelial lesion (HSIL); Cervical intraepithelial neoplasia 1 (CIN1); Cervical Intraepithelial neoplasia 2 (CIN2); Cervical intraepithelial neoplasia 3 (CIN3); Carcinoma in situ (CIS); Invasive Cervical Carcinoma (ICC).
The term“about” when used in reference to numerical ranges, cut-offs, or specific values is used to indicate that the recited values may vary by up to as much as 10% from the listed value. As many of the numerical values used herein are experimentally determined, it should be understood by those skilled in the art that such determinations can, and often times will, vary among different experiments. The values used herein should not be considered unduly limiting by virtue of this inherent variation. Thus, the term“about” is used to encompass variations of ± 10% or less, variations of ± 5% or less, variations of ± 1% or less, variations of ± 0.5% or less, or variations of ± 0.1% or less from the specified value.
As used herein,“treating” and like terms refer to reducing the severity and/or frequency of symptoms, eliminating symptoms and/or the underlying cause of said symptoms, reducing the frequency or likelihood of symptoms and/or their underlying cause, delaying, preventing and/or slowing the progression of diseases and/or disorders, such as cancers or benign proliferative disorders, and improving or remediating damage caused, directly or indirectly, by the diseases and/or disorders such as cancers or benign proliferative disorders.
As used herein, the phrase“therapeutically effective dose” refers to an amount of a composition comprising at least one active pharmaceutical ingredient, for example comprising lopinavir and ritonavir, as described herein, effective to achieve a particular biological or therapeutic result such as, but not limited to, biological or therapeutic results disclosed, described, or exemplified herein. The therapeutically effective dose may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the composition to cause a desired response in a subject. Such results include, but are not limited to, the reduction, remission, and/or regression of the benign or malignant disease or prevention of the development of the benign or malignant disease, as determined by any means suitable in the art.
As used herein,“subject” includes a vertebrate, mammal, domestic animal or a human being.
ANHYDROUS COMPOSITIONS
According to a first aspect of the invention, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient;
b. a hydrophilic muco-adhesive agent; and
c. a solvent for the at least one active pharmaceutical ingredient;
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient;
b. a hydrophilic muco-adhesive agent; c. a solvent for the at least one active pharmaceutical ingredient; and d. a thickener;
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient;
b. a hydrophilic muco-adhesive agent;
c. a solvent for the at least one active pharmaceutical ingredient;
d. a thickener;
e. a stiffening agent;
f. a blending agent; and
g. an antioxidant;
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition; and wherein the anhydrous composition is a semi-solid at room temperature.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition; and wherein the anhydrous composition is an ointment. In one embodiment, there is provided an anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition; wherein the anhydrous composition is a semi-solid at room
temperature; and wherein the muco-adhesive composition is a semi-solid at room
temperature.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition; wherein the anhydrous composition is an ointment; and wherein the muco-adhesive composition is a gel.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition and wherein the muco-adhesive composition is a gel.
In one embodiment, there is provided an anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition and wherein the muco-adhesive composition has an increase in adhesiveness compared to the anhydrous composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition and wherein the muco-adhesive composition provides prolonged retention of the at least one active pharmaceutical ingredient at the site of application.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition and wherein the muco-adhesive composition provides prolonged retention of the at least one active pharmaceutical ingredient at the site of application for at least 15 minutes, preferably at least 30 minutes, preferably at least 1 hour, preferably at least 90 minutes, preferably at least 2 hours, preferably at least 4 hours, preferably at least 6 hours, preferably at least 8 hours, preferably at least 10 hours, preferably at least 12 hours, or preferably at least 24 hours.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition and wherein the muco-adhesive composition has a greater viscosity than the anhydrous composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient; and
b. hydroxypropylmethylcellulose;
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. ritonavir;
b. lopinavir; and
c. a hydrophilic muco-adhesive agent, such as hydroxypropylmethylcellulose; wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. ritonavir;
b. lopinavir;
c. hydroxypropylmethylcellulose;
d. oleic acid;
e. stearic acid; and
f. butylated hydroxytoluene
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
In one embodiment, the anhydrous composition comprises:
a. about 1.2 to about 1.4% by weight of ritonavir;
b. about 9 to about 11% by weight of lopinavir;
c. about 0.5 to 1.5% by weight of hydroxypropylmethylcellulose;
d. about 55 to about 65% by weight of oleic acid;
e. about 28 to about 32% by weight of stearic acid; and
f. about 0.05 to about 0.5% by weight of butylated hydroxytoluene
wherein all % are by weight based upon the total weight of the composition; and wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
In one embodiment, the anhydrous composition comprises:
a. about 0.5 to about 0.7% by weight of ritonavir;
b. about 4 to about 6% by weight of lopinavir;
c. about 0.5 to 1.5% by weight of hydroxypropylmethylcellulose;
d. about 55 to about 65% by weight of oleic acid;
e. about 28 to about 32% by weight of stearic acid; and
f. about 0.05 to about 0.5% by weight of butylated hydroxytoluene
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
In one embodiment, the anhydrous composition comprises:
a. ritonavir;
b. lopinavir;
c. hydroxypropylmethylcellulose;
d. oleic acid;
e. stearic acid;
f. butylated hydroxytoluene;
g. mono diglyceride;
h. ceresin wax;
i. hydrogenated vegetable oil;
j. polyoxyl 100 stearate; and
k. glycerol monooleate;
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
In one embodiment, the anhydrous composition comprises:
a. about 0.9 to about 1.1% by weight of ritonavir;
b. about 9 to about 11% by weight of lopinavir; c. about 0.5 to about 1.5% by weight of hydroxypropylmethylcellulose;
d. about 55 to about 65% by weight of oleic acid;
e. about 4 to about 5% of stearic acid;
f. about 0.1 to about 0.3% by weight of butylated hydroxytoluene;
g. about 4 to about 6% by weight of mono diglyceride;
h. about 5 to about 7% by weight of ceresin wax;
i. about 9 to about 11% by weight of hydrogenated vegetable oil;
j. about 1 to about 3% by weight of polyoxyl 100 stearate; and
k. about 2 to about 4% by weight of glycerol monooleate;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
In one embodiment, the anhydrous composition comprises:
a. about 0.9 to about 1.1% by weight of ritonavir;
b. about 11 to about 13% by weight of lopinavir;
c. about 0.5 to about 1.5% by weight of hydroxypropylmethylcellulose; d. about 50 to about 60% by weight of oleic acid;
e. about 4 to about 5% of stearic acid;
f. about 0.1 to about 0.3% by weight of butylated hydroxytoluene;
g. about 4 to about 6% by weight of mono diglyceride;
h. about 5 to about 7% by weight of ceresin wax;
i. about 9 to about 11% by weight of hydrogenated vegetable oil;
j. about 1 to about 3% by weight of polyoxyl 100 stearate; and
k. about 2 to about 4% by weight of glycerol monooleate;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
In one embodiment, the anhydrous composition comprises:
a. about 0.4 to about 0.6% by weight of ritonavir;
b. about 4 to about 6% by weight of lopinavir; c. about 0.5 to about 1.5% by weight of hydroxypropylmethylcellulose;
d. about 55 to about 65% by weight of oleic acid;
e. about 4 to about 5% of stearic acid;
f. about 0.1 to about 0.3% by weight of butylated hydroxytoluene;
g. about 4 to about 6% by weight of mono diglyceride;
h. about 5 to about 7% by weight of ceresin wax;
i. about 9 to about 11% by weight of hydrogenated vegetable oil;
j. about 1 to about 3% by weight of polyoxyl 100 stearate; and
k. about 2 to about 4% by weight of glycerol monooleate;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
In one embodiment, the anhydrous composition comprises:
a. about 0.4 to about 0.6% by weight of ritonavir;
b. about 5 to about 7% by weight of lopinavir;
c. about 0.5 to about 1.5% by weight of hydroxypropylmethylcellulose; d. about 55 to about 65% by weight of oleic acid;
e. about 4 to about 5% of stearic acid;
f. about 0.1 to about 0.3% by weight of butylated hydroxytoluene;
g. about 4 to about 6% by weight of mono diglyceride;
h. about 5 to about 7% by weight of ceresin wax;
i. about 9 to about 11% by weight of hydrogenated vegetable oil;
j. about 1 to about 3% by weight of polyoxyl 100 stearate; and
k. about 2 to about 4% by weight of glycerol monooleate;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms into a muco- adhesive composition.
In one embodiment, the anhydrous composition for topical application comprises: a. at least one active pharmaceutical ingredient; and
b. about 0.1 to about 10% by weight of hydrophilic muco-adhesive agent; wherein all % are by weight based upon the total weight of the composition; and wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient;
b. about 0.1 to about 10% by weight of hydrophilic muco-adhesive agent; and c. about 25 to about 75% by weight of a solvent for the at least one active
pharmaceutical ingredient;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient;
b. about 0.1 to about 10% by weight of hydrophilic muco-adhesive agent;
c. about 25 to about 75% by weight of a solvent for the at least one active
pharmaceutical ingredient; and
d. about 0.1 to about 65% by weight of a stiffening agent;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient;
b. about 0.1 to about 10% by weight of hydrophilic muco-adhesive agent;
c. about 25 to about 75% by weight of a solvent for the at least one active
pharmaceutical ingredient; and
d. at least 0.1% by weight of a thickener;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition. In one embodiment, there is provided an anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient;
b. about 0.1 to about 10% by weight of hydrophilic muco-adhesive agent;
c. about 25 to about 75% by weight of a solvent for the at least one active
pharmaceutical ingredient; and
d. about 0.1 to about 40% by weight of a thickener;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient;
b. about 0.1 to about 10% by weight of hydrophilic muco-adhesive agent;
c. about 25 to about 75% by weight of a solvent for the at least one active
pharmaceutical ingredient; and
d. about 0.1 to about 40% by weight of a thickener; and
e. about 0.1 to about 65% by weight of a stiffening agent;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising:
a. at least one active pharmaceutical ingredient;
b. about 0.1 to about 10% by weight of hydrophilic muco-adhesive agent;
c. about 25 to about 75% by weight of a solvent for the at least one active
pharmaceutical ingredient;
d. about 0.1 to about 40% by weight of a thickener; and
e. about 0.1 to about 65% by weight of a stiffening agent;
f. about 0.1 to about 48% by weight of a blending agent; and
g. about 0.05 to about 0.5% of an antioxidant;
wherein all % are by weight based upon the total weight of the composition; and wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, the anhydrous composition for topical application comprises: a. at least one active pharmaceutical ingredient; and
b. about 0.1 to about 10% by weight of hydroxypropylmethylcellulose; wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
In one embodiment, there is provided an anhydrous composition for topical application comprising: a. about 25 to about 75% by weight of oleic acid;
b. about 0.1 to about 65% by weight of stearic acid;
c. ritonavir;
d. lopinavir;
e. about 0.1 to about 10% by weight of hydroxypropylmethylcellulose; f. about 0.05 to about 0.5% by weight of butylated hydroxytoluene; g. about 0.1 to about 10% by weight of mono diglyceride;
h. about 0.1 to about 10% by weight of ceresin wax;
i. about 0.1 to about 20% by weight of hydrogenated vegetable oil;
j. about 0.1 to about 38% by weight of polyoxyl 100 stearate; and
k. about 0.1 to about 10% by weight of glycerol monooleate;
wherein all % are by weight based upon the total weight of the composition; and
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
Advantageously, upon topical administration of the anhydrous composition to a site of application (such as preferably a mucosal membrane) the anhydrous composition transforms to a muco-adhesive composition. This in-situ transformation of the anhydrous composition to a muco-adhesive composition results in the muco-adhesive composition having different rheological behaviour, such as an increase in viscosity, and/or an increase in adhesiveness and/or increase in tackiness compared to the anhydrous composition.
In one embodiment, the muco-adhesive composition is a gel. In one embodiment, the muco-adhesive composition has an increase in adhesiveness and/or has a different rheological behaviour, such as an increase in viscosity, compared to the anhydrous composition. Advantageously, this increase in adhesiveness and/or different rheological behaviour means that the anhydrous composition is easier to handle during its manufacturing and/or during filing of dosing apparatus, such as a syringe, but can still provide upon application a muco-adhesive composition that is retained at the site of application for a prolonged period. In one embodiment, the muco-adhesive composition has a greater viscosity than the anhydrous composition.
The muco-adhesive compositions advantageously provide prolonged retention of the at least one active pharmaceutical ingredient at the site of application. This ensures that the composition remains at the site of application long enough to allow a significant proportion of the active pharmaceutical ingredient to be released from the composition and have the desired efficacious effect on the disease and/or disorder being treated.
The skilled person would be aware that various in-vitro techniques can be employed to measure the release of the active pharmaceutical ingredient from the composition. For example, the amount of active pharmaceutical ingredient delivered through a membrane can be determined using, for example, Vertical Diffusion Studies (“Topical and Transdermal Drug Products, Pharmacopeial Forum, Vol. 35(3) [May- June 2009]). The in-vitro release rate can be correlated to how the composition will perform in an in-vivo setting. In one embodiment, at least 50% by weight of the at least one active pharmaceutical ingredient is released from the composition, such as at least 60% by weight, such as at least 70% by weight, such as at least 80% by weight, such as at least 90% by weight, or such as at least 95% by weight within 2 hours, within 3 hours, within 4 hours, within 6 hours, or within 8 hours.
In one embodiment, the anhydrous composition remains at the site of application for at least 15 minutes, such as at least 30 minutes, such as at least 1 hour, such as at least 90 minutes, such as at least 2 hours, such as at least 4 hours, such as at least 6 hours, such as at least 8 hours, such as at least 10 hours, such as at least 12 hours, or such as at least 24 hours.
In one embodiment, the anhydrous composition remains at the site of application for up to 30 minutes, for up to 1 hour, for up to 90 minutes, for up to 2 hours, up to 4 hours, for up to 6 hours, for up to 8 hours, for up to 10 hours, for up to 12 hours, or for up to 24 hours.
A semi-solid is any material that can be present in a stationary material state until an external stress is applied resulting in flow of the material. The physical properties of a semi solid are a mixture of a solid and liquid. It will be apparent to the skilled person that the pharmaceutical composition is in a form of a semi-solid, for example by testing the pharmaceutical composition using a rheometer. From rheometric testing of the
pharmaceutical composition the yield stress, the storage modulus, the loss modulus and tan d can be determined. The yield stress is a direct measure for the initiation of material flow (solid state to fluid state) in response to an applied stress. The storage modulus is a direct measure of a fluid’s solid-like behaviour. The loss modulus is a direct measure of a fluid’s fluid-like behaviour. Tan d is the ratio of the loss modulus to the storage modulus and is a unitless measure. A tan d value of <1 defines a material as largely solid like, while a tan d value >1 defines a material as predominantly fluid-like. For example, the rheological behaviour of the semi-solid can be tested according to the methodology described in Example 6
In one embodiment, the anhydrous composition is a semi-solid at room temperature.
In another embodiment, the muco-adhesive composition is a semi-solid at room temperature. In another embodiment, the anhydrous composition is a semi-solid at room temperature and the muco-adhesive composition is a semi-solid at room temperature.
In one embodiment, the rheological behaviour of the anhydrous composition is different to the rheological behaviour of the muco-adhesive composition, for example, the compositions differ in respect to complex viscosity and/or storage modulus and/or loss modulus and/or tan delta.
Complex viscosity is defined as the frequency-dependent viscosity function determined for a viscoelastic fluid by subjecting it to oscillatory shear stress. Storage modulus is a measure of the stored energy within a material and represents the elastic portion. The loss modulus measures the energy dissipated as heat and represents the viscous portion. Tan delta is the ratio of loss modulus to the storage modulus. Thus, a tan delta value of greater than 1 means that the composition has a greater loss modulus than storage modulus.
In one embodiment, the complex viscosity of the muco-adhesive composition is greater than the complex viscosity of the anhydrous composition. In one embodiment, the muco-adhesive composition has a complex viscosity at least 50% greater than, such as at least 75% greater than, such as at least 100% greater than, or such as at least 110% greater than, the complex viscosity of the anhydrous composition and wherein the complex viscosity is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the anhydrous composition has a complex viscosity of between 1500 and 2000 cP.s, such as between 1600 and 1900 cP.s, or such as between 1700 and 1800 cP.s, and wherein the complex viscosity is measured at an angular frequency of 0.1 rad/s. In one embodiment, the muco-adhesive composition has a complex viscosity of between 3500 and 4000 cP.s, such as between 3600 and 3900 cP.s, or such as between 3800 and 3900 cP.s, and wherein the complex viscosity is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the anhydrous composition has a complex viscosity of between 1500 and 2000 cP.s, such as between 1600 and 1900 cP.s, or such as between 1700 and 1800 cP.s, and the muco-adhesive composition has a complex viscosity of between 3500 and 4000 cP.s, such as between 3600 and 3900 cP.s, or such as between 3800 and 3900 cP.s, and wherein the complex viscosity is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the storage modulus of the muco-adhesive composition is greater than the storage modulus of the anhydrous composition. In one embodiment, the muco- adhesive composition has a modulus of at least 100% greater than, such as at least 150% greater than, or such as at least 200% greater than, the storage modulus of the anhydrous composition and wherein the storage modulus is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the anhydrous composition has a storage modulus of between 50 and 150 Pa, such as between 75 and 125 Pa, or such as between 90 and 110 Pa, and wherein the storage modulus is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the muco-adhesive composition has a storage modulus of between 250 and 450 Pa, such as between 300 and 400 Pa, or such as between 300 and 350 Pa, and wherein the storage modulus is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the anhydrous composition has a storage modulus of between 50 and 150 Pa, such as between 75 and 125 Pa, or such as between 90 and 110 Pa, and the muco- adhesive composition has a storage modulus of between 250 and 450 Pa, such as between 300 and 400 Pa, or such as between 300 and 350 Pa, and wherein the storage modulus is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the loss modulus of the muco-adhesive composition is at least 20% greater than, such as at least 30% greater than, or such as at least 40% greater than, the loss modulus of the anhydrous composition and wherein the loss modulus is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the anhydrous composition has a loss modulus of between 130 and 170 Pa, such as between 140 and 160 Pa, or such as between 140 and 150 Pa, and the loss modulus is measured at an angular frequency of 0.1 rad/s. In one embodiment, the muco-adhesive composition has a loss modulus of between 190 and 230 Pa, such as between 200 and 220 Pa, or such as between 205 and 215 Pa, and the loss modulus is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the anhydrous composition has a loss modulus of between 130 and 170 Pa, such as between 140 and 160 Pa, or such as between 140 and 150 Pa, and the muco-adhesive composition has a loss modulus of between 190 and 230 Pa, such as between 200 and 220 Pa, or such as between 205 and 215 Pa, and the loss modulus is measured at an angular frequency of 0.1 rad/s.
In one embodiment, the tan delta value of the muco-adhesive composition is at least 15% less than, such as at least 20% less than, or such as at least 25% less than, the tan delta value of the anhydrous composition. In one embodiment, the anhydrous composition has a tan delta value greater than 1, such as greater than 1.2, such as greater than 1.3, or such as about 1.4, and wherein the loss modulus and the storage modulus are measured at an angular frequency of 0.1 rad/s. In one embodiment, the muco-adhesive composition has a tan delta value less than 1 , such as less than 0 8, such as greater than 0.7, or such as about 0.7, and wherein the loss modulus and the storage modulus are measured at an angular frequency of 0.1 rad/s. In one embodiment, the anhydrous composition has a tan delta value greater than 1, such as greater than 1.2, such as greater than 1.3, or such as about 1.4, and the muco-adhesive composition has a tan delta value less than I, such as less than 0.8, such as greater than 0.7, or such as about 0.7, and wherein the loss modulus and the storage modulus are measured at an angular frequency of 0.1 rad/s.
In one embodiment, the anhydrous composition comprises less than 1% by weight of water, such as less than 0.5% by weight, such as less than 0.2% by weight, such as less than 0.1% by weight, or such as less than 0.05% by weight.
In one embodiment, the anhydrous composition is an ointment or a paste. In one embodiment, the anhydrous composition is an ointment. In one embodiment, the ointment comprises a hydrocarbon base.
In one embodiment, the anhydrous composition is for intravaginal delivery. In one embodiment, the anhydrous composition is for intravaginal delivery using a syringe.
In one embodiment, the anhydrous composition is for topical delivery of the at least one active pharmaceutical ingredient to the cervix (e.g. as an ointment or paste) for preventing the development of, or treating, cervical cancer (e.g. caused by high-risk types of HPV such as HPV16).
muco-adhesive One advantage of the anhydrous formulation is that the hydrophilic muco-adhesive agent only becomes active upon topical application of the anhydrous composition to a site of application, such as preferably a mucosal membrane. In one embodiment, the site of application is an environment comprising water. Environments comprising water activate the hydrophilic muco-adhesive agent resulting in the anhydrous composition transforming to a muco-adhesive composition and in particular embodiments becoming a gel. The word gel describes the polymeric interaction between the hydrophilic muco-adhesive agent and the water. Advantageously, the resulting gel enables the composition to be retained at the site of application for a prolonged period.
In one embodiment, the site of application comprises a mucus membrane, for example the mucus membrane is selected from buccal mucosa, nasal mucosa, ocular mucosa, vaginal mucosa and rectal mucosa. The mucus membranes provide an aqueous environment and as such activates the hydrophilic muco-adhesive agent upon application of the anhydrous composition to the site of application, resulting in the prolonged retention of the at least one active pharmaceutical ingredient at the site of application, for example, activation of the hydrophilic muco-adhesive agent results in the anhydrous composition transforming into a muco-adhesive agent and becoming a gel.
In one embodiment, the hydrophilic muco-adhesive agent is present in the anhydrous composition at about 0.1 to about 20% by weight of the total anhydrous composition weight, such as about 0.1 to about 10% by weight, such as about 0.1 to about 5% by weight, such as about 0.2 to about 4% by weight, such as about 0.5 to about 3% by weight, such as about 0.5 to about 1.5% by weight, such as about 1% by weight, or such as 1% by weight.
In one embodiment, the hydrophilic muco-adhesive agent is a non-ionic polymer or an ionic polymer.
In one embodiment, the non-ionic polymer is a cellulose ether.
In one embodiment, the hydrophilic muco-adhesive agent is a cellulose ether or an ionic polymer.
In one embodiment, the cellulose ether is selected from methyl cellulose,
ethylcellulose and hydroxypropylmethylcellulose.
In one embodiment, the cellulose ether is hydroxypropylmethylcellulose.
In one embodiment, the hydroxypropylmethylcellulose has a degree of methoxy substitution of between 19 and 24% by weight and a degree of hydroxypropyl substitution of between 4 and 12% by weight.
In one embodiment, the ionic polymer is sodium polyacrylate. In one embodiment, the hydrophilic muco-adhesive has a viscosity of about 2000 to about 6000 cps, such as about 3000 to about 5000 cps, such as about 3500 to about 4500 cps, such as about 3800 to about 4200 cps, such as about 4000 cps, or such as 4000 cps.
Active Pharmaceutical Ingredient
In one embodiment, the at least one active pharmaceutical ingredient is a solid at room temperature. In one embodiment, the at least one active pharmaceutical ingredient is synthetically prepared. In one embodiment, the at least one active pharmaceutical ingredient is not a fatty acid (free or bound state).
In one embodiment, the at least one active pharmaceutical ingredient is present in a dissolved state in the anhydrous composition. In another embodiment, the at least one active pharmaceutical ingredient is present in a dispersed state in the anhydrous composition. In another embodiment, an amount of the least one active pharmaceutical ingredient is present in a dispersed state and an amount is present in a dissolved state in the anhydrous composition.
It will be apparent to the skilled person that the active pharmaceutical ingredient is dissolved or dispersed within the anhydrous composition by the use of techniques such as optical microscopy using polarised light filters, differential scanning calorimetry or micro FTIR. For example, a placebo anhydrous composition (i.e., a composition containing no active pharmaceutical ingredients) can be spiked with a crystalline active pharmaceutical ingredient. When viewed under an optical microscope using polarised light filters, the crystalline active pharmaceutical ingredient will exhibit birefringence. Thus, the spiked placebo composition can be used as a comparison standard in order to confirm there is no crystalline active pharmaceutical ingredient in the anhydrous composition, and thereby demonstrating the active pharmaceutical ingredient is dissolved within the anhydrous composition.
Alternatively, and/or additionally, micro FTIR can be used to confirm the active
pharmaceutical ingredient is dissolved in the anhydrous composition. In this case, spectra obtained for an anhydrous composition spiked with the active pharmaceutical ingredient and for the anhydrous composition can be compared and used to demonstrate the active pharmaceutical ingredient is dissolved in the anhydrous composition.
In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition. The compositions of the invention are particularly suitable for active pharmaceutical ingredients used in the composition that are prone to chemical or physical degradation. In one embodiment, the active pharmaceutical is prone to degradation due to hydrolysis. In one embodiment, the active pharmaceutical ingredient used in the composition is prone to degradation due to oxidation. In one embodiment, the active pharmaceutical ingredient used in the composition is prone to degradation which is accelerated by heat. In one embodiment, the active pharmaceutical ingredient used in the composition is prone to physical form changes, e.g. solid-state polymorphic transitions.
In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition for at least 3 months, such as at least 6 months, such as at least 9 months, such as at least 12 months, such as at least 18 months, such as at least 24 months, or such as at least 36 months, at a temperature of 5°C, 25°C, 30°C or at 40°C, and/or at a relative humidity of 60%, 65% or 75% RH.
In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition. In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed at room temperature. In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed at elevated temperatures, such as >30°C, such as >40°C, such as >50°C, such as >60°C, such as >65°C such as about 70°C. In one embodiment, the at least one active pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed under an inert atmosphere, such as a vacuum, such as a vacuum at about -0.5 bar. In one embodiment, the at least one active
pharmaceutical ingredient is stable within the anhydrous composition during a process to manufacture the anhydrous composition wherein the process is performed in the absence of water, for example wherein the process is performed under an insert atmosphere, such as a vacuum, such as a vacuum at about -0.5 bar.
In one embodiment, the at least one active pharmaceutical ingredient is present in the anhydrous composition between about 0.001 and about 50% by weight of the total anhydrous composition weight, such as between about 0.01 and about 50% by weight, such as between about 0.001 and about 5% by weight, such as between about 0.1 and about 25% by weight, such as between about 0.5 and about 15% by weight, such as between about 0.5 and about
10% by weight, such as between about 0.01 and about 2.5% by weight, such as between about
0.1 and about 2.5% by weight, such as between about 0.2 and about 2% by weight, such as between about 0.2 and about 1.5% by weight, such as between about 0.4 and about 1.5% by weight, such as between about 0.4 and about 0.6% by weight, such as about 0.5% by weight, such as 0.5% by weight, such as between about 0.5 and about 0.7% by weight, such as about 0.6% by weight, such as 0.6% by weight, such as between about 0.9 and about 1.1% by weight, such as about 1% by weight, such as 1% by weight, such as between about 1.2 and about 1.4% by weight, such as about 1.3% by weight, such as 1.3% by weight, such as between about 2 and about 20% by weight, such as between about 2 and about 15% by weight, such as between about 4 and about 12% by weight, such as between about 4 and about 6% by weight, such as about 5% by weight, such as 5% by weight, such as between about 5 and about 7% by weight, such as about 6% by weight, such as 6% by weight, such as between about 9 and about 11% by weight, such as about 10% by weight, such as 10% by weight, such as between about 11 and about 13% by weight, such as about 12% by weight, or such as 12% by weight.
In one embodiment, the at least one active pharmaceutical ingredient is classified as a Biopharmaceutics Classification System (BCS) Class II or a BCS Class IV active
pharmaceutical ingredient. A BCS class II active pharmaceutical ingredient is classed as an active ingredient having a high permeability and a low solubility. A BCS class IV active pharmaceutical ingredient is classed as an active ingredient having a low permeability and a low solubility. According to ICH guidelines (ICH guideline M9 on biopharmaceutics classification system based biowaivers; 6th August 2018) a drug substance is classified as highly soluble if the highest single therapeutic dose is completely soluble in 250 ml or less of aqueous media over the pH range of 1.2 - 6.8 at 37 ± l°C. The assessment of permeability should preferentially be based on the extent of absorption derived from human
pharmacokinetic studies, e.g., absolute bioavailability or mass balance. High permeability can be concluded when the absolute bioavailability is > 85%.
In one embodiment, the at least one active pharmaceutical ingredient is selected from a protease inhibitor, a retinoid, a vitamin D analog, an antileprosy active pharmaceutical ingredient, a calcineurin inhibitor, a cannabinoid, a 5 alpha-reductase inhibitor, an androgen receptor inhibitor, a peroxisome proliferator activated receptor activator, an antihistamine, a chloride channel activator, a tyrosine kinase inhibitor, a hormone, a protease inhibitor, and a mTOR kinase inhibitor.
In one embodiment, the at least one active pharmaceutical ingredient is selected from abacavir, efavirenz, enfuvirtide, nevirapine, ritonavir, lopinavir, tenofovir, adefovir, entecavir, ribavirin, acyclovir, famciclovir, penciclovir, valacyclovir, cidofovir, ganciclovir,
valganciclovir, oseltamivir, zanamivir, amprenavir, bexarotene, calcifediol, calcitriol, clofazimine, cyclosporin A, doxercalciferol, dronabinol, dutasteride, enzalutamide, fenofibrate, isotretinoin, loratadine, lubiprostone, nintedanib, paricalcitol, progesterone, saquinavir, sirolimus, tipranavir, tretinoin, atorvastatin, carvedilol, itraconazole, ketoprofen, and simvastatin.
In one embodiment, the at least one active pharmaceutical ingredient is selected from abacavir, efavirenz, enfuvirtide, nevirapine, ritonavir, lopinavir, tenofovir, adefovir, entecavir, ribavirin, acyclovir, famciclovir, penciclovir, valacyclovir, cidofovir, ganciclovir,
valganciclovir, oseltamivir and zanamivir.
In one embodiment, the anhydrous composition further comprises an active pharmaceutical ingredient synergist. In one embodiment, the active pharmaceutical ingredient synergist is a HIV protease enzyme inhibitor.
In one embodiment, the at least one active pharmaceutical ingredient is a HIV protease enzyme inhibitor. In one embodiment, the HIV protease enzyme inhibitor is selected from lopinavir and ritonavir.
Lopinavir (CAS# 192725-17-0) is a protease inhibitor chemically designated as [1S- [lR*(R*), 3R*, 4R*]]-N-[4-[(2,6-dimethylphenoxyacetyl]amino]-3-hydroxy-5-phenyl-l- (phenylmethyl)pentyl]tetrahydro-alpha-(l -methylethyl)-2-oxo- 1 (2H)-pyrimidineacetamide. It has the molecular formula C37H48N405 and a molecular weight of 628.80.
Ritonavir (CAS# 155214-67-5) is a protease inhibitor chemically designated as 10- Hydroxy-2-methyl-5-(l-methylethl)-l-[2-(l-methylethyl)-4-thiazolyl]-3,6-dioxo-8,
Hbis(phenylmethyl)-2,4,7,l2-tetraazatridecan-l3-oic acid, 5-thiazolylmethylester, [5S- (5R*,8R*,l0R8,l 1R*)]. It has the molecular formula C37H48N605S2 and a molecular weight of 720.95.
In one embodiment, the anhydrous composition comprises lopinavir and ritonavir. In one embodiment, the molar ratio of lopinavir to ritonavir present in the composition is between about 1 : 10 and about 18: 1, such as between about 1 : 10 and about 15: 1, such as between about 1 :5 and about 15: 1, such as between about 1 : 1 and about 15: 1, such as between about 2: 1 and about 15: 1, such as between about 4: 1 and about 15: 1, such as between about
8: 1 and about 14: 1, such as between about 9: 1 and about 14: 1, such as between about 10: 1 and about 14:1, such as between 10.5: 1 and about 18: 1, such as between 10.5: 1 and 18: 1, such as between about 10.5: 1 and about 14: 1, such as between about 11 : 1 to about 13: 1, such as between about 11.5 and about 17:1, such as between about 11.5: 1 and about 16.0: 1, such as between about 11.5: 1 and about 15: 1, such as about 14.5: 1, such as 14.5: 1, such as about
14: 1, such as 14: 1, such as about 13.8: 1, such as 13.8: 1, such as about 13.75: 1, such as
13.75: 1, such as about 13.5: 1, such as 13.5: 1, such as about 13: 1, such as 13:1, such as about
12.5: 1, such as 12.5: 1, such as about 12: 1, such as 12: 1, such as about 11.75: 1, such as 11.75: 1, such as about 9: 1, such as 9: 1, such as about 5: 1, such as 5: 1, such as about 4.6: 1, or such as 4.6: 1.
It will be appreciated that lopinavir has a molecular weight of 628.8 daltons and ritonavir has a molecular weight of 720.95 daltons. Accordingly molar ratios and w/w ratios will not be the same and a factor of 0.872 should be applied when converting molar ratios to w/w.
In one embodiment, lopinavir is present in the anhydrous composition at about 0.01 to about 50% of the total anhydrous composition weight, such as about 0.1 to about 25% by weight, such as about 2 to about 20% by weight, such as about 2 to about 15% by weight, such as about 4 to about 12% by weight, such as about 4 to about 6% by weight, such as at about 5% by weight, such as 5% by weight, such as about 5 to about 7% by weight, such as about 6% by weight, such as 6% by weight, such as about 9 to about 11% by weight, such as at about 10% by weight, such as 10% by weight, such as about 11 to about 13% by weight, such as about 12% by weight, or such as 12% by weight.
In one embodiment, ritonavir is present in the anhydrous composition at about 0.001 to about 5% by weight of the total anhydrous composition weight, such as about 0.01 to about 2.5% by weight, such as about 0.1 to about 2.5% by weight, such as about 0.2 to about 2% by weight, such as about 0.2 to about 1.5% by weight, such as about 0.4 to about 1.5% by weight, such as about 0.4 to about 0.6% by weight, such as at about 0.5% by weight, such as at 0.5% by weight, such as about 0.9 to about 1.1% by weight, such as about 1% by weight, such as 1% by weight, such as about 1.2 to 1.4% by weight, such as about 1.3% by weight, such as 1.3% by weight, such as about 0.5 to about 0.7% by weight, such as about 0.6% by weight, or such as 0.6% by weight.
Solvent for the at least one active pharmaceutical ingredient
In one embodiment, the anhydrous composition further comprises a solvent for the at least one active pharmaceutical ingredient.
In one embodiment, the solvent is selected from an unsaturated free fatty acid, PEG castor oil, diethylene glycol, monoethyl ether, propylene glycol, polyethylene glycol, and a medium chain triglyceride. Fatty acids are usually derived from triglycerides or
phospholipids. Glycerol has three hydroxyl functional groups, which can be esterified with one, two, or three fatty acids to form mono-, di-, or triglycerides respectively. Phospholipid molecules consist of two hydrophobic fatty acid“tails” and a hydrophilic“head” consisting of a phosphate group. These two components are joined together by a glycerol molecule. Both triglyceride and phospholipids comprise fatty acids in a bound state. Conversely, free fatty acids are fatty acids which are not bound, that is they are not esterified. An unsaturated free fatty acid is a free fatty acid wherein there is at least one double bond between carbon atoms in the fatty acid. In one embodiment, the solvent is an unsaturated free fatty acid.
In one embodiment, the unsaturated free fatty acid is selected from oleic acid, linoleic acid, alpha-linoleic acid, palmitoleic acid, gondoic acid, and ricinoleic acid.
In one embodiment, the unsaturated free fatty acid is oleic acid.
In one embodiment, of the total unsaturated fatty acid (bound and free form
unsaturated fatty acid) present within the composition, at least 90% by weight, such as at least 95% by weight, such as at least 98% by weight, such as at least 99% by weight, or such as at least 99.5% by weight, is in the free form, i.e., not esterified or bound to other components such as glycerol. The skilled person would be aware of methods used to determine the free fatty acid content versus the total fatty acid content. For example, the free fatty acid content can be measured by reacting the free fatty acid with a chromogeneous compound, thus changing the frequency that the chromogeneous compound absorbs electromagnetic radiation. Thus, the concentration of the chromogeneous compound reacted can be determined by monitoring the chromogeneous compound using a suitable wavelength which in turn can be used to determine the free fatty acid content in the sample.
It is to be understood that free fatty acids products that are commercially available may contain small amounts of other free fatty acids. For example, oleic acid typically contains 7-12% saturated free fatty acids, such as stearic and palmitic acid, together with other unsaturated free fatty acids, such as linoleic acid (Handbook of Pharmaceutical Excipients, 2nd Edition, see entry for Oleic acid). The terms saturated free fatty acid or unsaturated free fatty acid are to be understood as meaning the saturated free fatty acid or the unsaturated free fatty acid are of Pharmacopeia grade, such as the ETS Pharmacopeia and/or the British
Pharmacopeia, and that the saturated free fatty acid or unsaturated free fatty acid may contain small amounts of other free fatty acids.
In one embodiment, the unsaturated free fatty acid is not in the form of a triglyceride or polysorbate.
In one embodiment, the at least one active pharmaceutical ingredient has a solubility measured at ambient temperature of at least 1% w/v in the solvent, such as at least 5% w/v in the solvent, such as at least 10% w/v in the solvent, such as at least 12% w/v in the solvent, such as at least 15% w/v in the solvent, or such as at least 18% w/v in the solvent.
In one embodiment, the solvent is present in the anhydrous composition at a level of at least 25% by weight of the total anhydrous composition weight, such as at least 35% by weight, such as at least 45% by weight, such as about 25 to about 75% by weight, such as about 55 to about 75% by weight, such as about 45 to about 75% by weight, such as about 50 to about 70% by weight, such as about 55 to about 70% by weight, such as about 55 to about 65% by weight, such as about 50 to about 60% by weight, such as about 25 to about 60% by weight, or such as about 50 to about 65% by weight.
In one embodiment, the solvent is present in the anhydrous composition at a level of about 55% by weight of the total anhydrous composition weight, such as about 57% by weight, such as about 58% by weight, such as about 60% by weight, such as about 62% by weight, such as about 63% by weight, such as about 65% by weight, such as about 67% by weight, or such as about 68% by weight.
Figure imgf000032_0001
Optionally, additional excipients may be included in the composition according to the present invention providing that inclusion of such excipients does not unacceptably impact the ability of the composition, upon topical administration to a site of application, to transform into a muco-adhesive composition.
In one embodiment, the anhydrous composition further comprises a thickener. A thickener is an excipient which when added to a mixture increases the viscosity of the mixture and confers the anhydrous composition with greater physical stability and/or control during delivery of the active pharmaceutical ingredient to the site of application.
In one embodiment, the thickener is selected from mono di glyceride, ceresin wax, and hydrogenated vegetable oil or a combination thereof.
In one embodiment, the anhydrous composition comprises mono di glyceride, ceresin wax, and hydrogenated vegetable oil.
In one embodiment, the anhydrous composition further comprises a stiffening agent. The stiffening agent is an excipient used to stiffening the composition so that the anhydrous composition is a semi-solid at room temperature. In one embodiment, the stiffening agent is a solid at room temperature. Conveniently, the stiffening agent is a saturated free fatty acid, such as a C10-C38 saturated free fatty acid, such as a C16-C22 saturated free fatty acid. A saturated free fatty acid, is a free fatty acid (i.e., the fatty acid is not bound to another molecule, such as glycerol) wherein there are no double bonds between the carbon atoms in the fatty acid.
In one embodiment, the stiffening agent is selected from capric acid, undecylic acid, lauric acid, tridecylic acid, myristic acid, pentadecylic acid, palmitic acid, margaric acid, stearic acid, nonadecylic acid, arachidic acid, heneicosylic acid, behenic acid, tricosylic acid, lignoceric acid, pentacosylic acid, cerotic acid, heptacosylic acid, montanic acid, nonacosylic acid, melissic acid, henatriacontylic acid, lacceroic acid, psyllic acid, geddic acid, ceroplastic acid, hexatriacontylic acid, heptatriacontanoic acid and octatriacontanoic acid.
In one embodiment, the stiffening agent is stearic acid. Stearic acid is a straight chained saturated fatty acid with an 18-carbon chain.
In one embodiment, of the total saturated fatty acid (bound and free form saturated fatty acid) present within the composition, at least 90% by weight, such as at least 95% by weight such as at least 98% by weight, such as at least 99% by weight, or such as at least 99.5% by weight, is in the free form, i.e., not esterified or bound to other components such as glycerol. The skilled person would be aware of methods used to determine the free fatty acid content versus the total fatty acid content. For example, the free fatty acid content can be measured by reacting the free fatty acid with a chromogeneous compound, thus changing the frequency that the chromogeneous compound absorbs electromagnetic radiation. Thus, the concentration of the chromogeneous compound reacted can be determined by monitoring the chromogeneous compound using a suitable wavelength which in turn can be used to determine the free fatty acid content in the sample.
In one embodiment, the saturated free fatty acid is not in the form of a triglyceride or polysorbate.
In one embodiment, of the total fatty acid (unsaturated and saturated fatty acids in the bound and free states) present within the composition, at least 90% by weight, such as at least 95% by weight, such as at least 98% by weight, such as at least 99% by weight, or such as at least 99.5% by weight, is in the free form, i.e., not esterified or bound to other components such as glycerol. The skilled person would be aware of methods used to determine the free fatty acid content versus the total fatty acid content. For example, the free fatty acid content can be measured by reacting the free fatty acid with a chromogeneous compound, thus changing the frequency that the chromogeneous compound absorbs electromagnetic radiation. Thus, the concentration of the chromogeneous compound reacted can be determined by monitoring the chromogeneous compound using a suitable wavelength which in turn can be used to determine the free fatty acid content in the sample.
In one embodiment, the saturated free fatty acid and the unsaturated free fatty acid are not in the form of a triglyceride or polysorbate.
In one embodiment, the anhydrous composition further comprises oleic acid and stearic acid. In one embodiment, the anhydrous composition further comprises oleic acid, stearic acid, and hydroxypropylmethylcellulose.
In one embodiment, the anhydrous composition further comprises oleic acid, stearic acid, mono di glyceride, ceresin wax, and hydrogenated vegetable oil.
In one embodiment, the amount of thickener present in the anhydrous composition is at least about 0.1% of the total anhydrous composition weight, such as at least 1% by weight, such as at least about 3% by weight, such as about 0.1 to about 40% by weight, such as about 3 to about 40% by weight, such as about 3 to about 35% by weight, such as about 5 to about 21% by weight, such as about 4 to about 12% by weight, such as about 4 to about 7% by weight, such as about 0.1 to about 10% by weight, such as about 0.1 to about 20% by weight, such as about 5% by weight, such as 5% by weight, such as about 6% by weight, such as 6% by weight, such as about 8 to about 12% by weight, such as about 10% by weight, such as 10% by weight, such as about 10 to about 35% by weight, such as about 15 to about 35% by weight, such as about 20 to about 30% by weight, such as about 21% by weight, or such as 21% by weight.
In one embodiment, the mono di glyceride is present in the anhydrous composition at about 0.1 to about 30% by weight of the total anhydrous composition weight, such as about 0.1 to about 10% by weight, such as about 1 to about 10% by weight, such as about 3 to about 7% by weight, such as about 4 to about 6% by weight, such as about 5% by weight, or such as 5% by weight.
In one embodiment, the ceresin wax is present in the anhydrous composition at about 0.1 to about 30% by weight of the total anhydrous composition weight, such as about 0.1 to about 10% by weight, such as about 1 to about 10% by weight, such as about 4 to about 8% by weight, such as about 5 to about 7% by weight, such as about 5% by weight, such as about 6% by weight, or such as 6% by weight.
In one embodiment, the hydrogenated vegetable oil is present in the anhydrous composition at about 0.1 to about 30% by weight of the total anhydrous composition weight, such as about 0.1 to about 20% by weight, such as about 5 to about 15% by weight, such as about 8 to about 12% by weight, such as about 9 to about 11% by weight, such as about 10% by weight, or such as 10% by weight.
In one embodiment, the stiffening agent is present in the anhydrous composition at about 0.1 to about 65% by weight of the total anhydrous composition weight, such as about 3 to about 60% by weight, such as about 3 to about 50% by weight, such as about 3 to about
40% by weight, such as about 3 to about 35% by weight, such as about 3 to about 6% by weight, such as about 4 to about 5% by weight, such as about 4.5% by weight, such as 4.5% by weight, such as about 15 to about 25% by weight, such as about 18 to about 22% by weight, such as about 20% by weight, such as about 20 to about 30% by weight, such as about 24 to about 30% by weight, such as about 26% by weight, such as about 25 to about 35% by weight, such as about 28 to about 32% by weight, such as about 30% by weight, such as 30% by weight, such as about 32% by weight, or such as about 34% by weight.
In one embodiment, the stearic acid is present in the anhydrous composition at about 0.1 to about 65% by weight of the total anhydrous composition weight, such as about 3 to about 40% by weight, such as about 3 to about 35% by weight, such as about 3 to about 6% by weight, such as about 4 to about 5% by weight, such as about 4.5% by weight, such as 4.5% by weight, such as about 15 to about 25% by weight, such as about 18 to about 22% by weight, such as about 20% by weight, such as about 20 to about 30% by weight, such as about 24 to about 30% by weight, such as about 26% by weight, such as about 25 to about 35% by weight, such as about 28 to about 32% by weight, such as about 30% by weight, such as 30% by weight, such as about 32% by weight, or such as about 34% by weight.
In one embodiment, the anhydrous composition further comprises a blending agent. A blending agent is an agent which promotes uniformity within the anhydrous composition, for example it promotes uniformity for organoleptic attributes within the anhydrous composition.
In one embodiment, the blending agent is selected from polyoxy 100 stearate and glycerol monooeleate. In one embodiment, the composition comprises polyoxy 100 stearate and glycerol monooeleate.
In one embodiment, the blending agent is present in the anhydrous composition at at least 0.1% by weight of the total anhydrous composition weight, such as at least 1% by weight, such as at least 1.5% by weight, such as about 0.1 to about 48% by weight, such as about 0.1 to about 38% by weight, such as about 0.1 to about 10% by weight, such as 0.1 to about 3% by weight, such as about 1 to about 10% by weight, such as about 2 to about 5% by weight, such as about 2% by weight, such as 2% by weight, such as about 3% by weight, such as 3% by weight, such as about 5% by weight, or such as 5% by weight.
In one embodiment, the polyoxy 100 stearate is present in the anhydrous composition at about 0.1 to about 38% by weight of the total anhydrous composition weight, such as at about 0.1 to about 10% by weight, such as about 0.5 to about 5% by weight, such as about 1 to about 3% by weight, such as about 2% by weight, or such as 2% by weight.
In one embodiment, the glycerol monooleate is present in the anhydrous composition at about 0.1 to about 10% by weight of the total anhydrous composition weight, such as about 0.5 to about 5% by weight, such as about 2 to about 4% by weight, such as about 3% by weight, or such as 3% by weight.
In one embodiment, the anhydrous composition comprises an antioxidant. In one embodiment, the antioxidant is butylated hydroxytoluene. In one embodiment, the antioxidant is present in the anhydrous composition at about 0.05 to about 0.5% by weight of the total anhydrous composition weight, such as about 0.1 to about 0.3% by weight, such as about 0.2% by weight, such as 0.2% by weight, such as about 0.1% by weight, or such as 0.1% by weight.
In one embodiment, the anhydrous composition further comprises oleic acid, stearic acid, mono di glyceride, ceresin wax, hydrogenated vegetable oil, polyoxyl 100 stearate, glycerol monooleate, and butylatedhydroxytoluene.
MEDICAL USES
In one embodiment, the anhydrous composition is for use as a medicament.
In one embodiment, the anhydrous composition is for use as a medicament wherein the anhydrous composition is applied topically. In one embodiment, the anhydrous composition is for use as a medicament wherein the anhydrous composition is topically applied to the part of the body in need of the treatment. In one embodiment, the anhydrous composition is applied topically to an environment comprising water. In one embodiment, the anhydrous composition is topically applied to skin. In one embodiment, the anhydrous composition is topically applied to mucosa. In one embodiment, the anhydrous composition is for use as a medicament wherein the anhydrous composition is topically applied to the cervix. This is particularly advantageous as the patient can self-administer the anhydrous composition and/or treatment with the anhydrous composition negates the need of treatment by surgery.
In one embodiment, the anhydrous compositions are useful in the treatment and/or prevention of diseases and/or disorders. In one embodiment, the anhydrous compositions are useful in the treatment of benign proliferative disorders.
In one embodiment, the anhydrous compositions are useful in the treatment of cancer and particularly useful for preventing the development of cancers. Accordingly, normal subjects (i.e. subjects with no detectable cancer), subjects with pre-malignant cells or particularly cancer prone subjects may be treated by topical administration of the anhydrous compositions according to the invention with a view to preventing the development of cancer. When used to treat (or prevent the development of) cervical cancer, the anhydrous compositions are pastes or ointments and may be applied directly to the cervix by techniques known to the art.
Disclosed herein are anhydrous compositions comprising lopinavir and ritonavir for use as a medicament in the treatment of cancer or benign proliferative disorders (e.g. warts) or in the prevention of the development of cancer.
The invention, to the extent that it is applicable to the prevention and treatment of cancer, may be applied to a wide range of cancers such as ovarian carcinoma, breast carcinoma, lung carcinoma, uterine carcinoma, cervical carcinoma and thyroid carcinoma. It is also applicable to cancer prone conditions. The invention is applicable particularly, but by no means exclusively, to pre-cancerous conditions and cancers caused by oncogenic viruses, e.g. high-risk or even low-risk forms of human papilloma viruses (HPVs).
Conveniently, the compositions may be administered to treat, and particularly prevent, the development of cervical cancer. Conveniently, the inhibitors are used to treat, or prevent the development of cervical cancers caused by HPV (particularly high-risk types of HPV such as HPV16 or HPV 18).
The compositions may be used to prevent or treat cancer as a monotherapy (i.e.
including the use of the anhydrous composition comprising two or more active
pharmaceutical ingredients) or in combination with other compounds or treatments used in cancer therapy (e.g. chemotherapeutic agents, radiotherapy).
Conveniently, the compositions are used to treat humans. However, it will be appreciated that the compositions may also have some veterinary use.
DOSING
It will be appreciated that the amount of the at least one active pharmaceutical ingredient required is determined by biological activity and bioavailability, which in turn depends, in part, on the precise mode of administration, the physicochemical properties of the anhydrous composition employed, and whether the anhydrous compositions are being used as a monotherapy or in a combined therapy with other oral or topical medicines. Indeed, it is also possible that the at least one active pharmaceutical ingredient could be topically applied in addition to oral dosing of the same compounds or other active pharmaceutical ingredient(s). The frequency of administration will also be influenced by the abovementioned factors and particularly the half-life of the active pharmaceutical ingredients within the subject being treated. Daily doses may be given as a single administration. Alternatively, administration may be twice or more times during a day. As an example, the anhydrous compositions may be topically administered at least once a day, such as once a day, or such as twice a day.
Optimal dosages to be administered may be determined by those skilled in the art, and will vary with the strength of the preparation, the mode of administration, and the
advancement of the disease condition. Additional factors depending on the particular subject being treated will result in a need to adjust dosages, including, for example, subject age, weight, gender, diet, and time of administration.
Suitable amounts of the at least one active pharmaceutical ingredient to be given as a daily dose are of about 0.01 mg to about 10 g, such as about 0.1 mg to about 10 g, such as about 1 mg to about 5g, such as about 1 mg to about 1 g, such as about 5 mg to about 2 g, such as about 10 mg to about 1 g, such as about 5 mg to about 500 mg, such as about 10 mg to about 500 mg, such as about 10 mg to about 400 mg, such as about 5 mg to about 200 mg, such as about 5 mg to about 50 mg, such as about 10 mg to about 40 mg, such as about 20 mg to about 40 mg, such as about 25 mg to about 35 mg, such as about 27 mg to about 32 mg, such as about 29 mg, such as 29 mg, such as about 28.7 mg, such as 28.7 mg, such as about 15 mg to about 35 mg, such as about 20 mg to about 30 mg, such as about 23 mg to about 27 mg, such as about 25 mg, such as 25 mg, such as about 5 mg to about 25 mg, such as about 10 mg to about 20 mg, such as about 12 mg to about 16 mg, such as about 14 mg, such as 14 mg, such as about 14.3 mg, such as about 14.3 mg, such as about 11 mg to about 15 mg, such as about 13 mg, such as 13 mg, such as about 12.5 mg, such as 12.5 mg, such as about 100 mg to about 400 mg, such as about 200 mg to about 400 mg, such as about 250 mg to about 350 mg, such as about 280 mg to about 320 mg, such as about 290 mg to about 310 mg, such as about 300 mg, such as 300 mg, such as about 25 mg to about 325 mg, such as about 50 mg to about 250 mg, such as about 125 mg to about 175 mg, such as about 140 mg to about 160 mg, such as about 150 mg, or such as 150 mg.
Suitable amounts of lopinavir to be given as a daily dose of about 0.01 mg to about 10 g, such as about 0.1 mg to about 10 g, such as about 1 mg to about 5 g, such as about 5 mg to about 2 g, such as about 10 mg to about 1 g, such as about 10 mg to about 500 mg, such as about 10 mg to about 400 mg, such as about 100 mg to about 400 mg, such as about 200 mg to about 400 mg, such as about 250 mg to about 350 mg, such as about 280 mg to about 320 mg, such as about 290 mg to about 310 mg, such as about 300 mg, such as 300 mg, such as about 25 mg to about 325 mg, such as about 50 mg to about 250 mg, such as about 125 mg to about 175 mg, such as about 140 mg to about 160 mg, such as about 150 mg, or such as 150 mg.
Suitable amounts of ritonavir to be given as a daily dose of about 0.01 mg to about 10 g, such as about 0.1 mg to about 10 g, such as about 1 mg to about 5 g, such as about 1 mg to about 1 g, such as about 5 mg to about 500 mg, such as about 5 mg to about 200 mg, such as about 5 mg to about 50 mg, such as about 10 mg to about 40 mg, such as about 20 mg to about 40 mg, such as about 25 mg to about 35 mg, such as about 27 mg to about 32 mg, such as about 29 mg, such as 29 mg, such as about 28.7 mg, such as 28.7 mg, such as about 15 mg to about 35 mg, such as about 20 mg to about 30 mg, such as about 23 mg to about 27 mg, such as about 25 mg, such as 25 mg, such as about 5 mg to about 25 mg, such as about 10 mg to about 20 mg, such as about 12 mg to about 16 mg, such as about 14 mg, such as 14 mg, such as about 14.3 mg, such as 14.3 mg, such as about 11 mg to about 15 mg, such as about 13 mg, such as 13 mg, such as about 12.5 mg, or such as 12.5 mg.
In one embodiment, about 300 mg of lopinavir and about 29 mg ritonavir per day may be administered to the cervix of a woman.
In one embodiment, about 300 mg of lopinavir and about 25 mg ritonavir per day may be administered to the cervix of a woman.
In another embodiment, about 150 mg of lopinavir and about 14 mg ritonavir per day may be administered to treat the cervix of a woman.
In another embodiment, about 150 mg of lopinavir and about 13 mg ritonavir per day may be administered to treat the cervix of a woman.
In an embodiment about 3.0 g of the anhydrous composition may be administered to a subject per day. Such dosage forms may comprise about 300 mg of lopinavir and about 29 mg of ritonavir; or about 150 mg of lopinavir and about 14 mg of ritonavir.
In another embodiment, about 2.5 g of the anhydrous composition may be
administered to a subject per day. Such dosage forms may comprise about 300 mg of lopinavir and about 25 mg of ritonavir; or about 150 mg of lopinavir and about 12.5 mg of ritonavir.
In an embodiment about 3.0 g of the anhydrous composition disclosed in Table 3 or Table 4 is administered to the cervix by a syringe applicator as a once per day application (preferably in the evening before retiring for the night).
In a most preferred embodiment about 2.5 g of the anhydrous composition disclosed in Table 6 or Table 7 is administered to the cervix by a syringe applicator as a once per day application (preferably in the evening before retiring for the night). TREATMENT REGIMENS
The anhydrous composition may be administered to a subject for as long as treatment is required. The length of time for which treatment will be required will depend upon the exact condition being treated or prevented and its severity. A skilled person will appreciate that treatment should be maintained in view of a number of factors which will include any requirement to eradicate the disease or disorder.
In one embodiment, a course of treatment may be for 2 - 4 weeks, 7-21 days or for about 14 days. After this time a clinician may assess whether the course of treatment has been successful. A decision may then be made whether or not to continue treatment.
It will be appreciated that a clinician may wish to take into account menstruation when deciding on a treatment regimen for women with conditions relating to the cervix.
Accordingly, a treatment regimen may be for about 14 - 21 days and can be administered between menses. A clinician may elect to stop topical treatment of the cervix during menses and recommence a new course of treatment in the next menstrual cycle. By way of example, a treatment regimen can be: (1) 14 - 21 days of administration; (2) followed by 1 - 14 days without treatment (during which menses may occur if treating the cervix); and (3) a further cycle of 14 -21 days of treatment if this is considered medically necessary.
TREATMENT OF HPV RELATED DYSPLASIA
Conveniently, the anhydrous compositions may be used to treat female subjects having an HPV related dysplasia of the cervix.
As used herein,“dysplasia” encompasses pre-invasive lesions and cancer. HPV related pre-invasive lesions include high grade squamous intraepithelial lesion (HSIL), atypical squamous cells of undetermined significance (ASCUS), and low grade squamous intraepithelial lesion (LSIL). HPV related cancers include, for example, cervical
intraepithelial neoplasia (CIN) and invasive cervical cancer (ICC).
The disclosed methods and treatment regimens can be used to treat HPV related dysplasia. In some aspects, for example, the disclosed methods and treatment regimens can be used to treat HSIL. In some aspects, the disclosed methods and treatment regimens can be used to treat ASCUS. In other aspects, the disclosed methods and treatment regimens can be used to treat LSIL. In other aspects, the disclosed methods and treatment regimens can be used to treat CIN. In yet other embodiments, the disclosed methods and treatment regimens can be used to treat ICC. Additionally, the disclosed methods and treatment regimens can be used to inhibit the progression of HPV related dysplasia. In some aspects, for example, the disclosed methods and treatment regimens can be used to inhibit the progression of HSIL. In some aspects, the disclosed methods and treatment regimens can be used to inhibit the progression of ASCUS. In other aspects, the disclosed methods and treatment regimens can be used to inhibit the progression of LSIL. In other aspects, the disclosed methods and treatment regimens can be used to inhibit the progression of CIN. In yet other embodiments, the disclosed methods and treatment regimens can be used to inhibit the progression of ICC.
In one embodiment, there is provided a method of treating a patient having an HPV related dysplasia of the cervix comprising administering intravaginally to said patient a therapeutically effective dose of an anhydrous composition according to the first aspect.
In one embodiment, the anhydrous composition reduces the severity of the HPV related dysplasia. In one embodiment, the severity of the HPV is reduced from CIN3 to CIN2, from CIN3 to CIN1, from CIN3 to HPV negative, from CIN2 to CIN1, from CIN2 to HPV negative, or from CIN1 to HPV negative.
In one embodiment, the patient has a cervical cytology of high grade squamous intraepithelial lesion (HSIL), atypical squamous cells of undetermined significance (ASCUS), or low grade squamous intraepithelial lesion (LSIL).
In one embodiment, the anhydrous composition reduces the cervical cytology from HSIL to a normal cytology, from HSIL to ACSUS, from HSIL to LSIL, from ACSUS to a normal cytology, or from LSIL to a normal cytology.
In some embodiments, the composition induces apoptosis of HPV infected cells.
PROCESS OF MANUFACTURE
In a second aspect, there is provided a method of manufacturing the anhydrous composition of the first aspect comprising the step incorporating (e.g. mixing) a hydrophilic muco-adhesive agent into the anhydrous composition.
In one embodiment, there is provided a method of manufacturing the anhydrous composition of the first aspect comprising the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition.
In one embodiment, the step of mixing uses a low shear mixing process or a high pressure homogenizing process. Advantageously, using high pressure homogenizing (HPH) in the mixing step results in greater dispersion of the muco-adhesive agent within the anhydrous composition and a decrease in particle size resulting in the HPH generated anhydrous composition having an increase in product surface area and an increase in viscosity relative to the low shear generated anhydrous composition. This means that a lower level of hydroxypropylmethylcellulose can be used within the HPH generated anhydrous composition compared to the low shear generated anhydrous composition in order obtain compositions with equivalent rheological behaviour and/or viscosity.
In one embodiment, the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition is performed at room temperature. This has the advantage of the process being particularly well suited to active pharmaceutical ingredients which are unstable when exposed to temperatures above room temperature, for example >30°C, >40°C, >50°C, and >60°C.
In one embodiment, the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition is performed at a temperature greater than 30°C, such as greater than 40°C, such as greater than 50°C, such as greater than 60°C, such as greater than 65°C, such as about 70°C.
In one embodiment, the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition is performed at a temperature greater than the melting point of the thickener and/or stiffening agent, such as greater than 50°C, such as greater than 60°C, such as greater than 65°C, such as about 70°C.
In one embodiment, the method of manufacturing the anhydrous composition further comprises the step of mixing the at least one active pharmaceutical ingredient, a solvent and a stiffening agent together.
In one embodiment, the step of mixing the at least one active pharmaceutical ingredient, a solvent and a stiffening agent together uses low shear mixing.
In one embodiment, the step of mixing the at least one active pharmaceutical ingredient, a solvent and a stiffening agent together is performed at a temperature greater than the melting point of the stiffening agent.
In one embodiment, the step of mixing the at least one active pharmaceutical ingredient, a solvent and a stiffening agent together is performed at a temperature greater than the melting point of the stiffening agent and until a clear and transparent melt is obtained.
In one embodiment, the method of manufacturing the anhydrous composition further comprises the step of mixing the at least one active pharmaceutical ingredient, a solvent a thickener, and a stiffening agent together.
In one embodiment, the step of mixing the at least one active pharmaceutical ingredient, a solvent, a thickener, and a stiffening agent together uses low shear mixing. In one embodiment, the step of mixing the at least one active pharmaceutical ingredient, a solvent, a thickener, and a stiffening agent together is performed at a temperature greater than the melting point of the thickener and/or stiffening agent.
In one embodiment, the step of mixing the at least one active pharmaceutical ingredient, a solvent, a thickener, and a stiffening agent together is performed at a temperature greater than the melting point of the thickener and/or stiffening agent and until a clear and transparent melt is obtained.
In one embodiment, the step of mixing a hydrophilic muco-adhesive agent into the anhydrous composition using a low shear mixing process is performed until the muco- adhesive agent is dispersed within the anhydrous composition.
In one embodiment, the process further comprises a cooling step. In one embodiment, the cooling step comprises cooling the mixture to a temperature less than 70°C, such as less than 60°C, such as less than 50°C, such as about 45°C.
In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent and a stiffening agent together; and
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i.
In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent and a stiffening agent together using low shear mixing; and
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing.
In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent and a stiffening agent together using low shear mixing at a temperature greater than the melting point of the stiffening agent; and
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing. In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent and a stiffening agent together using low shear mixing at a temperature greater than the melting point of a stiffening agent;
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing; and
iii. cooling the mixture from step ii. while maintaining low shear mixing.
In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent and a stiffening agent together using low shear mixing at a temperature greater than the melting point of the stiffening agent;
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing so that the muco-adhesive is dispersed within the anhydrous composition; and
iii. cooling the mixture from step ii. while maintaining low shear mixing.
In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent and a stiffening agent together using low shear mixing at a temperature greater than the melting point of the stiffening agent until a clear and transparent melt is obtained;
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing so that the muco-adhesive is dispersed within the anhydrous composition; and
iii. cooling the mixture from step ii. while maintaining low shear mixing.
In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent, a thickener and a stiffening agent together; and
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent, a thickener and a stiffening agent together using low shear mixing; and
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing.
In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent, a thickener and a stiffening agent together using low shear mixing at a temperature greater than the melting point of the stiffening agent and/or thickener; and
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing.
In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent, a thickener and a stiffening agent together using low shear mixing at a temperature greater than the melting point of the stiffening agent and/or thickener; ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing; and
iii. cooling the mixture from step ii. while maintaining low shear mixing.
In one embodiment, the method of manufacturing the anhydrous composition of the first aspect comprises:
i. mixing at least one active pharmaceutical ingredient, a solvent and a stiffening agent together using low shear mixing at a temperature greater than the melting point of the stiffening agent and the thickener until a clear and transparent melt is obtained;
ii. mixing a hydrophilic muco-adhesive agent into the mixture from step i. using low shear mixing so that the muco-adhesive is dispersed within the anhydrous composition; and
iii. cooling the mixture from step ii. while maintaining low shear mixing. In one embodiment, the method of manufacture is performed under an inert atmosphere. In one embodiment, the inert atmosphere is provided by a vacuum. In a further embodiment, the vacuum is about -0.5 bar. Performing the method of manufacture under an inert atmosphere has the advantage that any components of the anhydrous composition, such as the at least one active pharmaceutical ingredient, that are prone to degradation, such as oxidative or hydrolysis degradation, are less likely to degrade during the manufacturing process.
DOSING APPARATUS
In one aspect, there is provided a syringe comprising the anhydrous composition of the first aspect. In one embodiment, the syringe is for vaginal dosing of the anhydrous composition.
In a further aspect, there is provided a kit of parts, the kit comprising instructions for use and a syringe comprising the anhydrous composition of the first aspect. In one embodiment, there is provided a kit of parts, the kit comprising instructions for use and a syringe comprising the anhydrous composition, the syringe for vaginal dosing of the anhydrous composition.
In a further aspect, there is provided a tube comprising the anhydrous composition of the first aspect. In one embodiment, the tube is an aluminium tube.
In a further aspect, there is provided a kit of parts, the kit comprising instructions for use and a tube comprising the anhydrous composition of the first aspect. In one embodiment, the tube is an aluminium tube.
EXAMPLES
The following abbreviations have been used in the examples:
API - active pharmaceutical ingredient
HPMC - hydroxypropylmethylcellulose
PEG - polyethylene glycol
Example 1: Solubility of Ritonavir and/or Lopinavir
The solubility of Ritonavir and Lopinavir, and Ritonavir alone in a range of pharmaceutically acceptable solvents is presented below. As can be seen, Ritonavir and Lopinavir, and Ritonavir alone have high solubility in oleic acid.
Figure imgf000047_0001
'Empirically determined, by mass weighing, at ambient temperature, using Lopinavir/Ritonavir API blend molar ratio of 9: 1 2Based on oleic acid specific gravity of 0.9
3 Solubility for Ritonavir only, measured at 37°C, Lei et al.Development of a novel self-micro-emulsifying drug delivery system (SMEDD S) for reducing HIV protease inhibitor-induced intestinal epithelial barrier dysfunction”, Mol. Pharm. 2010 Jun 7:7(3) 844-853
Example 2: Ritonavir and Lopinavir chemical and physical stability
It is known from the literature that potency loss for Lopinavir and Ritonavir is thermolytically driven and Ritonavir is particularly prone to hydrolysis due to its carbomate functional group (Donato et al,“LC Method for Studies on the Stability of Lopinavir and Ritonavir in Soft Gelatin Capsules”, Chromatographia, April 2006, 63, 437-443). Ritonavir is also known to be oxygen labile. A preferable manufacturing route for compositions comprising
Lopinavir/Ritonavir is thus one that excludes heat, water, and/or oxygen.
Example 3: Preparation of formulations
For all formulations presented below, all materials used are pharmaceutical grade (either US Pharmacopeia or European Pharmacopeia) except for white ceresin wax, which is Japanese Pharmaceutical Excipient grade. The oleic acid used was super refined grade (Supplier: Croda). Formulation 3a: i mixture (oleic acid and stearic ; full and half strength- molar ratio of lo
Figure imgf000048_0001
to ritonavir 9:1
The manufacture of binary mixture of oleic acid and stearic acid active batch with rheology suitable for topical dosing is described below in accordance with Table 1 :- i. Into main vessel add 1, 4. Stir at 60 rpm and periodically homogenize at 600 rpm to dissolve. -0.5 bar vacuum applied.
ii. Into main vessel add 5, 6. Stir at 60 rpm and periodically homogenize at 600 rpm to dissolve. -0.5 bar vacuum applied.
iii. Into main vessel add 2. Stir at 60 rpm and periodically homogenize at 600 rpm to disperse. -0.5 bar vacuum applied.
iv. Into main vessel add 3. Stir at 60 rpm and periodically homogenize at 600 rpm to combine. -0.5 bar vacuum applied.
v. Discharge the product into well labelled polypail, awaiting packing into aluminium tubes.
Formulation 3b: Binary mixture (oleic acid and stearic acid); placebo
The manufacture of a placebo batch containing a binary mixture of oleic acid and stearic acid with rheology suitable for topical dosing is described below in accordance with Table 2:- i. Into main vessel add 1, 4. Stir at 60 rpm and periodically homogenize at 600 rpm to dissolve. -0.5 bar vacuum applied.
ii. Into main vessel add 2. Stir at 60 rpm and periodically homogenize at 600 rpm to disperse. -0.5 bar vacuum applied.
iii. Into main vessel add 3. Stir at 60 rpm and periodically homogenize at 600 rpm to combine. -0.5 bar vacuum applied.
iv. Discharge the product into well labelled polypail, awaiting packing into aluminium tubes.
Formulation 3c: Mixture (oleic acid, stearic acid and thickeners); full strength - molar ratio of lopinavir to ritonavir 12:1
The manufacture of a vaginal dosage form full strength active batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 3:- i. Add into the mixer the following materials - 3,4,5,6,7,8,9,1,10,11
ii. Exclude air from the interior of the vessel
iii. Heat to 70 °C while low shear mixing, to achieve a clear, transparent melt. iv. Add into the mixer the following material - 2
v. Exclude air from the interior of the vessel
vi. Mix via low shear, to finely disperse the HPMC within the melt
vii. Reduce the content temperature to 45°C while low shear mixing
viii. Discharge to storage vessel, and exclude air during storage.
Formulation 3d: Mixture (oleic acid, stearic acid and thickeners); half strength - molar ratio of lopinavir to ritonavir 12:1
The manufacture of a vaginal dosage form half strength active batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 4:- i. Add into the mixer the following materials - 3,4,5,6,7,8,9,1,10 1 ii. Exclude air from the interior of the vessel
iii. Heat to 70 °C while low shear mixing, to achieve a clear, transparent melt.
iv. Add into the mixer the following material - 2
v. Exclude air from the interior of the vessel
vi. Mix via low shear, to finely disperse the HPMC within the melt
vii. Reduce the content temperature to 45°C while low shear mixing
viii. Discharge to storage vessel, and exclude air during storage.
Formulation 3e: Mixture (oleic acid, stearic acid and thickeners): placebo
The manufacture of a vaginal dosage form placebo batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 5:- i. Add into the mixer the following materials - 3, 4, 5, 6, 7, 8, 9,1
ii. Exclude air from the interior of the vessel
iii. Heat to 70 °C while low shear mixing, to achieve a clear, transparent melt.
iv. Add into the mixer the following material - 2
v. Exclude air from the interior of the vessel
vi. Mix via low shear, to finely disperse the HPMC within the melt
vii. Reduce the content temperature to 45°C while low shear mixing
viii. Discharge to storage vessel, and exclude air during storage. Formulation 3f: Mixture (oleic acid, stearic acid, HPMC and thickeners); full strength molar ratio of lopinavir to ritonavir 13.8:1
The manufacture of a vaginal dosage form placebo batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 6:- i. Add into the mixer the following materials - 3, 4, 5, 6, 7, 8, 9, 1, 10, 11
ii. Exclude air from the interior of the vessel
iii. Heat to 70 °C while low shear mixing, to achieve a clear, transparent melt.
iv. Add into the mixer the following material - 2
v. Exclude air from the interior of the vessel
vi. Mix via low shear, to finely disperse the HPMC within the melt
vii. Reduce the content temperature to 45°C while low shear mixing
viii. Discharge to storage vessel, exclude air during storage.
ix. Pack product into aluminium tubes, suitable for 2.5 g dispensing so as to deliver 300 mg Lopinavir and 25 mg Ritonavir
Formulation 3g; Mixture (oleic acid, stearic acid. HPMC and thickeners): half strength
- molar ratio of lopinavir to ritonavir 13.8:1
The manufacture of a vaginal dosage form placebo batch exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 7:- i. Add into the mixer the following materials - 3,4,5,6,7,8,9,1,10,11
ii. Exclude air from the interior of the vessel
iii. Heat to 70 °C while low shear mixing, to achieve a clear, transparent melt.
iv. Add into the mixer the following material - 2
v. Exclude air from the interior of the vessel
vi. Mix via low shear, to finely disperse the HPMC within the melt
vii. Reduce the content temperature to 45°C while low shear mixing
viii. Discharge to storage vessel, exclude air during storage.
ix. Pack product into aluminium tubes, suitable for 2.5 g dispensing so as to deliver 150 mg Lopinavir and 12.5 mg Ritonavir. Formulation 3h: Mixture (oleic acid, stearic acid, HPMC and thickeners); full strength molar ratio of lopinavir to ritonavir 13.8:1; cold process
The manufacture of a vaginal dosage form exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 8:- i. Into medicine mixer add 1, 9, 10, 11
ii. Lower the lid and purge with nitrogen
iii. Mix without heating for several hours until a clear, transparent solution is achieved iv. Raise the lid and add 7
v. Lower the lid and purge with nitrogen
vi. Mix without heating until a clear, transparent solution is achieved
vii. Raise the lid and add 2, 3, 4, 5, 6, 8
viii. Lower the lid and purge with nitrogen
ix. Mix without heating for several hours
x. Discharge the product into bulk storage vessel, awaiting packing into aluminium tubes
Formulation 3i: Binary mixture (oleic acid and stearic acid): full strength- molar ratio of lopinavir to ritonavir 13.8:1
The manufacture of binary mixture of oleic acid and stearic acid active batch is described below in accordance with Table 9:- i. Into main vessel add 1, 4. Stir to dissolve
ii. Into main vessel add 5, 6. Stir to dissolve
iii. Into main vessel add 2. Stir to disperse
iv. Into main vessel add 3. Stir to blend. Aim for a very smooth consistency.
v. Discharge the product into a well labelled polypail.
Formulation 3i: Binary mixture (oleic acid and stearic acid); full strength- molar ratio of lopinavir to ritonavir 9.2:1
The manufacture of binary mixture of oleic acid and stearic acid active batch is described below in accordance with Table 10:- i. Into main vessel add 1, 4. Stir to dissolve
ii. Into main vessel add 5, 6. Stir to dissolve
iii. Into main vessel add 2. Stir to disperse
iv. Into main vessel add 3. Stir to blend. Aim for a very smooth consistency. v. Discharge the product into a well labelled polypail.
Formulation 3k: Mixture (oleic acid, stearic acid, HPMC and thickeners); full strength molar ratio of lopinavir to ritonavir 13.8:1; cold process
The manufacture of a vaginal dosage form exhibiting rheology suitable for syringe applicator vaginal dosing is described below in accordance with Table 11 :-
1 Into medicine mixer add 1, 9, 10, 11
ii. Lower the lid and purge with nitrogen
iii. Mix without heating for several hours until a clear, transparent solution is achieved iv. Raise the lid and add 7
v. Lower the lid and purge with nitrogen
vi. Mix without heating until a clear, transparent solution is achieved
vii. Raise the lid and add 2, 3, 4, 5, 6, 8
viii. Lower the lid and purge with nitrogen
ix. Mix overnight, without heating
x. Discharge the product and pass through a colloid mill
xi. Filter the milled product through a 400pm GAF filter into a storage vessel.
xii. Purge the headspace with nitrogen, and seal the lid, awaiting packing into aluminium tubes.
Table 1
Formulation Stearic acid as a stiffening agent, no
Rationale thickeners present - no heat
Batch
number: N/A
Batch size: 2000 g
Active
supplier: Cipla
Figure imgf000053_0001
*N2 PURGE ANY BULK
SUPPLY AFTER USE
** PROTECT FROM UV
LIGHT
Table 2
Formulation Stearic acid as a stiffening agent,
Rationale no thickeners present - no heat
Batch
number: N/A
Batch size: 1775 g
Active
supplier: Cipla
Figure imgf000054_0001
*N2 PURGE ANY BULK
SUPPLY AFTER USE
** PROTECT
FROM UV LIGHT
Table 3
Figure imgf000055_0001
* Melt completely /mix source supply prior to dispensing
**N2 purge source supply after sampling
* «Protect from uv light
Figure imgf000055_0002
Table 4
Figure imgf000056_0001
* Melt completely/mix source supply prior to dispensing
**N2 purge source supply after sampling
* «Protect from uv light
Figure imgf000056_0002
Table 5
PLACEBO ointment
Product name:
Figure imgf000057_0001
* Melt completely/mix source
supply prior to dispensing
**N2 purge source supply after sampling
* «Protect from uv
light
Table 6
Figure imgf000058_0001
* Melt completely /mix source supply prior to dispensing
**N2 purge source supply after sampling
* «Protect from uv light
Figure imgf000058_0002
Table 7
Figure imgf000059_0001
* Melt completely /mix source supply prior to dispensing
**N2 purge source supply after sampling
* «Protect from uv light
Figure imgf000059_0002
Table 8
Figure imgf000060_0001
* *N2 purge source supply after sampling
* * * Protect from UV
light
Figure imgf000060_0002
Table 9
Formulation Stearic acid as a stiffening agent, no
Rationale thickeners present - no heat
Batch number: N/A
Batch size: 1500 g
Figure imgf000061_0001
*N2 PURGE SOURCE SUPPLY
AFTER SAMPLING
** PROTECT FROM UV LIGHT
Table 10
Formulation Stearic acid as a stiffening agent, no
Rationale thickeners present - no heat
Batch number: N/A
Batch size: 1500.0 g
Figure imgf000062_0001
*N2 PURGE ANY BULK SUPPLY
AFTER USE
** PROTECT FROM
UV LIGHT
(Whenever possible, once wetted, apply vacuum or N2 blanket to reduce oxygen exposure and aeration)
Table 11
Figure imgf000063_0001
* *N2 purge source supply after sampling
* * * Protect from UV
light
Figure imgf000063_0002
Example 4: assessment of the Formulations
The formulations described in Example 3 are suitable for topical dosing and syringe dosing. Organoleptic assessments of formulations 3a, 3b, 3c, 3d, 3e, 3f, 3g, 3i, 3j, 3h and 3k were conducted.
Organoleptic assessment of Formulations 3a and 3b
Dosing Procedure:
1 Remove the cap from the tube
2 Dispense a small amount of the ointment onto a clean fingertip.
3 With the fingertip, apply the ointment to the affected skin area, rubbing gently.
4 Replace the cap on the tube
Observations
The opaque ointment presents as easy to dispense from the tube and is controllable on the skin during rubbing. After a short period of rubbing, the ointment clarifies to leave the skin non- tacky and with minimal greasiness. Approximately 10 minutes after dosing, the skin presents as silky, with negligible greasiness. These attributes denote an ointment as particularly suitable for topical application.
Organoleptic assessment of Formulations 3c, 3d and 3e
Dosing Procedure
1 Remove the cap from the tube
2 Thread the dosing syringe applicator onto the tube orifice
3 Squeeze the tube to force product into the syringe applicator until the plunger rises to the 3 g mark.
4 Discharge the ointment from the syringe applicator onto a petri dish.
5 Add 10 drops of water onto the dispended ointment
6 With the fingertip rub gently to blend in the water.
7 Replace the cap on the tube
Observations
The opaque ointment presents as easy to dispense from the tube into the syringe applicator, after a short period of rubbing the ointment becomes increasingly more opaque, increasing whiteness, embodied, gelatinous, and cream-like. These attributes denote an ointment that is compatible with a water environment, and as such, particularly suitable for topical application to an area of the body where water is present, for example vaginal application.
Organoleptic assessment of Formulations 3c, 3d, 3f, 3g, 3i, 3i, 3h, and 3k
An organoleptic assessment was performed for formulations 3c, 3d, 3f, 3g, 3i, 3j, 3h and 3k using the following methodology.
Dosing Procedure:
• Remove the cap from tube.
• Dispense a small amount of the ointment onto a clean fingertip.
• With the fingertip, apply the ointment to an area of skin, rubbing gently.
· Replace the cap on the tube.
• Record observations for colour, transparency, homogeneity, material state, and textural attributes.
The organoleptic assessment results for formulations 3c, 3d, 3f, 3g, 3i, 3j, 3h and 3k are presented in Table 12 below:
Table 12
Figure imgf000065_0001
Figure imgf000066_0002
Example 5: Stability studies
The compositions according to Example 3 have been proven to be stable during the process employed to manufacture them (i.e. processes described in Example 3). Additionally, the compositions according to Example 3 have been found to be stable upon storage.
Formulations 3c, 3d, 3f, 3g, 3i, 3j, and 3h were stored under different time and stability conditions, and subsequently analysed. The stability data is presented in Tables 13 and 14. A stable product is defined as one demonstrating 90-110% w/w assay for both Lopinavir and Ritonavir. Based on the data generated, the hot processed product
(Formulations 3f and 3g) when stored at 5°C (range 2-8°C) affords at least 10 months stability. Formulation 3h (prepared using a cold process) demonstrates greater stability than that for formulations 3c and 3d when stored for 3 months stored at 25°C. This is thought to be due to the absence of thermal exposure of Ritonavir and Lopinavir during the manufacture. Forward projection of the stability data suggest at least 6 months/25°C is achievable for Formulation 3h. To get stability beyond 6 months for Formulation 3h, cold storage (5°C) may be required.
Table 13
Figure imgf000066_0001
Figure imgf000067_0001
* All batches put to stability are packed in to phenolic epoxy lined aluminium tubes **Formulation 3j was an early formulation. A stability assessment was not taken at the time of manufacture. For stability trending, formulation 3i (a close variant of formulation 3j) was manufactured.
Table 14
Figure imgf000068_0001
* All batches put to stability are packed in to phenolic epoxy lined aluminium tubes
**Formulation 3j was an early formulation. A stability assessment was not taken at the time of manufacture. For stability trending, formulation 3i (a close variant of formulation 3j) was manufactured. The following analytical methodology was used to analyse the formulations. UHPLC:
Equipment Parameters:
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0002
Gradient Elution Program
Figure imgf000071_0001
Example 6: Rheological characterisation of the in-situ transition provided by the hydrophilic muco-adhesive when the anhydrous composition is exposed to an
environment comprising water
The following methodology was used to determine the rheological response of the anhydrous composition upon exposure to an environment comprising water. A Discovery Hybrid Rheometer (Model HR-3, TA instruments) was used. The sample was introduced onto the peltier plate (base) in excess (l-2g), and the spindle lowered to make contact with the sample. The excess sample was cleaned up. The spindle was then rotated in a predetermined fashion to exert a series of shear forces on the sample. The various sample shear force response parameters (complex viscosity, storage and loss moduli, tan 5) were captured and then plotted in order to make conclusions regarding inherent sample attributes.
Instrument parameters for the purpose of capturing complex viscosity, storage modus and loss modulus are as follows :- Peltier temperature: 37 °C
Sample size: Approximately 1 g
Oscillation frequency method parameters:
Speed range 0.1 to 1 rad/s
Strain 0.5 %
Spindle 40 mm parallel plate
Gap setting 1000 pm
The range of HPMC level investigated was 0 - 10 % w/w, see Formulations 6a, 6b, 6c and 6d.
Formulation 6a: 0%
Figure imgf000072_0001
The manufacture of a composition comprising 0% HPMC is described below in accordance with Table 15:- i. Into vessel A of known mass add 8, 9, 10, 11, 12, 13, 1
ii. Heat the contents of vessel A on a hot plate just enough to melt (ceresin wax MP
75°C) with stirring
iii. Transfer the vessel A to town water bath and begin cooling to ambient with stirring.
Once at ambient temperature continue stirring to maintain shear thinned state.
iv. Into vessel B add 2, and dissolve the required amount 14
v. Dissolve into the vessel B the required amount 15 and 16 with magnetic stirring and then carefully remove the magnetic stirrer.
vi. Under inert atmosphere (NPD glovebox) and non-actinic light, while stirring vessel A add in the contents of vessel B into vessel A and mix well to obtain uniformity of mixture
vii. Under inert atmosphere (NPD glovebox) and non-actinic light, pack (aim 25 g) into ABL tubes and seal.
Formulation 6b: 1% HPMC - Manufactured using a low shear mixing process The manufacture of a composition comprising 1% HPMC is described below in accordance with Table 16:- i. Into main vessel add 4, 5, 6, 7, 8, 9, 10, 1
ii. Heat the contents to 85°C, stirring as required to combine (e.g. 60 rpm)
iii. Cool content to 60°C, stirring as required (e.g. 60 rpm)
iv. Into a separate SS vessel add 2 and dissolve the required amount 11 with overhead stirring.
v. Dissolve into the same SS vessel the required amount 12, 13 with overhead stirring.
Solution must be clear and transparent.
vi. Disperse into the same SS vessel the required amount of 3 with stirring. Confirm absence of clumping.
vii. Lift the main vessel lid and introduce the solvent phase contents into the main vessel. viii. Under vacuum (e.g. -0.5 to -0.9 bar), stir very well to combine the two phases while cooling to ambient. Aim for a very smooth consistency free from lumps. Faster /longer (e.g 90 rpm 120 mins) stirring can help to achieve this. Use homogenizer periodically at 600 rpm to assist.
ix. Discharge the product into well labelled polypail, awaiting packing into aluminium tubes.
Formulation 6c: 1 % HPMC - Manufactured using high pressure homogenizing
The manufacture of a composition comprising 1% HPMC is described below in accordance with Table 17:- i. Into main vessel add 4, 5, 6, 7, 8, 9, 10, 1
ii. Heat the contents to 85°C, stirring as required to combine (e.g. 60 rpm)
iii. Cool content to 60°C, stirring as required (e.g. 60 rpm)
iv. Into a separate SS vessel add 2 and dissolve the required amount 11 with overhead stirring
v. Dissolve into the solvent the required amount 12, 13 with overhead stirring vi. Disperse into the same SS vessel the required amount of 3 with stirring. Confirm absence of clumping
vii. Warm the solvent phase to 60°C with stirring
viii. Lift the main vessel lid and introduce the solvent phase contents into the main vessel ix. Under vacuum (e.g. -0.5 to -0.9 bar), stir for 30 minutes to combine the two phases while reducing the heat to 40°C. Use homogenizer periodically at 600 rpm to assist. x. Discharge the product into well labelled polypail and pass through the homogenizer as per next step
xi. Pass 2 x 1L portions through the high pressure homogeniser (Panda) (single pass only) at pressures of approximately 250 bar and/or 1000 bar. Record actual pressure used xii. Allow portions to set overnight.
Formulation 6d: 10% HPMC
The manufacture of a composition comprising 10% HPMC is described below in accordance with Table 18:- i. Into main vessel add 4, 5, 6, 7, 8, 9, 10, 1
ii. Heat the contents of just enough to melt (ceresin wax MP 75 °C), stirring as required. iii. Cool content to 40 °C, stirring as required.
iv. Into a separate SS vessel add 2 and dissolve the required amount 11 with overhead stirring.
v. Disperse into the same SS vessel the required amount of 3 with stirring. Confirm
absence of clumping.
vi. Lift the main vessel lid and introduce the solvent phase contents into the main vessel. vii. Under vacuum, assess ability to effect stirring to combine the two phases and cool to ambient.
Table 15
Formulation
Rationale 0% HPMC
Batch size: 100 g
Figure imgf000075_0001
*N2 PURGE ANY BULK SUPPLY ALTER USE Table 16
Formulation
Rationale 1% HPMC-low shear mixing
Batch size: 2 kg
Figure imgf000076_0001
*N2 PURGE ANY BULK SUPPLY
AFTER USE
** PROTECT
FROM UV LIGHT Table 17
Formulation 1 % HPMC - HPH
Rationale
Batch size: 2 kg
Figure imgf000077_0001
*N2 PURGE ANY BULK SUPPLY
AFTER USE
** PROTECT FROM
UV LIGHT Table 18
Formulation Rationale !0% HPMC
Batch size: 2 kg
Figure imgf000078_0001
*N2 PURGE ANY BU K SUPPLY
AFTER USE
** PROTECT FROM
UV LIGHT Assessment of Rheological response
The muco-adhesive effect has been interpreted via the rheological response of the formulation in the presence of water, as demonstrated in the oscillation frequency plot as shown in Figure 1 for a prototype vaginal dosage form ointment containing 1% w/w HPMC (formulation 6b). From Figure 1, it can be seen that once 2-3 drops of water are introduced to the product environment, a shift in the rheological response occurs, consistent with the forming of an in situ gel dispersion. This drives an increase in both the complex viscosity (h) and the storage modulus (G’). The storage modulus is a direct measure of a fluid’s solid-like behaviour. The ratio of the loss modulus (G”) to the storage modulus (G’) is a unitless measure, Tan 5. A Tan d value of <1 defines a material as largely solid-like; >1 classifies the material as predominately fluid-like. In the presence of water, the increase in complex viscosity and storage modulus can be interpreted as defining a product with increased adherence potential and resistance to movement.
A prototype vaginal ointment without HMPC (formulation 6a) does not exhibit the same upward trend for complex viscosity or storage modulus. In fact, as per Figure 2, the complex viscosity and storage modulus both decrease, which, together with the increasing loss modulus, results in raising Tan d. Such a product can be interpreted as having low potential to adhere within the vaginal cavity.
High pressure homogenizing (HPH) is an alternate manufacturing process step, found to achieve greater product dispersion, a decrease in particle size, leading to an increase in product surface area, and potentially an increase in viscosity. As per Figure 3, a prototype vaginal ointment with 1% w/w HPMC processed via HPH (formulation 6c) demonstrates the ability to achieve an increase in product viscosity when compared to the same formulation processed via traditional low shear (formulation 6b).
Work has been conducted to assess the rheology response when the HPH processed formulation (formulation 6c) is introduced to a water environment. While driving no further increase in complex viscosity beyond that already evidenced by the alternate HPH
manufacturing process, the product does nonetheless, still display the same transition from fluid-like to solid-like, as per Figure 4. The storage modulus switches from a position of below the loss modulus, to above that of the loss modulus; Tan d moves from >1 to be <1, and the product demonstrates solid-like behaviour.
Example 7: Varying grade of HPMC
Formulation 7 The manufacture of a composition comprising HPMC 2910 (4000 cps) as the mucosa adhesive is described below in accordance with Table 19:- i. Into main vessel add materials except 2
ii. Apply 3x nitrogen blanket
iii. Mix as required, with heating, until clear solution (e.g. 60 rpm, homogenizer 600 rpm) iv. Into main vessel add 2
v. Apply 3x nitrogen blanket
vi. Mix as required until well dispersed (e.g. 60 rpm, homogenizer 600 rpm)
vii. Reduce temperature to ambient while mixing as required (e.g. 60 rpm, homogenizer 600 rpm)
viii. Discharge the product and confirm no lumps, especially around homogenizer ix. Pack into 50 mL aluminium tubes, sufficient for rheological analysis
Rheological assessment of Formulation 7
The rheological response of Formulation 7 was determined using the methodology as described in Example 6.
Rheological assessment of Formulation 7 reveals an increase in viscosity and decrease in tan delta when water is added, see Figure 5. This data demonstrates that HPMC 2910 (4000 cps) acts as a mucosa adhesive. The muco-adhesive effect has been interpreted via the rheological response of the formulation in the presence of water, as demonstrated in the oscillation frequency plot as shown in Figure 5 for a prototype vaginal dosage form ointment containing 1% w/w HPMC (formulation 7). From Figure 5, it can be seen that once 2-3 drops of water are introduced to the product environment, over a period of time (60 minutes) a shift in the rheological response occurs, consistent with the forming of an in situ gel dispersion. This drives an increase in both the complex viscosity (h) and the storage modulus (G’), and a reduction in the Tan d value. The storage modulus is a direct measure of a fluid’s solid-like behaviour, while the ratio of the loss modulus (G”) to the storage modulus (G’) is a unitless measure, Tan d. A Tan d value of <1 defines a material as largely solid-like; >1 classifies the material as predominately fluid-like. In the presence of water, the increase in complex viscosity and storage modulus, and reduction in Tan d can be interpreted as defining a product with increased adherence potential and resistance to movement. Formulation Rationale HPMC variation, hot process
Batch size: 3000 g
Figure imgf000081_0001
**N2 PURGE ANY BULK SUPPLY
AFTER USE
* «PROTECT FROM
UV LIGHT Example 8: Excipient One Factor At A Time (OF AT) analysis
One-Factor- At-a-Time (OF AT) excipient level variation analysis of centre-point full- strength (Formulation 8a) /half- strength (Formulation 8b) formulations, sometimes referred to as Design-Of-Experiments (DOE) has been conducted for each of the materials as per the below table, via seventeen additional Formulations 8c to 8s.
The impact for each change was assessed by rheological response, specifically the recording of the maximum complex viscosity (q) observed across a frequency sweep 0.1-1 radians/second at 0.5 % strain via TA Instruments DHR Rheometer fitted with 40mm parallel plate, a gap setting of 1000 pm, and a temperature of 37 °C.
Aside from the API solvent, which attracts its own set of selection criteria (namely the minimum amount required to achieve dissolved state for Lopinavir and Ritonavir), and antioxidant (with and without), the lower levels for other excipients were selected based on being, in essence, just above zero. The upper level of each excipient was based on
understanding of the likelihood for the anomaly to exert an effect, while still maintaining a doseable semisolid.
The centre-point full-strength (Formulation 8a) /half- strength (Formulation 8b) formulations yield viscosities of approximately 2000-l500cps. The half strength contains a higher solvent level than the full-strength, which translates to a somewhat lower viscosity (Note: It is the full-strength formulation, with 13 % total drug substance, that dictates the minimum amount of solvent i.e. 55.3 %. The half strength formulation, with only 6.5 % total drug substance, thus contains the balance of the material added to the solvent, which commensurately rises to 61.8 %).
Formulation 8d (q = 288cps) is too runny for dosing, while Formulation 8s (q = 9l5cps) is easily stiff enough for dosing. Formulation 8c (q = l8085cps) is so stiff as to be barely dose-able via the intended 6 mm tube orifice. A larger tube orifice (8-10 mm), or even use of pre-charged dosing applicators would facilitate easier dosing of product at this upper viscosity.
A summary table of the rheological viscosity outputs for the range finding
formulations, incorporating a Material Risk Assessment (MRA) is presented in Table 20. Table 20
o
O
4
Os
n H w
O
O C/I
Figure imgf000083_0001
4
00
Formulation 8a: Full strength ointment
The manufacture of Formulation 8a is described below in accordance with Table 21 i. Into main vessel add all materials except for 2
ii. Heat the contents to 75°C, mixing as required until clear solution e.g. 60 rpm, homogenizer 600 rpm
iii. Into the main vessel add 2 to disperse, mixing for e.g. 10 minutes, 60 rpm,
homogenizer 600 rpm
iv. Cool contents to 45°C, mixing as required e.g. 60 rpm, homogenizer 600 rpm v. Discharge the product into HDPE bag lined polypail, well labelled
Formulation 8b: Half strength ointment
The manufacture of Formulation 8b is described below in accordance with Table 22:- i. Into main vessel add all materials except for 2
ii. Exclude air from the interior via vacuum -80kPa. Replace with nitrogen iii. Repeat step ii two more times
iv. Heat the contents to 75°C, mixing as required until clear solution e.g. 60 rpm, homogenizer 600 rpm
v. Into the main vessel add 2
vi. Repeat step ii three times
vii. Mix for e.g. 10 minutes, 60 rpm, homogenizer 600 rpm to disperse
viii. Cool contents to 45°C, mixing as required e.g. 60 rpm, homogenizer 600 rpm ix. Discharge the product into HDPE bag lined polypail, well labelled
x. Pack 75 g into each of 30 tubes with Darex seal
Formulations 8c. 8d, 8e, 8f„ 8g, 8h„ 8i„ 8i, 8k. 81. 8m. 8n, 8o, 8p. 8q, 8r„ 8s and 8t
The manufacture of Formulations 8c, 8d, 8e, 8f, 8g, 8h, 8j, 8k, 81, 8m, 8n, 8o, 8p, 8q, 8r, 8s and 8t are described below in accordance with Table 23 :- i. Into main vessel add all materials except for 2
ii. Exclude air from the interior via vacuum -80kPa. Replace with nitrogen iii. Repeat step ii two more times
iv. Heat the contents to 75°C, mixing as required until clear solution e.g. 60 rpm, homogenizer 600 rpm
v. Into the main vessel add 2
vi. Repeat step ii three times vii. Mix for e.g. 10 minutes, 60 rpm, homogenizer 600 rpm to disperse viii. Cool content to 45°C, mixing as required e.g. 60 rpm, homogenizer 600 rpm ix. Discharge the product into HDPE bag lined polypail, well labelled
x. Pack 75 g into each of 3 tubes with Latex seal, keep the rest as bulk, well sealed.
Table 21
Figure imgf000085_0001
* *N2 purge source supply after
sampling
*** Protect from UV light
Figure imgf000085_0002
Table 22
Figure imgf000086_0001
* *N2 purge source supply after sampling
* * * Protect from UV light
Figure imgf000086_0002
Table 23
Figure imgf000087_0002
Figure imgf000087_0001
Example 9: A Phase 1, single centre, double blind, randomised, parallel group, ascending single and multiple dose, safety and tolerability, pharmacokinetic (PK) and pharmacodynamic (PD) study of Formulations 3c, 3d and 3e in healthy women volunteers.
The compositions according to Example 3 (Formulations 3c, 3d, and 3e) were investigated according to the clinical trial described below.
Study Obiectives
1. To evaluate the safety, PK and PD of compositions in healthy women volunteers after multiple doses of Formulations 3c and 3d.
2. To observe the rates of side effects reported by women using the compositions compared to placebo.
Investigational Plan/Studv Design
This study comprised two cohorts, both in healthy volunteers with no cervical pathology. There were 9 participants per Cohort, of whom 6 received active and 3 received placebo. For all formulations tested, the amount of the composition administered per dose was 3 g. For Formulation 3c this equates to 300 mg of lopinavir and 28.7 mg of ritonavir being
administered to the patients per dose. For Formulation 3d this equates to 150 mg of lopinavir and 14.3 mg of ritonavir administered per dose.
Cohort 1:
Period 1 : Single dose of Formulation 3d or placebo Formulation 3e followed by confinement. PK blood sampling during confinement.
Period 2: 21 daily doses of Formulation 3d or placebo Formulation 3e followed by PK blood sampling.
Cohort 2:
Period 1 : Single dose of Formulation 3c or placebo Formulation 3e followed by confinement. PK blood sampling during confinement.
Period 2: 21 daily doses of Formulation 3c or placebo Formulation 3e followed by PK blood sampling.
Participation Criteria
Inclusion Criteria: a. Women, 20 to 45 years old, with an intact uterus and vagina.
b. Generally, in good health with no clinically significant pulmonary, cardiac,
gastroenterological, pancreatic, neurologic, renal, musculoskeletal, rheumatologic, metabolic, neoplastic, or endocrine disease.
c. BMI of =>l9 and <= 30.0
d. ECG and vital signs within normal ranges
e. Agree to no Alcohol from 48 hours prior to dosing in period 1 until 7 days after receiving the final dose in period 2.
f. Abstain from food or beverages containing grapefruit, starfruit, pomegranate, pineapple, or pomelo for the entire study
g. Able and willing to abstain from sexual intercourse +/- 6 hours around dosing within Periods 1 and 2
h. Able and willing to use stringent methods of contraception after required abstinence
period through to Day 29 (7 days after receiving the final dose in period 2), including the use of a non-latex condom (for partner protection) and a second acceptable contraception method; vasectomy, contraceptive pill, contraceptive implants or IUDs are allowed (note: IUDs should have been inserted at least 1 month prior to enrolment and not because of the involvement in this study)
i. Agree to abstain from activities such as vaginal douching or insertion of any vaginal
products other than the study drug for at least 48 hours prior to enrolment and throughout the study.
j. Negative Pap test at screening or within 3 years of enrolment and no history of cervical intraepithelial lesions within the previous 3 years
k. Able and willing to return to the clinic for all study procedures.
1. Able and willing to provide informed consent.
Exclusion Criteria:
a. Women who are pregnant, plan to become pregnant in the next 3 months, or lactating females.
b. History of genital herpes with >3 outbreaks per year, or active non-HPV vaginal infection c. Positive result for Hep B, Hep C or HIV.
d. Have an active pelvic infection (positive urine screen for gonorrhoea or chlamydial
infection, positive test and symptoms for bacterial vaginosis, Candida vaginitis or trichomonal vaginitis) e. Current or recent abnormal vaginal discharge and /or abnormal vaginal bleeding, within the 3 months prior to randomization as accessed by Investigator.
f. Had an abortion or miscarriage within the 3 months prior to randomization
g. Currently taking any of the following medications: oral corticosteroids, inhaled salmeterol and fluticasone; immunomodulatory treatments, over the counter (OTC) intra-vaginal preparation, or any prescription that in the opinion of the Investigator could interfere with the interpretation of the results.
h. Currently taking any of the medications listed here - Alfuzosin, Amiodarone,
dronedarone, Ranolazine, Fusidic Acid, Colchicine, Astemizole, terfenadine, Lurasidone, Pimozide, Quetiapine, Dihydroergotamine, ergonovine, ergotamine, methylergonovine, Cisapride, Lovastatin, simvastatin, Avanafil, Sildenafil, Vardenafil, Oral midazolam, triazolam, St. John's wort.
i. Recent history (within previous 3 months) of Stevens- Johnson syndrome, erythema
multiforme, urticaria, angioedema, deep vein thrombosis, tinnitus, vertigo, blood glucose disorders, pancreatitis, haemophilia.
j. Hypersensitivity to any component of R131 vaginal ointment excipients
k. Participation in any clinical study with an experimental medication or device within 30 days or 5 half-lives (whichever is longer) of enrolment.
l. Current alcohol or substance abuse as assessed by the Investigator.
m. An employee or first-degree family member of an employee, the Sponsor, the CRO or study site.
n. Not having a GP
Screening Evaluations:
The screening evaluations must have been made within 3 months of randomization into the study. Screening consisted of the following components:
Demographic/Medical History
A complete medical history was taken from each participant.
Physical Examination
The physical examination consisted of a review of body systems with height and weight (in indoor clothing).
Blood Tests The following laboratory blood tests were performed:
• Electrolytes (sodium and potassium), ALT, GGT, ALP, albumin, total protein, total bilirubin, urea, uric acid, serum creatinine, TFT, fasting lipids, amylase, glucose, and HbAlc
· Haemoglobin, red cell count, PCV, MCV, MCH, platelet count, white cell count, neutrophils, lymphocytes, monocytes, eosinophils and basophils. CD4/CD8 counts
• HIV and hepatitis B and C.
The measurement at screening serve as a baseline to monitor any abnormalities that may manifest as a result of dosing
Other Tests
Drugs of abuse testing were carried out on all participants as part of the screening procedures. A urine sample was required to test for cannabinoids (marijuana), amphetamines,
benzodiazepines and opiates (i.e. morphine, heroin and codeine).
Urinalysis dipstick to check for protein, leucocytes, nitrites, pH, specific gravity, glucose, ketones, and blood.
Vaginal swabs for microbiology (gonorrhoea, Chlamydia, bacterial vaginosis, Candida) and HPV genotyping
Alcohol breath testing was carried out at the Clinical Site on the first night of each
confinement period. Serum HCG testing was carried out on all participants as part of the screening procedures and within 3 days before the lst dose.
Vital Signs
Vital signs were recorded and consisted of blood pressure (supine and sitting), heart rate, temperature and respiratory rate. Participants’ vital signs should be within the following limits:
Heart rate > 60 or < 99 beats/minute
Supine:
Systolic Blood Pressure > 90 or < 160 mm Hg; Diastolic Blood Pressure > 50 or < 90 mm Hg Sitting:
Systolic Blood Pressure > 90 or < 160 mm Hg; Diastolic Blood Pressure > 50 or < 90 mm Hg
Temperature > 36°C or < 37.7°C
Respiratory Rate > 12 or < 20 breaths/minute
Summary of Study Activities/Schedule of Events
Informed consent was needed from each participant. Participants were screened to confirm study eligibility.
Randomisation
Participants were randomized following the Principal Investigator or their delegates documented acceptance of participants following review of completed screening procedures.
Study Confinement
Participants arrived at approximately 5pm on Day 1 and Day 22. The duration of study confinement was approximately 27 hours. Participants were released from the clinical site once the 24-hour post dose assessments had been completed.
Dosing
Dosing began at approximately 8pm on each day dosing was scheduled. Participants were instructed to insert the medication in private. Dosing applicators were returned to study staff and examined to ensure the full dose has been applied and for reconciliation of study drug. Sample collection
Vaginal swabs were self-administered by the participants.
PK Blood samples: blood samples (8 mL) were drawn through venous catheters and transferred into vacutainers containing sodium heparin as the anti-coagulant. The time of collection was recorded as the time the full 8 mL of blood is collected. The venous catheters were kept patent by flushing with 1.5 mL-2.0 mL of heparinized saline following each sample (0-24 hours). The sampling intervals were at: Day 1-2: 0, 1, 2, 4, 8, 12, 24 hours; Day 22-23: 0, 1, 2, 4, 8, 12, 24 hours. Samples were collected at their due time. Any deviation was noted.
Sample processing and storage Plasma: Plasma was separated by centrifugation at 3500 rpm for 5 minutes at about 4°C. No aids for separation of plasma from red cells was used. The plasma sample was transferred with clean pipettes. The assay was determined using a validated Analytical method.
Each plasma sample was placed into a polypropylene storage tube with a screw cap. The plasma was stored frozen at -60°C or colder at the clinical site pending transfer to a
Laboratory for assay.
End of Study
Within one week after the last study day, each participant was required to provide a blood sample for analysis. Any abnormalities as compared to initial screening were monitored and followed up until they return to normal.
Participants were assessed for the occurrence of adverse events from consent until the last study day in each cohort.
Vital signs (blood pressure, heart rate, respiratory rate and temperature) at last study visit.
• Laboratory tests (haematology (CBC, CD4+/CD8+ peripheral lymphocyte count, biochemistry (RFT, LFT, electrolytes, TFT, fasting lipids, HbAlc, amylase), Serum
HCG Pregnancy and urinalysis (dipstick), at last study visit.
• A follow-up phone call to each participant was made within 7 days (+ 2 days) of the end of the study to record any possible Adverse Events (AE) post study. Any events were recorded in source documentation.
All AE’s were followed-up until resolution, or until the Investigator was of the opinion that follow-up was no longer required, or until 30 days from the last dose (as long as the
Investigator was satisfied that follow-up is no longer required), whichever is earlier.
Adverse Events
During confinement the designated Supervisor for the study or a delegated representative must have been present at the study site throughout the study. Principal Investigator or at least one delegated Trial Physician was on call throughout the studies. On all study visits each participant was asked how they felt. This occured at each sampling point throughout the study. AE’s were recorded in source documentation.
Each AE was classified by the Principal Investigator as serious adverse event (SAE) or non- serious. Non- serious adverse events were assessed as being mild, moderate, or severe to describe the maximum intensity of the AE. The Principal Investigator also provided the possible relationship between the AE and the study medication as highly probable, probable, possible, remotely or not ("no") related to the study medication.
The Principal Investigator should have stated if the cause of the AE is related to the concurrent non-investigational medication(s) if any are being taken, an underlying disease, a combination of these factors or is unknown.
Safety Results:
109 adverse events in total reported by 18 participants, see Table 24.
• 1 event‘related’ to study medication
• 6 events‘probably related’ to study medication
· 82 events‘possibly related’ to study medication
• 6 events‘probably not related’ to study medication
• 14 events‘not related’ to study medication
• No SAEs
Conclusion:
Formulations 3c and 3d are deemed to be well-tolerated since all AEs were either minor or not related to administration of study medication
Table 24: Adverse event summary:
Figure imgf000094_0001
Figure imgf000095_0001
Pharmacokinetic Analyses:
Pharmacokinetic parameters:
The area under the plasma drug concentration time curve (AUC), the peak plasma drug concentration (Cmax) and the time to maximum drug concentration (Tmax) were determined for lopinavir and ritonavir for each subject receiving active treatment.
The plasma drug concentration (C) versus the real sampling time (t) data were analysed by a "noncompartmental" method to obtain the pharmacokinetic parameters. Initially the plasma data in the post distribution phase of the plasma concentration - time plot were fitted using linear regression to: ·
In C = In Co - t.Kel
where Co is the zero-time intercept of the extrapolated terminal phase and Kel is the terminal elimination rate constant. The area (AUCo-t) from time zero to the last determined concentration-time point (t) in the post distribution phase was calculated using the trapezoidal rule.
Lopinavir and ritonavir concentration and pharmacokinetic parameters
The mean lopinavir and ritonavir plasma concentration-time data for each sampling time is listed in Tables 25 and 26. The pharmacokinetic parameters for lopinavir and ritonavir are summarised in Tables 27 and 28.
Table 25: Mean (±SD) Plasma Lopinavir and Ritonavir Concentration Data vs Sampling Times (Formulation 3d) Vaginal Ointment)
Figure imgf000096_0001
Table 26: Mean (±SD) Plasma Lopinavir and Ritonavir Concentration Data vs Sampling Times (Formulation 3c) Vaginal Ointment)
Figure imgf000097_0001
Table 27: Pharmacokinetic Parameters for Lopinavir
Figure imgf000098_0001
Table 28: Pharmacokinetic Parameters for Ritonavir
Figure imgf000099_0001
Discussion of results:
Cmax:
Following an oral dose of 400 mg lopinavir (as Kaletra 400 mg/lOO mg tablets)* twice daily for 2 weeks, the mean Cmax for lopinavir was 12.3 ± 5.4 pg/mL (SPMC Kaletra) . Adjusting for dose comparison with a 300 mg dose administered topically in ointment form, the mean Cmax would be 9.23 ± 4.1 pg/mL.
Following a topical dose of 300 mg lopinavir daily for 21 days as 2.5 g ointment containing 12% w/w lopinavir, the mean Cmax was 396.3 ± 297.3 pg/mL.
The ratio of Cmax oral/Cmax topical is >23,000 indicating that less than 0.004% of the topical dose is available systemically. AUC O-t:
Following an oral dose of 400 mg lopinavir (as Kaletra 400 mg/lOO mg tablets)* twice daily for 2 weeks, the AUCO-t for lopinavir was 113.2 ± 60.5 pg h/mL (SPMC Kaletra). Adjusting for dose comparison with a 300 mg dose administered topically in ointment form, the AUCO-t would be 84.9 ± 45.4 pg h/mL.
Following a topical dose of 300 mg lopinavir daily for 21 days as 2.5 g ointment containing 12% w/w lopinavir, the AUCO-t was 7368.1 ± 4973.1 pg/mL.
The ratio of AUC oral/AUC topical is >11,500 indicating that less than 0.009% of the topical dose is available systemically.
Conclusion:
The combined Cmax and AUC data indicate that systemic absorption of lopinavir from topical administration of the ointment is negligible.
Example 10: A Phase lb. Multicentre. Open Label. Study of the Efficacy. Safety and Tolerability of Formulation 3h in Women with Cvtological Abnormalities of the Uterine Cervix.
The composition according to Example 3 (Formulations 3h) is investigated according to the clinical trial described below.
Study Objectives
Efficacy Objectives · Demonstrate histological clearance of cytological abnormalities following application of
Formulation 3h in women with high-grade or low-grade CIN (Cervical intra-epithelial neoplasia).
• Demonstrate changes to colposcopic appearance of the uterine cervix following
application of Formulation 3h; · Assess changes in HPV status following application of Formulation 3h ointment.
Safety Objective
• Assess the incidence of AEs following the application of Formulation 3h ointment.
Tolerability Objective • Assess the tolerability of Formulation 3h, measured by compliance with dosing schedule of Formulation 3h during 21 consecutive days of treatment for up to 3 treatment cycles.
Study Design:
This study is designed as a Phase lb multicentre, open label study investigating the efficacy, safety and tolerability of Formulation 3h ointment in women with cytological abnormalities of the uterine cervix.
In this single arm study, participants are stratified according to their grade of cytological abnormality:
• Biopsy proven high-grade cytological abnormalities of the uterine cervix defined as CIN 2 and above;
• Low-grade cytological abnormalities of the uterine cervix defined as CIN l/LSIL.
Formulation 3h ointment is self-applied to the vagina once a day for 21 consecutive days in up to 3 treatment cycles. Participants attend a clinic visit 7 days after administration of the final dose of investigational product. Participants complete a daily diary card and Vaginal Irritation Questionnaire to capture compliance with investigational product administration,
AEs and changes to concomitant medication.
Participation in this study includes a screening visit, up to 3 treatment cycles and an end of study visit as follows:
• Screening Visit: Day -28 to Day 0.
· Treatment Cycle 1 :
Day 1* to Day 21 : Treatment Cycle 1 - investigational product applied once daily for 21 days;
Day 8, 15, 22: telephone follow up; check AEs, conmeds, compliance, dosing issues;
Day 28: visual assessment of disease, if no disease detected#, treatment is stopped, and a biopsy is performed 6 weeks later (Day 70).
· Treatment Cycle 2:
Day 29** to Day 49: Treatment Cycle 2 - non-responders, identified at Day 28, will continue investigational application once daily for 21 days;
Day 36, 43, 50: telephone follow up; check AEs, conmeds, compliance, dosing issues; Day 56: visual assessment of disease, if no disease detected#, treatment is stopped, and a biopsy is performed 6 weeks later (Day 98).
• Treatment Cycle 3 :
Day 57** to Day 77: Treatment Cycle 3 non-responders, identified at Day 56, can continue investigational application once daily for 21 days OR be referred to their primary physician;
Day 64, 71, 78: telephone follow up; check AEs, conmeds, compliance, dosing issues;
Day 84: visual assessment of disease and biopsy performed 6 weeks later (Day 126)
• Post-treatment assessment visit (PTAV) / Early termination visit (ETV): Day 70, Day 98 or Day 126 depending on response.
*Day 1 commences at the end of a participant’s menstrual cycle.
** Day 29 and 57 may be delayed to the end of the participants’ menstrual cycle if required.
#If absence of disease, defined as no colposcopic evidence of CIN, participants are considered responders. If disease is detected, defined as ongoing colposcopic evidence of CIN, participants are considered non-responders.
Participation Criteria
Inclusion Criteria:
To be eligible for study entry participants must satisfy all of the following criteria:
1. Provision of written informed consent prior to any study specific procedures;
2. Female participants aged 25-45 years inclusive at the time of screening visit;
3. Positive result for cervical high-risk HPV (types 16, 18 or‘other’);
4. High-grade cytological abnormality of the uterine cervix defined as CIN 2 as proven by colposcopic biopsy collected at screening
OR
low-grade cytological abnormality of the uterine cervix defined as CIN l/LSIL, as demonstrated by colposcopic biopsy within 6 months prior to screening.
Participants will be stratified according to their grade of cytological abnormality;
5. Transformation zone needs to be fully visible; 6. Generally, in good health with no clinically significant disease as determined by the investigator;
7. Regular menstrual cycle with an approximate 28-day cycle
OR
women who are amenorrhoeic due to effective contraception (such as Mirena, Jadelle, or continuous COC)
8. Agree to abstain from activities such as vaginal douching or insertion of any vaginal products other than the study drug for at least 48 hours prior to enrolment and throughout the study. Tampons may be used during the menstrual cycle only.
9. Women of childbearing potential (WOCBP) must use a highly effective form of birth control (confirmed by the Investigator). Rhythm methods will not be considered as highly effective methods of birth control. Highly effective forms of birth control include:
• True sexual abstinence (defined as refraining from heterosexual intercourse for the duration of the study and a minimum of 30 days following the last dose of study drug);
• Vasectomised partner (provided that the partner is the sole sexual partner of the female participant with childbearing potential and that the vasectomised partner has received medical assessment of the surgical success);
• Oral or transdermal combined (oestrogen and progestogen containing) hormonal contraception associated with inhibition of ovulation;
• Oral, injectable or implantable progestogen-only hormone contraception associated with inhibition of ovulation (Depo-Provera™, Implanon);
• Any effective intrauterine device/levonorgestrel intrauterine system;
• Female sterilisation by tubal occlusion;
• Evra Patch™.
WOCBP must agree to use a highly effective method of birth control, as defined above, from enrolment, and at least 14 days prior to Day 1, throughout the study duration and within 30 days after the last dose of IMP.
WOCBP are defined as women who are neither permanently sterilised (hysterectomy, bilateral oophorectomy, or bilateral salpingectomy), nor who are postmenopausal. Women will be considered post-menopausal if they have been amenorrhoeic for 12 months or more without an alternative biological or medical cause e.g. contraceptive method such as Mirena.
10. Male partners of female participants must agree to use condoms during sexual intercourse from the first dose of investigational product until 30 days after the participants last dose to avoid potential transfer of investigational product.
11. Able and willing to abstain from sexual intercourse from 6 hours prior to dosing until 6 hours after dosing;
12. Ability and willingness to attend the necessary visits to the study centre;
13. Ability to comprehend all study related documentation, including written informed consent form, and complete all study-related tasks including daily diary;
14. Be willing and able to adhere to the prohibitions and restrictions specified in the protocol.
Exclusion Criteria:
Participants are excluded from the study if one or more of the following criteria are applicable:
1. Any significant disease or disorder (e.g. cardiovascular, pulmonary, gastrointestinal, hepatic, renal, neurological, musculoskeletal, endocrine, metabolic, malignant, psychiatric, major physical impairment) which, in the opinion of the investigator, may either put the participant at risk because of participation in the study, or may influence the results of the study, or the participant’s ability to participate in the study;
2. Any clinically significant abnormal findings in physical examination, vital signs, haematology, clinical chemistry, or urinalysis during screening and at baseline, which in the opinion of the investigator, may put the participant at risk because of her participation in the study, or may influence the results of the study, or the participant’s ability to complete entire duration of the study;
3. Pregnant, breastfeeding, or lactating women (WOCBP must have a negative serum pregnancy test at screening and a negative urine pregnancy test at the start of each treatment period [i.e. Day 1, Day 28, Day 56]);
4. Women who plan to become pregnant in the next 6 months; 5. History of genital herpes with >3 outbreaks per year, or active non-HP V vaginal infection;
6. Active pelvic infection (positive for gonorrhoea or chlamydial infection, positive test for bacterial vaginosis, Candida vaginitis or trichomonal vaginitis). Participants with positive results can be re tested once during screening;
7. Positive bimanual exam consistent with pelvic inflammatory disease;
8. Positive result for hepatitis B, hepatitis C or human immunodeficiency virus;
9. Current or recent abnormal vaginal discharge and /or abnormal vaginal bleeding, within the 3 months prior to Day 1 as assessed by the investigator;
10. Had an abortion or miscarriage or taken the morning-after pill within the 3 months prior to enrolment;
11. Currently taking immunosuppressants, intra-vaginal preparations, or any prescription that in the opinion of the investigator could be a potential safety issue or interfere with the interpretation of the results;
12. Previous exposure to lopinavir/ritonavir (within 3 months prior to screening), contraindication to the use of lopinavir/ritonavir or known allergy, hypersensitivity, or intolerance to any component of lopinavir/ritonavir ointment excipients;
13. Previous HPV vaccination;
14. Recent history (within 3 months prior to screening) of Stevens- Johnson syndrome, erythema multiforme, urticaria, angioedema, deep vein thrombosis, tinnitus, vertigo, blood glucose disorders, pancreatitis, haemophilia;
15. Receipt of any investigational product within 30 days or 5 half-lives prior to dosing;
16. Employees of the clinical study team or family members (first-degree relatives) of such individuals or anyone involved in the planning and/or conduct of the study. Clinical study team refers to employees directly involved in the study who have been delegated study- related tasks accordingly;
17. Participants who, in the opinion of the Investigator, do not understand the information and procedures of the study, or would not be compliant with them (in particular the study restrictions and risks involved).
Figure imgf000105_0001
Investigational product is administered every day for 21 days for up to 3 cycles as follows:
• Cycle 1 (daily at 8pm from Day 1 to Day 21);
• Cycle 2 (daily at 8pm from Day 29 to Day 49);
• Cycle 3 (daily at 8pm from Day 57 to Day 77).
There is a 7-day treatment cessation in between cycles to allow for menstruation.
Investigational product is self-administered by participants at approximately 8pm (±1 hour) each evening. Participants are provided dosing instructions and important application instructions.
Participants record details of investigational product application in a diary card each day to monitor compliance. Participants are also asked to note in their diary card if there is any waste/spillage.
Participants are required to bring their investigational product and diary card to each study site visit. The tubes are to be weighed prior to dispensing to the participant and again at each clinic visit to assess compliance.
There are no fasting requirements associated with the timing of application of investigational product.
Safety Assessments
The following safety assessments are performed at timepoints outlined in the Schedule of Events, see Table 25.
• Medical History
• Physical Examination
• Vital Signs
• Body Weight and Height
• 12-lead Electrocardiogram
• Clinical Laboratory Safety Tests (Haematology, Biochemistry, ETrinalysis, Vaginal Microbiology, Viral Serology, Drugs of Abuse Screen, Alcohol Screen, Pregnancy Screen) Efficacy Assessments
The efficacy of the investigational product are assessed by improvements in the cytological abnormalities of the uterine cervix. Efficacy assessments are performed at timepoints outlined in the Schedule of Events, see Table 25.
• Colposcopic visual assessment
• Colposcopic Biopsy
• Cytological Sampling
• HPV Genotyping
Table 29
Figure imgf000108_0001
Figure imgf000108_0002
Figure imgf000109_0001

Claims

1. An anhydrous composition for topical application comprising: a. at least one active pharmaceutical ingredient; and
b. a hydrophilic muco-adhesive agent;
wherein upon topical administration of the anhydrous composition to a site of application the anhydrous composition transforms into a muco-adhesive composition.
2. The anhydrous composition according to claim 1 wherein the muco-adhesive
composition provides prolonged retention of the at least one active pharmaceutical ingredient at the site of application.
3. The anhydrous composition according to claim 2 wherein the muco-adhesive
composition provides retention of the at least one active pharmaceutical ingredient at the site of application for at least 15 minutes, at least 30 minutes, at least 1 hour, at least 90 minutes, at least 2 hours, at least 4 hours, at least 6 hours, at least 8 hours, at least 10 hours, at least 12 hours, or at least 24 hours.
4. The anhydrous composition according any preceding claim wherein the muco- adhesive composition has a greater viscosity than the anhydrous composition.
5. The anhydrous composition according to any preceding claim wherein the muco- adhesive composition is a gel.
6. The anhydrous composition according to any preceding claim wherein the hydrophilic muco-adhesive agent is present in the anhydrous composition at about 0.1 to about 20% by weight of the total anhydrous composition weight, such as about 0.1 to about 10% by weight, such as about 0.1 to about 5% by weight, such as about 0.2 to about 4% by weight, such as about 0.5 to about 3% by weight, such as about 0.5 to about 1.5% by weight, or such as about 1% by weight.
7. The anhydrous composition according to any preceding claim wherein the hydrophilic muco-adhesive agent is a cellulose ether or an ionic polymer.
8. The anhydrous composition according to claim 7 wherein the hydrophilic muco- adhesive agent is a cellulose ether selected from methyl cellulose, ethylcellulose and hydroxypropylmethylcellulose.
9. The anhydrous composition according to claim 8 wherein the cellulose ether is hydroxypropylmethylcellulose.
10. The anhydrous composition according to claim 9 wherein the
hydroxypropylmethylcellulose has a degree of methoxy substitution of between 19 and 24% by weight and a degree of hydroxypropyl substitution of between 4 and 12% by weight.
11. The anhydrous composition according to any preceding claim wherein the at least one active pharmaceutical ingredient is present in a dissolved state within the anhydrous composition.
12. The anhydrous composition according to any preceding claim wherein the at least one active pharmaceutical ingredient is stable within the anhydrous composition.
13. The anhydrous composition according to any preceding claim wherein the at least one active pharmaceutical ingredient is a HIV protease enzyme inhibitor.
14. The anhydrous composition according to any preceding claim wherein the HIV
protease enzyme inhibitor is selected from lopinavir and ritonavir.
15. The anhydrous composition according to any preceding claim, wherein the anhydrous composition comprises lopinavir and ritonavir.
16. The anhydrous composition according to claim 15 wherein the molar ratio of lopinavir to ritonavir present in the composition is between about 1:10 and about 18:1, such as between about 1:10 and about 15:1, such as between about 1:5 and about 15:1, such as between about 1:1 and about 15:1, such as between about 2:1 and about 15:1, such as between about 4:1 and about 15:1, such as between about 8:1 and about 14:1, such as between about 9:1 and about 14:1, such as between about 10:1 and about 14:1, such as between 10.5:1 and about 18:1, such as between 10.5:1 and 18:1, such as between about 10.5:1 and about 14:1, such as between about 11:1 to about 13:1, such as between about 11.5 and about 17:1, such as between about 11.5:1 and about 16.0:1, such as between about 11.5:1 and about 15:1, such as about 14.5:1, such as 14.5:1, such as about 14:1, such as 14:1, such as about 13.8:1, such as 13.8:1, such as about 13.75:1, such as 13.75:1, such as about 13.5:1, such as 13.5:1, such as about 13:1, such as 13:1, such as about 12.5:1, such as 12.5:1, such as about 12:1, such as 12:1, such as about 11.75:1, such as 11.75:1, such as about 9:1, such as 9:1, such as about
5:1, such as 5:1, such as about 4.6:1, or such as 4.6:1.
17. The anhydrous composition according to any preceding claim further comprising a solvent for the at least one active pharmaceutical ingredient.
18. The anhydrous composition according to claim 17 wherein the solvent is selected from an unsaturated free fatty acid, PEG castor oil, diethylene glycol, monoethyl ether, propylene glycol, polyethylene glycol, and medium chain triglyceride.
19. The anhydrous composition according to claim 18 wherein the solvent is an
unsaturated free fatty acid.
20. The anhydrous composition according to claim 19 wherein the unsaturated free fatty acid is oleic acid.
21. The anhydrous composition according to claims 17-20 wherein the at least one active pharmaceutical ingredient has a solubility of at least 1% w/v in the solvent.
22. The anhydrous composition according to any preceding claim further comprising a stiffening agent.
23. The anhydrous composition according to claim 22 wherein the stiffening agent is a saturated free fatty acid.
24. The anhydrous composition according to claim 23 wherein the saturated free fatty acid is stearic acid.
25. The anhydrous composition according to any preceding claim further comprising a thickener.
26. The anhydrous composition according to claim 25 wherein the thickener is selected from a mono di glyceride, ceresin wax, and hydrogenated vegetable oil or a combination thereof.
27. The anhydrous composition according to any preceding claim comprising oleic acid, stearic acid and hydroxypropylmethylcellulose.
28. The anhydrous composition according to any preceding claim further comprising a blending agent and/or an antioxidant.
29. The anhydrous composition according to any preceding claim wherein the site of application is a mucosal membrane such as the vaginal wall.
30. The anhydrous composition according to any preceding claim for use as a
medicament.
31. The anhydrous composition according to any preceding claim for use in prevention or treatment of cancerous conditions, for the prevention or treatment of oncogenic viral infections or for the prevention or treatment of benign proliferative disorders.
32. A method of manufacturing the anhydrous composition of claims 1-31 comprising the step of incorporating (e.g. mixing) a hydrophilic muco-adhesive agent into the anhydrous composition.
PCT/IB2019/054289 2018-05-24 2019-05-23 Pharmaceutical compositions WO2019224776A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1808567.0 2018-05-24
GBGB1808567.0A GB201808567D0 (en) 2018-05-24 2018-05-24 Pharmaceutical compositions

Publications (1)

Publication Number Publication Date
WO2019224776A1 true WO2019224776A1 (en) 2019-11-28

Family

ID=62812390

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2019/054289 WO2019224776A1 (en) 2018-05-24 2019-05-23 Pharmaceutical compositions

Country Status (2)

Country Link
GB (1) GB201808567D0 (en)
WO (1) WO2019224776A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11738024B2 (en) 2018-05-24 2023-08-29 Douglas Pharmaceuticals Limited Lopinavir and ritonavir for the treatment of cervix disorders

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998005303A1 (en) * 1996-08-02 1998-02-12 Farmigea S.P.A. Bioadhesive complexes of polycarbophil and azole antifungal or antiprotozoal drugs
WO2003099264A1 (en) * 2002-05-23 2003-12-04 Umd, Inc. Compositions and method for transmucosal drug delivery and cryoprotection
WO2004016245A1 (en) * 2002-08-16 2004-02-26 Mcneil-Ppc Inc. Optimal volume of intra-vaginal semisolid dosage forms for treating vaginal infections
WO2004067063A2 (en) * 2003-01-22 2004-08-12 Umd, Inc. Improved formulations for transmucosal vaginal delivery of bisphosphonates
US20050214230A1 (en) * 2004-03-23 2005-09-29 J. B. Chemicals & Pharmaceuticals Ltd. Novel stomatological gel
EP1782794A2 (en) * 2005-09-12 2007-05-09 Mcneil-PPC, Inc Anhydrous composition containing an acid-acid buffer system
EP1872775A1 (en) * 2006-06-29 2008-01-02 Polichem S.A. Use of a hydrophilic matrix comprising a polyacrylic acid derivative, a cellulose ether and a disintegrant for the manufacture of a medicament for treating female genital disorders
WO2008002353A1 (en) * 2006-06-27 2008-01-03 Dow Pharmaceutical Sciences Two-phase mucoadhesive composition
WO2013006608A1 (en) * 2011-07-05 2013-01-10 Novan, Inc. Topical compositions
WO2015059485A1 (en) * 2013-10-23 2015-04-30 The University Of Manchester Treatment of cancer and benign proliferative disorders

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998005303A1 (en) * 1996-08-02 1998-02-12 Farmigea S.P.A. Bioadhesive complexes of polycarbophil and azole antifungal or antiprotozoal drugs
WO2003099264A1 (en) * 2002-05-23 2003-12-04 Umd, Inc. Compositions and method for transmucosal drug delivery and cryoprotection
WO2004016245A1 (en) * 2002-08-16 2004-02-26 Mcneil-Ppc Inc. Optimal volume of intra-vaginal semisolid dosage forms for treating vaginal infections
WO2004067063A2 (en) * 2003-01-22 2004-08-12 Umd, Inc. Improved formulations for transmucosal vaginal delivery of bisphosphonates
US20050214230A1 (en) * 2004-03-23 2005-09-29 J. B. Chemicals & Pharmaceuticals Ltd. Novel stomatological gel
EP1782794A2 (en) * 2005-09-12 2007-05-09 Mcneil-PPC, Inc Anhydrous composition containing an acid-acid buffer system
WO2008002353A1 (en) * 2006-06-27 2008-01-03 Dow Pharmaceutical Sciences Two-phase mucoadhesive composition
EP1872775A1 (en) * 2006-06-29 2008-01-02 Polichem S.A. Use of a hydrophilic matrix comprising a polyacrylic acid derivative, a cellulose ether and a disintegrant for the manufacture of a medicament for treating female genital disorders
WO2013006608A1 (en) * 2011-07-05 2013-01-10 Novan, Inc. Topical compositions
WO2015059485A1 (en) * 2013-10-23 2015-04-30 The University Of Manchester Treatment of cancer and benign proliferative disorders

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11738024B2 (en) 2018-05-24 2023-08-29 Douglas Pharmaceuticals Limited Lopinavir and ritonavir for the treatment of cervix disorders

Also Published As

Publication number Publication date
GB201808567D0 (en) 2018-07-11

Similar Documents

Publication Publication Date Title
US10668082B2 (en) Vaginal inserted estradiol pharmaceutical compositions and methods
US11266661B2 (en) Vaginal inserted estradiol pharmaceutical compositions and methods
US11246875B2 (en) Vaginal inserted estradiol pharmaceutical compositions and methods
US20190343771A1 (en) Vaginal inserted estradiol pharmaceutical compositions and methods
US11738024B2 (en) Lopinavir and ritonavir for the treatment of cervix disorders
BR112019025914A2 (en) pharmaceutical compositions of estradiol for vaginal insertion and methods
TWI574689B (en) Lower dosage strength imiquimod formulations and short dosing regimens for treating genital and perianal warts
WO2019224777A1 (en) Pharmaceutical compositions
WO2019224776A1 (en) Pharmaceutical compositions
US20230285287A1 (en) Pharmaceutical compositions
US20240148729A1 (en) Lopinavir and ritonavir for the treatment of cervix disorders
US20220257771A1 (en) Compositions comprising lopinavir and treatment of conditions
AU2014252196B2 (en) Progesteron receptor modulators for use in the therapy of uterine fibroids
KR20180100567A (en) Vaginal insertion type estradiol pharmaceutical composition and method
RU2713888C2 (en) Pharmaceutical compositions and methods based on oestradiol for intravaginal introduction
SA01210754B1 (en) Pharmaceutical combination of DROSPIRENONE and estrogen for hormone replacement therapy

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: 19731796

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19731796

Country of ref document: EP

Kind code of ref document: A1