WO2013148682A1 - Procédés et compositions pour l'administration contrôlée d'agents phytochimiques - Google Patents

Procédés et compositions pour l'administration contrôlée d'agents phytochimiques Download PDF

Info

Publication number
WO2013148682A1
WO2013148682A1 PCT/US2013/033877 US2013033877W WO2013148682A1 WO 2013148682 A1 WO2013148682 A1 WO 2013148682A1 US 2013033877 W US2013033877 W US 2013033877W WO 2013148682 A1 WO2013148682 A1 WO 2013148682A1
Authority
WO
WIPO (PCT)
Prior art keywords
polymeric matrix
biocompatible polymeric
curcumin
cancer
phytochemical
Prior art date
Application number
PCT/US2013/033877
Other languages
English (en)
Inventor
Ramesh C. Gupta
Manicka V. Vadhanam
Farrukh Aqil
Original Assignee
University Of Louisville Research Foundation, Inc.
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
Priority claimed from US13/429,601 external-priority patent/US8858995B2/en
Application filed by University Of Louisville Research Foundation, Inc. filed Critical University Of Louisville Research Foundation, Inc.
Publication of WO2013148682A1 publication Critical patent/WO2013148682A1/fr

Links

Classifications

    • 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
    • A61K47/40Cyclodextrins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the presently-disclosed subject matter relates to methods and compositions for the controlled delivery of phytochemical agents.
  • the presently-disclosed subject matter relates to methods and compositions for treating cancer wherein a phytochemical agent is incorporated into and released in a controlled manner from a biocompatible and biodegradable polymeric matrix.
  • chemotherapeutic agents are administered orally or intravenously in bolus doses to elicit a response.
  • Oral delivery of the chemotherapeutic agents often presents a bioavailability problem, as a large part of the agents is destroyed in the gut and the remainder is, at least in part, detoxified during first pass in the liver. Systemic delivery overcomes this hurdle, but this route of administration often leads to undesirable dose spikes.
  • chemotherapeutic agents is that high doses of the agents are usually required in order to achieve an effective tissue or plasma concentration, but the high doses generally result in undesirable toxicity.
  • a composition that comprises a biocompatible polymeric matrix incorporating an effective amount of a phytochemical agent.
  • the composition is capable of releasing a controlled low dose of the phytochemical agent over a time period. In some embodiments, the time period is at least about 18 months.
  • the biocompatible polymeric matrix comprises a polymer that can be selected from polycaprolactone, cyclodextrin, F68, polyethylene glycol, and combinations thereof.
  • the biocompatible polymeric matrix is comprised of polycaprolactone in combination with a second polymer that, in some embodiments, can be selected from cyclodextrin, F68, and polyethylene glycol.
  • the polycaprolactone is combined with the second polymer in a ratio of about 4: 1 or about 9: 1 to form the biocompatible polymeric matrix.
  • the biocompatible polymeric matrix comprises
  • the biocompatible polymeric matrix comprises polycaprolactone and polyethylene glycol in a ratio of about 65:35. In some embodiments, the biocompatible matrix is biodegradable.
  • the phytochemical agent comprises about 2% to about 50% of the weight of the polymer or polymers forming the biocompatible polymeric matrix.
  • the phytochemical agent is selected from curcumin, green tea polyphenols,
  • biocompatible polymeric matrix is a combination of one or more individual phytochemical agents.
  • the phytochemical agent is a combination of: oltipraz and curcumin; curcumin, ellagic acid, co-enzyme Q-10, and lycopene; curcumin, green tea polyphenols, diindolylmethane, and punicalagin; delphinidin, petunidin, malvidin, peonidin, and cyanidin; or punicalagin and delphinidin.
  • each phytochemical agent can be incorporated into the same biocompatible polymeric matrix or, in some embodiments, each phytochemical agent one or more additional biocompatible polymeric matrices such that the one or more additional biocompatible polymeric matrices each incorporate a single phytochemical agent.
  • a second biocompatible matrix is provided that incorporates at least one phytochemical agent.
  • compositions of the presently-disclosed subject matter can further comprise an effective amount of an anti-inflammatory agent or an effective amount of a chemotherapeutic agent.
  • the anti-inflammatory agents and the chemotherapeutic agents are incorporated into the same biocompatible polymeric matrix that also incorporates a phytochemical agent.
  • the antiinflammatory agents and the chemotherapeutic agents are incorporated into a second biocompatible polymeric matrix.
  • the compositions further include a polymeric coating that surrounds the biocompatible polymeric matrix that incorporates an effective amount of the phytochemical agent.
  • the polymeric coating is comprised of polycaprolactone.
  • the polymeric coating comprises one or more layers, such as, in some embodiments, about 5 to about 40 layers.
  • the biocompatible polymeric matrix itself, which incorporates the effective amount of the phytochemical agents is provided in the form of a coating such that the compositions of the presently-disclosed subject matter can be utilized as a coating for additional compositions.
  • a method for treating a cancer comprises administering to a subject in need thereof an effective amount of a
  • U 024:UN106:923010:1 :LOUISVILLE - composition comprising a biocompatible polymeric matrix incorporating a phytochemical agent, where the phytochemical agent is incorporated into and released over a time period from the biocompatible polymeric matrix at a controlled low dose of the phytochemical agent.
  • the time period is at least about 18 months.
  • a method of treating a cancer further comprises administering an effective amount of an anti-inflammatory agent or a
  • chemotherapeutic agent that is incorporated into and released over a time period from the biocompatible polymeric matrix or a second biocompatible polymeric matrix.
  • the cancer is selected from breast cancer, lung cancer, or cervical cancer.
  • the subject is at an increased risk of developing a primary cancer, which, in some embodiments, is breast cancer, lung cancer, or cervical cancer.
  • the subject is at an increased risk of developing a secondary cancer.
  • a method for treating a cancer wherein a phytochemical agent is administered to a subject by subcutaneous implantation of a biocompatible matrix incorporating a phytochemical agent.
  • the phytochemical agent is administered by implantation of a biocompatible polymeric matrix incorporating that phytochemical agent at a site distant from the site of the cancer.
  • the implantation occurs at a site of a cancer or at a site suspected of developing a cancer.
  • the device for insertion into a cervical canal.
  • the device comprises: a substantially cylindrical shaft having a proximal end and a distal end, and defining a canal that extends from the proximal end to the distal end; and, a cap attached to the proximal end of the cylindrical shaft that includes a central opening in registry with the canal defined by the shaft.
  • the device is comprised of a biocompatible polymeric matrix incorporating an effective amount of a phytochemical agent in accordance with the presently-disclosed subject matter.
  • a device where the cylindrical shaft is suitably adapted for engaging the walls of a cervical canal and where a bottom surface of the cap is suitably adapted for engaging an external orifice of a uterus.
  • a device is provided where the canal defined by the cylindrical shaft allows for the passage of bodily fluids from the uterus.
  • the bottom surface of the cap has a concave shape such that the bottom surface of the cap suitably engages an external orifice of the uterus.
  • a device for cervical insertion is provided where the distal end of the cylindrical shaft is tapered.
  • a device for cervical insertion is provided where the cylindrical shaft is about 19 to about 25 mm in length and about 9 to 11 mm in diameter; the cap is about 20 to about 25 mm in diameter; and, the canal is about 4 mm to about 5 mm in diameter.
  • FIGURE 1 is a picture of exemplary compositions of the presently-disclosed subject matter, and including an image of a biocompatible polymeric matrix without the incorporation of a phytochemical agent (Sham) and images of biocompatible polymeric matrices incorporating various phytochemical agents including curcumin, ellagic acid, oltipraz, and green tea polyphenols (GTPs).
  • a phytochemical agent Sham
  • GTPs green tea polyphenols
  • FIGURE 2 includes graphs depicting the release of curcumin from a
  • FIG. 2A is a graph depicting the daily release of curcumin from a biocompatible polymeric matrix where the amount ⁇ g) of curcumin released per day (y-axis) and the percentage of curcumin released from the matrix per day (y- axis) are plotted against a release period measured in days (x-axis).
  • Figure 2B is a graph depicting the cumulative release of curcumin from a biocompatible polymeric matrix where the total amount ⁇ g) of curcumin released from the matrix (y-axis) and the total percentage of curcumin released from the matrix (y-axis) are plotted against a release period measured in days (x-axis).
  • FIGURE 3 includes graphs depicting the release of green tea polyphenols (GTPs) from a biocompatible polymeric matrix in vitro.
  • Figure 3A is a graph depicting the daily release of GTPs from a biocompatible polymeric matrix where the amount ⁇ g) of GTPs released per day (y-axis) and the percentage of GTPs released from the matrix per day (y- axis) are plotted against a release period measured in days (x-axis).
  • Figure 3B is a graph depicting the cumulative release of GTPs from a biocompatible polymeric matrix where the
  • U 024:UN106:923010:1 :LOUISVILLE - total amount ⁇ g) of GTPs released from the matrix (y-axis) and the total percentage of GTPs released from the matrix (y-axis) are plotted against a release period measured in days (x- axis).
  • FIGURE 4 includes graphs depicting the release of diindolylmethane (DIM) from a biocompatible polymeric matrix in vitro.
  • Figure 4A is a graph depicting the daily release of DIM from a biocompatible polymeric matrix where the amount ⁇ g) of DIM released per day (y-axis) and the percentage of DIM released from the matrix per day (y-axis) are plotted against a release period measured in days (x-axis).
  • Figure 4B is a graph depicting the cumulative release of DIM from a biocompatible polymeric matrix where the total amount ⁇ g) of DIM released from the matrix (y-axis) and the total percentage of DIM released from the matrix (y-axis) are plotted against a release period measured in days (x-axis).
  • FIGURE 5 includes graphs depicting the release of punicalagins from a biocompatible polymeric matrix in vitro.
  • Figure 5A is a graph depicting the daily release of punicalagins from a biocompatible polymeric matrix where the amount ⁇ g) of punicalagins released per day (y-axis) and the percentage of punicalagins released from the matrix per day (y-axis) are plotted against a release period measured in days (x-axis).
  • Figure 5B is a graph depicting the cumulative release of punicalagins from a biocompatible polymeric matrix where the total amount ⁇ g) of punicalagins released from the matrix (y-axis) and the total percentage of punicalagins released from the matrix (y-axis) are plotted against a release period measured in days (x-axis).
  • FIGURE 6 includes graphs depicting the release of oltipraz from a biocompatible polymeric matrix in vitro.
  • Figure 6A is a graph depicting the daily release of oltipraz from a biocompatible polymeric matrix where the amount ⁇ g) of oltipraz released per day (y-axis) is plotted against a release period measured in days (x-axis).
  • Figure 6B is a graph depicting the cumulative release of oltipraz from a biocompatible polymeric matrix where the total amount ⁇ g) of oltipraz released from the matrix (y-axis) is plotted against a release period measured in days (x-axis).
  • FIGURE 7 includes graphs depicting the release of lycopene from a
  • Figure 7A is a graph depicting the daily release of lycopene from a biocompatible polymeric matrix where the amount ⁇ g) of lycopene released per day (y-axis) is plotted against a release period measured in days (x-axis).
  • Figure 7B is a graph depicting the cumulative release of lycopene from a biocompatible polymeric matrix where the total amount ⁇ g) of lycopene released from the matrix (y-axis) is plotted against a release period measured in days (x-axis).
  • FIGURE 8 includes graphs depicting the release of resveratrol from a
  • Figure 8A is a graph depicting the daily release of resveratrol from a biocompatible polymeric matrix where the amount ⁇ g) of resveratrol released per day (y-axis) is plotted against a release period measured in days (x-axis).
  • Figure 8B is a graph depicting the cumulative release of resveratrol from a biocompatible polymeric matrix where the total amount ⁇ g) of resveratrol released from the matrix (y-axis) is plotted against a release period measured in days (x-axis).
  • FIGURE 9 includes graphs depicting the effect of incorporating various loads of curcumin into a biocompatible polymeric matrix in vitro, including incorporating 2% w/w of curcumin ( ⁇ ), 5% w/w of curcumin ( ), and 20% w/w of curcumin (A) into separate biocompatible polymeric matrices.
  • Figure 9A is a graph depicting the daily release of curcumin from each of the biocompatible polymeric matrices where the amount ⁇ g) of curcumin released per day (y-axis) from each biocompatible polymeric matrix is plotted against a release period measured in days (x-axis).
  • Figure 9B is a graph depicting the cumulative release of curcumin from each biocompatible polymeric matrix where the total amount ⁇ g) of curcumin released from each matrix (y-axis) is plotted against a release period measured in days (x-axis).
  • FIGURE 10 includes graphs depicting the effect of incorporating various loads of GTPs into a biocompatible polymeric matrix in vitro, including incorporating 1% w/w of GTPs ( ⁇ ), 2.5% w/w of GTPs (is), 5% w/w of GTPs (A), and 10% w/w of GTPs ( «) into a separate biocompatible polymeric matrices comprised of polycaprolactone, MW 65K polymers and polycaprolactone, MW 15K polymers that were combined in a 1 :4 ratio .
  • Figure 1 OA is a graph depicting the daily release of GTPs from each of the biocompatible polymeric matrices where the amount ⁇ g) of GTPs released per day (y-axis) from each biocompatible polymeric matrix is plotted against a release period measured in days (x-axis).
  • Figure 10B is a graph depicting the cumulative release of GTPs from each biocompatible polymeric matrix where the total amount ⁇ g) of GTPs released from each matrix (y-axis) is plotted against a release period measured in days (x-axis).
  • FIGURE 1 1 is a graph depicting the short-term in vivo release of curcumin from a biocompatible polymeric matrix where the total amount (mg) of curcumin released (y-axis) and the total percentage of curcumin released (y-axis) from the matrix are plotted against a release period measured in days (x-axis).
  • FIGURE 12 includes graphs depicting the levels of curcumin (ng/g) in lung tissue (Figure 12A) and liver tissue (Figure 12B) of rats (y-axis) at different time intervals (x-axis),
  • FIGURE 13 includes graphs depicting the long-term in vivo release of curcumin from a biocompatible polymeric matrix.
  • Figure 13A is a graph depicting the daily release of curcumin from a biocompatible polymeric matrix where the total amount (mg) of curcumin released per week (y-axis) and the total percentage of curcumin released per week (y-axis) from the matrix are plotted against a treatment period measured in weeks (x-axis).
  • Figure 13B is a graph depicting the cumulative release of curcumin from a biocompatible polymeric matrix where the total amount (mg) of curcumin released (y-axis) and the total percentage of curcumin released (y-axis) are plotted against a treatment period measured in weeks (x-axis).
  • FIGURE 14 includes graphs depicting the long-term in vivo release of GTPs from a biocompatible polymeric matrix.
  • Figure 14A is a graph depicting the daily release of GTPs from a biocompatible polymeric matrix where the total amount (mg) of GTPs released per week (y-axis) and the total percentage of GTPs released per week (y-axis) from the matrix are plotted against a treatment period measured in weeks (x-axis).
  • Figure 14B is a graph depicting the cumulative release of GTPs from a biocompatible polymeric matrix where the total amount (mg) of GTPs released (y-axis) and the total percentage of GTPs released (y- axis)are plotted against a treatment period measured in weeks (x-axis).
  • FIGURE 15 includes graphs depicting the long-term in vivo release of DIM from a biocompatible polymeric matrix.
  • Figure 15A is a graph depicting the daily release of DIM from a biocompatible polymeric matrix where the total amount (mg) of DIM released per week (y-axis) and the total percentage of DIM released per week (y-axis) from the matrix are plotted against a treatment period measured in weeks (x-axis).
  • Figure 15B is a graph depicting the cumulative release of DIM from a biocompatible polymeric matrix where the total amount (mg) of DIM released (y-axis) and the total percentage of DIM released (y-axis) are plotted against a treatment period measured in weeks (x-axis).
  • FIGURE 16 includes graphs depicting the long-term in vivo release of punicalagins from a biocompatible polymeric matrix.
  • Figure 16A is a graph depicting the daily release of punicalagins from a biocompatible polymeric matrix where the total amount (mg) of punicalagins released per week (y-axis) and the total percentage of punicalagins released per week (y-axis) from the matrix are plotted against a treatment period measured in weeks (x-axis).
  • Figure 16B is a graph depicting the cumulative release of punicalagins from a biocompatible polymeric matrix where the total amount (mg) of punicalagins released (y-
  • FIGURE 17 is a graph depicting the effect of curcumin, administered by subcutaneous implantation of a biocompatible polymeric matrix incorporating 20% w/w curcumin or by diet, on the formation of DNA adducts in liver and lung tissue of rats treated with a single bolus dose of benzo[a]pyrene.
  • FIGURE 18 is a graph depicting the effect of oltipraz, administered by subcutaneous implantation of a biocompatible polymeric matrix incorporating 10% w/w oltipraz, on the formation of DNA adducts in liver tissue of rats treated with a bolus dose of dibenzo[a, 1 ]pyrene.
  • FIGURE 19 is a graph depicting the effect of GTPs, administered by
  • FIGURE 20 is a graph depicting the effect of curcumin, administered by subcutaneous implantation of a 1 cm biocompatible polymeric matrix incorporating 20% w/w curcumin, on the formation of DNA adducts in liver and lung tissue of rats treated with a sustained low dose of benzo[a]pyrene.
  • FIGURE 21 is a graph depicting the effect of GTPs, administered by
  • FIGURE 22 includes graphs depicting the effect of GTPs, administered by subcutaneous implantation of two 2-cm biocompatible polymeric matrices incorporating 10% w/w GTPs, on expression of various cytochrome P450 (CYP) mRNA in liver tissue of rats treated with a sustained low dose of benzo[a]pyrene.
  • Figure 22A is a graph depicting the effect of GTPs on the expression of CYP 1A1.
  • Figure 22B is a graph depicting the effect of GTPs on the expression of CYP 1B 1.
  • FIGURE 23 includes graphs depicting the modulation of 17 -estradiol-induced mammary tumor volume (Figure 23 A) and mammary tumor multiplicity (Figure 23B) by silastic tubing implants incorporating either ellagic acid, lycopene, or curcumin.
  • FIGURE 24 includes graphs depicting the inhibition of 17 -estradiol-induced mammary tumor multiplicity (Figure 24A) and mammary tumor volume (Figure 24B) by
  • U 024:UN106:923010:1 :LOUISVILLE - ellagic acid administered by a silastic tubing implant incorporating ellagic acid (slow release) or by dietary supplementation with ellagic acid.
  • FIGURE 25 is a graph depicting the effect of a biocompatible silastic tubing implant incorporating fennel extract on 17p-estradiol-induced mammary cell proliferation as measured by cytological evaluation of histological sections for cellular markers of proliferation, including proliferating cell nuclear antigen (PCNA) and the Ki-67 protein.
  • PCNA proliferating cell nuclear antigen
  • FIGURE 26 includes graphs depicting the inhibition of 17 -estradiol-induced mammary tumor volume (Figure 26A) and mammary tumor multiplicity (Figure 26B) by the combined administration of phytochemical agents incorporated into biocompatible silastic tubing implants and implanted subcutaneously (A: sham implant; B: one implant each of oltipraz and curcumin; and, C: one implant each of curcumin, ellagic acid, coenzyme-Q 10, and lycopene), where the numbers on the bars represent percent reduction.
  • A sham implant
  • B one implant each of oltipraz and curcumin
  • C one implant each of curcumin, ellagic acid, coenzyme-Q 10, and lycopene
  • FIGURE 27 is a graph depicting the effect of combined administration of curcumin, GTPs, DIM, and punicalagin implants, comprised of a biocompatible polymeric matrix, on mammary cell proliferation in rats treated with 17 -estradiol, where the percentage of PCNA positive cells is plotted against treatment groups at different time intervals.
  • FIGURE 28 is a graph depicting the synergistic activity of various phytochemical agents in inhibiting growth of HI 299 human lung cancer cells, where the various phytochemical agents (y-axis) are plotted against the half maximal inhibitory concentration (IC 50 ) for each agent (x-axis).
  • FIGURE 29 is a graph depicting the synergistic activity of various phytochemical agents in inducing apoptosis in HI 299 human lung cancer cells where the various phytochemical agents (y-axis) are plotted against the percentage of cells undergoing apoptosis (x-axis).
  • FIGURE 30 is a side view of a device for insertion into a cervical canal made in accordance with the presently-disclosed subject matter.
  • FIGURE 31 is a side view of a device for insertion into a cervical canal made in accordance with the presently-disclosed subject matter and depicting exemplary dimensions of features of the device.
  • FIGURE 32 includes perspective views ( Figures 32A, 32B, 32C, and 32D) of an exemplary device for cervical insertion.
  • FIGURE 33 includes schematic diagrams depicting the placement of an exemplary device for cervical insertion.
  • Figure 33A is a schematic diagram depicting a front
  • Figure 33B is a schematic diagram depicting a side view of the device and showing the device engaging the walls of a cervical canal and the external orifice of a uterus.
  • FIGURE 34 includes graphs depicting the release of curcumin from an exemplary device for cervical insertion in vitro.
  • Figure 34A is a graph depicting the daily release of curcumin from an exemplary device for cervical insertion where the amount ⁇ g) of curcumin released per day (y-axis) and the percentage of curcumin released from the device per day (y- axis) are plotted against a release period measured in days (x-axis).
  • Figure 34B is a graph depicting the cumulative release of curcumin from an exemplary device for cervical insertion where the total amount ⁇ g) of curcumin released from the device (y-axis) and the total percentage of curcumin released from the device (y-axis) are plotted against a release period measured in days (x-axis).
  • FIGURE 36 includes graphs depicting the average daily release of curcumin from a 2-cm biocompatible polymeric matrix incorporating 40 mg of curcumin under in vivo conditions, where the biocompatible polymeric matrix was formed from polycaprolactone- 12 IK and polyethylene glycol-8K in a 65:35 ratio, where the compositions were
  • FIGURE 37 is a graph depicting curcumin levels in the plasma of ACI rats treated with biocompatible polymeric matrix compositions formed from polycaprolactone-121K and polyethylene glycol-8K in a 65:35 ratio and incorporating curcumin (two 2-cm implants; 20 mg curcumin/cm) or treated with a curcumin diet (1000 ppm), where 1.5 ml plasma was extracted from each time point after pooling and analyzed by HPLC using a fluorescence detector.
  • FIGURE 38 is a graph depicting liver curcumin levels in ACI rats treated with biocompatible polymeric matrix compositions formed from polycaprolactone-121K and polyethylene glycol-8K in a 65:35 ratio and incorporating curcumin (two 2-cm implants, 20 mg/cm implant) or treated with a curcumin diet (1000 ppm), where, based on about a 10 g
  • the total curcumin administered over 90 days corresponded to 900 mg (or approximately 50 mg/kg b.wt.) as opposed to 38 mg delivered by both the implants combined (2.1 1 mg/kg b.wt.), and where tissues (approximately 500 mg) from individual animals were extracted and analyzed by HPLC coupled with a fluorescence detector.
  • FIGURE 39 is a graph depicting brain curcumin levels in ACI rats treated with biocompatible polymeric matrix compositions formed from polycaprolactone-121K and polyethylene glycol-8K in a 65:35 ratio and incorporating curcumin (two 2-cm implants, 20 mg/cm) or treated with dietary curcumin (1000 ppm), where tissues (approximately 500 mg) from individual animals were extracted and analyzed by HPLC coupled with a fluorescence detector.
  • FIGURE 40 is a graph depicting the effect of curcumin administered by biocompatible polymeric matrix compositions formed from polycaprolactone-121K and polyethylene glycol-8K in a 65:35 ratio and incorporating curcumin (two 2-cm implants, 20 mg/cm) or by diet (1000 ppm) on CYP1A1 protein expression in hepatic microsomes of ACI rats, where sham implants (blank implants prepared without curcumin) and curcumin diet groups were compared with untreated controls, and where curcumin implants were compared with sham implants for all statistical purposes at a significance level of p value ⁇ 0.05.
  • FIGURE 41 is a graph showing the effect of curcumin administered by biocompatible polymeric matrix compositions formed from polycaprolactone-121K and polyethylene glycol-8K in a 65:35 ratio and incorporating curcumin (two 2-cm implants, 20 mg/cm) or by diet (1000 ppm) on CYP3A4 activity in hepatic microsomes of ACI rats, where sham implants (blank implants prepared without curcumin) and curcumin diet groups were compared with untreated controls, and where curcumin implants were compared with sham implants for all statistical purposes at a significance level ⁇ value ⁇ 0.05.
  • FIGURE 42 is a graph showing the effect of coating each biocompatible polymeric matrix composition incorporating curcumin with various numbers of layers of blank polycaprolactone to minimize the initial burst release, where the compositions were coated with a 10% solution of polycaprolactone-80K (mol. wt. 80,000) in dichloromethane with intermittent drying.
  • FIGURE 43 includes graphs depicting the percent average in vitro daily and cumulative release of oltipraz (Figure 43A), withaferin A ( Figure 43B), and curcumin (Figure 43C) from biocompatible polymeric matrices prepared by coating 20 to 30 layers of polycaprolactone incorporating the phytochemical agents (2 cm length, 2.6 mm dia), where the load of phytochemical agent was 20%, except for withaferin A which was 3%, and where
  • FIGURE 44 includes graphs depicting the percent average in vitro daily and cumulative release of curcuminoid I ( Figure 44A), curcuminoid II ( Figure 44B), curcuminoid III ( Figure 44C), and mixture of three curcuminoids in natural ratio as presented in curcumin (75:20:5; Figure 44D) from biocompatible polymeric matrices incorporating the
  • phytochemical agents and prepared by the coating procedure (2 cm length, 2.6 mm dia; 10% drug load), where the compositions were agitated in a shaker incubator in PBS supplemented with 10% bovine serum and the media was changed daily, and where the release was measured spectrophotometrically against the standard curve of individual curcuminoids.
  • FIGURE 45 includes graphs depicting the rate of in vitro and in vivo release of curcumin, oltipraz and withaferin A (Figure 45A) from coated biocompatible polymeric matrices grafted subcutaneously in female A/J mice where the animals were euthanized after 3 weeks and the implants recovered were solvent extracted to measure the residual amount as described in text, and depicting the in vivo cumulative release of withaferin A (Figure 45B) from coated biocompatible polymeric matrices (1.5 cm, 5% drug load) grafted
  • FIGURE 46 includes images ( Figure 46A) and a graph ( Figure 46B) depicting the inhibition of dibenzo[a,/]pyrene (DBP)-DNA adducts in lung tissue of female A/J mice by phytochemical agents delivered via biocompatible polymeric matrices of the presently- disclosed subject matter, where animals were treated with DBP by subcutaneous polymeric implants or implants of oltipraz and withaferin A (two 1.5 cm, 2.6 mm dia; 20% and 5% loads, respectively), and where, three weeks later, animals were euthanized and lung DNA adducts were analyzed by 32 P-postlabeling.
  • DBP dibenzo[a,/]pyrene
  • FIGURE 47 includes graphs depicting curcuminoid levels in lung (Figure 47 A), liver ( Figure 47B) and brain (Figure 47C) of female A/J mice treated with curcumin via biocompatible polymeric matrices (two, 1.5-cm implants; 10% drug load) for 21 days, where samples were analyzed by ultra performance liquid chromatography (UPLC).
  • UPLC ultra performance liquid chromatography
  • FIG. 48 includes graphs and an image depicting the effects of polymeric implants incorporating withaferin A and prepared by a coating procedure on the inhibition of human lung cancer A549 cell xenografts in nude mice, where, following inoculation with human lung cancer A549 cells (2.5 x 10 6 cells), when tumor xenografts grew to over 50 mm 3 ,
  • U 024:UN106:923010:1 :LOUISVILLE - are approximations that can vary depending upon the desired properties sought to be obtained by the presently-disclosed subject matter.
  • the term "about,” when referring to a value or to an amount of mass, weight, time, volume, concentration or percentage is meant to encompass variations of in some embodiments ⁇ 20%, in some embodiments ⁇ 10%, in some embodiments ⁇ 5%, in some embodiments ⁇ 1%, in some embodiments ⁇ 0.5%, and in some embodiments ⁇ 0.1% from the specified amount, as such variations are appropriate to perform the disclosed method.
  • phytochemical agents are known to have many health-promoting properties, including reducing cancer mortality, their application as an effective agent for the prophylaxis and treatment of cancer has been limited by the delivery of these agents orally in a capsule, tablet, or powdered format.
  • the biggest drawback of phytochemical agents in the prophylaxis and treatment of cancer is that oral delivery of these agents results in limited bioavailability. Similar to traditional chemotherapeutic agents, when
  • phytochemical agents are administered orally, a large part of the agent is destroyed in the gut and a large portion of the remaining phytochemical agent is detoxified during first pass through the liver. As such, a high oral dose of the phytochemical agent is required to achieve a reasonable therapeutic effect.
  • Systemic delivery overcomes this hurdle, but again, like traditional chemotherapeutic agents, systemic delivery can lead to undesirable dose spikes and potential toxicity. To that end, the presently-disclosed subject matter provides new compositions, and methods of using the same, for administering phytochemical agents in a controlled low dose over a period of time.
  • a composition that comprises a biocompatible polymeric matrix incorporating an effective amount of a phytochemical agent.
  • the composition is capable of releasing a controlled low dose of the phytochemical agent over a time period. In some embodiments, the time period is at least about 18 months.
  • biocompatible is used herein to refer to a composition that is substantially non-toxic in the in vivo environment of its intended use, and that is not substantially rejected by the subject's physiological system (i.e., is non-antigenic).
  • biocompatibility of a particular composition can be gauged by the composition's toxicity, infectivity, pyrogenicity, irritation potential, reactivity, hemolytic activity, carcinogenicity, and/or immunogenicity.
  • a biocompatible composition When introduced into a majority of subjects, a biocompatible composition will not cause an undesirably adverse, long-lived, or escalating biological reaction or response, and is distinguished from a mild, transient inflammation, which typically accompanies surgery or implantation of foreign objects into a living organism.
  • a biocompatible polymeric matrix of the presently-disclosed subject matter can be fabricated from a variety of polymeric materials.
  • polymeric materials are broadly classified as synthetic, natural, or blends thereof, and within these broad classes, the polymeric materials can be further classified as biodegradable or biostable.
  • Biodegradable polymers degrade in vivo as a function of chemical composition, method of manufacture, and composition structure.
  • Biostable polymers on the other hand, remain intact in vivo for extended periods of time ranging from several years to more and include polymers such as ethylene-vinyl acetate copolymers, polyurethanes, polyacrylonitriles, and certain
  • a biocompatible polymeric matrix is provided that is biodegradable.
  • both synthetic and natural polymers can be used, with synthetic polymers being preferred due to a more uniform and reproducible degradation, as well as other physical properties.
  • synthetic biodegradable polymers capable of being used in accordance with the presently-disclosed subject matter include, but are not limited to, polycaprolactone, polyanhydrides, polyhydroxyacids such as polylactic acid, polyglycolic acids and copolymers thereof, polyesters, polyamides, polyorthoesters, and certain polyphazenes. Examples of naturally-occurring biodegradable polymers capable of being
  • U 024:UN106:923010:1 :LOUISVILLE - used in accordance with the presently-disclosed subject matter include, but are not limited to, proteins and polysaccharides such as collagen, hylauronic acid, albumin, and gelatin.
  • a biocompatible polymeric matrix is provided that is comprised of a polymers selected from polycaprolactone, cyclodextrin, F68, polyethylene glycol, and combinations thereof.
  • the biocompatible polymeric matrix comprises polycaprolactone in combination with a second polymer.
  • the polycaprolactone and the second polymer are combined in a ratio of about 4: 1 or about 9: 1.
  • the second polymer is selected from cyclodextrin, F68, and polyethylene glycol.
  • the biocompatible polymeric matrix comprises polycaprolactone and F68 in a ratio of about 4: 1.
  • the biocompatible polymeric matrix comprises polycaprolactone and polyethylene glycol in a ratio of about 65:35 as, in some embodiments, such a combination and/or ratio of polymers is useful for increasing the release of a phytochemical or other therapeutic agent from the biocompatible polymeric matrices of the presently-disclosed subject matter.
  • a composition of the presently-disclosed subject matter can be formulated by dissolving one or more polymers in an appropriate solvent to initiate the formation of the biocompatible polymeric matrix.
  • One or more phytochemical agents are then dissolved in either the same solvent as the one or more polymers or another appropriate solvent depending on the solubility of the particular phytochemical agent.
  • the solvents can then be evaporated under reduced pressure to produce a composition where the phytochemical agents are embedded or entrapped within a polymeric matrix.
  • phytochemical agents generally have been delivered orally through the diet of a subject. Indeed, almost all published animal studies to date have used a dietary route for administering phytochemical agents, with the dosages of the phytochemical agents ranging from a 10-1000 mg/kg per day in a given diet. When these dose ranges are extrapolated for use in clinical studies, however, the dosage range becomes alarmingly high and thus imposes a risk of toxicity to the subject.
  • compositions that effectively releases phytochemical agents at levels that are significantly lower relative to levels provided by a dietary route (i.e., oral delivery) and furthers provides constant, steady- state dosages of the phytochemical agents over a time period, or in other words, provides a "controlled low dose" of the phytochemical agents.
  • a dietary route i.e., oral delivery
  • U 024:UN106:923010:1 :LOUISVILLE - amount of a phytochemical agent into a biocompatible polymeric matrix of the presently- disclosed subject matter allows for similar amounts of the phytochemical agent to be released each day as the agent diffuses from the matrix and the biocompatible polymeric matrix degrades in a uniform manner.
  • the presently-disclosed compositions are capable of lowering the effective dose delivered to a given subject, if desired, to thereby minimize toxicity while also delivering the controlled dose at a constant, steady-state level over extended time periods to increase bioavailability and to elicit a biological response.
  • the controlled low dose of the phytochemical agent is released over a time period of at least about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, or about 24 months from the time of administration.
  • a bolus dose of the phytochemical agent can be released from the biocompatible polymeric matrix over a time period of about 1 to about 2 weeks from the time of administration, and the controlled low dose of the phytochemical agent is released over a time period beginning at about 2 to about 3 weeks after administration and extending to about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 1 1, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, or about 24 months from the time of administration.
  • a composition of the presently-disclosed subject matter is provided that releases a controlled low dose of the phytochemical agent over a time period of at least about 18 months.
  • phytochemical agent refers to a non-nutritive plant- derived compound, or an analog thereof, that is capable of “treating” a cancer, as defined herein below.
  • phytochemical agents include, but are not limited to compounds such as monophenols; flavonoids, such as flavonols, flavanones, flavones, flavan-3-ols, anthocyanins, anthocyanidins, isoflavones, dihydroflavonols, chalcones, and coumestans; phenolic acids; hydroxycinnamic acids; lignans; tyrosol esters; stillbenoids; hydrolysable tannins; carotenoids, such as carotenes and xanthophylls; monoterpenes; saponins; lipids, such as phytosterols, tocopherols, and omega-3,6,9 fatty acids; diterpenes; triterpinoids
  • the phytochemical agent can be an analog of a plant-derived compound, such as oltipraz, which is an analog of l,2-dithiol-3-thione, a compound that is found in many cruciferous
  • Table 1 provides a list of specific phytochemical agents that are exemplary of the broader classes of phytochemical agents described herein above.
  • the phytochemical agent is selected from curcumin, green tea polyphenols, punicalagin, diindolylmethane, oltipraz, tocotrienol, tocopherol, plumbagin, cyanidin, delphinidin, lycopene, lupeol, curcurbitacin-B, Withaferin A, indole-3-carbinol, genestein, equol, resveratrol, co-enzyme Q-10, ellagic acid, petunidin, malvidin, peonidin, fennel extract, and combinations thereof.
  • the phytochemical agent is a combination of oltipraz and curcumin; curcumin, ellagic acid, co-enzyme Q-10, and lycopene; curcumin, green tea polyphenols, diindolylmethane, and punicalagin; delphinidin, petunidin, malvidin, peonidin, and cyanidin; or punicalagin and delphinidin.
  • one or more phytochemical agents are incorporated into and a released from a single biocompatible polymeric matrix.
  • a second biocompatible polymeric matrix can be provided that incorporates at least one phytochemical agent such that multiple biocompatible polymeric matrices are provided, wherein each biocompatible polymeric matrix incorporates at least one phytochemical agent.
  • a composition that comprises a biocompatible matrix comprising polycaprolactone and F68 in a ratio of about 4: 1 that incorporates an effective amount of curcumin such that the composition releases a controlled low dose of curcumin over a time period.
  • the phytochemical agent comprises about 2% to about 50% of the weight of the one or more polymers comprising the biocompatible polymeric matrix.
  • the phytochemical agents comprises about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 18%, about 19%, about 20%, about 21%, about 22%, about 25%, about 30%, about 35%, about 40%, about 45%, about 46%, about 47%, about 48%, about 49%, or about 50% of the weight of the one or more polymers.
  • the phytochemical agents are added to between about 2% to about 20% of the polymer weight.
  • the optimum amount of each phytochemical agent that is incorporated into a biocompatible polymeric matrix can vary depending on the particular phytochemical agents used and the desired dosage to be achieved. Determination and adjustment of the amount of a
  • phytochemical agent to be used in a particular composition or application as well as when and how to make such adjustments, can be ascertained using only routine experimentation.
  • Any phytochemical agent of the presently-disclosed subject matter can be provided in the form of a pharmaceutically acceptable salt or solvate.
  • a salt can be formed using a suitable acid and/or a suitable base.
  • Suitable acids that are capable of forming salts with the phytochemical agents of the presently disclosed subject matter include inorganic acids such as trifluoroacetic acid (TFA), hydrochloric acid (HQ), hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, phosphoric acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, fumaric acid, anthranilic acid, cinnamic acid, naphthalene sulfonic acid, sulfanilic acid, or the like.
  • inorganic acids such as trifluoroacetic acid (TFA), hydrochloric acid (HQ), hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, phosphoric acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, ox
  • Suitable bases capable of forming salts with the phytochemical agents of the presently disclosed subject matter include inorganic bases such as sodium hydroxide, ammonium hydroxide, potassium hydroxide and the like; and organic bases such as mono-, di- and tri-alkyl and aryl amines (e.g. triethylamine, diisopropyl amine, methyl amine, dimethyl amine, and the like), and optionally substituted ethanolamines (e.g. ethanolamine, diethanolamine, and the like).
  • inorganic bases such as sodium hydroxide, ammonium hydroxide, potassium hydroxide and the like
  • organic bases such as mono-, di- and tri-alkyl and aryl amines (e.g. triethylamine, diisopropyl amine, methyl amine, dimethyl amine, and the like), and optionally substituted ethanolamines (e.g. ethanolamine, diethanolamine, and the like).
  • solvate refers to a complex or aggregate formed by one or more molecules of a solute, e.g. a phytochemical agent or a pharmaceutically-acceptable salt thereof, and one or more molecules of a solvent.
  • a solute e.g. a phytochemical agent or a pharmaceutically-acceptable salt thereof
  • solvents include, but are not limited to, water, methanol, ethanol, isopropanol, acetic acid, and the like.
  • the solvent is water, the solvate formed is a hydrate.
  • pharmaceutically-acceptable salt or solvate thereof is intended to include all permutations of salts and solvates, such as a solvate of a pharmaceutically-acceptable salt of a
  • a composition that includes a biocompatible polymeric matrix incorporating an effective amount of a phytochemical agent where the composition further comprises a polymeric coating surrounding the initial composition.
  • the polymeric coating is comprised of polycaprolactone.
  • the polymeric coating is comprised of about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50 layers, about 75, about 100, about 125, about 150, about 175, or up to about 200 layers of the polymer around the initial composition.
  • the polymeric coating is comprised of about 5 to about 40 layers of the polymer around the initial composition.
  • providing a polymeric coating around the compositions of the presently-disclosed subject matter prevents or reduces an amount of phytochemical
  • U 024:UN106:923010:1 :LOUISVILLE - agent that is initially released from the compositions in a high concentration (i.e., a burst release) such that the polymeric coating provides a more sustained release and/or prevents toxicity that may otherwise occur as a result of the high initial release of the phytochemical agents from the compositions.
  • compositions themselves i.e., the biocompatible matrices incorporating the phytochemical agents
  • the compositions are provided in the form of a coating.
  • the compositions can be used to surround or otherwise envelop an additional composition, and thus, act as a coating while still providing a sustained release of the phytochemical agents.
  • composition further comprises an effective amount of a chemotherapeutic agent that is incorporated into and released from a biocompatible polymeric matrix that also incorporates an effective amount of a chemotherapeutic agent
  • chemotherapeutic agents include, but are not limited to, platinum coordination compounds such as cisplatin, carboplatin or oxalyplatin; taxane compounds, such as paclitaxel or docetaxel; topoisomerase I inhibitors such as camptothecin compounds for example irinotecan or topotecan; topoisomerase II inhibitors such as anti-tumor podophyllotoxin derivatives for example etoposide or teniposide; anti-tumor vinca alkaloids for example vinblastine, vincristine or vinorelbine; anti -tumor nucleoside derivatives for example 5- fluorouracil, gemcitabine or capecitabine; alkylating agents, such as nitrogen mustard or nitrosourea for example cyclophosphamide, chlorambucil, carmustine or lomustine; antitumor anthracycline derivatives for example da
  • composition further comprises an effective amount of a anti-inflammatory agent that is incorporated into and released from a biocompatible polymeric matrix that also incorporates an effective amount of a
  • U 024:UN106:923010:1 :LOUISVILLE - phytochemical agents or from a second biocompatible polymeric matrix examples include, but are not limited to, non-steroidal antiinflammatory agents ( SAIDS), such as aspirin, diclofenac, indomethacin, sulindac, ketoprofen, flurbiprofen, ibuprofen, naproxen, piroxicam, tenoxicam, tolmetin, ketorolac, oxaprosin, mefenamic acid, fenoprofen, nambumetone, acetaminophen, and combinations thereof; COX-2 inhibitors, such as nimesulide, flosulid, celecoxib, rofecoxib, parecoxib sodium, valdecoxib, etoricoxib, etodolac, meloxi
  • SAIDS non-steroidal antiinflammatory agents
  • COX-2 inhibitors such as nimesulide,
  • glucocorticoids such as hydrocortisone, cortisone, prednisone, prednisolone,
  • methylprednisolone meprednisone, triamcinolone, paramethasone, fluprednisolone, betamethasone, dexamethasone, fludrocortisone, desoxycorticosterone, rapamycin; or others or analogues of these agents or combinations thereof.
  • a method for treating a cancer comprises administering to a subject in need thereof an effective amount of a composition of the presently-disclosed subject matter comprising a biocompatible polymeric matrix incorporating a phytochemical agent, wherein the phytochemical agent is incorporated into and released over a time period from the biocompatible polymeric matrix at a controlled low dose of the phytochemical agent.
  • the terms “treating” or “treatment” relate to any treatment of a cancer including, but are not limited to, therapeutic treatment and prophylactic treatment of a cancer.
  • therapeutic treatment of a cancer the terms “treating” or “treatment” include, but are not limited to, inhibiting the progression of a cancer, arresting the development of a cancer, reducing the severity of a cancer, ameliorating or relieving one or more symptoms associated with a cancer, and causing a regression or a cancer or one or more symptoms associated with a cancer.
  • the terms “treating” or “treatment,” as used herein, further include the prophylactic treatment of a cancer including, but not limited to, any action that occurs before the development of a cancer. It is understood that the degree of prophylaxis need not be absolute (e.g. the complete prophylaxis of a cancer such that the subject does not develop a cancer at all), and that intermediate levels of prophylaxis, such as increasing the time required for at least one symptom resulting from a cancer to develop, reducing the severity or spread of a cancer in a subject, or reducing the time that at least one
  • U 024:UN106:923010:1 :LOUISVILLE - adverse health effect of a cancer is present within a subject, are all examples of prophylactic treatment of a cancer.
  • treating a cancer can include, but is not limited to, killing cancer cells, inhibiting the development of cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the available blood supply to a tumor or cancer cells, promoting an immune response against a tumor or cancer cells, reducing or inhibiting the initiation or progression of a cancer, increasing the lifespan of a subject with a cancer, or inhibiting or reducing the formation of DNA adducts by chemical carcinogens.
  • a method for treating a cancer comprising inhibiting or reducing the formation of DNA adducts.
  • DNA adducts i.e. carcinogens covalently bound to DNA
  • Many carcinogens are known to induce the formation of DNA adducts (Hemminki, 1995) and the presence of DNA adducts in humans has been strongly correlated with an increased risk for cancer development (Santella, 1997).
  • human studies have shown a higher accumulation of tissue DNA adducts in cigarette smokers than in non- smokers or individuals who have never smoked, indicating that DNA adduct formation is a viable target for the treatment of cancer.
  • cancer refers to all types of cancer, neoplasm, or malignant tumors found in subjects, including leukemias, carcinomas, and sarcomas.
  • cancers include, but are not limited to, cancer of the brain, bladder, breast, cervix, colon, head and neck, kidney, lung, non-small cell lung, melanoma, mesothelioma, ovary, prostate, sarcoma, stomach, uterus, skin, esophagus, pancreas, and medulloblastoma.
  • the cancer is selected from the group consisting of breast cancer, lung cancer, and cervical cancer.
  • breast cancer is meant to refer to any cancer of the breast and includes, but is not limited to, mammary carcinoma, adenocarcinoma, lobular (small cell) carcinoma, intraductal carcinoma, medullary breast cancer, mucinous breast cancer, tubular breast cancer, papillary breast cancer, Paget's disease, inflammatory breast cancer, and hormone- dependent and hormone -independent tumors of the breast.
  • cervical cancer is meant to refer to any cancer of the cervix and includes, but is not limited to, squamous cell carcinomas, adenocarcinomas, and adenosquamous carcinomas, or mixed carcinomas.
  • lung cancer is meant to refer to any cancer of the lung and includes non-small cell lung carcinomas and small cell lung carcinomas.
  • non-small cell lung carcinomas include, but are not limited to, squamous cell lung carcinomas, adenocarcinomas, bronchioalveolar carcinomas, adenosquamous carcinomas, papillary adenocarcinomas, mucoepidermoid carcinomas, adenoid cystic carcinomas, large cell carcinomas, and giant cell and spindle cell carcinomas.
  • the subject is at an increased risk of developing a primary cancer.
  • the primary cancer is selected from the group consisting of breast cancer, lung cancer, and cervical cancer.
  • the phrase "increased risk” is used herein to refer to those subjects whose likelihood of developing a cancer in their lifetime is increased, as compared to a normal subject. Such subjects may be identified by factors that include, but are not limited to: a genetic pre-disposition to certain cancers; life style choices such as tobacco smoking, tobacco chewing, or dietary habits; medical treatments such as immunosuppression; or exposure to carcinogenic agents in the environment, such as viruses, chemicals, medications, and radiation, or age-related factors, such as hormone levels.
  • subjects at an increased risk of developing breast cancer may be identified by factors such as age, gender, early age of menarche, late age of menopause, nulliparity, use of oral contraceptives, prolonged estrogen replacement therapy, breast density, or mutations in two known breast- cancer related genes, BRCA1 and BRCA2.
  • subjects at risk of developing cervical cancer may be identified by factors such as human papilloma virus (HPV) infection, life style choices such as tobacco smoking or dietary habits, human immunodeficiency virus (HIV) infection, use of oral contraceptives, multiple pregnancies, hormonal therapies, genetic predispositions to cervical cancer, and cervical dysplasia.
  • HPV human papilloma virus
  • HAV human immunodeficiency virus
  • the term "primary cancer” is meant to refer to an original tumor or cancer cell in a subject. Such primary cancers are usually named for the part of the body in which the primary cancer originates.
  • a “secondary cancer” is used herein to refer to a cancer which has spread, or metastasized, from an initial site (i.e. a primary cancer site) to another site in the body of a subject, a cancer which represents a residual primary cancer, or a cancer that has originated from treatment with an antineoplastic agent(s) or radiation or both.
  • the term “secondary cancer” is thus not limited to any one particular type of cancer, including the type of primary cancer from which it derived.
  • U 024:UN106:923010:1 :LOUISVILLE - preventing or treating a cancer is further provided where the subject is at risk of developing a secondary cancer.
  • the term "subject” includes both human and animal subjects.
  • veterinary therapeutic uses are provided in accordance with the presently disclosed subject matter.
  • the presently-disclosed subject matter provides for the treatment of mammals such as humans, as well as those mammals of importance due to being endangered, such as Siberian tigers; of economic importance, such as animals raised on farms for consumption by humans; and/or animals of social importance to humans, such as animals kept as pets or in zoos.
  • Examples of such animals include but are not limited to: carnivores such as cats and dogs; swine, including pigs, hogs, and wild boars; ruminants and/or ungulates such as cattle, oxen, sheep, giraffes, deer, goats, bison, and camels; and horses.
  • carnivores such as cats and dogs
  • swine including pigs, hogs, and wild boars
  • ruminants and/or ungulates such as cattle, oxen, sheep, giraffes, deer, goats, bison, and camels
  • horses are also provided.
  • domesticated fowl i.e., poultry, such as turkeys, chickens, ducks, geese, guinea fowl, and the like, as they are also of economic importance to humans.
  • livestock including, but not limited to, domesticated swine, ruminants, ungulates, horses (including
  • the phytochemical agent is administered to a subject by subcutaneous implantation of the biocompatible polymeric matrix incorporating the phytochemical agent.
  • the phytochemical agent is administered to a subject by implantation of a composition of the presently-disclosed subject matter at a site distant from the site of the
  • U 024:UN106:923010:1 :LOUISVILLE - cancer a composition of the presently-disclosed subject matter can be implanted subcutaneously in the arm of a subject such that the phytochemical agents, upon their release into systemic circulation, are capable of treating a breast, cervical, or lung cancer, which is located at a site distant from the site of the implantation of the presently- disclosed composition.
  • the phytochemical agent is administered to a subject by implantation of the biochemical polymeric matrix incorporating the phytochemical agent at a site of a cancer or at a site suspected of developing a cancer.
  • the biocompatible polymeric composition can be implanted at the site of a tumor, either following surgical removal or resection of the tumor, or can be implanted into the surrounding tissue.
  • the composition need not be implanted internally within the body of a subject, but can be implanted into an external orifice of the subject where a cancer is present.
  • a composition of the presently-disclosed subject matter can be implanted into the cervix of a subject by insertion of the biocompatible polymeric matrix incorporating the phytochemical agent into the cervical canal of a subject.
  • a biocompatible matrix incorporating a phytochemical agent can be administered to a site suspected of developing a cancer by the inserting the composition into the cervical canal of the subject such that an effective amount of the phytochemical agent is administered to the cervix of the subject.
  • the term "effective amount” is used herein to refer to an amount of the composition (e.g., a composition comprising a phytochemical agent that is incorporated into and released from a biocompatible polymeric composition) sufficient to produce a measurable biological response (e.g., inhibition of the development of tumor cells or a reduction in the number of tumor cells).
  • a measurable biological response e.g., inhibition of the development of tumor cells or a reduction in the number of tumor cells.
  • Actual dosage levels of active ingredients in a therapeutic composition of the presently-disclosed subject matter can be varied so as to administer an amount of the active compound(s) that is effective to achieve the desired therapeutic response for a particular subject and/or application.
  • the selected dosage level will depend upon a variety of factors including the activity of the therapeutic composition, formulation, the route of administration, combination with other drugs or treatments, severity of the condition being treated, and the physical condition and prior medical history of the subject being treated.
  • a minimal dose is administered, and dose is escalated in the absence of dose-limiting toxicity to a minimally effective amount. Determination and
  • a device 10 for insertion into a cervical canal includes: a substantially cylindrical shaft 20 having a proximal end 22 and a distal end 24, and defining a canal 26 that extends from the proximal end 22 to the distal end 24; and, a cap 30 attached to the proximal end 22 of the cylindrical shaft 20, said cap including a central opening 32 in registry with the canal defined by the shaft.
  • an exemplary device 10 can be inserted into the cervix of a subject to "treat" a cancer, as defined herein.
  • a device 10 for insertion into a cervical canal is provided wherein the cylindrical shaft 20 is suitably adapted for engaging the walls of a cervical canal and the bottom surface 34 of the cap 30 is suitably adapted for engaging an external orifice of the uterus.
  • the bottom surface 34 of the cap 30 has a concave shape such that the bottom surface 34 of the cap 30 suitably engages the external orifice of the uterus.
  • a device 10 for insertion into a cervical canal wherein the distal end 24 of the shaft 20 is tapered such that the device 10 is easily inserted into the cervix of a subject.
  • the canal 26 defined by the shaft 20 allows for the passage of bodily fluids from the uterus.
  • the device is designed and fabricated such that the device contacts a transformation zone of a cervix of a subject.
  • transformation zone refers to a region of a cervix that separates the ectocervical region, which is comprised of squamous cells, from the endocervical region, which is comprised of columnar cells.
  • the transformation zone is typically found on the outside of the external orifice of the uterus.
  • the transformation zone decreases in size and reaches the cervical canal.
  • a device for cervical insertion is provided that can be utilized to treat cervical cancer in female subjects of all ages by maintaining contact with the transformation zone.
  • an exemplary device for insertion into a cervical canal can be adapted for suitable insertion into the cervix of a human subject.
  • a device is provided wherein the cylindrical shaft is about 19 to about 25 mm in length and about 9 to about 11 mm in diameter, the cap is about 20 to 25 mm in diameter, and the canal is about 4 mm to about 5 mm in diameter.
  • an exemplary device of the presently-disclosed subject matter can be comprised of a biocompatible polymeric matrix incorporating an effective amount of a phytochemical agent, as described herein.
  • an exemplary device can be fabricated by extruding a dissolved mixture of one or more polymers and one or more phytochemical agents through a suitable mold and dried to provide a suitably shaped device. Further shaping of the device can additionally be performed during the drying process.
  • different shaped molds can be provided such that other devices can be fabricated or adapted to provide suitably shaped or sized devices for insertion or implantation at other anatomical locations.
  • the phytochemical agent is selected from curcumin, green tea polyphenols, punicalagin, diindolylmethane, oltipraz, tocotrienol, tocopherol, plumbagin, cyanidin, delphinidin, lycopene, lupeol, curcurbitacin-B, Withaferin A, indole-3-carbinol, genestein, equol, resveratrol, co-enzyme Q-10, ellagic acid, petunidin, malvidin, peonidin, fennel extract, and combinations thereof.
  • the phytochemical agent comprises about 2% to about 50% of the polymer weight.
  • a device for cervical insertion where the biocompatible polymeric matrix is comprised of polymers selected from polycaprolactone,
  • the biocompatible polymeric matrix comprises polycaprolactone in combination with a second polymer that, in certain embodiments, can be selected from cyclodextrin, F68, and polyethylene glycol. In some embodiments the polycaprolactone and the second polymer are combined in a ratio of about 4: 1 or about 9: 1. In some embodiments, the biocompatible matrix comprises polycaprolactone and F68 in a ratio of about 4: 1 and, in some
  • the phytochemical agent comprises curcumin.
  • the biocompatible polymeric matrix is biodegradable.
  • compositions were prepared by first dissolving two polymers in an appropriate solvent: e.g. a water-insoluble, biodegradable, and biocompatible
  • polycaprolactone (MW 65,000) and a water-soluble cyclodextrin.
  • Polycaprolactone and cyclodextrin ( in a 4: 1 or 9: 1 ratio) were dissolved in 2-5 volumes of dichloromethane (g/ml).
  • Some preparations comprising polycaprolactone as a first polymer included either F68 or polyethylene glycol instead of cyclodextrin.
  • Desired phytochemical agent(s) approximately 2% to 20% or more of the polymer weight, were then dissolved either in the same solvent as the polymer or another appropriate solvent (e.g., ethanol, acetone, tetrahydrofuran), depending upon the phytochemical agent's solubility.
  • phytochemicals incorporated into a biocompatible polymeric matrix ranged from less than 2 mg to as much as 20 mg of phytochemical agent per 100 mg of polymer. Depending on the particular phytochemical agent employed, however, it was possible to incorporate as much as 50 mg or more of the phytochemical agent per 100 mg of polymer.
  • compositions comprising a biocompatible polymeric matrix by itself (sham) or incorporating curcumin, ellagic acid, oltipraz, or green tea polyphenols (GTPs) are shown in Figure 1.
  • compositions which were approximately 1 cm in length and approximately 1 mm to 2.2 mm in diameter, comprising a biocompatible polymeric matrix incorporating a desired phytochemical agent (20% w/w) were placed in a release medium (5-20 ml phosphate-buffered-saline (pH 7.4) containing 10% bovine serum) at 37°C with constant agitation in a water bath to simulate an in vivo scenario.
  • the medium was replaced daily and the amount of phytochemical agents released from the compositions per day was measured spectrophotometrically against a standard curve and expressed as micrograms per day.
  • Figures 2 to 8 show the amount of exemplary phytochemical agents released per day and the total amount of phytochemical agents released from a biocompatible polymeric matrix of the presently-disclosed subject matter over a particular time period, including data for curcumin ( Figures 2A and 2B), GTPs ( Figures 3A and 3B),
  • U 024:UN106:923010:1 :LOUISVILLE - biocompatible polymeric matrix is dependent on the lipophilicity of the phytochemical agent, with a higher initial release for highly water-soluble compounds (e.g. green tea polyphenols) and a somewhat lower release for more lipophilic agents (e.g. curcumin).
  • highly water-soluble compounds e.g. green tea polyphenols
  • more lipophilic agents e.g. curcumin
  • Example 3 Effect of Phytochemical Agent Load on In Vitro Release of Phytochemicals from a Biocompatible Polymeric Matrix
  • a biocompatible polymeric matrix comprised of polycaprolactone, MW 65K and polycaprolactone, MW 15K combined in a 1 :4 ratio, and placed in a release medium (phosphate-buffered-saline (pH 7.4) containing 10% bovine serum) at 37°C with constant agitation in a water bath.
  • a release medium phosphate-buffered-saline (pH 7.4) containing 10% bovine serum
  • phytochemical agent can be effectively released from a biocompatible polymeric matrix in vivo and that this release is similar to the release kinetics observed for a phytochemical agent in vitro.
  • the total amount released at each time interval was determined by subtracting the unreleased amount of the phytochemical remaining in each matrix from the initial amount implanted into the rats, and this amount was divided by the initial amount implanted into the rats to determine the total percentage of the phytochemical released from the matrix at each time interval.
  • Figures 13 to 16 show the amount of exemplary phytochemical agents released per day and the total amount of phytochemical agents released from a biocompatible polymeric matrix of the presently-disclosed subject matter in vivo over an extended time period of 18 weeks, including data for the long-term release of curcumin (Figures 13A and 13B), GTPs ( Figures 14A and 14B), diindolylmethane (DIM, Figures 15A and 15B), and punicalagins ( Figures 16A and 16B). As shown in Figures 13 to 16, a greater amount of each phytochemical was initially released from the biocompatible polymeric matrix in the first few weeks following implantation.
  • the phytochemical agents were released at consistently lower levels indicating that a controlled low dose of the phytochemical agents can be sustainably released from a biocompatible polymeric matrix incorporating a phytochemical reagent following subcutaneous implantation of the compositions. This greater initial release of the phytochemical agent, followed by a sustained lower release, is consistent with the in vitro data depicted in Figures 2 to 5.
  • Intervention with phytochemical agents is a useful strategy to inhibit or reduce cancer development.
  • phytochemical agents must generally be administered in the diet at high doses in order to achieve effective plasma concentration and increased bioavailability.
  • U 024:UN106:923010:1 :LOUISVILLE - adducts by P-postlabeling 30 days post B[a]P treatment showed 65% and 83% inhibition of adducts in the lung and liver (9 ⁇ 5 and 4 ⁇ 0.7 adducts/10 9 nucleotides), respectively compared with sham treatment (26 ⁇ 5 and 23 ⁇ 12 adducts/10 9 nucleotides).
  • the average dose of curcumin over the duration of the study was approximately 1 10 ⁇ g/rat/day based on the amount left in the implants recovered from animals.
  • Example 7 Effect of Chemotherapeutic Compositions on Inhibition of Tissue DNA Adducts in Subjects Treated with a Sustained Low Dose of a Carcinogen
  • biocompatible polymeric matrices comprised of
  • polycaprolactone and incorporating benzo[a]pyrene were first prepared by dissolving both B[a]P and polycaprolactone in an appropriate solvent, melting the polymer following removal of the solvent, and extruding the molten material through a mold.
  • the B[a]P-PCL implant resulted initially in low adduct level in the lung (13 ⁇ 3) but the levels increased progressively after 15 days (23 ⁇ 12) and 30 days (26 ⁇ 5), indicating that animals were exposed to B[a]P continuously.
  • Adducts were also detected in the liver (1 1 ⁇ 2, 21 ⁇ 6 and 23 ⁇ 4, respectively) and mammary tissue (16 ⁇ 2, 18 ⁇ 5, 23 ⁇ 5, respectively) and increased steadily with time. Measurement of the residual B[a]P in implants recovered from the animals showed that ⁇ 1 mg of total B[a]P was released over 30 days. This release was consistent with an average daily release of 26 ⁇ 9 ⁇ g B[a]P when implants were stirred at 37 °C in phosphate-buffered-saline, pH 7.4 containing 10% serum.
  • the sustained, low-doses of B[a]P resulted in a similar degree of DNA adduct accumulation in all of the three tissues analyzed as compared with the bolus-dose, which resulted in higher adduct accumulation in the liver.
  • This data shows that the polymer-based delivery system is an effective means to study sustained, low-dose carcinogenesis.
  • compositions Following the establishment of the presently-disclosed compositions as an effective means to study sustained, low-dose carcinogenesis, the ability of the presently- disclosed compositions to inhibit tissue DNA adducts in subjects treated with sustained low carcinogen doses was determined by examining the effect of curcumin administered via a biocompatible polymeric matrix on the inhibition of tissue DNA adduct formation induced by a sustained low dose of B[a]P. Briefly, one group of Sprague-Dawley rats received a 1 cm biocompatible polymeric matrix incorporating 20% w/w of curcumin, while a second group of rats received a sham biocompatible polymeric matrix that did not incorporate an agent. Both groups were maintained on a control diet.
  • Cytochrome P450 proteins are a diverse family of hemoproteins and are widely regarded as an important enzyme in the metabolism and formation of carcinogens.
  • liver tissue was collected from Sprague-Dawley rats that subcutaneously received a 1 cm biocompatible polymeric matrix incorporating B[a]P (10% carcinogen load) and either a 1 cm biocompatible polymeric matrix incorporating 10% w/w of GTPs or a sham biocompatible polymeric matrix that did not incorporate a phytochemical agent, as described in Example 7.
  • silastic implants used in the experiments described below were fabricated by filling silastic tubing with a desired agent.
  • the silastic implants are not biodegradable like compositions disclosed herein comprising biocompatible polymeric matrices, the silastic implants provide proof of concept that a reduced dose of phytochemical agents administered systemically can be effective in inhibiting mammary carcinogenesis.
  • ellagic acid was administered either in the diet
  • the total amount of ellagic acid administered during the entire duration of the study by diet was 800 mg/rat while the subcutaneous implantation of the biocompatible silastic implant incorporating ellagic acid delivered only 1.48 ⁇ 0.87 mg per implant, as determined by HPLC measurement of the material left in implants recovered after termination.
  • an approximately 135-fold reduction of the ellagic acid dose produced similar biological responses when delivered by the slow release system. This approach thus opens new avenues for phytochemical agents whose use has been discontinued for therapeutic purposes, either due to lack of bioavailability or toxicity due to high doses.
  • Example 10 Inhibition of Mammary Cell Carcinogenesis by Combined Administration of Phytochemical Agents
  • Groups A-C 8 animals/group.
  • Group A subcutaneously received sham implants e.g., biocompatible silastic implants without a phytochemical agent
  • groups B and C received implants of 3 cm biocompatible silastic implants incorporating various phytochemical agents.
  • Group B received one implant each of oltipraz and curcumin
  • Group C received one implant each of oltipraz and curcumin
  • U 024:UN106:923010:1 :LOUISVILLE - of curcumin, ellagic acid, co-enzyme-QlO, and lycopene Two weeks later, all groups received implants of 17p-estradiol implants comprised of a biocompatible silastic implant incorporating the 17p-estradiol. Animals received standard rodent chow diet and water ad libitum. Six months later, animals were euthanized and mammary tumors were counted and their sizes were measured with a caliper.
  • Example 1 1 - Inhibition of Mammary Cell Proliferation by Combined Administration of Phytochemical Agents
  • Increased cellular proliferation is an integral part of the cancer phenotype and agents that have the potential to slow the growth of these phenotypes are often considered prophylactic (Dorai T & Aggarwal BB, 2004).
  • human lung cancer cells H1299 were incubated with vehicle only, with phytochemical agents individually, or with a combination of phytochemical agents. Following incubation for 1-3 days, the cells were incubated with 3- (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT).
  • the resultant formazan product was solubilized in dimethyl sulfoxide and the absorbance was measured at 540 nm to determine the extent of cellular proliferation.
  • incubation of the H1299 cells with a mixture of delphinidin, petunidin, malvidin, peonidin, and cyanidin inhibited the growth of HI 299 cells at a much lower concentration as compared to phytochemical agents administered individually.
  • Example 13 Formulation and Use of Device for Cervical Insertion
  • a device for cervical insertion (Figure 30) was made of biodegradable polymers (polycaprolactone: F68 in a 4: 1 ratio) incorporating a desired phytochemical agent (see, e.g., Figures 32A-32D showing perspective view of an exemplary device for cervical insertion comprising a biocompatible polymeric matrix incorporating curcumin).
  • the device was capable of incorporating a phytochemical agent load of 0.5% to as much as 20% or more and was prepared by extruding a molten polymer formulation through a plastic mold made of a polypropylene transfer pipet, a round spoon and a 10-ml plastic syringe. The cut-piece of the pipet mold was narrow on one end and broad on the other end.
  • the typical device weighed approximately 2.5 to 4 g, depending upon the size of the mold.
  • the device for cervical insertion is capable of delivering phytochemical agents to uterine cervix locally.
  • the device can be inserted into a cervix such that the shaft of the device engages the walls of the cervical canal and the bottom surface of the cap engages the external orifice of the uterus.
  • the device is capable of delivering an effective amount of a phytochemical agent locally to a cervix or the device can also be used to deliver phytochemical agents via systemic delivery to other organs like the uterus and ovary or to an organ more distant from the site of insertion such as the lungs.
  • An exemplary device for cervical insertion is relatively non-invasive and can include a tapered distal end to allow for easy insertion into a cervix as well as a canal through the device to allow for the passage of normal bodily fluids from the uterus.
  • an exemplary device for cervical insertion can be fabricated such that the cylindrical shaft is about 19 to about 25 mm in length and about 9 to about 11 mm in
  • the cap is about 20 to about 25 mm in diameter
  • the canal is about 4 mm to about 5 mm in diameter.
  • a cervical insertion device comprising a biocompatible polymeric matrix incorporating curcumin was placed in a release medium (phosphate-buffered-saline (pH 7.4) containing 10% bovine serum) at 37°C with constant agitation in a water bath to simulate an in vivo scenario.
  • the medium was replaced daily and the amount of curcumin released from the compositions per day was measured spectrophotometrically against a standard curve and expressed as micrograms per day.
  • Figures 34A and 34B show the amount of curcumin released per day and the total amount of curcumin released from an exemplary device for cervical insertion over a release period of 18 days.
  • This data which is similar to the data described herein for the in vitro and in vivo release of phytochemical agents, shows that over the initial five days following the placement of the device into the release medium a greater amount of curcumin is released from the device. However, after approximately 5 days in the release medium, the amount of curcumin being released decreased and approached lower steady-state levels indicating that an exemplary device for cervical insertion can effectively be used to release a controlled low dose of a phytochemical agent over a time period.
  • U 024:UN106:923010:1 :LOUISVILLE - nude mice are injected with 1-10 million tumor cells (lung, breast or cervical tumor cells) subcutaneously on the dorsal flank in 50-100 ⁇ of PBS or growth media according to established protocols (Morton CL & Houghton PJ, 2007).
  • the mice also receive a 2 cm biocompatible polymeric matrix incorporating 2-20% (w/w) of a phytochemical agent as a subcutaneous implant. Tumors start appearing at the site of implantation after approximately 2-4 weeks. The tumor growth is then monitored twice a week and is measured using a digital caliper.
  • the tumor tissue is isolated for examination of biochemical and molecular parameters. Different phytochemical agents are examined for their antitumor efficacy and the efficacy of combinations of different phytochemical agents are also examined by administering the combinations as a single implant or in multiple implants. Analysis of tumor tissue also reveals the mechanism by which the phytochemical agents are able to exert their effects.
  • mice receiving biocompatible polymeric implants incorporating an effective amount of a phytochemical agent As compared with animals receiving sham implants, a reduction in tumor volume is observed in mice receiving biocompatible polymeric implants incorporating an effective amount of a phytochemical agent, indicating that a composition and method that includes or makes use of a biocompatible polymeric matrix incorporating an effective amount of a phytochemical agent is useful for treating breast, lung, and cervical cancer.
  • Phytochemical agents are often used in adjuvant therapy with conventional chemotherapeutic drugs to enhance their therapeutic efficacy and are recommended in adjuvant therapy with selective Cox-2 inhibitors (like celecoxib) for adenomatous polyposis.
  • Selective Cox-2 inhibitors often leave other critical carcinogenic pathways unaltered, however, and phytochemicals can augment their activity by inhibiting such alternative pathways involved in carcinogenesis.
  • Previous studies have found that silibinin sensitizes the hormone refractory DU145 prostate carcinoma cells to cisplatin and carboplatin-induced cell growth inhibition and apoptotic death, and can be combined to enhance the therapeutic efficacy of these compounds.
  • certain chemotherapeutics like paclitaxel induce expression of antiapoptotic (XIAP, IAP-1, IAP-2, Bcl-2, and Bcl-xL), proliferative
  • cyclooxygenase 2, c-Myc, and cyclin Dl cyclooxygenase 2, c-Myc, and cyclin Dl
  • metastatic proteins vascular endothelial growth factor, matrix metalloproteinase-9, and intercellular adhesion molecule- 1 which can
  • U 024:UN106:923010:1 :LOUISVILLE - be inhibited by addition of phytochemical agents like curcumin. Additionally, certain agents like linalool moderately inhibit cell proliferation, but are known to exert synergistic effects with chemotherapeutics like doxorubicin by potentiating the cytotoxicity and proapoptotic effects of doxorubicin. Previous studies have shown that linalool increases doxorubicin accumulation and decrease Bcl-xL levels which results in its higher cytotoxicity for malignant cells. Phytochemical agents like licochalcone and licorice were also found to inhibit cisplatin induced hepatotoxicity and nephrotoxicity.
  • biocompatible polymeric matrix can effectively be used in adjuvant therapy to administer a phytochemical agent with a chemotherapeutic agent and/or an anti-inflammatory agent, either in the same implant or in separate implants, and to mitigate the toxicity, increase the efficacy, or both of
  • nude mice are injected with 1-10 million tumor cells (lung, breast or cervical tumor cells) subcutaneously on the dorsal flank in 50-100 ⁇ of PBS or growth media according to established protocols (Morton CL & Houghton PJ, 2007). When the tumors start appearing at the site of injection
  • mice then receive a 2 cm biocompatible polymeric matrix incorporating 2-20% (w/w) of a phytochemical agent and a chemotherapeutic agent and/or anti-inflammatory agents as subcutaneous implant(s).
  • the biocompatible polymeric matrix incorporating the chemotherapeutic agent and/or anti-inflammatory agents is prepared by the same method that is used to prepare the biocompatible polymeric matrices incorporating an effective amount of a phytochemical agent, which is described herein above in Example 1.
  • the tumor growth is monitored twice a week and is measured using a digital caliper.
  • the tumor is allowed to reach approximately 1.5 cm in diameter in the control group. At this point, all the animals are euthanized and the tumor tissue is isolated for the examination of biochemical and molecular parameters.
  • U 024:UN106:923010:1 :LOUISVILLE - anti-inflammatory agent is also incorporated into a biocompatible polymeric matrix, and is administered in conjunction with a biocompatible polymeric matrix incorporating a phytochemical agent and a chemotherapeutic agent, a reduction in the local inflammatory reaction to the chemotherapeutic agent is observed, indicating that the inclusion of an antiinflammatory agent is useful for overcoming any local inflammatory reaction to the chemotherapeutic agent.
  • mice To determine the effect of a biocompatible polymeric matrix incorporating a phytochemical agent on inhibiting chemically-induced lung tumors, female A/J mice (4-5 weeks old) receive either a sham implant or a biocompatible polymeric matrix implant incorporating 2-20% (w/w) of one or more phytochemical agents. Two weeks after receiving the implants, the mice receive 11.65 mg/kg body weight of 4-(methylnitrosamino)-l-(3- pyridyl)- 1 -butanone ( K) by gavage three times a week for 10 weeks. Six mice are then euthanatized at 5-weeks after the first carcinogen treatment and 1-week after the last carcinogen treatment (1 1 weeks) for intermediate biomarker analysis.
  • mice Six weeks after the last carcinogen treatment (16 weeks), all mice are euthanatized, lung tissue is removed, surface adenomas are counted, and tissue is serially sectioned to count internal tumors. Part of the lung is also snap frozen for biomarker analysis. A reduction in the tumor multiplicity, both on the surface of the lung and internally, and a reduction in the conversion of adenomas to carcinomas is observed in the group receiving a biocompatible polymeric matrix
  • compositions are useful for treating chemically-induced lung tumors.
  • U 024:UN106:923010:1 :LOUISVILLE - classified as an adenocarcinoma starts appearing around 30 days post-carcinogen administration and at the end of 35 days post MNU administration, the cumulative incidence of palpable carcinomas is around 60-90%.
  • a second group of rats receives an estradiol implant (1.2 cm silastic implant containing 9 mg) that is implanted subcutaneously on the back of ACI rats at 8-9 weeks of age.
  • the first palpable tumor appears around 3-4 months and nearly 100% of the rats develop palpable mammary tumors by 8 months.
  • one or more phytochemical agents are delivered via a biocompatible polymeric matrix two weeks before the chemical carcinogen treatment.
  • the rats are monitored weekly for palpable tumors.
  • postponement of the appearance of the first tumor by approximately 3 to 5 weeks, as well as a reduction in tumor multiplicity and burden by 50-80 percent is observed in rats receiving a biocompatible polymeric matrix incorporating one or more phytochemical agents, indicating that a composition and method that includes or makes use of a
  • biocompatible polymeric matrix incorporating an effective amount of a phytochemical agent is useful for inhibiting the growth of chemically -induced breast tumors.
  • compositions of the presently-disclosed subject matter which incorporate an effective amount of a phytochemical agent into a biocompatible polymeric matrix
  • studies where undertaken to assess tissue levels of curcumin achieved via the polymeric matrices as compared with the delivery of cucrumin via the traditional dietary route.
  • the biochemical efficacy of curcumin to alter expression and/or activity of various hepatic xenobiotic -metabolizing enzymes was also determined and compared with the different delivery routes.
  • PCL-121 medical grade poly ( ⁇ -caprolactone) 121,000 molecular weight (PCL-121) was purchased from SurModics Pharmaceuticals (Birmingham, AL), dichloromethane, acetonitrile, anhydrous citric acid and phosphate-buffered-saline (PBS) were from Sigma- Aldrich (St.
  • the curcumin implants i.e., the biocompatible polymeric matrices incorporating curcumin
  • the curcumin implants were then prepared by first dissolving curcumin (20% w/w) in ethanol, and the polymers (PCL-121K and PEG-8K in 65:35 ratios) in dichloromethane. Both solutions were then mixed together to prepare a homogenous solution of drug and polymers. The solvents were evaporated on a water bath maintained at 65 °C followed by overnight drying under vacuum at 65 °C to remove residual solvents to prepare an amorphous molecular dispersion of drug in polymer.
  • the dried molten polymer was then extruded through silastic tubing mold (internal diameter 3.4 mm) attached to a syringe. After a few minutes, the cylindrical implants were removed from the tubing and excised into desired lengths (2.0 cm, 200 mg containing 40 mg drug) and stored at -20 °C under argon until used.
  • Average daily release (residual amount at T n - Residual amount at Tn_i)/ T n -T n _i (Days) where Ti, T 2 , T 3 ,.. .Tn are different time intervals.
  • Plasma pooled from all the animals was mixed with 200 ⁇ of 0.5 M sodium acetate to reduce the pH to 5. Plasma was then extracted three times with 3 ml ethyl acetate. Ethyl acetate extracts were pooled and dried under vacuum. The dried residue was reconstituted in 100 ⁇ of acetonitrile, and one-half of the solution was analyzed by HPLC. Similarly, liver or brain tissue (approximately 500 mg) from each animal was homogenized in 3 ml PBS (pH 7.4) containing 200 ⁇ sodium acetate (0.5 M). The homogenate was then extracted twice with two volumes of ethyl acetate.
  • the three curcuminoids were separated by using acetonitrile and 1% citric acid (adjusted to pH 2.5) at a flow rate of 1 ml/ min with a gradient elution in which acetonitrile concentration was increased from 0% to 30% in first 5 min, followed by an increase to 45% in next 5-20 min. Acetonitrile was then maintained at this ratio till 36 min. Curcuminoids were detected using 410 and 500 nm as excitation and emission maxima, respectively, in the fluorescence detector.
  • liver samples from the animals were homogenized in 0.25 M sucrose buffer (pH 7.4) at 3000 rpm with Polytron. The homogenate was centrifuged at 3000 g for 20 min at 4 °C to
  • Microsomal proteins were subsequently quantified using a bicinchoninic acid (BCA) method and a BCATM Protein Assay kit (Thermo Scientific, Rockford, IL) and were resolved by sodium dodecyl sulfatepolyacrylamide gel electrophoresis (SDS-PAGE) on a 10% gel. Protein bands were then transferred to poly vinyli dene diflouride (PVDF) membrane, which was incubated with 5% non-fat dried milk in Tris-buffered-saline for 1 h at room temperature (25 °C) to block the non-specific binding sites.
  • BCA bicinchoninic acid
  • BCATM Protein Assay kit Thermo Scientific, Rockford, IL
  • SDS-PAGE sodium dodecyl sulfatepolyacrylamide gel electrophoresis
  • the membrane was then incubated with primary antibodies for CYP1A1 and GSTM, followed by horseradish peroxidase-conjugated secondary antibody.
  • the bands were detected by enhanced chemiluminescence using Pierce ® ECL Western-Blotting Substrate (Thermo Scientific, Rockford, IL) and quantified using VersaDoc Imaging System (BioRad Laboratories, Hercules, CA).
  • curcumin During in vivo release, extracellular fluid from the site of implantation enters into the polymeric matrix, dissolves the drug and diffuses out into the surrounding tissue. Curcumin from surrounding tissues then enters into the systemic circulation and is distributed to different tissues. Therefore, tissue concentrations of curcumin delivered via both diet and via implants was further measured in plasma, liver and brain. As noted above, curcumin was
  • U 024:UN106:923010:1 :LOUISVILLE - extracted from the tissues using solvent extraction and analyzed using HPLC coupled with a fluorescence detector with an extraction efficiency of approximately 90% from plasma and approximately 70% from tissues (liver and brain). Fluorescence detector was used due to very high fluorescence of curcumin, which not only increased the specificity of curcumin detection but also increased the sensitivity by at least 4- to 5 -fold. The lower limit of detection in plasma was found to be 125 pg (340 pM), and limit of quantification was approximately 200 pg (540 pM) by this method.
  • Curcumin diet on the other hand showed different kinetics in that curcumin was detected only on day 4 at a level of 0.3 ng/ml (815 pM) decreasing to 0.2 ng/ml after 12 days (543 pM) and was undetectable on 25 and 90 days. It was counterintuitive that instead of the fact that curcumin diet consumption was constant at all the time periods, curcumin was not observed on day 1 and a decline in plasma curcumin concentrations was observed after day 4, suggesting time- and exposure-dependent absorption kinetics. Furthermore, curcumin due to its lipophilic nature exhibits biphasic elimination kinetics with rapid equilibration and distribution of plasma curcumin into various tissues.
  • curcumin due to high tissue distribution of curcumin, initially almost all curcumin equilibrated into tissues and was not observed in plasma on day 1. However, with time, as the lipophilic tissues became saturated, curcumin was observed in plasma after 4 days on dietary administration.
  • curcumin possesses poor oral bioavailability owing to its low aqueous solubility and rapid metabolism both in intestine and in liver. Therefore, to further understand the kinetics of curcumin delivered via both the routes, its concentration in the liver was also measured (Figure 38). Curcumin delivered via both routes was found to be at similar levels in liver and was 25-30 ng on day 1. The levels increased slightly, though insignificant, on day 4 after which the levels declined to 10-15 ng after 12 days. This trend was found to exactly mimic the plasma concentrations suggestive of hepatic regulation of plasma curcumin levels at least via dietary route. Curcumin is known to induce the
  • curcumin concentration was found to be much higher (10- to 20-fold) in the liver as compared to plasma via both routes, again suggestive of its high tissue distribution.
  • High curcumin concentrations in hepatic tissues via dietary administration were expected as most of the orally administered curcumin gets absorbed into the liver, where it undergoes rapid metabolism.
  • similar behavior from the implant route was unexpected as it bypasses the hepatic first-pass metabolism and almost all of the administered curcumin directly reaches into the systemic circulation.
  • curcumin concentration in the whole brain tissue was further analyzed as the brain is also one of the highly perfused organs. Brain tissue was also selected because of curcumin' s known potent activities against Alzheimer's disease and against brain gliomas, where continuous localized/systemic delivery of this compound could make a significant improvement in the life of such patients.
  • curcumin delivery to the brain via implant route showed biphasic kinetics similar to liver. Since drug concentration that reaches a tissue is determined by rate and extent of its perfusion by systemic circulation, the brain showed rather high curcumin levels (30 ⁇ 7.3 ng/g tissue), almost equal to liver concentration on day 1 of implantation. This concentration dropped slowly to around 7.26 ⁇ 2.45 ng/g tissue after 12 days and stayed almost at the same levels even after 90 days. It was noted that the brain concentration was slightly less than the liver concentration from 12 to 90 days (a constant drug concentration period), which may be attributed, physiologically, to the liver receiving approximately 23% of cardiac output and brain receiving slightly less than liver ( approximately 18%). This finding again showed that tissue distribution from the implant route was perfusion rate limited and was dictated by cardiac output to that particular organ. It may also be noted that curcumin concentration in the brain tissue was approximately 3.5- fold higher by the implant route.
  • curcumin' s effective concentration is 5-20 ⁇ depending upon the cell line used. Since the plasma and liver concentrations observed in the foregoing in vivo study (78-95 nM) were much below these, it was also determined whether those concentrations were effective to modulate hepatic cytochromes as studies have shown that curcumin is a natural agonist of aryl hydrocarbon receptor (AhR)/pregnane xenobiotic receptor (PXR) pathway and induces the expression of phase I enzymes like CYP1A1
  • curcumin also interacts with the keapl protein to lower its affinity for nuclear factor (erythroid-derived 2)-like 2 (rf2) protein followed by Nrf2
  • phase II enzymes like glutathione-S-transferase ⁇
  • GSTM glutathione-S-transferase ⁇
  • curcumin implants were found to induce expression of CYPlAl by approximately 2-fold after 4, 12, 25 and 90 days of treatment. Although a 2- fold increase in CYPlAl was observed on day 1 also, yet the effect was masked by PEG- 8000 as a similar effect was also observed in sham implants (blank implants prepared with only PCL-121 and PEG- 8 K without any drug) at this time point and this effect from the sham implants was absent at all other time points. This effect from sham implants was presumably due to release of significant amounts of PEG-8000 on the first day
  • Curcumin diet increased the CYPlAl levels only slightly on day 1 (1.43-fold), which returned to basal levels after 4 days, and in fact, the levels were downregulated after 25 and 90 days of treatment. Since the hepatic curcumin levels were similar by both the routes tested, such route-dependent differences in induction of CYP 1A1 expression were counterintuitive. It is known that various chemopreventives like indole-3-carbinol (13C) are the most effective in inducing CYPlAl when given orally as compared to when administered systemically.
  • indole-3-carbinol like diindolylmethane (DIM) formed under stomach acidic conditions are more effective AhR ligands as compared to the parent compound, thus resulting in higher efficacy of indole-3-carbinol.
  • Curcumin is also known to degrade under slightly basic conditions of intestine forming various active degradation products like ferulic acid and vanillin, and it was possible that one or more of those metabolites might have AhR antagonistic activity resulting in blunting of curcumin's CYPlAl upregulating activity at initial time points and eventual downregulation after 25 and 90 days of treatment.
  • GSTM was not found to get modulated via either of the routes.
  • curcumin implants were also found to increase the activity from day 1 to day 12 of treatment as compared to untreated animals; however, this increase became significant only after 25 days when compared with sham implants. This enhanced activity remained consistently upregualated even after 3 months of treatment (in contrast to sham implants) and did not reduce to basal levels after 25 days.
  • curcumin diet showed slightly higher activity on day 1 of treatment, which was significant higher after 4 days as compared to untreated animals and again reduced to basal levels at 12 days of treatment. It thus appeared that continuous delivery of curcumin directly into the systemic circulation by the implant route was more effective in increasing and maintaining the higher CYP3A4 activity as compared to dietary route.
  • curcumin delivered directly into the systemic circulation was found to be more efficacious in inducing CYP1A1 and CYP3A4 enzymes required for its chemopreventive activity against various
  • curcumin has shown significant potential in various in situ and cell culture studies.
  • Example 20 Analysis of Coated Implant Formulations
  • the extruded implant formulation described herein above involved a heating step (70°C) to evaporate the solvent(s) as well as for melting of the polymer matrix containing the desired agent for extruding it into cylindrical implants.
  • the technology was not easily adapted for heat-labile compounds.
  • a further polymeric matrices formulation was developed in which blank PCL implants (about 1.2 mm diameter), prepared by the extrusion method, were coated with 20-40 layers of 5-15% solution of PCL in dichloromethane containing a desired amount of a particular agent (e.g., a phytochemical agent) dissolved either in dichloromethane or another appropriate solvent which was miscible with dichloromethane.
  • ⁇ -polycaprolactone (mol. wt. 80,000; P-80) was purchased from Sigma-Aldrich (St. Louis, MO, USA); Pluronic ® F68 (F68) was used from BASF Corp. (Florham Park, NJ, USA); silastic tubing (1.4 mm internal dia) was purchased from Allied Biomedical (Ventura, CA, USA); disposable syringes (5 ml) were purchased from BD
  • U 024:UN106:923010:1 :LOUISVILLE - Biosciences (Franklin lakes, NJ, USA); penicillin-streptomycin solution was purchased from Invitrogen (Carlsbad, CA); amber vials (20 ml) were purchased from National Scientific (Rockwood, TN, USA); curcumin and the three curcuminoids, curcuminoid I, curcuminoid II (demethoxycurcumin) and curcuminoid III (bisdemethoxycurcumin) were generous gifts of Sabinsa Corp.
  • blank P-80 implants (1.4 mm dia) were then prepared by dissolving P-80:F68 (9: 1) in dichloromethane, followed by evaporation of the solvent in a Petri dish under hood and extrusion of the molten polymer through a silastic tubing mold, similar to the methodologies described herein above.
  • Those blank implants were coated with about 20-30 layers of 10% P-80 solution in dichloromethane containing 1.0% oltipraz, curcumin, or individual curcuminoids I, II and III, except for withaferin A which was present at 0.3 and 0.5%.
  • one end of the blank implant (2.5 - 3.5 cm) was inserted into a silastic tube plug (6 mm long, 1.4 mm dia) while the other end of the plug was attached with a pipet tip. Coatings were then added by dipping the blank implant into the solution with intermittent drying with cool air using a commercial hair dryer and placing them under hood. Twenty to 30 layers (i.e., dippings) generally resulted in 2.6 mm diameter coated implants as measured by a digital caliper. Implants formulated in this manner had a 10% drug load of the test agents, except for withaferin A which represented 3% and 5% load. The assembly was placed under hood overnight to remove the residual DCM, implants were excised in 1 or 2 cm lengths and stored in amber vials under argon at -20°C until used.
  • DBP dibenzo[a,/]pyrene
  • Polymeric implants (1-cm, 1.4 mm dia, 5% DBP load) at the same time. Implants were grafted subcutaneously, and animal were euthanized after 3 weeks. Two additional groups of mice received only withaferin A implants to determine the rate of release in vivo and those animals were euthanized after 2 and 8 weeks. In both cases, animals were euthanized by CO 2 asphyxiation and selected tissues (lung, liver and brain) were collected and stored at -80°C until processing. Implants were also recovered, dried under vacuum and stored in amber vials under argon at 4°C until analysis.
  • DBP dibenzo[a,/]pyrene
  • tissue levels were measured by solvent extraction, where the eluates were analyzed by ultra performance liquid chromatography (UPLC). Briefly, liver and brain tissues (500 and 400 mg, respectively) from each animal were homogenized in 3 ml PBS (pH 7.4) containing 200 ⁇ of 0.5 M sodium acetate, and extracted with ethyl acetate. After evaporation of the pooled extracts, the residue was reconstituted in 100 ⁇ acetonitrile and analyzed by UPLC using Shimadzu UPLC system (Kyoto, Japan), with a Shim-Pack XR-ODS II reverse phase column
  • demethoxycurcumin and bisdemethoxycurcumin were separated by using acetonitrile and 1% citric acid (pH 2.5) at a flow rate of 0.75 ml/min with a linear gradient elution in which acetonitrile concentration was increased from 2 to 30% in first 2 min, followed by an increase to 45% from 2 to 6.4 min; the latter ratio was then maintained till 11.5 min and finally decreased to 2% in 13 min.
  • the curcuminoids were detected using 410 and 500 nm as excitation and emission maxima, respectively, in the fluorescence detector and total curcumin concentration was calculated from standard curves of individual
  • biocompatible polymeric matrices incorporating amounts of curcumin, green tea polyphenols and many other chemopreventive phytochemical agents, when grafted subcutaneously in rodents, provide controlled systemic delivery for long durations.
  • That polymeric implant formulation (the "extrusion” method), however, was not readily adaptable to some heat-labile compounds.
  • the implants prepared by those method generally accompanied an initial burst release of the compound. From the foregoing described experiments, however, it has been found that the coated implant formulations circumvent the limitations of the implants formulated by the extrusion method.
  • the "coated" polymeric implant formulation was developed using structurally diverse compounds, namely, oltipraz, curcumin and its three constituents, curcuminoids I, II and III, and withaferin A.
  • Oltipraz an analogue of thiol-3- thione, a constituent of cruciferous vegetables, has been reported as an effective
  • U 024:UN106:923010:1 :LOUISVILLE - Withaferin A a triterpenoid, represents a the principal bioactive of the herb Withania somenifera, commonly known as "ashwagandha", as part of the 'ayurvedic' folklore medicine in India.
  • the coated implants were formulated by coating multiple layers of the polymer (P80) solution in dichloromethane, with or without test agents, with intermittent drying with cool air on blank P80 implants (1.4 mm dia). Twenty or 30 coatings of 20% and 10% polymer solution, respectively resulted in cylindrical implants with about 2.6 mm diameter. The number of coatings depended upon the polymer solution
  • coated implants were prepared from individual constituents of curcumin, i.e., curcuminoid I, curcuminoid II (demethoxycurcumin) and curcuminoid III (bisdemethoxycurcumin). While no noticeable differences were found during the formulation of the coated implants, the three curcuminoids were released at different rates, with the cumulative release over 3 weeks being in the following descending order: curcuminoid I (24.8%), curcuminoid II (41.8%) and
  • curcuminoid III (51.4%) ( Figures 44A, 44B, and 44C, respectively).
  • DNA adducts The identity of DNA adducts has previously been established as guanine and adenine derivatives of the electrophilic metabolites, DBP-l l,12-diol-13,14-epoxides. No qualitative differences were observed in the adduct profiles in the absence or presence of test chemopreventives.
  • curcumin As described herein above, it is known that when curcumin is administered orally, it exhibits poor bioavailability and rapid metabolism both in the intestine as well as in the liver. To measure tissue the distribution of curcuminoids delivered by the implant route,
  • U 024:UN106:923010:1 :LOUISVILLE - curcuminoids were extracted from the lung, liver and brain tissues by solvent extraction and UPLC-UV. All the curcuminoids were readily measurable even after 21 days of grafting the implants. When delivered individually, the three curcuminoids I, II and III were found to be present in essentially the same ratio. In the liver, curcuminoid I and III levels were similar (5- 6 ng/g tissue); however, curcuminoid II was present in 4-5 times lower levels (Figure 47 A and 47B), suggesting this analog underwent more rapid metabolism.
  • the liver levels of the three curcuminoids I, II and III were found to be present essentially the same ratio as present in the original mixture and the total levels were significantly higher (15 ng/g tissue) than when given individually.
  • the coated polymeric implant formulations allowed embedding any compound in the polymeric matrix as long the compound was soluble in dichloromethane or any other solvent (e.g.,
  • the polymeric implant formulation accommodated heat-labile compounds, circumvented the problem of oral bioavailability, delivered continuous sustained low doses for long durations (e.g., months), and allowed in vivo efficacy assessment of minor plant constituents and synthetic metabolites which otherwise remains uninvestigated due to their insufficient quantities available.
  • Resveratrol Role of Nuclear Factor-kB, Cyclooxygenase 2, and Matrix
  • Estrogen receptors alpha and beta have similar activities in multiple endothelial cell pathways. Endocrinology. 143:3785-95.
  • Licochalcone A inhibits the growth of colon carcinomas and attenuates cisplatin- induced toxicity without a loss of chemotherapeutic efficacy in mice.
  • BMJ 321 624-628.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Dermatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Inorganic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

L'invention concerne des compositions qui comprennent une matrice polymère biocompatible comprenant une quantité efficace d'un agent phytochimique, une combinaison d'agents phytochimiques, ou un agent phytochimique et un ou plusieurs agents thérapeutiques supplémentaires. Des procédés de traitement d'un cancer utilisant les compositions sont également décrits.
PCT/US2013/033877 2012-03-26 2013-03-26 Procédés et compositions pour l'administration contrôlée d'agents phytochimiques WO2013148682A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13/429,601 2012-03-26
US13/429,601 US8858995B2 (en) 2008-03-10 2012-03-26 Methods and compositions for controlled delivery of phytochemical agents

Publications (1)

Publication Number Publication Date
WO2013148682A1 true WO2013148682A1 (fr) 2013-10-03

Family

ID=49261168

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/033877 WO2013148682A1 (fr) 2012-03-26 2013-03-26 Procédés et compositions pour l'administration contrôlée d'agents phytochimiques

Country Status (1)

Country Link
WO (1) WO2013148682A1 (fr)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2564438C1 (ru) * 2014-07-07 2015-09-27 Закрытое акционерное общество "ИльмиксГрупп" (далее - ЗАО "ИльмиксГрупп") Способ лечения простатической интраэпителиальной неоплазии (пин)
KR20170045223A (ko) * 2014-08-19 2017-04-26 더 리젠츠 오브 더 유니버시티 오브 캘리포니아 국소 약물 전달을 위한 임플란트 및 이의 사용 방법
CN107375191A (zh) * 2017-07-28 2017-11-24 吕鹏威 一种用于长效降血糖弹性体皮下埋植棒
CN110917243A (zh) * 2019-12-30 2020-03-27 南京晓庄学院 一种含植物提取物的凝胶膏剂及其制备方法
US11173291B2 (en) 2020-03-20 2021-11-16 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery
US11338119B2 (en) 2020-03-20 2022-05-24 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery
US11344526B2 (en) 2020-03-20 2022-05-31 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050042293A1 (en) * 1997-10-29 2005-02-24 The University Of British Columbia Polymeric systems for drug delivery and uses thereof
US20050129731A1 (en) * 2003-11-03 2005-06-16 Roland Horres Biocompatible, biostable coating of medical surfaces
US7153520B2 (en) * 2000-12-07 2006-12-26 Samyang Corporation Composition for sustained delivery of hydrophobic drugs and process for the preparation thereof
US20100076542A1 (en) * 2007-02-21 2010-03-25 Eurocor Gmbh Coated expandable system
US20100086599A1 (en) * 2006-09-16 2010-04-08 Kairosmed Gmbh Oral modified release formulations

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050042293A1 (en) * 1997-10-29 2005-02-24 The University Of British Columbia Polymeric systems for drug delivery and uses thereof
US7153520B2 (en) * 2000-12-07 2006-12-26 Samyang Corporation Composition for sustained delivery of hydrophobic drugs and process for the preparation thereof
US20050129731A1 (en) * 2003-11-03 2005-06-16 Roland Horres Biocompatible, biostable coating of medical surfaces
US20100086599A1 (en) * 2006-09-16 2010-04-08 Kairosmed Gmbh Oral modified release formulations
US20100076542A1 (en) * 2007-02-21 2010-03-25 Eurocor Gmbh Coated expandable system

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2564438C1 (ru) * 2014-07-07 2015-09-27 Закрытое акционерное общество "ИльмиксГрупп" (далее - ЗАО "ИльмиксГрупп") Способ лечения простатической интраэпителиальной неоплазии (пин)
KR20170045223A (ko) * 2014-08-19 2017-04-26 더 리젠츠 오브 더 유니버시티 오브 캘리포니아 국소 약물 전달을 위한 임플란트 및 이의 사용 방법
EP3597176A1 (fr) * 2014-08-19 2020-01-22 The Regents Of The University Of California Implants pour administration localisée de médicaments et leurs procédés d'utilisation
US10912933B2 (en) 2014-08-19 2021-02-09 The Regents Of The University Of California Implants for localized drug delivery and methods of use thereof
US11324935B2 (en) 2014-08-19 2022-05-10 The Regents Of The University Of California Implants for localized drug delivery and methods of use thereof
KR102444092B1 (ko) 2014-08-19 2022-09-16 더 리젠츠 오브 더 유니버시티 오브 캘리포니아 국소 약물 전달을 위한 임플란트 및 이의 사용 방법
US11918770B2 (en) 2014-08-19 2024-03-05 The Regents Of The University Of California Implants for localized drug delivery and methods of use thereof
CN107375191A (zh) * 2017-07-28 2017-11-24 吕鹏威 一种用于长效降血糖弹性体皮下埋植棒
CN110917243A (zh) * 2019-12-30 2020-03-27 南京晓庄学院 一种含植物提取物的凝胶膏剂及其制备方法
US11173291B2 (en) 2020-03-20 2021-11-16 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery
US11338119B2 (en) 2020-03-20 2022-05-24 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery
US11344526B2 (en) 2020-03-20 2022-05-31 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery

Similar Documents

Publication Publication Date Title
AU2009223653B2 (en) Methods and compositions for controlled delivery of phytochemical agents
US8858995B2 (en) Methods and compositions for controlled delivery of phytochemical agents
WO2013148682A1 (fr) Procédés et compositions pour l'administration contrôlée d'agents phytochimiques
Esmeeta et al. Plant-derived bioactive compounds in colon cancer treatment: An updated review
Turrini et al. Potential effects of pomegranate polyphenols in cancer prevention and therapy
Kunihiro et al. Curcumin, but not curcumin-glucuronide, inhibits Smad signaling in TGFβ-dependent bone metastatic breast cancer cells and is enriched in bone compared to other tissues
Zavatti et al. Experimental study on Satureja montana as a treatment for premature ejaculation
Shwter et al. Chemoprevention of colonic aberrant crypt foci by Gynura procumbens in rats
El-Naggar et al. Nigella sativa L. seed extract modulates the neurotransmitter amino acids release in cultured neurons in vitro
Aqil et al. Multi-layer polymeric implants for sustained release of chemopreventives
Kundu et al. Prospect of natural products in glioma: a novel avenue in glioma management
Pellegrini et al. Naringenin modulates skeletal muscle differentiation via estrogen receptor α and β signal pathway regulation
Ullah et al. Suppression of cisplatin-induced vomiting by cannabis sativa in pigeons: neurochemical evidences
AU2014276836B2 (en) Composition for enhancing semen quality in a male subject
Cao et al. Phytochemical-mediated glioma targeted treatment: drug resistance and novel delivery systems
Choi et al. α-Terpineol induces fatty liver in mice mediated by the AMP-activated kinase and sterol response element binding protein pathway
KR20210081334A (ko) 님나무 (azadirachta indica) 조성물 및 암 치료 방법
Geerts et al. Sho–Saiko–To: The right blend of traditional oriental medicine and liver cell biology
Swanson Flavonoids, inflammation and cancer
Ali et al. Nephroprotective effects of Helianthus annuus seeds extract in gentamicin induced nephrotoxic male mice
Norfazlina et al. Cytotoxicity study of Nigella sativa and Zingiber zerumbet extracts, thymoquinone and zerumbone isolated on human myeloid leukemia (HL60) cell
JP2016506924A (ja) 閉経に関連する顔面潮紅発症の治療/軽減に使用するためのn−メチルセロトニンおよび関連物質
Pires et al. Pre-clinical interaction of ayahuasca, a brew used in spiritual movements, with morphine and propofol
El-Gendy The beneficial effect of Trifolium flower extracts on paracetamol-intoxicated male rats
Kashyap et al. Nephroprotective Effect of Luteolin against Gentamicin-induced Nephrooxicity in Albino Rats

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: 13768471

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: 13768471

Country of ref document: EP

Kind code of ref document: A1