EP2037954A2 - Method for modulating the pharmacokinetics and metabolism of a therapeutic agent - Google Patents
Method for modulating the pharmacokinetics and metabolism of a therapeutic agentInfo
- Publication number
- EP2037954A2 EP2037954A2 EP07812192A EP07812192A EP2037954A2 EP 2037954 A2 EP2037954 A2 EP 2037954A2 EP 07812192 A EP07812192 A EP 07812192A EP 07812192 A EP07812192 A EP 07812192A EP 2037954 A2 EP2037954 A2 EP 2037954A2
- Authority
- EP
- European Patent Office
- Prior art keywords
- agent
- enzyme
- therapeutic agent
- pharmaceutical composition
- effective amount
- 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.)
- Withdrawn
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y302/00—Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
- C12Y302/01—Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
- C12Y302/01031—Beta-glucuronidase (3.2.1.31)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/43—Enzymes; Proenzymes; Derivatives thereof
- A61K38/46—Hydrolases (3)
- A61K38/47—Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
Definitions
- the present invention is directed to a method for enhancing the activity of a therapeutic agent.
- the invention is a method for modulating the pharmacokinetics and metabolism of a therapeutic agent by the administration to a subject in need thereof a therapeutic agent in combination with an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- the present invention is further directed to a combination product comprising a therapeutic agent and an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- the present invention is also directed to a method for administering to a subject in need thereof a therapeutic agent in combination with an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- Biotransformation of a therapeutic agent to a metabolite of the agent by drug metabolizing enzymes plays an important role in determining the pharmacokinetic profile and/or toxico logical effects of many pharmaceutical compounds.
- the natural role of metabolizing enzymes is to enable the clearance and in some cases, detoxification, of xenobiotics.
- a drug-metabolizing enzyme can also limit the bioavailability of the therapeutic agent or produce metabolites of the therapeutic agent, which metabolites may cause toxicity. From both a clinical and drug development perspective, extensive metabolism can limit the exposure levels of the therapeutic agent that can be achieved, making it difficult to characterize the dose response and making it more difficult to evaluate toxicity and safety.
- the liver was once thought to be the dominant site for the pass metabolism of compounds delivered by the oral route.
- the intestine also has been shown to play a central role in the metabolism of a number of different drugs.
- CYP3A4 enzymes in the small intestine have been shown to play an important role in metabolism of drugs such as midazolam, saquanavir and oxybutinin.
- UDP- glucuronosyltransferase (uridine 5-diphosphate-glucuronosyltransferase) in the intestine has been shown to play an important role in the metabolism of drugs such as the non-steroidal anti-inflammatory drug diclofenac and the immunosuppressive agent mycophenolate mofetil.
- Chemical based approaches include structural modification of the parent compound, coadministration of an enzyme inhibitor or co-administration of a buffering agent.
- Drug delivery based approaches include bypassing duodenal metabolism and delivering a compound to the colon or bypassing the liver through transdermal or parenteral routes. Although both chemical based and drug delivery based approaches can be effective, they also have inherent limitations.
- CYP450's and beta glucuronidase oral administration of these enzymes has not been considered.
- One explanation for this is that the enzyme may be inactivated by the acidic conditions in the stomach, prior to reaching the small intestine.
- Another object of the present invention is a method for modulating the pharmacokinetics and metabolism of a therapeutic agent by the administration of a therapeutic agent in combination with an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- a further object of the present invention is to increase the bioavailability and/or enhance the pharmacokinetic profile of a therapeutic agent and/or its metabolites through the administration of an enzyme, whereby administering the enzyme transforms a metabolite of the agent back to the agent.
- a further object of the present invention is to increase the bioavailability and/or enhance the pharmacokinetic profile (e.g. increase the duration of action) of a therapeutic agent and/or its metabolites through the oral administration of an enzyme.
- Another object of the present invention is to provide a combination product comprising a therapeutic agent and an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- Another object of the present invention is to provide a method for the coadministration of the therapeutic agent with an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- Another object of the present invention is to provide a method for administering to a subject in need thereof a therapeutic agent in combination with an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- the methods of the present invention increase the bioavailability of a therapeutic agent, which agent is subject to extensive glucuronidation and enterohepatic re-circulation.
- the bioavailability of a potential cancer therapeutic agent is increased, which agent is subject to extensive glucuronidation and enterohepatic recirculation, by transforming the metabolite of the agent back to the agent; e.g. by administering a drug metabolizing enzyme such as ⁇ -glucuronidase.
- Figure 1 is a schematic diagram illustrating the effect of oral administration of ⁇ -glucuronidase on pharmacokinetics and metabolism of a therapeutic agent that undergoes glucuronidation.
- Section (A) represents duodenal delivery of a therapeutic agent and/or enzyme;
- Section (B) represents absorption of the agent and intestinal metabolism resulting in formation of a glucuronidated metabolite;
- Section (C) represents hepatic metabolism resulting in formation of additional glucuronide metabolite;
- Section (D) represents transport of the agent and glucuronide metabolite into systemic circulation and distribution;
- Section (E) represents enterohepatic recirculation leading to biliary excretion of the agent and glucuronide metabolite into the duodenum;
- Section (F) represents cleavage of the glucuronide metabolite to the agent and subsequent reabsorption and return to Section (B);
- Section (G) represents elimination of the agent and metabol
- the present invention is directed to providing a method for enhancing the activity of a therapeutic agent.
- the present invention is directed to a method for modulating the pharmacokinetics and metabolism of a therapeutic agent by the administration to a subject in need thereof a therapeutic agent in combination with an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- the present invention is further directed to a combination product comprising a therapeutic agent and an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- the present invention is also directed to a method for administering to a subject in need thereof a therapeutic agent in combination with an enzyme, whereby a metabolite of the agent is transformed back to the agent.
- the method of the present invention is to increase the bioavailability and/or enhance the pharmacokinetic profile of a metabolized compound and/or its metabolites through the coadministration of an enzyme.
- An embodiment of the method of the present invention further comprises the administration of the therapeutic agent in combination with an enzyme for transforming the metabolite of the agent back to the agent, whereby the enzyme releases the agent from a metabolic conjugate form in the small intestine.
- An example of the method includes modulating the pharmacokinetics and metabolism of a therapeutic agent by the administration of the therapeutic agent in combination with an enzyme, whereby the therapeutic effectiveness of the agent is boosted.
- Another example of the method includes a substantially reduced dose of the therapeutic agent, whereby the therapeutic effectiveness of the agent is below a heretofore known recommended dose and well below toxic levels, thus consequently improving the therapeutic window of said agent.
- Another embodiment of the method of the present invention further comprises modulating the pharmacokinetics and metabolism of a therapeutic agent by the administration of the agent in combination with ⁇ -glucuronidase, whereby ⁇ -glucuronidase releases the agent from a glucuronide conjugate form in the small intestine, thereby modifying the pharmacokinetics and metabolic disposition of the agent.
- An advantage of the present invention is that the effective dose of a therapeutic agent can be substantially reduced, since the de-conjugated agent is allowed to enterohepatically recirculate and be reabsorbed into the enterocyte, i.e. the cells of the epithelium, thus subjecting the de-conjugated agent to "second" pass gut and hepatic metabolism.
- a further embodiment of the method of the present invention further comprises oral coadministration of the enzyme ⁇ -glucuronidase and a therapeutic agent, wherein, as shown in Figure 1, the ⁇ -glucuronidase catalyzes hydrolysis of the extensively glucuronidated conjugate agent, thereby increasing the bioavailability of the agent.
- a further example of the method includes increasing the bioavailability of a therapeutic agent that typically undergoes glucuronidation in the small intestine and enterohepatic re-circulation, for instance:
- an anticholesterol agent including, but not limited to, ZETIA ® brand of ezetimibe (Harris M, Davis W, et al., Ezetimibe, Drugs of Today, 2003, 39(4): 229-
- a cancer therapeutic agent including, but not limited to, irrinotecan, ZARNESTRA ® (brand of tipifarnib), histone-deacetylases (HDAC) or (6,7-dimethoxy- 2,4-dihydro-indeno[l,2-c]pyrazol-3-yl)-(3-fluoro-phenyl)-amine;
- an analgesic agent including, but not limited to, acetominophen, morphine (Fisher MB, Campanale K, et al, In vitro glucuronidation using human liver microsomes and the pore-forming peptide alamethicin, Drug Metab. Dispos., 2000, 28(5): 560-6), codeine (Vree TB, van Dongen RT, et al., Codeine analgesia is due to codeine-6-glucuronide, not morphine, Int. J. Clin.
- an anti-psychotic agent including, but not limited to, risperidone
- a selective estrogen receptor modulator including, but not limited to, tamoxifen or raloxifme; - a retinoid (Fisher, et al.);
- a fluoroquinolone including, but not limited to, ciprofloxacin (Tanimura H, Tominaga S, et al., Transfer of ciprofloxacin to bile and determination of biliary metabolites in humans, Arzneistoffforschung, 1986, 36(9): 1417-20; and, Tachibana M, Tanaka M, et al., Acyl glucuronidation of fluoroquinolone antibiotics by the UDP- glucuronosyltransferase IA subfamily in human liver microsomes, Drug Metab. Dispos., 2005, 33(6): 803-11); - an anticonvulsant agent including, but not limited to, topiramate, carisbamate, valproic acid or GW273293;
- an anti-HIV agent including, but not limited to, zidovudine
- a blood glucose lowering agent including, but not limited to, troglitazone or muraglitazar
- - an antidiabetic agent including, but not limited to, muraglitzar
- - an anti-Parkinson's disease therapeutic agent including, but not limited to, tolcapone
- an antismoking agent including, but not limited to, nicotine.
- a further example of the method includes increasing the bioavailability of a therapeutic agent by administering one of the enzymes selected from the group consisting of N-oxide reductase, sulfoxide reductase, esterase, amidase, glucosidase, ⁇ -glucoronidase and sulfatase.
- a further example of the method includes increasing the bioavailability of a therapeutic agent by administering a combination product of an esterase enzyme and an ester prodrug.
- the present invention is directed to a method for transforming a metabolite of a therapeutic agent back to the agent by the administration of an enzyme.
- An embodiment of the method for transforming the metabolite of the agent back to the agent includes oral, intra-duodenal or rectal administration of the enzyme.
- the systemic concentration or exposure to the therapeutic agent is increased.
- an enzyme such as beta- glucuronidase will allow the adminstration of a lower dose of the agent, thus for example, mitigating potential toxicities were a higher dose to be administered.
- agents shown to be therapeutically effective only at doses, which exceed recommendation, at which patients are at risk because serious adverse effects have been demonstrated at such doses may now be made available at lower, safer doses when coadministered with an enzyme according to an embodiment of the present invention.
- administering refers to the application of a therapeutic agent or to application of an enzyme.
- coadministering refers to the application of an enzyme or in combination with an enzyme.
- Such application includes administering an effective amount of said agent with one or more enzymes separately at different times during the course of a therapy or concurrently in a combination form, such as in a combination product.
- coadministering further includes therapeutically or prophylactically administering an effective amount of a combination product comprising a therapeutic agent and an enzyme, wherein the agent and enzyme may be administered at different times during the course of a therapy or concurrently in a combination form.
- An example of the administration of the combination product includes and is not limited to the separate delivery of either the agent or the enzyme.
- the scope of the method for use described herein may only involve the step of delivery of an enzyme formulation or enzyme-inducing agent, whereby the delivery of the enzyme transforms the metabolite of the hormone back to the hormone.
- An advantage of the method of the present invention includes greater specificity in modulating the metabolism of a therapeutic agent.
- the use of the present invention is not limited to naturally occurring endogenous or exogenous enzymes, it also includes the use of bioengineered enzymes.
- the scope of the combination product and method for use thereof described herein includes, without limitation, a combination of chemistry, delivery and enzymatic approaches to modulate the levels of therapeutic agent and metabolites.
- delivery modes in some cases, may involve separate delivery of enzyme system and drug and in the cases of endogenous hormones may only involve the delivery of an enzyme formulation or inducing agent.
- Such various delivery modes would be expected by one skilled in the art to achieve the aim of the invention and are, thus included without limitation.
- enzyme refers to a protein molecule that catalyzes chemical reactions of other substances without it being destroyed or substantially altered upon completion of the reactions.
- the scope of the term includes, without limitation, endogenous and exogenous enzymes.
- the term further includes naturally occurring enzymes and bioengineered enzymes or mixtures thereof.
- Suitable enzymes for administration include, without limitation, cytochrome P450, glucuronyl transferase, sulfontransferase, carboxyl esterase, ⁇ -glucuronidase, soluble epoxide hydrolase, 11- ⁇ -hydroxysteroid dehydrogenases, aldehyde oxidase, xanthine oxidase and the like.
- Bioengineered enzymes including bioengineered enzymes that have altered activity compared to the naturally occurring enzymes, such as those generated using molecular biology techniques, may also be suitable and are included in the scope of the invention.
- enzyme inducing agent refers to an agent for use in converting an inactive form of an enzyme to one possessing metabolic activity.
- agents include, without limitation, activator ions, cofactors, coenzymes, proenzymes, zymogens (enzyme precursors converted to an active enzyme) and the like.
- subject refers to a patient, such as an animal, a mammal or a human, who has been the object of treatment, observation or experiment.
- therapeutically effective amount refers to that amount of a therapeutic agent that elicits the biological or medicinal response in an animal or human, that is being sought by a researcher, veterinarian, medical doctor, or other clinician, which includes alleviation of the symptoms of the disease, disorder or condition being treated.
- Constitutively effective amount refers to that amount of an enzyme that is required to maintain a therapeutically effective amount of the agent, whereby the metabolite of the agent is transformed back to the agent.
- the constitutively effective amount of enzyme includes, and is not limited to, that amount of enzyme that maximizes the extent of in vivo conversion of a metabolite of the therapeutic agent to a "de- metabolized" therapeutic agent.
- the stoichiometric ratio of enzyme to agent may range from a catalytic amount to an excess amount, as estimated by a ratio of an amount of agent converted (mmoles converted/minute/kg) based on enzyme units dosed relative to an amount of agent dosed (mmoles/kg).
- enzyme unit refers to unit for the amount of a particular enzyme. One U is defined as that amount of the enzyme that catalyzes the conversion of 1 micro mole of substrate per minute based on specific conversion conditions, such as at a temperature of 30 0 C and a pH and substrate concentration that yields the maximal substrate conversion rate.
- An embodiment of the present invention includes, an amount of ⁇ - glucuronidase in a range of from about 0.1 to about 10,000 enzyme units dosed relative to an amount of agent dosed; or, a range of from about 1.0 to about 2,000 units; or, a range of from about 10 to about 2,000 units; or, a range of from about 100 to about 2,000 units; or, a range of from about 1.0 to about 1,000 units; or, a range of from about 10 to about 1,000 units; or, a range of from about 100 to about 1,000 units.
- the amount of the enzyme administered will allow administration of a therapeutic agent below a toxic level while maintaining the therapeutic window of the agent such that the therapeutically effective amount is maintained throughout the period of absorption.
- an agent that may normally have a small therapeutic window can be given at a lower dose while maintaining therapeutic effectiveness.
- the availability of certain therapeutic agents previously discarded due to an inability to administer same at an other than toxic dose may now be administered using the present invention.
- therapeutic window refers to the dose range of an agent or of its concentration in a bodily system that provides safe, effective therapy.
- combination product refers to the use of a combination product comprising a therapeutic agent and an enzyme transforming the metabolite of the therapeutic agent back into the therapeutic agent.
- metabolite refers to the intermediate or products of a metabolic pathway in the body.
- the term "enterohepatic recirculation" is the process whereby a substance re- enters the gastrointestinal tract via the bile either in an unchanged form or after having been metabolized, e.g. being conjugated.
- the therapeutically effective amount of a therapeutic agent according to the present invention may be a reduced amount of the therapeutic agent compared to the effective amount of the agent otherwise recommended for treating a disease, disorder or condition. It is contemplated that the agent may be administered to a subject before, during or after the time the enzyme is administered.
- therapeutic agent refers to an agent or a combination of agents used to treat a disease, disorder or condition and includes, without limitation, facilitating the eradication of, inhibiting the progression of or promoting stasis of a disease, disorder or condition and includes, without limitation, pro-drugs that are cleaved in situ by a co-administered enzyme.
- the present invention includes a pharmaceutical composition comprising an admixture of a therapeutic agent and one or more pharmaceutically acceptable excipients for coadministration with an enzyme, wherein the enzyme is used for transforming the metabolite of the therapeutic agent back into the therapeutic agent, as a combination product to a subject in need thereof.
- the present invention includes a process for making a pharmaceutical composition of a therapeutic agent and an optional pharmaceutically acceptable carrier for coadministration with an enzyme.
- the present invention includes a pharmaceutical composition resulting from the process of mixing the agent and an optional pharmaceutically acceptable carrier.
- Contemplated processes include both conventional and unconventional pharmaceutical techniques.
- the present invention includes a product containing (a) a pharmaceutical composition containing a therapeutically effective amount of a therapeutic agent and (b) a pharmaceutical composition containing a constitutively effective amount of an enzyme as a combined preparation for simultaneous, separate or sequential use.
- An embodiment of the present invention includes a product containing (a) a pharmaceutical composition containing a therapeutically effective amount of a therapeutic agent that is subject to glucuronidation and (b) a pharmaceutical composition containing a constitutively effective amount of ⁇ -glucuronidase as a combined preparation for simultaneous, separate or sequential use.
- An embodiment of the present invention includes an enzyme selected from the group consisting of N-oxide reductase, sulfoxide reductase, esterase, amidase, glucosidase, ⁇ -glucoronidase and sulfatase.
- Another embodiment of the present invention includes the enzyme ⁇ -glucoronidase.
- An embodiment of the present invention includes a product containing (a) a pharmaceutical composition containing a therapeutically effective amount of a therapeutic agent that is subject to glucuronidation selected from the group consisting of ezetimibe, irrinotecan, tipifarnib, acetominophen, morphine, codeine, hydromorphone, testosterone, dihydrotestosterone, estradiol, 17-alpha-ethynylestradiol, estriol, tamoxifen, raloxifme, ciprofloxacin, valproic acid, GW273293, zidovudine, troglitazone, muraglitazar, muraglitzar, tolcapone, nicotine and (6,7-dimethoxy-2,4- dihydro-indeno[l,2-c]pyrazol-3-yl)-(3-fluoro-phenyl)-amine and (b) a pharmaceutical composition containing a constitutively effective amount of ⁇ -glucuronidase
- An embodiment of the present invention includes a product containing (a) a pharmaceutical composition containing a therapeutically effective amount of (6,7- dimethoxy-2,4-dihydro-indeno[l,2-c]pyrazol-3-yl)-(3-fluoro-phenyl)-amine and (b) a pharmaceutical composition containing a constitutively effective amount of ⁇ -glucuronidase as a combined preparation for simultaneous, separate or sequential use.
- Said pharmaceutical composition may take a wide variety of forms to effectuate mode of administration, wherein the mode includes, and is not limited to, site specific administration in the gastrointestinal tract (e.g. through a duodenal or colonic catheter) or in an enteric coated capsule or a controlled release delivery system, would be used to bypass the stomach and deliver both the agent and an enzyme.
- the composition may be in a dosage unit such as a tablet, pill, capsule, powder, granule, sterile solution, suspension or liposomal delivery system for a plurality of administration modes.
- compositions suitable for oral administration include solid forms such as pills, tablets, caplets, capsules (each including immediate release, timed release and sustained release formulations), granules and powders; and, liquid forms such as solutions, syrups, elixirs, emulsions and suspensions.
- forms useful for parenteral administration include sterile solutions, emulsions and suspensions.
- Formulations useful for liposomal delivery for either the enzyme or the therapeutic agent are known to those skilled in the art and include those employed to stabilize the enzymes, making them amenable to oral delivery such as, without limitation, enteric coated or controlled release formulations, conjugation to polymers or peptides, encapsulation in lipid-based carriers and the like.
- the pharmaceutical composition may be presented in a form suitable for once-weekly or once-monthly administration at various times using techniques known to those skilled in the art.
- the dosage form contains an effective amount of the therapeutic agent necessary to be therapeutically or prophylactically effective as described above.
- the pharmaceutical composition may contain from about 0.001 mg to about 1000 mg, preferably from about 0.001 to about 500 mg, of the therapeutic agent and may be constituted into any form suitable for the mode of administration selected for the subject in need.
- An example of a contemplated effective amount of the therapeutic agent for a pharmaceutical composition containing the therapeutic agent of the present invention may range from about 0.001 mg to about 300 mg/kg of body weight per day. In another example, the range is from about 0.003 to about 100 mg/kg of body weight per day. In another example, the range is from about 0.005 to about 15 mg/kg of body weight per day.
- the pharmaceutical composition, medicine or medicament containing the therapeutic agent may be administered according to a dosage regimen of from about 1 to about 5 times per day.
- An example of a contemplated effective constitutive amount of the enzyme for a pharmaceutical composition containing the enzyme of the present invention may be in a range of from about 0.1 to about 10,000 enzyme units dosed relative to an amount of agent dosed; or, a range of from about 1.0 to about 2,000 units; or, a range of from about 10 to about 2,000 units; or, a range of from about 100 to about 2,000 units; or, a range of from about 1.0 to about 1,000 units; or, a range of from about 10 to about 1,000 units; or, a range of from about 100 to about 1,000 units.
- the pharmaceutical composition, medicine or medicament containing the enzyme may be administered according to a dosage regimen of from about 1 to about 5 times per day.
- the pharmaceutical composition may also contain the above effective amounts of both the therapeutic agent and the enzyme.
- the pharmaceutical composition is preferably in the form of a tablet or capsule containing, e.g., 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 150, 200, 250, 500, and 1000 milligrams of the therapeutic agent for the symptomatic adjustment of the dosage to the patient to be treated.
- Optimal dosages will vary depending on factors associated with the particular patient being treated (e.g., age, weight, diet and time of administration), the severity of the condition being treated, the particular compound being used, the mode of administration and the strength of the preparation. The use of either daily administration or post-periodic dosing may be employed.
- a catheterized rat model was used in which the agent alone or agent and ⁇ -glucuronidase enzyme was administered intraduodenally or intracolonically.
- the intraduodenal and intraco Ionic dosing was intended to prevent the potential for loss of enzymatic activity when orally dosing the ⁇ -glucuronidase (e.g. by degradation in the stomach).
- the C max of the agent without enzyme present was 75 + 30 ng/ml (as shown in Table 8) and increased to a C max of 129 + 41 ng/ml with enzyme present (as shown in Table 7).
- the primary glucuronide metabolite AUC increased from 1216 + 371 (ng.hr)/ml with no enzyme present to an AUC of 8940 + 2144 (ng.hr)/ml with enzyme present.
- the resulting pharmacokinetic data suggested that when the agent and its primary glucuronide (i.e. metabolite) were secreted into the duodenum by the bile duct and were exposed to intraduodenally administered ⁇ -glucuronidase in the gut lumen, the glucuronide was cleaved back to the agent, thus allowing the agent to be reabsorbed.
- the agent used as the test compound (6,7-dimethoxy-2,4-dihydro-indeno[l,2- c]pyrazol-3-yl)-(3-fluoro-phenyl)-amine (hereinafter referred to as Compound 1) and two of its glucuronide metabolites, GIuA and GIuB, were tested.
- GIu A was found to be relatively unstable in DMSO, especially after freezing and thawing, therefore, GIu A was prepared fresh.
- Oral Jugular and PEG-400 75 7.5 10 Gavage Portal Vein
- Compound 1 was prepared in neat PEG-400.
- Compound 1 was added to neat Tween 80, the mixture was sonicated for thirty minutes at 45°C until Compound 1 was completely dissolved and then deionized (DI) water was added to form a solution consisting of 20% Tween 80 and 80% DI water by weight.
- DI deionized
- Compound 1 was prepared by addition to DI water containing 20% hydroxypropyl ⁇ -cyclodextran by weight.
- ⁇ -glucuronidase was added to the final formulation at a concentration of 57,600 units/ml.
- mice Male Sprague Dawley rats weighing 300-400 g were purchased from Charles River Labs. All rat experiments were conducted under approved animal protocols. The rats were catheterized at Charles River Labs and had one or more of the following chronic catheters for dosing and sampling: jugular/carotid, duodenal, and colonic. Rats were limited to only one GI catheter and one vascular catheter. For IV dosing, rats were purchased with jugular and carotid catheters for dosing in the jugular catheter and blood sampling through the carotid catheter.
- rats had one of the GI catheters for dosing and a carotid catheter for blood sampling. The surgeries were done two weeks prior to start of testing. Rats were fed 4 hours post dose administration. All rats were dosed without anesthesia and sacrificed at the end of the study.
- the two glucuronide conjugates were prepared and purified to permit accurate quantification of the levels of intact parent Compound 1 versus Cpd 1-GluA and Cpd 1-GluB.
- Each standard curve for Compound 1, Cpd 1-GluA, and Cpd 1-GluB generated from rat plasma included ten concentration levels.
- the lowest limit of quantification for each sample of Compound 1, Cpd 1-GluA, and Cpd 1-GluB was 1.0 ng/ml, with linearity demonstrated to 1000 ng/ml.
- the quantitative procedure for each sample included: sample preparation, HPLC separation and mass spectrometry detection.
- the rat plasma was prepared using protein precipitation techniques. Reference standards and quality control samples in rat plasma for Compound 1 were transferred to a 96 deep-well plate. The precipitation solution contained 100 ng/ml of internal standard in acetonitrile. 300 ⁇ l of precipitation solution were added to 100 ⁇ l of each standard, quality control and plasma sample. Samples were vortexed for 10 mins, then centrifuged at 4500 RPM for 10 mins. 10 ⁇ l of each sample were injected for LC/MS/MS analysis.
- the HPLC column chosen for the bioanalysis was purchased from Thermo Electron (Hypersil BetaBasic C 18, 5cm X 2.1mm).
- the eluting solvents were composed of Solvent A: 0.1% formic acid in H 2 O and Solvent B: 0.1% formic acid in acetonitrile.
- a step gradient was used and flow rate was 300 ⁇ l/min.
- the total run time was 3.2 mins.
- Solvent B was increased from 35% at 0 min to 75% at 2.1 mins and decreased back to 35% at 2.2 mins.
- the retention times for Compound 1, phenacetin, Cpd 1-GluA and Cpd 1-GluB were 1.1 min, 0.9 min, 1.3 min, and 1.6 min, respectively.
- API3000 with a Turbo Ionspray interface from Applied Biosystems was used for the detection of Compound 1, the internal standard and major metabolites. Electrospray ionization was performed in the positive ion mode heated nebulizer with a temperature of 300 0 C. LC/MS/MS chromatograms were acquired in MRM (multiple reaction mode). The MRM transitions for Compound 1, the internal standard, Cpd 1-GluA and Cpd 1-GluB were from 326.3 to 190.6, 181.1 to 110.3, 502.2 to 368.3 and 502.2 to 326.4, respectively. Pharmacokinetic Data Analysis
- the intestinal absorption ratio (F a ) was calculated by Equation (2): 7 7 _ / O (AUCjugular - AUCportal) P a — L - 2 ⁇ Jb Un vp
- Equation (2) Qb is the blood flow and was estimated to be 15.3 mL/min per body weight (250 g) by measurement with a flowmeter.
- Compound 1 has a MW of 325.35 g/mole and is poorly soluble in aqueous media at pH 7.0 ( ⁇ 0.002 mg/mL) and pH 3.0 ( ⁇ 0.006 mg/mL).
- a solubility of 7.5 mg/mL was achieved in neat PEG-400 for the oral gavage study.
- Improved solubility of 10 mg/mL for in vivo dosing studies was achieved by formulating Compound 1 in a solution containing 20/80-weight% Tween 80/DI water and with the exception of the IV dosing.
- Compound 1 exhibited high absorption (13 XlO "6 cm/sec).
- the cyclic increase and decrease in the portal vein concentration of Compound 1 between 0.5 and 4 hours are consistent with enterohepatic circulation of the glucuronide conjugates, conversion of the conjugates back to Compound 1, followed by re-absorption across the gut wall.
- the values represented in each table for the presence of compound and conjugates are the mean (rounded up to the first decimal place) for a treatment group of 4 animals.
- Tables 2, 3 and 4 show a summary of the PK parameters following IV and oral gavage dosing of Compound 1 at 2 and 75 mg/kg, respectively.
- GIu A and B were present in plasma, with GIu A being the predominant form having almost 66 times higher systemic exposure than GIu B following oral gavage dosing.
- Plasma levels of Compound 1 are 2% and 6% the level of the metabolites following IV and oral administration respectively, showing that Compound 1 undergoes extensive glucuronidation, irrespective of the route of administration.
- the local absorption ratio (Fa) i.e. fraction of Compound 1 absorbed in an intact form from the intestinal tract into the portal system
- the systemic oral bioavailability of Compound 1 following oral administration of Compound 1 at 75 mg/kg was estimated using Equation (2) to be 19 ⁇ 5 %.
- the calculated hepatic recovery ratio (Fh) using Equation (3) was 40 %.
- the mean ratio of GIu A to the Compound 1 was 20, while the ratio of GIu B to the Compound 1 was only 0.2.
- the magnitude of these values agreed with the ratios of metabolites in the jugular vein, where the ratio of GIu A to Compound 1 was 78 and the ratio of GIu B to Compound 1 was 1.
- the results have demonstrated that the therapeutic effectiveness of a therapeutic agent can be boosted by such recirculation.
- pH 7.4 buffer is representative of the in vivo pH range in the GI tract.
- Tables 5 (pH 5.0) and Table 6 (pH 7.4) The results of each treatment are shown in Tables 5 (pH 5.0) and Table 6 (pH 7.4).
- GIu A was prepared fresh in these experiments.
- a time-dependent incubation of GIu A and GIu B was carried out for 2, 4 and 6 hours.
- GIu-A 100% GIu-A 60% GIu-A 34% GIu-A 0% GIu-A
- GIu-A 100% GIu-A 67% GIu-A 48% GIu-A 23% GIu-A
- GIu-A was hydrolyzed back to the Compound 1 following incubation with ⁇ -glucuronidase in a time-dependent manner at both pH 5.0 and pH 7.4. There was 100% conversion to Compound 1 by 18 hours of incubation at pH 5.0 and 77% conversion at 6 hours at pH 7.4. Comparatively, the conversion of GIu A to Compound 1 is less rapid at pH 7.4 than at pH 5.0.
- GIu-B was not converted at either pH (100% GIu B remaining) after 6 or 18 hours of incubation.
- both GIu-A and GIu-B were stable and Compound 1 was not observed. ///. Pharmacokinetics Following In Vivo Co-administration of ⁇ -glucuronidase
- Duodenal administration of ⁇ -glucuronidase demonstrated a statistically significant increase ("boosting") in the levels of Compound 1, GIu A and GIu B.
- boosting a statistically significant increase in the levels of Compound 1, GIu A and GIu B.
- At a dose of 10 mg/kg there was a 2.9 fold increase in the Compound 1 AUC from 60 + 18 ng.hr/ml (as shown in Table 8) with no enzyme present to 177 ⁇ 55 with enzyme present (p ⁇ 0.005) (as shown in Table 7).
- the C max of Compound 1 increased from 75 ⁇ 30 ng/ml (as shown in Table 8) with no enzyme present to 129 ⁇ 41 ng/ml with enzyme present (as shown in Table 7).
- the T max of Compound 1 at 30 minutes remained unchanged.
- the AUC of GIu A increased by more than a factor of 7 from 1216 ⁇ 371 ng.hr/ml with no enzyme present to 8940 ⁇ 2144 ng.hr/ml with enzyme present (p ⁇ 0.005).
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Immunology (AREA)
- Gastroenterology & Hepatology (AREA)
- Zoology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Description
Claims
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US81522306P | 2006-06-20 | 2006-06-20 | |
PCT/US2007/071510 WO2007149827A2 (en) | 2006-06-20 | 2007-06-19 | Method for modulating the pharmacokinetics and metabolism of a therapeutic agent |
Publications (2)
Publication Number | Publication Date |
---|---|
EP2037954A2 true EP2037954A2 (en) | 2009-03-25 |
EP2037954A4 EP2037954A4 (en) | 2011-09-14 |
Family
ID=38834295
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP07812192A Withdrawn EP2037954A4 (en) | 2006-06-20 | 2007-06-19 | Method for modulating the pharmacokinetics and metabolism of a therapeutic agent |
Country Status (2)
Country | Link |
---|---|
EP (1) | EP2037954A4 (en) |
WO (1) | WO2007149827A2 (en) |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN102097026B (en) * | 2009-12-14 | 2013-04-10 | 中国科学院上海药物研究所 | Device and method for simulating pharmacokinetics characteristics in vitro |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5279824A (en) * | 1988-04-14 | 1994-01-18 | Merck Patent Gmbh | Pharmaceutical formulation containing heparin and endo-beta-glucuronidase, useful for the treatment of thrombosis |
US20040071685A1 (en) * | 2002-10-09 | 2004-04-15 | Devin Houston | Compositions and methods for increasing the bioavailability of plant polyphenols |
US20050260186A1 (en) * | 2003-03-05 | 2005-11-24 | Halozyme, Inc. | Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases |
WO2006042056A2 (en) * | 2004-10-08 | 2006-04-20 | The Ohio State University | Methods and compositions for enzyme-specific activation of carbohydrate-conjugated prodrugs |
Family Cites Families (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
DE4236237A1 (en) * | 1992-10-27 | 1994-04-28 | Behringwerke Ag | Prodrugs, their preparation and use as medicines |
EP0648503B1 (en) * | 1993-09-22 | 2000-07-26 | Hoechst Aktiengesellschaft | Pro-prodrugs, their production and use |
JP3805365B2 (en) * | 1994-12-23 | 2006-08-02 | シンジェンタ リミテッド | Compound |
AU2003234567A1 (en) * | 2002-05-15 | 2003-12-02 | Janssen Pharmaceutica N.V. | N-substituted tricyclic 3-aminopyrazoles as pdfg receptor inhibitors |
CA2529027C (en) * | 2003-06-13 | 2013-09-10 | Immunomedics, Inc. | D-amino acid peptides |
-
2007
- 2007-06-19 EP EP07812192A patent/EP2037954A4/en not_active Withdrawn
- 2007-06-19 WO PCT/US2007/071510 patent/WO2007149827A2/en active Application Filing
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5279824A (en) * | 1988-04-14 | 1994-01-18 | Merck Patent Gmbh | Pharmaceutical formulation containing heparin and endo-beta-glucuronidase, useful for the treatment of thrombosis |
US20040071685A1 (en) * | 2002-10-09 | 2004-04-15 | Devin Houston | Compositions and methods for increasing the bioavailability of plant polyphenols |
US20050260186A1 (en) * | 2003-03-05 | 2005-11-24 | Halozyme, Inc. | Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases |
WO2006042056A2 (en) * | 2004-10-08 | 2006-04-20 | The Ohio State University | Methods and compositions for enzyme-specific activation of carbohydrate-conjugated prodrugs |
Non-Patent Citations (3)
Title |
---|
HOUBA P H ET AL: "Pronounced antitumor efficacy of doxorubicin when given as the prodrug DOX-GA3 in combination with a monoclonal antibody beta-glucuronidase conjugate.", INTERNATIONAL JOURNAL OF CANCER. JOURNAL INTERNATIONAL DU CANCER 15 FEB 2001 LNKD- PUBMED:11251980, vol. 91, no. 4, 15 February 2001 (2001-02-15), pages 550-554, XP002655272, ISSN: 0020-7136 * |
See also references of WO2007149827A2 * |
YAN Z ET AL: "N-glucuronidation of the platelet-derived growth factor receptor tyrosine kinase inhibitor 6,7-(dimethoxy-2,4-dihydroindeno[1,2-c]pyr a zol-3-yl)-(3-fluoro-phenyl)-amine by human UDP-glucuronosyltransferases", DRUG METABOLISM AND DISPOSITION, vol. 34, no. 5, May 2006 (2006-05), pages 748-755, XP002655273, ISSN: 0090-9556 * |
Also Published As
Publication number | Publication date |
---|---|
EP2037954A4 (en) | 2011-09-14 |
WO2007149827A3 (en) | 2009-04-23 |
WO2007149827A2 (en) | 2007-12-27 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Liu et al. | Absorption and metabolism of flavonoids in the caco-2 cell culture model and a perused rat intestinal model | |
US9415034B2 (en) | Inhibitors and enhancers of uridine diphosphate-glucuronosyltransferase 2B (UGT2B) | |
JP5133064B2 (en) | Inhibitors and promoters of uridine diphosphate (UDP) -glucuronyltransferase 2B (UGT2B) | |
JP2002537343A (en) | Multi-particle formulation | |
JP2002538173A (en) | Use of gallic esters to increase the bioavailability of orally administered drug compounds | |
McMillan | Leukotrienes in respiratory disease | |
JP2023506208A (en) | Tissue-catalyzed growth of polymers as epithelial linings for therapeutics | |
JP2019048848A (en) | Methods for treating GI tract disorders | |
US20230046803A1 (en) | Combination therapies comprising targeted therapeutics | |
US5567432A (en) | Masking of liposomes from RES recognition | |
US20060040875A1 (en) | Inhibitors and enhancers of uridine diphosphate-glucuronosyltransferase 2B (UGT2B) | |
EP2037954A2 (en) | Method for modulating the pharmacokinetics and metabolism of a therapeutic agent | |
EP3995480A1 (en) | Akr1c3 inhibitor and medical use | |
US20200239472A1 (en) | Targeted therapeutics | |
US20080031866A1 (en) | Method for modulating the pharmacokinetics and metabolism of a therapeutic agent | |
EP1625854A1 (en) | Albumin conjugates containing a glucuronic linker | |
US9566341B1 (en) | Compounds including Cox inhibitor moiety and enhanced delivery of active drugs using same | |
WO2022178821A1 (en) | Use of akr1c3-activated compound | |
EP1142589A1 (en) | Medicinal preparations | |
Bodd et al. | Oral administration of codeine in the presence of ethanol: a pharmacokinetic study in man | |
US20240156832A1 (en) | Methods of metabolizing metopimazine and its salts | |
JP5766121B2 (en) | Delivery system for delivery of substances into the oral cavity | |
JPH1121250A (en) | Pharmaceutical preparation | |
JP2003501384A (en) | Use of verapamil and verapamil derivatives for the manufacture of a medicament having a β-glucuronidase inhibitory action in human tissues | |
CN104140422A (en) | Positively charged water-soluble oxicam with fast skin penetration speed and prodrug of related compound of oxicam |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
17P | Request for examination filed |
Effective date: 20081219 |
|
AK | Designated contracting states |
Kind code of ref document: A2 Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR |
|
AX | Request for extension of the european patent |
Extension state: AL BA HR MK RS |
|
R17D | Deferred search report published (corrected) |
Effective date: 20090423 |
|
DAX | Request for extension of the european patent (deleted) | ||
RIC1 | Information provided on ipc code assigned before grant |
Ipc: A61K 38/46 20060101ALI20110804BHEP Ipc: A61K 38/44 20060101ALI20110804BHEP Ipc: A61K 38/43 20060101AFI20110804BHEP |
|
A4 | Supplementary search report drawn up and despatched |
Effective date: 20110816 |
|
17Q | First examination report despatched |
Effective date: 20130301 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN |
|
18D | Application deemed to be withdrawn |
Effective date: 20130712 |