METHODS AND COMPOSITIONS FOR ORAL DELIVERY OF FTS

Information

  • Patent Application
  • 20160271089
  • Publication Number
    20160271089
  • Date Filed
    June 01, 2016
    8 years ago
  • Date Published
    September 22, 2016
    7 years ago
Abstract
Disclosed are oral dosage forms containing a Ras antagonist including FTS and structural analogs thereof, and at least one pharmaceutically acceptable excipient other than a cyclodextrin, and methods of orally administering same to treat diseases and disorders responsive to the Ras antagonists.
Description
BACKGROUND OF THE INVENTION

U.S. Pat. No. 5,705,528 teaches farnesylthiosalicyclic acid (FTS) and analogs thereof and their utility as anti-cancer agents. U.S. Pat. No. 6,462,086 teaches additional therapeutic utilities of these compounds, namely in connection with treatment of non-malignant diseases, pathological states or other disorders that feature or otherwise include Ras-induced proliferation of cells. The patent also teaches that these compounds are inactive when administered orally, but that this shortcoming can be overcome by making a salt of the compound (i.e., salification), formulating the salt in cyclodextrin, and then preparing a buccal tablet (which will dissolve in the mouth when held against the mucous membrane).


SUMMARY OF THE INVENTION

Applicants have discovered that FTS and its analogs do not require salification or formulation in cyclodextrin to be active upon oral administration.


A first aspect of the present invention is directed to an oral dosage form comprising an amount of a Ras antagonist effective to treat a responsive disease or disorder involving abnormal cell proliferation, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula (I)




embedded image


wherein:


R1 represents farnesyl, geranyl or geranyl-geranyl;

    • R2 is COOR7, or CONR7R8, wherein R7 and R8 are each independently hydrogen, alkyl or alkenyl;


      R3, R4, R5 and R6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto; and


      X represents S. The oral dosage forms are tablets or capsules. In preferred embodiments, the Ras antagonist is not in the form of a salt.


A second aspect of the invention is directed to method of treating a treating a disease or disorder involving abnormal cell proliferation, comprising administering to a human subject in need thereof an oral dosage form comprising an amount of a Ras antagonist effective to treat the disease or disorder, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula




embedded image


wherein:


R1 represents farnesyl, geranyl or geranyl-geranyl;


R2 is COOR7, or CONR7R8, wherein R7 and R8 are each independently hydrogen, alkyl or alkenyl;


R3, R4, R5 and R6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto; and


X represents S.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a graph showing concentrations (ng/mL) of FTS in plasma of mice over time following oral (e.g., gavage) administration of FTS in a corn oil carrier.



FIG. 2 is a graph showing concentrations (ng/mL) of FTS in plasma of mice over time following oral (gavage) administration of FTS in aqueous carboxymethylcellulose (CMC) carrier.



FIG. 3 is a bar graph showing average tumor weight in mice (in grams) following oral (gavage) administration of different amounts of FTS in carboxymethylcellulose, as compared to a control.



FIG. 4 is a bar graph showing FTS plasma concentration (ng/ml) over 1-24 hours after oral administration of 200 mg/kg of FTS to rats.





DETAILED DESCRIPTION

Ras antagonists useful in the present invention are represented by formula I:




embedded image


wherein


R1 represents farnesyl, geranyl or geranyl-geranyl;


R2 is COOR7, or CONR7R8, wherein R7 and R8 are each independently hydrogen, alkyl or alkenyl;


R3, R4, R5 and R6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto; and


X represents S.


These compounds represent farnesyl-thiosalicylic acid (FTS) (i.e., S-trans, trans-FTS) and its analogs. The structure of FTS is as follows:




embedded image


FTS analogs embraced by formula I include 5-fluoro-FTS, 5-chloro-FTS, 4-chloro-FTS and S-farnesyl-thiosalicylic acid methyl ester (FTSME). Structures of these compounds are set forth below.




embedded image


Pharmaceutically acceptable salts of the Ras antagonists of formula I may be useful. In preferred embodiments, however, the Ras antagonist is not in the form of a salt (i.e., non-salified).


Methods of preparing the compounds of formula I are disclosed in U.S. Pat. Nos. 5,705,528 and 6,462,086. Those disclosures (e.g., Example 7 in the '086 patent) are incorporated herein by reference.


Oral dosage forms useful in the present invention include tablets (e.g., including film-coated, sugar-coated, controlled or sustained release), and capsules, e.g., hard gelatin capsules (including controlled or sustained release), and soft gelatin capsules.


Oral dosage forms may be prepared by mixing the active pharmaceutical ingredient, which in this case are the Ras antagonists of formula I, with one or more appropriate carriers (excipients), and then formulating the composition into the desired dosage form e.g., compressing the composition into a tablet or filling into a capsule. The proviso is that the oral dosage forms do not contain a cyclodextrin. Typical excipients useful as bulking agents or diluents, binders, buffers or pH adjusting agents, disintegrants (including crosslinked and super disintegrants such as croscarmellose), glidants, and/or lubricants include lactose, starch, mannitol, microcrystalline cellulose, ethyl cellulose, sodium carboxymethyl cellulose, hydroxypropylmethyl cellulose, dibasic calcium phosphate, acacia, gelatin, stearic acid, magnesium stearate, corn oil, vegetable oils, and polyethylene glycols, and others known to the pharmaceutical practitioner. Coating agents such as sugar, shellac, and synthetic polymers may be employed. Dyes and other colorants may be added as well. See, Remington's Pharmaceutical Sciences, The Science and Practice of Pharmacy, 20th Edition, (2000).


The oral dosage forms of the present invention are useful in treating diseases and disorders responsive to the Ras antagonists of formula I, e.g., diseases and disorders characterized or mediated, at least in part, by abnormal (e.g., uncontrolled) cell proliferation, such as Ras-induced cell proliferation. The proliferating cells may be malignant or non-malignant in nature. Of course, the term “responsive” does not require that a therapeutic response would be achieved in each and every patient, but rather what a skilled practitioner would reasonably expect based on existing data from patient populations. These diseases and disorders include cancers including breast cancer, colon cancer, glioblastoma, lung cancer (small cell and non-small cell lung cancer), melanoma, Merkel cell carcinoma, neuroblastoma, neurofibromatosis, ovarian cancer, pancreatic cancer and prostate cancer.


Examples of non-malignant diseases and disorders characterized by or involving abnormal (e.g., uncontrolled) cell proliferation include liver cirrhosis, restenosis after angioplasty (post-angioplasty restenosis), atherosclerosis, and graft rejection (e.g., graft-versus-host disease (GVHD)). Other examples include autoimmune diseases such as type-1 diabetes mellitus, systemic lupus erythematosis, rheumatoid arthritis, psoriasis, multiple sclerosis, Guillain-Barre syndrome and primary antiphospholipid syndrome.


As used herein, the term “effective amount” refers to a dosage of the Ras antagonist that inhibits disease onset or progression or ameliorates symptom(s) of the disease. The typical daily dose is 5 mg to 1000 mg, e.g., 100 mg to 1,000 mg per day, which may be administered once daily or as divided doses two or three times daily. Thus, the oral dosage forms of the present invention e.g., tablet, hard gelatin capsule, or soft gelatin capsule, typically contains from 5 mg to 500 mg of the Ras antagonist. In preferred embodiments, the oral dosage forms contain from 10 mg to 250 mg of active pharmaceutical ingredient. The oral dosage forms are typically administered to an individual diagnosed with the disease or disorder, although prophylactic administration may be useful to inhibit onset of a disease or disorder e.g., administration prior to an angioplasty procedure to inhibit onset of restenosis.


The oral dosage forms may be suitably packaged for distribution and sale, including printed instructions for administering them as described herein.


The present invention will now be described by way of the following non-limiting examples. They are presented solely for purposes of illustration, and are not intended to limit the invention in any way.


Example 1
Bioavailability of Orally Administered FTS

a. FTS Formulated in Corn Oil.


FTS (40 mg) was dispersed in corn oil (10 ml).


Aliquots of the resulting suspension were administered by gavage to CD-1 mice (eighteen) at a dose of 40 mg/ml (at a volume of 10 ml/kg of FTS suspension calculated for actual animal weight). Blood samples were collected in lithium heparin tubes via the retro-orbital plexus while under CO2/O2 anesthesia at 1, 2, 4, 8, 12 and 24 hours (3 animals at each time point) after dosing. Aliquots (100 μL) of plasma were mixed with water, pH 11 buffer solution and acetonitrile. After centrifugation, the supernatant was evaporated to dryness. The residue was resuspended in water/acetonitrile and centrifuged, and the supernatant was transferred to auto-sampler vials for FTS determination by liquid chromatography/mass spectrometry (LC/MS).


As shown in FIG. 1, mean plasma FTS concentrations were 2159 ng/ml at 1 hour and 1326 ng/ml after 2 hours. There was a rapid decrease to less than 3 ng/ml after 24 hours. These values indicated 55% oral bioavailabilty compared to intra-peritoneal injection of FTS in a control group of animals.


b. FTS Formulated in 0.5% Aqueous Carboxymethylcellulose.


FTS (40 mg) was dispersed in 0.5% aqueous carboxymethylcellulose (10 ml). Aliquots of the resulting suspension were administered by gavage to CD-1 mice (eighteen) at a dose of 40 mg/ml (at a volume of 10 ml/kg of FTS suspension calculated for actual animal weight). Blood samples were collected in lithium heparin tubes via the retro-orbital plexus while under CO2/O2 anesthesia at 1, 2, 4, 8, 12, and 24 hours (3 animals at each time point) after dosing. Aliquots (100 μL) of plasma were mixed with water, pH 11 buffer solution, and acetonitrile. After centrifugation, the supernatant was evaporated to dryness. The residue was re-suspended in water/acetonitrile and centrifuged, and the supernatant was transferred to auto-sampler vials for FTS determination by LC/MS.


As shown in FIG. 2, mean plasma FTS concentrations were 3381 ng/ml at 1 hour, 1593 ng/ml after 2 hours, and decreased rapidly to less than 3 ng/ml after 24 hours. These values indicated 69% oral bioavailabilty compared to intra-peritoneal injection of FTS in a control group of animals.


These results, as illustrated in FIGS. 1 and 2, were unexpected, and in addition, show that therapeutic levels may be readily obtained upon oral administration.


Example 2
Inhibition of Pancreatic Tumor Growth with Orally

Administered FTS


Nude CD1-Nu Mice were implanted subcutaneously with 5.0×106 Panc-1 cells in 0.2 ml of PBS just above the right femoral joint. Eleven days after implantation, palpable tumors were observed.


FTS was dispersed in 0.5% aqueous carboxymethylcellulose at a concentration of 5 mg/ml. On the eleventh day after implantation, mice were segregated into three groups and were given vehicle or FTS suspension by gavage.


Group 1 (Controls, 6 mice) received 0.2 ml vehicle daily.


Group 2 (20 mg/kg FTS, 7 mice) received 0.1 ml FTS suspension daily.


Group 3 (40 mg/kg FTS, 6 mice) received 0.2 ml FTS suspension daily.


Mice were treated for ten days, and were sacrificed. Tumors were excised and weighed.


As shown in FIG. 3, average tumor weight for Group 1 (controls) was 1.02 g; average tumor weight for Group 2 (20 mg/kg FTS) was 0.68 g (32% lower than control); and average tumor weight for Group 3 (40 mg/kg FTS) was 0.47 g (53% lower than control).


Example 3
Compatibility of FTS with Pharmaceutical Excipients

Physical mixtures (1:1 w/w) of FTS and representative pharmaceutical excipients commonly used in oral formulations were stored at 40° C./75% RH accelerated stress conditions for four weeks in loosely capped vials. Samples were assayed by HPLC at the 0, 2-week and 4-week time points. The results are shown in the table below.













TABLE





Excipient
0 weeks
2 weeks
4 weeks
Observation







Lactose
97.94
97.23
98.09
Compatible


Dibasic calcium
97.95
95.75
97.08
Compatible


phosphate


Starch
97.88
97.30
98.11
Compatible


Hydroxypropylmethyl
97.93
98.06
98.12
Compatible


Cellulose


Microcrystalline
97.93
97.72
98.12
Compatible


Cellulose


Polyvinylpyrrolidone
97.71
92 .44
92.56
Potential






incompatibility


Croscarmellose
97.76
98.14
98.13
Compatible


Sodium


Sodium Carboxymethyl
97.76
98.12
98.17
Compatible


Cellulose


FTS Substance
97.75
97.10
97.76









Results of the accelerated stability evaluation demonstrated that many conventional pharmaceutical excipients may be employed in FTS tablets and capsules.


Example 4
FTS Tablets (200 mg)

FTS active pharmaceutical ingredient (2000 g), microcrystalline cellulose (2000 g), croscarmellose sodium (200 g), and magnesium stearate (100 g) are blended to uniformity and compressed into tablets weighing 430 mg. Assuming a 5% loss on material transfers and tablet press start-up, adjustment, and shut down, approximately 9,500 FTS 200 mg tablets are yielded.


Example 5
FTS Hard Gelatin Capsules (100 mg.)

FTS active pharmaceutical ingredient (1000 g), lactose (1000 g), microcrystalline cellulose (1000 g), and amorphous colloidal silicon dioxide (15 g) are blended to uniformity and filled into hard gelatin capsules. Assuming a 5% loss on material transfers and encapsulating machine start-up, adjustment, and shut down, approximately 9,500 FTS 100 mg capsules are yielded.


Example 6
FTS Soft Gelatin Capsules (50 mg)

FTS active pharmaceutical ingredient (500 g), is uniformly dispersed in a mixture of corn oil (3000 g), lecithin (30 g), and Tween-80 (150 g) and filled into soft gelatin capsules. Assuming a 5% loss on material transfers and soft gelatin encapsulating machine start-up, adjustment, and shut down, approximately 9,500 FTS 50 mg soft gelatin capsules are yielded.


Example 7
FTS Capsules (200 mg)

FTS active pharmaceutical ingredient (2841 g), microcrystalline cellulose (1421 g), starch (710 g), and magnesium stearate (29 g) are blended to uniformity and filled into size #0 hard-shell gelatin capsules, with a fill weight of 0.357 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 13,000 FTS 200 mg capsules are yielded.


Example 8
FTS Capsules (200 mg)

FTS active pharmaceutical ingredient (1000 g) and microcrystalline cellulose (125 g) are blended to uniformity and filled into size #1 hard-shell gelatin capsules, with a fill weight of 0.225 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 4,750 FTS 200 mg capsules are yielded.


Example 9
FTS Capsules (100 mg)

FTS active pharmaceutical ingredient (1000 g) and microcrystalline cellulose (125 g) are blended to uniformity and filled into size #2 hard-shell gelatin capsules, with a fill weight of 0.112.5 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 9,500 FTS 100 mg capsules are yielded.


Example 10
FTS Capsules (50 mg)

FTS active pharmaceutical ingredient (1000 g) and microcrystalline cellulose (125 g) are blended to uniformity and filled into size #3 hard-shell gelatin capsules, with a fill weight of 0.056 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 18,000 FTS 50 mg capsules are yielded.


Example 11
Bioavailability of Orally Administered FTS to Rats

FTS was dissolved in a mixture of corn oil (95%) and ethanol (5%). Aliquots of the resulting solution were administered to Crl:CD(SD)(IGS)BR rats (twenty-four) at a dose of 200 mg/kg. Blood samples were collected in lithium-heparin-containing tubes via the retro-orbital plexus while under CO2/O2 anesthesia at 1, 2, 4, 8, 12 and 24 hours after dosing. Concentration of FTS at each time point was determined by LC/MS. As shown in FIG. 4, mean plasma FTS concentrations were 4454 ng/ml at 1 hour, 4550 ng/ml at 2 hours, 3047 ng/ml at 4 hours, 2216 ng/ml at 8 hours, 685 ng/ml at 12 hours, and 140 ng/ml at 24 hours. These values indicate a high level of oral bioavailability.


All patent and non-patent publications cited in this specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All these publications are herein incorporated by reference to the same extent as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference.


Although the invention herein has been described with reference to particular embodiments, it is to be understood that these embodiments are merely illustrative of the principles and applications of the present invention. It is therefore to be understood that numerous modifications may be made to the illustrative embodiments and that other arrangements may be devised without departing from the spirit and scope of the present invention as defined by the appended claims.

Claims
  • 1. An oral dosage form comprising an amount of a Ras antagonist effective to treat a disease or disorder involving abnormal cell proliferation, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula
  • 2. The oral dosage form of claim 1, wherein the Ras antagonist is farnesyl-thiosalicyclic acid (FTS).
  • 3. The oral dosage form of claim 1, wherein the Ras antagonist is 5-fluoro-FTS.
  • 4. The oral dosage form of claim 1, wherein the Ras antagonist is 5-chloro-FTS.
  • 5. The oral dosage form of claim 1, wherein the Ras antagonist is 4-chloro-FTS.
  • 6. The oral dosage form of claim 1, wherein the Ras antagonist is S-farnesyl-thiosalicylic acid methyl ester.
  • 7. The oral dosage form of claim 1, wherein the effective amount is about 5 mg to about 500 mg.
  • 8. The oral dosage form of claim 1, wherein the effective amount is about 10 mg to about 250 mg.
  • 9. The oral dosage form of claim 1, wherein said carrier comprises one or more of a bulking agent, binder, disintegrant, glidant or lubricant.
  • 10. The oral dosage form of claim 1, wherein said tablet has a coating on an outer surface thereof.
  • 11. The oral dosage form of claim 1, which is in the form of a soft or hard gelatin capsule.
  • 12. A method of treating a disease or disorder involving abnormal cell proliferation, comprising administering to a human in need thereof an oral dosage form comprising an amount of a Ras antagonist effective to treat the disease or disorder, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula I
  • 13. The method of claim 12, wherein the disease is cancer.
  • 14. The method of claim 13, wherein the cancer is breast cancer.
  • 15. The method of claim 13, wherein the cancer is pancreatic cancer.
  • 16. The method of claim 12, wherein the disease or disorder is cirrhosis of the liver.
  • 17. The method of claim 12, wherein the disease or disorder is post-angioplasty restenosis.
  • 18. The method of claim 17, wherein the oral dosage form is administered prophylactically.
  • 19. The method of claim 12, wherein the disease or disorder is atherosclerosis.
  • 20. The method of claim 12, wherein the disease or disorder is graft rejection.
  • 21. The method of claim 12, wherein the disease or disorder is an autoimmune disease.
CROSS-REFERENCE TO RELATED APPLICATIONS

The present application is a continuation of U.S. patent application Ser. No. 14/662,420, filed Mar. 19, 2005, which is a continuation of U.S. patent application Ser. No. 13/326,598, filed on Dec. 15, 2011, which application is a divisional of U.S. patent application Ser. No. 11/659,582, filed on Feb. 7, 2007, now U.S. Pat. No. 8,088,756 issued Jan. 2, 2012 which application is a national phase entry under 35 U.S.C. §371 of International Application No. PCT/US2005/029389, filed Aug. 18, 2005, which claims priority from U.S. Provisional Application No. 60/602,361, filed Aug. 18, 2004, all of which are incorporated herein by reference.

Provisional Applications (1)
Number Date Country
60602361 Aug 2004 US
Divisions (1)
Number Date Country
Parent 11659582 Feb 2007 US
Child 13326598 US
Continuations (2)
Number Date Country
Parent 14662420 Mar 2015 US
Child 15170416 US
Parent 13326598 Dec 2011 US
Child 14662420 US