AU2017204046A1 - Compositions and methods for enhancing proteasome activity - Google Patents

Compositions and methods for enhancing proteasome activity Download PDF

Info

Publication number
AU2017204046A1
AU2017204046A1 AU2017204046A AU2017204046A AU2017204046A1 AU 2017204046 A1 AU2017204046 A1 AU 2017204046A1 AU 2017204046 A AU2017204046 A AU 2017204046A AU 2017204046 A AU2017204046 A AU 2017204046A AU 2017204046 A1 AU2017204046 A1 AU 2017204046A1
Authority
AU
Australia
Prior art keywords
compound
alkyl
proteasome
hydrogen
haloalkyl
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
AU2017204046A
Inventor
Daniel Finley
Timothy C. Gahman
Randall W. King
Byung-Hoon Lee
Min Jae Lee
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.)
Harvard College
Original Assignee
Harvard College
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2011210765A external-priority patent/AU2011210765A1/en
Application filed by Harvard College filed Critical Harvard College
Priority to AU2017204046A priority Critical patent/AU2017204046A1/en
Publication of AU2017204046A1 publication Critical patent/AU2017204046A1/en
Abandoned legal-status Critical Current

Links

Abstract

Abstract Proteinopathies result from the proteasome not acting efficiently enough to eliminate harmful proteins and prevent the formation of the pathogenic aggregates. As described herein, inhibition of proteasome-associated deubiquitinase Uspl4 results in increased proteasome efficiently. The present invention therefore provides novel composition and methods for inhibition of Uspl4, enhancement of proteasome activity and treatment of proteinpathies.

Description

Compositions and Methods for Enhancing Proteasome Activity
RELATED APPLICATIONS
This application claims the benefit of priority to United States Provisional Patent Application serial number 61/373,404, filed August 13,2010, and United States Provisional Patent Application serial number 61/336,959, filed January 28, 2010; the contents of both of which are hereby incorporated by reference.
GOVERNMENT SUPPORT
This invention was made with U.S. Government support under National Institutes of Health Grant Nos. GM065592, GM66492, and DK082906. The government has certain rights in the invention.
BACKGROUND
The proteasome is a large protein complex that contains 33 distinct subunits. Proteasome complexes function as proteases in part to degrade unneeded or misfoided proteins. Protcasomcs regulate many aspects of cell physiology, and proteasome dysfunction has been implicated in a variety of diseases, including cancer and neurodegenerative diseases (Finley D., (2009), Amur Rev. Biochem., 78, 477-513; Hoellcr and Dikic, (2009), Nature, 458, 438-444; Demarto and Gillette, (2007), Cell, 129, 659-662); Dahlmann, B. (2007) BCB Biochem 8, Suppl 1, S3; Schartz AL and Ciechanover A (2009) Ann Rev Pharmacol Toxicol 49, 73-96).
Most, but not all, proteasome substrates arc targeted for degradation via the covalent attachment of muitimcric chains of a small, highly-conserved protein called ubiquitin. Because longer ubiquitin chains interact more strongly with the proteasome than shorter chains (Thrower et a!. (2000), EMBO J. 19,94-102), processes that alter ubiquitin chain length frequently also affect substrate degradation rates. The length of ubiquitin chains attached to substrates tagged for proteasome degradation can be modulated by certain proteasome-associated deubiquitinating enzymes and ubiquitin ligases. These deubiquitinating enzymes and ligases appear to regulate proteasome activity by disassembling or extending protcasome-bound ubiquitin chains.
Mammalian protcasomcs contain three major deubiquitinating enzymes: Rpnl 1, Uch37, and Uspl4 (Finley D., (2009), Annu. Rev. Biochem,, 78, 477-513). Rpnl 1 removes ubiquitin from the tagged substrate by cutting at the junction between the ubiquitin chain and the substrate. Because the Rpnl 1-mediated cleavage occurs following a substrate's commitment to proteolysis, but prior to substrate degradation, Rpnl 1 helps to prevent ubiquitin from being degraded along with the substrate, thus minimizing fluctuations in cellular ubiquitin levels. Additionally, because the proteasome substrate must pass through a narrow translocation channel before encountering the protcasome’s sequestered proteolytic sites, removal of a bulky ubiquitin chain may also facilitate substrate translocation. Thus, removal of the ubiquitin chain by Rpnl 1 promotes substrate degradation through cn bloc removal of the ubiquitin chain at a relatively late step in the proteasome pathway (Verma et a/., (2002) Science, 298, 611-615; Yao and Cohen, (2002), Nature, 419, 403-407).
In contrast to Rpnl 1, Uch37 functions prior to the commitment of a substrate to proteasome degradation. Uch37 disassembles ubiquitin chains at the substrate-distal tip (Lam et«/., (1997), Nature, 385, 737-740), and its enzymatic activity shortens chains rather than remove them entirely. It lias been proposed that chain trimming by Uch37 increases the ability of the proteasome to discriminate between long and short multiubiquitin chains (Lam et al., (1997), Nature, 385, 737-740). Little is known about how Uch37 may regulate proteasome function in cells.
Very little is known about the function of Uspl4. However, the yeast ortholog of Uspl4, Ubp6, has been suggested to disassemble ubiquitin chains at the substrate-distal tip and to function prior to the commitment of a substrate to proteasome degradation. (Hanna eta!., (2006), Cell, 127(7), 1401-1413). Ubp6 is thought to act as a proteasome inhibitor, and prior work on Ubp6 has indicated a noncatalytic mode of proteasome inhibition (Hanna eta!., (2006), Cell, 127(7), 1401-1413).
SUMMARY
The present invention provides novel compositions and methods for the inhibition of Uspl4, the enhancement of proteasome activity and the treatment of proteinopathies and other diseases for which enhanced protein breakdown may be therapeutic. Aside from proteinopathies, the enhancment of proteasome activity may be therapeutic for any disease characterized by deficient proteasome activity, or deficient activity of other components of the ubiquitin-proteasome pathway, such as in von Hippel-Lindau disease, spinocerebellar ataxia 1, Angelman syndrome, giant axon neuropathy, inclusion body myopathy with Paget disease of bone and frontotemporal dementia (1BMPFD), and others (Lehman, N. L., (2009), Acta Ncuropathologica, 118(3), 329-347;Wcihl et al., (2007), Neuromuscular
Disorders, / 7, 87-87). Enhancing protcasome activity could also be therapeutic for diseases in which proteasomc substrates arc involved and contribute to pathology, but which do not satisfy a strict definition of proteopathies. For example, numerous oncoproteins are proteasome substrates and their ability to promote cancer could potentially be attenuated by enhancing proteasome activity.
One aspect of the invention relates to a compound represented by formula I, II, III, IV, V, VI, VII, or VIII, or pharmaceutically acceptable salt, solvate, hydrate, prodrug, chcmically-protcctcd form, enantiomer or stereoisomer thereof; wherein the formula are as defined below.
Another aspect of the invention relates to a method of inhibiting the dcubiquitination activity of a Uspl4 protein comprising contacting the Uspl4 protein with IU1 or a compound of formula I, II, III, IV, V, VI, VII, or VIII, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof.
Another aspect of the invention relates to a method of enhancing protein degradation by a proteasome in a cell comprising contacting the cell with 1U1 ora compound of formula I, II, III, IV, V, VI, VII, or VIII, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chcmically-protcctcd form, enantiomer or stereoisomer thereof.
Another aspect of the invention relates to a method of treating or preventing a proteinopathy in a subject comprising administering to the subject IU1 or a compound of formula I, II, III, IV, V, VI, VII, or VIII, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, or a pharmaceutical composition comprising the same.
Another aspect of the invention relates to a method of enhancing proteasome function in a subject comprising administering to the subject IU1 or a compound of formula I, II, III, IV, V, VI, VII, or VIII, ora pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, or a pharmaceutical composition comprising the same.
Another aspect of the invention relates to a method of increasing degradation of Tau, TDP-43 or ataxin-3 in a subject comprising administering to the subject IU1 or a compound of formula I, II, III, IV, V, VI, VII, or VIII, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, or a pharmaceutical composition comprising the same.
Another aspect of the invention relates to an isolated protcasomc lacking enzymatically active Uch37 and comprising enzymatically active Uspl4. In certain embodiments the proteasome comprises enzymatically inactive Uch37 and/or vinylsulfone-Uch37 adducts. In some embodiments the enzymatically active Uspl4 is a recombinant protein. In certain embodiments the proteasome is a human proteasome or a murine proteasome.
Another aspect of the invention relates to a method of generating a proteasome comprising enzymatically inactive Uch37 and further comprising enzymatically active Uspl4 comprising purifying a proteasome lacking Uspl4 but comprising Uch37, treating the purified protcasomc with a dcubiquitinase inhibitor, and reconstituting the purified proteasome with enzymatically active Usp] 4. In certain embodiments the proteasome is a human proteasome or a murine proteasome. in some embodiments the proteasome is purified from HEK293 cells. In some embodiments the dcubiquitinase inhibitor is ubiquitin-vinylsulfone. In certain embodiments the active Uspl4 is recombinantly produced.
Another aspect of the invention relates to a method of screening for an inhibitor of Uspl4 comprising providing a protcasomc comprising enzymatically inactive Uch37 and further comprising enzymatically active Uspl4, contacting the protcasomc with a test compound and a Usp 14 substrate, and determining whether the test compound inhibits the deubiquitination of the substrate. In certain embodiments, the substrate is coupled to a reporter that is detectable after cleavage by a deubiquitinase and/or is an ubiquitin-dependant proteasome substrate. In some embodiments the substrate is Ub-AMC or polyubiquitinated cycltn B. in certain embodiments, deubiquitination of the substrate is demonstrated by inhibition of substrate degradation. In some embodiments the proteasome comprises vinylsulfonc-Uch37 adducts. In certain embodiments the Usp 14 is a recombinant protein. In some embodiments the protcasomc is a human proteasome or a murine proteasome.
Another aspect of the invention relates to a kit comprising an isolated proteasome lacking enzymatically active Uch37 and comprising enzymatically active Usp 14, and instructions of use. In certain embodiments, the kit can comprise a UspI4 substrate. In some embodiments the Usp 14 substrate is Ub-AMC and/or polyubiquitinated cycltn B.
Additional aspects, embodiments, and advantages of the invention are discussed below in detail. Moreover, the foregoing information and the following detailed description are merely illustrative examples of various aspects and embodiments of the invention, and are intended to provide an overview or framework for understanding the nature and character of the claimed aspects and embodiments.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure IA shows an immunoblot that was performed using either recombinant Uspl4 protein (Purified Usp14) or affinity-purified Uspl4 deficient human protcasomcs (Human Protcasomc) and anti-Usp 14 antibody. The band corresponding to Uspl4 is indicated.
Figure IB shows an immunoblot that was performed using anti-Uch37 antibody and Usp 14-deficient purified human protcasomes (26S) either untreated (-VS) or treated with Ub-VS (+VS). The band corresponding to Uch37 is indicated. Nonspecific bands are indicated with an asterisk.
Figure 2A shows a nondenaturing gel analysis that had undergone in-gel suc-LLVY-AMC staining (indicating presence of protcasomcs) that was performed using commercially available human protcasomcs (Biomol), untreated, purified Usp 14 deficient human protcasomes (-Ub-VS) or Ub-VS treated purified Usp 14 deficient human protcasomcs (+Ub-VS).
Figure 2B shows a Coomassie Brilliant Blue (CBB) staining of purified, recombinant wild-type Usp 14 (Uspl4-wt) or catalytically inactive mutant Usp 14 (Usp 14-C114A) either with or without a GST tag, along with a GST control (GST) and a protein size marker (Marker).
Figure 2C shows the results of a gel-shift assay of protcasomcs alone (-), GST and protcasomcs (GST), GST tagged wild-type Usp 14 and protcasomcs (GST-Uspl4-wt), GST tagged catalytically inactive mutant Usp 14 and protcasomcs (GST-Uspl4-Cl 14A), untagged wild-type Usp 14 and protcasomcs (Uspl4-wt) or untagged catalytically inactive mutant Uspl4 and protcasomes (Uspl4-Cl 14A) that had either been stained with in gel suc-LLVY-AMC staining (top, to show the presence of protcasomes) or Coomassie Brilliant Blue (CBB) staining.
Figure 3 shows the results of a Ub-AMC hydrolysis assay for Usp 14 activity in the presence or Ub-VS treated human protcasomcs.
Figure 4A shows a plot of the linear kinetics (R2 > 0.99) of the initial rates of Ub- AMC hydrolysis by Uspl4 and protcasome at 1 μΜ Ub-AMC, I nM protcasomc, and the indicated concentration of Uspl4,
Figure 4B shows a Michaelis-Menten plot of Usp 14-dependent Ub-AMC hydrolysis in the presence of human proteasome for 25 minutes at 1 μΜ Ub-AMC, 1 nM proteasome, and the indicated concentration of Usp 14,
Figure 4C shows a plot of the linear kinetics (R2 > 0.99) of the initial rates of Ub-AMC hydrolysis by Usp 14 and proteasome at 4 nM Usp 14, I nM proteasome and the indicated concentration of Ub-AMC.
Figure 4D shows a Michaelis-Menten plot of concentration-dependent Ub-AMC hydrolysis in the presence of Usp 14 and human protcasomc for 30 minutes at 4 nM Usp 14, I nM protcasome and the indicated concentration of Ub-AMC.
Figure 5 shows an immunobfot that was performed using an antibody specific for Cyclin B, which also detects polyubiquitinated Cycltn B (Ubn-ClbB). In this experiment, Ub„-ClbB was treated with 26S human proteasome alone, human proteasome and wild-type Usp 14 (Uspl4-wt) or human proteasome and catalytically inactive UspI4 (Uspl4-CA), and subsequently analyzed by immunoblotting.
Figure 6A shows a diagram of human Usp 14, depicting the ubiquitin-like domain (UBL), the catalytic domain (CAT), the location of exon 4 and the position of Cysl 14.
Figure 6B shows immunoblots that were performed on cellular lysates from human 293 cells that co-cxprcsscd Tau along with cither wild-type Uspl4 (UspI4-wt), catalytically inactive Usp 14 (Uspl4-CA), short form Usp 14 (Uspl4-SF) or UBL domain deficient Uspl4 (Uspl4-AUBL) and stained using antibodies specific for Tau, UspI4 or Actin, as indicated.
Figure 7 shows immunoblots that were performed on cellular lysates from 293 ceils that co-expressed the indicated forms of flag-tagged Usp 14 along with tagged hRpnl 1 either before (Extract) or after (Purified proteasome) proteasome affinity purification and stained using anti-Flag antibody. Where indicated, Ub-VS was incubated with lysate prior to proteasome purification. Extract samples represent 5% of total. Asterisks, nonspecific signals. Protcasomc subunit Rpn 13, load control. Control samples, empty vector.
Figure 8A shows a statistical plot of the high-throughput large scale compound screening for inhibitors of Usp 14 catalytic inhibitors.
Figure 8B shows a frequency distribution curve used to determine AMC quenching compounds (control = bottom curve).
Figure 9A shows the chemical structure of Uspl4 inhibitor IU1.
Figure 9B shows a graph depicting (left) the inhibition of Uspl 4 loaded proteasomc (Uspl4-Ptsm) deubiquitinase activity by 1U1 (left bar = 0 μΜ, second to left bar = 4 μΜ, third bar = 8 μΜ, right bar = 17 μΜ) and (right) the lack of quenching of AMC fluorescence by 1U1 (left bar = 0 μΜ, right bar = 17 μΜ).
Figure 9C shows a graph depicting (left) the inhibition of Uspl 4 loaded proteasome (Uspl4-Ptsm) deubiquitinase activity by IU3 (left bar = 0 μΜ, middle bar = 8 μΜ, right bar = 17 μΜ) and (right) the lack of inhibition of Isopeptidasc T (IsoT) by !U1 (left bar = 0 μΜ, middle bar = 8 μΜ, right bar = 17 μΜ).
Figure 9D shows a graph depicting the lack of inhibition of Uspl 4 that has not been complcxcd to the proteasomc by cither vehicle (DMSO), 1U1 or 1UIC,
Figure 9E shows a graph depicting the lack of inhibition of 26S human proteasomes (Ptsm) that had not been treated with Ub-VS by either vehicle (DMSO) or IU1 (i.c, proteasomes which lack Uspl4).
Figure 10A shows the chemical structure of IU1C, an inactive control compound forlUl.
Figure I OB shows a plot comparing the deubiquitinase inhibition activity of IU 1 (bottom circles, bottom triangles) with the deubiquitinase inhibition activity of 1U1C (top circles, top triangles, squares).
Figure IOC shows the ineffectiveness of IU1C in promoting Tau degradation. Immunoblots were performed using lysates of MEF cells that co-expressed Tau and Uspl4 and that were treated with 0,25, 50,75 or 100 μΜ 1U1C and stained with antibodies specific for either Tau or Actin.
Figure 10D shows a graph depicting inhibition of the deubiquitinase activity of the indicated deubiquitinases by IU IC (left bars = 0 μΜ, right bars = 17 μΜ).
Figure 11A shows a plot depicting the deubiquitinase activity of proteasomc bound Uspl4, IsoT or Uch37 that had been treated with the indicated concentration of 1U1.
Figure 1 IB shows a graph depicting the inhibition of the deubiquitinase activity of the indicated deubiquitinases by IU1 (left bars = 0 μΜ, right bars = 17 μΜ).
Figure I2A shows immunoblots of purified 26S human proteasomes (~4 nM) that had been incubated witli or without Uspl4 (80 nM) and treated either with vehicle (DMSO), 1U1C or 1U1 at the indicated concentrations and stained with antibodies specific for either Uspl4 or AIpha7. The asterisk (*) denotes a nonspecific signal generated by the anti-Usp 14 antibody.
Figure 12B shows imniunoblots as in Figure 12A, except ---2-fold molar excess of Usp 14 was incubated with the proteasome in the absence or presence of the indicated compound (30 μΜ).
Figure 13 shows graphs depicting the deubiquitination activity of proteasome bound Usp 14 that had been treated with vehicle (control) or IU1 and subjected to the indicated number of rounds of ultrafiltration (spins, left panel; no spin = left bar, lx spin = middle bar, 3x spin = right bar), or spin-column gel filtration (right panel; control = left bar, IU1 - right bar).
Figure 14A shows a plot depicting an IC50 curve of proteasome bound Usp 14 treated with the indicated concentration of 1U1 for 45 minutes.
Figure 14B shows a plot depicting an IC50 curve of proteasome bound Usp 14 treated with the indicated concentration of IU 1 for 30 minutes.
Figure 15 shows an immunoblot that was performed using an antibody specific for Cyciin B and polyubiquitinated Cyclin B (Ub„-ClbB) that had been treated with 4 nM of 26S human proteasome, cither alone, with wild-type Usp 14 (Uspl4-wt), with 1U1, and/or with proteasome inhibitor as indicated. The immunoblot was cither subject to a long exposure (Long exp) or a short exposure (Short exp).
Figure I6A shows an immunoblot that was performed using an antibody specific for Cyclin B and polyubiquitinated Cyclin B (Ub„-ClbB) that had been treated with 4 nM of 26S human proteasome, either alone or in combination with wild-type Usp 14 (60 nM) along with either vehicle or 1U1 (34 μΜ).
Figure 16B shows an immunoblot that was performed using an antibody specific for T7-taggcd Sicln and polyubiquitinated Sicl(Ubn-Sicln) that had been treated with 5 nM of 26S human proteasome, cither alone or in combination with wild-type Usp 14 (75 nM) along with cither vehicle or 1U1 (75 μΜ).
Figure 17 shows plots depicting the ion counts of Liquid Chromatography/Mass Spectrometry (LC/MS) traces of lysates from MEF cells that had not been treated with IU 1 (No 1U1), or been treated with 1U1 for 1 or 24 hours. The bottom panel depicts the ion count for an 1U3 standard solution at l pg/mL.
Figure 18 shows plots depicting the ion counts of LC/MS traces of various concentrations of IU 1 standard.
Figure 19 shows the LTV spectrum of IU 1, depicting absorption maxima at 255 nm and 305 nm (left) and HPLC chromatograms showing the time-dependence of IU1 internalization into cells, followed at 300 nm (right).
Figure 20A shows a graph depicting the 1UI concentration in MEF cells after normalization by cell number as detected by UV absoiption assay.
Figure 20B shows a graph depicting the IU1 concentration in 293 cells after normalization by cell number as detected by UV absorption assay.
Figure 21A shows immunoblots that were performed using lysates of MEF cells that co-expresscd Tau and Uspl4 and that were treated with 0,25, 50, 75 or 100 μΜ IU 1 and stained with antibodies specific for Tau, LacZor Actin.
Figure 21B shows the result of quantitative RT-PCR analysis of Tau RNA levels in MEF cells that co-expressed Tau and Uspl4 and that were treated with 0, 25, 50, 75 or 100 μΜ 1U1 -
Figure 22A shows immunoblots that were performed using lysates of MEF cells that co-expressed TDP43!1:‘- and Uspl4 and that were treated with 75 μΜ lUlfor the indicated number of hours and stained with antibodies specific forTDP43llas, LacZ or Actin,
Figure 22B shows immunoblots that were performed using lysates of MEF cells that co-expressed cither Atx3-Q80 or Atx3-Q22 along with Uspl4 and that were treated with 0,50 or 100 μΜ 1U1 and stained with antibodies specific for Atx3 or Actin.
Figure 22C shows immunoblots that were performed using lysates of MEF cells that co-expressed cither wild-type GFAP (GFAP-wt), K63Q mutant GFAP (GFAP-K63Q) or E210K GFAP (GFAP-E210K) along with Uspl4 and that were treated with 0, 25,50 or 100 μΜ IU1 and stained with antibodies specific for GFAP or Actin.
Figure 22D shows the result of quantitative RT-PCR analysis of TDP-43 RNA levels in MEF cells that co-cxprcsscd TDP-43 and Usp-14 and that was treated with 75 μΜ IU1 for each indicated time.
Figure 23A shows immunoblots stained with anti-DNPH or anti-Actin antibodies that were performed on DNPH-treated lysates of MEF cells that were preincubated with vehicle or 75 μΜ IU 1 and treated with 63 μΜ menadione, as indicated.
Figure 23B shows immunoblots stained with anti-DNPH or anti-Actin antibodies that were performed on DNPH-treated lysates of HEK293 cells that were preincubated with 1U1 (75 μΜ) or protcasomc inhibitors (20 μΜ MG 132, 10 μΜ PS-341) for 4 h, then treated with menadione (300 μΜ) for 60 min.
Figure 24 shows a plot depicting MEF cell viability as assessed by MTT assay upon treatment with the indicated concentration of 1U1 for 48 hours.
Figure 25A shows a plot depicting MEF cell viability as assessed by MTT assay upon treatment with the indicated concentration of 1U1 for 6 hours.
Figure 25B shows a plot depicting MEF cell viability as assessed by MTT assay upon treatment with the indicated concentration of 1U1 for 12 hours.
Figure 25C shows a plot depicting MEF cell viability as assessed by MTT assay upon treatment with the indicated concentration of 1U1 for 24 hours.
Figure 26A shows a plot depicting 293 and HeLa ceil viability as assessed by MTT assay upon treatment with the indicated concentration of iUI for 6 hours.
Figure 26B shows a plot depicting 293 and HcLa cell viability as assessed by MTT assay upon treatment with the indicated concentration of IUI for 12 hours.
Figure 26C shows a plot depicting 293 and HeLa cell viability as assessed by MTT assay upon treatment with the indicated concentration of IU1 for 24 hours.
Figure 27A shows fluorescent microscopy images of TUNEL stained MEF cells that had been treated with 100 μΜ 1U1 or control for 6 hr.
Figure 27B shows a graph depicting the quantification of the TUNEL staining analysis depicted in Figure 31 A.
Figure 28 shows a plot depicting the percent confiucncy of MEF cells that had been treated with the indicated concentration of vehicle or IUI for the indicated period of time.
Figure 29 depicts one approach to compounds of the invention.
Figure 30 depicts a graph showing MTT assay for cell viability; specifically, 1U1 effects on cell survival upon oxidative stress. Experiment performed in HEK.293 cells with Menadione (dosc-dcpendcnt, 4hr) and 1U1 (50 uM, 6 hr). MEF cells show this effect as well. The effect on the ICso for menadione is almost 4-fold.
Figure 31 depicts a table of selected compounds of the invention, including some percent inhibition and ICjo values. Percent inhibition was measured at 8 μΜ from 1U1-1 to 1U-46, at 4 μΜ from 1U1 -47 to 1UI -96, and at 17 μΜ for CM to C9.
Figure 31A shows immunoblots that were performed using lysates of Uspl4-/~ MEF cells that co-expressed tau and LacZ and that were treated with 0, 25, 50, 75 or 100 μΜ IU 1 and stained with antibodies specific for tau, LacZ or Actin.
Figure 31B show's immunoblots that were performed using lysates of Usp 14-/-MEF cells that co-expressed TDP-43llas and LacZ and that were treated with 75 μΜ 1U1 for the indicated number of hours and stained with antibodies specific for TDP-43nag, LaeZ or Actin.
Figure 32 shows immunoblots that were performed using lysates of wild-type MEF cells that co-expressed tau and Ub-independent proteasome substrate cODC-EGFP and that were incubated with 50 μΜ IUI for 6 h and stained with antibodies specific for tau, cODC-EGFP or Actin. Proteasome inhibitors (30 μΜ MG 132, 10 μΜ PS-341) were treated 4 hr before lysis.
Figure 33 shows immunoblots that were performed using lysates of wild-type MEF cells that co-expressed HA-tagged Ub and/or Flag-tagged TDP-43 and that were incubated with 50 μΜ IUI for 6 hr and stained with antibodies specific for Flag-TDP-43, HA-Ub, or Actin. Proteasome inhibitors (20 μΜ MG 132, 10 μΜ PS-341) were added 4 hr before lysis. Arrows indicate likely ubiquitinated TDP-43 species. HC, heavy chain of antibody.
Figure 34A shows immunoblots that were performed using lysates of wild-type or UspI4 ! MEF cells that were treated with 1U1 (0,25, 50, 75, or 100 μΜ) for 6 hr and stained with antibodies specific for Ub, Actin, CP subunit a7, or RP subunit mRPT5.
Figure 34B shows quantification of ubiquitin levels in Figure 34A. Polyubiquitin and monoubiquitin levels from wild-type and Usp]4"‘‘ MEF cells were quantified after treatment of various concentration of IUI. Ub signals were normalized to that of endogenous actin. Quantification was achieved by densitometry of a film image (left bar = poly UB (+/+ MEF); second bar to the left = poly Ub (UspI4'f' MEF); third bar = mono Ub (+/+ MEF); right bar = mono Ub (Uspl4'A MEF)).
Figure 35A shows the specificity oflUl for USP14 which is observed independently of Ub-AMC concentration. Assays of Ub-AMC hydrolysis were done as in Figure l IB, except lower concentrations of Ub-AMC were used (left bar = 0 μΜ; right bar — 17 μΜ).
Figure 35B shows the summary of Km values for Ub-AMC of deubiquitinating enzymes in this study. Km values of DUBs used in the selectivity assays were obtained from the literature. Unknown Km values were determined in this study, as indicated. These values are significant because the DUB assays should be most sensitive to inhibition when substrate is at a low concentration as compared to the Km of the enzyme in question. CD, catalytic domain.
Figure36 shows that IUI inhibits proteasome-associated USP14 activity without a detectable lag period. 2.5 nM of human protcasome was mixed with 30 nM of recombinant USP14 protein, The reaction was then initiated by adding 1 μΜ Ub-AMC. After 30 min, IU1 (100 μΜ) or vehicle (DMSO) was added to the sample.
Figure 37 confirms Figure 13, except prolonged incubation (5 and 8 hr) was tested. The percent USP14 activity was normalized to 26S peptidase activity (i.c. LLVY-AMC hydrolysis). 1U1 was added to 100 μΜ (left bar = DMSO; right bar = IU1).
Figure 38 shows in vitro chain trimming assays that were performed with human proteasome purified in the presence of ADP (ADP prep) and assayed in the presence of ADP. Immunoblot was performed using an antibody specific for human cyclin B (CCNB). IU1 is effective at inhibition of chain trimming at approximately 5 μΜ, as expected from Ub-AMC hydrolysis data.
Figure 39 shows that 1U1 does not affect cyclin B degradation in the presence of USP14-CA. Assays were done as in Figure 5.
Figure 40A shows 'H-NMR spectroscopic data of IU1.
Figure 40B shows LC/MS analysis of 1U1. TIC, total ion count, SPC, shared peak count extracted from the peak with the indicated retention time.
Figure 41A shows 'H-NMR spectroscopic data of IUIC.
Figure 41B shows LC/MS analysis of IUIC. TIC, total ion count. SPC, shared peak count extracted from the peak with the indicated retention time.
Figure 42A shows a graph depicting rapid release of the internalized 1U1 from wild-type MEF cells. After wild-type MEFs were incubated with 50 μΜ of IU1 for one hour, the culture media were replaced with fresh media without IU 1. Internalized IUI was monitored at the indicated times and its concentration was normalization by cell number as detected by UV absorption assay.
Figure 42B shows the comparable concentration of IU1 from 1 hr to 48 hr in the scrum-containing media ofHEK293 cells.
Figure 42C shows the comparable concentration of IUI from 1 hr to 48 hr in the serum-containing media of Usp!4~f~ MEF cells.
Figure 43A shows the quantitative analysis of tau levels after IUI treatment in wild-type MEFs. Quantification was performed with infrared dye-conjugated secondary antibodies using Odyssey imaging system. Tau signal intensities were normalized to that of endogenous actin and relative amounts arc shown.
Figure 43B shows the quantitative analysis of tau levels after 1U1 treatment in Uspi4'~ MEFs. Quantification was performed as in Figure 43A.
Figure 44A shows that the immunofluorescence signal of transiently expressed mCherry-NBRl (top row) was significantly increased after treatment with 200 liM of bafiiomycin Ai (BafAQ, an autolysosome formation inhibitor, for 6 hr in wild-type MEFs.
Figure 44B shows that the stimulation of tau degradation by IU1 is not mediated by autophagy. Wild-type MEF cells were transfected with a plasmid expressing tau and then treated with 200 uM of BafA) and/or 75 liM of IU1 for 6 hrs, and analyzed by SDS-PAGE/immunoblot using the Odyssey infrared imaging system.
Figure 44C shows the quantification of normalized tau protein level from three independent experiments (mean ± SD) as performed in Figure 44B using Odyssey software.
Figure 45A shows that 1U1 treatment docs not affect the integrity of protcasomc. Total cell extracts (50 ug/lanc) before and after a 6-hr IU1 treatment (100 μΜ) were resolved by native PAGE, and the protcasomc was visualized using cither an in-gel activity stain with a fluorogenic peptide substrate (LLVY-AMC), or immunobiotting with antibodies specific to subunit a(\ RP2-CP and RP-CP indicate distinct forms of the 26S protcasomc.
Figure 45B shows that IU1 treatment docs not induce the transcription of PsmbS gene, a proteasome subunit, A luciferase reporter gene containing the murine PsmbS promoter (-1 kb to 0 kb) was transiently expressed in wild-type and Uspl4m>’ MEFs and promoter activity was assessed following incubation of 25 or 50 μΜ of IU1 for 8 hr. For normalization of luciferase activity, a control experiment using the promotcr-lcss pGL3 plasmid was performed. Values arc mean ± SD from three independent experiments. RLU, relative light units.
Figure 45C shows that IU1 treatment docs not induce the transcription of UbB, a ubiquitin gene. Quantitative RT-PCR was performed using total mRNA from +/+ (left panels) and UspI4J~ MEFs (right) after incubation with a graded doses of 1U1 for 6 hr.
Figure 45D shows that IU1 treatment does not induce the transcription of ad, a proteasome subunit. Quantitative RT-PCR was performed using total mRNA from +/+ (left panels) and Uspl4'u MEFs (right) after incubation with a graded doses of IU 1 for 6 hr.
Figure 45E shows that IU1 treatment does not induce the transcription of a7, a proteasome subunit. Quantitative RT-PCR was performed using total mRNA from +/+ {left panels) and Usp!4' ~ MEFs (right) after incubation with a graded doses of IU1 for 6 hr.
Figure 46A depicts a graph showing MTT assay for cell viability; specifically, IUIC effects on cell survival upon oxidative stress. Experiment performed in HEK293 ceils with Menadione (dose-dependent, 4hr) and IUIC (50 uM, 6 hr).
Figure 46B shows a graph depicting the internalized IUIC concentration in wild-type MEF cells. After the indicated time-course treatment of 50 uM of IUIC, IUIC levels were measured by LC/MS. The concentration shown was normalized by cell number as detected by UV absorption assay.
Figure 46C shows a graph depicting the internalized IUIC concentration in 293 cells, After the indicated time-course treatment of 50 uM of IU 1C, HJ1C levels were measured by LC/MS. The concentration shown was normalized by cell number as detected by UV absorption assay.
Figure 46D shows a graph depicting rapid release of the internalized IUIC from wild-type MEF cells. The experiment was performed as in Figure 42A.
Figure 46E shows a graph depicting rapid release of the internalized IUIC from 293 cells. The experiment was performed as in Figure 42A.
Figure 47 shows an immunoblot that were performed using lysates of wild-type MEF cells that transiently expressed Tau and that were treated with 0, 20,40,60 or 80 μΜ of IU 1-47, a more potent IU1 derivative, and stained with an antibody specific for Tau.
DETAILED DESCRIPTION
Proteinopathies are a class of diseases and disorders that result from the aggregation of abnormal or misfolded proteins. Often, and perhaps typically, such proteins are eliminated from cells through protcasome-mcdiatcd degradation. However, in the case of proteinopathies, the protcasome docs not act efficiently enough to eliminate all of the harmfi.il proteins and prevent the formation of the pathogenic aggregates.
As is demonstrated herein, under normal growth conditions, the protcasome is subject to tonic inhibition brought about by the trimming of substrate-bound ubiquitin chains by Uspl4. Ubiquitin chain trimming inhibits the protcasome because it removes from proteasome substrates the signal (a ubiquitin chain) that allows recognition by the proteasome; the proteasome-bound substrate can therefore escape without being degraded. Consequently, an inhibitor of chain trimming by Uspl4 promotes protein degradation by the protcasome. Thus, as a result of this inhibitory mechanism, the mammalian proteasome pathway does not ordinarily operate at fill! efficiency because the pathway is partially inhibited by Usp!4.
The methods and compositions of the present invention enhance proteasome activity by inhibiting the deubiquitinase activity of Uspl4. As demonstrated herein, this enhanced proteasome activity increases the ability of a cell to eliminate abnormal or misfolded proteins, including those associated with human disease. The methods and compositions of the present invention are therefore useful for the enhancement of proteasome function and the treatment of protcinopathics.
Definitions
In order for the present invention to be more readily understood, certain terms and phrases are defined below and throughout the specification.
The articles "a" and "an" are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.
The phrase “and/or,” as used herein in the specification and in the claims, should be understood to mean “cither or both” of the elements so conjoined, i.c., elements that arc conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with “and/or” should be construed in the same fashion, i.c., “one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
As used herein in the specification and in the claims, “or” should be understood to have the same meaning as “and/or” as defined above. For example, when separating items in a list, "or” or “and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as “only one of’ or “exactly one of,” or, when used in the claims, “consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e., “one or the other but not both”) when preceded by terms of exclusivity, such as '‘either,” “one of,” “only one of,” or “exactly one of.” “Consisting essentially of,” when used in the claims, shall have its ordinary meaning as used in the field of patent law.
As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements.
This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a nonlimiting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.
In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding ” “composed of,” and the like arc to be understood to be open-ended, i.c., to mean including but not limited to. Only the transitional phrases “consisting of’ and “consisting essentially of’ shall be closed or scmi-closcd transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.
The definition of each expression, e.g., alkyl, m, n, and the like, when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.
It will be understood that "substitution" or "substituted with" includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, c.g., a compound which docs not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, or other reaction.
The term "substituted" is also contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyciic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described herein below. The permissible substituents may be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the hctcroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This invention is not intended to be limited in any manner by the permissible substituents of organic compounds.
The term “lower” when appended to any of the groups listed below indicates that the group contains less than seven carbons (i.e. six carbons or less). For example “lower alkyl” refers to an alkyl group containing 1-6 carbons, and “lower alkenyl” refers to an alkycnyl group containing 2-6 carbons.
The term “saturated,” as used herein, pertains to compounds and/or groups which do not have any carbon-carbon double bonds or carbon-carbon triple bonds.
The term “unsaturated,” as used herein, pertains to compounds and/or groups which have at least one carbon-carbon double bond or carbon-carbon triple bond.
The term “aliphatic,” as used herein, pertains to compounds and/or groups which are linear or branched, but not cyclic (also known as “acyclic” or “open-chain” groups).
The term “cyclic,” as used herein, pertains to compounds and/or groups which have one ring, or two or more rings (e.g,, spiro, fused, bridged).
The term "aromatic" refers to a planar or polycyclic structure characterized by a cyclically conjugated molecular moiety containing 4n+2 electrons, wherein n is the absolute value of an integer. Aromatic molecules containing fused, or joined, rings also are referred to as bicylic aromatic rings. For example, bicyclic aromatic rings containing heteroatoms in a hydrocarbon ring structure are referred to as bicyclic heteroaryl rings.
The term “hydrocarbon” as used herein refers to an organic compound consisting entirely of hydrogen and carbon.
For purposes of this invention, the chemical elements arc identified in accordance with the Periodic Table of the Elements, CAS version. Handbook of Chemistry and Physics, 67th Ed., 1986-87, inside cover.
The term "heteroatom" as used herein is art-recognized and refers to an atom of any element other than carbon or hydrogen. Illustrative heteroatoms include boron, nitrogen, oxygen, phosphorus, sulfur and selenium.
The term "alkyl" means an aliphatic or cyclic hydrocarbon radical containing from 1 to 12 carbon atoms. Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tcrt-butyl, n-pcntyl, isopentyl, neopentyi, n-hexyl, 2-methylcyclopentyl, and l-cyclohcxylcthyl.
The term "substituted alkyl" means an aliphatic or cyclic hydrocarbon radical containing from 1 to 12 carbon atoms, substituted with 1, 2, 3, 4, or 5 substiuents independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, hatoaikyi, fluoroaikyl, hydroxy, alkoxy, aikyenyloxy, alkynyloxy, carbocyciyloxy, heterocyclyloxy, haloalkoxy, fluoroalkyloxy, sulfhydryl, alkylthio, haloalkylthio, fluoroalkylthio, alkyenylthio, aikynylthio, sulfonic acid, alkylsulfonyl, haioalkylsulfonyl, fluroralkylsulfonyl, alkcnyisulfonyl, alkynylsulfonyl, aikoxysulfonyi, haloalkoxysulfonyl, fluroralkoxysulfonyl, aikcnyloxysulfonyl, alkynyioxysulfony, aminosulfonyl, sulfinic acid, alkylsulfinyl, haloalkylsulflnyl, fluroralkylsulfinyl, alkcnylsulfinyl, alkynylsulfinyi, alkoxysulfinyl, haloalkoxysulfinyl, fluroralkoxysulfiiiyl, alkcnyloxysulfinyl, alkynyloxysulfiny, aminosulfinyl, formyl, alkylcarbonyl, haloalkylcarbonyl, fluoroalkylcarbonyl, alkenylcarbonyl, alkynylcarbonyl, carboxy, alkoxycarbonyl, haloalkoxycarbonyl, fluoroalkoxycarbonyl, alkenyloxycarbonyl, alkynyloxycarbonyl, alkylcarbonyloxy, haloalkylcarbonyloxy, fluoroalkylcarbonyloxy, alkenylcarbonyloxy, alkynylcarbonyloxy, alkylsulfonyloxy, haloalkylsulfonyloxy, fiuroralkyisulfonyloxy, alkenylsuifonyloxy, alkynylsulfonyloxy, haloalkoxysulfonyioxy, fluroralkoxysulfonyloxy, alkcnyloxysulfonyloxy, aikynyloxysulfbnyloxy, alkylsulfinyloxy, haloalkylsuifinyloxy, fluroralkylsulfinyloxy, alkcnylsulfinyloxy, alkynylsulfinyloxy, alkoxysulfinyloxy, haloalkoxysulfinyloxy, fkiroralkoxysulfinyloxy, alkenyloxysulfinyloxy, aikynyloxysulfinyloxy, aminosulfinyloxy, amino, amido, aminosulfonyl, aminosulfinyl, cyano, nitro, azido, phosphinyl, phosphoryl, silyl and silyloxy.
The term "alkylene" is art-recognized, and as used herein pertains to a bidentate moiety obtained by removing two hydrogen atoms of an alkyl group, as defined above.
The term "alkenyl" as used herein means a straight or branched chain hydrocarbon containing from 2 to 10 carbons and containing at least one carbon-carbon double bond formed by the removal of two hydrogens. Representative examples of alkenyl include, but are not limited to, ethenyl, 2-propenyl, 2-methyl-2-propenyl, 3-butenyl, 4-pentenyl, 5-hexcnyl, 2-heptenyl, 2-methyl-1-heptenyl, and 3-decenyl.
The term "alkynyl" as used herein means a straight or branched chain hydrocarbon group containing from 2 to 10 carbon atoms and containing at least one carbon-carbon triple bond. Representative examples of alkynyl include, but arc not limited, to acctylcnyl, I-propynyl, 2-propynyl, 3-butynyI, 2-pcntynyi, and 1-butynyl.
The term "carbocyclyl" as used herein means monocyclic or multicyclic (c.g., bicychc, tricyclic, etc.) hydrocarbons containing from 3 to 12 carbon atoms that is completely saturated or has one or more unsaturated bonds, and for the avoidance of doubt, the degree of unsaturation docs not result in an aromatic ring system (e.g. phenyl).
Examples of carbocyclyl groups include 1-eye lop ropy!, 1-cyclobutyl, 2-cyclopentyl, 1-cyclopentcnyl, 3-cyclohexyl, 1-cyclohexenyl and 2-cyclopentenylmethyl.
The term "hcterocyclyl", as used herein include non-aromatic, ring systems, including, but not limited to, monocyclic, bicyclic (e.g. fused and spirocyclic) and tricyclic rings, which can be completely saturated or which can contain one or more units of unsaturation, for the avoidance of doubt, the degree of unsaturation docs not result in an aromatic ring system, and have 3 to 12 atoms including at least one heteroatom, such as nitrogen, oxygen, or sulfur. For purposes of exemplification, which should not be construed as limiting the scope of this invention, the following are examples of heterocyclic rings: azepines, azetidinyi, morpholinyl, oxopiperidinyl, oxopyrrolidinyl, piperazinyl, pipcridinyl, pyrrolidinyl, quinicludinyl, thiomorpholinyl, tetrahydropyranyi and tctrahydrofuranyl. The hcterocyclyl groups of the invention arc substituted with 0, l, 2,3,4 or 5 substituents independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, alkycnyloxy, alkynyloxy, carbocyclyloxy, heterocyclyloxy, haloalkoxy, fluoroalkyloxy, sulfhydryl, afkylthio, haloalkylthio, fluoroaikyithio, alkyenylthio, alkynylthio, sulfonic acid, aikylsulfonyl, haioalkylsulfonyl, fluroraikylsulfonyl, alkenylsulfonyl, aikynylsulfonyl, alkoxysulfonyl, haloalkoxysulfonyl, fluroralkoxysulfonyi, alkenyloxysuifonyl, alkynyloxysulfony, aminosulfonyl, sulfinic acid, alkylsulfinyl, haloalkylsulfinyl, fluroralkylsulfinyl, alkenylsulfinyl, alkynylsulfinyl, alkoxysulfinyl, haloalkoxysulfinyl, fluroralkoxysulfinyl, alkcnyloxysulfmy], alkynyloxysulfiny, amtnosulfinyl, formyl, alkylcarbonyl, haloalkylcarbonyl, fluoroalkylcarbonyl, alkcnyicarbonyl, alkynylcarbony!, carboxy, alkoxycarbonyi, haloalkoxycarbonyl, fiuoroalkoxycarbonyl, alkenyloxycarbonyi, alkynyloxycarbonyl, alkylcarbonyloxy, haloalkylcarbonyloxy, fluoroalkylcarbonyloxy, alkenylcarbonyloxy, alkynylcarbonyloxy, alkylsulfonyloxy, haloalkylsulfonyloxy, fluroralkylsulfonyloxy, alkenylsulfonyloxy, alkynylsulfonyioxy, haioalkoxysuifonyloxy, fluroralkoxysulfonyloxy, alkenyloxysutfonyloxy, aikynyloxysulfonyloxy, alkylsulfinyloxy, haloalkylsulfinyloxy, fluroralkylsulfinyloxy, alkcnylsulfinyloxy, alkynylsulfinyloxy, alkoxysulfinyloxy, haloalkoxysulfinyloxy, Auroral koxysuifinyloxy, alkcnyloxysuffinyloxy, alkynyfoxysulflnyloxy, aminosulfinyloxy, amino, amido, aminosulfonyl, aminosulfinyl, cyano, nitro, azido, phospbinyl, phosphoryl, stlyl, silyloxy, and any of said substiucnts bound to the heterocyclyi group through an alkylene moiety (e.g. methylene).
The term “N-heterocyclyl” as used herein is a subset of heterocyclyi, as defined herein, which have at least one nitrogen atom through which the N-heterocyclyl moiety is bound to the parent moiety. Representative examples include pyrrolidin-l-yl, piperidin-1-yl, pipcrazin-l-yi, hexahydropyrimidin-l-yl, morpholin-l-yl, l,3-oxazinan-3-yl and 6-azaspiro[2.5]oct-6-yi. As with the heterocyclyi groups, the N-hctcrocyclyl groups of the invention are substituted with 0, 1, 2,3,4 or 5 substituents independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, alkyenyloxy, alkynyloxy, carbocyclyloxy, heterocyclyloxy, haloalkoxy, fluoroalkyloxy, sulfliydryl, alkylthio, haioalkylthio, fluoroalkylthio, alkyenylthio, alkynylthio, sulfonic acid, aikylsuifonyl, haloalkylsulfonyl, fluroralkylsulfonyl, alkenylsulfonyl, alkynylsulfonyl, aikoxysulfonyl, haloalkoxysulfonyl, fluroralkoxysulfonyl, alkcnyloxysulfonyl, alkynyloxysuifony, aminosulfonyl, suifinic acid, alkylsuifinyl, haloaikylsulfitly 1, fluroralkylsulfinyl, alkcnylsulfinyl, alkynylsulfinyl, alkoxysulfinyl, haloalkoxysulfinyl, fluroralkoxysulfinyl, alkcnyloxysulfinyl, alkynyloxysulfiny, aminosulfinyl, formyl, alkylcarbonyl, haloalkylcarbonyl, fluoroalkylcarbonyl, alkcnyicarbonyl, alkynylcarbony!, carboxy, alkoxycarbonyi, haloalkoxycarbonyl, fiuoroalkoxycarbonyl, alkenyloxycarbonyi, alkynyloxycarbonyl, alkylcarbonyloxy, haloalkylcarbonyloxy, fluoroalkylcarbonyloxy, alkenylcarbonyloxy, alkynylcarbonyloxy, alkylsulfonyloxy, haloalkylsulfonyloxy, fluroralkylsulfonyloxy, alkenylsulfonyloxy, alkynylsulfonyioxy, haioalkoxysuifonyloxy, fluroralkoxysulfonyloxy, alkcnyloxysulfonyloxy, aikynyloxysulfonyloxy, alkylsulfinyloxy, haloalkylsulfinyloxy, fluroralkylsulfinyloxy, alkcnylsulfinyloxy, alkynylsulfinyloxy. alkoxysuifinyloxy, haioaikoxysulfinyloxy, fluroralkoxysulfinyloxy, aikcnyloxysuifinyloxy, alkynyloxysulfinyloxy, aminosulfinyioxy, amino, amido, aminosulfonyi, aminosulfinyl, cyano, nitro, azido, phosphinyl, phosphoryl, siiyl, silyloxy, and any of said substituents bound to the N-heterocyc!yi group through an alkylene moiety (e,g. methylene).
The term "aryl," as used herein means a phenyl group, naphthyl or anihracenyl group. The aryl groups of the present invention can be optionally substituted with 1,2,3,4 or 5 substituents independently selected Ιτοιη the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, aikycnyloxy, alkynyloxy, carbocyciyloxy, hctcrocyclyloxy, haloalkoxy, fluoroalkyloxy, sulfhydryl, alkylthio, haloalkylthio, fluoroalkylthio, alkycnylthio, alkynylthio, sulfonic acid, alkylsulfonyl, haloalkylsulfonyl, fluroralkylsuifonyl, alkenylsuifonyl, alkynylsulfonyl, alkoxysulfonyl, haloalkoxysulfonyl, fluroralkoxysulfonyl, alkenyloxysulfonyl, alkynyloxysulfony, aminosulfonyi, sulfmic acid, alkylsulfinyl, haloalkylsulfinyi, fluroralkylsulfinyl, aikenylsulfinyi, alkynylsulfmyl, alkoxysulfinyi, haloalkoxysulfinyl, fluroralkoxysulfinyl, alkenyloxysulfinyl, alkynyloxysulfiny, aminosuffinyl, formyl, alkylcarbonyl, haloalkylcarbonyl, fluoroalkylcarbonyl, alkcnylcarbonyl, alkynylcarbonyl, carboxy, alkoxycarbonyl, haloalkoxycarbonyl, fluoroalkoxycarbonyl, alkcnyloxycarbonyl, alkynyioxycarbonyl, alkylcarbonyloxy, haloalkylcarbonyloxy, fluoroalkylcarbonyloxy, alkcnylcarbonyloxy, alkynylcarbonyioxy, alkylsulfonyloxy, haloalkylsulfonyloxy, fluroralkylsulfonyloxy, alkenylsulfonyloxy, alkynylsulfonyloxy, haloalkoxysulfonyloxy, fluroralkoxysulfonyloxy, alkenyloxysulfonyloxy, alkynyloxysulfonytoxy, alkylsulfinyloxy, haloalkylsulfinyloxy, fluroralkylsulfinyloxy, alkenylsulfinyloxy, aikynyisulfinyloxy, alkoxysuifinyloxy, haioaikoxysulfinyloxy, fluroralkoxysulfinyloxy, alkenyloxysulfinyloxy, alkynyloxysulfinyloxy, aminosulfinyioxy, amino, amido, aminosulfonyi, aminosulfinyl, cyano, nitro, azido, phosphinyl, phosphoryl, siiyl, silyioxy, and any of said substiucnts bound to the heterocyclyl group through an alkylene moiety (e.g. methylene).
The term "arylcnc," is art-recognizcd, and as used herein pertains to a bidentate moiety obtained by removing two hydrogen atoms of an aryl ring, as defined above.
The term "arylaikyl" or "aralkyl" as used herein means an aryl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Representative examples of aralkyl include, but are not limited to, benzyl, 2-phenylethyl, 3-phenylpropyl, and 2-naphth-2~yIethyl.
The term "biaryl," as used herein means an aryl-substituted aryl, an aryl-substituted hctcroaryl, a hctcroaryl-substitutcd aryl or a hctcroaryl-substitutcd hctcroaryl, wherein aryl and hctcroaryl arc as defined herein. Representative examples include 4-(phenyi)phenyl and 4-(4-fluorophenyl)pyridinyl.
The term "heteroaryl" as used herein include aromatic ring systems, including, but not limited to, monocyclic, bicyclic and tricyclic rings, and have 3 to 12 atoms including at least one heteroatom, such as nitrogen, oxygen, or sulfur. For purposes of exemplification, which should not be construed as limiting the scope of this invention: azaindolyl, bcnzo(b)thienyl, bcnzimidazolyl, bcnzofuranyl, bcnzoxazolyl, bcnzothiazolyl, bcnzothiadiazolyl, bcnzotriazolyl, bcnzoxadiazolyl, furanyl, imidazolyl, imidazopyridinyl, indolyl, indolinyl, indazolyl, isoindolinyl, isoxazolyl, isothiazolyl, isoquinolinyl, oxadtazolyl, oxazolyl, purinyl, pyranyl, pyrazinyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrrolyl, pynOlo[2,3-djpyrimidinyl, pyrazolo[3,4-d]pyrimidinyI, quinolinyl, quinazolinyl, triazolyl, thiazolyl, thiophenyl, tctrahydroindolyl, tetrazolyl, thiadiazolyl, thienyl, ihiomorpholinyl, triazolyl or tropanyl. The hctcroaryl groups of the invention are substituted with 0, 1,2,3,4 or 5 substituents independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, atkyenyloxy, alkynyloxy, carbocyclyloxy, hctcrocyclyloxy, haloalkoxy, fluoroalkyloxy, sulfhydryl, alkylthio, haloalkylthio, fluoroalkyllhio, alkycnylthio, alkynylthio, sulfonic acid, alkylsulfonyl, haloalkylsulfonyl, fluroralkylsulfonyl, alkenylsuifonyl, alkynylsuffonyl, alkoxysulfonyl, haloalkoxysulfonyl, fluroralkoxysulfonyt, alkenyloxysulfonyl, alkynyloxysulfony, aminosulfonyl, sulfinic acid, alkylsulfmyl, haloalkylsulfinyl, fluroralkylsulfinyl, alkenylsulfinyl, alkynylsulfinyl, alkoxysulfinyl, haloalkoxysulfinyl, fluroralkoxysulfinyl, alkenyloxysulfinyl, alkynyloxysulfiny, aminosulfinyl, formyl, aikylcarbonyi, haloalkylcarbonyl, fluoroalkylcarbonyl, aikcnyicarbonyi, alkynylcarbonyl, carboxy, alkoxycarbonyl, haloalkoxycarbonyl, fluoroalkoxycarbonyl, alkcnyloxycarbonyl, alkynyloxycarbonyl, alkylcarbonyloxy, haloalkylcarbonyloxy, fluoroalkylcarbonyloxy, alkcnylcarbonyloxy, alkynylcarbonyloxy, alkylsulfonyloxy, haloalkylsulfonyloxy, fluroralkylsulfonyloxy, alkenylsulfonyloxy, alkynylsulfonyloxy, haloalkoxysulfonyloxy, fluroralkoxysulfonyloxy, alkenyioxysulfonyloxy, alkynyloxysulfonyloxy, alkylsulfinyloxy, haloalkylsulfmyloxy, fluroralkylsulfinyloxy, alkenylsulfinyloxy, alkynylsulfinyloxy, alkoxysulfinyloxy, haloalkoxysulfinyloxy, fluroralkoxysulfinyloxy, alkenyloxysulfinyloxy, alkynyloxysulfinyloxy, aminosulfinyloxy, amino, amido, aminosulfonyl, aminosulfinyl. cyano, nitre, azido, phosphinyl, pliosphoryl, silyl, silyloxy, and any of said subsitucnts bound io the hetcroaryl group through an alkyicne moiety (c.g. mctliylenc).
The term "heteroarylene," is art-recognized, and as used herein pertains to a bidentate moiety obtained by removing two hydrogen atoms of a heteroaryl ring, as defined above.
The term "heteroarylalkyl" or "heteroaralkyl" as used herein means a heteroaryl, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Representative examples of heteroarylalkyl include, but arc not limited to, pyridin- 3-y I methyl and 2-(thicn-2-yl)ethyl,
The term "halo" or "halogen" means -Cl, -Br, -i or -F.
The term "haloalkyl" means an alkyl group, as defined herein, wherein at least one hydrogen is replaced with a halogen, as defined herein. Representative examples of haloalkyl include, but are not limited to, chloromethyl, 2-fluoroethyl, trifluoromethyl, pentafluoroethyl, and 2-chloro-3-f]uoropentyi.
The term “fluoroaikyl" means an alkyl group, as defined herein, wherein all the hydrogens arc replaced with fluorines.
The term "hydroxy” as used herein means an -OH group.
The term "alkoxy" as used herein means an alkyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, and hexyloxy. The terms “alkyenyloxy”, “alkynyloxy”, “carbocyclyloxy”, and “heterocyclyloxy” are likewise defined.
The term "haioalkoxy" as used herein means an alkoxy group, as defined herein, wherein at least one hydrogen is replaced with a halogen, as defined herein. Representative examples of haioalkoxy include, but arc not limited to, chloromcthoxy, 2-fluorocthoxy, trifluoromcthoxy, and pentafluoroethoxy. The term “fluoroalkyloxy” is likewise defined.
The term "aryloxy” as used herein means an aryl group, as defined herein, appended to the parent molecular moiety through an oxygen. The term "heteroaryloxy” as used herein means a heteroaryl group, as defined herein, appended to the parent molecular moiety through an oxygen. The terms “heteroaryloxy” is likewise defined.
The term "arylalkoxy" or "arylalkyloxy" as used herein means an arylalkyl group, as defined herein, appended to the parent molecular moiety through an oxygen. The term "hetcroarylalkoxy" is likewise defined. Representative examples of aryloxy and hctcroarylaikoxy include, but arc not limited to, 2-chlorophcnylmcthoxy, 3-tnfluoromethyl-phenylcthoxy, and 2,3-dimcthyIpyridinylmcthoxy.
The term "sulfhydryl" or “thio” as used herein means a -SH group.
The term "alkylthio" as used herein means an alkyl group, as defined herein, appended to the parent molecular moiety through a sulfur. Representative examples of alkylthio include, but are not limited, methylthio, ethylthio, tert-butylthio, and hexylthio. The terms “haloalkyithio”, “fluoroalkylthio”, “alkycnylthio”, “alkynylthio”, “carbocyclylthio”, and “hcterocyclylthio” arc likewise defined.
The term "arylthio” as used herein means an aryl group, as defined herein, appended to the parent molecular moiety through an sulfur. The term "hctcroarylthio” is likewise defined.
The term "arylalkylthio" or "aralkylthio" as used herein means an arylalkyl group, as defined herein, appended to the parent molecular moiety through an sulfur. The term "heteroarylalkylthio" is likewise defined.
The term “sulfonyl” as used herein refers to -S(=0)2- group.
The term “sulfonic acid” as used herein refers to -S(=0)20H.
The term "alkylsulfonyl" as used herein means an alkyl group, as defined herein, appended to the parent molecular moiety through a sulfonyl group, as defined herein. Representative examples of alkylsulfonyl include, but are not limited to, mcthylsulfonyl and ethylsulfonyl. The terms “haloalkylsulfonyl”, “AuroraIkyIsulfonyl”, “alkenyisulfonyl”, “alkynyisulfonyl”, “carbocyclylsulfonyl”, "heterocyctylsulfonyl”, “arylsulfonyl”, “aralkylsulfonyl”, “hcteroarylsulfonyl” and “heteroaralkylsulfonyl” are likewise defined.
The term "alkoxysulfonyl" as used herein means an alkoxy group, as defined herein, appended to the parent molecular moiety through a sulfonyl group, as defined herein. Representative examples of alkoxysulfonyl include, but arc not limited to, mcthoxysulfonyl, cthoxysulfonyl and propoxysulfonyl. The terms “haloalkoxysulfonyl”, “fiuroralkoxysulfonyl”, “alkcnyloxysulfonyl”, “alkynyloxysulfonyl”, “carbocyclyloxy.sulfonyl”, “heterocyclyloxysulfonyl”, “aryloxysulfonyl”, “aralkyloxysutfonyl”, “hetetOaryloxysulfonyl” and “heteroaralkyloxysulfonyl” are likewise defined.
The terms triflyl, tosyl, mesyl, and nonaflyl are art-recognized and refer to trifluoromcthancsulfonyl, p-tolucncsulfonyl, mcthancsulfonyl, and nonafluorobutanesulfonyl groups, respectively. Tlte terms inflate, tosylatc, mesylate, and nonaflatc are art-rccognizcd and refer to trifluoromcthanesulfonatc ester,/j-tolucncsulfonatc ester, mcthancsulfonate ester, and nonafinorobutanesuifonate ester functional groups and molecules that contain said groups, respectively.
The term “aminosulfonyl” as used herein means an amino group, as defined herein, appended to the parent molecular moiety through a sulfonyl group.
The term “sulfinyi” as used herein refers to -S{=0)- group. Sulfinyi groups are as defined above for sulfonyl groups. The term “sulfinic acid” as used herein refers to -S<-0)0H.
The term “oxy” refers to a -0- group.
The term "carbonyl" as used herein means a -C(-O)- group.
The term “thiocarbonyl” as used herein means a -C(=S)- group.
The term "formyl" as used herein means a -CY^OjH group.
The term "alkylcarbonyl" as used herein means an alkyl group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein. Representative examples of alkylcarbonyl include, but are not limited to, acetyl, 1-oxopropyl, 2,2-dimethyl-1-oxopropyl, l-oxobutyl, and 1-oxopcntyl. The terms ‘lialoaikylcarbonyr, “fluoroalkylcarbonyl”, “alkcnylcarbonyl”, “alkynylcarbonyl”, “carbocyciylcarbonyi”, “hctcrocyclylcarbonyl”, ltarylcarbonyP’, “aralkylcarbonyl”, “hctcroarylcarbonyl”, and ‘licleiOaralkylcarbonyl” arc likewise defined.
The term "carboxy" as used herein means a -CO2H group.
The term "alkoxycarbonyl" as used herein means an alkoxy group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein. Representative examples of alkoxycarbonyl include, but are not limited to, mcthoxycarbonyl, cthoxycarbonyl, and tert-butoxycarbonyl. The terms “haloalkoxycarbonyl”, “fhioroaikoxycarbonyP’, “alkcnyloxycarbonyl”, “alkynyloxycarbonyl”, “carbocyclyloxycarbonyl”, “hetcrocyclyloxycarbonyr, “aryloxycarbonyi”, “aralkyloxycarbonyl”, “bctcroaryloxycarbonyr, and “heteroaralkyloxycarbonyl” are likewise defined.
The term "alkylcarbonyloxy" as used herein means an alkylcarbonyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. Representative examples of alkylcarbonyloxy include, but are not limited to, acetyloxy, ethylearbonyloxy, and tert-butyicarbonyloxy. The terms “haloalkylcarbonyloxy”, “fiuoroalkylcarbonyloxy”, “alkenylcarbonyloxy”, “alkynylcarbonyloxy”, “carbocyclylcarbonyloxy” “hctcrocyclylcarbonyloxy”, “arylcarbonyloxy”, “aralkylcarbonyloxy”, “heteroarylcarbonyloxy", and “hctcroaralkylcarbonyloxy” are likewise defined.
The term “alkylsulfonyloxy” as used herein means an aikylsuifonyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. The terms “haloalkylsuifonyioxy”, “fluroralkylsulfonyloxy”, “alkenylsulfonyloxy”, “alkynylsulfonyloxy”, “carbocyclylsulfonyloxy”, “heterocyclylsulfonyloxy”, “arylsulfonyloxy”, “aralkylsulfonyloxy”, “hctcroarylsulfonyloxy", “hetcroaralkylsulfonyloxy”, “haloalkoxysulfonyloxy”, “fluroralkoxysulfonyloxy”, “alkcnyloxysulfonyloxy”, “alkynyloxysulfonyloxy”, “carbocyclyloxysulfonyloxy”, “hclcrocyclyloxysulfonyloxy”, “aryloxysulfonyloxy”, “aralkyloxysulfonyloxy”, “heteroaryloxysulfonyloxy” and “heteroaralkyloxysulfonyloxy·’
The term "amino" as used herein refers to -NH2 and substituted derivatives thereof wherein one or both of the hydrogens are independently replaced with substituents selected from the group consisting of alkyl, haloalkyl, fluoroalkyl, alkenyl, alkynyl, carbocyclyl, hctcrocyclyl, aryl, aralkyl, hctcroaryi, hctcroaralkyl, alkylcarbonyl, haloalkylcarbonyl, fluoroalkylcarbonyl, alkcnylcarbonyl, alkynylcarbonyl, carbocyclylcarbonyl, hctcrocyclylcarbonyl, arylcarbonyl, aralkylcarbonyl, hetcroarylcarnbonyl, hctcroaralkylcarbonyl and the sufonyl and sulfinyl groups defined above; or when both hydrogens together are replaced with an alkylcne group (to form a ring which contains the nitrogen). Representative examples include, but are not limited to methylamino, acetylamino, and dintethylamino.
The term “amido” as used herein means an amino group, as defined herein, appended to the parent molecular moiety through a carbonyl.
The term "cyano" as used herein means a -C=N group.
The term "nitro" as used herein means a -NOj group.
The term “azido” as used herein means a -N3 group.
The term "phosphinyl" as used herein includes -PH3 and substituted derivatives thereof wherein one, two or three of the hydrogens are independently replaced with substituents selected from the group consisting of alkyl, haloalkyl, fluoroalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, aralkyl, heteroaryl, heteroaralkyl, alkoxy, haloalkoxy, fluoroalkyloxy, aikenyloxy, alkynyloxy, carbocyclyloxy, hcterocyclyloxy, aryloxy, aralkyloxy, heteroaryloxy, heteroaralkyloxy, and amino.
The term “phosphoryl” as used herein refers to -P(=0)0I-T and substituted derivatives thereof wherein one or both of the hydroxyls are independently replaced with substituents selected from the group consisting of alkyl, haloalkyl, fhioroalkyl, alkenyl, alkynyl, carbocycfyl, heterocyclyl, aiyl, aralkyl, heteroaryl, heteroaralkyl, alkoxy, haloalkoxy, fluoroalkyloxy, alkenyloxy, alkynyloxy, carbocyclyloxy, heterocyclyloxy, aryloxy, aralkyloxy, heteroaryloxy, heteroaralkyloxy, and amino.
The term "silyl" as used herein includes HiSi- and substituted derivatives thereof wherein one, two or three of the hydrogens arc independently replaced with subsitutuents selected from alkyl, haloalkyl, ftuoroalkyl, alkenyl, alkynyl, carbocyciyl, heterocyclyl, aryl, aralkyl, hctcroaryl, and hcteroaralkyl, Rcprcscntitivc examples include trimcthylsilyl (TMS), tcrt-butyldiphcnylsilyl (TBDPS), tcrt-butyldimethylsilyi (TBS/TBDMS), triisopropylsiiyl (TIPS), and [2-(trimcthylsilyl)ethoxy]methyl (SEM).
The term "silyloxy" as used herein means a silyl group, as defined herein, is appended to the parent molecule through an oxygen atom.
The abbreviations Me, Et, Ph, Tf, Nf, Ts, and Ms represent methyl, ethyl, phenyl, trifluoromclhancsulfonyl, nonafluorobutancsulfonyl, p-tolucncsulfonyl and mcthancsulfony], respectively. A more comprehensive list of the abbreviations utilized by organic chemists of ordinary skill in the art appears in the first issue of each volume of the Journal of Organic Chemistry, this list is typically presented in a table entitled Standard List of Abbreviations.
As used herein, the term “administering" means providing a pharmaceutical agent or composition to a subject, and includes, but is not limited to, administering by a medical professional and self-administering.
As used herein, the phrases “ncurodcgcncrative disorder” and “ncurodcgcncrativc disease” refers to a wide range of diseases and/or disorders of the central and peripheral nervous system, such as ncuropathologics, and includes but is not limited to, Parkinson’s disease, Alzheimer’s disease (AD), amyotrophic lateral sclerosis (ALS), denervation atrophy, otosclerosis, stroke, dementia, multiple sclerosis, Huntington’s disease, encephalopathy associated with acquired immunodeficiency disease (AIDS), and other diseases associated with neuronal cell toxicity and cell death.
As used herein, the phrase "phamiaceuiically acceptable" refers to those agents, compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio,
As used herein, the phrase "pharmaceutically-acceptable carrier" means a pharmaceuticaliy-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material, involved in carrying or transporting an agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaccutically-acccptablc carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymcthyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl lauratc; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogcn-frcc water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) pH buffered solutions; (21) polyesters, polycarbonates and/or polyanhydridcs; and (22) other non-toxic compatible substances employed in pharmaceutical formulations.
As used herein, the phrase “pharmaceuticaliy-acceptable salts” refers to the relatively non-toxic, inorganic and organic salts of compounds.
As used herein, the phrase “protcinopathy” refers to any disease associated with the accumulation and/or aggregation of abnormal or misfoldcd proteins. Though protcinopathics arc frequently neurodcgcncrative diseases, protcinopathics also include diseases of other tissues, including the liver, muscle and heart, and include some cancers.
As used herein, the term “subject” means a human or non-human animal selected for treatment or therapy.
As used herein, the phrase “subject suspected of having” means a subject exhibiting one or more clinical indicators of a disease or condition. In certain embodiments, the disease or condition is cancer, a neurodegenerative disoi'dcr or pancreatitis.
As used herein, the phrase “subject in need thereof' means a subject identified as in need of a therapy or treatment of the invention.
As used herein, the phrase “therapeutic effect” refers to a local or systemic effect in animals, particularly mammals, and more particularly humans, caused by an agent. The phrases “therapeutical ly-effective amount” and “effective amount” mean the amount of an agent that produces some desired effect in at least a sub-population of cells. A therapeutically effective amount includes an amount of an agent that produces some desired local or systemic effect at a reasonable benefit/risk ratio applicable to any treatment. For example, certain agents used in the methods of the present invention may be administered in a sufficient amount to produce a reasonable benefit/risk ratio applicable to such treatment.
As used herein, the term “treating” a disease in a subject or “treating” a subject having or suspected of having a disease refers to subjecting the subject to a pharmaceutical treatment, e.g., the administration of an agent, such that at least one symptom of the disease is decreased or prevented from worsening.
As used herein, “any of the aforementioned compounds” is any compound of formula I, II, III, IV, V, VI, VII, and VIII.
Inhibitors of Uspl4
One aspect of the invention relates to a compound represented by formula I:
I or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof: wherein, independently for each occurrence, A is aryl, heteroaryl, carbocyclyi, heterocyclyl, or biaryl; R1 is hydrogen, alkyl, haloalkyl, fluoroalkyl, tower alkyoxy, halo or trifluoromethyl; G is -N= or -C(R2)=; Z is =C(RS)-, =C(R2)- or=N-; R2 is hydrogen, alkyl, haloalkyl, fluoroalkyl, lower alkyoxy, halo or trifluoromethyl; or, when G is -C(R2)= and Z is =C(R2)-, the two R2 taken together are
or hctcroaryl;
Y is -CH2NR2R·', -CH2(N-heterocyclyl), -CH2NH(CH2)„NH(alkyl), -CH2NH{CH2)i,N(alkyl)2, -CH2NH(CH2)j;(N-heterocyclyl), -CH2N(alkyl)(C'H2)nNH(a!kyl), -CH2N(alkyl)(CH2)nN(alkyl)2, -CH2N(aIkyl)(CH2)n(N-heterocyclyl), -CH2NH(CH2)nO{alkyl), -CH2N(alkyl)(CH2)nO(alkyi), -NR-R4, -NR-W'R7, -MR5(N-hctcrocyclyl), or -N-hetcrocyclyl; n is 1, 2, 3 or 4; R3 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, hctcroaryl, or hctcroaralkyl; R4 ivS hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; R3 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; R6 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, hctcroaryl, or heteroaralkyl; R7 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, hctcroaryl, or hctcroaralkyl;
Rs is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; R9 is alkyl; or two R9 taken together with the nitrogen to which they are bound are an N-heterocyclyl group; and R10 is hydrogen, alkyl, hatoalkyl, fluoroalkyl, alkyoxy, alkoxyalkyl, halo, trifluoromcthyl, sul foxy methyl, sulfonamido, amino, amido, N-hctcrocycIyl, aminoalkyl, amidoalkyl, or N-hetrocyclylalkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, provided that when A is 4-fluorophenyl, R1 is methyl, G is -C(R2)=, R2 is methyl, X is
and Y is -CH2(piperidin-l-yl), Z is not =C(H)-.
In certain embodiments, the present invention relates to any of the aforementioned compounds, provided that when A is 4-methylphenyl, R1 is methyl, G is -C(R2)=, R2 is methyl, X is
and Y is -CH2(4-rncthylpiperidin-l-yl), Z is not =C(H)- (i.e., Cl00).
In certain embodiments, the present invention relates to any of the aforementioned compounds, provided that when A is 4-chlorophcnyl, R1 is methyl, G is -N=, X is
and Y is -NH2, Z is not =N- (i.e., Cl21).
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein G is -N=.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein G is -C(R2)=. in certain embodiments, the present invention relates to any of the aforementioned compounds, wherein the compound is represented by formula II:
II or is a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof.
Another aspect of the invention relates to a compound represented by formula III:
III or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof; wherein, independently for each occurrence, A is aryl, hctcroaryl, carbocyclyl, hetcrocyclyl, or biaryl; Z is =C(R2)- or -N-; R2 is hydrogen, alkyl, haloaikyl, fluoroalkyl, lower alkyoxy, halo or trifluoromethyl; or, R2 and X taken together are
or
or hetcroaryl; Y is -CHzNRV, -CH2(N-heterocyc!yl), -CH2M4(CH2)n]vlH(alkyl), -CH2NH(CH2)JSN(alkyl)2, -CH2NH(CH2)1}(N-heterocyc]yl), -CH2N(a!kyl)(CH2)I1NH(alkyl), -CH2N(alkyl)(CH2}nN(alkyl)2! ~CH2N(alkyi)(CH2)n(N-heteiOcyciyl), -CkbNHfCHjXOCalkyl), -CH2N(alkyl)(CH2)nO(alkyi), -NR- R4, -NR5NRf'R7, -NR5(N-hctcrocyclyl), or -N-hctcrocyclyl; n is 1,2, 3 or 4; R·’ is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloaikyl, fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; R4 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloaikyl, fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; R* is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fiuoroalkyl, aryl, aralkyl, hctcroaryl, or heteroaralkyl; R6 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fiuoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl: R7 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fiuoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; R9 is alkyl; or two R9 taken together with the nitrogen to which they are bound are an N-hctcrocycly! group; and RI(J is hydrogen, alkyl, haloalkyl, fiuoroalkyl, alkyoxy, alkoxyalkyl, halo, trifluoromethyl, suifoxymethyl, sulfonamido, amino, amido, N-hcterocyclyi, aminoalkyl, amidoalkyl, or N-hctrocyciylaikyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, provided that the compound is not Cl2.
In certain embodiments, the present invention relates to any of the aforementioned compounds, provided that the compound is not C31.
Another aspect of the invention relates to a compound represented by formula IV:
IV or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof; wherein, independently for each occurrence, A is aryl, heteroaryl, carbocyclyl, heterocyclyl, or biaryl; and
Rn is hydrogen, alkyl, alkylcarbonyl, aralkyl, haloalkyl, fiuoroalkyl, alkoxyalkyl, trifluoromethyl, or silyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, provided that the compound is not C73.
Another aspect of the invention relates to a compound represented by formula V:
V or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chcmically-protcctcd form, enantiomer or stereoisomer thereof; wherein, independently for each occurrence, A is aryl, heteroaryl, carboeyclyl, hetcrocyclyl, or biaryl; R12 is hydrogen or alkyl; and
Rl' is hydrogen, alkyl, alkenyl, alkynyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, alkyenytoxy, alkynyloxy, carbocyclyloxy, heterocyclyloxy, haloalkoxy, fluoroalkyloxy, formyl, alkylcarbonyl, haloalkylcarbonyi, fluoroalkylcarbonyl, alkcnylcarbonyl, alkynylcarbonyl, carboxy, alkoxycarbonyl, haloalkoxycarbonyi, fluoroalkoxycarbonyl, alkcnyioxycarbonyl, alkynyloxycarbonyl, alkyicarbonyloxy, haloalkylcarbonyloxy, fltioroalkylcarbonyloxy, alkenylcarbonyloxy, alkynylcarbonyloxy, sulfoxymethyl, sulfonamido, amino, amido, azido, aminosulfonyi, aminosulfinyl, cyano, nitro, phosphinyl, phosphoryl, silyl, silyloxy, and any of said substiuents bound through a methylene or ethylene moiety; or one or two instances of Ru, and the carbon to which it is bound, taken together are -N=.
In certain embodiments, the present invention relates to any of the aforementioned compounds, provided that the compound is not Cl06.
Another aspect of the invention relates to a compound represented by formula VI:
VI or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof; wherein, independently for each occurrence, A is ary!, heteroaryl, carboeyclyl, hetcrocyclyl, or biaryl; R1 is hydrogen, alkyl, haloalkyl, fluoroalkyl, lower alkyoxy, halo or trifluoromethyl;
Rt4 is hydrogen orX; both R|J, taken together, arc
: and R1' is hydrogen, alkyl, alkenyl, alkynyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, alkycnyloxy, alkynyloxy, carbocyclyloxy, hcterocyclyloxy, haloalkoxy, fltioroalkyloxy, formyl, alkylcarbonyl, haloalkylcarbonyl, fluoroalkylcarbonyl, alkenylcarbonyl, alkynylcarbonyl, carboxy, alkoxycarbonyl, haloalkoxycarbonyl, fluoroalkoxycarbonyl, alkenyloxycarbonyl. alkynyloxycarbonyl, aikylcarbonyloxy, haloalkylcarbonyloxy, fluoroalkylcarbonyloxy, alkenylcarbonyloxy, alkynylcarbonyioxy, sulfoxymcthyl, sulfonamido, amino, amido, azido, aminosulfonyl, aminosulfinyi, cyano, nitro, phosphinyl, phosphoryl, silyl, silyioxy, and any of said substiuents bound through a methylene or ethylene moiety; or one or two instances of RL\ and the carbon to which it is bound, taken together are N.
In certain embodiments, the present invention relates to any of the aforementioned compounds, provided that the compound is not Cl 18.
Another aspect of the invention relates to a compound represented by formula VII:
VII or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chcmicaliy-protcctcd form, enantiomer or stereoisomer thereof; wherein, independently for each occurrence, A is aryl, heteroaryl, carbocyclyl, hcterocyclyl, or biaryl; and Y is -CHzNRV, -CH^-heterocyclyi), -CPI2NH (CH 2)nNH(aiky 1), -CH2NH(CH2)nN(alkyi)2, -CH2NH(CH2)n(N-heteiOcyclyl), -CH2N(alkyl)(CH2)nNH(alkyl), -CH2N(alkyl)(CH2)nN(alkyl)2l -CH2N(alkyl)(CH2)n(N-hcteiOcyclyl), -CH2NH(CH2),0(alkyI), -CH2N(alkyl)(CH2)plO(alkyI)1 -NR’R4, -NR5NRf'R7 or -NR5{N-hctcrocyclyl). in certain embodiments, the present invention relates to any of the aforementioned compounds, provided that the compound is not Cl33.
Another aspect of die invention relates to a compound represented by formula VIM:
VIIJ or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof; wherein, independently for each occurrence, A is aryl, hctcroaryl, carbocyclyl, heterocyclyl, or biaryl; and R12 is hydogen or alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, provided that the compound is not Cl39.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is aryl or heteroaryl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is phenyl, pyridin-2-yI, pyridin-3-yl or pyrimidin-2-yl, optionally substituted with 1,2, 3, 4 or 5 substituents independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, alkyenyloxy, alkynyloxy, carbocyclyloxy, hcterocyclyloxy, haloalkoxy, fluoroalkyloxy, formyl, alkylcarbonyl, haloalkylcarbonyl, fluoroalkyl carbonyl, alkenylcarbonyl, alkynylcarbonyl, carboxy, alkoxycarbonyl, haloalkoxycarbonyl, fluoroalkoxycarbonyi, alkenyloxycarbonyl, alkynyloxycarbonyl, alkylcarbonyloxy, haloalkylcarbonyloxy, fluoroalkylcarbonyloxy, alkcnylcarbonyloxy, alkynylcarbonyloxy, sulfoxymethyl, sulfonamido, amino, amido, azido, aminosulfonyl, aminosulfinyl, cyano, nitro, phosphinyl, phosphoryl, silyl, silyloxy, and any of said substiuents bound to the phenyl, pyridin-2yl, pyridin-3-yl or pyrimidin-2-yl through a methylene or ethylene moiety.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is phenyl, optionally substituted with 1, 2, 3,4 or 5 substituents independently selected from the group consisting of al kyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is phenyl substituted in the two position (ortho substituted) with a substituted selected from the group consisting of alky!, halo, haloalkyl, fluoroalkyl, hydroxy, aikoxy, haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is phenyl substituted in the three position (meta substituted) with a substituted selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, aikoxy, haloalkoxy, fluoroalkyloxy, amino, azido cyano, and nitro. in certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is phenyl substituted in the four position (para substituted) with a substituted selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, aikoxy, haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is phenyl substituted in the two and four positions with substitutents independently selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, aikoxy, haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is pyridin-2-yI, optionally substituted in the four position with a substituent selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is pyrimidin-2-yl, optionally substituted in the four position with a substituent selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is biaryl. in certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is 4-(phcnyl)phen-l~yl or 4-(2-pyridinyl)phen-1-yl, optionally substituted with 1, 2, 3, 4 or 5 substituents independently selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein A is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is haloalkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is fluoroalkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R! is methyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is halomethyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is fluoromcthyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R! is ethyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is haloethyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is fluorocthyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R2 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R2 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R2 is methyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R2 is ethyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is hydrogen; and R2 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is alkyl; and R2 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is methyl; and R2 is methyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R1 is ethyl; and R2 is ethyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is =C(R8)-; and R8 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is “CfR*)-; and Rs is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is =N-.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein X is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein X is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein X is or
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein X is heteroaiyl.
In certain embodiments, the present invention relates to any of (he aforementioned compounds, wherein X is pyriOlo[l,2-a]pyrazin-3-yi.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R9 is alkyl. In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R9 is methyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR3R4.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR’R4; and R3 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR'R4; and R3 is alkyl. In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR3R4; and R4 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR~'R4; and R4 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR3R4; and R4 is atkoxyalkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR',R4: R' is hydrogen; and R4 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR’R4; R3 is alkyl; and R4 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR''R4: R' is hydrogen; and R4 is alkoxyalkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NR3R4; RJ is alkyl; and R4 is alkoxyalkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2(N-heterocyclyl), which is optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of alkyl, haloalkyf, fluoroalkyl, halo, hydroxyl, alkoxy, haloalkoxy, fluoroalkoxy, amino and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2(pipcridin-l-yl), -CH2(pipcrazin-l-yl), -CH2(hexahydropyrimidin-l-yl), -CH2(morpholin-1 -yl) or -CH?( 1,3-oxazinan-3-yl), which is optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of alkyl, haloalkyl, fluoroalkyl, halo, hydroxyl, alkoxy, haloalkoxy, fluoroalkoxy, amino and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2(piperidin-l-yl) or -CH2(piperazin-l -yl), which is optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of alkyl, haloalkyl, fluoroalkyl, halo, hydroxyl, alkoxy, haloalkoxy, fluoroalkoxy, amino and nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NH(CH2)nNH(alkyl), -CH2NH(CH2)nN(alkyl)2, -CH2NH(CH2)nN(alkylcnc), -CH2N(alkyl)(CH2)nNH(alkyI), -CH.NfalkylXCHj^Nialkyl)^ or -CH2N(alkyl)(CH2)nN(aIkylene).
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -CH2NH(CH2)nO(alkyl) or -CH2N(alkyl)(CH2)nO(alkyl).
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein n is 1.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein n is 2.
In certain embodiments, the present invent ion relates to any of the aforementioned compounds, wherein n is 3.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein n is 4.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is or
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR3R4.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR3R4; and R3 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR"'R4; and R"' is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR3R4; and R4 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR'R4; and R4 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR'R4; R3 is hydrogen: and R4 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR'R4; R3 is hydrogen; and R4 is hydrogen. In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR'R4; R/' is alkyl; and R4 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR5NRCR7 or-NR5(N-hcterocyc!yl).
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR'^NR^R7; and R5 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR5NR6R7; and R5 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR5NR°R7; and R5, R6 and R7 are, independently, hydrogen or alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR5(N-heterocyciyl); and iV is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is -NR5(N-hcterocyclyi): and R5 is alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Y is
or
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is =C(R2)-; and the two R2 taken together are
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is =C(R2)-; and tire two R2 taken together are
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is =C(R2)-; and the two Rz taken together are
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is =C(R2)-; and the two R2 taken together are
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is =C(R2)-; and the two R2 taken together are
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Rl() is hydrogen, alkyl, haloalkyl, fluoroalkyl, alkyoxy, aikoxyalkyl, halo or trifluoromethyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R10 is hydrogen, amino, amido, N-heterocyclyl, aminoalkyl, amidoalkyf, orN-hetrocyclylalkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R!0 is hydrogen, halo or N-hctcrocyclyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R10 is hydrogen, chloro or pipcridin-l-yf.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Ri0 is hydrogen or N-heterocyclylalkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R10 is hydrogen or pipcridm-l-ylmcthyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R10 is hydrogen or alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R10 is hydrogen. in certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Rn is hydrogen or alkyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Rn is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R11 is methyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R12 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R12 is methyl. In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R13 is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein exactly one R13, and the carbon to which it is bound, is -N=.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein RM is hydrogen.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein R14 is X. In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is -C(R2)-; the two R2 taken together are
; and R1(1 is hydrogen, halo or N-heterocyclyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein Z is =C(R2)-; the two R2 taken together arc
; and R!i) is hydrogen or N-hetcrocyclylalkyl.
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, selected from the group consisting of
and
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, selected from the group consisting of
and
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, of the following formula
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, of the following formula
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prod rug, chemically-protected form, enantiomer or stereoisomer thereof, selected from the group consisting of
and
; wherein W is methyl, fluoro, chloro, nitro, methoxy, ethoxy, -SO2NH2 or -C(=0)NH2·
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, selected from the group consisting of
; wherein W is alkyl, fluoro, chloro, nitro, methoxy, ethoxy, -SO2NH2 or-C(=0)NH2.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein wherein W is methyl.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein wherein W is fluoro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein wherein W is chloro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein wherein W is nitro.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein wherein W is methoxy.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein wherein W is ethoxy.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein wherein W is -SO2NH2.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein wherein W is -C(=0)NH2.
Another aspect of the invention relates to a compound, ora pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, selected from the group consisting of
wherein W is alkyl, fluoro, chloro, nitro, methoxy, ethoxy, -SO2NH2 or-C(=0)NH2.
In certain embodiments, the present invention relates to any of the aforementioned compounds, wherein wherein W is chloro.
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chcmically-protcctcd form, enantiomer or stereoisomer thereof, selected from the group consisting of
and
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, selected from the group consisting of
, and
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, selected from the group consisting of
and
Another aspect of the invention relates to a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, selected from the group consisting of
and
Many of the compounds of the invention may be provided as salts with pharmaceutically compatible counterions (i.e., pharmaceutically acceptable salts). A “pharmaceutically acceptable salt” means any non-toxic salt that, upon administration to a recipient, is capable of providing, cither directly or indirectly, a compound or a prodrug of a compound of this invention. A “pharmaceutically acceptable counterion” is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient. Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms.
Acids commonly employed to form pharmaceutically acceptable salts include inorganic acids such as hydrogen bisulfide, hydrochloric, hydrobromic, hydroiodic, sulfuric and phosphoric acid, as well as organic acids such as para-toluencsulfonic, salicylic, tartaric, bitartaric, ascorbic, maleic, besylic, fumaric, gluconic, glucuronic, formic, glutamic, methancsulfonic, ethancsulfonic, bcnzcnesulfonic, lactic, oxalic, para-bromophenylsulfonic, carbonic, succinic, citric, benzoic and acetic acid, and related inorganic and organic acids. Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolatc, oxalate, malonatc, succinate, suberate, scbacate, fumaratc, malcate, butync-1,4-dioate, hcxync-l,6-dioatc, benzoate, chlorobenzoate, mcthylbcnzoatc, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthaiate, tercphathalatc, sulfonate, xylencsulfonatc, phcnylacctatc, phenylpropionatc, phenylbutyrate, citrate, lactate, .beta.-hydroxybutyrate, glycolate, maleate, tartrate, methanesulfonate, propanesulfonate, naphthalene-1-sulfonate, naphthalenc-2-sulfonate, mandelatc and the like salts. Pharmaceutically acceptable acid addition salts include those formed with mineral acids such as hydrochloric acid and hydrobromic acid, and those formed with organic acids such as maleic acid.
Suitable bases for forming pharmaceutically acceptable salts with acidic functional groups include, but arc not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxy-substituted mono-, di-, or trialkylamines; dicyclohcxylamine; tributyl amine; pyridine; N-methyl,N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-hydroxy-lowcr alkyl amines), such as mono-, bis-, or tris-(2-hydroxyethyl)amine, 2-hydiOxy-tert-butylamine, or tris-(hydroxymethyl)methylamine, N,N-di-lower alkyl-N-(hydroxy lower alkyl)-aminc$, such as N,N-dimcthyI-N-(2-hydroxycthyl)aminc, or tri-(2-hydroxycthyl)amine; N-mcthyl-D-glucaminc; and amino acids such as arginine, lysine, and the like.
Certain compounds of the invention and their sails may exist in more than one crystal form and the present invention includes each crystal form and mixtures thereof.
Certain compounds of the invention and their salts may also exist in the form of solvates, for example hydrates, and the present invention includes each solvate and mixtures thereof.
Certain compounds of the invention may contain one or more chiral centers, and exist tn different optically active forms. When compounds of the invention contain one chiral center, the compounds exist in two enantiomeric forms and the present invention includes both enantiomers and mixtures of enantiomers, such as racemic mixtures. The enantiomers may be resolved by methods known to those skilled in the art, for example by formation of diastereoisomeric salts which may be separated, for example, by crystallization; formation of diastereoisomeric derivatives or complexes which may be separated, for example, by crystallization, gas-liquid or liquid chromatography; selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic esterification; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support for example silica with a bound chiral ligand or in the presence of a chiral solvent. It will be appreciated that where the desired enantiomer is converted into another chemical entity by one of the separation procedures described above, a further step may be used to liberate the desired enantiomeric form. Alternatively, specific enantiomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer into the other by asymmetric transformation.
When a compound of the invention contains more than one chiral center, it may exist in diastereoisomeric forms. The diastereoisomeric compounds may be separated by methods knowm to those skilled in the art, for example chromatography or crystallization and the individual enantiomers may be separated as described above. The present invention includes each diastereoisomer of compounds of the invention and mixtures thereof.
Certain compounds of the invention may exist in different tautomeric forms or as different geometric isomers, and the present invention includes each tautomer and/or geometric isomer of compounds of the invention and mixtures thereof.
Certain compounds of the invention may exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers. The present invention includes each conformational isomer of compounds of the invention and mixtures thereof.
Certain compounds of the invention may exist in zwitterionic form and the present invention includes each zwitterionic form of compounds of the invention and mixtures thereof.
The present invention also includes pro-drugs. As used herein the term "pro-drug" refers to an agent which is converted into the parent diug in vivo by some physiological chemical process (e.g., a prodrug on being brought to the physiological pH is converted to the desired drag form). Pro-drags are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not. The prodrug may also have improved solubility in pharmacological compositions over the parent drug. An example, without limitation, of a pro-drug would be a compound of the present invention wherein it is administered as an ester (the "pro-drug") to facilitate transmittal across a cell membrane where water solubility is not beneficial, but then it is metabolically hydrolyzed to the carboxylic acid once inside the ceil where water solubility is beneficial. Pro-drugs have many useful properties. For example, a pro-drug may be more water soluble than the ultimate drag, thereby facilitating intravenous administration of the drag. A pro-drug may also have a higher level of oral bioavai lability than the ultimate drag. After administration, the prodrug is enzymatically or chemically cleaved to deliver the ultimate drug in the blood or tissue.
Exemplary pro-drugs upon cleavage release the corresponding free acid, and such hydrolyzable ester-forming residues of the compounds of this invention include but arc not limited to carboxylic acid substituents (e.g., -C(0)2H or a moiety that contains a carboxylic acid) wherein the free hydrogen is replaced by (Ct-Cfralkyl, (C2-Cu)alkanoyIoxymethyl, (C,j-C9)l-(aIkanoyioxy)ethyl, 1-methyl-l-(alkanoyIoxy)-cthyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1-(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1-(alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminoniethyl having from 3 to 9 carbon atoms, 1-(N-(alkoxycarbonyI)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4-crotonoiactonyl, gamma-butyrolacton-4-yl, di-N,N-(Cs-C2)alkylamino(C2-C_0alkyl (such as p-dimcthyiaminocthyl), carbamoyi-(Ci-C2)alkyl, N,N-di(Ci-C2)-alkylcarbamoy!-(Ci-C2)alkyl and piperidino-, pyrrolidino- or morpholino(C2-C;*)alkyl.
Other exemplary pro-drugs release an alcohol or amine of a compound of the invention wherein the free hydrogen of a hydroxyl or amine substituent is replaced by (Ci-Cfijalkanoyloxymethyl, I-((Ci-CV)alkanoyloxy)ethyl, 1 -methyl-1 -((Ct-C6)aIkanoyloxy)ethyl, (Ci-C6)alkoxycarbonyl-oxymethyl, N-(C|-C6)alkoxycarbonylamino-methyl, succinoyl, (C|-C6)alkanoyl, a-amino(Ci-C.))alkanoyl, arylactyl and α-aminoacyl, or α-aminoacyl-a-aminoacyl wherein said α-aminoacyl moieties are independently any of the naturally occurring L-amino acids found in proteins, -P(0)(0H)a, -P(0)(0(Ci-CY,)alkyl)2 or glycosyl (the radical resulting from detachment of the hydroxyl of the hcmiacctal of a carbohydrate).
The phrase "protecting group" as used herein means temporary substituents which protect a potentially reactive functional group from undesired chemical transformations. Examples of such protecting groups include esters of carboxyl ic acids, silyl ethers of alcohols, and acetals and ketals of aldehydes and ketones, respectively. The field of protecting group chemistry has been reviewed (Greene, T.W.; Wuts, P.G.M. Protective Groups in Organic Synthesis, 2nd cd.; Wiley: New York, 1991). Protected forms of the inventive compounds are included within the scope of this invention.
The term “chemically protected form,” as used herein, pertains to a compound in which one or more reactive functional groups are protected from undesirable chemical reactions, that is, are in the form of a protected or protecting group (also known as a masked or masking group). It may be convenient or desirable to prepare, purify, and/or handle the active compound in a chemically protected form.
By protecting a reactive functional group, reactions involving other unprotected reactive functional groups can be performed, without affecting the protected group; the protecting group may be removed, usually in a subsequent step, without substantially affecting the remainder of the molecule. Sec, for example, Protective Groups in Organic Synthesis (T, Green and P. Wuts, Wiley, 1991), and Protective Groups in Organic Synthesis (T. Green and P. Wuts; 3rd Edition; John Wiley and Sons, 1999),
For example, a hydroxy group may be protected as ail ether (-OR) or an ester (-0C(=0)R), for example, as: a t-butyl ether; a benzyl, benzhydryl (diphenylmethyl), or trityl (triphcnylmcthyl) ether; a trimcthylsilyl or t-butyldimethylsilyl ether; or an acetyl ester (-0C(=0)C'HJr0Ac).
For example, an aldehyde or ketone group may be protected as an acetal or ketal, respectively, in which the carbonyl group (0(=0)) is converted to a diethcr (C(0R)2), by reaction with, for example, a primary alcohol. The aldehyde or ketone group is readily regenerated by hydrolysis using a large excess of water in the presence of acid.
For example, an amine group may be protected, for example, as an amide (-NRC(=0)R) or a urethane (-NRC(=0)0R), for example, as: a methyl amide (-NHC(=0)CFh); a benzyloxy amide (-NHC(=0)0CH2C0H5NHCbz); as a t-butoxy amide (-NHC(=0)0C(CH2)i,-NHBoc); a 2-biphcnyi-2-propoxy amide (- NHC(=0}OC(CHj)2C'fiH^C6H5NHBoc), as a 9-fluorcnylmcthoxy amide (-NHFmoc), as a 6-nitrovcratryloxy amide (-NHNvoc), as a 2-trimcihylsilylcthyloxy amide (-NHTcoc), as a 2,2,2-trichloroethyloxy amide (-NHTroc), as an allyioxy amide (-NHAlloc), as a 2-(phenylsuifonyl)ethyloxy amide (-NHPsec); or, in suitable cases (e.g.. cyclic amines), as a nitroxide radical.
For example, a carboxylic acid group may be protected as an ester or an amide, for example, as: a benzyl ester; a t-butyl ester; a methyl ester; or a methyl amide.
For example, a thiol group may be protected as a thioethcr (-SR), for example, as: a benzyl thioethcr; or an acetamidomcthyl ether (-SCHiNHC^OjCH?).
Pharmaceutical Compositions
The invention provides pharmaceutical compositions comprising inhibitors of Uspl4. In one aspect, the present invention provides pharmaceutically acceptable compositions which comprise a therapeutical ly-effective amount of one or more of the compounds described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. In another aspect, the agents of the invention can be administered as such, or administered in mixtures with pharmaceutically acceptable carriers and can also be administered in conjunction with other agents. Conjunctive therapy thus includes sequential, simultaneous and separate, or coadministration of one or more compound of the invention, wherein the therapeutic effects of the first administered has not entirely disappeared when the subsequent compound is administered.
As described in detail below, the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; or (8) nasally.
As set out above, in certain embodiments, agents of the invention may be compounds containing a basic functional group, such as amino or alkylamino, and arc, thus, capable of forming pharmaccutically-acccptablc salts with pharmaccutically-acccptablc acids. These salts can be prepared in situ in the administration vehicle or the dosage form manufacturing process, or through a separate reaction of a purified compound of the invention in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed during subsequent purification. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylatc, mesylate, glucohcptonatc, lactobionate, and laurylsulphonatc salts and the like (see, for example, Berge et al. (1977) "Pharmaceutical Salts",,/. Pham. Sci. 66:1-19).
The pharmaceutically acceptable salts of the subject compounds include the conventional nontoxic salts or quaternary ammonium salts of the compounds, e.g.t from non-toxic organic or inorganic acids. For example, such conventional nontoxic salts include those derived from inorganic acids such as hydrochloride, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, palmitic, maleic, hydroxymalcic, phcnyiacctic, glutamic, benzoic, salicyclic, sulfanilic, 2-acctoxybcnzoic, fumaric, tolucncsulfonic, mcthancsulfonic, ethane disulfonic, oxalic, isothionic, and the like. in other cases, the compounds of the present invention may be compounds containing one or more acidic functional groups and, thus, are capable of forming pharmaceufically-acceptable salts with pharmaccutically-acccptablc bases. These salts can likewise be prepared in situ in the administration vehicle or the dosage form manufacturing process, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaccutically-acccptablc metal cation, with ammonia, or with a pharmaccutically-acccptablc organic primary, secondary or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like (see, for example, Berge et ai„ supra).
Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
Examples of pharmaceutical iy-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, cthylcncdiamine tctraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
The formulations of the compounds of the invention may be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated and the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the agent which produces a therapeutic effect.
In certain embodiments, a formulation of the present invention comprises an excipient, including, but not limited to, cyclodcxtrins, liposomes, micelle forming agents, e.g.s bile acids, and polymeric carriers, e.g., polyesters and polyanhydrides; and an agent of the present invention. In certain embodiments, an aforementioned formulation renders orally bioavailable a agent of the present invention.
Methods of preparing these formulations or compositions may include the step of bringing into association a compound of the present invention with the c airier and, optionally, one or more accessory ingredients.
Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microcmulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diiuents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, com, germ, olive, castor and sesame oils), glycerol, tctrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isosteary! alcohols, polyoxyethylene sorbitol and sorbitan esters, macrocrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oii-in-watcr or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and tiic like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A compound of the present invention may also be administered as a bolus, electuary or paste.
In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaccutically-acceptablc carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyirolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol, glycerol monostcaratc, and non-ionic surfactants; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a laic, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-shelled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like. A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch giycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. Compositions of the invention may also be formulated for rapid release, e.g., freeze-dried. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingrcdient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in microencapsulated form, if appropriate, with one or more of the above-described excipients.
Formulations of the pharmaceutical compositions of tire invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
Dosage forms for the topical or transderma] administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically-acceptabie earner, and with any preservatives, buffers, or propellants which may be required.
The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
Transderma! patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.
Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention.
Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically-acceptabie sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
Examples of suitable aqueous and nonaqueous earners which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the ease of dispersions, and by the use of surfactants.
In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form.
Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
Injectable depot forms arc made by forming microencapsulc matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microcmulsions which are compatible with body tissue.
Exemplary formulations comprising agents of the invention arc determined based on various properties including, but not limited to, chemical stability at body temperature, functional efficiency time of release, toxicity and optimal dose.
The preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given in forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories.
Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, arc formulated into pharmaceutically-acccptable dosage forms by conventional methods known to those of skill in the art.
Therapeutic Methods of the Invention
The present invention further provides novel therapeutic methods of treating proteinopathies and other diseases for which enhanced protein breakdown may be therapeutic, including ncurodegcncrativc diseases, comprising administering to a subject, (e.g., a subject in need thereof), an effective amount of a compound of the invention. A subject in need thereof may include, for example, a subject who has been diagnosed with a protcinopathy or a subject who has been treated for a protcinopathy, including subjects that have been refractory to the previous treatment.
The methods of the present invention may be used to treat any protcinopathy. Examples of such proteinophaties include, but are not limited to, Alzheimer’s disease, cerebral β-amyloid angiopathy, retinal ganglion cell degeneration, prion diseases (e.g. bovine spongiform encephalopathy, kuru, Creutzfeldt-Jakob disease, variant Creutzfeldt-Jakob disease, Gcrstmann-Strausslcr-Schcinkcr syndrome, fatal familial insomnia) tauopathics (e.g. frontotemporal dementia, Alzheimer’s disease, progressive supranuclear palsy, corticobasal dcgcration, frontotemporal lobar degeneration), frontcmporal lobar degeneration, amyotrophic lateral sclerosis, Huntington's disease, familial British dementia, Familial Danish dementia, hereditary cerebral hemorrhage with amyloidosis (Iclandic), CADASIL, Alexander disease, Seipinopathies, familial amyloidotlc neuropothy, senile systemic amyloidosis, seipinopathies, AL amyloidosis, AA amyloidosis, type II diabetes, aortic medial amyloidosis, ApoAl amyloidosis, Apoll amyloidosis, ApoAlV amyloidosis, familial amyloidosis of the Finish type, lysozyme amyloidosis, fibrinogen amyloidosis, dialysis amyloidosis, inclusion body myositis/myopathy, cataracts, medullary thyroid carcinoma, cardiac atrial amyloidosis, pituitary prolactinoma, hereditary lattice corneal dystrophy, cutaneous lichen amyloidosis, corneal lactoferrin amyloidosis, corneal lactofcrrin amyloidosis, pulmonary alveolar proteinosis, odontogenic tumor amylois, seminal vesical amyloid, cystric fibrosis, sickle cell disease, critical illness myopathy, von Hippcl-Lindau disease, spinocerebellar ataxia 1, Angelman syndrome, giant axon neuropathy, inclusion body myopathy with Paget disease of bone and frontotemporal dementia (IBMPFD). in some embodiments, the subject pharmaceutical compositions of the present invention will incorporate the substance or substances to be delivered in an amount sufficient to deliver to a patient a therapeutically effective amount of an incorporated therapeutic agent or other materia! as part of a prophylactic or therapeutic treatment. The desired concentration of the active agent will depend on absorption, inactivat ion, and excretion rates of the drug as well as the delivery rate of the compound. It is to be noted that dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. Typically, dosing will be determined using techniques known to one skilled in the art.
The dosage of the subject agent may be determined by reference to the plasma concentrations of the agent. For example, the maximum plasma concentration (Cmax) and the area under the plasma concentration-time curve from time 0 to infinity (AUC (0-4)) may be used. Dosages for the present invention include those that produce the above values for Cmax and AUC (0-4) and other dosages resulting in larger or smaller values for those parameters.
Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
The selected dosage level will depend upon a variety of factors including the activity of the particular agent employed, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could prescribe and/or administer doses of the agents of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. in general, a suitable daily dose of an agent of the invention will be that amount of the agent which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
If desired, the effective daily dose of the agent may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
The precise time of administration and amount of any particular agent that will yield the most effective treatment in a given patient will depend upon the activity, pharmacokinetics, and bioavailability of a particular agent, physiological condition of the patient (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage and type of medication), route of administration, and the like. The guidelines presented herein may be used to optimize the treatment, e.g., determining the optimum time and/or amount of administration, which will require no more than routine experimentation consisting of monitoring the subject and adjusting the dosage and/or timing.
While the subject is being treated, the health of the subject may be monitored by measuring one or more of the relevant indices at predetermined times during a 24-hour period. All aspects of the treatment, including supplements, amounts, times of administration and formulation, may be optimized according to the results of such monitoring. The patient may be periodically reevaluated to determine the extent of improvement by measuring the same parameters, the first such rcevaluation typically occurring at the end of four weeks from the onset of therapy, and subsequent reevafuations occurring every four to eight weeks during therapy and then every three months thereafter. Therapy may continue for several months or even years, with a minimum of one month being a typical length of therapy for humans. Adjustments, for example, to the amount(s) of agent administered and to the time of administration may be made based on these revaluations.
Treatment may be initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage may be increased by small increments until the optimum therapeutic effect is attained. In addition, the combined use an agent that modulates an autotrophy-associated gene product and a second agent, e.g. another agent useful for the treatment of the autophagy-rclatcd disease, may reduce the required dosage for any individual agent because the onset and duration of effect of the different compounds and/or agents may be complimentary.
One aspect of the invention relates method of inhibiting the dcubiquitination activity of a Uspl4 protein comprising contacting the Usp 14 protein with any one of the aforementioned compounds (including IU1), or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof.
Another aspect of the invention relates to a method of enhancing protein degradation by a proteasome in a cell comprising contacting the cell with any one of the aforementioned compounds (including IU1), or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof.
Another aspect of the invention relates to a method of treating or preventing a proteinopathy in a subject comprising administering to the subject any one of the aforementioned compounds (including Mil), or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof.
In certain embodiments, the present invention relates to any of the aforementioned methods, wherein the proteinopathy is selected from the group consisting of Alzheimer’s disease, cerebral β-amyloid angiopathy, retinal ganglion cell degeneration,, bovine spongiform encephalopathy, kuru. Crcutzfcfdt-Jakob disease, variant Crcutzfcidt-Jakob disease, Gcrstmann-Strausslcr-Schcinker syndrome, fatal familial insomnia, frontotemporal dementia, Alzheimer’s disease, progressive supranuclear palsy, corticobasal degcration, frontotemporal lobar degeneration, frontemporal lobar degeneration, amyotrophic lateral sclerosis, Huntington’s disease, familial British dementia, Familial Danish dementia, hereditary cerebral hemorrhage with amyloidosis (Iclandic), CADAS1L, Alexander disease, familial amyloidotic neuropothy, senile systemic amyloidosis, serpinopathies, AL amyloidosis, AA amyloidosis, type II diabetes, aortic medial amyloidosis, ApoAl amyloidosis, ApoII amyloidosis, ApoAl V amyloidosis, familial amyloidosis of the Finish type, lysozyme amyloidosis, fibrinogen amyloidosis, dialysis amyloidosis, inclusion body myositis/myopathy, cataracts, medullary thyroid carcinoma, cardiac atrial amyloidosis, pituitary prolactinoma, hereditary lattice corneal dystrophy, cutaneous lichen amyloidosis, corneal lactoferrin amyloidosis, corneal lactoferrin amyloidosis, pulmonary alveolar proteinosis, odontogenic tumor amylois, seminal vesical amyloid, cystric fibrosis, sickle cell disease and critical illness myopathy.
In certain embodiments, the present invention relates to any of the aforementioned methods, wherein the proteinopathy is Alzheimer’s disease, frontotemporal lobar degeneration, amyotrophic lateral sclerosis or Machado-Joscph disease.
Another aspect of the invention relates to a method of treating or preventing a disease, for which enhanced protein breakdown may be therapeutic, in a subject comprising administering to the subject any one of the aforementioned compounds (including IU1), or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, or a pharmaceutical composition thereof.
In certain embodiments, the present invention relates to any of the aforementioned methods, wherein the disease is selected from the group consisting of von Hippel-Lindau disease, spinocerebellar ataxia I, Angelman syndrome, giant axon neuropathy, inclusion body myopathy with Paget disease of bone and frontotemporal dementia (IBMPFD).
Another aspect of the invention relates to a method of enhancing proteasome function in a subject comprising administering to the subject any one of the aformentioned compounds(including IU1), or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, or a pharmaceutical composition thereof.
Another aspect of the invention relates to a method of increasing degradation of Tau, TDP-43 or ataxin-3 in a subject comprising administering to the subject any one of the aformentioned compounds(inciuding IU1), or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, or a pharmaceutical composition thereof.
In certain embodiments, the present invention relates to any of the aforementioned methods, wherein said subject is human.Isolated Reconstituted Proteasomes
Certain aspects of the invention relate to isolated proteasomes that lack enzymatically active Uch37 but comprise enzymatically active Uspl4. Sell proteasomes can be from any suitable organism. In certain embodiments the proteasomes of the invention arc mammalian proteasomes, such as human or murine proteasomes. Such proteasomes may contain enzymatically inactive Uch37 or may lack Uch37 altogether. The proteasomes of the invention arc useful, for example, in methods of screening for specific inhibitors of Uspl4. See, for example, International Patent Application Publication WO 2008/147536 Al, hereby incorporated by reference in its entirety.
In certain embodiments, the proteasomes of the invention include enzymatically inactive Uch37. Uch37 can be rendered inactive through any method known in the art, including, for example, through mutation of its enzymatic site, through treatment with a Uch37 specific inhibitor, or through treatment with a non-specific deubiquitinasc inhibitor (e,g., through treatment with ubiquitin-vinylsulfone). Treatment of Uch37 with ubiquitin-vinylsulfone results in the generation of viny!sulfone-Uch37 adducts, which are inactive for deubiquitinase activity.
Another aspect of the invention relates to methods of generating proteasomes of the invention. Such methods may include steps of purifying a proteasome lacking Uspl4 but comprising Uch37, treating the purified proteasome with a deubiquitinasc inhibitor, and/or reconstituting the purified proteasome with enzymatically active (JspI4.
Purification of the protcasomc lacking Uspl4 but comprising Uch37 can be done using any suitable method known in the art. For example, human proteasomes can be affinity-purified from a HEK.293 cell line that expresses HTBH-tagged hRpnl 1. The cells can be lysed and the proteasomes affinity purified with NeutrAvidin agarose resin to produce proteasomes lacking Uspl4 but containing Uch37.
Any suitable Uch37 inhibitor can be used in the methods of the invention, including Uch37 specific inhibitors and non-specific deubiquitinase inhibitor (e.g., ubiquitin-vinylsulfone).
The active Usp!4 that is used to reconstitute the proteasomes of the invention can be from any suitable source, including, for example, Uspl4 purified from a mammalian cell or rccombinantly produced Uspl4,
Another aspect of the invention relates to methods of screening for inhibitors of Uspl4 comprising providing a proteasomc of the invention, contacting the protcasome with a test compound and a Uspl4 substrate, and determining whether the test compound inhibits the deubiquitination of the substrate.
Dcubiquitination of the substrate can be detected cither directly or indirectly using any suitable method. For example, in certain embodiments, the substrate is coupled to a reporter that is detectable after cleavage by a deubiquitinase and/or is an ubiquitin-dependant protcasomc substrate (e.g, Ub-AMC). In other certain embodiments, deubuquitination of the substrate is demonstrated by inhibition of substrate degradation.
Another aspect of the invention relates to a kit comprising the isolated proteasome of the invention, instructions of use, and/or a Lisp 14 substrate. In some embodiments the Uspl4 substrate is Ub-AMC and/or polyubiquitinated cyciin B.
EXAMPLES
The invention now being generally described, it will be more readily understood by reference to the following, which is included merely for purposes of illustration of certain aspects and embodiments of the present invention, and is not intended to limit the invention.
Example 1 -- Synthesis oflnhibitors
Figure 29 depicts one approach to the preparation of pyrroles of the invention. By forming a 1,3-diazoie, instead of a pyrrole, similar diazole componds may be prepared. By varying the ring substitution on aryl amine la, or substituting an alkyl amine, hctcroaryl amine, aralkyl amine, etc., a wide variety of compounds may be prepared. Likewise, compouind le may be reacted with a number of nucleophiles to provide a wide variety of compounds. Experimental procedures corresponding to the compounds shown in Figure 29 are provided below.
Synthesis of l-(4-ehlorophenyl)~2,5-dimethylpvrrole (lc). A mixture of la (7.65g, 60-Ommo!) and lb (34.2 g, 300.0 mmol) in acetic acid (40 mL) was heated to 100 "C for 1 hour, the solvent was then evaporated and the residue was purified by silica column chromatography to give lc (11.07 g, yield: 89.8%).
Synthesis of 2-ehloro-l-[l-(4-chIorophenyl)-2,5-dimelhylpym>l-3-yl|ethan-i-one (le). To a solution of AICI.< (7.98 g, 60.0 mmol) in 1,2-dichloroethane (50 mL) was added Id (6.78 g, 60.0 mmol) at 0 °C. The resulting mixture was stirred for 30 min and added to a solution of lc (6.17 g, 30.0 mmoL) in 1,2-dichiorocthane (50 mL) at 0 °C. The reaction mixture was then warmed to room temperature for 2 hour and poured into ice-water (20 mL). The mixture was extracted with dichloromcthane (15 mL x 3), dried over MgSCL and purified by silica column chromatography to give le, 3.37 g, yield: 39.9%
Synthesis of 1-|l-(4-ch!orophenyI)-2,5-dimethylpyrroI-3-yl|-2-piperidyIethan-l-one (1). To a solution of le (85 mg, 0.3 mmol) and tricthyfamine(61 mg, 0.6 mmol) in acetonitrile(10 mL) was added If (28 mg, 0.33 mmol). After being heated to reflux for lhour, the mixture was concentrated and the residue was dissolved in dichloromcthane (30 mL), washed with sat. NaHCCb(10 mL), dried over MgS04 and purified by silica column chromatography to give 1 (83 mg, yield: 83.8%). LC'/MS: 331.1 (ΜΉ) . IHNMR (CDCh, 300 MHz): 7.45-7.49 (2H, dd), 7.10-7.13 (2H, dd), 6.39 (2H, ds), 3.56(214, s), 2.53-2.56 (4H, m), 2.30 (3H, s), 1.98 (3H, s), 1.62-1.70 (4H, m), 1.44-1.49 (2H, m). Example 2 — Uspl4 Mediates Substrate Deubiquitination
To test whether Uspl4 is a potent inhibitor of human protcasomes, a purification procedure was developed to generate protcasomes that lack dctcctible levels of dcubiquitinasc Usp 14 (modified from Wang et al., (2007), Biochemistry, 46, 3553-3565). Briefly, human protcasomes were affinity-purified on a large scale from a stable HEK293 cell line harboring HTBH-tagged hRpnl 1. The cells were Dounce-homogenized in lysis buffer (50 mM NaH2PCL [pH 7.5], 100 raM NaCl, 10% glycerol, 5 mM MgCl2, 0.5% NP- 40,5 mM ATP, and 1 mM DTT) containing protease inhibitors. Lysates were cleared, then incubated with NeutrAvidin agarose resin (Thermo Scientific) overnight at 4 °C. The beads were then washed with excess lysis buffer followed by the wash buffer (50 mM Tris-HCl [pH 7.5], 1 mM MgCh and 1 mM ATP). For VS-proteasomcs, 1 to 1.5 μΜ of Ub-VS (Boston Biochcm) was added to the resin and incubated at 30 °C for 2 h. Residual Ub-VS was removed by washing the beads with at least 20 bed vol of wash buffer. 26S proteasomes were eluted from the beads by cleavage, using TEV protease (Invitrogen).
Using this proteasome purification procedure, Human proteasomes were affinity-purified from a hRpnl 1-tagged line of HEK293 cells. Purification of proteasomes lacking Uspl4 but containing related deubiquitinase Uch37 was confirmed by western blot using an anti-Usp]4 and anti-Uch37 antibodies (Figure 1A and IB, respectively). The purified UspI4-frce proteasome (also described as 26S proteasomes) retained high levels of dcubiquitinating activity that could be irreversibly inhibited by treating the proteasome with ubiquitin-vinylsulfonc (Ub-VS, Yao et ai, (2006) Nat, Cell Biol., rS% 994-1002). Ub-VS inhibits deubiquitination of substrates by forming adducts with the Cys amino acid located in the active site of thiol protease class dcubiquitinating enzymes. As demonstrated in Figure IB, addition of Ub-VS to 26S proteasomes resulted in enzymatically inactive VS-Uch37 adducts forming with all detectable Uch37.
In order to generate pure, recombinant Uspl4 enzyme, GST-Uspl4 (WT and Cl 14A variants) was expressed in E. coll strain Rosetta 2 (DE3) cells (Novagcn). Cultures were grown at 37 °C until ODf,ou reached 0.6 to 0.8, and expression was induced overnight with 1 mM IPTG at room temperature. Ceils were then harvested in PBS containing protease inhibitors and lysed by French press. The cleared lysates were incubated with GST Sepharose 4B resin (GE Healthcare) at 4°C for 1 h, and subsequently washed with excess PBS, followed by PBS containing 100 mM NaCl. The GST moiety was removed by thrombin in the cleavage buffer (50 mM Tris-HCl [pH 8.0], 150 mM NaCl, 2.5 mM CaCl?, and 0.1% 2-mercaptoethanol) for 3 h at room temperature. GST-tagged Uspl4 proteins for proteasome binding assays were eluted before thrombin cleavage using elution buffer (10 mM reduced glutathione in 50 mM Tris-HCl [pH 8.0]).
The inhibited “VS-proteasomes” described above, which lack endogenous deubiquitination activity due to Ub-VS treatment, were successfully reconstituted with recombinant Uspl4 (Figure 2). An Ub-AMC hydrolysis assay was performed with 1 nM of Ub-VS treated human proteasome (VS-Proteasome) alone, 400nM of Uspl4 alone, or VS-proteasome that had been reconstituted with 4 or 40 nM of recombinant Uspl4 protein. As has been described above, the deubiquitination activity of the VS-proteasome was almost completely inhibited (Figure 3). In contrast, the reconstituted Uspl4/VS proteasome demonstrated substantial deubiquitination activity (Figure 3). In fact, the Uspl4/VS protcasome demonstrated an 800-fold increase in Ub-AMC hydrolyzing activity over that of isolated Uspl4 alone (Figure 3). Thus, the enzymatic activity of Uspl4 is increased by its complexing with the protcasome. Therefore, the Ub-AMC assay allows the success of reconstitution to be followed.
The Ub-AMC assay was also used to examine the kinetics of Ub-AMC hydrolysis by the reconstituted Uspl4-proteasome complexes. Ub-AMC hydrolysis by Uspl4/VS proteasomes that had been reconstituted with various amounts of UspJ4 was monitored over a period of 30 minutes (Figure 4). Analysis of the results of this assay demonstrated the affinity of Uspl4 for the protcasome is approximately 4 nM.
Example 3 — Uspl4 Inhibits Proteasomai Degradation
The effect of UspJ4 on the degradation of ubiquitinated substrates was examined using an in vitro degradation assay using the ubiquitin-dependant protcasome substrate polyubiquitinated cyclin B (Ubn-ClnB). In these experiments, Ubn-ClnB was incubated with human proteasomes (4 nM), containing either wild-type or catalytically inactive Uspl4 (60 nM). The catalytically inactive Usp used in these assays was UspI4-Cl 14A, which contains a mutation in UspI4’s active site for deubiquitination. Notably, both wild-type Usp 14 and Uspl4-Cl I4A are able to bind to 26S mammalian proteasomes (Figure 2). As demonstrated in Figure 5, Usp 14 strongly inhibits the degradation of cyclin B, while the active site mutant of Uspl4 showed little inhibitory effect. The lack of inhibition of Ubn-ClnB degradation by the active site mutant indicates that the ubiquitin chain trimming activity of wild-type UspJ4 is required for Uspt4\s inhibition of proteasome degradation. Indeed, extensive trimming of the ubiquitin groups from cyclin B was evident by immunoblot analysis in the samples containing wild-type Usp 14, but was nearly eliminated when catalytically inactive Usp!4 was used (Figure 5).
An effect of Uspl4 on Tau degradation in human cells was observed in the human cell line, HEK293. Tau was cocxprcsscd with exogenous wild-type or catalytically inactive Usp 14 and Tau protein levels were determined by western blot. Expression of wild-type Uspl4, but not enzymatically inactive Usp 14, stabilized Tau in the human cell tine (Figure 6). In fact, expression of enzymatically inactive Uspl4 in HEK293 cells resulted in accelerated Tau degradation (Figure 6B). This dominant negative effect likely reflects the displacement of endogenous, wild-type Usp 14 from the proteasome. This hypothesis was confirmed using a mutant form of Usp 14 that lacks the N-tcrminal UBL domain (Usp 14-AUBL). The N-tcrminal UBL domain (Figure 6A) is the principal proteasome-binding site on Uspi4. Deletion of the UBL attenuated the dominant negative effect (Figure 7), indicating that protcasome binding is required for the mediation of this effect.
The short form (SF) of lisp] 4 is an endogenous Usp 14 splice variant that is expressed from mRNA that lacks a junctional exon (exon 4) between the N-terminal ubiquitin-like domain of Usp 14 and its catalytic domain (Wilson eta!., (2002), Nat. Genet., 32, 420-425; Figure 6A). Like the catalytically inactive mutant ofUspM, Uspl4-SF exhibited a dominant negative effect on Tau stability in HEK293 cells (Figure 6A). This suggests that Uspl4-SF may be an endogenous inhibitor of Usp 14, Consistent with this possibility, Uspl4-SF is able to bind protcasome, but unlike the wild-type enzyme, it is not activated enzymatically by protcasome binding (Figure 7).
Example 4 -- Specific Inhibitors of Uspl4
As demonstrated above, chain trimming at the proteasome by Usp 14 is a key regulatory step in the ubiquitin-dependent proteolytic pathway. Therefore, in order to identify enhancers of proteasome function, a high-throughput screen for small molecule Usp 14 inhibitors was performed using VS-proteasomes reconstituted with recombinant Usp 14 and assayed with Ub-AMC (Figure 8).
Compounds were screened for Uspl4/26S inhibition in 384-wcll low-volume plates in duplicate. Data processing was done by a robust Z-scorc method and each compound was plotted using Spotflre software. Compounds over the cut-off of Z > 5 were mostly autofluoresccnt and were therefore not counted. To exclude quenching compounds that only affect AMC fluorescence, 312 primary hits were tested for quenching of AMC amine, and pure quenchers were scored as false-positives and excluded from further analysis (Figure 8B). Of the 63,052 compounds analyzed in the high-throughput screen, 215 were identified as true inhibitors of Usp 14.
In order to identify compounds that specifically inhibited Uspl4 but were not general dcubiquitinase inhibitors, the 215 hit compounds were countcrscreencd against a panel of dcubiquitinating enzymes. Among the hit compounds that inhibited the activity of Uspl4 but not any other tested dcubiquitinase, l-[l-(4-fluotOphenyl)-2,5-dimethlypynOl-3-yl]-2-pyiroIidin-l-ylethanone (1U1, Figure 9) was selected for further analysis.
Example 5 -- Specific Inhibition of Uspl4 by IU1
Additional studies were performed on the specific Usp 14 inhibitor IU1 (Figure 10). To serve as a negative control, a compound that is structurally similar to IU1, termed “IU1C” (Figure 9B) which docs not inhibit Usp 14 dcubiquitinase (Figure 10B) or enhance protcasome function (Figure 1OC) was also identified.
The specificity of IUI for Uspl4 was determined by testing its ability to inhibit the activity of eight deubiquitinating enzymes of human origin. As seen in Figures 11 and 9C\ despite being a potent inhibitor of proteasome-bound Usp14, JU1 failed to significantly inhibit the other tested deubiquitinating enzymes, including Uch37. Note that Figure 9E shows that IUI does not inhibit the proteasome-bound form of Uch37. Furthermore, IU1 also failed to inhibit the activity of Uspl4 that had not been loaded onto a protcasome (Figure 9D), indicating that IUI specifically inhibits the proteasome-bound, activated form of Uspl4.
As the binding of Uspl4 to the protcasome enhances Uspl4 activity, it was possible that IUI inhibited UspI4 activity by interfering with the Uspl4/proteasome interaction. Therefore, ability of IUI to interfere with the ability of Uspl4 to bind to the proteasome was examined. Purified human proteasomes were incubated with recombinant Uspl4 either in the presence or the absence of various concentrations of IU1. As seen in Figure 12,1UI did not antagonize Uspl4 complcxing with the protcasome, indicating that the inhibitory activity of IU I is not the result of an inhibition of the formation of Usp 14/protcasomc complexes.
The reversibility of IU 1 inhihibition of Uspl4 was next assayed. Usp 14/protcasomc complexes were treated with IUI, followed by centrifugation with a Micron-YM3 filter up to three times. After each spin, the protein complex was tested for dcubiquitinase activity. As demonstrated in Figure 13, the activity of Usp 14 returned following centrifugation, thereby indicating that inhibition of Uspl4 by IUI is rapidly reversible. Consistent with this observation, mass spectrometry analysis of 1U1 inhibited UspI4 failed to detect any covalent IU1 -Usp 14 adducts.
The Usp 14 inhibitory activity of IUI was further quantified by generating two independent IC50 curves for Uspl4/26S protcasome complexes treated with various concentrations of IUI for either 45 minutes (Figure 14A) or 30 minutes (Figure I4B). The data plot of each experiment was fit into a four parameter logistic model (the Hill-slope model) based on guidelines from the NIH Chemical Genomics Center. The results of these experiments indicated that the IC50 value of IUI is 2-5 μΜ (Figure 14).
Using methods similar to those described in Example 3, Cyclin B was used as a substrate to test whether 1U1 influenced the trimming of ubiquitin chains by protcasome complexes. To separate chain trimming from substrate degradation, these assays were done in the presence of protcasome inhibitors. The effectiveness of the protcasomc inhibitors is evidenced by the accumulation of unmodified cyclin B in the assay (Figure 15). When proteasomes that lack Uspl4 were tested, IUI had little or no effect on the release of ubiquitin chains from cyclin B (Figure 15), which is likely mediated by another deubiquitinating enzyme on the proteasome, Rpnl 1. Upon the addition of Uspl4, however, chain trimming by the proteasome complexes was strongly enhanced, as apparent from the increased electrophoretic mobility of the ubiquitinated forms of cyclin B. The further addition of 1U1 to the Uspl4/profcasome complexes reversed this effect, and reduced the chain trimming to a level similar to that of the proteasome complexes that lacked Uspl4 (Figure 15).
It was next tested whether IUI could serve as an enhancer of substrate degradation by the proteasome. Using the methods described in Example 3, an in vitro Ubn-ClnB degradation assay was performed, but this time in the absence or presence of 34 μΜ IU1. The addition of IUI to proteasomes that lack Uspl4 had no effect on substrate degradation or chain trimming, Confirming the results described above, addition of Uspl4 to the protcasomc complex enhanced chain trimming and dramatically inhibited substrate degradation. The addition of IUI stimulated the activity of UspI4-containing proteasomes in degrading Ub-cyclin B and inhibited ubiquitin chain trimming (Figure 16).
Example 6 — Cellular Entry oflUl
The IUI experiments described above were performed in vitro. In order for 1U1 to enhance proteasome degradation in vivo, it is necessary the IUI be able to enter ceils. In order to examine this, entry of IU 1 into cells was assayed by electrospray mass spectrometry using an Agilent scries 1200 LC/6130 system with a rcvcrscd-phasc Cis column. IUI was added to MEFs at 50 μΜ for various periods of time. Cell lysates were coilctcd and ethyl acetate extraction was used to prepare mass spectrometer samples. Ion count of LC/MS traces (m/z at 301) at 0 hr, I hr and 24 hr arc shown (Figure 17). This assay revealed that, when added to the medium at 50 μΜ, IU 1 reached a steady-state concentration of ~19 μΜ within cells by 1 hour, and maintained approximately the same level over the time course of the experiment (Figures 17 and 18). Similar results, extending through two days, were obtained using a separate UV absorption assay (Figure 19). Additionally, IUI concentrations were maintained in the medium as for at least two days. These results indicate that IUI is stable compound within both cells and standard media.
Example 7 -- Enhancement of in vivo Proteasomal Degradation by IU1
To determine whether IUI could enhance proteasomc function in living cells, Tau was expressed in M EF cells, which were then treated with 1U1 at concentrations from 25 to 100 μΜ. Specifically, after 36 hours of Tau and LacZV5 expression, MEF cells were incubated with 0,25,50, 75 or 100 μΜ of 1U1 for 6 hours. As seen in Figure 21 A, JU1 reduced Tau levels at all concentrations tested. No effect was seen on Tau mRNA levels (Figure 2IB).
Other proteins that have been implicated in protcotoxic mechanisms were also tested. Using similar methods to those described above, it was demonstrated that TDP-43 (implicated in frontotemporal lobar degeneration and amyotrophic lateral sclerosis), ataxin-3 (implicated in Machado-Joseph disease) and glial fibrillary acidic protein (GFAP, implicated in Alexander disease) were similarly depleted from cells upon 1U1 treatment (Figure 22A-C). On the other hand, IUI had little or no effect upon the in vivo degradation of the ubiquitin-independent proteasome substrate, GFP-ODC. Together, these results indicate that 1U1 is a general enhancer of the ubiquitin-mediated proteasome degradation.
Oxidized proteins form another class of proteasome substrates that play an important role in human health. Harmful oxidized proteins accumulate upon ageing and arc implicated in a variety of age-related diseases and disorders (Stadtman (2006) Protein oxidation and aging. Free Radic. Res. 40, 1250-1258; Ahmed et ai. (2007) Protein oxidative modifications and replicative senescence of WI-38 human embryonic fibroblasts. Ann. NY Acad. Sci., 1119, 88-96; Moskovitz et ai (2001). Methionine sulfoxide reductase (MsrA) is a regulator of antioxidant defense and lifespan in mammals. Proc. Nat. Acad.
Sci. USA, /967, 12920-)2925.). Protein oxidation was induced by treating cells with menadione, and oxidized species were visualized using an antibody specific for protein carbonyls. Specifically, MEFs were prcincubatcd with vehicle or 75 μΜ 1U1 for 4 hours and then treated with 63 μΜ menadione for 45 minutes. The cells were lysed and lysates were incubated with DNPH and immunoblottcd with anti-DNPH antibody to assay for oxidized proteins. Accumulation of oxidized proteins was reduced in cells treated with IUI than in untreated cells (Figure 23). When proteasome inhibitor PS-341 was added together with IUI, the effect of IUI was eliminated, indicating that IUI does not prevent oxidation reaction, but rather it enhances the proteasomal degradation of the oxidized proteins. These data indicate that there is a Uspl4-inhibitcd ubiquitin-dependent mechanism for the degradation of proteins damaged by reactive oxygen species. Menadione is toxic to cells, and IU1 treatment reduced this toxicity substantially in HEK293 cells (Fig. 30), strongly supporting the hypothesis that proteins are critical targets of oxidative damage in cells. IU1 also reduced the toxicity of an unrelated oxidizing agent, hydrogen peroxide (data not shown). IU1C, the 1U1 variant that is inactive against Uspl4, failed to reduce menadione cytotoxicity (data not shown). Importantly, these experiments indicate that IU1 can promote cell survival during proteotoxic stress.
Example 8 — The Effects of IU 1 on Cellular Proliferation ami Viability
The effect of IU1 on cell viability was next examined by MTT assay. IU1 was added to MEF. HEK293 and HeLa cells at various concentrations, followed by addition of MTT solution after 6, 12,24 or 48 hours of IU 1 incubation. Effects on cell viability became apparent at concentrations over 100 μΜ, well above the doses required to enhance the degradation ofTau, TDP-43, ataxin-3, and oxidized proteins (Figures 21-23).
Moreover, IU1 did not noticeably induce apoptosis in MEF cells, as assessed by TUMEL assay (Figure 31).
Cell proliferation of MEFs (Figure 28) and 293 cells that had been exposed to various concentrations of 1U1 was measured by microscopy in real time. The results of this assay revealed only a slight inhibition in cellular proliferation at 120 μΜ, but no apparent inhibition at lower concentrations (Figure 28). Taken together with the results of the cell viability assays presented above, this indicates that lUl’s inhibition ofubiquitin chain trimming by Uspl4 does not grossly compromise cell function.
EQUIVALENTS
The present invention provides, in part, methods for the enhancement of protein turnover by the protcasome and the treatment of diseases involving either proteasome substrates, upstream components of the ubiquitin-proteasomc pathway, or the proteasome itself. While specific embodiments of the subject invention have been discussed, the above specification is illustrative and not restrictive. Many variations of the invention will become apparent to those skilled in the art upon review of this specification. The appended claims are not intended to claim all such embodiments and variations, and the full scope of the invention should be determined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.
All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.

Claims (108)

  1. Wc claim:
    1. A compound represented by formula II:
    or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof: wherein, independently for each occurrence, A is aryl, hctcroaryl, carbocyclyl, hcterocyclyi, or biaryl; R1 is hydrogen, alkyl, haloalkyl, fluoroalkyl, lower alkyoxy, halo or trifiuoromethyl; Z is =C(RS)-, =C(R2)- or =N~; R2 is hydrogen, alkyl, haloalkyl, fluoroalkyl, lower alkyoxy, halo or trifiuoromethyl; or, when Z is =C(R2)-, the two R2 taken together arc
    or heteroaryl; Y is -CH.NR’R^, -CH2(N-hcterocyclyl), -CH2NH(CH2)nNH(alkyl), -CH2NH(CH2)„N(alkyl)2, -CH2NH{CH2)„(N-hctcrocyclyI), -CH2N(alkyl)(CH2)nNH(alkyl), -CH2N(alkyl)(CH2)„N(alkyl)2l -CH-2N(alkyl)(CH2)n(N-hetcrocyclyl), -CH2NH(CH2)1,0(alkyl)J -CH2N(alkyl)(CH2)nO(alkyl), -NR3R4, -NR5NR6R7 or -NR5(N-hctcrocyclyl); n is 1, 2, 3 or 4; R‘ is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; is hydrogen, alkyl, substituted alkyl, alkoxyalkyt, haloalkyl, fluoroalkyl, aryl, aralkyl, hcteroaryl, or hctcroarallcyi; R5 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; R6 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl. fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; R7 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; R8 is hydrogen, alkyl, substituted alkyl, alkoxyalkyl, haloalkyl, fluoroalkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl; Ry is alkyl; or two R9 taken together with the nitrogen to which they arc bound arc an N-heterocyclyl group; and R10 is hydrogen, alkyl, haloalkyl, fluoroalkyl, alkyoxy, alkoxyalkyl, halo, trifluoromethyi, amino, amido, N-heterocycly 1, aminoalkyl, amidoalkyi, or N-hetrocyclylalkyl; provided that when A is 4-fluorophenyl, R1 is methyl, R2 is methyl, X is
    and Y is -CHjfpiperidin-l-yl), Z is not =C(H)-; and that when A is 4-mcthylphcnyl, R1 is methyl, R2 is methyl, X is
    and Y is -CH2(4-mcthylpipcridm-!-yl), Z is not=C(H)-..
  2. 2. The compound of claim 1, wherein A is aryl or heteroaryl.
  3. 3. The compound of claim i, wherein A is phenyl, pyridin-2-yl, pyridin-3-yl or pynmidin-2-yl, optionally substituted with 1, 2, 3, 4 or 5 substituents independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, alkyenyloxy, alkynyloxy, carbocyclyloxy, heterocyclyloxy, haloalkoxy, fluoroalkyloxy, formyl, alkylcarbonyl, haioalkylcarbonyl, fluoroaikylcarbonyl, alkenylcarbonyl, alkynylcarbonyl, carboxy, alkoxycarbonyl, haloalkoxycarbony!, fluoroalkoxycarbonyi, alkenyloxycarbonyf, alkynyloxycarbonyl, aJkylcarbonyloxy, haloaikylcarbonyloxy, fluoroalkylcarbonyloxy, alkenylcarbonyloxy, aikynylcarbonyloxy, amino, amido, azido, aminosuifonyl, aminosulfmyl, cyano, nitro, phosphinyl, phosphoryl, silyl, silyloxy, and any of said substiucnts bound to the hcterocyciyl group through a methylene or ethylene moiety.
  4. 4. The compound of claim 1, wherein A is phenyl, optionally substituted with 1,2, 3, 4 or 5 substituents independently selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haioalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
  5. 5. The compound of claim 1, wherein A is
  6. 6. The compound of claim 1, wherein A is phenyl substituted in the two position (ortho substituted) with a substitutent selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haioalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
  7. 7. The compound of claim 1, wherein A is
  8. 8. The compound of claim 1, wherein A is phenyl substituted in the three position (meta substituted) with a substitutent selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haioalkoxy, fluoroalkyloxy, amino, azido cyano, and nitro.
  9. 9. The compound of claim 1, wherein A is
    or
  10. 10. The compound of claim 1, wherein A is phenyl substituted in the four position (para substituted) with a substitutent selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haioalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
  11. 11. The compound of claim 1, wherein A is
  12. 12. The compound of claim 1, wherein A is phenyl substituted in the two and four positions with substitutents independently selected from the group consisting of atkyl, halo, haloalkyl, fluoroatkyl, hydroxy, alkoxy, haloalkoxy, fhioroalkyloxy, amino, azido, cyano, and nitro.
  13. 13. The compound of claim 1, wherein A is
  14. 14. The compound of claim 1, wherein A is pyridin-2-yl, optionally substituted in the four position with a substituent selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
  15. 15. The compound of claim 1, wherein A is
  16. 16. The compound of claim 1, wherein A is pyrimidin-2-yl, optionally substituted in the four position with a substituent selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy, haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro.
  17. 17. The compound of claim 1, wherein A is
  18. 18. The compound of claim 1, wherein A is biaryl.
  19. 19. The compound of claim 1, wherein A is 4-(phcnyi)phen-l-y! or 4-(2- pyridinyl)phcn-l-yi, optionally substituted with 1, 2, 3,4 or 5 substituents independently selected from the group consisting of alkyl, halo, haloalkyl, fluoroalkyl, hydroxy, alkoxy. haloalkoxy, fluoroalkyloxy, amino, azido, cyano, and nitro,
  20. 20. The compound of claim 1, wherein A is
  21. 21. The compound of any one of claims 1 -20, wherein R! is hydrogen.
  22. 22. The compound of any one of claims 1 -20, R1 is alkyl, haloalkyl or fluoroalkyl.
  23. 23. The compound of any one of claims 1-20, R1 is methyl, halomcthyl or fluoromethyl.
  24. 24. The compound of any one of claims 1-20, R1 is ethyl, haloethyl or fluoroethyl.
  25. 25. The compound of any one of claims 1-20, R2 is hydrogen.
  26. 26. The compound of any one of claims 1-20, R2 is alkyl, haloalkyl or fluoroalkyl.
  27. 27. The compound of any one of claims 1-20, R2 is methyl, halomcthyl or fluoromethyl.
  28. 28. The compound of any one of claims 1-20, R2 is ethyl, haloethyl or fluoroethyl.
  29. 29. The compound of any one of claims 1-20, R1 is hydrogen; and R2 is hydrogen.
  30. 30. The compound of any one of claims 1-20, R1 is alkyl; and R2 is alkyl.
  31. 31. The compound of any one of claims 1-20, R1 is methyl; and R2 is methyl.
  32. 32. The compound of any one of claims 1-20, R1 is ethyl; and R2 is ethyl.
  33. 33. The compound of any one of claims 1-32, Z is =C(RS)-; and R8 is hydrogen.
  34. 34. The compound of any one of claims 1 -32, Z is =C(Rh)-; and R8 is alkyl.
  35. 35. The compound of any one of claims 1-32, Z is =N-,
  36. 36. The compound of any one of claims 1-24, wherein Z is =C(R2)-; and the two R2 taken together are
  37. 37. The compound of any one of claims 1-32, X is
  38. 38. The compound of any one of claims 1-32, X is
  39. 39. The compound of any one of claims 1-32, X is
  40. 40. The compound of any one of claims 1 -39, Y is -CHiNR’R4.
  41. 41. The compound of any one of claims 1 -39, Y is -CH2NR''R4; and R"’ is hydrogen.
  42. 42. The compound of any one of claims 1-39, Y is -CH2NR3R4; and R'’ is alkyl.
  43. 43. The compound of any one of claims 1-39, Y is -CH2NR‘'R4; and R4 is hydrogen.
  44. 44. The compound of any one of claims 1-39, Y is -CI-kNR’R4; and R4 is alkyl.
  45. 45. The compound of any one of claims 1-39, Y is -CI-RNR^R4; R"’ is hydrogen; and R4 is alkyl.
  46. 46. The compound of any one of claims 1-39, Y is -CH^NR-'R4; R'4 is alkyl; and R4 is alkyl.
  47. 47. The compound of any one of claims 1 -38, Y is
  48. 48. The compound of any one of claims 1-39, Y is -CThiN-heterocydyl), which is optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of alkyl, haloalkyl, fluoroalkyl, halo, hydroxyl, alkoxy, haloalkoxy, fluoroaikoxy, amino and nitro.
  49. 49. The compound of any one of claims 1-39, Y is -CIT>(pipcndin-l-yl), -Ckkfpipcrazin-] -yl), -CHafhcxahydropyrimidin-l-yl), -Ckbimorpholin- 1-yl) or -ChkfJ,3-oxazinan-3-yl), which is optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of alkyl, haloalkyl, fluoroalkyl, halo, hydroxyl, alkoxy, haloalkoxy, fluoroaikoxy, amino and nitro.
  50. 50. The compound of any one of claims 1 -39, Y is -CH2(pipcridin-l-yl) or -CFhfpiperazin-l-yl), which is optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of alkyl, haloalkyl, fluoroalkyi, halo, hydroxyl, aikoxy, haioalkoxy, fluoroaikoxy, amino and nitro.
  51. 51. The compound of any one of claims 1 -39, Y is
  52. 52. The compound of any one of claims 1-39, Y is -CH2NH(CH2)nNH(alkyf), -CH2NH(CH2)nN(alkyl)2, -CHzNH(CH2)nN(alkylcne), -CH2N(alkyl)(CH2)„NH(alkyl), -CH2N(aIkyl)(CH2)„N(alkyl)2 or -CH2N(alkyl)(CH2)„N(alkylene).
  53. 53. The compound of any one of claims 1-38, Y is -CH2NH(CH2)nO(alkyl) or -CH2N(alkyl)(CH2)nO(alkyl).
  54. 54. The compound of claim 52 or 53, wherein n is I.
  55. 55. The compound of claim 52 or 53, wherein n is 2.
  56. 56. The compound of claim 52 or 53, wherein n is 3.
  57. 57. The compound of claim 52 or 53, wherein n is 4.
  58. 58. The compound of any one of claims 1*39, Y is
  59. 59. The compound of any one of claims 1-39, Y is -NRfR'1,
  60. 60. The compound of any one of claims 1-39, Y is -NR'R'1: and R' is hydrogen,
  61. 61. The compound of any one of claims 1-39, Y is -NR^R4; and R’’ is alkyl.
  62. 62. The compound of any one of claims 1-39, Y is -NR’R4; and RJ is hydrogen.
  63. 63. The compound of any one of claims 1 -39, Y is -NR^R^; and R* is alkyl.
  64. 64. The compound of any one of claims 1-39, Y is -NR’R14; R~1 is hydrogen; and R"1 is alkyl,
  65. 65. The compound of any one of claims 1 -39, Y is -NR'R'1; R"' is alkyl; and R^ is alkyl,
  66. 66. The compound of any one of claims 1-39, Y is -NR5NR6R7 or -NR:,(N- hctcrocyciyl),
  67. 67. The compound of any one of claims 1-39, Y is -NR5NRflR7; and R^ is hydrogen.
  68. 68. The compound of any one of claims 3-39, Y is -NR^NR^R7; and R^ is alkyl.
  69. 69. The compound of any one of claims ! -39, Y is -NRiNRf’R7; and R\ Rf’ and R7 are, independently, hydrogen or alkyl.
  70. 70. The compound of any one of claims 1-39, Y is -’NR'XN-hctcrocycly!); and R5 is hydrogen.
  71. 71. The compound of any one of claims 1-39, Y is -NR^N-hctcrocyclyl); and R’^ is alkyl.
  72. 72. The compound of any one of claims 1-39, Y is
    or
  73. 73. A compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, selected from the group consisting of
    and
    , wherein W is methyl, fluoro, chloro, nitro, methoxy, ethoxy, -SO2NH2 or -0(=0)14¾.
  74. 74. A pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, of any one of claims 1 -73, or 1U1, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, and a pharmaceutically acceptable excipient.
  75. 75. A method of inhibiting the dcubiquitination activity of a Uspl4 protein comprising contacting the Uspl4 protein with a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, of any one of claims 1-73, or IU1, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof.
  76. 76. A method of enhancing protein degradation by a proteasome in a cell comprising contacting the cell with a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, of any one of claims 1-73, or IU1, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof.
  77. 77. A method of treating or preventing a proteinopathy in a subject comprising administering to the subject a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, of any one of claims 1-73, or IU1, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof or a pharmaceutical composition of claim 74.
  78. 78. The method of claim 77, wherein the proteinopathy is selected from the group consisting of Alzheimer’s disease, cerebral β-amyloid angiopathy, retinal ganglion cell degeneration,, bovine spongiform encephalopathy, kuru, Crcutzfcldt-Jakob disease, variant Crcutzfcldt-Jakob disease, Gcrstmann-Straussicr-Schcinkcr syndrome, fatal familial insomnia, frontotemporal dementia, Alzheimer’s disease, progressive supranuclear palsy, corticobasal degeration. frontotemporal lobar degeneration, frontemporai lobar degeneration, amyotrophic lateral sclerosis, Huntington's disease, familial British dementia, Familial Danish dementia, hereditary cerebral hemorrhage with amyloidosis (Iclandic), CADASIL, Alexander disease, familial amyloidotic ncuropothy, senile systemic amyloidosis, scrpinopathics, AL amyloidosis, AA amyloidosis, type 11 diabetes, aortic media! amyloidosis, ApoAl amyloidosis, Apoll amyloidosis, ApoAlV amyloidosis, familial amyloidosis of the Finish type, lysozyme amyloidosis, fibrinogen amyloidosis, dialysis amyloidosis, inclusion body myositis/myopathy, cataracts, medullary thyroid carcinoma, cardiac atrial amyloidosis, pituitary prolactinoma, hereditary lattice corneal dystrophy, cutaneous lichen amyloidosis, corneal lactofeirin amyloidosis, corneal lactoferrin amyloidosis, pulmonary alveolar proteinosis, odontogenic tumor amylois, seminal vesical amyloid, cystric fibrosis, sickle cell disease and critical illness myopathy,
  79. 79. The method of claim 77, wherein the protcinopathy is Alzheimer’s disease, frontotemporal lobar degeneration, amyotrophic lateral sclerosis or Machado-Joseph disease.
  80. 80. A method of treating or preventing a disease, for which enhanced protein breakdown may be therapeutic, in a subject comprising administering to the subject a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chcmically-protcctcd form, enantiomer or stereoisomer thereof, of any one of claims 1-73, or 1UI, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, or a pharmaceutical composition of claim 74.
  81. 81. The method of claim 80, wherein the disease is selected from the group consisting of von Hippet-Lindau disease, spinocerebellar ataxia 1, Angelman syndrome, giant axon neuropathy, inclusion body myopathy with Paget disease of bone and frontotemporal dementia (IBMPFD).
  82. 82. A method of enhancing proteasomc function in a subject comprising administering to the subject a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodnig, chemically-protected form, enantiomer or stereoisomer thereof, of any one of claims 1-73, or 1U1, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, or a pharmaceutical composition of claim 74.
  83. 83. A method of increasing degradation of Tau, TDP-43 or ataxin-3 in a subject comprising administering to the subject a compound, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, of any one of claims 1-73, or IU1, or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, chemically-protected form, enantiomer or stereoisomer thereof, or a pharmaceutical composition of claim 74,
  84. 84. The method of any one of claims 77-83, wherein said subject is human.
  85. 85. An isolated proteasome comprising enzymatically inactive Uch37 and further comprising enzymatically active Uspl4.
  86. 86. The proteasome of claim 85, wherein said proteasome comprises vinylsulfone-Uch37 adducts.
  87. 87. The proteasome of claim 85, wherein said Uspl4 is a recombinant protein.
  88. 88. The proteasome of claim 85, wherein said proteasome is a human proteasome or a murine proteasome.
  89. 89. An isolated proteasome comprising enzymatically active Uspl4 and lacking enzymatically active Uch37.
  90. 90. The proteasome of claim 89, wherein said Usp34 is a recombinant protein.
  91. 91. The proteasome of claim 89, wherein said proteasome is a human proteasome or a murine proteasome.
  92. 92. A method of generating a proteasome comprising enzymatically inactive Uch37 and further comprising enzymatically active Usp 14 comprising: (a) purifying a proteasome lacking Usp 14 but comprising Uch37; (b) treating said purified proteasome with a deubiquitinasc inhibitor; and (c) reconstituting said purified proteasome with enzymatically active Usp 14.
  93. 93. The method of claim 92, wherein said proteasome is a human proteasome or a murine proteasome.
  94. 94. The method of claim 92, wherein said proteasome lacking Usp 14 but comprising Uch37 is purified from HEK293 colls. Any Docket No.: HMV-194.25
  95. 95. The method of claim 92, wherein said dcubiquitinasc inhibitor is ubiquitin-vinylsulfonc.
  96. 96. The method of claim 92, wherein said active Uspi4 is recombinantly produced.
  97. 97. A method of screening for an inhibitor of Uspl4 comprising: (a) providing a proteasome comprising enzymatically inactive Uch37 and further comprising enzymatically active Usp 14; (b) contacting said proteasome with a test compound and a UspI4 substrate; and (c) determining whether said test compound inhibits the dcubiquitination of said substrate.
  98. 98. The method of claim 97, wherein said substrate is coupled to a reporter that is detectable after cleavage by a dcubiquitinasc.
  99. 99. The method of claim 98, wherein said substrate is Ub-AMC.
  100. 100. The method of claim 97. wherein said substrate is an ubiquitin-dependant proteasome substrate.
  101. 101. The method of claim 100, wherein deubuquitination of said substrate is demonstrated by inhibition of substrate degradation.
  102. 102. The method of claim 101, wherein said substrate is polyubiquitinated cyclin B,
  103. 103. The method of claim 97, wherein said proteasome comprises viny!sulfone-Uch37 adducts.
  104. 104. The method of claim 97, wherein said Uspl4 is a recombinant protein.
  105. 105. The method of claim 97, wherein said proteasome is a human proteasome or a murine proteasome.
  106. 106. A kit comprising an isolated proteasome of any one of claim 85-9!, and instructions of use.
  107. 107. The kit of claim 106 further comprising a Usp 14 substrate
  108. 108. The kit of claim 107, wherein said Usp 14 substrate is Ub-AMC or polyubiquitinated cyclin B.
AU2017204046A 2010-01-28 2017-06-15 Compositions and methods for enhancing proteasome activity Abandoned AU2017204046A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2017204046A AU2017204046A1 (en) 2010-01-28 2017-06-15 Compositions and methods for enhancing proteasome activity

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US33695910P 2010-01-28 2010-01-28
US61/336,959 2010-01-28
US37340410P 2010-08-13 2010-08-13
US61/373,404 2010-08-13
AU2011210765A AU2011210765A1 (en) 2010-01-28 2011-01-28 Compositions and methods for enhancing proteasome activity
PCT/US2011/022929 WO2011094545A2 (en) 2010-01-28 2011-01-28 Compositions and methods for enhancing proteasome activity
AU2015249184A AU2015249184A1 (en) 2010-01-28 2015-10-30 Compositions and methods for enhancing proteasome activity
AU2017204046A AU2017204046A1 (en) 2010-01-28 2017-06-15 Compositions and methods for enhancing proteasome activity

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2015249184A Division AU2015249184A1 (en) 2010-01-28 2015-10-30 Compositions and methods for enhancing proteasome activity

Publications (1)

Publication Number Publication Date
AU2017204046A1 true AU2017204046A1 (en) 2017-07-06

Family

ID=54605507

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2015249184A Abandoned AU2015249184A1 (en) 2010-01-28 2015-10-30 Compositions and methods for enhancing proteasome activity
AU2017204046A Abandoned AU2017204046A1 (en) 2010-01-28 2017-06-15 Compositions and methods for enhancing proteasome activity

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU2015249184A Abandoned AU2015249184A1 (en) 2010-01-28 2015-10-30 Compositions and methods for enhancing proteasome activity

Country Status (1)

Country Link
AU (2) AU2015249184A1 (en)

Also Published As

Publication number Publication date
AU2015249184A1 (en) 2015-11-19

Similar Documents

Publication Publication Date Title
EP2528911B1 (en) Compositions and methods for enhancing proteasome activity
US9981981B2 (en) Tricyclic proteasome activity enhancing compounds
US11104675B2 (en) PTPN11 inhibitors
CA3099151A1 (en) Substituted heterocyclic inhibitors of ptpn11
CN112312904A (en) Spiro compounds
WO2020156242A1 (en) Shp2 inhibitor and application thereof
AU2008226466A1 (en) Aminopyrimidines useful as inhibitors of protein kinases
EA028175B1 (en) Pyrazolo-pyrrolidin-4-one derivatives as bet inhibitors and their use in the treatment of disease
WO2017147312A1 (en) Combination therapies for treatment of spinal muscular atrophy
CA3018992A1 (en) Fused hexacyclic imidazole derivatives as modulators of tnf activity
CA3019245A1 (en) Fused pentacyclic imidazole derivatives as modulators of tnf activity
CN104337812B (en) Substituted heteroaryl compound and its application method and purposes
CA3128917A1 (en) Acryl-containing nuclear transport modulators and uses thereof
AU2017204046A1 (en) Compositions and methods for enhancing proteasome activity
AU2018359413A1 (en) Kinase inhibitors for the treatment of central and peripheral nervous system disorders
CA3181351A1 (en) Nampt modulators
US20190382396A1 (en) Salts of bicyclo[1.1.1]pentane inhibitors of dual leucine zipper (dlk) kinase for the treatment of disease
CA3193745A1 (en) Imidazopiperazine inhibitors of transcription activating proteins
WO2024011294A1 (en) Anti-giardial compounds
CA2957566A1 (en) Indoline sulfonamide inhibitors of dape and ndm-1 and use of the same

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application