US20110178138A1 - Inhibitors of protein prenyltransferases - Google Patents

Inhibitors of protein prenyltransferases Download PDF

Info

Publication number
US20110178138A1
US20110178138A1 US13/056,077 US200813056077A US2011178138A1 US 20110178138 A1 US20110178138 A1 US 20110178138A1 US 200813056077 A US200813056077 A US 200813056077A US 2011178138 A1 US2011178138 A1 US 2011178138A1
Authority
US
United States
Prior art keywords
compound
compounds
ggtase
group
alkyl
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.)
Abandoned
Application number
US13/056,077
Other languages
English (en)
Inventor
Ohyun Kwon
Fuyuhiko Tamanoi
Hanna Fiji
Masaru Watanabe
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 California
Original Assignee
University of California
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 California filed Critical University of California
Assigned to THE REGENTS OF THE UNIVERSITY OF CALIFORNIA reassignment THE REGENTS OF THE UNIVERSITY OF CALIFORNIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WATANABE, MASARU, KWON, OHYUN, FIJI, HANNAH, TAMANOI, FUYUHIKO
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF CALIFORNIA LOS ANGELES
Assigned to THE REGENTS OF THE UNIVERSITY OF CALIFORNIA reassignment THE REGENTS OF THE UNIVERSITY OF CALIFORNIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FIJI, HANNAH D.G., KWON, OHYUN, WATANABE, MASARU, TAMANOI, FUYUHIKO
Publication of US20110178138A1 publication Critical patent/US20110178138A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/92Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with a hetero atom directly attached to the ring nitrogen atom
    • C07D211/96Sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/20Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/46Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with hetero atoms directly attached to the ring nitrogen atom
    • C07D207/48Sulfur atoms

Definitions

  • Embodiments of the present invention were made with Government support of Grant Nos. CA32737, GM071779, and GM081282, awarded by the NIH. The U.S. Government may have certain rights in this invention.
  • Protein prenyltransferases such as protein geranylgeranyltransferase type I (GGTase-I) and Rab geranylgeranytransferase (RabGGTase), catalyze posttranslational modification of proteins, often involving the addition of isoprenoids (1-5).
  • protein farnesylation involves the addition of a C15 farnesyl group to proteins ending with the C-terminal CAAX motif (C is cysteine, A is an aliphatic amino acid and X is usually serine, methionine, glutamine, cysteine or alanine).
  • Farnesylated proteins include Ras proteins, Rheb proteins, nuclear lamins and Hdj2.
  • Protein geranylgeranylation involves the addition of a longer isoprenoid, C20 geranylgeranyl group. Protein geranylgeranylation is critical for the function of a number of proteins such as RhoA, Rac and Rab.
  • Rho family proteins such as RhoA, Cdc42 and Rac as well as the ⁇ -subunit of heterotrimeric G-proteins are geranylgeranylated at a cysteine within the CAAL motif (similar to the CAAX motif but the C-terminal amino acid is leucine or phenylalanine) at their C-termini.
  • Rab proteins involved in protein transport across the secretory pathway and endocytosis pathway are also geranylgeranylated. These proteins usually end with CC (two cysteines) or CXC at the C-termini and both cysteines are geranylgeranylated.
  • Geranylgeranyl transferase type I catalyzes mono geranylgeranylation of proteins such as Rho, Rac and Cdc42.
  • This enzyme is a heterodimer consisting of alpha and beta subunits (15).
  • RabGGTase or GGTase-II
  • GGTase-II catalyzes digeranylgeranylation of Rab proteins (16,17).
  • This enzyme also contains alpha and beta subunits, but contains an additional subunit Rab Escort Protein (REP) (16,18).
  • the REP subunit binds to the substrate Rab protein (19).
  • the alpha and beta subunits share homology with corresponding subunits of GGTase-I.
  • FTase protein farnesyltransferase
  • GGTase I protein geranylgeranyltransferase type I
  • FTase catalyzes the transfer of a C15 farnesyl group from farnesyl pyrophosphate, an intermediate in cholesterol biosynthesis, to proteins such as Ras, Rheb, nuclear lamins, CENP-E, F and protein tyrosine phosphatases pRL1-3. See Tamanoi, F., Gau, C. L., Edamatsu, H., Jiang, C. and Kato-Stankiewicz, J. (2001) Protein farnesylation in mammalian cells, Cell Mol. Life. Sci. 58, 1-14. These proteins end with the CaaX motif that is recognized by FTase.
  • GGTase I catalyzes the transfer of a C20 geranylgeranyl group from geranylgeranyl pyrophosphate to proteins ending with the CaaL motif.
  • Geranylgeranylated proteins include Rho, Rac, Cdc42 as well as gamma-subunit of heterotrimeric G-proteins.
  • Certain protein prenyltransferases have been implicated in cancer processes, including GGTase I, RabGGTase, and FTase. See Tamanoi, F., Gau, C. L., Edamatsu, H., Jiang, C. and Kato-Stankiewicz, J. (2001) Protein farnesylation in mammalian cells, Cell Mol. Life. Sci. 58, 1-14; Carrico, D., Blaskovich, M. A., Bucher, C. J., Sebti, S. M., Hamilton, A. D.
  • RhoA protein was recently found to be activated downstream of Ras in most pancreatic cancer cells harboring oncogenic K-ras mutation (7).
  • RalB plays critical roles in the survival pathway (8).
  • RhoC is overexpressed in metastatic cancer and RhoC knockout mice exhibit defect in metastasis (9,10).
  • Overexpression of Rab25 in breast and ovarian cancer cells has been reported, and this mutation is a determinant for aggressiveness of these cancers (11,12).
  • Rab25 is also upregulated in prostate cancer and transitional-cell bladder cancer (11). Overexpression of other Rab proteins such as Rab5a and Rab7 in cancer has been reported (13,14).
  • FTIs Farnesyltransferase inhibitors
  • Ras proteins are farnesylated and that farnesylation and membrane association of the Ras proteins is critical for their ability to transform cells.
  • GGTIs geranylgeranyltransferase type I inhibitors
  • Diversity-oriented synthesis of a chemical compound library provides a powerful means to identify small molecule inhibitors against medically relevant targets.
  • Initial screens of a pilot library followed by diversification using solid phase synthesis can yield potent inhibitors of enzymes in a relatively short period of time.
  • J is hydrogen or is 1-2 substituents independently selected from the group consisting of halogen, C 1 -C 3 alkyl, OR′, SR′, and NR′ 2 , where R′ is alkyl.
  • E is hydrogen or is 1-2 substituents selected from the group consisting of halogen, C 1 -C 3 alkyl, OR′, SR′, and NR′ 2 , where R′ is alkyl,
  • W is selected from the group consisting of hydrogen, cyclic, linear, or branched alkyl of from 2 to 8 carbons, unsubstituted phenyl, and phenyl substituted with C 1 -C 3 alkyl, halogen, OR′, SR', and NR' 2 , where R′ is alkyl,
  • A is selected from the group consisting of:
  • M is selected from the group consisting of OH, OR′′, NH 2 , NHOH, NHOR′′, wherein R′′ is methyl or ethyl, or any other group that has a polar metal binder
  • R corresponds to an alpha-substituent of natural or non-natural alpha-amino acid
  • Z is S-U
  • the compound inhibits the activity of a protein prenyltransferase.
  • the invention is also directed to pharmaceutical compositions comprising the compound of the invention and a pharmaceutically acceptable carrier or diluent.
  • Some embodiments of the invention are directed a method comprising administering the compound of the invention to a cell in an amount sufficient to inhibit the activity of GGTase I, RabGGTase, or both GGTase I and RabGGTase.
  • Some embodiments of the invention are directed to a method comprising administering a compound of the invention in an amount sufficient to inhibit the growth of a cancer cell.
  • the cancer can be, but is not limited, pancreatic, leukemia, breast, lung, colon, ovarian, stomach, and prostate cancer.
  • the cancer cell comprises GGTase I modified proteins and/or RabGGTase modified proteins.
  • Some embodiments of the invention are directed to a method comprising administering to a subject in need of treatment for a cancer a pharmaceutical composition of the present invention in an amount sufficient to inhibit the activity of a protein prenyltransferase.
  • Some embodiments of the invention are directed to a method comprising measuring the GGTase I and/or RabGGTase inhibiting activity of a compound of the invention.
  • FIG. 1 presents a pilot library of compounds.
  • FIG. 2 presents the structure of GGTIs identified from the pilot library.
  • FIG. 3 illustrates that GGTI effects are not influenced by the addition of detergents.
  • FIG. 4 presents the structure of FTIs identified from the pilot library.
  • FIG. 5 presents an illustration of synthetic steps.
  • the steps may include (a) allenoic acid, Mukaiyama's reagent, DIPEA or Et 3 N, DCM, room temperature, for 12 hours; (b) PPH 3 or PBu 3 , imine, benzene or DCM at room temperature or 60° C.; (c) 2.5% TFA/DCM for 12 hours; (d) thiol, n-BuLi, at ⁇ 25° C., THF.
  • FIG. 6 presents illustrations of chemical steps in building block preparation.
  • FIG. 7 presents ⁇ -substituted allenoic acid building blocks.
  • FIG. 8 presents ⁇ -substituted allenoic acid building blocks.
  • FIG. 9 presents N-sulfonylimine building blocks.
  • FIG. 10 presents thiol building blocks.
  • FIGS. 11A-H present structures and numbers of the library compounds.
  • FIG. 12 presents tetrahydropyridine scaffold compounds with GGTI activity.
  • FIG. 13 presents piperidine scaffold compounds with GGTI activity.
  • FIG. 14 presents piperidine scaffold compounds with GGTI activity.
  • FIGS. 15A and 15B present dose dependency of GGTase I inhibition by two GGTI compounds.
  • FIG. 16 presents a comparison of GGTase inhibition and FTase inhibition by three compounds.
  • FIGS. 17A and 17B present GGTI compounds with preferential inhibition of K-Ras4B-driven GGTase I activity.
  • FIG. 18 presents focused future libraries based on SAR analysis.
  • FIGS. 19-1 through 19 - 27 present the measured Kras inhibitory activities of individual compounds.
  • FIGS. 20A-20C illustrate specific inhibition of GGTase I by P3-E5 and P5-H6.
  • FIGS. 20D-F show the structure of GGTase I (D), FTase (E) and GGTase II (F).
  • FIGS. 21A-21D illustrate that GGTIs compete with substrate protein but do not compete with GGPP, with a scheme shown in 21E.
  • FIGS. 22A and 22B show inhibition of germaylgeranylation in cells treated with P3-E5.
  • FIG. 23 illustrates inhibition of proliferation in K562 cells by GGTIs.
  • FIG. 24A illustrates that GGTIs induce G1 cell cycle arrest in K562 cells with a scheme shown in 24B.
  • FIGS. 25A and 25B illustrate that mP5-H6 inhibits geranylgeranylation in vivo.
  • FIG. 26 illustrates that mP5-H6 exhibits increased potency to inhibit proliferation of Panc-1 and Jurkat cells.
  • FIG. 27 illustrates exemplary GGTIs of the present invention (mP5-H6, P5-H6, and P3-E5) and some other GGTI compounds.
  • FIG. 28 illustrates structures of GGTIs with dihydropyrrole scaffold 6.
  • FIGS. 29A to 29C illustrate structures of GGTIs with pyrrolidine scaffold 10.
  • FIGS. 30A to 30E illustrate structures and members of a library of compounds.
  • FIG. 31 illustrates a method of synthesizing GGTIs.
  • FIG. 32 illustrates a formula for exemplary compounds of the present invention.
  • FIG. 33 shows Western blots of cell lysates with geranylgeranylated Rheb, using compounds 22 and 23.
  • FIG. 34 shows the effect of P3-E5 (A) and P5-H6 (B) on the enzymatic activity of GGTase-I (left), RabGGTase (middle) and FTase (right). Varying concentrations of compounds were added to each enzyme reaction. Data represent the mean+/ ⁇ S.D. of two measurements from two independent experiments.
  • FIG. 35 shows a kinetic analysis of GGTase-I inhibition. Double reciprocal plots were obtained from substrate velocity curves for the inhibition of GGTase-I by P3-E5 (left) and P5-H6 (right).
  • A shows varying GGPP concentrations with a fixed RhoA protein concentration were used.
  • B shows varying RhoA protein concentrations with a fixed GGPP concentration. The amount of GGTI used is indicated in the figure.
  • FIG. 36 shows the cellular activity of modified GGTI compounds.
  • A shows the molecular structure of P5-H6 and modified P5-H6 compounds. K562 cells were treated with modified compounds for 72 hours and then cell number was counted. IC 50 values of cell viability relative to the DMSO were measured.
  • B shows the inhibitory effect of 12.5 ⁇ M P5-H6 or P61-A6 on PANC-1 and Jurkat cell viability. Data represent the mean+/ ⁇ S.D. of two measurements from two independent experiments. *, P ⁇ 0.05 compared with the value for DMSO.
  • C-E P5-H6 or P61-A6 treatment inhibits Rap1 geranylgeranylation in NIH3T3 cells.
  • FIG. 37 shows the effects of P61-A6 or P61-B6 on cell proliferation and cell cycle in MCF-7 cells.
  • A shows the inhibition of proliferation of MCF-7 by P61-A6 and P61-B6.
  • MCF-7 cells were treated with P61-A6, P61-B6 or DMSO for 72 hours. Cell viability relative to the DMSO control (100% value) is plotted.
  • B MCF-7 cells were treated with indicated concentrations ( ⁇ M) of P61-A6, P61-B6 or DMSO for 48 hours. Cell cycle profiles were monitored by flow cytometry. Percentages of cells in each phase of the cell cycle are indicated by different shades.
  • NIH3T3 cells were transfected with p21 CIPI/WAFI -luciferase or empty vector. Cells were treated with P61-A6 or P61-B6 compound at indicated concentrations or with DMSO for 48 hours and luciferase assay was performed. Data represent the mean+/ ⁇ S.D. of two measurements from two independent experiments. *, P ⁇ 0.05; **, P ⁇ 0.005 compared with the value for DMSO.
  • FIG. 38 shows the dual specificity inhibitors of GGTase-I and RabGGTase.
  • A shows the molecular structure of dual specificity inhibitors.
  • B shows the IC 50 values of dual specificity inhibitors against GGTase-I, RabGGTase and FTase.
  • C shows the Inhibitory effect of 25 ⁇ M P49-F5 compound on in vitro activities of GGTase-I, RabGGTase and FTase.
  • FIG. 39 shows RabGGTase preferential inhibitors.
  • A shows the molecular structure of RabGGTase inhibitors.
  • B shows IC 50 values of RabGGTase preferential inhibitors against GGTase-I, RabGGTase and FTase.
  • C shows the inhibitory effect of 25 ⁇ M P49-F6 compound on in vitro activities of GGTase-I, RabGGTase and FTase.
  • FIG. 40 shows a characterization of P49-F6 inhibition of RabGGTase.
  • in vitro RabGGTase assay was carried out with varying concentration of GGPP (A) or Rab7 (B).
  • Each reaction mixture contained recombinant RabGGTase and recombinant REP-1 protein. They were mixed with a low concentration of GGPP (0.125 ⁇ M) to form an active enzyme first. Duplicate reactions were carried out. The amount of RabGGTI used is indicated.
  • FIG. 41 Primary FACS data. MCF-7 cells were treated with 10 ⁇ M of P61-A6, P61-B6 or DMSO for 48 hours. Data shown here are representative of two independent experiments for each treatment.
  • FIG. 42 shows the inhibition of RabGGTase activity in cells.
  • A shows that P49-F6 treatment inhibits Rab5b geranylgeranylation in NIH3T3 cells.
  • Whole cell lysates from cells treated with DMSO or P49-F6 for 48 hours were prepared and processed for immunoblot analysis using antibody against Rab5b (upper panel) or actin (lower panel).
  • B shows whole cell lysates from NIH3T3 cells treated with DMSO or P49-F6 for 48 hours were prepared and processed for immunoblot analysis using antibody against unprenylated form of Rap1 (upper panel), total-Rap1 (upper middle panel), H-Ras (lower middle panel) or actin (lower panel).
  • FIG. 43 shows the effects of RabGGTI on membrane association of Rab5b protein.
  • Rho GDI and Na + /K + ATPase were used as marker proteins for the soluble and membrane fractions, respectively. Bars indicate intensity of protein bands after normalization using loading control.
  • FIG. 44 shows that P61A6 inhibits PANC-1 xenograft tumor growth in SCID mice.
  • FIG. 45 shows an examination of mouse body weight indicating that the GGTI treatment did not cause adverse effects.
  • FIG. 46 shows that P61A6 inhibits PANC-1 xenograft tumor growth in SCID mice.
  • FIG. 47 shows that the PANC-1 cell tumor size was reduced by administering P61A6 six times per week.
  • FIG. 48 shows dose dependent inhibition of a xenograft tumor by P61A6.
  • FIG. 49 shows that the administration of P61A6 did not significantly effect mouse body weight.
  • FIG. 50 shows the serum levels of P61A6 over time.
  • FIG. 51 shows an example synthesis method for preparing the second-generation library.
  • the following reaction conditions can be used: (a) allenoic acid, Mukaiyama's reagent, DIPEA or Et 3 N, DCM, rt, 12 h; (b) PBu 3 , imine, benzene, 60° C. (c) PBu 3 , imine, DCM, rt (d) 2.5% TFA/DCM, 12 h.
  • FIG. 52 shows the synthesis of the building blocks for the focused second-generation library.
  • FIG. 53 shows the structure of an example gamma-substituted allenoic acid used in the synthesis of the focused second-generation library.
  • FIG. 54 shows the structures of example alpha-substituted allenoic acids used in the synthesis of the focused second-generation library.
  • FIG. 55 shows the structures of example imines used in the synthesis of the focused second-generation library.
  • FIG. 56 shows the structures and numbers of the focused second-generation library compounds.
  • FIG. 57 shows the structures of the building blocks used in the synthesis of the focused second-generation library compounds with scaffold 6.
  • FIG. 58 shows the structures of the building blocks used in the synthesis of the focused second-generation library compounds with scaffold 7.
  • FIG. 59 shows tetrahydropyridine scaffold (7) with GGTI activity that exceeds the GGTI activity of P3-E5 (UC23).
  • FIG. 60 shows dihydropyrrole scaffold (6) with GGTI activity that exceeds the GGTI activity of P5-H6 (UC22).
  • Rho proteins such as RhoA and Rac are critical in enhancing transformation.
  • peptidomimetic inhibitors of GGTase I have shown promise in inhibiting proliferation of cancer cells. An arrest of the cell cycle at the G0/G1 phase was consistently observed with GGTase I inhibitors.
  • RhoC geranylgeranylated proteins
  • GGTIs that specifically inhibit K-Ras4B geranylgeranylation but do not inhibit geranylgeranylation of RhoA are of interest, as they may provide a way to overcome one of the major shortcomings of currently available FTIs. While FTIs can potently inhibit FTase, they are incapable of inhibiting K-Ras, as this protein undergoes modification by GGTase I. See Tamanoi, F., Gau, C. L., Edamatsu, H., Jiang, C. and Kato-Stankiewicz, J. (2001) Protein farnesylation in mammalian cells, Cell Mol. Life. Sci. 58, 1-14.
  • embodiments of the invention are directed to the identification and characterization of small molecule inhibitors of GGTase-I with novel scaffolds from a library consisting of allenoate derived compounds. These compounds can exhibit specific inhibition of GGTase-I, often by competing with a substrate protein. It was discovered that derivatizing a carboxylic acid emanating from the core ring of one of the GGTI compounds improved their cellular activity. The improved GGTI compounds inhibit proliferation of a variety of human cancer cell lines and cause G 1 cell cycle arrest and induction of p21 CIPI/WAFI .
  • Embodiments of the invention are also directed to the identification and characterization of novel small molecule inhibitors of RabGGTase. These compounds were identified by screening the GGTI compounds described herein for compounds that also exhibited RabGGTase inhibition. This screening led to the discovery of a common structural feature for RabGGTase inhibitors; the presence of a characteristic six-atom aliphatic tail attached to the penta-substituted pyrrolidine core. Additional screening and research led to the identification of compounds with preferential inhibition of RabGGTase. It is believed that these compounds inhibit RabGGTase activity by competing with the protein substrate.
  • inhibitor means that a compound stops or otherwise prevents at least one function of a target compound.
  • a GGTase I inhibitor can stop or otherwise prevent at least one activity a target compound, for example, an activity of the enzyme GGTase I.
  • the target compounds for the inhibitors described herein is a protein prenyltransferase. Inhibition can occur in vitro and/or in vivo using a predetermined amount of an inhibitor.
  • alkyl denotes branched or unbranched hydrocarbon chains, preferably having about 1 to about 8 carbons, such as, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, 2-methylpentyl pentyl, hexyl, isohexyl, heptyl, 4,4-dimethyl pentyl, octyl, 2,2,4-trimethylpentyl and the like.
  • Substituted alkyl includes an alkyl group optionally substituted with one or more functional groups which may be attached to such chains, such as, hydroxyl, bromo, fluoro, chloro, iodo, mercapto or thio, cyano, alkylthio, heterocyclyl, aryl, heteroaryl, carboxyl, carbalkoyl, alkyl, alkenyl, nitro, amino, alkoxyl, amido, and the like to form alkyl groups such as trifluoro methyl, 3-hydroxyhexyl, 2-carboxypropyl, 2-fluoroethyl, carboxymethyl, cyanobutyl and the like.
  • “Bulky alkyl” includes cycloalkyl and branched chain alkyls with 4-8 carbons.
  • cycloalkyl as employed herein alone or as part of another group includes saturated or partially unsaturated (containing 1 or more double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocyclicalkyl, bicyclicalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons forming the rings, preferably 3 to 10 carbons, forming the ring and which may be fused to 1 or 2 aromatic rings as described for aryl, which include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl and cyclododecyl, cyclohexenyl.
  • “Substituted cycloalkyl” includes a cycloalkyl group optionally substituted with 1 or more substituents such as halogen, alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol and/or alkylthio and/or any of the substituents included in the definition of “substituted alkyl.” For example,
  • alkenyl refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons, and more preferably 2 to 8 carbons in the normal chain, which include one or more double bonds in the normal chain, such as vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4-pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 3-octenyl, 3-nonenyl, 4-decenyl, 3-undecenyl, 4-dodecenyl, 4,8,12-tetradecatrienyl, and the like.
  • Substituted alkenyl includes an alkenyl group optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • alkynyl refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons and more preferably 2 to 8 carbons in the normal chain, which include one or more triple bonds in the normal chain, such as 2-propynyl, 3-butynyl, 2-butynyl, 4-pentynyl, 3-pentynyl, 2-hexynyl, 3-hexynyl, 2-heptynyl, 3-heptynyl, 4-heptynyl, 3-octynyl, 3-nonynyl, 4-decynyl, 3-undecynyl, 4-dodecynyl and the like.
  • Substituted alkynyl includes an alkynyl group optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • arylalkyl refers to alkyl, alkenyl and alkynyl groups as described above having an aryl substituent.
  • Representative examples of arylalkyl include, but are not limited to, benzyl, 2-phenylethyl, 3-phenylpropyl, phenethyl, benzhydryl and naphthylmethyl and the like.
  • Substituted arylalkyl includes arylalkyl groups wherein the aryl portion is optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • arylalkyl refers to alkyl, alkenyl and alkynyl groups as described above having an aryl substituent.
  • Representative examples of arylalkyl include, but are not limited to, benzyl, 2-phenylethyl, 3-phenylpropyl, phenethyl, benzhydryl and naphthylmethyl and the like.
  • Substituted arylalkyl includes arylalkyl groups wherein the aryl portion is optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • Salt, crystalline, and other forms of the chemical compounds depicted in the formulas and structures shown and described herein are contemplated within the meaning of “compound” of the invention.
  • the compounds described herein may be used in their salt form (e.g., a sodium, potassium, or other pharmaceutically acceptable salt) or in a crystalline form.
  • a salt cannot be readily prepared using conventional methods but, as one of skill in the art will appreciate, alternative methods may be used to prepare a salt.
  • the salt or crystalline forms of the compounds described herein may be useful as part of a pharmaceutical composition.
  • halogen or “halo” as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine.
  • halogenated alkyl refers to “alkyl”, “alkenyl” and “alkynyl” which are substituted by one or more atoms selected from fluorine, chlorine, bromine, fluorine, and iodine.
  • aryl or “Ar” as employed herein alone or as part of another group refers to monocyclic and polycyclic aromatic groups containing 6 to 10 carbons in the ring portion (such as phenyl or naphthyl including 1-naphthyl and 2-naphthyl) and may optionally include one to three additional rings fused to a carbocyclic ring or a heterocyclic ring (such as aryl, cycloalkyl, heteroaryl or cycloheteroalkyl rings).
  • “Substituted aryl” includes an aryl group optionally substituted with one or more functional groups, such as halo, haloalkyl, alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, trifluoromethyl, trifluoromethoxy, alkynyl, cycloalkyl-alkyl, cycloheteroalkyl, cycloheteroalkylalkyl, aryl, heteroaryl, arylalkyl, aryloxy, aryloxyalkyl, arylalkoxy, alkoxycarbonyl, arylcarbonyl, arylalkenyl, aminocarbonylaryl, arylthio, arylsulfinyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl, heteroaryloxy, hydroxy, nitro, cyano, amino, substituted amino wherein the amino wherein
  • heterocyclic or “heterocycle”, as used herein, represents an unsubstituted or substituted stable 5- to 10-membered monocyclic ring system which may be saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from N, O or S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocyclic groups include, but is not limited to, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, oxopyrrolidinyl, oxoazepinyl, azepinyl, pyrrolyl, pyrrolidinyl, furanyl, thienyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isooxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, thiadiazolyl, tetrahydropyranyl, thiamorpholinyl, thiamorpholinyl,
  • heterocyclic aromatic refers to a 5- or 7-membered aromatic ring which includes 1, 2, 3 or 4 hetero atoms such as nitrogen, oxygen or sulfur and such rings fused to an aryl, cycloalkyl, heteroaryl or heterocycloalkyl ring (e.g. benzothiophenyl, indolyl), and includes possible N-oxides.
  • “Substituted heteroaryl” includes a heteroaryl group optionally substituted with 1 to 4 substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.” Examples of heteroaryl groups include the following:
  • polar metal binder means a polar group that is capable of binding, e.g., chelating, to a metal.
  • Non-limiting examples have been disclosed in the chemical structures included in this application and its figures.
  • A can be any of the following (with or without the double bond within the ring):
  • the “linker” of Formula I can be any of the following:
  • inventive compounds with the novel linkers and scaffolds described herein can be produced using variations of a pilot library.
  • a pilot library was constructed using allenoate as a multireactive core molecule. This pilot library was screened for compounds which inhibited protein farnesyltransferase (FTase), that is, farnesyltransferase inhibitors (FTIs), and for compounds which inhibited protein geranylgeranyltransferase type I (GGTase I), that is geranylgeranyltransferase type I inhibitors (GGTIs). Because these enzymes catalyze modification of signaling proteins such as Ras, Rheb and Rho, small molecule inhibitors can be anti-cancer therapeutics. 4,314 compounds were synthesized and screened for compounds with improved potency of inhibition. As discussed further below, structure activity relationship studies of these compounds pointed to the significance of certain substitution patterns on the aromatic substitutions of the scaffold ring structure of initial hits.
  • the pilot library was constructed by reacting allenoates with imines, aldehydes and maleimides under phosphine catalysis conditions to produce an array of compounds including dihydropyrroles, tetrahydropyridines, bicyclic succinimides, dioxanylidenes and dihydrofurans.
  • the 171 compounds produced for the pilot library are illustrated in FIG. 1 .
  • RhoA and K-Ras4B are two very different substrates of this enzyme and it is of interest to identify small molecule inhibitors exhibiting preferential inhibition on reaction driven by one substrate over another.
  • P2-F10 demonstrates significant inhibition of GGTase I when K-Ras4B is used as a substrate, while virtually no inhibition was observed when RhoA was used as a substrate.
  • known GGTI compound GGTI-298 and GGTI-2166 inhibit RhoA-driven GGTase I activity slightly better than K-Ras4B-driven GGTase I activity ( FIG. 17 ).
  • This group of compounds possesses 5-alkyl-2-aryl-N-tosy-2,5-dihydropyrrole-3-carboxylic acid structure, which is distinct from the previously described tetrahydropyridine motif.
  • the FTI P1-F 11 resembles the GGTI P1-F10, while the FTI P2-B7 resembles the GGTI P2-C10.
  • the FTI P2-E9 is similar to the GGTI P2-G3. Similarity of some GGTI and FTI compounds may point to common structural features between the two closely related enzymes. As described before, FTase and GGTase I have similar three-dimensional structures and a shared alpha subunit. On the other hand, FTase specific inhibitors may recognize a region(s) that is different between the two enzymes. The FTIs identified here exhibit a weaker inhibition of GGTase I at the concentration that inhibits FTase.
  • An advantage of using the diversity oriented library described herein is that it is possible to quickly synthesize a large number of compounds that are related to lead compounds.
  • Split-and-pool synthesis on solid support is one of the fastest and most efficient ways of generating a large number of spatially segregated compounds. See (a) Furka, A.; Sebestyen, F.; Asgedom, M.; Dibó (1988), in Highlights of Modern Biochemistry, Proceedings of the 14 th International Congress of Biochemistry, Prague, Czechoslovakia (VSP, Utrecht, Netherlands), 13, 47. (b) Furka, A.; Sebestyén, F.; Asgedom, M.; Dibó (1991), Int. J. Pept. Protein Res. 37, 487.
  • the SynPhase Lantern consists of a grafted mobile surface polymer (such as polystyrene) onto a rigid and unreactive base polymer that is cylindrical in appearance.
  • the SynPhase Lantern is available in three different sizes, with loadings of 15 ⁇ mol, 35 ⁇ mol, and 75 ⁇ mol. Considering that a typical assay requires 1 nmol of a small organic molecule, 15 ⁇ 75 ⁇ mol of compound provides a large enough quantity of chemicals for multiple assays.
  • the rigid polymeric support of lanterns beneath a grafted mobile phase makes weighing unnecessary and handling easier than with resins.
  • Heterocycles 4 and 5 were cleaved off the resin in order to furnish acids 6 and 7 in 91 ⁇ 92% yield (based on a theoretical loading of 15 ⁇ M/Lantern) after flash column chromatography (FCC).
  • FCC flash column chromatography
  • the Michael addition of thiols to 4 and 5 with n-butyl lithium as a base provided 8 and 9, which, upon trifluoroacetic acid (TFA)-mediated cleavage, provided 10 and 11.
  • TFA trifluoroacetic acid
  • ⁇ -substituted allenoic acid 21 was prepared by saponification of ester 20.
  • N-sulfonylimine 24 was formed by the azeotropic removal of water from a refluxing toluene mixture of appropriate sulfonamide 22, aldehyde 23, and catalytic acid.
  • FIG. 7 The building blocks synthesized as illustrated in FIG. 6 are shown in FIG. 7 ( ⁇ -substituted allenoic acid 17), FIG. 8 ( ⁇ -substituted allenoic acid 21), and FIG. 9 (N-sulfonylimine 24).
  • FIG. 10 The other building blocks, thiols, which were purchased for the library synthesis, are shown in FIG. 10 .
  • building blocks were synthesized, they were tested to determine whether or not they would be incorporated into the synthesis of the focused library of GGTIs. Building blocks were chosen so that only the ones that provided high purity (by 1 H NMR and LC/MS analysis) for the final crude products would be used in the synthesis of the library. The building blocks chosen for the synthesis of the library and the resulting number of compounds are shown in FIG. 11 . Scaffolds that require distinctive set of reaction conditions are drawn separately.
  • 5-alkyl-2-aryl-4-mercapto-N-sulfonyl-2,5-dihydropyrrole-3-carboxylic acid 10 7 allenoic acids (A05-A11), 25 imines (C01-C06, C09-C14, C16, C20, C21, C23, C24, C26, C27, C29, C30, C33, C35, C40, C41), and 19 thiols (E01, E02, E04, E06, E07, E16-E25, E28-E30, E32) were chosen to give 3,325 (7 ⁇ 25 ⁇ 19) 5-alkyl-2-aryl-4-mercapto-N-sulfonyl-2,5-dihydropyrrole-3-carboxylic acids ( FIG. 11C ).
  • the next step was the split-and-pool synthesis of a library of heterocycles (shown in FIG. 5 ).
  • the first split step of the proposed library synthesis is the coupling of 23 different allenoic acids to the benzyl alcohol of the Wang resin.
  • resin-bound allenoates MCMs
  • imines under nucleophilic phosphine catalysis conditions to generate two distinctive heterocycles 4 and 5.
  • the last split step involves use of the ⁇ , ⁇ -unsaturated ester moiety (MCF) in stereoselective Michael addition using thiols to further increase structural diversity, resulting in two additional scaffolds 8 and 9.
  • a pair of numbers below the structure of a GGTI is % activities of GGTase I for proteins K-Ras4B/RhoA in the presence of the GGTI compound. Smaller numbers indicate better inhibition. Activities below 10% (over 90% inhibition) for both proteins are underlined. Significantly better inhibition for RhoA over K-Ras4B is in parenthesis.
  • the “label” column provides annotation (e.g., B01C01T means a compound made of B01 and C01 and the Tebbe reaction, and B01C01E21 means a compound made of building blocks B01, C01, and E21) of each compound for each entry.
  • the “Kras” column provides % activity of GGTase I for protein K-Ras4B in the presence of the compound of that entry.
  • the % activity of GGTase I was determined as follows: a working solution that contained each compound in DMSO at the concentration of 1 mM was prepared. The working solution was added to GGTase I reaction mixture so that the final concentration of the compound is 50 ⁇ M. GGTase I activity was assayed by incubating [ 3 H]GGPP with K-Ras4B or RhoA protein in the presence of GGTase I and examining radioactivity incorporated into the substrate protein by spotting onto a filter paper. After washing with TCA, ethanol and acetone, the radioactivity retained on the filter was determined by the use of a scintillation counter.
  • GGTase I activity in the presence of DMSO was taken as a 100% value and the activity in the presence of the compound was shown as percent activity compared with the 100% value.
  • the table in FIG. 12 shows 24 2,6-diaryl-N-sulfonyl-1,2,5,6-tetrahydropyridine-3-carboxylic acids 7 that were selected out of 367 compounds. None of the enones 13 were active indicating that the carboxylic acid moiety is crucial for the GGTI activity. For 6-aryl-4-mercapto-N-sulfonyl-piperidine-3-carboxylic acid 11 the 4-mercapto substituent appeared critical to endowing GGTase inhibitory activity.
  • IC50 values for the inhibition of GGTase I using RhoA as a substrate were 5 and 7 ⁇ M for P3-D9 and P3-E5, respectively (see FIG. 12 for the structures of P3-D9 and P3-E5). Under the same reaction condition, IC 50 values of 1.6 and 0.50M were determined for the known GGTIs, GGTI-298 and GGTI-2166, respectively. Specificity of GGTase I inhibition by P3-D9 and P3-E5 was examined. As can be seen in FIG.
  • FIG. 17 lists GGTI compounds with reproducibly stronger inhibition of K-Ras4B driven GGTase I over RhoA driven activity.
  • the novel compound P4-F1 inhibits K-Ras4B driven GGTase I activity, but does not inhibit RhoA-driven GGTase I activity. Accordingly, some embodiments of the present invention are directed to compounds that inhibit K-Ras4B.
  • FIG. 12 Analysis of the structures in FIG. 12 suggest construction of a library of 2,6-diaryl-N-sulfonyl-1,2,5,6-tetrahydropyridine-3-carboxylic acids 7 consisting of B02 through B07 and new imines C47 through C49 for further improvement of GGTI activity ( FIG. 18 ).
  • J is hydrogen or is 1-2 substituents independently selected from the group consisting of halogen, C 1 -C 3 alkyl, OR′, SR', and NR' 2 , where R′ is alkyl,
  • E is hydrogen or is 1-2 substituents selected from the group consisting of halogen, C 1 -C 3 alkyl, OR′, SR′, and NR′ 2 , where R′ is alkyl,
  • W is selected from the group consisting of hydrogen, cyclic, linear, or branched alkyl of from 2 to 8 carbons, unsubstituted phenyl, and phenyl substituted with C 1 -C 3 alkyl, halogen, OR′, SR′, and NR′ 2 , where R′ is alkyl,
  • A is selected from the group consisting of:
  • M is selected from the group consisting of OH, OR′′, NH 2 , NHOH, NHOR′′, wherein R′′ is methyl or ethyl, or any other group that has a polar metal binder
  • R corresponds to an alpha-substituent of natural or non-natural alpha-amino acid
  • Z is S-U
  • X-Y is any one of the following:
  • substituents can be selected from those described herein.
  • linker 3 of Formula I is used and the remaining substituents can be selected from those described herein.
  • these compounds may be based on scaffolds 6, 7, 10, or 11. These scaffolds have the following structures:
  • the compounds constructed on scaffold 6 may include substituents at R1, P and R3.
  • R1 may be selected from the groups described in relation to W herein.
  • P is a protecting group. While many protecting groups may be used at P, the following protecting group is preferred:
  • E is selected from the groups previously described herein for E.
  • R3 can be, for example, an unsubstituted phenyl or a substituted phenyl ring.
  • the substitution also represented as J
  • J can be at any position on the phenyl ring.
  • compound UC-22 also referred to as P5-H6
  • This compound has the following chemical structure:
  • FIG. 2 Further compounds of interest that can be based on scaffold 6 are listed in FIG. 2 . These compounds include P2-F10, P2-F12, P2-G3 and P2-G6.
  • GGTase I inhibiting compounds can be based on scaffold 7.
  • the compounds based on scaffold 7 may include substituents at R3, P and R2.
  • R3 is represented herein as W and any of the groups described in relation to W may be used at position R3.
  • P is a protecting group as described above.
  • R2 can be, for example, an unsubstituted phenyl or a substituted phenyl ring.
  • the substitution also represented as J
  • J can be at any position on the phenyl ring.
  • compound UC-23 also referred to as P3-E5
  • This compound has the following chemical structure:
  • FIG. 2 Further compounds of interest that can be based on scaffold 7 are listed in FIG. 2 . These compounds include P2-D5, P2-D7, P2-D8, P2-D9, and P2-D10.
  • R1 may be selected from the groups described as W herein.
  • P is a protecting group as described above.
  • R3 can be, for example, an unsubstituted phenyl or a substituted phenyl ring.
  • the substitution also represented as J
  • J can be at any position on the phenyl ring.
  • SR4 can be represented as thiols, with R4 being selected from the group consisting of hydrogen, alkyl of from 2 to 10 carbons.
  • GGTase I inhibiting compounds can be based on scaffold 7.
  • the compounds based on scaffold 11 may include substituents at R3, P R2, and SR4.
  • R3 is represented herein as W and any of the groups described in relation to W may be used at position R3.
  • P is a protecting group as described above.
  • R2 can be, for example, an unsubstituted phenyl or a substituted phenyl ring.
  • the substitution also represented as J
  • J can be at any position on the phenyl ring.
  • SR4 can be represented as thiols, with R4 being selected from the group consisting of hydrogen, alkyl of from 2 to 10 carbons. Compounds prepared using scaffold 11 are disclosed herein.
  • the GGTI compound can be selected from the following additional examples of GGTIs that have been made by the methods described herein.
  • compound P61-A6 One example embodiment of the compounds that may be built from Formula 1 and/or the scaffolds disclosed herein is compound P61-A6. This compound has the following chemical structure and has been found to inhibit GGTase I activity:
  • Some exemplary compounds that inhibit GGTase I activity based on Formula 1 include those listed in Table 3 below. In Table 3 in vitro IC 50 values for the GGTase I activity are reported. In Table 3 the isolated yield of each synthetic step for the synthesis of corresponding compounds are also reported.
  • Some exemplary compounds that inhibit GGTase I activity based on Formula 1 include those listed in Table 4.
  • Table 4 in cell GI 50 values for the growth inhibition of Jurkat cells are reported.
  • the GI 50 value is the concentration of a test compound needed to produce 50% growth inhibition in the cell type listed.
  • Table 4 the isolated yield of each synthetic step for the synthesis of corresponding compounds are also reported.
  • One embodiment of the compounds that may be built from Formula 1 is compound P61-H7. This compound has the following chemical structure:
  • Some exemplary compounds that inhibit GGTase I activity based on Formula 1 include those listed in Table 5.
  • Table 5 in vitro IC 50 values for the GGTase I activity are reported.
  • Table 5 the isolated yield of each synthetic step for the synthesis of corresponding compounds are also reported.
  • One embodiment of the compounds that may be built from Formula 1 is compound P61-D3. This compound has the following chemical structure:
  • One embodiment of the compounds that may be built from Formula 1 is compound P61-E7. This compound has the following chemical structure:
  • Some exemplary compounds that inhibit GGTase I activity based on Formula 1 include those listed in Table 6.
  • Table 6 in cell GI 50 values for the growth inhibition of Jurkat cells are reported.
  • Table 6 the isolated yield of each synthetic step for the synthesis of corresponding compounds are also reported.
  • Some exemplary compounds that inhibit GGTase I activity based on Formula 1 include those listed in Tables 7-15.
  • Tables 7-15 in vitro IC 50 values for the GGTase I activity are reported.
  • Tables 7-15 in cell GI 50 values for the growth inhibition of a variety of cancer cell lines e.g., Jurkat, K562, MDA-MB-231, BT474, MCF7, MiaPaCa2, Aspc-1, Panc-1, Capan-2, CFpac-1, and MCF10a
  • protein prenyltransferase inhibiting compounds described herein can be effective as inhibitors of RabGGTase. As shown in the examples below, some of these compounds can exhibit dual specificity and can inhibit both RabGGTase and GGTase I.
  • RabGGTase inhibitors Prior to the present compounds, only a handful of RabGGTase inhibitors have been identified in the past (26). Commonly used inhibitors are bisphosphonate type compounds, however the inhibition of RabGGTase requires about mM concentration of the compounds. In contrast, in some embodiments, the compounds described herein surprisingly inhibit RabGGTase at a ⁇ M concentration. Accordingly, the RabGGTase inhibitors can be used at a concentration of about 1 ⁇ M up to about 50 ⁇ M, for example at concentrations between 1 ⁇ M and 25 ⁇ M.
  • these RabGGTase inhibiting compounds use the scaffolds of the previously described GGTI compounds but possess an extra hydrophobic tail emanating from the core ring.
  • Such hydrophic tails have been illustrated throughout the application. As one of skill in the art will appreciate, these compounds can also use the previously described substituent groups in addition to the hydrophic tail.
  • Embodiments of the present invention are also directed to RabGGTase compounds with a six-atom aliphatic tail attached to the penta-substituted pyrrolidine core via thioether linkage to the 5-member ring core. These compounds can have an unexpectedly potent effect in vivo and/or in vitro on cancer cell proliferation. In some embodiments, these compounds can have dual specificity for RabGGTase and GGTase I.
  • RabGGTIs may be valuable as anticancer drugs.
  • a study using siRNA showed that the inhibition of RabGGTase leads to apoptosis induction in human cancer cells (42). Elevated levels of RabGGTase are detected in a number of human cancers (42).
  • FTI compounds which inhibit RabGGTase induce mislocalization of Rab protein and apoptosis (42). Accordingly, these compounds can be useful as cancer therapeutics.
  • compounds described as GGTase I inhibitors or RabGGTase inhibitors are not limited to only that characterization.
  • a compound can have dual specificity to inhibit both GGTase I and RabGGTase. Therefore, as one of skill in the art will appreciate, the compounds described herein are protein prenyltransferase inhibitors capable of inhibiting GGTase I, RabGGTase, other protein prenyltransferases, and combinations thereof.
  • the compounds described herein can be made using any method known to one of skill in the art or as described herein. Suitable methods have previously been disclosed in relation to Formula 1 in section I above and additional methods are described to follow. Accordingly, the present invention is also directed to novel, improved methods of synthesizing protein prenyl transferase inhibitors.
  • R is, an alpha-substituent of a natural or non-natural amino acid; comprising, reacting a compound according to formula I′
  • compositions having linker 1 can be prepared using the following example reaction scheme:
  • compositions having linker 2 can be prepared using the following example reaction scheme:
  • compositions having linker 3 can be prepared using the following example reaction scheme:
  • the compounds of the invention are useful as pharmaceutical compositions prepared with a therapeutically effective amount of a compound of the invention, as defined herein, and a pharmaceutically acceptable carrier or diluent.
  • the compounds of the invention can be formulated as pharmaceutical compositions and administered to a subject in need of treatment, for example a mammal, such as a human patient, in a variety of forms adapted to the chosen route of administration, for example, orally, nasally, intraperitoneally, or parenterally, by intravenous, intramuscular, topical or subcutaneous routes, or by injection into tissue.
  • Suitable oral forms for administering the compounds include, lozenges, troches, tablets, capsules, effervescent tablets, orally disintegrating tablets, floating tablets designed to increase gastric retention times, buccal patches, and sublingual tablets.
  • the compounds of the invention may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier, or by inhalation or insufflation. They may be enclosed in coated or uncoated hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet.
  • a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier, or by inhalation or insufflation.
  • the compounds may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • compositions suitable for administration to humans are meant to include, but is not limited to, those ingredients described in Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, 21st ed. (2006) (hereinafter Remington's), which is herein incorporated by reference in its entirety.
  • compositions and preparations should contain at least 0.1% compounds.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2% to about 60% of the weight of a given unit dosage form.
  • the amount of compounds in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • the tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • a syrup or elixir may contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the compounds may be incorporated into sustained-release preparations and devices.
  • the compounds may be incorporated into time release capsules, time release tablets, and time release pills.
  • the composition is administered using a dosage form selected from the group consisting of effervescent tablets, orally disintegrating tablets, floating tablets designed to increase gastric retention times, buccal patches, and sublingual tablets.
  • the compounds may also be administered intravenously or intraperitoneally by infusion or injection.
  • Solutions of the compounds can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations can contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the compounds which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, buffers or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • the compounds may be applied in pure form. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
  • a dermatologically acceptable carrier which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Other solid carriers include nontoxic polymeric nanoparticles or microparticles.
  • Useful liquid carriers include water, alcohols or glycols or water/alcohol/glycol blends, in which the compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver the compounds to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508), all of which are hereby incorporated by reference.
  • Useful dosages of the compounds of formula 1 can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949, which is hereby incorporated by reference.
  • the concentration of the compounds in a liquid composition can be from about 0.1-25% by weight, or from about 0.5-10% by weight.
  • concentration in a semi-solid or solid composition such as a gel or a powder can be about 0.1-5% by weight, or about 0.5-2.5% by weight.
  • the amount of the compounds required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
  • Effective dosages and routes of administration of agents of the invention are conventional.
  • the exact amount (effective dose) of the agent will vary from subject to subject, depending on, for example, the species, age, weight and general or clinical condition of the subject, the severity or mechanism of any disorder being treated, the particular agent or vehicle used, the method and scheduling of administration, and the like.
  • a therapeutically effective dose can be determined empirically, by conventional procedures known to those of skill in the art. See, e.g., The Pharmacological Basis of Therapeutics , Goodman and Gilman, eds., Macmillan Publishing Co., New York.
  • an, effective dose can be estimated initially either in cell culture assays or in suitable animal models. The animal model may also be used to determine the appropriate concentration ranges and routes of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • a therapeutic dose can also be selected by analogy to dosages for comparable therapeutic agents.
  • the pharmaceutical compositions described herein contain a therapeutically effective dose of the compound.
  • the term “therapeutically effective amount” of the compounds disclosed herein, when used in a method of treating a cancer refers to that dose of the compound that lessens or prevents the occurrence of cancer when administered to a mammal in need of such treatment.
  • the therapeutically effective amount will vary depending on the needs of the subject, but this amount can readily be determined by one of skill in the art, for example, a physician.
  • Treatment may involve daily or multi-daily doses of compound(s) over a period of a few days to months, or even years.
  • a suitable dose will be in the range of from about 0.001 to about 100 mg/kg, e.g., from about 0.01 to about 100 mg/kg of body weight per day, such as above about 0.1 mg per kilogram, or in a range of from about 1 to about 10 mg per kilogram body weight of the recipient per day.
  • a suitable dose may be about 1 mg/kg, 10 mg/kg, or 50 mg/kg of body weight per day.
  • the compounds are conveniently administered in unit dosage form; for example, containing 0.05 to 10000 mg, 0.5 to 10000 mg, 5 to 1000 mg, or about 100 mg of active ingredient per unit dosage form.
  • the dosage unit contains about 1 mg, about 10 mg, about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 750 mg, or about 1000 mg of active ingredient.
  • the compounds can be administered to achieve peak plasma concentrations of, for example, from about 0.5 to about 75 ⁇ M, about 1 to 50 ⁇ M, about 2 to about 30 ⁇ M, or about 5 to about 25 ⁇ M.
  • Exemplary desirable plasma concentrations include at least or no more than 0.25, 0.5, 1, 5, 10, 25, 50, 75, 100 or 200 ⁇ M.
  • plasma levels may be from about 1 to 100 micromolar or from about 10 to about 25 micromolar. This may be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the compounds, optionally in saline, or orally administered as a bolus containing about 1-100 mg of the compounds.
  • Desirable blood levels may be maintained by continuous infusion to provide about 0.00005-5 mg per kg body weight per hour, for example at least or no more than 0.00005, 0.0005, 0.005, 0.05, 0.5, or 5 mg/kg/hr.
  • such levels can be obtained by intermittent infusions containing about 0.0002-20 mg per kg body weight, for example, at least or no more than 0.0002, 0.002, 0.02, 0.2, 2, 20, or 50 mg of the compounds per kg of body weight.
  • the compounds may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator.
  • Some embodiments of the present invention are directed to methods of using the compounds described herein. These compounds, as illustrated in the examples below and throughout the specification, are useful in inhibiting the geranylgeranylation of signaling proteins by inhibiting the activity of at least GGTase I and/or RabGGTase. However, because GGTIs such as those described herein can inhibit geranylgeranylation of a number of signaling proteins the compounds described herein are effective for the treatment of a wide range of human cancers. For example, mP5-H6 inhibits proliferation of a leukemic cell line (Jurkat), breast cancer cell lines (BT474 and MDA-MB231) and pancreatic cancer cell lines (Panc-1 and MiaPaCa2).
  • Pancreatic cancer is of particular interest, as recent studies (Lim, K-H et al., Current Biology 16, 2385; Lim, K-H et al., Cancer Cell 7, 533) have established the importance of RalA and RalB proteins (both are geranylgeranylated) in pancreatic cancer.
  • RalA is commonly activated in a panel of cell lines from pancreatic cancer.
  • Studies using siRNA showed that inhibition of RalA reduced tumor growth.
  • RalB is found to be important for metastasis.
  • the RalA activation occurs downstream of K-ras activation that is seen in more than 80% of pancreatic cancer cases. Since K-ras prenylation can be inhibited by the combination of GGTI and FTI, GGTIs may be particularly important for pancreatic cancer.
  • GGTIs have been shown to inhibit proliferation of breast cancer cells (Vogt, A. et al., J. Biol. Chem. 272, 27224). This is accompanied by the accumulation of G1 phase cells and the increase of p21.
  • a geranylgeranylated protein Rac3 is reported to be overactivated in breast cancer cells (Mira J-P et al., PNAS 97, 185).
  • RhoA or RhoC by siRNA inhibited proliferation and invasiveness of breast cancer cells in vitro and in vivo (Pille, J-Y et al., Molecular Therapy 11, 267).
  • GGTIs and/or RabGGTase inhibitors may also be valuable in inhibiting cancer metastasis. This is based, in part, on findings that indicate geranylgeranylated proteins play important roles in metastasis.
  • RhoC another geranylgeranylated protein RhoC plays essential roles in cancer metastasis (Hakem A. et al., Genes & Dev. 19, 1974; Clark, E. A. et al., Nature 406, 532).
  • the compounds described herein can be used in methods of treating cancer and/or in methods of reducing the size of a cancerous tumor.
  • the method of treating cancer involves inhibiting a protein prenyltransferase by administering the compound described herein to a subject in need of treatment.
  • the methods of treating cancer can be applied to any cancer that is activated through a signaling pathway incorporating GGTase I and/or RabGGTase.
  • Rho protein activation e.g., RhoA and Rac in cancer cells.
  • the cancer is selected from the group consisting of pancreatic, leukemia, breast, and prostate.
  • the cancer is selected from the group consisting of pancreatic, leukemia, breast, prostate, colon, ovarian, lung, and stomach cancer.
  • the compounds described herein are also useful in methods of inhibiting the activity of GGTase I and/or RabGGTase by administering the compounds described herein to a cell. These methods can be applied either in vivo or in vitro. For example, these compounds can be used for therapeutic purposes associated with inhibiting the activity of GGTase I and/or RabGGTase in a subject in need of treatment thereof. These compounds can also be used in research methods designed to develop such therapeutics, for example, as part of a library screening as described herein.
  • the compounds described herein are also useful in methods of cytostatically inhibiting the growth of a cancer cell. These methods can be used as a stand alone therapeutic or in conjunction with a cytotoxic treatment or a surgical procedure.
  • the compounds described herein can be administered following surgery to remove a tumor or following chemotherapy designed to kill the tumor cells to control the growth of any cancer cells that these treatments may have missed. These compounds, when administered in these embodiments, may function as a preventative measure designed to reduce the likelihood of remission.
  • the compounds herein are also useful in assays for measuring the GGTase I and/or RabGGTase inhibiting activity of a compound.
  • the compounds disclosed herein can be used as controls to evaluate new compounds for potential GGTase I and/or RabGGTase inhibiting activity.
  • the compounds disclosed herein can be used as part of assays to determine therapeutic compounds for use in the methods disclosed herein as well.
  • kits comprising a compound of the invention in the form of a pharmaceutically acceptable dosage form or as a compound.
  • kits can include one or more containers filled with one or more of the ingredients of the pharmaceutical dosage forms.
  • the kit comprises a container for the dosage form or compound.
  • suitable containers include, for example, a bottle, a box, a blister card, a foil packet, or a combination thereof.
  • the kit also contains directions for properly administering the dosage form or for properly using the compound, for example, as part of an assay.
  • the kits can also be designed in a manner such that they are tamper resistant or designed to indicate if tampering has occurred.
  • the kit can contain the dosage form or compound with another pharmaceutical composition or compound, for example, an FTI.
  • kits can be a notice or printed instructions.
  • printed instructions can be in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of the manufacture, use, or sale for human administration to treat a condition that could be treated by the compounds and dosage forms described herein.
  • the kit further comprises printed matter, which, e.g., provides information on the use of the dosage form to treat a condition or disease or a pre-recorded media device which, e.g., provides information on the use of the dosage form to treat a condition or disease, or a planner, integrally linked to the particular methods of using the compositions.
  • Print matter can be, for example, one of a book, booklet, brochure or leaflet.
  • the printed matter can describe the use of the dosage forms described herein to treat a condition or disease, for example, to treat a cancer involving GGTase I modification of proteins.
  • Possible formats include, but are not limited to, a bullet point list, a list of frequently asked questions (FAQ) or a chart. Additionally, the information to be imparted can be illustrated in non-textual terms using pictures, graphics, or other symbols.
  • Pre-recorded media device can be, for example, a visual media device, such as a videotape cassette, a DVD (digital video disk), filmstrip, 35 mm movie, or any other visual media device.
  • pre-recorded media device can be an interactive software application, such as a CD-ROM (compact disk-read only memory) or floppy disk.
  • pre-recorded media device can be, for example, an audio media device, such as a record, audiocassette, or audio compact disk.
  • the information contained on the pre-recorded media device can describe the use of the dosage forms and compounds described herein to treat a condition or disease, for example, to treat a cancer involving GGTase I modification of proteins.
  • a “planner” can be, for example, a weekly, a monthly, a multi-monthly, a yearly, or a multi-yearly planner.
  • the planner can be used as a diary to monitor dosage amounts, to keep track of dosages administered, or to prepare for future events wherein taking a regularly administered dosage form as described herein.
  • the planner can be a calendar which will provide a means to monitor when a dosage has been taken and when it has not been taken. This type of planner will be particularly useful for patients having unusual schedules for administering medication to themselves. Additionally, the planner can be useful for the elderly, children, or other patient group who may administer medication to themselves and may become forgetful.
  • One skilled in the art will appreciate the variety of planning tools that would be appropriate for use with compounds and dosage forms described herein.
  • the kit can also include a container for storing the other components of the kit.
  • the container can be, for example, a bag, box, envelope or any other container that would be suitable for use with the compounds and dosage forms described herein.
  • the container is large enough to accommodate each component and/or any administrative devices that may be accompany the dosage form of the present invention. However, in some cases, it may be desirable to have a smaller container which can be hidden in a patient's pocketbook, briefcase, or pocket.
  • the present invention includes a kit comprising a pharmaceutical dosage form described herein.
  • the kit further comprises printed instructions for its use.
  • the kit further comprises a printed matter, a pre-recorded media device, or a planner describing the use of the pharmaceutical dosage form of the present invention to treat or prevent a condition which could be aided by taking the compositions disclosed herein.
  • the present invention provides a method of delivering a pharmaceutical dosage form described herein, to a patient in need thereof, the method comprising:
  • the access to the pharmaceutical dosage form is a prescription.
  • Still other aspects of the present invention include a method of educating a consumer regarding the pharmaceutical dosage forms described herein, the method comprising distributing the oral pharmaceutical dosage form to a consumer with consumer information at a point of sale.
  • the consumer information is presented in a format selected from the group consisting of: English language text, a foreign language text, a visual image, a chart, a telephone recording, a website, and access to a live customer service representative.
  • the consumer information is a direction for use, appropriate age use, indication, contraindication, appropriate dosing, warning, telephone number, or website address.
  • the method of educating the consumer further comprises providing professional information to a relevant person in a position to answer a consumer question regarding the pharmaceutical dosage form.
  • the relevant person is a physician, physician assistant, nurse practitioner, pharmacist, or customer service representative.
  • the distributing of the pharmaceutical dosage form is to a location with a pharmacist or a health care provider.
  • This example demonstrates phosphine catalysis of polymer-bound allenoates and a combinatorial library approach to the development of potent inhibitors of protein geranylgeranyltransferase type I (GGTase-I). These methods are applicable to synthesizing the “left hand” (e.g., the A group in Formula 1) of the compounds of the present invention having the particular scaffolds described herein.
  • a collection of 138 heterocycles was screened for their ability to inhibit the activity of human GGTase-I to geranylgeranylate K-Ras4B or RhoA.
  • Purified GGTase-I was incubated with its substrate protein K-Ras4B or RhoA, [ 3 H]GGPP, and the 138 compounds. After 30 min, the degree of incorporation of tritiated geranylgeranyl groups was measured using a scintillation counter.
  • GGTIs A number of compounds were identified as GGTIs including the following compounds numbered 1 and 2:
  • the phosphine-catalyzed annulation between solid-bound polymer-supported allenoates 5 and N-tosylimines proceeded smoothly.
  • the allenoates 5a and 5b were treated with N-tosyltolualdimine and 50 mol % of PPh 3 (for 5a) or PBu 3 (for 5b) in benzene at 60° C. to provide the polymer-bound dihydropyrroles 6.
  • Tetrahydropyridines 7 were formed from the reactions of 5c and 5d with N-tosyltolualdimine in the presence of 50 mol % of PBu 3 at room temperature for 2 and 4 days, respectively.
  • the ⁇ , ⁇ -unsaturated enoate functionalities in 6 and 7 were utilized to further increase the modularity and number of analogs.
  • the Michael additions of thiols to 6 and 7 using n-butyllithium as base provided 10 and 11, respectively, which upon TFA-mediated cleavage yielded 12 and 13, respectively, in 77-95% yield (Scheme 2).
  • Scheme 2 The two-step sequences occurred with high diastereoselectivities, providing the pentasubstituted pyrrolidine 12 and the tetrasubstituted piperidine 13 as single diastereoisomeric products.
  • ⁇ -Substituted allenoic acids B were prepared through saponification of the esters 19.
  • the N-sulfonylimines C were formed simply through azeotropic removal of water from a mixture of the appropriate sulfonamide 20, aldehyde 21, and BF 3 .OEt 2 under reflux in toluene.
  • Chart 1 presents the building blocks synthesized as illustrated in Scheme 3 and the commercially available thiol building blocks D.
  • Chart I Eleven ⁇ -substituted allenoic acids A, 12 ⁇ -substituted allenoic acids B, 46 N-sulfonimines C, and 32 thiols D.
  • the 4288 lanterns were inserted into 4288 vials and treated with 2.5% TFA in CH 2 Cl 2 for 12 h; the lanterns were then removed and rinsed with CH 2 Cl 2 .
  • the resulting solution was concentrated and further co-evaporated with CHCl 3 to effectively remove TFA.
  • the cleaved compounds were weighed and redissolved in CHCl 3 ; a portion (2 mop of each compound was transferred into 54 96-well plates (80 compounds per well; two columns of wells in each plate were left empty to accommodate controls in subsequent assays) and the solvents were left to evaporate.
  • the products were redissolved in DMSO and analyzed in the same assay for activity against GGTase-I.
  • FIG. 33 shows Western blot of the cell lysate qualitatively detecting the amount of germanylgeranylated Rheb.
  • the descriptors P and U designate processed and unprocessed Rheb, respectively.
  • Examples 2-8 illustrate assays of bioactivity. These methods are applicable to the compounds of the present invention having the particular linkers and scaffolds described herein, to determine their activity.
  • compounds P3-E5 and P5-H6 specifically inhibit GGTase I.
  • the graph illustrates the effect of P3-E5 and P5-H6 on the enzymatic activity of GGTase-I (A), FTase (B) GGTase-II (C). Varying concentrations of the two compounds were added to each enzyme reaction.
  • FTase protein farnesyltransferase
  • K-Ras4B as a substrate protein
  • FTase JENA Bioscience, San Diego, Calif.
  • [3H]farnesyl pyrophosphate was carried out using K-Ras4B as a substrate protein, FTase (JENA Bioscience, San Diego, Calif.) and [3H]farnesyl pyrophosphate. Incubation was for 30 min at 37° C.
  • the GGTase-I assay was carried out using RhoA as a substrate protein, GGTase-I (JENA Bioscience) and [3H]geranylgeranyl pyrophosphate. Incubation was for 30 min at 37° C.
  • GGTase-II (or RabGGTase) assay was carried out using YPT1 as a substrate protein, GGTase-II (Calbiochem, San Diego, Calif.) plus REP1 (Calbiochem) and [3H]geranylgeranyl pyrophosphate. Incubation was for 30 min at 37° C.
  • FIGS. 20D-20F show the three dimensional structure of GGTase I (D), FTsse (E) and GGTase II (F) which can be obtained from X-ray diffraction studies of protein crystals.
  • FIGS. 21A-21D P3-E5 and P5-H6 compete with a substrate protein but do not compete with GGPP.
  • the graphs in FIG. 21 show double reciprocal plots obtained from substrate velocity curves for the inhibition of GGTase-I by P3-E5 (left) and P5-H6 (right).
  • the upper figures show varying GGPP concentrations with a fixed RhoA protein concentration were used.
  • the lower figures show varying RhoA protein concentrations with a fixed GGPP concentration was used.
  • the following scale was used to prepare the graphs: Y-axis: 1/v, fmol/min.
  • X-axis 1/s, mM.
  • P3-ES inhibits geranylgeranylation in cells.
  • Human embryonic kidney (HEK) 293 cells expressing Myc-HA tagged-Rheb-CVSL (upper) or Myc-HA tagged-Rheb-WT (lower) were treated with P3-ES for 48 hours and then lysed.
  • the cell lysates were run on a SDS polyacrylamide gel and immunoblotted using anti-Myc or anti-Actin antibody.
  • the descriptors P and U indicate processed and unprocessed Rheb, respectively. Similar results have been obtained with P5-H6 but are not depicted in FIG. 22 .
  • K562 cells are a cell line of human erythroleukemia cells.
  • the K562 cells were treated with P3-ES, PS-H6 or GGTI-298 (a known GGTI compound illustrated in FIG. 27 ) for 72 hours and then cell number was counted using cell counting kit-8 (Dojindo, Gaithersburg, Md.). Cell viability relative to the DMSO control and the known GGTI compound is plotted in FIG. 23 . It can be seen that both P3-ES and P5-H6 inhibit the growth of the K562 cells.
  • P3-E5 induces G1 arrest in K562 cells.
  • K562 cells were treated with indicated concentrations (mM) of P3-E5 (ranging from 0 to 10 mM) or GGTI-298 (ranging from 0 to 10 mM) for 48 hours.
  • Cell cycle profiles were monitored by flow cytometry. Percentages of cells in each phase of the cell cycle are indicated by different colors. Red: G0/G1 phase. Black: S phase. Gray: G2/M phase. Similar results were obtained with P5-H6.
  • FIG. 24B illustrates an exemplary mechanism to explain the cell cycle effect of GGTIs.
  • RhoA is known to inhibit expression of p21 WAF.
  • GGTI may function by inhibiting geranylgeranylation of RhoA leading to the increase in p21WAF expression. This could result in G1 arrest.
  • mP5-H6 inhibits geranylgeranylation in vivo.
  • Human embryonic kidney (HEK) 293 cells expressing Myc-HA tagged-Rheb-CVSL (upper) or Myc-HA tagged-Rheb-WT (lower) were treated with mP5-H6a (also referred to as mP5-H6), or GGTI-298 for 48 hours and then lysed. Lysates were run on a SDS polyacrylamide gel and immunoblotted with anti-Myc or anti-Actin antibody. Processed and unprocessed Rheb may be used in the method described in this example as shown in FIG. 22 .
  • mP5-H6 exhibits increased potency to inhibit proliferation relative to P5-H6.
  • Panc-1 cells Pancreatic cancer cell line
  • Jurkat cells T-cell leukemia
  • the cell number was counted as described in Example 5.
  • the graph shows that mP5-H6a inhibits the growth of Panc-1 cells and Jurkat cells better than P5-H6.
  • NIH3T3 cells were maintained in Dulbecco's modified Eagle's medium (DMEM; Cellgro, Herndon, Va.) supplemented with 10% (v/v) fetal bovine serum (FBS; Hyclone, Logan, Utah), 2% L-glutamine, 1% penicillin, and 1% streptomycin stock solutions (Life Technologies, Gaithersburg, Md.).
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • streptomycin stock solutions Life Technologies, Gaithersburg, Md.
  • K562 cells were maintained in RPMI-1640 medium (Cellgro) supplemented with 10% (v/v) FBS and penicillin/streptomycin.
  • PANC-1 cells were maintained in DMEM/F12 medium (Invitrogen, Grand Island, N.Y.) supplemented with 10% (v/v) FBS and penicillin/streptomycin.
  • MCF-7 cells were maintained in Eagle's Minimum Essential Medium (EMEM; Cellgro) supplemented with 10% (v/v) FBS and penicillin/streptomycin.
  • the allenoate derived compounds library including P3-E5 and P5-H6 were synthesized as described in (28). Examples of suitable methods for the synthesis of P63-F10, P63-C7, P63-E11, P62-A5, P62-C11, P62-E4 and modified P5-H6 compounds (P61-A2, P61-A5, P61-A6, P61-A7 and P61-B4) have been described above.
  • FTase or GGTase-I 50 nM were used to initiate reactions containing 0.4 ⁇ M of [ 3 H]-FPP or 0.5 ⁇ M of [ 3 H]-GGPP and 2 ⁇ M of MBP-tagged substrates (K-Ras4B for FTase; RhoA for GGTase-I) in 20 ⁇ l of buffer ⁇ 50 mM Tris-HCl (pH 7.5), 10 mM MgCl 2 , 50 mM ZnCl 2 and 5 mM DTT ⁇ . Inhibitors were added at the indicated concentrations. The final DMSO concentration was 2.5% for all samples.
  • the reaction contained the following components in 20 ⁇ l; 0.625 ⁇ l of [ 3 H]-GGPP (0.7 ⁇ M), 25 nM RabGGTase, 0.6 ⁇ M REP-1, 0.6 ⁇ M purified Rab7 or Ypt1 protein, 40 mM HEPES (pH 7.5), 150 mM NaCl, 5 mM DTT, 3 mM MgCl 2 and 0.3% CHAPS. Reactions were carried out for 20 min at 37° C. and the products were analyzed as described above for the GGTase-I reaction. Graphing and Michaelis-Menten analysis were performed using Prism versions (GraphPad, San Diego Calif.).
  • FIG. 43 The inhibition of Rab geranylgeranylation in cells was examined according to (29). Briefly, whole cell lysates were subjected to 15% SDS-PAGE containing 4M urea followed by immunoblotting with the antibody against Rab5b (Santa Cruz: sc-598) or actin. Subcellular fractionations ( FIG. 44 ) were performed as described by Gomes et al. (30). Briefly, cells were treated with P49-F6 for 48 hours. After osmotic lysis, cell debris were removed by centrifugation at 500 ⁇ g for 10 min, and the supernatant was subjected to ultracentrifugation at 100,000 ⁇ g for 60 min.
  • Cell viability was determined by Cell Counting Kit-8 (CCK-8, Dojindo, Kumamoto, Japan) as described previously (31). Briefly, cells (5 ⁇ 10 3 ) were plated onto 96-well plates and treated with the appropriate inhibitor as indicated in figure legends. Cell viability was calculated relative to the DMSO control. Cell cycle profile was analyzed by flow cytometry as described previously (32).
  • NIH3T3 cells were transfected with p21 WAFI/CIPI promoter-Luc or vector plasmids (33) (both plasmids are provided by Dr. Genhong Cheng). Cells were treated with GGTI compounds. Promega luciferase assay kit was used according to the manufacturer's protocol.
  • Allenoic acid based chemical compound library and identification of GGTase-I inhibitors As described above, we have reported construction of a library of allenoate derived compounds and identification of novel GGTI compounds (28). The library construction involved the use of allenoates as multireactive core molecules. Using the second set of building blocks (imines, aldehydes and maleimides) that react with allenoates under similar reaction conditions (phosphine catalysis), we have produced diverse compounds including dihydropyrroles (34), tetrahydropyridines (35), bicyclic succinimides (unpublished results), dioxanylidenes (36) and alpha-pyrones (37).
  • building blocks imines, aldehydes and maleimides
  • phosphine catalysis we have produced diverse compounds including dihydropyrroles (34), tetrahydropyridines (35), bicyclic succinimides (unpublished results), dioxanylidenes (36) and alpha-pyrones (37).
  • GGTI compounds from within this library was carried out by screening a 171-compound pilot library using an in vitro assay with RhoA protein as a substrate. Scaffolds that initially showed activity were optimized by solid-phase split-and-pool combinatorial synthesis. This enabled us to identify at least two types of novel compounds; one group containing a tetrahydropyridine ring as its core scaffold and the other group having a dihydropyrrole ring as its core scaffold.
  • P3-E5 and P5-H6 were further characterized. As described herein, P3-E5 and P5-H6 inhibit GGTase-I with IC 50 values of 313 and 466 nM, respectively.
  • the specificity of GGTase-I inhibition by P3-E5 and P5-H6 was examined by assaying their ability to inhibit two closely related enzymes, FTase and RabGGTase. As can be seen, no significant inhibition of FTase activity was observed by these compounds even when the concentration was increased to 50,000 nM ( FIG. 34 ). Similarly, P3-E5 showed little inhibition against RabGGTase even at 50,000 nM. P5-H6 showed little inhibition against RabGGTase up to 10,000 nM.
  • GGTIs compete with substrate protein.
  • Michaelis-Menten analysis of the inhibition of GGTase-I is shown in FIG. 35 .
  • the upper panels show the data derived from the results obtained using varying concentrations of geranylgeranyl pyrophosphate (GGPP), while the lower panels show data derived from the results obtained using varying concentrations of the substrate protein, RhoA.
  • GGPP geranylgeranyl pyrophosphate
  • RhoA the substrate protein
  • P3-E5 and P5-H6 are competitive inhibitors with respect to the protein substrate and uncompetitive inhibitors with respect to GGPP.
  • P3-E5 and P5-H6 compete for binding of the protein substrate but not the isoprenoid substrate of GGTase-I. Ki values of 187 ⁇ 13 nM and 408 ⁇ 32 nM, respectively, were calculated for P3-E5 and P5H6.
  • the improved potency of these compounds to inhibit cell proliferation correlates with their increased ability to inhibit protein geranylgeranylation inside the cell.
  • Results illustrating this correlation using P5-H6 and P61-A6 are shown in FIG. 36C .
  • the inhibition of protein geranylgeranylation can be evaluated using an antibody that specifically detects unprenylated Rap1.
  • Treatment with P5-H6 or P61-A6 led to the appearance of the unprocessed Rap1 band in a dose dependent manner.
  • the appearance of the Rap1 band is observed at 2.5 ⁇ M concentration with P61-A6, while this is not seen with P5-H6, reflecting improvement in the potency of P61-A6 to inhibit protein geranylgeranylation.
  • P61-A6 did not inhibit protein farnesylation. This was examined by using a farnesylated protein H-Ras. While farnesyltransferase inhibitor (FTI) (BMS-225975) slowed the mobility of H-Ras protein on a SDS polyacrylamide gel, no such mobility shift was observed with P61-A6 or another GGTI compound, GGTI-298. Similarly, P61-A6 did not inhibit geranylgeranylation of Rab5b, as a slow migrating band representing that of unmodified Rab5b was detected only after the treatment with RabGGTase inhibitors (P49-F6) and not with P61-A6 ( FIG. 36E ).
  • FTI farnesyltransferase inhibitor
  • P61-A6 exhibits improved ability to inhibit geranylgeranylation in cells, its ability to inhibit GGTase-I enzyme was less than that of P5-H6, as the IC 50 value for the enzyme inhibition was 1 ⁇ M. No significant inhibition of FTase or RabGGTase activity was observed by P61-A6 compound even when the concentration was increased to 100,000 nM (data not shown).
  • GGTI compounds inhibit proliferation of various human cancer cell lines and cause G 1 cell cycle arrest. As shown in Table S1, inhibition of cellular proliferation was observed in a variety of human cancer cell lines. These results indicate that a broad range of human cancer cell lines can be inhibited using the GGTI compounds described herein.
  • the inhibition of cellular proliferation by GGTI can be due to the inhibition of cell cycle progression.
  • treatment of a breast cancer cell line (MCF-7) with P61-A6 or P61-B6 caused dose-dependent inhibition of proliferation. This is associated with a significant dose-dependent enrichment of G 1 phase cells, while the percentage of S-phase cells decreased ( FIG. 37B , FIG. 41 ).
  • Similar G 1 enrichment was observed with leukemic cell line, Jurkat, two pancreatic cancer cell lines, PANC-1 and MiaPaCa2, as well as with another breast cancer cell line MDA-MB-231 (Table S2).
  • GGTI Another mechanism through which GGTI can effect cell cycle progression is to inhibit RhoA which negatively regulates expression of a Cdk inhibitor p21 CIPI/WAFI .
  • RhoA a Cdk inhibitor
  • luciferase transcriptional activation from the p21 CIPI/WAFI promoter (33) was measured.
  • Transient expression systems with NIH3T3 cells were used to examine the ability of P61-A6 to induce p21 CIPI/WAFI -luciferase expression.
  • P61-A6 induced significant (4-fold) inductions of luciferase activity versus DMSO in a dose dependence manner ( FIG. 37C ).
  • FIG. 38 shows four examples of compounds (P8-G7, P8-H6, P8-H7 and P49-F5) that exhibit inhibition of both GGTase-I and RabGGTase at a single ⁇ M range. On the other hand, these compounds do not inhibit FTase even with more than 100 ⁇ M concentration ( FIGS. 39B and C).
  • these compounds share a common structural feature.
  • These dual specificity compounds all have a characteristic six-atom aliphatic tail attached to the penta-substituted pyrrolidine core via thioether linkage (putative RabGGTI feature) to the 5-member ring core.
  • FIG. 39 shows the structure and IC 50 values for five example RabGGTase preferential inhibitors, P23-D6, P47-D11, P49-A6, P49-F6 and P50-E11.
  • n-hexylmercapto substituent at C4 of the pyrrolidine ring.
  • n-pentyl thioether P47-D11
  • para-methoxyphenyl thioether P23-D6
  • these compounds inhibit RabGGTase with an IC so value of about 2-5 ⁇ M, while the inhibition of GGTase-I required more than 50 ⁇ M. No significant inhibition of FTase was observed using 100 ⁇ M of these compounds.
  • RabGGTI competes with the substrate protein.
  • Kinetic analysis was carried out to examine whether our RabGGTI compounds compete with substrate protein.
  • Rab geranylgeranyltransferase consists of a tightly bound core complex, the alpha and beta subunits, and the third subunit REP protein. These subunits were first mixed together in the presence of a low concentration of GGPP and then the concentration of each substrate was altered and the effect of inhibition by RabGGTI was examined.
  • a RabGGTI compound P49-F6 inhibits RabGGTase activity with respect to the substrate protein Rab7 ( FIG. 40B ).
  • increasing concentration of GGPP did not influence the inhibition by RabGGTI ( FIG. 8A ), showing that they are uncompetitive inhibitors with respect to GGPP.
  • a Ki value of 1.36 ⁇ 0.38 ⁇ M was calculated from these experiments.
  • the improvement of the cellular activity of our GGTI compound can be correlated with the increase in the ability to inhibit protein geranylgeranylation, as detected by the appearance of unprenylated Rapt protein.
  • the modification did not improve potency of these compounds to inhibit GGTase-I enzyme. Therefore, the improvement of cellular activity may reflect increased cellular uptake or stability of the compound.
  • the GGTI compounds described herein exhibit inhibition of proliferation of human cancer cell lines including leukemic, pancreatic cancer and breast cancer cell lines.
  • This class of inhibitors causes cell cycle arrest at the G I phase (38,39).
  • These inhibitors exhibit significant G I arrest with the human cancer cell lines examined ( FIG. 37B , Table S2).
  • dramatic G 1 arrest is observed with a breast cancer cell line MCF-7.
  • our GGTI compounds induce p21 CIPI/WAFI expression, as observed by using a luciferase reporter assay.
  • GGTase-I and RabGGTase share similar active site structures (16,26); both enzymes have a core structure that consists of alpha and beta subunits. In addition, the corresponding subunits in these enzymes share significant homology. Therefore, in some embodiments, these inhibitors bind to similar pockets in these enzymes.
  • RabGGTI compounds possess a structural feature that is unique to this group of inhibitors. They contain a characteristic long aliphatic tail attached to the penta-substituted pyrrolidine core. While not wishing to be bound to a single theory, it is believed that the aliphatic tail fits into a pocket or interferes with an enzymatic process that is specific to RabGGTase, but not to GGTase-I. For example, the significance of lipid binding pockets of REP for RabGGTase reaction has been suggested (18,43).
  • GGTI's can inhibit PANC-1 tumor growth in mice.
  • Seven SCID mice (six weeks old) received a subcutaneous implantation of 5 million PANC-1 cells. The mice were supplied with food and water for 14 days before treatment was started. The treatment group received 160 uM GGTI (p61A6) in 0.25 ml 0.9% NaCl while the control group received 0.25 ml 0.9% NaCl. Each dosage was injected 3 times per week. These dosage were designed to administer p61A6 at approximately 1.16 mg/kg of body weight. Also, since the mouse has about 2 ml blood, the final concentration of p61A6 in vivo was approximately 20 uM. The final DMSO concentration in the control group was approximately 0.8%.
  • FIGS. 44 and 45 and table S3 Results from the test are shown in FIGS. 44 and 45 and table S3 below. These results indicate that GGTI P61A6 inhibits PANC-1 xenograft tumor growth in SCID mice.
  • FIG. 45 and table S3 show examinations of body weight as well as hematological and biochemical examinations indicating that the GGTI treatment did not cause adverse effects.
  • mice Six SCID mice (six weeks old) received a subcutaneous implantation of 3 million PANC-1 cells. The mice were supplied with food and water for 14 days before treatment began. The mice were divided into the following treatment groups (all non-control groups administered P61-A6 at the dose listed): (1) control: 0.25 ml 0.9% Nacl; (2) treatment group 1: 1.16 mg/kg of body weight in 0.25 ml 0.9% Nacl, 1/week, i.p. (20 uM); (3) treatment group 2: 1.16 mg/kg of body weight in 0.25 ml 0.9% Nacl, 6/week, i.p.
  • treatment group 3 0.58 mg/kg of body weight in 0.25 ml 0.9% Nacl, 3/week, i.p. (10 uM); and (5) treatment group 5: 0.29 mg/kg of body weight in 0.25 ml 0.9% Nacl, 3/week, i.p. (5 uM). Every two weeks each mouse's body weight and tumor volume was measured.
  • compositions of the invention can be administered is a dosage regimen that is highly convenient, e.g., once weekly instead of daily.
  • the compositions of the present invention can be administered every 1, 2, 3, 4, 5, 6, or 7 days, or may be administered otherwise.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
US13/056,077 2008-07-28 2008-07-28 Inhibitors of protein prenyltransferases Abandoned US20110178138A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2008/009106 WO2010014054A1 (fr) 2008-07-28 2008-07-28 Inhibiteurs de prényltransférases de protéine

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/009106 A-371-Of-International WO2010014054A1 (fr) 2008-07-28 2008-07-28 Inhibiteurs de prényltransférases de protéine

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/614,789 Continuation US8815935B2 (en) 2008-07-28 2012-09-13 Inhibitors of protein prenyltransferases

Publications (1)

Publication Number Publication Date
US20110178138A1 true US20110178138A1 (en) 2011-07-21

Family

ID=41610593

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/056,077 Abandoned US20110178138A1 (en) 2008-07-28 2008-07-28 Inhibitors of protein prenyltransferases
US13/614,789 Active US8815935B2 (en) 2008-07-28 2012-09-13 Inhibitors of protein prenyltransferases

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/614,789 Active US8815935B2 (en) 2008-07-28 2012-09-13 Inhibitors of protein prenyltransferases

Country Status (2)

Country Link
US (2) US20110178138A1 (fr)
WO (1) WO2010014054A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019076269A1 (fr) 2017-10-16 2019-04-25 清华大学 Inhibiteur de la voie de l'acide mévalonique et composition pharmaceutique correspondante

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012097371A2 (fr) * 2011-01-14 2012-07-19 The Regents Of The University Of California Inhibiteurs chimiques de la biosynthèse du cholestérol et de l'angiogenèse
EP2712655B1 (fr) 2011-04-28 2019-12-18 The Broad Institute, Inc. Inhibiteurs de l'histone désacétylase
CA2880117C (fr) 2012-07-27 2021-04-06 The Broad Institute, Inc. Inhibiteurs d'histone-desacetylases
US9914717B2 (en) * 2012-12-20 2018-03-13 The Broad Institute, Inc. Cycloalkenyl hydroxamic acid derivatives and their use as histone deacetylase inhibitors
WO2016107542A1 (fr) * 2014-12-29 2016-07-07 成都先导药物开发有限公司 Composé pyrrole amide, son procédé de préparation et son utilisation
CN105801464B (zh) 2014-12-29 2019-05-28 成都先导药物开发有限公司 吡咯酰胺类化合物及其制备方法与用途
CN105732459B (zh) * 2014-12-29 2019-05-31 成都先导药物开发有限公司 吡咯酰胺类化合物及其制备方法与用途
CN105732597B (zh) * 2014-12-29 2019-01-01 成都先导药物开发有限公司 一种制备吡咯酰胺类化合物的中间体化合物及其制备方法与应用
EP3852755A4 (fr) * 2018-09-20 2022-06-08 The Regents of the University of California Énantiomère (r) d'aplexone
CN112574090B (zh) * 2020-11-23 2022-04-29 浙大城市学院 一种含硫的多取代吡咯类化合物及其制备方法

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559157A (en) * 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
US4608392A (en) * 1983-08-30 1986-08-26 Societe Anonyme Dite: L'oreal Method for producing a non greasy protective and emollient film on the skin
US4820508A (en) * 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
US4938949A (en) * 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
US4992478A (en) * 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same
WO2007111948A2 (fr) * 2006-03-22 2007-10-04 The Regents Of The University Of California Inhibiteurs de prényltransférases protéiques

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003259717A1 (en) * 2002-08-07 2004-02-25 Bristol-Myers Squibb Company Modulators of rabggt and methods of use thereof
CA2528652A1 (fr) 2003-06-19 2004-12-23 Pfizer Products Inc. Antagoniste de nk1

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559157A (en) * 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
US4608392A (en) * 1983-08-30 1986-08-26 Societe Anonyme Dite: L'oreal Method for producing a non greasy protective and emollient film on the skin
US4820508A (en) * 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
US4992478A (en) * 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same
US4938949A (en) * 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
WO2007111948A2 (fr) * 2006-03-22 2007-10-04 The Regents Of The University Of California Inhibiteurs de prényltransférases protéiques
US8093274B2 (en) * 2006-03-22 2012-01-10 The Regents Of The University Of California Inhibitors of protein prenyltransferases

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Castellano, et al., J.A.C.S., Vol. 129, pp. 5843-5845, published April 17, 2007. *
Watanabe, et al., J. Biological Chem., 283(15), pp. 9571-9579, published April 2008. *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019076269A1 (fr) 2017-10-16 2019-04-25 清华大学 Inhibiteur de la voie de l'acide mévalonique et composition pharmaceutique correspondante

Also Published As

Publication number Publication date
US8815935B2 (en) 2014-08-26
US20130102639A1 (en) 2013-04-25
WO2010014054A1 (fr) 2010-02-04

Similar Documents

Publication Publication Date Title
US8815935B2 (en) Inhibitors of protein prenyltransferases
US8093274B2 (en) Inhibitors of protein prenyltransferases
Li Petri et al. Pyrrolidine in drug discovery: a versatile scaffold for novel biologically active compounds
Bai et al. Peptidomimetic α-acyloxymethylketone warheads with six-membered lactam P1 glutamine mimic: SARS-CoV-2 3CL protease inhibition, coronavirus antiviral activity, and in vitro biological stability
Crisman et al. KEAP1‐NRF2 protein–protein interaction inhibitors: Design, pharmacological properties and therapeutic potential
US9162991B2 (en) Cinnamoyl inhibitors of transglutaminase
US20110201643A1 (en) Novel n-substituted tetrahydroisoquinoline/isoindoline hydroxamic acid compounds
Liang et al. Structure, functions and selective inhibitors of HDAC6
US9221761B2 (en) Method for inhibition of deubiquitinating activity
Zhu et al. Structure-based discovery of selective BRPF1 bromodomain inhibitors
EA017533B1 (ru) Соли (е)-n-(2-аминофенил)-3-{1-[4-(1-метил-1h-пиразол-4-ил)бензолсульфонил]-1h-пиррол-3-ил}акриламида
Li et al. Heat shock protein 90 (Hsp90)/Histone deacetylase (HDAC) dual inhibitors for the treatment of azoles-resistant Candida albicans
Matys et al. Imidazolidine-4-one derivatives in the search for novel chemosensitizers of Staphylococcus aureus MRSA: Synthesis, biological evaluation and molecular modeling studies
D'Amico et al. Targeting TRIM proteins: a quest towards drugging an emerging protein class
MX2007011072A (es) Hidroxamatos como inhibidores de la histona deacetilasa y formulaciones farmaceuticas que los contienen.
Ross et al. Targeting MYC: from understanding its biology to drug discovery
Hafez et al. Novel benzothiazole derivatives as multitargeted-directed ligands for the treatment of Alzheimer’s disease
Liu et al. Natural product-based proteolysis targeting chimeras (PROTACs)
Dai et al. Target fishing reveals a novel mechanism of 1, 2, 4-oxadiazole derivatives targeting Rpn6, a subunit of 26S proteasome
Kanada et al. Discovery of DS-9300: a highly potent, selective, and once-daily oral EP300/CBP histone acetyltransferase inhibitor
US20200278355A1 (en) Conjugated proteins and uses thereof
Saha et al. Novel triazole based inhibitors of Ras farnesyl transferase
Wang et al. Cysteine derivatives as acetyl lysine mimics to inhibit zinc-dependent histone deacetylases for treating cancer
Barbaraci et al. Discovery of first novel sigma/HDACi dual-ligands with a potent in vitro antiproliferative activity
Akella Development of New Drugs for the Treatment of Oral Cancers

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE REGENTS OF THE UNIVERSITY OF CALIFORNIA, CALIF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KWON, OHYUN;TAMANOI, FUYUHIKO;FIJI, HANNAH;AND OTHERS;SIGNING DATES FROM 20090422 TO 20090427;REEL/FRAME:023360/0923

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF CALIFORNIA LOS ANGELES;REEL/FRAME:025750/0205

Effective date: 20110203

AS Assignment

Owner name: THE REGENTS OF THE UNIVERSITY OF CALIFORNIA, CALIF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KWON, OHYUN;FIJI, HANNAH D.G.;TAMANOI, FUYUHIKO;AND OTHERS;SIGNING DATES FROM 20110329 TO 20110402;REEL/FRAME:026137/0357

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION