WO2014205393A1 - Nuclear transport modulators and uses thereof - Google Patents

Nuclear transport modulators and uses thereof Download PDF

Info

Publication number
WO2014205393A1
WO2014205393A1 PCT/US2014/043484 US2014043484W WO2014205393A1 WO 2014205393 A1 WO2014205393 A1 WO 2014205393A1 US 2014043484 W US2014043484 W US 2014043484W WO 2014205393 A1 WO2014205393 A1 WO 2014205393A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
heteroaryl
heterocyclyl
compound
substituted
Prior art date
Application number
PCT/US2014/043484
Other languages
French (fr)
Inventor
Erkan Baloglu
Sharon Shacham
Dilara Mccauley
Trinayan KASHYAP
William SENAPEDIS
Yosef Landesman
Gali Golan
Ori Kalid
Sharon Shechter
Original Assignee
Karyopharm Therapeutics Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Karyopharm Therapeutics Inc. filed Critical Karyopharm Therapeutics Inc.
Publication of WO2014205393A1 publication Critical patent/WO2014205393A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • Restoration of appropriate nuclear localization of functional p53 protein can normalize some properties of neoplastic cells (Cai et al, 2008; Hoshino et al 2008; Lain et al 1999a; Lain et al 1999b; Smart et al 1999), can restore sensitivity of cancer cells to DNA damaging agents (Cai et al, 2008), and can lead to regression of established tumors (Sharpless & DePinho 2007, Xue et al, 2007). Similar data have been obtained for other tumor suppressor proteins such as forkhead (Turner and Sullivan 2008) and c-Abl (Vignari and Wang 2001).
  • CRM1 inhibition may provide particularly interesting utility in familial cancer syndromes (e.g., Li-Fraumeni Syndrome due to loss of one p53 allele, BRCA1 or 2 cancer syndromes), where specific tumor suppressor proteins (TSP) are deleted or dysfunctional and where increasing TSP levels by systemic (or local) administration of CRM1 inhibitors could help restore normal tumor suppressor function.
  • familial cancer syndromes e.g., Li-Fraumeni Syndrome due to loss of one p53 allele, BRCA1 or 2 cancer syndromes
  • TSP tumor suppressor proteins
  • RNAs are carried into and out of the nucleus by specialized transport molecules, which are classified as importins if they transport molecules into the nucleus, and exportins if they transport molecules out of the nucleus (Terry et al, 2007; Sorokin et al 2007). Proteins that are transported into or out of the nucleus contain nuclear import/localization (NLS) or export (NES) sequences that allow them to interact with the relevant transporters. Chromosomal Region Maintenance 1 (Crml), which is also called exportin-1 or Xpol , is a major exportin.
  • Nrml Chromosomal Region Maintenance 1
  • Crml blocks the exodus of tumor suppressor proteins and/or growth regulators such as p53, c-Abl, p21, p27, pRB, BRCA1 , IkB, ICp27, E2F4, KLF5, YAP1, ZAP, KLF5, HDAC4, FID AC 5 or forkhead proteins (e.g. FOX03a) from the nucleus that are associated with gene expression, cell proliferation, angiogenesis and epigenetics.
  • Crml inhibitors have been shown to induce apoptosis in cancer cells even in the presence of activating oncogenic or growth stimulating signals, while sparing normal (untransformed) cells.
  • LMB Crml inhibitor Leptomycin B
  • Crml In addition to tumor suppressor proteins, Crml also exports several key proteins that are involved in many inflammatory processes. These include IkB, NF-kB, Cox-2, RXRa, Commdl, HIFl , HMGBl, FOXO, FOXP and others.
  • IkB protein inhibitor of NF-kB
  • a protein inhibitor of NF-kB binds to NF-kB in the nucleus and the complex IkB-NF-kB renders the NF-kB transcriptional function inactive.
  • IkB dissociates from the IkB-NF-kB complex, which releases NF-kB and unmasks its potent transcriptional activity.
  • Many signals that activate NF-kB do so by targeting IkB for proteolysis (Phosphorylation of IkB renders it "marked” for ubiquitination and then proteolysis).
  • the nuclear IkBa-NF-kB complex can be exported to the cytoplasm by Crml where it dissociates and NF-kB can be reactivated. Ubiquitinated IkB may also dissociate from the NF-kB complex, restoring NF-kB transcriptional activity. Inhibition of Crml induced export in human neutrophils and macrophage like cells (U937) by LMB not only results in accumulation of transcriptionally inactive, nuclear IkBa-NF-kB complex but also prevents the initial activation of NF-kB even upon cell stimulation (Ghosh 2008, Huang 2000).
  • COMMDl is another nuclear inhibitor of both NF-kB and hypoxia- inducible factor 1 (HIFl) transcriptional activity. Blocking the nuclear export of COMMDl by inhibiting Crml results in increased inhibition of NF-kB and HIFl transcriptional activity (Muller 2009).
  • RXRa Retinoid X receptor a
  • RXRa Retinoid X receptor a
  • Lep B is able to prevent IL- ⁇ induced cytoplasmic increase in RXRa levels in human liver derived cells (Zimmerman 2006).
  • CRM1 Inhibition affects gene expression by inhibiting/activating a series of transcription factors like ICp27, E2F4, KLF5, YAP1, ZAP
  • Crml inhibition has potential therapeutic effects across many dermatologic syndromes including inflammatory dermatoses (atopy, allergic dermatitis, chemical dermatitis, psoriasis), sun-damage (Ultraviolet / UV damage), and infections.
  • CRMl inhibition best studied with LMB, showed minimal effects on normal keratinocytes, and exerted anti-inflammatory activity on keratinocytes subjected to UV, TNFa, or other inflammatory stimuli (Kobayashi & Shinkai 2005, Kannan & Jaiswal 2006).
  • NRF2 nuclear factor erythroid-related factor 2
  • LMB induces apoptosis in keratinocytes infected with oncogenic human papillomavirus (HPV) strains such as HPV16, but not in uninfected keratinocytes (Jolly et al, 2009).
  • HPV human papillomavirus
  • Crml also mediates the transport of key neuroprotectant proteins that may be useful in neurodegenerative diseases including Parkinson's Disease (PD), Alzheimer's Disease, and Amyotrophic Lateral Sclerosis. For example, (1) forcing nuclear retention of key neuroprotective regulators such as NRF2 (Wang 2009), FOXA2 (Kittappa et al, 2007), parking in neuronal cells and/or by (2) inhibiting NFKB transcriptional activity by
  • HIV human immunodeficiency virus
  • adenovirus adenovirus
  • simian retrovirus type 1 Borna disease virus
  • influenza usual strains as well as H1N1 and avian H5N1 strains
  • HBV hepatitis B
  • HCV human papillomavirus
  • RSV respiratory syncytial virus
  • MCV Merkel cell polyomavirus
  • the HIV-1 Rev protein which traffics through nucleolus and shuttles between the nucleus and cytoplasm, facilitates export of unspliced and singly spliced HIV transcripts containing Rev Response Elements (RRE) RNA by the CRMl export pathway.
  • RRE Rev Response Elements
  • Inhibition of Rev-mediated RNA transport using CRMl inhibitors such as LepB or PKF050-638 can arrest the HIV-1 transcriptional process, inhibit the production of new HIV-1 virions, and thereby reduce HIV-1 levels (Pollard 1998, Daelemans 2002).
  • Dengue virus is the causative agent of the common arthropod-borne viral disease, dengue fever (DF), and its more severe and potentially deadly dengue hemorrhagic fever (DHF). DHF appears to be the result of an over exuberant inflammatory response to DENV.
  • NS5 is the largest and most conserved protein of DENV. CRMl regulates the transport of NS5 from the nucleus to the cytoplasm, where most of the NS5 functions are mediated.
  • HSV type 1 tegument protein VP 13/14, or hUL47
  • human CMV protein pp65 the SARS Coronavirus ORF 3b Protein
  • M RSV matrix
  • HCC hepatocellular carcinoma
  • cervical cancer due to HPV
  • CRMl inhibitors could therefore have beneficial effects on both the viral infectious process as well as on the process of neoplastic transformation due to these viruses.
  • CRMl controls the nuclear localization and therefore activity of multiple DNA metabolizing enzymes including histone deacetylases (HDAC), histone acetyltransferases (HAT), and histone methyltransferases (HMT). Suppression of cardiomyocyte hypertrophy with irreversible CRMl inhibitors has been demonstrated and is believed to be linked to nuclear retention (and activation) of HDAC 5, an enzyme known to suppress a hypertrophic genetic program (Monovich et al, 2009). Thus, CRMl inhibition may have beneficial effects in hypertrophic syndromes, including certain forms of congestive heart failure and hypertrophic cardiomyopathies.
  • HDAC histone deacetylases
  • HAT histone acetyltransferases
  • HMT histone methyltransferases
  • CRMl has also been linked to other disorders.
  • Leber's disorder a hereditary disorder characterized by degeneration of retinal ganglion cells and visual loss, is associated with inaction of the CRMl switch (Gupta N 2008).
  • Leber's disorder a hereditary disorder characterized by degeneration of retinal ganglion cells and visual loss.
  • the present invention relates to compounds, and pharmaceutically acceptable salts thereof, useful as nuclear transport modulators; pharmaceutically acceptable compositions comprising compounds of the present invention or their pharmaceutically acceptable salts; and methods of using said compounds, salts and compositions in the treatment of various disorders.
  • the compounds of the invention have general formula I:
  • the compounds of the invention have general formula II:
  • the compounds of the invention have general formula III:
  • Compounds of the present invention are useful for treating a variety of diseases, disorders or conditions associated with abnormal cellular responses triggered by improper nuclear transport.
  • one embodiment of the invention is use of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treating a variety of diseases, disorders or conditions associated with abnormal cellular responses triggered by improper nuclear transport.
  • Another embodiment of the invention is a method for treating a variety of diseases, disorders or conditions associated with CRM1 activity in a subject in need thereof, the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt or composition thereof.
  • diseases, disorders, or conditions include those described herein.
  • Compounds provided by this invention are also useful for the study of nuclear transport modulation in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated, for example, by kinases; and the comparative evaluation of new nuclear transport modulators.
  • each of the sections directed to compounds of Structural Formulas I, II and III begins with a first embodiment.
  • a statement in a particular section e.g. , "Compounds of Structural Formula ⁇ "
  • a statement that the values for the remaining variables are as described in the first embodiment, or any aspect thereof refers only to the embodiment of the identified number (e.g, the first embodiment) in the same section (e.g., "Compounds of Structural Formula II").
  • a first embodiment of a compound of Structural Formula I is a compound of Structural Formula I:
  • X is -N- or -C(H)-
  • R a and R b are each independently halo or hydrogen, wherein at least one of R a and R b is halo;
  • each R 1 is independently selected from halo; haloalkyl; -(CH 2 )i- 4 R°; -(CH 2 )o -4 OR°; -0-(CH 2 )o -4 C(0)OR°; -(CH 2 ) 0-4 CH(OR°) 2 ; -(CH 2 )o- 4 SR°; -(CH 2 ) 0 - 4 -carbocyclyl, which may be substituted with R°; -(CH 2 )o -4 -aryl, which may be substituted with R°;
  • each R° is independently hydrogen, C 1-6 aliphatic, -CH 2 -carbocyclyl,
  • each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Cj-Q alkyl, -NH 2 , -N0 2 , -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted C1-C3 alkyl) 2 , -O-C1-C3 alkyl, -C(0)OH,
  • R 2 is selected from -C(0)-0-R 3 , -C(S)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(S)-N(R 5 )(R 6 ),
  • R 3 is selected from C 1 -C4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl;
  • R 4 is selected from -N(H)(C 3 -C 6 cycloalkyl), -N(C r C 4 alkyl)(C 3 -C 6 cycloalkyl),
  • R 5 and R 6 are each independently selected from hydrogen, C1-C4 alkyl, C 2 -C 4 alkenyl,
  • R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
  • each R 7 is independently hydrogen or C 1 -C alkyl; and n is 0, 1, 2, 3, 4 or 5; wherein unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
  • R a and R b are each independently halo.
  • the values for the remaining variables are as described in the first embodiment.
  • R a and R b are the same.
  • the values for the remaining variables are as described in the first embodiment, or first aspect thereof.
  • the halo of R a and R b is chlorine or bromine.
  • the values for the remaining variables are as described in the first embodiment, or first or second aspect thereof.
  • R a and R b are the same and are each chlorine or bromine.
  • the values for the remaining variables are as described in the first embodiment, or first through third aspects thereof.
  • X is -C(H)-.
  • the values for the remaining variables are as described in the first embodiment, or first through fourth aspects thereof.
  • n is 0, 1 or 2.
  • the values for the remaining variables are as described in the first embodiment, or first through fifth aspects thereof.
  • each R 1 is independently selected from -CF 3 , -CN, halo, - OH, C1-C3 alkyl, C 3 -C 6 cycloalkyl, C 3 -C 12 heterocycloalkyl, halo-Q- C 3 alkyl, -NH 2 , -N0 2 , -NH(Ci-C 3 alkyl), -N(d-C 3 alkyl)(Ci-C 3 alkyl), -C(0)OH,
  • each R 1 is independently selected from halo, -Ci-C 4 alkyl, -d-d haloalkyl and -0-Ci-C 4 alkyl, or is absent.
  • the values for the remaining variables are as described in the first embodiment, or first through seventh aspects thereof.
  • R is selected from -C(0)-0-R , -C(0)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-S(0)i -2 -R 4 , and heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through eighth aspects thereof.
  • R 2 is -C(0)-0-R 3 , and R 3 is selected from optionally substituted C1-C4 alkyl and C2-C4 alkenyl; or R 2 is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or R 2 is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted heteroaryl; or R 2 is -C(0)-NH-NH-C(0)-R 4 or
  • R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C 6 alkyl, -(C 0 -C 4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl; or R 2 is optionally substituted C 5 -C 6 heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through ninth aspects thereof.
  • R 2 is -C(0)-0-CH(CH 3 ) 2 ,
  • each R 7 is hydrogen.
  • the values for the remaining variables are as described in the first embodiment, or first through eleventh aspects thereof.
  • R 5 is selected from hydrogen and C1-C4 alkyl; and R 6 is selected from C1-C4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through twelfth aspects thereof.
  • R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through thirteenth aspects thereof.
  • R J is selected from optionally substituted C1-C4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through fourteenth aspects thereof.
  • R 4 is selected from -N(R 8 )(C 3 -C 6 cycloalkyl), -C 3 -C 6 alkyl, -(Co-Q alkylene)-heterocyclyl, and -(C 0 -Ci alkylene)-heteroaryl, wherein R 8 is hydrogen or -C 1 -C 4 alkyl; any alkyl or alkylene portion of R 4 is optionally and independently substituted with one or more substituents selected from the group consisting of oxo and -N(R 9 ) 2 , wherein each R 9 is independently selected from hydrogen and Q-C 4 alkyl; any heterocyclyl portion of R 4 comprises at least one nitrogen atom in a ring, and is optionally substituted with one or more substituents selected from the group consisting of Q- C 4 alkyl and oxo; and any heteroaryl portion of R 4 comprises at least one nitrogen atom in a
  • R is selected from -C(0)-0-R , -C(0)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , and heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through sixteenth aspects thereof.
  • R 2 is -C(0)-0-R 3 , and R 3 is selected from optionally substituted C r C 4 alkyl; or R 2 is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C 3 -C 7 heterocyclyl; or R 2 is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted C 5 -C 6 heteroaryl; or R 2 is -C(0)-NH-NH-C(0)-R 4 or
  • R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C 6 alkyl, -(C 0 -C 4 alkylene)-(C 3 - C 7 )heterocyclyl and -(C 0 -C 4 alkylene)-(C5-C 6 )heteroaryl; or R 2 is optionally substituted C 5 -C 6 heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through seventeenth aspects thereof.
  • a second embodiment of a compound of Structural Formula I is a compound represented by Structural Formula la:
  • R la and R lb are each independently selected from halo; haloalkyl; -(CH 2 )i. 4 R°; -(CH 2 ) 0-4 OR°; -O-(CH 2 ) 0-4 C(O)OR°; -(CH 2 ) 0 - 4 CH(OR°) 2 ; -(CH 2 ) 0 . 4 SR°;
  • each R° is independently hydrogen, Ci -6 aliphatic, -CH 2 -carbocyclyl,
  • each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Ci-C 3 alkyl, -NH 2 , -N0 2 , -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted C1 -C3 alkyl) 2 , -0-C,-C 3 alkyl, -C(0)OH, -C(0)0-(unsubstituted C1-C3 alkyl), -C(0)-(unsubstituted C 1 -C3 alkyl), -0-(unsubstituted Q-Q3 alkyl), and -S-(unsubstituted Ci-C 3 alkyl); and
  • m 0 or 1.
  • R l is -C1-C4 haloalkyl.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment.
  • R is -C 1 -C 4 haloalkyl.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first aspect thereof.
  • R l is -CF 3 and R lb is -CF 3 .
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first or second aspect thereof.
  • R is -C 1 -C 4 haloalkyl or -0-C 1 -C 4 alkyl or absent.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through third aspects thereof.
  • R la is halo or -Cj-C 4 haloalkyl.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fourth aspects thereof.
  • m is 1.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fifth aspects thereof.
  • a third embodiment of a compound of Structural Formula I is a compound represented by Structural Formu
  • R 2 is selected from -C(0)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 ,
  • R a and R b are each independently halo.
  • the values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third embodiment, or first aspect thereof.
  • R a and R b are the same.
  • the values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third embodiment, or first or second aspect thereof.
  • the halo of R a and R b is chlorine or bromine.
  • the values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third embodiment, or first through third aspects thereof.
  • R 2 is -C(0)-0-R 3 , and R 3 is selected from optionally substituted Ci-C 4 alkyl and C 2 -C 4 alkenyl; or R 2 is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or R 2 is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted heteroaryl; or R 2 is -C(0)-NH-NH-C(0)-R 4 or
  • R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C 6 alkyl, -(C 0 -C 4 alkylene)-heterocyclyl and -(C 0 -C 4 alkylene)-heteroaryl; or R 2 is optionally substituted C5-C6 heteroaryl.
  • the values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third embodiment, or first through fourth aspects thereof.
  • R 2 is -C(0)-0-CH(CH 3 ) 2 ,
  • a fourth embodiment of a compound of Structural Formula I is a compound of Structural Formula I, or a pharmaceutically acceptable salt thereof, wherein:
  • X is -N- or -C(H)-
  • R a and R b are each independently halo or hydrogen, wherein at least one of R a and R b is halo;
  • each R 1 is independently selected from halo; haloalkyl; -(CH 2 )i- 4 R 0 ; -(CH 2 )o -4 OR°;
  • -CH CH-heteroaryl, which may be substituted with R°; -N0 2 ; -CN; -N 3 ; -(CH 2 ) 0-4 N(R°) 2 ;
  • each R° is independently hydrogen, C 1-6 aliphatic, -CH 2 -carbocyclyl,
  • each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C 1 -C 3 alkyl, halo-Ci-C 3 alkyl, -NH 2 , -N0 2 , -NH(unsubstituted C 1 -C 3 alkyl), -N(unsubstituted d-C 3 alkyl) 2 , -O-C 1 -C 3 alkyl, -C(0)OH,
  • R 2 is selected from -C(0)-0-R 3 , -C(S)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(S)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(S)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 ,
  • R 3 is selected from hydrogen, C1-C4 alkyl, C 2 -C4 alkenyl, C 2 -C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl;
  • R 4 is selected from -N(H)(C 3 -C 6 cycloalkyl), -N(C r C 4 alkyl)(C 3 -C 6 cycloalkyl), -C r C 6 alkyl, -(C 0 -C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl, -(C0-C4
  • R 5 and R 6 are each independently selected from hydrogen, C1-C4 alkyl, Q-C4 alkenyl, C 2 -C 4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or
  • R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
  • each R 7 is independently hydrogen or C 1 -C4 alkyl
  • n 0, 1, 2, 3, 4 or 5; wherein unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
  • Alternative values for the variables in Structural Formula I are as described in the first through third embodiments, or any aspect thereof.
  • R 2 is -C(0)-0-R 3 , and R 3 is hydrogen or is selected from optionally substituted C1-C4 alkyl and C 2 -C4 alkenyl; or R 2 is
  • R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or R is
  • R 6 is an optionally substituted heteroaryl
  • R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl), -d-C 6 alkyl, -(C 0 -C 4 alkylene)-heterocyclyl and -(C 0 -C4 alkylene)-heteroaryl; or R 2 is optionally substituted C 5 -C 6 heteroaryl.
  • the values for the remaining variables are as described in the first through third embodiments, or any aspect thereof, or the fourth embodiment.
  • R 3 is hydrogen or is selected from optionally substituted C 1 -C 4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl.
  • the values for the remaining variables are as described in the first through third embodiments, or any aspect thereof, or the fourth embodiment, or first aspect thereof.
  • R 2 is -C(0)-0-R 3 , and R 3 is hydrogen or is selected from optionally substituted C C 4 alkyl; or R 2 is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C 3 -C 7 heterocyclyl; or R 2 is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted C 5 -C 6 heteroaryl; or R 2 is -C(0)-NH-NH-C(0)-R 4 or
  • R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl), -C 1 -C 6 alkyl, -(C 0 -C 4 alkylene)-(C 3 - C )heterocyclyl and -(C 0 -C 4 alkylene)-(C5-C 6 )heteroaryl; or R 2 is optionally substituted C5-C6 heteroaryl.
  • the values for the remaining variables are as described in the first through third embodiments, or any aspect thereof, or the fourth embodiment, or first or second aspect thereof.
  • a fifth embodiment of a compound of Structural Formula I is a compound of Structural Formula lb, or a pharmaceutically acceptable salt thereof, wherein the values for R a , R b and R 2 are as described in the fourth embodiment.
  • Alternative values for the variables in Structural Formula lb are as described in the first through fourth embodiments, or any aspect thereof.
  • R 2 is -C(0)-0-R 3 , and R 3 is hydrogen or is selected from optionally substituted Ci-C 4 alkyl and C 2 -C 4 alkenyl; or R 2 is
  • R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or R 2 is
  • R 6 is an optionally substituted heteroaryl
  • R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(C C 4 alkyl)(C 3 -C 6 cycloalkyl), -C r C 6 alkyl, -(C 0 -C 4 alkylene)-heterocyclyl and -(C 0 -C 4 alkylene)-heteroaryl; or R 2 is optionally substituted C5-C6 heteroaryl.
  • the values for the remaining variables are as described in the first through fourth embodiments, or any aspect thereof, or the fifth embodiment.
  • a first embodiment of a compound of Structural Formula II is a compound of Structural Formula II:
  • X is -N- or -C(H)-
  • each R 1 is independently selected from halo; haloalkyl; -(CH 2 )i-4R°; -(CH 2 )o- 4 0R°;
  • -(CH 2 )o -4 -heteroaryl which may be substituted with R°
  • -CH CH-carbocyclyl, which may be substituted with R°
  • -CH CH-aryl, which may be substituted with R°
  • -CH CH-heterocyclyl, which may be substituted with R°;
  • -CH CH-heteroaryl, which may be substituted with R°; -N0 2 ; -CN; -N 3 ;
  • each R° is independently hydrogen, Ci -6 aliphatic, -CH 2 -carbocyclyl, -CH 2 -aryl, -CH 2 -heterocyclyl, -CH 2 -heteroaryl, -0(CH 2 )o-i-carbocyclyl, -0(CH 2 )o-i-aryl, -0(CH 2 )o-i-heterocyclyl, -O(CH 2 ) 0 -i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
  • each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C 1 -C3 alkyl, halo-Ci-C 3 alkyl, -NH 2 , -N0 2 , -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted d-C 3 alkyl) 2 , -0-Ci-C 3 alkyl, -C(0)OH, -C(0)0-(unsubstituted C r C 3 alkyl),
  • R 2 is halo, cyano, -CF3 or -CHF 2 ;
  • R a is selected from -CN, -C(0)-0-R 3 , -C(S)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(S)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(S)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(S)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(S)-R 4 ,
  • R 3 is selected from hydrogen, Cj-C 4 alkyl, C -C 4 alkenyl, C 2 -C 4 alkynyl,
  • R 4 is selected from -N(H)(C 3 -C 6 cycloalkyl), -N(C r C 4 alkyl)(C 3 -C 6 cycloalkyl),
  • R 5 and R 6 are each independently selected from hydrogen, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; each R 7 is independently hydrogen or C1-C4 alkyl; and
  • n 0, 1, 2, 3, 4 or 5;
  • each alkyl, alkenyl, alkynyl, alkyl ene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted;
  • the compound is not (E)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4- triazol-l-yl)-3-bromo-l-(3,3-difluoroazetidin-l-yl)prop-2-en-l-one.
  • R 2 is bromo or chloro. Values for the remaining variables are as defined in the first embodiment.
  • R is fluoro, bromo or chloro. Values for the remaining variables are as defined in the first embodiment, or first aspect thereof.
  • R a is selected from -C(0)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , and
  • R a is selected from -C(0)-0-R 3 , -C(S)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(S)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ) and
  • R a is selected from -C(0)-0-R 3 , -C(0)-N(R 5 )(R 6 ), and -C(0)-N(R 7 )-N(R 5 )(R 6 ). Values for the remaining variables are as defined in the first embodiment, or first through fourth aspects thereof.
  • X is -C(H)-. Values for the remaining variables are as defined in the first embodiment, or first through fifth aspects thereof.
  • n is 0, 1 or 2. Values for the remaining variables are as defined in the first embodiment, or first through sixth aspects thereof.
  • each R 1 is independently selected from -CF 3 , -CN, halo, - OH, C C 3 alkyl, C 3 -C 6 cycloalkyl, C 3 -Ci 2 heterocycloalkyl, halo-C r C 3 alkyl, -NH 2 , -N0 2 , -NH(Ci-C 3 alkyl), -N(C C 3 alkyl)(d-C 3 alkyl), -C(0)OH,
  • each R 1 is independently selected from halo, -C1-C4 alkyl, -C1-C4 haloalkyl and -0-CrC4 alkyl, or is absent. Values for the remaining variables are as defined in the first embodiment, or first through eighth aspects thereof.
  • R a is -C(0)-0-R 3 , and R 3 is selected from optionally substituted C1-C4 alkyl and C 2 - C4 alkenyl; or
  • R a is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
  • R a is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted heteroaryl; or
  • R a is -C(0)-NH-NH-C(0)-R 4 or -C(0)-NH-NH-S(0)i -2 -R 4
  • R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(d-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C 6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C 0 -C4 alkylene)-heteroaryl.
  • R a is -C(0)-0-R 3 , and R 3 is selected from optionally substituted d-C alkyl; or R a is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C 3 - C 7 heterocyclyl; or
  • R a is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted C 5 -C 6 heteroaryl; or R a is -C(0)-NH-NH-C(0)-R 4 or -C(0)-NH-NH-S(0) 1-2 -R 4 , and R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(C 1 -C 4 alkyl)(C 3 -C 6 cycloalkyl), -CpC 6 alkyl, -(C0-C4 alkylene)-(C 3 -C7)heterocyclyl and -(C0-C4 alkylene)-(C5-C6)heteroaryl.
  • R a is selected from 3,3- difluoroazetidin-l-ylcarbonyl, N'-(pyrazin-2-yl)-hydrazinylcarbonyl, and -C(0)OCH(CH 3 ) 2 .
  • Values for the remaining variables are as defined in the first embodiment, or first through eleventh aspects thereof.
  • each R 7 is hydrogen.
  • the values for the remaining variables are as described in the first embodiment, or first through twelfth aspects thereof.
  • R 5 is selected from hydrogen and C1-C4 alkyl; and R 6 is selected from C 1 -C 4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through thirteenth aspects thereof.
  • R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through fourteenth aspects thereof.
  • R 3 is selected from optionally substituted CpC 4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through fifteenth aspects thereof.
  • R is selected from -N(R )(C 3 -C 6 cycloalkyl), -C 3 -C 6 alkyl, -(Co- alkylene)-heterocyclyl, and -(QrQ alkylene)-heteroaryl, wherein R 8 is hydrogen or -C 1 -C 4 alkyl; any alkyl or alkylene portion of R 4 is optionally and independently substituted with one or more substituents selected from the group consisting of oxo and -N(R 9 ) 2 , wherein each R 9 is independently selected from hydrogen and CrC 4 alkyl; any heterocyclyl portion of R 4 comprises at least one nitrogen atom in a ring, and is optionally substituted with one or more substituents selected from the group consisting of Ci- C 4 alkyl and oxo; and any heteroaryl portion of R 4 comprises at least one nitrogen atom in a ring and is optionally substituted
  • R is halo.
  • the values for the remaining variables are as described in the first embodiment, or first through seventeenth aspects thereof.
  • X is -N-. Values for the remaining variables are as defined in the first embodiment, or first through eighteenth aspects thereof.
  • X is -N- or -C(H)-
  • R la and R lb are each independently selected from halo; haloalkyl; -(CH 2 )i -4 R°; -(CH 2 )o -4 OR 0 ; -O-(CH 2 ) 0-4 C(O)OR°; -(CH 2 ) 0 - 4 CH(OR o ) 2; -(CH 2 ) 0-4 SR° ;
  • each R° is independently hydrogen, C 1-6 aliphatic, -CH 2 -carbocyclyl,
  • each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Ci-C 3 alkyl, -NH 2 , -N0 2 , -NH(unsubstituted Ci-C 3 alkyl), -N(unsubstituted Ci-C 3 alkyl) 2 , -O-C1-C3 alkyl, -C(0)OH,
  • n 0 or 1 ;
  • R 2 is halo, cyano, -CF 3 or -CHF 2 ;
  • R a is selected from -CN, -C(0)-0-R 3 , -C(S)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(S)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(S)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(S)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(S)-R 4 ,
  • R 3 is selected from hydrogen, C 1-C4 alkyl, C 2 -C4 alkenyl, C 2 -C 4 alkynyl,
  • R 4 is selected from -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl),
  • R 5 and R 6 are each independently selected from hydrogen, C1-C4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; and
  • each R 7 is independently hydrogen or C1-C4 alkyl, wherein: unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
  • the compound is not (E)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4- triazol- 1 -yl)-3 -bromo- 1 -(3 ,3-difluoroazetidin- 1 -yl)prop-2-en- 1 -one.
  • R la is -Cj-C 4 haloalkyl. Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment.
  • R lb is -Ci-C 4 haloalkyl. Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment, or first aspect thereof.
  • R la is -CF 3 and R lb is -CF 3 .
  • Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment, or first or second aspect thereof.
  • R lb is -Ci-C 4 haloalkyl or -0-Ci-C 4 alkyl or absent.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through third aspects thereof.
  • R la is halo or -Ci-C 4 haloalkyl.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fourth aspects thereof.
  • m is 1.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fifth aspects thereof.
  • a third embodiment of a compound of Structural Formula II is a compound represented by Structural Formula lib:
  • R 2 is halo, cyano, -CF 3 or -CHF 2 ;
  • R a is selected from -C(0)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ),
  • R is selected from hydrogen, C C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl,
  • R 4 is selected from -N(H)(C 3 -C 6 cycloalkyl), -N(C r C 4 alkyl)(C 3 -C 6 cycloalkyl),
  • R 5 and R 6 are each independently selected from hydrogen, Cj-C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; and
  • each R 7 is independently hydrogen or Cj-C 4 alkyl, wherein:
  • each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted;
  • the compound is not (E)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l ,2,4- triazol- 1 -yl)-3 -bromo- 1 -(3 ,3-difluoroazetidin- 1 -yl)prop-2-en- 1 -one
  • R 2 is bromo or chloro. Values for the remaining variables are as defmed in the first embodiment, or any aspect thereof or the third embodiment.
  • R a is -C(0)-0-R 3 , and R 3 is selected from optionally substituted Ci-C 4 alkyl and C 2 - C 4 alkenyl; or R a is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
  • R a is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted heteroaryl; or
  • R a is -C(0)-NH-NH-C(0)-R 4 or -C(0)-NH-NH-S(0) 1 -2 -R 4
  • R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(d-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C 6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl.
  • R a is -C(0)-0-R 3 , and R 3 is selected from optionally substituted C1-C4 alkyl; or R a is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C 3 - C 7 heterocyclyl; or
  • R a is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted C 5 -C 6 heteroaryl; or R a is -C(0)-NH-NH-C(0)-R 4 or -C(0)-NH-NH-S(0)i -2 -R 4 , and R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C 6 alkyl, -(C0-C4 alkylene)-(C3-C 7 )heterocyclyl and -(C0-C4 alkylene)-(C 5 -C6)heteroaryl.
  • R a is selected from -C(0)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , and
  • R a is selected from -C(0)-0-R 3 , -C(S)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(S)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ) and
  • R a is selected from -C(0)-0-R 3 , -C(0)-N(R 5 )(R 6 ), and -C(0)-N(R 7 )-N(R 5 )(R 6 ). Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the third embodiment, or first through fifth aspects thereof
  • R 2 is halo. Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the third
  • a first embodiment of a compound of Structural Formula III is a compound of Structural Formula III:
  • X is -N- or -C(H)-
  • each R 1 is independently selected from halo; haloalkyl; -(CH 2 ) 1-4 R°;
  • -CH CH-heteroaryl, which may be substituted with R°; -N0 2 ; -CN; -N 3 ;
  • each R° is independently hydrogen, C 1-6 aliphatic, -CH 2 -carbocyclyl, -CH 2 -aryl, -CH 2 -heterocyclyl, -CH 2 -heteroaryl, -0(CH 2 )o-i-carbocyclyl, -0(CH 2 )o-i-aryl, -0(CH 2 )o-i
  • each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C r C 3 alkyl, halo-C C 3 alkyl, -NH 2 , -N0 2 ,
  • R 2 is halo, -CF 3 , -CHF 2 , or -CN;
  • R a is selected from -CN, -C(0)-0-R 3 , -C(S)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(S)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(S)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(S)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(S)-R 4 ,
  • R 3 is selected from hydrogen, Ci-C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl,
  • R is selected from -N(H)(C 3 -C 6 cycloalkyl), -N(C r C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-Ce alkyl, -(C 0 -C 4 alkylene)-carbocyclyl, -(C 0 -C 4 alkylene)- eterocyclyl, -(C 0 -C 4 alkylene)-aryl, and -(Co-C 4 alkylene)- eteroaryl;
  • R 5 and R 6 are each independently selected from hydrogen, Q-Q alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or
  • R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
  • each R is independently hydrogen or Q-C4 alkyl
  • n 0, 1 , 2, 3, 4 or 5;
  • each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted;
  • the compound is not (E)-isopropyl 2-fluoro-3-(3-(3-methoxy-5- (trifluoromethyl)phenyl)- lH- l,2,4-triazol-l-yl)acrylate.
  • R 2 is halo.
  • the values for the remaining variables are as defined in the first embodiment.
  • R 2 is bromo or chloro.
  • the values for the remaining variables are as defined in the first embodiment, or first aspect thereof.
  • R 2 is bromo, chloro, -CF 3 , -CHF 2 , or - CN.
  • the values for the remaining variables are as defined in the first embodiment, or first or second aspect thereof.
  • R a is selected from -CN, -C(0)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , and
  • R is selected from -CN, -C(0)-0-R 3 , -C(S)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(S)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), and
  • R a is selected from is selected from -CN, -C(0)-0-R 3 , -C(0)-N(R 5 )(R 6 ), and -C(0)-N(R 7 )-N(R 5 )(R 6 ).
  • the values for the remaining variables are as defined in the first embodiment, or first through fifth aspects thereof.
  • R is selected from 3,3- difluoroazetidin-l-ylcarbonyl, N'-(pyrazin-2-yl)-hydrazinylcarbonyl, -C(0)OCH(CH 3 ) 2 , -C(0)NH 2 , -COOH, and -CN.
  • the values for the remaining variables are as defined in the first embodiment, or first through sixth aspects thereof.
  • X is -N-.
  • the values for the remaining variables are as defined in the first embodiment, or first through seventh aspects thereof.
  • X is -CH-.
  • the values for the remaining variables are as defined in the first embodiment, or first through eighth aspects thereof.
  • n is 0, 1 or 2.
  • the values for the remaining variables are as defined in the first embodiment, or first through ninth aspects thereof.
  • each R 1 is independently selected from -CF 3 , -CN, halo, - OH, Ci-C 3 alkyl, C 3 -C 6 cycloalkyl, C 3 -Ci 2 heterocycloalkyl, halo-Ci- C 3 alkyl, -NH 2 , -N0 2 , -NH(Ci-C 3 alkyl), -N(Ci-C 3 alkyl)(Ci-C 3 alkyl), -C(0)OH,
  • each R 1 is independently selected from halo, -C1-C4 alkyl, -C 1 -C 4 haloalkyl and -O-C1-C4 alkyl, or is absent.
  • the values for the remaining variables are as defined in the first embodiment, or first through eleventh aspects thereof.
  • R a is -CN; or R a is -C(0)-0-R 3 , and R 3 is selected from hydrogen and optionally substituted C1-C4 alkyl or C 2 -C4 alkenyl; or R a is -C(0)NH 2 ; or R is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated
  • R a is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted heteroaryl; or R is -C(0)-NH-NH-C(0)-R 4 or -C(0)-NH-NH-S(0)i -2 -R 4 , and R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(d-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C 6 alkyl, -(C 0 - C4 alkylene) -heterocyclyl and -(C 0 -C 4 alkylene)-heteroaryl.
  • the values for the remaining variables are as defined in the first embodiment, or first through twelfth aspects thereof.
  • R a is -C(0)-0-R 3 , and R 3 is selected from hydrogen and optionally substituted Ci-C 4 alkyl; or
  • R a is -C(0)NH 2 ;
  • R a is -C(0)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C 3 - C 7 heterocyclyl; or
  • R is -C(0)-NH-NH(R 6 ), and R 6 is an optionally substituted C 5 -C 6 heteroaryl; or R a is -C(0)-NH-NH-C(0)-R 4 or -C(0)-NH-NH-S(0)i -2 -R 4 , and R 4 is selected from optionally substituted -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C 6 alkyl, -(C 0 -C 4 alkylene)-(C 3 -C 7 )heterocyclyl and -(C 0 -C 4 alkylene)-(C5-C 6 )heteroaryl.
  • R a is selected from -C(0)OH, -C(0)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , and
  • each R 7 is hydrogen.
  • the values for the remaining variables are as described in the first embodiment, or first through fifteenth aspects thereof.
  • R 5 is selected from hydrogen and Ci-C 4 alkyl
  • R 6 is selected from hydrogen, Ci-C 4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through sixteenth aspects thereof.
  • R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl.
  • the values for the remaining variables are as described in the first embodiment, or first through seventeenth aspects thereof.
  • R 3 is selected from hydrogen and optionally substituted Ci-C 4 alkyl, carbocyclyl, aryl, heterocyclyl or heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through eighteenth aspects thereof.
  • R 4 is selected from -N(R 8 )(C 3 -C 6 cycloalkyl), -C 3 -C 6 alkyl, -(C 0 -Ci alkylene)-heterocyclyl, and -(C 0 -Ci alkylene)-heteroaryl,
  • R is hydrogen or -Q-C4 alkyl; any alkyl or alkyl ene portion of R is optionally and independently substituted with one or more substituents selected from the group consisting of oxo and -N(R 9 ) 2 , wherein each R 9 is independently selected from hydrogen and Q-C4 alkyl; any heterocyclyl portion of R 4 comprises at least one nitrogen atom in a ring, and is optionally substituted with one or more substituents selected from the group consisting of Q- C 4 alkyl and oxo; and any heteroaryl portion of R 4 comprises at least one nitrogen atom in a ring and is optionally substituted with one or more Q-C4 alkyl.
  • the values for the remaining variables are as described in the first embodiment, or first through sixteenth aspects thereof.
  • a second embodiment of a compound of Structural Formula III is a compound represented by Structural Formula Ilia:
  • R la and R lb are each independently selected from halo; haloalkyl; -(CH 2 )i.4R°; -(CH 2 )o- 4 OR°; -0-(CH 2 )o-4C(0)OR° ; -(CH 2 )o-4CH(OR°) 2 ; -(CH 2 ) 0 - 4 SR°;
  • each R° is independently hydrogen, C 1-6 aliphatic, -CH 2 -carbocyclyl,
  • each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted Q-C 3 alkyl, halo-Ci-C 3 alkyl, -NH 2 , -N0 2 , -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted C r C 3 alkyl) 2 , -0-Ci-C 3 alkyl, -C(0)OH,
  • n 0 or 1 ;
  • X is -N- or -C(H)-
  • R 2 is halo, -CF 3 , -CHF 2 , or -CN;
  • R a is selected from -CN, -C(0)-0-R 3 , -C(S)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(S)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(S)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(S)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , -C(0)-N(R 7 )-N(R 7 )-C(S)-R 4 ,
  • R 3 is selected from hydrogen, C1-C4 alkyl, C 2 -C 4 alkenyl, C 2 -C alkynyl,
  • R 4 is selected from -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C 6 alkyl, -(C 0 -C 4 alkylene)-carbocyclyl, -(C 0 -C 4 alkylene)-heterocyclyl, -(C0-C 4 alkylene)-aryl, and -(C 0 -C 4 alkylene)-heteroaryl;
  • R 5 and R 6 are each independently selected from hydrogen, C 1 -C 4 alkyl, C2-C 4 alkenyl, C2-C 4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or
  • R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
  • each R 7 is independently hydrogen or C 1 -C 4 alkyl, wherein:
  • each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted;
  • the compound is not (E)-isopropyl 2-fluoro-3-(3-(3-methoxy-5- (trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)acrylate .
  • R l a is -C 1 -C 4 haloalkyl.
  • the values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment.
  • R lb is -C1-C 4 haloalkyl.
  • the values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment, or first aspect thereof.
  • R la is -CF 3 and R lb is -CF 3 .
  • the values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment, or first or second aspect thereof.
  • R lb is -C 1 -C 4 haloalkyl or -O-C 1 -C 4 alkyl or absent.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through third aspects thereof.
  • R la is halo or -Cj-C 4 haloalkyl.
  • the values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fourth aspects thereof.
  • m is 1. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fifth aspects thereof.
  • a third embodiment of a compound of Structural Formula III is a compound represented by Structural Formula Illb:
  • R 2 is halo, -CF 3 , -CHF 2 , or -CN;
  • R a is selected from -CN, -C(0)-0-R 3 , -C(0)-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 5 )(R 6 ), -C(0)-N(R 7 )-N(R 7 )-C(0)-R 4 , and -C(0)-N(R 7 )-N(R 7 )-S(0)i -2 -R 4 , wherein:
  • R 3 is selected from hydrogen, C1-C4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl,
  • R 4 is selected from -N(H)(C 3 -C 6 cycloalkyl), -N(Ci-C 4 alkyl)(C 3 -C 6 cycloalkyl), -Ci-C6 alkyl, -(C 0 -C 4 alkylene)-carbocyclyl, -(C 0 -C 4 alkylene)-heterocyclyl, -(C 0 -C 4 alkylene)-aryl, and -(C 0 -C 4 alkylene)-heteroaryl;
  • R 5 and R 6 are each independently selected from hydrogen, Ci-C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; and
  • each R 7 is independently hydrogen or Ci-C 4 alkyl, wherein:
  • carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted;
  • the compound is not (E)-isopropyl 2-fluoro-3-(3-(3-methoxy-5- (trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)acrylate.
  • Compounds of the present invention may have asymmetric centers, chiral axes, and chiral planes (e.g., as described in: E. L. Eliel and S. H. Wilen, Stereo-chemistry of Carbon Compounds, John Wiley & Sons, New York, 1994, pages 1119-1190), and occur as racemates, racemic mixtures, and as individual diastereomers or enantiomers, with all possible isomers and mixtures thereof, including optical isomers, being included in the present invention.
  • aliphatic or "aliphatic group,” as used herein, denotes a monovalent hydrocarbon radical that is straight-chain (i.e., unbranched), branched, or cyclic (including fused, bridged, and spiro-fused polycyclic).
  • An aliphatic group can be saturated or can contain one or more units of unsaturation, but is not aromatic. Unless otherwise specified, aliphatic groups contain 1-12 carbon atoms. However, in some embodiments, an aliphatic group contains 1-6 or 2-8 carbon atoms. In some embodiments, aliphatic groups contain 1-4 carbon atoms and, in yet other embodiments, aliphatic groups contain 1-3 carbon atoms.
  • Suitable aliphatic groups include, but are not limited to, linear or branched, alkyl, alkenyl, and alkynyl groups, and hybrids thereof, such as (cycloalkyl) alkyl, (cycloalkenyl)alkyl or (cy clo alkyl) alkenyl .
  • alkyl as used herein, unless otherwise indicated, means straight or branched saturated monovalent hydrocarbon radicals, typically C C 12 , preferably Q-Q.
  • Ci-C alkyl means a straight or branched saturated monovalent hydrocarbon radical having from one to six carbon atoms (e.g., 1 , 2, 3, 4, 5 or 6).
  • alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, and t-butyl.
  • alkoxy means an "alkyl-O-" group, wherein alkyl is defined above. Examples of alkoxy include methoxy and ethoxy.
  • alkenyl means a saturated straight chain or branched non-cyclic hydrocarbon having from 2 to 12 carbon atoms and having at least one carbon- carbon double bond. Alkenyl groups may be optionally substituted with one or more substituents.
  • alkenyl encompasses radicals having carbon-carbon double bonds in the "cis” and “trans” or, alternatively, the "E” and “Z” configurations. If an alkenyl group includes more than one carbon-carbon double bond, each carbon-carbon double bond is independently a cis or trans double bond, or a mixture thereof.
  • alkynyl means a saturated straight chain or branched non-cyclic hydrocarbon having from 2 to 12 carbon atoms and having at least one carbon- carbon triple bond. Alkynyl groups may be optionally substituted with one or more substituents.
  • alkylene refers to an alkyl group having from 2 to 12 carbon atoms and two points of attachment to the rest of the compound.
  • alkylene groups include methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), n-propylene (-CH2CH2CH2-), isopropylene (-CH 2 CH(CH 3 )-), and the like.
  • Alkylene groups may be optionally substituted with one or more substituents.
  • amino refers to a chemical moiety having the formula -N(R) 2 , wherein each R is independently selected from hydrogen and Q-Q alkyl.
  • aryl alone or in combination, as used herein, means a carbocyclic aromatic system containing one or more rings, which may be attached together in a pendent manner or may be fused. In particular embodiments, aryl is one, two or three rings. In one aspect, the aryl has six to twelve ring atoms.
  • aryl encompasses aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl and acenaphthyl. An aryl group can be optionally substituted as defined and described herein.
  • cycloaliphatic refers to a saturated or partially unsaturated cyclic aliphatic monocyclic or bi cyclic ring system, as described herein, having from 3 to 12 members, wherein the aliphatic ring system is optionally substituted as defined and described herein.
  • Cycloaliphatic groups include, without limitation, cycloalkyl, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl and cycloalkenyl, for example cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl and cyclooctadienyl.
  • cycloaliphatic also include aliphatic rings that are fused to one or more aromatic or nonaromatic rings, such as decahydronaphthyl, tetrahydronaphthyl, decalin, or bicyclo[2.2.2]octane.
  • cycloalkyl means saturated cyclic hydrocarbons, i.e. compounds where all ring atoms are carbons.
  • examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
  • cycloalkyl can optionally be substituted with one or more substituents selected from -OH, -SH, halogen, amino, nitro, cyano, Ci-Ci 2 alkyl, C2-C12 alkenyl or C2-C12 alkynyl group, C -C ⁇ 2 alkoxy, Ci-C ⁇ haloalkyl, and Ci-C 12 haloalkoxy.
  • halo or halogen as used herein means halogen and includes, for example, and without being limited thereto, fluoro, chloro, bromo, iodo and the like, in both radioactive and non-radioactive forms.
  • halo is selected from the group consisting of fluoro, chloro and bromo.
  • haloalkyl includes an alkyl substituted with one or more F, CI, Br, or I, wherein alkyl is defined above.
  • heteroaryl refers to an aromatic group containing one or more heteroatoms (e.g. , one or more heteroatoms independently selected from O, S and N).
  • a heteroaryl group can be monocyclic or polycyclic, e.g. a monocyclic heteroaryl ring fused to one or more carbocyclic aromatic groups or other monocyclic heteroaryl groups.
  • the heteroaryl groups of this invention can also include ring systems substituted with one or more oxo moieties.
  • heteroaryl has five to fifteen ring atoms and, preferably, 5 or 6 ring atoms.
  • heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, quinolyl, isoquinolyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, purinyl, oxadiazolyl, thiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl,
  • heteroaryl groups may be C-attached or N-attached (where such is possible).
  • a group derived from pyrrole may be pyrrol- 1-yl (N-attached) or pyrrol-3-yl (C-attached).
  • Heterocyclyl means a cyclic 3-12 membered saturated or unsaturated aliphatic ring containing 1 , 2, 3, 4 or 5 heteroatoms (e.g. , one or more heteroatoms independently selected from O, S and N). When one heteroatom is S, it can be optionally mono- or di-oxygenated (i.e. -S(O)- or -S(0) 2 -).
  • the heterocyclyl can be monocyclic or polycyclic, in which case the rings can be attached together in a pendent manner or can be fused or spiro.
  • a heterocyclyl is a three- to seven-membered ring system.
  • Exemplary heterocyclyls include, for example, and without being limited thereto, piperidinyl, piperazinyl, pyrrolidinyl, tetrahydrofuranyl and the like.
  • Hydroxyl means -OH.
  • Thioalkoxy means -S-alkyl, wherein alkyl is defined as above.
  • substituents and substitution patterns on the compounds of the invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below.
  • substituted whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent.
  • an “optionally substituted group” can have a suitable substituent at each substitutable position of the group and, when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent can be either the same or different at every position.
  • an "optionally substituted group” can be unsubstitued.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. If a substituent is itself substituted with more than one group, it is understood that these multiple groups can be on the same carbon atom or on different carbon atoms, as long as a stable structure results.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • each R° may be substituted as defined below and is independently hydrogen, C ⁇ 6 aliphatic, -CH 2 Ph, -0(CH 2 )o ⁇ iPh, -CH 2 -(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl monocyclic or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, which may be substituted as defined below.
  • Suitable monovalent substituents on R° are independently halogen, -(CH 2 ) 0-2 R e , -(haloR e ), -(CH 2 ) 0 réelle 2 OH, -(CH 2 )o_ 2 OR e , -(CH 2 ) 0 2 CII(OR e ) ; -O(haloR'), -CN, ⁇ N 3 , -(CH 2 ) 0 ⁇ 2 C(O)R e , -(CH 2 ) 0 _ 2 C(O)OH, -(CH 2 ) 0 _ 2 C(O)OR e , -(CH 2 ) 0 _ 2 SR e , -(CH 2 )o- 2 SH, -(CH 2 )o- 2 NH 2 , ⁇ (CH 2 )o_ 2 N
  • Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted” group include: -0(CR* 2 ) 2 - 3 0-, wherein each independent occurrence of R* is selected from hydrogen, Q-e aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • Suitable substituents on the aliphatic group of R* include halogen, -R", -(haloR*), -OH, -OR', -O(haloR'), -CN, -C(0)OH, -C(0)OR', -NH 2 , -NHR", -NR* 2 , and -N0 2 , wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci ⁇ aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • Suitable substituents on a substitutable nitrogen of an "optionally substituted group” include -R ⁇ , -NR ⁇ 2 , -C(0)R ⁇ , -C(0)OR ⁇ , -C(0)C(0)R ⁇ , -C(0)CH 2 C(0)R ⁇ , - S(0) 2 R ⁇ , -S(0) 2 NR ⁇ 2 , -C(S)NR ⁇ 2 , -C(NH)NR ⁇ 2 , and -N(R ⁇ )S(0) 2 R ⁇ ; wherein each R ⁇ is independently hydrogen, Ci_ 6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or, notwithstanding the definition above, two independent occurrences of R ⁇ , taken together with their intervening atom
  • Suitable substituents on the aliphatic group of R ⁇ are independently halogen, -R*, -(haloR e ), -OH, -OR*, -0(haloR e ), -CN, -C(0)OH, -C(0)OR e , -NH 2 , -NHR ⁇ -NR* 2 , or -N0 2 , wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently C ⁇ aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having C heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • Preferred substituents on heteroaryl can be selected from the group consisting of -OH, -SH, nitro, halogen, amino, cyano, Cj-Cn alkyl, C 2 -Ci 2 alkenyl, C 2 -C 12 alkynyl, Ci-C 12 alkoxy, Cj-Cn haloalkyl, Cj-Cn haloalkoxy and Ci-C thioalkoxy.
  • Preferred substituents on alkyl, alkylene and heterocyclyl include the preferred substituents on heteroaryl and oxo.
  • the substituent on an alkyl, alkylene, heterocyclyl or heteroaryl is an amino group having the formula -N(R) 2 , wherein each R is independently selected from hydrogen and Ci-C 4 alkyl.
  • the term "pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, trifluoroacetic acid (2,2,2-trifiuoroacetic acid), oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate,
  • benzenesulfonate benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (Ci_ 4 alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereoisomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
  • each of Structural Formulas II and III (Structural Formula II, Ila, lib, etc.), used herein, is meant to include compounds having a carbon-carbon double bond (e.g. , an exocyclic double bond) with a configuration that is cis (or Z), trans (or E), or a mixture of cis and trans.
  • Structural Formula II For exam le, Structural Formula II:
  • exocyclic double bond refers to the carbon-carbon double bond in a compound of Structural Formula II and a compound of Structural Formula III indicated
  • the exocyclic double bond is in a cis configuration. In other embodiments, the exocyclic double bond is in a trans configuration.
  • the application reflects whether a carbon-carbon double bond in a particular compound exists in a cis or trans configuration by indicating the configuration of the double bond in the chemical name associated with the compound.
  • cis or "cis configuration” refers to a carbon-carbon double bond, typically an exocyclic double bond, that is predominantly cis. In some embodiments, greater than about 85% of compound molecules in a mixture of the compound have a carbon- carbon double bond (e.g. , an exocyclic double bond) that is cis. In some embodiments, greater than about 90%, greater than about 95%, greater than about 98%, greater than about 99%, greater than about 99.5% or greater than about 99.8% of compound molecules in a mixture of the compound have a carbon-carbon double bond (e.g. , an exocyclic double bond) that is cis.
  • a carbon-carbon double bond e.g. , an exocyclic double bond
  • trans or “trans configuration” refers to a carbon-carbon double bond, typically an exocyclic double bond, that is predominantly trans. In some embodiments, greater than about 85% of compound molecules in a mixture of the compound have a carbon- carbon double bond (e.g. , an exocyclic double bond) that is cis. In some embodiments, greater than about 90%), greater than about 95%, greater than about 98%>, greater than about 99%), greater than about 99.5% or greater than about 99.8%) of compound molecules in a mixture of the compound have a carbon-carbon double bond (e.g. , an exocyclic double bond) that is cis.
  • a carbon-carbon double bond e.g. , an exocyclic double bond
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 14 C-enriched carbon are within the scope of this invention.
  • Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention.
  • pharmaceutically acceptable salt means either an acid addition salt or a basic addition salt which is compatible with the treatment of patients.
  • exemplary inorganic acids which form suitable salts include, but are not limited thereto, hydrochloric, hydrobromic, sulfuric and phosphoric acid and acid metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate.
  • Illustrative organic acids which form suitable salts include the mono-, di- and tricarboxylic acids.
  • Illustrative of such acids are, for example, acetic, trifluoro acetic acid (2,2,2-trifluoroacetic acid), glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxybenzoic,
  • Either the mono- or di-acid salts can be formed, and such salts can exist in either a hydrated, solvated or substantially anhydrous form.
  • the acid addition salts of these compounds are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms.
  • Other non-pharmaceutically acceptable salts, e.g., oxalates may be used, for example, in the isolation of compounds described herein for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • a "pharmaceutically acceptable basic addition salt” is any non-toxic organic or inorganic base addition salt of the acid compounds described herein or any of its
  • Illustrative inorganic bases which form suitable salts include, but are not limited thereto, lithium, sodium, potassium, calcium, magnesium or barium hydroxides.
  • Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as methylamine, trimethyl amine and picoline or ammonia. The selection of the appropriate salt may be important so that an ester functionality, if any, elsewhere in the molecule is not hydrolyzed. The selection criteria for the appropriate salt will be known to one skilled in the art.
  • Acid addition salts of the compounds described herein are most suitably formed from pharmaceutically acceptable acids, and include, for example, those formed with inorganic acids, e.g., hydrochloric, sulphuric or phosphoric acids and organic acids, e.g., succinic, maleic, acetic, trifluoroacetic or fumaric acid.
  • Other non-pharmaceutically acceptable salts, e.g., oxalates may be used for example in the isolation of compounds described herein for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • base addition salts such as sodium, potassium and ammonium salts
  • solvates and hydrates of compounds of the invention are also included within the scope of the invention. The conversion of a given compound salt to a desired compound salt is achieved by applying standard techniques, well known to one skilled in the art.
  • stereoisomers is a general term for all isomers of the individual molecules that differ only in the orientation of their atoms in space. It includes mirror image isomers (enantiomers), geometric (cis/trans) isomers and isomers of compounds with more than one chiral centre that are not mirror images of one another (diastereomers).
  • treat or “treating” means to alleviate symptoms, eliminate the causation of the symptoms either on a temporary or permanent basis, or to prevent or slow the appearance of symptoms of the named disorder or condition.
  • a therapeutically effective amount is an amount that promotes healing of a wound.
  • promoting wound healing means treating a subject with a wound and achieving healing, either partially or fully, of the wound. Promoting wound healing can mean, e.g., one or more of the following: promoting epidermal closure;
  • terapéuticaally effective amount means an amount of the compound which is effective in treating or lessening the severity of one or more symptoms of a disorder or condition.
  • pharmaceutically acceptable carrier means a non-toxic solvent, dispersant, excipient, adjuvant or other material which is mixed with the active ingredient in order to permit the formation of a pharmaceutical composition, i.e., a dosage form capable of being administered to a subject.
  • a pharmaceutical composition i.e., a dosage form capable of being administered to a subject.
  • pharmaceutically acceptable oil typically used for parenteral administration.
  • Pharmaceutically acceptable carriers are well known in the art.
  • the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the amount of compound in compositions of this invention is such that is effective to measurably inhibit CRM1, in a biological sample or in a patient.
  • a composition of this invention is formulated for administration to a patient in need of such composition.
  • patient means an animal.
  • the animal is a mammal.
  • the patient is a veterinary patient (i.e., a non-human mammal patient).
  • the patient is a dog.
  • the patient is a human.
  • compositions of this invention refers to a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose- based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxy
  • compositions of the present invention may be administered orally, parenterally (including subcutaneous, intramuscular, intravenous and intradermal), by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • provided compounds or compositions are administrable intravenously and/or intraperitoneally.
  • parenteral includes subcutaneous, intravenous, intramuscular, intraocular, intravitreal, intra-articular, intra- synovial, intrasternal, intrathecal, intrahepatic, intraperitoneal intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, subcutaneously, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a nontoxic parenterally acceptable diluent or solvent for example as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are examples of sterile, fixed oils.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • a provided oral formulation is formulated for immediate release or sustained/delayed release.
  • the composition is suitable for buccal or sublingual administration, including tablets, lozenges and pastilles.
  • a provided compound can also be in microencapsulated form.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • Pharmaceutically acceptable compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically- transdermal patches may also be used.
  • compositions may be formulated as micronized suspensions or in an ointment such as petrolatum.
  • compositions of this invention may also be administered by nasal aerosol or inhalation.
  • compositions of this invention are formulated for intra-peritoneal administration.
  • compositions of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration.
  • provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
  • the dosage is from about 0.5 to about 100 mg/kg of body weight, or between 1 mg and 1000 mg/dose, every 4 to 120 hours, or according to the requirements of the particular drug.
  • the pharmaceutical compositions of this invention will be administered from about 1 to about 6 times per day.
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
  • a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary.
  • the dosage or frequency of administration, or both may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level.
  • Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms
  • the present invention provides a method for treating a CRM 1 -mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
  • the compounds and compositions described herein can also be administered to cells in culture, e.g. in vitro or ex vivo, or to a subject, e.g., in vivo, to treat, prevent, and/or diagnose a variety of disorders, including those described herein below.
  • CRM1 -mediated disorder or condition means any disease or other deleterious condition in which CRM1 is known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which CRM1 is known to play a role.
  • the present invention provides methods of treating a disease associated with expression or activity of p53, p73, p21, pRB, p27, MB, NFKB, c-Abl, FOXO proteins, COX- 2, or an HDAC (histone deacetylases) in a subject comprising administering to the patient a therapeutically effective amount of a compound described herein.
  • the present invention relates to a method of treating or lessening the severity of a disease or condition selected from a proliferative disorder (e.g., cancer), an inflammatory disorder, an autoimmune disorder, a viral infection, an ophthalmological disorder or a neurodegenerative disorder wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention.
  • the present invention relates to a method of treating or lessening the severity of cancer. Specific examples of the above disorders are set forth in detail below.
  • Cancers treatable by the compounds of this invention include, but are not limited to, hematologic malignancies (leukemias, lymphomas, myelomas including multiple myeloma, myelodysplastic and myeloproliferative syndromes) and solid tumors (carcinomas such as prostate, breast, lung, colon, pancreatic, renal, ovarian as well as soft tissue and osteosarcomas, and stromal tumors).
  • Breast cancer (BC) can include basal-like breast cancer (BLBC), triple negative breast cancer (TNBC) and breast cancer that is both BLBC and TNBC.
  • breast cancer can include invasive or non-invasive ductal or lobular carcinoma, tubular, medullary, mucinous, papillary, cribriform carcinoma of the breast, male breast cancer, recurrent or metastatic breast cancer, phyllodes tumor of the breast and Paget' s disease of the nipple.
  • Inflammatory disorders treatable by the compounds of this invention include, but are not limited to, multiple sclerosis, rheumatoid arthritis, degenerative joint disease, systemic lupus, systemic sclerosis, vasculitis syndromes (small, medium and large vessel), atherosclerosis, inflammatory bowel disease, irritable bowel syndrome, Crohn's disease, mucous colitis, ulcerative colitis, gastritis, sepsis, psoriasis and other dermatological inflammatory disorders (such as eczema, atopic dermatitis, contact dermatitis, urticaria, scleroderma, and dermatosis with acute inflammatory components, pemphigus, pemphigoid, allergic dermatitis), and urticarial syndromes.
  • multiple sclerosis rheumatoid arthritis, degenerative joint disease, systemic lupus, systemic sclerosis, vasculitis syndromes (small, medium and large vessel), atherosclerosis,
  • Viral diseases treatable by the compounds of this invention include, but are not limited to, acute febrile pharyngitis, pharyngoconjunctival fever, epidemic
  • keratoconjunctivitis infantile gastroenteritis, Coxsackie infections, infectious mononucleosis, Burkitt lymphoma, acute hepatitis, chronic hepatitis, hepatic cirrhosis, hepatocellular carcinoma, primary HSV-1 infection (e.g., gingivostomatitis in children, tonsillitis and pharyngitis in adults, keratoconjunctivitis), latent HSV-1 infection (e.g., herpes labialis and cold sores), primary HSV-2 infection, latent HSV-2 infection, aseptic meningitis, infectious mononucleosis, Cytomegalic inclusion disease, Kaposi's sarcoma, multicentric Castleman disease, primary effusion lymphoma, AIDS, influenza, Reye syndrome, measles,
  • primary HSV-1 infection e.g., gingivostomatitis in children, tonsillitis and pharyngitis in
  • Viral diseases treatable by the compounds of this invention also include chronic viral infections, including hepatitis B and hepatitis C.
  • Exemplary ophthalmology disorders include, but are not limited to, macular edema (diabetic and nondiabetic macular edema), aged related macular degeneration wet and dry forms, aged disciform macular degeneration, cystoid macular edema, palpebral edema, retina edema, diabetic retinopathy, chorioretinopathy, neovascular maculopathy, neovascular glaucoma, uveitis, ulceris, retinal vasculitis, endophthalmitis, panophthalmitis, metastatic ophthalmia, choroiditis, retinal pigment epitheliitis, conjunctivitis, cyclitis, scleritis, episcleritis, optic neuritis, retrobulbar optic neuritis, keratitis, blepharitis, exudative retinal detachment, corneal ulcer, conjunctival ulcer, chronic nummular
  • Neurodegenerative diseases treatable by a compound of Formula I include, but are not limited to, Parkinson's, Alzheimer's, and Huntington's, and Amyotrophic lateral sclerosis (ALS/Lou Gehrig's Disease).
  • Compounds and compositions described herein may also be used to treat disorders of abnormal tissue growth and fibrosis including dilative cardiomyopathy, hypertrophic cardiomyopathy, restrictive cardiomyopathy, pulmonary fibrosis, hepatic fibrosis,
  • PWD polycystic kidney disorder
  • Compounds and compositions described herein may also be used to treat disorders related to food intake such as obesity and hyperphagia.
  • a compound or composition described herein may be used to treat or prevent allergies and respiratory disorders, including asthma, bronchitis, pulmonary fibrosis, allergic rhinitis, oxygen toxicity, emphysema, chronic bronchitis, acute respiratory distress syndrome, and any chronic obstructive pulmonary disease (COPD).
  • allergies and respiratory disorders including asthma, bronchitis, pulmonary fibrosis, allergic rhinitis, oxygen toxicity, emphysema, chronic bronchitis, acute respiratory distress syndrome, and any chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • the disorder or condition associated with CRM1 activity is muscular dystrophy, arthritis, for example, osteoarthritis and rheumatoid arthritis, ankylosing spondilitis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, musclewasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary dystrophy, arthritis, for example, osteoarthritis and rheumatoid arthritis, ankylosing spondilitis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type
  • nephritis familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy,
  • the disorder or condition associated with CRM1 activity is head injury, uveitis, inflammatory pain, allergen induced asthma, non-allergen induced asthma, glomerular nephritis, ulcerative colitis, necrotizing enterocolitis, hyperimmunoglobulinemia D with recurrent fever (HIDS), TNF receptor associated periodic syndrome (TRAPS), cryopyrin-associated periodic syndromes, Muckle- Wells syndrome (urticaria deafness amyloidosis),familial cold urticaria, neonatal onset multisystem inflammatory disease (NOMID), periodic fever, aphthous stomatitis, pharyngitis and adenitis (PFAPA syndrome), Blau syndrome, pyogenic sterile arthritis, pyoderma gangrenosum,acne (PAPA), deficiency of the interleukin-1 -receptor antagonist (DIRA), subarachnoid hemorrhage, polycy
  • the present invention provides a use of a compound described herein for the manufacture of a medicament for the treatment of a disease associated with expression or activity of p53, p73, p21, pRB, p27, ⁇ , NFKB, c-Abl, FOXO proteins, COX- 2 or an HDAC in a subject.
  • the present invention provides a use of a compound described herein in the manufacture of a medicament for the treatment of any of cancer and/or neoplastic disorders, angiogenesis, autoimmune disorders, inflammatory disorders and/or diseases, epigenetics, hormonal disorders and/or diseases, viral diseases, neurodegenerative disorders and/or diseases, wounds, and ophthalmologic disorders.
  • the present invention provides a method for inhibiting CRM1 in a biological sample comprising contacting the biological sample with, or administering to the patient, a pharmaceutically acceptable salt of a compound of the invention, or pharmaceutically acceptable composition thereof.
  • a compound or composition described herein can be used to treat a neoplastic disorder.
  • a "neoplastic disorder” is a disease or disorder characterized by cells that have the capacity for autonomous growth or replication, e.g., an abnormal state or condition characterized by proliferative cell growth.
  • Exemplary neoplastic disorders include:
  • carcinoma sarcoma
  • metastatic disorders e.g., tumors arising from prostate, brain, bone, colon, lung, breast, ovarian, and liver origin
  • hematopoietic neoplastic disorders e.g., leukemias, lymphomas, myeloma and other malignant plasma cell disorders, and metastatic tumors.
  • Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers.
  • Treatment with the compound can be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
  • the disclosed methods are useful in the prevention and treatment of cancer, including for example, solid tumors, soft tissue tumors, and metastases thereof, as well as in familial cancer syndromes such as Li Fraumeni Syndrome. Familial Breast-Ovarian Cancer (BRCA1 or BRAC2 mutations) Syndromes, and others.
  • the disclosed methods are also useful in treating non-solid cancers.
  • Exemplary solid tumors include malignancies (e.g., sarcomas, adenocarcinomas, and carcinomas) of the various organ systems, such as those of lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary (e.g., renal, urothelial, or testicular tumors) tracts, pharynx, prostate, and ovary.
  • malignancies e.g., sarcomas, adenocarcinomas, and carcinomas
  • gastrointestinal e.g., colon
  • genitourinary e.g., renal, urothelial, or testicular tumors
  • Exemplary adenocarcinomas include colorectal cancers, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, and cancer of the small intestine.
  • Exemplary cancers described by the National Cancer Institute include: Acute Lymphoblastic Leukemia, Adult; Acute Lymphoblastic Leukemia, Childhood; Acute
  • Ependymoma Childhood; Epithelial Cancer, Ovarian; Esophageal Cancer; Esophageal Cancer, Childhood; Ewing's Family of Tumors; Extracranial Germ Cell Tumor, Childhood; Extragonadal Germ Cell Tumor; Extrahepatic Bile Duct Cancer; Eye Cancer, Intraocular Melanoma; Eye Cancer, Retinoblastoma; Gallbladder Cancer; Gastric (Stomach) Cancer; Gastric (Stomach) Cancer, Childhood; Gastrointestinal Carcinoid Tumor; Germ Cell Tumor, Extracranial, Childhood; Germ Cell Tumor, Extragonadal; Germ Cell Tumor, Ovarian;
  • Hodgkin's Lymphoma Adult; Hodgkin's Lymphoma, Childhood; Hodgkin's Lymphoma During Pregnancy; Hypopharyngeal Cancer; Hypothalamic and Visual Pathway Glioma, Childhood; Intraocular Melanoma; Islet Cell Carcinoma (Endocrine Pancreas); Kaposi's Sarcoma; Kidney Cancer; Laryngeal Cancer; Laryngeal Cancer, Childhood; Leukemia, Acute Lymphoblastic, Adult; Leukemia, Acute Lymphoblastic, Childhood; Leukemia, Acute Myeloid, Adult; Leukemia, Acute Myeloid, Childhood; Leukemia, Chronic Lymphocytic; Leukemia, Chronic Myelogenous; Leukemia, Hairy Cell; Lip and Oral Cavity Cancer; Liver Cancer, Adult (Primary); Liver Cancer, Childhood (Primary); Lung Cancer, Non-Small Cell; Lung Cancer, Small Cell; Lymphoblastic Leuk
  • Lymphoma Central Nervous System (Primary); Lymphoma, Cutaneous T-Cell; Lymphoma, Hodgkin's, Adult; Lymphoma, Hodgkin's, Childhood; Lymphoma, Hodgkin's During
  • Lymphoma Non-Hodgkin's, Adult
  • Lymphoma Non- Hodgkin's, Childhood
  • Lymphoma Non-Hodgkin's During Pregnancy; Lymphoma, Primary Central Nervous System; Macroglobulinemia, Waldenstrom's; Male Breast Cancer; Malignant Mesothelioma, Adult; Malignant Mesothelioma, Childhood; Malignant Thymoma; Medulloblastoma, Childhood; Melanoma; Melanoma, Intraocular; Merkel Cell Carcinoma; Mesothelioma, Malignant; Metastatic Squamous Neck Cancer with Occult Primary; Multiple Endocrine Neoplasia Syndrome, Childhood; Multiple Myeloma/Plasma Cell Neoplasm; Mycosis Fungoides; Myelodysplastic Syndromes; Myelogenous Leukemia, Chronic; Myeloid
  • Ovarian Epithelial Cancer Ovarian Germ Cell Tumor; Ovarian Low Malignant Potential Tumor; Pancreatic Cancer; Pancreatic Cancer, Childhood; Pancreatic Cancer, Islet Cell;
  • Further exemplary cancers include diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL). Yet further exemplary cancers include endocervical cancer, B- cell ALL, T-cell ALL, B- or T-cell lymphoma, mast cell cancer, glioblastoma,
  • Exemplary sarcomas include fibrosarcoma, alveolar soft part sarcoma (ASPS), liposarcoma, leiomyosarcoma, chondrosarcoma, synovial sarcoma, chordoma, spindle cell sarcoma, histiocytoma, rhabdomyosarcoma, Ewing's sarcoma, neuroectodermal sarcoma, phyllodes/osteogenic sarcoma and chondroblastic osteosarcoma.
  • Metastases of the aforementioned cancers can also be treated or prevented in accordance with the methods described herein.
  • a compound described herein is administered together with an additional "second" therapeutic agent or treatment.
  • second therapeutic agent may be made from any agent that is typically used in a monotherapy to treat the indicated disease or condition.
  • the term “administered together” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention.
  • a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.
  • the present invention provides a single unit dosage form comprising a compound of the invention, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the effective amount of the compound of this invention is less than its effective amount would be where the second therapeutic agent is not administered. In another embodiment, the effective amount of the second therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized. Other potential advantages (including without limitation improved dosing regimens and/or reduced drug cost) will be apparent to those of skill in the art.
  • the additional agents may be administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds of this invention in a single composition.
  • a compound described herein is administered together with an additional cancer treatment.
  • additional cancer treatments include, for example: chemotherapy, targeted therapies such as antibody therapies, kinase inhibitors, immunotherapy, and hormonal therapy, epigenetic therapy, proteosome inhibitors, and anti- angiogenic therapies. Examples of each of these treatments are provided below.
  • the term "combination,” “combined,” and related terms refer to the simultaneous or sequential administration of therapeutic agents in accordance with this invention.
  • a compound of the present invention can be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present invention provides a single unit dosage form comprising a compound of the invention, an additional therapeutic agent, and a
  • compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of a compound of the invention can be administered.
  • a compound described herein is administered with a chemotherapy.
  • Chemotherapy is the treatment of cancer with drugs that can destroy cancer cells. "Chemotherapy” usually refers to cytotoxic drugs which affect rapidly dividing cells in general, in contrast with targeted therapy. Chemotherapy drugs interfere with cell division in various possible ways, e.g., with the duplication of DNA or the separation of newly formed chromosomes. Most forms of chemotherapy target all rapidly dividing cells and are not specific for cancer cells, although some degree of specificity may come from the inability of many cancer cells to repair DNA damage, while normal cells generally can.
  • chemotherapeutic agents used in cancer therapy include, for example, antimetabolites (e.g., folic acid, purine, and pyrimidine derivatives) and alkylating agents (e.g., nitrogen mustards, nitrosoureas, platinum, alkyl sulfonates, hydrazines, triazenes, aziridines, spindle poison, cytotoxic agents, topoisomerase inhibitors and others).
  • antimetabolites e.g., folic acid, purine, and pyrimidine derivatives
  • alkylating agents e.g., nitrogen mustards, nitrosoureas, platinum, alkyl sulfonates, hydrazines, triazenes, aziridines, spindle poison, cytotoxic agents, topoisomerase inhibitors and others.
  • agents include Aclarubicin, Actinomycin, Alitretinoin, Altretamine, Aminopterin, Aminolevulinic acid, Amrubicin, Amsacrine, Anagrelide, Arsenic trioxide, Asparaginase, Atrasentan, Belotecan, Bexarotene, Bendamustin, Bleomycin, Bortezomib, Busulfan, Camptothecin, Capecitabine, Carboplatin, Carboquone, Carmofur, Carmustine, Celecoxib, Chlorambucil, Chlormethine, Cisplatin, Cladribine, Clofarabine, Crisantaspase, Cyclophosphamide, Cytarabine, dacarbazine, Dactinomycin, Daunorubicin, Decitabine, Demecolcine, Docetaxel, Doxorubicin, Efaproxiral, Elesclomol, Elsamitrucin, En
  • Procarbazine Raltitrexed, Ranimustine, Rubitecan, Sapacitabine, Semustine, Sitimagene ceradenovec, Strataplatin, Streptozocin, Talaporfin, Tegafur-uracil, Temoporfin,
  • Temozolomide Teniposide, Tesetaxel, Testolactone, Tetranitrate, Thiotepa, Tiazofurine, Tioguanine, Tipifarnib, Topotecan, Trabectedin, Triaziquone, Triethylenemelamine,
  • Tretinoin Triplatin, Tretinoin, Treosulfan, Trofosfamide, Uramustine, Valrubicin, Verteporfin, Vinblastine, Vincristine, Vindesine, Vinflunine, Vinorelbine, Vorinostat, Zorubicin, and other cytostatic or cytotoxic agents described herein.
  • the chemotherapy agents can be used in combination with a compound described herein.
  • Targeted therapy constitutes the use of agents specific for the deregulated proteins of cancer cells.
  • Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell.
  • Prominent examples are the tyrosine kinase inhibitors such as Axitinib, Bosutinib, Cediranib, desatinib, erolotinib, imatinib, gefitinib, lapatinib, Lestaurtinib, Nilotinib, Semaxanib, Sorafenib, Sunitinib, and Vandetanib, and also cyclin-dependent kinase inhibitors such as Alvocidib and Seliciclib.
  • Monoclonal antibody therapy is another strategy in which the therapeutic agent is an antibody which specifically binds to a protein on the surface of the cancer cells.
  • the therapeutic agent is an antibody which specifically binds to a protein on the surface of the cancer cells.
  • Examples include the anti-HER2/neu antibody trastuzumab (Herceptin®) typically used in breast cancer, and the anti-CD20 antibody rituximab and Tositumomab typically used in a variety of B-cell malignancies.
  • Other exemplary antibodies include Cetuximab, Panitumumab, Trastuzumab, Alemtuzumab, Bevacizumab, Edrecolomab, and Gemtuzumab.
  • Exemplary fusion proteins include Aflibercept and Denileukin diftitox.
  • the targeted therapy can be used in combination with a compound described herein, e.g., Gleevec (Vignari and Wang 2001).
  • Targeted therapy can also involve small peptides as '3 ⁇ 4oming devices" which can bind to cell surface receptors or affected extracellular matrix surrounding the tumor.
  • Radionuclides which are attached to these peptides eventually kill the cancer cell if the nuclide decays in the vicinity of the cell.
  • RGDs Radionuclides which are attached to these peptides
  • An example of such therapy includes BEXXAR®.
  • Compounds and methods described herein may be used to treat or prevent a disease or disorder associated with angiogenesis.
  • Diseases associated with angiogenesis include cancer, cardiovascular disease and macular degeneration.
  • Angiogenesis is the physiological process involving the growth of new blood vessels from pre-existing vessels. Angiogenesis is a normal and vital process in growth and development, as well as in wound healing and in granulation tissue. However, it is also a fundamental step in the transition of tumors from a dormant state to a malignant one.
  • Angiogenesis may be a target for combating diseases characterized by either poor vascularisation or abnormal vasculature.
  • VEGF Vascular endothelial growth factor
  • Tumors induce blood vessel growth (angiogenesis) by secreting various growth factors ⁇ e.g., VEGF).
  • growth factors such as bFGF and VEGF can induce capillary growth into the tumor, which some researchers suspect supply required nutrients, allowing for tumor expansion.
  • Angiogenesis represents an excellent therapeutic target for the treatment of cardiovascular disease. It is a potent, physiological process that underlies the natural manner in which our bodies respond to a diminution of blood supply to vital organs, namely the production of new collateral vessels to overcome the ischemic insult.
  • VEGF vascular endothelial growth factor
  • VEGF causes increased permeability in blood vessels in addition to stimulating angiogenesis.
  • VEGF causes proliferation of capillaries into the retina. Since the increase in angiogenesis also causes edema, blood and other retinal fluids leak into the retina, causing loss of vision.
  • Anti-angiogenic therapy can include kinase inhibitors targeting vascular endothelial growth factor (VEGF) such as sunitinib, sorafenib, or monoclonal antibodies or receptor "decoys" to VEGF or VEGF receptor including bevacizumab or VEGF-Trap, or thalidomide or its analogs (lenalidomide, pomalidomide), or agents targeting non-VEGF angiogenic targets such as fibroblast growth factor (FGF), angiopoietins, or angiostatin or endo statin.
  • VEGF vascular endothelial growth factor
  • FGF fibroblast growth factor
  • angiopoietins angiostatin or endo statin.
  • Epigenetics is the study of heritable changes in phenotype or gene expression caused by mechanisms other than changes in the underlying DNA sequence.
  • epigenetic changes in eukaryotic biology is the process of cellular differentiation. During morphogenesis, stem cells become the various cell lines of the embryo which in turn become fully differentiated cells. In other words, a single fertilized egg cell changes into the many cell types including neurons, muscle cells, epithelium, blood vessels etc. as it continues to divide. It does so by activating some genes while inhibiting others.
  • Epigenetic changes are preserved when cells divide. Most epigenetic changes only occur within the course of one individual organism's lifetime, but, if a mutation in the DNA has been caused in sperm or egg cell that results in fertilization, then some epigenetic changes are inherited from one generation to the next. Specific epigenetic processes include paramutation, bookmarking, imprinting, gene silencing, X chromosome inactivation, position effect, reprograrnming, transvection, maternal effects, the progress of carcinogenesis, many effects of teratogens, regulation of histone modifications and heterochromatin, and technical limitations affecting parthenogenesis and cloning.
  • Exemplary diseases associated with epigenetics include ATR-syndrome, fragile X-syndrome, ICF syndrome, Angelman's syndrome, Prader-Wills syndrome, BWS, Rett syndrome, ot-thalassaemia, cancer, leukemia, Rubinstein-Taybi syndrome and Coffin-Lowry syndrome.
  • the first human disease to be linked to epigenetics was cancer.
  • DNA methylation occurs at CpG sites, and a majority of CpG cytosines are methylated in mammals.
  • CpG islands stretches of DNA near promoter regions that have higher concentrations of CpG sites (known as CpG islands) that are free of methylation in normal cells.
  • CpG islands become excessively methylated in cancer cells, thereby causing genes that should not be silenced to turn off.
  • This abnormality is the trademark epigenetic change that occurs in tumors and happens early in the development of cancer. Hypermethylation of CpG islands can cause tumors by shutting off tumor-suppressor genes. In fact, these types of changes may be more common in human cancer than DNA sequence mutations.
  • MGMT 06-methylguanine-DNA methyltransferase
  • CDKN2B MLH1 cyclin-dependent kinase inhibitor 2B
  • RASSF1A RASSF1A
  • hypermethylation of the promoter of MGMT causes the number of G-to-A mutations to increase.
  • Microsatellites are common in normal individuals, and they usually consist of repeats of the dinucleotide CA. Too much methylation of the promoter of the DNA repair gene MLH1 can make a micro satellite unstable and lengthen or shorten it. Microsatellite instability has been linked to many cancers, including colorectal, endometrial, ovarian, and gastric cancers.
  • Fragile X syndrome is the most frequently inherited mental disability, particularly in males. Both sexes can be affected by this condition, but because males only have one X chromosome, one fragile X will impact them more severely. Indeed, fragile X syndrome occurs in approximately 1 in 4,000 males and 1 in 8,000 females. People with this syndrome have severe intellectual disabilities, delayed verbal development, and "autistic-like" behavior.
  • Fragile X syndrome gets its name from the way the part of the X chromosome that contains the gene abnormality looks under a microscope; it usually appears as if it is hanging by a thread and easily breakable. The syndrome is caused by an abnormality in the FMR1 (fragile X mental retardation 1) gene. People who do not have fragile X syndrome have 6 to 50 repeats of the trinucleotide CGG in their FMR1 gene. However, individuals with over 200 repeats have a full mutation, and they usually show symptoms of the syndrome. Too many CGGs cause the CpG islands at the promoter region of the FMR1 gene to become
  • Fragile X syndrome is not the only disorder associated with mental retardation that involves epigenetic changes. Other such conditions include Rubenstein-Taybi, Coffin- Lowry, Prader-Willi, Angelman, Beckwith- Wiedemann, ATR-X, and Rett syndromes.
  • Epigenetic therapies include inhibitors of enzymes controlling epigenetic modifications, specifically DNA methyltransferases and histone deacetylases, which have shown promising anti-tumorigenic effects for some malignancies, as well as antisense oligonucleotides and siRNA.
  • a compound described herein is administered with an immunotherapy.
  • Cancer immunotherapy refers to a diverse set of therapeutic strategies designed to induce the patient's own immune system to fight the tumor.
  • Contemporary methods for generating an immune response against tumors include intravesicular BCG immunotherapy for superficial bladder cancer, prostate cancer vaccine Provenge, and use of interferons and other cytokines to induce an immune response in renal cell carcinoma and melanoma patients.
  • Allogeneic hematopoietic stem cell transplantation can be considered a form of immunotherapy, since the donor's immune cells will often attack the tumor in a graft-versus- tumor effect.
  • the immunotherapy agents can be used in combination with a compound described herein.
  • a compound described herein is administered with a hormonal therapy.
  • the growth of some cancers can be inhibited by providing or blocking certain hormones.
  • hormone- sensitive tumors include certain types of breast and prostate cancers, as well as certain types of leukemia which respond to certain retinoids/retinoic acids. Removing or blocking estrogen or testosterone is often an important additional treatment.
  • administration of hormone agonists, such as progestogens may be therapeutically beneficial.
  • the hormonal therapy agents can be used in combination with a compound described herein.
  • Hormonal therapy agents include the administration of hormone agonists or hormone antagonists and include retinoids/retinoic acid, compounds that inhibit estrogen or testosterone, as well as administration of progestogens.
  • the compounds and methods described herein may be used to treat or prevent a disease or disorder associated with inflammation, particularly in humans and other mammals.
  • a compound described herein may be administered prior to the onset of, at, or after the initiation of inflammation.
  • the compounds are preferably provided in advance of any inflammatory response or symptom. Administration of the compounds can prevent or attenuate inflammatory responses or symptoms.
  • Exemplary inflammatory conditions include, for example, multiple sclerosis, rheumatoid arthritis, psoriatic arthritis, degenerative joint disease, spondouloarthropathies, other seronegative inflammatory arthridities, polymyalgia rheumatica, various vasculidities (e.g., giant cell arteritis, ANCA+ vasculitis), gouty arthritis, systemic lupus erythematosus, juvenile arthritis, juvenile rheumatoid arthritis, osteoarthritis, osteoporosis, diabetes (e.g., insulin dependent diabetes mellitus or juvenile onset diabetes), menstrual cramps, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, Crohn's disease, mucous colitis, ulcerative colitis, gastritis, esophagitis, pancreatitis, peritonitis, Alzheimer's disease, shock, ankylosing spondylitis, gastritis,
  • Exemplary inflammatory conditions of the skin include, for example, eczema, atopic dermatitis, contact dermatitis, urticaria, schleroderma, psoriasis, and dermatosis with acute inflammatory components.
  • a compound or method described herein may be used to treat or prevent allergies and respiratory conditions, including asthma, bronchitis, pulmonary fibrosis, allergic rhinitis, oxygen toxicity, emphysema, chronic bronchitis, acute respiratory distress syndrome, and any chronic obstructive pulmonary disease (COPD).
  • the compounds may be used to treat chronic hepatitis infection, including hepatitis B and hepatitis C.
  • a compound or method described herein may be used to treat autoimmune diseases and/or inflammation associated with autoimmune diseases, such as organ-tissue autoimmune diseases (e.g., Raynaud's syndrome), scleroderma, myasthenia gravis, transplant rejection, endotoxin shock, sepsis, psoriasis, eczema, dermatitis, multiple sclerosis, autoimmune thyroiditis, uveitis, systemic lupus erythematosis, Addison's disease, autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), and Grave's disease.
  • the compounds described herein can be used to treat multiple sclerosis.
  • a compound described herein may be administered alone or in combination with other compounds useful for treating or preventing inflammation.
  • anti-inflammatory agents include, for example, steroids ⁇ e.g., Cortisol, cortisone, fludrocortisone, prednisone, 6[alpha]-methylprednisone, triamcinolone, betamethasone or dexamethasone), nonsteroidal antiinflammatory drugs (NSAIDS (e.g., aspirin,
  • the other therapeutic agent is an antibiotic (e.g. , vancomycin, penicillin, amoxicillin, ampicillin, cefotaxime, ceftriaxone, cefixime, rifampinmetronidazole, doxycycline or streptomycin).
  • the other therapeutic agent is a PDE4 inhibitor (e.g., roflumilast or rolipram).
  • the other therapeutic agent is an antihistamine (e.g.
  • the other therapeutic agent is an anti-malarial (e.g., artemisinin, artemether, artsunate, chloroquine phosphate, mefloquine hydrochloride, doxycycline hyclate, proguanil hydrochloride, atovaquone or halofantrine).
  • the other compound is drotrecogin alfa.
  • anti-inflammatory agents include, for example, aceclofenac, acemetacin, e-acetamidocaproic acid, acetaminophen, acetaminosalol, acetanilide, acetylsalicylic acid, S-adenosylmethionine, alclofenac, alclometasone, alfentanil, algestone, allylprodine, alminoprofen, aloxiprin, alphaprodine, aluminum bis(acetylsalicylate), amcinonide, amfenac, aminochlorthenoxazin, 3-amino-4- hydroxybutyric acid, 2-amino-4- picoline, aminopropylon, aminopyrine, amixetrine, ammonium salicylate, ampiroxicam, amtolmetin guacil, anileridine, antipyrine, antrafenine, apazone, beclomethasone, bendazac, benory
  • chloroprednisone chlorthenoxazin, choline salicylate, cinchophen, cinmetacin, ciramadol, clidanac, clobetasol, clocortolone, clometacin, clonitazene, clonixin, clopirac, cloprednol, clove, codeine, codeine methyl bromide, codeine phosphate, codeine sulfate, cortisone, cortivazol, cropropamide, crotethamide, cyclazocine, deflazacort, dehydrotestosterone, desomorphine, desonide, desoximetasone, dexamethasone, dexamethasone-21- isonicotinate, dexoxadrol, dextromoramide, dextropropoxyphene, deoxycorticosterone, dezocine, diampromide, diamorphone, diclofenac, difen
  • a compound described herein may be administered with a selective COX-2 inhibitor for treating or preventing inflammation.
  • selective COX-2 inhibitors include, for example, deracoxib, parecoxib, celecoxib, valdecoxib, rofecoxib, etoricoxib, and lumiracoxib.
  • a provided compound is administered in combination with an anthracycline or a Topo II inhibitor.
  • a provided compound is administered in combination with Doxorubicin (Dox).
  • a provided compound is administered in combination with bortezomib (and more broadly including carfilzomib). It was surprisingly found that a provided compound in combination with Dox or bortezomib resulted in a synergystic effect (i.e., more than additive).
  • Compounds and methods described herein may be used to treat or prevent a disease or disorder associated with a viral infection, particularly in humans and other mammals.
  • a compound described herein may be administered prior to the onset of, at, or after the initiation of viral infection.
  • the compounds are preferably provided in advance of any viral infection or symptom thereof.
  • Exemplary viral diseases include acute febrile pharyngitis, pharyngoconjunctival fever, epidemic keratoconjunctivitis, infantile gastroenteritis, Coxsackie infections, infectious mononucleosis, Burkitt lymphoma, acute hepatitis, chronic hepatitis, hepatic cirrhosis, hepatocellular carcinoma, primary HSV-1 infection (e.g., gingivostomatitis in children, tonsillitis and pharyngitis in adults, keratoconjunctivitis), latent HSV-1 infection (e.g., herpes labialis and cold sores), primary HSV-2 infection, latent HSV-2 infection, aseptic meningitis, infectious mononucleosis, Cytomegalic inclusion disease, Kaposi's sarcoma, multicentric Castleman disease, primary effusion lymphoma, AIDS, influenza, Reye syndrome, measles, postin
  • Exemplary viral influenza A strains include H1N1, H3N2, H5N1 , H7N3, H7N9.
  • a compound described herein can also be used to treat or prevent influenza B.
  • Exemplary viral pathogens include Adenovirus, Coxsackievirus, Dengue virus, Encephalitis Virus, Epstein-Barr virus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus.
  • Herpes simplex virus type 1 Herpes simplex virus type 2, cytomegalovirus, Human herpesvirus type 8, Human immunodeficiency virus, Influenza virus, measles virus, Mumps virus, Human papillomavirus, Parainfluenza virus, Poliovirus, Rabies virus, Respiratory syncytial virus, Rubella virus, Varicella-zoster virus, West Nile virus, Dungee, and Yellow fever virus.
  • Viral pathogens may also include viruses that cause resistant viral infections.
  • Antiviral drugs are a class of medications used specifically for treating viral infections. Antiviral action generally falls into one of three mechanisms: interference with the ability of a virus to infiltrate a target cell (e.g., amantadine, rimantadine and pleconaril), inhibition of the synthesis of virus (e.g., nucleoside analogues, e.g., acyclovir and zidovudine (AZT), and inhibition of the release of virus (e.g., zanamivir and oseltamivir).
  • a target cell e.g., amantadine, rimantadine and pleconaril
  • inhibition of the synthesis of virus e.g., nucleoside analogues, e.g., acyclovir and zidovudine (AZT)
  • ZT zidovudine
  • ophthamology disorders include macular edema
  • age related macular degeneration wet and dry forms, aged disciform macular degeneration, cystoid macular edema, palpebral edema, retina edema, diabetic retinopathy, chorioretinopathy, neovascular maculopathy, neovascular glaucoma, uveitis, ulceris, retinal vasculitis, endophthalmitis, panophthalmitis, metastatic ophthalmia, choroiditis, retinal pigment epithelitis, conjunctivitis, cyclitis, scleritis, episcleritis, optic neuritis, retrobulbar optic neuritis, keratitis, blepharitis, exudative retinal detachment, corneal ulcer, conjunctival ulcer, chronic nummular keratitis, ophthalmic disease associated with hypoxia or ischemia, retinopathy of prematur
  • ophthalmology disorders treatable using the compounds and methods described herein include proliferative vitreoretinopathy and chronic retinal detachment.
  • Inflammatory eye diseases are also treatable using the compounds and methods described herein.
  • Neurodegeneration is the umbrella term for the progressive loss of structure or function of neurons, including death of neurons.
  • Many neurodegenerative diseases including Parkinson's, Alzheimer's, and Huntington's occur as a result of neurodegenerative processes.
  • Many similarities appear which relate these diseases to one another on a sub-cellular level. Discovering these similarities offers hope for therapeutic advances that could ameliorate many diseases simultaneously.
  • There are many parallels between different neurodegenerative disorders including atypical protein assemblies as well as induced cell death.
  • Alzheimer's disease is characterized by loss of neurons and synapses in the cerebral cortex and certain subcortical regions. This loss results in gross atrophy of the affected regions, including degeneration in the temporal lobe and parietal lobe, and parts of the frontal cortex and cingulate gyrus.
  • Huntington's disease causes astrogliosis and loss of medium spiny neurons. Areas of the brain are affected according to their structure and the types of neurons they contain, reducing in size as they cumulatively lose cells. The areas affected are mainly in the striatum, but also the frontal and temporal cortices.
  • the striatum's subthalamic nuclei send control signals to the globus pallidus, which initiates and modulates motion. The weaker signals from subthalamic nuclei thus cause reduced initiation and modulation of movement, resulting in the characteristic movements of the disorder.
  • Exemplary treatments for Huntington's disease include tetrabenazine, neuroleptics, benzodiazepines, amantadine, remacemide, valproic acid, selective serotonin reuptake inhibitors (SSRIs), mirtazapine and antipsychotics.
  • SSRIs selective serotonin reuptake inhibitors
  • the mechanism by which the brain cells in Parkinson's are lost may consist of an abnormal accumulation of the protein alpha-synuclein bound to ubiquitin in the damaged cells.
  • the alpha-synuclein-ubiquitin complex cannot be directed to the proteosome.
  • This protein accumulation forms proteinaceous cytoplasmic inclusions called Lewy bodies.
  • the latest research on pathogenesis of disease has shown that the death of dopaminergic neurons by alpha-synuclein is due to a defect in the machinery that transports proteins between two major cellular organelles— the endoplasmic reticulum (ER) and the Golgi apparatus. Certain proteins like Rabl may reverse this defect caused by alpha-synuclein in animal models.
  • Exemplary Parkinson's disease therapies include levodopa, dopamine agonists such as include bromocriptine, pergolide, pramipexole, ropinirole, piribedil, cabergoline,
  • apomorphine and lisuride dopa decarboxylate inhibitors
  • MAO-B inhibitors such as selegilene and rasagilene
  • anticholinergics and amantadine apomorphine and lisuride
  • MAO-B inhibitors such as selegilene and rasagilene
  • anticholinergics and amantadine apomorphine and lisuride
  • MAO-B inhibitors such as selegilene and rasagilene
  • Amyotrophic lateral sclerosis is a disease in which motor neurons are selectively targeted for degeneration.
  • exemplary ALS therapies include riluzole, baclofen, diazepam, trihexyphenidyl and amitriptyline.
  • neurodegenerative therapeutics include antisense
  • Wounds are a type of condition characterized by cell or tissue damage. Wound healing is a dynamic pathway that optimally leads to restoration of tissue integrity and function.
  • the wound healing process consists of three overlapping phases.
  • the first phase is an inflammatory phase, which is characterized by homeostasis, platelet aggregation and degranulation. Platelets as the first response, release multiple growth factors to recruit immune cells, epithelial cells, and endothelial cells.
  • the inflammatory phase typically occurs over days 0-5.
  • the second stage of wound healing is the proliferative phase during which macrophages and granulocytes invade the wound. Infiltrating fibroblasts begin to produce collagen.
  • the principle characteristics of this phase are epithelialization, angiogenesis, granulation tissue formation and collagen production.
  • the proliferative phase typically occurs over days 3-14.
  • the third phase is the remodeling phase where matrix formation occurs.
  • the fibroblasts, epithelial cells, and endothelial cells continue to produce collagen and collagenase as well as matrix metalloproteases (MMPs) for remodeling. Collagen crosslinking takes place and the wound undergoes contraction.
  • MMPs matrix metalloproteases
  • the remodeling phase typically occurs from day 7 to one year.
  • the present invention provides a method for promoting wound healing in a subject, comprising administering to the subject a therapeutically effective amount of a compound (e.g., a CRM1 inhibitor), or
  • the method need not achieve complete healing or closure of the wound; it is sufficient for the method to promote any degree of wound closure. In this respect, the method can be employed alone or as an adjunct to other methods for healing wounded tissue.
  • the compounds and compositions described herein can be used to treat wounds during the inflammatory (or early) phase, during the proliferative (or middle) wound healing phase, and/or during the remodeling (or late) wound healing phase.
  • the subject in need of wound healing is a human or an animal, for example, a dog, a cat, a horse, a pig, or a rodent, such as a mouse.
  • the compounds and compositions described herein useful for wound healing are administered topically, for example, proximate to the wound site, or systemically.
  • a therapeutically effective amount of a compound or composition described herein can be administered (optionally in combination with other agents) to the wound site by coating the wound or applying a bandage, packing material, stitches, etc., that are coated or treated with the compound or composition described herein.
  • the compounds and compositions described herein can be formulated for topical administration to treat surface wounds.
  • Topical formulations include those for delivery via the mouth (buccal) and to the skin such that a layer of skin (i. e., the epidermis, dermis, and/or subcutaneous layer) is contacted with the compound or composition described herein.
  • Topical delivery systems may be used to administer topical formulations of the compounds and compositions described herein.
  • the compounds and compositions described herein can be administered at or near the wound site by, for example, injection of a solution, injection of an extended release formulation, or introduction of a biodegradable implant comprising the compound or composition described herein.
  • the compounds and compositions described herein can be used to treat acute wounds or chronic wounds.
  • a chronic wound results when the normal reparative process is interrupted.
  • Chronic wounds can develop from acute injuries as a result of unrecognized persistent infections or inadequate primary treatment.
  • chronic lesions are the end stage of progressive tissue breakdown owing to venous, arterial, or metabolic vascular disease, pressure sores, radiation damage, or tumors.
  • Chronic wounds include, but are not limited to the following: chronic ischemic skin lesions; scleroderma ulcers; arterial ulcers; diabetic foot ulcers; pressure ulcers; venous ulcers; non-healing lower extremity wounds; ulcers due to inflammatory conditions; and/or long-standing wounds.
  • Other examples of chronic wounds include chronic ulcers, diabetic wounds, wounds caused by diabetic neuropathy, venous insufficiencies, and arterial insufficiencies, and pressure wounds and cold and warm burns.
  • Yet other examples of chronic wounds include chronic ulcers, diabetic wounds, wounds caused by diabetic neuropathy, venous insufficiencies, arterial insufficiencies, and pressure wounds.
  • Acute wounds include, but are not limited to, post-surgical wounds, lacerations, hemorrhoids and fissures.
  • the compounds and compositions described herein can be used for diabetic wound healing or accelerating healing of leg and foot ulcers secondary to diabetes or ischemia in a subject.
  • the wound is a surface wound.
  • the wound is a surgical wound (e.g., abdominal or gastrointestinal surgical wound).
  • the wound is a burn.
  • the wound is the result of radiation exposure.
  • the compounds and compositions described herein can also be used for diabetic wound healing, gastrointestinal wound healing, or healing of an adhesion due, for example, to an operation.
  • the compounds and compositions described herein can also be used to heal wounds that are secondary to another disease.
  • inflammatory skin diseases such as psoriasis and dermatitis
  • the compounds and compositions described herein can be used to heal wounds that are secondary to these diseases, for example, inflammatory skin diseases, such as psoriasis and dermatitis.
  • the wound is an internal wound.
  • the internal wound is a chronic wound.
  • the wound is a vascular wound.
  • the internal wound is an ulcer.
  • Examples of internal wounds include, but are not limited to, fistulas and internal wounds associated with cosmetic surgery, internal indications, Crohn's disease, ulcerative colitis, internal surgical sutures and skeletal fixation.
  • Other examples of internal wounds include, but are not limited to, fistulas and internal wounds associated with cosmetic surgery, internal indications, internal surgical sutures and skeletal fixation.
  • wounds include, but are not limited to, abrasions, avulsions, blowing wounds (i.e., open pneumothorax), burn wounds, contusions, gunshot wounds, incised wounds, open wounds, penetrating wounds, perforating wounds, puncture wounds, seton wounds, stab wounds, surgical wounds, subcutaneous wounds, diabetic lesions, or tangential wounds.
  • blowing wounds i.e., open pneumothorax
  • burn wounds i.e., contusions, gunshot wounds, incised wounds, open wounds, penetrating wounds, perforating wounds, puncture wounds, seton wounds, stab wounds, surgical wounds, subcutaneous wounds, diabetic lesions, or tangential wounds.
  • wounds that can be treated by the compounds and compositions described herein include acute conditions or wounds, such as thermal burns, chemical burns, radiation burns, burns caused by excess exposure to ultraviolet radiation ⁇ e.g., sunburn); damage to bodily tissues, such as the perineum as a result of labor and childbirth; injuries sustained during medical procedures, such as episiotomies; trauma- induced injuries including cuts, incisions, excoriations; injuries sustained from accidents; post-surgical injuries, as well as chronic conditions, such as pressure sores, bedsores, conditions related to diabetes and poor circulation, and all types of acne.
  • acute conditions or wounds such as thermal burns, chemical burns, radiation burns, burns caused by excess exposure to ultraviolet radiation ⁇ e.g., sunburn
  • damage to bodily tissues such as the perineum as a result of labor and childbirth
  • injuries sustained during medical procedures such as episiotomies
  • trauma- induced injuries including cuts, incisions, excoriations
  • injuries sustained from accidents post-surgical injuries,
  • the wound can include dermatitis, such as impetigo, intertrigo, folliculitis and eczema, wounds following dental surgery; periodontal disease; wounds following trauma; and tumor- associated wounds.
  • dermatitis such as impetigo, intertrigo, folliculitis and eczema
  • wounds following dental surgery periodontal disease
  • wounds following trauma and tumor- associated wounds.
  • wounds include animal bites, arterial disease, insect stings and bites, bone infections, compromised skin/muscle grafts, gangrene, skin tears or lacerations, skin aging, surgical incisions, including slow or non-healing surgical wounds, intracerebral hemorrhage, aneurysm, dermal asthenia, and post-operation infections.
  • the wound is selected from the group consisting of a burn wound, an incised wound, an open wound, a surgical or post surgical wound, a diabetic lesion, a thermal burn, a chemical burn, a radiation burn, a pressure sore, a bedsore, and a condition related to diabetes or poor circulation.
  • the wound is selected from the group consisting of an incised wound, an open wound, a surgical or post surgical wound, a diabetic lesion, a pressure sore, a bedsore, and a condition or wound related to diabetes or poor circulation.
  • the wound is selected from the group consisting of a non- radiation burn wound, an incised wound, an open wound, a surgical or post surgical wound, a diabetic lesion, a thermal burn, a chemical burn, a pressure sore, a bedsore, and a condition related to diabetes or poor circulation.
  • the wound is selected from the group consisting of an incised wound, an open wound, a surgical or post surgical wound, a diabetic lesion, a pressure sore, a bedsore, and a condition related to diabetes or poor circulation.
  • the present disclosure also relates to methods and compositions of reducing scar formation during wound healing in a subject.
  • the compounds and compositions described herein can be administered directly to the wound or to cells proximate the wound at an amount effective to reduce scar formation in and/or around the wound.
  • a method of reducing scar formation during wound healing in a subject comprising administering to the subject a therapeutically effective amount of a compound described herein (e.g., a CRM1 inhibitor), or a pharmaceutically acceptable salt thereof.
  • the wound can include any injury to any portion of the body of a subject.
  • methods are provided to ameliorate, reduce, or decrease the formation of scars in a subject that has suffered a burn injury. According to preferred embodiments, methods are provided to treat, reduce the occurrence of, or reduce the probability of developing hypertrophic scars in a subject that has suffered an acute or chronic wound or injury.
  • Compounds and compositions described herein may also be used to treat disorders of abnormal tissue growth and fibrosis including dilative cardiomyopathy, hypertrophic cardiomyopathy, restrictive cardiomyopathy, pulmonary fibrosis, hepatic fibrosis, glomerulonephritis, and other renal disorders.
  • Radiation therapy is the medical use of high-energy radiation (e.g. , x-rays, gamma rays, charged particles) to shrink tumors and kill malignant cells, and is generally used as part of cancer treatment. Radiation therapy kills malignant cells by damaging their DNA.
  • high-energy radiation e.g. , x-rays, gamma rays, charged particles
  • Radiation therapy can be delivered to a patient in several ways.
  • radiation can be delivered from an external source, such as a machine outside the patient's body, as in external beam radiation therapy.
  • External beam radiation therapy for the treatment of cancer uses a radiation source that is external to the patient, typically either a radioisotope, such as 60 Co, 137 Cs, or a high energy x-ray source, such as a linear accelerator.
  • the external source produces a collimated beam directed into the patient to the tumor site.
  • External-source radiation therapy avoids some of the problems of internal- source radiation therapy, but it undesirably and necessarily irradiates a significant volume of non-tumorous or healthy tissue in the path of the radiation beam along with the tumorous tissue.
  • the adverse effect of irradiating of healthy tissue can be reduced, while maintaining a given dose of radiation in the tumorous tissue, by projecting the external radiation beam into the patient at a variety of "gantry" angles with the beams converging on the tumor site.
  • the particular volume elements of healthy tissue, along the path of the radiation beam, change, reducing the total dose to each such element of healthy tissue during the entire treatment.
  • the irradiation of healthy tissue also can be reduced by tightly collimating the radiation beam to the general cross section of the tumor taken perpendicular to the axis of the radiation beam.
  • Numerous systems exist for producing such a circumferential collimation some of which use multiple sliding shutters which, piecewise, can generate a radio-opaque mask of arbitrary outline.
  • the amount can be at least about 1 Gray (Gy) fractions at least once every other day to a treatment volume.
  • the radiation is administered in at least about 2 Gray (Gy) fractions at least once per day to a treatment volume.
  • the radiation is administered in at least about 1 Gray (Gy) fractions at least once per day to a treatment volume.
  • At least about 2 Gray (Gy) fractions at least once per day to a treatment volume for five consecutive days per week.
  • radiation is administered in 10 Gy fractions every other day, three times per week to a treatment volume.
  • a total of at least about 20 Gy is administered to a patient in need thereof.
  • at least about 30 Gy is administered to a patient in need thereof.
  • at least about 40 Gy is
  • the patient receives external beam therapy four or five times a week.
  • An entire course of treatment usually lasts from one to seven weeks depending on the type of cancer and the goal of treatment. For example, a patient can receive a dose of 2 Gy/day over 30 days.
  • Internal radiation therapy is localized radiation therapy, meaning the radiation source is placed at the site of the tumor or affected area.
  • Internal radiation therapy can be delivered by placing a radiation source inside or next to the area requiring treatment.
  • Internal radiation therapy is also called brachytherapy.
  • Brachytherapy includes intercavitary treatment and interstitial treatment.
  • intracavitary treatment containers that hold radioactive sources are put in or near the tumor. The sources are put into the body cavities.
  • interstitial treatment the radioactive sources alone are put into the tumor. These radioactive sources can stay in the patient permanently. Typically, the radioactive sources are removed from the patient after several days. The radioactive sources are in containers.
  • the radiopharmaceutical agent can be administered by targeted delivery or by systemic delivery of targeted radioactive conjugates, such as a radiolabeled antibody, a radiolabeled peptide and a liposome delivery system.
  • targeted delivery the radiolabeled pharmaceutical agent can be a radiolabeled antibody. See, for example, Ballangrud A. M., et al. Cancer Res., 2001 ; 61 :2008-2014 and Goldenber, D.M. J Nucl. Med. , 2002; 43(5):693-713, the contents of which are incorporated by reference herein.
  • the radiopharmaceutical agent can be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or
  • the radiolabeled pharmaceutical agent can be a radiolabeled peptide.
  • a radiolabeled peptide See, for example, Weiner RE, Thakur ML. Radiolabeled peptides in the diagnosis and therapy of oncological diseases. Appl Radiat Isot 2002 Nov;57(5):749-63, the contents of which are incorporated by reference herein.
  • bracytherapy can be used to deliver the radiopharmaceutical agent to the target site.
  • Brachytherapy is a technique that puts the radiation sources as close as possible to the tumor site. Often the source is inserted directly into the tumor.
  • the radioactive sources can be in the form of wires, seeds or rods. Generally, cesium, iridium or iodine are used.
  • Systemic radiation therapy is another type of radiation therapy and involves the use of radioactive substances in the blood.
  • Systemic radiation therapy is a form of targeted therapy.
  • a patient typically ingests or receives an injection of a radioactive substance, such as radioactive iodine or a radioactive substance bound to a monoclonal antibody.
  • a "radiopharmaceutical agent,” as defined herein, refers to a pharmaceutical agent which contains at least one radiation- emitting radioisotope. Radiopharmaceutical agents are routinely used in nuclear medicine for the diagnosis and/or therapy of various diseases.
  • the radiolabeled pharmaceutical agent for example, a radiolabeled antibody, contains a radioisotope (RI) which serves as the radiation source.
  • RI radioisotope
  • the term "radioisotope” includes metallic and non-metallic radioisotopes. The radioisotope is chosen based on the medical application of the radiolabeled pharmaceutical agents.
  • the radioisotope is a metallic radioisotope
  • a chelator is typically employed to bind the metallic radioisotope to the rest of the molecule.
  • the radioisotope is a non-metallic radioisotope
  • the non-metallic radioisotope is typically linked directly, or via a linker, to the rest of the molecule.
  • a "metallic radioisotope” is any suitable metallic radioisotope useful in a therapeutic or diagnostic procedure in vivo or in vitro.
  • Suitable metallic radioisotopes include, but are not limited to: Actinium-225, Antimony-124, Antimony- 125, Arsenic-74, Barium- 103, Barium- 140, Beryllium-7, Bismuth-206, Bismuth-207, Bismuth212, Bismuth213, Cadmium-109, Cadmium-115m, Calcium-45, Cerium-139, Cerium-141, Cerium-144, Cesium-137, Chromium-51, Cobalt-55, Cobalt-56, Cobalt-57, Cobalt-58, Cobalt-60, Cobalt-64, Copper-60, Copper-62, Copper-64, Copper-67, Erbium- 169,
  • a "non-metallic radioisotope” is any suitable nonmetallic radioisotope (non-metallic radioisotope) useful in a therapeutic or diagnostic procedure in vivo or in vitro.
  • Suitable non-metallic radioisotopes include, but are not limited to: Iodine- 131, Iodine-125, Iodine-123, Phosphorus-32, Astatine-211, Fluorine-18, Carbon-11, Oxygen- 15, Bromine-76, and Nitrogen-13.
  • Identifying the most appropriate isotope for radiotherapy requires weighing a variety of factors. These include tumor uptake and retention, blood clearance, rate of radiation delivery, half-life and specific activity of the radioisotope, and the feasibility of large-scale production of the radioisotope in an economical fashion.
  • the key point for a therapeutic radiopharmaceutical is to deliver the requisite amount of radiation dose to the tumor cells and to achieve a cytotoxic or tumoricidal effect while not causing unmanageable side-effects.
  • the physical half-life of the therapeutic radioisotope be similar to the biological half-life of the radiopharmaceutical at the tumor site. For example, if the half-life of the radioisotope is too short, much of the decay will have occurred before the radiopharmaceutical has reached maximum target/background ratio. On the other hand, too long a half-life could cause unnecessary radiation dose to normal tissues. Ideally, the radioisotope should have a long enough half-life to attain a minimum dose rate and to irradiate all the cells during the most radiation sensitive phases of the cell cycle. In addition, the half-life of a radioisotope has to be long enough to allow adequate time for
  • the target receptor sites in tumors are typically limited in number. As such, it is preferred that the radioisotope have high specific activity. The specific activity depends primarily on the production method. Trace metal contaminants must be minimized as they often compete with the radioisotope for the chelator and their metal complexes compete for receptor binding with the radiolabeled chelated agent.
  • the type of radiation that is suitable for use in the methods of the present invention can vary.
  • radiation can be electromagnetic or particulate in nature.
  • Electromagnetic radiation useful in the practice of this invention includes, but is not limited to, x-rays and gamma rays.
  • Particulate radiation useful in the practice of this invention includes, but is not limited to, electron beams (beta particles), protons beams, neutron beams, alpha particles, and negative pi mesons.
  • the radiation can be delivered using conventional radiological treatment apparatus and methods, and by intraoperative and stereotactic methods. Additional discussion regarding radiation treatments suitable for use in the practice of this invention can be found throughout Steven A. Leibel et al., Textbook of Radiation Oncology (1998) (publ. W.
  • Radiation can also be delivered by other methods such as targeted delivery, for example by radioactive "seeds," or by systemic delivery of targeted radioactive conjugates.
  • Alpha particles are particularly good cytotoxic agents because they dissipate a large amount of energy within one or two cell diameters.
  • the ⁇ -particle emitters have relatively long penetration range (2-12 mm in the tissue) depending on the energy level. The long-range penetration is particularly important for solid tumors that have heterogeneous blood flow and/or receptor expression.
  • the ⁇ -particle emitters yield a more homogeneous dose distribution even when they are heterogeneously distributed within the target tissue.
  • therapeutically effective amounts of the compounds and compositions described herein are administered in combination with a therapeutically effective amount of radiation therapy to treat cancer ⁇ e.g., lung cancer, such as non-small cell lung cancer).
  • a therapeutically effective amount of radiation therapy to treat cancer ⁇ e.g., lung cancer, such as non-small cell lung cancer.
  • the amount of radiation necessary can be determined by one of skill in the art based on known doses for a particular type of cancer. See, for example, Cancer Medicine 5 th ed., Edited by R.C. Bast et al, July 2000, BC Decker.
  • Example 4 Assays. Certain compounds of the invention were tested in various assays. MTT Cell Proliferation Assay
  • the MTT cell proliferation assay was used to study the cytotoxic properties of the compounds.
  • the assay was performed according to the method described by Roche
  • the assay is based on the cleavage of the tetrazolium salt, MTT, in the presence of an electron-coupling reagent.
  • the water-insoluble formazan salt produced must be solubilized in an additional step.
  • Cells grown in a 96-well tissue culture plate were incubated with the MTT solution for approximately 4 hours. After this incubation period, a water-insoluble formazan dye formed. After solubilization, the formazan dye was quantitated using a scanning multi-well spectrophotometer (ELISA reader). The absorbance revealed directly correlates to the cell number.
  • the cells were seeded at 5,000-10,000 cells in each well of 96-well plate in 100 ⁇ iL of fresh culture medium and were allowed to attach overnight.
  • the stock solutions of the compounds were diluted in 100xL cell culture medium to obtain eight concentrations of each test compound, ranging from 1 nM to 30 ⁇ .
  • 20 xL of CellTiter 96 Aqueous One Solution Reagent (Promega, G358B) was added to each well and the plate was returned to the incubator (37 °C; 5% C0 2 ) until an absolute OD of 1.5 was reached for the control cells. All optical densities were measured at 490 run using a Vmax Kinetic Microplate Reader (Molecular Devices).
  • Inhibition (%) (1- (OD o /OD)) X 100.
  • the compounds were tested against Zl 38, MMl S and 3T3 cells.
  • the Zl 38 cell line is a mature B-cell acute lymphoblastic leukemia cell line derived from a patient with chronic lumphocytic leukemia.
  • the MMl S cell line was established from the peripheral blood of a human multiple myeloma patient.
  • 3T3 cells are standard fibroblast cells; they were originally isolated from Swiss mouse embryo tissue.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to compounds of Structural Formulas (I), (II) and (III): or their pharmaceutically acceptable salts; pharmaceutical compositions comprising the compounds of Structural Formulas (I), (II) and (III); and methods of using said compounds, salts and compositions in the treatment of various disorders associated with CRMl activity. The values for the variables in Structural Formulas (I), (II) and (III) are as described and defined herein.

Description

NUCLEAR TRANSPORT MODULATORS AND USES THEREOF
RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application No.
61/838,129, filed on June 21, 2013; U.S. Provisional Application No. 61/838,135, filed on June 21, 2013; U.S. Provisional Application No. 61/838,153, filed on June 21, 2013; and U.S. Provisional Application No. 61/842,475, filed on July 3, 2013. The entire teachings of these applications are incorporated herein by reference.
BACKGROUND OF THE INVENTION
[0002] Cells from most maj or human solid and hematologic malignancies exhibit abnormal cellular localization of a variety of oncogenic proteins, tumor suppressor proteins, and cell cycle regulators (Cronshaw et al, 2004, Falini et al 2006). For example, certain p53 mutations lead to localization in the cytoplasm rather than in the nucleus. This results in the loss of normal growth regulation, despite intact tumor suppressor function. In other tumors, wild-type p53 is sequestered in the cytoplasm or rapidly degraded, again leading to loss of its suppressor function. Restoration of appropriate nuclear localization of functional p53 protein can normalize some properties of neoplastic cells (Cai et al, 2008; Hoshino et al 2008; Lain et al 1999a; Lain et al 1999b; Smart et al 1999), can restore sensitivity of cancer cells to DNA damaging agents (Cai et al, 2008), and can lead to regression of established tumors (Sharpless & DePinho 2007, Xue et al, 2007). Similar data have been obtained for other tumor suppressor proteins such as forkhead (Turner and Sullivan 2008) and c-Abl (Vignari and Wang 2001). In addition, abnormal localization of several tumor suppressor and growth regulatory proteins may be involved in the pathogenesis of autoimmune diseases (Davis 2007, Nakahara 2009). CRM1 inhibition may provide particularly interesting utility in familial cancer syndromes (e.g., Li-Fraumeni Syndrome due to loss of one p53 allele, BRCA1 or 2 cancer syndromes), where specific tumor suppressor proteins (TSP) are deleted or dysfunctional and where increasing TSP levels by systemic (or local) administration of CRM1 inhibitors could help restore normal tumor suppressor function.
[0003] Specific proteins and RNAs are carried into and out of the nucleus by specialized transport molecules, which are classified as importins if they transport molecules into the nucleus, and exportins if they transport molecules out of the nucleus (Terry et al, 2007; Sorokin et al 2007). Proteins that are transported into or out of the nucleus contain nuclear import/localization (NLS) or export (NES) sequences that allow them to interact with the relevant transporters. Chromosomal Region Maintenance 1 (Crml), which is also called exportin-1 or Xpol , is a major exportin.
[0004] Overexpression of Crml has been reported in several tumors, including human ovarian cancer (Noske et al, 2008), cervical cancer (van der Watt et al, 2009), pancreatic cancer (Huang et al, 2009), hepatocellular carcinoma (Pascale et al, 2005) and osteosarcoma (Yao et al, 2009) and is independently correlated with poor clinical outcomes in these tumor types.
[0005] Inhibition of Crml blocks the exodus of tumor suppressor proteins and/or growth regulators such as p53, c-Abl, p21, p27, pRB, BRCA1 , IkB, ICp27, E2F4, KLF5, YAP1, ZAP, KLF5, HDAC4, FID AC 5 or forkhead proteins (e.g. FOX03a) from the nucleus that are associated with gene expression, cell proliferation, angiogenesis and epigenetics. Crml inhibitors have been shown to induce apoptosis in cancer cells even in the presence of activating oncogenic or growth stimulating signals, while sparing normal (untransformed) cells. Most studies of Crml inhibition have utilized the natural product Crml inhibitor Leptomycin B (LMB). LMB itself is highly toxic to neoplastic cells, but poorly tolerated with marked gastrointestinal toxicity in animals (Roberts et al, 1986) and humans (Newlands et al, 1996). Derivatization of LMB to improve drug-like properties leads to compounds that retain antitumor activity and are better tolerated in animal tumor models (Yang et al, 2007, Yang et al, 2008, Mutka et al, 2009). Therefore, nuclear export inhibitors could have beneficial effects in neoplastic and other proliferative disorders. To date, however, small- molecule, drug-like Crml inhibitors for use in vitro and in vivo are uncommon.
[0006] In addition to tumor suppressor proteins, Crml also exports several key proteins that are involved in many inflammatory processes. These include IkB, NF-kB, Cox-2, RXRa, Commdl, HIFl , HMGBl, FOXO, FOXP and others. The nuclear factor kappa B (NF-kB/rel) family of transcriptional activators, named for the discovery that it drives immunoglobulin kappa gene expression, regulate the mRNA expression of variety of genes involved in inflammation, proliferation, immunity and cell survival. Under basal conditions, a protein inhibitor of NF-kB, called IkB, binds to NF-kB in the nucleus and the complex IkB-NF-kB renders the NF-kB transcriptional function inactive. In response to inflammatory stimuli, IkB dissociates from the IkB-NF-kB complex, which releases NF-kB and unmasks its potent transcriptional activity. Many signals that activate NF-kB do so by targeting IkB for proteolysis (Phosphorylation of IkB renders it "marked" for ubiquitination and then proteolysis). The nuclear IkBa-NF-kB complex can be exported to the cytoplasm by Crml where it dissociates and NF-kB can be reactivated. Ubiquitinated IkB may also dissociate from the NF-kB complex, restoring NF-kB transcriptional activity. Inhibition of Crml induced export in human neutrophils and macrophage like cells (U937) by LMB not only results in accumulation of transcriptionally inactive, nuclear IkBa-NF-kB complex but also prevents the initial activation of NF-kB even upon cell stimulation (Ghosh 2008, Huang 2000). In a different study, treatment with LMB inhibited IL-Ιβ induced NF-kB DNA binding (the first step in NF-kB transcriptional activation), IL-8 expression and intercellular adhesion molecule expression in pulmonary microvascular endothelial cells (Walsh 2008). COMMDl is another nuclear inhibitor of both NF-kB and hypoxia- inducible factor 1 (HIFl) transcriptional activity. Blocking the nuclear export of COMMDl by inhibiting Crml results in increased inhibition of NF-kB and HIFl transcriptional activity (Muller 2009).
[0007] Crml also mediates Retinoid X receptor a (RXRa) transport. RXRa is highly expressed in the liver and plays a central role in regulating bile acid, cholesterol, fatty acid, steroid and xenobiotic metabolism and homeostasis. During liver inflammation, nuclear RXRa levels are significantly reduced, mainly due to inflammation-mediated nuclear export of RXRa by Crml. Lep B is able to prevent IL-Ιβ induced cytoplasmic increase in RXRa levels in human liver derived cells (Zimmerman 2006).
[0008] The role of Crml -mediated nuclear export in NF-kB, HIF- 1 and RXRa signalling suggests that blocking nuclear export can be potentially beneficial in many inflammatory processes across multiple tissues and organs including the vasculature (vasculitis, arteritis, polymyalgia rheumatic, atherosclerosis), dermatologic (see above), rheumatologic
(rheumatoid and related arthritis, psoriatic arthritis, spondyloarthropathies, crystal arthropathies, systemic lupus erythematosus, mixed connective tissue disease, myositis syndromes, dermatomyositis, inclusion body myositis, undifferentiated connective tissue disease, Sjogren's syndrome, scleroderma and overlap syndromes, etc.).
[0009] CRM1 Inhibition affects gene expression by inhibiting/activating a series of transcription factors like ICp27, E2F4, KLF5, YAP1, ZAP
[0010] Crml inhibition has potential therapeutic effects across many dermatologic syndromes including inflammatory dermatoses (atopy, allergic dermatitis, chemical dermatitis, psoriasis), sun-damage (Ultraviolet / UV damage), and infections. CRMl inhibition, best studied with LMB, showed minimal effects on normal keratinocytes, and exerted anti-inflammatory activity on keratinocytes subjected to UV, TNFa, or other inflammatory stimuli (Kobayashi & Shinkai 2005, Kannan & Jaiswal 2006). Crml inhibition also upregulates NRF2 (nuclear factor erythroid-related factor 2) activity, which protects keratinocytes (Schafer et al, 2010, Kannan & Jaiswal 2006) and other cell types (Wang et al, 2009) from oxidative damage. LMB induces apoptosis in keratinocytes infected with oncogenic human papillomavirus (HPV) strains such as HPV16, but not in uninfected keratinocytes (Jolly et al, 2009).
[0011] Crml also mediates the transport of key neuroprotectant proteins that may be useful in neurodegenerative diseases including Parkinson's Disease (PD), Alzheimer's Disease, and Amyotrophic Lateral Sclerosis. For example, (1) forcing nuclear retention of key neuroprotective regulators such as NRF2 (Wang 2009), FOXA2 (Kittappa et al, 2007), parking in neuronal cells and/or by (2) inhibiting NFKB transcriptional activity by
sequestering ΙκΒ to the nucleus in glial cells, Crml inhibition could slow or prevent neuronal cell death found in these disorders. There is also evidence linking abnormal glial cell proliferation to abnormalities in CRMl levels or CR1 function (Shen 2008).
[0012] Intact nuclear export, primarily mediated through CRMl, is also required for the intact maturation of many viruses. Viruses where nuclear export, and/or CRMl itself, has been implicated in their lifecycle include human immunodeficiency virus (HIV), adenovirus, simian retrovirus type 1, Borna disease virus, influenza (usual strains as well as H1N1 and avian H5N1 strains), hepatitis B (HBV) and C (HCV) viruses, human papillomavirus (HPV), respiratory syncytial virus (RSV), Dungee, Severe Acute Respiratory Syndrome coronavirus, yellow fever virus, West Nile Virus, herpes simplex virus (HSV), cytomegalovirus (CMV), and Merkel cell polyomavirus (MCV). (Bhuvanakantham 2010, Cohen 2010, Whittaker 1998). It is anticipated that additional viral infections reliant on intact nuclear export will be uncovered in the near future.
[0013] The HIV-1 Rev protein, which traffics through nucleolus and shuttles between the nucleus and cytoplasm, facilitates export of unspliced and singly spliced HIV transcripts containing Rev Response Elements (RRE) RNA by the CRMl export pathway. Inhibition of Rev-mediated RNA transport using CRMl inhibitors such as LepB or PKF050-638 can arrest the HIV-1 transcriptional process, inhibit the production of new HIV-1 virions, and thereby reduce HIV-1 levels (Pollard 1998, Daelemans 2002).
[0014] Dengue virus (DENV) is the causative agent of the common arthropod-borne viral disease, dengue fever (DF), and its more severe and potentially deadly dengue hemorrhagic fever (DHF). DHF appears to be the result of an over exuberant inflammatory response to DENV. NS5 is the largest and most conserved protein of DENV. CRMl regulates the transport of NS5 from the nucleus to the cytoplasm, where most of the NS5 functions are mediated. Inhibition of CRMl mediated export of NS5 results in altered kinetics of virus production and reduces induction of the inflammatory chemokine interleukin-8 (IL-8), presenting a new avenue for the treatment of diseases caused by DENV and other medically important flaviviruses including Hepatitis C virus (Rawlinson 2009).
[0015] Other virus-encoded RNA-binding proteins that use CRMl to exit the nucleus include the HSV type 1 tegument protein (VP 13/14, or hUL47), human CMV protein pp65, the SARS Coronavirus ORF 3b Protein, and the RSV matrix (M) protein (Williams 2008, Sanchez 2007, Freundt 2009, Ghildyal 2009).
[0016] Interestingly, many of these viruses are associated with specific types of human cancer including hepatocellular carcinoma (HCC) due to chronic HBV or HCV infection, cervical cancer due to HPV, and Merkel cell carcinoma associated with MCV. CRMl inhibitors could therefore have beneficial effects on both the viral infectious process as well as on the process of neoplastic transformation due to these viruses.
[0017] CRMl controls the nuclear localization and therefore activity of multiple DNA metabolizing enzymes including histone deacetylases (HDAC), histone acetyltransferases (HAT), and histone methyltransferases (HMT). Suppression of cardiomyocyte hypertrophy with irreversible CRMl inhibitors has been demonstrated and is believed to be linked to nuclear retention (and activation) of HDAC 5, an enzyme known to suppress a hypertrophic genetic program (Monovich et al, 2009). Thus, CRMl inhibition may have beneficial effects in hypertrophic syndromes, including certain forms of congestive heart failure and hypertrophic cardiomyopathies.
[0018] CRMl has also been linked to other disorders. Leber's disorder, a hereditary disorder characterized by degeneration of retinal ganglion cells and visual loss, is associated with inaction of the CRMl switch (Gupta N 2008). There is also evidence linking
neurodegenerative disorders to abnormalities in nuclear transport. [0019] In view of the above, the discovery of compounds that modulate nuclear transport is desirable.
SUMMARY OF THE INVENTION
[0020] The present invention relates to compounds, and pharmaceutically acceptable salts thereof, useful as nuclear transport modulators; pharmaceutically acceptable compositions comprising compounds of the present invention or their pharmaceutically acceptable salts; and methods of using said compounds, salts and compositions in the treatment of various disorders.
[0021 ] In one embodiment, the compounds of the invention have general formula I:
Figure imgf000007_0001
wherein each variable is as defined and described herein.
[0022] In another embodiment, the compounds of the invention have general formula II:
Figure imgf000007_0002
wherein each variable is as defined and described herein.
[0023] In yet another embodiment, the compounds of the invention have general formula III:
Figure imgf000007_0003
wherein each variable is as defined and described herein.
[0024] Compounds of the present invention (compounds of Structural Formula I, Structural Formula II, Structural Formula III, etc.) and pharmaceutically acceptable salts and compositions thereof are useful for treating a variety of diseases, disorders or conditions associated with abnormal cellular responses triggered by improper nuclear transport.
Therefore, one embodiment of the invention is use of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treating a variety of diseases, disorders or conditions associated with abnormal cellular responses triggered by improper nuclear transport.
[0025] Another embodiment of the invention is a method for treating a variety of diseases, disorders or conditions associated with CRM1 activity in a subject in need thereof, the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt or composition thereof. Such diseases, disorders, or conditions include those described herein.
[0026] Compounds of the invention, and pharmaceutically acceptable salts thereof, are also useful in the manufacture of a medicament for the treatment of a variety of diseases, disorders or conditions associated with abnormal cellular responses triggered by improper nuclear transport. Such diseases, disorders, or conditions include those described herein.
[0027] Compounds provided by this invention are also useful for the study of nuclear transport modulation in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated, for example, by kinases; and the comparative evaluation of new nuclear transport modulators.
DETAILED DESCRIPTION OF THE INVENTION
Compounds of the Invention
[0028] Values for the variables in Structural Formulas I, II and III are as described in the sections directed to compounds of Structural Formulas I, II and III, respectively. For example, R1, R2, Ra, Rb, X and n in Structural Formula I are as described in the section below entitled, "Compounds of Structural Formula I." R1, R2, Ra, X and n in Structural Formula II are as described in the section entitled, "Compounds of Structural Formula II," and R , R , Ra, X and n in Structural Formula III are as described in the section entitled, "Compounds of Structural Formula III."
[0029] The numbering of the embodiments in each of the sections directed to compounds of Structural Formulas I, II and III, as well as cross-references to the numbering of the embodiments, is made with respect to that particular section. For example, each of the sections directed to compounds of Structural Formulas I, II and III begins with a first embodiment. Thus, a statement in a particular section (e.g. , "Compounds of Structural Formula Π"), such as a statement that the values for the remaining variables are as described in the first embodiment, or any aspect thereof, refers only to the embodiment of the identified number (e.g, the first embodiment) in the same section (e.g., "Compounds of Structural Formula II").
Compounds of Structural Formula I
[0030] A first embodiment of a compound of Structural Formula I is a compound of Structural Formula I:
Figure imgf000009_0001
or a pharmaceutically acceptable salt thereof, wherein:
X is -N- or -C(H)-;
Ra and Rb are each independently halo or hydrogen, wherein at least one of Ra and Rb is halo;
each R1 is independently selected from halo; haloalkyl; -(CH2)i-4R°; -(CH2)o-4OR°; -0-(CH2)o-4C(0)OR°; -(CH2)0-4CH(OR°)2; -(CH2)o-4SR°; -(CH2)0-4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°;
-(CH2)o-4-heterocyclyl, which may be substituted with R°; -(CH2)0-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°; -CH=CH-heterocyclyl, which may be substituted with R°; -CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3; -(CH2)0-4N(R°)2; -(CH2)o-4N(R0)C(0)R0 ; -(CH2)o-4N(R0)C(S)R°; -(CH2)0-4N(R°)C(O)NR°2;
-(CH2)o-4N(R0)C(S)NR0 2; -(CH2)o-4N(R0)C(0)OR0; -(CH2)o-4N(R0)N(R°)C(0)R°;
-(CH2)o-4N(R0)N(R0)C(0)NR°2; -(CH2)0-4N(Ro)N(Ro)C(O)OR°; -(CH2)0-4C(O)R°;
-(CH2)o-4C(S)R°; -(CH2)o-4C(0)OR°; -(CH2)0-4C(O)SR°; -(CH2)0-4OC(O)R°;
-(CH2)0-4OC(0)(CH2)o-4SR°, -(CH2)0-4SC(S)SR°; -(CH2)0-4SC(O)R°; -(CH2)o-4C(0)NR0 2; -(CH2)o.4C(S)NR°2; -(CH2)0-4C(S)SR°; -(CH2)0-4OC(O)NR°2; -(CH2)0-4C(O)N(ORo)R°; -(CH2)o-4C(0)C(0)R°; -(CH2)0-4C(O)CH2C(O)R°; -(CH2)0-4C(NOR°)R°; -(CH2)0-4SSR°; -(CH2)0-4S(O)2R°; -(CH2)o-4S(0)2OR°; -(CH2)0-4OS(O)2R°; -(CH2)0-4S(O)2NR°2;
-(CH2)0-4S(O)R°; -(CH2)0-4N(R°)S(O)2NR°2; -(CH2)0-4N(Ro)S(O)2R°; -(CH2)0-4N(OR°)R°;
-(CH2)0-4C(NH)NR°2; -(CH2)0-4P(O)2Ro; -(CH2)0-4P(O)R°2; -(CH2)0-4OP(O)R°2;
-(CH2)0-4OP(O)(OR°)2; -(CH2)0.4ON(R°)2; and -(CH2)0-4C(O)O-N(R°)2, wherein:
each R° is independently hydrogen, C1-6 aliphatic, -CH2-carbocyclyl,
-CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -O(CH2)0-i-carbocyclyl,
-0(CH2)o-i-aryl, -0(CH2)o-1-heterocyclyl, -0(CH2)o-i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Cj-Q alkyl, -NH2, -N02, -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted C1-C3 alkyl)2, -O-C1-C3 alkyl, -C(0)OH,
-C(0)0-(unsubstituted C1-C3 alkyl), -C(0)-(unsubstituted CrC3 alkyl),
-0-(unsubstituted C1-C3 alkyl), and -S-(unsubstituted C1-C3 alkyl);
R2 is selected from -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6),
-C(0)-N(R7)-N(R5)(R6), -C(S)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4,
-C(S)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-C(S)-R4, -C(S)-N(R7)-N(R7)-C(S)-R4,
-C(0)-N(R7)-N(R7)-S(0)1-2-R4, -C(S)-N(R7)-N(R7)-S(0)1-2-R4 and heteroaryl, wherein:
R3 is selected from C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(CrC4 alkyl)(C3-C6 cycloalkyl),
-Ci-C6 alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl, -(C0-C4 alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl,
C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or
R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
each R7 is independently hydrogen or C1-C alkyl; and n is 0, 1, 2, 3, 4 or 5; wherein unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
[0031] In a first aspect of the first embodiment, Ra and Rb are each independently halo. The values for the remaining variables are as described in the first embodiment.
[0032] In a second aspect of the first embodiment, Ra and Rb are the same. The values for the remaining variables are as described in the first embodiment, or first aspect thereof.
[0033] In a third aspect of the first embodiment, the halo of Ra and Rb is chlorine or bromine. The values for the remaining variables are as described in the first embodiment, or first or second aspect thereof.
[0034] In a fourth aspect of the first embodiment, Ra and Rb are the same and are each chlorine or bromine. The values for the remaining variables are as described in the first embodiment, or first through third aspects thereof.
[0035] In a fifth aspect of the first embodiment, X is -C(H)-. The values for the remaining variables are as described in the first embodiment, or first through fourth aspects thereof.
[0036] In a sixth aspect of the first embodiment, n is 0, 1 or 2. The values for the remaining variables are as described in the first embodiment, or first through fifth aspects thereof.
[0037] In a seventh aspect of the first embodiment, each R1 is independently selected from -CF3, -CN, halo, - OH, C1-C3 alkyl, C3-C6 cycloalkyl, C3-C12 heterocycloalkyl, halo-Q- C3 alkyl, -NH2, -N02, -NH(Ci-C3 alkyl), -N(d-C3 alkyl)(Ci-C3 alkyl), -C(0)OH,
-C(0)0-(Ci-C6 alkyl), -C(0)-(CrC3 alkyl), -0-(CrC3 alkyl), -0-(Ci-C3 haloalkyl), and -S-( Cj-C3 alkyl), or is absent. The values for the remaining variables are as described in the first embodiment, or first through sixth aspects thereof
[0038] In an eighth aspect of the first embodiment, each R1 is independently selected from halo, -Ci-C4 alkyl, -d-d haloalkyl and -0-Ci-C4 alkyl, or is absent. The values for the remaining variables are as described in the first embodiment, or first through seventh aspects thereof.
2 3
[0039] In a ninth aspect of the first embodiment, R is selected from -C(0)-0-R , -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-S(0)i-2-R4, and heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through eighth aspects thereof.
[0040] In a tenth aspect of the first embodiment, R2 is -C(0)-0-R3, and R3 is selected from optionally substituted C1-C4 alkyl and C2-C4 alkenyl; or R2 is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or R2 is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or R2 is -C(0)-NH-NH-C(0)-R4 or
-C(0)-NH-NH-S(0)1-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl; or R2 is optionally substituted C5-C6 heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through ninth aspects thereof.
[0041] In an eleventh aspect of the first embodiment, R2 is -C(0)-0-CH(CH3)2,
-C(0)-NH-NH-C(0)-C(CH3)3 or
Figure imgf000012_0001
. The values for the remaining variables are as described in the first embodiment, or first through tenth aspects thereof.
[0042] In a twelfth aspect of the first embodiment, each R7 is hydrogen. The values for the remaining variables are as described in the first embodiment, or first through eleventh aspects thereof.
[0043] In a thirteenth aspect of the first embodiment, R5 is selected from hydrogen and C1-C4 alkyl; and R6 is selected from C1-C4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through twelfth aspects thereof.
[0044] In a fourteenth aspect of the first embodiment, R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through thirteenth aspects thereof.
[0045] In a fifteenth aspect of the first embodiment, RJ is selected from optionally substituted C1-C4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through fourteenth aspects thereof. [0046] In a sixteenth aspect of the first embodiment, R4 is selected from -N(R8)(C3-C6 cycloalkyl), -C3-C6 alkyl, -(Co-Q alkylene)-heterocyclyl, and -(C0-Ci alkylene)-heteroaryl, wherein R8 is hydrogen or -C1-C4 alkyl; any alkyl or alkylene portion of R4 is optionally and independently substituted with one or more substituents selected from the group consisting of oxo and -N(R9)2, wherein each R9 is independently selected from hydrogen and Q-C4 alkyl; any heterocyclyl portion of R4 comprises at least one nitrogen atom in a ring, and is optionally substituted with one or more substituents selected from the group consisting of Q- C4 alkyl and oxo; and any heteroaryl portion of R4 comprises at least one nitrogen atom in a ring and is optionally substituted with one or more Ci-C4 alkyl. The values for the remaining variables are as described in the first embodiment, or first through fifteenth aspects thereof.
[0047] In a seventeenth aspect of the first embodiment, R is selected from -C(0)-0-R , -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, and heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through sixteenth aspects thereof.
[0048] In an eighteenth aspect of the first embodiment, R2 is -C(0)-0-R3, and R3 is selected from optionally substituted CrC4 alkyl; or R2 is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C3-C7 heterocyclyl; or R2 is -C(0)-NH-NH(R6), and R6 is an optionally substituted C5-C6 heteroaryl; or R2 is -C(0)-NH-NH-C(0)-R4 or
-C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-(C3- C7)heterocyclyl and -(C0-C4 alkylene)-(C5-C6)heteroaryl; or R2 is optionally substituted C5-C6 heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through seventeenth aspects thereof.
[0049] A second embodiment of a compound of Structural Formula I is a compound represented by Structural Formula la:
Figure imgf000013_0001
a pharmaceutically acceptable salt thereof, wherein: Rla and Rlb are each independently selected from halo; haloalkyl; -(CH2)i.4R°; -(CH2)0-4OR°; -O-(CH2)0-4C(O)OR°; -(CH2)0-4CH(OR°)2; -(CH2)0.4SR°;
-(CH2)o-4-carbocyclyl, which may be substituted with R°; -(CH2)0.4-aryl, which may be substituted with R°; -(CH2)0-4-heterocyclyl, which may be substituted with R°; -(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°;
-CH=CH-heterocyclyl, which may be substituted with R°; -CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3; -(CH2)o-4N(R0)2; -(CH2)0-4N(Ro)C(O)R°; -(CH2)0-4N(R°)C(S)R°; -(CH2)0.4N(Ro)C(O)NR°2; -(CH2)0- N(R°)C(S)NR°2;
-(CH2)o-4N(R0)C(0)OR0; -(CH2)o-4N(R0)N(R°)C(0)R0;
-(CH2)0- N(R°)N(Ro)C(O)NR°2; -(CH2)0-4N(Ro)N(Ro)C(O)OR°; -(CH2)0-4C(O)R°; -(CH2)o.4C(S)R°; -(CH2)0-4C(O)OR°; -(CH2)0-4C(O)SR°; -(CH2)0-4OC(O)R°;
-(CH2)o-4OC(0)(CH2)o-4SR°, -(CH2)0-4SC(S)SR°; -(CH2)0- SC(O)R°;
-(CH2)0- C(O)NR°2; -(CH2)0-4C(S)NRo 2; -(CH2)0.4C(S)SR°; -(CH2)0-4OC(O)NRo 2; -(CH2)o-4C(0)N(OR0)R0 ; -(CH2)o.4C(0)C(0)R°; -(CH2)0-4C(O)CH2C(O)R°;
-(CH2)0-4C(NOR°)R°; -(CH2)0- SSR°; -(CH2)0- S(O)2R°; -(CH2)0-4S(O)2OR°;
-(CH2)0-4OS(O)2; -(CH2)o- S(0)2NR°2; -(CH2)0-4S(O)Ro;
-(CH2)o-4N(R0)S(0)2NR0 2; -(CH2)0-4N(Ro)S(O)2R°; -(CH2)0-4N(ORo)R°;
-(CH2)0-4C(NH)NR°2; -(CH2)0.4P(O)2R°; -(CH2)0-4P(O)R°2; -(CH2)0-4OP(O)R°2;
-(CH2)0- OP(O)(OR°)2; -(CH2)0-4ON(R°)2; and -(CH2)0-4C(O)O-N(R°)2, wherein: each R° is independently hydrogen, Ci-6 aliphatic, -CH2-carbocyclyl,
-CH2-aryl, -CH2-heterocyclyl, -CH -heteroaryl, -O(CH2)0-i-carbocyclyl,
-0(CH2)o-i-aryl, -0(CH2)o-i -heterocyclyl, -O(CH2)0-i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Ci-C3 alkyl, -NH2, -N02, -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted C1 -C3 alkyl)2, -0-C,-C3 alkyl, -C(0)OH, -C(0)0-(unsubstituted C1-C3 alkyl), -C(0)-(unsubstituted C1-C3 alkyl), -0-(unsubstituted Q-Q3 alkyl), and -S-(unsubstituted Ci-C3 alkyl); and
m is 0 or 1.
The values for the remaining variables are as described in the first embodiment, or any aspect thereof.
[0050] In a first aspect of the second embodiment, Rl is -C1-C4 haloalkyl. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment.
lb
[0051] In a second aspect of the second embodiment, R is -C1-C4 haloalkyl. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first aspect thereof.
[0052] In a third aspect of the second embodiment, Rl is -CF3 and Rlb is -CF3. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first or second aspect thereof.
[0053] In a fourth aspect of the second embodiment, R is -C1-C4 haloalkyl or -0-C1-C4 alkyl or absent. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through third aspects thereof.
[0054] In a fifth aspect of the second embodiment, Rla is halo or -Cj-C4 haloalkyl. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fourth aspects thereof.
[0055] In a sixth aspect of the second embodiment, m is 1. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fifth aspects thereof.
[0056] A third embodiment of a compound of Structural Formula I is a compound represented by Structural Formu
Figure imgf000015_0001
(lb), or a pharmaceutically acceptable salt thereof, wherein the values for Ra, Rb and R2 are as described in the first or second embodiment, or any aspect thereof.
[0057] In a first aspect of the third embodiment, R2 is selected from -C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4,
7 7 4
-C(0)-N(R')-N(R')-S(0)i-2-R , and heteroaryl. The values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third embodiment.
[0058] In a second aspect of the third embodiment, Ra and Rb are each independently halo. The values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third embodiment, or first aspect thereof.
[0059] In a third aspect of the third embodiment, Ra and Rb are the same. The values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third embodiment, or first or second aspect thereof.
[0060] In a fourth aspect of the third embodiment, the halo of Ra and Rb is chlorine or bromine. The values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third embodiment, or first through third aspects thereof.
[0061] In a fifth aspect of the third embodiment, R2 is -C(0)-0-R3, and R3 is selected from optionally substituted Ci-C4 alkyl and C2-C4 alkenyl; or R2 is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or R2 is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or R2 is -C(0)-NH-NH-C(0)-R4 or
-C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl; or R2 is optionally substituted C5-C6 heteroaryl. The values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third embodiment, or first through fourth aspects thereof.
[0062] In a sixth aspect of the third embodiment, R2 is -C(0)-0-CH(CH3)2,
-C(0)-NH-NH-C(0)-C(CH3)3 or
Figure imgf000016_0001
. The values for the remaining variables are as described in the first or second embodiment, or any aspect thereof, or the third
embodiment, or first through fifth aspects thereof. [0063] A fourth embodiment of a compound of Structural Formula I is a compound of Structural Formula I, or a pharmaceutically acceptable salt thereof, wherein:
X is -N- or -C(H)-;
Ra and Rb are each independently halo or hydrogen, wherein at least one of Ra and Rb is halo;
each R1 is independently selected from halo; haloalkyl; -(CH2)i-4R0; -(CH2)o-4OR°;
-0-(CH2)o_4C(0)OR°; -(CH2)0-4CH(ORo)2; -(CH2)0-4SRo; -(CH2)0-4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°;
-(CH2)o-4-heterocyclyl, which may be substituted with R°; -(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°; -CH=CH-heterocyclyl, which may be substituted with R°;
-CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3; -(CH2)0-4N(R°)2;
-(CH2)0-4N(R°)C(O)R°; -(CH2)0-4N(R°)C(S)R°; -(CH2)0-4N(R°)C(O)NR°2;
-(CH2)0-4N(R°)C(S)NR°2; -(CH2)0-4N(R°)C(O)OR°; -(CH2)0-4N(Ro)N(Ro)C(O)R°;
-(CH2)o-4N(R°)N(R°)C(0)NR°2; -(CH2)0.4N(Ro)N(Ro)C(O)OR°; -(CH2)0_4C(O)R°;
-(CH2)o-4C(S)R°; -(CH2)o.4C(0)OR°; -(CH2)0-4C(O)SR°; -(CH2)0.4OC(O)R°;
-(CH2)o-40C(0)(CH2)o.4SR0, -(CH2)0-4SC(S)SRo; -(CH2)0-4SC(O)Ro; -(CH2)0-4C(O)NRo 2;
-(CH2)0-4C(S)NRo 2; -(CH2)0-4C(S)SRo; -(CH2)04OC(O)NRo 2; -(CH2)o-4C(0)N(OR0)R°;
-(CH2)o-4C(0)C(0)R°; -(CH2)0-4C(O)CH2C(O)R°; -(CH2)o-4C(NOR0)R0; -(CH2)0-4SSRo;
-(CH2)o-4S(0)2R°; -(CH2)0-4S(O)2OR°; -(CH2)0.4OS(O)2R°; -(CH2)0-4S(O)2NR°2;
-(CH2)o-4S(0)R°; -(CH2)o-4N(R°)S(0)2NR°2; -(CH2)0-4N(Ro)S(O)2R°; -(CH2)0.4N(OR°)R°;
-(CH2)o-4C(NH)NR°2; -(CH2)0-4P(O)2Ro ; -(CH2)0.4P(O)Ro 2; -(CH2)0-4OP(O)R°2;
-(CH2)o-4OP(0)(OR°)2; -(CH2)0-4ON(Ro)2; and -(CH2)0-4C(O)O-N(Ro)2, wherein:
each R° is independently hydrogen, C1-6 aliphatic, -CH2-carbocyclyl,
-CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -O^H^o-i-carbocyclyl,
-0(CH2)o-i-aryl, -0(CH2)o-i-heterocyclyl, -0(CH2)o-i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Ci-C3 alkyl, -NH2, -N02, -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted d-C3 alkyl)2, -O-C1-C3 alkyl, -C(0)OH,
-C(0)0-(unsubstituted C1-C3 alkyl), -C(0)-(unsubstituted C1-C3 alkyl),
-0-(unsubstituted C1-C3 alkyl), and -S-(unsubstituted C1-C3 alkyl);
R2 is selected from -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(S)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4,
-C(S)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-C(S)-R4, -C(S)-N(R7)-N(R7)-C(S)-R4, -C(0)-N(R7)-N(R7)-S(0)i-2-R4, -C(S)-N(R7)-N(R7)-S(0)]-2-R4 and heteroaryl, wherein:
R3 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(CrC4 alkyl)(C3-C6 cycloalkyl), -Cr C6 alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl, -(C0-C4
alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, C1-C4 alkyl, Q-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or
R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
each R7 is independently hydrogen or C1-C4 alkyl; and
n is 0, 1, 2, 3, 4 or 5; wherein unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted. Alternative values for the variables in Structural Formula I are as described in the first through third embodiments, or any aspect thereof.
[0064] In a first aspect of the fourth embodiment, R2 is -C(0)-0-R3, and R3 is hydrogen or is selected from optionally substituted C1-C4 alkyl and C2-C4 alkenyl; or R2 is
-C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or R is
-C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or R2 is
-C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -d-C6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl; or R2 is optionally substituted C5-C6 heteroaryl. The values for the remaining variables are as described in the first through third embodiments, or any aspect thereof, or the fourth embodiment.
[0065] In a second aspect of the fourth embodiment, R3 is hydrogen or is selected from optionally substituted C1-C4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl. The values for the remaining variables are as described in the first through third embodiments, or any aspect thereof, or the fourth embodiment, or first aspect thereof.
[0066] In a third aspect of the fourth embodiment, R2 is -C(0)-0-R3, and R3 is hydrogen or is selected from optionally substituted C C4 alkyl; or R2 is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C3-C7 heterocyclyl; or R2 is -C(0)-NH-NH(R6), and R6 is an optionally substituted C5-C6 heteroaryl; or R2 is -C(0)-NH-NH-C(0)-R4 or
-C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -C1-C6 alkyl, -(C0-C4 alkylene)-(C3- C )heterocyclyl and -(C0-C4 alkylene)-(C5-C6)heteroaryl; or R2 is optionally substituted C5-C6 heteroaryl. The values for the remaining variables are as described in the first through third embodiments, or any aspect thereof, or the fourth embodiment, or first or second aspect thereof.
[0067] A fifth embodiment of a compound of Structural Formula I is a compound of Structural Formula lb, or a pharmaceutically acceptable salt thereof, wherein the values for Ra, Rb and R2 are as described in the fourth embodiment. Alternative values for the variables in Structural Formula lb are as described in the first through fourth embodiments, or any aspect thereof.
[0068] In a first aspect of the fifth embodiment, R2 is -C(0)-0-R3, and R3 is hydrogen or is selected from optionally substituted Ci-C4 alkyl and C2-C4 alkenyl; or R2 is
-C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or R2 is
-C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or R2 is
-C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(C C4 alkyl)(C3-C6 cycloalkyl), -CrC6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl; or R2 is optionally substituted C5-C6 heteroaryl. The values for the remaining variables are as described in the first through fourth embodiments, or any aspect thereof, or the fifth embodiment. [0069] Exemplary compounds of Structural Formula I are set forth in Table 1. Compounds of Structural Formula II
[0070] A first embodiment of a compound of Structural Formula II is a compound of Structural Formula II:
Figure imgf000020_0001
or a pharmaceutically acceptable salt thereof, wherein:
X is -N- or -C(H)-;
each R1 is independently selected from halo; haloalkyl; -(CH2)i-4R°; -(CH2)o-40R°;
-0-(CH2)o-4C(0)OR0; -(CH2)0-4CH(ORo)2; -(CH2)0-4SR°; -(CH2)0-4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°; -(CH2)o-4-heterocyclyl, which may be substituted with R°;
-(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°; -CH=CH-heterocyclyl, which may be substituted with R°;
-CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3;
-(CH2)0-4N(R°)2; -(CH2)o.4N(R°)C(0)R°; -(CH2)0-4N(R°)C(S)R°;
-(CH2)o-4N(R0)C(0)NR°2; -(CH2)0-4N(Ro)C(S)NR°2; -(CH2)0-4N(Ro)C(O)OR°; -(CH2)o-4N(R0)N(R°)C(0)R0; -(CH2)o-4N(R0)N(R0)C(0)NR0 2;
-(CH2)o-4N(R0)N(R°)C(0)OR0; -(CH2)0- C(O)R°; -(CH2)0.4C(S)R°;
-(CH2)0-4C(O)OR°; -(CH2)0-4C(O)SR°; -(CH2)0-4OC(O)R°;
-(CH2)0-4OC(0)(CH2)o.4SR0, -(CH2)0.4SC(S)SR°; -(CH2)0.4SC(O)Ro; -(CH2)0-4C(O)NR°2; -(CH2)0-4C(S)NR°2; -(CH2)0-4C(S)SR°; -(CH2)0-4OC(O)NR°2; -(CH2)o-4C(0)N(OR°)R°; -(CH2)0-4C(O)C(O)R°; -(CH2)0-4C(O)CH2C(O)R°; -(CH2)o-4C(NOR°)R°; -(CH2)0-4SSRo; -(CH2)o-4S(0)2R°; -(CH2)0- S(O)2OR°; -(CH2)o- OS(0)2R°; -(CH2)0-4S(O)2NRo 2; -(CH2)0-4S(O)R°;
-(CH2)o- N(R°)S(0)2NR°2; -(CH2)0-4N(Ro)S(O)2R°; -(CH2)0- N(ORo)R°; -(CH2)o.4C(NH)NR°2; -(CH2)0-4P(O)2R°; -(CH2)0-4P(O)R°2; -(CH2)0-4OP(O)R°2;
-(CH2)0-4OP(O)(ORo)2; -(CH2)0-4ON(R°)2; and -(CH2)0-4C(O)O-N(R°)2, wherein:
each R° is independently hydrogen, Ci-6 aliphatic, -CH2-carbocyclyl, -CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i-carbocyclyl, -0(CH2)o-i-aryl, -0(CH2)o-i-heterocyclyl, -O(CH2)0-i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Ci-C3 alkyl, -NH2, -N02, -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted d-C3 alkyl)2, -0-Ci-C3 alkyl, -C(0)OH, -C(0)0-(unsubstituted CrC3 alkyl),
-C(0)-(unsubstituted C1-C3 alkyl), -0-(unsubstituted CrC3 alkyl), and
-S-(unsubstituted C1-C3 alkyl);
R2 is halo, cyano, -CF3 or -CHF2;
Ra is selected from -CN, -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(S)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, -C(S)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-C(S)-R4,
-C(S)-N(R7)-N(R7)-C(S)-R4, -C(0)-N(R7)-N(R7)-S(0)1-2-R4, and
-C(S)-N(R7)-N(R7)-S(0)1-2-R4, wherein:
R3 is selected from hydrogen, Cj-C4 alkyl, C -C4 alkenyl, C2-C4 alkynyl,
carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(CrC4 alkyl)(C3-C6 cycloalkyl),
-Ci-C6 alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl,
-(C0-C4 alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; each R7 is independently hydrogen or C1-C4 alkyl; and
n is 0, 1, 2, 3, 4 or 5; wherein:
unless otherwise designated, each alkyl, alkenyl, alkynyl, alkyl ene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
the compound is not (E)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4- triazol-l-yl)-3-bromo-l-(3,3-difluoroazetidin-l-yl)prop-2-en-l-one.
[0071] In a first aspect of the first embodiment, R2 is bromo or chloro. Values for the remaining variables are as defined in the first embodiment.
[0072] In a second aspect of the first embodiment, R is fluoro, bromo or chloro. Values for the remaining variables are as defined in the first embodiment, or first aspect thereof.
[0073] In a third aspect of the first embodiment, Ra is selected from -C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, and
-C(0)-N(R7)-N(R7)-S(0)i.2-R4. Values for the remaining variables are as defined in the first embodiment, or first or second aspect thereof.
[0074] In a fourth aspect of the first embodiment, Ra is selected from -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6) and
-C(S)-N(R7)-N(R5)(R6). Values for the remaining variables are as defined in the first embodiment, or first through third aspects thereof.
[0075] In a fifth aspect of the first embodiment, Ra is selected from -C(0)-0-R3, -C(0)-N(R5)(R6), and -C(0)-N(R7)-N(R5)(R6). Values for the remaining variables are as defined in the first embodiment, or first through fourth aspects thereof.
[0076] In a sixth aspect of the first embodiment, X is -C(H)-. Values for the remaining variables are as defined in the first embodiment, or first through fifth aspects thereof.
[0077] In a seventh aspect of the first embodiment, n is 0, 1 or 2. Values for the remaining variables are as defined in the first embodiment, or first through sixth aspects thereof.
[0078] In an eighth aspect of the first embodiment, each R1 is independently selected from -CF3, -CN, halo, - OH, C C3 alkyl, C3-C6 cycloalkyl, C3-Ci2 heterocycloalkyl, halo-Cr C3 alkyl, -NH2, -N02, -NH(Ci-C3 alkyl), -N(C C3 alkyl)(d-C3 alkyl), -C(0)OH,
-C(0)0-(Ci-C6 alkyl), -C(0)-(CrC3 alkyl), -0-(Ci-C3 alkyl), -0-(C C3 haloalkyl), and -S-( C1-C3 alkyl), or is absent. Values for the remaining variables are as defined in the first embodiment, or first through seventh aspects thereof.
[0079] In a ninth aspct of the first embodiment, each R1 is independently selected from halo, -C1-C4 alkyl, -C1-C4 haloalkyl and -0-CrC4 alkyl, or is absent. Values for the remaining variables are as defined in the first embodiment, or first through eighth aspects thereof.
[0080] In a tenth aspect of the first embodiment:
Ra is -C(0)-0-R3, and R3 is selected from optionally substituted C1-C4 alkyl and C2- C4 alkenyl; or
Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or
Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(d-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl.
Values for the remaining variables are as defined in the first embodiment, or first through ninth aspects thereof.
[0081] In an eleventh aspect of the first embodiment:
Ra is -C(0)-0-R3, and R3 is selected from optionally substituted d-C alkyl; or Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C3- C7 heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted C5-C6 heteroaryl; or Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)1-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(C1-C4 alkyl)(C3-C6 cycloalkyl), -CpC6 alkyl, -(C0-C4 alkylene)-(C3-C7)heterocyclyl and -(C0-C4 alkylene)-(C5-C6)heteroaryl.
Values for the remaining variables are as defined in the first embodiment, or first through tenth aspects thereof. [0082] In a twelfth aspect of the first embodiment, Ra is selected from 3,3- difluoroazetidin-l-ylcarbonyl, N'-(pyrazin-2-yl)-hydrazinylcarbonyl, and -C(0)OCH(CH3)2. Values for the remaining variables are as defined in the first embodiment, or first through eleventh aspects thereof.
[0083] In a thirteenth aspect of the first embodiment, each R7 is hydrogen. The values for the remaining variables are as described in the first embodiment, or first through twelfth aspects thereof.
[0084] In a fourteenth aspect of the first embodiment, R5 is selected from hydrogen and C1-C4 alkyl; and R6 is selected from C1-C4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through thirteenth aspects thereof.
[0085] In a fifteenth aspect of the first embodiment, R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through fourteenth aspects thereof.
[0086] In a sixteenth aspect of the first embodiment, R3 is selected from optionally substituted CpC4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through fifteenth aspects thereof.
4 8
[0087] In a seventeenth aspect of the first embodiment, R is selected from -N(R )(C3-C6 cycloalkyl), -C3-C6 alkyl, -(Co- alkylene)-heterocyclyl, and -(QrQ alkylene)-heteroaryl, wherein R8 is hydrogen or -C1-C4 alkyl; any alkyl or alkylene portion of R4 is optionally and independently substituted with one or more substituents selected from the group consisting of oxo and -N(R9)2, wherein each R9 is independently selected from hydrogen and CrC4 alkyl; any heterocyclyl portion of R4 comprises at least one nitrogen atom in a ring, and is optionally substituted with one or more substituents selected from the group consisting of Ci- C4 alkyl and oxo; and any heteroaryl portion of R4 comprises at least one nitrogen atom in a ring and is optionally substituted with one or more CpC4 alkyl. The values for the remaining variables are as described in the first embodiment, or first through sixteenth aspects thereof.
[0088] In an eighteenth aspect of the first embodiment, R is halo. The values for the remaining variables are as described in the first embodiment, or first through seventeenth aspects thereof. [0089] In a nineteenth aspect of the first embodiment, X is -N-. Values for the remaining variables are as defined in the first embodiment, or first through eighteenth aspects thereof.
[0090] In a second embodiment of a compound of Structural Formula II, the compound is represented by Structural Formula Ila:
or a pharmaceutically acceptable salt thereof, wherein:
X is -N- or -C(H)-;
Rla and Rlb are each independently selected from halo; haloalkyl; -(CH2)i-4R°; -(CH2)o-4OR0; -O-(CH2)0-4C(O)OR°; -(CH2)0-4CH(ORo)2; -(CH2)0-4SR°;
-(CH2)o-4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°; -(CH2)o-4-heterocyclyl, which may be substituted with R°; -(CH2)o-4-heteroaryl, which may be substituted with R°; -CH==CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°;
-CH=CH-heterocyclyl, which may be substituted with R°; -CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3; -(CH2)0-4N(R°)2; -(CH2)0-4N(Ro)C(O)Ro; -(CH2)o-4N(R0)C(S)R°; -(CH2)o-4N(R0)C(0)NR0 2; -(CH2)o-4N(R0)C(S)NR°2;
-(CH2)o-4N(R0)C(0)OR0; -(CH2)o-4N(R°)N(R0)C(0)R°;
-(CH2)o-4N(R0)N(R°)C(0)NR0 2; -(CH2)0-4N(Ro)N(Ro)C(O)OR°; -(CH2)0-4C(O)R°; -(CH2)o-4C(S)R°; -(CH2)o-4C(0)OR°; -(CH2)0-4C(O)SR°; -(CH2)0-4OC(O)R°;
-(CH2)o-4OC(0)(CH2)o-4SR°, -(CH2)0-4SC(S)SRo; -(CH2)0-4SC(O)R°;
-(CH2)o-4C(0)NR0 2; -(CH2)0-4C(S)NRo 2; -(CH2)0-4C(S)SRo; -(CH2)0-4OC(O)NR°2; -(CH2)o-4C(0)N(OR°)R°; -(CH2)0-4C(O)C(O)R°; -(CH2)0-4C(O)CH2C(O)R°;
-(CH2)0-4C(NOR°)R°; -(CH2)0-4SSR°; -(CH2)0-4S(O)2R°; -(CH2)0-4S(O)2OR°;
-(CH2)0-4OS(O)2R°; -(CH2)0.4S(O)2NR°2; -(CH2)0- S(O)R°;
-(CH2)o-4N(R°)S(0)2NR°2; -(CH2)0-4N(Ro)S(O)2R°; -(CH2)0-4N(ORo)R°;
-(CH2)o- C(NH)NR°2; -(CH2)0-4P(O)2R°; -(CH2)0-4P(O)Ro 2; -(CH2)0-4OP(O)Ro 2;
-(CH2)o-4OP(0)(OR°)2; -(CH2)0-4ON(R°)2; and -(CH2)0-4C(O)O-N(R°)2, wherein: each R° is independently hydrogen, C1-6 aliphatic, -CH2-carbocyclyl,
-CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i -carbocyclyl,
-0(CH2)o-i-aryl, -O(CH2)0-i -heterocyclyl, -0(CH2)o-i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Ci-C3 alkyl, -NH2, -N02, -NH(unsubstituted Ci-C3 alkyl), -N(unsubstituted Ci-C3 alkyl)2, -O-C1-C3 alkyl, -C(0)OH,
-C(0)0-(unsubstituted d-C3 alkyl), -C(0)-(unsubstituted C1-C3 alkyl),
-0-(unsubstituted Cj-C3 alkyl), and -S-(unsubstituted Ci-C3 alkyl);
m is 0 or 1 ;
R2 is halo, cyano, -CF3 or -CHF2; and
Ra is selected from -CN, -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(S)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, -C(S)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-C(S)-R4,
-C(S)-N(R7)-N(R7)-C(S)-R4, -C(0)-N(R7)-N(R7)-S(0)1-2-R4, and
-C(S)-N(R7)-N(R7)-S(0)i-2-R4, wherein:
R3 is selected from hydrogen, C 1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl,
carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl),
-Ci-C6 alkyl, -(C0-C4 alkyl ene)-carbocyclyl, -(C0-C4 alky lene) -heterocyclyl,
-(C0-C4 alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; and
each R7 is independently hydrogen or C1-C4 alkyl, wherein: unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
the compound is not (E)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4- triazol- 1 -yl)-3 -bromo- 1 -(3 ,3-difluoroazetidin- 1 -yl)prop-2-en- 1 -one.
Alternative values for the variables of structural formula Ila are as defined in the first embodiment, or any aspect thereof.
[0091] In a first aspect of the second embodiment, Rla is -Cj-C4 haloalkyl. Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment.
[0092] In a second aspect of the second embodiment, Rlb is -Ci-C4 haloalkyl. Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment, or first aspect thereof.
[0093] In a third aspect of the second embodiment, Rla is -CF3 and Rlb is -CF3. Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment, or first or second aspect thereof.
[0094] In a fourth aspect of the second embodiment, Rlb is -Ci-C4 haloalkyl or -0-Ci-C4 alkyl or absent. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through third aspects thereof.
[0095] In a fifth aspect of the second embodiment, Rla is halo or -Ci-C4 haloalkyl. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fourth aspects thereof.
[0096] In a sixth aspect of the second embodiment, m is 1. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fifth aspects thereof.
[0097] A third embodiment of a compound of Structural Formula II is a compound represented by Structural Formula lib:
Figure imgf000028_0001
or a pharmaceutically acceptable salt thereof, wherein:
R2 is halo, cyano, -CF3 or -CHF2; and
Ra is selected from -C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6),
-C(0)-N(R7)-N(R7)-C(0)-R4 and -C(0)-N(R7)-N(R7)-S(0)i-2-R4, wherein: R is selected from hydrogen, C C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl,
carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(CrC4 alkyl)(C3-C6 cycloalkyl),
-Ci-C6 alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl,
-(C0-C4 alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, Cj-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; and
each R7 is independently hydrogen or Cj-C4 alkyl, wherein:
unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
the compound is not (E)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l ,2,4- triazol- 1 -yl)-3 -bromo- 1 -(3 ,3-difluoroazetidin- 1 -yl)prop-2-en- 1 -one
Alternative values for the variables of Structural Formula lib are as defined in the first embodiment, or any aspect thereof.
[0098] In a first aspect of the third embodiment, R2 is bromo or chloro. Values for the remaining variables are as defmed in the first embodiment, or any aspect thereof or the third embodiment.
[0099] In a second aspect of the third embodiment:
Ra is -C(0)-0-R3, and R3 is selected from optionally substituted Ci-C4 alkyl and C2- C4 alkenyl; or Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or
Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)1 -2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(d-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl.
Values for the remaining variables are as defined in the first embodiment, or any aspect thereof or the third embodiment, or first aspect thereof.
[00100] In a third aspect of the third embodiment:
Ra is -C(0)-0-R3, and R3 is selected from optionally substituted C1-C4 alkyl; or Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C3- C7 heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted C5-C6 heteroaryl; or Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-(C3-C7)heterocyclyl and -(C0-C4 alkylene)-(C5-C6)heteroaryl.
Values for the remaining variables are as defined in the first embodiment, or any aspect thereof or the third embodiment, or first or second aspect thereof.
[00101] In a fourth aspect of the third embodiment, Ra is selected from -C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, and
-C(0)-N(R7)-N(R7)-S(0)i_2-R4. Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the third embodiment, or first through third aspects thereof.
[00102] In a fifth aspect of the third embodiment, Ra is selected from -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6) and
-C(S)-N(R7)-N(R5)(R6). Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the third embodiment, or first through fourth aspects thereof. [00103] In a sixth aspect of the third embodiment, Ra is selected from -C(0)-0-R3, -C(0)-N(R5)(R6), and -C(0)-N(R7)-N(R5)(R6). Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the third embodiment, or first through fifth aspects thereof
[00104] In a seventh aspect of the third embodiment, R2 is halo. Values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the third
embodiment, or first through sixth aspects thereof.
[00105] Exemplary compounds of Structural Formula II are set forth in Table 2. Compounds of Structural Formula III
[00106] A first embodiment of a compound of Structural Formula III is a compound of Structural Formula III:
Figure imgf000030_0001
or a pharmaceutically acceptable salt thereof, wherein:
X is -N- or -C(H)-;
each R1 is independently selected from halo; haloalkyl; -(CH2)1-4R°;
-(CH2)o.4OR°; -0-(CH2)o-4C(0)OR°; -(CH2)0-4CH(OR°)2; -(CH2)0-4SR°; -(CH2)o-4-carbocyclyl, which may be substituted with R°; -(CH2)0-4-aryl, which may be substituted with R°; -(CH2)0-4-heterocyclyl, which may be substituted with R°; -(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°; -CH=CH-heterocyclyl, which may be substituted with R°;
-CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3;
-(CH2)0-4N(R°)2; -(CH2)0-4N(R°)C(O)R°; -(CH2)0-4N(R°)C(S)R°;
-(CH2)0-4N(R°)C(O)NR°2; -(CH2)0-4N(R°)C(S)NR°2; -(CH2)0-4N(R°)C(O)OR°; -(CH2)0-4N(R°)N(Ro)C(O)R°; -(CH2)0-4N(Ro)N(R°)C(O)NRo 2;
-(CH2)o-4N(R0)N(R°)C(0)OR°; -(CH2)0-4C(O)R°; -(CH2)0-4C(S)R°;
-(CH2)0-4C(O)OR°; -(CH2)0-4C(O)SR°; -(CH2)0-4OC(O)R°; -(CH2)o-4OC(0)(CH2)o-4SR0, -(CH2)0-4SC(S)SR°; -(CH2)0-4SC(O)R°;
-(CH2)o-4C(0)NR°2; -(CH2)0-4C(S)NRo 2; -(CH2)0-4C(S)SR°; -(CH2)0-4OC(O)NR°2; -(CH2)o-4C(0)N(OR0)R°; -(CH2)o-4C(0)C(0)R0; -(CH2)0-4C(O)CH2C(O)R°;
-(CH2)0.4C(NOR°)R°; -(CH2)0.4SSR°; -(CH2)0-4S(O)2R°; -(CH2)0-4S(O)2OR°; -(CH2)0-4OS(O)2R°; -(CH2)0-4S(O)2NR°2; -(CH2)0-4S(O)R°;
-(CH2)0-4N(R°)S(O)2NR°2; -(CH2)0-4N(Ro)S(O)2R°; -(CH2)0-4N(ORo)R°;
-(CH2)0-4C(NH)NR°2; -(CH2)0.4P(O)2R°; -(CH2)0.4P(O)R°2; -(CH2)0-4OP(O)R°2; -(CH2)0-4OP(O)(OR°)2; -(CH2)0-4ON(R°)2; and -(CH2)0-4C(O)O-N(R°)2, wherein: each R° is independently hydrogen, C1-6 aliphatic, -CH2-carbocyclyl, -CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i-carbocyclyl, -0(CH2)o-i-aryl, -0(CH2)o-i-heterocyclyl, -0(CH2)o-i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted CrC3 alkyl, halo-C C3 alkyl, -NH2, -N02,
-NH(unsubstituted CrC3 alkyl), -N(unsubstituted C C3 alkyl)2, -0-C C3 alkyl, -C(0)OH, -C(0)0-(unsubstituted CrC3 alkyl), -C(0)-(unsubstituted C C3 alkyl), -0-(unsubstituted C C3 alkyl), and -S-(unsubstituted Cj-C3 alkyl);
R2 is halo, -CF3, -CHF2, or -CN;
Ra is selected from -CN, -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(S)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, -C(S)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-C(S)-R4,
-C(S)-N(R7)-N(R7)-C(S)-R4, -C(0)-N(R7)-N(R7)-S(0)1-2-R4, and
-C(S)-N(R7)-N(R7)-S(0)i-2-R4, wherein:
R3 is selected from hydrogen, Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl,
carbocyclyl, aryl, heterocyclyl and heteroaryl; R is selected from -N(H)(C3-C6 cycloalkyl), -N(CrC4 alkyl)(C3-C6 cycloalkyl), -Ci-Ce alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)- eterocyclyl, -(C0-C4 alkylene)-aryl, and -(Co-C4 alkylene)- eteroaryl;
R5 and R6 are each independently selected from hydrogen, Q-Q alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or
R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
each R is independently hydrogen or Q-C4 alkyl; and
n is 0, 1 , 2, 3, 4 or 5; wherein:
unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
the compound is not (E)-isopropyl 2-fluoro-3-(3-(3-methoxy-5- (trifluoromethyl)phenyl)- lH- l,2,4-triazol-l-yl)acrylate.
[00107] In a first aspect of the first embodiment, R2 is halo. The values for the remaining variables are as defined in the first embodiment.
[00108] In a second aspect of the first embodiment, R2 is bromo or chloro. The values for the remaining variables are as defined in the first embodiment, or first aspect thereof.
[00109] In a third aspect of the first embodiment, R2 is bromo, chloro, -CF3, -CHF2, or - CN. The values for the remaining variables are as defined in the first embodiment, or first or second aspect thereof.
[00110] In a fourth aspect of the first embodiment, Ra is selected from -CN, -C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, and
-C(0)-N(R7)-N(R7)-S(0)1-2-R4. The values for the remaining variables are as defined in the first embodiment, or first through third aspects thereof.
[00111] In a fifth aspect of the first embodiment, R is selected from -CN, -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), and
-C(S)-N(R7)-N(R5)(R6). The values for the remaining variables are as defined in the first embodiment, or first through fourth aspects thereof.
[00112] In a sixth aspect of the first embodiment, Ra is selected from is selected from -CN, -C(0)-0-R3, -C(0)-N(R5)(R6), and -C(0)-N(R7)-N(R5)(R6). The values for the remaining variables are as defined in the first embodiment, or first through fifth aspects thereof. [00113] In a seventh aspect of the first embodiment, R is selected from 3,3- difluoroazetidin-l-ylcarbonyl, N'-(pyrazin-2-yl)-hydrazinylcarbonyl, -C(0)OCH(CH3)2, -C(0)NH2, -COOH, and -CN. The values for the remaining variables are as defined in the first embodiment, or first through sixth aspects thereof.
[00114] In an eighth aspect of the first embodiment, X is -N-. The values for the remaining variables are as defined in the first embodiment, or first through seventh aspects thereof.
[00115] In a ninth aspect of the first embodiment, X is -CH-. The values for the remaining variables are as defined in the first embodiment, or first through eighth aspects thereof.
[00116] In a tenth aspect of the first embodiment, n is 0, 1 or 2. The values for the remaining variables are as defined in the first embodiment, or first through ninth aspects thereof.
[00117] In an eleventh aspect of the first embodiment, each R1 is independently selected from -CF3, -CN, halo, - OH, Ci-C3 alkyl, C3-C6 cycloalkyl, C3-Ci2 heterocycloalkyl, halo-Ci- C3 alkyl, -NH2, -N02, -NH(Ci-C3 alkyl), -N(Ci-C3 alkyl)(Ci-C3 alkyl), -C(0)OH,
-C(0)0-(Ci-C6 alkyl), -C(0)-(Ci-C3 alkyl), -0-(Ci-C3 alkyl), -0-(Ci-C3 haloalkyl), and -S-( Ci-C3 alkyl), or is absent. The values for the remaining variables are as defined in the first embodiment, or first through tenth aspects thereof.
[00118] In a twelfth aspect of the first embodiment, each R1 is independently selected from halo, -C1-C4 alkyl, -C1-C4 haloalkyl and -O-C1-C4 alkyl, or is absent. The values for the remaining variables are as defined in the first embodiment, or first through eleventh aspects thereof.
[00119] In a thirteenth aspect of the first embodiment, Ra is -CN; or Ra is -C(0)-0-R3, and R3 is selected from hydrogen and optionally substituted C1-C4 alkyl or C2-C4 alkenyl; or Ra is -C(0)NH2; or R is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated
heterocyclyl; or Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or R is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(d-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0- C4 alkylene) -heterocyclyl and -(C0-C4 alkylene)-heteroaryl. The values for the remaining variables are as defined in the first embodiment, or first through twelfth aspects thereof.
[00120] In a fourteenth aspect of the first embodiment: Ra is -C(0)-0-R3, and R3 is selected from hydrogen and optionally substituted Ci-C4 alkyl; or
Ra is -C(0)NH2; or
Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C3- C7 heterocyclyl; or
R is -C(0)-NH-NH(R6), and R6 is an optionally substituted C5-C6 heteroaryl; or Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-(C3-C7)heterocyclyl and -(C0-C4 alkylene)-(C5-C6)heteroaryl.
The values for the remaining variables are as defined in the first embodiment, or first through thirteenth aspects thereof.
[00121] In a fifteenth aspect of the first embodiment, Ra is selected from -C(0)OH, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, and
-C(0)-N(R7)-N(R7)-S(0)1-2-R4. The values for the remaining variables are as defined in the first embodiment, or first through fourteenth aspects thereof.
[00122] In a sixteenth aspect of the first embodiment, each R7 is hydrogen. The values for the remaining variables are as described in the first embodiment, or first through fifteenth aspects thereof.
[00123] In a seventeenth aspect of the first embodiment, R5 is selected from hydrogen and Ci-C4 alkyl; and R6 is selected from hydrogen, Ci-C4 alkyl, carbocyclyl, aryl, heterocyclyl and heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through sixteenth aspects thereof.
[00124] In an eighteenth aspect of the first embodiment, R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through seventeenth aspects thereof.
[00125] In a nineteenth aspect of the first embodiment, R3 is selected from hydrogen and optionally substituted Ci-C4 alkyl, carbocyclyl, aryl, heterocyclyl or heteroaryl. The values for the remaining variables are as described in the first embodiment, or first through eighteenth aspects thereof. [00126] In a twentieth aspect of the first embodiment, R4 is selected from -N(R8)(C3-C6 cycloalkyl), -C3-C6 alkyl, -(C0-Ci alkylene)-heterocyclyl, and -(C0-Ci alkylene)-heteroaryl,
8 4
wherein R is hydrogen or -Q-C4 alkyl; any alkyl or alkyl ene portion of R is optionally and independently substituted with one or more substituents selected from the group consisting of oxo and -N(R9)2, wherein each R9 is independently selected from hydrogen and Q-C4 alkyl; any heterocyclyl portion of R4 comprises at least one nitrogen atom in a ring, and is optionally substituted with one or more substituents selected from the group consisting of Q- C4 alkyl and oxo; and any heteroaryl portion of R4 comprises at least one nitrogen atom in a ring and is optionally substituted with one or more Q-C4 alkyl. The values for the remaining variables are as described in the first embodiment, or first through sixteenth aspects thereof.
[00127] A second embodiment of a compound of Structural Formula III is a compound represented by Structural Formula Ilia:
Figure imgf000035_0001
or a pharmaceutically acceptable salt thereof, wherein:
Rla and Rlb are each independently selected from halo; haloalkyl; -(CH2)i.4R°; -(CH2)o-4OR°; -0-(CH2)o-4C(0)OR°; -(CH2)o-4CH(OR°)2; -(CH2)0-4SR°;
-(CH2)0-4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°; -(CH2)o-4-heterocyclyl, which may be substituted with R°; -(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°;
-CH=CH-heterocyclyl, which may be substituted with R°; -CH==CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3; -(CH2)0-4N(Ro)2; -(CH2)0-4N(Ro)C(O)Ro; -(CH2)o-4N(R0)C(S)R0; -(CH2)o-4N(R0)C(0)NR0 2; -(CH2)0-4N(R°)C(S)NR°2;
-(CH2)o-4N(R0)C(0)OR°; -(CH2)o.4N(R0)N(R0)C(0)R0;
-(CH2)o.4N(R°)N(R°)C(0)NR°2; -(CH2)o-4N(R0)N(R0)C(0)OR°; -(CH2)0-4C(O)R°; -(CH2)o-4C(S)R°; -(CH2)0-4C(O)OR°; -(CH2)0-4C(O)SR°; -(CH2)0-4OC(O)R°;
-(CH2)o-40C(0)(CH2)o-4SR0, -(CH2)0-4SC(S)SR°; -(CH2)o-4SC(0)R°; -(CH2)0-4C(O)NR°2; -(CH2)0-4C(S)NR°2; -(CH2)0-4C(S)SR°; -(CH2)0.4OC(O)NR°2; -(CH2)o-4C(0)N(OR0)R°; -(CH2)o-4C(0)C(0)R0; -(CH2)0-4C(O)CH2C(O)R°;
-(CH2)0-4C(NOR°)R°; -(CH2)0-4SSR°; -(CH2)0-4S(O)2R°; -(CH2)0-4S(O)2OR°;
-(CH2)o-4OS(0)2R°; -(CH2)0-4S(O)2NR°2; -(CH2)0-4S(O)R°;
-(CH2)0-4N(R°)S(O)2NR°2; -(CH2)0-4N(Ro)S(O)2R°; -(CH2)0-4N(OR°)R°;
-(CH2)0-4C(NH)NR°2; -(CH2)0.4P(O)2R°; -(CH2)0-4P(O)R°2; -(CH2)0-4OP(O)R°2;
-(CH2)0-4OP(O)(OR°)2; -(CH2)0-4ON(R°)2; and -(CH2)0-4C(O)O-N(R°)2, wherein: each R° is independently hydrogen, C1-6 aliphatic, -CH2-carbocyclyl,
-CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i-carbocyclyl,
-O(CH2)0-1-aryl, -O(CH2)0-i -heterocyclyl, -0(CH2)o-i -heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted Q-C3 alkyl, halo-Ci-C3 alkyl, -NH2, -N02, -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted CrC3 alkyl)2, -0-Ci-C3 alkyl, -C(0)OH,
-C(0)0-(unsubstituted Q-C3 alkyl), -C(0)-(unsubstituted Q-C3 alkyl),
-0-(unsubstituted C C3 alkyl), and -S-(unsubstituted C C3 alkyl);
m is 0 or 1 ;
X is -N- or -C(H)-;
R2 is halo, -CF3, -CHF2, or -CN; and
Ra is selected from -CN, -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(S)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, -C(S)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-C(S)-R4,
-C(S)-N(R7)-N(R7)-C(S)-R4, -C(0)-N(R7)-N(R7)-S(0)i-2-R4, and
-C(S)-N(R7)-N(R7)-S(0)i-2-R4, wherein:
R3 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C alkynyl,
carbocyclyl, aryl, heterocyclyl and heteroaryl; R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl, -(C0-C4 alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or
R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; and
each R7 is independently hydrogen or C1-C4 alkyl, wherein:
unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
the compound is not (E)-isopropyl 2-fluoro-3-(3-(3-methoxy-5- (trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)acrylate .
Alternative values for the variables in Structural Formula Ilia are as described in the first embodiment, or any aspect thereof.
[00128] In a first aspect of the second embodiment, Rl a is -C1-C4 haloalkyl. The values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment.
[00129] In a second aspect of the second embodiment, Rlb is -C1-C4 haloalkyl. The values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment, or first aspect thereof.
[00130] In a third aspect of the second embodiment, Rla is -CF3 and Rlb is -CF3. The values for the remaining variables are as defined in the first embodiment, or any aspect thereof, or the second embodiment, or first or second aspect thereof.
[00131] In a fourth aspect of the second embodiment, Rlb is -C1-C4 haloalkyl or -O-C1-C4 alkyl or absent. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through third aspects thereof.
[00132] In a fifth aspect of the second embodiment, Rla is halo or -Cj-C4 haloalkyl. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fourth aspects thereof. [00133] In a sixth aspect of the second embodiment, m is 1. The values for the remaining variables are as described in the first embodiment, or any aspect thereof, or the second embodiment, or first through fifth aspects thereof.
[00134] A third embodiment of a compound of Structural Formula III is a compound represented by Structural Formula Illb:
Figure imgf000038_0001
(Illb),
or a pharmaceutically acceptable salt thereof, wherein:
R2 is halo, -CF3, -CHF2, or -CN; and
Ra is selected from -CN, -C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, and -C(0)-N(R7)-N(R7)-S(0)i-2-R4, wherein:
R3 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl,
carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl, -(C0-C4 alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; and
each R7 is independently hydrogen or Ci-C4 alkyl, wherein:
unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene,
carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
the compound is not (E)-isopropyl 2-fluoro-3-(3-(3-methoxy-5- (trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)acrylate.
Alternative values for the variables in Structural Formula Illb are as described in the first embodiment, or any aspect thereof.
[00135] Exemplary compounds of Structural Formula III are set forth in Table 3. Compounds and Definitions
[00136] Compounds of this invention include those described generally above, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Additionally, general principles of organic chemistry are described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausalito: 1999, and "March's Advanced Organic Chemistry", 5th Ed., Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[00137] Unless specified otherwise within this specification, the nomenclature used in this specification generally follows the examples and rules stated in Nomenclature of Organic Chemistry, Sections A, B, C, D, E, F, and H, Pergamon Press, Oxford, 1979, which is incorporated by reference herein for its exemplary chemical structure names and rules on naming chemical structures. Optionally, a name of a compound may be generated using a chemical naming program: ACD/ChemSketch, Version 5.09/September 2001 , Advanced Chemistry Development, Inc., Toronto, Canada.
[00138] Compounds of the present invention may have asymmetric centers, chiral axes, and chiral planes (e.g., as described in: E. L. Eliel and S. H. Wilen, Stereo-chemistry of Carbon Compounds, John Wiley & Sons, New York, 1994, pages 1119-1190), and occur as racemates, racemic mixtures, and as individual diastereomers or enantiomers, with all possible isomers and mixtures thereof, including optical isomers, being included in the present invention.
[00139] The term "aliphatic" or "aliphatic group," as used herein, denotes a monovalent hydrocarbon radical that is straight-chain (i.e., unbranched), branched, or cyclic (including fused, bridged, and spiro-fused polycyclic). An aliphatic group can be saturated or can contain one or more units of unsaturation, but is not aromatic. Unless otherwise specified, aliphatic groups contain 1-12 carbon atoms. However, in some embodiments, an aliphatic group contains 1-6 or 2-8 carbon atoms. In some embodiments, aliphatic groups contain 1-4 carbon atoms and, in yet other embodiments, aliphatic groups contain 1-3 carbon atoms. Suitable aliphatic groups include, but are not limited to, linear or branched, alkyl, alkenyl, and alkynyl groups, and hybrids thereof, such as (cycloalkyl) alkyl, (cycloalkenyl)alkyl or (cy clo alkyl) alkenyl .
[00140] The term "alkyl," as used herein, unless otherwise indicated, means straight or branched saturated monovalent hydrocarbon radicals, typically C C12, preferably Q-Q. As such, "Ci-C alkyl" means a straight or branched saturated monovalent hydrocarbon radical having from one to six carbon atoms (e.g., 1 , 2, 3, 4, 5 or 6). Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, and t-butyl.
[00141] The term "alkoxy," as used herein, means an "alkyl-O-" group, wherein alkyl is defined above. Examples of alkoxy include methoxy and ethoxy.
[00142] As used herein, the term "alkenyl" means a saturated straight chain or branched non-cyclic hydrocarbon having from 2 to 12 carbon atoms and having at least one carbon- carbon double bond. Alkenyl groups may be optionally substituted with one or more substituents. The term "alkenyl" encompasses radicals having carbon-carbon double bonds in the "cis" and "trans" or, alternatively, the "E" and "Z" configurations. If an alkenyl group includes more than one carbon-carbon double bond, each carbon-carbon double bond is independently a cis or trans double bond, or a mixture thereof.
[00143] As used herein, the term "alkynyl" means a saturated straight chain or branched non-cyclic hydrocarbon having from 2 to 12 carbon atoms and having at least one carbon- carbon triple bond. Alkynyl groups may be optionally substituted with one or more substituents.
[00144] As used herein, the term "alkylene" refers to an alkyl group having from 2 to 12 carbon atoms and two points of attachment to the rest of the compound. Non-limiting examples of alkylene groups include methylene (-CH2-), ethylene (-CH2CH2-), n-propylene (-CH2CH2CH2-), isopropylene (-CH2CH(CH3)-), and the like. Alkylene groups may be optionally substituted with one or more substituents.
[00145] The term "amino," as used herein, refers to a chemical moiety having the formula -N(R)2, wherein each R is independently selected from hydrogen and Q-Q alkyl.
[00146] The term "aryl," alone or in combination, as used herein, means a carbocyclic aromatic system containing one or more rings, which may be attached together in a pendent manner or may be fused. In particular embodiments, aryl is one, two or three rings. In one aspect, the aryl has six to twelve ring atoms. The term "aryl" encompasses aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl and acenaphthyl. An aryl group can be optionally substituted as defined and described herein.
[00147] The terms "cycloaliphatic," "carbocyclyl," "carbocyclo," and "carbocyclic," used alone or as part of a larger moiety, refer to a saturated or partially unsaturated cyclic aliphatic monocyclic or bi cyclic ring system, as described herein, having from 3 to 12 members, wherein the aliphatic ring system is optionally substituted as defined and described herein. Cycloaliphatic groups include, without limitation, cycloalkyl, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl and cycloalkenyl, for example cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl and cyclooctadienyl. The terms "cycloaliphatic," "carbocyclyl," "carbocyclo," and "carbocyclic" also include aliphatic rings that are fused to one or more aromatic or nonaromatic rings, such as decahydronaphthyl, tetrahydronaphthyl, decalin, or bicyclo[2.2.2]octane.
[00148] The term "cycloalkyl", as used herein, means saturated cyclic hydrocarbons, i.e. compounds where all ring atoms are carbons. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. In some embodiments, cycloalkyl can optionally be substituted with one or more substituents selected from -OH, -SH, halogen, amino, nitro, cyano, Ci-Ci2 alkyl, C2-C12 alkenyl or C2-C12 alkynyl group, C -C\2 alkoxy, Ci-C^ haloalkyl, and Ci-C12 haloalkoxy.
[00149] The term "halo" or "halogen" as used herein means halogen and includes, for example, and without being limited thereto, fluoro, chloro, bromo, iodo and the like, in both radioactive and non-radioactive forms. In a preferred embodiment, halo is selected from the group consisting of fluoro, chloro and bromo.
[00150] The term "haloalkyl", as used herein, includes an alkyl substituted with one or more F, CI, Br, or I, wherein alkyl is defined above.
[00151] The term "heteroaryl", as used herein, refers to an aromatic group containing one or more heteroatoms (e.g. , one or more heteroatoms independently selected from O, S and N). A heteroaryl group can be monocyclic or polycyclic, e.g. a monocyclic heteroaryl ring fused to one or more carbocyclic aromatic groups or other monocyclic heteroaryl groups. The heteroaryl groups of this invention can also include ring systems substituted with one or more oxo moieties. In one aspect, heteroaryl has five to fifteen ring atoms and, preferably, 5 or 6 ring atoms. Examples of heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, quinolyl, isoquinolyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, purinyl, oxadiazolyl, thiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzotriazolyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, dihydroquinolyl, tetrahydroquinolyl,
dihydroisoquinolyl, tetrahydroisoquinolyl, benzofuryl, furopyridinyl, pyrolopyrimidinyl, and azaindolyl. The foregoing heteroaryl groups may be C-attached or N-attached (where such is possible). For instance, a group derived from pyrrole may be pyrrol- 1-yl (N-attached) or pyrrol-3-yl (C-attached).
[00152] "Heterocyclyl" means a cyclic 3-12 membered saturated or unsaturated aliphatic ring containing 1 , 2, 3, 4 or 5 heteroatoms (e.g. , one or more heteroatoms independently selected from O, S and N). When one heteroatom is S, it can be optionally mono- or di-oxygenated (i.e. -S(O)- or -S(0)2-). The heterocyclyl can be monocyclic or polycyclic, in which case the rings can be attached together in a pendent manner or can be fused or spiro. In one aspect, a heterocyclyl is a three- to seven-membered ring system. Exemplary heterocyclyls include, for example, and without being limited thereto, piperidinyl, piperazinyl, pyrrolidinyl, tetrahydrofuranyl and the like.
[00153] "Hydroxyl" means -OH.
[00154] "Oxo" means =0.
[00155] "Thioalkoxy" means -S-alkyl, wherein alkyl is defined as above.
[00156] It is understood that substituents and substitution patterns on the compounds of the invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below. In general, the term "substituted," whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an "optionally substituted group" can have a suitable substituent at each substitutable position of the group and, when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent can be either the same or different at every position. Alternatively, an "optionally substituted group" can be unsubstitued.
[00157] Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. If a substituent is itself substituted with more than one group, it is understood that these multiple groups can be on the same carbon atom or on different carbon atoms, as long as a stable structure results. The term "stable," as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
[00158] Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted group" are independently halogen; -(CH2)( R°; -(CH2)0^OR°; -O(CH2)0-4RO, -0-(CH2)o^C(0)OR°; -(CH2)( CH(OR0)2; -(CH2)04SR°; -(CH^Ph, which may be substituted with R°; -(CH2)o^O(CH2)o»iPh which may be substituted with R°; -CH=CHPh, which may be substituted with R°; -(CH2)o~40(CH2)o_i-pyridyl which may be substituted with R°; -N02; -CN; -N3 ; -(CH2)< N(R°)2; -(CH2)0^N(R°)C(O)R°; -N(R°)C(S)R°;
-(CH2)o-4N(R°)C(0)NR°2; -N(R°)C(S)NR°2; -(CH2)0^N(R°)C(O)OR°; -N(R°)N(R°)C(0)R°; -N(R°)N(R°)C(0)NR°2; -N(R°)N(R°)C(0)OR°; -(CH2)0^C(O)R°; -C(S)R°;
-(CH2)o-4C(0)OR°; -(CH2)o-4C(0)SR°; -(CH2)( C(0)OSiR°3; -(CH2)0^OC(O)R°;
-OC(0)(CH2)o^SR-, SC(S)SR°; -(CH2)( SC(0)R0; -(CH2)( C(0)NR0 2; -C(S)NR°2;
-C(S)SR°; -SC(S)SR°, -(CH2)(MOC(0)NR0 2; -C(0)N(OR°)R°; -C(0)C(0)R°;
-C(0)CH2C(0)R°; -C(NOR°)Ro;-(CH2)0^SSR°; -(CH2)0^S(O)2R°; -(CH2)o_4S(0)2OR°; -(CH2)o-40S(0)2R°; -S(0)2NR°2; -(CH2)0^S(O)R°; -N(R°)S(0)2NR°2; -N(R°)S(0)2R°; -N(OR°)R°; -C(NH)NR°2; -P(0)2R°; -P(0)R°2; -OP(0)R°2; -OP(0)(OR°)2; SiR°3; -(CH straight or branched alkylene)0-N(R°)2; or -(Ci^ straight or branched
alkylene)C(0)0-N(R°)2, wherein each R° may be substituted as defined below and is independently hydrogen, C^6 aliphatic, -CH2Ph, -0(CH2)o~iPh, -CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl monocyclic or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, which may be substituted as defined below.
[00159] Suitable monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), are independently halogen, -(CH2)0-2Re, -(haloRe), -(CH2)02OH, -(CH2)o_2ORe, -(CH2)0 2CII(ORe) ; -O(haloR'), -CN, ~N3, -(CH2)0^2C(O)Re, -(CH2)0_2C(O)OH, -(CH2)0_2C(O)ORe, -(CH2)0_ 2SRe, -(CH2)o-2SH, -(CH2)o-2NH2, ~(CH2)o_2NHRe, -(CH2)0_2NRe 2, -N02, -SiRe 3, -OSiR\ -C(0)SR' -(Ci^ straight or branched alkylene)C(0)ORe, or ~SSRe wherein each Re is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently selected from Q- aliphatic, -CH2Ph, -0(CH2)o iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =0 and =S.
[00160] Suitable divalent substituents on a saturated carbon atom of an "optionally substituted group" include the following: =0, =S, =NNR*2, =NNHC(0)R*, =NNHC(0)OR*, =NNHS(0)2R*, =NR*, =NOR*, -0(C(R*2))2_30- and -S(C(R*2))2_3S- wherein each independent occurrence of R* is selected from hydrogen, aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted" group include: -0(CR*2)2-30-, wherein each independent occurrence of R* is selected from hydrogen, Q-e aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00161] Suitable substituents on the aliphatic group of R* include halogen, -R", -(haloR*), -OH, -OR', -O(haloR'), -CN, -C(0)OH, -C(0)OR', -NH2, -NHR", -NR*2, and -N02, wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci^ aliphatic, -CH2Ph, -0(CH2)o-iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00162] Suitable substituents on a substitutable nitrogen of an "optionally substituted group" include -R, -NR 2, -C(0)R, -C(0)OR, -C(0)C(0)R, -C(0)CH2C(0)R, - S(0)2R, -S(0)2NR 2, -C(S)NR 2, -C(NH)NR 2, and -N(R)S(0)2R; wherein each R is independently hydrogen, Ci_6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl monocyclic or bicyclic ring having C heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00163] Suitable substituents on the aliphatic group of R are independently halogen, -R*, -(haloRe), -OH, -OR*, -0(haloRe), -CN, -C(0)OH, -C(0)ORe, -NH2, -NHR\ -NR*2, or -N02, wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently C^ aliphatic, -CH2Ph, -0(CH2)o-iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having C heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00164] Preferred substituents on heteroaryl can be selected from the group consisting of -OH, -SH, nitro, halogen, amino, cyano, Cj-Cn alkyl, C2-Ci2 alkenyl, C2-C12 alkynyl, Ci-C12 alkoxy, Cj-Cn haloalkyl, Cj-Cn haloalkoxy and Ci-C thioalkoxy. Preferred substituents on alkyl, alkylene and heterocyclyl include the preferred substituents on heteroaryl and oxo. In one embodiment, the substituent on an alkyl, alkylene, heterocyclyl or heteroaryl is an amino group having the formula -N(R)2, wherein each R is independently selected from hydrogen and Ci-C4 alkyl.
[00165] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases.
Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, trifluoroacetic acid (2,2,2-trifiuoroacetic acid), oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate,
benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate,
trifluoroacetate (2,2,2-trifluoroacetate), undecanoate, valerate salts, and the like.
[00166] Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(Ci_4alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further
pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
[00167] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereoisomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
[00168] Unless specifically indicated (by a chemical name or other indicator designating double bond geometry, for example), each of Structural Formulas II and III (Structural Formula II, Ila, lib, etc.), used herein, is meant to include compounds having a carbon-carbon double bond (e.g. , an exocyclic double bond) with a configuration that is cis (or Z), trans (or E), or a mixture of cis and trans. For exam le, Structural Formula II:
Figure imgf000046_0001
Figure imgf000047_0001
, and mixtures thereof. Similarly, the following structural formula:
Figure imgf000048_0001
is meant to denote both and
Figure imgf000048_0002
, and mixtures thereof.
[00169] As used herein, "exocyclic double bond" refers to the carbon-carbon double bond in a compound of Structural Formula II and a compound of Structural Formula III indicated
with an arrow in the following structures:
Figure imgf000048_0003
respectively. In some embodiments described herein, the exocyclic double bond is in a cis configuration. In other embodiments, the exocyclic double bond is in a trans configuration.
[00170] The configuration of a carbon-carbon double bond, typically an exocyclic double bond in a compound of Structural Formula II or III, can be established by x-ray
crystallography. The application reflects whether a carbon-carbon double bond in a particular compound exists in a cis or trans configuration by indicating the configuration of the double bond in the chemical name associated with the compound.
[00171] As used herein, "cis" or "cis configuration" refers to a carbon-carbon double bond, typically an exocyclic double bond, that is predominantly cis. In some embodiments, greater than about 85% of compound molecules in a mixture of the compound have a carbon- carbon double bond (e.g. , an exocyclic double bond) that is cis. In some embodiments, greater than about 90%, greater than about 95%, greater than about 98%, greater than about 99%, greater than about 99.5% or greater than about 99.8% of compound molecules in a mixture of the compound have a carbon-carbon double bond (e.g. , an exocyclic double bond) that is cis.
[00172] As used herein, "trans" or "trans configuration" refers to a carbon-carbon double bond, typically an exocyclic double bond, that is predominantly trans. In some embodiments, greater than about 85% of compound molecules in a mixture of the compound have a carbon- carbon double bond (e.g. , an exocyclic double bond) that is cis. In some embodiments, greater than about 90%), greater than about 95%, greater than about 98%>, greater than about 99%), greater than about 99.5% or greater than about 99.8%) of compound molecules in a mixture of the compound have a carbon-carbon double bond (e.g. , an exocyclic double bond) that is cis.
[00173] Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention.
[00174] The term "pharmaceutically acceptable salt" means either an acid addition salt or a basic addition salt which is compatible with the treatment of patients.
[00175] In some embodiments, exemplary inorganic acids which form suitable salts include, but are not limited thereto, hydrochloric, hydrobromic, sulfuric and phosphoric acid and acid metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate. Illustrative organic acids which form suitable salts include the mono-, di- and tricarboxylic acids. Illustrative of such acids are, for example, acetic, trifluoro acetic acid (2,2,2-trifluoroacetic acid), glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxybenzoic,
phenylacetic, cinnamic, salicylic, 2-phenoxybenzoic, p-toluenesulfonic acid and other sulfonic acids such as methanesulfonic acid and 2-hydroxyethanesulfonic acid. Either the mono- or di-acid salts can be formed, and such salts can exist in either a hydrated, solvated or substantially anhydrous form. In general, the acid addition salts of these compounds are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms. Other non-pharmaceutically acceptable salts, e.g., oxalates may be used, for example, in the isolation of compounds described herein for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
[00176] A "pharmaceutically acceptable basic addition salt" is any non-toxic organic or inorganic base addition salt of the acid compounds described herein or any of its
intermediates. Illustrative inorganic bases which form suitable salts include, but are not limited thereto, lithium, sodium, potassium, calcium, magnesium or barium hydroxides. Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as methylamine, trimethyl amine and picoline or ammonia. The selection of the appropriate salt may be important so that an ester functionality, if any, elsewhere in the molecule is not hydrolyzed. The selection criteria for the appropriate salt will be known to one skilled in the art.
[00177] Acid addition salts of the compounds described herein are most suitably formed from pharmaceutically acceptable acids, and include, for example, those formed with inorganic acids, e.g., hydrochloric, sulphuric or phosphoric acids and organic acids, e.g., succinic, maleic, acetic, trifluoroacetic or fumaric acid. Other non-pharmaceutically acceptable salts, e.g., oxalates may be used for example in the isolation of compounds described herein for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt. Also included within the scope of the invention are base addition salts (such as sodium, potassium and ammonium salts), solvates and hydrates of compounds of the invention. The conversion of a given compound salt to a desired compound salt is achieved by applying standard techniques, well known to one skilled in the art.
[00178] The term "stereoisomers" is a general term for all isomers of the individual molecules that differ only in the orientation of their atoms in space. It includes mirror image isomers (enantiomers), geometric (cis/trans) isomers and isomers of compounds with more than one chiral centre that are not mirror images of one another (diastereomers).
[00179] The term "treat" or "treating" means to alleviate symptoms, eliminate the causation of the symptoms either on a temporary or permanent basis, or to prevent or slow the appearance of symptoms of the named disorder or condition. In the case of wound healing, a therapeutically effective amount is an amount that promotes healing of a wound.
[00180] As used herein, "promoting wound healing" means treating a subject with a wound and achieving healing, either partially or fully, of the wound. Promoting wound healing can mean, e.g., one or more of the following: promoting epidermal closure;
promoting migration of the dermis; promoting dermal closure in the dermis; reducing wound healing complications, e.g., hyperplasia of the epidermis and adhesions; reducing wound dehiscence; and promoting proper scab formation.
[00181] The term "therapeutically effective amount" means an amount of the compound which is effective in treating or lessening the severity of one or more symptoms of a disorder or condition.
[00182] The term "pharmaceutically acceptable carrier" means a non-toxic solvent, dispersant, excipient, adjuvant or other material which is mixed with the active ingredient in order to permit the formation of a pharmaceutical composition, i.e., a dosage form capable of being administered to a subject. One example of such a carrier is pharmaceutically acceptable oil typically used for parenteral administration. Pharmaceutically acceptable carriers are well known in the art.
[00183] When introducing elements disclosed herein, the articles "a", "an", "the", and "said" are intended to mean that there are one or more of the elements. The terms
"comprising", "having", "including" are intended to be open-ended and mean that there may be additional elements other than the listed elements.
Uses, Formulation and Administration
Pharmaceutically Acceptable Compositions
[00184] According to another embodiment, the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in compositions of this invention is such that is effective to measurably inhibit CRM1, in a biological sample or in a patient. In certain embodiments, a composition of this invention is formulated for administration to a patient in need of such composition. The term "patient", as used herein, means an animal. In some embodiments, the animal is a mammal. In certain embodiments, the patient is a veterinary patient (i.e., a non-human mammal patient). In some embodiments, the patient is a dog. In other embodiments, the patient is a human.
[00185] The term "pharmaceutically acceptable carrier, adjuvant, or vehicle" refers to a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose- based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
[00186] Compositions of the present invention may be administered orally, parenterally (including subcutaneous, intramuscular, intravenous and intradermal), by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. In some embodiments, provided compounds or compositions are administrable intravenously and/or intraperitoneally.
[00187] The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intraocular, intravitreal, intra-articular, intra- synovial, intrasternal, intrathecal, intrahepatic, intraperitoneal intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, subcutaneously, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are
conventionally employed as a solvent or suspending medium.
[00188] Pharmaceutically acceptable compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added. In some embodiments, a provided oral formulation is formulated for immediate release or sustained/delayed release. In some embodiments, the composition is suitable for buccal or sublingual administration, including tablets, lozenges and pastilles. A provided compound can also be in microencapsulated form.
[00189] Alternatively, pharmaceutically acceptable compositions of this invention may be administered in the form of suppositories for rectal administration. Pharmaceutically acceptable compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
[00190] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically- transdermal patches may also be used.
[00191] For ophthalmic use, provided pharmaceutically acceptable compositions may be formulated as micronized suspensions or in an ointment such as petrolatum.
[00192] Pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation.
[00193] In some embodiments, pharmaceutically acceptable compositions of this invention are formulated for intra-peritoneal administration.
[00194] The amount of compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. In one embodiment, provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions. In another embodiment, the dosage is from about 0.5 to about 100 mg/kg of body weight, or between 1 mg and 1000 mg/dose, every 4 to 120 hours, or according to the requirements of the particular drug. Typically, the pharmaceutical compositions of this invention will be administered from about 1 to about 6 times per day.
[00195] It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
[00196] Upon improvement of a patient's condition, a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary.
Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms
Uses of Compounds and Pharmaceutically Acceptable Compositions
[00197] Compounds and compositions described herein are generally useful for the inhibition of CRM1 and are therefore useful for treating one or more disorders associated with activity of CRM1. Thus, in certain embodiments, the present invention provides a method for treating a CRM 1 -mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof. The compounds and compositions described herein can also be administered to cells in culture, e.g. in vitro or ex vivo, or to a subject, e.g., in vivo, to treat, prevent, and/or diagnose a variety of disorders, including those described herein below.
[00198] The activity of a compound utilized in this invention as an inhibitor of CRM1 may be assayed in vitro, in vivo or in a cell line. Detailed conditions for assaying a compound utilized in this invention as an inhibitor of CRM1 are set forth in the Examples below.
[00199] As used herein, the term "CRM1 -mediated" disorder or condition, as used herein, means any disease or other deleterious condition in which CRM1 is known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which CRM1 is known to play a role. In some embodiments, the present invention provides methods of treating a disease associated with expression or activity of p53, p73, p21, pRB, p27, MB, NFKB, c-Abl, FOXO proteins, COX- 2, or an HDAC (histone deacetylases) in a subject comprising administering to the patient a therapeutically effective amount of a compound described herein. In another embodiment, the present invention relates to a method of treating or lessening the severity of a disease or condition selected from a proliferative disorder (e.g., cancer), an inflammatory disorder, an autoimmune disorder, a viral infection, an ophthalmological disorder or a neurodegenerative disorder wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention. In a more specific
embodiment, the present invention relates to a method of treating or lessening the severity of cancer. Specific examples of the above disorders are set forth in detail below.
[00200] Cancers treatable by the compounds of this invention include, but are not limited to, hematologic malignancies (leukemias, lymphomas, myelomas including multiple myeloma, myelodysplastic and myeloproliferative syndromes) and solid tumors (carcinomas such as prostate, breast, lung, colon, pancreatic, renal, ovarian as well as soft tissue and osteosarcomas, and stromal tumors). Breast cancer (BC) can include basal-like breast cancer (BLBC), triple negative breast cancer (TNBC) and breast cancer that is both BLBC and TNBC. In addition, breast cancer can include invasive or non-invasive ductal or lobular carcinoma, tubular, medullary, mucinous, papillary, cribriform carcinoma of the breast, male breast cancer, recurrent or metastatic breast cancer, phyllodes tumor of the breast and Paget' s disease of the nipple.
[00201] Inflammatory disorders treatable by the compounds of this invention include, but are not limited to, multiple sclerosis, rheumatoid arthritis, degenerative joint disease, systemic lupus, systemic sclerosis, vasculitis syndromes (small, medium and large vessel), atherosclerosis, inflammatory bowel disease, irritable bowel syndrome, Crohn's disease, mucous colitis, ulcerative colitis, gastritis, sepsis, psoriasis and other dermatological inflammatory disorders (such as eczema, atopic dermatitis, contact dermatitis, urticaria, scleroderma, and dermatosis with acute inflammatory components, pemphigus, pemphigoid, allergic dermatitis), and urticarial syndromes.
[00202] Viral diseases treatable by the compounds of this invention include, but are not limited to, acute febrile pharyngitis, pharyngoconjunctival fever, epidemic
keratoconjunctivitis, infantile gastroenteritis, Coxsackie infections, infectious mononucleosis, Burkitt lymphoma, acute hepatitis, chronic hepatitis, hepatic cirrhosis, hepatocellular carcinoma, primary HSV-1 infection (e.g., gingivostomatitis in children, tonsillitis and pharyngitis in adults, keratoconjunctivitis), latent HSV-1 infection (e.g., herpes labialis and cold sores), primary HSV-2 infection, latent HSV-2 infection, aseptic meningitis, infectious mononucleosis, Cytomegalic inclusion disease, Kaposi's sarcoma, multicentric Castleman disease, primary effusion lymphoma, AIDS, influenza, Reye syndrome, measles,
postinfectious encephalomyelitis, Mumps, hyperplastic epithelial lesions (e.g., common, flat, plantar and anogenital warts, laryngeal papillomas, epidermodysplasia verruciformis), cervical carcinoma, squamous cell carcinomas, croup, pneumonia, bronchiolitis, common cold, Poliomyelitis, Rabies, influenza-like syndrome, severe bronchiolitis with pneumonia, German measles, congenital rubella, Varicella, and herpes zoster. Viral diseases treatable by the compounds of this invention also include chronic viral infections, including hepatitis B and hepatitis C.
[00203] Exemplary ophthalmology disorders include, but are not limited to, macular edema (diabetic and nondiabetic macular edema), aged related macular degeneration wet and dry forms, aged disciform macular degeneration, cystoid macular edema, palpebral edema, retina edema, diabetic retinopathy, chorioretinopathy, neovascular maculopathy, neovascular glaucoma, uveitis, iritis, retinal vasculitis, endophthalmitis, panophthalmitis, metastatic ophthalmia, choroiditis, retinal pigment epitheliitis, conjunctivitis, cyclitis, scleritis, episcleritis, optic neuritis, retrobulbar optic neuritis, keratitis, blepharitis, exudative retinal detachment, corneal ulcer, conjunctival ulcer, chronic nummular keratitis, ophthalmic disease associated with hypoxia or ischemia, retinopathy of prematurity, proliferative diabetic retinopathy, polypoidal choroidal vasculopathy, retinal angiomatous proliferation, retinal artery occlusion, retinal vein occlusion, Coats' disease, familial exudative vitreoretinopathy, pulseless disease (Takayasu's disease), Eales disease, antiphospholipid antibody syndrome, leukemic retinopathy, blood hyperviscosity syndrome, macroglobulinemia, interferon- associated retinopathy, hypertensive retinopathy, radiation retinopathy, corneal epithelial stem cell deficiency or cataract.
[00204] Neurodegenerative diseases treatable by a compound of Formula I include, but are not limited to, Parkinson's, Alzheimer's, and Huntington's, and Amyotrophic lateral sclerosis (ALS/Lou Gehrig's Disease). [00205] Compounds and compositions described herein may also be used to treat disorders of abnormal tissue growth and fibrosis including dilative cardiomyopathy, hypertrophic cardiomyopathy, restrictive cardiomyopathy, pulmonary fibrosis, hepatic fibrosis,
glomerulonephritis, polycystic kidney disorder (PKD) and other renal disorders.
[00206] Compounds and compositions described herein may also be used to treat disorders related to food intake such as obesity and hyperphagia.
[00207] In another embodiment, a compound or composition described herein may be used to treat or prevent allergies and respiratory disorders, including asthma, bronchitis, pulmonary fibrosis, allergic rhinitis, oxygen toxicity, emphysema, chronic bronchitis, acute respiratory distress syndrome, and any chronic obstructive pulmonary disease (COPD).
[00208] In some embodiments, the disorder or condition associated with CRM1 activity is muscular dystrophy, arthritis, for example, osteoarthritis and rheumatoid arthritis, ankylosing spondilitis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, musclewasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary
hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, behcet's disease, incontinentia pigmenti, tuberculosis, asthma, Crohn's disease, colitis, ocular allergy, appendicitis, paget's disease, pancreatitis, periodonitis, endometriosis, inflammatory bowel disease, inflammatory lung disease, silica-induced diseases, sleep apnea, AIDS, HIV-1, autoimmune diseases, antiphospholipid syndrome, lupus, lupus
nephritis, familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy,
inflammatory neuropathy, parkinson's disease, multiple sclerosis, alzheimer's disease, amyotropic lateral sclerosis, huntington's disease, cataracts, or hearing loss.
[00209] In other embodiments, the disorder or condition associated with CRM1 activity is head injury, uveitis, inflammatory pain, allergen induced asthma, non-allergen induced asthma, glomerular nephritis, ulcerative colitis, necrotizing enterocolitis, hyperimmunoglobulinemia D with recurrent fever (HIDS), TNF receptor associated periodic syndrome (TRAPS), cryopyrin-associated periodic syndromes, Muckle- Wells syndrome (urticaria deafness amyloidosis),familial cold urticaria, neonatal onset multisystem inflammatory disease (NOMID), periodic fever, aphthous stomatitis, pharyngitis and adenitis (PFAPA syndrome), Blau syndrome, pyogenic sterile arthritis, pyoderma gangrenosum,acne (PAPA), deficiency of the interleukin-1 -receptor antagonist (DIRA), subarachnoid hemorrhage, polycystic kidney disease, transplant, organ transplant, tissue transplant, myelodysplastic syndrome, irritant-induced inflammation, plant irritant-induced
inflammation, poison ivy/ urushiol oil-induced inflammation, chemical irritant-induced inflammation, bee sting-induced inflammation, insect bite-induced inflammation, sunburn, burns, dermatitis, endotoxemia, lung injury, acute respiratory distress syndrome, alcoholic hepatitis, or kidney injury caused by parasitic infections.
[00210] In further aspects, the present invention provides a use of a compound described herein for the manufacture of a medicament for the treatment of a disease associated with expression or activity of p53, p73, p21, pRB, p27, ΙκΒ, NFKB, c-Abl, FOXO proteins, COX- 2 or an HDAC in a subject. In some embodiments, the present invention provides a use of a compound described herein in the manufacture of a medicament for the treatment of any of cancer and/or neoplastic disorders, angiogenesis, autoimmune disorders, inflammatory disorders and/or diseases, epigenetics, hormonal disorders and/or diseases, viral diseases, neurodegenerative disorders and/or diseases, wounds, and ophthalmologic disorders.
[00211] In some embodiments, the present invention provides a method for inhibiting CRM1 in a biological sample comprising contacting the biological sample with, or administering to the patient, a pharmaceutically acceptable salt of a compound of the invention, or pharmaceutically acceptable composition thereof.
Neoplastic Disorders
[00212] A compound or composition described herein can be used to treat a neoplastic disorder. A "neoplastic disorder" is a disease or disorder characterized by cells that have the capacity for autonomous growth or replication, e.g., an abnormal state or condition characterized by proliferative cell growth. Exemplary neoplastic disorders include:
carcinoma, sarcoma, metastatic disorders, e.g., tumors arising from prostate, brain, bone, colon, lung, breast, ovarian, and liver origin, hematopoietic neoplastic disorders, e.g., leukemias, lymphomas, myeloma and other malignant plasma cell disorders, and metastatic tumors. Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers. Treatment with the compound can be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
[00213] The disclosed methods are useful in the prevention and treatment of cancer, including for example, solid tumors, soft tissue tumors, and metastases thereof, as well as in familial cancer syndromes such as Li Fraumeni Syndrome. Familial Breast-Ovarian Cancer (BRCA1 or BRAC2 mutations) Syndromes, and others. The disclosed methods are also useful in treating non-solid cancers. Exemplary solid tumors include malignancies (e.g., sarcomas, adenocarcinomas, and carcinomas) of the various organ systems, such as those of lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary (e.g., renal, urothelial, or testicular tumors) tracts, pharynx, prostate, and ovary. Exemplary adenocarcinomas include colorectal cancers, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, and cancer of the small intestine.
[00214] Exemplary cancers described by the National Cancer Institute include: Acute Lymphoblastic Leukemia, Adult; Acute Lymphoblastic Leukemia, Childhood; Acute
Myeloid Leukemia, Adult; Adrenocortical Carcinoma; Adrenocortical Carcinoma,
Childhood; AIDS-Related Lymphoma; AIDS-Related Malignancies; Anal Cancer;
Astrocytoma, Childhood Cerebellar; Astrocytoma, Childhood Cerebral; Bile Duct Cancer, Extrahepatic; Bladder Cancer; Bladder Cancer, Childhood; Bone Cancer,
Osteosarcoma/Malignant Fibrous Histiocytoma; Brain Stem Glioma, Childhood; Brain Tumor, Adult; Brain Tumor, Brain Stem Glioma, Childhood; Brain Tumor, Cerebellar Astrocytoma, Childhood; Brain Tumor, Cerebral Astrocytoma/Malignant Glioma, Childhood; Brain Tumor, Ependymoma, Childhood; Brain Tumor, Medulloblastoma, Childhood; Brain Tumor, Supratentorial Primitive Neuroectodermal Tumors, Childhood; Brain Tumor, Visual Pathway and Hypothalamic Glioma, Childhood; Brain Tumor, Childhood (Other); Breast Cancer; Breast Cancer and Pregnancy; Breast Cancer, Childhood; Breast Cancer, Male;
Bronchial Adenomas/Carcinoids, Childhood; Carcinoid Tumor, Childhood; Carcinoid Tumor, Gastrointestinal; Carcinoma, Adrenocortical; Carcinoma, Islet Cell; Carcinoma of Unknown Primary; Central Nervous System Lymphoma, Primary; Cerebellar Astrocytoma, Childhood; Cerebral Astrocytoma/Malignant Glioma, Childhood; Cervical Cancer;
Childhood Cancers; Chronic Lymphocytic Leukemia; Chronic Myelogenous Leukemia; Chronic Myeloproliferative Disorders; Clear Cell Sarcoma of Tendon Sheaths; Colon Cancer; Colorectal Cancer, Childhood; Cutaneous T-Cell Lymphoma; Endometrial Cancer;
Ependymoma, Childhood; Epithelial Cancer, Ovarian; Esophageal Cancer; Esophageal Cancer, Childhood; Ewing's Family of Tumors; Extracranial Germ Cell Tumor, Childhood; Extragonadal Germ Cell Tumor; Extrahepatic Bile Duct Cancer; Eye Cancer, Intraocular Melanoma; Eye Cancer, Retinoblastoma; Gallbladder Cancer; Gastric (Stomach) Cancer; Gastric (Stomach) Cancer, Childhood; Gastrointestinal Carcinoid Tumor; Germ Cell Tumor, Extracranial, Childhood; Germ Cell Tumor, Extragonadal; Germ Cell Tumor, Ovarian;
Gestational Trophoblastic Tumor; Glioma, Childhood Brain Stem; Glioma, Childhood Visual Pathway and Hypothalamic; Hairy Cell Leukemia; Head and Neck Cancer; Hepatocellular (Liver) Cancer, Adult (Primary); Hepatocellular (Liver) Cancer, Childhood (Primary);
Hodgkin's Lymphoma, Adult; Hodgkin's Lymphoma, Childhood; Hodgkin's Lymphoma During Pregnancy; Hypopharyngeal Cancer; Hypothalamic and Visual Pathway Glioma, Childhood; Intraocular Melanoma; Islet Cell Carcinoma (Endocrine Pancreas); Kaposi's Sarcoma; Kidney Cancer; Laryngeal Cancer; Laryngeal Cancer, Childhood; Leukemia, Acute Lymphoblastic, Adult; Leukemia, Acute Lymphoblastic, Childhood; Leukemia, Acute Myeloid, Adult; Leukemia, Acute Myeloid, Childhood; Leukemia, Chronic Lymphocytic; Leukemia, Chronic Myelogenous; Leukemia, Hairy Cell; Lip and Oral Cavity Cancer; Liver Cancer, Adult (Primary); Liver Cancer, Childhood (Primary); Lung Cancer, Non-Small Cell; Lung Cancer, Small Cell; Lymphoblastic Leukemia, Adult Acute; Lymphoblastic Leukemia, Childhood Acute; Lymphocytic Leukemia, Chronic; Lymphoma, AIDS- Related;
Lymphoma, Central Nervous System (Primary); Lymphoma, Cutaneous T-Cell; Lymphoma, Hodgkin's, Adult; Lymphoma, Hodgkin's, Childhood; Lymphoma, Hodgkin's During
Pregnancy; Lymphoma, Non-Hodgkin's, Adult; Lymphoma, Non- Hodgkin's, Childhood; Lymphoma, Non-Hodgkin's During Pregnancy; Lymphoma, Primary Central Nervous System; Macroglobulinemia, Waldenstrom's; Male Breast Cancer; Malignant Mesothelioma, Adult; Malignant Mesothelioma, Childhood; Malignant Thymoma; Medulloblastoma, Childhood; Melanoma; Melanoma, Intraocular; Merkel Cell Carcinoma; Mesothelioma, Malignant; Metastatic Squamous Neck Cancer with Occult Primary; Multiple Endocrine Neoplasia Syndrome, Childhood; Multiple Myeloma/Plasma Cell Neoplasm; Mycosis Fungoides; Myelodysplastic Syndromes; Myelogenous Leukemia, Chronic; Myeloid
Leukemia, Childhood Acute; Myeloma, Multiple; Myeloproliferative Disorders, Chronic; Nasal Cavity and Paranasal Sinus Cancer; Nasopharyngeal Cancer; Nasopharyngeal Cancer, Childhood; Neuroblastoma; Non-Hodgkin's Lymphoma, Adult; Non-Hodgkin's Lymphoma, Childhood; Non- Hodgkin's Lymphoma During Pregnancy; Non-Small Cell Lung Cancer; Oral Cancer, Childhood; Oral Cavity and Lip Cancer; Oropharyngeal Cancer;
Osteo sarcoma/Malignant Fibrous Histiocytoma of Bone; Ovarian Cancer, Childhood;
Ovarian Epithelial Cancer; Ovarian Germ Cell Tumor; Ovarian Low Malignant Potential Tumor; Pancreatic Cancer; Pancreatic Cancer, Childhood; Pancreatic Cancer, Islet Cell;
Paranasal Sinus and Nasal Cavity Cancer; Parathyroid Cancer; Penile Cancer;
Pheochromocytoma; Pineal and Supratentorial Primitive Neuroectodermal Tumors,
Childhood; Pituitary Tumor; Plasma Cell Neoplasm/Multiple Myeloma; Pleuropulmonary Blastoma; Pregnancy and Breast Cancer; Pregnancy and Hodgkin's Lymphoma; Pregnancy and Non-Hodgkin's Lymphoma; Primary Central Nervous System Lymphoma; Primary Liver Cancer, Adult; Primary Liver Cancer, Childhood; Prostate Cancer; Rectal Cancer; Renal Cell (Kidney) Cancer; Renal Cell Cancer, Childhood; Renal Pelvis and Ureter, Transitional Cell Cancer; Retinoblastoma; Rhabdomyosarcoma, Childhood; Salivary Gland Cancer; Salivary Gland Cancer, Childhood; Sarcoma, Ewing's Family of Tumors; Sarcoma, Kaposi's; Sarcoma (Osteo sarcoma)/Malignant Fibrous Histiocytoma of Bone; Sarcoma, Rhabdomyosarcoma, Childhood; Sarcoma, Soft Tissue, Adult; Sarcoma, Soft Tissue, Childhood; Sezary
Syndrome; Skin Cancer; Skin Cancer, Childhood; Skin Cancer (Melanoma); Skin Carcinoma, Merkel Cell; Small Cell Lung Cancer; Small Intestine Cancer; Soft Tissue Sarcoma, Adult; Soft Tissue Sarcoma, Childhood; Squamous Neck Cancer with Occult Primary, Metastatic; Stomach (Gastric) Cancer; Stomach (Gastric) Cancer, Childhood; Supratentorial Primitive Neuroectodermal Tumors, Childhood; T- Cell Lymphoma, Cutaneous; Testicular Cancer; Thymoma, Childhood; Thymoma, Malignant; Thyroid Cancer; Thyroid Cancer, Childhood; Transitional Cell Cancer of the Renal Pelvis and Ureter; Trophoblastic Tumor, Gestational; Unknown Primary Site, Cancer of, Childhood; Unusual Cancers of Childhood; Ureter and Renal Pelvis, Transitional Cell Cancer; Urethral Cancer; Uterine Sarcoma; Vaginal Cancer; Visual Pathway and Hypothalamic Glioma, Childhood; Vulvar Cancer; Waldenstrom's Macro globulinemia; and Wilms' Tumor.
[00215] Further exemplary cancers include diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL). Yet further exemplary cancers include endocervical cancer, B- cell ALL, T-cell ALL, B- or T-cell lymphoma, mast cell cancer, glioblastoma,
neuroblastoma, follicular lymphoma and Richter's syndrome. [00216] Exemplary sarcomas include fibrosarcoma, alveolar soft part sarcoma (ASPS), liposarcoma, leiomyosarcoma, chondrosarcoma, synovial sarcoma, chordoma, spindle cell sarcoma, histiocytoma, rhabdomyosarcoma, Ewing's sarcoma, neuroectodermal sarcoma, phyllodes/osteogenic sarcoma and chondroblastic osteosarcoma.
[00217] Metastases of the aforementioned cancers can also be treated or prevented in accordance with the methods described herein.
Combination therapies
[00218] In some embodiments, a compound described herein is administered together with an additional "second" therapeutic agent or treatment. The choice of second therapeutic agent may be made from any agent that is typically used in a monotherapy to treat the indicated disease or condition. As used herein, the term "administered together" and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present invention provides a single unit dosage form comprising a compound of the invention, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
[00219] In one embodiment of the invention, where a second therapeutic agent is administered to a subject, the effective amount of the compound of this invention is less than its effective amount would be where the second therapeutic agent is not administered. In another embodiment, the effective amount of the second therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized. Other potential advantages (including without limitation improved dosing regimens and/or reduced drug cost) will be apparent to those of skill in the art. The additional agents may be administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds of this invention in a single composition.
Cancer Combination Therapies
[00220] In some embodiments, a compound described herein is administered together with an additional cancer treatment. Exemplary additional cancer treatments include, for example: chemotherapy, targeted therapies such as antibody therapies, kinase inhibitors, immunotherapy, and hormonal therapy, epigenetic therapy, proteosome inhibitors, and anti- angiogenic therapies. Examples of each of these treatments are provided below. As used herein, the term "combination," "combined," and related terms refer to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a compound of the present invention can be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present invention provides a single unit dosage form comprising a compound of the invention, an additional therapeutic agent, and a
pharmaceutically acceptable carrier, adjuvant, or vehicle.
[00221] The amount of both a compound of the invention and additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that can be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Preferably, compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of a compound of the invention can be administered.
Chemotherapy
[00222] In some embodiments, a compound described herein is administered with a chemotherapy. Chemotherapy is the treatment of cancer with drugs that can destroy cancer cells. "Chemotherapy" usually refers to cytotoxic drugs which affect rapidly dividing cells in general, in contrast with targeted therapy. Chemotherapy drugs interfere with cell division in various possible ways, e.g., with the duplication of DNA or the separation of newly formed chromosomes. Most forms of chemotherapy target all rapidly dividing cells and are not specific for cancer cells, although some degree of specificity may come from the inability of many cancer cells to repair DNA damage, while normal cells generally can.
[00223] Examples of chemotherapeutic agents used in cancer therapy include, for example, antimetabolites (e.g., folic acid, purine, and pyrimidine derivatives) and alkylating agents (e.g., nitrogen mustards, nitrosoureas, platinum, alkyl sulfonates, hydrazines, triazenes, aziridines, spindle poison, cytotoxic agents, topoisomerase inhibitors and others). Exemplary agents include Aclarubicin, Actinomycin, Alitretinoin, Altretamine, Aminopterin, Aminolevulinic acid, Amrubicin, Amsacrine, Anagrelide, Arsenic trioxide, Asparaginase, Atrasentan, Belotecan, Bexarotene, Bendamustin, Bleomycin, Bortezomib, Busulfan, Camptothecin, Capecitabine, Carboplatin, Carboquone, Carmofur, Carmustine, Celecoxib, Chlorambucil, Chlormethine, Cisplatin, Cladribine, Clofarabine, Crisantaspase, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Daunorubicin, Decitabine, Demecolcine, Docetaxel, Doxorubicin, Efaproxiral, Elesclomol, Elsamitrucin, Enocitabine, Epirubicin, Estramustine, Etoglucid, Etoposide, Floxuridine, Fludarabine, Fluorouracil (5FU), Fotemustine, Gemcitabine, Gliadel implants, Hydroxycarbamide, Hydroxyurea, Idarubicin, Ifosfamide, Irinotecan, Irofulven, Ixabepilone, Larotaxel, Leucovorin, Liposomal doxorubicin, Liposomal daunorubicin, Lonidamine, Lomustine, Lucanthone, Mannosulfan, Masoprocol, Melphalan, Mercaptopurine, Mesna, Methotrexate, Methyl aminolevulinate, Mitobronitol, Mitoguazone, Mitotane, Mitomycin, Mitoxantrone, Nedaplatin, Nimustine, Oblimersen, Omacetaxine, Ortataxel, Oxaliplatin, Paclitaxel, Pegaspargase, Pemetrexed, Pentostatin, Pirarubicin, Pixantrone, Plicamycin, Porfimer sodium, Prednimustine,
Procarbazine, Raltitrexed, Ranimustine, Rubitecan, Sapacitabine, Semustine, Sitimagene ceradenovec, Strataplatin, Streptozocin, Talaporfin, Tegafur-uracil, Temoporfin,
Temozolomide, Teniposide, Tesetaxel, Testolactone, Tetranitrate, Thiotepa, Tiazofurine, Tioguanine, Tipifarnib, Topotecan, Trabectedin, Triaziquone, Triethylenemelamine,
Triplatin, Tretinoin, Treosulfan, Trofosfamide, Uramustine, Valrubicin, Verteporfin, Vinblastine, Vincristine, Vindesine, Vinflunine, Vinorelbine, Vorinostat, Zorubicin, and other cytostatic or cytotoxic agents described herein.
[00224] Because some drugs work better together than alone, two or more drugs are often given at the same time. Often, two or more chemotherapy agents are used as combination chemotherapy. In some embodiments, the chemotherapy agents (including combination chemotherapy) can be used in combination with a compound described herein.
Targeted therapy
[00225] Targeted therapy constitutes the use of agents specific for the deregulated proteins of cancer cells. Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell. Prominent examples are the tyrosine kinase inhibitors such as Axitinib, Bosutinib, Cediranib, desatinib, erolotinib, imatinib, gefitinib, lapatinib, Lestaurtinib, Nilotinib, Semaxanib, Sorafenib, Sunitinib, and Vandetanib, and also cyclin-dependent kinase inhibitors such as Alvocidib and Seliciclib. Monoclonal antibody therapy is another strategy in which the therapeutic agent is an antibody which specifically binds to a protein on the surface of the cancer cells. Examples include the anti-HER2/neu antibody trastuzumab (Herceptin®) typically used in breast cancer, and the anti-CD20 antibody rituximab and Tositumomab typically used in a variety of B-cell malignancies. Other exemplary antibodies include Cetuximab, Panitumumab, Trastuzumab, Alemtuzumab, Bevacizumab, Edrecolomab, and Gemtuzumab. Exemplary fusion proteins include Aflibercept and Denileukin diftitox. In some embodiments, the targeted therapy can be used in combination with a compound described herein, e.g., Gleevec (Vignari and Wang 2001).
[00226] Targeted therapy can also involve small peptides as '¾oming devices" which can bind to cell surface receptors or affected extracellular matrix surrounding the tumor.
Radionuclides which are attached to these peptides (e.g., RGDs) eventually kill the cancer cell if the nuclide decays in the vicinity of the cell. An example of such therapy includes BEXXAR®.
Angiogenesis
[00227] Compounds and methods described herein may be used to treat or prevent a disease or disorder associated with angiogenesis. Diseases associated with angiogenesis include cancer, cardiovascular disease and macular degeneration.
[00228] Angiogenesis is the physiological process involving the growth of new blood vessels from pre-existing vessels. Angiogenesis is a normal and vital process in growth and development, as well as in wound healing and in granulation tissue. However, it is also a fundamental step in the transition of tumors from a dormant state to a malignant one.
Angiogenesis may be a target for combating diseases characterized by either poor vascularisation or abnormal vasculature.
[00229] Application of specific compounds that may inhibit or induce the creation of new blood vessels in the body may help combat such diseases. The presence of blood vessels where there should be none may affect the mechanical properties of a tissue, increasing the likelihood of failure. The absence of blood vessels in a repairing or otherwise metabolically active tissue may inhibit repair or other essential functions. Several diseases, such as ischemic chronic wounds, are the result of failure or insufficient blood vessel formation and may be treated by a local expansion of blood vessels, thus bringing new nutrients to the site, facilitating repair. Other diseases, such as age-related macular degeneration, may be created by a local expansion of blood vessels, interfering with normal physiological processes.
[00230] Vascular endothelial growth factor (VEGF) has been demonstrated to be a major contributor to angiogenesis, increasing the number of capillaries in a given network. Upregulation of VEGF is a major component of the physiological response to exercise and its role in angiogenesis is suspected to be a possible treatment in vascular injuries. In vitro studies clearly demonstrate that VEGF is a potent stimulator of angiogenesis because, in the presence of this growth factor, plated endothelial cells will proliferate and migrate, eventually forming tube structures resembling capillaries.
[00231] Tumors induce blood vessel growth (angiogenesis) by secreting various growth factors {e.g., VEGF). Growth factors such as bFGF and VEGF can induce capillary growth into the tumor, which some researchers suspect supply required nutrients, allowing for tumor expansion.
[00232] Angiogenesis represents an excellent therapeutic target for the treatment of cardiovascular disease. It is a potent, physiological process that underlies the natural manner in which our bodies respond to a diminution of blood supply to vital organs, namely the production of new collateral vessels to overcome the ischemic insult.
[00233] Overexpression of VEGF causes increased permeability in blood vessels in addition to stimulating angiogenesis. In wet macular degeneration, VEGF causes proliferation of capillaries into the retina. Since the increase in angiogenesis also causes edema, blood and other retinal fluids leak into the retina, causing loss of vision.
[00234] Anti-angiogenic therapy can include kinase inhibitors targeting vascular endothelial growth factor (VEGF) such as sunitinib, sorafenib, or monoclonal antibodies or receptor "decoys" to VEGF or VEGF receptor including bevacizumab or VEGF-Trap, or thalidomide or its analogs (lenalidomide, pomalidomide), or agents targeting non-VEGF angiogenic targets such as fibroblast growth factor (FGF), angiopoietins, or angiostatin or endo statin.
Epigenetics
[00235] Compounds and methods described herein may be used to treat or prevent a disease or disorder associated with epigenetics. Epigenetics is the study of heritable changes in phenotype or gene expression caused by mechanisms other than changes in the underlying DNA sequence. One example of epigenetic changes in eukaryotic biology is the process of cellular differentiation. During morphogenesis, stem cells become the various cell lines of the embryo which in turn become fully differentiated cells. In other words, a single fertilized egg cell changes into the many cell types including neurons, muscle cells, epithelium, blood vessels etc. as it continues to divide. It does so by activating some genes while inhibiting others.
[00236] Epigenetic changes are preserved when cells divide. Most epigenetic changes only occur within the course of one individual organism's lifetime, but, if a mutation in the DNA has been caused in sperm or egg cell that results in fertilization, then some epigenetic changes are inherited from one generation to the next. Specific epigenetic processes include paramutation, bookmarking, imprinting, gene silencing, X chromosome inactivation, position effect, reprograrnming, transvection, maternal effects, the progress of carcinogenesis, many effects of teratogens, regulation of histone modifications and heterochromatin, and technical limitations affecting parthenogenesis and cloning.
[00237] Exemplary diseases associated with epigenetics include ATR-syndrome, fragile X-syndrome, ICF syndrome, Angelman's syndrome, Prader-Wills syndrome, BWS, Rett syndrome, ot-thalassaemia, cancer, leukemia, Rubinstein-Taybi syndrome and Coffin-Lowry syndrome.
[00238] The first human disease to be linked to epigenetics was cancer. Researchers found that diseased tissue from patients with colorectal cancer had less DNA methylation than normal tissue from the same patients. Because methylated genes are typically turned off, loss of DNA methylation can cause abnormally high gene activation by altering the arrangement of chromatin. On the other hand, too much methylation can undo the work of protective tumor suppressor genes.
[00239] DNA methylation occurs at CpG sites, and a majority of CpG cytosines are methylated in mammals. However, there are stretches of DNA near promoter regions that have higher concentrations of CpG sites (known as CpG islands) that are free of methylation in normal cells. These CpG islands become excessively methylated in cancer cells, thereby causing genes that should not be silenced to turn off. This abnormality is the trademark epigenetic change that occurs in tumors and happens early in the development of cancer. Hypermethylation of CpG islands can cause tumors by shutting off tumor-suppressor genes. In fact, these types of changes may be more common in human cancer than DNA sequence mutations.
[00240] Furthermore, although epigenetic changes do not alter the sequence of DNA, they can cause mutations. About half of the genes that cause familial or inherited forms of cancer are turned off by methylation. Most of these genes normally suppress tumor formation and help repair DNA, including 06-methylguanine-DNA methyltransferase (MGMT), MLH1 cyclin-dependent kinase inhibitor 2B (CDKN2B), and RASSF1A. For example,
hypermethylation of the promoter of MGMT causes the number of G-to-A mutations to increase.
[00241] Hypermethylation can also lead to instability of micro satellites, which are repeated sequences of DNA. Microsatellites are common in normal individuals, and they usually consist of repeats of the dinucleotide CA. Too much methylation of the promoter of the DNA repair gene MLH1 can make a micro satellite unstable and lengthen or shorten it. Microsatellite instability has been linked to many cancers, including colorectal, endometrial, ovarian, and gastric cancers.
[00242] Fragile X syndrome is the most frequently inherited mental disability, particularly in males. Both sexes can be affected by this condition, but because males only have one X chromosome, one fragile X will impact them more severely. Indeed, fragile X syndrome occurs in approximately 1 in 4,000 males and 1 in 8,000 females. People with this syndrome have severe intellectual disabilities, delayed verbal development, and "autistic-like" behavior.
[00243] Fragile X syndrome gets its name from the way the part of the X chromosome that contains the gene abnormality looks under a microscope; it usually appears as if it is hanging by a thread and easily breakable. The syndrome is caused by an abnormality in the FMR1 (fragile X mental retardation 1) gene. People who do not have fragile X syndrome have 6 to 50 repeats of the trinucleotide CGG in their FMR1 gene. However, individuals with over 200 repeats have a full mutation, and they usually show symptoms of the syndrome. Too many CGGs cause the CpG islands at the promoter region of the FMR1 gene to become
methylated; normally, they are not. This methylation turns the gene off, stopping the FMR1 gene from producing an important protein called fragile X mental retardation protein. Loss of this specific protein causes fragile X syndrome. Although a lot of attention has been given to the CGG expansion mutation as the cause of fragile X, the epigenetic change associated with FMR1 methylation is the real syndrome culprit.
[00244] Fragile X syndrome is not the only disorder associated with mental retardation that involves epigenetic changes. Other such conditions include Rubenstein-Taybi, Coffin- Lowry, Prader-Willi, Angelman, Beckwith- Wiedemann, ATR-X, and Rett syndromes.
[00245] Epigenetic therapies include inhibitors of enzymes controlling epigenetic modifications, specifically DNA methyltransferases and histone deacetylases, which have shown promising anti-tumorigenic effects for some malignancies, as well as antisense oligonucleotides and siRNA.
Immunotherapy
[00246] In some embodiments, a compound described herein is administered with an immunotherapy. Cancer immunotherapy refers to a diverse set of therapeutic strategies designed to induce the patient's own immune system to fight the tumor. Contemporary methods for generating an immune response against tumors include intravesicular BCG immunotherapy for superficial bladder cancer, prostate cancer vaccine Provenge, and use of interferons and other cytokines to induce an immune response in renal cell carcinoma and melanoma patients.
[00247] Allogeneic hematopoietic stem cell transplantation can be considered a form of immunotherapy, since the donor's immune cells will often attack the tumor in a graft-versus- tumor effect. In some embodiments, the immunotherapy agents can be used in combination with a compound described herein.
Hormonal therapy
[00248] In some embodiments, a compound described herein is administered with a hormonal therapy. The growth of some cancers can be inhibited by providing or blocking certain hormones. Common examples of hormone- sensitive tumors include certain types of breast and prostate cancers, as well as certain types of leukemia which respond to certain retinoids/retinoic acids. Removing or blocking estrogen or testosterone is often an important additional treatment. In certain cancers, administration of hormone agonists, such as progestogens may be therapeutically beneficial. In some embodiments, the hormonal therapy agents can be used in combination with a compound described herein.
[00249] Hormonal therapy agents include the administration of hormone agonists or hormone antagonists and include retinoids/retinoic acid, compounds that inhibit estrogen or testosterone, as well as administration of progestogens.
Inflammation and Autoimmune Disease
[00250] The compounds and methods described herein may be used to treat or prevent a disease or disorder associated with inflammation, particularly in humans and other mammals. A compound described herein may be administered prior to the onset of, at, or after the initiation of inflammation. When used prophylactically, the compounds are preferably provided in advance of any inflammatory response or symptom. Administration of the compounds can prevent or attenuate inflammatory responses or symptoms. Exemplary inflammatory conditions include, for example, multiple sclerosis, rheumatoid arthritis, psoriatic arthritis, degenerative joint disease, spondouloarthropathies, other seronegative inflammatory arthridities, polymyalgia rheumatica, various vasculidities (e.g., giant cell arteritis, ANCA+ vasculitis), gouty arthritis, systemic lupus erythematosus, juvenile arthritis, juvenile rheumatoid arthritis, osteoarthritis, osteoporosis, diabetes (e.g., insulin dependent diabetes mellitus or juvenile onset diabetes), menstrual cramps, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, Crohn's disease, mucous colitis, ulcerative colitis, gastritis, esophagitis, pancreatitis, peritonitis, Alzheimer's disease, shock, ankylosing spondylitis, gastritis, conjunctivitis, pancreatis (acute or chronic), multiple organ injury syndrome (e.g., secondary to septicemia or trauma), myocardial infarction, atherosclerosis, stroke, reperfusion injury (e.g., due to cardiopulmonary bypass or kidney dialysis), acute glomerulonephritis, thermal injury (i.e., sunburn), necrotizing enterocolitis, granulocyte transfusion associated syndrome, and/or Sjogren's syndrome. Exemplary inflammatory conditions of the skin include, for example, eczema, atopic dermatitis, contact dermatitis, urticaria, schleroderma, psoriasis, and dermatosis with acute inflammatory components.
[00251] In another embodiment, a compound or method described herein may be used to treat or prevent allergies and respiratory conditions, including asthma, bronchitis, pulmonary fibrosis, allergic rhinitis, oxygen toxicity, emphysema, chronic bronchitis, acute respiratory distress syndrome, and any chronic obstructive pulmonary disease (COPD). The compounds may be used to treat chronic hepatitis infection, including hepatitis B and hepatitis C.
[00252] Additionally, a compound or method described herein may be used to treat autoimmune diseases and/or inflammation associated with autoimmune diseases, such as organ-tissue autoimmune diseases (e.g., Raynaud's syndrome), scleroderma, myasthenia gravis, transplant rejection, endotoxin shock, sepsis, psoriasis, eczema, dermatitis, multiple sclerosis, autoimmune thyroiditis, uveitis, systemic lupus erythematosis, Addison's disease, autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), and Grave's disease. [00253] In a particular embodiment, the compounds described herein can be used to treat multiple sclerosis.
Combination therapy
[00254] In certain embodiments, a compound described herein may be administered alone or in combination with other compounds useful for treating or preventing inflammation. Exemplary anti-inflammatory agents include, for example, steroids {e.g., Cortisol, cortisone, fludrocortisone, prednisone, 6[alpha]-methylprednisone, triamcinolone, betamethasone or dexamethasone), nonsteroidal antiinflammatory drugs (NSAIDS (e.g., aspirin,
acetaminophen, tolmetin, ibuprofen, mefenamic acid, piroxicam, nabumetone, rofecoxib, celecoxib, etodolac or nimesulide). In another embodiment, the other therapeutic agent is an antibiotic (e.g. , vancomycin, penicillin, amoxicillin, ampicillin, cefotaxime, ceftriaxone, cefixime, rifampinmetronidazole, doxycycline or streptomycin). In another embodiment, the other therapeutic agent is a PDE4 inhibitor (e.g., roflumilast or rolipram). In another embodiment, the other therapeutic agent is an antihistamine (e.g. , cyclizine, hydroxyzine, promethazine or diphenhydramine). In another embodiment, the other therapeutic agent is an anti-malarial (e.g., artemisinin, artemether, artsunate, chloroquine phosphate, mefloquine hydrochloride, doxycycline hyclate, proguanil hydrochloride, atovaquone or halofantrine). In one embodiment, the other compound is drotrecogin alfa.
[00255] Further examples of anti-inflammatory agents include, for example, aceclofenac, acemetacin, e-acetamidocaproic acid, acetaminophen, acetaminosalol, acetanilide, acetylsalicylic acid, S-adenosylmethionine, alclofenac, alclometasone, alfentanil, algestone, allylprodine, alminoprofen, aloxiprin, alphaprodine, aluminum bis(acetylsalicylate), amcinonide, amfenac, aminochlorthenoxazin, 3-amino-4- hydroxybutyric acid, 2-amino-4- picoline, aminopropylon, aminopyrine, amixetrine, ammonium salicylate, ampiroxicam, amtolmetin guacil, anileridine, antipyrine, antrafenine, apazone, beclomethasone, bendazac, benorylate, benoxaprofen, benzpiperylon, benzydamine, benzylmorphine, bermoprofen, betamethasone, betamethasone- 17-valerate, bezitramide, [alpha] -bisabolol, bromfenac, p- bromoacetanilide, 5-bromosalicylic acid acetate, bromosaligenin, bucetin, bucloxic acid, bucolome, budesonide, bufexamac, bumadizon, buprenorphine, butacetin, butibufen, butorphanol, carbamazepine, carbiphene, caiprofen, carsalam, chlorobutanol,
chloroprednisone, chlorthenoxazin, choline salicylate, cinchophen, cinmetacin, ciramadol, clidanac, clobetasol, clocortolone, clometacin, clonitazene, clonixin, clopirac, cloprednol, clove, codeine, codeine methyl bromide, codeine phosphate, codeine sulfate, cortisone, cortivazol, cropropamide, crotethamide, cyclazocine, deflazacort, dehydrotestosterone, desomorphine, desonide, desoximetasone, dexamethasone, dexamethasone-21- isonicotinate, dexoxadrol, dextromoramide, dextropropoxyphene, deoxycorticosterone, dezocine, diampromide, diamorphone, diclofenac, difenamizole, difenpiramide, diflorasone, diflucortolone, diflunisal, difluprednate, dihydrocodeine, dihydrocodeinone enol acetate, dihydromorphine, dihydroxyaluminum acetylsalicylate, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, diprocetyl, dipyrone, ditazol, droxicam, emorfazone, enfenamic acid, enoxolone, epirizole, eptazocine, etersalate, ethenzamide, ethoheptazine, ethoxazene, ethylmethylthiambutene, ethylmorphine, etodolac, etofenamate, etonitazene, eugenol, felbinac, fenbufen, fenclozic acid, fendosal, fenoprofen, fentanyl, fentiazac, fepradinol, feprazone, floctafenine, fluazacort, flucloronide, flufenamic acid, flumethasone, flunisolide, flunixin, flunoxaprofen, fluocmolone acetonide, fluocinonide, fluocmolone acetonide, fluocortin butyl, fluocoitolone, fluoresone, fluorometholone, fluperolone, flupirtine, fluprednidene, fluprednisolone, fluproquazone, flurandrenolide, flurbiprofen, fluticasone, formocortal, fosfosal, gentisic acid, glafenine, glucametacin, glycol salicylate, guaiazulene, halcinonide, halobetasol, halometasone, haloprednone, heroin, hydrocodone, hydro cortamate, hydrocortisone, hydrocortisone acetate, hydrocortisone succinate, hydrocortisone hemi succinate, hydrocortisone 21 -lysinate, hydrocortisone cypionate, hydromorphone, hydroxypethidine, ibufenac, ibuprofen, ibuproxam, imidazole salicylate, indomethacin, indoprofen, isofezolac, isoflupredone, isoflupredone acetate, isoladol, isomethadone, isonixin, isoxepac, isoxicam, ketobemidone, ketoprofen, ketorolac, p- lactophenetide, lefetamine, levallorphan, levorphanol, levophenacyl-morphan, lofentanil, lonazolac, lornoxicam, loxoprofen, lysine acetylsalicylate, mazipredone, meclofenamic acid, medrysone, mefenamic acid, meloxicam, meperidine, meprednisone, meptazinol,
mesalamine, metazocine, methadone, methotrimeprazine, methylprednisolone,
methylprednisolone acetate, methylprednisolone sodium succinate, methylprednisolone suleptnate, metiazinic acid, metofoline, metopon, mofebutazone, mofezolac, mometasone, morazone, morphine, morphine hydrochloride, morphine sulfate, morpholine salicylate, myrophine, nabumetone, nalbuphine, nalorphine, 1-naphthyl salicylate, naproxen, narceine, nefopam, nicomorphine, nifenazone, niflumic acid, nimesulide, 5'-nitro-2'- propoxyacetanilide,norlevorphanol, normethadone, normorphine, norpipanone, olsalazine, opium, oxaceprol, oxametacine, oxaprozin, oxycodone, oxymorphone, oxyphenbutazone, papaveretum, paramethasone, paranyline, parsalmide, pentazocine, perisoxal, phenacetin, phenadoxone, phenazocine, phenazopyridine hydrochloride, phenocoll, phenoperidine, phenopyrazone, phenomorphan, phenyl acetylsalicylate, phenylbutazone, phenyl salicylate, phenyramidol, piketoprofen, piminodine, pipebuzone, piperylone, pirazolac, piritramide, piroxicam, pirprofen, pranoprofen, prednicarbate, prednisolone, prednisone, prednival, prednylidene, proglumetacin, proheptazine, promedol, propacetamol, properidine, propiram, propoxyphene, propyphenazone, proquazone, protizinic acid, proxazole, ramifenazone, remifentanil, rimazolium metilsulfate, salacetamide, salicin, salicylamide, salicylamide o- acetic acid, salicylic acid, salicylsulfuric acid, salsalate, salverine, simetride, sufentanil, sulfasalazine, sulindac, superoxide dismutase, suprofen, suxibuzone, talniflumate, tenidap, tenoxicam, terofenamate, tetrandrine, thiazolinobutazone, tiaprofenic acid, tiaramide, tilidine, tinoridine, tixocortol, tolfenamic acid, tolmetin, tramadol, triamcinolone, triamcinolone acetonide, tropesin, viminol, xenbucin, ximoprofen, zaltoprofen and zomepirac.
[00256] In one embodiment, a compound described herein may be administered with a selective COX-2 inhibitor for treating or preventing inflammation. Exemplary selective COX-2 inhibitors include, for example, deracoxib, parecoxib, celecoxib, valdecoxib, rofecoxib, etoricoxib, and lumiracoxib.
[00257] In some embodiments, a provided compound is administered in combination with an anthracycline or a Topo II inhibitor. In certain embodiments, a provided compound is administered in combination with Doxorubicin (Dox). In certain embodiments, a provided compound is administered in combination with bortezomib (and more broadly including carfilzomib). It was surprisingly found that a provided compound in combination with Dox or bortezomib resulted in a synergystic effect (i.e., more than additive).
Viral infections
[00258] Compounds and methods described herein may be used to treat or prevent a disease or disorder associated with a viral infection, particularly in humans and other mammals. A compound described herein may be administered prior to the onset of, at, or after the initiation of viral infection. When used prophylactically, the compounds are preferably provided in advance of any viral infection or symptom thereof.
[00259] Exemplary viral diseases include acute febrile pharyngitis, pharyngoconjunctival fever, epidemic keratoconjunctivitis, infantile gastroenteritis, Coxsackie infections, infectious mononucleosis, Burkitt lymphoma, acute hepatitis, chronic hepatitis, hepatic cirrhosis, hepatocellular carcinoma, primary HSV-1 infection (e.g., gingivostomatitis in children, tonsillitis and pharyngitis in adults, keratoconjunctivitis), latent HSV-1 infection (e.g., herpes labialis and cold sores), primary HSV-2 infection, latent HSV-2 infection, aseptic meningitis, infectious mononucleosis, Cytomegalic inclusion disease, Kaposi's sarcoma, multicentric Castleman disease, primary effusion lymphoma, AIDS, influenza, Reye syndrome, measles, postinfectious encephalomyelitis, Mumps, hyperplastic epithelial lesions (e.g., common, flat, plantar and anogenital warts, laryngeal papillomas, epidermodysplasia verruciformis), cervical carcinoma, squamous cell carcinomas, croup, pneumonia, bronchiolitis, common cold, Poliomyelitis, Rabies, influenza-like syndrome, severe bronchiolitis with pneumonia, German measles, congenital rubella, Varicella, and herpes zoster.
[00260] Exemplary viral influenza A strains include H1N1, H3N2, H5N1 , H7N3, H7N9. A compound described herein can also be used to treat or prevent influenza B.
[00261] Exemplary viral pathogens include Adenovirus, Coxsackievirus, Dengue virus, Encephalitis Virus, Epstein-Barr virus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus. Herpes simplex virus type 1, Herpes simplex virus type 2, cytomegalovirus, Human herpesvirus type 8, Human immunodeficiency virus, Influenza virus, measles virus, Mumps virus, Human papillomavirus, Parainfluenza virus, Poliovirus, Rabies virus, Respiratory syncytial virus, Rubella virus, Varicella-zoster virus, West Nile virus, Dungee, and Yellow fever virus. Viral pathogens may also include viruses that cause resistant viral infections.
[00262] Antiviral drugs are a class of medications used specifically for treating viral infections. Antiviral action generally falls into one of three mechanisms: interference with the ability of a virus to infiltrate a target cell (e.g., amantadine, rimantadine and pleconaril), inhibition of the synthesis of virus (e.g., nucleoside analogues, e.g., acyclovir and zidovudine (AZT), and inhibition of the release of virus (e.g., zanamivir and oseltamivir).
Ophthalmology
[00263] Compounds and methods described herein may be used to treat or prevent an ophthamology disorder. Exemplary ophthamology disorders include macular edema
(diabetic and nondiabetic macular edema), age related macular degeneration wet and dry forms, aged disciform macular degeneration, cystoid macular edema, palpebral edema, retina edema, diabetic retinopathy, chorioretinopathy, neovascular maculopathy, neovascular glaucoma, uveitis, iritis, retinal vasculitis, endophthalmitis, panophthalmitis, metastatic ophthalmia, choroiditis, retinal pigment epithelitis, conjunctivitis, cyclitis, scleritis, episcleritis, optic neuritis, retrobulbar optic neuritis, keratitis, blepharitis, exudative retinal detachment, corneal ulcer, conjunctival ulcer, chronic nummular keratitis, ophthalmic disease associated with hypoxia or ischemia, retinopathy of prematurity, proliferative diabetic retinopathy, polypoidal choroidal vasculopathy, retinal angiomatous proliferation, retinal artery occlusion, retinal vein occlusion, Coats' disease, familial exudative vitreoretinopathy, pulseless disease (Takayasu's disease), Eales disease, antiphospholipid antibody syndrome, leukemic retinopathy, blood hyperviscosity syndrome, macro globulinemia, interferon- associated retinopathy, hypertensive retinopathy, radiation retinopathy, corneal epithelial stem cell deficiency and cataract.
[00264] Other ophthalmology disorders treatable using the compounds and methods described herein include proliferative vitreoretinopathy and chronic retinal detachment.
[00265] Inflammatory eye diseases are also treatable using the compounds and methods described herein.
Neurodegenerative disease
[00266] Neurodegeneration is the umbrella term for the progressive loss of structure or function of neurons, including death of neurons. Many neurodegenerative diseases including Parkinson's, Alzheimer's, and Huntington's occur as a result of neurodegenerative processes. As research progresses, many similarities appear which relate these diseases to one another on a sub-cellular level. Discovering these similarities offers hope for therapeutic advances that could ameliorate many diseases simultaneously. There are many parallels between different neurodegenerative disorders including atypical protein assemblies as well as induced cell death.
[00267] Alzheimer's disease is characterized by loss of neurons and synapses in the cerebral cortex and certain subcortical regions. This loss results in gross atrophy of the affected regions, including degeneration in the temporal lobe and parietal lobe, and parts of the frontal cortex and cingulate gyrus.
[00268] Huntington's disease causes astrogliosis and loss of medium spiny neurons. Areas of the brain are affected according to their structure and the types of neurons they contain, reducing in size as they cumulatively lose cells. The areas affected are mainly in the striatum, but also the frontal and temporal cortices. The striatum's subthalamic nuclei send control signals to the globus pallidus, which initiates and modulates motion. The weaker signals from subthalamic nuclei thus cause reduced initiation and modulation of movement, resulting in the characteristic movements of the disorder. Exemplary treatments for Huntington's disease include tetrabenazine, neuroleptics, benzodiazepines, amantadine, remacemide, valproic acid, selective serotonin reuptake inhibitors (SSRIs), mirtazapine and antipsychotics.
[00269] The mechanism by which the brain cells in Parkinson's are lost may consist of an abnormal accumulation of the protein alpha-synuclein bound to ubiquitin in the damaged cells. The alpha-synuclein-ubiquitin complex cannot be directed to the proteosome. This protein accumulation forms proteinaceous cytoplasmic inclusions called Lewy bodies. The latest research on pathogenesis of disease has shown that the death of dopaminergic neurons by alpha-synuclein is due to a defect in the machinery that transports proteins between two major cellular organelles— the endoplasmic reticulum (ER) and the Golgi apparatus. Certain proteins like Rabl may reverse this defect caused by alpha-synuclein in animal models. Exemplary Parkinson's disease therapies include levodopa, dopamine agonists such as include bromocriptine, pergolide, pramipexole, ropinirole, piribedil, cabergoline,
apomorphine and lisuride, dopa decarboxylate inhibitors, MAO-B inhibitors such as selegilene and rasagilene, anticholinergics and amantadine,
[00270] Amyotrophic lateral sclerosis (ALS/Lou Gehrig's Disease) is a disease in which motor neurons are selectively targeted for degeneration. Exemplary ALS therapies include riluzole, baclofen, diazepam, trihexyphenidyl and amitriptyline.
[00271] Other exemplary neurodegenerative therapeutics include antisense
oligonucleotides and stem cells.
Wound Healing
[00272] Wounds are a type of condition characterized by cell or tissue damage. Wound healing is a dynamic pathway that optimally leads to restoration of tissue integrity and function. The wound healing process consists of three overlapping phases. The first phase is an inflammatory phase, which is characterized by homeostasis, platelet aggregation and degranulation. Platelets as the first response, release multiple growth factors to recruit immune cells, epithelial cells, and endothelial cells. The inflammatory phase typically occurs over days 0-5. The second stage of wound healing is the proliferative phase during which macrophages and granulocytes invade the wound. Infiltrating fibroblasts begin to produce collagen. The principle characteristics of this phase are epithelialization, angiogenesis, granulation tissue formation and collagen production. The proliferative phase typically occurs over days 3-14. The third phase is the remodeling phase where matrix formation occurs. The fibroblasts, epithelial cells, and endothelial cells continue to produce collagen and collagenase as well as matrix metalloproteases (MMPs) for remodeling. Collagen crosslinking takes place and the wound undergoes contraction. The remodeling phase typically occurs from day 7 to one year.
[00273] Compounds and compositions described herein can be used for promoting wound healing (e.g. , promoting or accelerating wound closure and/or wound healing, mitigating scar fibrosis of the tissue of and/or around the wound, inhibiting apoptosis of cells surrounding or proximate to the wound). Thus, in certain embodiments, the present invention provides a method for promoting wound healing in a subject, comprising administering to the subject a therapeutically effective amount of a compound (e.g., a CRM1 inhibitor), or
pharmaceutically acceptable salt or composition thereof. The method need not achieve complete healing or closure of the wound; it is sufficient for the method to promote any degree of wound closure. In this respect, the method can be employed alone or as an adjunct to other methods for healing wounded tissue.
[00274] The compounds and compositions described herein can be used to treat wounds during the inflammatory (or early) phase, during the proliferative (or middle) wound healing phase, and/or during the remodeling (or late) wound healing phase.
[00275] In some embodiments, the subject in need of wound healing is a human or an animal, for example, a dog, a cat, a horse, a pig, or a rodent, such as a mouse.
[00276] In some embodiments, the compounds and compositions described herein useful for wound healing are administered topically, for example, proximate to the wound site, or systemically.
[00277] More specifically, a therapeutically effective amount of a compound or composition described herein can be administered (optionally in combination with other agents) to the wound site by coating the wound or applying a bandage, packing material, stitches, etc., that are coated or treated with the compound or composition described herein. As such, the compounds and compositions described herein can be formulated for topical administration to treat surface wounds. Topical formulations include those for delivery via the mouth (buccal) and to the skin such that a layer of skin (i. e., the epidermis, dermis, and/or subcutaneous layer) is contacted with the compound or composition described herein. Topical delivery systems may be used to administer topical formulations of the compounds and compositions described herein.
[00278] Alternatively, the compounds and compositions described herein can be administered at or near the wound site by, for example, injection of a solution, injection of an extended release formulation, or introduction of a biodegradable implant comprising the compound or composition described herein.
[00279] The compounds and compositions described herein can be used to treat acute wounds or chronic wounds. A chronic wound results when the normal reparative process is interrupted. Chronic wounds can develop from acute injuries as a result of unrecognized persistent infections or inadequate primary treatment. In most cases however, chronic lesions are the end stage of progressive tissue breakdown owing to venous, arterial, or metabolic vascular disease, pressure sores, radiation damage, or tumors.
[00280] In chronic wounds, healing does not occur for a variety of reasons, including improper circulation in diabetic ulcers, significant necrosis, such as in burns, and infections. In these chronic wounds, viability or the recovery phase is often the rate-limiting step. The cells are no longer viable and, thus, initial recovery phase is prolonged by unfavorable wound bed environment.
[00281] Chronic wounds include, but are not limited to the following: chronic ischemic skin lesions; scleroderma ulcers; arterial ulcers; diabetic foot ulcers; pressure ulcers; venous ulcers; non-healing lower extremity wounds; ulcers due to inflammatory conditions; and/or long-standing wounds. Other examples of chronic wounds include chronic ulcers, diabetic wounds, wounds caused by diabetic neuropathy, venous insufficiencies, and arterial insufficiencies, and pressure wounds and cold and warm burns. Yet other examples of chronic wounds include chronic ulcers, diabetic wounds, wounds caused by diabetic neuropathy, venous insufficiencies, arterial insufficiencies, and pressure wounds.
[00282] Acute wounds include, but are not limited to, post-surgical wounds, lacerations, hemorrhoids and fissures.
[00283] In a particular embodiment, the compounds and compositions described herein can be used for diabetic wound healing or accelerating healing of leg and foot ulcers secondary to diabetes or ischemia in a subject.
[00284] In one embodiment, the wound is a surface wound. In another embodiment, the wound is a surgical wound (e.g., abdominal or gastrointestinal surgical wound). In a further embodiment, the wound is a burn. In yet another embodiment, the wound is the result of radiation exposure.
[00285] The compounds and compositions described herein can also be used for diabetic wound healing, gastrointestinal wound healing, or healing of an adhesion due, for example, to an operation.
[00286] The compounds and compositions described herein can also be used to heal wounds that are secondary to another disease. For example, in inflammatory skin diseases, such as psoriasis and dermatitis, there are numerous incidents of skin lesions that are secondary to the disease, and are caused by deep cracking of the skin, or scratching of the skin. The compounds and compositions described herein can be used to heal wounds that are secondary to these diseases, for example, inflammatory skin diseases, such as psoriasis and dermatitis.
[00287] In a further embodiment, the wound is an internal wound. In a specific aspect, the internal wound is a chronic wound. In another specific aspect, the wound is a vascular wound. In yet another specific aspect, the internal wound is an ulcer. Examples of internal wounds include, but are not limited to, fistulas and internal wounds associated with cosmetic surgery, internal indications, Crohn's disease, ulcerative colitis, internal surgical sutures and skeletal fixation. Other examples of internal wounds include, but are not limited to, fistulas and internal wounds associated with cosmetic surgery, internal indications, internal surgical sutures and skeletal fixation.
[00288] Examples of wounds include, but are not limited to, abrasions, avulsions, blowing wounds (i.e., open pneumothorax), burn wounds, contusions, gunshot wounds, incised wounds, open wounds, penetrating wounds, perforating wounds, puncture wounds, seton wounds, stab wounds, surgical wounds, subcutaneous wounds, diabetic lesions, or tangential wounds. Additional examples of wounds that can be treated by the compounds and compositions described herein include acute conditions or wounds, such as thermal burns, chemical burns, radiation burns, burns caused by excess exposure to ultraviolet radiation {e.g., sunburn); damage to bodily tissues, such as the perineum as a result of labor and childbirth; injuries sustained during medical procedures, such as episiotomies; trauma- induced injuries including cuts, incisions, excoriations; injuries sustained from accidents; post-surgical injuries, as well as chronic conditions, such as pressure sores, bedsores, conditions related to diabetes and poor circulation, and all types of acne. In addition, the wound can include dermatitis, such as impetigo, intertrigo, folliculitis and eczema, wounds following dental surgery; periodontal disease; wounds following trauma; and tumor- associated wounds. Yet other examples of wounds include animal bites, arterial disease, insect stings and bites, bone infections, compromised skin/muscle grafts, gangrene, skin tears or lacerations, skin aging, surgical incisions, including slow or non-healing surgical wounds, intracerebral hemorrhage, aneurysm, dermal asthenia, and post-operation infections.
[00289] In preferred embodiments, the wound is selected from the group consisting of a burn wound, an incised wound, an open wound, a surgical or post surgical wound, a diabetic lesion, a thermal burn, a chemical burn, a radiation burn, a pressure sore, a bedsore, and a condition related to diabetes or poor circulation. In more preferred embodiments, the wound is selected from the group consisting of an incised wound, an open wound, a surgical or post surgical wound, a diabetic lesion, a pressure sore, a bedsore, and a condition or wound related to diabetes or poor circulation.
[00290] In some embodiments, the wound is selected from the group consisting of a non- radiation burn wound, an incised wound, an open wound, a surgical or post surgical wound, a diabetic lesion, a thermal burn, a chemical burn, a pressure sore, a bedsore, and a condition related to diabetes or poor circulation. In some embodiments, the wound is selected from the group consisting of an incised wound, an open wound, a surgical or post surgical wound, a diabetic lesion, a pressure sore, a bedsore, and a condition related to diabetes or poor circulation.
[00291] The present disclosure also relates to methods and compositions of reducing scar formation during wound healing in a subject. The compounds and compositions described herein can be administered directly to the wound or to cells proximate the wound at an amount effective to reduce scar formation in and/or around the wound. Thus, in some embodiments, a method of reducing scar formation during wound healing in a subject is provided, the method comprising administering to the subject a therapeutically effective amount of a compound described herein (e.g., a CRM1 inhibitor), or a pharmaceutically acceptable salt thereof.
[00292] The wound can include any injury to any portion of the body of a subject.
According to embodiments, methods are provided to ameliorate, reduce, or decrease the formation of scars in a subject that has suffered a burn injury. According to preferred embodiments, methods are provided to treat, reduce the occurrence of, or reduce the probability of developing hypertrophic scars in a subject that has suffered an acute or chronic wound or injury.
Other disorders
[00293] Compounds and compositions described herein may also be used to treat disorders of abnormal tissue growth and fibrosis including dilative cardiomyopathy, hypertrophic cardiomyopathy, restrictive cardiomyopathy, pulmonary fibrosis, hepatic fibrosis, glomerulonephritis, and other renal disorders.
Combination Radiation Therapy
[00294] Compounds and compositions described herein are useful as radiosensitizers. Therefore, compounds and compositions described herein can be administered in
combination with radiation therapy. Radiation therapy is the medical use of high-energy radiation (e.g. , x-rays, gamma rays, charged particles) to shrink tumors and kill malignant cells, and is generally used as part of cancer treatment. Radiation therapy kills malignant cells by damaging their DNA.
[00295] Radiation therapy can be delivered to a patient in several ways. For example, radiation can be delivered from an external source, such as a machine outside the patient's body, as in external beam radiation therapy. External beam radiation therapy for the treatment of cancer uses a radiation source that is external to the patient, typically either a radioisotope, such as 60Co, 137Cs, or a high energy x-ray source, such as a linear accelerator. The external source produces a collimated beam directed into the patient to the tumor site. External-source radiation therapy avoids some of the problems of internal- source radiation therapy, but it undesirably and necessarily irradiates a significant volume of non-tumorous or healthy tissue in the path of the radiation beam along with the tumorous tissue.
[00296] The adverse effect of irradiating of healthy tissue can be reduced, while maintaining a given dose of radiation in the tumorous tissue, by projecting the external radiation beam into the patient at a variety of "gantry" angles with the beams converging on the tumor site. The particular volume elements of healthy tissue, along the path of the radiation beam, change, reducing the total dose to each such element of healthy tissue during the entire treatment.
[00297] The irradiation of healthy tissue also can be reduced by tightly collimating the radiation beam to the general cross section of the tumor taken perpendicular to the axis of the radiation beam. Numerous systems exist for producing such a circumferential collimation, some of which use multiple sliding shutters which, piecewise, can generate a radio-opaque mask of arbitrary outline.
[00298] For administration of external beam radiation, the amount can be at least about 1 Gray (Gy) fractions at least once every other day to a treatment volume. In a particular embodiment, the radiation is administered in at least about 2 Gray (Gy) fractions at least once per day to a treatment volume. In another particular embodiment, the radiation is
administered in at least about 2 Gray (Gy) fractions at least once per day to a treatment volume for five consecutive days per week. In another particular embodiment, radiation is administered in 10 Gy fractions every other day, three times per week to a treatment volume. In another particular embodiment, a total of at least about 20 Gy is administered to a patient in need thereof. In another particular embodiment, at least about 30 Gy is administered to a patient in need thereof. In another particular embodiment, at least about 40 Gy is
administered to a patient in need thereof.
[00299] Typically, the patient receives external beam therapy four or five times a week. An entire course of treatment usually lasts from one to seven weeks depending on the type of cancer and the goal of treatment. For example, a patient can receive a dose of 2 Gy/day over 30 days.
[00300] Internal radiation therapy is localized radiation therapy, meaning the radiation source is placed at the site of the tumor or affected area. Internal radiation therapy can be delivered by placing a radiation source inside or next to the area requiring treatment. Internal radiation therapy is also called brachytherapy. Brachytherapy includes intercavitary treatment and interstitial treatment. In intracavitary treatment, containers that hold radioactive sources are put in or near the tumor. The sources are put into the body cavities. In interstitial treatment, the radioactive sources alone are put into the tumor. These radioactive sources can stay in the patient permanently. Typically, the radioactive sources are removed from the patient after several days. The radioactive sources are in containers.
[00301] There are a number of methods for administration of a radiopharmaceutical agent. For example, the radiopharmaceutical agent can be administered by targeted delivery or by systemic delivery of targeted radioactive conjugates, such as a radiolabeled antibody, a radiolabeled peptide and a liposome delivery system. In one particular embodiment of targeted delivery, the radiolabeled pharmaceutical agent can be a radiolabeled antibody. See, for example, Ballangrud A. M., et al. Cancer Res., 2001 ; 61 :2008-2014 and Goldenber, D.M. J Nucl. Med. , 2002; 43(5):693-713, the contents of which are incorporated by reference herein.
[00302] In another particular embodiment of targeted delivery, the radiopharmaceutical agent can be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or
phosphatidylcholines. See, for example, Emfietzoglou D, Kostarelos , Sgouros G. An analytical dosimetry study for the use of radionuclide-liposome conjugates in internal radiotherapy. J Nucl Med 2001; 42:499-504, the contents of which are incorporated by reference herein.
[00303] In yet another particular embodiment of targeted delivery, the radiolabeled pharmaceutical agent can be a radiolabeled peptide. See, for example, Weiner RE, Thakur ML. Radiolabeled peptides in the diagnosis and therapy of oncological diseases. Appl Radiat Isot 2002 Nov;57(5):749-63, the contents of which are incorporated by reference herein.
[00304] In addition to targeted delivery, bracytherapy can be used to deliver the radiopharmaceutical agent to the target site. Brachytherapy is a technique that puts the radiation sources as close as possible to the tumor site. Often the source is inserted directly into the tumor. The radioactive sources can be in the form of wires, seeds or rods. Generally, cesium, iridium or iodine are used.
[00305] Systemic radiation therapy is another type of radiation therapy and involves the use of radioactive substances in the blood. Systemic radiation therapy is a form of targeted therapy. In systemic radiation therapy, a patient typically ingests or receives an injection of a radioactive substance, such as radioactive iodine or a radioactive substance bound to a monoclonal antibody.
[00306] A "radiopharmaceutical agent," as defined herein, refers to a pharmaceutical agent which contains at least one radiation- emitting radioisotope. Radiopharmaceutical agents are routinely used in nuclear medicine for the diagnosis and/or therapy of various diseases. The radiolabeled pharmaceutical agent, for example, a radiolabeled antibody, contains a radioisotope (RI) which serves as the radiation source. As contemplated herein, the term "radioisotope" includes metallic and non-metallic radioisotopes. The radioisotope is chosen based on the medical application of the radiolabeled pharmaceutical agents. When the radioisotope is a metallic radioisotope, a chelator is typically employed to bind the metallic radioisotope to the rest of the molecule. When the radioisotope is a non-metallic radioisotope, the non-metallic radioisotope is typically linked directly, or via a linker, to the rest of the molecule.
[00307] As used herein, a "metallic radioisotope" is any suitable metallic radioisotope useful in a therapeutic or diagnostic procedure in vivo or in vitro. Suitable metallic radioisotopes include, but are not limited to: Actinium-225, Antimony-124, Antimony- 125, Arsenic-74, Barium- 103, Barium- 140, Beryllium-7, Bismuth-206, Bismuth-207, Bismuth212, Bismuth213, Cadmium-109, Cadmium-115m, Calcium-45, Cerium-139, Cerium-141, Cerium-144, Cesium-137, Chromium-51, Cobalt-55, Cobalt-56, Cobalt-57, Cobalt-58, Cobalt-60, Cobalt-64, Copper-60, Copper-62, Copper-64, Copper-67, Erbium- 169,
Europium-152, Gallium-64, Gallium-67, Gallium-68, Gadoliniuml53, Gadolinium- 157 Gold-195, Gold-199, Hafnium-175, Hafnium- 175-181, Holmium-166, Indium-110, Indium- I l l, Iridium-192, Iron 55, Iron-59, Krypton85, Lead-203, Lead-210, Lutetium-177,
Manganese-54, Mercury- 197, Mercury203, Molybdenum-99, Neodymium-147, Neptunium- 237, Nickel-63, Niobium95, Osmium-185+191, Palladium- 103, Palladium- 109, Platinum- 195m, Praseodymium- 143, Promethium-147, Promethium-149, Protactinium-233, Radium- 226, Rhenium-186, Rhenium-188, Rubidium-86, Ruthenium-97, Ruthenium- 103,
Ruthenium- 105, Ruthenium- 106, Samarium-153, Scandium-44, Scandium-46, Scandium-47, Selenium-75, Silver-l l Om, Silver-I l l , Sodium- 22, Strontium-85, Strontium-89, Strontium- 90, Sulfur-35, Tantalum-182, Technetium-99m, Tellurium- 125, Tellurium- 132, Thallium- 204, Thorium-228, Thorium-232, Thallium-170, Tin-113, Tin-114, Tin-117m, Titanium-44, Tungsten-185, Vanadium-48, Vanadium-49, Ytterbium- 169, Yttrium-86, Yttrium-88, Yttrium-90, Yttrium-91 , Zinc-65, Zirconium-89, and Zirconium-95.
[00308] As used herein, a "non-metallic radioisotope" is any suitable nonmetallic radioisotope (non-metallic radioisotope) useful in a therapeutic or diagnostic procedure in vivo or in vitro. Suitable non-metallic radioisotopes include, but are not limited to: Iodine- 131, Iodine-125, Iodine-123, Phosphorus-32, Astatine-211, Fluorine-18, Carbon-11, Oxygen- 15, Bromine-76, and Nitrogen-13.
[00309] Identifying the most appropriate isotope for radiotherapy requires weighing a variety of factors. These include tumor uptake and retention, blood clearance, rate of radiation delivery, half-life and specific activity of the radioisotope, and the feasibility of large-scale production of the radioisotope in an economical fashion. The key point for a therapeutic radiopharmaceutical is to deliver the requisite amount of radiation dose to the tumor cells and to achieve a cytotoxic or tumoricidal effect while not causing unmanageable side-effects.
[00310] It is preferred that the physical half-life of the therapeutic radioisotope be similar to the biological half-life of the radiopharmaceutical at the tumor site. For example, if the half-life of the radioisotope is too short, much of the decay will have occurred before the radiopharmaceutical has reached maximum target/background ratio. On the other hand, too long a half-life could cause unnecessary radiation dose to normal tissues. Ideally, the radioisotope should have a long enough half-life to attain a minimum dose rate and to irradiate all the cells during the most radiation sensitive phases of the cell cycle. In addition, the half-life of a radioisotope has to be long enough to allow adequate time for
manufacturing, release, and transportation.
[00311] Other practical considerations in selecting a radioisotope for a given application in tumor therapy are availability and quality. The purity has to be sufficient and reproducible, as trace amounts of impurities can affect the radiolabeling and radiochemical purity of the radiopharmaceutical.
[00312] The target receptor sites in tumors are typically limited in number. As such, it is preferred that the radioisotope have high specific activity. The specific activity depends primarily on the production method. Trace metal contaminants must be minimized as they often compete with the radioisotope for the chelator and their metal complexes compete for receptor binding with the radiolabeled chelated agent.
[00313] The type of radiation that is suitable for use in the methods of the present invention can vary. For example, radiation can be electromagnetic or particulate in nature. Electromagnetic radiation useful in the practice of this invention includes, but is not limited to, x-rays and gamma rays. Particulate radiation useful in the practice of this invention includes, but is not limited to, electron beams (beta particles), protons beams, neutron beams, alpha particles, and negative pi mesons. The radiation can be delivered using conventional radiological treatment apparatus and methods, and by intraoperative and stereotactic methods. Additional discussion regarding radiation treatments suitable for use in the practice of this invention can be found throughout Steven A. Leibel et al., Textbook of Radiation Oncology (1998) (publ. W. B. Saunders Company), and particularly in Chapters 13 and 14. Radiation can also be delivered by other methods such as targeted delivery, for example by radioactive "seeds," or by systemic delivery of targeted radioactive conjugates. J. Padawer et ah, Combined Treatment with Radioestradiol lucanthone in Mouse C3HBA Mammary
Adenocarcinoma and with Estradiol lucanthone in an Estrogen Bioassay, Int. J. Radiat.
Oncol. Biol. Phys. 7:347-357 (1981). Other radiation delivery methods can be used in the practice of this invention.
[00314] For tumor therapy, both a and β-particle emitters have been investigated. Alpha particles are particularly good cytotoxic agents because they dissipate a large amount of energy within one or two cell diameters. The β -particle emitters have relatively long penetration range (2-12 mm in the tissue) depending on the energy level. The long-range penetration is particularly important for solid tumors that have heterogeneous blood flow and/or receptor expression. The β-particle emitters yield a more homogeneous dose distribution even when they are heterogeneously distributed within the target tissue.
[00315] In a particular embodiment, therapeutically effective amounts of the compounds and compositions described herein are administered in combination with a therapeutically effective amount of radiation therapy to treat cancer {e.g., lung cancer, such as non-small cell lung cancer). The amount of radiation necessary can be determined by one of skill in the art based on known doses for a particular type of cancer. See, for example, Cancer Medicine 5th ed., Edited by R.C. Bast et al, July 2000, BC Decker.
[00316] The above disclosure generally describes the present invention. A more complete understanding can be obtained by reference to the following specific Examples. These Examples are described solely for purposes of illustration and are not intended to limit the scope of the invention. Changes in form and substitution of equivalents are contemplated as circumstances may suggest or render expedient. Although specific terms have been employed herein, such terms are intended in a descriptive sense and not for purposes of limitation.
EXEMPLIFICATION
Abbreviations
aq. Aqueous
DABCO 1 ,4-Diazabicyclo[2.2.2]octane
DIPEA Diisopropylethylamine
DMF N,N-Dimethylformamide DMSO Dimethylsulfoxide
EDCI 3-(ethyliminomethyleneamino)-N,N-dimethylpropan-l-amine
eq. equivalent(s)
Et Ethyl
EtOAc Ethyl acetate
g gram
GC Gas chromatography
h hour(s)
HOBt 1 -Hydroxybenzotriazole
HPLC High performance liquid chromatography
IPA isopropyl alcohol
kg kilogram(s)
L Liter(s)
LCMS Liquid Chromatography Mass Spectrometry
M Molar
Me methyl
mg milligram(s)
min minute(s)
mL milliliters
MTBE methyl rt-butyl ether
N Normal
NMPv Nuclear magnetic resonance
z'Pr isopropyl
THF Tetrahydrofuran
¾ Retention time
[00317] Throughout the following description of such processes it is to be understood that, where appropriate, suitable protecting groups will be added to, and subsequently removed from, the various reactants and intermediates in a manner that will be readily understood by one skilled in the art of organic synthesis. Conventional procedures for using such protecting groups as well as examples of suitable protecting groups are described, for example, in "Protective Groups in Organic Synthesis", T.W. Green, P.G.M. Wuts, Wiley-Interscience, New York, (1999). It is also to be understood that a transformation of a group or substituent into another group or substituent by chemical manipulation can be conducted on any intermediate or final product on the synthetic path toward the final product, in which the possible type of transformation is limited only by inherent incompatibility of other functionalities carried by the molecule at that stage to the conditions or reagents employed in the transformation. Such inherent incompatibilities, and ways to circumvent them by carrying out appropriate transformations and synthetic steps in a suitable order, will be readily understood to the one skilled in the art of organic synthesis. Examples of
transformations are given below, and it is to be understood that the described transformations are not limited only to the generic groups or substituents for which the transformations are exemplified. References and descriptions on other suitable transformations are given in "Comprehensive Organic Transformations - A Guide to Functional Group Preparations" R. C. Larock, VHC Publishers, Inc. (1989). References and descriptions of other suitable reactions are described in textbooks of organic chemistry, for example, "Advanced Organic Chemistry", March, 4th ed. McGraw Hill (1992) or, "Organic Synthesis", Smith, McGraw Hill, (1994). Techniques for purification of intermediates and final products include for example, straight and reversed phase chromatography on column or rotating plate, recrystallization, distillation and liquid-liquid or solid-liquid extraction, which will be readily understood by the one skilled in the art. The definitions of substituents and groups are as in formula I except where defined differently. The term "room temperature" and "ambient temperature" shall mean, unless otherwise specified, a temperature between 16 and 25 °C. The term "reflux" shall mean, unless otherwise stated, in reference to an employed solvent a temperature at or above the boiling point of named solvent.
Example 1. Synthetic Procedures: Compounds of Structural Formula I
Synthesis of (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid
(8).
Figure imgf000089_0001
Figure imgf000089_0002
Synthesis of 3,5-bis(trifluoromethyl)benzothioamide (2).
[00318] 3,5-Bis(trifluoromethyl)benzonitrile (1) (200 g, 836.4 mmol) was dissolved in DMF (1 L). Sodium hydrosulphide hydrate (123.7 g, 1678 mmol) and magnesium chloride hexahydrate (186.7 g, 920 mmol) were added and the reaction mixture was stirred at room temperature for 3 h. The reaction mixture was transferred into iced water (10 L) and extracted with EtOAc (3 X 1L). The combined organic layers were washed with brine (3 X 500 mL), dried over anhydrous Na2S04 and concentrated under reduced pressure to afford 3,5-bis(trifluoromethyl)benzothioamide (2) (Yield: 205 g, 90%). The product was used without further purification in the following step. LCMS: m/z 21 '4.04 [M+H]+, ¾ = 2.78 min.
Synthesis of 3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazole (3).
[00319] 3,5-bis(trifluoromethyl)benzothioamide (2) (205.65 g, 752 mmol) was dissolved in DMF (1.028 L) and hydrazine hydrate monohydrate (71 mL) was added drop wise. The reaction mixture was stirred at room temperature for 1 h followed by the drop wise addition of formic acid (1.028 L) over 45 min and heated at 90 °C for 3 h. The reaction mixture was allowed to cool to room temperature, transferred into iced water (10 L), and extracted with EtOAc (3 X 1L). The combined organic layers were washed with brine (3 X 500 mL), dried over anhydrous Na2S04 and concentrated under reduced pressure and crystallized from petroleum ether to give 3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazole (3) (Yield: 160 g, 75 %). 1H NMR (400 MHz, OMSO-d6) δ 14.52 (s, 1H), 8.77 (d, J = 5.4 Hz, 1H), 8.53 (s, 2H), 8.18 (s, 1H). LCMS: m/z 282.19 [M+H]+, tR = 2.69 min.
Synthesis of isopropyl propiolate (5).
[00320] Propiolic acid (4) (1000 g, 14.28 mol) was dissolved in /-PrOH (8 L). BF3-etherate (4.54 kg, 25.7 mol) was added slowly at 25 °C over a period of 30 minutes. The temperature of the reaction mixture was gradually increased up to 90 °C and stirred for 3 h. GC monitoring after 3 h showed the completion of the reaction. The reaction mixture was cooled to room temperature, quenched with 20 L of ice cold water and stirred for 30 minutes. 10 L of dichloromethane was added to the reaction mixture and stirred for an additional 30 min. The organic layers were separated and the aqueous layer was re-extracted with 5 L of dichloromethane. The combined organic layers were washed with 10 L of brine, dried over anhydrous Na2S04, and concentrated under reduced pressure at 35 - 40 °C (product is volatile) to obtain isopropyl propiolate (5) as a brown liquid (Yield: 1.32 kg, 81 %). Purity 89.67 % (GC); 1H NMR (300 MHz, CDC13) δ 4.98-5.05 (m, 1H), 2.85 (s, 1H), 1.22 (d, J = 6.6 Hz, 6H).
Synthesis of (Z)-isopropyl 3-iodoacrylate (6).
[00321] Isopropyl propiolate (5) (1000 g, 8.92 mol) was added in acetic acid (3.7 L) at 25 °C and the reaction mixture was stirred for 10 min. Sodium iodide (2.138 kg, 13.98 mol) was added (a dark brown color was observed) while stirring. The temperature was increased to 110 °C and stirred for 1.5 h. GC monitoring showed the completion of the reaction after 1.5 h. The reaction mixture was cooled to room temperature, quenched with iced water (18.75 L) and stirred for 30 min. MTBE (5 L) was added to the reaction mixture and stirred for additional 30 min. The organic layers were separated and the aqueous layer was re-extracted with MTBE (5 L). The combined organic layers were washed with NaHC03 (2 x 10 L), NaHS03 (2 x 5 L), brine (5.2 L), dried over anhydrous sodium sulfate and concentrated under reduced pressure at 35 °C to obtain (Z)-isopropyl 3-iodoacrylate (6) as a brown liquid. Yield (1.49 kg, 70%). Purity 87.34 % (GC); 1H NMR (300 MHz, CDC13) δ 7,38 (d, J= 8.7 Hz, 1H), 6.83 (d, J= 8.7 Hz, 1H), 5.08-5.131 (m, 1H), 1.28 (d, 6H, J= 6.3 Hz).
Synthesis of (Z)-isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l- yl)acrylate (7).
[00322] 3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazole (3) (160 g, 569 mmol) was added in DMF (0.96 L). DABCO (127.74 g, 1138 mmol) was then introduced and the reaction mixture was stirred for 30 min at room temperature. Iodo ester (6) (150.32 g, 626 mmol) was added drop wise over a period of 1 h at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for additional 2 h. The reaction mixture was quenched with cold water (5 L), stirred for 15 min, and extracted with ethyl acetate (3 X 1 L). The combined organic layers were washed with saturated solution of sodium thiosulphate (2 X 500 mL) followed by brine (3 X 500 mL), dried over anhydrous sodium sulfate (100 g) and concentrated under reduced pressure. The crude product (250 g) was stirred in methanol (625 mL) and maintained at 0-5 °C for 1 h. The precipitated solid was filtered and washed with cold methanol (100 mL), to obtain (Z)-isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH- l,2,4-triazol-l-yl)acrylate (7) as while solid. Yield (138 g, 61%). Ή NMR (400 MHz, DMSO-dfi) δ 9.75 (s, 1 H), 8.63 (s, 2H), 7.95 (s, 1H), 7.30 (d, J = 10.8 Hz, 1H), 5.77 (d, J = 10.8 Hz, 1 H), 5.13-5.19 (m, 1H), 1.34 (d, J = 6.4 Hz, 6H). LCMS: m/z 394.24 [M+H]+, tR = 2.95 min.
Synthesis ( )-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid (8).
[00323] (Z)-Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylate (7) (4 g, 10.17 mmol) was added to a mixture of THF:H20 (1 : 1, 80 mL) at room temperature. The reaction mixture was cooled to 0 °C and cold lithium hydroxide mono hydrate solution (2.13 g in 10 mL water) was added over a period of 30 min. The reaction mixture was slowly warmed to room temperature and stirred for 3 h. The reaction mixture was quenched with iced water (40 mL) and stirred for 30 min. The pH was adjusted to 1-2 using 3M HC1 solution (25 mL) and the reaction mixture was extracted with ethyl acetate (3 x 70 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain the crude compound which was crystallized using petroleum ether (25 mL). The solid precipitated was filtered and washed with petroleum ether (10 mL), dried under reduced pressure to give (Z)-3-(3-(3,5- bis(trifluoromethyl)phenyl)-lH-l ,2,4-triazol-l-yl)acrylic acid (8) as a white solid. Yield (3 g, 84%). Ή NMR (400 MHz, DMSO- ) δ 13.01 (bs, 1H), 9.37 (s, 1H), 8.51 (s, 2H), 8.28 (s, 1H), 7.49 (d, J = 10 Hz, 1H), 5.97 (d, J = 10 Hz, 1H).
Synthesis of isopropyl 3-(3-(3, 5-bis (trifluoromethyl) phenyl)-l//-l, 2, 4-triazoI-l-yl)-2, 3-dibromopropanoate (300).
General Procedure 1. Bromination.
Figure imgf000092_0001
7
[00324] (Z)-Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylate (7) (20 g, 50.85 mmol) was dissolved in dichloromethane (200 mL) at room temperature. Bromine (5.24 mL, 101.71 mmol) was added dropwise over 15 min at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 6 h, transferred into iced water, and extracted with CH2C12 (3 x 500 mL). The combined organic layers were washed with saturated aqueous sodium bisulphite (500 mL) solution followed by brine, dried over anhydrous Na2S04 and concentrated under reduced pressure to give the crude product, which was purified by silica gel chromatography (0-15% ethyl acetate/n-hexane) to yield isopropyl 3-(3-(3,5-bis(trifluoro methyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dibromopropanoate (300). (Yield: 21 g, 75%). 1H NMR (400 MHz, DMSO-c/d) <S 9.1 1 (s, 1H), 8.50 (s, 2H), 8.31 (s, 1H), 7.41 (d, J = 9.6 Hz, 1H), 5.54 (d, J = 9.6 Hz, 1H), 4.82- 4.79 (m, 1H), 1.05-1.00 (m, 6H). LCMS: m/z 554.09 [M+H]+, ¾ = 3.12 min.
Synthesis of isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dichloropropanoate (301).
Figure imgf000092_0002
[00325] (Z)-Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl-lH-l,2,4-triazol-l-yl) (7) (0.5 g, 1.17 mmol) was dissolved in CH2C12 (10 mL) and cooled to 0 °C. Chlorine gas was purged into the reaction mixture for 1 h at 0 °C. The reaction mixture was transferred into water (300 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over anhydrous Na2S0 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (50% EtOAc in hexane) to give isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dichloropropanoate (301). Yield (0.4 g, 68%), 1H NMR (400 MHz, DMSO- 6) δ 9.12 (s, 1H), 8.52 (s, 2H), 8.32 (s, 1 H), 7.36 (d, J = 8.0 Hz, 1 H), 5.70 (d, J = 8.0 Hz, 1H), 4.89- 4.86 (m, 1H), 1.08 (d, J = 28 Hz, 6H); LCMS: m/z 464.1 [M+H]+,/R = 3.22 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3-dibromo-l- (3,3-difluoroazetidin-l-yI)propan-l-one (302).
Figure imgf000093_0001
Synthesis of (Z)-3-(3-(3,5-bis(trifluoromethyI)phenyl)-lH-l,2,4-triazoI-l-yl)-l-(3,3- difluoroazetidin-l-yI)prop-2-en-l-one (9).
[00326] (Z)-3-(3-(3,5-Bis(trifluoromethyl)phenyl)- \H- 1,2,4-triazol- l-yl)acrylic acid (8) (30 g, 85.47 mmol) was dissolved in CH2C12 (600 mL) and cooled to 0 °C. 3,3- Difluoroazitidine hydrochloride (13.29 g, 102. 56 mmol), EDCI (14.24 g, 128.20 mmol) and HOBt (13.78 g, 102.56 mmol) were added at 0 °C followed by a dropwise addition of DIPEA (21.74 mL, 102.56 mmol) over 15 min. The reaction mixture was allowed to warm to room temperature and stirred for 4, transferred into water (250 mL) and extracted with CH2C12 (3 x 250 mL). The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure to give crude product, which was purified by silica gel chromatography (0.5 % MeOH in CH2C12) to give (Z)-3-(3-(3,5- bis(trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)-l -(3,3 -difiuoroazetidin- 1 -yl)prop-2-en- 1 - one (9) (Yield: 12 g, 33%). 1H NMR (400 MHz, CDC13) δ 9.63 (s, 1H), 8.61 (s, 2H), 7.95 (s, 1H), 7.26 (d, J= 10.4 Hz, 1H), 5.68 (d, J= 10.8 Hz, 1H), 4.46-4.60 (m, 4H). LCMS: m/z 427.39 [M+H]+, tR = 3.04 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyI)-lH-l,2,4-triazol-l-yl)-2,3-dibromo-l- (3,3-difluoroazetidin-l-yI)propan-l-one (302).
1003271 3-(3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3-dibromo-l-(3,3- difluoroazetidin-l-yl)propan-l-one (302) was synthesized using general procedure 1 (Yield: 0.4 g, 54%). !H NMR (400 MHz, DMSO-c¾ δ 9.22 (s, 1H), 8.59 (s, 2H), 7.38 (d, J= 8 Hz, 1H), 5.65 (d, J= 12 Hz, 1H), 5.06 (t, J= 12 Hz, 1H), 4.77-4.74 (m, 2H), 4.59-4.52 (m, 2H): LCMS : m/z 586.84 [M+H]+, tR = 2.80 min.
Synthesis of (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dichloro-l-(3,3-difluoroazetidin-l-yl)propan-l-one (303):
Figure imgf000094_0001
[00328] (Z)-3-(3-(3, 5-Bis (trifluoromethyl) phenyl)-lH-l, 2, 4-triazol-l-yl)-l-(3, 3- difluoroazetidin-l-yl) prop-2-en-l-one (9) (0.2 g, 0.4 mmol) was dissolved in CH2CI2 (10 mL) and cooled to 0 °C. Chlorine gas was purged in to reaction mixture for 1 h at 0 °C and then poured into water (25 mL) and extracted with EtOAc (3 x 25 mL). The combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (50% EtOAc in hexane) to obtain (Z)-3 - (3 -(3 ,5 -bis(trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)-2,3 -dichloro- 1 -(3 , 3 - difluoroazetidin-l-yl) propan-l-one (303). Yield (0.1 g, 42 %). 1H NMR (400 MHz, CDCI3) δ 8.65 (s, 2H), 8.42 (s, 1H), 7.98 (s, 1H), 6.53 (d, J= 12 Hz, 1H), 5.06 (d, J= 12 Hz, 1Ή), 4.88-4.76 (m, 2H), 4.59-4.53 (m, 2H). LCMS: m/z 497.1 [M+H] +, tR: 2.89 min.
Synthesis of isopropyl 3-(3-(3,5-bis(trifluoroniethyl)phenyl)-l H-l ,2,4-triazol-l -yl)-2- bromopropanoate (304).
Figure imgf000094_0002
Syn thesis of isopropyl 2,3-dibromopropanoate (11).
[00329] 2,3-dibromopropanoic acid (10) (0.250 g, 10.78 mmol) was dissolved in isopropyl alcohol (2 mL) at room temperature. BF3.Etherate (0.3 mL, 21.56 mmol) was added to the reaction mixture and heated at 70-80 °C for 3 h. The reaction mixture was allowed to cool to room temperature, transferred into iced water, and extracted with ethyl acetate (3 x 20 mL). The combined organic layers were washed with saturated solution of sodium bicarbonate, brine, and dried over anhydrous Na2S04. The organic layer was concentrated under reduced pressure to give pure isopropyl 2,3-dibromopropanoate (11) (Yield: 0.20 g, 68%) which was used in the next step without further purification. !H NMR (400 MHz, CDC13) δ 5.18-5.1 1 (m, 1H), 4.52-4.39 (m, 1H), 3.96-3.91 (m, 1H), 3.73-3.67 (m, 1H), 1.34- 1.27 (m, 6H).
Synthesis of isopropyl 3-(3-(3,5-bis(tritluoromethyI)phenyI)-l//-l ,2,4-triazol-l-yl)-2- bromopropanoate (304).
[00330] 3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazole (3) (0.170 g, 0.60 mmol) was dissolved in dichloromethane (10 mL) at room temperature. The reaction mixture was cooled to 0 °C, and isopropyl 2,3-dibromopropanoate (0.200 g, 0.72 mmol) was added followed by a dropwise addition of DIPEA (0.125 mL, 0.72 mmol) at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 2 h. The reaction mixture was transferred into iced water and extracted with ethyl acetate (3 x 25 mL). The combined organic layers were washed with brine and dried over anhydrous Na2S04. The organic layer was concentrated under reduced pressure to give the crude product which was purified by silica gel chromatography (0-5 % ethyl acetate/ n-hexane) to obtain isopropyl 3- (3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-bromopropanoate (304). (Yield: 3 mg, 1%) 1H NMR (400 MHz, DMSO^) δ 8.57 (s, 2H), 8.24 (s, 1H), 7.92 (s, 1H), 5.13-5.09 (m, 1H), 4.91-4.86 (m, 1 H), 4.76-4.73 (m, 1H), 1.31 (d, J = 6.4 Hz, 1H), 1.29 (d, J = 6.4 Hz, 6H). LCMS: m/z 476.27 [M+2], fR = 3.01 min.
Synthesis of isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dibromo-7V-pivaloylpropanehydrazide (305).
Figure imgf000096_0001
[00331] ( l-S-CS-CS^-bisCtrifluoromethy^ heny^-lH-l^^-triazol-l-yl)-^- pivaloylacrylohydrazide (12) (0.2 g, 0.448 mmol) was dissolved in dichloromethane (10 mL) at room temperature. The reaction mixture was cooled to 0 °C and bromine (0.045 mL, 0.89 mmol) was added dropwise. The reaction mixture was stirred for 15 minutes at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 6 hours. The reaction mixture was transferred into iced water and extracted with dichloromethane (3 x 50 mL). The combined organic layers were washed with brine and dried over anhydrous Na2S04. The organic layer was concentrated under reduced pressure to give crude compound, which was purified by silica gel chromatography (0-15% ethyl acetate/n- hexane) to obtain isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dibromo-TV-pivaloylpropanehydrazide (305) (Yield: 0.1 g, 38 %). 1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 9.54 (s, 1H), 9.01 (s, 1H), 8.56 (s, 2H), 8.28 (s, 1H), 7.35 (d, J = 10 Hz, 1H), 5.37 (d, J = 10 Hz, 1H), 1.03 (s, 9H). LCMS: m/z 607.9 [M]\ tR = 2.66 mm.
Example 2. Synthetic Procedures: Compounds of Structural Formula II
Synthesis of (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid
(8).
Figure imgf000097_0001
Figure imgf000097_0002
Figure imgf000097_0003
Synthesis of 3,5-bis(trifluoromethyl)benzothioamide (2).
[00332] 3,5-Bis(trifluoromethyl)benzonitrile (1) (200 g, 836.4 mmol) was dissolved in DMF (1 L). Sodium hydrosulphide hydrate (123.7 g, 1678 mmol) and magnesium chloride hexahydrate (186.7 g, 920 mmol) were added and the reaction mixture was stirred at room temperature for 3 h. The reaction mixture was transferred into iced water (10 L) and extracted with EtOAc (3 X 1L). The combined organic layers were washed with brine (3 X 500 mL), dried over anhydrous Na2S04 and concentrated under reduced pressure to afford 3,5-bis(trifluoromethyl)benzothioamide (2) (yield: 205 g, 90%). The product was used without further purification in the following step. LCMS: m/z 274.04 [M+H]+, ¾ = 2.78 min.
Synthesis of 3-(3,5-bis(trifIuoromethyl)phenyl)-lH-l,2,4-triazole (3).
[00333] 3,5-Bis(trifluoromethyl)benzothioamide (2) (205.65 g, 752 mmol) was dissolved in DMF (1.028 L) and hydrazine hydrate monohydrate (71 mL) was added dropwise. The reaction mixture was stirred at room temperature for 1 h, then formic acid (1.028 L) was added dropwise over 45 min and the reaction mixture was heated at 90 °C for 3 h. The reaction mixture was allowed to cool to room temperature, transferred into iced water (10 L), and extracted with EtOAc (3 X 1L). The combined organic layers were washed with brine (3 X 500 mL), dried over anhydrous Na2S04 and concentrated under reduced pressure and crystallized from petroleum ether to give 3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4- triazole (3) (yield: 160 g, 75%). ¾ NMR (400 MHz, DMSO-c¾) δ 14.52 (s, 1H), 8.77 (d, J = 5.4 Hz, 1 H), 8.53 (s, 2H), 8.18 (s, 1H). LCMS: m/z 282.19 [M+H]+, tR = 2.69 min. Synthesis of isopropyl propiolate (5).
[00334] Propiolic acid (4) (1000 g, 14.28 mol) was dissolved in -PrOH (8 L). BF3- etherate (4.54 kg, 25.7 mol) was added slowly at 25 °C over a period of 30 minutes. The temperature of the reaction mixture was gradually increased to 90 °C and stirred for 3 h. GC monitoring after 3 h showed completion of the reaction. The reaction mixture was cooled to room temperature, quenched with 20 L of ice cold water and stirred for 30 minutes. 10 L of dichloromethane was added to the reaction mixture and stirred for an additional 30 min. The organic layers were separated and the aqueous layer was re-extracted with 5 L of
dichloromethane. The combined organic layers were washed with 10 L of brine, dried over anhydrous Na2S04, and concentrated under reduced pressure at 35 - 40 °C (product is volatile) to obtain isopropyl propiolate (5) as a brown liquid (yield: 1.32 kg, 81%). Purity 89.67% (GC); 1H NMR (300 MHz, CDC13) δ 4.98-5.05 (m, 1H), 2.85 (s, 1H), 1.22 (d, J= 6.6 Hz, 6H).
Synthesis of ( )-isopropyl 3-iodoacrylate (6).
[00335] Isopropyl propiolate (5) (1000 g, 8.92 mol) was added in acetic acid (3.7 L) at 25 °C and the reaction mixture was stirred for 10 min. Sodium iodide (2.138 kg, 13.98 mol) was added (a dark brown color was observed) while stirring. The temperature was increased to 110 °C and stirred for 1.5 h. GC monitoring showed completion of the reaction after 1.5 h. The reaction mixture was cooled to room temperature, quenched with iced water (18.75 L) and stirred for 30 min. MTBE (5 L) was added to the reaction mixture and stirred for an additional 30 min. The organic layers were separated and the aqueous layer was re-extracted with MTBE (5 L). The combined organic layers were washed with NaHC03 (2 x 10 L), NaHS03 (2 x 5 L), brine (5.2 L), dried over anhydrous sodium sulfate and concentrated under reduced pressure at 35 °C to obtain (Z)-isopropyl 3-iodoacrylate (6) as a brown liquid. Yield (1.49 kg, 70%). Purity 87.34% (GC); 1H NMR (300 MHz, CDC13) δ 7.38 (d, J= 8.7 Hz, 1H), 6.83 (d, J= 8.7 Hz, 1H), 5.08-5.131 (m, 1H), 1.28 (d, 6H, J= 6.3 Hz).
Synthesis of (Z)-isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l- yl)acrylate (7).
[00336] 3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazole (3) (160 g, 569 mmol) was added in DMF (0.96 L). DAB CO (127.74 g, 1138 mmol) was then introduced and the reaction mixture was stirred for 30 min at room temperature. Iodoester (6) (150.32 g, 626 mmol) was added dropwise over a period of 1 h at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for an additional 2 h. The reaction mixture was quenched with cold water (5 L), stirred for 15 min, and extracted with ethyl acetate (3 X 1 L). The combined organic layers were washed with saturated solution of sodium thiosulphate (2 X 500 mL) followed by brine (3 X 500 mL), dried over anhydrous sodium sulfate (100 g) and concentrated under reduced pressure. The crude product (250 g) was stirred in methanol (625 mL) and maintained at 0-5 °C for 1 h. The precipitated solid was filtered and washed with cold methanol (100 mL), to obtain (Z)-isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH- l,2,4-triazol-l-yl)acrylate (7) as while solid. Yield (138 g, 61%). 1H NMR (400 MHz, DMSC ¾ δ 9.75 (s, 1H), 8.63 (s, 2H), 7.95 (s, 1H), 7.30 (d, J = 10.8 Hz, 1H), 5.77 (d, J = 10.8 Hz, 1H), 5.13-5.19 (m, 1H), 1.34 (d, J = 6.4 Hz, 6H). LCMS: m/z 394.24 [M+H]+, 7R = 2.95 min.
Synthesis of (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid (8).
General procedure 1. Ester hydrolysis.
[00337] (Z)-Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylate (7) (4 g, 10.17 mmol) was added to a mixture of THF:H20 (1 : 1, 80 mL) at room temperature. The reaction mixture was cooled to 0 °C and cold lithium hydroxide monohydrate solution (2.13 g in 10 mL water) was added over a period of 30 min. The reaction mixture was slowly warmed to room temperature and stirred for 3 h. The reaction mixture was quenched with iced water (40 mL) and stirred for 30 min. The pH was adjusted to 1-2 using 3M HC1 solution (25 mL) and the reaction mixture was extracted with ethyl acetate (3 x 70 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain the crude compound which was crystallized using petroleum ether (25 mL). The solid precipitate was filtered and washed with petroleum ether (10 mL), dried under reduced pressure to give (Z)-3-(3-(3,5- bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid (8) as a white solid. Yield (3 g, 84%). ¾ NM (400 MHz, DMSO-<¾ δ 13.01 (bs, 1H), 9.37 (s, 1H), 8.51 (s, 2H), 8.28 (s, 1H), 7.49 (d, J = 10 Hz, 1H), 5.97 (d, J = 10 Hz, 1H). Syntheses of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-3-bromo- V- (pyrazin-2-yl)acrylohydrazide (100), isopropyl -3-(3-(3,5-bis(trifluoromethyl)phenyI)- -l,2,4-triazol-l-yl)-3-bromoacrylate (101).
Figure imgf000100_0001
Synthesis of isopropyl 3-(3-(3, 5-bis (trifluoromethyl) phenyl)- lH-1 , 2, 4-triazol-l-yl)-2, 3-dibromopropanoate (9).
General Procedure 2. Bromination.
[00338] (Z)-Isopropyl 3 -(3 -(3 ,5-bis(trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)acrylate (7) (20 g, 50.85 mmol) was dissolved in dichloromethane (200 mL) at room temperature. Bromine (5.24 mL, 101.71 mmol) was added dropwise over 15 min at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 6 h, transferred into iced water, and extracted with CH2C12 (3 x 500 mL). The combined organic layers were washed with saturated aqueous sodium bisulphite (500 mL) solution followed by brine, dried over anhydrous Na2S04 and concentrated under reduced pressure to give the crude product, which was purified by silica gel chromatography (0-15% ethyl acetate/n-hexane) to yield isopropyl 3-(3 -(3,5-bis(trifluoro methyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)-2,3 - dibromopropanoate (9). (Yield: 21 g, 75%). 1H NMR (400 MHz, DMSO- 6) 5 9.11 (s, 1H), 8.50 (s, 2H), 8.31 (s, 1H), 7.41 (d, J = 9.6 Hz, 1H), 5.54 (d, J = 9.6 Hz, 1H), 4.82- 4.79 (m, 1H), 1.05-1.00 (m, 6H). LCMS: m/z 554.09 [M+H]+, ¾ = 3.12 min.
Synthesis of isopropyl 3-(3-(3, 5-bis (trifluoromethyl) phenyl)-lH-l,2,4-triazol-l-yl)-2- bromoacrylate (10) and isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol- l-yl)-3-bromoacrylate (101).
General Procedure 3. Elimination. [00339] Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dibromopropanoate (9) (12 g, 21.7 mmol) was dissolved in tetrahydrofuran (120 mL) and 3M lithium hydroxide monohydrate solution in water (14.4 mL, 43.5 mmol) was added at -20 °C. The reaction mixture was stirred at -20 °C for 2 h. The reaction mixture was diluted with water (120 mL) and extracted with ethyl acetate (3 x 200 mL). The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (4-8% EtOAc in hexanes) to give isopropyl 3-(3-(3,5-bis(trifluoromethyl) phenyl)-lH-l,2,4-triazol-
1- yl)-2-bromoacrylate (10) (yield: 3.56 g, 34%); Ή NMR (400 MHz, DMSO-i¾) δ 9.47 (s, 1H), 8.93 (s, 1H), 8.58 (s, 2H), 8.34 (s, 1H), 5.12-5.09 (m,lH), 1.34- 1.25 (m,6H) and isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l ,2,4-triazol-l-yl)-3-bromoacrylate (101) (Yield: 6 g, 58 %); Ή NMR (400 MHz, DMSO-i¾) δ 8.97 (s, 1H), 8.46 (s, 2H), 8.32 (s, 1H), 8.10 (s, 1H), 5.12-5.06 (m, 1H), 2.51-2.50 (m, 6H).
Synthesis of 3-(3-(3,5-bis(trifluoronicthyl)phcnyl)-lH-l,2,4-triazol-l-yl)-3- bromoacrylic acid (11).
[00340] Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-3- bromoacrylate (101) (0.4 g, 0.84 mmol) was dissolved in THF (10 mL) at room temperature. 3N lithium hydroxide solution in water (0.85 mL, 2.54 mmol) was added at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 4 h, transferred into iced water, and acidified to pH 1-2 using 3M HC1 solution (3 mL) and extracted with ethyl acetate (3 x 25 mL). The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure and purified by silica gel column chromatography (5% MeOH in CH2C12) to give 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH- l,2,4-triazol-l-yl)-3-bromoacrylic acid (11). Yield: 0.2 g, 55%; 'H NMR (400 MHz, DMSO- d6) δ 14.16 (s, 1H), 9.02 (s, 1H), 8.49 (s, 2H), 8.29 (s, 1H), 7.90 (s, 1H). LCMS: m/z 430.06 [M+H]+, tR = 2.47 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyI)phenyl)-lH-l,2,4-triazol-l-yl)-3-bromo-N'- (pyrazin-2-yl)acrylohydrazide (100).
[00341] 3-(3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-3-bromo-A^-(pyrazin-
2- yl)acrylohydrazide (100) was synthesized using General Procedure 4. Yield (54%). Ή NMR (400 MHz, DMSO d6) δ 10.63 (s, 1H), 9.44 (s, 1H), 9.20 (s, 1H), 8.77 (s, 1H), 8.59 (s, 2H), 8.35 (s, 1H), 8.13-8.12 (m, 2H), 7.98 (s, 1H). LCMS: m/z 523.89 [M+H]+, tR = 2.502 min.
Synthesis of 3-(3-(3,5-bis(trifluoroinethyl)phenyl)-lH-l,2,4-triazol-l-yl)-3-broino-l- (3,3-difluoroazetidin-l -yl)prop-2-en-l-one (102).
Figure imgf000102_0001
Synthesis of (Z)-3-(3-(3,5-bis(trifluoroinethyl)phenyl)-lH-l,2,4-triazol-l-yl)-l-(3,3- difluoroazetidin-l-yl)prop-2-en-l-one (12).
General Procedure 4: Amide bond formation.
[00342] (Z)-3-(3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid (8) (30 g, 85.47 mmol) was dissolved in CH2C12 (600 mL) and cooled to 0 °C. 3,3- Difluoroazitidine hydrochloride (13.29 g, 102. 56 mmol), EDCI (14.24 g, 128.20 mmol) and HOBt (13.78 g, 102.56 mmol) were added at 0 °C followed by a dropwise addition of DIPEA (21.74 mL, 102.56 mmol) over 15 min. The reaction mixture was allowed to warm to room temperature and stirred for 4 h, then was transferred into water (250 mL) and extracted with CH2C12 (3 x 250 mL). The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure to give crude product, which was purified by silica gel chromatography (0.5% MeOH in CH2C12) to give (Z)-3 -(3 -(3 , 5 -bis(trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)-l -(3,3 -difluoroazetidin- 1 - yl)prop-2-en-l-one (12) (Yield: 12 g, 33%). 1H NMR (400 MHz, CDC13) δ 9.63 (s, 1H), 8.61 (s, 2H), 7.95 (s, 1H), 7.26 (d, J= 10.4 Hz, 1H), 5.68 (d, J= 10.8 Hz, 1H), 4.46-4.60 (m, 4H). LCMS: m/z 427.39 [M+H]+, ¾ = 3.04 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3-dibromo-l- (3,3-difluoroazetidin-l-yl)propan-l-one (13). [00343] 3-(3-(3,5-Bis(trifluoromethyl)phenyl) H-l,2,4-triazol-l-yl)-2,3-dibromo-l-(3,3- difluoroazetidin-l-yl)propan-l-one (13) was synthesized using General Procedure 2 (Yield: 0.4 g, 54%). 1H NMR (400 MHz, OMSO-d6) δ 9.22 (s, 1H), 8.59 (s, 2H), 7.38 (d, J- 8 Hz, 1H), 5.65 (d, J= 12 Hz, 1H), 5.06 (t, J= 12 Hz, 1H), 4.77-4.74 (m, 2H), 4.59-4.52 (m, 2H): LCMS: m/z 586.84 [M+H]+, tR = 2.802 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-bromo-l-(3,3- difluoroazetidin-l-yl)prop-2-en-l-one (14) and 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH- l,2,4-triazol-l-yl)-3-bromo-l-(3,3-difluoroazetidin-l-yl)prop-2-en-l-one (102).
[00344] 3-(3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-bromo-l-(3,3- difluoroazetidin-l -yl)prop-2-en-l -one (14) and 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH- 1 ,2,4-triazol- 1 -yl)-3 -bromo- 1 -(3 ,3-difluoroazetidin- 1 -yl)prop-2-en- 1 -one (102) were synthesized using General Procedure 3 (Yield: 0.2 g, 53%). 3-(3-(3,5- bis(trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)-2-bromo- 1 -(3 ,3-difluoroazetidin- 1 - yl)prop-2-en-l-one (14): 1H NMR (400 MHz, CDC13) δ 9.23 (s, 1H), 8.63 (s, 2H), 8.52 (s, 1H), 7.98 (s, 1H), 4.71 -4.70 (m, 4H). LCMS: m/z 505.1 [M]",/R = 2.72 min. 3-(3-(3,5- bis(trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)-3 -bromo- 1 -(3 ,3 -difluoroazetidin- 1 - yl)prop-2-en-l-one (102) (Yield: 0.05 g, 13%); 1H NMR (400 MHz, CDC13) δ 8.56 (s, 2H), 8.41 (s, 1H), 7.97 (s, 1H), 7.44 (s, 1H), 4.61-4.49 (m, 4H), LCMS: m/z 507.1
[M+H]+, /R = 2.54 min.
Synthesis of (3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-3-chloro-l- (3,3-difluoroazetidin-l-yl)prop-2-en-l-one (103).
Figure imgf000103_0001
Synthesis of (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dichloro-l-(3,3-difluoroazetidin-l-yl)propan-l-one (J 5).
[00345] (Z)-3-(3-(3, 5-Bis (trifluoromethyl) phenyl)-lH-l, 2, 4-triazol-l-yl)-l-(3, 3- difluoroazetidin-l -yl) prop-2-en-l -one (12) (0.2 g, 0.4 mmol) was dissolved in CH2C12 (10 mL) and cooled to 0 °C. Chlorine gas was purged into the reaction mixture for 1 h at 0 °C. The reaction mixture was then poured into water (25 mL) and extracted with EtOAc (3 x 25 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (50% EtOAc in hexane) to obtain (Z)-3-(3-(3,5-bis(trifiuoromethyl)phenyl)-lH-l,2,4- triazol-l -yl)-2, 3 -dichloro-1 -(3, 3 -difluoroazetidin-l-yl) propan-l-one (15) (Yield: 0.1 g, 42%). !H NMR (400 MHz, CDC13) δ 8.65 (s, 2H), 8.42 (s, 1H), 7.98 (s, 1H), 6.53 (d, J = 12 Hz, 1 H), 5.06 (d, J= 12 Hz, 1H), 4.88-4.76 (m, 2H), 4.59-4.53 (m, 2H). LCMS: m/z 497.1 [M+H] +, tR: 2.89 min.
Synthesis of 3-(3-(3,5-bis(trifIuoromethyI)phenyI)-lH-l,2,4-triazoI-l-yI)-3-chIoro- l-(3,3-difluoroazetidin-l-yl)prop-2-en-l-one (103) and 3-(3-(3,5- bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-chloro-l-(3,3- difluoroazetidin-l-y])prop-2-en-l-one (16).
[00346] 3 -(3 -(3 , 5-Bis(trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)-2,3 -dichloro- 1 -(3 ,3- difluoroazetidin -l-yl)propan-l-one (15) (0.1 g, 0.2 mmol) was dissolved in methanol (10 mL) and cooled to 0 °C, at which temperature triethylamine (0.06 mL, 0.4 mmol) was added. The reaction mixture was allowed to warm to room temperature and stirred for 2 h, transferred into water (30 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure to obtain the crude product, which was purified by silica gel chromatography (50-60% EtOAc in hexanes) to give 3-(3-(3,5-bis(trifluoromethyl)phenyl)- lH- l ,2,4- triazol- l -yl)-3-chloro- l -(3,3-difluoroazetidin- l -yl)prop-2-en-l -one (103) (Yield: 0.01 g, 1 1 %); Ή NMR (400 MHz, CDC13) δ 8.561 (s, 2H), 8.51 (s, 1 H), 7.97 (s, 1H), 7.37 (s, 1 H), 4.60-4.54 (m, 4H) and 3-(3-(3 ,5-bis(trifluoromethyl)phenyl)- lH- l ,2,4-triazol-l - yl)-2-chloro-l -(3,3-difluoroazetidin-l -yl)prop-2-en-l -one (16) (Yield: 0.02 g, 21 %); Ή NMR (400 MHz, CDCI3) δ 9.12 (s, 1H), 8.64 (s, 2H), 8.44 (s, 1 H), 7.98 (s, 1H), 4.72- 4.74 (m, 4H).
Synthesis of (3-(3-(3,5-bis(trifluoromethyI)phenyl)-lH-l,2,4-triazol-l-yl)-3-chIoro- '-(pyrazin-2-yl)acrylohydrazide (104).
Figure imgf000105_0001
Synthesis of isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dichloropropanoate (17).
[00347] (Z)-Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl-lH-l,2,4-triazol-l-yl) (7) (0.5 g, 1.17 mmol) was dissolved in CH2C12 (10 mL) and cooled to 0 °C. Chlorine gas was purged into the reaction mixture for 1 h at 0 °C. The reaction mixture was transferred into water (300 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (50% EtOAc in hexane) to give isopropyl 3 -(3 -(3 ,5-bis(trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)-2,3 - dichloropropanoate (17). Yield (0.4 g, 68%), 1H NMR (400 MHz, DMSO-c/6) ^5 9.12 (s, 1H), 8.52 (s, 2H), 8.32 (s, 1H), 7.36 (d, J = 8.0 Hz, 1H), 5.70 (d, J = 8.0 Hz, 1H), 4.89- 4.86 (m, 1H), 1.08 (d, J = 28 Hz, 6H); LCMS: m/z 464.1 [M+H]VR = 3.22 min.
Synthesis of isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2- chloroacrylate (18) and isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol- l-yl)-3-chloroacrylate (19).
[00348] Isopropyl 3 -(3 -(3 ,5 -bis(trifluoromethyl)phenyl)- IH- 1 ,2,4-triazol- 1 -yl)-2,3 - dichloropropanoate (17) (1 g, 2.15 mmol) was dissolved in methanol (10 mL) and cooled to 0 °C. Triethylamine (0.6 mL, 4.31 mmol) was added at 0 °C. The reaction mixture was allowed to warm to room temperature, stirred for 2 h, transferred into water (300 mL), and extracted with EtOAc (3 x 100 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (50% EtOAc in hexane) to obtain isopropyl 3-(3-(3,5- bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-chloroacrylate (18) (Yield: 0.45 g, 48%); 1H NMR (400 MHz, DMSO-i 6) δ 9.47 (s, 1H), 8.93 (s, 1H), 8.58 (s, 2H), 8.34 (s, 1 H), 5.12-5.09 (m, 1H), 1.34-1.25 (m, 6H). LCMS : w/z 462.10 [M+H]", iR = 3.28 min and isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l ,2,4-triazol-l-yl)-3-chloroacrylate (19) (Yield: 0.2 g, 21%); NMR (400 MHz, DMSO-i/6) δ 8.95 (s, 1 H), 8.45 (s, 2H), 8.31 (s, 1 H), 8.10 (s, 1H), 5.12-5.09 (m, 1H), 1.34-1.25 (m, 6H). LCMS: m/z 462.10 [M+H]", tK = 3.30 min.
Synthesis of isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-3- chloroacrylic acid (20).
[00349] Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH- l ,2,4-triazol-l-yl)-3- chloroacrylic acid (20) was synthesized using General Procedure 1. Yield (0.25 g, 58%). 1H NMR (400 MHz, DMSO-t¾ δ 14.02 (s, 1H), 8.97 (s, 1H), 8.42 (s, 2H), 8.32 (s, 1H), 8.32 (s, 1H). LCMS: m/z 386.05 [M+H]+, ¾ = 2.19 min.
Synthesis of (3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-3-chloro- 7V-(pyrazin-2-yI)acrylohydrazide (104).
[00350] (3-(3-(3,5-Bis(trifluoromethyl)phenyl)-l H- l ,2,4-triazol-l-yl)-3-chloro-N'- (pyrazin-2-yl)acrylohydrazide (104) was synthesized using General Procedure 4. Yield (0.1 g, 40%). 1H NMR (400 MHz, DMSO-i¾ δ 10.71 (s, 1H), 9.45 (s, 1H), 9.20 (s, 1H), 8.59 (d, J = 6Hz, 3H), 8.34 (s, 1H), 8.13-8.1 1 (m, 2H), 7.96 (s, 1H). LCMS: m/z All.15 [M+H]+, 2.34 min.
Example 3. Synthetic Procedures: Compounds of Structural Formula III
Synthesis (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid (8).
Figure imgf000107_0001
Figure imgf000107_0002
Synthesis of 3,5-bis(trifluoromethyl)benzothioamide (2).
[00351] 3,5-Bis(trifluoromethyl)benzonitrile (1) (200 g, 836.4 mmol) was dissolved in DMF (1 L). Sodium hydrosulphide hydrate (123.7 g, 1678 mmol) and magnesium chloride hexahydrate (186.7 g, 920 mmol) were added and the reaction mixture was stirred at room temperature for 3 h. The reaction mixture was transferred into iced water (10 L) and extracted with EtOAc (3 X 1L). The combined organic layers were washed with brine (3 X 500 mL), dried over anhydrous Na2S04 and concentrated under reduced pressure to afford 3,5-bis(trifluoromethyl)benzothioamide (2) (Yield: 205 g, 90%). The product was used without further purification in the following step. LCMS: m/z 21 M [M+H]+, tR = 2.78 min
Synthesis of 3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazole (3).
[00352] 3,5-Bis(trifluoromethyl)benzothioamide (2) (205.65 g, 752 mmol) was dissolved in DMF (1.028 L) and hydrazine hydrate monohydrate (71 mL) was added dropwise. The reaction mixture was stirred at room temperature for 1 h followed by the dropwise addition of formic acid (1.028 L) over 45 min and heated at 90 °C for 3 h. The reaction mixture was allowed to cool to room temperature, transferred into iced water (10 L), and extracted with EtOAc (3 X 1L). The combined organic layers were washed with brine (3 X 500 mL), dried over anhydrous Na2S04 and concentrated under reduced pressure and crystallized from petroleum ether to give 3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazole (3) (Yield: 160 g, 75 %). 1H NMR (400 MHz, DMSO-< 6) δ 14.52 (s, 1H), 8.77 (d, J = 5.4 Hz, 1H), 8.53 (s, 2H), 8.18 (s, 1H). LCMS: m/z 282.19 [M+H]+, ¾ = 2.69 min.
Synthesis of isopropyl propiolate (5).
[00353] Propiolic acid (4) (1000 g, 14.28 mol) was dissolved in z-PrOH (8 L). BF3- etherate (4.54 kg, 25.7 mol) was added slowly at 25 °C over a period of 30 minutes. The temperature of the reaction mixture was gradually increased up to 90 °C and stirred for 3 h. GC monitoring after 3 h showed completion of the reaction. The reaction mixture was cooled to room temperature, quenched with 20 L of ice cold water and stirred for 30 minutes. 10 L of dichloromethane was added to the reaction mixture and stirred for an additional 30 min. The organic layers were separated and the aqueous layer was re-extracted with 5 L of dichloromethane. The combined organic layers were washed with 10 L of brine, dried over anhydrous Na2S04, and concentrated under reduced pressure at 35 - 40 °C (product is volatile) to obtain isopropyl propiolate (5) as a brown liquid (Yield: 1.32 kg, 81 %). Purity 89.67 % (GC); 1H NMR (300 MHz, CDC13) δ 4.98-5.05 (m, 1H), 2.85 (s, lH), 1.22 (d, J = 6.6 Hz, 6H).
Synthesis of (Z)-isopropyl 3-iodoacrylate (6).
[00354] Isopropyl propiolate (5) (1000 g, 8.92 mol) was added in acetic acid (3.7 L) at 25 °C and the reaction mixture was stirred for 10 min. Sodium iodide (2.138 kg, 13.98 mol) was added (a dark brown color was observed) while stirring. The temperature was increased to 1 10 °C and stirred for 1.5 h. GC monitoring showed completion of the reaction after 1.5 h. The reaction mixture was cooled to room temperature, quenched with iced water (18.75 L) and stirred for 30 min. MTBE (5 L) was added to the reaction mixture and stirred for an additional 30 min. The organic layers were separated and the aqueous layer was re-extracted with MTBE (5 L). The combined organic layers were washed with NaHC03 (2 x 10 L), NaHS03 (2 x 5 L), brine (5.2 L), dried over anhydrous sodium sulfate and concentrated under reduced pressure at 35 °C to obtain (Z)-isopropyl 3-iodoacrylate (6) as a brown liquid. Yield (1.49 kg, 70%). Purity 87.34 % (GC); Ή NMR (300 MHz, CDC13) δ 7.38 (d, J= 8.7 Hz, 1H), 6.83 (d, J= 8.7 Hz, 1H), 5.08-5.131 (m, 1H), 1.28 (d, 6H, J= 6.3 Hz).
Synthesis of ( )-isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l- yl)acrylate (7).
[00355] 3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazole (3) (160 g, 569 mmol) was added in DMF (0.96 L). DABCO (127.74 g, 1 138 mmol) was then introduced and the reaction mixture was stirred for 30 min at room temperature. Iodoester (6) (150.32 g, 626 mmol) was added drop wise over a period of 1 h at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for an additional 2 h. The reaction mixture was quenched with cold water (5 L), stirred for 15 min, and extracted with ethyl acetate (3 X 1 L). The combined organic layers were washed with a saturated solution of sodium thiosulphate (2 X 500 mL) followed by brine (3 X 500 mL), dried over anhydrous sodium sulfate (100 g) and concentrated under reduced pressure. The crude product (250 g) was stirred in methanol (625 mL) and maintained at 0-5 °C for 1 h. The precipitated solid was filtered and washed with cold methanol (100 mL) to obtain (Z)-isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)- lH-l ,2,4-triazol-l-yl)acrylate (7) as white solid. Yield (138 g, 61%). 1H NMR (400 MHz, DMSO- 5) δ 9.75 (s, 1 H), 8.63 (s, 2H), 7.95 (s, 1H), 7.30 (d, J = 10.8 Hz, 1H), 5.77 (d, J = 10.8 Hz, 1 H), 5.13-5.19 (m, 1H), 1.34 (d, J = 6.4 Hz, 6H). LCMS: m/z 394.24 [M+H]+, tR = 2.95 min.
Synthesis (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid (8). General procedure 1. Ester hydrolysis.
[00356] (Z)-Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylate (7) (4 g, 10.17 mmol) was added to a mixture of THF:H20 (1 : 1, 80 mL) at room temperature. The reaction mixture was cooled to 0 °C and cold lithium hydroxide monohydrate solution (2.13 g in 10 mL water) was added over a period of 30 min. The reaction mixture was slowly warmed to room temperature and stirred for 3 h. The reaction mixture was quenched with iced water (40 mL) and stirred for 30 min. The pH was adjusted to 1-2 using 3M HC1 solution (25 mL) and the reaction mixture was extracted with ethyl acetate (3 x 70 mL). The combined organic layers were washed with brine (50 mL). dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain the crude compound which was crystallized using petroleum ether (25 mL). The solid precipitate was filtered and washed with petroleum ether (10 mL), dried under reduced pressure to give (Z)-3-(3-(3,5- bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid (8) as a white solid. Yield (3 g, 84%). 1H NMR (400 MHz, DMSO-d6) δ 13.01 (bs, 1 H), 9.37 (s, 1H), 8.51 (s, 2H), 8.28 (s, 1H), 7.49 (d, J = 10 Hz, 1 H), 5.97 (d, J = 10 Hz, 1 H). Syntheses of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-bromo-V-(pyrazin-2-yl)acrylohydrazide (200) and isopropyl 3-(3-(3,5- bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-bromoacrylic acid (204).
Figure imgf000110_0001
Synthesis of isopropyl 3-(3-(3, 5-bis (trifluoromethyl) phenyl)-lH-l, 2, 4-triazol-l-yl)-2, 3-dibromopropanoate (9).
General Procedure 2. Bromination.
[00357] (Z)-Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylate (7) (20 g, 50.85 mmol) was dissolved in dichloromethane (200 mL) at room temperature. Bromine (5.24 mL, 101.71 mmol) was added dropwise over 15 min at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 6 h, transferred into iced water and extracted with CH2CI2 (3 x 500 mL). The combined organic layers were washed with saturated aqueous sodium bisulphite (500 mL) solution followed by brine, dried over anhydrous Na2S04 and concentrated under reduced pressure to give the crude product, which was purified by silica gel chromatography (0-15% ethyl acetate/n-hexane) to yield isopropyl 3-(3-(3,5-bis(trifluoro methyl)phenyl)-lH-l ,2,4-triazol-l-yl)-2,3- dibromopropanoate (9). Yield: 21 g, 75%. 1H NMR (400 MHz, DMSO-< ) δ 9.1 1 (s, 1H), 8.50 (s, 2H), 8.31 (s, 1H), 7.41 (d, J = 9.6 Hz, 1H), 5.54 (d, J = 9.6 Hz, 1H), 4.82-4.79 (m, 1H), 1.05-1.00 (m, 6H). LCMS: m/z 554.09 [M+H]+, tR = 3.12 min.
Synthesis of isopropyl 3-(3-(3, 5-bis (trifluoromethyl) phenyl)-lH-l,2,4-triazol-l-yl)-2- bromoacrylate (201) and isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4- triazol- 1 -yl)-3-bromoacrylate (10).
General Procedure 3. Elimination.
[00358] Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l ,2,4-triazol-l-yl)-2,3- dibromopropanoate (9) (12 g, 21.7 mmol) and lithium hydroxide monohydrate (1.83 g, 43.5 mmol) were added in tetrahydrofuran (120 mL) at -20 °C. The reaction mixture was stirred at -20 °C for 2 h. The reaction mixture was diluted with water (120 mL) and extracted with ethyl acetate (3 x 200 mL). The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (4-8% EtOAc in hexanes) to give isopropyl 3-(3-(3,5- bis(trifluoromethyl) phenyl)- lH-l ,2,4-triazol-l-yl)-2-bromoacrylate (201) (Yield: 35.6 g, 34%), 1H NMR (400 MHz, DMSO-i¾ δ 9.47 (s, 1 H), 8.93 (s, 1 H), 8.58 (s, 2H), 8.34 (s, 1H), 5.12-5.09 (m, 1H), 1.34-1.25 (m, 6H) and isopropyl 3-(3-(3,5- bis(trifluoromethyl)phenyl)-lH-l ,2,4-triazol-l-yl)-3-bromoacrylate (101); yield (6 g, 58 %), !H NMR (400 MHz, DMSO-<¾) δ 8.97 (s, 1 H), 8.46 (s, 2H), 8.32 (s, 1 H), 8.10 (s, 1 H), 5.12-5.06 (m, 1H), 2.51 -2.50 (m, 6H).
Synthesis of isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2- bromoacrylic acid (204).
[00359] Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2- bromoacrylate (201) (15 g, 31.7 mmol) and 3M lithium hydroxide solution (31.7 mL, 95.3 mmol) were added in tetrahydrofuran (120 mL) at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 4 h, diluted with water (500 mL), neutralized by using 3M HC1 solution (30 mL), and extracted with ethyl acetate (3 x 300 mL). The combined organic layers were washed with brine, dried over anhydrous Na2S04, concentrated under reduced pressure, and purified by silica gel chromatography (5% MeOH in C¾C12) to give isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l ,2,4-triazol-l-yl)-2- bromoacrylic acid (204). Yield (7.5 g, 52%). 1H NMR (400 MHz, DMSO-i¾ δ 14.03 (s, 1H), 9.44. (s, 1H), 8.92 (s, 1H), 8.57 (s, 1 H), 8.32 (s, 1H), LCMS: m/z 428.06 [M+H]+, ¾ = 2.33 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazoI-l-yl)-2-bromo- N'-(pyrazin-2-yl)acrylohydrazide (200).
[00360] 3-(3-(3,5-bis(trifiuoromethyl)phenyl)-lH-l ,2,4-triazol-l -yl)-2-bromo-N,- (pyrazin-2-yl)acrylohydrazide (200) was synthesized using General Procedure 4. Yield (54%). 1H NMR (400 MHz, DMSO d6) δ 10.62 (s, 1H), 9.44 (s, 1H), 9.2 (s, 1H), 8.77 (s, 1H), 8.59 (S, 2H), 8.34 (s, 1H), 8.13-8.1 1 (m, 1H), 7.98 (s, 1H). LCMS: m/z 522.2
[Μ+Η]+,¾ = 2.50 min. Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazoI-l-yl)-2-bromo-l- (3,3-difluoroazetidin-l -yl)prop-2-cn-l -one (202).
Figure imgf000112_0001
11 12
-20 °C LiOH.H20,
THF, H20
Figure imgf000112_0002
Synthesis of (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-l-(3,3- difluoroazetidin-l-yl)prop-2-en-l-one (11).
General Procedure 4: Amide bond formation.
[00361] (Z)-3-(3-(3,5-Bis(trifluorornethyl)phenyl)-lH-l,2,4-triazol-l-yl)acrylic acid (8) (30 g, 85.47 mmol) was dissolved in CH2C12 (600 mL) and cooled to 0 °C. 3,3- Difluoroazetidine hydrochloride (13.29 g, 102.56 mmol), EDCI (14.24 g, 128.20 mmol) and HOBt (13.78 g, 102.56) were added to this reaction mixture at 0 °C followed by dropwise addition of DIPEA (21.74 mL, 102.56 mmol) within 15 min. The reaction mixture was slowly warmed to room temperature and allowed to stir further for 4 h. The reaction mixture was transferred into water (250 mL) and extracted with CH2C12 (3 x 250 mL). The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure to give crude product which was purified by silica gel
chromatography (0.5% MeOH in CH2C12) to give (2 -3-(3-(3,5-bis(trifluoromethyl)phenyl)- lH-l ,2,4-triazol-l-yl)-l-(3,3-difluoroazetidin-l-yl)prop-2-en-l-one (11) (Yield: 12 g, 33%); 1H NMR (400 MHz, CDC13) 9.63 (s, 1H), 8.61 (s, 2H), 7.95 (s, 1H), 7.26 (d, J= 10.4 Hz, 1H), 5.68 (d, J= 10.8 Hz, 1H), 4.46-4.60 (m, 4H). LCMS: m/z 427.39 [M+H]+, ¾ = 3.04 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3-dibromo-l- (3,3-difluoroazetidin-l-yl)propan-l-one (12). [00362] 3-(3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3-dibromo-l-(3,3- difluoroazetidin-l-yl)propan-l-one (12) was synthesized using General Procedure 2. Yield (0.4 g, 54%). 1H NMR (400 MHz, OMSO-d6) δ 9.22 (s, 1 H), 8.59 (s, 2H), 7.38 (d, J= 8 Hz, 1 H), 5.65 (d, J= 12 Hz, 1 H), 5.06 (t, J= 12 Hz, 1H), 4.77-4.74 (m, 2H), 4.59-4.52 (m, 2H): LCMS: m/z 586.84 [M+H]+, tR = 2.80 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-bromo-l-(3,3- difluoroazetidin-l-yl)prop-2-en-l-one (202) and 3-(3-(3,5-bis(trifluoromethyl)phenyl)- lH-l,2,4-triazol-l-yl)-3-bromo-l-(3,3-difluoroazetidin-l-yl)prop-2-en-l-one (13).
[00363] Compounds 202 and 13 were synthesized using General Procedure 3. 3-(3-(3,5- bis(trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)-2-bromo- 1 -(3 ,3 -difluoroazetidin- 1 - yl)prop-2-en-l-one (202) (Yield: 0.2 g, 53%); 1H NMR (400 MHz, CDC13) δ 9.23 (s, 1H), 8.63 (s, 2H), 8.52 (s, 1 H), 7.98 (s, 1H), 4.71-4.70 (m, 4H). LCMS: m/z 505.00 [M]"JR = 2.72 min. and 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-3-bromo-l-(3,3- difluoroazetidin-l-yl)prop-2-en-l-one (13) (Yield: 0.05 g, 13 %); 1H NMR (400 MHz, CDC13) δ 8.56 (s, 2H), 8.41 (s, 1H), 7.97 (s, 1H), 7.44 (s, 1H), 4.61 -4.49 (m, 4H), LCMS: m/z 507.13 [M]+, tR = 2.54 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2- bromoacrylamide (203).
Figure imgf000113_0001
[00364] Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2- bromoacrylic acid (204) (6.0 g, 13.95 mmol) was dissolved in THF (60 mL) and isobutyl chloroformate (2.86 g, 20.93 mmol), N-methylmorpholine (1.98 g, 19.53 mmol) were added at 0 °C. The reaction mixture was slowly warmed to room temperature and stirred at room temperature for 30 min. The reaction mixture was filtered and the filtrate was purged with ammonia gas for 15 min at 0 °C. The reaction mixture was transferred into iced water and extracted with ethyl acetate (3 x 100 mL). The combined organic layers were washed with brine and dried over anhydrous Na2S04. The organic layer was concentrated under reduced pressure to give crude product, which was purified by silica gel chromatography (5% MeOH in CH2C12) to give 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)- 2-bromoacrylamide (203) (Yield: 4.5 g, 75%); 'H NMR (400 MHz, DMSO-c¾ δ 9.41 (s, 1H), 8.71 (s, 1H), 8.57 (s, 1H), 8.33 (s, 1H), 8.01 (s, 1H), 7.95 (s, 1 H): LCMS: m/z 427.09 [M+H]-, tR = 6.87 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyI)phenyI)-lH-l,2,4-triazoI-l-yl)-2- bromoacrylonitrile (205).
Figure imgf000114_0001
203 205
[00365] 3-(3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-bromoacrylamide (203) (100 mg, 0.23 mmol) was dissolved in dimethylformamide (2 mL) and phosphorus oxychloride (0.5 mL) was added at 0 °C. The reaction mixture was stirred at 0 °C for 1 h. The reaction mixture was transferred into iced water (10 mL) and extracted with ethyl acetate (3 10 mL). The combined organic layers were washed with brine and dried over anhydrous Na2S04. The organic layer was concentrated under reduced pressure to give the crude product, which was purified by silica gel chromatography (40% EtOAc in hexane) to give 3- (3-(3,5-bis(trifiuoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-bromoacrylonitrile (205) (Yield: 0.05 g, 53%); 1H NMR (400 MHz, DMSO-i¾ δ 9.24 (s, 1H), 9.12 (s, 1H), 8.57 (s, 2H), 8.37 (s, 1H). LCMS: m/z 41 1.1 [M]+, tR = 2.89 min. Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-chloro-l- (3,3-difluoroazetidin-l- l)prop-2-en-l-one (206).
Figure imgf000115_0001
Synthesis of (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dichloro-l-(3,3-difluoroazetidin-l-yl)propan-l-one (14).
[00366] (Z)-3-(3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-l-(3,3- difluoroazetidin-l-yl)prop-2-en-l-one (11) (0.2 g, 0.4 mmol) was dissolved in CH2C12 (10 mL) and cooled to 0 °C. Chlorine gas was purged into the reaction mixture for 1 h at 0 °C. The reaction mixture was then poured into water (25 mL) and extracted with EtOAc (3 x 25 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (50% EtOAc in hexanes) to obtain (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4- triazol-l-yl)-2,3-dichloro-l-(3,3-difluoroazetidin-l-yl) propan-l -one (14) (Yield: 0.1 g, 42%); 1H NMR (400 MHz, CDC13) δ 8.65 (s, 2H), 8.42 (s, IH), 7.98 (s, IH), 6.53 (d, J= 12 Hz, IH), 5.06 (d, J= 12 Hz, IH), 4.88-4.76 (m, 2H), 4.59-4.53 (m, 2H). LCMS: m/z 497.1 [M+H] +, tR: 2.89 min. Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-3-chloro- l-(3,3-difluoroazetidin-l-yI)prop-2-en-l-one (15) and 3-(3-(3,5- bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-chloro-l-(3,3- difluoroazetidin-l-yl)prop-2-en-l-one (206).
[00367] 3-(3-(3,5-Bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3-dichloro-l-(3,3 difluoroazetidin -l -yl)propan-l-one (14) (0.1 g, 0.2 mmol) was dissolved in methanol (10 mL) and cooled to 0 °C, at which temperature triethylamine (0.06 mL, 0.4 mmol) was added. The reaction mixture was allowed to warm to room temperature and stirred for 2 h, transferred into water (30 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure to obtain the crude product, which was purified by silica gel chromatography (50-60% EtOAc in hexanes) to give 3-(3-(3 ,5-bis(trifluoromethyl)phenyl)- lH- 1 ,2,4- triazol-l-yl)-3-chloro- l -(3,3-difluoroazetidin- l -yl)prop-2-en-l -one (15) (Yield: 0.01 g, 1 1%); 1H NMR (400 MHz, CDC13) δ 8.561 (s, 2H), 8.51 (s, 1H), 7.97 (s, 1H), 7.37 (s, 1H), 4.60-4.54 (m, 4H) and 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH- l ,2,4-triazol- l - yl)-2-chloro-l -(3,3-difluoroazetidin- l -yl)prop-2-en-l -one (206) (Yield: 0.02 g, 21 %); 1H NMR (400 MHz, CDC13) δ 9.12 (s, 1H), 8.64 (s, 2H), 8.44 (s, 1H), 7.98 (s, 1H), 4.72- 4.74 (m, 4H).
Synthesis o 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-chloro- '-(pyrazin-2-yl)acrylohydrazide (207).
Figure imgf000116_0001
Synthesis of isopropyi 3-(3-(3,5-bis(trifluoromethyi)phenyl)-lH-l,2,4-triazoi-l-yl)-2,3- dichloropropanoate (16).
[00368] (Z)-Isopropyl 3-(3-(3,5-bis(trifluoromet yl)phenyl-lH-l,2,4-triazol-l-yl) (7) (0.5 g, 1.17 mmol) was dissolved in CH2CI2 (10 mL) and cooled to 0 °C. Chlorine gas was purged into the reaction mixture for 1 h at 0 °C. The reaction mixture was transferred into water (300 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (50% EtOAc in hexane) to give isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dichloropropanoate (16) (Yield: 0.4 g, 68%); Ή NMR (400 MHz, OMSO-d6) δ 9.12 (s, 1 H), 8.52 (s, 2H), 8.32 (s, 1H), 7.36 (d, J = 8.0 Hz, 1H), 5.70 (d, J = 8.0 Hz, 1H), 4.89- 4.86 (m, 1H), 1.08 (d, J = 28 Hz, 6H); LCMS: m/z 464.1 [M+H]+,¾ = 3.22 min.
Synthesis of isopropyl 3-(3-(3, 5-bis (trifluoromethyl) phenyl)-lH-l, 2, 4-triazol-l-yl)-2- chloroacrylate (17) and isopropyl 3-(3-(3, 5-bis (trifluoromethyl) phenyl)-lH-l, 2, 4- triazol-l-yl)-3-chloroacrylate (18).
[00369] Isopropyl 3-(3-(3,5-bis(trifiuoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2,3- dichloropropanoate (16) (1 g, 2.15 mmol) was dissolved in methanol (10 mL) and cooled to 0 °C. Triethylamine (0.6 mL, 4.31 mmol) was added at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 2 h, transferred into water (300 mL), and extracted with EtOAc (3 x 100 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (50% EtOAc in hexane) to obtain isopropyl 3-(3- (3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2-chloroacrylate (17) (Yield: 0.45 g, 48%); 1H NMR (400 MHz, DMSO-J^) δ 9.47 (s, 1H), 8.93 (s, 1H), 8.58 (s, 2H), 8.34 (s, 1H), 5.12-5.09 (m, 1H), 1.34-1.25 (m, 6H). LCMS: m/z 462.10 [M+H]\ fR = 3.28 min and isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-l/J-l ,2,4-triazol-l-yl)-3-chloroacrylate (18) (Yield: 0.2 g, 21%); NMR (400 MHz, DMSO-di) δ 8.95 (s, 1H), 8.45 (s, 2H), 8.31 (s, 1H), 8.10 (s, 1H), 5.12-5.09 (m, 1H), 1.34-1.25 (m, 6H). LCMS: m/z 462.10 [M+H]\ fR = 3.30 min.
Synthesis of isopropyl 3-(3-(3, 5-bis (trifluoromethyl) phenyl)-lH-l, 2, 4-triazol-l-yl)-2- chloroacrylic acid (19).
[00370] Isopropyl 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazol-l-yl)-2- chloroacrylic acid (19) was synthesized using General Procedure 1. Yield: 0.25 g, 58%. H NMR (400 MHz, DMSO-i 6) δ 14.03 (s, 1H), 9.44 (s, 1H), 8.92 (s, 2H), 8.57 (s, 1H), 8.32 (s, 1H). LCMS: m/z 386.05 [M+H]+, ¾ = 2.33 min.
Synthesis of 3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4-triazoI-l-yl)-2-chloro- A'-(pyrazin-2-yl)acrylohydrazide (207).
[00371] 3-(3-(3,5-Bis(tπfluoromethyl)phenyl)-lH-l ,2,4-triazol-l-yl)-2-chloro-N,- (pyrazin-2-yl)acrylohydrazide (207) was synthesized using General Procedure 4. Yield (0.05 g, 20%). 1H NMR (400 MHz, DMSO- ) δ 10.70 (s, 1H), 9.45 (s, 1H), 9.20 (s, 1H), 8.60 (d, J = 6 Hz, 3H), 8.37 (s, 1H), 8.12-8.10- (m, 2H), 7.96 (s, 1H). LCMS: m/z 476.18 [M+H]+, ¾ = 2.35 min.
Example 4. Assays. Certain compounds of the invention were tested in various assays. MTT Cell Proliferation Assay
[00372] The MTT cell proliferation assay was used to study the cytotoxic properties of the compounds. The assay was performed according to the method described by Roche
Molecular Biochemicals, with minor modifications. The assay is based on the cleavage of the tetrazolium salt, MTT, in the presence of an electron-coupling reagent. The water-insoluble formazan salt produced must be solubilized in an additional step. Cells grown in a 96-well tissue culture plate were incubated with the MTT solution for approximately 4 hours. After this incubation period, a water-insoluble formazan dye formed. After solubilization, the formazan dye was quantitated using a scanning multi-well spectrophotometer (ELISA reader). The absorbance revealed directly correlates to the cell number. The cells were seeded at 5,000-10,000 cells in each well of 96-well plate in 100 \iL of fresh culture medium and were allowed to attach overnight. The stock solutions of the compounds were diluted in 100xL cell culture medium to obtain eight concentrations of each test compound, ranging from 1 nM to 30 μΜ. After incubation for approximately 64-72 hours, 20 xL of CellTiter 96 Aqueous One Solution Reagent (Promega, G358B) was added to each well and the plate was returned to the incubator (37 °C; 5% C02) until an absolute OD of 1.5 was reached for the control cells. All optical densities were measured at 490 run using a Vmax Kinetic Microplate Reader (Molecular Devices). In most cases, the assay was performed in duplicate and the results were presented as a mean percent inhibition to the negative control±SE. The following formula was used to calculate the percent of inhibition: Inhibition (%) = (1- (ODo/OD)) X 100. [00373] The compounds were tested against Zl 38, MMl S and 3T3 cells. The Zl 38 cell line is a mature B-cell acute lymphoblastic leukemia cell line derived from a patient with chronic lumphocytic leukemia. The MMl S cell line was established from the peripheral blood of a human multiple myeloma patient. 3T3 cells are standard fibroblast cells; they were originally isolated from Swiss mouse embryo tissue.
[00374] The results of the MTT assay are reported in Tables 1 , 2 and 3.
Table 1. MTT Cell Proliferation Assay Results for Exemplary Compounds of Structural Formula I (A = <100 nM; B = 100 nM to <5 μΜ; C = 5 μΜ to 30 μΜ; D =
Figure imgf000119_0001
able 2. MTT Cell Proliferation Assay Results for Exemplary Compounds of
Figure imgf000120_0001
Table 3. MTT Cell Proliferation Assay Results for Exemplary Compounds of
Figure imgf000120_0002
Figure imgf000121_0001
BIBLIOGRAPHY
[00375] Cronshaw JM and Matunis MJ. 2004. The nuclear pore complex: disease associations and functional correlations TRENDS Endocrin Metab. 15:34-39 [00376] Falini B et al. 2006. Both carboxy-terminus NES motif and mutated tryptophan(s) are crucial for aberrant nuclear export of nucleophosmin leukemic mutants in NPMc+ AML Blood. 107:4514-4523.
[00377] Cai X and Liu X. 2008. Inhibition of Thr-55 phosphorylation restores p53 nuclear localization and sensitizes cancer cells to DNA damage. PNAS. 105: 16958-16963.
[00378] Daelemans D, Afonina E, Nilsson J 2002 A synthetic HIV-1 Rev inhibitor interfering with the CRM 1 -mediated nuclear export. Proc Natl Acad Sci U S A
99(22): 14440-5.98052-2517.
[00379] Davis JR et al. 2007. Controlling protein compartmentalization to overcome disease Pharmaceut Res. 24: 17-27.
[00380] Freundt E, Yu L, Park E, et al 2009 Molecular determinants for subcellular localization of the severe acute respiratory syndrome coronavirus open reading frame 3b protein. J Virol 83(13):6631-40.
[00381] Ghildyal R, Ho A, Dias M, et al 2009 The respiratory syncytial virus matrix protein possesses a Crml-mediated nuclear export mechanism. J Virol 83(1 1):5353-62.
[00382] Ghosh CC et al 2008 Analysis of nucleocytoplasmic shuttling of NF kappa B proteins in human leukocytes. Methods Mol Biol. 457:279-92.
[00383] Gupta N et al 2008 Retinal tau pathology in human glaucomas. Can J
Ophthalmol. 2008 Feb;43(l):53-60.
[00384] HoshinoL et al. 2008. Combined effects of p53 gene therapy and leptomycin B in human esophageal squamous cell carcinoma. Oncology. 75: 1 13-1 19.
[00385] Lain S et al. 1999a An inhibitor of nuclear export activates the p53 response and induces the localization of HDM2 and p53 to Ul A-positive nuclear bodies associated with the PODs Exp Cell Res. 248:457-472.
[00386] Lain S et al. 1999b. Accumulating active p53 in the nucleus by inhibition of nuclear export: a novel strategy to promote the p53 tumor suppressor function Exp Cell Res. 253 :315.
[00387] Muller PA et al. 2009 Nuclear-cytosolic transport of COMMD 1 regulates NF- kappaB and HIF-1 activity. Traffic 10(5):514-27.
[00388] Mutka S 2007 Nuclear Export Inhibitors (NEIs) as novel cancer therapies AACR Annual Meeting. Poster 5609. [00389] Mutka S, Yang W, Dong S, et al. 2009. Identification of nuclear export inhibitors with potent anticancer activity in vivo. Cancer Res. 69: 510-7.
[00390] Nakahara J et al. 2009. Abnormal expression of TIP30 and arrested
nucleocytoplasmic transport within oligodendrocyte precursor cells in multiple sclerosis J Clin Invest. 1 19: 169-181.
[00391] Noske A et al. 2008. Expression of the nuclear export protein chromosomal region maintenance/exportin 1/Xpol is a prognostic factor in human ovarian cancer. Cancer.
112: 1733-1743.
[00392] Pollard V & Malim M. 1998 The HIV-1 Rev protein Annu Rev Microbiol 52:491- 532.
[00393] Rawlinson S, Pryor M, Wright P, Jans D 2009 CRM 1 -mediated nuclear export of dengue virus RNA polymerase NS5 modulates interleukin-8 induction and virus production. J Biol Chem 284(23): 15589-97.
[00394] Sanchez V, Mahr J, Orazio N, et al 2007 Nuclear export of the human
cytomegalovirus tegument protein pp65 requires cyclin-dependent kinase activity and the Crml exporter. J Virol 81(21): 1 1730-6..
[00395] Sorokin AV et al. 2007. Nucleocytoplasmic transport of proteins . Biochemistry 72: 1439-1457.
[00396] Terry LJ et al. 2007. Crossing the nuclear envelope: hierarchical regulation of nucleocytoplasmic transport. Science 318: 1412-1416.
[00397] Van der Watt PJ et al. 2008. The Karyopherin proteins, Crml and Karyopherin betal, are overexpressed in cervical cancer and are critical for cancer cell survival and proliferation. Int J Cane. 124: 1829-1840.
[00398] Walsh MD et al. 2008 Exportin 1 inhibition attenuates nuclear factor-kappaB- dependent gene expression. Shock 29: 160-166.
[00399] Williams P, Verhagen J, Elliott G 2008 Characterization of a CRM 1 -dependent nuclear export signal in the C terminus of herpes simplex virus type 1 tegument protein UL47. J Virol 82(21): 10946-52.
[00400] Yang W 2007 Anti-tumor activity of novel nuclear export inhibitors (NEIs) in multiple murine leukemia models. AACR Annual Meeting. Poster 5597.
[00401] Yao Y et al. 2009. The expression of CRM1 is associated with prognosis in human osteosarcoma. Oncol Rep. 21 :229-35. [00402] Zimmerman TL et al 2006 Nuclear export of retinoid X receptor alpha in response to interleukin-1 beta-mediated cell signaling: roles for INK and SER260. J Biol
Chem281 : 15434-15440.
[00403] The teachings of all patents, published applications and references cited herein are incorporated by reference in their entirety.
[00404] While this invention has been particularly shown and described with references to example embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims

A compound of Structural Formula I:
Figure imgf000125_0001
(I),
or a pharmaceutically acceptable salt thereof, wherein:
X is -N- or -C(H)-;
Ra and R are each independently halo or hydrogen, wherein at least one of Ra and Rb is halo;
each R1 is independently selected from halo; haloalkyl; -(CH2)i-4R°; -(CH2)o-4OR°;
-O-(CH2)0-4C(O)OR°; -(CH2)0-4CH(OR°)2; -(CH2)0-4SR°; -(CH2)0.4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°; -(CH2)o-4-heterocyclyl, which may be substituted with R°;
-(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°; -CH=CH-heterocyclyl, which may be substituted with R°;
-CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3;
-(CH2)0-4N(R°)2; -(CH2)o.4N(R0)C(0)R°; -(CH2)0-4N(Ro)C(S)R°;
-(CH2)0-4N(R°)C(O)NR°2; -(CH2)0-4N(Ro)C(S)NR°2; -(CH2)0.4N(R°)C(O)OR°; -(CH2)o-4N(R°)N(R0)C(0)R°; -(CH2)0.4N(Ro)N(Ro)C(O)NRo 2;
-(CH2)o-4N(R°)N(R°)C(0)OR0 ; -(CH2)0-4C(O)R°; -(CH2)0-4C(S)R°;
-(CH2)0-4C(O)OR°; -(CH2)0-4C(O)SR°; -(CH2)0.4OC(O)R°;
-(CH2)o- OC(0)(CH2)0-4SR°, -(CH2)0-4SC(S)SRo ; -(CH2)0.4SC(O)R°;
-(CH2)0-4C(O)NR°2; -(CH2)0-4C(S)NR°2; -(CH2)0.4C(S)SR°; -(CH2)0-4OC(O)NR°2; -(CH2)o-4C(0)N(OR°)R°; -(CH2)0-4C(O)C(O)Ro; -(CH2)0-4C(O)CH2C(O)R°; -(CH2)o-4C(NOR0)R°; -(CH2)0-4SSR°; -(CH2)0-4S(O)2R°; -(CH2)0-4S(O)2OR°; -(CH2)0-4OS(O)2R°; -(CH2)0- S(O)2NR°2; -(CH2)0-4S(O)R°;
-(CH2)o-4N(R°)S(0)2NR°2; -(CH2)0-4N(Ro)S(O)2; -(CH2)0.4N(ORo)Ro ; -(CH2)o-4C(NH)NR°2; -(CH2)0-4P(O)2R°; -(CH2)0-4P(O)R°2; -(CH2)0-4OP(O)R°2; -(CH2)o-4OP(0)(OR0)2; -(CH2)0-4ON(Ro)2; and -(CH2)0-4C(O)O-N(R°)2, wherein: each R° is independently hydrogen, C1-6 aliphatic, -CH2-carbocyclyl,
-CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i-carbocyclyl,
-0(CH2)o-i-aryl, -O(CH2)0-i-heterocyclyl, -0(CH2)o-i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Q-Cs alkyl, -NH2, -N02,
-NH(unsubstituted Ci-C3 alkyl), -N(unsubstituted C1-C3 alkyl)2, -0-Ci-C3 alkyl, -C(0)OH, -C(0)0-(unsubstituted Ci-C3 alkyl), -C(0)-(unsubstituted Cx-C3 alkyl), -0-(unsubstituted Ci-C3 alkyl), and -S-(unsubstituted C1-C3 alkyl);
R2 is selected from -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(S)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, -C(S)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-C(S)-R4,
-C(S)-N(R7)-N(R7)-C(S)-R4, -C(0)-N(R7)-N(R7)-S(0)i-2-R4,
-C(S)-N(R7)-N(R7)-S(0)1 -2-R4 and heteroaryl, wherein:
R3 is selected from hydrogen, C1 -C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl, -(C0- C4 alkylene)-aryl, and -(C0-C4 alkyl ene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or
R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
each R7 is independently hydrogen or C1-C4 alkyl; and
n is 0, 1 , 2, 3, 4 or 5; wherein: unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
2. The compound of claim 1, wherein Ra and Rb are each independently halo.
3. The compound of claim 1 , wherein Ra and Rb are the same.
4. The compound of any one of claims 1-3, wherein the halo of Ra and Rb is chlorine or bromine.
5. The compound of any one of claims 1-4, wherein X is -C(H)-.
6. The compound of any one of claims 1 -5, wherein n is 0, 1 or 2.
7. The compound of any one of claims 1-6, wherein each R is independently selected from -CF3, -CN, halo, -OH, d-C3 alkyl, C3-C6 cycloalkyl, C3-Ci2 heterocycloalkyl, halo-Ci-C3 allcyl, -NH2, -N02, -NH(C,-C3 alkyl), -N(C,-C3 alkyl)(C,-C3 alkyl), -C(0)OH, -C(0)0-(Ci-C6 alkyl), -C(0)-(Ci-C3 alkyl), -0-(C!-C3 alkyl), -0-(Ci-C3 haloalkyl), and -S-(Ci-C3 alkyl), or is absent.
8. The compound of any one of claims 1-6, wherein each R1 is independently selected from halo, -Ci-C4 alkyl, -Ci-C4 haloalkyl and -O-C1-C4 alkyl, or is absent.
9. The compound of any one of claims 1 -5, represented by Structural Formula la:
Figure imgf000127_0001
or a pharmaceutically acceptable salt thereof, wherein: Rla and Rlb are each independently selected from halo; haloalkyl; -(CH^ R0; -(CH2)o-4OR0; -0-(CH2)o-4C(0)OR°; -(CH2)0-4CH(OR°)2; -(CH2)0-4SR°;
-(CH2)0-4-carbocyclyl, which may be substituted with R°; -(CH2)0-4-aryl, which may be substituted with R°; -(CH2)0-4-heterocyclyl, which may be substituted with R°; -(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°; -CH=CH-heterocyclyl, which may be substituted with R°; -CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3; -(CH2)0-4N(R°)2; -(CH2)0-4N(Ro)C(O)Ro; -(CH2)o-4N(R°)C(S)R0 ; -(CH2)o-4N(R0)C(0)NR°2; -(CH2)0-4N(R°)C(S)NR°2;
-(CH2)o-4N(R0)C(0)OR°; -(CH2)o-4N(R°)N(R°)C(0)R°;
-(CH2)o-4N(R0)N(R0)C(0)NR°2; -(CH2)0.4N(Ro)N(R°)C(O)OR°; -(CH2)0-4C(O)R°; -(CH2)0-4C(S)R°; -(CH2)o.4C(0)OR°; -(CH2)0-4C(O)SR°; -(CH2)0-4OC(O)R°;
-(CH2)0-40C(0)(CH2)o-4SR°, -(CH2)o-4SC(S)SR0; -(CH2)0-4SC(O)R°;
-(CH2)0.4C(O)NR°2; -(CH2)0-4C(S)NR°2; -(CH2)0.4C(S)SR°; -(CH2)0-4OC(O)NR°2; -(CH2)0-4C(O)N(OR°)R°; -(CH2)o-4C(0)C(0)R0; -(CH2)0-4C(O)CH2C(O)R°;
-(CH2)0-4C(NOR°)R°; -(CH2)0-4SSR°; -(CH2)0-4S(O)2R°; -(CH2)0-4S(O)2OR°;
-(CH2)o-4OS(0)2R0; -(CH2)0-4S(O)2NR°2; -(CH2)0.4S(O)R°;
-(CH2)o_4N(R°)S(0)2NR0 2; -(CH2)0-4N(Ro)S(O)2R°; -(CH2)0-4N(OR°)Ro;
-(CH2)o-4C(NH)NR°2; -(CH2)0-4P(O)2R°; -(CH2)0_4P(O)R°2; -(CH2)0-4OP(O)R°2;
-(CH2)0-4OP(O)(OR°)2; -(CH2)0-4ON(Ro)2; and -(CH2)0-4C(O)O-N(R°)2, wherein: each R° is independently hydrogen, Ci-6 aliphatic, -CH2-carbocyclyl,
-CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i-carbocyclyl,
-0(CH2)o-i-aryl, -0(CH2)o-i -heterocyclyl, -0(CH2)o-i -heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted CrC3 alkyl, halo-Ci-C3 alkyl, -NH2, -N02, -NH(unsubstituted C 1-C3 alkyl), -N(unsubstituted C1-C3 alkyl)2, -0-d-C3 alkyl, -C(0)OH, -C(0)0-(unsubstituted CrC3 alkyl), -C(0)-(unsubstituted Cj-Cs alkyl),
-0-(unsubstituted Ci-C3 alkyl), and -S-(unsubstituted Cj-C3 alkyl); and
m is 0 or 1.
10. The compound of claim 9, wherein Rla is -Ci-C4 haloalkyl.
11. The compound of claim 9 or 10, wherein Rlb is -C]-C4 haloalkyl.
12. The compound of claim 9, wherein Rla is -CF3 and Rlb is -CF3.
13. The compound of any one of claims 1-12, wherein R2 is selected from -C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4,
-C(0)-N(R7)-N(R7)-S(0)1-2-R4, and heteroaryl.
14. The compound of any one of claims 1-13, wherein R3 is selected from Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl.
15. The compound of any one of claims 1-14, wherein:
R2 is -C(0)-0-R3, and R3 is selected from optionally substituted CpC4 alkyl and C2-C4 alkenyl; or
R2 is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
R2 is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or
R2 is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0),-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(CrC4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl; or
R2 is optionally substituted C5-C6 heteroaryl.
16. The compound of any one of claims 1-13, wherein: R2 is -C(0)-0-R3, and R3 is hydrogen or is selected from optionally substituted C 1-C4 alkyl and C2-C4 alkenyl; or
R2 is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
R2 is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or
R2 is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C alkylene)-heterocyclyl and -(C0-C
alkylene)-heteroaryl; or
R2 is optionally substituted C5-C6 heteroaryl.
The compound of any one of claims 1-16, wherein R is -C(0)-0-CH(CH3)2,
-C(0)-NH-NH-C(0)-C(CH3)3 or
Figure imgf000130_0001
18. A compound represented by Structural Formula lb:
Figure imgf000130_0002
or a pharmaceutically acceptable salt thereof, wherein:
Ra and R are each independently halo or hydrogen, wherein at least one of Ra and R is halo; and
R2 is selected from -C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6),
-C(0)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-S(0)i-2-R4, and heteroaryl, wherein:
R is selected from hydrogen, Cj-C alkyl, C2-C4 alkenyl, C2-C4 alkynyl,
carbocyclyl, aryl, heterocyclyl and heteroaryl; R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(CrC4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl, -(Co-C4 alkylene)-aryl, and -(C0-C4 alkyl ene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or
R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; and
each R7 is independently hydrogen or Ci-C4 alkyl; wherein:
unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
19. The compound of claim 18, wherein Ra and Rb are each independently halo.
20. The compound of claim 18, wherein Ra and Rb are the same.
21. The compound of any one of claims 18-20, wherein the halo of Ra and Rb is chlorine or bromine.
22. The compound of any one of claims 18-21, wherein R is selected from Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl.
23. The compound of any one of claims 18-22, wherein:
R2 is -C(0)-0-R3, and R3 is selected from optionally substituted Cj-C alkyl and C2- C4 alkenyl; or
R2 is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
R2 is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or
R2 is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)I-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(d-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4
alkylene)-heteroaryl; or
R2 is optionally substituted C5-C6 heteroaryl.
The compound of any one of claims 18-21, wherein:
R2 is -C(0)-0-R3, and R3 is hydro gen or is selected from optionally substituted C1-C4 alkyl and C2-C4 alkenyl; or
R2 is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
R2 is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or
R2 is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)1 -2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4
alkylene)-heteroaryl; or
R2 is optionally substituted C5-C6 heteroaryl.
The compound of any one of claims 18-24, wherein R2 is -C(0)-0-CH(CH3)2,
-C(0)-NH-NH-C(0)-C(CH3)3 or
Figure imgf000132_0001
com ound represented by any one of the following structural formulas
Figure imgf000132_0002
Figure imgf000133_0001
or a pharmaceutically acceptable salt of any of the foregoing. 27. A compound of Structural Formula II:
Figure imgf000133_0002
or a pharmaceutically acceptable salt thereof, wherein:
X is -N- or -C(H)-;
each R1 is independently selected from halo; haloalkyl; -(CH2)i-4R°; -(CH2)o-4OR°;
-0-(CH2)o.4C(0)OR°; -(CH2)0-4CH(OR°)2; -(CH2)0-4SR°; -(CH2)0.4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°; -(CH2)o-4-heterocyclyl, which may be substituted with R°;
-(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°; -CH=CH-heterocyclyl, which may be substituted with R°;
-CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3;
-(CH2)o-4N(R°)2; -(CH2)o-4N(R°)C(0)R0; -(CH2)0-4N(R°)C(S)Ro;
-(CH2)0-4N(R°)C(O)NR°2; -(CH2)0-4N(Ro)C(S)NR°2; -(CH2)0-4N(R°)C(O)OR°; -(CH2)o-4N(R0)N(R°)C(0)R0; -(CH2)0-4N(Ro)N(Ro)C(O)NR°2; -(CH2)0-4N(R°)N(Ro)C(O)OR°; -(CH2)0-4C(O)R°; -(CH2)0-4C(S)R°;
-(CH2)0-4C(O)OR°; -(CH2)0-4C(O)SR°; -(CH2)MOC(0)R°;
-(CH2)0-4OC(0)(CH2)o-4SR°, -(CH2)0-4SC(S)SR°; -(CH2)0.4SC(O)R°;
-(CH2)0-4C(O)NR°2; -(CH2)0-4C(S)NR°2; -(CH2)0.4C(S)SR°; -(CH2)0-4OC(O)NR°2;
-(CH2)0_4C(O)N(ORo)R°; -(CH2)0_4C(O)C(O)Ro; -(CH2)0.4C(O)CH2C(O)R°;
-(CH2)o-4C(NOR°)R°; -(CH2)0-4SSRo; -(CH2)0-4S(O)2R°; -(CH2)0-4S(O)2OR°;
-(CH2)0-4OS(O)2R°; -(CH2)0-4S(O)2NR°2; -(CH2)0-4S(O)R°;
-(CH2)o_4N(R°)S(0)2NR°2; -(CH2)0-4N(R°)S(O)2R°; -(CH2)0-4N(ORo)R°;
-(CH2)o-4C(NH)NR°2; -(CH2)0.4P(O)2R°; -(CH2)0-4P(O)Ro 2; -(CH2)0-4OP(O)R°2;
-(CH2)0-4OP(O)(OR°)2; -(CH2)0-4ON(R°)2; and -(CH2)0.4C(O)O-N(R°)2, wherein:
each R° is independently hydrogen, Ci_6 aliphatic, -CH2-carbocyclyl, -CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i-carbocyclyl, -0(CH2)o-i-aryl, -0(CH2)o-i-heterocyclyl, -O(CH2)0-1-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Ci-C3 alkyl, -NH2, -N02, -NH(unsubstituted C!-C3 alkyl), -N(unsubstituted C C3 alkyl)2, -O-C 1-C3 alkyl, -C(0)OH, -C(0)0-(unsubstituted C1-C3 alkyl),
-C(0)-(unsubstituted Ci-C3 alkyl), -0-(unsubstituted C1-C3 alkyl), and
-S-(unsubstituted C1-C3 alkyl);
R2 is halo, cyano, -CF3 or -CHF2;
Ra is selected from -CN, -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(S)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, -C(S)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-C(S)-R4,
-C(S)-N(R7)-N(R7)-C(S)-R4, -C(0)-N(R7)-N(R7)-S(0)i-2-R4, and
-C(S)-N(R7)-N(R7)-S(0)i-2-R4, wherein: R3 is selected from hydrogen, Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl),
-Ci-C6 alkyl, -(C0-C4 alkylene)-carbocyclyl, -(C0-C4 alkylene)-heterocyclyl,
-(C0-C4 alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or Rs and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
each R7 is independently hydrogen or C1-C4 alkyl; and
n is 0, 1, 2, 3, 4 or 5; wherein:
unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
the compound is not (E)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4- triazol-l-yl)-3-bromo-l-(3,3-difluoroazetidin-l-yl)prop-2-en-l-one.
28. The compound of claim 27, wherein Ra is selected from -C(0)-0-R3,
-C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, and -C(0)-N(R7)-N(R7)-S(0)i-2-R4.
29. The compound of claim 27, wherein Ra is selected from -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6) and
-C(S)-N(R7)-N(R5)(R6).
30. The compound of claim 29, wherein Ra is selected from -C(0)-0-R3,
-C(0)-N(R5)(R6), and -C(0)-N(R7)-N(R5)(R6).
31. The compound of any one of claims 27-30, wherein R2 is bromo or chloro.
32. The compound of any one of claims 27-31, wherein X is -C(H)-
33. The compound of any one or claims 27-32, wherein X is -N-.
34. The compound of any one of claims 27-33, wherein n is 0, 1 or 2.
35. The compound of any one of claims 27-34, wherein each R1 is independently selected from -CF3, -CN, halo, - OH, C1-C3 alkyl, C3-C6 cycloalkyl, C3-C]2 heterocycloalkyl, halo-Ci-C3 alkyl, -NH2, -N02, -NH(C,-C3 alkyl), -N(C,-C3 alkyl)(CrC3 alkyl), -C(0)OH, -C(0)0-(Ci-C6 alkyl), -C(0)-(d-C3 alkyl), -0-(CrC3 alkyl), -0-(d-C3 haloalkyl), and -S-( C]-C3 alkyl), or is absent.
36. The compound of any one of claims 27-35, wherein each R1 is independently selected from halo, -C1-C4 alkyl, -C]-C4 haloalkyl and -0-Ci-C4 alkyl, or is absent.
37. The compound of any one o Structural Formula Ila:
Figure imgf000136_0001
or a pharmaceutically acceptable salt thereof, wherein:
Rla and Rlb are each independently selected from halo; haloalkyl; -(CH2)i-4R°; -(CH2)0-4OR°; -0-(CH2)o- C(0)OR°; -(CH2)0- CH(OR°)2; -(CH2)0-4SR°;
-(CH2)o-4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°; -(CH2)0.4-heterocyclyl, which may be substituted with R°; -(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°;
-CH=CH-heterocyclyl, which may be substituted with R°; -CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3; -(CH2)0-4N(R°)2; -(CH2)0-4N(R°)C(O)R°;
-(CH2)o-4N(R0)C(S)R°; -(CH2)0-4N(Ro)C(O)NR°2; -(CH2)0-4N(R°)C(S)NR°2;
-(CH2)o-4N(R0)C(0)OR°; -(CH2)0-4N(Ro)N(Ro)C(O)R°;
-(CH2)0-4N(Ro)N(Ro)C(O)NR°2; -(CH2)0-4N(Ro)N(Ro)C(O)OR°; -(CH2)0- C(O)R°; -(CH2)o-4C(S)R0; -(CH2)o-4C(0)OR°; -(CH2)0-4C(O)SR°; -(CH2)0-4OC(O)R°;
-(CH2)0-4OC(0)(CH2)o-4SR°, -(CH2)0.4SC(S)SR°; -(CH2)0.4SC(O)Ro;
-(CH2)o-4C(0)NR°2; -(CH2)0-4C(S)NR°2; -(CH2)0.4C(S)SR°; -(CH2)0-4OC(O)NR°2; -(CH2)o-4C(0)N(OR0)R0; -(CH2)o-4C(0)C(0)R°; -(CH2)o-4C(0)CH2C(0)R°;
-(CH2)o.4C(NOR0)R0; -(CH2)0.4SSR°; -(CH2)0_4S(O)2R°; -(CH2)0. S(O)2OR°;
-(CH2)o-4OS(0)2R°; -(CH2)0- S(O)2NR°2; -(CH2)0-4S(O)R°;
-(CH2)o-4N(R0)S(0)2NR°2; -(CH2)0_4N(Ro)S(O)2R°; -(CH2)0-4N(OR°)R°;
-(CH2)0-4C(NH)NR°2; -(CH2)0-4P(O)2R°; -(CH2)0-4P(O)R°2; -(CH2)0-4OP(O)R°2; -(CH2)o.4OP(0)(OR°)2; -(CH2)0-4ON(R°)2; and -(CH2)0.4C(O)O-N(Ro)2, wherein: each R° is independently hydrogen, Ci-6 aliphatic, -CH2-carbocyclyl, -CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i-carbocyclyl,
-O(CH2)0-i-aryl, -0(CH2)o-i -heterocyclyl, -0(CH2)o-i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted C1-C3 alkyl, halo-Ci-C3 alkyl, -NH2, -N02, -NH(unsubstituted C1-C3 alkyl), -N(unsubstituted C1-C3 alkyl)2, -O-C1-C3 alkyl, -C(0)OH,
-C(0)0-(unsubstituted CrC3 alkyl), -C(0)-(unsubstituted C1-C3 alkyl),
-0-(unsubstituted C1-C3 alkyl), and -S-(unsubstituted C1-C3 alkyl); and
m is 0 or 1.
38. The compound of claim 37, wherein Rla is -Cj-C4 haloalkyl.
39. The compound of claim 37 or 38, wherein RIb is -Ci-C haloalkyl.
40. The compound of claim 37, wherein Rla is -CF3 and Rlb is -CF3.
41. The compound of any one of claims 27-40, wherein: Ra is -C(0)-0-R3, and R3 is selected from optionally substituted Q-Q alkyl and C2- C4 alkenyl; or
Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or
Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i.2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), - - alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alky lene)-hetero aryl .
42. The compound of claim 41, wherein:
Ra is -C(0)-0-R3, and R3 is selected from optionally substituted Ci-C4 alkyl; or Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C3- C7 heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted C5-C6 heteroaryl; or Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(C1-C4 alkyl)(C3-C6 cycloalkyl), -Q-Q alkyl, -(C0-C4 alkylene)-(C3-C7)heterocyclyl and -(C0-C4 alkylene)-(C5-C6)heteroaryl.
The compound of claim 42, wherein Ra is selected from 3,3-difluoroazetidm-l- ylcarbonyl, N'-(pyrazin-2-yl)-hydrazinylcarbonyl, and -C(0)OCH(CH3)2.
44. A compound represented by Structural Formula lib:
Figure imgf000138_0001
or a pharmaceutically acceptable salt thereof, wherein: R2 is halo, cyano, -CF3 or -CHF2;
Ra is selected from -C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6),
-C(0)-N(R7)-N(R7)-C(0)-R4, and -C(0)-N(R7)-N(R7)-S(0)1-2-R4, wherein: R is selected from hydrogen, C1 -C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl,
carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl),
-Ci-C6 alkyl, -(C0-C4 alkyl ene)-carbocyclyl, -(C0-C4 alkyl ene)-heterocyclyl,
-(C0-C4 alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl; and
each R7 is independently hydrogen or C[-C4 alkyl; wherein:
unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
the compound is not (E)-3-(3-(3,5-bis(trifluoromethyl)phenyl)-lH-l,2,4- triazol- 1 -yl)-3-bromo- 1 -(3,3-difluoroazetidin- 1 -yl)prop-2-en- 1 -one.
45. The compound of claim 44, wherein R2 is bromo or chloro.
46. The compound of claim 44 or claim 45, wherein:
Ra is -C(0)-0-R3, and R3 is selected from optionally substituted C1-C4 alkyl and C2- C4 alkenyl; or
Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or
Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(CrC4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl.
47. The compound of claim 46, wherein:
Ra is -C(0)-0-R3, and R3 is selected from optionally substituted C1-C4 alkyl; or Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C3-
C7 heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted C5-C6 heteroaryl; or Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(C]-C4 alkyl)(C3-C6 cycloalkyl), -Cj-C6 alkyl, -(C0-C4 alkylene)-(C3-C7)heterocyclyl and -(C0-C4 alkylene)-(C5-C6)heteroaryl,
48. The compound of claim 47, wherein Ra is selected from 3,3-difluoroazetidin-l- ylcarbonyl, N'-(pyrazin-2-yl)-hydrazinylcarbonyl, and -C(0)OCH(CH3)2.
49. A compound represented by any one of the following structural formulas:
Figure imgf000140_0001
or a pharmaceutically acceptable salt of any of the foreg A compound represented by Structural Formula III:
Figure imgf000141_0001
III),
or a pharmaceutically acceptable salt thereof, wherein:
X is -N- or -C(H)-;
each R1 is independently selected from halo; haloalkyl; -(CH2)1-4R°;
-(CH2)o-4OR°; -0-(CH2)o-4C(0)OR°; -(CH2)0-4CH(OR°)2; -(CH2)0-4SR°;
-(CH2)o-4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°; -(CH2)o-4-heterocyclyl, which may be substituted with R°; -(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°; -CH=CH-heterocyclyl, which may be substituted with R°;
-CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3;
-(CH2)o_4N(R°)2; -(CH2)0-4N(R°)C(O)R°; -(CH2)0-4N(R°)C(S)R°;
-(CH2)o.4N(R°)C(0)NR°2; -(CH2)0-4N(Ro)C(S)NR°2; -(CH2)0-4N(R°)C(O)OR°; -(CH2)0.4N(Ro)N(R°)C(O)R°; -(CH2)0-4N(R°)N(Ro)C(O)NRo 2;
-(CH2)0-4N(R°)N(R°)C(O)OR°; -(CH2)0-4C(O)Ro; -(CH2)0.4C(S)Ro;
-(CH2)0-4C(O)OR°; -(CH2)0-4C(O)SR°; -(CH2)0-4OC(O)R°;
-(CH2)0-4OC(0)(CH2)o-4SR°, -(CH2)0-4SC(S)SR°; -(CH2)0-4SC(O)R°;
-(CH2)o.4C(0)NR°2; -(CH2)0-4C(S)NR°2; -(CH2)0-4C(S)SR°; -(CH2)0-4OC(O)NR°2; -(CH2)o-4C(0)N(OR0)R0; -(CH2)o-4C(0)C(0)R°; -(CH2)o-4C(0)CH2C(0)R0;
-(CH2)0-4C(NOR°)R°; -(CH2)0-4SSR°; -(CH2)0-4S(O)2R°; -(CH2)0-4S(O)2OR°; -(CH2)0-4OS(O)2R°; -(CH2)0.4S(O)2NR°2; -(CH2)0-4S(O)R°;
-(CH2)o-4N(R°)S(0)2NR°2; -(CH2)0-4N(R°)S(O)2R°; -(CH2)0.4N(OR°)R°;
-(CH2)0-4C(NH)NR°2; -(CH2)0-4P(O)2Ro; -(CH2)0-4P(O)R°2; -(CH2)0-4OP(O)R°2; -(CH2)0-4OP(O)(OR°)2; -(CH2)0-4ON(R°)2; and -(CH2)0-4C(O)O-N(R°)2, wherein: each R° is independently hydrogen, C1-6 aliphatic, -CH2-carbocyclyl, -CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i-carbocyclyl, -O(CH2)0-i-aryl, -0(CH2)o-i-heterocyclyl, -0(CH2)o-i-heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted CrC3 alkyl, halo-Ci-C3 alkyl, -NH2, -N02,
-NH(unsubstituted CrC3 alkyl), -N(unsubstituted C C3 alkyl)2, -0-C C3 alkyl, -C(0)OH, -C(0)0-(unsubstituted C1 -C3 alkyl): -C(0)-(unsubstituted C1-C3 alkyl), -0-(unsubstituted Ci-C3 alkyl), and -S-(unsubstituted Ci-C3 alkyl);
R2 is halo, -CF3, -CHF2, or -CN;
Ra is selected from -CN, -C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(S)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, -C(S)-N(R7)-N(R7)-C(0)-R4, -C(0)-N(R7)-N(R7)-C(S)-R4,
-C(S)-N(R7)-N(R7)-C(S)-R4, -C(0)-N(R7)-N(R7)-S(0)i-2-R4, and
-C(S)-N(R7)-N(R7)-S(0)1-2-R4, wherein:
R3 is selected from hydrogen, C 1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl,
carbocyclyl, aryl, heterocyclyl and heteroaryl;
R4 is selected from -N(H)(C3-C6 cycloalkyl), -N(C,-C4 alkyl)(C3-C6 cycloalkyl),
-CrC6 alkyl, -(C0-C4 alkyl ene)-carbocyclyl, -(C0-C4 alkyl ene)-heterocyclyl,
-(Co-C4 alkylene)-aryl, and -(C0-C4 alkylene)-heteroaryl;
R5 and R6 are each independently selected from hydrogen, Cj-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl; or R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a heterocyclyl or heteroaryl;
each R7 is independently hydrogen or Cj-C4 alkyl; and
n is 0, 1, 2, 3, 4 or 5; wherein: unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted; and
the compound is not (E)-isopropyl 2-fluoro-3-(3-(3-methoxy-5- (trifluoromethyl)phenyl)- 1 H- 1 ,2,4-triazol- 1 -yl)acrylate.
51. The compound of claim 50, wherein R2 is halo, -CF3 or -CHF2.
52. The compound of claim 50 or claim 51 , wherein R2 is halo.
53. The compound of claim 52, wherein R2 is bromo or chloro.
54. The compound of claim 50, wherein R2 is bromo, chloro, -CF3 or -CHF2.
55. The compound of any one of claims 50-54, wherein Ra is selected from -CN,
-C(0)-0-R3, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, and -C(0)-N(R7)-N(R7)-S(0)i-2-R4.
56. The compound of any one of claims 50-54, wherein Ra is selected from -CN,
-C(0)-0-R3, -C(S)-0-R3, -C(0)-N(R5)(R6), -C(S)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), and -C(S)-N(R7)-N(R5)(R6).
57. The compound of claim 56, wherein Ra is selected from is selected from -CN,
-C(0)-0-R3, -C(0)-N(R5)(R6), and -C(0)-N(R7)-N(R5)(R6).
58. The compound of claim 57, wherein Ra is selected from 3,3-difluoroazetidin-l- ylcarbonyl, N'-(pyrazin-2-yl)-hydrazinylcarbonyl, -C(0)OCH(CH3)2, -C(0)NH2, -COOH, and -CN.
59. The compound of any one of claims 50-54, wherein Ra is selected from -C(0)OH, -C(0)-N(R5)(R6), -C(0)-N(R7)-N(R5)(R6), -C(0)-N(R7)-N(R7)-C(0)-R4, and
-C(0)-N(R7)-N(R7)-S(0)i-2-R4.
60. The compound of any one of claims 50-59, wherein:
Ra is -CN; or
Ra is -C(0)-0-R3, and R3 is selected from hydrogen and optionally substituted C1-C4 alkyl or C2-C4 alkenyl; or
Ra is -C(0)NH2; or
Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted heteroaryl; or
Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(Ci-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C alkyl, -(C0-C4 alkylene)-heterocyclyl and -(C0-C4 alkylene)-heteroaryl.
61. The compound of claim 60, wherein:
Ra is -C(0)-0-R3, and R3 is selected from hydrogen and optionally substituted -C4 alkyl; or
Ra is -C(0)NH2; or
Ra is -C(0)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated C3- C7 heterocyclyl; or
Ra is -C(0)-NH-NH(R6), and R6 is an optionally substituted C5-C6 heteroaryl; or Ra is -C(0)-NH-NH-C(0)-R4 or -C(0)-NH-NH-S(0)i-2-R4, and R4 is selected from optionally substituted -N(H)(C3-C6 cycloalkyl), -N(C1-C4 alkyl)(C3-C6 cycloalkyl), -Ci-C6 alkyl, -(C0-C4 alkylene)-(C3-C7)heterocyclyl and -(C0-C4 alkylene)-(C5-C6)heteroaryl.
62. The compound of any one of claims 50-61 , wherein X is -N-
63. The compound of any one of claim 50-61 , wherein X is -CH-
64. The compound of any one of claims 50-63, wherein n is 0, 1 or 2.
65. The compound of any one of claims 50-64, wherein each R1 is independently selected from -CF3, -CN, halo, - OH, C]-C3 alkyl, C3-C6 cycloalkyl, C3-Ci2 heterocycloalkyl, halo-C-C3 alkyl, -NH2, -N02, -NH(Ci-C3 alkyl), -N(Ci-C3 alkyl)(Ci-C3 alkyl), -C(0)OH, -C(0)0-(C,-C6 alkyl), -C(0)-(CrC3 alkyl), -0-(d-C3 alkyl), -0-(d-C3 haloalkyl), and -S-( Ci-C3 alkyl), or is absent.
66. The compound of any one of claims 50-64, wherein each R1 is independently selected from halo, -Q-C4 alkyl, -C1-C4 haloalkyl and -O-C1-C4 alkyl, or is absent.
67. The compound of any one of claims 50-63, represented by Structural Formula Ilia:
Figure imgf000145_0001
or a pharmaceutically acceptable salt thereof, wherein:
Rla and Rlb are each independently selected from halo; haloalkyl; -(CH2)|.4R°; -(CH2)o-4OR0; -0-(CH2)o-4C(0)OR°; -(CH2)0-4CH(ORo)2; -(CH2)0-4SRo;
-(CH2)o-4-carbocyclyl, which may be substituted with R°; -(CH2)o-4-aryl, which may be substituted with R°; -(CH2)o-4-heterocyclyl, which may be substituted with R°; -(CH2)o-4-heteroaryl, which may be substituted with R°; -CH=CH-carbocyclyl, which may be substituted with R°; -CH=CH-aryl, which may be substituted with R°;
-CH=CH-heterocyclyl, which may be substituted with R°; -CH=CH-heteroaryl, which may be substituted with R°; -N02; -CN; -N3; -(CH2)o-4N(R°)2; -(CH2)0-4N(Ro)C(O)Ro; -(CH2)o.4N(R0)C(S)R0 ; -(CH2)o-4N(R0)C(0)NR0 2; -(CH2)o-4N(R0)C(S)NR0 2;
-(CH2)o-4N(R0)C(0)OR°; -(CH2)o-4N(R0)N(R0)C(0)R0;
-(CH2)o-4N(R0)N(R0)C(0)NR0 2; -(CH2)o-4N(R0)N(R0)C(0)OR0; -(CH2)o-4C(0)R0 ; -(CH2)o-4C(S)R°; -(CH2)o-4C(0)OR°; -(CH2)0-4C(O)SRo; -(CH2)0-4OC(O)Ro;
-(CH2)0-4OC(0)(CH2)o-4SR0, -(CH2)0-4SC(S)SR°; -(CH2)0-4SC(O)Ro; -(CH2)o-4C(0)NR0 2; -(CH2)0-4C(S)NR°2; -(CH2)0-4C(S)SR°; -(CH2)0-4OC(O)NR°2; -(CH2)o.4C(0)N(OR0)R0; -(CH2)o.4C(0)C(0)R0; -(CH2)o-4C(0)CH2C(0)R°;
-(CH2)o.4C(NOR°)R°; -(CH2)0-4SSR°; -(CH2)0_4S(O)2R°; -(CH2)0-4S(O)2OR°;
-(CH2)0-4OS(O)2R°; -(CH2)0-4S(O)2NRo 2; -(CH2)0-4S(O)R°;
-(CH2)o_4N(R°)S(0)2NR°2; -(CH2)0-4N(Ro)S(O)2Ro; -(CH2)0-4N(ORo)R°;
-(CH2)o.4C(NH)NR°2; -(CH2)0-4P(O)2R°; -(CH2)0-4P(O)R°2; -(CH2)0-4OP(O)R°2; -(CH2)0-4OP(O)(OR°)2; -(CH2)0.4ON(Ro)2; and -(CH2)0-4C(O)O-N(R°)2, wherein: each R° is independently hydrogen, C1-6 aliphatic, -CH2-carbocyclyl, -CH2-aryl, -CH2-heterocyclyl, -CH2-heteroaryl, -0(CH2)o-i-carbocyclyl,
-O(CH2)0-i-aryl, -O(CH2)0-] -heterocyclyl, -O(CH2)0-i -heteroaryl, carbocyclyl, aryl, heterocyclyl or heteroaryl, or two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered carbocyclyl, aryl, heterocyclyl or heteroaryl; and
each R° and each ring formed from two independent occurrences of R°, taken together with their intervening atom(s), are optionally and independently substituted with one or more substituents selected from the group consisting of halo, CN, OH, unsubstituted Cj-C3 alkyl, halo-C]-C3 alkyl, -NH2, -N02, -NH(unsubstituted d-C3 alkyl), -N(unsubstituted C C3 alkyl)2, -0-Ci-C3 alkyl, -C(0)OH,
-C(0)0-(unsubstituted C 1 -C3 alkyl), -C(0)-(unsubstituted Ci-C3 alkyl),
-0-(unsubstituted Ci-C3 alkyl), and -S-(unsubstituted Ci-C3 alkyl); and
m is 0 or 1.
68. The compound of claim 67, wherein Rla is -Cj-C4 haloalkyl.
69. The compound of claim 67 or 68, wherein Rlb is -Ci-C4 haloalkyl.
70. The compound of claim 67, wherein Rla is -CF3 and Rlb is -CF3.
71. The compound of any one of claims 50-61, represented by Structural Formula Illb:
Figure imgf000147_0001
harmaceutically acceptable salt thereof.
Figure imgf000147_0002
or a pharmaceutically acceptable salt of any of the foregoing.
73. The compound of any one of claims 27-72, wherein the exocyclic double bond is in a trans configuration.
74. The compound of any one of claims 27-72, wherein the exocyclic double bond is in a cis configuration.
75. A pharmaceutically acceptable composition comprising a compound of any one of claims 1 -74, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
76. A method for treating a disorder associated with CRM1 activity, the method
comprising administering to a subject in need thereof a therapeutically effective amount of a compound of any one of claims 1-74, or a pharmaceutically acceptable salt thereof, or a composition of claim 75.
77. The method according to claim 76, wherein the disorder is selected from a
proliferative disorder, cancer, an inflammatory disorder, an autoimmune disorder, a viral infection, an ophthalmological disorder, a neurodegenerative disorder, a disorder of abnormal tissue growth, a disorder related to food intake, an allergy, and a respiratory disorder.
78. The method according to claim 77, wherein the disorder is cancer.
79. A method for promoting wound healing in a subject in need thereof, comprising
administering to the subject in need thereof a therapeutically effective amount of a compound of any one of claims 1 -74, or a pharmaceutically acceptable salt thereof, or a composition of claim 75.
PCT/US2014/043484 2013-06-21 2014-06-20 Nuclear transport modulators and uses thereof WO2014205393A1 (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201361838153P 2013-06-21 2013-06-21
US201361838135P 2013-06-21 2013-06-21
US201361838129P 2013-06-21 2013-06-21
US61/838,129 2013-06-21
US61/838,135 2013-06-21
US61/838,153 2013-06-21
US201361842475P 2013-07-03 2013-07-03
US61/842,475 2013-07-03

Publications (1)

Publication Number Publication Date
WO2014205393A1 true WO2014205393A1 (en) 2014-12-24

Family

ID=51177202

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/043484 WO2014205393A1 (en) 2013-06-21 2014-06-20 Nuclear transport modulators and uses thereof

Country Status (1)

Country Link
WO (1) WO2014205393A1 (en)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9206158B2 (en) 2011-07-29 2015-12-08 Karyopharm Therapeutics Inc. Hydrazide containing nuclear transport modulators and uses thereof
US9266843B2 (en) 2012-05-09 2016-02-23 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US9303000B2 (en) 2011-01-17 2016-04-05 Karyopharm Therapeutics Inc. Olefin containing nuclear transport modulators and uses thereof
US9428490B2 (en) 2011-07-29 2016-08-30 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US9550757B2 (en) 2010-03-05 2017-01-24 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
CN106831731A (en) * 2017-01-17 2017-06-13 广州市闻皓生物科技有限公司 A kind of novel method for synthesizing of Selinexor bulk drugs
US9738624B2 (en) 2013-06-21 2017-08-22 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
WO2018050801A1 (en) * 2016-09-16 2018-03-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of systemic mastocytosis
US10202366B2 (en) 2013-03-15 2019-02-12 Karyopharm Therapeutics Inc. Methods of promoting wound healing using CRM1 inhibitors
WO2019232724A1 (en) * 2018-06-06 2019-12-12 Xw Laboratories, Inc. Compounds as nuclear transport modulators and uses thereof
US10519139B2 (en) 2014-08-15 2019-12-31 Karyopharm Therapeutics Inc. Polymorphs of Selinexor
US10526295B2 (en) 2015-12-31 2020-01-07 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US10618886B1 (en) 2018-09-30 2020-04-14 Xw Laboratories Inc. Compounds as neuronal histamine receptor-3 antagonists and uses thereof
US10640476B2 (en) 2017-03-30 2020-05-05 Xw Laboratories Inc. Bicyclic heteroaryl derivatives and preparation and uses thereof
US10683262B2 (en) 2018-01-10 2020-06-16 Xw Laboratories Inc. Ketamine derivatives and compositions thereof
US10709706B2 (en) 2015-12-31 2020-07-14 Karopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
WO2020173417A1 (en) * 2019-02-26 2020-09-03 微境生物医药科技(上海)有限公司 Acryloyl-containing nuclear transport regulator and uses thereof
US10774031B2 (en) 2015-09-23 2020-09-15 Xw Laboratories Inc. Prodrugs of gamma-hydroxybutyric acid, compositions and uses thereof
WO2020223678A1 (en) * 2019-05-01 2020-11-05 Karyopharm Therapeutics Inc. Process for preparing xpo1 inhibitors and intermediates for use in the preparation of xp01 inhibitors
US11279669B2 (en) 2019-12-20 2022-03-22 XWPharma Ltd. Methods of synthesizing 4-valyloxybutyric acid
US11304906B2 (en) 2020-06-18 2022-04-19 XWPharma Ltd. Controlled release granulations of water-soluble active pharmaceutical ingredients
US11357734B2 (en) 2020-06-18 2022-06-14 XWPharma Ltd. Pharmaceutical granulations of water-soluble active pharmaceutical ingredients
CN114634456A (en) * 2022-03-22 2022-06-17 赣南师范大学 5-nitroimino-4H-1, 2, 4-triazole compound and preparation method and application thereof
WO2022143779A1 (en) * 2020-12-29 2022-07-07 南京明德新药研发有限公司 Alkenyl-containing compound and application thereof
US11395801B2 (en) 2020-10-05 2022-07-26 XWPharma Ltd. Modified release compositions of a gamma-hydroxybutyric acid derivative
US11510892B2 (en) 2021-03-19 2022-11-29 XWPharma Ltd. Pharmacokinetics of combined release formulations of a γ-hydroxybutyric acid derivative
US11602530B2 (en) 2016-11-28 2023-03-14 Biogen Ma Inc. CRM1 inhibitors for treating epilepsy
US11690811B2 (en) 2021-08-13 2023-07-04 XWPharma Ltd. Pharmaceutical compositions and oral dosage forms of ketamine derivatives
CN116514773A (en) * 2023-04-24 2023-08-01 重庆汉佩生物科技有限公司 Verdinexor (KPT-335) and synthesis method of hydrochloride thereof
US11896573B2 (en) 2020-07-24 2024-02-13 XWPharma Ltd. Pharmaceutical compositions and pharmacokinetics of a gamma-hydroxybutyric acid derivative
WO2024053502A1 (en) * 2022-09-09 2024-03-14 ダイキン工業株式会社 METHOD FOR PRODUCING α-CHLOROACRYLIC ACID ESTER

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011109799A1 (en) * 2010-03-05 2011-09-09 Karyopharm Therapeutics, Inc. Nuclear transport modulatiors and uses thereof
WO2012099807A1 (en) * 2011-01-17 2012-07-26 Karyopharm Therapeutics, Inc. Olefin containing nuclear transport modulators and uses thereof
WO2013019548A1 (en) * 2011-07-29 2013-02-07 Karyopharm Therapeutics, Inc. Hydrazide containing nuclear transport modulators and uses thereof
WO2013019561A1 (en) * 2011-07-29 2013-02-07 Karyopharm Therapeutics, Inc. Nuclear transport modulators and uses thereof
WO2013170068A2 (en) * 2012-05-09 2013-11-14 Karyopharm Therapeutics, Inc. Nuclear transport modulators and uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011109799A1 (en) * 2010-03-05 2011-09-09 Karyopharm Therapeutics, Inc. Nuclear transport modulatiors and uses thereof
WO2012099807A1 (en) * 2011-01-17 2012-07-26 Karyopharm Therapeutics, Inc. Olefin containing nuclear transport modulators and uses thereof
WO2013019548A1 (en) * 2011-07-29 2013-02-07 Karyopharm Therapeutics, Inc. Hydrazide containing nuclear transport modulators and uses thereof
WO2013019561A1 (en) * 2011-07-29 2013-02-07 Karyopharm Therapeutics, Inc. Nuclear transport modulators and uses thereof
WO2013170068A2 (en) * 2012-05-09 2013-11-14 Karyopharm Therapeutics, Inc. Nuclear transport modulators and uses thereof

Non-Patent Citations (47)

* Cited by examiner, † Cited by third party
Title
"Advanced Organic Chemistry", 1992, MCGRAW HILL
"Cancer Medicine", July 2000, BC DECKER
"Handbook of Chemistry and Physics"
"March's Advanced Organic Chemistry", 2001, JOHN WILEY & SONS
"Nomenclature of Organic Chemistry", 1979, PERGAMON PRESS
BALLANGRUD A. M. ET AL., CANCER RES., vol. 61, 2001, pages 2008 - 2014
CAI X; LIU X.: "Inhibition of Thr-55 phosphorylation restores p53 nuclear localization and sensitizes cancer cells to DNA damage", PNAS, vol. 105, 2008, pages 16958 - 16963
CRONSHAW JM; MATUNIS MJ: "The nuclear pore complex: disease associations and functional correlations", TRENDS ENDOCRIN METAB, vol. 15, 2004, pages 34 - 39
DAELEMANS D; AFONINA E; NILSSON J: "A synthetic HIV-1 Rev inhibitor interfering with the CRM1-mediated nuclear export", PROC NATL ACAD SCI U S A, vol. 99, no. 22, 2002
DAVIS JR ET AL.: "Controlling protein compartmentalization to overcome disease", PHARMACEUT RES., vol. 24, 2007, pages 17 - 27
E. L. ELIEL; S. H. WILEN: "Stereo-chemistry of Carbon Compounds", 1994, JOHN WILEY & SONS, pages: 1119 - 1190
EMFIETZOGLOU D; KOSTARELOS K; SGOUROS G: "An analytical dosimetry study for the use of radionuclide-liposome conjugates in internal radiotherapy", J NUCL MED, vol. 42, 2001, pages 499 - 504
FALINI B ET AL.: "Both carboxy-terminus NES motif and mutated tryptophan(s) are crucial for aberrant nuclear export of nucleophosmin leukemic mutants in NPMc+ AML", BLOOD, vol. 107, 2006, pages 4514 - 4523
FREUNDT E; YU L; PARK E ET AL.: "Molecular determinants for subcellular localization of the severe acute respiratory syndrome coronavirus open reading frame 3b protein", J VIROL, vol. 83, no. 13, 2009, pages 6631 - 40
GHILDYAL R; HO A; DIAS M ET AL.: "The respiratory syncytial virus matrix protein possesses a Crml-mediated nuclear export mechanism", J VIROL, vol. 83, no. 11, 2009, pages 5353 - 62
GHOSH CC ET AL.: "Analysis of nucleocytoplasmic shuttling of NF kappa B proteins in human leukocytes", METHODS MOL BIOL., vol. 457, 2008, pages 279 - 92
GOLDENBER, D.M., J NUCL. MED., vol. 43, no. 5, 2002, pages 693 - 713
GUPTA N ET AL.: "Retinal tau pathology in human glaucomas", CAN J OPHTHALMOL., vol. 43, no. 1, February 2008 (2008-02-01), pages 53 - 60
HOSHINOL ET AL.: "Combined effects of p53 gene therapy and leptomycin B in human esophageal squamous cell carcinoma", ONCOLOGY, vol. 75, 2008, pages 113 - 119
J. PADAWER ET AL.: "Combined Treatment with Radioestradiol lucanthone in Mouse C3HBA Mammary Adenocarcinoma and with Estradiol lucanthone in an Estrogen Bioassay", INT. J. RADIAT. ONCOL. BIOL. PHYS., vol. 7, 1981, pages 47 - 57
J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
LAIN S ET AL.: "Accumulating active p53 in the nucleus by inhibition of nuclear export: a novel strategy to promote the p53 tumor suppressor function", EXP CELL RES., vol. 253, 1999, pages 315
LAIN S ET AL.: "An inhibitor of nuclear export activates the p53 response and induces the localization of HDM2 and p53 to UIA-positive nuclear bodies associated with the PODs", EXP CELL RES., vol. 248, 1999, pages 457 - 472
MULLER PA ET AL.: "Nuclear-cytosolic transport of COMMD regulates NF-kappaB and HIF-1 activity", TRAFFIC, vol. 10, no. 5, 2009, pages 514 - 27
MUTKA S: "Nuclear Export Inhibitors (NEIs) as novel cancer therapies AACR Annual Meeting", POSTER, 2007, pages 5609
MUTKA S; YANG W; DONG S ET AL.: "Identification of nuclear export inhibitors with potent anticancer activity in vivo", CANCER RES., vol. 69, 2009, pages 510 - 7
NAKAHARA J ET AL.: "Abnormal expression ofTIP30 and arrested nucleocytoplasmic transport within oligodendrocyte precursor cells in multiple sclerosis", J CLIN INVEST., vol. 119, 2009, pages 169 - 181
NOSKE A ET AL.: "Expression of the nuclear export protein chromosomal region maintenance/exportin 1/Xpol 1 is a prognostic factor in human ovarian cancer", CANCER, vol. 112, 2008, pages 1733 - 1743
POLLARD V; MALIM M: "The HIV-1 Rev protein", ANNU REV MICROBIOL, vol. 52, 1998, pages 491 - 532
R. C. LAROCK: "Comprehensive Organic Transformations - A Guide to Functional Group Preparations", 1989, VHC PUBLISHERS, INC.
RAWLINSON S; PRYOR M; WRIGHT P; JANS D: "CRM1-mediated nuclear export of dengue virus RNA polymerase NS5 modulates interleukin-8 induction and virus production", J BIOL CHEM, vol. 284, no. 23, 2009, pages 15589 - 97
ROBERT T. BUCKLER ET AL: "Synthesis and antiinflammatory activity of some 1,2,3- and 1,2,4-triazolepropionic acids", JOURNAL OF MEDICINAL CHEMISTRY, vol. 21, no. 12, 1 December 1978 (1978-12-01), pages 1254 - 1260, XP055024397, ISSN: 0022-2623, DOI: 10.1021/jm00210a015 *
SANCHEZ V; MAHR J; ORAZIO N ET AL.: "Nuclear export of the human cytomegalovirus tegument protein pp65 requires cyclin-dependent kinase activity and the Crml exporter", J VIROL, vol. 81, no. 21, 2007, pages 11730 - 6
SMITH: "Organic Synthesis", 1994, MCGRAW HILL
SOROKIN AV ET AL.: "Nucleocytoplasmic transport of proteins", BIOCHEMISTRY, vol. 72, 2007, pages 1439 - 1457
STEVEN A. LEIBEL ET AL.: "Textbook of Radiation Oncology", 1998, W. B. SAUNDERS COMPANY
T.W. GREEN; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1999, WILEY-INTERSCIENCE
TERRY LJ ET AL.: "Crossing the nuclear envelope: hierarchical regulation of nucleocytoplasmic transport", SCIENCE, vol. 318, 2007, pages 1412 - 1416
THOMAS SORRELL: "Organic Chemistry", 1999, UNIVERSITY SCIENCE BOOKS
VAN DER WATT PJ ET AL.: "The Karyopherin proteins, Crm1 and Karyopherin betal, are overexpressed in cervical cancer and are critical for cancer cell survival and proliferation", INT J CANC., vol. 124, 2008, pages 1829 - 1840
VAN NECK T ET AL: "Inhibition of the CRM1-mediated nucleocytoplasmic transport by N-azolylacrylates: Structure-activity relationship and mechanism of action", BIOORGANIC & MEDICINAL CHEMISTRY, PERGAMON, GB, vol. 16, no. 21, 1 November 2008 (2008-11-01), pages 9487 - 9497, XP025545774, ISSN: 0968-0896, [retrieved on 20080920], DOI: 10.1016/J.BMC.2008.09.051 *
WALSH MD ET AL.: "Exportin 1 inhibition attenuates nuclear factor-kappaB-dependent gene expression", SHOCK, vol. 29, 2008, pages 160 - 166
WEINER RE; THAKUR ML: "Radiolabeled peptides in the diagnosis and therapy of oncological diseases", APPL RADIAT ISOT, vol. 57, no. 5, November 2002 (2002-11-01), pages 749 - 63
WILLIAMS P; VERHAGEN J; ELLIOTT G: "Characterization of a CRMI -dependent nuclear export signal in the C terminus of herpes simplex virus type 1 tegument protein UL47", J VIROL, vol. 82, no. 21, 2008, pages 10946 - 52
YANG W: "Anti-tumor activity of novel nuclear export inhibitors (NEIs) in multiple murine leukemia models", AACR ANNUAL MEETING. POSTER, 2007, pages 5597
YAO Y ET AL.: "The expression of CRM1 is associated with prognosis in human osteosarcoma", ONCOL REP., vol. 21, 2009, pages 29 - 5
ZIMMERMAN TL ET AL.: "Nuclear export of retinoid X receptor alpha in response to interleukin-lbeta-mediated cell signaling: roles for JNK and SER260", J BIOL CHEM, vol. 281, 2006, pages 15434 - 15440

Cited By (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9550757B2 (en) 2010-03-05 2017-01-24 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US9303000B2 (en) 2011-01-17 2016-04-05 Karyopharm Therapeutics Inc. Olefin containing nuclear transport modulators and uses thereof
US10544108B2 (en) 2011-07-29 2020-01-28 Karyopharm Therapeutics Inc. Hydrazide containing nuclear transport modulators and uses thereof
US9428490B2 (en) 2011-07-29 2016-08-30 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US11034660B2 (en) 2011-07-29 2021-06-15 Karyopharm Therapeutics Inc. Hydrazide containing nuclear transport modulators and uses thereof
US11787771B2 (en) 2011-07-29 2023-10-17 Karyopharm Therapeutics Inc. Hydrazide containing nuclear transport modulators and uses thereof
US9206158B2 (en) 2011-07-29 2015-12-08 Karyopharm Therapeutics Inc. Hydrazide containing nuclear transport modulators and uses thereof
US9714226B2 (en) 2011-07-29 2017-07-25 Karyopharm Therapeutics Inc. Hydrazide containing nuclear transport modulators and uses thereof
US10173987B2 (en) 2011-07-29 2019-01-08 Karyopharm Therapeutics Inc. Hydrazide containing nuclear transport modulators and uses thereof
US11318120B2 (en) 2012-05-09 2022-05-03 Biogen Ma Inc. Nuclear transport modulators and uses thereof
US10617677B2 (en) 2012-05-09 2020-04-14 Biogen Ma Inc. Nuclear transport modulators and uses thereof
US10058535B2 (en) 2012-05-09 2018-08-28 Biogen Ma Inc. Nuclear transport modulators and uses thereof
US10925859B2 (en) 2012-05-09 2021-02-23 Biogen Ma Inc. Nuclear transport modulators and uses thereof
US10335393B2 (en) 2012-05-09 2019-07-02 Biogen Ma Inc. Nuclear transport modulators and uses thereof
US9266843B2 (en) 2012-05-09 2016-02-23 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US9861614B2 (en) 2012-05-09 2018-01-09 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US9585874B2 (en) 2012-05-09 2017-03-07 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US10202366B2 (en) 2013-03-15 2019-02-12 Karyopharm Therapeutics Inc. Methods of promoting wound healing using CRM1 inhibitors
US9738624B2 (en) 2013-06-21 2017-08-22 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US10407405B2 (en) 2013-06-21 2019-09-10 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US11124493B2 (en) 2013-06-21 2021-09-21 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US11945794B2 (en) 2013-06-21 2024-04-02 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US11753401B2 (en) 2014-08-15 2023-09-12 Karyopharm Therapeutics Inc. Polymorphs of Selinexor
US11078190B2 (en) 2014-08-15 2021-08-03 Karyopharm Therapeutics Inc. Polymorphs of selinexor
US10519139B2 (en) 2014-08-15 2019-12-31 Karyopharm Therapeutics Inc. Polymorphs of Selinexor
US11746102B2 (en) 2014-08-15 2023-09-05 Karyopharm Therapeutics Inc. Polymorphs of selinexor
US11807629B2 (en) 2014-08-15 2023-11-07 Karyopharm Therapeutics Inc. Polymorphs of Selinexor
US10941107B2 (en) 2015-09-23 2021-03-09 Xw Laboratories Inc. Prodrugs of gamma-hydroxybutyric acid, compositions and uses thereof
US10774031B2 (en) 2015-09-23 2020-09-15 Xw Laboratories Inc. Prodrugs of gamma-hydroxybutyric acid, compositions and uses thereof
US10526295B2 (en) 2015-12-31 2020-01-07 Karyopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
US10709706B2 (en) 2015-12-31 2020-07-14 Karopharm Therapeutics Inc. Nuclear transport modulators and uses thereof
WO2018050801A1 (en) * 2016-09-16 2018-03-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of systemic mastocytosis
US11065230B2 (en) 2016-09-16 2021-07-20 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods and pharmaceutical compositions for the treatment of systemic mastocytosis
US11602530B2 (en) 2016-11-28 2023-03-14 Biogen Ma Inc. CRM1 inhibitors for treating epilepsy
CN106831731B (en) * 2017-01-17 2019-11-08 广州市闻皓生物科技有限公司 A kind of synthetic method of Selinexor bulk pharmaceutical chemicals
CN106831731A (en) * 2017-01-17 2017-06-13 广州市闻皓生物科技有限公司 A kind of novel method for synthesizing of Selinexor bulk drugs
US10882832B2 (en) 2017-03-30 2021-01-05 Xw Laboratories Inc. Bicyclic heteroaryl derivatives and preparation and uses thereof
US11639337B2 (en) 2017-03-30 2023-05-02 XWPharma Ltd. Bicyclic heteroaryl derivatives and preparation and uses thereof
US11401249B2 (en) 2017-03-30 2022-08-02 XWPharma Ltd. Bicyclic heteroaryl derivatives and preparation and uses thereof
US10640476B2 (en) 2017-03-30 2020-05-05 Xw Laboratories Inc. Bicyclic heteroaryl derivatives and preparation and uses thereof
US11440874B2 (en) 2018-01-10 2022-09-13 XWPharma Ltd. Ketamine derivatives and compositions thereof
US10836714B2 (en) 2018-01-10 2020-11-17 Xw Laboratories Inc. Ketamine derivatives and compositions thereof
US10683262B2 (en) 2018-01-10 2020-06-16 Xw Laboratories Inc. Ketamine derivatives and compositions thereof
WO2019232724A1 (en) * 2018-06-06 2019-12-12 Xw Laboratories, Inc. Compounds as nuclear transport modulators and uses thereof
JP7028996B2 (en) 2018-06-06 2022-03-02 エックスダブリューファルマ リミテッド Compounds and their use as nuclear transport modulators
WO2019233447A1 (en) 2018-06-06 2019-12-12 Xw Laboratories, Inc. Compounds as nuclear transport modulators and uses thereof
JP2021525781A (en) * 2018-06-06 2021-09-27 エックスダブリューファルマ リミテッド Compounds and their use as nuclear transport modulators
JP2022078109A (en) * 2018-06-06 2022-05-24 エックスダブリューファルマ リミテッド Compounds as nuclear transport modulators and uses thereof
AU2022201688B2 (en) * 2018-06-06 2023-03-30 XWPharma Ltd. Compounds as nuclear transport modulators and uses thereof
US11767313B2 (en) 2018-06-06 2023-09-26 XWPharma Ltd. Compounds as nuclear transport modulators and uses thereof
US10618886B1 (en) 2018-09-30 2020-04-14 Xw Laboratories Inc. Compounds as neuronal histamine receptor-3 antagonists and uses thereof
US10968202B2 (en) 2018-09-30 2021-04-06 Xw Laboratories Inc. Compounds as neuronal histamine receptor-3 antagonists and uses thereof
US11420955B2 (en) 2018-09-30 2022-08-23 XWPharma Ltd. Compounds as neuronal histamine receptor-3 antagonists and uses thereof
US10730853B2 (en) 2018-09-30 2020-08-04 Xw Laboratories Inc. Compounds as neuronal histamine receptor-3 antagonists and uses thereof
WO2020173417A1 (en) * 2019-02-26 2020-09-03 微境生物医药科技(上海)有限公司 Acryloyl-containing nuclear transport regulator and uses thereof
US11286247B2 (en) 2019-02-26 2022-03-29 Wigen Biomedicine Technology (shanghai) Co., Ltd. Acryloyl-containing nuclear transport regulators and uses thereof
WO2020223678A1 (en) * 2019-05-01 2020-11-05 Karyopharm Therapeutics Inc. Process for preparing xpo1 inhibitors and intermediates for use in the preparation of xp01 inhibitors
US11279669B2 (en) 2019-12-20 2022-03-22 XWPharma Ltd. Methods of synthesizing 4-valyloxybutyric acid
US11746081B2 (en) 2019-12-20 2023-09-05 XWPharma Ltd. Methods of synthesizing 4-valyloxybutyric acid
US11357734B2 (en) 2020-06-18 2022-06-14 XWPharma Ltd. Pharmaceutical granulations of water-soluble active pharmaceutical ingredients
US11304906B2 (en) 2020-06-18 2022-04-19 XWPharma Ltd. Controlled release granulations of water-soluble active pharmaceutical ingredients
US11896573B2 (en) 2020-07-24 2024-02-13 XWPharma Ltd. Pharmaceutical compositions and pharmacokinetics of a gamma-hydroxybutyric acid derivative
US11395801B2 (en) 2020-10-05 2022-07-26 XWPharma Ltd. Modified release compositions of a gamma-hydroxybutyric acid derivative
US11925710B2 (en) 2020-10-05 2024-03-12 XWPharma Ltd. Modified release compositions of a GAMMA-hydroxybutyric acid derivative
WO2022143779A1 (en) * 2020-12-29 2022-07-07 南京明德新药研发有限公司 Alkenyl-containing compound and application thereof
US11510892B2 (en) 2021-03-19 2022-11-29 XWPharma Ltd. Pharmacokinetics of combined release formulations of a γ-hydroxybutyric acid derivative
US11944597B2 (en) 2021-03-19 2024-04-02 XWPharma Ltd. Pharmacokinetics of combined release formulations of a gamma-hydroxybutyric acid derivative
US11690811B2 (en) 2021-08-13 2023-07-04 XWPharma Ltd. Pharmaceutical compositions and oral dosage forms of ketamine derivatives
CN114634456A (en) * 2022-03-22 2022-06-17 赣南师范大学 5-nitroimino-4H-1, 2, 4-triazole compound and preparation method and application thereof
CN114634456B (en) * 2022-03-22 2024-02-20 赣南师范大学 5-nitroimino-4H-1, 2, 4-triazole compound and preparation method and application thereof
WO2024053502A1 (en) * 2022-09-09 2024-03-14 ダイキン工業株式会社 METHOD FOR PRODUCING α-CHLOROACRYLIC ACID ESTER
CN116514773A (en) * 2023-04-24 2023-08-01 重庆汉佩生物科技有限公司 Verdinexor (KPT-335) and synthesis method of hydrochloride thereof

Similar Documents

Publication Publication Date Title
US11945794B2 (en) Nuclear transport modulators and uses thereof
US11318120B2 (en) Nuclear transport modulators and uses thereof
US10526295B2 (en) Nuclear transport modulators and uses thereof
WO2014205393A1 (en) Nuclear transport modulators and uses thereof
US10709706B2 (en) Nuclear transport modulators and uses thereof
WO2014152263A1 (en) Exo olefin-containing nuclear transport modulators and uses thereof
WO2017117406A1 (en) Substituted benzofuranyl and benzoxazolyl compounds and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14738999

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14738999

Country of ref document: EP

Kind code of ref document: A1