WO2015192119A1 - Pyrimidine compounds and methods using the same - Google Patents

Pyrimidine compounds and methods using the same Download PDF

Info

Publication number
WO2015192119A1
WO2015192119A1 PCT/US2015/035735 US2015035735W WO2015192119A1 WO 2015192119 A1 WO2015192119 A1 WO 2015192119A1 US 2015035735 W US2015035735 W US 2015035735W WO 2015192119 A1 WO2015192119 A1 WO 2015192119A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
formula
methyl
combine
chloro
Prior art date
Application number
PCT/US2015/035735
Other languages
French (fr)
Inventor
Gregory D. Cuny
Marcie A. Glicksman
Kevin J. Hodgetts
Steven L. MATHIEU
Yukari Y. PERRELLA
Vincent DARMENCY
Hrvoje Lusic
Original Assignee
Yuma Therapeutics, Inc.
The Brigham And Women's Hospital, 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 Yuma Therapeutics, Inc., The Brigham And Women's Hospital, Inc. filed Critical Yuma Therapeutics, Inc.
Priority to AU2015274285A priority Critical patent/AU2015274285C1/en
Priority to JP2017518041A priority patent/JP6779867B2/en
Priority to EP15806816.3A priority patent/EP3154547B1/en
Priority to US15/317,999 priority patent/US10336768B2/en
Priority to CA2952230A priority patent/CA2952230C/en
Priority to ES15806816T priority patent/ES2954453T3/en
Priority to CN201580043036.0A priority patent/CN106687114B/en
Publication of WO2015192119A1 publication Critical patent/WO2015192119A1/en
Priority to US16/459,138 priority patent/US10961254B2/en
Priority to US17/216,076 priority patent/US20210363154A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/78Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 2
    • C07D239/84Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D251/00Heterocyclic compounds containing 1,3,5-triazine rings
    • C07D251/02Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings
    • C07D251/12Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D251/14Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hydrogen or carbon atoms directly attached to at least one ring carbon atom
    • C07D251/16Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hydrogen or carbon atoms directly attached to at least one ring carbon atom to only one ring carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D251/00Heterocyclic compounds containing 1,3,5-triazine rings
    • C07D251/02Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings
    • C07D251/12Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D251/14Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hydrogen or carbon atoms directly attached to at least one ring carbon atom
    • C07D251/22Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hydrogen or carbon atoms directly attached to at least one ring carbon atom to two ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/10Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to pyrimidine-based small molecule inhibitors of heat shock protein 90 (Hsp90) and pharmaceutical compositions thereof.
  • the invention further relates to methods of treatment of a subject having a neurodegenerative disease.
  • Hsp90 proteins are implicated in stabilizing protein conformations, maintaining the function of many cell- signaling proteins, and ATPase activity. Hsp90 activity is also required for the proper folding, stabilization, activation, and localization of oncoproteins involved in tumor progression.
  • the /V-terminus ATP binding domain is responsible for the ATPase activity of this protein: this adenine nucleotide binding pocket is highly conserved among all Hsp90 proteins from bacteria to mammals but is not present in other chaperones.
  • Hsp90 protein has emerged as an important target in cancer treatment, as many Hsp90 client proteins themselves were identified as targets for cancer therapies.
  • the exemplary Hsp90 client proteins that are associated with cancer include HER2 (breast cancer), Raf-1/mutant BRAF (melanoma), Mutant EGFR (non-small cell lung cancer, glioblastoma), c-Kit (GIST), c-Met (gastric, lung, glioblastoma), HIF-1 a (renal cancer), Zap70 (chronic lymphocytic leukemia), Bcr-Abl (chronic myelogenous leukemia), mBcr-Abl (chronic myelogenous leukemia), Flt-3 (acute myeloid leukemia), IGF-1 R/Akt (myeloma), NMP-ALK
  • Hsp90 provides a compelling target for treating different classes of tumors. Increased levels of Hsp90 have also been implicated in neurodegenerative disorders, including
  • Tauopathies are atypical neurotrophic factor, Parkinson's, and Huntington's disease, and tauopathies. Tauopathies are atypical neurotrophic factor, Parkinson's, and Huntington's disease, and tauopathies. Tauopathies are atypical neurotrophic factor, Parkinson's, and Huntington's disease, and tauopathies. Tauopathies are atypical neurotrophic factor, Parkinson's, and Huntington's disease, and tauopathies. Tauopathies are examples of tauopathies.
  • Hsp90 stabilizes p35, an activator of cdk5, leading to increased tau phosphorylation. It has also been shown that Hsp90 inhibition activates heat shock factor 1 (HSF1 ), which in turn increases the expression of Hsp70. Increased expression of Hsp70 promotes tau solubility and binding to microtubules, inhibits ⁇ peptide aggregation, and enhances ⁇ peptide degradation.
  • HSF1 heat shock factor 1
  • Hsp90 has also emerged as a target for treating viral, fungal, and bacterial infections.
  • an Hsp90 inhibitor (geldanamycin) has been shown to delay the growth of influenza virus in cell culture.
  • Other viruses that rely on Hsp90 dependent processes include those belonging to the families:
  • Herpesviridae e.g., herpes simplex virus-1 , herpes simplex virus-2, herpes herpesvirus-5, Kaposi's sarcoma-associated herpesvirus, varicella zoster virus, or Epstein-Barr virus
  • Polyomaviridae e.g., SV40
  • Poxviridae e.g., vaccinia virus
  • Reoviridae e.g., rotavirus
  • Birnaviridae e.g., infectious bursal disease virus
  • picornaviridae e.g., poliovirus, rhinovirus, or coxsackievirus
  • flaviviridae e.g., hepatitis C virus or dengue virus
  • arenaviridae e.g., lymphocytic choriomeningitis virus
  • Hepeviridae e.g., Hepatitis E virus
  • Hsp90 inhibitors have also been used in vivo for the treatment of fungal infectious diseases, e.g., treatment of Candida albicans, Aspergillus fumigates, or Pneumocystis jiroveci. Moreover, Hsp90 inhibitors are also useful in the treatment of bacterial infections, e.g., mycobacteria, anthrax, or bacterial pneumonia.
  • inhibitors of Hsp90 represent beneficial therapeutics for the treatment of disorders, e.g., cancer, neurodegenerative diseases, and infectious diseases.
  • the invention features a compound according to formula (I):
  • Z 1 is -OR 7 , -N(R 10 )R 7 , -SR 7 , or -C(R 10 )(R 11 )R 7 ;
  • each R 1 and R 2 is, independently, H or optionally substituted Ci -3 alkyl (e.g., Ci -3 acyl);
  • R 3 is H, halogen, cyano, optionally substituted Ci -6 alkyl, optionally substituted Ci -3 alkoxy, or optionally substituted amino
  • R 4 is halogen, cyano, optionally substituted C 1-6 alkyl, optionally substituted C 1-3 alkoxy, optionally substituted amino, optionally substituted C 1-6 thioalkoxy, or optionally substituted C 6- io aryl, or R 3 and R 4 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising from one to three heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur, where the nitrogen is optionally substituted with R 9 ;
  • each R 5 and R 6 is, independently, H, optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkoxy, halogen, or CN;
  • R 7 is optionally substituted C 1-3 alkyl, optionally substituted C 1-3 alkcycloalkyl, optionally substituted C 1-3 alkheterocyclyl, or optionally substituted C 1-3 alkaryl
  • R 8 is H
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- , six-, or seven- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur
  • R 9 is H, optionally substituted C 1 -3 alkyl, optionally substituted C 3 . 8 cycloalkyl, optionally substituted C 6- io aryl, optionally substituted C 2 . 9 heteroaryl, optionally substituted C 2 . 9 heterocyclyl, optionally substituted Ci -3 alkcycloalkyl, optionally substituted Ci -3 alkheterocyclyl, or optionally substituted Ci- 3 alkaryl;
  • R m is H, halogen, cyano, optionally substituted Ci -4 alkyl (e.g., Ci -4 acyl), or optionally substituted Ci -3 alkoxy.
  • R m is H (e.g., the compound of formula (I) has the following structure:
  • Z 1 is not methoxy.
  • Z 2 is not unsubstituted Ci -3 alkoxy.
  • R 3 is H
  • R 4 is optionally substituted Ci -6 thioalkoxy
  • each of R 5 and R 6 is halogen (e.g., chloro)
  • Z 1 is not cyanomethoxy or aminomethoxy.
  • Z 1 is not cyanomethoxy or aminomethoxy.
  • Z 1 is not cyanomethoxy or aminomethoxy.
  • Z 1 is not cyanomethoxy or aminomethoxy.
  • R 3 is H
  • R 4 is substituted thioalkoxy
  • Z 1 is not substituted Ci alkoxy.
  • R 3 is H
  • R 4 is substituted thioalkoxy
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is methyl, dialkylaminoethyl, optionally substituted C 1 -3 alkcycloalkyl, optionally substituted C 1 -3 alkheterocyclyl, or optionally substituted C 1-3 alkaryl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • each of R 5 and R 6 is chloro, R 3 is H, and R 4 is halogen (e.g., chloro),
  • Z 1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy.
  • Z 1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy.
  • each of R 5 and R 6 is chloro, R 3 is H, and R 4 is halogen (e.g., chloro),
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is dimethylaminoethyl, optionally substituted C 1 -3 alkcycloalkyl, or optionally substituted C 1 -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • each of R 5 and R 6 is chloro, R 3 is H, and R 4 is halogen (e.g., chloro),
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is optionally substituted d_ 3 alkcycloalkyl, or optionally substituted C 1 -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is optionally substituted d_ 3 alkcycloalkyl, or optionally substituted Ci -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
  • R 7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C 2 alkheteroaryl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , Z 2 is CR 3 , and R 3 and R 4 combine to form a group according to formula (lla),
  • R 7 is not alkyl or unsubstituted C 2 alkheteroaryl.
  • R 7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C 2 alkheterocyclyl.
  • R 7 is not alkyl or C 2 alkheterocyclyl.
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 7 is not methyl.
  • R 7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
  • R 7 is not unsubstituted alkyi, substituted alkyi, unsubstituted alkheteroaryl, or substituted alkheteroaryl.
  • R 7 is not unsubstituted alkyi, substituted alkyi, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
  • R 9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl.
  • R 9 is H or -C(0)-N(H)-(linear d_ 3 alkyi).
  • Z is -OR 7 , R 7 is not methyl or 2-chloroethyl, and R 9 is H or -C(0)-N(H)-(linear C 1 -3 alkyi).
  • R 5 when R 5 is methoxy, R 6 is methyl, Z 1 is -OR 7 , Z 2 is CR 3 , and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not methyl.
  • R 7 is not methyl.
  • R 7 is not methyl or 2-(N,N-diethylamino)ethyl.
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • Z 1 is -OR 7 , and R 7 and R 8 combine to form -CH 2 -CH 2 -.
  • R 7 when R 7 is methyl, R 5 is chloro, Z 1 is -OR 7 , Z 2 is CR 3 , and R 3 and R 4 combine to form a group according to formula (Ilia), HN ⁇ (
  • R 6 is not bromo.
  • R 7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl.
  • R 8 is not H.
  • each R 5 and R 6 is chloro
  • R 7 is methyl and R 8 is H, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 1 nor R 2 is 2-(N,N-diethylamino)ethyl.
  • each R 1 and R 2 is H.
  • R 7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
  • R 7 is not substituted alkyl, heterocyclyl, alkheterocyclyl, or alkaryl.
  • R 7 is not 2-methoxyethyl or benzyl.
  • R 7 is not substituted alkyl or alkaryl.
  • R 7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl.
  • R 7 is not substituted alkyl.
  • R 7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl.
  • R 7 is not substituted alkyl or alkheterocyclyl.
  • R 7 is not benzyl.
  • R 7 is not alkaryl.
  • R 7 is not methyl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , Z 2 is CR 3 , and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
  • R 7 is not alkyl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , Z 2 is CR 3 , and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • each R 1 and R 2 is H.
  • each R 3 is H, and each R 5 and R 6 is chloro,
  • R 7 is not methyl.
  • the compound is according to formula (la):
  • Z 1 is -OR 7 , -N(R 10 )R 7 , -SR 7 , or -C(R 10 )(R 1 1 )R 7 ;
  • each R 1 and R 2 is, independently, H or optionally substituted Ci -3 alkyl
  • R 3 is H, halogen, optionally substituted Ci -3 alkyl, or optionally substituted Ci -3 alkoxy
  • R 4 is halogen, optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkoxy, optionally substituted amino, optionally substituted C 1-6 thioalkyl, or optionally substituted C 6- io aryl, or R 3 and R 4 , together with the atoms to which each is attached, join to form an optionally substituted five-, six-, or seven- membered ring optionally comprising from one to three heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur, where the nitrogen is optionally substituted with R 9 ;
  • each R 5 and R 6 is, independently, H, optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkoxy, halogen, or CN;
  • R 7 is optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkcycloalkyl, optionally substituted Ci- 3 alkheterocyclyl, or optionally substituted Ci -3 alkaryl, and R 8 is H; or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five-, six-, or seven- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur;
  • R 9 is H, optionally substituted C 1 -3 alkyl, optionally substituted C 3 . 8 cycloalkyl, optionally substituted C 6- io aryl, optionally substituted C 2 . 9 heteroaryl, optionally substituted C 2 . 9 heterocyclyl, optionally substituted Ci -3 alkcycloalkyl, optionally substituted Ci -3 alkheterocyclyl, or optionally substituted Ci- 3 alkaryl;
  • R 1 and R 2 when each of R 1 and R 2 is H, R 3 is H, R 4 is methyl or chloro, and each of R 5 and R 6 is chloro,
  • Z 1 is not methoxy.
  • R 3 is H
  • R 4 is halogen (e.g., chloro)
  • Z 1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy.
  • R 5 and R 6 when each of R 5 and R 6 is chloro, R 3 is H, and R 4 is halogen (e.g., chloro),
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is dimethylaminoethyl, optionally substituted Ci -3 alkcycloalkyl, or optionally substituted Ci -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 and R 6 when each of R 5 and R 6 is chloro, R 3 is H, and R 4 is halogen (e.g., chloro),
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is optionally substituted C 1 -3 alkcycloalkyl, or optionally substituted C 1 -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 3 is H
  • R 4 is halogen (e.g., chloro)
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is optionally substituted C 1 -3 alkcycloalkyl, or optionally substituted C 1 -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ma): (Ma),
  • R 7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C 2 alkheteroaryl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not alkyl or unsubstituted C 2 alkheteroaryl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C 2 alkheterocyclyl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not alkyl or C 2 alkheterocyclyl.
  • R 5 is chloro
  • R 6 is bromo
  • Z 1 is -OR 7
  • R 3 and R 4 combine to form a group according to formula (Ma)
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 when R 5 is chloro, R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (Ma),
  • Z is -OR 7 , and R 7 and R 8 combine to form -CH 2 -CH 2 -.
  • R 5 and R 6 when each R 5 and R 6 is bromo, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not methyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
  • R 7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheteroaryl, or substituted alkheteroaryl.
  • R 7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
  • R 9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , R 7 and R 8 combine to form -CH 2 -CH 2 -, and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 9 is H or -C(0)-N(H)-(linear d_ 3 alkyl).
  • R 5 and R 6 when each R 5 and R 6 is halo, and R 3 and R 4 combine to form a group according to formula (Ma), Z is -OR 7 , R 7 is not methyl or 2-chloroethyl, and R 9 is H or -C(0)-N(H)-(linear C 1 -3 alkyl).
  • R 5 when R 5 is methoxy, R 6 is methyl, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not methyl.
  • R 5 when R 5 is chloro, R 6 is ethyl, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not methyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Mb) or (lie),
  • R 7 is not methyl or 2-(N,N-diethylamino)ethyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Mb) or (lie),
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • each R 5 and R 6 is chloro, and R 3 and R 4 combine to form a group according to formula (lib) or (lie),
  • Z 1 is -OR 7 , and R 7 and R 8 combine to form -CH 2 -CH 2 -.
  • R 7 when R 7 is methyl, R 5 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ilia),
  • R 6 is not bromo.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (Ilia),
  • R 7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl.
  • Z 1 is -OR 7
  • R 3 and R 4 combine to form a group according to formula (Ilia)
  • R 8 is not H.
  • each R 5 and R 6 is chloro
  • R 7 is methyl and R 8 is H, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 when R 5 is chloro, R 6 is methoxy, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (1Mb):
  • R 1 nor R 2 is 2-(N,N-diethylamino)ethyl.
  • R 5 when R 5 is chloro, R 6 is methoxy, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (1Mb),
  • each R 1 and R 2 is H.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (IVa):
  • R 7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
  • R 7 is not substituted alkyl, heterocyclyl, alkheterocyclyl, or alkaryl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (IVb):
  • R 7 is not 2-methoxyethyl or benzyl.
  • R 7 is not substituted alkyl or alkaryl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (IVc):
  • R 7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl.
  • R 7 is not substituted alkyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (IVd):
  • R 7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl.
  • R 7 is not substituted alkyl or alkheterocyclyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (IVe)
  • R 7 is not benzyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (IVe) or (IVf),
  • R 7 is not alkaryl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
  • R 7 is not methyl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
  • R 7 is not alkyl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 when R 5 is chloro, R 6 is bromo, and Z 1 is -OR 7 ,
  • R 3 and R 4 do not combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk).
  • each R 1 and R 2 is H. In certain embodiments, when R 3 is H, and each R 5 and R 6 is chloro,
  • R 7 is not methyl.
  • R 3 and R 4 when R 3 and R 4 , together with the atoms to which each is attached, join to form a substituted five-membered ring comprising one nitrogen, the five-membered ring is not substituted with oxo. In particular embodiments, when R 3 and R 4 , together with the atoms to which each is attached, join to form a substituted five-membered ring comprising one sulfur, the five-membered ring is not substituted with hydroxy or Ci -3 alkoxy.
  • R 3 and R 4 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered saturated ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- of six-membered ring optionally comprising one or two heteroatoms selected from nitrogen and oxygen.
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring.
  • a compound is according to formula (lb):
  • each of R 1 and R 2 is, independently, H or optionally substituted C 1-3 alkyl
  • R 3 is H, halogen, optionally substituted Ci -3 alkyl, or optionally substituted Ci -3 alkoxy
  • R 4 is halogen, optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkoxy, optionally substituted amino, optionally substituted Ci -6 thioalkoxy, or optionally substituted C 6- io aryl, or R 3 and R 4 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one sulfur, or one oxygen, where the nitrogen is optionally substituted with R 9 ;
  • each of R 5 and R 6 is, independently, optionally substituted C 1-3 alkyl (e.g., optionally substituted Ci -3 acyl), optionally substituted Ci -3 alkoxy, halogen, or CN;
  • R 7 is optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkcycloalkyl, optionally substituted Ci- 3 alkheterocyclyl, or optionally substituted Ci -3 alkaryl, and R 8 is H; or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring; and
  • R 9 is H, optionally substituted C 1 -3 alkyl, optionally substituted C 3 . 8 cycloalkyl, optionally substituted C 6- io aryl, optionally substituted C 2 . 9 heteroaryl, optionally substituted C 2 . 9 heterocyclyl, optionally substituted C 1-3 alkcycloalkyl, optionally substituted C 1 -3 alkheterocyclyl, or optionally substituted Ci- 3 alkaryl.
  • R 1 and R 2 when each of R 1 and R 2 is H, R 3 is H, R 4 is methyl or chloro, and each of R 5 and R 6 is chloro,
  • R 7 is not methyl.
  • R 3 is H
  • R 4 is halogen (e.g., chloro)
  • R 7 is not 2-amino-2oxoethyl, 2-(N,N-diethylamino)ethyl, methyl, or benzyl.
  • R 7 is dimethylaminoethyl, optionally substituted Ci -3 alkcycloalkyl, or optionally substituted Ci -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 7 is optionally substituted C 1 -3 alkcycloalkyl, or optionally substituted C 1-3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 3 is H
  • R 4 is halogen (e.g., chloro)
  • R 7 is optionally substituted C 1 -3 alkcycloalkyl, or optionally substituted C 1-3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 when R 5 is chloro, R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (lla):
  • R 7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)-ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
  • R 5 when R 5 is chloro, R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (lla),
  • R 7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C 2 alkheteroaryl.
  • R 5 when R 5 is chloro, R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (lla),
  • R 7 is not alkyl or C 2 alkheteroaryl.
  • R 5 when R 5 is chloro, R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (lla),
  • R 7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C 2 alkheterocyclyl.
  • R 5 when R 5 is chloro, R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (lla),
  • R 7 is not alkyl or C 2 alkheterocyclyl.
  • R 5 is chloro
  • R 6 is bromo
  • R 3 and R 4 combine to form a group according to formula (lla)
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring comprising one oxygen atom and optionally comprising one more heteroatom selected from nitrogen, oxygen, and sulfur.
  • R 5 is chloro
  • R 6 is bromo
  • R 3 and R 4 combine to form a group according to formula (Ma)
  • R 7 and R 8 combine to form -CH 2 -CH 2 -.
  • R 5 and R 6 when each R 5 and R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not methyl.
  • R 7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
  • R 7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheteroaryl, or substituted alkheteroaryl.
  • R 7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
  • R 7 and R 8 when each R 5 and R 6 is chloro, R 7 and R 8 combine to form -CH 2 -CH 2 -, and R 3 and R 4 combine to form a group according to formula (lla),
  • R 9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl.
  • R 7 and R 8 combine to form -CH 2 -CH 2 -
  • R 3 and R 4 combine to form a group according to formula (lla)
  • R 9 is H or -C(0)-N(H)-(linear d_ 3 alkyl).
  • R 9 is H or -C(0)-N(H)-(linear d_ 3 alkyl).
  • R 5 when R 5 is methoxy, R 6 is methyl, and R 3 and R 4 combine to form a group according to formula (lla),
  • R 7 is not methyl.
  • R 5 when R 5 is chloro, R 6 is ethyl, and R 3 and R 4 combine to form a group according to formula (Ma),
  • R 7 is not methyl.
  • each R 5 and R 6 is chloro, and R 3 and R 4 combine to form a group according to formula (Mb) or (lie),
  • R 7 is not methyl or 2-(N,N-diethylamino)ethyl.
  • each R 5 and R 6 is chloro and R 3 and R 4 combine to form a group according to formula (Mb) or (lie),
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring comprising one oxygen atom and optionally comprising one more heteroatom selected from nitrogen, oxygen, and sulfur.
  • each R 5 and R 6 is chloro and R 3 and R 4 combine to form a group according to formula (lib) or (lie),
  • R 7 and R 8 combine to form -CH 2 -CH 2 -.
  • R 7 when R 7 is methyl, R 5 is chloro, and R 3 and R 4 combine to form a group according to formula (Ilia),
  • R 6 is not bromo.
  • R 7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl.
  • R 8 is not H.
  • each R 5 and R 6 is chloro
  • R 7 is methyl
  • R 8 is H, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring comprising one oxygen atom and optionally comprising one more heteroatom selected from nitrogen, oxygen, and sulfur.
  • R 5 when R 5 is chloro, R 6 is methoxy, and R 3 and R 4 combine to form a group according to formula (1Mb):
  • R 1 nor R 2 is 2-(N,N-diethylamino)ethyl.
  • R 5 when R 5 is chloro, R 6 is methoxy, and R 3 and R 4 combine to form a group according to formula (1Mb),
  • each R 1 and R 2 is H.
  • R 5 and R 6 when each R 5 and R 6 is chloro, and R 3 and R 4 combine to form a group according to formula (IVa):
  • R 7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
  • R 7 is not substituted alkyl, heterocyclyl, alkheterocyclyl, or alkaryl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, and R 3 and R 4 combine to form a group according to formula (IVb):
  • R 7 is not 2-methoxyethyl or benzyl.
  • R 7 is not substituted alkyl or alkaryl.
  • R 7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, and R 3 and R 4 combine to form a group according to formula (IVc),
  • R 7 is not substituted alkyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, and R 3 and R 4 combine to form a group according to formula (IVd):
  • R 7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, and R 3 and R 4 combine to form a group according to formula (IVd),
  • R 7 is not substituted alkyl or alkheterocyclyl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, and R 3 and R 4 combine to form a group according to formula (IVe) or (IVf):
  • R 7 is not benzyl.
  • R 7 is not substituted alkyl or alkaryl.
  • R 5 when R 5 is chloro, R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
  • R 7 is not methyl.
  • R 5 when R 5 is chloro, R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk), R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 when R 5 is chloro, R 6 is bromo, and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk), R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 when R 5 is chloro, R 6 is bromo,
  • R 3 and R 4 do not combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk).
  • each R 1 and R 2 is H.
  • R 7 is not methyl.
  • R 3 is H, halogen, optionally substituted C 1 -3 alkyl (e.g., optionally substituted C 1-3 acyl), or optionally substituted C 1-3 alkoxy
  • R 4 is halogen, optionally substituted C 1 -3 alkyl (e.g., optionally substituted C 1 -3 acyl), optionally substituted C 1-3 alkoxy, optionally substituted amino, optionally substituted Ci -6 thioalkyl, or optionally substituted C 6- io aryl, or R 3 and R 4 , together with the atoms to which each is attached, join to form an optionally substituted five- or six- membered ring optionally comprising one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R 9 .
  • R 7 is optionally substituted C 1 -3 alkyl, optionally substituted C 1 -3 alkcycloalkyl, optionally substituted C 1 -3 alkheterocyclyl, or optionally substituted C 1-3 alkaryl, and R 8 is H; or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 3 and R 4 when R 3 and R 4 combine to form a group according formula (Mb) or (lie), the carbonyl group of formula (lib) or (lie) is proximal to C 6 . In other embodiments, when R 3 and R 4 combine to form a group according to formula (lib) or (lie), the carbonyl group of formula (lib) or (lie) is proximal to C 5 or C 6 .
  • the N atom of the group according to formula (Ilia) is proximal to C 5 .
  • the N atom of the group according to formula (lllb) is proximal to C 5 or C 6 .
  • the N atom of the group according to formula (lllb) is proximal to C 6 .
  • the N atom of the group according to formula (lllb) is proximal to C 5 or C 6 .
  • the S atom of the group according to any one of formulae (IVa)-(IVk) is proximal to C 6 .
  • R 3 and R 4 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring.
  • R 3 and R 4 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring.
  • R 3 and R 4 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur, R 7 and R 8 combine to form a -CH 2 CH 2 - group.
  • R 7 and R 8 together with the atoms to which each is attached, join to form a five- or six-membered ring.
  • R 7 is optionally substituted Ci -3 alkyl. In other embodiments, R 7 is
  • Ci -3 alkylamino-C ⁇ -alkyl e.g., Ci -3 haloalkylamino-C ⁇ -alkyl (e.g., Ci -3 fluoroalkylamino-C 1 -3 -alkyl)
  • di-(C 1 -3 alkyl)amino-C 1-3 -alkyl e.g., R Y1 N(R Y2 )-(C 1-3 alkyl)-, where each of R Y1 and R Y2 is, independently, unsubstituted C 1 -3 alkyl or C 1 -3 haloalkylamino-C 1 -3 -alkyl, (e.g., C 1 -3 fluoroalkylamino-C 1 -3 -alkyl), or C 1 -3 haloalkyl (e.g., C 1 -3 fluoroalkyl).
  • R 7 is unsubstituted C 1 -3 alkyl, C 1-3 alkylamino-C 1 -3 -alkyl (e.g., C 1 -3 haloalkylamino-C 1 -3 -alkyl, e.g., C 1-3 fluoroalkylamino-Ci -3 -alkyl), or di-(Ci -3 alkyl)amino-Ci -3 -alkyl (e.g., R Y1 N(R Y2 )-(d.
  • each of R Y1 and R Y2 is, independently, unsubstituted Ci -3 alkyl, or d_ 3 haloalkylamino-Ci -3 -alkyl, e.g., Ci -3 fluoroalkylamino-Ci -3 -alkyl).
  • R 7 is optionally substituted C 1 -3 alkyl (e.g., R 7 is methyl), or R 7
  • each of R 24 and R 25 is, independently, H or optionally substituted C 1-3 alkyl (e.g., each of R 24 and R 25 is, independently, optionally substituted C 1 -3 alkyl, e.g., each of R 24 and R 25 is, independently, C 1 -3 haloalkyl (e.g., C 1 -3 fluoroalkyl); alternatively each of R 24 and R 25 is, independently, unsubstituted Ci -3 alkyl, e.g., methyl).
  • R 3 and R 4 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one nitrogen, an optional substituent on the ring is not oxo. In certain embodiments, when R 3 and R 4 , together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur, an optional substitutent on the ring is not hydroxyl or C 1-3 alkoxy.
  • each R 1 and R 2 is H.
  • each R 3 and R 4 is, independently, optionally substituted Ci -3 alkyl or optionally substituted Ci -3 alkoxy; or R 3 and R 4 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one sulfur, or one oxygen, where the nitrogen is optionally substituted with R 9 .
  • R 3 and R 4 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring.
  • R 3 and R 4 combine to
  • R 3 and R 4 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one nitrogen.
  • the N atom is proximal to C 5 of Formula (I).
  • R 3 and R 4 together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur.
  • the S atom is proximal to C 6 of the compound of the invention.
  • R 13 is optionally substituted Ci -3 alkyl, e.g., R 13B is -C(0)-R 13C , where R 13C is optionally substituted C 1-3 alkoxy or optionally substituted amino.
  • R 4 is C 1-3 alkyl. In specific embodiments, R 4 is methyl, ethyl, or isopropyl. In certain embodiments, R 4 is C 1 -3 alkoxy (e.g., R 4 is methoxy). In other embodiments, R 4 is optionally substituted C 1-6 thioalkoxy (e.g., R 4 is 4-amino-4-oxobutyl). In yet other embodiments, R 4 is optionally substituted amino (e.g., R 4 is methylamino). In still other embodiments, R 4 is halogen (e.g., R 4 is chloro).
  • R 3 is hydrogen or d_ 3 alkyl (e.g., R 3 is hydrogen, methyl, or ethyl).
  • R 3 and R 4 combine to form -X 1 -X 2 -X 3 -, where
  • each R 16 , R 19 , R 20 , R 21 , R 22 , and R 23 is, independently, H, or optionally substituted d_ 3 alkyl; and n is 1 or 2.
  • the chain of atoms -X 1 -X 2 -X 3 - includes no more than one heteroatom, the heteroatom being selected from nitrogen, oxygen, and sulfur.
  • X 1 is -(CR 14 R 15 )-.
  • each R 14 and R 15 is H.
  • R 16 is H.
  • X 1 is - N(R 9 )-.
  • R 9 is H or optionally substituted Ci -3 alkyl.
  • R 9 is hydrogen, methyl, or ethyl.
  • X 3 is -N(R 9 )-. In yet other embodiments, X 3 is halogen, optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkoxy, optionally substituted C 1-6 thioalkoxy, or optionally substituted C 6- io aryl.
  • Z 1 and R 8 combine to form -Z 3 -Y 1 -Y 2 -, where
  • each R 20 , R 21 , and R 22 is, independently, H or optionally substituted C 1 -3 alkyl;
  • n 1 or 2;
  • R 7 and R 8 form a group -Y 1 -Y 2 -, where:
  • each R 26 and R 27 is, independently, H or optionally substituted Ci -3 alkyl
  • n 1 or 2.
  • Z 3 is oxygen.
  • Y 1 is -(CR 26 R 27 ) m - or optionally substituted Ci -3 alkyl.
  • Y 1 is -(CR 26 R 27 ) m -
  • Y 1 is optionally substituted Ci -3 alkyl (e.g., Y 1 is methyl).
  • Y 1 is -(CH 2 ) k -N(R 24 )R 25 , where k is 2 or 3, and where each R 24 and R 25 is, independently, H or optionally substituted Ci -3 alkyl. In still other embodiments, k is 2. In further embodiments, each R and R is, independently, optionally substituted d.3 alkyl (e.g., each R 24 and R 25 is methyl). In certain embodiments, Y 2 is -(CR 26 R 27 )- or H. In other embodiments, Y 2 is -(CR 26 R 27 )-
  • each R 1 and R 2 is H
  • a compound of the invention has a structure according to formula (Va):
  • a compound of the invention has a structure according to formula (Vb):
  • each R 1 and R 2 is, independently, H or optionally substituted d -3 alkyl
  • each R 5 and R 6 is, independently, optionally substituted d -3 alkyl, optionally substituted Ci -3 alkoxy, halogen, or CN;
  • each R 14 , R 15 , R 16 , R 17 , R 17 , R 18 , R 20 , R 19 , R 20 , R 21 , R 22 , R 23 , R 26 , and R 27 is, independently, H or optionally substituted C 1-3 alkyl;
  • n 1 or 2;
  • n 1 or 2.
  • R 5 is chloro
  • R 6 is bromo
  • -X 1 -X 2 -X 3 - forms a group according to formula (lla):
  • Y 1 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)-ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
  • R 5 when R 5 is chloro, R 6 is bromo, and -X 1 -X 2 -X 3 - forms a group according to formula (lla),
  • Y 1 is not optionally substituted C 1 -3 alkyl or optionally substituted C 2 alkheteroaryl.
  • R 5 when R 5 is chloro, R 6 is bromo, and -X 1 -X 2 -X 3 - forms a group according to formula (lla),
  • Y 1 is not optionally substituted Ci -3 alkyl or optionally substituted C 2 alkheterocyclyl.
  • Y 2 is not H.
  • R 5 when R 5 is chloro, R 6 is bromo, and -X 1 -X 2 -X 3 - forms a group according to formula (lla),
  • each Y 1 and Y 2 is -CH 2 -.
  • each R 5 and R 6 is bromo, and -X 1 -X 2 -X 3 - forms a group according to formula (lla),
  • Y 1 is not methyl.
  • Y 1 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol-1 - yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
  • Y 1 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheteroaryl, or substituted alkheteroaryl.
  • R 5 and R 6 when each R 5 and R 6 is chloro, and -X 1 -X 2 -X 3 - forms a group according to formula (Ma),
  • Y 1 is not unsubstituted alkyi, substituted alkyi, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
  • each Y 1 and Y 2 is -CH 2 -, and -X 1 -X 2 -X 3 - forms a group according to formula (Ma),
  • R 9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl.
  • R 7 and R 8 combine to form -CH 2 -CH 2 -, and -X 1 - X 2 -X 3 - forms a group according to formula (Ma),
  • R 9 is H or -C(0)-N(H)-(linear d_ 3 alkyi).
  • R 9 is H or -C(0)-N(H)-(linear d_ 3 alkyi).
  • R 5 when R 5 is methoxy, R 6 is methyl, and -X 1 -X 2 -X 3 - forms a group according to formula (Ma),
  • Y 1 is not methyl.
  • R 5 when R 5 is chloro, R 6 is ethyl, and -X 1 -X 2 -X 3 - forms a group according to formula (Ma),
  • Y 1 is not methyl.
  • each R 5 and R 6 is chloro, and -X 1 -X 2 -X 3 - forms a group according to formula (Mb) or (lie),
  • Y 1 is not methyl or 2-(N,N-diethylamino)ethyl.
  • each R 5 and R 6 is chloro and -X 1 -X 2 -X 3 - forms a group according to formula (Mb) or (lie),
  • Y 2 is not H.
  • each R 5 and R 6 is chloro and -X 1 -X 2 -X 3 - forms a group according to formula (Mb) or (lie),
  • each Y 1 and Y 2 is -CH 2 -.
  • R 7 when R 7 is methyl, R 5 is chloro, and -X 1 -X 2 -X 3 - forms a group according to formula (Ilia), HN ⁇ (ma),
  • R 6 is not bromo.
  • R 7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl.
  • each R 5 and R 6 is chloro
  • Y 1 is methyl and Y 2 is H, or
  • each R 5 and R 6 is chloro
  • Y 2 is not H.
  • R 1 nor R 2 is 2-(N,N-diethylamino)ethyl.
  • R 5 when R 5 is chloro, R 6 is methoxy, and -X 1 -X 2 -X 3 - forms a group according to formula (1Mb),
  • each R 1 and R 2 is H.
  • Y 1 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
  • Y 1 is not heterocyclyl, alkheterocyclyl, or alkaryl.
  • Y 1 is not 2-methoxyethyl or benzyl.
  • Y 1 is not 2-methoxyethyl or benzyl.
  • each R 5 and R 6 is chloro, and -X 1 -X 2 -X 3 - forms a group according to formula (IVb),
  • Y 1 is not substituted alkyl or alkaryl.
  • Y 1 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl.
  • Y 1 is not substituted alkyl.
  • Y 1 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl.
  • Y 1 is not substituted alkyl or alkheterocyclyl.
  • each R 5 and R 6 is chloro, and -X 1 -X 2 -X 3 - forms a group according to formula (IVe) or (IVf): Y 1 is not benzyl.
  • Y 1 is not alkaryl or substituted alkyl.
  • R 5 when R 5 is chloro, R 6 is bromo, and -X 1 -X 2 -X 3 - forms a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
  • Y 1 is not methyl.
  • R 5 when R 5 is chloro, R 6 is bromo, -X 1 -X 2 -X 3 - forms a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
  • Y 2 is not H.
  • R 5 when R 5 is chloro, R 6 is bromo,
  • R 3 and R 4 do not combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk).
  • R 6 is methoxy
  • each R 1 and R 2 is H.
  • Y 1 is not methyl.
  • R 10 is H, optionally substituted C 1-3 alkyl, optionally substituted C 3 . 8 cycloalkyl, optionally substituted C 1-3 alkcycloalkyl, optionally substituted C 1 -3 alkheterocyclyl, or optionally substituted Ci- 3 alkaryl.
  • R 10 is H, optionally substituted Ci -3 alkyl, optionally substituted Ci- 3 alkcycloalkyl, optionally substituted Ci -3 alkheterocyclyl, or optionally substituted Ci -3 alkaryl.
  • X 1 is -N(R 9 )-
  • X 3 C(R 23 )-
  • each of R 17 , R 19 , and R 23 is H
  • Y 1 is -(CR 26 R 27 ) m -
  • Y 2 is -(CR 26 R 27 )-.
  • X 1 is -N(R 9 )-
  • X 3 C(R 23 )-
  • each of R 9 , R 19 , and R 23 is H
  • each of Y 1 and Y 2 is -CH 2 -.
  • X 1 is -N(R 9 )-
  • X 3 C(R 23 )-
  • each of R 19 and R 23 is H
  • Y 1 is - (CR 26 R 27 ) m -
  • Y 2 is -(CR 26 R 27 )-.
  • X 1 is -N(R 9 )-
  • X 3 C(R 23 )-
  • Y 2 is H
  • each of R 5 and R 6 is -CI
  • Y 1 is Me.
  • Y 1 when X 1 is H, and each of R 5 and R 6 is CI, Y 1 is -(CR 26 R 27 ) m - and Y 2 is -(CR 26 R 27 )-. In particular embodiments, when X 1 is H, and each of R 5 and R 6 is hal, Y 1 is -(CR 26 R 27 ) m - and Y 2 is -(CR 26 R 27 )- (e.g., each of Y 1 and Y 2 is -CH 2 -).
  • Y 1 is -(CR 26 R 27 ) m - and Y 2 is -(CR 26 R 27 )- (e.g., each of Y 1 and Y 2 is -CH 2 -).
  • each of R 5 and R 6 is, independently, halo, or optionally substituted Ci -3 alkyl, e.g., each of R 5 and R 6 is halo (e.g., each of R 5 and R 6 is -CI).
  • R 26 is H. In other embodiments, R 27 is H. In certain embodiments, n is 1 . In other embodiments, m is 1 . In particular embodiments, Y 1 is optionally substituted C 1-3 alkyl. In other embodiments, Y 1 is unsubstituted C 1 -3 alkyl, C 1 -3 alkylamino-C 1-3 -alkyl (e.g., C 1 -3 haloalkylamino-C 1 -3 -alkyl, e.g., C 1-3 fluoroalkylamino-C 1-3 -alkyl), di-(C 1 -3 alkyl)amino-C 1-3 -alkyl (e.g., R Y1 N(R Y2 )-(d_ 3 alkyl)-, where each of R Y1 and R Y2 is, independently, unsubstituted C 1 -3 alkyl, or C 1 -3 haloalkylamino-
  • Y 1 is unsubstituted d. 3 alkyl, d_ 3 alkylamino-Ci -3 -alkyl (e.g., d. 3 haloalkylamino-Ci -3 -alkyl, e.g., Ci -3 fluoroalkylamino-d- 3 -alkyl), or di-(Ci -3 alkyl)amino-Ci -3 -alkyl (e.g., R Y1 N(R Y2 )-(d_ 3 alkyl)-, where each of R Y1 and R Y2 is, independently, unsubstituted C 1 -3 alkyl, or C 1 -3 haloalkylamino-C 1-3 -alkyl, e.g., C 1-3 fluoroalkylamino-C 1 -3 -alkyl).
  • d_ 3 alkylamino-Ci -3 -alkyl e.g.
  • a compound of the invention has the formula as shown in Table 1 Table!.
  • the compound is not compound 38 or compound 39.
  • each of R 1 and R 2 is H, R 5 is CI, and R 6 is F or CI.
  • R 3 is H, halogen, optionally substituted C 1 -3 alkyl, or optionally substituted C 1-3 alkoxy
  • R 4 is halogen, optionally substituted C 1-3 alkyl, optionally substituted C 1 -3 alkoxy, optionally substituted amino, or optionally substituted C 1 -6 thioalkoxy, or R 3 and R 4 , together with the atoms to which each is attached, join to form an optionally substituted five- or six- membered ring optionally comprising one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R 9 .
  • R 3 is H, optionally substituted alkyl, or optionally substituted C 1 -3 alkoxy
  • R 4 is halogen, optionally substituted C 1-3 alkyl, optionally substituted C 1-3 alkoxy, optionally substituted amino, or optionally substituted C 1 -6 thioalkoxy, or R 3 and R 4 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R 9 .
  • R 3 is H, optionally substituted Ci alkyl, or optionally substituted Ci- 3 alkoxy
  • R 4 is halogen, optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkoxy, optionally substituted amino, or optionally substituted C 1 -6 thioalkoxy, or R 3 and R 4 join to form one of the following groups:
  • Z 2 is N, and R 4 is halogen, optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkoxy, optionally substituted amino, or optionally substituted Ci -6 thioalkoxy.
  • Z 2 is N, and R 4 is optionally substituted C 1-3 alkoxy or optionally substituted C 1 -6 thioalkoxy.
  • R 9 is H or d_ 3 alkyl.
  • the compound of the invention has a molecular weight of less than about 500 g/mol (e.g., less than about 450 g/mol, or less than about 400 g/mol).
  • the compound of the invention exhibits apical to basal (A ⁇ B) permeability in MDR1 -MDCK assay of greater than about 1 x 10 ⁇ 7 cm/sec (e.g., greater than about 5 x 10 ⁇ 7 cm/sec, greater than about 1 x 1 0 "6 cm/sec, or greater than about 3 x 1 0 "6 cm/sec).
  • the compound of the invention exhibits the (B ⁇ A)/(A ⁇ B) ratio of less than about 30 (e.g., less than about 10, less than about 5, or less than about 3).
  • the invention features a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, and one or more of pharmaceutically acceptable carriers or excipients.
  • the composition is formulated for administration orally, intradermally, intramuscularly, parenterally, intravenously, intra-arterially, intracranially, subcutaneously, intraorbitally, intraventricularly, intraspinally, intraperitoneally, or intranasally.
  • the composition is formulated for oral administration.
  • the invention features a method of treating a disorder in a mammal (e.g., a caused by the action of heat shock protein 90 (Hsp90).
  • the method involves administering to the mammal an effective amount of a compound according to formula (I):
  • Z 1 is -OR 7 , -N(R 10 )R 7 , -SR 7 , or -C(R 10 )(R 1 1 )R 7 ;
  • each R 1 and R 2 is, independently, H or optionally substituted Ci -3 alkyl
  • R 3 is H, halogen, cyano, optionally substituted C 1 -6 alkyl, optionally substituted C 1-3 alkoxy, or optionally substituted amino
  • R 4 is halogen, cyano, optionally substituted C 1 -6 alkyl, optionally substituted C 1 -3 alkoxy, optionally substituted amino, optionally substituted C 1 -6 thioalkoxy, or optionally substituted C 6 -i 0 aryl, or R 3 and R 4 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R 9 ;
  • each R 5 and R 6 is, independently, optionally substituted Ci -3 alkyl, optionally substituted Ci -3 alkoxy, halogen, or CN;
  • R 7 is optionally substituted C 1 -3 alkyl, optionally substituted C 1 -3 alkcycloalkyl, optionally substituted C 1 -3 alkheterocyclyl, or optionally substituted C 1-3 alkaryl, and R 8 is H; or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur;
  • R 9 is H, optionally substituted Ci -3 alkyl, optionally substituted C 3 . 8 cycloalkyl, optionally substituted C 6 -i 0 aryl, optionally substituted C 2 - 9 heteroaryl, optionally substituted C 2 - 9 heterocyclyl, optionally substituted Ci -3 alkcycloalkyl, optionally substituted Ci -3 alkheterocyclyl, or optionally substituted C 1 -3 alkaryl;
  • R m is H, halogen, optionally substituted Ci -4 alkyl, or optionally substituted Ci -3 alkoxy;.
  • Z 1 is not methoxy.
  • R 3 is H
  • R 4 is methyl or halogen (e.g., chloro)
  • each of R 5 and R 6 is halogen (e.g., chloro)
  • Z 2 is not unsubstituted Ci -3 alkoxy.
  • R 3 is H
  • R 4 is optionally substituted Ci -6 thioalkoxy
  • each of R 5 and R 6 is halogen (e.g., chloro)
  • Z 1 is not cyanomethoxy or aminomethoxy.
  • Z 1 is not cyanomethoxy or aminomethoxy.
  • Z 1 is not cyanomethoxy or aminomethoxy.
  • Z 1 is not cyanomethoxy or aminomethoxy.
  • Z 1 is not substituted Ci alkoxy.
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is methyl, dialkylaminoethyl, optionally substituted C 1 -3 alkcycloalkyl, optionally substituted C 1 -3 alkheterocyclyl, or optionally substituted C 1-3 alkaryl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • each of R 5 and R 6 is chloro, R 3 is H, and R 4 is halogen (e.g., chloro),
  • Z 1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy.
  • Z 1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy.
  • each of R 5 and R 6 is chloro, R 3 is H, and R 4 is halogen (e.g., chloro),
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is dimethylaminoethyl, optionally substituted C 1 -3 alkcycloalkyl, or optionally substituted C 1 -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 and R 6 is chloro
  • R 3 is H
  • R 4 is halogen (e.g., chloro)
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is optionally substituted d_ 3 alkcycloalkyl, or optionally substituted Ci -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • Z 1 is -OR 7 , -N(R 7 )R 10 , -SR 7 , or -C(R 7 )(R 10 )R 1 1 , in which R 7 is optionally substituted d_ 3 alkcycloalkyl, or optionally substituted Ci -3 alkheterocyclyl, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
  • R 7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C 2 alkheteroaryl.
  • R 7 is not alkyl or unsubstituted C 2 alkheteroaryl.
  • R 7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C 2 alkheterocyclyl.
  • R 7 is not alkyl or C 2 alkheterocyclyl.
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • Z is -OR 7 , and R 7 and R 8 combine to form -CH 2 -CH 2 -.
  • R 7 is not methyl.
  • R 7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
  • R 7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheteroaryl, or substituted alkheteroaryl.
  • R 7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
  • R 9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl.
  • R 9 is H or -C(0)-N(H)-(linear d_ 3 alkyl).
  • Z is -OR 7 , R 7 is not methyl or 2-chloroethyl, and R 9 is H or -C(0)-N(H)-(linear C 1 -3 alkyl).
  • R 5 is methoxy
  • R 6 is methyl
  • Z 1 is -OR 7
  • Z 2 is CR 3
  • R 3 and R 4 combine to form a group according to formula (Ma)
  • R 7 is not methyl.
  • R 5 is chloro
  • R 6 is ethyl
  • Z 1 is -OR 7
  • Z 2 is CR 3
  • R 3 and R 4 combine to form a group according to formula (Ma)
  • R 7 is not methyl.
  • R 7 is not methyl or 2-(N,N-diethylamino)ethyl.
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • Z 1 is -OR 7 , and R 7 and R 8 combine to form -CH 2 -CH 2 -.
  • R 6 is not bromo.
  • R 7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl.
  • Z 1 is -OR 7
  • Z 2 is CR 3
  • R 3 and R 4 combine to form a group according to formula (Ilia)
  • R 8 is not H.
  • each R 5 and R 6 is chloro
  • R 7 is methyl and R 8 is H, or R 7 and R 8 , together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • R 5 is chloro
  • R 6 is methoxy
  • Z 1 is -OR 7
  • Z 2 is CR 3
  • R 3 and R 4 combine to form a group according to formula (1Mb):
  • R 1 nor R 2 is 2-(N,N-diethylamino)ethyl.
  • each R 1 and R 2 is H.
  • R 7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
  • R 7 is not substituted alkyl, heterocyclyl, alkheterocyclyl, or alkaryl.
  • R 7 is not substituted alkyl or alkaryl.
  • Z 1 is -OR 7
  • Z 2 is CR 3
  • R 3 and R 4 combine to form a group according to formula (IVc): NH
  • R 7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl.
  • R 7 is not substituted alkyl.
  • R 7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl.
  • R 7 is not substituted alkyl or alkheterocyclyl.
  • R 7 is not benzyl.
  • R 7 is not alkaryl.
  • R 7 is not methyl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , Z 2 is CR 3 , and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
  • R 7 is not alkyl.
  • R 5 when R 5 is chloro, R 6 is bromo, Z 1 is -OR 7 , Z 2 is CR 3 , and R 3 and R 4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
  • R 7 and R 8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
  • each R 1 and R 2 is H.
  • each R 3 is H, and each R 5 and R 6 is chloro,
  • R 7 is not methyl.
  • R m is H (e.g., the compound of formula (I) has the following structure:
  • the substituents are defined as described herein.
  • the method involves administering to the mammal an effective amount of a compound according to formula (I).
  • the method involves administering to the mammal an effective amount of a compound according to formula (lb).
  • the method involves administering to the mammal an effective amount of a compound according to formula (Va).
  • the method involves administering to the mammal an effective amount of a compound according to formula (Vb).
  • the compound may be selected from Table 2, (e.g., any one of compounds 2, 5-16, 18-27, 29, 33-36, 40-48, and 58-77, or a or a pharmaceutically acceptable salt thereof).
  • the method involves administering to the mammal an effective amount of compound 38 or 39 or a pharmaceutically acceptable salt thereof.
  • the method involves treating a mammal having a neurodegenerative disorder by administering to the mammal the compound of formula (I) (e.g., formula (la), (lb), (Va), or (Vb)).
  • the compound of formula (I) e.g., formula (la), (lb), (Va), or (Vb)
  • the disorder is a neurodegenerative disorder (e.g., a tauopathy).
  • a neurodegenerative disorder e.g., a tauopathy
  • neurodegenerative disorder may be Alzheimer's disease, Huntington's disease, progressive supranuclear palsy, Parkinson's syndrome, Pick's disease, corticobasal degeneration, chronic traumatic
  • the neurodegenerative disorder is Alzheimer's disease.
  • the disorder is a proliferative disorder (e.g., a cancer, e.g., acute myeloid leukemia, gastrointestinal stromal tumor, gastric cancer, glioblastoma, lung cancer, lymphoma, melanoma, myeloma, non-small cell lung cancer, renal cancer, small cell lung cancer, blast-phase chronic myelogenous leukemia, leukemia, lymphoproliferative disorder, metastatic melanoma, relapsed multiple myeloma, refractory multiple myeloma, myeloproliferative disorders, pancreatic cancer, small intestine cancer, or solid tumor).
  • a proliferative disorder e.g., a cancer, e.g., acute myeloid leukemia, gastrointestinal stromal tumor, gastric cancer, glioblastoma, lung cancer, lymphoma, melanoma, myelom
  • the disorder is an inflammatory or autoimmune disease (e.g., rheumatoid arthritis, systemic lupus erythermatosus, or asthma).
  • the disorder is a cardiovascular disease (e.g., atherosclerosis or cardiomyoapthy).
  • the disorder is an allergy.
  • the invention features a method of treating an infectious disease in a mammal by administering an effective amount of the compound of the invention (e.g., compounds of the aspects described above), or a pharmaceutically acceptable salt thereof to the mammal.
  • the infectious disease is a viral infection.
  • the viral infection is a virus of Herpesviridae family (e.g., herpes simplex virus-1 , herpes simplex virus-2, herpes herpesvirus-5, Kaposi's sarcoma-associated herpesvirus, varicella zoster virus, or Epstein-Barr virus), Polyomaviridae family (e.g., SV40), Poxviridae family (e.g., vaccinia virus), Reoviridae family (e.g., rotavirus), Birnaviridae family (e.g., infectious bursal disease virus), Picornaviridae family (e.g., poliovirus, rhinovirus, or coxsackievirus), Flaviviridae family (e.g., hepatitis C virus or dengue virus), Arenaviridae family (e.g., lymphocytic choriomeningitis virus),
  • Rhabdoviridae family e.g., vesicular stomatitis virus
  • Paramoxyviridae family e.g., human parainfluenza virus 2, human parainfluenza virus 3, SV5, SV41 , measles virus, or Sendai virus
  • Bunyaviridae family e.g., La Crosse virus
  • Orthomoxyviridae family e.g., influenza A virus
  • Filoviridae family e.g., Ebola virus
  • Retroviridae family e.g., HTLV1 or HIV1
  • Hepadnaviridae family e.g., hepatitis B virus
  • the infectious disease is a fungal infection (e.g., Candida albicans, Aspergillus fumigates, or Pneumocystis jiroveci).
  • the infectious disease is a bacterial infection (e.g., mycobacteria, anthrax, or bacterial pneumonia).
  • the compound is administered orally, sublingually, buccally, transdermal ⁇ , intradermally, intramuscularly, parenterally, intravenously, intra-arterially, intracranial ⁇ , subcutaneously, intraorbitally, intraventricularly, intraspinally,
  • the compound is administered orally.
  • the mammal is human.
  • the invention features a method of inhibiting Hsp90 by contacting a cell with the compound of the invention, or a pharmaceutically acceptable salt thereof.
  • the method is carried out in vitro. In other embodiments, the method is carried out in vivo.
  • the invention features a kit containing:
  • the invention features the compounds of the invention for use in treating a disorder caused by the action of heat shock protein 90 (Hsp90).
  • the disorder is any one of the disorders described in the aspect featuring methods of the invention.
  • the invention features the compounds of the invention for use in treating an infectious disease.
  • the infectious disease is any one of the infectious diseases described in the aspect featuring methods of the invention.
  • the invention features uses of a compound of the invention in treating a disorder caused by the action of heat shock protein 90 (Hsp90).
  • the disorder can be any one of the disorders described in the aspect featuring methods of the invention.
  • the invention features uses of a compound of the invention in treating an infectious disease.
  • the infectious disease is any one of the infectious diseases described in the aspect featuring methods of the invention.
  • the invention features uses of a compound of the invention in the manufacture of a medicament for treating a disorder caused by the action of heat shock protein 90 (Hsp90).
  • the disorder can be any one of the disorders described in the aspect featuring methods of the invention.
  • the invention features uses of a compound of the invention in the manufacture of a medicament for treating an infectious disease.
  • the infectious disease is any one of the infectious diseases described in the aspect featuring methods of the invention.
  • acyl or "alkanoyl,” as used interchangeably herein, represent an alkyl group, as defined herein, or hydrogen attached to the parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl, acetyl, propionyl, butanoyl, and the like.
  • exemplary unsubstituted acyl groups include from 2 to 7 carbons.
  • Acyl can be unsubstituted or substituted in the same as defined for alkyl.
  • alkaryl represents a chemical substituent of formula -(alkylene)-(aryl), where each group is as defined herein and may be substituted or unsubstituted according to each respective definition.
  • alkheterocyclyl represents a chemical substituent of formula -(alkylene)- (heterocyclyl), where each group is as defined herein and may be substituted or unsubstituted according to each respective definition.
  • alkoxy represents a chemical substituent of formula -OR, where R is a C 1 -6 alkyl group, unless otherwise specified (e.g., R is C 1 -3 alkyl).
  • Alkoxy may be unsubstituted or substituted with one, two, or three substituents independently selected from the group consisting of: (1 ) alkoxy of one to six carbons; (2) hydroxyl; (3) amino; (4) alkylamino of one to six carbons; (5) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (6) cycloalkyl of three to eight carbons; (7) oxo; (8) hal; (9) alkylsulfonyl of one to six carbon atoms; (10) thioalkoxy of one to six carbon atoms; (1 1 ) aryl; (12) -C0 2 R A , where R A is selected from the group consisting of (a) hydrogen, (b) alkyl, where R A is
  • alkoxyalkyl represents an alkyl group that is substituted with an alkoxy group.
  • exemplary unsubstituted alkoxyalkyl groups include from 2 to 9 carbons.
  • the alkyl and the alkoxy each can be further substituted with 1 , 2, 3, 4, or 5 substituent groups as defined herein for each respective group.
  • alkyl as used herein, are inclusive of both straight chain and branched chain saturated groups of from 1 to 6 carbons, unless otherwise specified (e.g., from 1 to 3 carbons).
  • Alkyl groups are exemplified by methyl, ethyl, n- and iso-propyl, and may be optionally substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four substituents, unless otherwise specified, independently selected from the group consisting of: (1 ) alkoxy of one to six carbons; (2) hydroxyl; (3) amino; (4) alkylamino of one to six carbons; (5) dialkylamino, where each of alkyl groups is,
  • alkylene and the prefix "alk-,” as used herein, represent a saturated divalent CM O hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, propylene, isopropylene, and the like.
  • C x . y alkylene and the prefix "C x . y alk-” represent alkylene groups having between x and y carbons. Exemplary values for x are 1 , 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • the alkylene can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the alkyl group.
  • alkylsulfonyl represents an alkyl group attached to the parent molecular group through an -S(0) 2 - group.
  • exemplary unsubstituted alkylsulfonyl groups are of from 1 to 6 carbons.
  • the alkyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein.
  • amino represents a chemical substituent of formula -NH 2 .
  • Amino group may be singly substituted with, e.g., an alkyl, alkanoyl, aryl, aryloyl, cycloalkyi, heterocyclyl, or alkheterocyclyl group (e.g., "alkylamino” having formula -NH-(optionally substituted Ci -6 alkyl), e.g., -NH-(unsubstituted Ci- 6 alkyl)), or doubly substituted with, e.g., alkyl, alkanoyl, aryl, aryloyl, cycloalkyi, heterocyclyl, or alkheterocyclyl group (e.g., "dialkylamino” having formula -NR'R", where each of R' and R" is, independently, optionally substituted C 1 -6 alkyl, e.g., -NR'
  • Optionally substituted C 1-6 alkyl group may be a C 1-6 haloalkyl, e.g., a C 1 -6 fluoroalkyl.
  • aminoalkyl represents an alkyl group that is substituted with an amino group. Each of the alkyl and amino groups may be, independently, substituted or unsubstituted as defined herein for each respective group.
  • aryl represents a mono-, bicyclic, or multicyclic carbocyclic ring system having between three and twelve carbons and having one or two aromatic rings.
  • Non-limiting examples of aryl groups include phenyl, naphthyl, 1 ,2-dihydronaphthyl, 1 ,2,3,4-tetrahydronaphthyl, fluorenyl, indanyl, indenyl, and the like.
  • An aryl group may be optionally substituted with one, two, three, four, or five substituents independently selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (7) cycloalkyi of three to eight carbons; (8) oxo; (9) hal; (10) alkylsulfonyl of one to six carbon atoms; (1 1 ) thioalkoxy of one to six carbon atoms; (12) aryl; (13) alkaryl, where alkylene group is one to six carbon atoms; (14) -C0 2 R A , where R A is selected from the group consisting of (a) alkyl, (b) cycloalkyi, (c) alkcycloalkyi, (d) alkaryl
  • aryloyl represents an aryl group attached to the oarent molecular group through an alkyl group.
  • cyano as used herein represents -CN group.
  • cyanoalkyl represents an alkyl group that is substituted with cyano.
  • Exemplary unsubstituted cyanoalkyl groups include from 2 to 9 carbons.
  • the alkyl group can be further substituted with 1 , 2, 3, 4, or 5 substituent groups as defined herein for each respective group.
  • cycloalkyi represents a monovalent saturated or unsaturated non-aromatic cyclic hydrocarbon group of from three to eight carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[2.2.1 .Jheptyl, and the like. Cycloalkyi groups may be optionally substituted with, for example, one, two, three, or four substituents,
  • alkyl of one to six carbons independently, selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (7) cycloalkyi of three to eight carbons; (8) oxo; (9) halo; (10) alkylsulfonyl of one to six carbon atoms; (1 1 ) thioalkoxy of one to six carbon atoms; (12) aryl; (13) alkaryl, where alkylene group is one to six carbon atoms; (14) -C0 2 R A , where R A is selected from the group consisting of (a) alkyl, (b) cycloalkyi, (c) alkcycloalkyi, (d) alkaryl, and (e) hydrogen, where the alkylene group is
  • five-membered ring represent a saturated or unsaturated aromatic or non- aromatic group having five atoms in a cyclic array, where, unless otherwise specified, four atoms are carbons and the remaining atom is selected from the group consisting of carbon, nitrogen, sulfur, and oxygen.
  • a five-membered ring may be fused to another cyclic group selected from heterocyclyl, heteroaryl, cycloalkyi, and aryl.
  • a five-membered ring may be unsubstituted or substituted with, for example, one, two, three, or four substituents, independently selected from the group consisting of: (1 ) alkoxy of one to six carbons; (2) hydroxyl; (3) amino; (4) alkylamino of one to six carbons; (5) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (6) cycloalkyi of three to eight carbons; (7) oxo; (8) alkylsulfonyl of one to six carbon atoms; (9) thioalkoxy of one to six carbon atoms; (10) aryl; (1 1 ) alkaryl, where alkylene group is one to six carbon atoms; (12) optionally substituted alkyl of one to six carbons (e.g., unsubstituted alkyl, alkoxyalkyl, hydroxyalkyl, haloalkyl, or cyanoalkyl); (13)
  • fluoroalkyl represents an alkyl group, as defined herein, where one or more hydrogen radicals (e.g., 1 , 2, 3, 4, or 5, or more hydrogen radicals) bound to the alkyl group have been replaced by a fluorine radical.
  • fluoroalkyl group may be perfluoroalkyl.
  • Prefix "fluoro” indicates that the group in question is substituted by one or more (e.g., 1 , 2, 3, 4, or 5, or more) fluorine radicals.
  • haloalkyl represents an alkyl group, as defined herein, substituted by a halogen group (i.e., F, CI, Br, or I).
  • a Ci -6 haloalkyl may be substituted with one two, three, or in the case of alkyl groups of two carbons or more, four or five halogens.
  • Haloalkyl groups include perfluoroalkyls.
  • haloalkyl is fluoroalkyl.
  • the C 1-6 haloalkyl group may be further substituted with 1 , 2, 3, or 4 substituent groups as described herein for alkyl groups.
  • halogen represents a group selected from fluorine (-F), chlorine (-CI), bromine (-Br), and iodine (-I).
  • halo indicates that the group in question is substituted by a halogen group (i.e., F, CI, Br, or I).
  • heterocyclyl represents a 5-, 6- or 7-membered ring, unless otherwise specified, containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur.
  • the 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds.
  • heterocyclyl also represents a heterocyclic compound having a bridged multicyclic structure in which one or more carbons and/or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group.
  • heterocyclyl also includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one, two, or three carbocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, or another monocyclic heterocyclic ring, e.g., indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, benzofuryl, benzothienyl and the like.
  • fused heterocyclyls include tropanes and 1 ,2,3,5,8,8a-hexahydroindolizine.
  • Heterocyclics include pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, piperidinyl, homopiperidinyl, pyrazinyl, piperazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidiniyl, morpholinyl, thiomorpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, indolyl, quinolinyl, isoquinolinyl,
  • the heterocyclyl group may be optionally substituted with one, two, three, four, or five substituents independently selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is,
  • R A is selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyl, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms; or (15) - C(0)NR B R c , where each of R B and R c is, independently, selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyl, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms; or (15) - C(0)NR B R c , where each of R B and R c is, independently, selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyl, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms
  • heteroaryl represents that subset of heterocyclyls, as defined herein, which are aromatic: i.e., they contain 4n+2 pi electrons within the mono- or multicyclic ring system.
  • the heteroaryl is substituted with, e.g., 1 , 2, 3, or 4 substituent groups independently selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (7) cycloalkyl of three to eight carbons; (8) oxo; (9) hal; (10) alkylsulfonyl of one to six carbon atoms; (1 1 ) thioalkoxy of one to six carbon atoms; (12) aryl; (13) alkaryl; (14) -C
  • hydroxy represents an -OH group.
  • hydroxyalkyl represents an alkyl group, as defined herein, substituted by one or two hydroxy groups, with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group and is exemplified by hydroxymethyl, dihydroxypropyl, and the like.
  • /V-protected amino refers to an amino group, as defined herein, to which is attached an /V-protecting group, as defined herein.
  • V-protecting group represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used /V-protecting groups are disclosed in Greene, Protective Groups in Organic Synthesis, 3 rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference.
  • N-protecting groups include acyl, aroyl, or carbamyl groups, e.g., formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4- bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries, e.g., protected or unprotected D, L or D, L-amino acids, e.g., alanine, leucine, phenylalanine, and the like; sulfonyl groups, e.g., benzenesulfonyl, p- toluenesulfonyl, and the
  • phenylthiocarbonyl, and the like arylalkyi groups, e.g., benzyl, triphenylmethyl, benzyloxymethyl, and the like and silyl groups, e.g., trimethylsilyl, and the like.
  • Preferred N-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc), and
  • six-membered ring represent a saturated or unsaturated aromatic or non- aromatic group having six atoms in a cyclic array, where, unless otherwise specified, five atoms are carbons and the remaining atom is selected from the group consisting of carbon, nitrogen, sulfur, and oxygen.
  • a six-membered ring may be fused to another cyclic group selected from heterocyclyl, heteroaryl, cycloalkyl, and aryl.
  • a six-membered ring may be unsubstituted or substituted with, for example, one, two, three, or four substituents, independently selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (7) cycloalkyl of six to eight carbons; (8) oxo; (9) alkylsulfonyl of one to six carbon atoms; (10) thioalkoxy of one to six carbon atoms; (1 1 ) aryl; (12) alkaryl, where alkylene group is one to six carbon atoms; (13) - C0 2 R A , where R A is selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyi,
  • thioalkoxy represents a chemical substituent of formula -SR, where R is an alkyl group. In some embodiments, the alkyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as described herein for alkyl groups.
  • thiol represents a chemical substituent of formula -SH.
  • Asymmetric or chiral centers may exist in any of the compounds of the present invention.
  • the present invention contemplates the various stereoisomers and mixtures thereof.
  • Individual stereoisomers of compounds of the present invention are prepared synthetically from commercially available starting materials that contain asymmetric or chiral centers or by preparation of mixtures of enantiomeric compounds followed by resolution well known to those of ordinary skill in the art.
  • chiral compounds can be prepared by an asymmetric synthesis that favors the preparation of one enantiomer over the other.
  • a chiral pool synthesis (starting with an enantiomerically pure building block) can be used where the chiral group or center is retained in the intermediate or final product.
  • Enantiomers are designated herein by the symbols "R,” or “S,” depending on the configuration of substituents around the chiral atom.
  • enantiomers are designated as (+) or (-) depending on whether a solution of the enantiomer rotates the plane of polarized light clockwise or counterclockwise, respectively.
  • Geometric isomers may also exist in the compounds of the present invention.
  • the present invention contemplates the various geometric isomers and mixtures thereof resulting from the arrangement of substituents around a carbon-carbon double bond and designates such isomers as of the Z or E configuration, where the term "Z" represents substituents on the same side of the carbon-carbon double bond and the term “E” represents substituents on opposite sides of the carbon-carbon double bond. It is also recognized that for structures in which tautomeric forms are possible, the description of one tautomeric form is equivalent to the description of both, unless otherwise specified.
  • Each position in the compounds of the invention may include elements in their natural isotopic abundance. Alternatively, one or more positions in the compound of the invention may include an element enriched in a naturally occurring or a synthetic isotope.
  • one or more positions of the compound of the invention including hydrogen may be enriched with, e.g. , deuterium or tritium .
  • one or more positions of the compound of the invention including carbon may be enriched with, e.g. , 14 C or 13 C.
  • one or more positions of the compound of the invention including nitrogen may be enriched with, e.g., 15 N.
  • one or more positions of the compound of the invention including oxygen may be enriched with, e.g., 18 0, 17 0, or 15 0.
  • one or more positions of the compound of the invention including fluorine may be enriched with, e.g., 18 F.
  • one or more positions of the compound of the invention including carbon may be enriched with , e.g., 32 S, 33 S, 34 S, 35 S, or 36 S.
  • one or more positions of the compound of the invention including chlorine may be enriched with, e.g., 35 CI, 36 CI, or 37 CI.
  • ⁇ peptide e.g., inhibition of Hsp90
  • ⁇ peptide e.g., inhibition of aggregation of ⁇ peptide
  • tau protein e.g., taurin
  • the increases and decreases related to administration an effective amount of a compound are relative to levels or symptoms, as applicable, in a subject that has not been administered a compound of the invention or relative to the subject prior to administration of a compound of the invention.
  • element refers to a substance consisting of a single type of atoms, that is, each nucleus of each atom of a single element contains the same number of protons.
  • neurodegeneration refers to the progressive loss of structure or function of neurons, including death of neurons.
  • neurodegenerative disease refers to diseases in which neurodegeneration is, at least in part, a cause, symptom, or phenotype.
  • patient refers to any animal (e.g., a mammal, e.g., a human).
  • a subject to be treated according to the methods described herein may be one who has been diagnosed with a neurodegenerative disease, e.g., tauopathy (e.g., Alzheimer's disease), or a proliferative disease as having such a condition or one at risk of developing the condition. Diagnosis may be performed by any method or technique known in the art.
  • a subject to be treated according to the present invention may have been subjected to standard tests or may have been identified, without examination, as one at high risk due to the presence of one or more risk factors, e.g., increased total level of tau protein or an increased level of phosphorylated tau protein in a sample (e.g., cerebrospinal fluid) from the subject in comparison to the levels in a sample from a healthy subject.
  • risk factors e.g., increased total level of tau protein or an increased level of phosphorylated tau protein in a sample (e.g., cerebrospinal fluid) from the subject in comparison to the levels in a sample from a healthy subject.
  • composition represents a composition containing a compound described herein, formulated with a pharmaceutically acceptable excipient, and manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal.
  • Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup) ; for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other formulation described herein.
  • pharmaceutically acceptable excipient or “pharmaceutically acceptable carrier,” as used interchangeably herein, refers to any ingredient other than the compounds described herein (e.g., a vehicle capable of suspending or dissolving the active compound) and having the properties of being nontoxic and non-inflammatory in a patient.
  • Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, or waters of hydration.
  • excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine,
  • BHT butylated hydroxytoluene
  • prodrugs as used herein, represents those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use.
  • pharmaceutically acceptable salt represents those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and 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,
  • salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1 -19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008.
  • the salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.
  • Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate,
  • benzenesulfonate benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, 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, picrate, pivalate
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
  • pharmaceutically acceptable solvate as used herein means a compound as described herein wherein molecules of a suitable solvent are incorporated in the crystal lattice. A suitable solvent is physiologically tolerable at the dosage administered.
  • solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof.
  • suitable solvents are ethanol, water (for example, mono-, di-, and tri- hydrates), /V-methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), ⁇ /, ⁇ /'-dimethylformamide (DMF), ⁇ /, ⁇ /'-dimethylacetamide (DMAC), 1 ,3-dimethyl-2-imidazolidinone (DMEU), 1 ,3-dimethyl-3,4,5,6- tetrahydro-2-(1 H)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like.
  • water is the solvent
  • the molecule is referred to as a "hydrate.”
  • PSA polar surface area of a molecule
  • polar surface area of a molecule, as used interchangeably herein, are defined as the surface sum over all polar atoms.
  • the units of PSA are A 2 (angstrom squared).
  • prevent refers to prophylactic treatment or treatment that prevents one or more symptoms or conditions of a disease, disorder, or conditions described herein.
  • Preventive treatment that includes administration of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, can be acute, short-term, or chronic. The doses administered may be varied during the course of preventative treatment.
  • prodrug represents compounds that are rapidly transformed in vivo to the parent compound of the above formula, for example, by hydrolysis in blood.
  • Prodrugs of the compounds described herein may be conventional esters. Some common esters that have been utilized as prodrugs are phenyl esters, aliphatic (CrCg Or C 8 -C 24 ) esters, cholesterol esters, acyloxymethyl esters, carbamates, and amino acid esters. For example, a compound that contains an OH group may be acylated at this position in its prodrug form.
  • prodrugs of the compounds of the present invention are suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use.
  • proliferative disease refers to both cancer and non-cancer disease.
  • the proliferative disease is one characterized by unregulated proliferation of cells of a certain type (e.g., astrocytes).
  • the tumor cells associated with a proliferative disease e.g., cancer
  • Proliferative diseases to be treated using compounds of the invention and according to the methods of the invention may include glioma, meningioma, pituitary adenoma, nerve sheath tumor (e.g., schwannoma or neurofibroma).
  • Proliferative diseases within the scope of the present invention may be a cancer, e.g., acute myeloid leukemia, gastrointestinal stromal tumor, gastric cancer, lung cancer, lymphoma, melanoma, myeloma, non-small cell lung cancer, renal cancer, small cell lung cancer, blast-phase chronic myelogenous leukemia, leukemia, lymphoproliferative disorder, metastatic melanoma, relapsed multiple myeloma, refractory multiple myeloma, myeloproliferative disorders, pancreatic cancer, small intestine cancer, or solid tumor.
  • a cancer e.g., acute myeloid leukemia, gastrointestinal stromal tumor, gastric cancer, lung cancer, lymphoma, melanoma, myeloma, non-small cell lung cancer, renal cancer, small cell lung cancer, blast-phase chronic myelogenous leukemia, leukemia, lymphoproliferative disorder, metastatic
  • a proliferative disease to be treated using compounds of the invention according to the methods of the invention may include brain tumors (e.g., malignant brain tumors).
  • the brain tumors that may be treated with blood-brain-barrier penetrant compounds of the invention can be glioma or meningioma, in particular, glioma (e.g., glioblastoma), or a malignant version thereof.
  • the cancers that may be treated according to the present invention may also be a cancer that has metastasized to the brain (e.g., lung cancer, breast cancer, melanoma, colon cancer, renal cancer, and thyroid cancer).
  • treatment is an approach for obtaining beneficial or desired results, e.g., clinical results.
  • beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions; diminishment of extent of disease, disorder, or condition; stabilized (i.e., not worsening) state of disease, disorder, or condition; preventing spread of disease, disorder, or condition; delay or slowing the progress of the disease, disorder, or condition; amelioration or palliation of the disease, disorder, or condition; and remission (whether partial or total), whether detectable or undetectable.
  • “Palliating" a disease, disorder, or condition means that the extent and/or undesirable clinical manifestations of the disease, disorder, or condition are lessened and/or time course of the progression is slowed or lengthened, as compared to the extent or time course in the absence of treatment.
  • Figure 1 shows a 500 MHz 1 H NMR spectrum of compound 20 in CDCI 3 .
  • Figure 2 shows a 500 MHz 1 H NMR spectrum of compound 34 in CDCI 3 .
  • Figure 3 shows a 500 MHz 1 H NMR spectrum of compound 36 in CDCI 3 .
  • Figure 4 shows five graphs providing IC 50 data for compounds 20, 36, 37, and 39 and for a known Hsp90 inhibitor, as measured using a fluorescence polarization assay described in Example 2.
  • Figure 5A shows a graph comparing relative association/dissociation rates to the concentration of compound 20.
  • Figure 5B is a photograph of a gel demonstrating increase of the expression of Hsp70 at higher concentrations of compound 20 in a cell based functional assay. Further, Figure 5B shows that expression of Hsp90 remains unchanged relative to variation in the concentration of compound 20.
  • Figure 6A shows a histogram comparing % cell viability in viable cells (VC), cells contacted with compound 20, and cells contacted with a control compound (JNK inhibitor).
  • Figure 6B shows a graph comparing pTau231 levels in viable cells (VC), cells contacted with compound 20, and cells contacted with a control compound (JNK inhibitor).
  • Figure 6C shows a graph comparing pTau396 levels in viable cells (VC), cells contacted with compound 20, and cells contacted with a control compound (JNK inhibitor).
  • Figure 7 shows a graph of the concentration of compound 20 in mouse plasma. The data in this graph excludes the plasma level of compound 20 in animal IRN 12.
  • Figure 8 shows a graph of the concentration of compound 20 in mouse plasma.
  • the data in this graph includes all mouse plasma data points.
  • Figure 9 shows a graph of the concentration of compound 20 in mouse brain tissue. The data in this graph excludes the plasma level of compound 20 in animal IRN 12.
  • Figure 10 shows a graph of the concentration of compound 20 in mouse brain tissue.
  • the data in this graph includes all mouse brain tissue data points.
  • the invention features novel aminopyrimidines and related compounds having Hsp90 inhibitory activity, pharmaceutical compositions containing them, and their medical use (e.g., treatment of a proliferative disease (e.g., cancer) or a neurodegenerative disease (e.g., a tauopathy)).
  • a proliferative disease e.g., cancer
  • a neurodegenerative disease e.g., a tauopathy
  • the compounds of the invention are capable to penetrate blood-brain-barrier. Therefore, medical use of these compounds encompasses diseases and conditions afflicting mammalian (e.g., human) brain.
  • a non-limiting example of a pharmaceutically acceptable salt of a compound of the invention is:
  • the compounds of the invention can be prepared by processes analogous to those established in the art, for example, by the reaction sequence shown in Scheme 1 .
  • the numbering system used for the general schemes does not necessarily correspond to that employed elsewhere in the description or in the claims.
  • one strategy to access compounds of the invention (C) is to utilize standard cross-coupling reactions (e.g., Suzuki coupling, Hiyama coupling, Stille coupling, Negishi coupling, Tamao-Kumada coupling, or Murahashi coupling), where a nucleophile A and an electrophile B are coupled in the presence of a metal salt, e.g., a palladium, copper, iron, or nickel salt (e.g., PdCI 2 , Pd(OAc) 2 , CuBr, Cul, (CuOTf) 2 -toluene complex, Fe(OTf) 3 , FeCI 3 , FeBr 3 , NiCI 2 , or NiBr 2 ).
  • a metal salt e.g., a palladium, copper, iron, or nickel salt
  • ligands e.g., a phosphine (e.g., PPh 3 , P(2-furyl) 3 , P(f-Bu) 3 , dppf, dppb, or BINAP), an /V-heterocyclic carbene (e.g., SIMes or SIPr), or di-pyridine (e.g., 2,2'-bipyridyl or 1 ,10-phenanthroline), may be added to promote the reaction.
  • an organometallic complex e.g., Pd(PPh 3 ) 4 or (dppf)PdCI 2 , may be employed directly with or without additional ligands.
  • Additives e.g., tetrabutylammonium fluoride, LiCI, KOAc, or AgOTf, may be added to minimize dehalogenation or to facilitate the cross-coupling reaction.
  • solvents used in cross-coupling reactions are water, ethanol, acetone, tetrahydrofuran, toluene, 1 ,4-dioxane, and mixtures thereof.
  • conditions and catalysts that can be used to prepare a compound of the invention according to formula C using cross-coupling chemistry, see Miyaura et al., "Cross-Coupling Reactions: A Practical Guide" in
  • a compound of formula A may be an electrophile and have a leaving group X instead of M
  • compound of formula B may be a nucleophile and have a metal or metalloid group M instead of X.
  • MgCI, MgBr, Mgl, Li such as -OMs, -OTf,
  • a compound of formula A may be prepared according any method known in the art, e.g., metal-halogen (e.g., lithium-halogen) exchange (with or without a subsequent addition of boron-based, silicon-based, tin- based, zinc-based, or magnesium-based agents), preparation of Grignard reagent, Sandmeyer reaction, or cross-coupling with di-metalloid agent (e.g., Miyaura borylation reaction).
  • metal-halogen e.g., lithium-halogen
  • a compound of formula B may be prepared according to any method known in the art, e.g., Biginelli reaction followed by oxidation of the resulting 2-aminodihydropyrimidine derivative.
  • Biginelli reaction followed by oxidation of the resulting 2-aminodihydropyrimidine derivative.
  • Scheme 3 a synthetic approach outlines in Scheme 3 can be used to access a compound of formula B.
  • a compound of formula H may undergo condensation with a compound of formula I to give a compound of formula J.
  • the hydroxyl group in the compound of formula K may then be converted to a halogen in the compound of formula L according to any method known in the art, e.g., using dehydrating-halogenating reagents (e.g., POCI 3 , PCI 5 , SOCI 2 , S0 2 CI 2 , and brominating or iodinating variants thereof).
  • dehydrating-halogenating reagents e.g., POCI 3 , PCI 5 , SOCI 2 , S0 2 CI 2 , and brominating or iodinating variants thereof.
  • the hydroxyl group of the compound of formula K may be converted into a pseudohalogen in the compound of formula L using reagents, e.g., Tf 2 0, PhNTf 2 , PhNNf 2 , or P(0)(OR) 2 CI, and, optionally, a base (e.g., Et 3 N, ( Pr) 2 NEt, or pyridine) and/or catalyst (e.g., 4-dimethylaminopyridine).
  • /V-protecting group P may be removed from the compound L before or after the cross coupling shown in Scheme 1 according to methods known in the art (see, e.g., Greene, Protective Groups in Organic Synthesis, 3 rd Edition (John Wiley & Sons, New York, 1999)).
  • halogenation sulphonylation, nitration, formylation, hydrolysis, and coupling procedures.
  • These procedures can be used to introduce a functional group onto the parent molecule (e.g., the nitration or sulphonylation of aromatic rings) or to couple two molecules together (for example to couple an amine to a carboxylic acid to afford an amide; or to form a carbon-carbon bond between two heterocycles).
  • alcohol or phenol groups can be converted to ether groups by coupling a phenol with an alcohol in a solvent, e.g., tetrahydrofuran in the presence of a phosphine (e.g., triphenylphosphine) and a dehydrating agent (e.g., diethyl-, diisopropyl-, or dimethylazodicarboxylate).
  • a phosphine e.g., triphenylphosphine
  • a dehydrating agent e.g., diethyl-, diisopropyl-, or dimethylazodicarboxylate
  • ether groups can be prepared by deprotonation of an alcohol, using a suitable base (e.g., sodium hydride) followed by the addition of an alkylating agent (e.g., an alkyl halide or an alkylsulphonate).
  • a primary or secondary amine can be alkylated using a reductive alkylation process.
  • the amine can be treated with an aldehyde and a borohydride (e.g., sodium
  • a solvent e.g., a halogenated hydrocarbon, for example, dichloromethane, or an alcohol, for example, ethanol
  • an acid e.g., acetic acid
  • -OH groups may be generated from the corresponding ester, acid, acid chloride or aldehyde by reduction with a suitable reducing agent, e.g., a complex metal hydride, e.g., lithium aluminium hydride in a solvent (e.g., tetrahydrofuran).
  • a suitable reducing agent e.g., a complex metal hydride, e.g., lithium aluminium hydride in a solvent (e.g., tetrahydrofuran).
  • hydroxy groups can be converted into leaving groups, e.g., halogen atoms or sulphonyloxy groups (e.g., alkylsulphonyloxy, e.g.,
  • trifluoromethylsulphonyloxy or arylsuphonyl, e.g., p-toluenesulphonyloxy
  • arylsuphonyl e.g., p-toluenesulphonyloxy
  • an aliphatic alcohol can be reacted with thionyl chloride in a halogenated hydrocarbon (e.g., dichloromethane) to afford the corresponding alkylchloride.
  • a base e.g., triethylamine
  • ester groups can be converted to the corresponding carboxylic acid by acid- or base- catalysed hydrolysis depending on the nature of the ester group.
  • Acid catalysed hydrolysis can be achieved by treatment with an organic or inorganic acid (e.g., trifluoroacetic acid in an aqueous solvent, or a mineral acid, e.g., hydrochloric acid in a solvent, e.g., dioxan).
  • Base catalysed hydrolysis can be achieved by treatment with an alkali metal hydroxide (e.g., lithium hydroxide in an aqueous alcohol, e.g. methanol).
  • aromatic halogen substituents in the compounds may be subjected to halogen-metal exchange by treatment with a base (e.g., a lithium base, e.g., n-butyl or f-butyl lithium) optionally at a low temperature (e.g., -78°C) in a solvent (e.g., tetrahydrofuran) and the mixture may then quenched with an electrophile to introduce a desired substituent.
  • a base e.g., a lithium base, e.g., n-butyl or f-butyl lithium
  • a solvent e.g., tetrahydrofuran
  • Aromatic halogen substituents can also be subjected to palladium catalysed reactions to introduce groups, e.g., carboxylic acids, esters, cyano, or amino substituents.
  • aromatic halogen substituents in the compounds may participate in a range of metal catalysed reactions to introduce alternative functional groups, e.g., amines, amides, ethers, thiols, aryl groups, or heteroaryl groups.
  • aldehyde groups can be prepared by oxidation of the corresponding alcohol using conditions well known to those skilled in the art.
  • an alcohol can be treated with an oxidising agent (e.g., the Dess-Martin reagent) in a solvent (e.g., a halogenated hydrocarbon, for example dichloromethane).
  • oxidising agent e.g., the Dess-Martin reagent
  • solvent e.g., a halogenated hydrocarbon, for example dichloromethane
  • Alternative oxidising conditions can be used, e.g., treatment with oxalyl chloride and an activating amount of dimethylsulphoxide and subsequent quenching by the addition of an amine (e.g., triethylamine).
  • Such a reaction can be carried out in an appropriate solvent (e.g., a halogenated hydrocarbon, for example dichloromethane) and under appropriate conditions (e.g., cooling below room temperature, e.g., to -78°C followed by warming to room temperature).
  • a halogenated hydrocarbon for example dichloromethane
  • sulphur atoms can be oxidised to the corresponding sulphoxide or sulphone using an oxidising agent (e.g., a peroxy acid, e.g., 3-chloroperoxybenzoic acid) in an inert solvent (e.g., a halogenated hydrocarbon, e.g., dichloromethane) at around ambient temperature.
  • an oxidising agent e.g., a peroxy acid, e.g., 3-chloroperoxybenzoic acid
  • an inert solvent e.g., a halogenated hydrocarbon, e.g., dichloromethan
  • Particular reduction approaches include the removal of oxygen atoms from particular functional groups, saturation (or partial saturation) of unsaturated compounds including aromatic rings.
  • primary alcohols can be generated from the corresponding ester or aldehyde by reduction, using a metal hydride (e.g., lithium aluminium hydride or sodium borohydride in a solvent, e.g., methanol).
  • a metal hydride e.g., lithium aluminium hydride or sodium borohydride in a solvent, e.g., methanol
  • -OH groups can be generated from the corresponding carboxylic acid by reduction, using a metal hydride (e.g., lithium aluminium hydride in a solvent, e.g., tetrahydrofuran).
  • a nitro group may be reduced to an amine by catalytic hydrogenation in the presence of a metal catalyst (e.g., palladium on a solid support, e.g., carbon) in a solvent (e.g., an ether, e.g., tetrahydrofuran, or an alcohol, e.g., methanol), or by chemical reduction using a metal (e.g., tin or iron) in the presence of an acid (e.g., hydrochloric acid).
  • a metal catalyst e.g., palladium on a solid support, e.g., carbon
  • a solvent e.g., an ether, e.g., tetrahydrofuran, or an alcohol, e.g., methanol
  • an acid e.g., hydrochloric acid
  • an amine can be obtained by reduction of a nitrile, e.g., by catalytic hydrogenation in the presence of a metal catalyst (e.g., palladium on a solid support, e.g., carbon), or Raney nickel in a solvent (e.g., tetrahydrofuran) and under suitable conditions (e.g., cooling to below room temperature, e.g., to -78°C, or heating, e.g., to reflux).
  • a metal catalyst e.g., palladium on a solid support, e.g., carbon
  • Raney nickel e.g., tetrahydrofuran
  • compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo.
  • Pharmaceutical compositions typically include a compound as described herein and a pharmaceutically acceptable excipient.
  • the compounds described herein can also be used in the form of the free base, in the form of salts, zwitterions, solvates, or as prodrugs, or pharmaceutical compositions thereof. All forms are within the scope of the invention.
  • the compounds, salts, zwitterions, solvates, prodrugs, or pharmaceutical compositions thereof may be administered to a patient in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art.
  • the compounds used in the methods described herein may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump, or transdermal administration, and the pharmaceutical compositions formulated accordingly.
  • Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal, and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.
  • compositions for use in accordance with the present invention thus can be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries that facilitate processing of compounds of Formula (I), (la), (lb), (Va), or (Vb) into preparations which can be used pharmaceutically.
  • compositions which can contain one or more
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container.
  • an excipient serves as a diluent, it can be a solid, semisolid, or liquid material (e.g., normal saline), which acts as a vehicle, carrier or medium for the active ingredient.
  • the compositions can be in the form of tablets, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, and soft and hard gelatin capsules.
  • the type of diluent can vary depending upon the intended route of administration.
  • the resulting compositions can include additional agents, e.g., preservatives.
  • the excipient or carrier is selected on the basis of the mode and route of administration. Suitable pharmaceutical carriers, as well as pharmaceutical necessities for use in pharmaceutical formulations, are described in Remington: The Science and Practice of Pharmacy, 21 st Ed., Gennaro, Ed., Lippencott Williams & Wilkins (2005), a well-known reference text in this field, and in the USP/NF (United States Pharmacopeia and the National Formulary).
  • excipients examples include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents, e.g., talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents, e.g., methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • lubricating agents e.g., talc, magnesium stearate, and mineral oil
  • wetting agents emulsifying and suspending agents
  • preserving agents e.g., methyl- and propylhydroxy-benzoates
  • sweetening agents and flavoring agents.
  • compositions can be manufactured in a conventional manner, e.g., by
  • the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
  • the dosage of the compound used in the methods described herein, or pharmaceutically acceptable salts or prodrugs thereof, or pharmaceutical compositions thereof can vary depending on many factors, e.g., the pharmacodynamic properties of the compound; the mode of administration; the age, health, and weight of the recipient; the nature and extent of the symptoms; the frequency of the treatment, and the type of concurrent treatment, if any; and the clearance rate of the compound in the animal to be treated.
  • One of skill in the art can determine the appropriate dosage based on the above factors.
  • the compounds used in the methods described herein may be administered initially in a suitable dosage that may be adjusted as required, depending on the clinical response.
  • a suitable daily dose of a compound of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • a compound of the invention may be administered to the patient in a single dose or in multiple doses. When multiple doses are administered, the doses may be separated from one another by, for example, 1 - 24 hours, 1 -7 days, 1 -4 weeks, or 1 -12 months.
  • the compound may be administered according to a schedule or the compound may be administered without a predetermined schedule.
  • An active compound may be administered, for example, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 1 0, 1 1 , or 12 times per day, every 2 nd , 3 rd , 4 th , 5 th , or 6 th day, 1 , 2, 3, 4, 5, 6, or 7 times per week, 1 , 2, 3, 4, 5, or 6 times per month, or 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , or 12 times per year. It is to be understood that, for any particular subject, specific dosage regimes should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions.
  • an effective amount of a compound of the invention may be, for example, a total daily dosage of, e.g., between 0.05 mg and 3000 mg of any of the compounds described herein.
  • the dosage amount can be calculated using the body weight of the patient.
  • Such dose ranges may include, for example, between 10-1 000 mg (e.g., 50-800 mg).
  • 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1 000 mg of the compound is administered.
  • the time period during which multiple doses of a compound of the invention are administered to a patient can vary.
  • doses of the compounds of the invention are administered to a patient over a time period that is 1 -7 days; 1 -12 weeks; or 1 -3 months.
  • the compounds are administered to the patient over a time period that is, for example, 4-1 1 months or 1 -30 years.
  • the compounds are administered to a patient at the onset of symptoms.
  • the amount of compound that is administered may vary during the time period of administration. When a compound is administered daily, administration may occur, for example, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , or 12 times per day.
  • a compound identified as capable of treating any of the conditions described herein, using any of the methods described herein, may be administered to patients or animals with a pharmaceutically- acceptable diluent, carrier, or excipient, in unit dosage form.
  • the chemical compounds for use in such therapies may be produced and isolated by any standard technique known to those in the field of medicinal chemistry.
  • Conventional pharmaceutical practice may be employed to provide suitable formulations or compositions to administer the identified compound to patients suffering from a disease in which necrosis occurs. Administration may begin before the patient is symptomatic.
  • Exemplary routes of administration of the compounds e.g., the compounds having Formula (I), (la), (lb), (Va) or (Vb)), or pharmaceutical compositions thereof, used in the present invention include
  • oral dosage forms can be, for example, in the form of tablets, capsules, a liquid solution or suspension, a powder, or liquid or solid crystals, which contain the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients.
  • excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, pregelatinized starch, microcrystalline cellulose, magnesium aluminum silicate,
  • inert diluents or fillers e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lac
  • lubricating agents e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc.
  • Other pharmaceutically acceptable excipients can be colorants, flavoring agents, plasticizers, humectants, buffering agents, and the like.
  • Formulations for oral administration may also be presented as chewable tablets, as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose,
  • Oil medium for example, peanut oil, liquid paraffin, or olive oil.
  • Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.
  • Controlled release compositions for oral use may be constructed to release the active drug by controlling the dissolution and/or the diffusion of the active drug substance. Any of a number of strategies can be pursued in order to obtain controlled release and the targeted plasma concentration versus time profile.
  • controlled release is obtained by appropriate selection of various formulation parameters and ingredients, including, e.g., various types of controlled release compositions and coatings. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, nanoparticles, patches, and liposomes.
  • compositions include biodegradable, pH, and/or temperature-sensitive polymer coatings.
  • Dissolution or diffusion controlled release can be achieved by appropriate coating of a tablet, capsule, pellet, or granulate formulation of compounds, or by incorporating the compound into an appropriate matrix.
  • a controlled release coating may include one or more of the coating substances mentioned above and/or, e.g., shellac, beeswax, glycowax, castor wax, carnauba wax, stearyl alcohol, glyceryl
  • the matrix material may also include, e.g., hydrated methylcellulose, carnauba wax and stearyl alcohol, carbopol 934, silicone, glyceryl tristearate, methyl acrylate-methyl methacrylate, polyvinyl chloride, polyethylene, and/or halogenated fluorocarbon.
  • liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils, e.g., cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • aqueous solutions suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils, e.g., cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • Dosages for buccal or sublingual administration typically are 0.1 to 500 mg per single dose as required.
  • the physician determines the actual dosing regimen which is most suitable for an individual patient, and the dosage varies with the age, weight, and response of the particular patient.
  • the above dosages are exemplary of the average case, but individual instances exist wherein higher or lower dosages are merited, and such are within the scope of this invention.
  • compositions may take the form of tablets, lozenges, etc. formulated in a conventional manner.
  • Liquid drug formulations suitable for use with nebulizers and liquid spray devices and electrohydrodynamic (EHD) aerosol devices will typically include a compound of the invention with a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier is a liquid, e.g., alcohol, water, polyethylene glycol, or a perfluorocarbon.
  • another material may be added to alter the aerosol properties of the solution or suspension of compounds of the invention. Desirably, this material is liquid, e.g., an alcohol, glycol, polyglycol, or a fatty acid.
  • compositions for nasal administration also may conveniently be formulated as aerosols, drops, gels, and powders.
  • the formulations may be provided in a single or multidose form.
  • dosing may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension.
  • this may be achieved, for example, by means of a metering atomizing spray pump.
  • the compounds may further be formulated for aerosol administration, particularly to the respiratory tract by inhalation and including intranasal administration.
  • the compound will generally have a small particle size for example on the order of five (5) microns or less. Such a particle size may be obtained by means known in the art, for example by micronization.
  • the active ingredient is provided in a pressurized pack with a suitable propellant, e.g., a chlorofluorocarbon (CFC), for example, dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, or carbon dioxide, or other suitable gas.
  • a suitable propellant e.g., a chlorofluorocarbon (CFC), for example, dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, or carbon dioxide, or other suitable gas.
  • the aerosol may conveniently also contain a surfactant,
  • the dose of drug may be controlled by a metered valve.
  • the active ingredients may be provided in a form of a dry powder, e.g., a powder mix of the compound in a suitable powder base, e.g., lactose, starch, and starch derivatives, e.g., hydroxypropylmethyl cellulose, and polyvinylpyrrolidine (PVP).
  • the powder carrier will form a gel in the nasal cavity.
  • the powder composition may be presented in unit dose form for example in capsules or cartridges of e.g., gelatin or blister packs from which the powder may be administered by means of an inhaler.
  • Aerosol formulations typically include a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device.
  • the sealed container may be a unitary dispensing device, e.g., a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal after use.
  • the dosage form comprises an aerosol dispenser, it will contain a propellant, which can be a compressed gas, e.g., compressed air or an organic propellant, e.g.,
  • the aerosol dosage forms can also take the form of a pump-atomizer.
  • compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the compounds of the invention may be dissolved or suspended in a parenterally acceptable liquid vehicle.
  • acceptable vehicles and solvents water, water adjusted to a suitable pH by addition of an appropriate amount of hydrochloric acid, sodium hydroxide or a suitable buffer, 1 ,3-butanediol, Ringer's solution and isotonic sodium chloride solution.
  • the aqueous formulation may also contain one or more preservatives, for example, methyl, ethyl or n-propyl p-hydroxybenzoate. Additional information regarding parenteral formulations can be found, for example, in the United States Pharmacopeia-National
  • the parenteral formulation can be any of the five general types of preparations identified by the USP-NF as suitable for parenteral administration:
  • a liquid preparation that is a drug substance (e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)), or a solution thereof;
  • a drug substance e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)
  • drug for Injection the drug substance (e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)) as a dry solid that will be combined with the appropriate sterile vehicle for parenteral administration as a drug injection ;
  • the drug substance e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)
  • a liquid preparation of the drug substance e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)
  • a suitable emulsion medium e.g., a liquid preparation of the drug substance (e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)) that is dissolved or dispersed in a suitable emulsion medium;
  • a liquid preparation of the drug substance e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb) suspended in a suitable liquid medium;
  • exemplary formulations for parenteral administration include solutions of the compound prepared in water suitably mixed with a surfactant, e.g., hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • Formulations for parenteral administration may, for example, contain excipients, sterile water, or saline, polyalkylene glycols, e.g., polyethylene glycol, oils of vegetable origin, or hydrogenated napthalenes.
  • Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene- polyoxypropylene copolymers may be used to control the release of the compounds.
  • Other potentially useful parenteral delivery systems for compounds include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
  • Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene- 9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel.
  • the parenteral formulation can be formulated for prompt release or for sustained/extended release of the compound.
  • exemplary formulations for parenteral release of the compound include: aqueous solutions, powders for reconstitution, cosolvent solutions, oil/water emulsions, suspensions, oil-based solutions, liposomes, microspheres, and polymeric gels.
  • the compounds and compositions described herein can be used in the treatment of conditions and disorders in which Hsp90 has been implicated, e.g., cell proliferative disorders, e.g., cancers, neurodegenerative diseases, e.g., tauopathies (e.g., Alzheimer's disease), and infectious diseases.
  • cell proliferative disorders e.g., cancers
  • neurodegenerative diseases e.g., tauopathies (e.g., Alzheimer's disease)
  • infectious diseases e.g., cell proliferative disorders, e.g., cancers, neurodegenerative diseases, e.g., tauopathies (e.g., Alzheimer's disease), and infectious diseases.
  • Hsp90 has emerged as a key therapeutic target for cancer therapy due to the involvement of this multichaperone complex in various pathogenic cellular processes.
  • Hsp90 client proteins include those implicated in: acute myeloid leukemia (Flt-3), breast cancer (HER2), chronic lymphoid leukemia (Zap70), chronic myeloid leukemia (Bcr-Abl or mBcr-Abl), gastrointestinal stromal tumor (c-Kit), gastric cancer (c- Met), glioblastoma (mutant EGFR or c-Met), lung cancer (c-Met), lymphoma (NMP-ALK), melanoma (Raf- 1 /mutant BRAF), myeloma (IGF-1 R/Akt), non-small cell lung cancer (mutant EGFR), renal cancer, (HIF- 1 a), and small cell lung cancer (Akt).
  • Flt-3 acute myeloid leukemia
  • HER2 breast cancer
  • Zap70 chronic my
  • Brain tumors that may be treated using compounds of the invention include glioma or meningioma, in particular, glioma (e.g., glioblastoma) or neuroblastoma.
  • the brain tumors (e.g., brain cancers) that may be treated using compounds of the invention according to the methods of the invention may include primary tumors (those tumors that originated in the brain) and metastatic tumors (those tumors that originated in tissues other than brain tissues and spread to the brain).
  • Still other cell proliferative disorders that may be treated by the inhibition of Hsp90 include: blast-phase chronic myelogenous leukemia, leukemia,
  • lymphoproliferative disorder metastatic melanoma, multiple myeloma (e.g., relapsed or refractory multiple myeloma), myeloproliferative disorders, pancreatic cancer, small intestine cancer, and solid tumor.
  • multiple myeloma e.g., relapsed or refractory multiple myeloma
  • myeloproliferative disorders pancreatic cancer, small intestine cancer, and solid tumor.
  • cancer cells have been shown to be more sensitive to Hsp90 inhibition than non-pathogenic cells. Accordingly, the compounds described herein may be useful treatments for cell proliferative disorders.
  • Hsp90 have been implicated in neurodegenerative disorders.
  • tauopathies which are conditions characterized by accumulation of abnormal Tauproteins (e.g., hyperphosphorylated and aggregated Tau).
  • AD Alzheimer's disease
  • argyrophilic grain disease amyotrophic lateral sclerosis
  • corticobasal degeneration dementia pugislistica, Down's syndrome
  • familial British dementia familial British dementia
  • frontal lobe degeneration (dementia lacking distinctive histopathological features)
  • chronic traumatic encephalopathy traumatic brain injury
  • frontotemporal dementia e.g., fronto-temporal dementia with parkinsonism linked to chromosome 17 (FTDP-17)
  • hippocampal tauopathy in cerebral aging myotonic dystrophy of type I , Niemann-Pick disease of type C, Parkinson's disease (e.
  • the compounds described herein may be useful in treating a neurodegenerative disorder, e.g., tauopathy (e.g., tauopathy),
  • Hsp90 has emerged as a therapeutic target for treating infectious diseases, e.g., viral infections, fungal infections, and bacterial infections. Many pathogens (e.g., viruses, fungi, and bacteria) rely on Hsp90-dependent processes (see, e.g., Geller et al., Biochim. Biophys. Acta - Mol. Cell Res., 1823:698-706, 2012; the disclosure of which is incorporate herein in its entirety). Thus, inhibition of Hsp90 provides a therapeutic benefit to a patient afflicted with an infection that relies on the activity of Hsp90. For example, an Hsp90 inhibitor (geldanamycin) was shown to delay the growth of influenza virus in cell culture.
  • Herpesviridae e.g., herpes simplex virus-1 , herpes simplex virus-2, herpes herpesvirus-5, Kaposi's sarcoma-associated herpesvirus, varicella zoster virus, or Epstein-Barr virus
  • Polyomaviridae e.g., SV40
  • Poxviridae e.g., vaccinia virus
  • Reoviridae e.g., rotavirus
  • Birnaviridae e.g., infectious bursal disease virus
  • Picornaviridae e.g., poliovirus, rhinovirus, or coxsackievirus
  • Flaviviridae e.g., hepatitis C virus or dengue virus
  • Arenaviridae e.g., lymphocytic choriomeningitis virus
  • Hepeviridae e.g., Hepatitis E virus
  • Rhabdoviridae e.g., vesicular stomatitis virus
  • Paramoxyviridae e.g., human parainfluenza virus 2, human parainfluenza virus 3, SV5, SV41 , measles virus, or Sendai virus
  • Bunyaviridae e.g., La Crosse virus
  • Orthomoxyviridae e.g., influenza A virus
  • Filoviridae e.g., Ebola virus
  • Retroviridae e.g., HTLV1 or HIV1
  • Hsp90 inhibitors have also been used in vivo for the treatment of fungal infectious diseases, e.g., treatment of Candida albicans, Aspergillus fumigates, or Pneumocystis jiroveci. Moreover, Hsp90 inhibitors are also useful in the treatment of bacterial infections, e.g., mycobacteria, anthrax, or bacterial pneumonia. A discussion of the diseases that may be treated with Hsp90 inhibitors is provided in the U.S. Patent Application Publication
  • the compounds of the invention may be used according to the method of the invention to treat infectious diseases, e.g., viral infections, fungal infections, or bacterial infections.
  • Hsp90 has been shown to play a role in antigen presentation, activation of lymphocytes, macrophages, maturation of dendritic cells, and in the enhanceosome mediated induction of inflammation. Hsp90 inhibition is associated with blockage of components of inflammation, e.g., reduction of cytokine and NO production, as well as blockage of NFKB nuclear translocation. Further, considerable body of work indicates that chaperones, such as Hsp90, may be capable of inducing the production of proinflammatory cytokines by the monocyte-macrophage system and the activation and maturation of dendritic cells via the TLR2-and 4-signal transduction pathways. Thus, Hsp90 apparently can function as a potent activator of the innate immune system.
  • Hsp90 elevated levels were detected in the serum of systemic lupus erythematosus patients. Autoantibodies and cells reactive to Hsp have been detected in patients with rheumatoid arthritis. Antiinflammatory effect of inhibiting Hsp90 was also observed to reduce airway inflammation in murine model of asthma.
  • the compounds of the invention may be applicable to the treatment of inflammatory or autoimmune diseases in a patient. Moreover, anti-inflammatory effect of Hsp90 inhibition can have therapeutic application in the treatment of allergies. Thus, the compounds of the invention may be used in the treatment of allergy.
  • Hsp90 has recently been implicated in etiology of cardiovascular disorders, such as atherosclerosis and cardiomyopathy.
  • cardiovascular diseases e.g., atherosclerosis or cardiomyopathy.
  • kits containing (i) a pharmaceutical composition of the invention, and (ii) instructions for use of the pharmaceutical composition to treat a disorder in a mammal caused by the action of Hsp90, e.g., a neurodegenerative disorder, a proliferative disorder, or an infectious disease, as described herein.
  • Kits of the invention may include instructions explaining how a practitioner (e.g., a physician, a nurse, a care-giver, or a patient) may administer the composition contained therein.
  • the pharmaceutical composition within the kit of the invention may be provided in a container (e.g., a bottle, an ampule, a tube, or a blister pack).
  • kits may also include additional components, e.g., instructions or administration schedules for a patient suffering from a neurodegenerative disease or a proliferative disease, and optionally, a device(s) for administering the pharmaceutical composition (e.g., a syringe).
  • additional components e.g., instructions or administration schedules for a patient suffering from a neurodegenerative disease or a proliferative disease
  • a device(s) for administering the pharmaceutical composition e.g., a syringe
  • Compounds 34 and 36 were prepared according to methods known in the art, e.g., those described herein.
  • the 1 H NMR spectra (CDCI 3 ) for compounds 34 and 36 are provided in Figures 2 and 3, respectively.
  • Compounds 40-48 can be prepared according to methods known in the art, e.g., those described herein.
  • reaction was quenched by addition of 1 N NaOH aq. (1 mL) and stirred at rt for 1 h.
  • the reaction was taken up in DCM (10 mL) and washed with sat. NaHC0 3 aq. (1 0 mL).
  • the organic layer was dried over Na 2 S0 4 , filtered, and volatiles were evaporated.
  • the residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product was obtained in 66 % yield as a yellow solid.
  • the reaction was taken up in DCM (10 mL) and washed with sat. NaHC0 3 aq. (10 mL). The organic layer was dried over Na 2 S0 4 , filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product was obtained in 81 % yield as a yellow solid.
  • Step 1 Synthesis of precursor 2,5-dioxopyrrolidin-1-yl 2-amino-4-(5,7-dichloro-2,3- dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidine-6-carboxylate.
  • the filtrand (57 mg, 0.15 mmol) was dissolved in dry DMF (1 mL), cooled to 0 °C, and to the reaction vessel were added N-hydroxysuccinimide (23 mg, 0.2 mmol) and 1 -ethyl-3-(3- dimethylaminopropyl)carbodiimide HCI (38 mg, 0.2 mmol).
  • the reaction was left to stir warming to rt over 8h.
  • the solution was taken up into DCM (20 mL) and washed with sat. NH 4 CI aq. (3 ⁇ 20 mL).
  • the organic layer was dried over Na 2 S0 4 , filtered, and volatiles were evaporated.
  • Hsp90 inhibitory activity of the compounds of the invention was assessed using a fluorescence polarization (FP) assay using FITC-labeled geldanamycin and truncated alpha-Hsp90 protein.
  • FP fluorescence polarization
  • Hsp90 inhibitory activity of the compounds of the invention was assessed using a robust and reproducible assay based on the AlphaLISA format was developed (PerkinElmer, Inc., Waltham, MA; the format of the assay is described in, e.g., ELISA to AlphaLISA Conversion Guide, PerkinElmer, Inc., published in August, 2012).
  • This assay employs biotinylated geldanamycin, His-tagged-Hsp90, and Ni +2 coated beads. This assay is homogenous and miniaturized to 384-well plates. Measurement of binding activity was performed on an Envision reader.
  • Hsp90 cell based functional assays Cells were treated with compounds of the invention for 24 h and then lysed in a buffer containing NP-40, orthovanadate, and protease inhibitors. Western blots were performed with antibodies specific to Hsp70, Hsp90, or actin (as control).

Abstract

The present invention relates to 2-amino-4-arylpyrimidine and 2-amino-4-aryltriazine compounds as inhibitors of heat shock protein 90 family of chaperone proteins. The invention also features pharmaceutical compositions and kits that include the compounds and compositions of the invention. The invention further relates to the medical use of these compounds and compositions for the treatment of a disorder in a subject. For example, the disorder is a neurodegenerative disease.

Description

PYRIMIDINE COMPOUNDS AND METHODS USING THE SAME
Statement as to Federally Funded Research
This invention was made with government support under Grant No. STTR 1 R41 G042205-01 awarded by the National Institutes of Health. The government has certain rights in the invention.
Field of the Invention
In general, the present invention relates to pyrimidine-based small molecule inhibitors of heat shock protein 90 (Hsp90) and pharmaceutical compositions thereof. The invention further relates to methods of treatment of a subject having a neurodegenerative disease.
Background of the Invention
Hsp90 proteins are implicated in stabilizing protein conformations, maintaining the function of many cell- signaling proteins, and ATPase activity. Hsp90 activity is also required for the proper folding, stabilization, activation, and localization of oncoproteins involved in tumor progression. The /V-terminus ATP binding domain is responsible for the ATPase activity of this protein: this adenine nucleotide binding pocket is highly conserved among all Hsp90 proteins from bacteria to mammals but is not present in other chaperones. Hsp90 protein has emerged as an important target in cancer treatment, as many Hsp90 client proteins themselves were identified as targets for cancer therapies. The exemplary Hsp90 client proteins that are associated with cancer include HER2 (breast cancer), Raf-1/mutant BRAF (melanoma), Mutant EGFR (non-small cell lung cancer, glioblastoma), c-Kit (GIST), c-Met (gastric, lung, glioblastoma), HIF-1 a (renal cancer), Zap70 (chronic lymphocytic leukemia), Bcr-Abl (chronic myelogenous leukemia), mBcr-Abl (chronic myelogenous leukemia), Flt-3 (acute myeloid leukemia), IGF-1 R/Akt (myeloma), NMP-ALK
(lymphoma), and Akt (small cell lung cancer). Overexpression of mutated Hsp90 client or amplification of its clients, such as HER2, leads to the increased dependency of tumor cells on Hsp90 chaperone function. Accordingly, Hsp90 provides a compelling target for treating different classes of tumors. Increased levels of Hsp90 have also been implicated in neurodegenerative disorders, including
Alzheimer's, Parkinson's, and Huntington's disease, and tauopathies. Tauopathies are
neurodegenerative diseases characterized by tau protein abnormalities, which then result in the accumulation of hyperphosphorylated and aggregated tau protein. It has been proposed that hyperphosphorylated tau in Alzheimer's disease is a pathogenic process caused by aberrant activation of kinases, particularly cdk5 and GSK β3. Studies have shown that Hsp90 stabilizes p35, an activator of cdk5, leading to increased tau phosphorylation. It has also been shown that Hsp90 inhibition activates heat shock factor 1 (HSF1 ), which in turn increases the expression of Hsp70. Increased expression of Hsp70 promotes tau solubility and binding to microtubules, inhibits Αβ peptide aggregation, and enhances Αβ peptide degradation.
Hsp90 has also emerged as a target for treating viral, fungal, and bacterial infections. For example, an Hsp90 inhibitor (geldanamycin) has been shown to delay the growth of influenza virus in cell culture. Other viruses that rely on Hsp90 dependent processes include those belonging to the families:
Herpesviridae (e.g., herpes simplex virus-1 , herpes simplex virus-2, herpes herpesvirus-5, Kaposi's sarcoma-associated herpesvirus, varicella zoster virus, or Epstein-Barr virus), Polyomaviridae (e.g., SV40), Poxviridae (e.g., vaccinia virus), Reoviridae (e.g., rotavirus), Birnaviridae (e.g., infectious bursal disease virus), picornaviridae (e.g., poliovirus, rhinovirus, or coxsackievirus), flaviviridae (e.g., hepatitis C virus or dengue virus), arenaviridae (e.g., lymphocytic choriomeningitis virus), Hepeviridae (e.g., Hepatitis E virus), Rhabdoviridae (e.g., vesicular stomatitis virus), Paramoxyviridae (e.g., human parainfluenza virus 2, human parainfluenza virus 3, SV5, SV41 , measles virus, or Sendai virus), Bunyaviridae (e.g., La Crosse virus), Orthomoxyviridae (e.g., influenza A virus), Filoviridae (e.g., Ebola virus), Retroviridae (e.g., HTLV1 or HIV1 ), and Hepadnaviridae (e.g., hepatitis B virus). Hsp90 inhibitors have also been used in vivo for the treatment of fungal infectious diseases, e.g., treatment of Candida albicans, Aspergillus fumigates, or Pneumocystis jiroveci. Moreover, Hsp90 inhibitors are also useful in the treatment of bacterial infections, e.g., mycobacteria, anthrax, or bacterial pneumonia.
In view of the above, inhibitors of Hsp90 represent beneficial therapeutics for the treatment of disorders, e.g., cancer, neurodegenerative diseases, and infectious diseases.
Summary of the Invention
In one aspect, the invention features a compound according to formula (I):
Figure imgf000004_0001
(I), or a pharmaceutically acceptable salt thereof,
where
Z1 is -OR7, -N(R10)R7, -SR7, or -C(R10)(R11)R7;
Z2 is -N= or -C(R3)=;
each R1 and R2 is, independently, H or optionally substituted Ci-3 alkyl (e.g., Ci-3 acyl);
R3 is H, halogen, cyano, optionally substituted Ci-6 alkyl, optionally substituted Ci-3 alkoxy, or optionally substituted amino, and R4 is halogen, cyano, optionally substituted C1-6 alkyl, optionally substituted C1-3 alkoxy, optionally substituted amino, optionally substituted C1-6 thioalkoxy, or optionally substituted C6-io aryl, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising from one to three heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur, where the nitrogen is optionally substituted with R9;
each R5 and R6 is, independently, H, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, halogen, or CN;
R7 is optionally substituted C1-3 alkyl, optionally substituted C1-3 alkcycloalkyl, optionally substituted C1-3 alkheterocyclyl, or optionally substituted C1-3 alkaryl, and R8 is H; or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- , six-, or seven- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur; R9 is H, optionally substituted C1 -3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2.9 heteroaryl, optionally substituted C2.9 heterocyclyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl;
R10 is H, optionally substituted Ci-3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2.9 heteroaryl, optionally substituted C2.9 heterocyclyl, optionally substituted C1-3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1 -3 alkaryl, and R11 is H, optionally substituted C1-3 alkyl, or R10 and R1 1 combine to form =0 or =S; and
Rm is H, halogen, cyano, optionally substituted Ci-4 alkyl (e.g., Ci-4 acyl), or optionally substituted Ci-3 alkoxy.
In particular embodiments of formula (I), Rm is H (e.g., the compound of formula (I) has the following structure:
Figure imgf000005_0001
(I), or a pharmaceutically acceptable salt thereof).
In certain embodiments of formula (I), when Z2 is CR3,each of R1 and R2 is H, R3 is H, R4 is methyl or halogen (e.g., chloro), and each of R5 and R6 is halogen (e.g., chloro),
Z1 is not methoxy.
In certain embodiments of formula (I), when Z2 is CR3, R3 is H, R4 is methyl or halogen (e.g., chloro), each of R5 and R6 is halogen (e.g., chloro),
Z2 is not unsubstituted Ci-3 alkoxy.
In particular embodiments of formula (I), when Z2 is N, R3 is H, R4 is optionally substituted Ci-6 thioalkoxy, and each of R5 and R6 is halogen (e.g., chloro),
Z1 is not cyanomethoxy or aminomethoxy.
In other embodiments of formula (I), when Z2 is N, R3 is H, each of R5 and R6 is halogen (e.g., chloro), R4 is substituted Ci-6 thioalkoxy,
Z1 is not cyanomethoxy or aminomethoxy.
In some embodiments of formula (I), when Z2 is N, R3 is H, R4 is optionally substituted Ci-6 thioalkoxy, Z1 is not cyanomethoxy or aminomethoxy.
In certain embodiments of formula (I), when Z2 is N, R3 is H, R4 is substituted C1 -6 thioalkoxy,
Z1 is not cyanomethoxy or aminomethoxy. In further embodiments of formula (I), when Z2 is N, R3 is H, R4 is substituted thioalkoxy,
Z1 is not substituted Ci alkoxy. In particular embodiments of formula (I), when Z2 is N, R3 is H, R4 is substituted thioalkoxy,
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is methyl, dialkylaminoethyl, optionally substituted C1 -3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1-3 alkaryl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In other embodiments of formula (I), when Z2 is CR3, each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy.
In yet other embodiments of formula (I), when Z2 is CR3, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy.
In still other embodiments of formula (I), when Z2 is CR3, each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is dimethylaminoethyl, optionally substituted C1 -3 alkcycloalkyl, or optionally substituted C1 -3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In further embodiments of formula (I), when Z2 is CR3, each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is optionally substituted d_3 alkcycloalkyl, or optionally substituted C1 -3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In particular embodiments of formula (I), when Z2 is CR3, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is optionally substituted d_3 alkcycloalkyl, or optionally substituted Ci-3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In some embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma) :
Figure imgf000007_0001
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
In certain embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lla),
R7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C2 alkheteroaryl. In some embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lla),
R7 is not alkyl or unsubstituted C2 alkheteroaryl.
In particular embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lla),
R7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C2 alkheterocyclyl.
In other embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lla),
R7 is not alkyl or C2 alkheterocyclyl.
In still other embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lla),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In yet other embodiments of formula (I), when R5 is chloro, R6 is bromo, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lla),
-OR7, and R7 and R8 combine to form -CH2-CH2-
In some embodiments of formula (I), when each R5 and R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lla),
R7 is not methyl.
In particular embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lla),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not unsubstituted alkyi, substituted alkyi, unsubstituted alkheteroaryl, or substituted alkheteroaryl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not unsubstituted alkyi, substituted alkyi, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
In certain embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to form -CH2-CH2-, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to form -CH2-CH2-, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R9 is H or -C(0)-N(H)-(linear d_3 alkyi).
In particular embodiments of formula (I), when each R5 and R6 is halo, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
Z is -OR7, R7 is not methyl or 2-chloroethyl, and R9 is H or -C(0)-N(H)-(linear C1 -3 alkyi).
In some embodiments of formula (I), when R5 is methoxy, R6 is methyl, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl.
In certain embodiments of formula (I), when R5 is chloro, R6 is ethyl, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl.
In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
Figure imgf000008_0001
R7 is not methyl or 2-(N,N-diethylamino)ethyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In certain embodiments of formula (I), when each R5 and R6 is chloro, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lib) or (lie),
Z1 is -OR7, and R7 and R8 combine to form -CH2-CH2-.
In some embodiments of formula (I), when R7 is methyl, R5 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ilia), HN^ (|||a) i
R6 is not bromo.
In particular embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ilia),
R7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl.
In certain embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ilia),
R8 is not H.
In some embodiments of formula (I), when Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ilia),
each R5 and R6 is chloro, and
R7 is methyl and R8 is H, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In other embodiments of formula (I), when R5 is chloro, R6 is methoxy, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (1Mb):
Figure imgf000009_0001
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl.
In certain embodiments of formula (I), when R5 is chloro, R6 is methoxy, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (1Mb),
each R1 and R2 is H.
In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVa):
Figure imgf000010_0001
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
In particular embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVa),
R7 is not substituted alkyl, heterocyclyl, alkheterocyclyl, or alkaryl.
In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R combine to form a group according to formula (IVb):
c(IVb),
R7 is not 2-methoxyethyl or benzyl.
In certain embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVb),
R7 is not substituted alkyl or alkaryl.
In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R combine to form a group according to formula (IVc):
Figure imgf000010_0002
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVc),
R7 is not substituted alkyl.
In particular embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula IVd :
Figure imgf000010_0003
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl.
In certain embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVd),
R7 is not substituted alkyl or alkheterocyclyl.
In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVe) or (IVf):
Figure imgf000011_0001
R7 is not benzyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVe) or (IVf),
R7 is not alkaryl.
In certain embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a rou accordin to formula IV IVh IVi IV or IVk :
Figure imgf000011_0002
R7 is not methyl.
In certain embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
R7 is not alkyl.
In some embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In particular embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, and Z2 is CR3, R3 and R4 do not combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk).
In particular embodiments of formula (I), when R6 is methyl,
each R1 and R2 is H. In certain embodiments of formula (I), when R3 is H, and each R5 and R6 is chloro,
R7 is not methyl.
In particular embodiments, the compound is according to formula (la):
Figure imgf000012_0001
la), or a pharmaceutically acceptable salt thereof,
where
Z1 is -OR7, -N(R10)R7, -SR7, or -C(R10)(R1 1)R7;
each R1 and R2 is, independently, H or optionally substituted Ci-3 alkyl;
R3 is H, halogen, optionally substituted Ci-3 alkyl, or optionally substituted Ci-3 alkoxy, and R4 is halogen, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, optionally substituted amino, optionally substituted C1-6 thioalkyl, or optionally substituted C6-io aryl, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-, six-, or seven- membered ring optionally comprising from one to three heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur, where the nitrogen is optionally substituted with R9;
each R5 and R6 is, independently, H, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, halogen, or CN;
R7 is optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl, and R8 is H; or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five-, six-, or seven- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur;
R9 is H, optionally substituted C1 -3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2.9 heteroaryl, optionally substituted C2.9 heterocyclyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl;
R10 is H, optionally substituted Ci-3 alkyl (e.g., optionally substituted Ci-3 acyl), optionally substituted C3.8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2.9 heteroaryl, optionally substituted C2.9 heterocyclyl, optionally substituted C1-3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1-3 alkaryl, and R1 1 is H, optionally substituted C1 -3 alkyl, or R10 and R1 1 combine to form =0 or =S.
In other embodiments, when each of R1 and R2 is H, R3 is H, R4 is methyl or chloro, and each of R5 and R6 is chloro,
Z1 is not methoxy. In yet other embodiments, when R3 is H, and R4 is halogen (e.g., chloro),
Z1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy. In still other embodiments, when each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is dimethylaminoethyl, optionally substituted Ci-3 alkcycloalkyl, or optionally substituted Ci-3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In further embodiments, when each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is optionally substituted C1 -3 alkcycloalkyl, or optionally substituted C1 -3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In particular embodiments, when R3 is H, and R4 is halogen (e.g., chloro),
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is optionally substituted C1 -3 alkcycloalkyl, or optionally substituted C1 -3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In some embodiments, when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma):
Figure imgf000013_0001
(Ma),
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
In certain embodiments, when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C2 alkheteroaryl.
In some embodiments, when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not alkyl or unsubstituted C2 alkheteroaryl.
In particular embodiments, when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C2 alkheterocyclyl.
In other embodiments, when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not alkyl or C2 alkheterocyclyl. In still other embodiments, when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In yet other embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (Ma),
Z is -OR7, and R7 and R8 combine to form -CH2-CH2-.
In some embodiments, when each R5 and R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl.
In particular embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
In other embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheteroaryl, or substituted alkheteroaryl.
In other embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
In certain embodiments, when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to
form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (Ma),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl.
In other embodiments, when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (Ma),
R9 is H or -C(0)-N(H)-(linear d_3 alkyl).
In particular embodiments, when each R5 and R6 is halo, and R3 and R4 combine to form a group according to formula (Ma), Z is -OR7, R7 is not methyl or 2-chloroethyl, and R9 is H or -C(0)-N(H)-(linear C1 -3 alkyl).
In some embodiments, when R5 is methoxy, R6 is methyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl.
In certain embodiments, when R5 is chloro, R6 is ethyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl.
In some embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
Figure imgf000015_0001
R7 is not methyl or 2-(N,N-diethylamino)ethyl.
In other embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In certain embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (lib) or (lie),
Z1 is -OR7, and R7 and R8 combine to form -CH2-CH2-.
In some embodiments, when R7 is methyl, R5 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ilia),
Figure imgf000015_0002
R6 is not bromo.
In particular embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ilia),
R7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl. In certain embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ilia),
R8 is not H.
In some embodiments, when R3 and R4 combine to form a group according to formula (Ilia), each R5 and R6 is chloro, and
R7 is methyl and R8 is H, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In other embodiments, when R5 is chloro, R6 is methoxy, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (1Mb):
Figure imgf000016_0001
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl.
In certain embodiments, when R5 is chloro, R6 is methoxy, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (1Mb),
each R1 and R2 is H.
In some embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVa):
Figure imgf000016_0002
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
In particular embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVa),
R7 is not substituted alkyl, heterocyclyl, alkheterocyclyl, or alkaryl.
In some embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVb):
Figure imgf000016_0003
R7 is not 2-methoxyethyl or benzyl.
In certain embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVb),
R7 is not substituted alkyl or alkaryl.
In some embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVc):
Figure imgf000017_0001
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl.
In other embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVc),
R7 is not substituted alkyl.
In particular embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVd):
Figure imgf000017_0002
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl.
In certain embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVd),
R7 is not substituted alkyl or alkheterocyclyl.
In some embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVe)
Figure imgf000017_0003
R7 is not benzyl.
In other embodiments, when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVe) or (IVf),
R7 is not alkaryl.
In certain embodiments, when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
Figure imgf000017_0004
Figure imgf000018_0001
R7 is not methyl.
In certain embodiments, when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
R7 is not alkyl.
In some embodiments, when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In particular embodiments, when R5 is chloro, R6 is bromo, and Z1 is -OR7,
R3 and R4 do not combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk).
In particular embodiments, when R6 is methyl,
each R1 and R2 is H. In certain embodiments, when R3 is H, and each R5 and R6 is chloro,
R7 is not methyl.
In certain embodiments, when R3 and R4, together with the atoms to which each is attached, join to form a substituted five-membered ring comprising one nitrogen, the five-membered ring is not substituted with oxo. In particular embodiments, when R3 and R4, together with the atoms to which each is attached, join to form a substituted five-membered ring comprising one sulfur, the five-membered ring is not substituted with hydroxy or Ci-3 alkoxy. In further embodiments, when R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur, R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In particular embodiments, R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur. In other embodiments, R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered saturated ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur. In yet other
embodiments, R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- of six-membered ring optionally comprising one or two heteroatoms selected from nitrogen and oxygen. In certain embodiments, R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring.
In some embodiments, a compound is according to formula (lb):
Figure imgf000019_0001
(lb), or a pharmaceutically acceptable salt thereof,
where
each of R1 and R2 is, independently, H or optionally substituted C1-3 alkyl;
R3 is H, halogen, optionally substituted Ci-3 alkyl, or optionally substituted Ci-3 alkoxy, and R4 is halogen, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, optionally substituted amino, optionally substituted Ci-6 thioalkoxy, or optionally substituted C6-io aryl, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one sulfur, or one oxygen, where the nitrogen is optionally substituted with R9;
each of R5 and R6 is, independently, optionally substituted C1-3 alkyl (e.g., optionally substituted Ci-3 acyl), optionally substituted Ci-3 alkoxy, halogen, or CN;
R7 is optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl, and R8 is H; or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring; and
R9 is H, optionally substituted C1 -3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2.9 heteroaryl, optionally substituted C2.9 heterocyclyl, optionally substituted C1-3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl.
In other embodiments, when each of R1 and R2 is H, R3 is H, R4 is methyl or chloro, and each of R5 and R6 is chloro,
R7 is not methyl.
In yet other embodiments, when R3 is H, and R4 is halogen (e.g., chloro),
R7 is not 2-amino-2oxoethyl, 2-(N,N-diethylamino)ethyl, methyl, or benzyl.
In still other embodiments, when each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro), R7 is dimethylaminoethyl, optionally substituted Ci-3 alkcycloalkyl, or optionally substituted Ci-3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In further embodiments, when each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro), R7 is optionally substituted C1 -3 alkcycloalkyl, or optionally substituted C1-3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In particular embodiments, when R3 is H, and R4 is halogen (e.g., chloro),
R7 is optionally substituted C1 -3 alkcycloalkyl, or optionally substituted C1-3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In some embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (lla):
N
9
Ra (Ma),
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)-ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
In certain embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (lla),
R7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C2 alkheteroaryl.
In other embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (lla),
R7 is not alkyl or C2 alkheteroaryl.
In particular embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (lla),
R7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C2 alkheterocyclyl.
In some embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (lla),
R7 is not alkyl or C2 alkheterocyclyl. In still other embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (lla),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring comprising one oxygen atom and optionally comprising one more heteroatom selected from nitrogen, oxygen, and sulfur. In yet other embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (Ma),
R7 and R8 combine to form -CH2-CH2-. In some embodiments, when each R5 and R6 is bromo, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl.
In particular embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (lla),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
In other embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (lla),
R7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheteroaryl, or substituted alkheteroaryl.
In other embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (lla),
R7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
In certain embodiments, when each R5 and R6 is chloro, R7 and R8 combine to form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (lla),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl. In other embodiments, when each R5 and R6 is chloro, R7 and R8 combine to form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (lla),
R9 is H or -C(0)-N(H)-(linear d_3 alkyl).
In particular embodiments, when each R5 and R6 is halo, and R3 and R4 combine to form a group according to formula (lla),
and R9 is H or -C(0)-N(H)-(linear d_3 alkyl).
In some embodiments, when R5 is methoxy, R6 is methyl, and R3 and R4 combine to form a group according to formula (lla),
R7 is not methyl.
In certain embodiments, when R5 is chloro, R6 is ethyl, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl.
In some embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
Figure imgf000022_0001
R7 is not methyl or 2-(N,N-diethylamino)ethyl.
In other embodiments, when each R5 and R6 is chloro and R3 and R4 combine to form a group according to formula (Mb) or (lie),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring comprising one oxygen atom and optionally comprising one more heteroatom selected from nitrogen, oxygen, and sulfur. In certain embodiments, when each R5 and R6 is chloro and R3 and R4 combine to form a group according to formula (lib) or (lie),
R7 and R8 combine to form -CH2-CH2-.
In some embodiments, when R7 is methyl, R5 is chloro, and R3 and R4 combine to form a group according to formula (Ilia),
Figure imgf000022_0002
R6 is not bromo.
In particular embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (Ilia),
R7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl.
In particular embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (Ilia),
R8 is not H.
In some embodiments, when R3 and R4 combine to form a group according to formula (Ilia),
each R5 and R6 is chloro,
R7 is methyl, and R8 is H, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring comprising one oxygen atom and optionally comprising one more heteroatom selected from nitrogen, oxygen, and sulfur.
In other embodiments, when R5 is chloro, R6 is methoxy, and R3 and R4 combine to form a group according to formula (1Mb):
Figure imgf000023_0001
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl.
In certain embodiments, when R5 is chloro, R6 is methoxy, and R3 and R4 combine to form a group according to formula (1Mb),
each R1 and R2 is H.
In some embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVa):
Figure imgf000023_0002
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
In particular embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVa),
R7 is not substituted alkyl, heterocyclyl, alkheterocyclyl, or alkaryl.
In some embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVb):
Figure imgf000023_0003
R7 is not 2-methoxyethyl or benzyl.
In certain embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVb),
R7 is not substituted alkyl or alkaryl.
In some embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVc):
Figure imgf000023_0004
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl. In other embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVc),
R7 is not substituted alkyl. In particular embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVd):
Figure imgf000024_0001
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl. In certain embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVd),
R7 is not substituted alkyl or alkheterocyclyl.
In some embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVe) or (IVf):
Figure imgf000024_0002
R7 is not benzyl.
In other embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVe) or (IVf),
R7 is not substituted alkyl or alkaryl.
In certain embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
Figure imgf000024_0003
R7 is not methyl.
In some embodiments, when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk), R7 and R8 together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur. In particular embodiments, when R5 is chloro, R6 is bromo,
R3 and R4 do not combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk).
In particular embodiments, when R6 is methoxy,
each R1 and R2 is H.
In certain embodiments, when R3 is H, and each R5 and R6 is chloro,
R7 is not methyl.
In some embodiments of formula (I), (la), or (lb), R3 is H, halogen, optionally substituted C1 -3 alkyl (e.g., optionally substituted C1-3 acyl), or optionally substituted C1-3 alkoxy, and R4 is halogen, optionally substituted C1 -3 alkyl (e.g., optionally substituted C1 -3 acyl), optionally substituted C1-3 alkoxy, optionally substituted amino, optionally substituted Ci-6 thioalkyl, or optionally substituted C6-io aryl, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six- membered ring optionally comprising one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R9.
In further embodiments of formula (I), (la), or (lb), R7 is optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1-3 alkaryl, and R8 is H; or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In particular embodiments, when R3 and R4 combine to form a group according formula (Mb) or (lie), the carbonyl group of formula (lib) or (lie) is proximal to C6. In other embodiments, when R3 and R4 combine to form a group according to formula (lib) or (lie), the carbonyl group of formula (lib) or (lie) is proximal to C5 or C6.
In certain embodiments, when R3 and R4 combine to form a group according to formula (Ilia), the N atom of the group according to formula (Ilia) is proximal to C5. In particular embodiments, when R3 and R4 combine to form a group according to formula (Ilia), the N atom of the group according to formula (lllb) is proximal to C5 or C6.
In some embodiments, when R3 and R4 combine to form a group according to formula (lllb), the N atom of the group according to formula (lllb) is proximal to C6. In other embodiments, when R3 and R4 combine to form a group according to formula (lllb), the N atom of the group according to formula (lllb) is proximal to C5 or C6. In particular embodiments, when R3 and R4 combine to form a group according to any one of formulae (IVa)-(IVk), the S atom of the group according to any one of formulae (IVa)-(IVk) is proximal to C6. In particular embodiments, when R3 and R4 combine to form a group according to any one of formulae (IVa)- (IVk), the S atom of the group according to any one of formulae (IVa)-(IVk) is proximal to C5 or C6.
In some embodiments, when R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur, R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring. In certain embodiments, when R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur, R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring. In particular embodiments, when R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur, R7 and R8 combine to form a -CH2CH2- group.
In other embodiments, R7 and R8, together with the atoms to which each is attached, join to form a five- or six-membered ring.
In particular embodiments, R7 is optionally substituted Ci-3 alkyl. In other embodiments, R7 is
unsubstituted Ci-3 alkyl, Ci-3 alkylamino-C^-alkyl (e.g., Ci-3 haloalkylamino-C^-alkyl (e.g., Ci-3 fluoroalkylamino-C1 -3-alkyl)), di-(C1 -3 alkyl)amino-C1-3-alkyl (e.g., RY1 N(RY2)-(C1-3 alkyl)-, where each of RY1 and RY2 is, independently, unsubstituted C1 -3 alkyl or C1 -3 haloalkylamino-C1 -3-alkyl, (e.g., C1 -3 fluoroalkylamino-C1 -3-alkyl), or C1 -3 haloalkyl (e.g., C1 -3 fluoroalkyl). In particular embodiments, R7 is unsubstituted C1 -3 alkyl, C1-3 alkylamino-C1 -3-alkyl (e.g., C1 -3 haloalkylamino-C1 -3-alkyl, e.g., C1-3 fluoroalkylamino-Ci-3-alkyl), or di-(Ci-3 alkyl)amino-Ci-3-alkyl (e.g., RY1 N(RY2)-(d.3 alkyl)-, where each of RY1 and RY2 is, independently, unsubstituted Ci-3 alkyl, or d_3 haloalkylamino-Ci-3-alkyl, e.g., Ci-3 fluoroalkylamino-Ci-3-alkyl).
In still other embodiments, R7 is optionally substituted C1 -3 alkyl (e.g., R7 is methyl), or R7
is -(CH2)k-N(R24)R25, where k is 2 or 3 (e.g., k is 2), and where each of R24 and R25 is, independently, H or optionally substituted C1-3 alkyl (e.g., each of R24 and R25 is, independently, optionally substituted C1 -3 alkyl, e.g., each of R24 and R25 is, independently, C1 -3 haloalkyl (e.g., C1 -3 fluoroalkyl); alternatively each of R24 and R25 is, independently, unsubstituted Ci-3 alkyl, e.g., methyl). In other embodiments, when R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one nitrogen, an optional substituent on the ring is not oxo. In certain embodiments, when R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur, an optional substitutent on the ring is not hydroxyl or C1-3 alkoxy.
In yet other embodiments, each R1 and R2 is H. In particular embodiments, each R3 and R4 is, independently, optionally substituted Ci-3 alkyl or optionally substituted Ci-3 alkoxy; or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one sulfur, or one oxygen, where the nitrogen is optionally substituted with R9. In some embodiments, R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring. In certain embodiments, R3 and R4 combine to
form -CH2CH2CH2-. In particular embodiments, R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one nitrogen. In some embodiments, R3 and R4 combine to form -N(R9)-CH=CH- (e.g., R9 is H or optionally substituted C1 -3 alkyl, e.g., C1 -3 haloalkyl (e.g., C1 -3 fluoroalkyl); alternatively R9 is H). In certain embodiments, the N atom is proximal to C5 of Formula (I). In particular embodiments, R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur. In yet other embodiments, R3 and R4 combine to form -C(R13A)=C(R13B)-S- where R13A is H, and R13B is H or optionally substituted Ci-3 alkyl. In particular embodiments, the S atom is proximal to C6 of the compound of the invention. In some embodiments, R13 is optionally substituted Ci-3 alkyl, e.g., R13B is -C(0)-R13C, where R13C is optionally substituted C1-3 alkoxy or optionally substituted amino. In other embodiments, R4 is C1-3 alkyl. In specific embodiments, R4 is methyl, ethyl, or isopropyl. In certain embodiments, R4 is C1 -3 alkoxy (e.g., R4 is methoxy). In other embodiments, R4 is optionally substituted C1-6 thioalkoxy (e.g., R4 is 4-amino-4-oxobutyl). In yet other embodiments, R4 is optionally substituted amino (e.g., R4 is methylamino). In still other embodiments, R4 is halogen (e.g., R4 is chloro). In particular embodiments, R3 is hydrogen or d_3 alkyl (e.g., R3 is hydrogen, methyl, or ethyl). In further embodiments, R3 and R4 combine to form -C(R13A)=C(R13B)-S- group, where R13A is H, and R13B is H or -C(0)-R13C, where R13C is optionally substituted C1-3 alkyl, optionally substituted C1-3 alkoxy, optionally substituted amino, or optionally substituted C2.9 heterocyclyl.
In some embodiments, R3 and R4 combine to form -X1-X2-X3-, where
X1 is -S-, -0-, -(CR14R15)-, -C(R16)=, -N(R9)-, -N=, H, or optionally substituted d_3 alkyl;
X2 is absent,-(CR17R18) -, -S-, -0-, -N=, -N(R9)-, -C(R19)=, =N-, =C(R20)-, or =C(R21 )-
C(R22)=;
X3 is -(CR14R15)- -S-, -0-, -N(R9)-, =N-, =C(R23)-, halogen, optionally substituted d_3 alkyl, optionally substituted C1-6 thioalkoxy, optionally substituted C1-3 alkoxy, or optionally substituted C6-io aryl; each R14 and R15 is, independently, H or optionally substituted C1 -3 alkyl, or R14 and R15 combine to form =0 or =S;
each R17 and R18 is, independently, H or optionally substituted C1 -3 alkyl, or R17 and R18 combine to form =0 or =S;
each R16, R19, R20, R21 , R22, and R23 is, independently, H, or optionally substituted d_3 alkyl; and n is 1 or 2.
In some embodiments, when X2 is not absent,
the chain of atoms -X1-X2-X3- includes no more than one heteroatom, the heteroatom being selected from nitrogen, oxygen, and sulfur.
In particular embodiments, X1 is -(CR14R15)-, -C(R16)=, -N(R9)-, -N=, or optionally substituted d_3 alkyl. In certain embodiments, X1 is -(CR14R15)-. In particular embodiments, each R14 and R15 is H. In other embodiments, X1 is -C(R16)=. In yet other embodiments, R16 is H. In still other embodiments, X1 is - N(R9)-. In some embodiments, R9 is H or optionally substituted Ci-3 alkyl. In certain embodiments, R9 is hydrogen, methyl, or ethyl. In particular embodiments, X1 is -N=. In other embodiments, X1 is optionally substituted d.3 alkyl. In yet other embodiments, X2 is absent, -(CH2)n- -N(R9)-, -C(H)=, =C(R20)-, or =C(H)-C(H)=. In still other embodiments, X2 is -C(H)=. In further embodiments, X2 is -N(R9)-. In some embodiments, R9 is H. In certain embodiments, R9 is optionally substituted C1 -3 alkyl (e.g., R9 is -C(O)- N(H)-Et). In particular embodiments, X2 is =C(R20)-. In other embodiments, R20 is optionally substituted C1 -3 alkyl. In yet other embodiments, X2 is absent. In still other embodiments, X3 is -CH2- -S-, =C(H)-, -N(R9)-, halogen, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, optionally substituted Ci-6 thioalkoxy, optionally substituted C6-io aryl. In certain embodiments, X3 is -CH2- In particular embodiments, X3 is -S-. In some embodiments, X3 is =C(H)-. In other embodiments, X3 is -N(R9)-. In yet other embodiments, X3 is halogen, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, optionally substituted C1-6 thioalkoxy, or optionally substituted C6-io aryl.
In certain embodiments, Z1 and R8 combine to form -Z3-Y1-Y2-, where
Z3 is -0-, -N(R10)-, -N=, -S-, or -(CR11 R12)-;
Y1 is -0-, -N(R10)-, -S-, -(CR26R27)m-, -C(R20)=, =C(R20)-, =C(R21 )-C(R22)=, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkheterocyclyl, optionally substituted Ci-3 alkcycloalkyl, or optionally substituted Ci-3 alkaryl; and
Y2 is -0-, -S-, -N(R10)-, -(CR26R27)-, =C(R20)-, =N-, or H; where
each R20, R21 , and R22 is, independently, H or optionally substituted C1 -3 alkyl; and
each R26 and R27 is, independently, H or optionally substituted C1 -3 alkyl, or R26 and R27 combine to form =0 or =S;
m is 1 or 2; and
where, when Y2 is H,
the chain of atoms - Z3-Y1-Y2- comprises no more than two heteroatoms, the heteroatom selected from nitrogen, oxygen, and sulfur. In yet other embodiments, R7 and R8 form a group -Y1-Y2-, where:
Y1 is -(CR26R27)m- -C(R20)=, optionally substituted C1-3 alkyl, optionally substituted C1-3 alkheterocyclyl, optionally substituted C1 -3 alkcycloalkyl, or optionally substituted C1 -3 alkaryl; and
Y2 is -(CR26R27)-, =C(R20)-, or H; where
each R26 and R27 is, independently, H or optionally substituted Ci-3 alkyl; and
m is 1 or 2.
In particular embodiments, Z3 is oxygen. In some embodiments, Y1 is -(CR26R27)m- -C(R20)=, optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkheterocyclyl, optionally substituted C1-3 alkcycloalkyl, or optionally substituted C1 -3 alkaryl. In further embodiments, Y1 is -(CR26R27)m- or optionally substituted Ci-3 alkyl. In other embodiments, Y1 is -(CR26R27)m- In other embodiments, Y1 is optionally substituted Ci-3 alkyl (e.g., Y1 is methyl). In yet other embodiments, Y1 is -(CH2)k-N(R24)R25, where k is 2 or 3, and where each R24 and R25 is, independently, H or optionally substituted Ci-3 alkyl. In still other embodiments, k is 2. In further embodiments, each R and R is, independently, optionally substituted d.3 alkyl (e.g., each R24 and R25 is methyl). In certain embodiments, Y2 is -(CR26R27)- or H. In other embodiments, Y2 is -(CR26R27)-
In some embodiments of any aspect, each R1 and R2 is H
In certain embodiments of formula (I) or (la), a compound of the invention has a structure according to formula (Va):
Figure imgf000029_0001
(Va), or a pharmaceutically acceptable salt thereof, where all substituents are as defined herein.
In particular embodiments of formula (I), (la), or (lb), a compound of the invention has a structure according to formula (Vb):
Figure imgf000029_0002
b), or a pharmaceutically acceptable salt thereof,
where
X1 is -S-, -0-, -(CR14R15)-, -C(R16)=, -N(R9)-, -N=, -H, or optionally substituted d_3 alkyl; X2 is absent,-(CR17R18) -, -S-, -0-, -N=, -N(R9)-, -C(R19)=, =N- =C(R20)-, or =C(R21 )-
C(R22)=;
X3 is -(CR14R15)- -S-, -0-, -N(R9)-, =N- =C(R23)-, halogen, optionally substituted d_3 alkyl, optionally substituted C^e thioalkoxy, optionally substituted C1-3 alkoxy, or optionally substituted C6-io aryl;
Y1 is -(CR26R27)m- -C(R20)=, =C(R20)-, =C(R21 )-C(R22)=, optionally substituted d_3 alkyl, optionally substituted C1-3 alkheterocyclyl, optionally substituted C1-3 alkcycloalkyl, or optionally substituted d-3 alkaryl;
Y2 is -0-, -S-, -N(R10)-, -(CR26R27)-, =C(R20)-, =N-, or H;
each R1 and R2 is, independently, H or optionally substituted d-3 alkyl;
each R5 and R6 is, independently, optionally substituted d-3 alkyl, optionally substituted Ci-3 alkoxy, halogen, or CN;
each R14, R15, R16, R17, R17, R18, R20, R19, R20, R21 , R22, R23 , R26, and R27 is, independently, H or optionally substituted C1-3 alkyl;
n is 1 or 2; and
m is 1 or 2. In some embodiments, when R5 is chloro, R6 is bromo, and -X1-X2-X3- forms a group according to formula (lla):
R9 (Ma),
Y1 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)-ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
In certain embodiments, when R5 is chloro, R6 is bromo, and -X1-X2-X3- forms a group according to formula (lla),
Y1 is not optionally substituted C1 -3 alkyl or optionally substituted C2 alkheteroaryl.
In particular embodiments, when R5 is chloro, R6 is bromo, and -X1-X2-X3- forms a group according to formula (lla),
Y1 is not optionally substituted Ci-3 alkyl or optionally substituted C2 alkheterocyclyl.
In still other embodiments, when R5 is chloro, R6 is bromo, and -X1-X2-X3- forms a group according to formula (lla),
Y2 is not H.
In yet other embodiments, when R5 is chloro, R6 is bromo, and -X1-X2-X3- forms a group according to formula (lla),
each Y1 and Y2 is -CH2-. In some embodiments, when each R5 and R6 is bromo, and -X1-X2-X3- forms a group according to formula (lla),
Y1 is not methyl.
In particular embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (lla),
Y1 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol-1 - yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
In other embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (lla),
Y1 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheteroaryl, or substituted alkheteroaryl. In some embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (Ma),
Y1 is not unsubstituted alkyi, substituted alkyi, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
In certain embodiments, when each R5 and R6 is chloro, each Y1 and Y2 is -CH2-, and -X1-X2-X3- forms a group according to formula (Ma),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl. In other embodiments, when each R5 and R6 is chloro, R7 and R8 combine to form -CH2-CH2-, and -X1- X2-X3- forms a group according to formula (Ma),
R9 is H or -C(0)-N(H)-(linear d_3 alkyi).
In particular embodiments, when each R5 and R6 is halo, and -X1-X2-X3- forms a group according to formula (Ma),
and R9 is H or -C(0)-N(H)-(linear d_3 alkyi).
In some embodiments, when R5 is methoxy, R6 is methyl, and -X1-X2-X3- forms a group according to formula (Ma),
Y1 is not methyl.
In certain embodiments, when R5 is chloro, R6 is ethyl, and -X1-X2-X3- forms a group according to formula (Ma),
Y1 is not methyl.
In some embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (Mb) or (lie),
Figure imgf000031_0001
Y1 is not methyl or 2-(N,N-diethylamino)ethyl.
In other embodiments, when each R5 and R6 is chloro and -X1-X2-X3- forms a group according to formula (Mb) or (lie),
Y2 is not H.
In certain embodiments, when each R5 and R6 is chloro and -X1-X2-X3- forms a group according to formula (Mb) or (lie),
each Y1 and Y2 is -CH2-.
In some embodiments, when R7 is methyl, R5 is chloro, and -X1-X2-X3- forms a group according to formula (Ilia), HN^ (ma),
R6 is not bromo.
In particular embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (Ilia),
R7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl.
In some embodiments, when -X1-X2-X3- forms a group according to formula (Ilia),
each R5 and R6 is chloro, and
Y1 is methyl and Y2 is H, or
Y1 is -(CR26R27)m- -C(R20)=, =C(R20)-, or =C(R21 )-C(R22)=, and Y2 is -0-, -S-, -N(R10)-, - (CR26R27)-, =C(R20)-, or =N-.
In some embodiments, when -X1-X2-X3- forms a group according to formula (Ilia),
each R5 and R6 is chloro, and
Y2 is not H.
In other embodiments, when R5 is chloro, R6 is methoxy, and -X1-X2-X3- forms a group according to formula (1Mb):
Figure imgf000032_0001
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl.
In certain embodiments, when R5 is chloro, R6 is methoxy, and -X1-X2-X3- forms a group according to formula (1Mb),
each R1 and R2 is H.
In some embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (IVa):
Figure imgf000032_0002
Y1 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
In particular embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (IVa),
Y1 is not heterocyclyl, alkheterocyclyl, or alkaryl.
In some embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (I Vb):
Figure imgf000033_0001
Y1 is not 2-methoxyethyl or benzyl. In certain embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (IVb),
Y1 is not substituted alkyl or alkaryl.
In some embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (I Vc):
Figure imgf000033_0002
Y1 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl.
In other embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (IVc),
Y1 is not substituted alkyl.
In particular embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (I Vd):
Figure imgf000033_0003
Y1 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl.
In certain embodiments, when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVd),
Y1 is not substituted alkyl or alkheterocyclyl.
In some embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (IVe) or (IVf):
Figure imgf000033_0004
Y1 is not benzyl.
In other embodiments, when each R5 and R6 is chloro, and -X1-X2-X3- forms a group according to formula (IVe) or (IVf),
Y1 is not alkaryl or substituted alkyl.
In certain embodiments, when R5 is chloro, R6 is bromo, and -X1-X2-X3- forms a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
(IVj), or
Figure imgf000034_0001
Y1 is not methyl.
In some embodiments, when R5 is chloro, R6 is bromo, -X1-X2-X3- forms a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
Y2 is not H.
In particular embodiments, when R5 is chloro, R6 is bromo,
R3 and R4 do not combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk). In particular embodiments, when R6 is methoxy,
each R1 and R2 is H.
In certain embodiments, when X1 is H, and each R5 and R6 is chloro,
Y1 is not methyl.
In some embodiments, R10 is H, optionally substituted C1-3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C1-3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl. In other embodiments, R10 is H, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl.
In particular embodiments, when X1 is -(CR14R15)- X2 is -N(R9)-, X3 is -(CR14R15)-, and each of R14 and R15 is H, R9 is H or -C(0)-N(H)-Et, and each of Y1 and Y2 is -CH2-. In other embodiments, when X1 is - (CR14R15)- X2 is -N(R9)-, and X3 is -(CR14R15)-, R9 is H or -C(0)-N(H)-Et, and each of Y1 and Y2 is - CH2-. In some embodiments, when X1 is -N(R9)-, X2 is -C(R19)=, X3 is =C(R23)-, and each of R17, R19, and R23 is H, Y1 is -(CR26R27)m- and Y2 is -(CR26R27)-. In other embodiments, when X1 is -N(R9)-, X2 is -C(R19)=, X3 is =C(R23)-, and each of R9, R19, and R23 is H, each of Y1 and Y2 is -CH2-. In some embodiments, when X1 is -N(R9)-, X2 is -C(R19)=, X3 is =C(R23)-, and each of R19 and R23 is H, Y1 is - (CR26R27)m-, and Y2 is -(CR26R27)-. In certain embodiments, when X1 is -N(R9)-, X2 is -C(R19)=, X3 is =C(R23)-, Y2 is H, and each of R5 and R6 is -CI, Y1 is Me. In particular embodiments, when X1 is - C(R16)=, X2 is =C(R20)-, and X3 is -S-, R16 is H. In some embodiments, when X1 is -C(R16)=, X2 is
=C(R20)-, and X3 is -S-, Y1 is -(CR26R27)m- and Y2 is -(CR26R27)- In certain embodiments, when X1 is -C(R16)=, X2 is =C(R20)-, and X3 is -S-, each of Y1 and Y2 is -CH2-. In some embodiments, when X1 is - C(R16)=, X2 is =C(R20)-, and X3 is -S-, each of R5 and R6 is -CI. In certain embodiments, when X1 is H, and each of R5 and R6 is CI, Y1 is -(CR26R27)m- and Y2 is -(CR26R27)-. In particular embodiments, when X1 is H, and each of R5 and R6 is hal, Y1 is -(CR26R27)m- and Y2 is -(CR26R27)- (e.g., each of Y1 and Y2 is -CH2-). In particular embodiments, when X1 is H, Y1 is -(CR26R27)m- and Y2 is -(CR26R27)- (e.g., each of Y1 and Y2 is -CH2-).
In particular embodiments, X1 is -(CR14R15)-, -C(R16)=, -N(R9)-, or optionally substituted d_3 alkyl (e.g.,-CH2-, -C(H)=, -N=, optionally substituted d_3 alkyl, or-N(R9)- (e.g., R17 is H or optionally substituted d_3 alkyl (e.g.,R9 is H, -Me, or -Et).
In some embodiments, X2 is absent, -(CH2)n- -N(R9)-, -C(H)=, =C(R20)-, or =C(H)-C(H)=. In certain embodiments, X2 is absent, -C(H)=, -N(R9)- (e.g., R9 is H or optionally substituted d-3 alkyl (e.g., R9 is d.3 haloalkyl, e.g., d_3 fluoroalkyl), or R9 is -C(0)-N(H)-Et), or =C(R20)-, where R20 is, e.g., optionally substituted C1 -3 alkyl.
In certain embodiments, X3 is -CH2-, -S-, =C(H)-, or optionally substituted C1 -3 alkyl. In some embodiments, each of R5 and R6 is, independently, halo, or optionally substituted Ci-3 alkyl, e.g., each of R5 and R6 is halo (e.g., each of R5 and R6 is -CI).
In some embodiments, R26 is H. In other embodiments, R27 is H. In certain embodiments, n is 1 . In other embodiments, m is 1 . In particular embodiments, Y1 is optionally substituted C1-3 alkyl. In other embodiments, Y1 is unsubstituted C1 -3 alkyl, C1 -3 alkylamino-C1-3-alkyl (e.g., C1 -3 haloalkylamino-C1 -3-alkyl, e.g., C1-3 fluoroalkylamino-C1-3-alkyl), di-(C1 -3 alkyl)amino-C1-3-alkyl (e.g., RY1 N(RY2)-(d_3 alkyl)-, where each of RY1 and RY2 is, independently, unsubstituted C1 -3 alkyl, or C1 -3 haloalkylamino-C1-3-alkyl, e.g., C1-3 fluoroalkylamino-Ci-3-alkyl), or d_3 haloalkyl (e.g., d.3 fluoroalkyl). In particular embodiments, Y1 is unsubstituted d.3 alkyl, d_3 alkylamino-Ci-3-alkyl (e.g., d.3 haloalkylamino-Ci-3-alkyl, e.g., Ci-3 fluoroalkylamino-d-3-alkyl), or di-(Ci-3 alkyl)amino-Ci-3-alkyl (e.g., RY1 N(RY2)-(d_3 alkyl)-, where each of RY1 and RY2 is, independently, unsubstituted C1 -3 alkyl, or C1 -3 haloalkylamino-C1-3-alkyl, e.g., C1-3 fluoroalkylamino-C1 -3-alkyl).
In certain embodiments, a compound of the invention has the formula as shown in Table 1 Table!.
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
or a pharmaceutically acceptable salt thereof.
In particular embodiments a com ound of the invention has the formula:
Figure imgf000039_0002
Figure imgf000040_0001

Figure imgf000041_0001
In some embodiments a com ound of the invention has the formula:
Figure imgf000041_0002
Figure imgf000042_0001
or a pharmaceutically acceptable salt thereof.
In further embodiments, the compound is not compound 38 or compound 39.
In certain embodiments of any formula described herein, each of R1 and R2 is H, R5 is CI, and R6 is F or CI.
In other embodiments of formula (I), (la), or (lb), R3 is H, halogen, optionally substituted C1 -3 alkyl, or optionally substituted C1-3 alkoxy, and R4 is halogen, optionally substituted C1-3 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted amino, or optionally substituted C1 -6 thioalkoxy, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six- membered ring optionally comprising one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R9. In yet other embodiments of formula (I), (la), or (lb), R3 is H, optionally substituted alkyl, or optionally substituted C1 -3 alkoxy, and R4 is halogen, optionally substituted C1-3 alkyl, optionally substituted C1-3 alkoxy, optionally substituted amino, or optionally substituted C1 -6 thioalkoxy, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R9.
In still other embodiments of formula (I), (la), or (lb), R3 is H, optionally substituted Ci alkyl, or optionally substituted Ci-3 alkoxy, and R4 is halogen, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, optionally substituted amino, or optionally substituted C1 -6 thioalkoxy, or R3 and R4 join to form one of the following groups:
-N(R9)-CH=CH- -CH2CH2CH2- -CH2CH2CH2CH2- and -C(R13A)=C(R13B)-S- where N is proximal to position 5, and R9 is H or d_3 alkyl.
In certain embodimentsof formula (I), Z2 is N, and R4 is halogen, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, optionally substituted amino, or optionally substituted Ci-6 thioalkoxy.
In particular embodiments of formula (I), Z2 is N, and R4 is optionally substituted C1-3 alkoxy or optionally substituted C1 -6 thioalkoxy. In other embodiments, X1 is -N(R9)-, -(CR14R15)-, -C(R16)=, H, or optionally substituted d alkyl. In yet other embodiments, X2 is absent,-(CR17R18)n- -C(R19)=, or =C(R20)-. In still other embodiments, X3 is - (CR14R15)- -S-, =C(R23)-, halogen, optionally substituted d_3 alkyl, optionally substituted d_6 thioalkoxy, or optionally substituted d-3 alkoxy. In certain embodiments, R9 is H or d_3 alkyl. In some embodiments, the compound of the invention has a molecular weight of less than about 500 g/mol (e.g., less than about 450 g/mol, or less than about 400 g/mol). In other embodiments, the compound of the invention exhibits apical to basal (A→B) permeability in MDR1 -MDCK assay of greater than about 1 x 10~7 cm/sec (e.g., greater than about 5 x 10 ~7 cm/sec, greater than about 1 x 1 0"6 cm/sec, or greater than about 3 x 1 0"6 cm/sec). In particular embodiments, the compound of the invention exhibits the (B→A)/(A→B) ratio of less than about 30 (e.g., less than about 10, less than about 5, or less than about 3).
In another aspect, the invention features a pharmaceutical composition comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, and one or more of pharmaceutically acceptable carriers or excipients. In certain embodiments, the composition is formulated for administration orally, intradermally, intramuscularly, parenterally, intravenously, intra-arterially, intracranially, subcutaneously, intraorbitally, intraventricularly, intraspinally, intraperitoneally, or intranasally. Preferably, the composition is formulated for oral administration.
In yet another aspect, the invention features a method of treating a disorder in a mammal (e.g., a caused by the action of heat shock protein 90 (Hsp90). The method involves administering to the mammal an effective amount of a compound according to formula (I):
Figure imgf000044_0001
(I), or a pharmaceutically acceptable salt thereof,
where
Z1 is -OR7, -N(R10)R7, -SR7, or -C(R10)(R1 1)R7;
Z2 is -N= or -C(R3)=;
each R1 and R2 is, independently, H or optionally substituted Ci-3 alkyl;
R3 is H, halogen, cyano, optionally substituted C1 -6 alkyl, optionally substituted C1-3 alkoxy, or optionally substituted amino, and R4 is halogen, cyano, optionally substituted C1 -6 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted amino, optionally substituted C1 -6 thioalkoxy, or optionally substituted C6-i0 aryl, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R9;
each R5 and R6 is, independently, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, halogen, or CN;
R7 is optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1-3 alkaryl, and R8 is H; or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur;
R9 is H, optionally substituted Ci-3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-i0 aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted C1 -3 alkaryl;
R10 is H, optionally substituted C1 -3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-10 aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted C1-3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl, and R11 is H, optionally substituted Ci-3 alkyl, or R10 and R1 1 combine to form =0 or =S and
Rm is H, halogen, optionally substituted Ci-4 alkyl, or optionally substituted Ci-3 alkoxy;.
In certain embodiments of formula (I), when Z2 is CR3,each of R1 and R2 is H, R3 is H, R4 is methyl or halogen (e.g., chloro), and each of R5 and R6 is halogen (e.g., chloro),
Z1 is not methoxy. In certain embodiments of formula (I), when Z2 is CR3, R3 is H, R4 is methyl or halogen (e.g., chloro), each of R5 and R6 is halogen (e.g., chloro),
Z2 is not unsubstituted Ci-3 alkoxy.
In particular embodiments of formula (I), when Z2 is N, R3 is H, R4 is optionally substituted Ci-6 thioalkoxy, and each of R5 and R6 is halogen (e.g., chloro),
Z1 is not cyanomethoxy or aminomethoxy.
In other embodiments of formula (I), when Z2 is N, R3 is H, each of R5 and R6 is halogen (e.g., chloro), R4 is substituted Ci-6 thioalkoxy,
Z1 is not cyanomethoxy or aminomethoxy.
In some embodiments of formula (I), when Z2 is N, R3 is H, R4 is optionally substituted Ci-6 thioalkoxy, Z1 is not cyanomethoxy or aminomethoxy.
In certain embodiments of formula (I), when Z2 is N, R3 is H, R4 is substituted C1 -6 thioalkoxy,
Z1 is not cyanomethoxy or aminomethoxy.
In further embodiments of formula (I), when Z2 is N, R3 is H, R4 is substituted Ci-6 thioalkoxy,
Z1 is not substituted Ci alkoxy.
In particular embodiments of formula (I), when Z2 is N, R3 is H, R4 is substituted C1 -6 thioalkoxy,
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is methyl, dialkylaminoethyl, optionally substituted C1 -3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1-3 alkaryl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In other embodiments of formula (I), when Z2 is CR3, each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy.
In yet other embodiments of formula (I), when Z2 is CR3, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is not 2-amino-2oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy.
In still other embodiments of formula (I), when Z2 is CR3, each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is dimethylaminoethyl, optionally substituted C1 -3 alkcycloalkyl, or optionally substituted C1 -3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur. In further embodiments of formula (I), when Z2 is CR3, each of R5 and R6 is chloro, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is optionally substituted d_3 alkcycloalkyl, or optionally substituted Ci-3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In particular embodiments of formula (I), when Z2 is CR3, R3 is H, and R4 is halogen (e.g., chloro),
Z1 is -OR7, -N(R7)R10, -SR7, or -C(R7)(R10)R1 1 , in which R7 is optionally substituted d_3 alkcycloalkyl, or optionally substituted Ci-3 alkheterocyclyl, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In some embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma) :
R9 (Ma),
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl.
In certain embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C2 alkheteroaryl.
In some embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not alkyl or unsubstituted C2 alkheteroaryl.
In particular embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not substituted alkyl, unsubstituted alkyl, or unsubstituted C2 alkheterocyclyl.
In other embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not alkyl or C2 alkheterocyclyl.
In still other embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In yet other embodiments of formula (I), when R5 is chloro, R6 is bromo, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
Z is -OR7, and R7 and R8 combine to form -CH2-CH2-.
In some embodiments of formula (I), when each R5 and R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl.
In particular embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl- methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl-1 ,2,4-oxadiazol-5-yl- methyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheteroaryl, or substituted alkheteroaryl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not unsubstituted alkyl, substituted alkyl, unsubstituted alkheterocyclyl, or substituted alkheterocyclyl.
In certain embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to form -CH2-CH2-, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to form -CH2-CH2-, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R9 is H or -C(0)-N(H)-(linear d_3 alkyl).
In particular embodiments of formula (I), when each R5 and R6 is halo, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
Z is -OR7, R7 is not methyl or 2-chloroethyl, and R9 is H or -C(0)-N(H)-(linear C1 -3 alkyl). In some embodiments of formula (I), when R5 is methoxy, R6 is methyl, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl. In certain embodiments of formula (I), when R5 is chloro, R6 is ethyl, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl.
In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lib) or (lie),
Figure imgf000048_0001
R7 is not methyl or 2-(N,N-diethylamino)ethyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lib) or (lie),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In certain embodiments of formula (I), when each R5 and R6 is chloro, Z2 is CR3, and R3 and R4 combine to form a group according to formula (lib) or (lie),
Z1 is -OR7, and R7 and R8 combine to form -CH2-CH2-.
In some embodiments of formula (I), when R7 is methyl, R5 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ilia),
of
(Ilia),
R6 is not bromo.
In particular embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ilia),
R7 is not isopropyl, 3,3,3-trifluoropropyl, or 2-(N,N-dimethylamino)ethyl. In certain embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ilia),
R8 is not H.
In some embodiments of formula (I), when Z2 is CR3, and R3 and R4 combine to form a group according to formula (Ilia),
each R5 and R6 is chloro, and
R7 is methyl and R8 is H, or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur. In other embodiments of formula (I), when R5 is chloro, R6 is methoxy, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (1Mb):
Figure imgf000049_0001
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl.
In certain embodiments of formula (I), when R5 is chloro, R6 is methoxy, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (1Mb),
each R1 and R2 is H.
In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVa):
Figure imgf000049_0002
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl.
In particular embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVa),
R7 is not substituted alkyl, heterocyclyl, alkheterocyclyl, or alkaryl.
In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVb):
Figure imgf000049_0003
nethoxyethyl or benzyl.
In certain embodiments of formula (I), when each R and R is chloro, Z is -OR , Z is CR , and R and R4 combine to form a group according to formula (IVb),
R7 is not substituted alkyl or alkaryl. In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVc): NH
CF3 (IVc),
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVc),
R7 is not substituted alkyl.
In particular embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula IVd :
Figure imgf000050_0001
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl.
In certain embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVd),
R7 is not substituted alkyl or alkheterocyclyl.
In some embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVe) or (IVf):
Figure imgf000050_0002
R7 is not benzyl.
In other embodiments of formula (I), when each R5 and R6 is chloro, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVe) or (IVf),
R7 is not alkaryl.
In certain embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a rou accordin to formula IV IVh IVi IV or IVk :
Figure imgf000050_0003
Figure imgf000051_0001
R7 is not methyl.
In certain embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
R7 is not alkyl.
In some embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, Z2 is CR3, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk),
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
In particular embodiments of formula (I), when R5 is chloro, R6 is bromo, Z1 is -OR7, and Z2 is CR3, R3 and R4 do not combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk).
In particular embodiments of formula (I), when R6 is methyl,
each R1 and R2 is H. In certain embodiments of formula (I), when R3 is H, and each R5 and R6 is chloro,
R7 is not methyl.
In particular embodiments of formula (I), Rm is H (e.g., the compound of formula (I) has the following structure:
Figure imgf000051_0002
(I), or a pharmaceutically acceptable salt thereof.
In certain embodiments of formula (I),
In some embodiments of the methods of the invention, the substituents are defined as described herein. In particular embodiments of the methods of the invention, the method involves administering to the mammal an effective amount of a compound according to formula (I). In certain embodiments of the methods of the invention, the method involves administering to the mammal an effective amount of a compound according to formula (lb). In other embodiments, the method involves administering to the mammal an effective amount of a compound according to formula (Va). In certain embodiments, the method involves administering to the mammal an effective amount of a compound according to formula (Vb). In particular embodiments, the compound may be selected from Table 2, (e.g., any one of compounds 2, 5-16, 18-27, 29, 33-36, 40-48, and 58-77, or a or a pharmaceutically acceptable salt thereof). In further embodiments, the method involves administering to the mammal an effective amount of compound 38 or 39 or a pharmaceutically acceptable salt thereof.
In certain embodiments of the methods of the invention, the method involves treating a mammal having a neurodegenerative disorder by administering to the mammal the compound of formula (I) (e.g., formula (la), (lb), (Va), or (Vb)).
In some embodiments, the disorder is a neurodegenerative disorder (e.g., a tauopathy). The
neurodegenerative disorder may be Alzheimer's disease, Huntington's disease, progressive supranuclear palsy, Parkinson's syndrome, Pick's disease, corticobasal degeneration, chronic traumatic
encephalopathy, traumatic brain injury, or frontotemporal dementia. Preferably, the neurodegenerative disorder is Alzheimer's disease. In other embodiments, the disorder is a proliferative disorder (e.g., a cancer, e.g., acute myeloid leukemia, gastrointestinal stromal tumor, gastric cancer, glioblastoma, lung cancer, lymphoma, melanoma, myeloma, non-small cell lung cancer, renal cancer, small cell lung cancer, blast-phase chronic myelogenous leukemia, leukemia, lymphoproliferative disorder, metastatic melanoma, relapsed multiple myeloma, refractory multiple myeloma, myeloproliferative disorders, pancreatic cancer, small intestine cancer, or solid tumor).
In particular embodiments, the disorder is an inflammatory or autoimmune disease (e.g., rheumatoid arthritis, systemic lupus erythermatosus, or asthma). In certain embodiments, the disorder is a cardiovascular disease (e.g., atherosclerosis or cardiomyoapthy). In other embodiments, the disorder is an allergy.
In yet another aspect, the invention features a method of treating an infectious disease in a mammal by administering an effective amount of the compound of the invention (e.g., compounds of the aspects described above), or a pharmaceutically acceptable salt thereof to the mammal.
In some embodiments, the infectious disease is a viral infection. In certain embodiments, the viral infection is a virus of Herpesviridae family (e.g., herpes simplex virus-1 , herpes simplex virus-2, herpes herpesvirus-5, Kaposi's sarcoma-associated herpesvirus, varicella zoster virus, or Epstein-Barr virus), Polyomaviridae family (e.g., SV40), Poxviridae family (e.g., vaccinia virus), Reoviridae family (e.g., rotavirus), Birnaviridae family (e.g., infectious bursal disease virus), Picornaviridae family (e.g., poliovirus, rhinovirus, or coxsackievirus), Flaviviridae family (e.g., hepatitis C virus or dengue virus), Arenaviridae family (e.g., lymphocytic choriomeningitis virus), Hepeviridae family (e.g., Hepatitis E virus),
Rhabdoviridae family (e.g., vesicular stomatitis virus), Paramoxyviridae family (e.g., human parainfluenza virus 2, human parainfluenza virus 3, SV5, SV41 , measles virus, or Sendai virus), Bunyaviridae family (e.g., La Crosse virus), Orthomoxyviridae family (e.g., influenza A virus), Filoviridae family (e.g., Ebola virus), Retroviridae family (e.g., HTLV1 or HIV1 ), or Hepadnaviridae family (e.g., hepatitis B virus). In particular embodiments, the infectious disease is a fungal infection (e.g., Candida albicans, Aspergillus fumigates, or Pneumocystis jiroveci).
In other embodiments, the infectious disease is a bacterial infection (e.g., mycobacteria, anthrax, or bacterial pneumonia).
In some embodiments of any aspect of the methods of the invention, the compound is administered orally, sublingually, buccally, transdermal^, intradermally, intramuscularly, parenterally, intravenously, intra-arterially, intracranial^, subcutaneously, intraorbitally, intraventricularly, intraspinally,
intraperitoneal^, intranasally, by inhalation, and topically. Preferably, the compound is administered orally.
In certain embodiments of any aspect of the methods of the invention, the mammal is human. In a further aspect, the invention features a method of inhibiting Hsp90 by contacting a cell with the compound of the invention, or a pharmaceutically acceptable salt thereof. In some embodiments, the method is carried out in vitro. In other embodiments, the method is carried out in vivo.
In another aspect, the invention features a kit containing:
(i) the pharmaceutical composition of the invention; and
(ii) instructions for use of the pharmaceutical compositions of (i) to treat a disorder in a mammal (e.g., a human) caused by the action of Hsp90.
In yet another aspect, the invention features the compounds of the invention for use in treating a disorder caused by the action of heat shock protein 90 (Hsp90). In some embodiments of this aspect, the disorder is any one of the disorders described in the aspect featuring methods of the invention. In a related aspect, the invention features the compounds of the invention for use in treating an infectious disease. In certain embodiments of this aspect, the infectious disease is any one of the infectious diseases described in the aspect featuring methods of the invention.
In still another aspect, the invention features uses of a compound of the invention in treating a disorder caused by the action of heat shock protein 90 (Hsp90). In some embodiments of this aspect, the disorder can be any one of the disorders described in the aspect featuring methods of the invention. In a related aspect, the invention features uses of a compound of the invention in treating an infectious disease. In certain embodiments of this aspect, the infectious disease is any one of the infectious diseases described in the aspect featuring methods of the invention.
In a further aspect, the invention features uses of a compound of the invention in the manufacture of a medicament for treating a disorder caused by the action of heat shock protein 90 (Hsp90). In some embodiments of this aspect, the disorder can be any one of the disorders described in the aspect featuring methods of the invention. In a related aspect, the invention features uses of a compound of the invention in the manufacture of a medicament for treating an infectious disease. In certain embodiments of this aspect, the infectious disease is any one of the infectious diseases described in the aspect featuring methods of the invention.
Definitions
Chemical Substituents
The term "about," as used herein, represents a value that is ±10% of the recited value.
The term "acyl" or "alkanoyl," as used interchangeably herein, represent an alkyl group, as defined herein, or hydrogen attached to the parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl, acetyl, propionyl, butanoyl, and the like. Exemplary unsubstituted acyl groups include from 2 to 7 carbons. Acyl can be unsubstituted or substituted in the same as defined for alkyl.
The term "alkaryl," as defined herein, represents a chemical substituent of formula -(alkylene)-(aryl), where each group is as defined herein and may be substituted or unsubstituted according to each respective definition.
The term "alkheterocyclyl," as defined herein, represents a chemical substituent of formula -(alkylene)- (heterocyclyl), where each group is as defined herein and may be substituted or unsubstituted according to each respective definition.
The term "alkoxy," as used herein, represents a chemical substituent of formula -OR, where R is a C1 -6 alkyl group, unless otherwise specified (e.g., R is C1 -3 alkyl). Alkoxy may be unsubstituted or substituted with one, two, or three substituents independently selected from the group consisting of: (1 ) alkoxy of one to six carbons; (2) hydroxyl; (3) amino; (4) alkylamino of one to six carbons; (5) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (6) cycloalkyl of three to eight carbons; (7) oxo; (8) hal; (9) alkylsulfonyl of one to six carbon atoms; (10) thioalkoxy of one to six carbon atoms; (1 1 ) aryl; (12) -C02RA, where RA is selected from the group consisting of (a) hydrogen, (b) alkyl, (c) cycloalkyl, (d) alkcycloalkyl, and (e) alkaryl, where the alkylene group is of one to six carbon atoms; or (13) - C(0)NRBRc, where each of RB and Rc is, independently, selected from the group consisting of (a) hydrogen, (b) alkyl, (c) cycloalkyl, (d) alkcycloalkyl, and (e) alkaryl, where the alkylene group is of one to six carbon atoms; with the proviso that no more than one substituent (2)-(5) may be attached to a single carbon atom of the alkyl group, and none of the substituents (2)-(5) may be attached to the carbon connected to the oxygen atom of the alkoxy group. No more than one oxo substituent may be attached to a single carbon of the alkoxy group, and no more than two oxo substituents may be found in any of the alkoxy group as defined herein.
The term "alkoxyalkyl," as used herein, represents an alkyl group that is substituted with an alkoxy group. Exemplary unsubstituted alkoxyalkyl groups include from 2 to 9 carbons. In some embodiments, the alkyl and the alkoxy each can be further substituted with 1 , 2, 3, 4, or 5 substituent groups as defined herein for each respective group. The terms "alkyl," as used herein, are inclusive of both straight chain and branched chain saturated groups of from 1 to 6 carbons, unless otherwise specified (e.g., from 1 to 3 carbons). Alkyl groups are exemplified by methyl, ethyl, n- and iso-propyl, and may be optionally substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four substituents, unless otherwise specified, independently selected from the group consisting of: (1 ) alkoxy of one to six carbons; (2) hydroxyl; (3) amino; (4) alkylamino of one to six carbons; (5) dialkylamino, where each of alkyl groups is,
independently, one to six carbons; (6) cycloalkyi of three to eight carbons; (7) oxo; (8) hal; (9) alkylsulfonyl of one to six carbon atoms; (10) thioalkoxy of one to six carbon atoms; (1 1 ) aryl; (12) - C02RA, where RA is selected from the group consisting of (a) hydrogen, (b) alkyl, (c) cycloalkyi, (d) alkcycloalkyi, and (e) alkaryl, where the alkylene group is of one to six carbon atoms; (13) -C(0)NRBRc, where each of RB and Rc is, independently, selected from the group consisting of (a) hydrogen, (b) alkyl, (c) cycloalkyi, (d) alkcycloalkyi, and (e) alkaryl, where the alkylene group is of one to six carbon atoms, or RB and Rc combine to form C2-9 heterocyclyl; and (14) cyano; with the proviso that no more than one substituent (2)-(5) may be attached to a single carbon atom of the alkyl group. No more than one oxo substituent may be attached to a single carbon of the alkyl group, and no more than two oxo substituents may be found in any of the alkyl group as defined herein.
The term "alkylene" and the prefix "alk-," as used herein, represent a saturated divalent CMO hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, propylene, isopropylene, and the like. The term "Cx.y alkylene" and the prefix "Cx.y alk-" represent alkylene groups having between x and y carbons. Exemplary values for x are 1 , 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, or 10. In some embodiments, the alkylene can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the alkyl group.
The term "alkylsulfonyl," as used herein, represents an alkyl group attached to the parent molecular group through an -S(0)2- group. Exemplary unsubstituted alkylsulfonyl groups are of from 1 to 6 carbons. In some embodiments, the alkyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein.
The term "amino," as used herein, represents a chemical substituent of formula -NH2. Amino group may be singly substituted with, e.g., an alkyl, alkanoyl, aryl, aryloyl, cycloalkyi, heterocyclyl, or alkheterocyclyl group (e.g., "alkylamino" having formula -NH-(optionally substituted Ci-6 alkyl), e.g., -NH-(unsubstituted Ci-6 alkyl)), or doubly substituted with, e.g., alkyl, alkanoyl, aryl, aryloyl, cycloalkyi, heterocyclyl, or alkheterocyclyl group (e.g., "dialkylamino" having formula -NR'R", where each of R' and R" is, independently, optionally substituted C1 -6 alkyl, e.g., -NR'R", where each of R' and R" is, independently, unsubstituted C1 -6 alkyl). Optionally substituted C1-6 alkyl group may be a C1-6 haloalkyl, e.g., a C1 -6 fluoroalkyl. The term "aminoalkyl," as used herein, represents an alkyl group that is substituted with an amino group. Each of the alkyl and amino groups may be, independently, substituted or unsubstituted as defined herein for each respective group. The term "aryl," as used herein, represents a mono-, bicyclic, or multicyclic carbocyclic ring system having between three and twelve carbons and having one or two aromatic rings. Non-limiting examples of aryl groups include phenyl, naphthyl, 1 ,2-dihydronaphthyl, 1 ,2,3,4-tetrahydronaphthyl, fluorenyl, indanyl, indenyl, and the like. An aryl group may be optionally substituted with one, two, three, four, or five substituents independently selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (7) cycloalkyi of three to eight carbons; (8) oxo; (9) hal; (10) alkylsulfonyl of one to six carbon atoms; (1 1 ) thioalkoxy of one to six carbon atoms; (12) aryl; (13) alkaryl, where alkylene group is one to six carbon atoms; (14) -C02RA, where RA is selected from the group consisting of (a) alkyl, (b) cycloalkyi, (c) alkcycloalkyi, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms; and (15) -C(0)NRBRc, where each of RB and Rc is, independently, selected from the group consisting of (a) alkyl, (b) cycloalkyi, (c) alkcycloalkyi, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms.
The term "aryloyl," as used herein, represents an aryl group attached to the oarent molecular group through an alkyl group.
The term "carbonyl," as used herein, represents a C(O) group, which can also be represented as C=0, and results from the combination of a tetravalent carbon atom and an oxo substituent.
The term "cyano," as used herein represents -CN group.
The term "cyanoalkyl," as used herein represents an alkyl group that is substituted with cyano.
Exemplary unsubstituted cyanoalkyl groups include from 2 to 9 carbons. In some embodiments, the alkyl group can be further substituted with 1 , 2, 3, 4, or 5 substituent groups as defined herein for each respective group.
The term "cycloalkyi," as used herein represents a monovalent saturated or unsaturated non-aromatic cyclic hydrocarbon group of from three to eight carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[2.2.1 .Jheptyl, and the like. Cycloalkyi groups may be optionally substituted with, for example, one, two, three, or four substituents,
independently, selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (7) cycloalkyi of three to eight carbons; (8) oxo; (9) halo; (10) alkylsulfonyl of one to six carbon atoms; (1 1 ) thioalkoxy of one to six carbon atoms; (12) aryl; (13) alkaryl, where alkylene group is one to six carbon atoms; (14) -C02RA, where RA is selected from the group consisting of (a) alkyl, (b) cycloalkyi, (c) alkcycloalkyi, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms; or (15) -C(0)NRBRc, where each of RB and Rc is, independently, selected from the group consisting of (a) alkyl, (b) cycloalkyi, (c) alkcycloalkyi, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms, or RB and Rc combine to form C2-9 heterocyclyl. The term "five-membered ring," as used herein, represent a saturated or unsaturated aromatic or non- aromatic group having five atoms in a cyclic array, where, unless otherwise specified, four atoms are carbons and the remaining atom is selected from the group consisting of carbon, nitrogen, sulfur, and oxygen. A five-membered ring may be fused to another cyclic group selected from heterocyclyl, heteroaryl, cycloalkyi, and aryl. A five-membered ring may be unsubstituted or substituted with, for example, one, two, three, or four substituents, independently selected from the group consisting of: (1 ) alkoxy of one to six carbons; (2) hydroxyl; (3) amino; (4) alkylamino of one to six carbons; (5) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (6) cycloalkyi of three to eight carbons; (7) oxo; (8) alkylsulfonyl of one to six carbon atoms; (9) thioalkoxy of one to six carbon atoms; (10) aryl; (1 1 ) alkaryl, where alkylene group is one to six carbon atoms; (12) optionally substituted alkyl of one to six carbons (e.g., unsubstituted alkyl, alkoxyalkyl, hydroxyalkyl, haloalkyl, or cyanoalkyl); (13) -C02RA, where RA is selected from the group consisting of (a) alkyl, (b) cycloalkyi, (c) alkcycloalkyl, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms; (14) -C(0)NRBRc, where each of RB and Rc is, independently, selected from the group consisting of (a) optionally substituted alkyl (e.g., unsubstituted alkyl, alkoxyalkyl, hydroxyalkyl, haloalkyl, or cyanoalkyl), (b) cycloalkyi, (c) alkcycloalkyl, (d) alkaryl, (e) heterocyclyl, (f) alkheterocyclyl, (g) alkoxy, and (h) hydrogen, where the alkylene group is of one to six carbon atoms, or RB and Rc combine to form C2-9 heterocyclyl; and (1 5) cyano.
The term "fluoroalkyl," as used herein, represents an alkyl group, as defined herein, where one or more hydrogen radicals (e.g., 1 , 2, 3, 4, or 5, or more hydrogen radicals) bound to the alkyl group have been replaced by a fluorine radical. In some embodiments, fluoroalkyl group may be perfluoroalkyl. Prefix "fluoro" indicates that the group in question is substituted by one or more (e.g., 1 , 2, 3, 4, or 5, or more) fluorine radicals.
The term "haloalkyl," as used herein, represents an alkyl group, as defined herein, substituted by a halogen group (i.e., F, CI, Br, or I). A Ci-6 haloalkyl may be substituted with one two, three, or in the case of alkyl groups of two carbons or more, four or five halogens. Haloalkyl groups include perfluoroalkyls. In certain embodiments, haloalkyl is fluoroalkyl. In some embodiments, the C1-6 haloalkyl group may be further substituted with 1 , 2, 3, or 4 substituent groups as described herein for alkyl groups.
The terms "halogen," "hal," or "halo," as used interchangeably herein, represent a group selected from fluorine (-F), chlorine (-CI), bromine (-Br), and iodine (-I). Prefix "halo" indicates that the group in question is substituted by a halogen group (i.e., F, CI, Br, or I).
The term "heterocyclyl," as used herein represents a 5-, 6- or 7-membered ring, unless otherwise specified, containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. The 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds. The term "heterocyclyl" also represents a heterocyclic compound having a bridged multicyclic structure in which one or more carbons and/or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group. The term "heterocyclyl" also includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one, two, or three carbocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, or another monocyclic heterocyclic ring, e.g., indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, benzofuryl, benzothienyl and the like. Examples of fused heterocyclyls include tropanes and 1 ,2,3,5,8,8a-hexahydroindolizine. Heterocyclics include pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, piperidinyl, homopiperidinyl, pyrazinyl, piperazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidiniyl, morpholinyl, thiomorpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, furyl, thienyl, thiazolidinyl, isothiazolyl, isoindazoyl, triazolyl, tetrazolyl, oxadiazolyl, uricyl, thiadiazolyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, dihydroindolyl, tetrahydroquinolyl, tetrahydroisoquinolyl, pyranyl, dihydropyranyl, dithiazolyl, benzofuranyl, benzothienyl and the like. The heterocyclyl group may be optionally substituted with one, two, three, four, or five substituents independently selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is,
independently, one to six carbons; (7) cycloalkyl of three to eight carbons; (8) oxo; (9) hal; (10) alkylsulfonyl of one to six carbon atoms; (1 1 ) thioalkoxy of one to six carbon atoms; (12) aryl; (13) alkaryl; (14) -C02RA, where RA is selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyl, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms; or (15) - C(0)NRBRc, where each of RB and Rc is, independently, selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyl, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms, or RB and Rc combine to form C2-9 heterocyclyl.
The term "heteroaryl," as used herein, represents that subset of heterocyclyls, as defined herein, which are aromatic: i.e., they contain 4n+2 pi electrons within the mono- or multicyclic ring system. In some embodiments, the heteroaryl is substituted with, e.g., 1 , 2, 3, or 4 substituent groups independently selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (7) cycloalkyl of three to eight carbons; (8) oxo; (9) hal; (10) alkylsulfonyl of one to six carbon atoms; (1 1 ) thioalkoxy of one to six carbon atoms; (12) aryl; (13) alkaryl; (14) -C02RA, where RA is selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyl, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms; or (15) - C(0)NRBRc, where each of RB and Rc is, independently, selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyl, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms.
The term "hydroxy," as used herein, represents an -OH group.
The term "hydroxyalkyl," as used herein, represents an alkyl group, as defined herein, substituted by one or two hydroxy groups, with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group and is exemplified by hydroxymethyl, dihydroxypropyl, and the like. The term "/V-protected amino," as used herein, refers to an amino group, as defined herein, to which is attached an /V-protecting group, as defined herein.
The term "/V-protecting group," as used herein, represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used /V-protecting groups are disclosed in Greene, Protective Groups in Organic Synthesis, 3rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference. N-protecting groups include acyl, aroyl, or carbamyl groups, e.g., formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4- bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries, e.g., protected or unprotected D, L or D, L-amino acids, e.g., alanine, leucine, phenylalanine, and the like; sulfonyl groups, e.g., benzenesulfonyl, p- toluenesulfonyl, and the like; carbamate forming groups, e.g., benzyloxycarbonyl, p- chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2- nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4-dimethoxybenzyloxycarbonyl,
3,5-dimethoxybenzyl oxycarbonyl, 2,4-dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2- nitro-4,5-dimethoxybenzyloxycarbonyl, 3,4,5-trimethoxybenzyloxycarbonyl, 1 -(p-biphenylyl)-l - methylethoxycarbonyl, a,a-dimethyl-3,5-dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t- butyloxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2, -trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9- methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl,
phenylthiocarbonyl, and the like, arylalkyi groups, e.g., benzyl, triphenylmethyl, benzyloxymethyl, and the like and silyl groups, e.g., trimethylsilyl, and the like. Preferred N-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc), and
benzyloxycarbonyl (Cbz).
The term "six-membered ring," as used herein, represent a saturated or unsaturated aromatic or non- aromatic group having six atoms in a cyclic array, where, unless otherwise specified, five atoms are carbons and the remaining atom is selected from the group consisting of carbon, nitrogen, sulfur, and oxygen. A six-membered ring may be fused to another cyclic group selected from heterocyclyl, heteroaryl, cycloalkyl, and aryl. A six-membered ring may be unsubstituted or substituted with, for example, one, two, three, or four substituents, independently selected from the group consisting of: (1 ) alkyl of one to six carbons; (2) alkoxy of one to six carbons; (3) hydroxyl; (4) amino; (5) alkylamino of one to six carbons; (6) dialkylamino, where each of alkyl groups is, independently, one to six carbons; (7) cycloalkyl of six to eight carbons; (8) oxo; (9) alkylsulfonyl of one to six carbon atoms; (10) thioalkoxy of one to six carbon atoms; (1 1 ) aryl; (12) alkaryl, where alkylene group is one to six carbon atoms; (13) - C02RA, where RA is selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyi, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms; (14) -C(0)NRBRc, where each of RB and Rc is, independently, selected from the group consisting of (a) alkyl, (b) cycloalkyl, (c) alkcycloalkyi, (d) alkaryl, and (e) hydrogen, where the alkylene group is of one to six carbon atoms, or RB and Rc combine to form C2-9 heterocyclyl; and (15) cyano. The term "thioalkoxy," as used herein, represents a chemical substituent of formula -SR, where R is an alkyl group. In some embodiments, the alkyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as described herein for alkyl groups. The term "thiol," as used herein, represents a chemical substituent of formula -SH.
When referring to numbered position within the compounds of the invention, the following numbering system is employed:
Figure imgf000060_0001
For example, the statement "R3 and R4 combine to form -N(R9)-CH=CH-" indicates that the nitrogen atom of -N(R9)-CH=CH- may be proximal to either C5 carbon or C6 carbon. The statement "R3 and R4 combine to form -N(R9)-CH=CH-" in combination with "the nitrogen atom of - 9)-CH=CH- is proximal to C5" indicates the following compound:
Figure imgf000060_0002
Asymmetric or chiral centers may exist in any of the compounds of the present invention. The present invention contemplates the various stereoisomers and mixtures thereof. Individual stereoisomers of compounds of the present invention are prepared synthetically from commercially available starting materials that contain asymmetric or chiral centers or by preparation of mixtures of enantiomeric compounds followed by resolution well known to those of ordinary skill in the art. These methods of resolution are exemplified by (1 ) attachment of a racemic mixture of enantiomers, designated (+/-), to a chiral auxiliary, separation of the resulting diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary or (2) direct separation of the mixture of optical enantiomers on chiral chromatographic columns or by chiral HPLC methods. Methods of chiral separations have been described previously (G.B. Cox (ed.) in Preparative Enantioselective
Chromatography, 2005, Blackwell Publishing). Alternatively, chiral compounds can be prepared by an asymmetric synthesis that favors the preparation of one enantiomer over the other. Alternatively a chiral pool synthesis (starting with an enantiomerically pure building block) can be used where the chiral group or center is retained in the intermediate or final product. Enantiomers are designated herein by the symbols "R," or "S," depending on the configuration of substituents around the chiral atom. Alternatively, enantiomers are designated as (+) or (-) depending on whether a solution of the enantiomer rotates the plane of polarized light clockwise or counterclockwise, respectively. Geometric isomers may also exist in the compounds of the present invention. The present invention contemplates the various geometric isomers and mixtures thereof resulting from the arrangement of substituents around a carbon-carbon double bond and designates such isomers as of the Z or E configuration, where the term "Z" represents substituents on the same side of the carbon-carbon double bond and the term "E" represents substituents on opposite sides of the carbon-carbon double bond. It is also recognized that for structures in which tautomeric forms are possible, the description of one tautomeric form is equivalent to the description of both, unless otherwise specified. Each position in the compounds of the invention may include elements in their natural isotopic abundance. Alternatively, one or more positions in the compound of the invention may include an element enriched in a naturally occurring or a synthetic isotope. For example, one or more positions of the compound of the invention including hydrogen may be enriched with, e.g. , deuterium or tritium . In some embodiments, one or more positions of the compound of the invention including carbon may be enriched with, e.g. , 14C or 13C. In other embodiments, one or more positions of the compound of the invention including nitrogen may be enriched with, e.g., 15N. In certain embodiments, one or more positions of the compound of the invention including oxygen may be enriched with, e.g., 180, 170, or 150. In particular embodiments, one or more positions of the compound of the invention including fluorine may be enriched with, e.g., 18F. In other embodiments, one or more positions of the compound of the invention including carbon may be enriched with , e.g., 32S, 33S, 34S, 35S, or 36S. In yet other
embodiments, one or more positions of the compound of the invention including chlorine may be enriched with, e.g., 35CI, 36CI, or 37CI.
Some abbreviations used herein :
BINAP - 2,2'-bis(diphenylphosphino)-1 , 1 '-binaphthyl ;
f-Bu - ieri-butyl or 1 ,1 -dimethylethyl;
cat - catecholato;
dppb - bis(diphenylphosphino)butane ;
dppf - bis(diphenylphosphino)ferrocene;
Et - ethyl;
Me - methyl;
OAc - acetate;
OMs - mesylate or methanesulfunoate;
ONf - nonaflate or nonafluoro-n-butylsulfonate;
OTf - triflate or trifluoromethanesulfonate;
pin -pinacolato;
i-Pr - isopropyl or 1 -methylethyl; and
n-Pr - n-propyl;
SIMes - 1 ,3-bis(2,4,6-trimethylphenyl)imidazolin-2-ylidene;
SIPr - 1 ,3,-bis(2,6-diisopropylphenyl)imidazolin-2-ylidene; and
THF - tetrahydrofuran. Other Terms
The term "about" is used herein to mean a value that is ±10% of the recited value.
The terms "effective amount" or "amount effective to" or "therapeutically effective amount" mean an amount of a compound of the invention sufficient to produce a desired result, for example, one or more of a decrease in Hsp90 activity (e.g., inhibition of Hsp90), an increase in expression of Hsp70, a decrease in aggregation of Αβ peptide (e.g., inhibition of aggregation of Αβ peptide), an increase in degradation of Αβ peptide, and a decrease in phosphorylation of tau protein, and/or a decrease in, or amelioration of symptoms of, a neurodegenerative disease (e.g., Alzheimer's disease) in a subject upon administration of a composition containing the compound of the invention. The increases and decreases related to administration an effective amount of a compound are relative to levels or symptoms, as applicable, in a subject that has not been administered a compound of the invention or relative to the subject prior to administration of a compound of the invention. The term "element," as used herein, refers to a substance consisting of a single type of atoms, that is, each nucleus of each atom of a single element contains the same number of protons.
The term "neurodegeneration," as used herein, refers to the progressive loss of structure or function of neurons, including death of neurons. The term "neurodegenerative disease" refers to diseases in which neurodegeneration is, at least in part, a cause, symptom, or phenotype.
The terms "patient" and "subject," as used interchangeably herein, refer to any animal (e.g., a mammal, e.g., a human). A subject to be treated according to the methods described herein may be one who has been diagnosed with a neurodegenerative disease, e.g., tauopathy (e.g., Alzheimer's disease), or a proliferative disease as having such a condition or one at risk of developing the condition. Diagnosis may be performed by any method or technique known in the art. One skilled in the art will understand that a subject to be treated according to the present invention may have been subjected to standard tests or may have been identified, without examination, as one at high risk due to the presence of one or more risk factors, e.g., increased total level of tau protein or an increased level of phosphorylated tau protein in a sample (e.g., cerebrospinal fluid) from the subject in comparison to the levels in a sample from a healthy subject.
The term "pharmaceutical composition," as used herein, represents a composition containing a compound described herein, formulated with a pharmaceutically acceptable excipient, and manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal. Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup) ; for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other formulation described herein. The term "pharmaceutically acceptable excipient" or "pharmaceutically acceptable carrier," as used interchangeably herein, refers to any ingredient other than the compounds described herein (e.g., a vehicle capable of suspending or dissolving the active compound) and having the properties of being nontoxic and non-inflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, or waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine,
methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.
The term "pharmaceutically acceptable prodrugs" as used herein, represents those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use.
The term "pharmaceutically acceptable salt," as use herein, represents those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and 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,
pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1 -19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate,
benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, 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, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. The term "pharmaceutically acceptable solvate" as used herein means a compound as described herein wherein molecules of a suitable solvent are incorporated in the crystal lattice. A suitable solvent is physiologically tolerable at the dosage administered. For example, solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. Examples of suitable solvents are ethanol, water (for example, mono-, di-, and tri- hydrates), /V-methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), Λ/,Λ/'-dimethylformamide (DMF), Λ/,Λ/'-dimethylacetamide (DMAC), 1 ,3-dimethyl-2-imidazolidinone (DMEU), 1 ,3-dimethyl-3,4,5,6- tetrahydro-2-(1 H)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like. When water is the solvent, the molecule is referred to as a "hydrate."
The abbreviation "PSA" and the term "polar surface area" of a molecule, as used interchangeably herein, are defined as the surface sum over all polar atoms. The units of PSA are A2 (angstrom squared).
The term "prevent," as used herein, refers to prophylactic treatment or treatment that prevents one or more symptoms or conditions of a disease, disorder, or conditions described herein. Preventive treatment that includes administration of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, can be acute, short-term, or chronic. The doses administered may be varied during the course of preventative treatment.
The term "prodrug," as used herein, represents compounds that are rapidly transformed in vivo to the parent compound of the above formula, for example, by hydrolysis in blood. Prodrugs of the compounds described herein may be conventional esters. Some common esters that have been utilized as prodrugs are phenyl esters, aliphatic (CrCg Or C8-C24) esters, cholesterol esters, acyloxymethyl esters, carbamates, and amino acid esters. For example, a compound that contains an OH group may be acylated at this position in its prodrug form. A thorough discussion is provided in Higuchi and Stella, Prodrugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and Judkins et al., Synthetic Communications 26(23) :4351 -4367, 1996, each of which is incorporated herein by reference. Preferably, prodrugs of the compounds of the present invention are suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use.
The term "proliferative disease," as used herein, refers to both cancer and non-cancer disease. The proliferative disease is one characterized by unregulated proliferation of cells of a certain type (e.g., astrocytes). Preferably, the tumor cells associated with a proliferative disease (e.g., cancer) respond to the inhibition of Hsp90 by apoptosis. Proliferative diseases to be treated using compounds of the invention and according to the methods of the invention may include glioma, meningioma, pituitary adenoma, nerve sheath tumor (e.g., schwannoma or neurofibroma). Proliferative diseases within the scope of the present invention may be a cancer, e.g., acute myeloid leukemia, gastrointestinal stromal tumor, gastric cancer, lung cancer, lymphoma, melanoma, myeloma, non-small cell lung cancer, renal cancer, small cell lung cancer, blast-phase chronic myelogenous leukemia, leukemia, lymphoproliferative disorder, metastatic melanoma, relapsed multiple myeloma, refractory multiple myeloma, myeloproliferative disorders, pancreatic cancer, small intestine cancer, or solid tumor. Preferably, a proliferative disease to be treated using compounds of the invention according to the methods of the invention may include brain tumors (e.g., malignant brain tumors). For example, the brain tumors that may be treated with blood-brain-barrier penetrant compounds of the invention can be glioma or meningioma, in particular, glioma (e.g., glioblastoma), or a malignant version thereof. The cancers that may be treated according to the present invention may also be a cancer that has metastasized to the brain (e.g., lung cancer, breast cancer, melanoma, colon cancer, renal cancer, and thyroid cancer). As used herein, and as well understood in the art, "treatment" is an approach for obtaining beneficial or desired results, e.g., clinical results. Beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions; diminishment of extent of disease, disorder, or condition; stabilized (i.e., not worsening) state of disease, disorder, or condition; preventing spread of disease, disorder, or condition; delay or slowing the progress of the disease, disorder, or condition; amelioration or palliation of the disease, disorder, or condition; and remission (whether partial or total), whether detectable or undetectable. "Palliating" a disease, disorder, or condition means that the extent and/or undesirable clinical manifestations of the disease, disorder, or condition are lessened and/or time course of the progression is slowed or lengthened, as compared to the extent or time course in the absence of treatment.
Brief Description of Drawings
Figure 1 shows a 500 MHz 1 H NMR spectrum of compound 20 in CDCI3.
Figure 2 shows a 500 MHz 1 H NMR spectrum of compound 34 in CDCI3.
Figure 3 shows a 500 MHz 1 H NMR spectrum of compound 36 in CDCI3.
Figure 4 shows five graphs providing IC50 data for compounds 20, 36, 37, and 39 and for a known Hsp90 inhibitor, as measured using a fluorescence polarization assay described in Example 2.
Figure 5A shows a graph comparing relative association/dissociation rates to the concentration of compound 20.
Figure 5B is a photograph of a gel demonstrating increase of the expression of Hsp70 at higher concentrations of compound 20 in a cell based functional assay. Further, Figure 5B shows that expression of Hsp90 remains unchanged relative to variation in the concentration of compound 20.
Figure 6A shows a histogram comparing % cell viability in viable cells (VC), cells contacted with compound 20, and cells contacted with a control compound (JNK inhibitor).
Figure 6B shows a graph comparing pTau231 levels in viable cells (VC), cells contacted with compound 20, and cells contacted with a control compound (JNK inhibitor). Figure 6C shows a graph comparing pTau396 levels in viable cells (VC), cells contacted with compound 20, and cells contacted with a control compound (JNK inhibitor). Figure 7 shows a graph of the concentration of compound 20 in mouse plasma. The data in this graph excludes the plasma level of compound 20 in animal IRN 12.
Figure 8 shows a graph of the concentration of compound 20 in mouse plasma. The data in this graph includes all mouse plasma data points.
Figure 9 shows a graph of the concentration of compound 20 in mouse brain tissue. The data in this graph excludes the plasma level of compound 20 in animal IRN 12.
Figure 10 shows a graph of the concentration of compound 20 in mouse brain tissue. The data in this graph includes all mouse brain tissue data points.
Detailed Description
The invention features novel aminopyrimidines and related compounds having Hsp90 inhibitory activity, pharmaceutical compositions containing them, and their medical use (e.g., treatment of a proliferative disease (e.g., cancer) or a neurodegenerative disease (e.g., a tauopathy)). In particular, the compounds of the invention are capable to penetrate blood-brain-barrier. Therefore, medical use of these compounds encompasses diseases and conditions afflicting mammalian (e.g., human) brain.
Compounds of the Invention
Exemplary compounds of the invention are shown in Table 2.
Table 2.
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
or a pharmaceutically acceptable salt thereof.
A non-limiting example of a pharmaceutically acceptable salt of a compound of the invention is:
Figure imgf000070_0002
Exemplary methods for synthesizing compounds of the invention are described herein.
68 Methods of Preparing Compounds of the Invention
The compounds of the invention can be prepared by processes analogous to those established in the art, for example, by the reaction sequence shown in Scheme 1 . The numbering system used for the general schemes does not necessarily correspond to that employed elsewhere in the description or in the claims.
As shown in Scheme 1 , one strategy to access compounds of the invention (C) is to utilize standard cross-coupling reactions (e.g., Suzuki coupling, Hiyama coupling, Stille coupling, Negishi coupling, Tamao-Kumada coupling, or Murahashi coupling), where a nucleophile A and an electrophile B are coupled in the presence of a metal salt, e.g., a palladium, copper, iron, or nickel salt (e.g., PdCI2, Pd(OAc)2, CuBr, Cul, (CuOTf)2-toluene complex, Fe(OTf)3, FeCI3, FeBr3, NiCI2, or NiBr2). Optionally ligands, e.g., a phosphine (e.g., PPh3, P(2-furyl)3, P(f-Bu)3, dppf, dppb, or BINAP), an /V-heterocyclic carbene (e.g., SIMes or SIPr), or di-pyridine (e.g., 2,2'-bipyridyl or 1 ,10-phenanthroline), may be added to promote the reaction. Alternatively, an organometallic complex, e.g., Pd(PPh3)4 or (dppf)PdCI2, may be employed directly with or without additional ligands. Additives, e.g., tetrabutylammonium fluoride, LiCI, KOAc, or AgOTf, may be added to minimize dehalogenation or to facilitate the cross-coupling reaction. One of skill in the art would be able to determine an appropriate solvent for the reaction through standard screening. Non-limiting examples of solvents used in cross-coupling reactions are water, ethanol, acetone, tetrahydrofuran, toluene, 1 ,4-dioxane, and mixtures thereof. For non-limiting examples of conditions and catalysts that can be used to prepare a compound of the invention according to formula C using cross-coupling chemistry, see Miyaura et al., "Cross-Coupling Reactions: A Practical Guide" in
Topics in Current Chemistry, Springer, 2002, and Nicolaou et al., Angew. Chem. Int. Ed., 44:4442-4489, 2005, which are incorporated herein by reference. Alternatively, a compound of formula A may be an electrophile and have a leaving group X instead of M, while compound of formula B may be a nucleophile and have a metal or metalloid group M instead of X.
Figure imgf000071_0001
M = B(OH)2, B(pin), B(cat), X = hal, e.g.,
SiMe3, SnBu3, -F, -CI, -Br, -I, or
ZnCI, ZnBr, Znl, pseudohalogen,
MgCI, MgBr, Mgl, Li, such as -OMs, -OTf,
where -ONf, or -OP(0)(OR)2,
cat = catecholato where
pin = pinacolato -OMs = mesylate,
-OTf = triflate,
-ONf = nonaflate.
A compound of formula A may be prepared according any method known in the art, e.g., metal-halogen (e.g., lithium-halogen) exchange (with or without a subsequent addition of boron-based, silicon-based, tin- based, zinc-based, or magnesium-based agents), preparation of Grignard reagent, Sandmeyer reaction, or cross-coupling with di-metalloid agent (e.g., Miyaura borylation reaction). Non-limiting examples of preparation of A (Sandmeyer reaction and lithium halogen exchange to prepare E, and Miyaura borylation reaction to prepare G) are shown in Scheme 2.
Figure imgf000072_0001
D E F B2(pin)2 = bis(pinacolato)diboron G
A compound of formula B may be prepared according to any method known in the art, e.g., Biginelli reaction followed by oxidation of the resulting 2-aminodihydropyrimidine derivative. Alternatively, a synthetic approach outlines in Scheme 3 can be used to access a compound of formula B.
Scheme 3. Preparation of electrophile B, e.g., an /V-protected B, i.e. L
Figure imgf000072_0002
H J K L
As shown in Scheme 3, a compound of formula H may undergo condensation with a compound of formula I to give a compound of formula J. The amino group of 2-aminopyrimidine derivative J may then be protected (P = a divalent /V-protecting group, two monovalent /V-protecting groups, or one monovalent /V-protecting group and one hydrogen) to furnish a compound of formula K. The hydroxyl group in the compound of formula K may then be converted to a halogen in the compound of formula L according to any method known in the art, e.g., using dehydrating-halogenating reagents (e.g., POCI3, PCI5, SOCI2, S02CI2, and brominating or iodinating variants thereof). Alternatively, the hydroxyl group of the compound of formula K may be converted into a pseudohalogen in the compound of formula L using reagents, e.g., Tf20, PhNTf2, PhNNf2, or P(0)(OR)2CI, and, optionally, a base (e.g., Et3N, ( Pr)2NEt, or pyridine) and/or catalyst (e.g., 4-dimethylaminopyridine). /V-protecting group P may be removed from the compound L before or after the cross coupling shown in Scheme 1 according to methods known in the art (see, e.g., Greene, Protective Groups in Organic Synthesis, 3rd Edition (John Wiley & Sons, New York, 1999)).
In the reactions described above, it may be necessary to protect reactive functional groups (e.g., hydroxy, amino, thio, or carboxy groups) to avoid their unwanted participation in the reactions. The incorporation of such groups, and the methods required to introduce and remove them are known to those skilled in the art (for example, Greene, supra). The deprotection step may be the final step in the synthesis such that the removal of protecting groups affords compounds of formula (la) as disclosed herein. Starting materials used in any of the schemes above can be purchased or prepared by methods described in the chemical literature, or by adaptations thereof, using methods known by those skilled in the art. The order in which the steps are performed can vary depending on the groups introduced and the reagents used, but would be apparent to those skilled in the art.
Compounds of any of formula (I), (la), (lb), (Va), or (Vb), or any of the intermediates described in the schemes above, can be further derivatized by using one or more standard synthetic methods known to those skilled in the art. Such methods can involve substitution, oxidation or reduction reactions. These methods can also be used to obtain or modify compounds of formula (I), (la), (lb), (Va), or (Vb), or any preceding intermediates by modifying, introducing or removing appropriate functional groups. Particular substitution approaches include alkylation, arylation, heteroarylation, acylation, thioacylation,
halogenation, sulphonylation, nitration, formylation, hydrolysis, and coupling procedures. These procedures can be used to introduce a functional group onto the parent molecule (e.g., the nitration or sulphonylation of aromatic rings) or to couple two molecules together (for example to couple an amine to a carboxylic acid to afford an amide; or to form a carbon-carbon bond between two heterocycles). For example, alcohol or phenol groups can be converted to ether groups by coupling a phenol with an alcohol in a solvent, e.g., tetrahydrofuran in the presence of a phosphine (e.g., triphenylphosphine) and a dehydrating agent (e.g., diethyl-, diisopropyl-, or dimethylazodicarboxylate). Alternatively, ether groups can be prepared by deprotonation of an alcohol, using a suitable base (e.g., sodium hydride) followed by the addition of an alkylating agent (e.g., an alkyl halide or an alkylsulphonate).
In another example, a primary or secondary amine can be alkylated using a reductive alkylation process. For example, the amine can be treated with an aldehyde and a borohydride (e.g., sodium
triacetoxyborohydride, or sodium cyanoborohydride) in a solvent (e.g., a halogenated hydrocarbon, for example, dichloromethane, or an alcohol, for example, ethanol) and, where necessary, in the presence of an acid (e.g., acetic acid).
In another example, -OH groups may be generated from the corresponding ester, acid, acid chloride or aldehyde by reduction with a suitable reducing agent, e.g., a complex metal hydride, e.g., lithium aluminium hydride in a solvent (e.g., tetrahydrofuran).
In another example, hydroxy groups (including phenolic OH groups) can be converted into leaving groups, e.g., halogen atoms or sulphonyloxy groups (e.g., alkylsulphonyloxy, e.g.,
trifluoromethylsulphonyloxy, or arylsuphonyl, e.g., p-toluenesulphonyloxy) using conditions known to those skilled in the art. For example, an aliphatic alcohol can be reacted with thionyl chloride in a halogenated hydrocarbon (e.g., dichloromethane) to afford the corresponding alkylchloride. A base (e.g., triethylamine) can also be used in the reaction.
In another example, ester groups can be converted to the corresponding carboxylic acid by acid- or base- catalysed hydrolysis depending on the nature of the ester group. Acid catalysed hydrolysis can be achieved by treatment with an organic or inorganic acid (e.g., trifluoroacetic acid in an aqueous solvent, or a mineral acid, e.g., hydrochloric acid in a solvent, e.g., dioxan). Base catalysed hydrolysis can be achieved by treatment with an alkali metal hydroxide (e.g., lithium hydroxide in an aqueous alcohol, e.g. methanol).
In another example, aromatic halogen substituents in the compounds may be subjected to halogen-metal exchange by treatment with a base (e.g., a lithium base, e.g., n-butyl or f-butyl lithium) optionally at a low temperature (e.g., -78°C) in a solvent (e.g., tetrahydrofuran) and the mixture may then quenched with an electrophile to introduce a desired substituent. Thus, for example, a formyl group can be introduced by using dimethylformamide as the electrophile. Aromatic halogen substituents can also be subjected to palladium catalysed reactions to introduce groups, e.g., carboxylic acids, esters, cyano, or amino substituents.
In another example, aromatic halogen substituents in the compounds may participate in a range of metal catalysed reactions to introduce alternative functional groups, e.g., amines, amides, ethers, thiols, aryl groups, or heteroaryl groups.
Particular oxidation approaches include dehydrogenations and aromatisation, and the addition of oxygen to certain functional groups. For example, aldehyde groups can be prepared by oxidation of the corresponding alcohol using conditions well known to those skilled in the art. For example, an alcohol can be treated with an oxidising agent (e.g., the Dess-Martin reagent) in a solvent (e.g., a halogenated hydrocarbon, for example dichloromethane). Alternative oxidising conditions can be used, e.g., treatment with oxalyl chloride and an activating amount of dimethylsulphoxide and subsequent quenching by the addition of an amine (e.g., triethylamine). Such a reaction can be carried out in an appropriate solvent (e.g., a halogenated hydrocarbon, for example dichloromethane) and under appropriate conditions (e.g., cooling below room temperature, e.g., to -78°C followed by warming to room temperature). In another example, sulphur atoms can be oxidised to the corresponding sulphoxide or sulphone using an oxidising agent (e.g., a peroxy acid, e.g., 3-chloroperoxybenzoic acid) in an inert solvent (e.g., a halogenated hydrocarbon, e.g., dichloromethane) at around ambient temperature.
Particular reduction approaches include the removal of oxygen atoms from particular functional groups, saturation (or partial saturation) of unsaturated compounds including aromatic rings. For example, primary alcohols can be generated from the corresponding ester or aldehyde by reduction, using a metal hydride (e.g., lithium aluminium hydride or sodium borohydride in a solvent, e.g., methanol). Alternatively, -OH groups can be generated from the corresponding carboxylic acid by reduction, using a metal hydride (e.g., lithium aluminium hydride in a solvent, e.g., tetrahydrofuran). In another example, a nitro group may be reduced to an amine by catalytic hydrogenation in the presence of a metal catalyst (e.g., palladium on a solid support, e.g., carbon) in a solvent (e.g., an ether, e.g., tetrahydrofuran, or an alcohol, e.g., methanol), or by chemical reduction using a metal (e.g., tin or iron) in the presence of an acid (e.g., hydrochloric acid). In a further example an amine can be obtained by reduction of a nitrile, e.g., by catalytic hydrogenation in the presence of a metal catalyst (e.g., palladium on a solid support, e.g., carbon), or Raney nickel in a solvent (e.g., tetrahydrofuran) and under suitable conditions (e.g., cooling to below room temperature, e.g., to -78°C, or heating, e.g., to reflux). Pharmaceutical Compositions
The compounds used in the methods described herein are preferably formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo. Pharmaceutical compositions typically include a compound as described herein and a pharmaceutically acceptable excipient.
The compounds described herein can also be used in the form of the free base, in the form of salts, zwitterions, solvates, or as prodrugs, or pharmaceutical compositions thereof. All forms are within the scope of the invention. The compounds, salts, zwitterions, solvates, prodrugs, or pharmaceutical compositions thereof, may be administered to a patient in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art. The compounds used in the methods described herein may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump, or transdermal administration, and the pharmaceutical compositions formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal, and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.
For human use, a compound of the invention can be administered alone or in admixture with a pharmaceutical carrier selected with regard to the intended route of administration and standard pharmaceutical practice. Pharmaceutical compositions for use in accordance with the present invention thus can be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries that facilitate processing of compounds of Formula (I), (la), (lb), (Va), or (Vb) into preparations which can be used pharmaceutically.
This invention also includes pharmaceutical compositions which can contain one or more
pharmaceutically acceptable carriers. In making the pharmaceutical compositions of the invention, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semisolid, or liquid material (e.g., normal saline), which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, and soft and hard gelatin capsules. As is known in the art, the type of diluent can vary depending upon the intended route of administration. The resulting compositions can include additional agents, e.g., preservatives. The excipient or carrier is selected on the basis of the mode and route of administration. Suitable pharmaceutical carriers, as well as pharmaceutical necessities for use in pharmaceutical formulations, are described in Remington: The Science and Practice of Pharmacy, 21 st Ed., Gennaro, Ed., Lippencott Williams & Wilkins (2005), a well-known reference text in this field, and in the USP/NF (United States Pharmacopeia and the National Formulary). Examples of suitable excipients are lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents, e.g., talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents, e.g., methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. Other exemplary excipients are described in Handbook of Pharmaceutical Excipients, 6th Edition, Rowe et al., Eds., Pharmaceutical Press (2009).
These pharmaceutical compositions can be manufactured in a conventional manner, e.g., by
conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes. Methods well known in the art for making formulations are found, for example, in Remington: The Science and Practice of Pharmacy, 21 st Ed., Gennaro, Ed., Lippencott Williams & Wilkins (2005), and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York. Proper formulation is dependent upon the route of administration chosen. The formulation and preparation of such compositions is well-known to those skilled in the art of pharmaceutical formulation. In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
Dosages
The dosage of the compound used in the methods described herein, or pharmaceutically acceptable salts or prodrugs thereof, or pharmaceutical compositions thereof, can vary depending on many factors, e.g., the pharmacodynamic properties of the compound; the mode of administration; the age, health, and weight of the recipient; the nature and extent of the symptoms; the frequency of the treatment, and the type of concurrent treatment, if any; and the clearance rate of the compound in the animal to be treated. One of skill in the art can determine the appropriate dosage based on the above factors. The compounds used in the methods described herein may be administered initially in a suitable dosage that may be adjusted as required, depending on the clinical response. In general, a suitable daily dose of a compound of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
A compound of the invention may be administered to the patient in a single dose or in multiple doses. When multiple doses are administered, the doses may be separated from one another by, for example, 1 - 24 hours, 1 -7 days, 1 -4 weeks, or 1 -12 months. The compound may be administered according to a schedule or the compound may be administered without a predetermined schedule. An active compound may be administered, for example, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 1 0, 1 1 , or 12 times per day, every 2nd, 3rd, 4th, 5th, or 6th day, 1 , 2, 3, 4, 5, 6, or 7 times per week, 1 , 2, 3, 4, 5, or 6 times per month, or 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , or 12 times per year. It is to be understood that, for any particular subject, specific dosage regimes should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. While the attending physician ultimately will decide the appropriate amount and dosage regimen, an effective amount of a compound of the invention may be, for example, a total daily dosage of, e.g., between 0.05 mg and 3000 mg of any of the compounds described herein. Alternatively, the dosage amount can be calculated using the body weight of the patient. Such dose ranges may include, for example, between 10-1 000 mg (e.g., 50-800 mg). In some embodiments, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1 000 mg of the compound is administered.
In the methods of the invention, the time period during which multiple doses of a compound of the invention are administered to a patient can vary. For example, in some embodiments doses of the compounds of the invention are administered to a patient over a time period that is 1 -7 days; 1 -12 weeks; or 1 -3 months. In other embodiments, the compounds are administered to the patient over a time period that is, for example, 4-1 1 months or 1 -30 years. In other embodiments, the compounds are administered to a patient at the onset of symptoms. In any of these embodiments, the amount of compound that is administered may vary during the time period of administration. When a compound is administered daily, administration may occur, for example, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , or 12 times per day.
Formulations
A compound identified as capable of treating any of the conditions described herein, using any of the methods described herein, may be administered to patients or animals with a pharmaceutically- acceptable diluent, carrier, or excipient, in unit dosage form. The chemical compounds for use in such therapies may be produced and isolated by any standard technique known to those in the field of medicinal chemistry. Conventional pharmaceutical practice may be employed to provide suitable formulations or compositions to administer the identified compound to patients suffering from a disease in which necrosis occurs. Administration may begin before the patient is symptomatic.
Exemplary routes of administration of the compounds (e.g., the compounds having Formula (I), (la), (lb), (Va) or (Vb)), or pharmaceutical compositions thereof, used in the present invention include
oral, sublingual, buccal, transdermal, intradermal, intramuscular, parenteral, intravenous, intra-arterial, intracranial, subcutaneous, intraorbital, intraventricular, intraspinal, intraperitoneal, intranasal, inhalation, and topical administration. The compounds desirably are administered with a pharmaceutically acceptable carrier. Pharmaceutical formulations of the compounds described herein formulated for treatment of the disorders described herein are also part of the present invention. Formulations for Oral Administration
The pharmaceutical compositions contemplated by the invention include those formulated for oral administration ("oral dosage forms"). Oral dosage forms can be, for example, in the form of tablets, capsules, a liquid solution or suspension, a powder, or liquid or solid crystals, which contain the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, pregelatinized starch, microcrystalline cellulose, magnesium aluminum silicate,
carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose, polyvinylpyrrolidone, or polyethylene glycol); and lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Other pharmaceutically acceptable excipients can be colorants, flavoring agents, plasticizers, humectants, buffering agents, and the like.
Formulations for oral administration may also be presented as chewable tablets, as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose,
microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.
Controlled release compositions for oral use may be constructed to release the active drug by controlling the dissolution and/or the diffusion of the active drug substance. Any of a number of strategies can be pursued in order to obtain controlled release and the targeted plasma concentration versus time profile. In one example, controlled release is obtained by appropriate selection of various formulation parameters and ingredients, including, e.g., various types of controlled release compositions and coatings. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, nanoparticles, patches, and liposomes. In certain embodiments, compositions include biodegradable, pH, and/or temperature-sensitive polymer coatings.
Dissolution or diffusion controlled release can be achieved by appropriate coating of a tablet, capsule, pellet, or granulate formulation of compounds, or by incorporating the compound into an appropriate matrix. A controlled release coating may include one or more of the coating substances mentioned above and/or, e.g., shellac, beeswax, glycowax, castor wax, carnauba wax, stearyl alcohol, glyceryl
monostearate, glyceryl distearate, glycerol palmitostearate, ethylcellulose, acrylic resins, dl-polylactic acid, cellulose acetate butyrate, polyvinyl chloride, polyvinyl acetate, vinyl pyrrolidone, polyethylene, polymethacrylate, methylmethacrylate, 2-hydroxymethacrylate, methacrylate hydrogels, 1 ,3 butylene glycol, ethylene glycol methacrylate, and/or polyethylene glycols. In a controlled release matrix formulation, the matrix material may also include, e.g., hydrated methylcellulose, carnauba wax and stearyl alcohol, carbopol 934, silicone, glyceryl tristearate, methyl acrylate-methyl methacrylate, polyvinyl chloride, polyethylene, and/or halogenated fluorocarbon.
The liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils, e.g., cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Formulations for Buccal Administration
Dosages for buccal or sublingual administration typically are 0.1 to 500 mg per single dose as required. In practice, the physician determines the actual dosing regimen which is most suitable for an individual patient, and the dosage varies with the age, weight, and response of the particular patient. The above dosages are exemplary of the average case, but individual instances exist wherein higher or lower dosages are merited, and such are within the scope of this invention.
For buccal administration, the compositions may take the form of tablets, lozenges, etc. formulated in a conventional manner. Liquid drug formulations suitable for use with nebulizers and liquid spray devices and electrohydrodynamic (EHD) aerosol devices will typically include a compound of the invention with a pharmaceutically acceptable carrier. Preferably, the pharmaceutically acceptable carrier is a liquid, e.g., alcohol, water, polyethylene glycol, or a perfluorocarbon. Optionally, another material may be added to alter the aerosol properties of the solution or suspension of compounds of the invention. Desirably, this material is liquid, e.g., an alcohol, glycol, polyglycol, or a fatty acid. Other methods of formulating liquid drug solutions or suspension suitable for use in aerosol devices are known to those of skill in the art (see, e.g., Biesalski, U.S. Pat. No. 5,1 12,598 and Biesalski, U.S. Pat. No. 5,556,61 1 , each of which is herein incorporated by reference). Formulations for Nasal or Inhalation Administration
The compounds may also be formulated for nasal administration. Compositions for nasal administration also may conveniently be formulated as aerosols, drops, gels, and powders. The formulations may be provided in a single or multidose form. In the case of a dropper or pipette, dosing may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension. In the case of a spray, this may be achieved, for example, by means of a metering atomizing spray pump.
The compounds may further be formulated for aerosol administration, particularly to the respiratory tract by inhalation and including intranasal administration. The compound will generally have a small particle size for example on the order of five (5) microns or less. Such a particle size may be obtained by means known in the art, for example by micronization. The active ingredient is provided in a pressurized pack with a suitable propellant, e.g., a chlorofluorocarbon (CFC), for example, dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, or carbon dioxide, or other suitable gas. The aerosol may conveniently also contain a surfactant, e.g., lecithin. The dose of drug may be controlled by a metered valve. Alternatively, the active ingredients may be provided in a form of a dry powder, e.g., a powder mix of the compound in a suitable powder base, e.g., lactose, starch, and starch derivatives, e.g., hydroxypropylmethyl cellulose, and polyvinylpyrrolidine (PVP). The powder carrier will form a gel in the nasal cavity. The powder composition may be presented in unit dose form for example in capsules or cartridges of e.g., gelatin or blister packs from which the powder may be administered by means of an inhaler.
Aerosol formulations typically include a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device. Alternatively, the sealed container may be a unitary dispensing device, e.g., a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal after use. Where the dosage form comprises an aerosol dispenser, it will contain a propellant, which can be a compressed gas, e.g., compressed air or an organic propellant, e.g.,
fluorochlorohydrocarbon. The aerosol dosage forms can also take the form of a pump-atomizer.
Formulations for Parenteral Administration
The compounds described herein for use in the methods of the invention can be administered in a pharmaceutically acceptable parenteral (e.g., intravenous or intramuscular) formulation as described herein. The pharmaceutical formulation may also be administered parenterally (intravenous, intramuscular, subcutaneous or the like) in dosage forms or formulations containing conventional, nontoxic pharmaceutically acceptable carriers and adjuvants. In particular, formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. For example, to prepare such a composition, the compounds of the invention may be dissolved or suspended in a parenterally acceptable liquid vehicle. Among acceptable vehicles and solvents that may be employed are water, water adjusted to a suitable pH by addition of an appropriate amount of hydrochloric acid, sodium hydroxide or a suitable buffer, 1 ,3-butanediol, Ringer's solution and isotonic sodium chloride solution. The aqueous formulation may also contain one or more preservatives, for example, methyl, ethyl or n-propyl p-hydroxybenzoate. Additional information regarding parenteral formulations can be found, for example, in the United States Pharmacopeia-National
Formulary (USP-NF), herein incorporated by reference.
The parenteral formulation can be any of the five general types of preparations identified by the USP-NF as suitable for parenteral administration:
(1 ) "Drug Injection:" a liquid preparation that is a drug substance (e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)), or a solution thereof;
(2) "Drug for Injection :" the drug substance (e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)) as a dry solid that will be combined with the appropriate sterile vehicle for parenteral administration as a drug injection ;
(3) "Drug Injectable Emulsion:" a liquid preparation of the drug substance (e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)) that is dissolved or dispersed in a suitable emulsion medium;
(4) "Drug Injectable Suspension:" a liquid preparation of the drug substance (e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)) suspended in a suitable liquid medium; and
(5) "Drug for Injectable Suspension:" the drug substance (e.g., a compound of Formula (I), (la), (lb), (Va) or (Vb)) as a dry solid that will be combined with the appropriate sterile vehicle for parenteral administration as a drug injectable suspension. Exemplary formulations for parenteral administration include solutions of the compound prepared in water suitably mixed with a surfactant, e.g., hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms. Conventional procedures and ingredients for the selection and preparation of suitable formulations are described, for example, in Remington: The Science and Practice of Pharmacy, 21 st Ed., Gennaro, Ed., Lippencott Williams & Wilkins (2005)and in The United States Pharmacopeia: The National Formulary (USP 36 NF31 ), published in 2013. Formulations for parenteral administration may, for example, contain excipients, sterile water, or saline, polyalkylene glycols, e.g., polyethylene glycol, oils of vegetable origin, or hydrogenated napthalenes. Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene- polyoxypropylene copolymers may be used to control the release of the compounds. Other potentially useful parenteral delivery systems for compounds include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene- 9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel. The parenteral formulation can be formulated for prompt release or for sustained/extended release of the compound. Exemplary formulations for parenteral release of the compound include: aqueous solutions, powders for reconstitution, cosolvent solutions, oil/water emulsions, suspensions, oil-based solutions, liposomes, microspheres, and polymeric gels. Methods of Treatment
The compounds and compositions described herein can be used in the treatment of conditions and disorders in which Hsp90 has been implicated, e.g., cell proliferative disorders, e.g., cancers, neurodegenerative diseases, e.g., tauopathies (e.g., Alzheimer's disease), and infectious diseases. Cell Proliferative Disorders
Hsp90 has emerged as a key therapeutic target for cancer therapy due to the involvement of this multichaperone complex in various pathogenic cellular processes. Hsp90 client proteins include those implicated in: acute myeloid leukemia (Flt-3), breast cancer (HER2), chronic lymphoid leukemia (Zap70), chronic myeloid leukemia (Bcr-Abl or mBcr-Abl), gastrointestinal stromal tumor (c-Kit), gastric cancer (c- Met), glioblastoma (mutant EGFR or c-Met), lung cancer (c-Met), lymphoma (NMP-ALK), melanoma (Raf- 1 /mutant BRAF), myeloma (IGF-1 R/Akt), non-small cell lung cancer (mutant EGFR), renal cancer, (HIF- 1 a), and small cell lung cancer (Akt). The compounds of the invention are particularly useful in the treatment of brain tumors due to their blood-brain-barrier penetrant properties. Brain tumors that may be treated using compounds of the invention include glioma or meningioma, in particular, glioma (e.g., glioblastoma) or neuroblastoma. The brain tumors (e.g., brain cancers) that may be treated using compounds of the invention according to the methods of the invention may include primary tumors (those tumors that originated in the brain) and metastatic tumors (those tumors that originated in tissues other than brain tissues and spread to the brain). Still other cell proliferative disorders that may be treated by the inhibition of Hsp90 include: blast-phase chronic myelogenous leukemia, leukemia,
lymphoproliferative disorder, metastatic melanoma, multiple myeloma (e.g., relapsed or refractory multiple myeloma), myeloproliferative disorders, pancreatic cancer, small intestine cancer, and solid tumor.
Moreover, cancer cells have been shown to be more sensitive to Hsp90 inhibition than non-pathogenic cells. Accordingly, the compounds described herein may be useful treatments for cell proliferative disorders.
Neurodegenerative Diseases
Increased levels of Hsp90 have been implicated in neurodegenerative disorders. For example, aberrant Hsp90 activity has been shown in tauopathies, which are conditions characterized by accumulation of abnormal Tauproteins (e.g., hyperphosphorylated and aggregated Tau). Accordingly, compounds and compositions described herein can be useful for the treatment of neurodegenerative diseases and tauopathies that include Alzheimer's disease (AD), argyrophilic grain disease, amyotrophic lateral sclerosis, corticobasal degeneration, dementia pugislistica, Down's syndrome, familial British dementia, frontal lobe degeneration (dementia lacking distinctive histopathological features), chronic traumatic encephalopathy, traumatic brain injury, frontotemporal dementia (FTD; e.g., fronto-temporal dementia with parkinsonism linked to chromosome 17 (FTDP-17)), hippocampal tauopathy in cerebral aging, myotonic dystrophy of type I , Niemann-Pick disease of type C, Parkinson's disease (e.g., parkinsonism- dementia complex of Guam, Parkinsonism with dementia of Guadeloupe, or postencephalitic
parkinsonism), Pick's disease (PiD), and progressive supranuclear palsy. Accordingly, the compounds described herein may be useful in treating a neurodegenerative disorder, e.g., tauopathy (e.g.,
Alzheimer's disease). Infectious Diseases
Hsp90 has emerged as a therapeutic target for treating infectious diseases, e.g., viral infections, fungal infections, and bacterial infections. Many pathogens (e.g., viruses, fungi, and bacteria) rely on Hsp90- dependent processes (see, e.g., Geller et al., Biochim. Biophys. Acta - Mol. Cell Res., 1823:698-706, 2012; the disclosure of which is incorporate herein in its entirety). Thus, inhibition of Hsp90 provides a therapeutic benefit to a patient afflicted with an infection that relies on the activity of Hsp90. For example, an Hsp90 inhibitor (geldanamycin) was shown to delay the growth of influenza virus in cell culture. Other viruses that rely on Hsp90 dependent processes include those belonging to the families: Herpesviridae (e.g., herpes simplex virus-1 , herpes simplex virus-2, herpes herpesvirus-5, Kaposi's sarcoma-associated herpesvirus, varicella zoster virus, or Epstein-Barr virus), Polyomaviridae (e.g., SV40), Poxviridae (e.g., vaccinia virus), Reoviridae (e.g., rotavirus), Birnaviridae (e.g., infectious bursal disease virus),
Picornaviridae (e.g., poliovirus, rhinovirus, or coxsackievirus), Flaviviridae (e.g., hepatitis C virus or dengue virus), Arenaviridae (e.g., lymphocytic choriomeningitis virus), Hepeviridae (e.g., Hepatitis E virus), Rhabdoviridae (e.g., vesicular stomatitis virus), Paramoxyviridae (e.g., human parainfluenza virus 2, human parainfluenza virus 3, SV5, SV41 , measles virus, or Sendai virus), Bunyaviridae (e.g., La Crosse virus), Orthomoxyviridae (e.g., influenza A virus), Filoviridae (e.g., Ebola virus), Retroviridae (e.g., HTLV1 or HIV1 ), and Hepadnaviridae (e.g., hepatitis B virus). Hsp90 inhibitors have also been used in vivo for the treatment of fungal infectious diseases, e.g., treatment of Candida albicans, Aspergillus fumigates, or Pneumocystis jiroveci. Moreover, Hsp90 inhibitors are also useful in the treatment of bacterial infections, e.g., mycobacteria, anthrax, or bacterial pneumonia. A discussion of the diseases that may be treated with Hsp90 inhibitors is provided in the U.S. Patent Application Publication
201 1 /0201587, the disclosure of which is incorporated herein by reference in its entirety. Therefore, the compounds of the invention may be used according to the method of the invention to treat infectious diseases, e.g., viral infections, fungal infections, or bacterial infections.
Inflammatory and Autoimmune Diseases, Allergy
Hsp90 has been shown to play a role in antigen presentation, activation of lymphocytes, macrophages, maturation of dendritic cells, and in the enhanceosome mediated induction of inflammation. Hsp90 inhibition is associated with blockage of components of inflammation, e.g., reduction of cytokine and NO production, as well as blockage of NFKB nuclear translocation. Further, considerable body of work indicates that chaperones, such as Hsp90, may be capable of inducing the production of proinflammatory cytokines by the monocyte-macrophage system and the activation and maturation of dendritic cells via the TLR2-and 4-signal transduction pathways. Thus, Hsp90 apparently can function as a potent activator of the innate immune system. Indeed, elevated levels of Hsp90 were detected in the serum of systemic lupus erythematosus patients. Autoantibodies and cells reactive to Hsp have been detected in patients with rheumatoid arthritis. Antiinflammatory effect of inhibiting Hsp90 was also observed to reduce airway inflammation in murine model of asthma. The compounds of the invention may be applicable to the treatment of inflammatory or autoimmune diseases in a patient. Moreover, anti-inflammatory effect of Hsp90 inhibition can have therapeutic application in the treatment of allergies. Thus, the compounds of the invention may be used in the treatment of allergy.
Cardiovascular Diseases
Hsp90 has recently been implicated in etiology of cardiovascular disorders, such as atherosclerosis and cardiomyopathy. Thus, the compounds of the present invention may be applicable to the treatment of cardiovascular diseases (e.g., atherosclerosis or cardiomyopathy).
Kits of the Invention
The present invention also provides kits containing (i) a pharmaceutical composition of the invention, and (ii) instructions for use of the pharmaceutical composition to treat a disorder in a mammal caused by the action of Hsp90, e.g., a neurodegenerative disorder, a proliferative disorder, or an infectious disease, as described herein. Kits of the invention may include instructions explaining how a practitioner (e.g., a physician, a nurse, a care-giver, or a patient) may administer the composition contained therein. The pharmaceutical composition within the kit of the invention may be provided in a container (e.g., a bottle, an ampule, a tube, or a blister pack). Furthermore, the kits may also include additional components, e.g., instructions or administration schedules for a patient suffering from a neurodegenerative disease or a proliferative disease, and optionally, a device(s) for administering the pharmaceutical composition (e.g., a syringe).
The following examples are meant to illustrate the invention. They are not meant to limit the invention in any way. Examples
Example 1. Synthesis of the Compounds of the Invention
Figure imgf000084_0001
Scheme 1. Reagents and conditions: (a) Pd(PPh3)4, Na2C03, dioxane/H20 (9/5), 90 aC, 72%.
4-(5,7-Dichloro-2,3-dihydrobenzofuran-4-yl)-5H-pyrrolo[3,2- /]pyrimidin-2-amine (20). A mixture of 5,7-dichloro-2,3-dihydro-1 -benzofuran-4-yl boronic acid (233 mg, 1 .0 mmol), 4-chloro-5/-/-pyrrolo[3,2- d]pyrimidin-2-amine (219 mg, 1 .3 mmol), sodium carbonate (318 mg, 3.0 mmol), palladium
tetrakis(triphenylphosphine), and dioxane/water (9/5, 14 ml) was stirred at 90 SC under argon for 20 h, then cooled down to room temperature, quenched with brine (25 ml), and extracted with ethyl acetate (30 ml x 2). The combined organic layers were dried over sodium sulfate and concentrated. The residue was purified by chromatography on silica gel using cyclohexane/ethyl acetate (100/0 to 30/70, 15 min) to give a product as a white solid (230 mg, 72%). 1 H NMR (500 MHz, CDCI3): δ 8.01 (s, 1 H), 7.45 (t, J = 3 Hz, 1 H), 7.34 (s, 1 H), 6.46 (m, 1 H), 4.83 (s, 2H), 4.71 (m, 2H), 3.60 (m, 1 H), 2.94 (m, 1 H); LCMS [M + H]+: 321 .0 (calcd for [C14H10CI2N4O + H]+: 321 .0).
The following compounds of the invention have been prepared according to the procedure described herein.
Figure imgf000084_0002
4-(5,7-Dichloro-2,3-dihydrobenzofuran-4-yl)-/V,6-dimethylpyrimidin-2-amine(3). 1 H NMR (500 MHz, CDCI3): δ 7.25 (s, 1 H), 6.57 (s, 1 H), 5.07 (d, J = 10 Hz, 1 H), 4.68 (t, J = 9 Hz, 2H), 3.29 (t, J = 9 Hz, 2H), 3.02 (d, J = 10 Hz, 3H), 2.40 (s, 3H).
Figure imgf000084_0003
4-(5,7-Dichloro-2,3-dihydrobenzofuran-4-yl)-W,W,6-trimethylpyrimidin-2-amine(4) Ή NMR (500 MHz, CDCI3): δ 7.25 (s, 1 H), 6.51 (s, 1 H), 4.67 (t, J = 9 Hz, 2H), 3.30 (t, J = 9 Hz, 2H), 3.20 (s, 6H), 2.40 (s, 3H).
Figure imgf000085_0001
4-(5,7-Dichloro-2,3-dihydrobenzofuran-4-yl)-6-methylpyrimidin-2-amine(5). 1 H NMR (500 MHz, CDCI3): δ 7.27 (s, 1 H), 6.64 (s, 1 H), 5.04 (s, 2H), 4.68 (t, J = 9 Hz, 2H), 3.25 (t, J = 9 Hz, 2H), 2.42 (s, 3H).
Figure imgf000085_0002
4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-6-ethylpyrimidin-2-amine (6). Ή NMR (500 MHz, CDCI3): δ 7.27 (s, 1 H), 6.65 (s, 1 H), 5.05 (s, 2H), 4.68 (t, J = 9 Hz, 2H), 3.26 (t, J = 8.5 Hz, 2H), 2.68 (q, J = 8 Hz, 2H), 1 .29 (t, J = 7.5 Hz, 3H); LCMS [M + H]+: 310.1 (calcd for [C14H13CI2N30 + H]+: 31 0.04).
Figure imgf000085_0003
4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-6-isopropylpyrimidin-2-amine (7). Ή NMR (500 MHz, CDCI3): δ 7.28 (s, 1 H), 6.66 (s, 1 H), 5.04 (s, 2H), 4.69 (t, J = 9 Hz, 2H), 3.27 (t, J = 9 Hz, 2H), 2.87 (m, 1 H), 1 .28 (d, J = 7 Hz, 6H); LCMS [M + H]+: 324.02 (calcd for [C15H15CI2N30 + H]+: 324.06).
Figure imgf000085_0004
4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-6-methoxypyrimidin-2-amine (8). 1 H NMR (500 MHz, CDCI3): δ 7.28 (s, 1 H), 6.19 (s, 1 H), 4.98 (s, 2H), 4.68 (t, J = 8.5 Hz, 2H), 3.94 (s, 3H), 3.25 (t, J = 9 Hz, 2H); LCMS [M + H]+: 31 1 .87 (calcd for [C13H1 1 CI2N302 + H]+: 312.02).
Figure imgf000086_0001
4-(2,4-Dichloro-5-methoxyphenyl)-5,6-dimethylpyrimidin-2-amine(9). 1 H NMR (500 MHz, CDCI3): δ 7.47 (s, 1 H), 6.83 (s, 1 H), 4.88 (s, 2H), 3.90 (s, 3H), 2.42 (s, 3H), 1 .95 (s, 3H).
Figure imgf000086_0002
4-(5,7-Dichloro-2,3-dihydrobenzofuran-4-yl)-5,6-dimethylpyrimidin-2-amine(10). Ή NMR (500 MHz, CDCI3): δ 7.25 (s, 1 H), 4.89 (s, 2H), 4.69 (t, J = 9 Hz, 2H), 3.25 (m, 1 H), 2.88 (m, 1 H), 2.42 (s, 3H), 1 .93 (s, 3H).
Figure imgf000086_0003
4-(2-chloro-4-fluoro-5-methoxyphenyl)-6,7-dihydro-5H-cyclopenta[d]pyrimidin-2-amine (11). H
NMR (500 MHz, CDCI3): δ 7.20 (d, J = 10.5 Hz, 1 H), 6.94(d, J = 9 Hz, 1 H), 4.99 (s, 2H), 3.90 (s, 3H), 2.91 (t, J = 8 Hz, 2H), 2.72 (t, J = 7.5 Hz, 2H), 2.08 (m, 2H); LCMS [M + H]+: 294.2 (calcd for [C14H13CIFN30 + H]+: 294.07).
Figure imgf000086_0004
4-(2,4-Dichloro-5-methoxyphenyl)-6,7-dihydro-5H-cyclopenta[ /]pyrimidin-2-amine(12). H NMR
(500 MHz, CDCI3): δ 7.47 (s, 1 H), 6.90 (s, 1 H), 4.98 (s, 2H), 3.91 (s, 3H), 2.92 (t, J = 7.5 Hz, 2H), 2.72 (t, J = 7.5 Hz, 2H), 2.08 (m, 2H).
Figure imgf000086_0005
4-(5,7-Dichloro-2,3-dihydrobenzofuran-4-yl)-6,7-dih^
1 H NMR (500 MHz, CDCI3): δ 7.25 (s, 1 H), 4.69 (m, 2H), 3.38 (m, 1 H), 2.99-2.81 (m, 4H), 2.49 (m, 1 H), 2.17-2.00 (m, 2H).
Figure imgf000087_0001
4-(2,4-dichloro-5-(2-(dimethylamino)ethoxy)phenyl)-6,7-dihydro-5H-cyclopenta[d]pyrimidin-2- amine (14). 1 H NMR (500 MHz, CDCI3): δ 7.46 (s, 1 H), 6.90 (s, 1 H), 4.97 (s, 2H), 4.13 (t, J = 6 Hz, 2H), 2.91 (t, J = 8 Hz, 2H), 2.80 (t, J = 6 Hz, 2H),2.71 (t, J = 7.5 Hz, 2H), 2.36 (s, 6H), 2.08 (m, 2H); LCMS [M + H]+: 367.1 (calcd for [C1 7H20CI2N4O + H]+: 367.10).
Figure imgf000087_0002
4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-5,6,7,8-tetrahydroquinazolin-2-amine (15). 1 H NMR
(500 MHz, CDCI3): δ 7.28 (s, 1 H), 4.88 (s, 2H), 4.69 (t, J = 9 Hz, 2H), 3.21 (m, 1 H), 2.89 (m, 1 H), 2.78 (m, 2H), 2.44 (m, 1 H), 2.15 (m, 1 H), 1 .86 (m, 2H), 1 .72 (m, 2H); LCMS [M + H]+: 336.1 (calcd for
[C16H15CI2N30 + H]+: 336.06).
Figure imgf000087_0003
4-(2,4-Dichloro-5-methoxyphenyl)-5H-pyrrolo[3,2- /]pyrimidin-2-amine (19). Ή NMR (500 MHz, CDCI3): δ 8.15 (s, 1 H), 7.54 (s, 1 H), 7.46 (m, 1 H), 7.15 (s, 1 H), 6.45 (m, 1 H), 4.86 (s, 2H), 3.94 (s, 3H).
Figure imgf000087_0004
4-(5,7-Dichloro-2,3-dihydrobenzofuran-4-yl)-5H^yrrolo[3,2- /]pyrimidin-2-amine (20). Ή NMR (500 MHz, CDCI3): δ 8.01 (s, 1 H), 7.45 (t, J = 3 Hz, 1 H), 7.34 (s, 1 H), 6.46 (m, 1 H), 4.83 (s, 2H), 4.71 (m, 2H), 3.60 (m, 1 H), 2.94 (m, 1 H); LCMS [M + H]+: 321 .0 (calcd for [C14H10CI2N4O + H]+: 321 .0). The 1 H NMR spectrum is shown in Figure 1 .
Figure imgf000088_0001
4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-5-methyl-5H^yrrolo[3,2-d]pyrimidin-2-am (21). 1 H
NMR (500 MHz, CDCI3): δ 7.32 (s, 1 H), 7.22 (d,J = 3 Hz, 1 H), 6.36(d, J = 3 Hz, 1 H), 4.81 (s, 2H), 4.72 (m, 2H), 3.40 (s, 3H), 3.34 (m, 1 H), 2.91 (m, 1 H); LCMS [M + H]+: 335.0 (calcd for [C15H12CI2N40 + H]+: 335.04).
4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-5-ethyl-5H-pyrrolo[3,2-d]pyrimidin-2-amine (22). H
NMR (500 MHz, CDCI3): 5 7.33 (m, 2H), 6.40 (d, J = 2.5 Hz, 1 H), 4.80 (s, 2H), 4.70 (m, 2H), 3.72 (d, J = 7 Hz, 2H), 3.33 (m, 1 H), 2.91 (m, 1 H), 1 .14 (t, J = 7.5 Hz, 3H); LCMS [M + H]+: 349.1 (calcd for
[C16H14CI2N40 + H]+: 349.05).
Figure imgf000088_0003
Ethyl-2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidine-6-carboxylate (23). 1 H NMR (500 MHz, CDCI3): δ 7.54 (s, 1 H), 7.35 (s, 1 H), 5.32 (s, 2H), 4.70 (m, 2H), 4.37 (m, 2H), 3.35 (m, 1 H), 2.92 (m, 1 H), 1 .38 (t, J = 7 Hz, 3H); LCMS [M + H]+: 41 0.0 (calcd for [C17H13CI2N303S + H]+: 410.01 ).
Figure imgf000088_0004
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-ethylthieno[2,3-d]pyrimidi
(24). 1 H NMR (500 MHz, CDCI3): δ 7.34 (s, 1 H), 7.21 (s, 1 H), 5.91 (s, 1 H), 5.28 (s, 2H), 4.70 (m, 2H), 4.48 (m, 2H), 3.36 (m, 1 H), 2.91 (m, 1 H), 1 .26 (t, J = 3.5 Hz, 3H); LCMS [M + H]+: 409.0 (calcd for
[C17H14CI2N402S + H]+: 409.02).
Compounds 34 and 36 were prepared according to methods known in the art, e.g., those described herein. The 1 H NMR spectra (CDCI3) for compounds 34 and 36 are provided in Figures 2 and 3, respectively. Compounds 40-48 can be prepared according to methods known in the art, e.g., those described herein.
Figure imgf000089_0001
4-chloro-6-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)pyrimidin-2-amine (49). 1 H NMR (400 MHz, DMSO-d6): δ = 7.48 (s, 1 H), 7.30 (s, 2 H), 6.78 (s, 1 H), 4.64 (t, J = 8.8 Hz, 2 H), 3.20 (t, J = 8.8 Hz, 2 H). LCMS: m/z calcd for C12H8CI3N30 [M+H]+: 31 6.0; found: 31 6.0.
Figure imgf000089_0002
50
4-trifluoromethyl-6-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)pyrimidin-2-amine (50). 1 H NMR (400 MHz, DMSO-d6): δ 7.520 (br s, 2H, NH2), 7.093 (s, 1 H), 4.655 (t, 2H, J = 8.8 Hz), 3.547 (s, 1 H), 3.221 (t, 2H, J = 8.8 Hz). LCMS: m/z calcd for C13H8CI2F3N30 [M+H]+: 350.1 ; found: 350.0.
Figure imgf000089_0003
51 4-thiomethyl-6-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)pyrimidin-2-amine (51). 1 H NMR (400 MHz, DMSO-d6): δ 7.438 (s, 1 H), 6.815 (br s, 2H, NH2), 6.549 (s, 1 H), 4.633 (t, 2H, J = 8.8 Hz), 3.178 (t, 2H, J = 8.8 Hz), 2.453 (s, 3H). LCMS: m/z calc +H]+: 328.2; found: 328.1 .
Figure imgf000090_0001
52
4-chloro-6-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)pyrimidine-2,5-diamine (52). 1 H NMR (400 MHz, DMSO-d6): δ = 7.46 (s, 1 H), 6.25 (s, 2 H), 4.67 (t, J = 8.8 Hz, 2 H), 4.30 (s, 2 H), 3.15-3.07 m, 2 H). HPLC/MS: m/z calcd for C12H9CI3N40 [M+H]+: 331 .0; found: 331 .1 .
Figure imgf000090_0002
53
2-amino-6-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)pyrimidin-4-ol (53). To a flask containing dioxane:1 N NaOH aq. (50:50; 1 ml_:1 mL) were added 4-chloro-6-(5,7-dichloro-2,3-dihydrobenzofuran-4- yl)pyrimidin-2-amine (49) (20 mg, 0.063 mmol), and DABCO (8 mg, 0.069 mmol), at rt. The reaction was subsequently heated at 80 °C. The reaction was cooled down, acidified by addition of 1 N HCI aq. (2 mL), taken up in ethyl acetate (5 mL) and washed with brine (3 x 5 mL). The organic layer was dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product was obtained in 77 % yield as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ = 7.39 (s, 1 H), 5.57 (s, 1 H), 4.62 (t, J = 8.8 Hz, 2 H), 3.20 (t, J = 8.8 Hz, 2 H). HPLC/MS: m/z calcd for C12H9CI2N302 [M+H]+: 298.0; found: 298.1 .
Figure imgf000090_0003
54
Methyl 2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-5H-pyrrolo[3,2-d]pyrimidine-5- carboxylate (54). To a flask containing 4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-5H-pyrrolo[3,2- d]pyrimidin-2-amine (20) (20 mg, 0.062 mmol) in DCM (1 mL) were added dry K2CO3 (30 mg, 0.22 mmol), and methyl chloroformate (0.014 mL, 0.186 mmol), at 0 °C. The reaction was allowed to stir for 8 h at rt. Afterwards, the reaction was quenched by addition of 1 N NaOH aq. (1 mL) and stirred at rt for 1 h. The reaction was taken up in DCM (10 mL) and washed with sat. NaHC03 aq. (1 0 mL). The organic layer was dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product was obtained in 66 % yield as a yellow solid.
1 H NMR (400 MHz, CDCI3): δ = 7.61 (s, 1 H), 7.23 (s, 1 H), 6.51 (s, 1 H), 4.63 (br s, 2 H), 3.77 (s, 3 H), 3.65 (br s, 1 H), 2.85 (br s, 1 H). HPLC/MS: m/z calcd for C16H12CI2N403 [M+H]+: 379.0; found: 379.1 .
Figure imgf000091_0001
1-(2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-y^
(55). To a flask containing 4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-5H-pyrrolo[3,2-d]pyrimidin-2-amine (20 mg, 0.062 mmol) in DCM (1 mL) were added dry K2C03 (30 mg, 0.22 mmol), and acetyl chloride (0.006 mL, 0.074 mmol), at 0 °C. The reaction was allowed to stir for 8 h at rt. Afterwards, the reaction was quenched by addition of 1 N NaOH aq. (1 mL) and stirred at rt for 1 h. The reaction was taken up in DCM (10 mL) and washed with sat. NaHC03 aq. (10 mL). The organic layer was dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product was obtained in 81 % yield as a yellow solid.
1 H NMR (400 MHz, CDCI3): δ = 7.57 (s, 1 H), 7.23 (s, 1 H), 6.55 (s, 1 H), 4.63 (br s, 2 H), 3.45 (br s, 1 H), 3.35 (s, 3 H), 2.85 (br s, 1 H). HPLC/MS: m/z calcd for C16H12CI2N402 [M+H]+: 363.0; found: 363.1 .
Figure imgf000091_0002
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-5H-pyrrolo[3,2-d]pyrimidine-7-carbaldehyde (56). To a flask containing dry THF (2 mL), were added dry DMF (0.1 mL) and POCI3 (0.015 mL, 0.16 mmol), at 0 °C. Reaction was stirred at 0 °C for 30 min, under argon, upon which 4-(5,7-dichloro-2,3- dihydrobenzofuran-4-yl)-5H-pyrrolo[3,2-d]pyrimidin-2-amine (20 mg, 0.062 mmol) in THF (1 mL) was added dropwise. The reaction was allowed to stir for 8 h warming to rt. To the reaction was then added 1 N NaOH aq. (2 mL) and heated to 80 °C for 1 h. The reaction was cooled to rt and taken up in EtOAc (20 mL) and washed with sat. NaHC03 aq. (20 mL). The organic layer was dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product was obtained in 58 % yield as a white solid.
1 H NMR (400 MHz, CDCI3): δ = 10.05 (s, 1 H), 7.96 (s, 1 H), 7.24 (s, 1 H), 4.63 (br s, 2 H), 3.31 (br s, 1 H), 2.85 (br s, 1 H). HPLC/MS: m/z calcd for C15H10CI2N4O2 [M+H]+: 349.0; found: 349.1 .
Figure imgf000092_0001
57
7-bromo-4-(2,4-dichloro-5-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-2-amine (57). To a flask containing 4-(2,4-dichloro-5-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-2-amine (20 mg, 0.065 mmol) in AcOH:tBuOH (50:50, 0.5 ml_:0.5 mL) were added LiBr (18 mg, 0.22 mmol), and Br2 (0.01 1 mL, 0.22 mmol), at 0 °C. The reaction was allowed to stir for 8 h at rt. Afterwards, the reaction was taken up in EtOAc (20 mL) and washed with sat. NaHC03 aq. (3 χ 20 mL), and Na2S203 (10 % wt. aq., 20 mL). The organic layer was dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product was obtained in 56 % yield as a yellow solid.
1 H NMR (400 MHz, DMSO-d6): δ = 7.75 (s, 1 H), 7.72 (s, 1 H), 7.26 (s, 1 H), 6.24 (s, 2 H), 3.86 (s, 3 H). HPLC/MS: m/z calcd for C13H9BrCI2N40 [M+H]+: 386.9; found: 387.0.
General procedure for synthesis of 2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3- d]pyrimidine-6-carboxamides:
Figure imgf000092_0002
Step 1 : Synthesis of precursor 2,5-dioxopyrrolidin-1-yl 2-amino-4-(5,7-dichloro-2,3- dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidine-6-carboxylate. To a flask containing degassed DMF:H20 (50:50; 2 mL:2 mL) were added ethyl 2-amino-4-chlorothieno[2,3-d]pyrimidine-6-carboxylate (100 mg, 0.39 mmol), (5,7-dichloro-2,3-dihydrobenzofuran-4-yl)boronic acid (91 mg, 0.39 mmol), NaHC03 (82 mg, 0.98 mmol), and Pd(PPh3)4 (22 mg, 0.02 mmol), at rt. The reaction was subsequently heated at 80 °C for 8 h under argon. The reaction was cooled down, taken up in ethyl acetate (20 mL) and washed with brine (3 χ 20 mL). The organic layer was dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product ethyl 2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidine-6- carboxylate was obtained in 38 % yield as a yellow solid. Ethyl 2-amino-4-(57-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidine-^ (60 mg,
0.15 mmol) was dissolved in THF (1 mL) and to the flask was added 1 N NaOH aq. (1 mL). The reaction was stirred for 8 h at rt. Subsequently, the solution was cooled to 0 °C and acidified by addition of 1 N HCI aq. (2 mL), resulting in formation of white precipitate, which was filtered and dried, affording 2-amino- 4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidine-6-carboxylic acid in quantitative yield, without further purification. The filtrand (57 mg, 0.15 mmol) was dissolved in dry DMF (1 mL), cooled to 0 °C, and to the reaction vessel were added N-hydroxysuccinimide (23 mg, 0.2 mmol) and 1 -ethyl-3-(3- dimethylaminopropyl)carbodiimide HCI (38 mg, 0.2 mmol). The reaction was left to stir warming to rt over 8h. The solution was taken up into DCM (20 mL) and washed with sat. NH4CI aq. (3 χ 20 mL). The organic layer was dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product 2,5- dioxopyrrolidin-1 -yl 2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidine-6- carboxylate was obtained as a white solid in 88% yield.
Figure imgf000093_0001
To a flask containing the 2,5-dioxopyrrolidin-1 -yl 2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4- yl)thieno[2,3-d]pyrimidine-6-carboxylate (1 0 mg, 0.02 mmol) was added dry DMF (0.5 mL), followed by addition of an amine (3 eq., 0.06 mmol), (e.g. ammonia, primary amine, or secondary amine). The reaction was allowed to stir at rt for 12 h, upon which it was taken up in DCM (5 mL) and washed with sat. NH4CI aq. (3 x 5 mL). The organic layer was dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The resulting amides were obtained in good to excellent yields.
Figure imgf000093_0002
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-(2-(pyrrolidin-1-yl)ethyl)thieno[2,3- d]pyrimidine-6-carboxamide (58). 1 H NMR (400 MHz, DMSO-d6): δ = 8.79 (s, 1 H), 7.61 (s, 1 H), 7.58 (s, 1 H), 7.28 (s, 2 H), 4.67 (t, J = 8.8 Hz, 2 H), 3.44-3.1 1 (m, 6 H), 2.90-2.75 (m, 4 H), 1 .77-1 .73 (m, 4 HPLC/MS: m/z calcd for C21 H21 CI2N502S [M+H]+: 478.1 ; found: 478.1
Figure imgf000094_0001
2-amino-N-(2-cyanoethyl)-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimid carboxamide (59). 1 H NMR (400 MHz, DMSO-d6): δ = 8.94 (t, J = 5.6 Hz, 1 H), 7.63 (s, 1 H), 7.60 (s, 1 H), 7.34 (s, 2 H), 4.69 (t, J = 8.8 Hz, 2 H), 3.46-3.41 (m, 2 H), 3.23-3.15 (m, 1 H), 3.03-2.97 (m, 1 H), 2.76 (t, J = 6.4 Hz, 2 H). HPLC/MS: m/z calcd for C18H13CI2N502S [M+H]+: 434.0; found: 434.1 .
Figure imgf000094_0002
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-(2-hydroxyethyl)thieno[2,3-d]pyrimidine-6- carboxamide (60). 1 H NMR (400 MHz, DMSO-d6): δ = 8.57 (t, J = 5.6 Hz, 1 H), 7.59 (s, 2 H), 7.26 (s, 2 H), 4.73 (t, J = 5.6 Hz, 1 H), 4.67 (t, J = 8.8 Hz, 2 H), 3.46-3.41 (m, 2 H), 3.26-3.12 (m, 3 H), 3.03-2.94 (m, 1 H). HPLC/MS: m/z calcd for C17H14CI2N403S [M+H]+: 425.0; found: 425.1 .
Figure imgf000094_0003
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-(2,2,2-trifluoroethyl)thieno[2,3-d]pyrimidi 6-carboxamide (61). 1 H NMR (400 MHz, DMSO-d6): δ = 9.18 (t, J = 6.0 Hz, 1 H), 7.72 (s, 1 H), 7.63 (s, 1 H), 7.38 (s, 2 H), 4.69 (t, J = 8.8 Hz, 2 H), 4.12-4.03 (m, 2 H), 3.26-3.18 (m, 1 H), 3.03-2.94 (m, 1 H). HPLC/MS: m/z calcd for C17HH CI2F3N4O2S [M+H]+: 463.0; found: 463.1 .
Figure imgf000095_0001
2-amino-N-cyclopropyl-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidine carboxamide (62). 1 H NMR (400 MHz, DMSO-d6): δ = 8.54 (s, 1 H), 7.61 (s, 1 H), 7.53 (s, 1 H), 7.29 (s, 2 H), 4.68 (t, J = 8.8 Hz, 2 H), 3.26-3.18 (m, 1 H), 3.03-2.94 (m, 1 H), 2.81 -2.71 (m, 1 H), 0.75-0.61 (m, 2 H), 0.55-0.40 (m, 2H). HPLC/MS: m/z calcd for C18H14CI2N402S [M+H]+: 421 .0; found: 421 .1 .
Figure imgf000095_0002
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-methylthieno[2,3-d]pyrimidine-6- carboxamide (63). 1 H NMR (400 MHz, DMSO-d6): δ = 8.62-8.59 (m, 1 H), 7.61 (s, 2 H), 7.27 (s, 2 H), 4.64 (t, J = 8.8 Hz, 2 H), 3.24 (s, 3 H), 3.23-3.18 (m, 1 H), 3.03-2.91 (m, 1 H). HPLC/MS: m/z calcd for C16H12CI2N402S [M+H]+: 395.0; found: 395.1 .
Figure imgf000095_0003
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-(2-me
carboxamide (64). 1 H NMR (400 MHz, DMSO-d6): δ = 8.51 -8.41 (m, 1 H), 7.59 (s, 1 H), 7.50 (s, 1 H), 7.26 (s, 2 H), 4.67 (t, J = 8.8 Hz, 2 H), 3.23-3.10 (m, 4 H), 3.03-2.91 (m, 2 H), 2.72-2.67 (m, 3 H).
HPLC/MS: m/z calcd for C18H16CI2N403S [M+H]+: 439.0; found: 439.1 .
Figure imgf000096_0001
2-amino-N-cyclobutyl-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidi carboxamide (65). 1 H NMR (400 MHz, DMSO-d6): δ = 8.68 (d, J = 6.8 Hz, 1 H), 7.61 (s, 1 H), 7.56 (s, 1 H), 7.26 (s, 2 H), 4.67 (t, J = 8.8 Hz, 2 H), 4.33-4.26 (m, 1 H), 3.23-3.18 (m, 1 H), 3.03-2.91 (m, 1 H), 2.16-1 .92 (m, 2 H), 1 .70-1 .60 (m, 1 H), 1 .33-1 .20 (m, 2 H), 0.90-0.78 (m, 1 H). HPLC/MS: m/z calcd for C19H16CI2N402S [M+H]+: 435.0; found: 435.1 .
Figure imgf000096_0002
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N,N-dimethylthieno[2,3-d]pyrimidine-6- carboxamide (66). 1 H NMR (400 MHz, DMSO-d6): δ = 7.54 (s, 1 H), 7.23 (s, 2 H), 7.15 (s, 1 H), 4.66 (t, J = 8.8 Hz, 2 H), 3.31 (s, 3 H), 3.23-2.90 (m, 5 H). HPLC/MS: m/z calcd for C17H14CI2N402S [M+H]+: 409.0; found: 409.1 .
Figure imgf000096_0003
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-(oxetan-3-yl)thieno[2,3-d]pyrimidine-6- carboxamide (67). 1 H NMR (400 MHz, DMSO-d6): δ = 9.17 (d, J = 6.8 Hz, 1 H), 7.64 (s, 1 H), 7.62 (s, 1 H), 7.31 (s, 2 H), 4.97-4.90 (m, 1 H), 4.75-4.65 (m, 4 H), 4.55-4.45 (m, 2 H), 3.20-3.1 1 (m, 1 H), 3.02-2.92 (m, 1 H). HPLC/MS: m/z calcd for C18H14CI2N403S [M+H]+: 437.0; found: 437.1 .
Figure imgf000097_0001
(2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidin-6- yl)(morpholino)methanone (68). 1 H NMR (400 MHz, DMSO-d6): δ = 7.54 (s, 1 H), 7.23 (s, 2 H), 7.12 (s, 1 H), 4.66 (t, J = 8.8 Hz, 2 H), 3.59 (br s, 8 H), 3.26-3.21 (m, 1 H), 3.02-2.92 (m, 1 H). HPLC/MS: m/z calcd for C19H16CI2N403S [M+H]+: 451 .0; found: 451 .1 .
Figure imgf000097_0002
(2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidin-6-yl)(pyrrolidin-1- yl)methanone (69). 1 H NMR (400 MHz, DMSO-d6): δ = 7.54 (s, 1 H), 7.23 (s, 2 H), 7.21 (s, 1 H), 4.66 (t, J = 8.8 Hz, 2 H), 3.68-3.64 (m, 2 H), 3.44 (br s, 2 H), 3.26-3.1 6 (m, 1 H), 3.02-2.92 (m, 1 H), 1 .91 -1 .76 (m, 4 H). HPLC/MS: m/z calcd for
Figure imgf000097_0003
[M+H]+: 435.0; found: 435.1 .
Figure imgf000097_0004
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-methyl-N-(2,2,2-trifluoroethyl)thieno[2,3- d]pyrimidine-6-carboxamide (70). 1 H NMR (400 MHz, DMSO-d6): δ = 7.54 (s, 1 H), 7.33 (s, 3 H), 4.66 (t, J = 8.8 Hz, 2 H), 4.41 -4.28 (m, 2 H), 3.27 (s, 3 H), 3.26-3.16 (m, 1 H), 3.02-2.92 (m, 1 H). HPLC/MS: m/z calcd for C18H13CI2F3N402S [M+H]+: 477.0; found: 477.1 .
Figure imgf000098_0001
2-amino-N-(2-cyanoethyl)-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-methylthieno[2,3- d]pyrimidine-6-carboxamide (71). 1 H NMR (400 MHz, DMSO-d6): δ = 7.54 (s, 1 H), 7.28 (s, 2 H), 7.20 (s, 1 H), 4.66 (t, J = 8.8 Hz, 2 H), 3.69 (br s, 2 H), 3.26-3.16 (m, 4 H), 3.02-2.92 (m, 1 H), 2.82-2.70 (m, 2 H). HPLC/MS: m/z calcd for
Figure imgf000098_0002
[M+H]+: 448.0; found: 448.1 .
Figure imgf000098_0003
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-methyl-N-(oxetan-3-yl)thieno[2,3- d]pyrimidine-6-carboxamide (72). 1 H NMR (400 MHz, DMSO-d6): δ = 7.55 (s, 1 H), 7.26 (s, 2 H), 7.16 (s, 1 H), 5.21 -5.16 (m, 1 H), 4.69-4.60 (m, 6 H), 3.26-3.14 (m, 4 H), 3.02-2.92 (m, 1 H). HPLC/MS: m/z calcd for C19H16CI2N403S [M+H]+: 451 .0; found: 451 .1 .
Figure imgf000098_0004
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-(2-hydroxy-2-methylpropyl)thieno[2,3- d]pyrimidine-6-carboxamide (73). 1 H NMR (400 MHz, DMSO-d6): δ = 8.44 (s, 1 H), 7.69 (s, 1 H), 7.59 (s, 1 H), 7.23 (s, 2 H), 4.67 (t, J = 8.8 Hz, 2 H), 4.45 (s, 1 H), 3.21 -3.12 (m, 3 H), 3.02-2.92 (m, 1 H), 1 .04 (s, 6 H). HPLC/MS: m/z calcd for C19H18CI2N403S [M+H]+: 453.0; found: 453.1 . 74 >
2-amino-N-cyclopropyl-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-methylthieno[2,3- d]pyrimidine-6-carboxamide (74). 1 H NMR (400 MHz, DMSO-d6): δ = 7.55 (s, 1 H), 7.36 (s, 1 H), 7.28 (s, 2 H), 4.70-4.62 (m, 2 H), 3.13-3.06 (m, 1 H), 3.00-2.91 (m, 4 H), 0.80-0.72 (m, 2 H), 0.68-0.63 (m, 2 H). HPLC/MS: m/z calcd for C19H16CI2N402S [M+H]+: 435.0; found: 435.1 .
Figure imgf000099_0001
2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-methoxy-N-methylthieno[2,3-d]pyrimidine- 6-carboxamide (75). 1 H NMR (400 MHz, DMSO-d6): δ = 7.57 (s, 1 H), 7.47 (s, 1 H), 7.37 (s, 2 H), 4.70- 4.62 (m, 2 H), 3.74 (s, 3 H), 3.27-3.19 (m, 4 H), 3.00-2.91 (m, 1 H). HPLC/MS: m/z calcd for
C17H14CI2N403S [M+H]+: 425.0; found: 425.1 .
Figure imgf000099_0002
1-(2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidin-6-yl)propan-1-one (76). To a flask containing 2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)-N-methoxy-N- methylthieno[2,3-d]pyrimidine-6-carboxamide (20 mg, 0.048 mmol) in dry THF (1 mL), was added EtMgBr (2.0 M in THF; 0.029 mL, 0.057 mmol), at 0 °C. The reaction was stirred for 8 h, upon which it was quenched by addition of sat. NH4CI aq (1 mL). The aqueous layer was extracted with EtOAc (3 x 2 mL), dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:10). The product was obtained in 91 % yield as a white solid.
1 H NMR (400 MHz, CDCI3): δ = 7.41 (s, 1 H), 7.34 (s, 1 H), 5.84 (br s, 2 H), 4.74-4.64 (m, 2 H), 3.87-3.76 (m, 1 H), 3.30-3.10 (m, 3 H), 1 .50-1 .41 (m, 3 H). HPLC/MS: m/z calcd for C17H13CI2N302S [M+H]+: 394.0; found: 394.1 .
Figure imgf000100_0001
2-(2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidin-6-yl)p (77).
To a flask containing ethyl 2-amino-4-(5,7-dichloro-2,3-dihydrobenzofuran-4-yl)thieno[2,3-d]pyrimidine-6- carboxylate (20 mg, 0.048 mmol) in dry THF (1 mL), was added MeMgBr (2.0 M in THF; 0.072 mL, 0.144 mmol), at 0 °C. The reaction was stirred for 8 h, upon which it was quenched by addition of sat. NH4CI aq (1 mL). The aqueous layer was extracted with EtOAc (3 x 2 mL), dried over Na2S04, filtered, and volatiles were evaporated. The residue was purified by silica gel chromatography using a gradient of DCM:MeOH (100:0 to 90:1 0). The product was obtained in 78 % yield as a white solid.
1 H NMR (400 MHz, DMSO-d6): δ = 7.53 (s, 1 H), 6.85 (s, 2 H), 6.54 (s, 1 H), 5.95 (s, 1 H), 4.64 (t, J = 8.8 Hz, 2 H), 3.20-3.12 (m, 1 H), 3.00-2.90 (m, 1 H), 1 .46 (s, 3 H), 1 .44 (s, 3H). HPLC/MS: m/z calcd for C17H15CI2N302S [M+H]+: 396.0; found: 396.1 .
Example 2. In Vitro Assays of the Compounds of the Invention
Hsp90 biochemical assay (fluorescence polarization)
Hsp90 inhibitory activity of the compounds of the invention was assessed using a fluorescence polarization (FP) assay using FITC-labeled geldanamycin and truncated alpha-Hsp90 protein.
Measurement of binding activity was performed on BMG CLARIOstar® reader (BMG Labtech, Ortenberg, Germany). This assay is homogeneous and is performed in 384-well plates performing consistently with known standards. The assay was validated with known Hsp90 inhibitors, e.g., PU-H71 (IC50 = 60 nM), which is in agreement with the reported IC50 (Luo et al., supra). The results of this assay for compounds 20, 36, 37, and 39 and for the known Hsp90 inhibitor are shown in Figure 4.
Hsp90 biochemical assay (AlphaLISA)
Hsp90 inhibitory activity of the compounds of the invention was assessed using a robust and reproducible assay based on the AlphaLISA format was developed (PerkinElmer, Inc., Waltham, MA; the format of the assay is described in, e.g., ELISA to AlphaLISA Conversion Guide, PerkinElmer, Inc., published in August, 2012). This assay employs biotinylated geldanamycin, His-tagged-Hsp90, and Ni+2 coated beads. This assay is homogenous and miniaturized to 384-well plates. Measurement of binding activity was performed on an Envision reader. The assay was validated with known Hsp90 inhibitors, including PU-H71 (IC50 = 60 nM) which is in agreement with the reported IC50 (Luo et al., supra). This assay is the primary assay for evaluating compounds of the invention. In this assay, compound 20 showed Hsp90 inhibitory activity (IC50 of 0.74 ± 0.1 μΜ; see Figure 5A). Hsp90 cell based functional assays Cells were treated with compounds of the invention for 24 h and then lysed in a buffer containing NP-40, orthovanadate, and protease inhibitors. Western blots were performed with antibodies specific to Hsp70, Hsp90, or actin (as control). Tau phosphorylation was assayed using, e.g., a method of Liu et al., Biochemistry, 49:4921 -4929, 2010, with SHSY5Y-hTau441 V337M/R406W cell line (Loeffler et al., J. Mol. Neurosci., 47:192-203, 2012). This cell line represents stable transfected cell line that has been shown to have hyper-phosphorylated tau by over-express the longest human tau isoform, hTAU441 with two mutations: V337M and R406W. The functional assay described herein provides an in vitro model of tauopathy and can be used to evaluate the effect of Hsp90 inhibitors on phosphorylated tau protein (p- Tau). In these assays, compound 20 showed increase in the expression of Hsp70 (see Figure 5B), significant decreases in the levels of pTau231 at 0.1 and 0.5 μΜ (see Figure 6B), and significant decreases in the levels of pTau396 at 0.05, 0.1 , and 0.5 μΜ (see Figure 6C).
Cytotoxicity assays
SH-SY5Y-hTAU441 cell viability was determined using the MTT assay. As shown in Figure 6A, compound 20 did not affect cell viability of SH-SY5Y-hTAU441 cells. SH-SY5Y cell viability may also be measured after 24 and 48 h with the CellTiter-Glo® assay that measures ATP-levels. In addition, cytotoxicity may be assessed using the Celigo® with live cell microscopic imaging and a GE InCell Analyzer 2000 to measure multiple outcomes including apoptosis markers, membrane permeability, and mitochondrial activity.
Mouse liver microsomal stability
Metabolic stability of the compounds of the invention was assessed by monitoring their degradation in mouse liver microsomes. Compound 20 demonstrated good microsomal stability (T1/2 = 26 min). Solubility
Compound solubility was determined in pH 7.4 buffer. Aqueous solubility of greater than or equal to about 0.5 μΜ (e.g., greater than or equal to about 1 μΜ, greater than or equal to about 2 μΜ, greater than or equal to about 5 μΜ, greater than or equal to about 10 μΜ, greater than or equal to about 20 μΜ, or greater than or equal to about 30 μΜ) may indicate a compound having acceptable solubility for medical use, e.g., for treatment of a neurodegenerative disorder. At pH of 7.4, compound 20 exhibits an aqueous solubility of 30 μΜ.
Cell permeability
Compounds may be assessed in MDR1 -MDCK permeability assay or Caco-2 permeability assay to determine their permeability. Apical (A) to basal (B) permeability >3 X1 0"6 cm/sec and B→A/A→B asymmetry < 3 for a compound are considered acceptable predictors of brain penetration, and compounds having such properties are unlikely to be P-glycoprotein (P-gp) substrates. Compound 20 has shown excellent permeability in MDR1 -MDCK assay (A-B = 28 x 10~6 cm/s) with low asymmetry (B→A/A→B asymmetry = 0.9).
Results of the above-described assays for certain compounds of the invention are summarized in Table 3. Table 3.
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
104
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
n Table , Hsp inhibi ory ac ivi y provides an assessmen of exemplary compounds for heir abili y o inhibit Hsp90. In particular, "-" indicates that the compound has IC50 of greater than about 10 μΜ, "+" indicates that the compound has IC50 in the range of about 4 μΜ to about 10 μΜ; "++" indicates that the compound has IC50 in the range of about 1 μΜ to about 4 μΜ; "+++" indicates that the compound has IC50 of less than about 1 μΜ. "Hsp 70 agonist activity" indicates EC50 (μΜ) of the exemplary compound at which Hsp70 is increased two-fold unless otherwise noted; "-" designates lack of observable effect at concentrations exceeding 10 μΜ. (1 ) Data according to AlphaLISA assay, FP assay result: >10 μΜ. (2)
Data according to AlphaLISA assay, FP assay result: 5.5 and 6.2 μΜ. (3) Data according to AlphaLISA assay, FP assay result: 1 and 1 .4 μΜ. (4) Data according to AlphaLISA assay, FP assay result: 2.3 and 2.4 μΜ. (5) Data according to AlphaLISA assay, FP assay result: 1 .7 μΜ. (6) Data according to AlphaLISA assay, FP assay result: 1 .8 μΜ. (7) Data according to AlphaLISA assay, FP assay result: 1 1 .1 μΜ. (8) Data according to AlphaLISA assay, FP assay result: 12.1 and 19.3 μΜ. (9) Data according to AlphaLISA assay, FP assay result: 8.5 and 1 1 .2 μΜ. Example 3. Pharmacokinetic Properties of the Compounds of the Invention
The pre-clinical study included 85 mice, distributed over 6 groups; a summary of treatment groups is given in Table 4.
Table 4.
Group n Sacrificed at Sample reference
A 5 Pre-dose A_0
B 3 15 minutes B 0.25
C 3 30 minutes C 0.5
D 3 1 hour D 1
E 3 2 hours E 2
F 3 8 hours F 8 Plasma and brain samples were taken pre-dose and at post dose time points of pre-dose, 15 and 30 minutes, 1 , 2, and 8 hours. The pre-dose group was represented by 5 animals, whereas the post-dose groups were represented by 3 animals.
Bioanalysis methods
Bioanalysis of mouse plasma samples for compound 20 was conducted by protein precipitation and LC- MS/MS with compound 22 as the internal standard. The method was based on a 'generic assay' and some method development was conducted to tailor that assay to the particular compound and internal standard. The eventual non-GLP assay was first tested by analyzing a bioanalytical run with spiked mouse plasma samples. The qualification run passed by the run acceptance criteria (see below).
Bioanalysis was then conducted of plasma sample extracts and of brain sample homogenate extracts, using a calibration and quality control samples spiked in mouse plasma. The assay is described below.
Assay of mouse plasma levels of compound 20
Sample treatment
Compound 20 and compound 22 were extracted from the mouse plasma matrix by protein precipitation. To 20.0 \}L of sample was added 10.0 μί of internal standard working solution (1000 ng/mL in MeOH) and 200 μί of MeCN. The mixture was vortexed (~5 sec) and centrifuged (14000 rpm, 5 min). The supernatant was then recovered and evaporated to dryness. The residue was redissolved in 100 μί of redissolving solution (80:20 v:v of mobile phases A:B). For analysis, 20.0 μί was injected in to the LC- MS/MS system. Chromatography
All chromatography was done with a type 1 100 liquid chromatograph (Agilent), equipped with an auto- injector. The analytical column was an Xbridge C18 3.5μιη 2.1 x 50 mm (Waters), employed at 50 °C. The mobile phase was a gradient, composed of solvent A :1 g/L ammonium acetate in Milli-Q water, and solvent B: MeCN. The gradient was as shown in Table 5.
Table 5.
Step Total time (min) Flow rate A (%) B (%)
(ML/min)
0 0.00 700 80 20
1 0.20 700 80 20
2 1 .00 700 0 100
3 2.00 700 0 100
4 2.10 700 80 20
5 5.00 700 80 20
Mass spectrometry
All experiments were done on an API 3000 triple quadrupole instrument (AB Sciex), operated in positive turbo-ionspray mode ('TIS+'). The instrument parameters had been optimized during method development. The MS/MS transitions employed were as shown in Table 6.
Table 6.
Compound Q1 , [M+H]+ (35CI2) Q3 [M+H-HCI]+ (35CI2)
(Da) (Da)
Compound 20 321 .1 285.0
Compound 22 349.2 318.9
Description of bioanalytical experiment and acceptance criteria
The calibration range for compound 20 was set up to cover concentrations between 0.988 and 20000 ng/mL. Two sets of calibration samples were used, one set placed before and the other placed after the study samples. In addition, QC samples at 5 levels (two samples at each level) were included in the run, as performance indicators and for run acceptance. Acceptance criteria for calibration and QC samples were applied, as follows:
• The absolute %RE (|%RE|) relative to the nominal concentration for individual calibration and QC samples should be within 20 % (or 25 % at LLOQ);
• A calibration level was considered valid when at least one of the calibration samples at that concentration level was accepted by the above |%RE| criterion;
· A QC level was considered valid when at least one of the QC samples at that concentration level was accepted by the above |%RE| criterion.
The lowest and highest accepted calibration concentration levels were adopted as the lower and upper limit of quantification, LLOQ and HLOQ, respectively.
Bioanalytical results
For compound 20, the highest calibration level (STD L, 20000 ng/mL) did not pass acceptance criteria as both calibration samples displayed too high a bias (see Table 7). All other calibration levels of compound 20, STD A through STD K, were accepted with single samples at STD C and STD G displaying too high bias. As a consequence, the lowest level (STD A, 0.988 ng/mL) was adopted as the LLOQ and the next higher level (STD K, 8000 ng/mL) was adopted as the HLOQ. One of the QC LLOQ and one of the QC Med sample results had too high a bias. The overall method performance in the bioanalytical run was accepted for compound 20.
Calibration and QC concentration levels employed are shown in Table 7:
Table 7.
Sample indicator Compound 20
concentration
(ng/mL)
STD A 0.988
STD B 2.37
STD C 5.93
STD D 14.8
STD E 35.6
STD F 88.9
STD G 222
STD H 533
STD 1 1333
STD J 3333
STD K 8000
STD L 20000
QCLLQ 2.37
QCLow 14.8
QCMed 88.9
QCHigh 1333
QCOC 8000 The bioanalysis results for plasma and brain samples are presented in Tables 8 and 9.
Table 8.
Group IRN Concentration
compound 20
(ng/mL)
A_0 2 0.00 a
A 0 4 2.16
A_0 6 0.00 a
A_0 8 0.00 a
A_0 10 0.00 a
B 0.25 12 2.66
B_0.25 14 17,900 b
B_0.25 16 12,000 b
C_0.5 18 1 1 ,500 D
C_0.5 20 10,500 b
C_0.5 22 1 1 ,100 b
D 1 24 6,890
D 1 26 7,130
D 1 28 7,470
E 2 30 4,150
E 2 32 4,180
E 2 34 4,440
F 8 36 99.3
F 8 38 22.5
F 8 40 21 .9
(a) "0.00" represents 'below limit of quantification' (LLOQ; the LLOQ was 0.988 ng/mL).
(b) Value is out of range (0.988 - 8000 ng/mL); two calibrators at 20000 ng/mL were rejected but showed an average response of 14000 ng/mL.
Table 9.
Group IRN Homogenate Brain weight Brain concentration concentration (g) of compound 20
(ng/mL) (ng/g)
A_0 2 0.00 a 0.453 0.00 a
A_0 4 0.00 a 0.483 0.00 a
A_0 6 0.00 a 0.451 0.00 a
A_0 8 0.00 a 0.487 0.00 a
A_0 10 0.00 a 0.472 0.00 a
B_0.25 12 0.00 a 0.463 0.00 a
B 0.25 14 2300 0.445 20674
B 0.25 16 1250 0.460 10870
C 0.5 18 1400 0.482 1 1618
C 0.5 20 844 0.458 7371
C 0.5 22 1200 0.458 10480
D 1 24 888 0.474 7494
D 1 26 859 0.459 7486
D 1 28 789 0.461 6846
E 2 30 410 0.455 3604
E 2 32 385 0.461 3341
E 2 34 294 0.467 2518
F 8 36 7.53 0.478 63.0
F 8 38 1 .83 0.489 15.0
F 8 40 1 .53 0.476 12.9
(a) "0.00" represents "below limit of quantification" (LLOQ; the LLOQ was 0.988 ng/mL).
In the plasma samples, the following observations were made:
• There was one minor response in a pre-dose sample (one A_0 sample; found at 2.16 ng/mL).
This response may have come from a minor contamination or from assay interference. Although selectivity of LC-MS/MS methods is generally high, selectivity for compound 20 in plasma was not tested. Conclusions can only be drawn after more elaborate method qualification or even method validation. At less than 3 times LLOQ, this pre-dose response is considered negligible here.
• Results for most measurements in time point B 0.25 and all measurements in time point C_0.5 are above the upper limit of quantification (ULOQ, at 8000 ng/mL). For obtaining more reliable results, these samples would have to be diluted prior to analysis. It is noted that a highest calibrator at 20000 ng/mL was included in the run but failed on bias. The mean back-calculated concentration of 20000 ng/mL was 14000 ng/mL., indicating a bias of -30 % at that level. The above ULOQ results were included in this Example as indicative values, in support of PK evaluation. However, the results from B_0.25 and C_0.5 time points should be treated with caution.
• Results from the first subject in the time point B 0.25 (IRN 12) are unlikely to be near the
observed 2.66 ng/mL, as that does not match with the relatively high concentrations observed in the other two subjects at this first post-dose time point(IRN 14 and 16). Provisionally, PK evaluation for this time point was conducted for both cases: (1 ) mean of 3 and (2) mean of two with exclusion of this BLOQ result.
In the brain (homogenate) samples one result appear different from expectation:
• results from the first subject in the time point B O.25 (IRN 12) are unlikely to be below LLOQ, as that does not match with the relatively high concentrations observed in the other two subjects at this first post-dose time point (IRN 14 and 16). Please note that this parallels the findings for plasma from this subject (IRN 12). Provisionally, PK evaluation for this time point was conducted for both cases: (1 ) mean of 3 and (2) mean of two with exclusion of this BLOQ result. Pharmacokinetic evaluation
Evaluation of pharmacokinetic parameters was conducted by calculation from the mean concentration (n=5 pre-dose, n=3 post-dose) at each time point. The pharmacokinetic results are summarized below.
Mouse Plasma
Results for pharmacokinetic evaluation are given in Tables 10-12 and Figures 7 and 8. No corrections were made for results above the ULOQ or for the inclusion of the result for animal IRN12 in group B O.25.
Table 10.
Group Time Point (h) Mean Group Concentration
(ng/mL)
A_0 0.00 0.432
B_0.25 0.25 9968 a
C_0.5 0.50 1 1033
D_1 1 .00 7163
E_2 2.00 4257
F 8 8.00 47.9
(a) Excluding IRN 12 result: 14950 ng/mL Tables 1 1 and 12 provide plasma pharmacokinetic profile of compound 20.
Table 11. (a)
Figure imgf000119_0001
Table 12. (a){
Figure imgf000119_0002
a nc u ng 2 resu t.
Mouse Brain Tissue
Results for brain tissue pharmacokinetic evaluation are given in Figures 9 and 10 and Tables 13-15. corrections were made for the inclusion of the odd result for animal IRN 12 in group B O.25. Table 13.
Group Time Point (h) Mean Group Concentration
Homogenate (ng/mL) Tissue (ng/g)
A 0 0.00 0.00 0.00
B 0.25 0.25 1 183 10515 a
C 0.5 0.50 1 148 9823
D 1 1 .00 845 7275
E 2 2.00 363 3154
F 8 8.00 3.63 30.0
(a) excluding I RN 12 result: 15772 ng/g.
Tables 14 and 15 provide brain tissue pharmacokinetic profile of compound 20.
Table 14. (a
Figure imgf000120_0001
(a) excluding I RN 12 result.
Table 15. w
Figure imgf000120_0002
Example 4. Effect of the Compounds of the Invention on the Total Level of Tau and Level of p-Tau in Cerebrospinal Fluid (CSF) and Brain in the Tau Transgenic Mouse Model (hTAU441)
To assess the effect of the compounds of the invention on p-tau accumulation, age-matched (e.g., 5 month old) transgenic mice humanized for the tau gene (hTAU mice) may be treated with low- or high- dose of a compound of the invention or vehicle by intraperitoneal administration for 7 days (N = 6 per arm). Dose of the compound of the invention may be calculated based on the PK results. hTAU transgenic mice (C57BL/6 background) over-express TAU441 bearing the missense mutations V337M and R406W under the control of the brain specific murine Thy-1 promoter. This human mutated tau isoform is expressed in high levels and the tau pathology and is visible at an early age starting at four months. Severity of the brain pathology correlates with increasing age and behavioral deficits, whereas no motor deficits occur. All animals may be sacrificed and quantified for soluble and insoluble tau and p- tau brain (hippocampus and cortex) using MSD multiarray p-tau (ThR211 ) immunosorbent assay (Meso Scale Discovery, Rockville, MD) to establish total tau and p-tau levels in CSF and Hsp70 levels in brain extracts. Specifically, the animals may be anaesthetized with ketamine/xylazin mix (note: isoflurane is known to influence p-tau levels), kept warm and in a horizontal position, prior to and during the collection of CSF followed by blood. The volume of CSF collected in hTAU441 is only 2-6 μί/ιηουεβ compared to some strains (2-15 μί/ιηουεβ). p-Tau (ThR211 ) and total tau levels may be assessed using phospho- PHF-Tau pThR211 (MSD duplex kit, Meso Scale Discovery, Rockville, MD).
Administration of a compound of the invention may lead to a decrease in p-tau levels in mice treated with a compound of the invention relative to p-tau levels in mice administered a vehicle.
Example 5. Effect of the Compounds of the Invention on Memory and Learning in the hTAU441 Transgenic Mouse Model
Five month old transgenic hTAU mice described in Example 3 may be administered intraperitoneally a low- or high-dose of a compound of the invention groups or a vehicle daily for 12 weeks (N = 15 per arm). A corresponding untreated group of mice will be analyzed as baseline. Behavioral testing, e.g., Probe Trial, Nose Poke Curiosity and Activity Test, and the Morris Water Maze task may be performed. Upon completion of the study, CSF may be collected and brain tissue may be harvested from these animals. Total tau levels and p-tau levels may be assessed in the CSF and brain. Additionally,
immunohistochemical determination of tau pathology may be conducted. Tau depositions may be determined using the monoclonal antibodies AT180 (Thermo Scientific Pierce Antibodies, Rockford, IL) and HT7 (Thermo Scientific Pierce Antibodies, Rockford, IL). Sodium selenate, a PP2A phosphatase activator that dephosphorylates tau and reverses memory deficits, is effective in the TMHT tau model (Corcoran et al., J. Clin. Neuroscience, 17:1025-1033, 2010). If chronic treatment with a compound of the invention is associated with a general improvement in memory function as measured by the Morris Maze, reduced levels of p-tau in the brain may be observed.
Other Embodiments
The invention is also described by the following numbered embodiments:
1 . A compound according to formula (I):
Figure imgf000121_0001
(I), or a pharmaceutically acceptable salt thereof,
where
Z1 is -OR7, -N(R10)R7, -SR7, or -C(R10)(R1 1)R7;
Z2 is -N= or -C(R3)=;
each R1 and R2 is, independently, H or optionally substituted C1 -3 alkyl;
R3 is H, halogen, cyano, optionally substituted Ci-6 alkyl, optionally substituted C^ alkoxy, or optionally substituted amino, and R4 is halogen, cyano, optionally substituted C1 -6 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted amino, optionally substituted C1 -6 thioalkoxy, or optionally substituted C6-10 aryl, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally including one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R9; each R5 and R6 is, independently, H, optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkoxy, halogen, or CN;
R7 is optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted alkaryl, and R8 is H; or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally including one or two heteroatoms selected from nitrogen, oxygen, and sulfur;
R9 is H, optionally substituted C1 -3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-10 aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted C1 -3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl;
R10 is H, optionally substituted Ci-3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted alkcycloalkyl, optionally substituted alkheterocyclyl, or optionally substituted C1 -3 alkaryl, and R1 1 is H, optionally substituted C1 -3 alkyl, or R10 and R1 1 combine to form =0 or =S;
Rm is H, halogen, optionally substituted C1 -4 alkyl, or optionally substituted C1 -3 alkoxy;
where,
when Z2 is CR3, each of R1 and R2 is H, R3 is H, R4 is methyl or chloro, and each of R5 and R6 is chloro,
Z1 is not methoxy;
when Z2 is N, each of R5 and R6 is chloro, R3 is H, R4 is substituted thioalkoxy,
Z1 is not cyanomethoxy or aminomethoxy;
when Z2 is CR3, each of R5 and R6 is chloro, R3 is H, and R4 is halogen,
Z1 is not 2-amino-2-oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy;
when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (lla):
Figure imgf000122_0001
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl;
when each R5 and R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (lla),
R7 is not methyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (lla),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl-methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl- 1 ,2,4-oxadiazol-5-yl-methyl;
when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (Ma),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl when R5 is methoxy, R6 is methyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when R5 is chloro, R6 is ethyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
Figure imgf000123_0001
R7 is not methyl or 2-(N,N-diethylamino)ethyl;
when R7 is methyl, R5 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ilia), HN^ (|||a) i
R6 is not bromo;
when R5 is chloro, R6 is methoxy, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (1Mb):
Figure imgf000123_0002
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVa):
Figure imgf000123_0003
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (I Vb):
Figure imgf000123_0004
R7 is not 2-methoxyethyl or benzyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVc):
Figure imgf000124_0001
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (I Vd):
Figure imgf000124_0002
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVe) or (IVf):
Figure imgf000124_0003
R7 is not benzyl;
when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
Figure imgf000124_0004
R7 is not methyl;
when R6 is methyl,
each R1 and R2 is H; and
when R3 is H, Z1 is -OR7, and each R5 and R6 is chloro,
R7 is not methyl.
2. The compound of embodiment 1 , where Rm is H.
3. A compound according to formula (la):
Figure imgf000125_0001
(la), or a pharmaceutically acceptable salt thereof, each R1 and R2 is, independently, H or optionally substituted C1 -3 alkyl;
R3 is H, halogen, cyano, optionally substituted C1 -6 alkyl, optionally substituted C1 -3 alkoxy, or optionally substituted amino, and R4 is halogen, cyano, optionally substituted C1 -6 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted amino, optionally substituted C1 -6 thioalkyl, or optionally substituted C6-io aryl, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally including one nitrogen, one oxygen, or one
9.
sulfur, where the nitrogen is optionally substituted with R
each R5 and R° is, independently, H, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkoxy, halogen, or CN;
R7 is optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1 -3 alkaryl, and R8 is H; or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally including one or two heteroatoms selected from nitrogen, oxygen, and sulfur;
R9 is H, optionally substituted Ci-3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted alkcycloalkyl, optionally substituted alkheterocyclyl, or optionally substituted C1 -3 alkaryl;
Z1 is -OR7, -N(R10)R7, -SR7, or -C(R10)(R1 1)R7; and
R10 is H, optionally substituted C1 -3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-10 aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted alkcycloalkyl, optionally substituted alkheterocyclyl, or optionally substituted Ci-3 alkaryl, and R1 1 is H, optionally substituted alkyl, or R10 and R1 1 combine to form =0 or =S;
where,
when each of R1 and R2 is H, R3 is H, R4 is methyl or chloro, and each of R5 and R6 is chloro, Z1 is not methoxy;
when each of R5 and R6 is chloro, R3 is H, and R4 is halogen,
Z1 is not 2-amino-2-oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy;
when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (lla):
Figure imgf000125_0002
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl;
when each R5 and R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl-methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl- 1 ,2,4-oxadiazol-5-yl-methyl;
when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (Ma),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl when R5 is methoxy, R6 is methyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when R5 is chloro, R6 is ethyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
Figure imgf000126_0001
R7 is not methyl or 2-(N,N-diethylamino)ethyl;
when R7 is methyl, R5 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ilia),
HN^ (ma),
R6 is not bromo;
when R5 is chloro, R6 is methoxy, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (1Mb):
Figure imgf000126_0002
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVa):
Figure imgf000126_0003
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (I Vb):
Figure imgf000127_0001
R7 is not 2-methoxyethyl or benzyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVc):
Figure imgf000127_0002
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (I Vd):
Figure imgf000127_0003
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVe) or (IVf):
Figure imgf000127_0004
R7 is not benzyl;
when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
Figure imgf000127_0005
R7 is not methyl;
when R6 is methyl, each R1 and R2 is H; and
when R3 is H, Z1 is -OR7, and each R5 and R6 is chloro,
R7 is not methyl. 4. A compound according to formula (lb):
Figure imgf000128_0001
or a pharmaceutically acceptable salt thereof,
where
each of R1 and R2 is, independently, H or optionally substituted alkyl;
R3 is H, halogen, cyano, optionally substituted Ci-6 alkyl, optionally substituted alkoxy, or optionally substituted amino, and R4 is halogen, cyano, optionally substituted Ci-6 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted amino, optionally substituted C1 -6 thioalkoxy, or optionally substituted C6-10 aryl, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally including one nitrogen, one sulfur, or one
9.
oxygen, where the nitrogen is optionally substituted with R
each of R5 and R° is, independently, H, optionally substituted C1 -3 alkyl, optionally substituted alkoxy, halogen, or CN;
R7 is optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1 -3 alkaryl, and R8 is H; or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring; and
R9 is H, optionally substituted C1 -3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted alkcycloalkyl, optionally substituted alkheterocyclyl, or optionally substituted Ci-3 alkaryl;
where,
when each of R1 and R2 is H, R3 is H, R4 is methyl or chloro, and each of R5 and R6 is chloro, R7 is not methyl; when each of R5 and R6 is chloro, R3 is H, and R4 is halogen,
R7 is not 2-amino-2-oxoethyl, 2-(N,N-diethylamino)ethyl, methyl, or benzyl;
when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (lla) :
Figure imgf000128_0002
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)-ethyl, difluoromethyl, or 2-(f-butylamino)ethyl; when each R5 and R6 is bromo, and R3 and R4 combine to form a group according to formula
(lla),
R7 is not methyl;
when each R5 and R6 is chloro, R8 is H, and R3 and R4 combine to form a group according to formula (lla),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl-methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl- 1 ,2,4-oxadiazol-5-yl-methyl;
when each R5 and R6 is chloro, R7 and R8 combine to form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (lla),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl;
when R5 is methoxy, R6 is methyl, and R3 and R4 combine to form a group according to formula
(lla),
R7 is not methyl;
when R5 is chloro, R6 is ethyl, and R3 and R4 combine to form a group according to formula (lla), R7 is not methyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
Figure imgf000129_0001
R7 is not methyl or 2-(N,N-diethylamino)ethyl;
when R7 is methyl, R5 is chloro, and R3 and R4 combine to form a group according to formula
(Ilia),
(Ilia),
R is not bromo;
when R5 is chloro, R6 is methoxy, and R3 and R4 combine to form a group according to formula
(1Mb) :
Figure imgf000129_0002
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula
(IVa):
Figure imgf000129_0003
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVb):
Figure imgf000130_0001
R7 is not 2-methoxyethyl or benzyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula
(IVc):
Figure imgf000130_0002
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula
(IVd):
Figure imgf000130_0003
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula
(IVe) or (IVf):
Figure imgf000130_0004
R7 is not benzyl;
when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula
(IVg), (IVh), (IVi), (IVj), or (IVk):
Figure imgf000130_0005
R7 is not methyl;
when R6 is methyl,
each R1 and R2 is H; and
when R3 is H, and each R5 and R6 is chloro, R7 is not methyl.
5. The compound of any one of embodiments 1 to 4, where R3 is H, halogen, optionally substituted alkyl, or optionally substituted alkoxy, and R4 is halogen, optionally substituted alkyl, optionally substituted Ci-3 alkoxy, optionally substituted amino, optionally substituted Ci-6 thioalkyl, or optionally substituted C6-10 aryl, or R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally including one nitrogen, one oxygen, or one sulfur, where the nitrogen is optionally substituted with R9. 6. The compound of any one of embodiments 1 to 5, where R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring.
7. The compound of any one of embodiments 1 to 5, where R3 and R4 combine to form -CH2CH2CH2- group.
8. The compound of any one of embodiments 1 to 5, where R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring including one nitrogen.
9. The compound of embodiment 8, where R3 and R4 combine to form -N(R9)-CH=CH- group.
10. The compound of embodiment 9, where R9 is H.
1 1 . The compounds of any one of embodiments 1 to 5, where R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring including one sulfur.
12. The compound of any one of embodiments 1 to 5, where R3 and R4 combine to form - C(R13A)=C(R13B)-S- group, where R13A is H, and R13B is H or optionally substituted d.3 alkyl.
13. The compound of embodiment 12, where R13B is optionally substituted C1 -3 alkyl.
14. The compound of embodiment 13, where R13B is -C(0)-R13C, where R13C is optionally substituted d. 3 alkoxy or optionally substituted amino.
15. The compound of any one of embodiments 1 to 5, where R3 and R4 combine to form - C(R13A)=C(R13B)-S- group, where R13A is H, and R13B is H or -C(0)-R13C, where R13C is optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted amino, or optionally substituted C2-9 heterocyclyl.
16. The compound of any one of embodiments 1 to 5, where R4 is alkyl.
17. The compound of embodiment 16, where R4 is methyl, ethyl, or isopropyl. 18. The compound of any one of embodiments 1 to 5, where R4 is alkoxy.
19. The compound of embodiment 18, where R4 is methoxy. 20. The compound of any one of embodiments 1 to 5, where R4 is optionally substituted Ci-6 thioalkoxy.
21 . The compound of embodiment 20, where R4 is 4-amino-4-oxobutyl.
22. The compound of any one of embodiments 1 to 5, where R4 is optionally substituted amino.
23. The compound of embodiment 22, where R4 is methylamino.
24. The compound of any one of embodiments 1 to 5, where R4 is halogen. 25. The compound of embodiment 24, where R4 is chloro.
26. The compound of any one of embodiments 1 -5 and 15-25, where R3 is hydrogen or alkyl.
27. The compound of embodiment 26, where R3 is hydrogen, methyl, or ethyl.
28. The compound of any one of embodiments 1 to 5, where R3 and R4 combine to form -X1-X2-X3-, where
X1 is -S-, -0-, -(CR14R15)-, -C(R16)=, -N(R9)-, -N=, H, or optionally substituted C1 -3 alkyl; X2 is absent,-(CR17R18) -, -S-, -0-, -N=, -N(R9)-, -C(R19)=, =N- =C(R20)-, or =C(R21)- C(R22)=;
X3 is -(CR14R15)-, -S-, -0-, -N(R9)-, =N- =C(R23)-, halogen, optionally substituted d_3 alkyl, optionally substituted Ci_s thioalkoxy, optionally substituted alkoxy, or optionally substituted C6-io aryl; each R14 and R15 is, independently, H or optionally substituted Ci-3 alkyl, or R14 and R15 combine to form =0 or =S;
each R17 and R18 is, independently, H or optionally substituted C1 -3 alkyl, or R17 and R18 combine to form =0 or =S;
each R16, R19, R20, R21 , R22, and R23 is, independently, H, or optionally substituted C1 -3 alkyl; and n is 1 or 2; and
where, when X2 is not absent,
the chain of atoms -X1-X2-X3- includes no more than one heteroatom, the heteroatom being selected from the group consisting of nitrogen, oxygen, and sulfur.
29. The compound of embodiment 28, where X1 is -(CR14R15)-, -C(R16)=, -N(R9)-, -N=, or optionally substituted C1 -3 alkyl.
30. The compound of embodiment 29, where X1 is -(CR14R15)- 31 . The compound of embodiment 30, where each R14 and R15 is H.
32. The compound of embodiment 29, where X1 is -C(R16)=. 33. The compound of embodiment 32, where R16 is H.
34. The compound of embodiment 29, where X1 is -N(R9)-.
35. The compound of embodiment 34, where R9 is H or optionally substituted C1 -3 alkyl.
36. The compound of embodiment 35, where R9 is hydrogen, methyl, or ethyl.
37. The compound of embodiment 29, where X1 is -N=. 38. The compound of embodiment 29, where X1 is optionally substituted C1 -3 alkyl.
39. The compound of any one of embodiments 28 to 37, where X2 is absent, -(CH2)n- -N(R9)-, -C(H)=, =C(R20)- or =C(H)-C(H)=. 40. The compound of embodiment 39, where X2 is -C(H)=.
41 . The compound of embodiment 39, where X2 is -N(R9)-.
42. The compound of embodiment 41 , where R9 is H.
43. The compound of embodiment 41 , where R9 is optionally substituted Ci.3 alkyl.
44. The compound of embodiment 43, where R9 is -C(0)-N(H)-Et. 45. The compound of embodiment 39, where X2 is =C(R20)-.
46. The compound of embodiment 45, where R20 is optionally substituted C1 -3 alkyl.
47. The compound of embodiment 39, where X2 is absent.
48. The compound of any one of embodiments 28-37 and 39-47, where X3 is -CH2- -S-, =C(H)-, - N(R9)-, halogen, optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted C1 -6 thioalkoxy, optionally substituted C6-10 aryl. 49. The compound of embodiment 48, where X3 is -CH2-
50. The compound of embodiment 48, where X3 is -S-. 51 . The compound of embodiment 48, where X3 is =C(H)-.
52. The compound of embodiment 48, where X3 is -N(R9)-.
53. The compound of embodiment 48, where X3 is halogen, optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted C1 -6 thioalkoxy, or optionally substituted C6-10 aryl.
54. The compound of any one of embodiments 1 to 53, where each R5 and R6 is, independently, halo or optionally substituted alkyl.
55. The compound of embodiment 54, where each R5 and R6 is halo.
56. The compound of embodiment 55, where each R5 and R6 is chloro.
57. The compound of any one of embodiments 1 to 56, where R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally including one or two heteroatoms selected from nitrogen, oxygen, and sulfur. 58. The compound of any one of embodiments 1 to 56, where R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered saturated ring optionally including one or two heteroatoms selected from nitrogen, oxygen, and sulfur.
59. The compound of any one of embodiments 1 to 56, where R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five- of six-membered ring optionally including one or two heteroatoms selected from nitrogen and oxygen.
60. The compound of any one of embodiments 1 to 56, where R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring.
61 . The compound of any one of embodiments 1 to 56, where the R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted saturated five-membered ring.
62. The compound of any one of embodiments 1 to 56, where the R7 and R8 combine to form a - CH2CH2- group.
63. The compound of any one of embodiments 1 to 56, where R7 is optionally substituted C1 -3 alkyl.
64. The compound of embodiment 63, where R7 is methyl.
65. The compound of embodiment 63, where R7 is -(CH2)k-N(R24)R25, where k is 2 or 3, and where each R24 and R25 is, independently, H or optionally substituted alkyl. 66. The compound of embodiment 65, where k is 2.
67. The compound of embodiment 65 or 66, where each R and R is, independently, optionally substituted Ci-3 alkyl.
68. The compound of any one of embodiments 65 to 67, where each R24 and R25 is methyl.
69. The compound of any one of embodiments 1 to 56, where R7 and R8 form a group -Y1-Y2-, where:
Y1 is -(CR26R27)m- or optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkheterocyclyl, optionally substituted alkcycloalkyl, or optionally substituted Ci-3 alkaryl; and
Y2 is -(CR26R27)- or H; where
each R26 and R27 is, independently, H or optionally substituted Ci-3 alkyl; and
m is 1 or 2.
70. The compound of any one of embodiments 1 to 5, where Z1 and R8 combine to form
-Z3-Y1-Y2-, where
Z3 is -0-, -N(R10)-, -N=, -S-, or -(CR14R15)-;
Y1 is -0-, -N(R10)-, -S-, -(CR26R27)m- -C(R20)=, =C(R20)-, =C(R21)-C(R22)=, optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkheterocyclyl, optionally substituted alkcycloalkyl, or optionally substituted C1 -3 alkaryl; and
Y2 is -0-, -S-, -N(R10)-, -(CR26R27)-, =C(R20)-, =N-, or H;
where
each R20, R21 , and R22 is, independently, H or optionally substituted C1 -3 alkyl; and
each R26 and R27 is, independently, H or optionally substituted Ci-3 alkyl, or R26 and R27 combine to form =0 or =S;
m is 1 or 2; and
where, when Y2 is H,
the chain of atoms -Z3-Y1-Y2- includes no more than two heteroatoms, the heteroatom selected from nitrogen, oxygen, and sulfur.
71 . The compound of embodiment 70, where Z3 is -0-.
72. The compound of embodiment 69 or 71 , where Y1 is -(CR26R27)m- or optionally substituted Ci-3 alkyl, optionally substituted alkheterocyclyl, optionally substituted alkcycloalkyl, or optionally substituted C1 -3 alkaryl.
73. The compound of any one of embodiments 69 to 72, where Y1 is -(CR26R27)m- or optionally substituted C1 -3 alkyl.
74. The compound of any one of embodiments 69 to 73, where Y2 is -(CR26R27)- or H. 75. The compound of any one of embodiments 69 to 74, where Y2 is -(CR26R27)-.
76. The compound of any one of embodiments 69 to 75, where R is H. 77. The compound of any one of embodiments 69 to 76, where R27 is H.
78. The compound of any one of embodiments 69 to 77, where m is 1 .
79. The compound of any one of embodiments 69 to 78, where Y1 is optionally substituted C1 -3 alkyl.
80. The compound of embodiment 79, where Y1 is methyl.
81 . The compound of embodiment 80, where Y1 is -(CH2)k-N(R24)R25, where k is 2 or 3, and where each R24 and R25 is, independently, H or optionally substituted C1 -3 alkyl.
82. The compound of embodiment 81 , where k is 2.
83. The compound of embodiment 81 or 82, where each R24 and R25 is, independently, optionally substituted Ci-3 alkyl.
84. The compound of any one of embodiments 81 to 83, where each R24 and R25 is methyl.
85. The compound of any one of embodiments 1 to 84, where each R1 and R2 is H. 86. A compound:
Figure imgf000136_0001
Figure imgf000137_0001

Figure imgf000138_0001
Figure imgf000139_0001

Figure imgf000140_0001
or a pharmaceutically acceptable salt thereof.
87. The compound of embodiment 84 having the formula:
Figure imgf000140_0002
Figure imgf000141_0001

Figure imgf000142_0001
88. A pharmaceutical composition including the compound of any one of embodiments 1 to 87, or a pharmaceutically acceptable salt thereof, and one or more of pharmaceutically acceptable carriers or excipients.
89. The pharmaceutical composition of embodiment 88, where the composition is formulated for administration orally, sublingually, buccally, transdermally, intradermally, intramuscularly, parenterally, intravenously, intra-arterially, intracranially, subcutaneously, intraorbitally, intraventricularly, intraspinally, intraperitoneally, intranasally, by inhalation, and topically.
90. The pharmaceutical composition of embodiment 89, where the composition is formulated for oral administration.
91 . A method of treating a disorder in a mammal caused by the action of heat shock protein 90 (Hsp90), the method including administering an effective amount of the compound of any one of embodiments 1 to 87 or a pharmaceutically acceptable salt thereof or the pharmaceutical composition of embodiment 88 to the mammal.
92. The method of embodiment 91 , where the disorder is a neurodegenerative disorder.
93. The method of embodiment 92, where the neurodegenerative disorder is a tauopathy. 94. The method of embodiment 92 or 93, where the neurodegenerative disorder is Alzheimer's disease, Huntington's disease, progressive supranuclear palsy, Parkinson's disease, Pick's disease, corticobasal degeneration, chronic traumatic encephalopathy, traumatic brain injury, or frontotemporal dementia.
95. The method of embodiment 94, where the neurodegenerative disorder is Alzheimer's disease.
96. The method of embodiment 91 , where the disorder is a proliferative disorder.
97. The method of embodiment 96, where the proliferative disorder is a cancer. 98. The method of embodiment 97, where the cancer is acute myeloid leukemia, gastrointestinal stromal tumor, gastric cancer, glioma, neuroblastoma, glioblastoma, lung cancer, lymphoma, melanoma, myeloma, non-small cell lung cancer, renal cancer, small cell lung cancer, blast-phase chronic myelogenous leukemia, leukemia, lymphoproliferative disorder, metastatic melanoma, relapsed multiple myeloma, refractory multiple myeloma, myeloproliferative disorders, pancreatic cancer, small intestine cancer, or solid tumor.
99. A method of treating an infectious disease in a mammal, the method including administering an effective amount of the compound of any one of embodiments 1 to 87 or a pharmaceutically acceptable salt or the pharmaceutical composition of embodiment 88 thereof to the mammal.
100. The method of embodiment 99, where the infectious disease is a viral infection.
101 . The method of embodiment 100, where the viral infection is an infection by a virus of a family selected from the group consisting of Herpesviridae, Polyomaviridae, Poxviridae, Reoviridae,
Birnaviridae, Picornaviridae, Flaviviridae, Arenaviridae, Hepeviridae, Rhabdoviridae, Paramoxyviridae, Bunyaviridae, Orthomoxyviridae, Filoviridae, Retroviridae, and Hepadnaviridae. 102. The method of embodiment 101 , where the virus of a family Herpesviridae is herpes simplex virus- 1 , herpes simplex virus-2, herpes herpesvirus-5, Kaposi's sarcoma-associated herpesvirus, varicella zoster virus, or Epstein-Barr virus. 103. The method of embodiment 101 , where the virus of Polyomaviridae family is SV40.
104. The method of embodiment 101 , where the virus of Poxviridae family is vaccinia virus.
105. The method of embodiment 101 , where the virus of Reoviridae family is rotavirus.
106. The method of embodiment 101 , where the virus of Birnaviridae family is infectious bursal disease virus.
107. The method of embodiment 101 , where the virus of Picornaviridae family is poliovirus, rhinovirus, or coxsackievirus.
108. The method of embodiment 101 , where the virus of Flaviviridae family is hepatitis C virus or dengue virus. 109. The method of embodiment 101 , where the virus of Arenaviridae family is lymphocytic
choriomeningitis virus.
1 10. The method of embodiment 101 , where the virus of Hepeviridae family is Hepatitis E virus. 1 1 1 . The method of embodiment 101 , where the virus of Rhabdoviridae family is vesicular stomatitis virus.
1 12. The method of embodiment 101 , where the virus of Paramoxyviridae family is human parainfluenza virus 2, human parainfluenza virus 3, SV5, SV41 , measles virus, or Sendai virus.
1 13. The method of embodiment 101 , where the virus of Bunyaviridae family is La Crosse virus.
1 14. The method of embodiment 101 , where the virus of Orthomoxyviridae family is influenza A virus. 1 15. The method of embodiment 101 , where the virus of Filoviridae family is Ebola virus.
1 16. The method of embodiment 101 , where the virus of Retroviridae family is HTLV1 or HIV1 .
1 17. The method of embodiment 101 , where the virus of Hepadnaviridae family is hepatitis B virus.
1 18. The method of embodiment 99, where the infectious disease is a fungal infection. 1 19. The method of embodiment 1 18, where the fungal infection is a Candida albicans infection, an Aspergillus fumigates infection, or Pneumocystis jiroveci infection.
120. The method of embodiment 99, where the infectious disease is a bacterial infection.
121 . The method of embodiment 120, where the bacterial infection is a mycobacteria infection or anthrax infection.
122. The method of embodiment 120, where the bacterial infection is a bacterial pneumonia.
123. The method of embodiment 91 , where the disorder an inflammatory or autoimmune disease.
124. The method of embodiment 123, where the inflammatory or autoimmune disease is rheumatoid arthritis, systemic lupus erythermatosus, or asthma.
125. The method of embodiment 91 , where the disorder is a cardiovascular disease.
126. The method of embodiment 125, where the cardiovascular disease is atherosclerosis or cardiomyopathy.
127. The method of embodiment 91 , where the disorder is an allergy.
128. The method of any one of embodiments 91 to 127, where the compound is administered orally, sublingually, buccally, transdermal^, intradermally, intramuscularly, parenterally, intravenously, intra- arterially, intracranially, subcutaneously, intraorbitally, intraventncularly, intraspinally, intrapentoneally, intranasally, by inhalation, and topically.
129. The method of embodiment 128, where the compound is administered orally. 130. The method of any one of embodiments 91 to 129, where the mammal is human.
131 . The method of any one of embodiments 91 to 130, where the compound is administered orally, sublingually, buccally, transdermal^, intradermally, intramuscularly, parenterally, intravenously, intra- arterially, intracranially, subcutaneously, intraorbitally, intraventncularly, intraspinally, intrapentoneally, intranasally, by inhalation, and topically.
132. The method of embodiment 131 , where the compound is administered orally.
133. The method of any one of embodiments 91 to 132, where the mammal is human.
134. A compound for use in treating a disorder in a mammal caused by the action of heat shock protein 90 (Hsp90), where the compound is the compound of any one of embodiments 1 to 87 or a pharmaceutically acceptable salt thereof.
135. The compound of embodiment 134, where the disorder is a neurodegenerative disorder. 136. The compound of embodiment 135, where the neurodegenerative disorder is a tauopathy.
137. The compound of embodiment 134 or 135, where the neurodegenerative disorder is Alzheimer's disease, Huntington's disease, progressive supranuclear palsy, Parkinson's disease, Pick's disease, corticobasal degeneration, chronic traumatic encephalopathy, traumatic brain injury, or frontotemporal dementia.
138. The compound of embodiment 137, where the neurodegenerative disorder is Alzheimer's disease.
139. The compound of embodiment 134, where the disorder is a proliferative disorder.
140. The compound of embodiment 139, where the proliferative disorder is a cancer.
141 . The compound of embodiment 140, where the cancer is acute myeloid leukemia, gastrointestinal stromal tumor, gastric cancer, glioblastoma, lung cancer, lymphoma, melanoma, myeloma, non-small cell lung cancer, renal cancer, small cell lung cancer, blast-phase chronic myelogenous leukemia, leukemia, lymphoproliferative disorder, metastatic melanoma, relapsed multiple myeloma, refractory multiple myeloma, myeloproliferative disorders, pancreatic cancer, small intestine cancer, or solid tumor.
142. The compound of embodiment 134, where the disorder is an infectious disease.
143. The compound of embodiment 142, where the infectious disease is a viral infection.
144. The compound of embodiment 143, where the viral infection is an infection by a virus of a family selected from the group consisting of Herpesviridae, Polyomaviridae, Poxviridae, Reoviridae,
Birnaviridae, Picornaviridae, Flaviviridae, Arenaviridae, Hepeviridae, Rhabdoviridae, Paramoxyviridae, Bunyaviridae, Orthomoxyviridae, Filoviridae, Retroviridae, and Hepadnaviridae.
145. The compound of embodiment 144, where the virus of a family Herpesviridae is herpes simplex virus-1 , herpes simplex virus-2, herpes herpesvirus-5, Kaposi's sarcoma-associated herpesvirus, varicella zoster virus, or Epstein-Barr virus.
146. The compound of embodiment 144, where the virus of Polyomaviridae family is SV40.
147. The compound of embodiment 144, where the virus of Poxviridae family is vaccinia virus.
148. The compound of embodiment 144, where the virus of Reoviridae family is rotavirus. 149. The compound of embodiment 144, where the virus of Birnaviridae family is infectious bursal disease virus.
150. The compound of embodiment 144, where the virus of Picornaviridae family is poliovirus, rhinovirus, or coxsackievirus.
141 . The compound of embodiment 144, where the virus of Flaviviridae family is hepatitis C virus or dengue virus. 152. The compound of embodiment 144, where the virus of Arenaviridae family is lymphocytic choriomeningitis virus.
153. The compound of embodiment 144, where the virus of Hepeviridae family is Hepatitis E virus. 154. The compound of embodiment 144, where the virus of Rhabdoviridae family is vesicular stomatitis virus.
155. The compound of embodiment 144, where the virus of Paramoxyviridae family is human parainfluenza virus 2, human parainfluenza virus 3, SV5, SV41 , measles virus, or Sendai virus.
156. The compound of embodiment 144, where the virus of Bunyaviridae family is La Crosse virus.
157. The compound of embodiment 144, where the virus of Orthomoxyviridae family is influenza A virus. 158. The compound of embodiment 144, where the virus of Filoviridae family is Ebola virus.
159. The compound of embodiment 144, where the virus of Retroviridae family is HTLV1 or HIV1 .
160. The compound of embodiment 144, where the virus of Hepadnaviridae family is hepatitis B virus.
161 . The compound of embodiment 134, where the infectious disease is a fungal infection.
162. The compound of embodiment 161 , where the fungal infection is a Candida albicans infection, an Aspergillus fumigates infection, or Pneumocystis jiroveci infection.
163. The compound of embodiment 134, where the infectious disease is a bacterial infection.
165. The compound of embodiment 163, where the bacterial infection is a mycobacteria infection or anthrax infection.
166. The compound of embodiment 163, where the bacterial infection is a bacterial pneumonia. 167. The compound of embodiment 134, where the disorder an inflammatory or autoimmune disease.
168. The compound of embodiment 167, where the inflammatory or autoimmune disease is rheumatoid arthritis, systemic lupus erythermatosus, or asthma.
169. The compound of embodiment 134, where the disorder is a cardiovascular disease.
170. The compound of embodiment 169, where the cardiovascular disease is atherosclerosis or cardiomyopathy.
171 . The compound of embodiment 170, where the disorder is an allergy.
172. Use of a compound in the manufacture of a medicament for treating a disorder in a mammal caused by the action of heat shock protein 90 (Hsp90), where the compound is the compound of any one of embodiments 1 to 87 or a pharmaceutically acceptable salt thereof.
173. Use of a compound for treating a disorder in a mammal caused by the action of heat shock protein 90 (Hsp90), where the compound is the compound of any one of embodiments 1 to 87 or a
pharmaceutically acceptable salt thereof.
174. The use of embodiment 172 or 173, where the disorder is a neurodegenerative disorder.
175. The use of embodiment 174, where the neurodegenerative disorder is a tauopathy. 176. The use of embodiment 174 or 175, where the neurodegenerative disorder is Alzheimer's disease, Huntington's disease, progressive supranuclear palsy, Parkinson's disease, Pick's disease, corticobasal degeneration, chronic traumatic encephalopathy, traumatic brain injury, or frontotemporal dementia.
177. The use of embodiment 176, where the neurodegenerative disorder is Alzheimer's disease.
178. The use of embodiment 172 or 173, where the disorder is a proliferative disorder.
179. The use of embodiment 178, where the proliferative disorder is a cancer. 180. The use of embodiment 179, where the cancer is acute myeloid leukemia, gastrointestinal stromal tumor, gastric cancer, glioblastoma, lung cancer, lymphoma, melanoma, myeloma, non-small cell lung cancer, renal cancer, small cell lung cancer, blast-phase chronic myelogenous leukemia, leukemia, lymphoproliferative disorder, metastatic melanoma, relapsed multiple myeloma, refractory multiple myeloma, myeloproliferative disorders, pancreatic cancer, small intestine cancer, or solid tumor.
181 . Use of a compound for treating, or in the manufacture of a meadicament for treating, an infectious disease, where the compound is the compound of any one of embodiments 1 to 87. 182. The use of embodiment 181 , where the infectious disease is a viral infection.
183. The use of embodiment 182, where the viral infection is an infection by a virus of a family selected from the group consisting of Herpesviridae, Polyomaviridae, Poxviridae, Reoviridae, Birnaviridae,
Picornaviridae, Flaviviridae, Arenaviridae, Hepeviridae, Rhabdoviridae, Paramoxyviridae, Bunyaviridae, Orthomoxyviridae, Filoviridae, Retroviridae, and Hepadnaviridae.
184. The use of embodiment 183, where the virus of a family Herpesviridae is herpes simplex virus-1 , herpes simplex virus-2, herpes herpesvirus-5, Kaposi's sarcoma-associated herpesvirus, varicella zoster virus, or Epstein-Barr virus.
185. The use of embodiment 183, where the virus of Polyomaviridae family is SV40. 186. The use of embodiment 183, where the virus of Poxviridae family is vaccinia virus.
187. The use of embodiment 183, where the virus of Reoviridae family is rotavirus.
188. The use of embodiment 183, where the virus of Birnaviridae family is infectious bursal disease virus.
189. The use of embodiment 183, where the virus of Picornaviridae family is poliovirus, rhinovirus, or coxsackievirus.
190. The use of embodiment 183, where the virus of Flaviviridae family is hepatitis C virus or dengue virus.
191 . The use of embodiment 183, where the virus of Arenaviridae family is lymphocytic choriomeningitis virus. 192. The use of embodiment 183, where the virus of Hepeviridae family is Hepatitis E virus.
193. The use of embodiment 183, where the virus of Rhabdoviridae family is vesicular stomatitis virus.
194. The use of embodiment 183, where the virus of Paramoxyviridae family is human parainfluenza virus 2, human parainfluenza virus 3, SV5, SV41 , measles virus, or Sendai virus.
195. The use of embodiment 183, where the virus of Bunyaviridae family is La Crosse virus.
196. The use of embodiment 183, where the virus of Orthomoxyviridae family is influenza A virus.
197. The use of embodiment 183, where the virus of Filoviridae family is Ebola virus. 198. The use of embodiment 183, where the virus of Retroviridae family is HTLV1 or HIV1 .
199. The use of embodiment 183, where the virus of Hepadnaviridae family is hepatitis B virus. 200. The use of embodiment 172 or 173, where the infectious disease is a fungal infection.
201 . The use of embodiment 200, where the fungal infection is a Candida albicans infection, an
Aspergillus fumigates infection, or Pneumocystis jiroveci infection. 202. The use of embodiment 172 or 173, where the infectious disease is a bacterial infection.
203. The use of embodiment 202, where the bacterial infection is a mycobacteria infection or anthrax infection. 204. The use of embodiment 203, where the bacterial infection is a bacterial pneumonia.
205. The use of embodiment 172 or 173, where the disorder an inflammatory or autoimmune disease.
206. The use of embodiment 205, where the inflammatory or autoimmune disease is rheumatoid arthritis, systemic lupus erythermatosus, or asthma.
207. The use of embodiment 172 or 173, where the disorder is a cardiovascular disease.
208. The use of embodiment 207, where the cardiovascular disease is atherosclerosis cardiomyopathy.
209. The use of embodiment 172 or 173, where the disorder is an allergy.
210. The compound of any one of embodiments 134 to 171 or the use of any one of embodiments 172 to 209, where the compound is formulated for administration by a route selected from the group consistin of oral, sublingual, buccal, transdermal, intradermal, intramuscular, parenteral, intravenous, intra-arterial, intracranial, subcutaneous, intraorbital, intraventricular, intraspinal, intraperitoneal, intranasal, by inhalation, and topical.
21 1 . The compound or the use of embodiment 128, where the compound is formulated for oral administration.
212. A method of inhibiting Hsp90, the method including contacting a cell with the compound of any one of embodiments 1 to 87 or a pharmaceutically acceptable salt thereof. 213. The method of embodiment 212, where the cell is in vitro.
214. A compound for use in inhibiting Hsp90, where the compound is the compound of any one of embodiments 1 to 87 or a pharmaceutically acceptable salt thereof.
215. Use of a compound for inhibiting Hsp90, where the compound is the compound of any one of embodiments 1 to 87 or a pharmaceutically acceptable salt thereof.
216. A kit including:
(i) the pharmaceutical composition of any one of embodiments 88 to 90; and
(ii) instructions for use of the pharmaceutical compositions of (i) to treat a disorder in a mammal caused by the action of Hsp90.
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each independent publication or patent application was specifically and individually indicated to be incorporated by reference. While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth, and follows in the scope of the claims.
Other embodiments are in the claims.

Claims

Claims
1 . A compound according to formula (I):
R! .R2
N
R N^N
Z1 (I), or a pharmaceutically acceptable salt thereof,
wherein
Z1 is -OR7, -N(R10)R7, -SR7, or -C(R10)(R11)R7;
Z2 is -C(R3)= or -N=;
each R1 and R2 is, independently, H, optionally substituted Ci-3 acyl, or optionally substituted C1 alkyl;
R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one to three heteroatoms selected from the group consisting of sulfur, oxygen, and nitrogen, wherein said nitrogen is optionally substituted with R9, or R3 is H, halogen, cyano, optionally substituted Ci-6 alkyl, optionally substituted alkoxy, or optionally substituted amino, and R4 is halogen, cyano, optionally substituted Ci-6 alkyl, optionally substituted Ci-3 alkoxy, optionally substituted amino, optionally substituted Ci-6 thioalkoxy, or optionally substituted C6-io aryl;
R5 is halogen, H, optionally substituted C1-3 alkyl, optionally substituted C1-3 alkoxy, or CN;
R6 is halogen, H, optionally substituted C1-3 alkyl, optionally substituted C1-3 alkoxy, or CN;
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five-, six-, or seven- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur; or R7 is optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl, and R8 is H;
R9 is H, optionally substituted Ci-3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-10 aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted C1-3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1-3 alkaryl;
R10 is H, optionally substituted C1-3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted alkcycloalkyl, optionally substituted alkheterocyclyl, or optionally substituted Ci-3 alkaryl, and R11 is H, optionally substituted alkyl, or R10 and R11 combine to form =0 or =S; and
Rm is H, halogen, cyano, optionally substituted C1-4 acyl, optionally substituted C1-4 alkyl, or optionally substituted C1-3 alkoxy;
wherein,
when Z2 is CR3, each of R1 and R2 is H, R3 is H, R4 is methyl or chloro, and each of R5 and R6 is chloro,
Z1 is not methoxy; when Z2 is N, each of R5 and R6 is chloro, R3 is H, R4 is substituted thioalkoxy, Z1 is not cyanomethoxy or aminomethoxy;
when Z2 is CR3, each of R5 and R6 is chloro, R3 is H, and R4 is halogen,
Z1 is not 2-amino-2-oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy;
when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma):
Figure imgf000153_0001
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl;
when each R5 and R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl-methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl- 1 ,2,4-oxadiazol-5-yl-methyl;
when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (Ma),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl when R5 is methoxy, R6 is methyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when R5 is chloro, R6 is ethyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
Figure imgf000153_0002
R7 is not methyl or 2-(N,N-diethylamino)ethyl;
when R7 is methyl, R5 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ilia),
HN^ (ma),
R6 is not bromo;
when R5 is chloro, R6 is methoxy, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (1 Mb) :
Figure imgf000154_0001
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl ;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVa) :
Figure imgf000154_0002
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (I Vb) :
Figure imgf000154_0003
R7 is not 2-methoxyethyl or benzyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVc) :
Figure imgf000154_0004
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (I Vd) :
Figure imgf000154_0005
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVe) or (IVf) :
Figure imgf000154_0006
R7 is not benzyl ; when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
Figure imgf000155_0001
R7 is not methyl;
when R6 is methyl,
each R1 and R2 is H; and
when R3 is H, Z1 is -OR7, and each R5 and R6 is chloro,
R7 is not methyl.
2. The compound of claim 1 , wherein Rm is H.
3. A compound according to formula (la):
Figure imgf000155_0002
or a pharmaceutically acceptable salt thereof,
wherein
each R1 and R2 is, independently, H or optionally substituted C1 -3 alkyl;
R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising from one to three heteroatoms selected from the group consisting of sulfur, oxygen, and nitrogen, wherein said nitrogen is optionally substituted with R9, or R3 is H, halogen, cyano, optionally substituted Ci_s alkyl, optionally substituted Ci-3 alkoxy, or optionally substituted amino, and R4 is halogen, cyano, optionally substituted Ci-6 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted amino, optionally substituted C1 -6 thioalkyl, or optionally substituted C6-10 aryl;
R5 is halogen, H, optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkoxy, or CN;
R6 is halogen, H, optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkoxy, or CN;
R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five-, six-, or seven- membered ring optionally comprising one or two heteroatoms selected from nitrogen, oxygen, and sulfur; or R7 is optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted Ci-3 alkaryl, and R8 is H;
R9 is H, optionally substituted C1 -3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-10 aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted C1 -3 alkcycloalkyl, optionally substituted C1 -3 alkheterocyclyl, or optionally substituted C1 -3 alkaryl;
Z1 is -OR7, -N(R10)R7, -SR7, or -C(R10)(R1 1)R7; and
R10 is H, optionally substituted Ci-3 acyl, optionally substituted Ci-3 alkyl, optionally substituted C3. 8 cycloalkyl, optionally substituted C6-io aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted C1 -3 alkaryl, and R1 1 is H, optionally substituted C1 -3 alkyl, or R10 and R1 1 combine to form =0 or =S;
wherein,
when each of R1 and R2 is H, R3 is H, R4 is methyl or chloro, and each of R5 and R6 is chloro, Z1 is not methoxy;
when each of R5 and R6 is chloro, R3 is H, and R4 is halogen,
Z1 is not 2-amino-2-oxoethoxy, 2-(N,N-diethylamino)ethoxy, methoxy, or benzyloxy;
when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma):
Figure imgf000156_0001
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)ethyl, difluoromethyl, or 2-(f-butylamino)ethyl;
when each R5 and R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl-methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl- 1 ,2,4-oxadiazol-5-yl-methyl;
when each R5 and R6 is chloro, Z1 is -OR7, R7 and R8 combine to form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (Ma),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl when R5 is methoxy, R6 is methyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when R5 is chloro, R6 is ethyl, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ma),
R7 is not methyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
Figure imgf000157_0001
R7 is not methyl or 2-(N,N-diethylamino)ethyl;
when R7 is methyl, R5 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (Ilia),
Figure imgf000157_0002
R6 is not bromo;
when R5 is chloro, R6 is methoxy, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (1Mb):
Figure imgf000157_0003
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVa):
Figure imgf000157_0004
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (I Vb):
Figure imgf000157_0005
R7 is not 2-methoxyethyl or benzyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVc):
Figure imgf000157_0006
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (I Vd):
Figure imgf000158_0001
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl;
when each R5 and R6 is chloro, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVe) or (IVf):
Figure imgf000158_0002
R7 is not benzyl;
when R5 is chloro, R6 is bromo, Z1 is -OR7, and R3 and R4 combine to form a group according to formula (IVg), (IVh), (IVi), (IVj), or (IVk):
(IVj), or
Figure imgf000158_0003
R7 is not methyl;
when R6 is methyl,
each R1 and R2 is H; and
when R3 is H, Z1 is -OR7, and each R5 and R6 is chloro,
R7 is not methyl.
4. A compound according to formula (lb):
Figure imgf000158_0004
(lb), or a pharmaceutically acceptable salt thereof,
wherein
each of R1 and R2 is, independently, H or optionally substituted alkyl;
R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one sulfur, or one oxygen, wherein the nitrogen is optionally substituted with R9, or R3 is H, halogen, cyano, optionally substituted alkyl, optionally substituted C1 -3 alkoxy, or optionally substituted amino, and R4 is halogen, cyano, optionally substituted alkyl, optionally substituted C1 -3 alkoxy, optionally substituted amino, optionally substituted C1 -6 thioalkoxy, or optionally substituted C6-10 aryl;
R5 is halogen, H, optionally substituted alkyl, optionally substituted alkoxy, or CN;
R6 is halogen, H, optionally substituted alkyl, optionally substituted alkoxy, or CN;
R7 is optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkcycloalkyl, optionally substituted Ci-3 alkheterocyclyl, or optionally substituted alkaryl, and R8 is H; or R7 and R8, together with the atoms to which each is attached, join to form an optionally substituted five-, six-, or seven- membered ring; and
R9 is H, optionally substituted C1 -3 alkyl, optionally substituted C3.8 cycloalkyl, optionally substituted C6-10 aryl, optionally substituted C2-9 heteroaryl, optionally substituted C2-9 heterocyclyl, optionally substituted alkcycloalkyl, optionally substituted alkheterocyclyl, or optionally substituted Ci-3 alkaryl;
wherein,
when each of R1 and R2 is H, R3 is H, R4 is methyl or chloro, and each of R5 and R6 is chloro, R7 is not methyl;
when each of R5 and R6 is chloro, R3 is H, and R4 is halogen,
R7 is not 2-amino-2-oxoethyl, 2-(N,N-diethylamino)ethyl, methyl, or benzyl;
when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (lla) :
r9 (Ma),
R7 is not methyl, ethyl, n-propyl, 2-(N-pyrazolyl)ethyl, 2-(N-imidazolyl)ethyl, 3-hydroxypropyl, cyanomethyl, 2-chloroethyl, 2-hydroxyethyl, 2-oxo-propyl, 2-(N,N-dimethylamino)-ethyl, difluoromethyl, or 2-(f-butylamino)ethyl;
when each R5 and R6 is bromo, and R3 and R4 combine to form a group according to formula
(lla),
R7 is not methyl;
when each R5 and R6 is chloro, R8 is H, and R3 and R4 combine to form a group according to formula (lla),
R7 is not methyl, 2-(N-imidazolyl)ethyl, methoxymethyl, 2-(N-pyrazolyl)ethyl, 2-(3-methylpyrazol- 1 -yl)ethyl, 2-pyridyl-methyl, 1 ,3-dimethyl-1 H-1 ,2,4-triazol-5-yl-methyl, 2-pyrimidinylmethyl, imidazol-2-yl-methyl, 5-methyl-isoxazol-3-yl-methyl, 4-methyl-imidazol-5-yl-methyl, or 3-methyl- 1 ,2,4-oxadiazol-5-yl-methyl;
when each R5 and R6 is chloro, R7 and R8 combine to form -CH2-CH2-, and R3 and R4 combine to form a group according to formula (lla),
R9 is not ethoxycarbonyl, cyclobutylaminocarbonyl, or cyclobutadienylaminocarbonyl;
when R5 is methoxy, R6 is methyl, and R3 and R4 combine to form a group according to formula
(lla),
R7 is not methyl;
when R5 is chloro, R6 is ethyl, and R3 and R4 combine to form a group according to formula (lla), R7 is not methyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (Mb) or (lie),
Figure imgf000160_0001
R7 is not methyl or 2-(N,N-diethylamino)ethyl;
when R7 is methyl, R5 is chloro, and R3 and R4 combine to form a group according to formula
(Ilia),
(Ilia),
R is not bromo;
when R5 is chloro, R6 is methoxy, and R3 and R4 combine to form a group according to formula
(1Mb) :
Figure imgf000160_0002
neither R1 nor R2 is 2-(N,N-diethylamino)ethyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula
(IVa):
Figure imgf000160_0003
R7 is not 3-(N-morpholinyl)propyl, benzyl, 1 -ethyl-pyrrolydin-3-yl, 1 -methyl-piperidin-4-yl, 2-(1 - methyl-pyrrolidin-2-yl)ethyl, or 3-(N,N-diethylamino)propyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula
(IVb):
Figure imgf000160_0004
R7 is not 2-methoxyethyl or benzyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula
(IVc):
Figure imgf000160_0005
R7 is not 2-(N,N-diethylamino)ethyl or 3-(N,N-dimethylamino)propyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula
(IVd):
Figure imgf000161_0001
R7 is not 2-(pyrrolidin-1 -yl)ethyl or 2-hydroxyethyl;
when each R5 and R6 is chloro, and R3 and R4 combine to form a group according to formula (IVe) or (IVf):
Figure imgf000161_0002
R7 is not benzyl;
when R5 is chloro, R6 is bromo, and R3 and R4 combine to form a group according to formula (IVg
Figure imgf000161_0003
R7 is not methyl;
when R6 is methyl,
each R1 and R2 is H; and
when R3 is H, and each R5 and R6 is chloro,
R7 is not methyl.
5. The compound of any one of claims 1 to 4, wherein R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five- or six-membered ring optionally comprising one nitrogen, one oxygen, or one sulfur, wherein said nitrogen is optionally substituted with R9, or R3 is H, halogen, optionally substituted C1 -3 acyl, optionally substituted C1 -3 alkyl, or optionally substituted C1 -3 alkoxy, and R4 is halogen, optionally substituted C1 -3 acyl, optionally substituted C1 -3 alkyl, optionally substituted Ci-3 alkoxy, optionally substituted amino, optionally substituted Ci-6 thioalkyl, or optionally substituted C6-io aryl.
6. The compounds of any one of claims 1 to 5, wherein R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one sulfur.
7. The compound of any one of claims 1 to 5, wherein R3 and R4 combine to form -C(R13A)=C(R13B)-S- group, wherein R13A is H, and R13B is optionally substituted Ci-3 alkyl or H.
8. The compound of claim 1 1 , wherein R13B is optionally substituted C1 -3 alkyl.
9. The compound of claim 12, wherein R13B is -C(0)-R13C, wherein R13C is optionally substituted amino or optionally substituted Ci-3 alkoxy.
10. The compound of any one of claims 1 to 5, wherein R3 and R4 combine to form -C(R13A)=C(R13B)-S- group, wherein R13A is H, and R13B is -C(0)-R13C,H, or C1 -6 aminoalkyl, wherein R13C is optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkoxy, optionally substituted amino, or optionally substituted C2-9 heterocyclyl.
1 1 . The compound of any one of claims 1 to 5, wherein R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring.
12. The compound of any one of claims 1 to 5, wherein R3 and R4 combine to form -CH2CH2CH2- group.
13. The compound of any one of claims 1 to 5, wherein R3 and R4, together with the atoms to which each is attached, join to form an optionally substituted five-membered ring comprising one nitrogen.
14. The compound of claim 13, wherein R3 and R4 combine to form -N(R9)-CH=CH- group.
15. The compound of any one of claims 1 to 5, wherein R3 and R4 combine to form -X1-X2-X3-, wherein
X1 is -S-, -0-, -(CR14R15)-, -C(R16)=, -N(R9)-, -N=, H, or optionally substituted C1 -3 alkyl; X2 is absent,-(CR17R18) -, -S-, -0-, -N=, -N(R9)-, -C(R19)=, =N- =C(R20)-, or =C(R21)-
C(R22)=;
X3 is -(CR14R15)-, -S-, -0-, -N(R9)-, =N- =C(R23)-, halogen, optionally substituted d_3 alkyl, optionally substituted Ci_s thioalkoxy, optionally substituted alkoxy, or optionally substituted C6-io aryl; each R14 and R15 is, independently, H or optionally substituted Ci-3 alkyl, or R14 and R15 combine to form =0 or =S;
each R17 and R18 is, independently, H or optionally substituted C1 -3 alkyl, or R17 and R18 combine to form =0 or =S;
each R16, R19, R20, R21 , R22, and R23 is, independently, H, or optionally substituted C1 -3 alkyl; and n is 1 or 2.
16. The compound of any one of claims 1 to 15, wherein R7 and R8 form a group -Y1-Y2-, wherein:
Y1 is -(CR26R27)m- or optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkheterocyclyl, optionally substituted C1 -3 alkcycloalkyl, or optionally substituted C1 -3 alkaryl; and
Y2 is -(CR26R27)- or H; wherein
each R26 and R27 is, independently, H or optionally substituted C1 -3 alkyl;
m is 1 , 2, or ; and
when Y1 is optionally substituted Ci-3 alkyl, optionally substituted Ci-3 alkheterocyclyl, optionally substituted Ci-3 alkcycloalkyl, or optionally substituted Ci-3 alkaryl, Y2 is H, and Y1 is not directly bonded to Y2.
17. The compound of any one of claims 1 to 5, wherein Z1 and R8 combine to form
-Z3-Y1-Y2-, wherein
Z3 is -0-, -N(R10)-, -N= -S-, or -(CR14R15)-;
Y1 is -0-, -N(R10)-, -S-, -(CR26R27)m- -C(R20)=, =C(R20)-, =C(R21)-C(R22)=, optionally substituted C1 -3 alkyl, optionally substituted C1 -3 alkheterocyclyl, optionally substituted C1 -3 alkcycloalkyl, or optionally substituted C1 -3 alkaryl; and
Y2 is -0-, -S-, -N(R10)-, -(CR26R27)-, =C(R20)-, =N-, or H;
wherein
each R20, R21 , and R22 is, independently, H or optionally substituted Ci-3 alkyl; and
each R26 and R27 is, independently, H or optionally substituted Ci-3 alkyl, or R26 and R27 combine to form =0 or =S;
m is 1 or 2; and
wherein, when Y2 is H,
the chain of atoms -Z3-Y1-Y2- comprises no more than two heteroatoms, each said heteroatom independently selected from nitrogen, oxygen, and sulfur.
18. A compound:
Figure imgf000163_0001
Figure imgf000164_0001
ı62
Figure imgf000165_0001
ı63
Figure imgf000166_0001
or a pharmaceutically acceptable salt thereof.
Figure imgf000167_0001
ı65
Figure imgf000168_0001
Figure imgf000169_0001
or a pharmaceutically acceptable salt thereof.
20. The compound of claim 19, wherein said compound is:
Figure imgf000169_0002
pharmaceutically acceptabl salt thereof.
21 . A pharmaceutical composition comprising the compound of any one of claims 1 to 20, or a pharmaceutically acceptable salt thereof, and one or more of pharmaceutically acceptable carriers or excipients.
22. The pharmaceutical composition of claim 21 , wherein said composition is formulated for administration orally, sublingually, buccally, transdermally, intradermally, intramuscularly, parenterally, intravenously, intra-arterially, intracranially, subcutaneously, intraorbitally, intraventricularly, intraspinally, intraperitoneally, intranasally, by inhalation, and topically.
23. The pharmaceutical composition of claim 22, wherein said composition is formulated for oral administration.
24. A method of treating a disorder in a mammal caused by the action of heat shock protein 90 (Hsp90), said method comprising administering an effective amount of the compound of any one of claims 1 to 20 or a pharmaceutically acceptable salt thereof or the pharmaceutical composition of claim 21 to said mammal.
25. The method of claim 25, wherein said disorder is a neurodegenerative disorder.
26. The method of claim 25, wherein said neurodegenerative disorder is a tauopathy.
27. The method of claim 24 or 25, wherein said neurodegenerative disorder is Alzheimer's disease, Huntington's disease, progressive supranuclear palsy, Parkinson's disease, Pick's disease, corticobasal degeneration, chronic traumatic encephalopathy, traumatic brain injury, or frontotemporal dementia.
28. The method of claim 27, wherein said neurodegenerative disorder is Alzheimer's disease.
29. The method of claim 24, wherein said disorder is a proliferative disorder.
30. The method of claim 29, wherein said proliferative disorder is a cancer.
31 . A method of treating an infectious disease in a mammal, said method comprising administering an effective amount of the compound of any one of claims 1 to 20 or a pharmaceutically acceptable salt or the pharmaceutical composition of claim 21 thereof to said mammal.
32. The method of claim 31 , wherein said infectious disease is a viral infection.
33. The method of claim 32, wherein said viral infection is an infection by a virus of a family selected from the group consisting of Herpesviridae, Polyomaviridae, Poxviridae, Reoviridae, Birnaviridae,
Picornaviridae, Flaviviridae, Arenaviridae, Hepeviridae, Rhabdoviridae, Paramoxyviridae, Bunyaviridae, Orthomoxyviridae, Filoviridae, Retroviridae, and Hepadnaviridae.
34. The method of claim 31 , wherein said infectious disease is a fungal infection.
35. The method of claim 31 , wherein said infectious disease is a bacterial infection.
36. The method of claim 24, wherein said disorder an inflammatory or autoimmune disease.
37. The method of claim 36, wherein said inflammatory or autoimmune disease is rheumatoid arthritis, systemic lupus erythermatosus, or asthma.
38. The method of claim 24, wherein said disorder is a cardiovascular disease.
39. The method of claim 38, wherein said cardiovascular disease is atherosclerosis.
40. The method of claim 24, wherein said disorder is an allergy.
41 . The method of any one of claims 24 to 40, wherein said compound is administered orally, sublingually, buccally, transdermally, intradermally, intramuscularly, parenterally, intravenously, intra- arterially, intracranially, subcutaneously, intraorbitally, intraventricularly, intraspinally, intraperitoneally, intranasally, by inhalation, and topically.
42. The method of claim 41 , wherein said compound is administered orally.
43. The method of any one of claims 24 to 42, wherein said mammal is human.
44. A method of inhibiting Hsp90, said method comprising contacting a cell with the compound of any one of claims 1 to 20 or a pharmaceutically acceptable salt thereof.
45. The method of claim 44, wherein said cell is in vitro.
46. A kit comprising:
(i) the pharmaceutical composition of any one of claims 21 to 23; and
(ii) instructions for use of the pharmaceutical compositions of (i) to treat a disorder in a mammal caused by the action of Hsp90.
PCT/US2015/035735 2014-06-13 2015-06-15 Pyrimidine compounds and methods using the same WO2015192119A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
AU2015274285A AU2015274285C1 (en) 2014-06-13 2015-06-15 Pyrimidine compounds and methods using the same
JP2017518041A JP6779867B2 (en) 2014-06-13 2015-06-15 Pyrimidine compounds and how to use them
EP15806816.3A EP3154547B1 (en) 2014-06-13 2015-06-15 Pyrimidine compounds and methods using the same
US15/317,999 US10336768B2 (en) 2014-06-13 2015-06-15 Pyrimidine compounds and methods using the same
CA2952230A CA2952230C (en) 2014-06-13 2015-06-15 Pyrimidine compounds and methods using the same
ES15806816T ES2954453T3 (en) 2014-06-13 2015-06-15 Pyrimidine compounds and methods using the same
CN201580043036.0A CN106687114B (en) 2014-06-13 2015-06-15 Pyrimidine compounds and methods of use thereof
US16/459,138 US10961254B2 (en) 2014-06-13 2019-07-01 Pyrimidine compounds and methods using the same
US17/216,076 US20210363154A1 (en) 2014-06-13 2021-03-29 Pyrimidine compounds and methods using the same

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462012152P 2014-06-13 2014-06-13
US62/012,152 2014-06-13

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/317,999 A-371-Of-International US10336768B2 (en) 2014-06-13 2015-06-15 Pyrimidine compounds and methods using the same
US16/459,138 Continuation US10961254B2 (en) 2014-06-13 2019-07-01 Pyrimidine compounds and methods using the same

Publications (1)

Publication Number Publication Date
WO2015192119A1 true WO2015192119A1 (en) 2015-12-17

Family

ID=54834482

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/035735 WO2015192119A1 (en) 2014-06-13 2015-06-15 Pyrimidine compounds and methods using the same

Country Status (8)

Country Link
US (3) US10336768B2 (en)
EP (1) EP3154547B1 (en)
JP (1) JP6779867B2 (en)
CN (1) CN106687114B (en)
AU (1) AU2015274285C1 (en)
CA (1) CA2952230C (en)
ES (1) ES2954453T3 (en)
WO (1) WO2015192119A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190096917A (en) * 2019-08-12 2019-08-20 씨제이헬스케어 주식회사 Heterocylic compound as a protein kinase inhibitor
US11524968B2 (en) 2017-10-18 2022-12-13 Hk Inno.N Corporation Heterocyclic compound as a protein kinase inhibitor

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3154547B1 (en) * 2014-06-13 2023-06-07 Yuma Therapeutics, Inc. Pyrimidine compounds and methods using the same
JP7156154B2 (en) * 2019-04-18 2022-10-19 株式会社島津製作所 Medium processing system and medium processing method
US20230390416A1 (en) * 2020-10-16 2023-12-07 The Brigham And Women's Hospital, Inc. Compositions for inducing tumor immunity and reducing drug tolerance

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5112598A (en) 1988-05-04 1992-05-12 Hermes Fabrik Pharmazeutischer Preparate Franz Gradinger Gmbh & Co. Kg Vitamin a aerosol-inhalate preparations
WO2008059368A2 (en) * 2006-11-17 2008-05-22 Pfizer Products Inc. Fused 2-amino pyrimidine compounds and their use for the treatment of cancer
US20100093696A1 (en) 2007-02-06 2010-04-15 Pfizer Inc. 2-amino pyrimidine compounds
US20110201587A1 (en) 2010-02-16 2011-08-18 Bio Holding, Inc. Hsp90 inhibitors and methods of use
WO2013177535A2 (en) * 2012-05-25 2013-11-28 Berg Pharma Llc Methods of treating a metabolic syndrome by modulating heat shock protein (hsp) 90-beta
US8642594B2 (en) * 2003-08-29 2014-02-04 Vernalis (R&D) Limited Substituted thieno[2,3-d]pyrimidines as HSP90 inhibitors

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2691655A (en) * 1952-05-24 1954-10-12 Burroughs Wellcome Co 2-amino-4-substituted amino-6-aryl pyrimidines and process of preparing same
TW440563B (en) * 1996-05-23 2001-06-16 Hoffmann La Roche Aryl pyrimidine derivatives and a pharmaceutical composition thereof
DE19735800A1 (en) * 1997-08-18 1999-02-25 Boehringer Ingelheim Pharma Use of known and new triazine derivatives
GB0416168D0 (en) * 2004-07-20 2004-08-18 Vernalis Cambridge Ltd Pyrmidothiophene compounds
WO2006090094A1 (en) 2005-02-28 2006-08-31 Vernalis R & D Ltd Pyrimidothiophene compounds for use as hsp90 inhibitors
DE102005022977A1 (en) * 2005-05-19 2006-12-07 Merck Patent Gmbh Phenylchinazolinderivate
US20090215777A1 (en) 2005-05-19 2009-08-27 Astex Therapeutics Ltd. Pyrimidine Derivatives As HSP90 Inhibitors
DE102006008880A1 (en) 2006-02-27 2007-09-06 Merck Patent Gmbh aminopyrimidine derivatives
AU2007226344A1 (en) 2006-03-11 2007-09-20 Vernalis (R & D) Ltd. Pyrrolopyrimidine derivatives used as HSP90 inhibitors
AR061185A1 (en) 2006-05-26 2008-08-13 Chugai Pharmaceutical Co Ltd HETEROCICLICAL COMPOUNDS AS INHIBITORS OF HSP90. PHARMACEUTICAL COMPOSITIONS.
JP2009067729A (en) * 2007-09-14 2009-04-02 Kyowa Hakko Kirin Co Ltd Hsp90 FAMILY PROTEIN INHIBITOR
WO2012101654A2 (en) 2011-01-25 2012-08-02 Sphaera Pharma Pvt. Ltd Novel triazine compounds
WO2013157021A1 (en) * 2012-04-20 2013-10-24 Advinus Therapeutics Limited Bicyclic compounds, compositions and medicinal applications thereof
KR102194646B1 (en) * 2012-11-27 2020-12-23 토마스 헬데이스 스티프텔스 퓌어 메디신스크 포르스닝 Pyrimidine-2,4-diamine derivatives for treatment of cancer
SG11201609981RA (en) 2014-06-04 2016-12-29 Thomas Helledays Stiftelse För Medicinsk Forskning Mth1 inhibitors for treatment of inflammatory and autoimmune conditions
EP3154547B1 (en) * 2014-06-13 2023-06-07 Yuma Therapeutics, Inc. Pyrimidine compounds and methods using the same
CA2961793C (en) * 2014-09-19 2021-03-16 Forma Therapeutics, Inc. Quinolinone pyrimidines compositions as mutant-isocitrate dehydrogenase inhibitors

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5112598A (en) 1988-05-04 1992-05-12 Hermes Fabrik Pharmazeutischer Preparate Franz Gradinger Gmbh & Co. Kg Vitamin a aerosol-inhalate preparations
US5556611A (en) 1988-05-04 1996-09-17 Hermes Fabrik Pharmazeutischer Praparate Vitamin A aerosol-inhalant preparations and method
US8642594B2 (en) * 2003-08-29 2014-02-04 Vernalis (R&D) Limited Substituted thieno[2,3-d]pyrimidines as HSP90 inhibitors
WO2008059368A2 (en) * 2006-11-17 2008-05-22 Pfizer Products Inc. Fused 2-amino pyrimidine compounds and their use for the treatment of cancer
US20100093696A1 (en) 2007-02-06 2010-04-15 Pfizer Inc. 2-amino pyrimidine compounds
US20110201587A1 (en) 2010-02-16 2011-08-18 Bio Holding, Inc. Hsp90 inhibitors and methods of use
WO2013177535A2 (en) * 2012-05-25 2013-11-28 Berg Pharma Llc Methods of treating a metabolic syndrome by modulating heat shock protein (hsp) 90-beta

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
"ELISA to AlphaLISA Conversion Guide", August 2012, PERKINELMER, INC.
"Encyclopedia of Pharmaceutical Technology", 1988, MARCEL DEKKER
"Handbook of Pharmaceutical Excipients", 2009, PHARMACEUTICAL PRESS
"Pharmaceutical Salts: Properties, Selection, and Use,", 2008, WILEY-VCH
"Remington: The Science and Practice of Pharmacy", 2005, LIPPENCOTT WILLIAMS & WILKINS
"The United States Pharmacopeia: The National Formulary (USP 36 NF31", 2013
BERGE ET AL.: "J. Pharmaceutical Sciences", vol. 66, 1977, pages: 1 - 19
CORCORAN ET AL., J. CLIN. NEUROSCIENCE, vol. 17, 2010, pages 1025 - 1033
GELLER ET AL., BIOCHIM. BIOPHYS. ACTA - MOL. CELL RES., vol. 1823, 2012, pages 698 - 706
GREENE: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
HIGUCHISTELLA: "Bioreversible Carriers in Drug Design", vol. 14, 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS, article "Prodrugs as Novel Delivery Systems"
JUDKINS ET AL., SYNTHETIC COMMUNICATIONS, vol. 26, no. 23, 1996, pages 4351 - 4367
LIU ET AL., BIOCHEMISTRY, vol. 49, 2010, pages 4921 - 4929
LOEFFLER ET AL., J. MOL. NEUROSCI., vol. 47, 2012, pages 192 - 203
MIYAURA ET AL.: "Topics in Current Chemistry", 2002, SPRINGER, article "Cross-Coupling Reactions: A Practical Guide"
NICOLAOU ET AL., ANGEW. CHEM. INT. ED., vol. 44, 2005, pages 4442 - 4489
See also references of EP3154547A4

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11524968B2 (en) 2017-10-18 2022-12-13 Hk Inno.N Corporation Heterocyclic compound as a protein kinase inhibitor
KR20190096917A (en) * 2019-08-12 2019-08-20 씨제이헬스케어 주식회사 Heterocylic compound as a protein kinase inhibitor
KR102112336B1 (en) 2019-08-12 2020-05-18 에이치케이이노엔 주식회사 Heterocyclic compound as a protein kinase inhibitor

Also Published As

Publication number Publication date
EP3154547B1 (en) 2023-06-07
ES2954453T3 (en) 2023-11-22
US20190389878A1 (en) 2019-12-26
US20210363154A1 (en) 2021-11-25
AU2015274285A1 (en) 2017-02-02
US10336768B2 (en) 2019-07-02
CN106687114A (en) 2017-05-17
CA2952230A1 (en) 2015-12-17
CN106687114B (en) 2021-08-31
JP2017521489A (en) 2017-08-03
EP3154547A4 (en) 2018-01-24
JP6779867B2 (en) 2020-11-04
EP3154547A1 (en) 2017-04-19
AU2015274285B2 (en) 2020-12-10
AU2015274285C1 (en) 2021-07-01
US20170107228A1 (en) 2017-04-20
CA2952230C (en) 2023-03-21
US10961254B2 (en) 2021-03-30

Similar Documents

Publication Publication Date Title
US10961254B2 (en) Pyrimidine compounds and methods using the same
DK2880035T3 (en) Hitherto UNKNOWN PYRROLOPYRIMIDINE COMPOUNDS AS PROTEIN CHINAS INHIBITORS
KR101792837B1 (en) Imidazopyrazines for use as kinase inhibitors
KR101828187B1 (en) Novel fused pyrimidine compound or salt thereof
CN113307811B (en) Tetrahydropyranyl amino-pyrrolopyrimidinones and methods of use thereof
EP2984088B1 (en) Quinazolines and azaquinazolines as dual inhibitors of ras/raf/mek/erk and pi3k/akt/pten/mtor pathways
CN110603256B (en) Pyrimidopyrimidinones useful as WEE-1 kinase inhibitors
CN111770914B (en) Compounds as neurokinin-1 receptor antagonists and uses thereof
US20180230152A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
AU2017204973A1 (en) Novel fluorinated quinazoline derivatives as EGFR inhibitors
WO2016139292A1 (en) Pyridinone compound, pharmaceutical composition containing the same and use
AU2018201295A1 (en) Benzhydrol-pyrazole derivatives having kinase inhibitory activity and uses thereof
CA3043288A1 (en) 3-amino-1,5-dihydro-pyrazolo[3,4-d]pyrimidin-4-ones as cyclin dependent kinase inhibitors
CN107686477B (en) Novel compounds as CDK4/6 inhibitors and uses thereof
AU2023209945A1 (en) Wee1 inhibitor, preparation therefor, and use thereof
EP3632912B1 (en) Pyridoquinazoline derivatives useful as protein kinase inhibitors
CN107652284B (en) CDK inhibitors for the treatment of proliferative diseases
CN111377922B (en) Fused tricyclic compounds and uses thereof
TW202402273A (en) WEE1 inhibitor, preparation therefor and use 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: 15806816

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15317999

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2017518041

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2952230

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015806816

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015806816

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2015274285

Country of ref document: AU

Date of ref document: 20150615

Kind code of ref document: A