AU2001253226B2 - Bile acid containing prodrugs with enhanced bioavailability - Google Patents

Bile acid containing prodrugs with enhanced bioavailability Download PDF

Info

Publication number
AU2001253226B2
AU2001253226B2 AU2001253226A AU2001253226A AU2001253226B2 AU 2001253226 B2 AU2001253226 B2 AU 2001253226B2 AU 2001253226 A AU2001253226 A AU 2001253226A AU 2001253226 A AU2001253226 A AU 2001253226A AU 2001253226 B2 AU2001253226 B2 AU 2001253226B2
Authority
AU
Australia
Prior art keywords
agent
biological activity
compound
prodrug
bile acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2001253226A
Other versions
AU2001253226A1 (en
Inventor
Andrew Coop
Kimberley A. Lentz
Dean Y. Maeda
James E. Polli
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Maryland at Baltimore
Original Assignee
University of Maryland at Baltimore
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Maryland at Baltimore filed Critical University of Maryland at Baltimore
Publication of AU2001253226A1 publication Critical patent/AU2001253226A1/en
Application granted granted Critical
Publication of AU2001253226B2 publication Critical patent/AU2001253226B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J43/00Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • C07J43/003Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton not condensed
    • 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/554Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being a steroid plant sterol, glycyrrhetic acid, enoxolone or bile acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J41/00Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring
    • C07J41/0033Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring not covered by C07J41/0005
    • C07J41/0055Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring not covered by C07J41/0005 the 17-beta position being substituted by an uninterrupted chain of at least three carbon atoms which may or may not be branched, e.g. cholane or cholestane derivatives, optionally cyclised, e.g. 17-beta-phenyl or 17-beta-furyl derivatives
    • C07J41/0061Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring not covered by C07J41/0005 the 17-beta position being substituted by an uninterrupted chain of at least three carbon atoms which may or may not be branched, e.g. cholane or cholestane derivatives, optionally cyclised, e.g. 17-beta-phenyl or 17-beta-furyl derivatives one of the carbon atoms being part of an amide group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • C07J9/005Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane containing a carboxylic function directly attached or attached by a chain containing only carbon atoms to the cyclopenta[a]hydrophenanthrene skeleton

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cardiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Botany (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Steroid Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicinal Preparation (AREA)

Description

WO 01/76531 PCT/US01/11327 BILE ACID CONTAINING PRODRUGS WITH ENHANCED BIOAVAILABILITY Background of the Invention Field of Invention This invention relates to the method of increasing the bioavailability and reducing the bioavailability variability of compounds by conjugating or linking a bile acid to the compound. This invention also relates to the new composition of matter obtained by attaching a bile acid to another compound to generate a prodrug. This invention further relates to the usage of the bile acid transporter to actively move a prodrug out of the lumen of the small intestine.
Description of the Related Art Insufficient or variable intestinal permeability is a reason for inadequate oral drug bioavailability. Many methods are available to increase bioavailability of various drugs While some of these methods improve the bioavailability of some drugs, not one method works for all compounds.
Some compounds that exhibit poor bioavailability, sub-optimal permeability, or variable bioavailability may have some or all of the following characteristics 1. Less than complete oral absorption.
2. Permeability less than a suitable reference marker metoprolol tartrate a permeability of 40 x 10-6 cm/sec across Caco-2 monolayers]).
3. Molecular weight greater than 500 Daltons.
4. Hydrogen donors greater than five.
-1- WO 01/76531 PCT/US01/11327 Hydrogen bond acceptors greater than 6. Being substrates for P-glycoprotein efflux, Multi-drug Resistanceassociated Protein (MRP) efflux, or other efflux systems.
7. Not being substrates of or ligands for a carrier or transporter.
One potential method to increase the bioavailability of compounds with these characteristics is to conjugate a bile acid to the drug or compound to create a prodrug.
A bile acid conjugated prodrug allows for an increase in bioavailability and/or a reduction in the variability of the compound because of active transport of the prodrug by the intestinal bile acid transporter (IBAT). The intestinal absorption of bile acids is a sodium dependent process involving the human intestinal bile acid transporter (hIBAT) and Na+/K ATP-ase The bile acid transporter is an ideal candidate for drug targeting because the human IBAT has a high transport capacity of 10 grams per day and bile acids are one of the largest molecules taken up by a carrier mediated system In light of this biology, the bile acid transporter appears to be a promising mechanism to improve oral drug absorption by incorporating a bile acid moiety with an active drug in a prodrug fashion. The bile acid prodrug approach to targeting the bile acid transporter in various tissues has been investigated with peptides 8, HMG- CoA reductase inhibitors and chlorambucil This work was directed either at extremely small molecules or for targeting delivery of drugs to the liver. Other attempts at drug therapy involve inhibiting the bile acid transporter to reduce cholesterol synthesis in the liver [11].
WO 01/76531 PCT/US01/11327 One compound that may benefit from bile acid conjugation to improve uptake in the small intestine is acyclovir. Acyclovir is an anti-viral compound, used to inhibit herpes virus proliferation. Its target is not the liver but all tissues within the body.
Typical treatment requires 200 mg doses administered five times daily, with a bioavailability after oral administration of 20% [12].
A different prodrug strategy has proved successful at improving the oral bioavailability of acyclovir [13, 14]. Valacyclovir, the L-valine ester prodrug of acyclovir, has an oral bioavailability of 54% This improved bioavailability for valacyclovir allows for a more convenient dosing regimen of 1000 mg, twice daily, with similar clinical efficacy as previously found for the acyclovir parent compound Valacyclovir is a substrate for the human intestinal peptide transporter (PepT1), with a Ki=4.08 mM in PepT1 expressing Xenopus laevis oocytes [17] and Ki= 1 .10 mM in stable lines of CHO/PepT1 [18].
In comparison to PepT1, IBAT has the potential advantages of higher capacity and micromolar affinity. Therefore, it is possible to further enhance the oral bioavailability of acyclovir by synthesizing a cholic acid ester prodrug to target the IBAT. It is also possible to increase the bioavailability of other compounds by conjugating the compounds to a bile acid.
Furthermore, animals other than humans have an intestinal bile acid transporter. One can use the animal intestinal bile acid transport and a bile acid conjugated prodrug to increase the adsorption of the prodrug and bioavailability of the compound in animals.
P:\WPDOCS\CRNV\MM7744590.ncw pages.2.doc- Summary of the Invention In a first broad form, the present invention relates to increasing the bioavailability of a compound by conjugating a compound to a bile acid to create a prodrug.
Accordingly, in a first aspect, the invention provides a compound represented by formula IX:
R
I R, wherein RI, R 2 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and betahydroxyl; x is a moiety containing a metabolically labile bond; and R4 is selected from the group consisting of an agent having biological, activity and a metabolic precursor of an agent having biological activity; or a pharmaceutically acceptable salt or solvate thereof.
According to a second aspect, the invention further provides a compound represented by formula X: Ri-R x wherein R 2 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R4 is a moiety that enhances binding of the compound to the intestinal bile acid transporter; X is a moiety containing a metabolically labile bond; -4- PAWPDOCS\CR AMMX7744590," pagcs.2.dwand Ri is selected from the group consisting of an agent having biological, activity or a metabolic precursor of an agent having biological activity; or a pharmaceutically acceptable salt or solvate thereof.
According to a third aspect, the invention further provides a compound represented by formula XI: R1
RXI
R2 Xl wherein R 1 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R 4 is a moiety that enhances binding of the compound to the intestinal bile acid transporter; X is a moiety containing a metabolically labile bond; and R 2 is selected from the group consisting of an agent having biological activity and a metabolic precursor of an agent having biological activity; or a pharmaceutically acceptable salt or solvate thereof.
According to a fourth aspect, the invention further provides a compound represented by formula XIII:
RI-X
R
2 R4 g R'2 XIII P:\WPDOCS\MDTSpcc'\7744590 doc23/05/06 -4b- Ct wherein R 2 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R4 is a solubility enhancing moiety; X is a moiety C, containing a metabolically labile bond; and R 1 is selected from the group consisting of an agent having biological activity and a metabolic precursor of an agent having biological IN 5 activity; or a pharmaceutically acceptable salt or solvate thereof.
According to a fifth aspect, the invention further provides a compound represented t^ by formula XIV: R2
XIV
wherein Ri and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R4 is a solubility enhancing moiety; X is a moiety containing a metabolically labile bond; and R 2 is selected from the group consisting of an agent having biological activity and a metabolic precursor of an agent having biological activity; or a pharmaceutically acceptable salt or solvate thereof.
According to a sixth aspect, the invention further provides a method of increasing the bioavailability of an agent having biological activity or a metabolic precursor of an agent having biological activity or reducing the variability of said bioavailability, said method comprising: conjugating a bile acid to said agent having biological activity or said metabolic precursor of an agent having biological activity through a metabolically labile bond to form a prodrug, and orally administering said prodrug to a subject in need thereof, wherein said prodrug is selected from the compounds according to the present invention.
According to a seventh aspect, the invention further provides a method of eliminating adverse interactions between an agent having biological activity or a metabolic precursor of an agent having biological activity and a compound, wherein said adverse interactions result from intestinal absorption of at least one of said agent having biological activity or said metabolic precursor of an agent having biological activity and said compound, said method comprising: conjugating a bile acid to said agent having biological P \WPDOCS\.MDTSpc\7744590 do-23105106 4c activity or said metabolic precursor of an agent having biological activity through a metabolically labile bond to form a prodrug, and orally administering said prodrug to a subject in need thereof, wherein said prodrug is selected from the compound according to the present invention.
According to an eighth aspect, the invention further provides a method of eliminating adverse interactions between an agent having biological activity or a metabolic precursor of an agent having biological activity and a compound, wherein said adverse interactions result from intestinal absorption of said compound or said agent having biological activity or metabolic precursor of an agent having biological activity, said method comprising: conjugating a bile acid to said agent having biological activity or said metabolic precursor of an agent having biological activity through a metabolically labile bond to form a prodrug, and orally administering said prodrug to a subject in need thereof, wherein said PAWPDOCS\CRNAMM\7744590ne pag2.domprodrug is selected from the compounds according the present invention.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or group of integers or steps but not the exclusion of any other integer or group of integers or steps.
The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.
In a preferred form, a compound of the invention is of the formula:
R,
R4 .Rj R2 wherein R 1
R
2 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl;
R
4 is selected from the group consisting of an agent having biological activity and a metabolic precursor of an agent having biological activity; or a pharmaceutically acceptable salt, solvent, or polymorph thereof.
In a further preferred form, a compound of the invention is of the formula: P.\WPDOCS\CRN\A1Nl\74459.new, Pages.2 docwherein R 2 and R 3 are independently selected from the group consisting of hydrogen, aipha-hydroxyl, and beta-hydroxyl; R, is selected from the group an agent having P:\WPDOCS\CRN\SET\Spec\7744590.new pages docbiological activity and a metabolic precursor of an agent having biological activity; R 4 is any chemical moiety that enhances binding of the compound to The intestinal bile acid transporter; or a pharmaceutically acceptable salt, solvent or polymorph thereof.
In yet another preferred form, a compound of the invention is of the formula: wherein R 1 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R 3 is selected from the group an agent having biological activity and a metabolic precursor of an agent having biological activity; R 4 is any chemical moiety that enhances binding of the compound to the intestinal bile acid transporter; or a pharmaceutically acceptable salt, solvent, or polymorph thereof.
Preferably, a further compound of the invention is of the formula:
R
2 wherein R 1 and R 2 are independently selected from The group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R 3 is selected from the group an agent having biological activity and a metabolic precursor of an agent having biological activity; R 4 is p:\WPDOCS\CRN\SET\SpA\7714590. pag.doxany chemical moiety that enhances binding of the compound to the intestinal bile acid transporter; or a pharmaceutically acceptable salt, solvent, or polymorph thereof In yet another preferred form, a compound of the invention is of the formula: wherein R 2 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; RI is selected from the group an agent having biological activity and a metabolic precursor of an agent having biological activity; R 4 is any chemical moiety that increases solubility of the compound; or a pharmaceutically acceptable salt solvent, or polymorph thereof.
In still yet another preferred form, a compound is of the formula: P:\WPDOCS CRN\SET\Spec\7744590.newpages.docwherein R 1 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R 2 is selected from the group an agent having biological activity and a metabolic precursor of an agent having biological activity; R 4 is any chemical moiety that increases solubility of the compound; or a pharmaceutically acceptable salt, solvent, or polymorph thereof.
In another preferred form, a compound of the invention is of the formula: wherein Ri and R 2 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R 3 is selected from the group an agent having biological activity and a metabolic precursor of an agent having biological activity; R 4 is any chemical moiety that increases solubility of the compound; or a pharmaceutically acceptable salt, solvent, or polymorph thereof.
In still yet another preferred form, a compound is of the formula: P:\WPDOCS\CRN\SETSpec\7744590.new pages.doc-
R
4
X
Preferably, the prodrug is administered orally to an animal or human.
Even more preferably, the intestinal bile acid transporter binds to the prodrug and moves the prodrug from the lumen of the small intestine into brush border cells.
In yet another preferred form, the prodrug or compound moves from inside the brush border cells into the blood stream.
In another preferred form, the bioavailability of a compound is increased by conjugating a compound to a bile acid to create a prodrug.
Preferably, a metabolically labile bond exists between the compound and the bile acid.
Even more preferably, the prodrug is administered orally to an animal or human.
In another preferred form, the intestinal bile acid transporter binds to the prodrug and moves the prodrug from the lumen of the small intestine into brush border cells.
In yet another preferred form, the prodrug or compound moves from inside the brush border cells into the blood stream.
In still yet another preferred form, the bioavailability of a compound is increased by linking a compound to a bile acid via a linker group to create a prodrug.
In still another preferred form, the prodrug is administered orally to an animal or human.
Preferably, the intestinal bile acid transporter binds to the prodrug and moves the prodrug from the lumen of the small intestine into brush border cells.
P\WPDOCS\CRN\SETSpe\7744590.new pagcs.doc- Even more preferably, the prodrug or compound moves from inside the brush border cells into the blood stream.
In still yet another preferred form, the linker group may be any bifunctional chemical moiety that achieves one or more of the following four functions: facilitates the synthesis of the prodrug, aid or enhance the binding of the prodrug to the intestinal bile acid transporter (IBAT), make it easier for the compound to disassociate from the bile acid after the prodrug has passed out of the lumen of the small intestine, enhance the solubility of the prodrug inside the body.
Preferably, the linker group may be any size, but more preferably be less than 200 daltons.
Even more preferably, a metabolically labile bond exists between the linker group and the compound, between the linker group and the bile acid, or within the linker group itself.
In a further preferred form, the bioavailability of a biologically active compound is increased by conjugating a biologically active compound to a bile acid to create a prodrug.
In another preferred form, a metabolically labile bond exists between the biologically active compound with the bile acid.
Preferably, the prodrug is administered orally to an animal or human.
Even more preferably, the intestinal bile acid transporter binds to the prodrug and moves the prodrug from the lumen of the small intestine into brush border cells.
In another preferred form, the prodrug or compound moves from inside the brush border cells into the blood stream.
In still yet another preferred form, a compound is conjugated to a bile acid to form a prodrug.
In another preferred form, the present invention relates to a pharmaceutical compound containing a compound and a bile acid.
In still yet another preferred form, the present invention relates to a pharmaceutical compound containing a compound, a metabolically labile bond, and a bile acid.
In a further preferred form, the present invention relates to a pharmaceutical compound containing an agent, a linker group, and a bile acid.
P:\WPDOCS\CRN\SE'Sp\7744590.ncw pagm.doc- Preferably, the present invention relates to a pharmaceutical compound containing an agent, a linker group, a bile acid, and a metabolically labile bond.
In another preferred form, the present invention relates to a pharmaceutical compound containing an agent with biological activity and a bile acid.
In yet another preferred form, the present invention relates to a pharmaceutical compound containing an agent with biological activity, a metabolically labile bond, and a bile acid.
In a further preferred form, the present invention relates to a pharmaceutical compound containing an agent with biological activity, a linker group, and a bile acid.
In still yet another preferred form, the present invention relates to a pharmaceutical compound containing an agent with biological activity, a linker group, a bile acid, and a metabolically labile bond.
Preferably, the present invention utilizes the intestinal bile acid transporter to actively uptake and remove from the lumen of the small intestine a bile acid containing prodrug.
In another preferred form, the present invention relates to a method for increasing the bioavailability of a compound by using the intestinal bile acid transporter to actively uptake and remove from the lumen of the small intestine a bile acid containing prodrug.
Preferably, the inventions uses a linker group to link a compound to a bile acid to create a prodrug.
Even more preferably, the linker group may be any bifunctional chemical moiety that achieves one or more of the following four functions: facilitates the synthesis of the prodrug, aid or enhance the binding of the prodrug to the intestinal bile acid transporter (IBAT), make it easier for the compound to disassociate from the bile acid after the prodrug has passed out of the lumen of the small intestine, enhance the solubility of the prodrug inside the body.
Even more preferably, the linker group may be any size, and even more preferably, less than 200 daltons.
Preferably, the linker group, when attached to the compound or the bile acid, results in a metabolically labile bond being present.
P:\WPDOCS\CRN\SET\Spc\7744590.new pages.doc- Even more preferably, the bioavailability variability of a compound is reduced by conjugating a compound to a bile acid to create a prodrug.
In a further preferred form, the prodrug is administered orally to an animal or human.
Preferably, the intestinal bile acid transporter binds to the prodrug and moves the prodrug from the lumen of the small intestine into brush border cells.
Even more preferably, the prodrug or compound moves from inside the brush border cells into the blood stream.
In yet another preferred form, the bioavailability variability of a compound is reduced by conjugating a compound to a bile acid to create a prodrug.
Preferably, a metabolically labile bond exists between the compound and the bile acid.
Even more preferably, the prodrug is administered orally to an animal or human.
In a further preferred form, the intestinal bile acid transporter binds to the prodrug and moves the prodrug from the lumen of the small intestine into brush border cells.
Even more preferably, the prodrug or compound moves from inside the brush border cells into the blood stream.
Preferably, the bioavailability variability of a compound is reduced by linking a compound to a bile acid via a linker group to create a prodrug.
Even more preferably, the prodrug is administered orally to an animal or human.
In a further preferred form, the intestinal bile acid transporter binds to the prodrug and moves the prodrug from the lumen of the small intestine into brush border cells.
In yet another preferred form, the prodrug or compound moves from inside the brush border cells into the blood stream.
In still yet another preferred form, the linker group may be a bifunctional chemical moiety.
Even more preferably, the linker group may be any size, but more preferably is less than 200 daltons.
Preferably, a metabolically labile bond exists between the linker group and the compound, between the linker group and the bile acid, or within the linker group itself.
P:\WPDOCS\CRN\SETSpC\774459Gf- page.do- Even more preferably, the bioavailability variability of a biologically active compound is reduced by conjugating a biologically active compound to a bile acid to create a prodrug.
In a further preferred form, a metabolically labile bond exist between the biologically active compound with the bile acid.
In another preferred form, the prodrug is administered orally to an animal or human.
In still yet another preferred form, the intestinal bile acid transporter binds to the prodrug and moves the prodrug from the lumen of the small intestine into brush border cells.
Even more preferably, the prodrug or compound moves from inside the brush border cells into the blood stream.
In another preferred form, an adverse drug-drug interaction is reduced or prevented by linking or conjugating a bile acid to at least one of the compounds which is involved in the adverse drug-drug interaction to create a prodrug.
Preferably, the intestinal bile acid transporter is used to uptake the prodrug, thereby avoiding a transporter that is involved in the adverse drug-drug interaction.
Even more preferably, an adverse drug-nutrient interaction is reduced or prevented by linking or conjugating a bile acid to a compound which is involved in the adverse drugnutrient interaction to create a prodrug.
In a further preferred form, the intestinal bile acid transporter is used to uptake the prodrug, thereby avoiding a transporter that is involved in the adverse drug-nutrient interaction.
In yet another preferred form, the lipophilicity of a compound is increased by linking or conjugating a bile acid to the compound.
Preferably, the increased lipophilicity will increase the bioavailability and reduce the variability of the bioavailability of the compound.
Even more preferably, the compound component of the prodrug will be cleaved from the prodrug after the prodrug binds to the intestinal bile acid transporter.
In another preferred form, the prodrug is coated with a substance that protects the prodrug from the acidic environment of the stomach and that does not inhibit absorption of the prodrug in the intestine.
?:\WPDOCS\CRN\SET\Sp\7744590.neW pagv.doc- In a further preferred form, the present invention relates to a pharmaceutical compound of acyclovir valyldeoxycholate.
o In still another preferred form, the present invention relates to a pharmaceutical N 5 compound of acyclovir valylchenodeoxycholate.
Even more preferably, the present invention relates to a pharmaceutical compound of atenolol cholic acid amide.
Description of Several Views of the Drawings Figure A illustrates the general structure of bile acids.
Figure 1B illustrates the general structure of the prodrug.
Figure 1C generalizes the synthesis of acyclovir valylchenodexycholate.
Figure ID generalizes the synthesis of acyclovir valyldeoxycholate.
Figure 1E generalizes the synthesis of atenolol cholic acid amide.
Figure 2 illustrates the competitive inhibition of uptake of 3 H-taurocholate by acyclovir valylchenodeoxycholate (acyclovir vCDC) chenodeoxycholate (CDC) and valacyclovir(A).
-lOc- WO 01/76531 PCT/US01/11327 Figure 3 illustrates the competitive inhibition of uptake of 3 H-taurocholate by acyclovir valyldeoxycholate (acyclovir vDC) deoxycholate (DC) and valacyclovir Figure 4 illustrates the competitive inhibition of uptake of 3 -taurocholate by atenolol cholic acid amide cholate and atenolol Figure 5 shows the concentration dependence of 3 H-taurocholate uptake in COShIBAT (COS cells transfected with hIBAT) in HBSS and in MHBSS (no sodium) Figure 6 illustrates the inhibition constant (Ki) of various bile acids and other agents by inhibiting 3 H-taurocholate uptake in COS cells transfected with hIBAT.
Detailed Description of the Invention In the preferred embodiment, this invention is a prodrug containing a bile acid attached to a compound. In an alternative embodiment, more than one bile acid can be attached to a compound. Alternatively, more than one compound can be attached to one bile acid. Alternatively, a complex of one or more bile acids can be attached to one or more compounds.
When used wherein, the word "compound" includes, but is not limited to, a pharmaceutical drug, a biologically active agent, a metabolic precursor to a pharmaceutical drug, a metabolic precursor to a biologically active agent, or any other agent which one would want to administer to an animal or human. A compound may be therapeutic or diagnostic in nature. A compound can also be nutritionally beneficial to an animal or human.
11 WO 01/76531 PCT/US01/11327 It is preferable that a metabolically labile bond exist between a compound and the bile acid to which the compound is attached or conjugated. By way of example only, a metabolically labile bond can be an ester, amide, carbamate, carbonate, ether, urea, anhydride, or sulfur containing derivatives, such as thioamides, thioesters, thiocarbamates, and thioureas.
In an alternative embodiment, a linker group exists between a compound and the bile acid to which the compound is attached. The linker group can be any bifunctional chemical moiety but preferably has a molecular weight less than 200 daltons. Four reasons exist for using a linker group; any bifunctional chemical moiety that achieves any one of these reasons is considered a linker group. First, the linker group can facilitates the synthesis of the prodrug. Second, the linker group can aid or enhance the binding of the prodrug to the intestinal bile acid transporter (IBAT).
Third, the linker group can make it easier for the compound to disassociate from the bile acid after the prodrug has passed out of the lumen of the small intestine. Fourth, the linker group can enhance the solubility of the prodrug inside the body.
Different types of linker groups can be used. Linker groups can be, for example, natural and unnatural amino acids, di-acids, di-amines, di-alcohols, sulphate, phosphate, sulfur containing moieties, amino alcohols, hydroxyacids, and polymers.
Examples of the amino acids are valine, glycine, taurine, alanine, leucine, tyrosine, aspartate, glutamate, lysine, arginine, asparagine, cysteine. Examples of the di-acids are oxalic acid, fumaric acid, succinic acid, maleic acid, and tartaric acid. Examples of the di-amines include ethylenediamine, propylenediamine, 1,3-diaminopropane, 1,4- -12- WO 01/76531 PCT/US01/11327 diaminobutane, 1,5-diaminopentane, piperazine, homopiperazine, and 3aminopiperidine. Examples of the di-alcohols are ethyleneglycol, propyleneglycol, 1,4-butanediol, polyethyleneglycol, and 1,5-pentanediol. Examples of the sulfur containing moieties are mercaptoacetic acid, mercaptopropanoic acid, mercaptobenzyl alcohol, 2-mercaptoethanol, 3-mercaptopropanol, and 4-mercaptobutanol. Examples of the amino alcohols are 2-aminoethanol, 3-aminopropanol, 4-aminobutanol, 4hydroxypiperidine, and 3-hydroxypiperidine. Examples of the hydroxyacids include 3-hydroxypropanoic acid, 4-hydroxybutanoic acid, and 5-hydroxypentanoic acid.
One example of polymers is polyethylene glycol.
It is preferably that the linker group has a metabolically labile bond for easy cleavage of the prodrug into the compound and the bile acid. By way of example only, a metabolically labile bond can be an ester, amide, carbamate, carbonate, ether, urea, anhydride, or sulfur containing derivatives, such as thioamides, thioesters, thiocarbamates, and thioureas.
These bile acid containing prodrugs are administered orally and absorbed in the small intestine. The intestinal bile acid transporter actively transports the prodrug from inside the lumen of the small intestine into brush border cells. Once inside brush border cells, the prodrug may pass passively through the cell membrane down a concentration gradient into the blood stream or be actively transported out of the brush border cells into the blood stream. This active transport of the prodrug out of the lumen of the small intestine occurs because of the affinity of the intestinal bile acid transporter for its ligand, bile acids.
-13- WO 01/76531 PCT/US01/11327 Intracellular or extracellular enzymes cleave one of the bonds between the bile acid and the compound either after the prodrug is inside the brush border cells, (2) attached to the outside of the brush border cells, located in proximity to the brush border cells, in the blood stream, or at other location inside the body, except cleavage should not occur in the stomach or lumen of the small intestine.
This active transport of a prodrug by the intestinal bile acid transport produces enhanced bioavailability of the compound when compared to the unconjugated compound. It also reduces bioavailability variability and provides for an alternative mechanism for compound permeation, including the avoidance of efflux pumps such as P-glycoprotein used by some drugs fexofenadine) and other drug transporters and/or nutrient transporters. By avoiding other drug transporters and/or nutrient transporters, drug-drug and/or drug-nutrient interactions can be reduced or eliminated.
Any bile acid may be conjugated or linked to a compound. More than one bile acid may be conjugated or linked to one compound. Also more than one compound may be conjugated or linked to one bile acid. The bile acids are, by way of example, cholate, glycocholate, taurocholate, deoxycholate, glycodeoxycholate, taurodeoxycholate, chenodeoxycholate, ursodeoxycholate, glycochenodeoxycholate, taurochenodeoxycholate, and lithocholate. The general structures of the bile acids are shown in Figure 1A.
The following structural requirements generally promote recognition of a bile acid by the intestinal bile acid transporter (IBAT): negatively charged side groups on -14- WO 01/76531 PCT/US01/11327 the bile acid; at least one alpha oriented hydroxyl group at the steroid nucleus at position 3, 7, or 12; and a cis configuration of rings A and B of the steroid nucleus By way of example, compounds that have improved bioavailability as a result of conjugating or linking the compound to a bile acid are those compounds which contain (Group 1) an alcohol function which can be chemically coupled to the bile acid to give an ester; or (Group 2) a primary or secondary amine function which can be chemically coupled to the bile acid to give an amide; or (Group 3) an acid function which can be chemically coupled to the bile acid to give an anhydride.
Compounds which contain an alcohol function (Group 1) may include, but are not limited to, anti-viral agents 6-deoxyacyclovir, ganciclovir, dihydroxybutylguanine, foscamet, penciclovir, famciclovir, zidovudine, idoxuridine, trifluorothymidine, vidarabine, cytarabine, ribavirin). Compounds which contain a primary or secondary amine function (Group 2) may include, but are not limited to, H-2 antagonists derivatives of cimetidine, ranitidine, nizatidine, famotidine, roxatidine). Compounds which contain an acid function (Group 3) may include, but are not limited to, bisphosphonates alendronate, etidronate, pamidronate, tiludronate, clonronate) and ACE inhibitors enalapril, captopril, lisonopril). This list is not exhaustive and is not meant to limit the scope of this invention, but is provided by way of illustration only.
Any salts, solvates, isomeric compositions, and physical forms of these compounds and/or their derivatives can be conjugated or linked to a bile acid to form a WO 01/76531 PCT/US01/11327 prodrug. One can also use known in the art field enteric coating to protect prodrugs from the acidity of the stomach.
Figure 1A illustrates the general structure of bile acids. Bile acids can vary in their substituents at R 1
R
2 and R 3 positions 12, 7, and 3, respectively). For example, for chenodeoxycholate, R 1 is H, R 2 is a-OH, and R 3 is a-OH. For deoxycholate, Ri is oc-OH, R 2 is H, and R 3 is a-OH. For cholate, R 1
R
2 and R 3 are all a-OH. Many natural and synthetic bile acids exist and are well-known in the art field. Any of these natural or synthetic bile acids can be used to create a prodrug.
However, it may be preferable to have at least one alpha orientated hydroxyl group in the bile acid and this alpha orientated hydroxyl group can be located at R 1
R
2 or R 3 Figure 1B illustrates the general structure of bile acid component of the prodrug. Either directly or via a linker group, a compound can be attached to any natural or synthetic bile acid at either R 1
R
2
R
3 or R4. While it may be preferable to have only one compound conjugated or linked to one bile acid, more than one compound can be conjugated or linked to the same bile acid. In addition, while it may be preferable to have the compound linked or conjugated to a bile acid at R 4 (because of potential structural requirements necessary for recognition of a bile acid by the intestinal bile acid transporter), it is possible to link or conjugate compounds to a bile acid at R 1
R
2 or R 3 When a compound is linked or conjugated to either R 1
R
2 or R 3 then it is preferable that R 4 be any chemical moiety which can aid or enhance the binding of the prodrug to the IBAT and/or can increase the solubility of the prodrug -16- WO 01/76531 PCT/US01/11327 inside the body. Examples of such chemical moieties include hydroxyl, any amino acid (such as glycyl, valyl, alanyl, tauryl, and leucyl), any di-amino acid (such as glycyl-valyl, glycyl-glycyl, valyl-glycyl, alanyl-glycyl, alanyl-valyl, and leucyl-valyl), any tri-amino acid (such as glycyl-glycyl-glycyl, glycyl-valyl-glycyl, glycyl-glycylalanyl, valyl-glycyl-leucyl, alanyl-glycyl-glycyl, alanyl-valyl-glycyl, and leucyl-valylvalyl). It is preferable that this chemical moiety have a molecular weight less than 1500 daltons.
Figures 1C, ID, and 1E generalize the synthesis of three prodrugs, acyclovir valylchenodeoxycholate, acyclovir valyldeoxycholate, and atenolol cholic acid amide, which have increased bioavailability and reduced bioavailability variability as a result of conjugating or linking the compounds, acyclovir and atenolol, to a bile acid. For these three prodrugs, the compounds are attached to a bile acid (chenodeoxycholate, deoxycholate, or cholic acid) at R 4 For acyclovir valylchenodeoxycholate and acyclovir valyldeoxycholate, valine is utilized as a linker group. Furthermore, both acyclovir valylchenodeoxycholate and acyclovir valyldeoxycholate use an ester as a metabolically labile bond that can be cleaved to release acyclovir. For atenolol cholic acid amide, no linker group is used, but rather atenolol is conjugated directly to cholic acid. Furthermore, atenolol cholic acid uses an amide as a metabolically labile bond that can be cleaved to release atenolol.
Acvclovir Valvyichenodeoxycholate Synthesis As in Figure 1 C, to synthesize acyclovir valylchenodeoxycholate, 1, isobutylchloroformate (iBuOCOCl; 130 gL, 1 mmol) is added dropwise to a cooled -17- WO 01/76531 PCT/US01/11327 oC) solution of chenodeoxycholate, 2, (1 mmol) and triethylamine (140 RL, 1 mmol) in N,N-dimethylformarnide (DMF) (10 mL) under an nitrogen (N 2 atmosphere. After 1.5 minutes, valacyclovir, 3, (0.42 g, 1.3 mmol) and triethylamine (NEt 3 280 jiL, 2 mmol) are then added to the reaction mixture as a solution in DMF (5 mL). The reaction is kept at -15 OC for 0.5 hours, then is warmed to room temperature for 1 hour. The triethylammonium chloride formed during the reaction is filtered off, and the filtrate is concentrated by rotary evaporation. The crude material is then purified using silica gel flash chromatograpy with MeOH/CHC13 250 mL) as the eluent.
The acyclovir valylchenodeoxycholate, 1, prodrug synthesis is monitored using thin layer chromotography (TLC) plates coated with silica gel GHLF-0.25 mm plates (60 F 254 manufactured by Analtech, Inc. (Newark, DE). Fast-atom bombardment mass spectrometry (FAB-MS) and high resolution mass spectrometry (HRMS) spectra are obtained on a Jeol SX 102 mass spectrometer in the positive ion mode. Proton nuclear magnetic resonance (NMR) spectrometry is performed in d 6 dimethyl sulfoxide (DMSO) on a 300 MHz General Electric Aquerius model spectrometer controlled by a Macintosh Power Mac 7100 using MacNMR v. software. The purity of the bile acid conjugate is determined by analysis on a Beckman System Gold high pressure liquid chromotography (HPLC) system consisting of a model 126 solvent module, model 168 detector, and model 507 autosampler. The HPLC column used is a Vydac analytical column (C 18 300 A, 18- WO 01/76531 PCT/US01/11327 4.6 x 250 mm) equipped with a guard cartridge. Solvent A is aqueous 0.1% trifluoroacetic acid (TFA) and solvent B is acetonitrile containing 0.1% TFA. The conjugate is eluted using a linear gradient of 5 to 75% B over 50 minutes at a flow rate of 1.0 mL/min and detected at 214 nm.
The amount of acyclovir valylchenodeoxycholate, 1, purified is 0.62 g Additionally, TLC RA(MeOH/CHC1 3 1:4) 0.46; HPLC Rt= 32.1 min (99.3 purity); and ESI-MS 700.4. HRMS (calculated for C 37
H
5 9 0 7
N
6 699.4445.
found 699.4454. The NMR spectra contained peaks consistent with both chenodeoxychloate and valacyclovir portions. Coupling through the amino acid amine (and not the aniline) was confirmed through the presence of the NH 2 signal at 5.3 ppm.
Acyclovir Valyldeoxycholate Synthesis As in Figure ID, to synthesize acyclovir valyldeoxycholate, 4, isobutylchloroformate (iBuOCOCI; 130 gL, 1 mmol) is added dropwise to a cooled (-15 solution of deoxycholate, 5, (1 mmol) and triethylamine (140 gL, 1 mmol) in N,N-dimethylformamide (DMF) (10 mL) under an nitrogen (N 2 atmosphere. After minutes, valacyclovir, 3, (0.42 g, 1.3 mmol) and triethylamine (NEt 3 280 pL, 2 mmol) are then added to the reaction mixture as a solution in DMF (5 mL). The reaction is kept at -15 oC for 0.5 hours, then is warmed to room temperature for 1 hour. The triethylammonium chloride formed during the reaction is filtered off, and -19- WO 01/76531 PCT/US01/11327 the filtrate is concentrated by rotary evaporation. The crude material is then purified using silica gel flash chromatograpy with MeOH/CHC1 3 250 mL) as the eluent.
The acyclovir valyldeoxycholate, 4, prodrug synthesis is monitored using thin layer chromotography (TLC) plates coated with silica gel GHLF-0.25 mm plates
F
25 4 manufactured by Analtech, Inc. (Newark, DE). Fast-atom bombardment mass spectrometry (FAB-MS) and high resolution mass spectrometry (HRMS) spectra are obtained on a Jeol SX 102 mass spectrometer in the positive ion mode. Proton nuclear magnetic resonance (NMR) spectrometry is performed in d 6 -dimethyl sulfoxide (DMSO) on a 300 MHz General Electric Aquerius model spectrometer controlled by a Macintosh Power Mac 7100 using MacNMR v. 5.0 software. The purity of the bile acid conjugate is determined by analysis on a Beckman System Gold high pressure liquid chromotography (HPLC) system consisting of a model 126 solvent module, model 168 detector, and model 507 autosampler. The HPLC column used is a Vydac analytical column (C 1 8 300 A, Sm, 4.6 x 250 mm) equipped with a guard cartridge. Solvent A is aqueous 0.1% trifluoroacetic acid (TFA) and solvent B is acetonitrile containing 0.1% TFA. The conjugate is eluted using a linear gradient of to 75% B over 50 minutes at a flow rate of 1.0 mL/min and detected at 214 nm.
The amount of acyclovir valyldeoxycholate, 4, purified is 0.59 g Additionally, TLC R(MeOH/CHC3, 1:4) 0.47; HPLC Rt 33.0 min (98.1 purity); FAB-MS 699.54; HRMS (calculated for C 37
H
59 0 6
N
7 699.4445, found: 699.4448. The NMR spectrum contained peaks consistent with both WO 01/76531 PCT/US01/11327 deoxycholic acid and valacyclovir portions. Coupling through the amino acid amine (and not the aniline) was confirmed through the presence of the NH 2 signal at 5.3ppm.
Atenolol Cholic Acid Amide Synthesis As in Figure 1E, to synthesize atenolol cholic acid amide, 6, isobutylchloroformate (iBuOCOC1; 130 IL, 1 mmol) is added dropwise to a cooled OC) solution of cholic acid, 7, (0.5 mmol) and triethylamine (140 gL, 1 mmol) in N,N-dimethylformamide (DMF) (10 mL) under an nitrogen (N 2 atmosphere. After minutes, atenolol, 8, (0.42 g, 1.3 mmol) and triethylamine (NEt 3 280 lL, 2 mmol) are then added to the reaction mixture as a solution in DMF (5 mL). The reaction is kept at -15 °C for 0.5 hours, then is warmed to room temperature for 1- hour. The triethylammonium chloride formed during the reaction is filtered off, and the filtrate is concentrated by rotary evaporation. The crude material is then purified using silica gel flash chromatograpy with MeOH/CHCl 3 250 mL) as the eluent.
The atenolol cholic acid amide, 6, prodrug synthesis is monitored using thin layer chromotography (TLC) plates coated with silica gel GHLF-0.25 mm plates
F
254 manufactured by Analtech, Inc. (Newark, DE). Fast-atom bombardment mass spectrometry (FAB-MS) and high resolution mass spectrometry (HRMS) spectra are obtained on a Jeol SX 102 mass spectrometer in the positive ion mode. Proton nuclear magnetic resonance (NMR) spectrometry is performed in d 6 -dimethyl sulfoxide (DMSO) on a 300 MHz General Electric Aquerius model spectrometer controlled by a Macintosh Power Mac 7100 using MacNMR v. 5.0 software. The -21- WO 01/76531 PCT/US01/11327 purity of the bile acid conjugate is determined by analysis on a Beckman System Gold high pressure liquid chromotography (HPLC) system consisting of a model 126 solvent module, model 168 detector, and model 507 autosampler. The HPLC column used is a Vydac analytical column (Cls, 300 A, Stm, 4.6 x 250 mm) equipped with a guard cartridge. Solvent A is aqueous 0.1% trifluoroacetic acid (TFA) and solvent B is acetonitrile containing 0.1% TFA. The conjugate is eluted using a linear gradient of to 75% B over 50 minutes at a flow rate of 1.0 mL/min and detected at 214 nm.
The amount ofatenolol cholic acid amide, 6, purified is 0.19 g Additionally, TLC Rf(AcOH/MeOH/CHCl3, 1:1:4) 0.47; HPLC Rt 34.9 min (98.3 purity); FAB-MS 657.6; HRMS (calculated for C 38
H
61
N
2 0 7 657.4479, found: 657.4470; IR (CHCl 3 3394, 2932, 2870, 1671, and 1610 cm- 1 Atenolol, 8, contains both a secondary alcohol and a secondary amine, and coupling to cholic acid, 7, through both of these nucleophiles could occur to give an ester or an amide, respectively. Only the amide is formed during the above reaction of cholic acid and atenolol which is confirmed through the presence of characteristic amide C=O stretching bands at 1671 cm' 1 and 1610 cm 1 in the IR spectrum. Also, NMR spectrum is consistent with that expected for a conjugate between cholic acid and atenolol the absence of the secondary amine proton signal at 4.92 ppm in the NMR). Coupling through the amine of atenolol (and not the alcohol) is also confirmed by a negative chloranil test, a test for secondary and primary amines. Atenolol is used as the positive control for the chloranil test.
-22- WO 01/76531 PCT/US01/11327 Bioavailability Assays To test the increase of bioavailability of acyclovir valylchenodeoxycholate, 1, acyclovir valyldeoxycholate, 4, and atenolol cholic acid amide, 6, the human intestinal bile acid transporter (hIBAT) cDNA (specifically, pCMVS-hIBAT expression plasmid) is transformed into competent DH5ca cells according to the Life Technologies protocol (Grand Island, NY), using LipofectAMINE 2000 transfection reagent (Life Technologies, Grand Island, NY). Cell dilutions are streaked on nutrient agar plates containing 50 gg/ml ampicillin, and incubated at 37 0 C overnight. Isolated colonies are selected aseptically, and are used to inoculate a 200 ml nutrient broth culture containing 50 pg/ml ampicillin. The culture is incubated for 24 hours at 225 RPM's and 37°C. cDNA is isolated from the broth cultures using a Quigan maxi-prep kit (DNA plasmid maxi kit #12162; Valencia, CA). After an isopropanol precipitation and ethanol wash, the DNA is reconstituted in 600 ll of sterile deionized water. DNA concentration is 1.18 gjg/ll, determined by spectrophotometry at 260 nm. The 260/280 absorbance ratio is 1.50, indicating the DNA is free from RNA contamination. pCMV5-hIBAT is also digested with the restriction endonucleases, BAMH1 and NDE, and results in bands of appropriate size (4238 bp and 1689 bp) when electrophoresed on a 1% agarose gel.
COS-7 cells are grown in T-75 flasks at 37 0 C, 5% CO 2 and 95% RH using Dulbecco's Modified Eagle's Medium (DMEM) supplemented with 10% FBS. Cells are passaged at 80-90% confluency using a 0.25% trypsin/0.20% EDTA solution, and -23- WO 01/76531 PCT/US01/11327 are plated at a density of 8x104 cells per well (1.88 cm 2 Cells are transfected at 24 hours post seeding. For each well transfected, 0.8 glg hIBAT cDNA is combined with LipofectAMINE 2000 reagent and incubated at room temperature for 20 minutes to allow complexes to form. A volume of 100 pl of hIBAT-lipid complex is added to each well of cells. Transfected cells are incubated at 37 0 C, 5% CO 2 and 95% RH for 24 hours, until ready for uptake assay.
Uptake studies are performed on COS-hIBAT cells at 24 hours posttransfection. Uptake buffer consists of either a Hank's Balanced Salts Solution (HBSS) containing 137 mM NaC1 or a Modified Hank's Balanced Salts Solution (MHBSS) that replaced the sodium chloride with 137 mM tetraethylammonium chloride Because bile acid transport is sodium dependent, this MHBSS approach allows for the simple modification of the uptake buffer to exclude all sources of sodium, and thus enables for a control uptake assay under sodium-free conditions.
For the Km and Vm, studies of taurocholate in the COS-hIBAT cells, cell culture medium is removed from each well and is replaced with 0.5 ml of uptake solution. 0.5 [tM 3 H-taurocholate is investigated in HBSS. Cells are incubated at 37 0 C and 100 RPM's for 10 minutes. Uptake solution is removed, and the cells are washed three times with ice cold HBSS. Cells are lysed, neutralized, and counted for associated radioactivity. Each well is analyzed for protein content using the Lowry method This uptake assay is performed in triplicate, under both sodium and sodium-free conditions.
-24- WO 01/76531 PCT/US01/11327 The saturable uptake of 3 H-taurocholate is determined using the following equation (Formula 1): dM Vm S d- V S kS (Formula 1) dt Km+S where Vmax and Km represent the Michaelis-Menten constants, kp is the passive uptake rate constant, S is the concentration of taurocholate, and dM/dt is the uptake rate oftaurocholate into the COS-hIBAT cells. This approach includes the contribution of passive uptake to the saturable kinetics of taurocholate uptake into the cells.
In the competitive inhibition studies (see Figures 2, 3, and the concentration of acyclovir valylchenodeoxycholate, 1, varies between 10 kM and 400 gM; the concentration of acyclovir valyldeoxycholate, 4, varies between 10 UM and 600 iM; and the concentration of atenolol cholic acid amide, 6, varies between 10 gM and 400 pM. Chenodeoxycholate, 2, deoxycholate, 5, and cholate inhibition studies are conduced as positive controls. Valacyclovir and atenolol inhibition studies are performed as negative controls.
Figure 2 illustrates the inhibition study of valacyclovir, 3, chenodeoxycholate (CDC), 2, and acyclovir valylchenodeoxycholate (acyclovir vCDC), 1, There is no inhibition of the bile acid transporter in COS-hIBAT cells after application of up to 600 gM valacyclovir, indicating that valacyclovir is not a substrate for hIBAT. A chenodeoxycholate inhibition study is performed as a WO 01/76531 PCT/US01/11327 positive control for this experiment, and results in Ki =5.4 jM, which indicates that the COS-hlBAT model is suitable for the inhibition study. The calculated Ki for acyclovir valylchenodeoxycholate is Ki =35.6 gM, indicating a very strong interaction of acyclovir valylchenodeoxycholate for the hIBAT transporter.
Similiarly for acyclovir valyldeoxycholate (Figure inhibition study of valacyclovir, 3, deoxycholate 5, and acyclovir valyldeoxycholate (acyclovir vDC), 4, are performed. Acyclovir valyldeoxycholate strongly interacts with hIBAT with a Ki=401 gM.
Likewise, in Figure 4, atenolol cholic acid amide, 6, strongly interacts with hIBAT with a Ki=160 gM, while atenolol, 8, itself does not interact. The natural bile acid cholate, 7, also strongly interacts with hIBAT [Ki=32.1 2.4) jiM].
Figure 5 shows both the sodium dependence and saturation of 3 H-taurocholate uptake. The uptake rate is measured at 3H-taurocholate concentrations from 0.1 to 125 jiM in HBSS with 137 mM NaCl and in MHBSS which contains no sodium. As expected, carrier mediated 3 H-taurocholate uptake is not present in the absence of sodium ions, thus establishing a baseline permeability due to the passive permeability of taurocholate into the cells. A passive uptake rate constant (kp) of 0.12 (pmoles/min mg protein)/RM is estimated using linear regression. Control uptake studies are also performed in untransfected COS-7 cells, and in COS-7 cells transfected with the antibiotic resistant vector, pcDNA3. The passive uptake of 3 H-taurocholate in these -26- WO 01/76531 PCT/US01/11327 experiments is the same as that obtained under sodium-free conditions (data not shown). The kinetic parameters for carrier mediated uptake are estimated using WinNonlin (version and yields a Km=12.0(±2.2) RM and a Vax=126.0(±5.9) pmoles/min/mg protein. These Michaelis-Menten parameters are in close agreement to those obtained previously [21].
Competitive inhibition studies are performed in HBSS containing 0.25 pM 3
H-
taurocholate, and varying the concentration of unlabeled bile acid from 1 to 100 gM.
Glycine, taurine, acyclovir, and atenolol inhibition studies are performed as negative controls. Incubation conditions and analysis are performed as described for the Km and Vmax studies. The following equation (Formula 2) is used to estimate the K i for a series of naturally occurring bile acids: dM VaX (K d= S I (Formula 2) dt S K K, where Vmax and Km are the Michaelis-Menten parameters for taurocholate uptake, S is 0.25 p.M 3 H-taurocholate, dM/dt is the uptake rate of taurocholate, and I is the concentration of inhibitor applied to the cells.
The effect of various bile acids (cholate, glycocholate, taurocholate, deoxycholate, taurodeoxycholate, glycodeoxycholate, chenodeoxycholate, glycochenodeoxycholate, taurochenodeoxycholate, ursodeoxycholate, and lithocholate) on the uptake of 3 H-taurocholate into COS-hIBAT cells is determined (see Figure 6).
Inhibition studies using glycine, taurine, valacyclovir, and atenolol are also performed -27- WO 01/76531 PCT/US01/11327 as negative controls. Figure 6 shows the Ki value SEM) for each compound tested.
All bile acids inhibit the uptake of 3 H-taurocholate into COS-hIBAT cells. However, the uptake of 3 H-taurocholate is not reduced in the presence of glycine or taurine.
Uptake of 3 H-taurocholate decreases in the presence of lithocholate, but never reaches 50% maximal velocity at a lithocholate concentration of 100 gLM. Lithocholate concentrations above 100 pM were not investigated. All bile acids inhibit the uptake of 3 H-taurocholate in COS-hIBAT cells. Valacyclovir and atenolol do not inhibit the uptake of 3 H-taurocholate. In these inhibition studies, 3 H-taurocholate concentration is held constant at 0.25 p.M and inhibitor concentration is varied between 1 and 100 pM; except for glycine and taurine, which range in concentration from 50 to 200 pgM, valacyclovir which range from 10 to 600 gM, and atenolol which range from 10 to 200 M. Inhibition studies are performed in triplicate. N/A denotes not applicable, because there was no evidence of inhibition.
Compounds can be conjugated or linked via a linker group to bile acids to improve the bioavailability and reduce the bioavailability variability of the compounds. It is preferable that a metabolically labile bond exist between the bile acid and the compound (with or without a linker group) for easy cleavage of the compound from the bile acid. While it is preferable that the compound or compound and linker group be attached to the R 4 of the bile acid, one can also attach the compound or compound and linker group at R 1
R
2 or R 3 Also, one can link or conjugate different -28- WO 01/76531 PCT/US01/11327 or the same compounds to multiple positions on the bile acid. Furthermore, one may attach more than one bile acid to a compound.
The prodrugs can be coated with various coating compounds known in the art field to protect the prodrug from the acidic environment in the stomach. These coating compounds dissolve in the basic environment in the small intestine, thereby permitting the prodrug to be available for uptake by the IBAT.
The prodrugs can also be converted into a pharmaceutically acceptable salt or pharmaceutically acceptable solvate or other physical forms polymorphs by way of example only and not limitation) via known in the art field methods.
Pharmaceutically acceptable carriers can be used along with the prodrugs. In making the compositions of the present invention, the prodrug can be mixed with an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the prodrug. Thus, the compositions can be in the form of tablets, pills, powers, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, soft and hard gelatin capsules, and other orally ingestible formulations.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; -29- WO 01/76531 PCT/US01/11327 emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxybenzoates, sweetening agents; and flavoring agents. The compositions of the present invention can also be formulated so as to provide quick, sustained or delayed release of the prodrug after administration to the patient by employing procedures known in the art.
The compositions are preferably formulated in a unit dosage form, each dosage containing from about 0.005 mg to 100 mg, more usually about 1.0 mg to about 300 mg, of the prodrug. Unit dosage form generally refers to physically discrete units suitable as unitary dosages for human and animal patients, each unit containing a predetermined quantity of the prodrug calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
A prodrug is used for treatment of human or animal patients which are in need of treatment with the compound that is contained in the prodrug. The specific purpose of the treatment, and the dose range to be administered, depends on the identity of the compound and the condition for which the patient is to be treated.
While a list of compounds for which this prodrug approach will work is too large to be contained here, below is a short list of compounds and their "drug" class: foscarnet, valacyclovir, acyclovir, ganciclovir, penciclovir, famciclovir (antivirals); alendronate, etidronate disodiumn, pamidronate, risedronate, tiludronate, clodronic acid (bisphosphonates); cimetidine, ranitidine (H-2 antagonists); enalaprilate, captopril, lisonopril (ACE inhibitors); losartan, E-3171 (angiotensin II antagonists); levofloxacin, norfloxacin (quinalone antibiotic WO 01/76531 PCT/USO1/11327 which have decreased absorption with antacids); formycin B; acetbutalol, pindolol, aiprenolol, atenolol, nadolol (beta adrenergic blockers); bretylium tosylate (antiarrhythmic agents); cefuroxime sodium (cephalosporins); chiorothiazide, hydrochiorothiazide, furosemide (diuretic agents); gabapentin, lamotrigine (anticonvulsant); didanosine (nucleoside reverse transcriptase inhibitors); neviriapine (non-nucleoside reverse transcriptase inhibitors); ritinavir, saquinavir, amprinavir (HIV protease inhibitors); tacrolinus, cyclosporin (immunosuppresants); zafirlukast (leukotriene receptor antagonists); leuprorelin actetate (LHRH analogues); dDAVP (1-deamino-8-Darginine-vasopressin; desmopressin), calcitonin, thyrotropin releasing hormone (polypeptide hormones); loratidine, cetirizine (non-sedating antihistamines); penicillin V, amoxicillin, cefacor, cefixime, cefuroxime axetil, cefuroxime sodium, ampicillin (antibiotics); terbutaline hcmisulfatc (adrcncrgic agonist agents); metformin (anti-diabetics); celecoxib, refecoxib (COX-2 inhibitors); sumatriptan, naratriptan, araztriptan, zolmitriptan (anti-migraines); 6mercaptopurine; ziprasidone; RGD mimetic (alpha lIb beta 3-antagonists); leuenkephalin analogues; aipha-methyldopa; 5-fluorouracil (fluoropoyrimidines); tacrine (acetyicholinesterase inhibitors); DZ-2640 (the ester-type oral carbapenem prodrug of an active parent compound, DU-668 1, and other carbapenems); vitamin B 12 (nutrients and minerals); 7-chiorokynurenic acid; oseltamivir or its active moiety; RGD (Arg-Gly-Asp) analogs (glycoprotein (GP) lIb/Illa agonists and antagonists; platelet aggregation inhibitors); -31- WO 01/76531 PCT/US01/11327 sibrafiban (oral platelet aggregation inhibitors); nelarabine, 9-beta-Darabinofuranosyl guanine (ara-G), and ara-G; mycophenolate mofetil (MMF) and its active immunosuppressant mycophenolic acid (MPA); nabumetone and its active metabolite 6-methoxy-2-naphthylacetic acid (anti-osteoarthritis agents); adefovir (9-[2-phosphonylmethoxyethyl]-adenine [PMEA]) and adefovir dipivoxil [bis-(POM)-PMEA], and cidofovir (antiviral nucleotides); cromoglicate lisetil and cromoglycic acid (anti-arthritis agents); oseltamivir or its active moiety; its parent Ro 64-0802 (inhibitors of influenza virus neuraminidasc); peptidomimctics; nucleic acids; This is for illustrative purposes only and is not meant to be exhaustive. Other compounds will have increased bioavailability and/or reduced bioavailability variability with bile acid conjugation.
All references cited herein are incorporated by reference in their entirety.
References: Remington: The Science and Practice of Pharmacy Gennaro, editor).
Mack Publishing Compant, Easton, PA. 1995.
Lipinski, Lombardo, Dominy, Feeney, P.J. Adv. Drug Del.
Rev. 1997, 23, 3-25.
L. Lack, Properties and biological significance of the ileal bile salt transport system, Environ Health Perspect 33 (1979) 79-90.
-32- WO 01/76531 PCT/US01/11327 W. Kramer, G. Wess, A. Enhsen, E. Falk, A. Hoffmann, G. Neckermann, G.
Schubert and M. Urmann, Modified bile acids as carriers for peptides and drugs, J Cont Rel 46 (1997) 17-30.
S. Matern and W. Gerok, Pathophysiology of the enterohepatic circulation of bile acids, Rev Physiol Biochem Pharmacol 85 (1979) 126-204.
G. Burckhardt, W. Kramer, G. Kurz and F.A. Wilson, Inhibition of bile salt transport in brush border membrane vesicles from rat small intestine by photoaffinity labeling, J Biol Chem 258 (1983) 3618-3622.
M. Kagedahl, P.W. Swaan, C.T. Redemann, M. Tang, C.S. Craik, F.C. Szoka, Jr. and S. Oie, Use of the intestinal bile acid transporter for the uptake of cholic acid conjugates with HIV-1 protease inhibitory activity, Pharm Res 14 (1997) 176-80.
W. Kramer, G. Wess, A. Enhsen, K. Bock, E. Falk, A. Hoffinann, G.
Neckermann, D. Gantz, S. Schulz and L. Nickau, Bile Acid derived HMG- CoA reductase inhibitors., Biochim Biophys Acta 1227 (1994) 137-154.
D.C. Kim, A.W. Harrison, M.J. Ruwart, K.F. Wilkinson, J.F. Fisher, I.J.
Hidalgo and R.T. Borchardt, Evaluation of the bile acid transporter in enhancing intestinal permeability to renin-inhibitory peptides., J Drug Target 1 (1993) 347-359.
[10] W. Kramer, G. Wess, G. Schubert, M. Bickel, F. Girbig, U. Gutjahr, S.
Kowalewski, K.H. Baringhaus, A. Enhsen and H.G. H, Liver-specific drug targeting by coupling to bile acids., J Biol Chem 267 (1992) 18598-18604.
-33- WO 01/76531 PCT/US01/11327 [11] M.C. Lewis, L.E. Brieaddy and C. Root, Effects of 2164U90 on ileal bile acid absorption and serum cholesterol in rats and mice, J Lipid Res 36 (1995) 1098- 1105.
[12] P. deMiranda and M.R. Blum, Pharmacokinetics of acyclovir after intravenous and oral administration, J Antimicrob Chemother 12 (1983) 29-37.
[13] M.A. Jacobson, Valaciclovir (BW256U87): the L-valyl ester of acyclovir., J Med Virol Supplement 1 (1993) 150-153.
[14] H. Gao, A.K. Mitra. Synthesis of acyclovir, gancyclovir, and their prodrugs: A review. Synthesis (2000): 329-359.
[15] J. Soul-Lawton, E. Seaber, N. On, R. Wootton, P. Rolan and J. Posner, Absolute bioavailability and metabolic disposition of valaciclovir, the L-valyl ester of acyclovir, following oral administration to humans, Antimicrob Agents Chemother 39 (1995) 2759-2764.
[16] K.H. Fife, R.A. Barbarash, T. Rudolph, B. Degregorio and R. Roth, Valaciclovir versus acyclovir in the treatment of first-episode genital herpes infection. Results of an international, multicenter, double-blind, randomized clinical trial, Sex Transm Dis 24 (1997) 481-486.
[17] P.V. Balimane, I. Tamai, A. Guo, T. Nakanishi, H. Kitada, F.H. Leibach, A.
Tsuji and P.J. Sinko, Direct evidence for peptide transporter (PepT1)mediated uptake of a nonpeptide prodrug, valacyclovir, Biochem Biophys Res Comm 250 (1998) 246-251.
-34- WO 01/76531 PCT/US01/11327 [18] H. Han, R.L. deVrueh, J.K. Rhie, K.M. Covitz, P.L. Smith, C.P. Lee, D.M.
Oh, W. Sadee and G.L. Amidon, 5'-Amino acid esters of antiviral nucleosides, acyclovir, and AZT are absorbed by the intestina; PEPT1 peptide transporter, Pharm Res 15 (1998) 1154-1159.
[19] C.E. Chandler, L.M. Zaccaro and J.B. Moberly, Transepithelial transport of cholyltaurine by Caco-2 cell monolayers is sodium dependent, Am J Physiol 264 (1993) G1118-25.
O.H. Lowry, N.J. Rosebrough, A.L. Farr and R.J. Randall, Protein measurement with the folin phenol reagent, J Biol Chem 193 (1951) 265-275.
[21] M.H. Wong, P. Oelkers and P.A. Dawson, Identification of a mutation in the ileal sodium-dependent bile acid transporter gene that abolishes transport activity, J Biol Chem 270 (1995) 27228-27234.
[22] F. Gaspari and M. Bonati, Correlation between n-octanol/water partition coefficient and liquid chromatographic retention for caffeine and its metabolites, and some structure-pharmacokinetic considerations, J Pharm Pharmacol 39 (1987) 252-60.
[23] K.A. Lentz, J. Hayashi, L.J. Lucisano and J.E. Polli, Development of a more rapid, reduced serum culture system for Caco-2 monolayers and application to the Biopharmaceutics Classification System, Int J Pharm 200 (2000) 41-51.
[24] G.A. Sawada, C.L. Barsuhn, B.S. Lutzke, M.E. Hooughton, G.E. Padbury, N.F.H. Ho and T.J. Raub, Increased lipophilicity and subsequent cell WO 01/76531 PCT/US01/11327 partitioning decrease passive transcellular diffusion of novel, highly lipophilic antioxidants, J Pharm Exp Ther 288 (1999) 1317-1326.
G.A. Sawada, L.R. Williams, B.S. Lutzke and T.J. Raub, Novel, highly lipophilic antioxidants readily diffuse across the blood-brain barrier and access intracellular sites, J Pharm Exp Ther 288 (1999) 1327-1333.
While the invention has been described in detail, and with reference to specific embodiments thereof, it will be apparent to one of ordinary skill in the art that various changes and modifications can be made therein without departing from the spirit and scope thereof. The artisan will further acknowledge that the Examples recited herein are demonstrative only and are not meant to be limiting.
-36-

Claims (14)

1. A compound represented by formula IX: wherein R 1 R 2 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and betahydroxyl; x is a moiety containing a metabolically labile bond; and R 4 is selected from the group consisting of an agent having biological, activity and a metabolic precursor of an agent having biological activity; or a pharmaceutically acceptable salt or solvate thereof.
2. A compound represented by formula X: wherein R 2 and R3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R 4 is a moiety that enhances binding of the compound to the intestinal bile acid transporter; X is a moiety containing a metabolically labile bond; and RI is selected from the group consisting of an agent having biological, activity or a metabolic precursor of an agent P:\WPDOCS\CRN\SESpcc\7744590.nw claims.2.doc- -38- having biological activity; or a pharmaceutically acceptable salt or solvate thereof.
3. A compound represented by formula XI: wherein R 1 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R 4 is a moiety that enhances binding of the compound to the intestinal bile acid transporter; X is a moiety containing a metabolically labile bond; and R 2 is selected from the group consisting of an agent having biological activity and a metabolic precursor of an agent having biological activity; or a pharmaceutically acceptable salt or solvate thereof.
4. A compound represented by formula XIII: RI-X R4 R4 32 XIII wherein R 2 and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; P \WPDOCS\MDT1Spcs\7744590 doc-23/0906 -39- R 4 is a solubility enhancing moiety; X is a moiety containing a metabolically labile bond; and R, is selected from the group consisting of an agent having biological activity and a metabolic precursor of an agent having biological activity; or a pharmaceutically acceptable salt or solvate thereof.
A compound represented by formula XIV: Rs 3 R2 XIV wherein RI and R 3 are independently selected from the group consisting of hydrogen, alpha-hydroxyl, and beta-hydroxyl; R4 is a solubility enhancing moiety; X is a moiety containing a metabolically labile bond; and R 2 is selected from the group consisting of an agent having biological activity and a metabolic precursor of an agent having biological activity; or a pharmaceutically acceptable salt or solvate thereof.
6. A method of increasing the bioavailability of an agent having biological activity or a metabolic precursor of an agent having biological activity or reducing the variability of said bioavailability, said method comprising: conjugating a bile acid to said agent having biological activity or said metabolic precursor of an agent having biological activity through a metabolically labile bond to form a prodrug, and orally administering said prodrug to a subject in need thereof, wherein said prodrug is selected from the compounds according to any one of claims P \WPDOCS\.MDT\Spm\7744590 dC.23.'006
7. The method of claim 6, wherein the absorption of said agent having biological activity or said metabolic precursor of an agent having biological activity is increased when attached to said bile acid.
8. A method of eliminating adverse interactions between an agent having biological activity or a metabolic precursor of an agent having biological activity and a compound, wherein said adverse interactions result from intestinal absorption of at least one of said agent having biological activity or said metabolic precursor of an agent having biological activity and said compound, said method comprising: conjugating a bile acid to said agent having biological activity or said metabolic precursor of an agent having biological activity through a metabolically labile bond to form a prodrug, and orally administering said prodrug to a subject in need thereof, wherein said prodrug is selected from the compound according to any one of claims
9. The method of claim 8, wherein the absorption of said agent having biological activity or said metabolic precursor of an agent having biological activity is increased when attached to said bile acid.
A method of eliminating adverse interactions between an agent having biological activity or a metabolic precursor of an agent having biological activity and a compound, wherein said adverse interactions result from intestinal absorption of said compound or said agent having biological activity or metabolic precursor of an agent having biological activity, said method comprising: conjugating a bile acid to said agent having biological activity or said metabolic precursor of an agent having biological activity through a metabolically labile bond to form a prodrug, and orally administering said prodrug to a subject in need thereof, wherein said prodrug is selected from the compounds according to any one of claims
11. The method of claim 10, wherein the absorption of said agent having biological activity or said metabolic precursor of an agent having biological activity is increased when attached to said bile acid. P \WPDOCSM1D'Specs\7744590 dc.24t05i06 V-41-
12. A compound represented by formula IX, as defined in claim 1, a compound t represented by formula X, as defined in claim 2, a compound represented by formula XI, as defined in claim 3, a compound represented by formula XIII, as defined in claim 4, a O compound represented by formula XIV, as defined in claim 5, substantially as hereinbefore C" 5 described, with reference to the accompanying Examples. I
13. A method of increasing the bioavailability of an agent having biological activity or a metabolic precursor of an agent having biological activity or reducing the I variability of said bioavailability, said method according to claim 6 and substantially as hereinbefore described, with reference to the accompanying Examples.
14. A method of eliminating adverse interactions between an agent having biological activity or a metabolic precursor of an agent having biological activity and a compound, wherein said adverse interactions result from intestinal absorption of at least one of said agent having biological activity or said metabolic precursor of an agent having biological activity and said compound, said method according to claim 8 and substantially as hereinbefore described, with reference to the accompanying Examples. A method of eliminating adverse interactions between an agent having biological activity or a metabolic precursor of an agent having biological activity and a compound, wherein said adverse interactions result from intestinal absorption of said compound or said agent having biological activity or metabolic precursor of an agent having biological activity, said method according to claim 10 and substantially as hereinbefore described, with reference to the accompanying Examples. DATED this twenty-fourth day of May 2006 University of Maryland, Baltimore By its Patent Attorneys DAVIES COLLISON CAVE
AU2001253226A 2000-04-07 2001-04-06 Bile acid containing prodrugs with enhanced bioavailability Ceased AU2001253226B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US19585400P 2000-04-07 2000-04-07
US60/195,854 2000-04-07
US26990601P 2001-02-21 2001-02-21
US60/269,906 2001-02-21
PCT/US2001/011327 WO2001076531A2 (en) 2000-04-07 2001-04-06 Bile acid containing prodrugs with enhanced bioavailability

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2006202213A Division AU2006202213A1 (en) 2000-04-07 2006-05-25 Bile acid containing prodrugs with enhanced bioavailability

Publications (2)

Publication Number Publication Date
AU2001253226A1 AU2001253226A1 (en) 2002-01-10
AU2001253226B2 true AU2001253226B2 (en) 2006-06-08

Family

ID=26891401

Family Applications (3)

Application Number Title Priority Date Filing Date
AU5322601A Pending AU5322601A (en) 2000-04-07 2001-04-06 Bile acid containing prodrugs with enhanced bioavailability
AU2001253226A Ceased AU2001253226B2 (en) 2000-04-07 2001-04-06 Bile acid containing prodrugs with enhanced bioavailability
AU2006202213A Abandoned AU2006202213A1 (en) 2000-04-07 2006-05-25 Bile acid containing prodrugs with enhanced bioavailability

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU5322601A Pending AU5322601A (en) 2000-04-07 2001-04-06 Bile acid containing prodrugs with enhanced bioavailability

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2006202213A Abandoned AU2006202213A1 (en) 2000-04-07 2006-05-25 Bile acid containing prodrugs with enhanced bioavailability

Country Status (5)

Country Link
EP (1) EP1267898A4 (en)
JP (1) JP2003530329A (en)
AU (3) AU5322601A (en)
CA (1) CA2405151A1 (en)
WO (1) WO2001076531A2 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002243204A1 (en) * 2000-10-06 2002-06-11 Xenoport, Inc. Bile-acid derived compounds for enhancing oral absorption and systemic bioavailability of drugs
US6992076B2 (en) * 2000-10-06 2006-01-31 Xenoport, Inc. Bile-acid derived compounds for providing sustained systemic concentrations of drugs after oral administration
EP1361847A2 (en) 2000-10-06 2003-11-19 Xenoport, Inc. Bile-acid conjugates for providing sustained systemic concentrations of drugs
AU2003303714B2 (en) 2003-01-13 2009-02-19 Bracco Imaging S.P.A. Improved linkers for radiopharmaceutical compounds
BRPI0418651A (en) * 2004-08-02 2007-05-29 Ambrilia Biopharma Inc lysine based compounds
JP2009530399A (en) * 2006-03-22 2009-08-27 シンデクサ ファーマシューティカルズ コーポレーション Compounds and methods for the treatment of diseases associated with ER stress
JP5201546B2 (en) * 2008-01-18 2013-06-05 学校法人近畿大学 N-acetylcysteine-conjugated bile acid, pharmaceutical composition containing the same and method for producing the same
ES2929516T3 (en) 2014-10-31 2022-11-29 Univ Utah Res Found Compositions and methods for particles with bile acids

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9212511D0 (en) * 1992-06-12 1992-07-22 Cortecs Ltd Pharmaceutical compositions
AUPO912997A0 (en) * 1997-09-11 1997-10-02 Commonwealth Scientific And Industrial Research Organisation Antiviral agents
GB9907048D0 (en) * 1999-03-27 1999-05-19 Karobio Ab Novel glucocorticoid receptor ligands for the treatment of meta bolic disorders
ATE363074T1 (en) * 1999-09-14 2007-06-15 Xenoport Inc SUBSTRATES AND SCREENING METHODS FOR TRANSPORT PROTEINS

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Kramer et al., Biochim Biophys Acta, 1994, 1227, 137-154 *
Kramer et al., J Biol Chem, 1992, 267(26) 18598-604 *
References 7-10 of admitted prior art described at page 2 of the specification *

Also Published As

Publication number Publication date
AU2006202213A1 (en) 2006-06-15
EP1267898A4 (en) 2006-02-01
JP2003530329A (en) 2003-10-14
EP1267898A2 (en) 2003-01-02
CA2405151A1 (en) 2001-10-18
AU5322601A (en) 2001-10-23
WO2001076531A2 (en) 2001-10-18
WO2001076531A3 (en) 2002-02-14

Similar Documents

Publication Publication Date Title
AU2006202213A1 (en) Bile acid containing prodrugs with enhanced bioavailability
US20230000993A1 (en) Silicon based drug conjugates and methods of using same
US10421773B2 (en) Bone-selective osteogenic oxysterol bisphosphonate analogs
US9884886B2 (en) Disulfide masked prodrug compositions and methods
AU2013356386B2 (en) Nucleoside kinase bypass compositions and methods
US20070167353A1 (en) Prodrug composition
US20070149446A1 (en) Tamandarin analogs and fragments thereof and methods of making and using
Zhang et al. Prodrug design targeting intestinal PepT1 for improved oral absorption: design and performance
IL263147A (en) Pegylated carfilzomib compounds
AU2001253226A1 (en) Bile acid containing prodrugs with enhanced bioavailability
US9550803B2 (en) Method to improve antiviral activity of nucleotide analogue drugs
US8940313B2 (en) Tyrosine-based prodrugs of antiviral agents
US20030212051A1 (en) Bile acid containing prodrugs with enhanced bioavailabilty
US20160303251A1 (en) Conjugates of garftase inhibitors
WO2021126990A1 (en) Oxytocin derivatives with improved properties
US20240245717A1 (en) Antiviral compounds, compositions, and uses thereof
CN113633780B (en) Small molecule conjugate and application thereof
Mehiri et al. A “ready-to-use” fluorescent-labelled-cysteine-TBTP (4-thiobutyltriphenylphosphonium) synthon to investigate the delivery of non-permeable PNA (peptide nucleic acids)-based compounds to cells
Roche et al. Synthesis and in vitro biological evaluation of valine-containing prodrugs derived from clinically used HIV-protease inhibitors
Etienne et al. Synthesis of camptothecin–amino acid carbamate linkers
NZ703894B2 (en) Substituted Aliphanes and Metallocenes Useful for Treating HCV Infections
NZ617102B2 (en) Substituted aliphanes, cyclophanes, heteraphanes, heterophanes and hetero-heteraphanes useful for treating hcv infections
NZ617102A (en) Substituted aliphanes, cyclophanes, heteraphanes, heterophanes and hetero-heteraphanes useful for treating hcv infections

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired