WO2008014236A1 - Dimeric iap inhibitors - Google Patents

Dimeric iap inhibitors Download PDF

Info

Publication number
WO2008014236A1
WO2008014236A1 PCT/US2007/074181 US2007074181W WO2008014236A1 WO 2008014236 A1 WO2008014236 A1 WO 2008014236A1 US 2007074181 W US2007074181 W US 2007074181W WO 2008014236 A1 WO2008014236 A1 WO 2008014236A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
independently
substituted
aryl
het
Prior art date
Application number
PCT/US2007/074181
Other languages
French (fr)
Inventor
Stephen M. Condon
Original Assignee
Tetralogic Pharmaceuticals Corporation
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 Tetralogic Pharmaceuticals Corporation filed Critical Tetralogic Pharmaceuticals Corporation
Priority to US12/374,731 priority Critical patent/US20100056495A1/en
Publication of WO2008014236A1 publication Critical patent/WO2008014236A1/en
Priority to US13/431,745 priority patent/US20120184530A1/en

Links

Classifications

    • 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
    • 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
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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

Definitions

  • Apoptosis is conserved across species and executed primarily by activated caspases, a family of cysteine proteases that cleave their substrates specifically at aspartate residues.
  • Caspases are produced in cells as catalytically inactive zymogens (procaspases) that are activated by proteolytic processing during the intitation of apoptosis. Once activated, effector caspases proteolytically activate a broad spectrum of cellular targets ultimately leading to cell death.
  • caspases In mammalian cells activation of the caspases is achieved through at least two independent mechanisms which are initiated by distinct caspases, but result in the activation of common executioner (effector) caspases.
  • the 'intrinsic pathway' is activated by cytochrome c which is released from mitochondria within the cell when apoptosis is initiated.
  • the 'extrinsic pathway' is intiated via activation of a death receptor located on the cell membrane.
  • death receptors such as, Fas (CD-95/Apol) and TNF-Rl , as well as other members of the TNF group of cytokine receptors, are activated by their corresponding ligands, Fas ligand (FasL/CD-95L) and TNF-alpha or Apo2 ⁇ igand/TNF-related apoptosis inducing ligand (Apo2L/TR ⁇ IL), respectfully.
  • Binding of procaspase-8 to an activated death receptor induces cleavage and removal of inhibitory domain of procaspase-8 releasing it from the receptor and allowing it to activate effector caspases-3. -6, and -7. The result is the proteolytic cleavage of cellular targets by the effector caspases and the induction of apoptosis.
  • IAPs inhibitors of apoptosis proteins
  • IAPs have been described in organisms ranging from Drosophila to Humans. All mammalian IAPs identified to date, including, for example, XIAP, cIAP-1 , cIAP-2, ML-IAP, NAIP, Bruce, and survivin exhibit anti- apoptotic activity in cell culture,
  • IAPs were originally discovered in Baculovirus by their ability to substitute for P35, an anti-apoptotic protein.
  • IAPs are made up of one to three Baculovirus IAP repeat (BlR) domains, and must also possess a carboxyl-terminal RING finger motif.
  • the BIR domain itself includes a zinc binding domain of about 70 residues made up of 4 alpha-helices and 3 beta strands.
  • the BlR domain itself is believed to inhibit apoptosis by interacting with the procaspase and inhibiting proteolytic activation of the procaspase.
  • IAPs are also known to be overexpressed in many human cancers. For example, XIAP is ubiquitously expressed in most adult and fetal tissues.
  • Smac/DIABLO-derived peptides have also been demonstrated to sensitize tumor cell lines to pro-apoptotic drugs. In non-tumorigenic cells signaled to undergo apoptosis, IAP- mediated inhibition of apoptosis must be eliminated, which is accomplished, at least in part, by Smac (second mitochondrial activator of caspases).
  • Smac. or DIABLO is synthesized in the cytoplasm as a 239 amino acid precursor protein, of which the N-terminal 55 residues serve as the mitochondria targeting sequence that is removed after import to the mitochondria. Mature Smac, containing 184 amino acids, accumulates in the inter-membrane space of the mitochondria where it has been shown to behave as an oligomer.
  • cytochrome c When apoptosis is induced, Smac is released from the mitochondria into the cytosol together with cytochrome c where it binds to IAPs eliminating the inhibitory effect of IAPs on protealysis of procaspases and enabling caspase activation. At the same time, cytochrome c induces multimerization of Apaf-1 to activate procaspase-9 and procaspase-3.
  • Smac interacts with essentially all IAPs identified to date including XIAP, c- IAPl , C-1AP2, ML-IAP, Bruce and survivin and may be a master regulator of apoptosis in mammals.
  • X-ray crystallography has shown that the first four amino acids (AVPI) of mature Smac bind to a portion of IAPs and this binding is thought to be essential for blocking the anti- apoptotic effects of IAPs. Therefore, Smac and various fragments of Smac, including AVPI peptides, have been proposed for use as targets for identification of therapeutic agents.
  • IAP antagonists such as Smac
  • Smac The basic biology of IAP antagonists, such as Smac, suggests that these proteins may complement or synergize other chemotherapeutic/anti -neoplastic agents and/or radiation.
  • Chemotherapeutic/anti-neoplastic agents and radiation would be expected to induce apoptosis as a result of DNA damage and/or the disruption of cellular metabolism.
  • Various embodiments of the invention are directed to a compound including a homodimer or heterodimer having monomeric units of formula (I):
  • each R 1 is, independently, H; C
  • -C 4 -alkynyl or C 3 -C 1O - cycloalkyl which are unsubstituted or substituted; each R 2 is, independently, H; C
  • -C 4 -alkenyl; C r C 4 -alkynyI or C 3 -C 1O - cycloalkyl which are unsubstituted or substituted; each R 3 is, independently, H; -CF 3 ; -C 2 H 5 ; Ci-C 4 -alkyl; C r C 4 -alkeny]; C r C 4 -alkynyl; - CH 2 -Z or any R 2 and R 3 together form a heterocyclic ring; each Z is, independently, H; -OH; F; Cl; -CH 3 ; -CF 3 ; -
  • each Zs is, independently, -N(R
  • R 5 is a residue of an amino acid, wherein the alkyl, cycloalkyl phenyl, and aryl substituents are unsubstituted or substituted; each U is, independently, as shown in structure (II):
  • each D is, independently, -CO-; -C(O)-C ⁇ -7 -alkylene or arylene; -CF 2 -; -0-; -S(0) r where r is 0-2; 1 ,3-dioxalane; or C 1-7 ⁇ alkyl-OH; where alkyl, alkylene, or arylene are unsubstituted or substituted with one or more halogens.
  • each D is, independently, N(R 11 ) wherein each Rh is, independently, H; unsubstituted or substituted C 1 -7 -alkyl; aryl; unsubstituted or substituted -O-(C
  • the compound of invention may have the formula (III):
  • R 1 and R 1 ' are each, independently, H; Ci-G t -alkyl; C
  • R 2 and R 2 ' are each, independently, H; Ci-C ⁇ -alkyl; C,-C4-alkenyl; C
  • R 3 and R 3 ' are each, independently, H; -CF 3 ; -C 2 Hs; C,-C 4 -alkyl; C,-C 4 -alkenyl; C r C 4 - alkynyi; or -CH 2 -Z or R 2 and R 3 together or independently with R 2 ' and R 3 ' form a heterocyclic ring; each Z is
  • R 4 and R 4 ' are each, independently, C ,-C ⁇ , straight or branched alkyl; C,-Ci 6 -alkenyl; C 1 - C 1 6-alkynyl; or C 3 -C,o-cycloaIkyI; -(CHi) 1 -6 -Zt; -(CHi ⁇ -aryl; or -(CH2)o-6-het; wherein alkyl, cycloalkyi, and phenyl are unsubstituted or substituted; each Zt is, independently, -N(R !0 )-C(O)-C M0 -alkyI; -N(R 1 O)-C(O)-(CH 2 WC 3-7 - cycloalkyl; -N(R,o)-C(0)-(CH 2 ) 0 .6-pheny]; -N ⁇ R, 0 )-C(O)-(CH 2 ) s .
  • each het is, independently, a 5-7 member heterocyclic ring containing 1-4 heteroatoms selected from N, O, and S, or an 8- 12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom;
  • each R 1 o is, independently, H; -CH 3 ; -CF 3 ; -CH 2 OH; or -CH 2 Cl;
  • each R) ⁇ and R1 2 is, independently, H; C ⁇ -alkyl; C 3-7 -cyclo
  • -8-alky] or alkylidene, and each R e is either ⁇ nsubstituted or substituted; each Q
  • each D is, independently, -CO-; -C(O)-C 1.7-alkylene or arylene; -CF 2 -; -0-; -S(0) r where r is 0-2; 1,3-dioxalane; or Ci.
  • 7 -alkyl-OH where alkyl, alkylene, or arylene are unsubstituted or substituted with one or more halogens, OH, -O-Ci- 6 -alkyl, -S-C 1-6 -alkyl; or -CF 3 ; or each D is, independently, N(Rh) wherein each Rh is, independently, H; unsubstituted or substituted C].
  • each R ⁇ , R?, R 8 , and R 9 is, independently, H, -Ci-io-alkyl; -C ⁇ jo-alkoxy; aryl-Ci.io- alkoxy; -OH; -O-C M0 -alkyl; -(CH 2 )o-6-C 3 -7-cycloalkyl; -O-(CH 2 ) 0 - 6 -aryl; phenyl; -(CH 2 ) i ⁇ -het; - O-(CH 2 ),.
  • each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; and any R 6 , R 7 , R 8 , and R 9 optionally together form a ring system; each R 13 and R 14 is, independently, H; C ⁇ io-alkyl; -(CH 2 ) 0-6 -C3 -7 -cycloalkyl; -(CH 2 ) O ⁇ - (CH)(M-(ary ⁇ ),.2; -C(O)-C M
  • L is one or more linkers covalently linking one or more of the positions R 4 , R 5 , or U, with R 4 ', R 5 ', or U'; or a pharmaceutically acceptable salt or hydrate thereof.
  • L may covalently link two identical monomeric units or L covalently links two non-identical monomeric units.
  • L may be selected from alkylene, alkenylene, alkynylene, cycloalkylene, cycloalkylalkyiene, aryl, arylalkylene, arylalkylalkylene, and heterocycloalkylene, heterocycloalkylalkylene, heteroaryl and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, optionally-substituted alkylene, alkenylene, alkynylene cycloalkylene, cycloalkylalkyiene, heterocycloalkylene, heterocycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, amino
  • the compounds of the invention may have a formula selected from a compound of formula (IV):
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ' , R 5 , R 5 ', U, and U' are defined as in claim 2;
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ' , R 5 , R 5 ', U, and U' are defined as in claim 2; and L links position R 4 with R 4 '; or a pharmaceutically acceptable salt thereof; and a compound of formula (VI): wherein
  • R 4 , R» ⁇ R 5 , Rs', U, and U' are defined as in claim 2;
  • L links position R 5 with R 5 '; or a pharmaceutically acceptable salt thereof.
  • the compounds of the invention may be a homodimer or heterodimer having monomelic units of general formula (XIII):
  • each R 3 is, independently, H; -CF 3 ; -C 2 F 5 ; -CH 2 -Z or any R 2 and R 3 together with the nitrogen form a C 3 -C 6 heteroaliphatic ring;
  • each Z is, independently, H; -OH; F; Cl; -CH 3 ; -CF 3 ; -CH 2 Cl; -CH 2 F; or CH 2 OH;
  • each R 4 is, independently, C
  • each Zi is, independently, -N(R 9 )-C(0)-C M o-a]kyl; -N(R 9 K(O)-(CH 2 ) L6 -C 3 .
  • each R 9 is, independently, H; -CH 3 ; -CF 3 ; -CH 2 OH; or -CH 2 Cl; each R 1 o and Rn is, independently, H; C 3 .
  • R 6 6 -C 3-7 -cycloalkyl; -C(O)- (CH 2 ) 0 -6-phenyI; -(CH 2 )o-6-C(0)-phenyl; -(CH 2 ) 1-6 -het; -C(O)-(CH 2 ) w-het; or R 5 an amino acid residue, wherein the alkyl, cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; each R 63 is, independently, H, methyl, ethyl.
  • each R a and Rg is independently, cis relative to acyl substituents at the one position of the ring and are each, independently, H, -C
  • R 12 or -S(O) 2 -NR 12 R 13 ; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; each R 12 and R 13 is, independently, H; -(CH 2 ) 0-6 -C 3 . 7 -cycloaikyl; -(CH 2 ) 0-6 - (CH)o-r(aryl)i.
  • each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; each n is 0, 1, or 2; and wherein substituted alkyl is substituted by one or more substituents selected from a double bond, halogen, OH 5 -O-C , -6 -alkyl, -S-d. 6 -alkyl, and -CF 3 ; substituted cycloalkyl is substituted by one or more substituents selected from a double bond, C 1 - 6 -alkyl, halogen, OH.-O-Ci ⁇ alkyl, -S ⁇ C,.
  • substituted phenyl or aryl is substituted by one or more substituents selected from halogen, hydroxy, C M -alkyl, C ⁇ -alkoxy, nitro, -CN, -0-C(O)-C M -alkyl, and --C(O)-O-C M - aryl; or a pharmaceutically acceptable salt or hydrate thereof.
  • the compounds of the invention may have the formula (XIV):
  • R 1 and Rf are each H;
  • R 2 and R 2 ' are each, independently, H or C]-C 4 -alkyl which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH 3 , -SCH 3 , -CN, - SCN, and nitro;
  • R 3 and R 3 ' are each, independently, H; -CF 3 ; -CiF 5 ; or -CH 2 -Z or R 2 and R 3 or R 2 ' and R 3 1 together with a nitrogen form a C 3 -C t , heteroaliphatic ring;
  • each Z is, independently, H; -OH; F; Cl; -CH 3 ; -CF 3 ; -CH 2 Cl; -CH 2 F; or -CH 2 OH;
  • I 1 Li and R 4 ' are each, independently, C
  • X and X' are each, independently, CH or N;
  • Rs and R 5 ' are each, independently, H; Ci.jo-alkyl; aryl; phenyl; C 3-7 -cycloalkyl; -(CH 2 ) 1 - 6 -C 3- 7-cycloalkyl; -C
  • R 5 and R& a are, independently, or together with R 5 ' and R ⁇ a ' together with a nitrogen are het;
  • R a , Rg, R a ', and R K ' are cis relative to an acyl substituent at position one of a ring to which they are attached and are each, independently, H, -C,.
  • L is one or more linkers covalently linking one or more of the positions R 4 , R 5 , R$, with a pharmaceutically acceptable salt or hydrate thereof.
  • L may covalently link two identical monomeric units or L covalently links two non-identical monomeric units.
  • L may be selected from alkylene, alkenylene, alkynylene, cycloalkylene, cycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, and heterocycloalkylene, heterocycloalkylalkylene, heteroaryl and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, optionally-substituted alkylene, alkenylene, alkynylene cycloalkylene, cycloalkylalkylene, heterocycloalkylene, heterocycioalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or
  • the compounds of the invention may have a formula selected from a compound of formula (XV): wherein
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ', R 5 , R 5 ', R 6a , R 6a ', R a , R a ', R 8 , R 8 ', X, X', n, and n' are defined as in claim 8;
  • R 4 ' , R 5 , R 5 ', R 6a , R 6a ', R a , R a ,', R 8 , R 8 ', X, X', n, and n' are defined as in claim 8;
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ', R 5 , R 5 ', R 6a , R 6a ' , R a , R a ', R 8 , R 8 ', X, X', n, and n' are defined as in claim 8;
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ', R 5 , R 5 ', R 6a , R 6a ' , R a , R a ', R 8 , R 8 ', X- X', n, and n' are defined as in claim 8;
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ', R 5 , R 5 ', R 6a , R 6a ' , R a , R a ', R 8 , R 8 ', X, X', n, and n' are defined as in claim 8;
  • L links position R 8 with R 8 '; or a pharmaceutically acceptable salt or hydrate thereof.
  • the compounds of the invention may have a formula selected from formula (VII):
  • Some embodiments of the invention include pharmaceutical compositions including a compound of formula (111):
  • R ⁇ and R t ' are each, independently, H; C 1 -C-i-alkyl; C
  • R 2 and R 2 ' are each, independently, H; Ci-C 4 -alkyI; Ci-C 4 -alkenyl; C j -C 4 -alkynyl; or C 3 - Cio-cycloalkyl which are unsubstituted or substituted;
  • R 3 and R 3 ' are each, independently, H; -CF 3 ; -C 2 H 5 ; C t -C 4 -alkyl; C 1 -C 4 -alkenyl; Ci-C 4 - alkynyl; or -CH 2 -Z or R 2 and R 3 together or independently with R 2 ' and R 3 ' form a heterocyclic ring; each Z is, independently, H
  • R 4 and R 4 ' are each, independently, Ci-Ci 6 straight or branched alkyl; C 1 -C t 6 -alkenyl; Ci- Ci 6 -alkynyl; or C 3 -Cio-cycloalkyl; -(CH 2 ) 1-6 -Z ⁇ ; -(C H 2 Wary I; or -(CH 2 )o ⁇ -het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each Z 1 is, independently, -N(R ⁇ o)-C(0)-C MO -alkyl; -N(R, 0 )-C(O)-(CH 2 ) 1-6 -C 3-7 - cycloalkyl; -N(R,o)-C(0) ⁇ (CH 2 ) 0 - 6 -phenyl ; -N(R, 0 )-C(O)-(CH 2 ) ⁇ -het
  • each het is, independently, a 5-7 member heterocyclic ring containing 1-4 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom;
  • each R 10 is, independently, H; -CH 3 ; -CF 3 ; -CH 2 OH; or -CH 2 Cl;
  • each Rn and R 12 is, independently, H; Ci- 4 -alkyl; C 3 - 7 -c
  • R5 and R5' are each, independently, H; C ⁇ io-alkyl; aryl; phenyl; C 3 -7-cycloalkyl; ⁇ (CH 2 ) ⁇ _ 6-C 3 . 7 -cycIoalkyl; -C,. l 0 -alkyl-aryl; -(CH 2 ) 0 - 6 -C 3-7 -cycloalkyI-(CH 2 )o- ⁇ -phenyl; -(CH 2 )( M - CHf(CH 2 ) M-phenyI] 2 ; indanyl; -C(O)-C 1 .
  • each An and Ar 2 is. independently, substituted or unsubstituted aryl or het; each R 1 and R 6 is, independently, H; -CVio-alkyi; C ⁇ o-alkylaryl; -OH; -O-C 1 .io-alkyl; - ⁇ CH 2 V6-C 3 -7-cycloaIkyl; -O-(CH 2 ) 0 - 6 -aryl; phenyl; aryl; phenyl-phenyl; -(CH 2 )i. 6 -het; -0-(CH 2 )i.
  • each D is, independently, -CO-; -C(0)-Ci.7-alkylene or arylene; -CF 2 -; -O- ; -S(O) 1 - where r is 0-2; 1,3-dioxalane; or d-7-alkyl-OH; where alkyl, alkylene, or arylene are unsubstituted or substituted with one or more halogens, OH, -O-C ⁇ -alkyl, -S-C
  • each R 43 , R 7 , R 8 , and R9 is, independently, H, -C
  • each alkyl, cycloalkyi, and aryl is unsubstituted or substituted; and any R ⁇ , R 7 , R 8 , and Rg optionally together form a ring system; each R
  • L is one or more linkers covalently linking one or more of the positions R 4 , R 5 , or U, with R 4 ', R 5 ', or U'; or a pharmaceutically acceptable salt or hydrate thereof; and a pharmaceutically acceptable excipient or carrier; or a compound of formula:
  • ' are each H;
  • R 2 and R 2 ' are each, independently, H or C 1 -C 4 -alkyI which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH 3 , -SCH 3 , -CN, - SCN, and nitro;
  • R 3 and R 3 ' are each, independently, H; -CF 3 ; -C 2 F 5 ; or -CH 2 -Z or R 2 and R 3 or R 2 ' and R 3 " together with a nitrogen form a C 3 -C 6 heteroaliphatic ring;
  • each Z is, independently, H; -OH; F; Cl; -CH 3 ; -CF 3 ; -CH 2 Cl; -CH 2 F; Or -CH 2 OH;
  • R 4 and R 4 ' are each, independently, C1-C 1 6 straight chain alkyl; C 3 -CiO branched chain alkyl; -(CH2) 0-6 -C 3 -C 7 -cycloalkyl; -(CH 2 ) 1-6 -Z 1 ; -(CH 2 ) 0-6 -phenyl; or -(CH 2 ) 0-6 -het; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; each Zi is, independently, -N(R 9 )-C(0)-C 1 -10 -alkyl; -N(R 9 )-C(O)-(CH 2 ) 1-6 -C 3-7 - cycloalkyl; -N(R 9 )-C(O)-(CH 2 ) 0-6 -phenyl; -N(R 9 )-C(OMCH 2 ),.
  • X and X' are each, independently, CH or N;
  • R5 and RjT are each, independently, 11; C].
  • R 5 is an amino acid residue, wherein each alkyl, cycioalky], phenyl, and aryl substituent is unsubstituted or substituted;
  • R 6a and K 6 ⁇ are each, independently, H, methyl, ethyl, -CF 3 , -CH 2 OH ,or -CH 2 Cl; or
  • R 5 and R ⁇ are, independently, or together with R 5 ' and R & ,' together with a nitrogen are het;
  • R a , Rg, R a ', and R ⁇ ' are cis relative to an acyl substituent at position one of a ring to which they are attached and are each, independently, H, -C 1 -io-alkyl; -OH; -O-C 1 -io-alkyl; - (CH 2 )o-6-C 3 .7-cycloalkyl; -O-(CH 2 ) 0 -6-aryl; phenyl; -(CH 2 ), ⁇ -het; -O-(CH 2 ) ⁇ - 6 -het; -N(R 12 )(R 13 ); - S-R, 2 ; -S(O)-R 12 ; -S(O) 2 -R 12 ; or -S(O) 2 -NR 12 R 13 ; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted;
  • R 12 and R 13 are each, independently, H; C M o-alkyl; -(CH 2 ) 0-6 -C 3-7 -cycloalkyl; -(CH 2 ) 0- ⁇ - (CH)o-r(aryI),.
  • each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; or R 12 and R) 3 together with a nitrogen atom form het; each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; n and n * are each, independently, O, ] , or 2; wherein each substituted alkyl is substituted by one or more substituents selected from a double bond, halogen, OH.-O-C ⁇ -alkyl, -S-C ⁇ -alkyl.
  • each substituted cycloalkyl is substituted by one or more substituents selected from a double bond, C )-6 -alkyl, halogen, OH,-O-C [-6 -alkyl, -S-C 1-6 -alkyl, and -CF 3 ; and each substituted phenyl or aryl is substituted by one or more substituents selected from halogen, hydroxy, C M -aikyl, C M -alkoxy, nitro, -CN, -0-C(O)-C M -alkyl, and -C(O)-O-C M - aryl; and
  • L is one or more linkers covalently linking one or more of the positions R 4 , R 5 , R ⁇ , with R 4 ', R 5 ⁇ R 8 ';or a pharmaceutically acceptable salt or hydrate thereof; and a pharmaceutically acceptable excipient or carrier.
  • L and L' are selected from alkyiene, alkenylene, alkynylene, cycloalkylene, cycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heterocycloalkylene, heterocycloalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, optionally-substituted alkylene, alkenylene, alkynylene cycloalkylene, cycloalkylalkylene, heterocycloalkylene, heterocycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, amino, substituted amino, oxygen atom, sulfide, sulfoxide, sulfone
  • the pharmaceutical compositions of the invention may include a second therapeutic agent, and in other embodiments, the compounds of the invention may provide therapy to an individual.
  • the pharmaceutical composition of the invention may further include a second therapeutic agent is selected from a chemotherapeutic agent, radiation, and a combination thereof, and in certain embodiemtns, the chemotherapeutic may be selected from an alkylating agent, a plant alkaloid, an antitumor antibiotic, an antimetabolite, a topoisomerase inhibitor, and a combination thereof.
  • the term "about” means plus or minus 10% of the numerical value of the number with which it is being used. Therefore, about 50% means in the range of 45%-55%.
  • peptide mimetic and “peptidomimetic” are used interchangeably herein, and generally refer to a peptide, partial peptide or non-peptide molecule that mimics the tertiary binding structure or activity of a selected native peptide or protein functional domain (e.g., binding motif or active site).
  • peptide mimetics include recombinantly or chemically produced peptides, recombinantly or chemically modified peptides, as well as non-peptide agents, such as small molecule drug mimetics, as further described below.
  • compositions, carriers, diluents and reagents are used interchangeably and represent that the materials are capable of administration upon a mammal without the production of undesirable physiological effects such as nausea, dizziness, rash, or gastric upset.
  • Providing when used in conjunction with a therapeutic means to administer a therapeutic directly into or onto a target tissue, or to administer a therapeutic to a patient whereby the therapeutic positively impacts the tissue to which it is targeted.
  • subject refers to an animal or mammal including, but not limited to, a human, dog, cat, horse, cow, pig, sheep, goat, chicken, monkey, rabbit, rat, or mouse, etc.
  • the term "therapeutic” means an agent utilized to treat, combat, ameliorate, prevent or improve an unwanted condition or disease of a patient.
  • Embodiments of the present invention are directed to promote apoptosis and, thus, cell death.
  • a therapeutically effective amount of a therapeutic compound is a predetermined amount calculated to achieve the desired effect, i.e., to effectiveiy promote apoptosis, preferably by eliminating IAP inhibition of apoptosis, more preferably by inhibiting an IAP binding to a caspase.
  • mimetic or “peptidomimetics” are interchangeable and refer to synthetic compounds having a three-dimensional structure (i.e. a "core peptide motif') based upon the three-dimensional structure of a selected peptide.
  • the peptide motif provides the mimetic compound with the desired biological activity, i.e., binding to IAP, wherein the binding activity of the mimetic compound is not substantially reduced, and is often the same as or greater than the binding affinity of the native peptide on which the mimetic is modeled.
  • portions of compounds based on peptides can be non-peptide like.
  • Peptidomimetic compounds can have additional characteristics that enhance their therapeutic application, such as increased cell permeability, greater affinity and/or avidity, and prolonged biological half-life.
  • Alkyl or “alkylene” unless otherwise specified, means a branched or unbranched, saturated aliphatic hydrocarbon group, having up to 12 carbon atoms. When used as part of another term, for example, “alkylamino,” the alkyl portion may be a saturated hydrocarbon chain.
  • alkyl groups include methyl, ethyl, n-propyl, isopropyl, n- butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, 2-methyibutyl, 2,2-dimethylpropyl, n-hexyi, 2- methylpentyl, 2,2-dimethylbutyl, n-heptyl, 3-heptyl, 2-methyIhexyf, and the like.
  • lower alkyl Ci-C 4 alkyl
  • alkyl of 1 to 4 carbon atoms are synonymous and used interchangeably to mean methyl, ethyl, 1 -propyl, isopropyl, cyclopropyi, 1 -butyl, sec-butyl or t- butyl.
  • substituted alkyl groups may contain one, two, three or four substituents which may be the same or different.
  • Substituenf * or substituted nucleic acid refer to a molecular group that replaces a hydrogen at any methyl group on a hydrocarbon.
  • Substituents include, for example, halo, pseudohaio, hydroxy, protected hydroxy, trityloxy, carboxy, carbonyl.
  • cyano nitro, acyl, acyloxy, acetyl, acetoxy, carbamoyl, carbamoyloxy, ally], allyloxy, oxo, thia, nitrile, formyl, mercapto, hydroxycarbonyi, hydroxycarbonylalkyl, alkyl, haloalkyl, polyhaloalkyl, aminoalkyl, diaminoalkyl, alkenyl containing 1 to 2 double bonds, alkynyi containing 1 to 2 triple bonds, cycloalkyl, cycloalkylalkyl, cycloalkyloxy, heterocyclyl, heterocyclylalkyl, aryl, aryloxy, arylalkyi, aralkenyl, aralkynyl, heteroaryl, heteroaryloxy, heteroarylalkyi, trialkylsilyl, diaikylarylsilyl, alkyldiarylsilyi,
  • substituted alkyi are substituted methyls, e.g., a methyl group substituted by the same substituents as the "substituted C n -C m alky! group.
  • “Substituted alkyi” may include aikyloxymethyl, such as, methoxymethyl, ethoxymethyl, and t-butoxymethyl; halomethyi, such as, chloromethyl, br ⁇ momethyl, iodomethy], and trifluoromethyl; hydroxymethyl; protected hydroxymethyl, such as, tetrahydropyranyloxymethyl; trityloxymethyl; cyanomethyl; nitromethyl; aminomethyl; carboxymethyl; alkyloxycarbonylmethyl; acetoxymethyl, carbamoyloxymethyl; allyloxycarbonylaminomethyl; propionyloxymethyl; acetoxymethyl; 6-hydroxyhexyl; 2,4-dichloro(n-butyl); 2-amino(is
  • Unsaturated hydrocarbons may have up to 12 carbon atoms and may be substituted by one or more of any of the substituents described hereinabove.
  • alkenylamino and "aikynylamino” the alky! portion may be an unsaturated hydrocarbon chain.
  • Amino denotes primary (i.e. -NH 2 ), secondary (i.e. -NRH), and tertiary (i.e. - NRR) amines.
  • Particular secondary and tertiary amines include, but are not limited to, alkylamine, dialkylamine, arylamine, diarylamine, arylalkylamine and diarylalkylamine including, for example, methylamine, ethylamine, propylamine, isopropylamine, phenylamine, benzylamine, dimethylamine, diethylamine, dipropylamine and disopropylamine.
  • Aryl when used alone or as part of another term, means a fused or unfused carbocyclic aromatic group having a designated number of carbon atoms, or if no number is designated, up to 14 carbon atoms.
  • Particular aryl groups include phenyl, naphthyl, biphenyl, phenanthrenyl, naphthacenyl, and the like (see, Lang's Handbook of Chemistry 13 th ed. (Dean, J. A., ed.) Table 7-2 [ 1985]).
  • a "fused ring system ' ' refers to two or more substituted or unsubstituted carbocyclic or carbocyclic aromatic groups that are fused together.
  • Substituted phenyl or substituted aryl denotes a phenyl or aryl group substituted with one, two, three, four or five substituents chosen from those described above, for example, halogen (F. Cl, Br, I), hydroxy, protected hydroxy, cyano, nitro, alkyi (such as CrC 6 alkyl), alkoxy (such as, Q- C(, alkoxy), benzyloxy, carboxy, protected carboxy, carboxymethyl, protected carboxy methyl, hydroxymethyl.
  • halogen F. Cl, Br, I
  • hydroxy protected hydroxy
  • cyano nitro
  • alkyi such as CrC 6 alkyl
  • alkoxy such as, Q- C(, alkoxy
  • benzyloxy carboxy, protected carboxy, carboxymethyl, protected carboxy methyl, hydroxymethyl.
  • substituted phenyls include, but are not limited to, mono- or di-halo-phenyl, such as, 2-chlorophenyl, 2-bromophenyi, 4-chlorophenyl, 2,6-dichlorophenyl, 2,5-dichlorophenyl, 3,4-dichlorophenyl, 3-chlorophenyI, 3-bromophenyl, 4- bromophenyl, 3,4-dibromophenyl, 3-chloro-4-fluorophenyl, 2-fluoropheny!, and the like; mono- or di-hydroxyphenyl, such as, 4-hydroxyphenyl, 3-hydroxyphenyl, 2,4-dihydroxyphenyl, protected-hydroxy derivatives thereof, and the like; nitrophenyl, such as, 3- or 4-nitrophenyl; cyanophenyl, for example, 4-cyanophenyl; mono- or di-lower alkyl
  • substituted phenyl may represent di-substituted phenyl groups where the substituents are different, such as, for example, 3-methyl-4-hydroxyphenyl, 3-chloro-4-hydroxyphenyl, 2- methoxy-4-bromopheny ⁇ , 4-ethyl-2-hydroxyphenyL 3-hydroxy-4-nitrophenyl, 2-hydroxy-4- chlorophenyl, and the like, as well as tri-substituted phenyl groups where the substituents are different, such as, for example, 3-methoxy-4-benzyloxy- ⁇ -methyl sulfonylamino, 3-methoxy-4- benzyloxy-6-phenyi sulfonylamino and the like and tetra-substituted phenyl groups where the substituents are different, such as, for example, 3-methoxy ⁇ 4-benzyloxy-5-methyl-6-phenyl sulfonylarnino.
  • Particular substituted phenyl groups include 2-ch3orophenyl, 2-aminophenyl, 2- bromophenyl, 3-methoxyphenyl, 3-ethoxy-phenyl, 4-benzyloxyphenyl, 4-methoxyphenyl, 3- ethoxy-4-benzyloxyphenyl, 3,4-diethoxyphenyI, 3-methoxy-4-benzyIoxyphenyl, 3-methoxy-4- ( 1-chloromethyl) benzyloxy-phenyl, 3-methoxy-4-( l-chloromethyl), and benzyloxy-6-methyl sulfonyl aminophenyl groups.
  • Fused aryl rings may also be substituted with one or more of any of the substituents specified herein, for example, fused aryl groups may contain 1 , 2 or 3 substituents in the same manner as substituted alkyl groups.
  • Heterocyclic group “heterocyclic”, “heterocycle”, “heterocyclyl”, or “heterocyclo” alone, and when used as a moiety in a complex group such as a heterocycloalkyl group, are used interchangeably and refer to any mono-, bi-, or tri-cyclic, saturated or unsaturated, aromatic (heteroaryl) or non-aromatic ring having the number of atoms designated, generally from 5 to about 14 ring atoms, where the ring atoms are carbon and at least one heteroatom (nitrogen, sulfur or oxygen). In a particular embodiment, the group incorporates 1 to 4 heteroatoms.
  • a 5- member ring has 0 to 2 double bonds and a 6- or 7-member ring has 0 to 3 double bonds; and the nitrogen or sulfur heteroatoms may optionally be oxidized (e.g. SO, SO 2 ), and any nitrogen heteroatom may optionally be quaternized.
  • non-aromatic heterocycles include morpholinyl (morpholino), pyrrolidinyl, oxiranyl, oxetanyl, tetrahydrofuranyl, 2,3-dihydrofuranyl, 2H-pyranyl, tetrahydropyranyl, th ⁇ ranyl, thietanyl, tetrahydrothietanyl, aziridinyl, azetidinyl, l-methyl-2-pyrrolyl, piperazinyl, and piperidinyl.
  • a "heterocycloalkyl” group is a heterocycle group as defined above, covalently bonded to an alkyl group as defined above.
  • Particular 5-membered heterocycles containing a sulfur or oxygen atom and one to three nitrogen atoms include thiazolyl, such as thiazol-2-yl and thiazol-2-yl N-oxide, thiadiazolyl, such as 1 ,3,4-thiadiazol-5-yl and l,2,4-thiadiazol-5-yl, oxazolyl, such as, oxazol-2- yl, and oxadiazolyK such as l,3,4-oxadiazol-5-yl, and l,2,4-oxadiazol-5-yl.
  • thiazolyl such as thiazol-2-yl and thiazol-2-yl N-oxide
  • thiadiazolyl such as 1 ,3,4-thiadiazol-5-yl and l,2,4-thiadiazol-5-yl
  • oxazolyl such as, oxazol-2- yl
  • oxadiazolyK such as
  • Particular 5- membered ring heterocycles containing 2 to 4 nitrogen atoms include imidazolyl, such as imidazol-2-yl; triazolyi, such as I ,3,4-triazol-5-yl, l,2,3-triazol ⁇ 5-yl, and 1 ,2,4-triazol-5-yl, and tetrazolyl such as lH-tetrazol-5-y ⁇ .
  • Particular benzo-fused 5-membered heterocycles are benzoxazol-2-yl, benzthiazol-2-yl, and benzimidazoi-2-yI.
  • Particular 6-membered heterocycles contain one to three nitrogen atoms and, optionally, a sulfur or oxygen atom, for example pyridyl, such as, pyrid-2-yl, pyrid-3-yl, and pyrid-4-yI; pyrimidyl, such as, pyrimid-2-yl and pyrimid-4-yl; triazinyl, such as, l,3,4-triazin-2-yl and l,3,5-triazin-4-yl; pyridazinyl, such as. pyridazin-3-yl, and pyrazinyl.
  • Substituents for optionally substituted heterocycles, and further examples of the 5- and 6-membered ring systems discussed above, can be found in U. S. Patent No. 4,278, 793 to W. Druckheimer et al.
  • Heteroaryl alone and when used as a moiety in a complex group such as a heteroarylalkyl group, refers to any mono-, bi-, or tri-cyciic aromatic ring system having the number of atoms designated where at least one ring is a 5-, 6- or 7-membered ring containing from one to four heteroatoms selected from the group nitrogen, oxygen, and sulfur (see Lang's Handbook of Chemistry, supra). Included in the definition are any bicyclic groups where any of the above heteroaryl rings are fused to a benzene ring.
  • heteroaryl whether substituted or unsubstituted group denoted by the term "heteroaryl”: thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, tetrazolyi, thiatriazolyl, oxatriazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, thiazinyl, oxazinyl, triazinyl, thiadiazinyl, oxadiazinyl, dithiazinyl, dioxazinyl, oxathiazinyl, tetrazinyl, thiatriazinyl, oxatriazinyl, dithiadiaziny
  • heteroaryls include: 1 ,3-thiazol-2-yl, 4-(carboxymethyl)-5-methyl, l ,3-thiazoI-2-yl, 4-(carboxymethyl)-5- methyl-l ,3-thiazol-2-yl sodium salt, l,2,4-thiadiazol-5-yl, 3-mefhyl-l ,2,4-thiadiazol-5-yl, 1 ,3,4- triazol-5-yl, 2-methyl- l,3,4-triazol-5-yl, 2-hydroxy-l,3,4-triazol-5-yl.
  • 2-carboxy-4-methyl- 1,3,4- triazol-5-yl sodium salt 2-carboxy-4-methyl-l,3,4-triazol-5-yl, l,3-oxazol-2-yI, 1 ,3,4-oxadiazol- 5-yl, 2-methyl-l ,3,4-oxadiazol-5-yl, 2-(hydroxymethyl)-l,3,4-oxadiazoI-5-yl, l,2,4-oxadiazol-5- yl, l,3,4-thiadiazoi-5-yl, 2-thiol- l,3,4-thiadiazol-5-yl, 2-(methylthio)-l,3,4-thiadiazol-5-yl, 2- amino-l ,3,4-thiadiazol-5-yl, lH-tetrazol-5-yl, 1 -methyl- l H-tetrazol-5-yl, 1-(1-
  • heteroaryl includes: 4-(carboxymethyI)-5-methyl-l, 3-thiazoi-2-yl, 4-(carboxymefhyl)-5- methyl-I ,3-thiazol-2-yl sodium salt, l ,3,4-triazol-5-yl, 2-methyl-l ,3,4-triazol-5-yl, I H-tetrazol- 5-yl, 1 -methyl- lH-tetrazol-5-yl, l-(l-(dimethy]amino)eth-2-yl)-lH-tetrazol-5-yi, 1- (carboxymethyl)-lH-tetrazol-5-yl, l-(carboxymethyl)-lH-tetrazol-5-yl sodium salt, 1- (methylsulfonic acid)-lH- tetrazol
  • a "linker” is a bond or linking group whereby two chemical moieties, such as, monomers of an active compound, are directly covalently linked to one another or are indirectly linked via a third chemical moiety to form a homo- or heterodimer.
  • the compounds set forth herein may include a single linker linking the two chemical moieties, or more than one linker linking the two chemical moieties at one or more position independently on each of the two chemical moieties.
  • a “linker” (L, Lj or L 2 ) may be a single or double covalent bond or a branched or unbranched, substituted or unsubstituted, hydrocarbon chain of 1 to about 100 atoms, typically, 1 to about 20 atoms, having a molecular weight up to about 500 MW.
  • a linker can be a bond, alkylene, alkenylene, alkynylene, cycloalkylene, cycloalkylalkyiene, heterocycloalkylene, heterocycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, or heteroarylalkylene, or an optionally-substituted alkylene, alkenylene, alkynyiene cycloalkylene, cycloalkylalkyiene, heterocycloalkylene, heterocycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, or heteroarylalkylene, of 2 to 12 atoms where one or more carbon atoms can be replaced with N, O, or S or an amino, substituted amino, oxygen atom, sulfide (-S-), sulfoxide (-SO-), sulfone (-SO 2 -),
  • linkers and linking groups are described in U.S. Patent Publication No. 20050197403, as well as in U.S. Patent Application Serial Number 1 1/363,387, filed February 27, 2006, both of which are incorporated herein by reference as though fully set forth.
  • particular "linkers” include, but are not limited to, -CH 2 CH 2 -, -CH 2 CHiCH 2 -, - CH-CH-, 1 ,4-phcnyl, 2,5-thiopheny], -CH(OH)CH(OH)-, -CH 2 CH-O-CHCH 2 -, and - CH 2 OCOCCH 2 -.
  • homodimer refers to a compound composed of two covalently bound monomeric units of a chemical moiety wherein the monomelic units are identical.
  • heterodimer refers to a compound composed of two covalently bound monomeric units of a chemical moiety wherein the monomeric units are different.
  • one monomeric unit of a heterodimer may include a substituent that is different from the other monomeric unit at one or more position.
  • Inhibitor means a compound which reduces or prevents a particular interaction or reaction. For example, the binding of IAP proteins to caspase proteins reduces or prevents the inhibition of apoptosis by an IAP protein.
  • “Pharmaceutically acceptable salts” include both acid and base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid, and the like.
  • Organic acids may be selected from aliphatic, cycloaliphatic, aromatic, arylaliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids, such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, p-toiuenesulfonic acid, salicyclic acid, and the like.
  • organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid
  • the present invention is generally directed to Smac peptidomimetics (herein referred to as "'Srnac mimetics" or "a Smac mimetic”) and the uses of Smac mimetics.
  • One embodiment of the invention is a therapeutic composition including a Smac mimetic.
  • Smac mimetics act as chemopotentiating or chemotherapeutic agents.
  • chemopotentiating agent refers to an agent that acts to increase the sensitivity of an organism, tissue, or cell to a chemical compound, or treatment namely "chemotherapeutic agents” or “chemo drugs” or radiation treatment.
  • a further embodiment of the invention is the therapeutic composition of a Smac mimetic, which acts as a chemopotentiating agent, and a biological agent, chemotherapeutic agent or radiation.
  • Another embodiment of the invention is a method of inhibiting tumor growth in vivo by administering a Smac mimetic.
  • Yet another embodiment is a method of inhibiting tumor growth in vivo by administering a Smac mimetic and a biologic agent, chemotherapeutic agent or radiation.
  • Still another embodiment of the invention is a method of treating an individual, such as, for example, patient with cancer, by administering Smac mimetics of the present invention alone, or in combination with, a biological agent, chemotherapeutic agent or radiation.
  • in situ cells or pathogenic cells in an individual, may be treated with a Smac mimetic or a Smac mimietic in combination with a secondary agent, such as, a biological agent, chemotherapeutic agent or radiation,
  • the contacting step is affected by administering a pharmaceutical composition including a therapeutically effective amount of the Smac mimetic, wherein the individual may be subject to concurrent or antecedent radiation or chemotherapy for treatment of a neoproliferative pathology.
  • Pathogenic cells may be of a tumor such as, but not limited to, bladder cancer, breast cancer, prostate cancer, lung cancer, pancreatic cancer, gastric cancer, colon cancer, ovarian cancer, renal cancer, hepatoma, melanoma, lymphoma, sarcoma, and combinations thereof.
  • autoimmune diseases are characterized in that the cells of the immune system produce antibodies against its own organs and molecules or directly attack tissues resulting in the destruction of these tissues. Failure of these self-reactive cells to undergo apoptosis leads to the manifestation of the disease. Defects in apoptosis regulation have been identified in autoimmune diseases such as systemic lupus erthematosus, or rheumatoid arthritis,
  • pathogenic cells may be those cells effected by an autoimmune disease or any disease whose symptoms include production of cells that are resistant to apoptosis.
  • affected cells are resistant to apoptosis due to the expression or overexpression of members of the Bcl-2 family of caspases.
  • autoimmune diseases include, but are not limited to, collagen diseases, such as, rheumatoid arthritis, systemic lupus erythematosus.
  • Sharp's syndrome CREST syndrome, calcinosis, Raynaud's syndrome, esophageal dysmotility, telangiectasia, dermatomyositis, vasculitis (Morbus Wegener's), and Sjogren's syndrome
  • renal diseases such as, Goodpasture's syndrome, rapidly-progressing glomerulonephritis, and membrano-proliferative glomerulonephritis type II
  • endocrine diseases such as, type-I diabetes, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), autoimmune parathyroidism, pernicious anemia, gonad insufficiency, idiopathic Morbus Addison's, hyperthyreosis, Hashimoto's thyroiditis, and primary myxedema
  • skin diseases such as, pemphigus vulgaris, bullous pemphigoid, herpes gestationis, epi
  • the IAP- binding peptides or mimetics are capable of potentiating apoptosis of cells.
  • the mimetics described herein are suitably small, and since structural features in relation to the IAP binding groove are well-characterized, a wide variety of mimetic compounds may be synthesized. Mimetics of the core IAP-binding portions are preferred. Added advantages of compounds of this size include improved solubility in aqueous solution and ease of delivery to selected targets in vivo.
  • IAP-binding compounds that may be prepared as dimers and dimers of these IAP-binding compounds.
  • dimers can be prepared using any synthetic technique available to persons of ordinary skill in the art, such as, for example, the dimeric Smac peptidomimetics disclosed in U.S. Patent Application Serial Number 1 1/363,387, filed 2/27/2006, which provides guidance on preparation of the dimers of the instant invention.
  • Various embodiments of the invention also include homodimers and heterodimer of monomeric units of general formula (1):
  • -C 4 -alkenyl; Ci-d-alkynyl; - CH 2 -Z or any R 2 and R 3 together form a heterocyclic ring; each Z is, independently, II; -OH; F; Cl; -CH 3 ; -CF 3 ; -CH 2 Cl; -CH 2 F Or -CH 2
  • each Zi is, independently, -N(R 10 )-C(0)-C
  • each het is, independently, a 5-7 member heterocyclic ring containing 1-4 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom;
  • each R 10 is, independently, H; -CH 3 ; -CF 3 ; -CH 2 OH; or -CH 2 Cl;
  • each R n and R 12 is, independently, H; Cu-alkyl; C 3 .
  • each R 5 is, independently, H; C
  • R 5 is a residue of an amino acid, wherein the alkyl, cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; each U is, independently, as shown in structure (II):
  • each D is, independently, -CO-; -C(O)-C ]_ 7 -alkylene or arylene; -CF 2 -; -0-; -S(O) r where r is 0-2; 1 ,3-dioxalane; or C
  • each D is, independently.
  • substituted phenyl or aryl of R i3 and Ru are substituted by one or more substituents selected from halogen, hydroxyl, C ⁇ -alkyl, C ⁇ - 4 -alkoxy, nitro, -CN, -0-C(O)-C M -alkyl, and -C(O)-O-C M -aryI; and pharmaceutically acceptable salts and hydrates thereof.
  • compounds of the invention are of general formula (III): wherein:
  • R 1 and Rf are each, independently, H; C]-C 4 -alkyl; C]-C 4 -alkenyl; C
  • R 2 and R 2 " are each, independently, H; C
  • R 3 and R 3 ' are each, independently, H; -CF 3 ; -C 2 H 5 ; C 3 -C 4 -a!kyl; C
  • R 4 and R 4 ' are each, independently, C1-C 16 straight or branched alkyl; CrC 1 ⁇ -alkenyl; Q- Ci6-alkynyl; or C 3 -C ⁇ o-cycloalkyl; ; -(CH 2 ) 0-6 -aryl; or - ⁇ CH 2 ) 0-6 -het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each Z 1 is, independently, -N(R !0 )-C(O)-C M0 -alkyl; -N(R S o)-C(O)-(CH 2 ) !
  • each het is, independently, a 5-7 member heterocyclic ring containing 1-4 heteroatoms selected from N, O, and S. or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S.
  • each R 1 o is, independently, H; -CH 3 ; -CF 3 ; -CH 2 OH; or -ClI 2 Cl; each Rj 1 and Rj 2 is, independently, H; C ⁇ -alkyl; C 3 _7-cycloalkyl; -(CH 2 ) s- 6 -C 3 .
  • R 5 and IV are each, independently, H; C
  • each D is, independently, N(Rh) wherein each Rh is, independently, H; unsubstituted or substituted C1.7-alk.yl; aryl; unsubstituted or substituted -O-(C
  • each R 6 , R7, Rg, and R9 is, independently, H, -C 1 .io-alkyl; -C
  • L is one or more linkers covalently linking one or more of the positions R 4 , R 5 , or U, with R 4 1 , R 5 ', or U'; and pharmaceutically acceptable salts and hydrates thereof.
  • the compounds of the invention are of a formula selected from compounds of formula (IV): wherein
  • R 4 , Rt', R5, R5', U, and U' are defined as described above;
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 - R 4 ', R5, R 5 ', U, and U' are defined as described above; and L links position R 4 with R 4 '; or pharmaceutically acceptable salts thereof; and a compound of formula (VI): wherein
  • Rs, Rs', V, and U " are defined as described above; and L links position R 5 with R 5 '; or pharmaceutically acceptable salts thereof.
  • each alkyl. cycloalkyl, and phenyl are unsubstituted or substituted; each Z, is, independently, -N(R 9 )-C(O)-C
  • R 12 or -S(O) 2 -NRJ 2 R 13 ; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; each R
  • 6 -alky s halogen, OH 7 -O-C , ⁇ -alky I, -S-C
  • Still other embodiments of the invention include compounds of formula (XIV):
  • R 2 and R 2 ' are each, independently, H or C]-C 4 -alkyl which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH 3 , -SCH 3 , -CN, - SCN, and nitro;
  • R 3 and R 3 ' are each, independently, H; -CF 3 ; -C 2 F 5 ; or -CH 2 -Z or R 2 and R 3 or R 2 ' and R 3 ' together with a nitrogen form a C 3 -C 6 heteroaliphatic ring; each Z is, independently, H; -OH; F; Cl; -CH 3 ; -CF 3 ; -CH 2 Cl; -CH 2 F; or -CH 2 OH;
  • R 4 and RV are each, independently, Cf-C 16 straight chain alkyl; C 3 -Ci O branched chain alkyl; -(CHiJo- ⁇ -Cj-CT-cycloalkyl; -(CH 2 ) I -6 -Zi; -(CH 2 )o- ⁇ -phenyl; or -(CH 2 )o- ⁇ -het; wherein each alkyl, cycloaikyl, and phenyl is unsubstituted or substituted; each Z, is, independently, -N(R 9 )-C(O)-C M() -alkyl; -N(Rg)-C(O)-(CH 2 )S -6 -C 3 .
  • each alkyl, cycioalkyl, and phenyl is unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S.
  • R5 and R s " are each, independently, 11; d-io-alkyl; aryl; phenyl; C 3-7 -cycloalkyl; -(CH 2 ) I - 6-C 3-7 -cycloalkyl; -C).
  • R 5 is an amino acid residue, wherein each alkyl, cycioalkyl, phenyl, and aryl substituent is unsubstituted or substituted;
  • R 63 and R 63 ' are each, independently, H, methyl, ethyl, -CF 3 , -CH 2 OH ,or -CH 2 Cl; or R 5 and R ⁇ are, independently, or together with R 5 ' and R 63 ' together with a nitrogen are het;
  • R 1 J, Rg, R a ⁇ and R «' are cis relative to an acyl substituent at position one of a ring to which they are attached and are each, independently, H, -C M o-alkyl; -OH; -O-Ci.
  • Rj 2 and R !3 are each, independently, H; CYio-alkyl; -(CH 2 )o ⁇ -C 3 . 7 -cycioaikyl; -(CH 2 )o- ⁇ - (CH)o-r(aryl) ⁇ . 2 ; -C(O)-C M0 -alkyl; -C(O)-(CH 2 ) N6 -C 3-7 -cycloalkyl; -C(O)-O-(CH 2 ) 0 ⁇ -aryl; - C(O)-(CH 2 V ⁇ -O-fluorenyl; -C(O)-NH-(CH 2 ) 0 .
  • each alky!, cycioalky], and aryl is unsubstituted or substituted; or R
  • L is one or more linkers covalently linking one or more of the positions R 4 , R 5 , R 8 , with R 4 ', R 5 ', R 8 '; and pharmaceutically acceptable salts and hydrates thereof.
  • the compounds of compounds of the invention may be selected from compounds of formula (XV):
  • R 1 , R 1 ' , R 2 , R 2 ' , R 3 , R 3 ', R 4 , R 4 ', R 5 , R 5 " , R* a , R 6a ', R 8 , R a ', R 8 , R 8 ', X, X', n, and n' are defined as described above;
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ', R 5 , R 5 ', R 6a , R 6a ', R a , R a ', R 8 , R 8 ', X, X', n. and n' are defined as described above;
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ', R 5 , R 5 ', R 6a , R 6a ', R a , R a ', R 8 , R 8 ', X, X', n, and n' are defined as described above;
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ', R 5 , R 5 ', R 6a , R 6a ' , R a , R a ', R 8 , R 8 ', X, X', n, and n' are defined as described above;
  • R 1 , R 1 ', R 2 , R 2 ', R 3 , R 3 ', R 4 , R 4 ', R 5 , R 5 ', R 6a , R 6a ' , R a , R a ', R 8 , R 8 ', X, X', n, and n' are defined as described above;
  • L links position R 8 with R 8 '; or pharmaceutically acceptable salts and hydrates thereof.
  • Still other embodiments of the invention include compouds of formulae (VIl), (VlIl). (IX), (X), (Xl) and (XII):
  • Mimetic, specifically, peptidomimetic design strategies are readily available in the art and can be easily adapted for use in the present invention (see, e.g., Ripka & Rich, Curr. Op. Chem. Biol. 2, 441-452, 1998; Hruby et al., Curr. Op. Chem. Biol. 1 , 1 14-1 19, 1997; Hruby & Balse, Curr. Med, Chem. 9, 945-970, 2000).
  • One class of mimetic mimics a backbone that is partially or completely non-peptide, but mimics the peptide backbone atom-for-atom and comprises side groups that likewise mimic the functionality of the side groups of the native amino acid residues.
  • peptidomimetics Several types of chemical bonds, e.g. ester, thioester, thioamide, retroamide, reduced carbonyl, dimethylene, and ketomethylene bonds, are known in the art to be generally useful substitutes for peptide bonds in the construction of protease-resistant peptidomimetics.
  • Another class of peptidomimetics comprises a small non-peptide molecule that binds to another peptide or protein, but which is not necessarily a structural mimetic of the native peptide.
  • Yet another class of peptidomimetics has arisen from combinatorial chemistry and the generation of massive chemical libraries.
  • the Smac mimetics of the invention are modified to produce peptide mimetics by replacement of one or more naturally occurring side chains of the 20 genetically encoded amino acids, or D-amino acids, with other side chains, for instance with groups, such as, alkyl, lower alkyl, cyclic 4-, 5-, 6-, to 7-membered alkyl, amide, amide lower alkyl, amide di-(lower alkyl), lower alkoxy, hydroxy, carboxy, and the lower ester derivatives thereof, and with 4-, 5-, 6-, to 7-membered heterocycles.
  • proline analogs can be made in which the ring size of the proline residue is changed from 5 members to 4, 6, or 7 members.
  • Cyclic groups can be saturated or unsaturated, and if unsaturated, can be aromatic or non-aromatic. Heterocyclic groups can contain one or more nitrogen, oxygen, and/or sulphur heteroatoms. Examples of such groups include furazanyl, imidazolidinyl, imidazolyl, imidazolinyl, isothiazolyl, isoxazolyl, morpholinyl (e.g. morpholino), oxazolyl, piperazinyl (e.g. 1 -piperazinyl), piperidyl (e.g.
  • These heterocyclic groups can be substituted or unsubstituted.
  • the substituent can be alkyi, alkoxy, halogen, oxygen, or substituted or unsubstituted phenyl.
  • Peptidomimetics may also have amino acid residues that have been chemically modified by phosphorylation, sulfonation, biotinylation, or the addition or removal of other moieties.
  • compositions encompass pharmaceutical compositions including a therapeutically effective amount of a Smac mimetic in dosage form and a pharmaceutically acceptable carrier, wherein the Smac mimetic inhibits the activity of an Inhibitor of Apoptosis protein (1 ⁇ P), thus promoting apoptosis.
  • the compositions include a therapeutically effective amount of a Smac mimetic in dosage form and a pharmaceutically acceptable carrier in combination with a chemotherapeutic and/or radiotherapy, wherein the Smac mimetic inhibits the activity of an IAP, thus promoting apoptosis and enhancing the effectiveness of the chemotherapeutic and/or radiotherapy.
  • a therapeutic composition for promoting apoptosis can be a therapeutically effective amount of a Smac mimetic which binds to at least one IAP.
  • the IAP can be XIAP, In another embodiment, the IAP can be ML-IAP, and in yet another embodiment, the IAP can be cIAP-1 or cIAP-2. fn further embodiments, the IAP can be multiple IAPs.
  • Embodiments of the invention also include methods for treating a patient having a condition characterized by inhibited apoptosis, wherein administration of a therapeutically effective amount of a Smac mimetic is delivered to the patient, and the Smac mimetic binds to at least one IAP,
  • the IAP can be XIAP.
  • the IAP can be ML-IAP, and in still another embodiment, the IAP can be cIAP-1 or cIAP-2.
  • the IAP can be multiple IAPs.
  • an additional chemotherapeutic agent (infra) or radiation may be administered prior to, along with, or following administration of the Smac mimetic.
  • chemotherapeutic agent include, but are not limited to, alkylating agents, antimetabolites, anti-tumor antibiotics, taxanes, hormonal agents, monoclonal antibodies, glucocorticoids, mitotic inhibitors, topoisomerase I inhibitors, topoisomerase II inhibitors, immunomodulating agents, cellular growth factors, cytokines, and nonsteroidal antiinflammatory compounds.
  • the Smac mimetics of the invention may be combined with a pharmaceutically acceptable carrier or excipient, and in some embodiments, the Smac mimetics of the invention may be combined with an additional chemotherapeutic agent and a pharmaceutically acceptable carrier or excipient.
  • pharmaceutically-acceptable carrier means one or more compatible solid or liquid fillers, diluents or encapsulating substances which are suitable for administration into a human.
  • carrier or “excipient” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • the components of the pharmaceutical compositions are also capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
  • the delivery systems of the invention are designed to include time-released, delayed release or sustained release delivery systems such that the delivering of the Smac mimetic occurs prior to, and with sufficient time, to cause sensitization of the site to be treated.
  • a Smac mimetic may be used in conjunction with radiation and/or additional anti-cancer chemical agents (infra). Such systems can avoid repeated administrations of the Smac mimetic compound, increasing convenience to the subject and the physician, and may be particularly suitable for certain compositions of the present invention.
  • release delivery systems include, but are not limited to, polymer base systems, such as, poly(lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides.
  • polymer base systems such as, poly(lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides.
  • Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Pat. No. 5,075,109.
  • Delivery systems also include non-polymer systems including: lipids including sterols, such as cholesterol, cholesterol esters and fatty acids or neutral fats, such as mono-, di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like.
  • lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats, such as mono-, di- and tri-glycerides
  • hydrogel release systems such as lipids including sterols, such as cholesterol, cholesterol esters and fatty acids or neutral fats, such as mono-, di- and tri-glycerides
  • sylastic systems such as cholesterol, cholesterol esters and fatty acids or neutral fats, such as mono-, di- and tri-glycerides
  • peptide based systems such as fatty acids or neutral fats, such as mono-, di- and tri-glycerides
  • Long-term sustained release is used herein, and means that the implant is constructed and arranged to deliver therapeutic levels of the active ingredient for at least about 30 days, and preferably about 60 days.
  • Long-term sustained release implants are well-known to those of ordinary skill in the art and include some of the release systems described above.
  • compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier that constitutes one or more accessory ingredients. In general, the compositions may be prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • compositions suitable for parenteral administration conveniently include a sterile aqueous preparation of a Smac mimetic which is preferably isotonic with the blood of the recipient.
  • This aqueous preparation may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may aiso be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1 ,3-butanediol.
  • acceptable vehicles and sêts that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • fatty acids such as oleic acid, may be used in the preparation of injectables.
  • Carrier formulation suitable for oral, subcutaneous, intravenous, intramuscular, etc. administrations can be found, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, PA which is incorporated herein in its entirety by reference thereto.
  • Smac peptidomimetics of the invention may be administered in effective amounts.
  • An effective amount is that amount of a preparation that alone, or together with further doses, produces the desired response. This may involve only slowing the progression of the disease temporarily, although it may involve halting the progression of the disease permanently or delaying the onset of or preventing the disease or condition from occurring. This can be monitored by routine methods known and practiced in the art.
  • doses of active compounds may be from about 0.01 mg/kg per day to about 1000 mg/kg per day, and in some embodiments, the dosage may be from 50-500 mg/kg.
  • the compounds of the invention may be administered intravenously, intramuscularly, or intradermally, and in one or several administrations per day.
  • the administration of the Smac peptidomimetic can occur simultaneous with, subsequent to, or prior to chemotherapy or radiation.
  • a dosage regimen of the Smac mimetic to reduce tumor growth can be oral administration of from about 1 mg to about 2000 mg/day, preferably about 1 to about 1000 mg/day, more preferably about 50 to about 600 mg/day, in two to four divided doses. Intermittent therapy (e.g., one week out of three weeks or three out of four weeks) may also be used.
  • Embodiments of the invention also include a method of treating a patient with cancer or an autoimmune disease by promoting apoptosis wherein administration of a therapeutically effective amount of a Smac mimetic and the Smac mimetic binds to at least one IAP.
  • the IAP can be XIAP.
  • the IAP can be ML- IAP, and in still another embodiment, the IAP can be cIAP-1 or cIAP-2.
  • the IAP can be multiple IAPs.
  • the method may further include concurrent administration of a chemotherapeutic agent including, but not limited to, alkylating agents, antimetabolites, anti-tumor antibiotics, taxanes, hormonal agents, monoclonal antibodies, glucocorticoids, mitotic inhibitors, topoisomerase I inhibitors, topoisomerase II inhibitors, immunomodulating agents, cellular growth factors, cytokines, and nonsteroidal antiinflammatory compounds.
  • a chemotherapeutic agent including, but not limited to, alkylating agents, antimetabolites, anti-tumor antibiotics, taxanes, hormonal agents, monoclonal antibodies, glucocorticoids, mitotic inhibitors, topoisomerase I inhibitors, topoisomerase II inhibitors, immunomodulating agents, cellular growth factors, cytokines, and nonsteroidal antiinflammatory compounds.
  • a variety of administration routes are available. The particular mode selected will depend, of course, upon the particular chemotherapeutic drug selected, the severity of the condition being treated, and the dosage required for therapeutic efficacy.
  • the methods of the invention may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of active compounds without causing clinically unacceptable adverse effects.
  • modes of administration include, but are not limited to, oral, rectal, topical, nasal, intradermal, inhalation, intra-peritoneal, or parenteral routes.
  • parenteral includes subcutaneous, intravenous, intramuscular, or infusion. Intravenous or intramuscular routes are particularly suitable for purposes of the present invention.
  • a Smac mimetic as described herein, with or without additional biological or chemotherapeutic agents or radiotherapy does not adversely affect normal tissues, while sensitizing tumor cells to the additional chemotherapeutic/radiation protocols.
  • the composition or method may be designed to allow sensitization of the cell or tumor to the chemotherapeutic or radiation therapy by administering at least a portion of the Smac mimetic prior to chemotherapeutic or radiation therapy.
  • the radiation therapy, and/or inclusion of chemotherapeutic agents may be included as part of the therapeutic regimen to further potentiate the tumor cell killing by the Smac mimetic.
  • a combination of a Smac mimetic and a chemotherapeutic/anti-neoplastic agent and/or radiation therapy of any type may be used in embodiments of the invention and may provide a more effective approach to destroying tumor cells.
  • Smac mimetics generally interact with IAPs, such as XlAP, cIAP-1, cIAP-2, ML-IAP, etc., and block the IAP mediated inhibition of apoptosis while chemotherapeutics/anti neoplastic agents and/or radiation therapy kills actively dividing cells by activating the intrinsic apoptotic pathway leading to apoptosis and cell death.
  • embodiments of the invention provide combinations of a Smac mimetic and chemotherapeutic/anti-neoplastic agents and/or radiation that may provide synergistic action against unwanted cell proliferation.
  • This synergistic action between a Smac mimetic and a chemotherapeutic/anti-neoplastic agent and/or radiation therapy can improve the efficiency of the chemotherapeutic/anti-neoplastic agent and/or radiation therapy.
  • chemotherapeutic agents include, but are not limited to the chemotherapeutic agents described in "Modern Pharmacology with Clinical Applications," Sixth Edition, Craig & Stitzel, Chpt. 56, pgs, 639-656 (2004), hereby incorporated by reference. This reference describes chemotherapeutic drugs including alkylating agents, antimetabolites, anti-tumor antibiotics, plant-derived products such as taxanes, enzymes, hormonal agents such as glucocorticoids, miscellaneous agents such as cisplatin, monoclonal antibodies, immunomodulating agents such as interferons, and cellular growth factors. Other suitable classifications for chemotherapeutic agents include mitotic inhibitors and nonsteroidal antiestrogenic analogs. Other suitable chemotherapeutic agents include toposiomerase ⁇ and II inhibitors and kinase inhibitors.
  • Suitable biological and chemotherapeutic agents include, but are not limited to, cisplatin, carmustine (BCNU), 5-flourouracil (5-FU), cytarabine (Ara-C), gemcitabine, methotrexate, daunorubicin, doxorubicin, dexamethasone, topotecan, etoposide, paciitaxel, vincristine, tamoxifen, TNF-alpha, TRAIL, interferon (in both its alpha and beta forms), thalidomide, and melphalan.
  • chemotherapeutic agents include nitrogen mustards such as cyclophosphamide, alkyl sulfonates, nitrosoureas, ethylenimines, triazenes, folate antagonists, purine analogs, pyrimidine analogs, anthracyclines, bleomycins, mitomycins, dactinomycins, plicamycin, vinca alkaloids, epipodophyllotoxins, taxanes, glucocorticoids, L-asparaginase, estrogens, androgens, progestins, luteinizing hormones, octreotide actetate, hydroxyurea, procarbazine, mitotane, hexamethylme ⁇ amme, carboplatin, mitoxantrone, monoclonal antibodies, levamisole, interferons, interleukins, filgrastim, and sargramostim.
  • Chemotherapeutic compositions also comprise other
  • the therapeutic compounds of the present invention may be administered with TRAIL or other chemical or biological agents which bind to and activate the TRAIL receptor(s).
  • TRAIL or other chemical or biological agents which bind to and activate the TRAIL receptor(s).
  • Many cancer cell types are sensitive to TRAIL-induced apoptosis, while most normal cells appear to be resistant to TRAIL-induced apoptosis.
  • TRA ⁇ L-resistant cells may arise by a variety of different mechanisms including loss of the receptor, presence of decoy receptors, or overexpression of FLIP which competes for zymogen caspase-8 binding during DISC formation.
  • Smac mimetics appear to increase tumor cell sensitivity to TRAIL leading to enhanced apoptosis, the clinical correlations of which are expected to be increased apoptotic activity in TRAIL resistant tumors, improved clinical response, increased response duration, and ultimately, enhanced patient survival rate.
  • reduction in XIAP levels by in vitro antisense treatment has been shown to cause sensitization of resistant melanoma cells and renal carcinoma cells to TRAIL (Chawla-Sarkar, et al., 2004).
  • the Smac mimetics disclosed herein may bind to IAPs and inhibit their interaction with caspases, therein potentiating TRAIL-induced apoptosis,
  • Another embodiment of the invention provides Smac mimetics that act synergisticaliy with a topoismerase inhibitor to potentiate their apoptotic inducing effect.
  • Topoisomerase inhibitors inhibit DNA replication and promote DNA damage by inhibiting the enzymes that are required in the DNA repair process thereby promoting apoptosis. Therefore, export of Smac from the mitochondria into the eel! cytosol is provoked by the DNA damage caused by topoisomerase inhibitors.
  • Topoisomerase inhibitors such as those of the Type I class, including camptothecin, topotecan, SN-38 (irinotecan active metabolite), and those of the Type II class including etoposide, show potent synergy with the Smac mimetics of the invention in a multi-resistant glioblastoma cell line (T98G), breast cancer line (MDA-MB-231), and ovarian cancer line (OVCAR-3) among others.
  • T98G multi-resistant glioblastoma cell line
  • MDA-MB-231 breast cancer line
  • OFVCAR-3 ovarian cancer line
  • topoisomerase inhibiting agents that may be used in embodiments of the invention include, but are not limited to irinotecan, topotecan, etoposide, amsacrine, exatecan, gimatecan, aclacinomycin A, camptothecin, daunorubicin, doxorubicin, ellipticine, epirubicin, and mitaxantrone.
  • a platinum containing compound may be used as chemotherapeutic/anti-neoplastic agent in combination with a Smac mimetic.
  • exemplary platinum containing compounds that may synergize with a Smac mimetic include, but are not limited to, cisplatin, carboplatin, and oxaliplatin.
  • taxanes may be used as the chemotherapeutic /anti-neoplastic agent that synergizes with a compound according to the invention.
  • Taxanes may act as, for example, anti-mitotic, mitotic inhibitors or microtubule polymerization agents and include, but are not limited to, docetaxel and paclitaxel. Taxanes are characterized as compounds that promote assembly of microtubules by inhibiting tubulin depolymerization, thereby blocking cell cycle progression.
  • Microtubules are highly dynamic cellular polymers made of alpha-beta-tubulin and associated proteins that play key roles during mitosis by participating in the organization and function of the spindle, assuring the integrity of the segregated DNA. Therefore, microtubules represent an effective target for cancer therapy, and taxanes may effectively attack this target by causing, for example, centrosomal impairment, induction of abnormal spindles, and suppression of spindle microtubule dynamics.
  • Another class of agents that may be utilized in embodiments of the invention includes microtubule poisons which, in contrast to taxanes, inhibit tubulin polymerization. These compounds include, but are not limited to vinca alkaloids, colchicine, and cryptophycines.
  • any agent that activates the intrinsic apoptotic pathway and/or causes the release of Smac or cytochrome c from the mitochondria has the potential to act synergistically with a Smac mimetic and may be used in combination with the compounds of embodiments of the invention.
  • Smac mimetic therapy may be used in connection with chemo-radiation or other radiation treatment protocols used to inhibit tumor cell growth.
  • Radiotherapy is the medical use of ionizing radiation as part of cancer treatment to control malignant cells and is suitable for use in embodiments of the present invention.
  • radiotherapy is often used as part of curative, primary, therapy, it is occasionally used as a palliative treatment where cure is not possible and the aim is for symptomatic relief.
  • Radiotherapy is commonly used for the treatment of tumors, and it is common to combine radiotherapy with surgery and/or chemotherapy.
  • the most common tumors treated with radiotherapy are breast cancer, prostate cancer, rectal cancer, head and neck cancers, gynecological tumors, bladder cancer, and lymphoma.
  • Radiation therapy is commonly applied just to the localized area involved with the tumor. Often the radiation fields include the draining lymph nodes.
  • Radiotherapy It is possible, but uncommon, to give radiotherapy to the whole body or entire skin surface. Radiation therapy is usually given daily for up to 35-38 fractions (a daily dose is a fraction). These small frequent doses allow healthy cells time to grow back, repairing damage inflicted by the radiation.
  • Three main divisions of radiotherapy are external beam radiotherapy, or teletherapy, brachytherapy or sealed source radiotherapy and unsealed source radiotherapy, which are all suitable examples of treatment protocol in the present invention. The differences relate to the position of the radiation source: external is outside the body, while sealed and unsealed source radiotherapy has radioactive material delivered internally. Brachytherapy sealed sources are usually extracted later, while unsealed sources are injected into the body. Administration of a Smac mimetic may occur prior to and/or concurrently with the treatment protocol. EXAMPLE
  • Annex in V/Propidium Iodide Stain ins Annexin V-fluorescein isothiocyanate staining shows the ability of dimeric Smac mimetics to induce apoptosis.
  • Cells are briefly exposed to various concentrations of dimeric Smac mimetics for 18-24 hours and removed from the assay plate by trypsinization. Cells are pelleted and resuspended in assay buffer (supplied by manufacturer).
  • Annexin V and propidium iodide are added to the cell preparations and incubated for 1 hour in the dark at room temperature. Following the incubation, additional buffer (200 ⁇ l) is added to each tube, and the samples are analyzed by flow cytometry.
  • Biological and chemotherapeutics/anti-neoplastic agents and radiation induce apoptosis by activating the extrinsic or intrinsic apoptotic pathways. Since Smac mimetics relieve inhibitors of apoptotic proteins (IAPs) and thus, remove the block in apoptosis, the combination of chemotherapeutics/anti-neoplastic agents and radiation with Smac mimetics should work synergistically to facilitate apoptosis.
  • IAPs inhibitors of apoptotic proteins
  • this potent synergy is that it makes possible the use of the dimeric Smac mimetics, which are IAP antagonists, to improve the efficacy of conventional chemotherapeutic agents, such as, marketed platinum containing compounds (cisplatin and carboplatin). This may be accomplished by lowering the required dose of the poorly tolerated platinum containing compounds and/or by improving the response rate at the marketed dose.
  • conventional chemotherapeutic agents such as, marketed platinum containing compounds (cisplatin and carboplatin). This may be accomplished by lowering the required dose of the poorly tolerated platinum containing compounds and/or by improving the response rate at the marketed dose.

Abstract

Compounds, compositions, and methods of using such compounds to modulate apoptosis including IAP antagonists are provided herein. Compositions including mimetics of the invention and, optionally, secondary agents, may be used to treat proliferative disorders such as, cancer and autoimmune diseases.

Description

A. Title:
DIMERlC IAP INHIBITORS
B. Cross-Reference to Related Applications:
[0001] This application claims priority to and benefit of U.S. Provisional Application No. 60/820,169 entitled "Dimeric IAP Inhibitors1' filed on July 24, 2006; the entire contents of which is hereby incorporated by reference in its entirety.
C. Government Interests: Not applicable
D. Parties to a Joint Research Agreement: Not applicable
E. Incorporation by Reference of Material submitted on a Compact Disc: Not applicable
F. Background
1. Field of Invention:
(0002] The invention presented herein provides compositions and methods for modulation of apoptotic signaling pathways.
2. Description of Related Art:
[0003J Apoptosis piays a central role in the development and homeostasis of all multicellular organisms. Alterations in apoptotic pathways have been implicated in many types of human pathologies, including developmental disorders, cancer, autoimmune diseases, and neurodegenerative disorders. One mode of action of chemotherapeutic drugs is cell death via apoptosis.
JΘ004] Apoptosis is conserved across species and executed primarily by activated caspases, a family of cysteine proteases that cleave their substrates specifically at aspartate residues. Caspases are produced in cells as catalytically inactive zymogens (procaspases) that are activated by proteolytic processing during the intitation of apoptosis. Once activated, effector caspases proteolytically activate a broad spectrum of cellular targets ultimately leading to cell death.
[0005] In mammalian cells activation of the caspases is achieved through at least two independent mechanisms which are initiated by distinct caspases, but result in the activation of common executioner (effector) caspases. The 'intrinsic pathway' is activated by cytochrome c which is released from mitochondria within the cell when apoptosis is initiated. The 'extrinsic pathway' is intiated via activation of a death receptor located on the cell membrane. During extrinsic activation, death receptors, such as, Fas (CD-95/Apol) and TNF-Rl , as well as other members of the TNF group of cytokine receptors, are activated by their corresponding ligands, Fas ligand (FasL/CD-95L) and TNF-alpha or Apo2 ϊigand/TNF-related apoptosis inducing ligand (Apo2L/TRΛIL), respectfully. Binding of procaspase-8 to an activated death receptor induces cleavage and removal of inhibitory domain of procaspase-8 releasing it from the receptor and allowing it to activate effector caspases-3. -6, and -7. The result is the proteolytic cleavage of cellular targets by the effector caspases and the induction of apoptosis.
[0006] In normal cells that have not received an apoptotic stimulus, most caspases remain inactive. Aberrantly activatation of caspases is inhibited by a family of evolutionarily conserved proteins called IAPs (inhibitors of apoptosis proteins). IAPs have been described in organisms ranging from Drosophila to Humans. All mammalian IAPs identified to date, including, for example, XIAP, cIAP-1 , cIAP-2, ML-IAP, NAIP, Bruce, and survivin exhibit anti- apoptotic activity in cell culture,
[0007] IAPs were originally discovered in Baculovirus by their ability to substitute for P35, an anti-apoptotic protein. Generally, IAPs are made up of one to three Baculovirus IAP repeat (BlR) domains, and must also possess a carboxyl-terminal RING finger motif. The BIR domain itself includes a zinc binding domain of about 70 residues made up of 4 alpha-helices and 3 beta strands. The BlR domain itself is believed to inhibit apoptosis by interacting with the procaspase and inhibiting proteolytic activation of the procaspase. IAPs are also known to be overexpressed in many human cancers. For example, XIAP is ubiquitously expressed in most adult and fetal tissues. However, overexpression of XIAP in tumor cells has been demonstrated to confer protection against a variety of apoptotic stimuli and promote resistance to chemotherapy. Consistent with this, a strong correlation between XIAP protein levels and survival of patients with acute myelogenous leukemia has been demonstrated. Down-regulation of XIAP expression by antisense oligonucleotides has been shown to sensitize tumor cells to a wide range of pro-apoptotic agents, both in vitro and in vivo.
[0008] Smac/DIABLO-derived peptides have also been demonstrated to sensitize tumor cell lines to pro-apoptotic drugs, In non-tumorigenic cells signaled to undergo apoptosis, IAP- mediated inhibition of apoptosis must be eliminated, which is accomplished, at least in part, by Smac (second mitochondrial activator of caspases). Smac. or DIABLO, is synthesized in the cytoplasm as a 239 amino acid precursor protein, of which the N-terminal 55 residues serve as the mitochondria targeting sequence that is removed after import to the mitochondria. Mature Smac, containing 184 amino acids, accumulates in the inter-membrane space of the mitochondria where it has been shown to behave as an oligomer. When apoptosis is induced, Smac is released from the mitochondria into the cytosol together with cytochrome c where it binds to IAPs eliminating the inhibitory effect of IAPs on protealysis of procaspases and enabling caspase activation. At the same time, cytochrome c induces multimerization of Apaf-1 to activate procaspase-9 and procaspase-3.
[0009] Smac interacts with essentially all IAPs identified to date including XIAP, c- IAPl , C-1AP2, ML-IAP, Bruce and survivin and may be a master regulator of apoptosis in mammals. X-ray crystallography has shown that the first four amino acids (AVPI) of mature Smac bind to a portion of IAPs and this binding is thought to be essential for blocking the anti- apoptotic effects of IAPs. Therefore, Smac and various fragments of Smac, including AVPI peptides, have been proposed for use as targets for identification of therapeutic agents.
[0010] The basic biology of IAP antagonists, such as Smac, suggests that these proteins may complement or synergize other chemotherapeutic/anti -neoplastic agents and/or radiation. Chemotherapeutic/anti-neoplastic agents and radiation would be expected to induce apoptosis as a result of DNA damage and/or the disruption of cellular metabolism. G. Brief summary of the invention: jθOll] Various embodiments of the invention are directed to a compound including a homodimer or heterodimer having monomeric units of formula (I):
Figure imgf000004_0001
wherein: each R1 is, independently, H; C|-C4-alky); Q-CValkenyl; C|-C4-alkynyl or C3-C1O- cycloalkyl which are unsubstituted or substituted; each R2 is, independently, H; C|-C4-aIkyl; C|-C4-alkenyl; CrC4-alkynyI or C3-C 1O- cycloalkyl which are unsubstituted or substituted; each R3 is, independently, H; -CF3; -C2H5; Ci-C4-alkyl; CrC4-alkeny]; CrC4-alkynyl; - CH2-Z or any R2 and R3 together form a heterocyclic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F or -CH2OH; each R4 is, independently, Ci-C 16 straight or branched alkyl; CrC]6-alkenyl; Ci-Ci6- alkynyl; C3-C;(rcycloalkyl; -(CH2)J-6-Z1; -{CH2)0-6-aryl; and -{Clh^-het; wherein alkyl, cycloalkyl, and pheny! are unsυbstituted or substituted; each Zs is, independently, -N(R|0)-C(O)-C|.i0-aikyl; -N(R1 O)-C(O)-(CII2)^-C3-7- cycloalkyl; -N(R,0)-C(O)-(CH3)0.6-phenyl; -N(R,0)-C(OMCH2),.6-het; -C(O)-N(Rn)(R12); - C(O)-O-C Mo-alkyl; -C(O)-O-(CH2), -Λ-v-cycloalkyl; -C(0)-0-(CH2)0-6'Phenyl; -C(O)-O- (CH2),^-het; -0-C(O)-C M0-alkyl; -0-C(O)-(CH2) w-C3.7-cycloaIkyl; -0-C(0)-(CH2)0-6-phenyl; - 0-C(O)-(CH2) i -6-het; wherein alkyl, cycloalkyl, and phenyl are unsυbstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1 -4 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom; each R1o is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each Rn and R12 is, independently, H; C^-alkyl; C3.7-cyc]oalkyl; -(CH2)^-C3-7- cycloaJkyl; (CH2)o-6-phenyl; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; or R1 j and R!2 together with the nitrogen form het; each R5 is, independently, H; CM0-alkyl; aryl; phenyl; C3-7-cycloalkyl; -(CH2) 1-0-C3-7- cycloalkyl; -CM0-alkyl-aryl; -(CH2)o-6-C3-7-cycloalky]-(CH2Wphenyl; -(CH2)<M-CH[(CI I2)M- phenyl]2; indanyl; -C(O)-C1-10-alkyϊ; -C(OHCH2)i.6-C3.7-cycloalky]; -C(O)-(CH2)0-6-phenyl; - (CH2)o-6-C(0)-phenyl; -(CH2)o-6-het; -C(O)-(CH2)i-6-het; or R5 is a residue of an amino acid, wherein the alkyl, cycloalkyl phenyl, and aryl substituents are unsubstituted or substituted; each U is, independently, as shown in structure (II):
Figure imgf000005_0001
wherein: each n is, independently, O to 5; each X is, independently, -CH or N; each Ra and R4, is, independently, an O, S. or N atom or Co.s-alkyl wherein one or more of the carbon atoms in the alkyl chain are optionally replaced by a heteroatom selected from O, S, or N, and where each alkyl is, independently, either unsubstituted or substituted; each Rd is, independently, selected from:
Figure imgf000006_0001
each R^ is, independently, H or any R^ and R4 together form a cycloalkyl or het; where if R(i and R^ form a cycloalkyl or het, R5 is attached to the formed ring at a C or N atom; each p and q is, independently, O or I ; each Re is, independently, C].g-alkyl or alkylidene, and each R^ is either unsubstituted or substituted; each Q is, independently, N, O, S, S(O), or S(O)2; each Arj and Ar2 is, independently, substituted or unsubstituted aryl or het; each Rf and Rg is, independently, H; -Cno-alkyl; CM 0-alkylaryl; -OH; -O-CM0-alkyl; - (CH2)o-6-C3-7-cycloalky]; -O-(CH2)0.6-aryi; phenyl; aryl; phenyl -phenyl; -(CH2)i.6-het; -O-(CH2)j. 6-het; -OR13; -C(0)-Ri3; -C(O)-N(R13)(R14); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2-Rn; -S(O)2- NR13R14; -NR13-S(O)2-R14; -S-Ct-10-aIkyl; aryl-CM-aikyl; or het-C M-alkyl wherein alkyl, cycloalkyl, het, and aryl are unsubstituted or substituted; -SO2-C |.2-alkyl; -SOa-C^-alkylphenyl; -O-Cι-4-alkyl; or any Rg and Rf together form a ring selected from het or aryl; each D is, independently, -CO-; -C(O)-C ι-7-alkylene or arylene; -CF2-; -0-; -S(0)r where r is 0-2; 1 ,3-dioxalane; or C1-7~alkyl-OH; where alkyl, alkylene, or arylene are unsubstituted or substituted with one or more halogens. OH, -O-C|-6-alkyl,
Figure imgf000006_0002
or -CF3; or each D is, independently, N(R11) wherein each Rh is, independently, H; unsubstituted or substituted C1 -7-alkyl; aryl; unsubstituted or substituted -O-(C|-7-cycloalkyl); -C(O)-C1-]0-aIkyI; - C(0)-C0-io-alkyl-aryl; -C-O-C M0-alkyl; -C-O-C0- lo-alkyl-aryl; -SO2-CM0-alkyI; or -SO2-(C0-Io- alkylaryl); each R6, R7, Re, and R9 is, independently, H, -CMo-alkyl; -Ci-io-alkoxy; aryl-C1 -!0- alkoxy; -OH; -O-CM0-aIkyl; -(CH3)0.6-C3.7-cycloalkyI; -0-(CH2Waryl; phenyl; -(CH2)] .6-het; - 0-(CH3),.6-het; -OR13; -C(O)-R13; -C(O)-N(R13)(RM); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2- R13; -S(O)2-NR13RH; or -NR13-S(O)2-R1^ wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; and any R6, R7, R8, and Rg optionally together form a ring system; each R13 and Rμ is, independently, H; C|_,o-alkyl; ~(CH2)o-ή-C3..7-cycloalkyi; -(CH2)O-O- (CH)o.r(aryl),.2; -C(O)-CM 0-alkyi; -C(O)-(CH2) ^-CYrcycioalkyl; -C(0)-0-(CH2)o-6-aryl; - C(0)-(CH2)o-6-0-fluorenyl; -C(O)-NH-(CH2)0^aryl; -C(0)-(CH2)o^-aryl; -C(O)-(CH2J0.6-het; - C(S)-CM0-alkyl; -C(S)-(CH2),. <,-C3-7-cycloalkyi; -C(S)-O-(CH2)0-6-aryl; -C(S)-(CH2V6-O- fluorenyl; -C(S)-NH-(CH2)0-6-aiyl; -C(S)-(CH2)o-ό-aryi; or -C(S)-(CH2)i^-hct; wherein each alkyl, cycloalkyi, and aryl is unsubstituted or substituted: or any Ro and R^ together with a nitrogen atom form het; wherein alkyl substituents of RB and R14 are unsubstituted or substituted and when substituted are substituted by one or more substituents selected from Ci-io-alkyl, halogen, OH,- O-C|-6-alkyl, -S-Ci^-alkyl, and -CF3; and substituted phenyl or aryl of Ro and R14 are substituted by one or more substituents selected from halogen, hydroxyl. C,.4-aikyl, C^-alkoxy, nitro, -CN, -0-C(O)-C, -4-alkyl, and -C(O)-O-C ,_r aryl; or a pharmaceutically acceptable salt or hydrate thereof.
[0012] In some embodiments, the compound of invention may have the formula (III):
Figure imgf000007_0001
wherein:
R1 and R1' are each, independently, H; Ci-Gt-alkyl; C|-C4-aJkenyl; C,-C4-alkynyl; or C3- Cio-cycloalkyl which are unsubstituted or substituted; R2 and R2' are each, independently, H; Ci-C^-alkyl; C,-C4-alkenyl; C|-C4-alkynyl; or C3- C,o-cycloalkyl which are unsubstituted or substituted; R3 and R3' are each, independently, H; -CF3; -C2Hs; C,-C4-alkyl; C,-C4-alkenyl; CrC4- alkynyi; or -CH2-Z or R2 and R3 together or independently with R2' and R3' form a heterocyclic ring; each Z is, independently, H; -OH; F; Ci; -CH3; -CF3; -CH2Cl; -CH2F; or -CH2OH;
R4 and R4' are each, independently, C ,-C^, straight or branched alkyl; C,-Ci6-alkenyl; C1- C 16-alkynyl; or C3-C,o-cycloaIkyI; -(CHi)1 -6-Zt; -(CHi^-aryl; or -(CH2)o-6-het; wherein alkyl, cycloalkyi, and phenyl are unsubstituted or substituted; each Zt is, independently, -N(R!0)-C(O)-CM0-alkyI; -N(R1O)-C(O)-(CH2WC3-7- cycloalkyl; -N(R,o)-C(0)-(CH2)0.6-pheny]; -N<R,0)-C(O)-(CH2)s.6-het; -C(O)-N(R1 1)(R12); - C(O)-O-C,. ,0-aIkyl; -C(O)-O-(CH2)1-6-C3.rcycIoalkyl; -C(O)-O-(CH2)0-6-phenyl; -C(O)-O- (CH2)I ^-het; -O-C(O)-CM0-alkyl; -O-C(O)-(CH2),.6-C3.7-cycloalkyl; -O-C(O)-(CH2)tM,-phenyl; or -O-C(O)-(CH2)t-6-het; wherein alkyl, cycloalkyi, and phenyl are unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1-4 heteroatoms selected from N, O, and S, or an 8- 12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom; each R1o is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each R) Ϊ and R12 is, independently, H; C^-alkyl; C3-7-cycloalkyl; -(CH2)I -O-Q-7- cycloalkyl; or (CH2)o-<j-phenyl; wherein alkyl, cycloalkyi, and phenyl are unsubstituted or substituted; or Rn and R12 together with a nitrogen form het; R8 and R5' are each, independently, H; Ci-io-alkyl; aryi; phenyl; C3-7-cycloalkyl; -(CH2)I- 6-C3.7-cycloalkyl; -Cuo-alkyl-aryl; -(CH2)o^-C3.7-cycloalkyl-(CH2)o-6-phenyl; -(CH2)O-4- CH[(CH2)M-phenyl]2; indanyl; -C(O)-C,.10-alkyl; -C(O)-(CH2)!-6-C3-7-cycloalkyl; -C(O)-(CH2V 6-phenyl; -(CH2)0-6-C(0)-phenyl; -(CH2)0-6-het; or -C(O)-(CH2) 1-6-het; wherein the alkyl. cycloalkyi, phenyl, and aryl substituents are unsubstituted or substituted; or R5 and R5' are, independently, an amino acid residue;
U and U* are each as shown in structure (II):
Figure imgf000008_0001
wherein: each X is, independently, -CH or N; each R3 and Rb is, independently, an O, S, or N atom or Co-s-alkyl wherein one or more of the carbon atoms in the alkyl chain are optionally replaced by a heteroatom selected from O, S, or N, and where each alkyl is, independently, either unsubstituted or substituted; each Rd is, independently, selected from:
Figure imgf000009_0001
Ar1-D-Ar2; each R0 is, independently, H or any Rc and Rj together form a cycloalkyl or het; where if R(j and Rc form a cycloalkyl or het, R5 is attached to the formed ring at a C or N atom; each p and q is, independently, 0 or 1 ; each Re is, independently, C|-8-alky] or alkylidene, and each Re is either υnsubstituted or substituted; each Q is, independently, N, O, S, S(O), or S(O)2; each Ari and Ar2 is, independently, substituted or unsubstituted aryl or het; each Rf and Rg is, independently, H; -C 1-io-alkyl: Ci-jo-alkylaryl; -OH; -O-C[-[0-alkyi; - (CH2)o-6-C3-7-cycloalkyI; -O-(CH2)0-6-aryl; phenyl; aryl; phenyl-phenyl; -(CH2) |.6-het; -O-(CH2)i. 6-het; -OR13; -C(O)-R13; -C(O)-N(R13)(RI4); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2-RI3; -S(O)2- NR13R14; -NR13-S(O)2-R14; -S-CM0-alkyl; aryl-CM-alkyl; or het-CM-alky] wherein alkyl, cycloalkyl, het, and aryl are unsubstituted or substituted; -Sθ2-C].2-alkyl; -Sθ2-C|.2-alkylphenyI; or -O-Ci-4-alkyl; or any Rg and Rf together form a ring selected from het or aryl; each D is, independently, -CO-; -C(O)-C 1.7-alkylene or arylene; -CF2-; -0-; -S(0)r where r is 0-2; 1,3-dioxalane; or Ci.7-alkyl-OH; where alkyl, alkylene, or arylene are unsubstituted or substituted with one or more halogens, OH, -O-Ci-6-alkyl, -S-C1-6-alkyl; or -CF3; or each D is, independently, N(Rh) wherein each Rh is, independently, H; unsubstituted or substituted C].7-alkyl; aryl; unsubstituted or substituted -O-(C|-7-cycloalkyl); -C(O)-C1.10-alkyl; - C(0)-Co-io-alkyl-aryl; -C-O-C M0-aikyl; -C-O-Co-io-alkyl-aryl; or -SO2-CM0-alkyl; or -SO3-(Co- 10-alkylaryi); each R^, R?, R8, and R9 is, independently, H, -Ci-io-alkyl; -Cμjo-alkoxy; aryl-Ci.io- alkoxy; -OH; -O-CM0-alkyl; -(CH2)o-6-C3-7-cycloalkyl; -O-(CH2)0-6-aryl; phenyl; -(CH2) i^-het; - O-(CH2),.6-het; -ORn; -C(O)-R13; -C(O)-N(R13)(R14); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2- R13; -S(O)2-NR13R14; or -NRI 3-S(O)2-RI4; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; and any R6, R7, R8, and R9 optionally together form a ring system; each R13 and R14 is, independently, H; Cμio-alkyl; -(CH2)0-6-C3-7-cycloalkyl; -(CH2)O^- (CH)(M-(aryϊ),.2; -C(O)-C M 0-alkyl; -C(O)-(CH2),.6-CY7-cycloalkyI; -C(O)-O-(CH2)0.6-aryl; - C(0)-(CH2)o-6-0-nuorenyl; -C(O)-NH-(CH2)0-6-aryl; -C(O)-(CH2Waryl; -C(0)-(CH2)o^-het; - C(S)-C |.10-alkyl; -C(S)-(CH2), ^-C^-cycloalkyl; -C(S)-O-(CH2)0-<)-aryh -C(S)-(CH2)0-6-O- fluorenyl; -C(S)-NH-(CH2)o-6-aryl; -C(SHCH2)o-6-aryi; or -C(S)-(CH2) ,,6-het; wherein each alkyi, cycloalkyl, and aryl is unsubstituted or substituted; or any R^ and Ru together with a nitrogen atom form het; wherein aikyl subsituents of R^ and Rj4 are unsubstituted or substituted and when substituted are substituted by one or more substituents selected from C|.ιo-alkyl, halogen, OH,- O-C;-«-alkyl, -S-C|-6-alkyl, and -CF3; and substituted phenyl or aryl of RB and R14 are substituted by one or more substituents selected from halogen, hydroxyl, C^-alkyl, CM-alkoxy, nitro, -CN, -0-C(O)-C M-alkyl, and -C(O)-O-C M-aryl;
L is one or more linkers covalently linking one or more of the positions R4, R5, or U, with R4', R5', or U'; or a pharmaceutically acceptable salt or hydrate thereof.
[0013] In some embodiments, L may covalently link two identical monomeric units or L covalently links two non-identical monomeric units. In various embodiments, L may be selected from alkylene, alkenylene, alkynylene, cycloalkylene, cycloalkylalkyiene, aryl, arylalkylene, arylalkylalkylene, and heterocycloalkylene, heterocycloalkylalkylene, heteroaryl and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, optionally-substituted alkylene, alkenylene, alkynylene cycloalkylene, cycloalkylalkyiene, heterocycloalkylene, heterocycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, amino, substituted amino, oxygen atom, sulfide, sulfoxide, sulfone, and disulfide, and in other embodiments, L may be selected from -CH2CH2-, -CH2CH2CH2-, -CH-CH-, 1,4-phenyl, 2,5- thiophenyl, -CH(OH)CH(OH)-, -CH2CH-O-CHCH2-, and -CH2C≡CC≡CCH2-.
[0014] In further embodiments, the compounds of the invention may have a formula selected from a compound of formula (IV):
Figure imgf000011_0001
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5 ', U, and U' are defined as in claim 2; and
L1 and L2, independently, link position R4 with R4' and R5 with R5'; or a pharmaceutically acceptable salt thereof; a compound of formula (V):
Figure imgf000011_0002
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5 ', U, and U' are defined as in claim 2; and L links position R4 with R4'; or a pharmaceutically acceptable salt thereof; and a compound of formula (VI):
Figure imgf000012_0001
wherein
R1, R1', R2, R2', R3, R3'. R4, R»\ R5, Rs', U, and U' are defined as in claim 2; and
L links position R5 with R5'; or a pharmaceutically acceptable salt thereof.
[0015] In various other embodiments, the compounds of the invention may be a homodimer or heterodimer having monomelic units of general formula (XIII):
Figure imgf000012_0002
wherein: each R] is, independently, II; each R2 is, independently, H or C|-C4-aIkyl which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH3, -SCH3. -CN, -SCN, and nitro; each R3 is, independently, H; -CF3; -C2F5; -CH2-Z or any R2 and R3 together with the nitrogen form a C3-C6 heteroaliphatic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; or CH2OH; each R4 is, independently, C|-Ct6 straight chain alkyl; C3-Ci0 branched chain alkyl; -
Figure imgf000012_0003
or -<CH2)0-6-het; wherein each alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each Zi is, independently, -N(R9)-C(0)-CMo-a]kyl; -N(R9K(O)-(CH2)L6-C3.?- cycioalkyl; -N(R9)-C(O)-(CH2)cwrphenyI; -N(R<,)-C(O)-(CH2)ι-6-het; -C(O)-N(R10)(R11); -C(O)- 0-CMo-a!kyl; -C(O)-U-(CH2)i-6-C3-7-cycloalkyl; -C(O)-O-(CH2)0-6-phenyi; -C(O)-O-(CH2),.*- het; -0-C(O)-C M0-a!kyl; -O-C(O)-(CH2)i-6-C3-7-cycloalkyl; -0-C(OMCH2)o-6-phenyl; or -O- C(O)-(CH2) ι^-het; wherein each alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted, and when substituted is substituted on a carbon atom by halogen, hydroxyl, C^-alkyl, d-4-alkoxy. nitro, -0-C(O)-Ci- 4-alkyl, ~C(O)-O-Ci_,-alkyl, or on a nitrogen by C1-4-alkyl, -0-C(O)-C M-alkyl, or -C(O)-O-C M- alkyl; each R9 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each R1o and Rn is, independently, H;
Figure imgf000013_0001
C3.7-cycloalkyl; -(CH2)I-6-C3-7- cycloalkyl; or (CH2)o-6-phenyl; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; or any R]0 and R1 \ together with a nitrogen form het; each X is, independently, CH or N; each R5 is, independently H; C|-10-alkyl; aryl; phenyl; C3-7-cycloalkyl; -(CH2)I-6-C3-7- cycloalkyl; -CM0-aIky!-aryI; -(CH2)o.ό-C3.7-cycloalkyl-(CH2)o-6-phenyl; -(CH2)(M-CHJ(CH2)M- phenyl]2; -(CH2)0^-CH(phenyl)2; -C(O)-CM0-aϊkyl; -C(OHCH2),.6-C3-7-cycloalkyl; -C(O)- (CH2)0-6-phenyI; -(CH2)o-6-C(0)-phenyl; -(CH2) 1-6-het; -C(O)-(CH2) w-het; or R5 an amino acid residue, wherein the alkyl, cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; each R63 is, independently, H, methyl, ethyl. -CF3, -CH2OH or -CH2Cl; or each any R5 and R^3 together with a nitrogen form a het; each Ra and Rg is independently, cis relative to acyl substituents at the one position of the ring and are each, independently, H, -C|.l0-alkyl; -OH; -O-C|.10-alkyl; -(CH2)0-6-C3.7-cycIoalkyl; -0-(CH2)o^aryl; phenyl; -(CII2)i-6-het; -O-(CH2)ι-6-het; -N(R12)(R13); -S-R13; -S(O)-R13; -S(O)2-
R12; or -S(O)2-NR12R13; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; each R12 and R13 is, independently, H;
Figure imgf000013_0002
-(CH2)0-6-C3.7-cycloaikyl; -(CH2)0-6- (CH)o-r(aryl)i.2; -C(O)-C,.,0-alkyJ;
Figure imgf000013_0003
-C(O)-O-(CH2WaryJ; - C(O)-(CH2y6-O-fluorenyl; -C(O)-NH-(CH2)0-6-aryl; -C(O)-(CH2)0-6-aryl; or -C(O)-(CH2) ,^-het; wherein each aikyl cycloalkyl, or aryl is unsubstituted or substituted; or any R12 and R13. together with a nitrogen atom form het; each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; each n is 0, 1, or 2; and wherein substituted alkyl is substituted by one or more substituents selected from a double bond, halogen, OH5-O-C ,-6-alkyl, -S-d.6-alkyl, and -CF3; substituted cycloalkyl is substituted by one or more substituents selected from a double bond, C 1-6-alkyl, halogen, OH.-O-Ci^alkyl, -S~C,.6-a]ky], and -CF3; and substituted phenyl or aryl is substituted by one or more substituents selected from halogen, hydroxy, CM-alkyl, C^-alkoxy, nitro, -CN, -0-C(O)-C M-alkyl, and --C(O)-O-CM- aryl; or a pharmaceutically acceptable salt or hydrate thereof.
[0016] In various other embodiments, the compounds of the invention may have the formula (XIV):
Figure imgf000014_0001
wherein:
R1 and Rf are each H; R2 and R2' are each, independently, H or C]-C4-alkyl which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH3, -SCH3, -CN, - SCN, and nitro; R3 and R3' are each, independently, H; -CF3; -CiF5; or -CH2-Z or R2 and R3 or R2' and R3 1 together with a nitrogen form a C3-Ct, heteroaliphatic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; or -CH2OH;
I1Li and R4' are each, independently, C|-C]6 straight chain alkyl; C3-C]O branched chain alkyl;
Figure imgf000014_0002
-(CH2)I-O-Zi ;
Figure imgf000014_0003
or -(CH2)o-ό-het; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; each Zi is, independently, -N(R9)-C(0)-Cuo-aikyl; -N(R9)-C(O)-(CH2),.6-C3.7- cycloalkyl; -N(R9)-C(0)-(CII2)o^-phenyl; -N(R9)-C(O)-(CH2)1-6-het; -C(O)-N(R10)(R1 i); -C(O)- O-Ci.io-alkyl; -C(O)-O-(CH2)^-C3.7-cycloa]kyl; -C(O)-O-(CH2)0-6-phenyl; -C(O)-O-(CH2),^- het; -O-C(O)-CM0-alkyl; -O-C(O)-(CH2)i-6-C3.7-cycloaIkyl; -O-C(O)-(CH2)0^-ρhenyl; or -O- C(O)-(CH2)I -6-het; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon atom by halogen, hydroxyl, CM-alkyl, CM-alkoxy, nitro, -0-C(O)-C M-alkyl, -C(O)-O-C M-alkyl, or on a nitrogen by CM-alkyI, -0-C(O)-C M-alky], or -C(O)-O-C1.4 -alkyl; each Ry is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each R1o and Rn is, independently, H; C^-alkyl; C3-7-cycIoalkyl; -(CH2)^-C3-7- cycloalkyl; or (CH2)o-ό-phenyl; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; or any R,o and Rn together with a nitrogen form het;
X and X' are each, independently, CH or N;
Rs and R5' are each, independently, H; Ci.jo-alkyl; aryl; phenyl; C3-7-cycloalkyl; -(CH2) 1- 6-C3-7-cycloalkyl; -C|-I0-aIkyl-aryl; -(CH2)0-6-C3-7-cycloalkyl-(CH2)0-6-ρhenyl; -(CH2)0^- CH[(CH2)M-phenyl]2; -(CH2)o-6-CH(phenyl)2; -C(O)-C, -i0-alkyl; -C(O)-(CH2)i-6-C3.7-cycloalkyl; -C(0)-(CH2)o-6-phenyl; -(CH2)0-6-C(O)-phenyl; -(CH2)ι^-het; or -C(O)-(CH2), ^-het; or R5 is an amino acid residue, wherein each alkyl, cycloalkyl, phenyl, and aryl substituent is unsubstituted or substituted; R6a and R63' are each, independently, H, methyl ethyl, -CF3, -CH2OH ,Or -CH2Cl; or
R5 and R&a are, independently, or together with R5' and R^a' together with a nitrogen are het;
Ra, Rg, Ra', and RK' are cis relative to an acyl substituent at position one of a ring to which they are attached and are each, independently, H, -C,.|0-alkyl; -OH; -0-C,-io-alkyl; - (CH2)0-6-C3-7-cycloalkyi; -O-(CH2Waryl; phenyl; -(CH2)M-het; -O-(CH2)i-6-het; -N(R12)(R13); - S-R12; -S(O)-R,2; -S(O)2-R12; or -S(O)2-NRI 2RI 3; wherein each alkyl, cycloalkyi, and aryl is unsubstituted or substituted; R12 and Ru are each, independently, H; Ct-io-alkyl; -(CH2)0-6-C3.7-cycloalkyl; -(CH2)o-ό~ (CH)o.i-(aryl),.2; -C(O)-C M0-alkyI; -C(O)-(CH2),^C3-7-cycIoalkyl; -C(O)-O-(CH2)(M,-^.; - C(O)-(CH2WO-fluorenyl; -C(O)-NI l-(CH2)0-6-aryl; -C(0)-(CH2)o-6-aryl; or -C(O)-(Cl I2)ι-6-het; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; or Rt2 and R^ together with a nitrogen atom form het; each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; n and n' are each, independently, O, 1, or 2; wherein each substituted alkyl is substituted by one or more substituents selected from a double bond, halogen, OH,-O-C!-6-alkyl, -S-C,-6-alkyl, and -CF3; each substituted cycloalkyl is substituted by one or more substituents selected from a double bond, Ci-6-alkyl, halogen, OH5-O-C !-6-alkyl, -S-Ct^-alkyl, and -CF3; and each substituted phenyl or aryl is substituted by one or more substituents selected from halogen, hydroxy, CM-alkyl, C^-alkoxy, nitro, -CN, -0-C(O)-C !-4-alkyl, and -C(O)-O-C1-4- aryl; and
L is one or more linkers covalently linking one or more of the positions R4, R5, R$, with
Figure imgf000016_0001
a pharmaceutically acceptable salt or hydrate thereof.
[0017] In some embodiments, L may covalently link two identical monomeric units or L covalently links two non-identical monomeric units. In various embodiments, L may be selected from alkylene, alkenylene, alkynylene, cycloalkylene, cycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, and heterocycloalkylene, heterocycloalkylalkylene, heteroaryl and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, optionally-substituted alkylene, alkenylene, alkynylene cycloalkylene, cycloalkylalkylene, heterocycloalkylene, heterocycioalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, amino, substituted amino, oxygen atom, sulfide, sulfoxide, sulfone, and disulfide, and in some other embodiments, L may be selected from -CH2CH2-, -CH2CH2CH2-- -CFT-CH-, 1 ,4-phenyl, 2,5-thiophenyl, -CH(OH)CH(OH)-, -CH2CH-O-CHCH2-, and -CH2C-CC≡CCH2-.
(0018) In certain embodiments, the compounds of the invention may have a formula selected from a compound of formula (XV):
Figure imgf000017_0001
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a', Ra, Ra', R8, R8', X, X', n, and n' are defined as in claim 8; and
L1 and L2, independently link position R4 with R4' and R5 with R5'; or a pharmaceutically acceptable salt or hydrate thereof; a compound of formula (XVl):
Figure imgf000017_0002
wherein
R1, R1', R2, R2' R3, R3', R4. R4' , R5 , R5', R6a, R6a', Ra, Ra,', R8, R8', X, X', n, and n' are defined as in claim 8; and
L1 and L2, independently link position R4 with R4' and R8 with R8'; or a pharmaceutically acceptable salt or hydrate thereof; a compound of formula (XVIl):
Figure imgf000018_0001
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a' , Ra, Ra', R8, R8', X, X', n, and n' are defined as in claim 8; and
L links position R4 with R4'; or a pharmaceutically acceptable salt or hydrate thereof; a compound of formula (XVIIl):
Figure imgf000018_0002
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a' , Ra, Ra', R8, R8', X- X', n, and n' are defined as in claim 8; and
L links position R5 with R5'; or a pharmaceutically acceptable salt or hydrate thereof; and a compound of formula (XIX):
Figure imgf000019_0001
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a' , Ra, Ra', R8, R8', X, X', n, and n' are defined as in claim 8; and
L links position R8 with R8'; or a pharmaceutically acceptable salt or hydrate thereof.
[0019| In particular embodiments, the compounds of the invention may have a formula selected from formula (VII):
Figure imgf000019_0002
formula (VIII):
formula (IX):
Figure imgf000020_0001
formula (X):
Figure imgf000021_0001
formula (XI):
Figure imgf000021_0002
formula (XII):
Figure imgf000021_0003
[0020] Some embodiments of the invention include pharmaceutical compositions including a compound of formula (111):
Figure imgf000022_0001
wherein:
Rι and Rt' are each, independently, H; C 1-C-i-alkyl;
Figure imgf000022_0002
C|-C4-a]kynyl; or C3- Cio-cycloalkyl which are unsubstituted or substituted; R2 and R2' are each, independently, H; Ci-C4-alkyI; Ci-C4-alkenyl; C j -C4-alkynyl; or C3- Cio-cycloalkyl which are unsubstituted or substituted; R3 and R3' are each, independently, H; -CF3; -C2H5; Ct-C4-alkyl; C 1-C4-alkenyl; Ci-C4- alkynyl; or -CH2-Z or R2 and R3 together or independently with R2' and R3' form a heterocyclic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; Or -CH3OH;
R4 and R4' are each, independently, Ci-Ci6 straight or branched alkyl; C 1-Ct 6-alkenyl; Ci- Ci6-alkynyl; or C3-Cio-cycloalkyl; -(CH2)1-6-Zι ; -(C H2 Wary I; or -(CH2)o^-het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each Z1 is, independently, -N(Rιo)-C(0)-CMO-alkyl; -N(R,0)-C(O)-(CH2)1-6-C3-7- cycloalkyl; -N(R,o)-C(0)~(CH2)0-6-phenyl; -N(R,0)-C(O)-(CH2)^-het; -C(O)-N(R11)(R12); - C(O)-O-C I-J o-alkyl; -C(O)-O-(CH2)1-6-C3-7-cycloalkyl; -C(O)-O-(CH2)0-t,-phenyl; -C(O)-O- (CH2)i.6-het; -O-C(O)-CM0-alkyl; -O-C(O)-(CH2)1.6-C3-7-cycloalkyl; -0-C(0)-(CH2)0-6-phenyl; or -0-C(O)-(CH2)I -6-het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1-4 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom; each R10 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each Rn and R12 is, independently, H; Ci-4-alkyl; C3-7-cyc!oalkyl; -(CIh)]^-Cw cycloalkyl; or (CH2)0-6-phenyl; wherein alky I, cycloalkyl. and phenyl are unsubstituted or substituted; or R| t and R12 together with a nitrogen form het;
R5 and R5' are each, independently, H; Cμio-alkyl; aryl; phenyl; C3-7-cycloalkyl; ~(CH2)ι_ 6-C3.7-cycIoalkyl; -C,.l 0-alkyl-aryl; -(CH2)0-6-C3-7-cycloalkyI-(CH2)o-ό-phenyl; -(CH2)(M- CHf(CH2) M-phenyI]2; indanyl; -C(O)-C1. l0-alkyl; -C(O)-(CH2) 1-6-C3.7-cycloa.kyl; -C(OMCH2)0. ό-phenyl; -(CH2)o^-C(0)-phenyi; -(CH2)o^-het; or -C(O)-(CH2)i-6-het; wherein the alkyl, cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; or R5 and R5' are, independently, an amino acid residue;
U and U' are each as shown in structure (II):
Figure imgf000023_0001
wherein; each X is, independently, -CH or N; each R3 and Rb is, independently, an O, S, or N atom or Co-s-alkyl wherein one or more of the carbon atoms in the alkyl chain are optionally replaced by a heteroatom selected from O, S, or N, and where each alkyl is, independently, either unsubstituted or substituted; each Rd is, independently, selected from; R,-Q-(Rf)p(Rg)q; and An-D-Ar2; each RH; is, independently, H or any Rc and R4 together form a cycloalkyl or het; where if Rd and Rc form a cycloalkyl or het, R5 is attached to the formed ring at a C or N atom; each p and q is, independently, O or 1 ; each R^ is, independently, Ci_s-alkyl or alkylidene, and each R^ is either unsubstituted or substituted; each Q is, independently, N, O, S, S(O). or S(O)2; each An and Ar2 is. independently, substituted or unsubstituted aryl or het; each R1 and R6 is, independently, H; -CVio-alkyi; CΗo-alkylaryl; -OH; -O-C 1.io-alkyl; - {CH2V6-C3-7-cycloaIkyl; -O-(CH2)0-6-aryl; phenyl; aryl; phenyl-phenyl; -(CH2)i.6-het; -0-(CH2)i. 6-het; -OR!3; -C(O)-R13; -C(O)-N(R13)(R14); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2-R13; -S(O)2- NR13R14; -NRI 3-S(O)2-RI4; -S-Ci-io-alkyl; aryl-CI-4-alkyI; or het-CM-alkyl wherein alkyl, cycloalkyi, het, and aryl are unsubstituted or substituted; -SO2-C1.2-al.cyl; -Sθ2-C).2-alkyiphenyI; or -O-Ci-4-alkyl; or any Rg and R1 together form a ring selected from het or aryi; each D is, independently, -CO-; -C(0)-Ci.7-alkylene or arylene; -CF2-; -O- ; -S(O)1- where r is 0-2; 1,3-dioxalane; or d-7-alkyl-OH; where alkyl, alkylene, or arylene are unsubstituted or substituted with one or more halogens, OH, -O-C^-alkyl, -S-C|-6-alkyi; Or -CF3; or each D is, independently, N(Rj1) wherein each Rh is, independently, H; unsubstituted or substituted C|-7-alkyl; aryl; unsubstituted or substituted -O-(C].7-cycloalkyl); -C(O)-Ci. to-alkyl; - C(O)-C0. i o-alkyl-aryl; -C-O-C1-10-alkyl; -C-O-Co-io-alkyl-aryl; or -S02-Ct-io-alkyl; or -SO2-(C0- 10-a!kylaryI); each R43, R7, R8, and R9 is, independently, H, -C|-to-alkyl; -Ct-10-alkoxy; aryl-Ci-10- alkoxy; -OH; -O-CM0-alkyl; -(CH2)0-6-C3.7-cycloalkyϊ; -O-(CH2)0-6-aryI; phenyl; -(CH2)1-6-het; - O-(CH2)t-6-het; -OR13; -C(O)-R13; -C(O)-N(R13)(Rt4); -N(Rj3)(R14); -S-R13; -S(O)-R13; -S(O)2-
R13; -S(O)2-NR13R14; or -NR13-S(O)2-R14; wherein each alkyl, cycloalkyi, and aryl is unsubstituted or substituted; and any R^, R7, R8, and Rg optionally together form a ring system; each R|3 and R14 is, independently, H; Ci-io-alkyl; -(CH2)0-6-C3-7-cycloalkyl; -(CH2)o-ό- (CH)o.t-(aryl)i.2; -C(O)-C, -10-alkyl; -C(O)-(CH2)1.6-C3.7-cycioalkyl; -C(0)-0-(CH2Waryl; - C(0)-(CH2W-0-fluorenyl; -C(O)-NH-(CH2)0-6-aryl; -C(0)-(CH2)o-6-aiyl; -C(0)-(CH2Whet; - C(S)-C ,.10-aIkyl; -C(S)-(CH2)f-6-C3-7-cycloalkyl; -C(S)-0-(CH2)o-6-aryI; -C(S)-(CH2WO- fluorenyl; -C(S)-NH-(CH2)o-6-aryl; -C(S)-(CH2WaTyI; or -C(S)-(CH2) 1-6-het; wherein each alkyl, cycloalkyi, and aryl is unsubstituted or substituted; or any R13 and R14 together with a nitrogen atom form het; wherein alkyl subsituents of R13 and R14 are unsubstituted or substituted and when substituted are substituted by one or more substituents selected from Cμio-alkyl, halogen, OH,- O-C|.6-alkyl, -S-C^-alkyl, and -CF3; and substituted phenyl or aryl of R13 and Rt4 are substituted by one or more substituents selected from halogen, hydroxy!, CM-alkyl, C^-alkoxy, nitro. -CN, -0-C(O)-C M-aIkyl, and -C(O)-O-C M-ary I;
L is one or more linkers covalently linking one or more of the positions R4, R5, or U, with R4', R5', or U'; or a pharmaceutically acceptable salt or hydrate thereof; and a pharmaceutically acceptable excipient or carrier; or a compound of formula:
Figure imgf000025_0001
wherein:
R1 and R| ' are each H; R2 and R2' are each, independently, H or C1-C4-alkyI which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH3, -SCH3, -CN, - SCN, and nitro; R3 and R3' are each, independently, H; -CF3; -C2F5; or -CH2-Z or R2 and R3 or R2' and R3" together with a nitrogen form a C3-C6 heteroaliphatic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; Or -CH2OH;
R4 and R4' are each, independently, C1-C16 straight chain alkyl; C3-CiO branched chain alkyl; -(CH2)0-6-C3-C7-cycloalkyl; -(CH2) 1-6-Z1 ; -(CH2)0-6-phenyl; or -(CH2)0-6-het; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; each Zi is, independently, -N(R9)-C(0)-C1 -10-alkyl; -N(R9)-C(O)-(CH2)1-6-C3-7- cycloalkyl; -N(R9)-C(O)-(CH2)0-6-phenyl; -N(R9)-C(OMCH2),.6-het; -C(O)-N(R10)(R1 1); -C(O)- O-CM0-alkyl; -C(O)-O-(CH2), -6-C3-7-cycloalkyl; -C(0)-0-(CH2)0-6-ρhenyl; -C(O)-O-(CH2), -6- het; -0-C(0)-C1 -10-alkyl; -0-C(O)-(CH2) 1-6-C3-7-cycloalkyl; -0-C(0)-(CH2)0-6-phenyl; or -O- C(O)-(CH2)1-6-het; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon atom by halogen, hydroxyl, C 1-4-alkyl, C 1-4-alkoxy, nitro, -O-C(O)-C1-4-alkyl, -C(O)-O-C 1-4-alkyl, or on a nitrogen by C 1-4-aIkyl, -0-C(O)-C 1-4-alkyl, or -C(O)-O-C 1-4-a!kyl; each R9 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2CI; each R1o and Rn is, independently, H; Q-4-alkyl; C3_7-cycloalky]; -(CH2)I -6-C3.7- cycloalkyh or (CH2 ^-phenyl; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; or any R]0 and Rf 1 together with a nitrogen form het;
X and X' are each, independently, CH or N;
R5 and RjT are each, independently, 11; C].|0-aikyl; aryl; phenyl; C3-7-cycloalkyl; -(CH2)]. 6-C3-7-cycloalkyI; -CM0-alky]-aryl; -(CH2)o-6-CY7-cycIoalkyI-(CH2)o,6-phenyi; -(CH2)(M- CH [(CH2), -4-phenyl]2; -(CH2)0-6-CH(phenyl)2; -C(O)-C M ralkyl; -C(O)-(CH2), -6-C3.7-cyc.oa.kyl; -C(0)-(CH2)o-6-phenyl; -(CH2)o-6-C(0)-pheny]; -(CH2),^-het; or -C(O)-(C H2) !-6-het; or R5 is an amino acid residue, wherein each alkyl, cycioalky], phenyl, and aryl substituent is unsubstituted or substituted; R6a and K6^ are each, independently, H, methyl, ethyl, -CF3, -CH2OH ,or -CH2Cl; or
R5 and R^ are, independently, or together with R5' and R&,' together with a nitrogen are het;
Ra, Rg, Ra', and R^' are cis relative to an acyl substituent at position one of a ring to which they are attached and are each, independently, H, -C 1-io-alkyl; -OH; -O-C 1-io-alkyl; - (CH2)o-6-C3.7-cycloalkyl; -O-(CH2)0-6-aryl; phenyl; -(CH2),^-het; -O-(CH2)ι-6-het; -N(R12)(R13); - S-R,2; -S(O)-R12; -S(O)2-R12; or -S(O)2-NR12R13; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted;
R12 and R13 are each, independently, H; CMo-alkyl; -(CH2)0-6-C3-7-cycloalkyl; -(CH2)0-ό- (CH)o-r(aryI),.2; -C(O)-Ci.,0-alkyl;
Figure imgf000026_0001
- C(0)-(CH2)o-6-0-fluoreny]; -C(0)-NH-(CH2)o-6-aryl; -C(O)-(CH2)0.6-aiyl; or -C(O)-(CH2) w-het; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; or R12 and R)3 together with a nitrogen atom form het; each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; n and n* are each, independently, O, ] , or 2; wherein each substituted alkyl is substituted by one or more substituents selected from a double bond, halogen, OH.-O-C^-alkyl, -S-C^-alkyl. and -CF3; each substituted cycloalkyl is substituted by one or more substituents selected from a double bond, C)-6-alkyl, halogen, OH,-O-C[-6-alkyl, -S-C1-6-alkyl, and -CF3; and each substituted phenyl or aryl is substituted by one or more substituents selected from halogen, hydroxy, CM-aikyl, CM-alkoxy, nitro, -CN, -0-C(O)-C M-alkyl, and -C(O)-O-CM- aryl; and
L is one or more linkers covalently linking one or more of the positions R4, R5, R^, with R4', R5 \ R8';or a pharmaceutically acceptable salt or hydrate thereof; and a pharmaceutically acceptable excipient or carrier.
[0021] The pharmaceutical composition of claim 14, wherein L and L' are selected from alkyiene, alkenylene, alkynylene, cycloalkylene, cycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heterocycloalkylene, heterocycloalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, optionally-substituted alkylene, alkenylene, alkynylene cycloalkylene, cycloalkylalkylene, heterocycloalkylene, heterocycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, amino, substituted amino, oxygen atom, sulfide, sulfoxide, sulfone, and disulfide.
[0022] In various embodiments, L and L' may be selected from -CH2CH2-, - CH2CH2CH2-, -CH=CH-, 1 ,4-phenyl, 2,5-thiophenyl, -CH(OH)CH(OH)-, -CH2CH-O-CHCH2-, and -CH2C≡CC≡CCH2-.
[0023J In some embodiments, the pharmaceutical compositions of the invention may include a second therapeutic agent, and in other embodiments, the compounds of the invention may provide therapy to an individual. In other embodiments, the pharmaceutical composition of the invention may further include a second therapeutic agent is selected from a chemotherapeutic agent, radiation, and a combination thereof, and in certain embodiemtns, the chemotherapeutic may be selected from an alkylating agent, a plant alkaloid, an antitumor antibiotic, an antimetabolite, a topoisomerase inhibitor, and a combination thereof. H. Description of Drawings: Not Applicable. I. Detailed Description:
[0024] It must be noted that, as used herein, and in the appended claims, the singular forms "a", "an" and "the" include plural reference unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein, have the same meanings as commonly understood by one of ordinary skill in the art. Although any methods similar or equivalent to those described herein can be used in the practice or testing of embodiments of the present invention, the preferred methods are now described. All publications and references mentioned herein are incorporated by reference. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
[0025] As used herein, the term "about" means plus or minus 10% of the numerical value of the number with which it is being used. Therefore, about 50% means in the range of 45%-55%.
[0026] The terms "mimetic", "peptide mimetic" and "peptidomimetic" are used interchangeably herein, and generally refer to a peptide, partial peptide or non-peptide molecule that mimics the tertiary binding structure or activity of a selected native peptide or protein functional domain (e.g., binding motif or active site). These peptide mimetics include recombinantly or chemically produced peptides, recombinantly or chemically modified peptides, as well as non-peptide agents, such as small molecule drug mimetics, as further described below.
[0027] As used herein, the terms "pharmaceutically acceptable", "physiologically tolerable" and grammatical variations thereof, as they refer to compositions, carriers, diluents and reagents, are used interchangeably and represent that the materials are capable of administration upon a mammal without the production of undesirable physiological effects such as nausea, dizziness, rash, or gastric upset.
[0028] "Providing" when used in conjunction with a therapeutic means to administer a therapeutic directly into or onto a target tissue, or to administer a therapeutic to a patient whereby the therapeutic positively impacts the tissue to which it is targeted.
[0029] As used herein, "subject", "patient" or "Individual" refers to an animal or mammal including, but not limited to, a human, dog, cat, horse, cow, pig, sheep, goat, chicken, monkey, rabbit, rat, or mouse, etc.
[0030] As used herein, the term "therapeutic" means an agent utilized to treat, combat, ameliorate, prevent or improve an unwanted condition or disease of a patient. Embodiments of the present invention are directed to promote apoptosis and, thus, cell death.
[0031] The terms "therapeutically effective amount" or "effective amount," as used herein, may be used interchangeably and refer to an amount of a therapeutic compound component of the present invention. For example, a therapeutically effective amount of a therapeutic compound is a predetermined amount calculated to achieve the desired effect, i.e., to effectiveiy promote apoptosis, preferably by eliminating IAP inhibition of apoptosis, more preferably by inhibiting an IAP binding to a caspase.
[0032] The terms "mimetics" or "peptidomimetics" are interchangeable and refer to synthetic compounds having a three-dimensional structure (i.e. a "core peptide motif') based upon the three-dimensional structure of a selected peptide. The peptide motif provides the mimetic compound with the desired biological activity, i.e., binding to IAP, wherein the binding activity of the mimetic compound is not substantially reduced, and is often the same as or greater than the binding affinity of the native peptide on which the mimetic is modeled. For example, in the mimetics of the present invention, we have found that portions of compounds based on peptides can be non-peptide like. Peptidomimetic compounds can have additional characteristics that enhance their therapeutic application, such as increased cell permeability, greater affinity and/or avidity, and prolonged biological half-life.
|0033] "Optional" or "optionally" may be taken to mean that the subsequently described structure, event or circumstance may or may not occur, and that the description includes instances where the events occurs and instances where it does not.
[Θ034J "Alkyl" or "alkylene" unless otherwise specified, means a branched or unbranched, saturated aliphatic hydrocarbon group, having up to 12 carbon atoms. When used as part of another term, for example, "alkylamino," the alkyl portion may be a saturated hydrocarbon chain. Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n- butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, 2-methyibutyl, 2,2-dimethylpropyl, n-hexyi, 2- methylpentyl, 2,2-dimethylbutyl, n-heptyl, 3-heptyl, 2-methyIhexyf, and the like. The terms "lower alkyl", "Ci-C4 alkyl", and "alkyl of 1 to 4 carbon atoms" are synonymous and used interchangeably to mean methyl, ethyl, 1 -propyl, isopropyl, cyclopropyi, 1 -butyl, sec-butyl or t- butyl. Unless specified, substituted alkyl groups may contain one, two, three or four substituents which may be the same or different.
|0035] "Substituenf* or "substituents" as used herein refer to a molecular group that replaces a hydrogen at any methyl group on a hydrocarbon. Substituents include, for example, halo, pseudohaio, hydroxy, protected hydroxy, trityloxy, carboxy, carbonyl. cyano, nitro, acyl, acyloxy, acetyl, acetoxy, carbamoyl, carbamoyloxy, ally], allyloxy, oxo, thia, nitrile, formyl, mercapto, hydroxycarbonyi, hydroxycarbonylalkyl, alkyl, haloalkyl, polyhaloalkyl, aminoalkyl, diaminoalkyl, alkenyl containing 1 to 2 double bonds, alkynyi containing 1 to 2 triple bonds, cycloalkyl, cycloalkylalkyl, cycloalkyloxy, heterocyclyl, heterocyclylalkyl, aryl, aryloxy, arylalkyi, aralkenyl, aralkynyl, heteroaryl, heteroaryloxy, heteroarylalkyi, trialkylsilyl, diaikylarylsilyl, alkyldiarylsilyi, triarylsilyi, alkylidene, arylalkylidene, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl, alkoxycarbonyl, alkoxycarbonylalkyl, aryloxycarbonyl, aryloxycarbonylalkyl, aralkoxycarbonyl, aralkoxycarbonyialkyl, arylcarbonylalkyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, arylaminocarbony], diarylaminocarbonyl, arylalkylaminocarbonyl, alkoxy, aryloxy, heteroaryloxy, heteroaralkoxy, heterocyciyioxy, heterocyclylsulfonyl, cycloalkoxy, perfϊuoroaikoxy, alkenyloxy, alkynyloxy, aralkoxy, alkylcarbonyloxy, arylcarbonyloxy, aralkylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, aralkoxycarbonyloxy, aminocarbonyloxy, alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkylaryiaminocarbonyloxy, diarylaminocarbonyloxy, guanidino, isothϊoureido, ureido, N-alkylureido, N-arylureido, N'-alkylureido, N*,N'-dialkylureido, N'- aikyl-N'-arylureido, N',N'-diarylureido. N'-arylureido, N'.N'-dialkylureido, N-alkyl-N'- arylureido, N-aryl-N'-alkylureido, N,N'-diarylureido, N, N',N'-trialkylureido, N,N'-dialkyl-N'- arylureido, N-alkyl-N\N'-diarylureido, N-aryI-N',N'-dialkylureido, N,N'-diaryl-N'-aIkylureido, N,N',N'-triarylureido, amidino, alkylamidino, arylamidino, aminothiocarbonyl, alkylaminothiocarbonyl, aryl aminothiocarbonyl, amino, protected amino, aminoalkyl, aminothio, acylamino, aminosulfinyl, aminosulfonyl, alkylaminoalkyl, dialkylaminoalkyl, arylaminoalkyl, diarylaminoalkyl, alkylarylaminoalkyi, alkylamino, dialkylamino, haloalkylamino, arylamino, diarylamino, alkylarylamino, alkylcarbonylamino, alkoxycarbonylamino, aralkoxycarbonylamino, arylcarbonyiamino, arylcarbonylaminoalkyl, aryioxycarbonylaminoalkyl, aryloxyarylcarbonylamino, aryloxycarbonylamino, alkylsulfonylamino, arylsulfonylamino, heteroarylsulfonylamino, heterocyclylsulfonylamino, heteroarylthio, azido, dialkylphosphonyl, alkylarylphosphonyl, diarylphosphonyl, hydroxyphosphonyl, alkylthio, arylthio, perfluoroalkylthio, hydroxycarbonylalkylthio, thiocyano, isothiocyano, alkylsuifinyl, alkylsulfmyloxy, alkylsullbnyl, alkylsulfonyloxy, arylsulflnyloxy, arylsulfonyloxy, hydroxysulfonyloxy, alkoxysulfonyloxy, aminosulfonyloxy, alkylaminosuifonyl alkylaminosuJfonyioxy, alkyiarylaminosulfonyloxy, alkylsulfonyl, arylsulfinyl, alkylsulfonylamino, arylsulfonyl, hydroxysulfonylo, alkoxysulfonyl, aminosulfonyl, alkylaminosuifonyl, diaikylaminosulfonyl, arylaminosulfonyl, diarylamiπosulfonyl, and alkylarylaminosutfonyl, alkyloxycarbonylamino, allyloxycarbony], allyloxycarbonylamino, and the like. For example, particular substituted alkyis are substituted methyls, e.g., a methyl group substituted by the same substituents as the "substituted Cn-Cm alky!" group. "Substituted alkyi" may include aikyloxymethyl, such as, methoxymethyl, ethoxymethyl, and t-butoxymethyl; halomethyi, such as, chloromethyl, brσmomethyl, iodomethy], and trifluoromethyl; hydroxymethyl; protected hydroxymethyl, such as, tetrahydropyranyloxymethyl; trityloxymethyl; cyanomethyl; nitromethyl; aminomethyl; carboxymethyl; alkyloxycarbonylmethyl; acetoxymethyl, carbamoyloxymethyl; allyloxycarbonylaminomethyl; propionyloxymethyl; acetoxymethyl; 6-hydroxyhexyl; 2,4-dichloro(n-butyl); 2-amino(iso- propyl); 2-carbamoyloxyethyl; carbocycle group, such as, for example, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, and cyclohexylmethyl groups, as well as the corresponding-ethyl, -propyl, -butyl, -pentyl, -hexyl groups, etc.
[0036] "Alkenyl" or "alkenylene" as used herein refers to an unsaturated, branched or unbranched, alphatic hydrocarbon having one or more double bond (-C=C-), and "alkynyl" or "alkynylene" as used herein refers to an unsaturated, branched or unbranched, alphatic hydrocarbon containing one or more triple bond (-C=C-). Unsaturated hydrocarbons may have up to 12 carbon atoms and may be substituted by one or more of any of the substituents described hereinabove. When used as part of another term, for example, "alkenylamino" and "aikynylamino" the alky! portion may be an unsaturated hydrocarbon chain.
[0037] "Amino" denotes primary (i.e. -NH2), secondary (i.e. -NRH), and tertiary (i.e. - NRR) amines. Particular secondary and tertiary amines include, but are not limited to, alkylamine, dialkylamine, arylamine, diarylamine, arylalkylamine and diarylalkylamine including, for example, methylamine, ethylamine, propylamine, isopropylamine, phenylamine, benzylamine, dimethylamine, diethylamine, dipropylamine and disopropylamine.
[0038] "Aryl", when used alone or as part of another term, means a fused or unfused carbocyclic aromatic group having a designated number of carbon atoms, or if no number is designated, up to 14 carbon atoms. Particular aryl groups include phenyl, naphthyl, biphenyl, phenanthrenyl, naphthacenyl, and the like (see, Lang's Handbook of Chemistry 13th ed. (Dean, J. A., ed.) Table 7-2 [ 1985]). As used herein a "fused ring system'' refers to two or more substituted or unsubstituted carbocyclic or carbocyclic aromatic groups that are fused together. Substituted phenyl or substituted aryl denotes a phenyl or aryl group substituted with one, two, three, four or five substituents chosen from those described above, for example, halogen (F. Cl, Br, I), hydroxy, protected hydroxy, cyano, nitro, alkyi (such as CrC6 alkyl), alkoxy (such as, Q- C(, alkoxy), benzyloxy, carboxy, protected carboxy, carboxymethyl, protected carboxy methyl, hydroxymethyl. protected hydroxymethyl, aminomethyl, protected aminomethyl, trifluoromethyl, alkylsulfonylamino, arylsulfonylamino, heterocyclylsulfonylamino, heterocyclyl, or aryl, and one or more methyne (CH) and/or methylene (CH2) groups in these substituents may be substituted with a group similar to those described above. Examples of "substituted phenyls" that may be utilized in embodiments of the invention include, but are not limited to, mono- or di-halo-phenyl, such as, 2-chlorophenyl, 2-bromophenyi, 4-chlorophenyl, 2,6-dichlorophenyl, 2,5-dichlorophenyl, 3,4-dichlorophenyl, 3-chlorophenyI, 3-bromophenyl, 4- bromophenyl, 3,4-dibromophenyl, 3-chloro-4-fluorophenyl, 2-fluoropheny!, and the like; mono- or di-hydroxyphenyl, such as, 4-hydroxyphenyl, 3-hydroxyphenyl, 2,4-dihydroxyphenyl, protected-hydroxy derivatives thereof, and the like; nitrophenyl, such as, 3- or 4-nitrophenyl; cyanophenyl, for example, 4-cyanophenyl; mono- or di-lower alkyl-phenyl group, such as, 4- methylphenyl, 2,4-dimethylphenyl, 2-methylphenyi, 4-(iso-propyl)phenyl, 4-ethyiphenyl, 3-(n- propyl)phenyl, and the like; a mono- or di-alkoxy-phenyl group, for example, 3,4- dimethoxyphenyl, 3-methoxy-4-benzyloxyphenyl, 3-methoxy-4-(l -chloromethyl) benzyloxy- phenyl, 3 -ethoxy phenyl, 4-(isopropoxy)phenyl, 4-(t-butoxy)phenyl, 3-ethoxy-4-methoxyphenyl and the like; 3- or 4-trifluoromethylphenyi; mono- or di-carboxyphenyl or protected carboxy phenyl, such as, 4-carboxyphenyl; mono- or di-hydroxymethyl-phenyl or protected hydroxymethyl phenyl, such as 3-(protected hydroxymethyl) phenyl or 3,4- di(hydroxymethyl)phenyl; mono- or di-(aminomethyl) phenyl or protected aminomethyl phenyl, such as 2-(aminomethyl)phenyl or 2,4-(protected aminomethyi) phenyl; or mono- or di-(N- (methylsulfonylamino)) phenyl, such as, 3-(N-methylsulfonylamino) phenyl. In addition, "substituted phenyl" may represent di-substituted phenyl groups where the substituents are different, such as, for example, 3-methyl-4-hydroxyphenyl, 3-chloro-4-hydroxyphenyl, 2- methoxy-4-bromophenyϊ, 4-ethyl-2-hydroxyphenyL 3-hydroxy-4-nitrophenyl, 2-hydroxy-4- chlorophenyl, and the like, as well as tri-substituted phenyl groups where the substituents are different, such as, for example, 3-methoxy-4-benzyloxy-ό-methyl sulfonylamino, 3-methoxy-4- benzyloxy-6-phenyi sulfonylamino and the like and tetra-substituted phenyl groups where the substituents are different, such as, for example, 3-methoxy~4-benzyloxy-5-methyl-6-phenyl sulfonylarnino. Particular substituted phenyl groups include 2-ch3orophenyl, 2-aminophenyl, 2- bromophenyl, 3-methoxyphenyl, 3-ethoxy-phenyl, 4-benzyloxyphenyl, 4-methoxyphenyl, 3- ethoxy-4-benzyloxyphenyl, 3,4-diethoxyphenyI, 3-methoxy-4-benzyIoxyphenyl, 3-methoxy-4- ( 1-chloromethyl) benzyloxy-phenyl, 3-methoxy-4-( l-chloromethyl), and benzyloxy-6-methyl sulfonyl aminophenyl groups. Fused aryl rings may also be substituted with one or more of any of the substituents specified herein, for example, fused aryl groups may contain 1 , 2 or 3 substituents in the same manner as substituted alkyl groups.
|0039] "Heterocyclic group", "heterocyclic", "heterocycle", "heterocyclyl", or "heterocyclo" alone, and when used as a moiety in a complex group such as a heterocycloalkyl group, are used interchangeably and refer to any mono-, bi-, or tri-cyclic, saturated or unsaturated, aromatic (heteroaryl) or non-aromatic ring having the number of atoms designated, generally from 5 to about 14 ring atoms, where the ring atoms are carbon and at least one heteroatom (nitrogen, sulfur or oxygen). In a particular embodiment, the group incorporates 1 to 4 heteroatoms. Typically, a 5- member ring has 0 to 2 double bonds and a 6- or 7-member ring has 0 to 3 double bonds; and the nitrogen or sulfur heteroatoms may optionally be oxidized (e.g. SO, SO2), and any nitrogen heteroatom may optionally be quaternized. Particular non-aromatic heterocycles include morpholinyl (morpholino), pyrrolidinyl, oxiranyl, oxetanyl, tetrahydrofuranyl, 2,3-dihydrofuranyl, 2H-pyranyl, tetrahydropyranyl, thϋranyl, thietanyl, tetrahydrothietanyl, aziridinyl, azetidinyl, l-methyl-2-pyrrolyl, piperazinyl, and piperidinyl. A "heterocycloalkyl" group is a heterocycle group as defined above, covalently bonded to an alkyl group as defined above. Particular 5-membered heterocycles containing a sulfur or oxygen atom and one to three nitrogen atoms include thiazolyl, such as thiazol-2-yl and thiazol-2-yl N-oxide, thiadiazolyl, such as 1 ,3,4-thiadiazol-5-yl and l,2,4-thiadiazol-5-yl, oxazolyl, such as, oxazol-2- yl, and oxadiazolyK such as l,3,4-oxadiazol-5-yl, and l,2,4-oxadiazol-5-yl. Particular 5- membered ring heterocycles containing 2 to 4 nitrogen atoms include imidazolyl, such as imidazol-2-yl; triazolyi, such as I ,3,4-triazol-5-yl, l,2,3-triazol~5-yl, and 1 ,2,4-triazol-5-yl, and tetrazolyl such as lH-tetrazol-5-yϊ. Particular benzo-fused 5-membered heterocycles are benzoxazol-2-yl, benzthiazol-2-yl, and benzimidazoi-2-yI. Particular 6-membered heterocycles contain one to three nitrogen atoms and, optionally, a sulfur or oxygen atom, for example pyridyl, such as, pyrid-2-yl, pyrid-3-yl, and pyrid-4-yI; pyrimidyl, such as, pyrimid-2-yl and pyrimid-4-yl; triazinyl, such as, l,3,4-triazin-2-yl and l,3,5-triazin-4-yl; pyridazinyl, such as. pyridazin-3-yl, and pyrazinyl. Substituents for optionally substituted heterocycles, and further examples of the 5- and 6-membered ring systems discussed above, can be found in U. S. Patent No. 4,278, 793 to W. Druckheimer et al.
[0040] "Heteroaryl" alone and when used as a moiety in a complex group such as a heteroarylalkyl group, refers to any mono-, bi-, or tri-cyciic aromatic ring system having the number of atoms designated where at least one ring is a 5-, 6- or 7-membered ring containing from one to four heteroatoms selected from the group nitrogen, oxygen, and sulfur (see Lang's Handbook of Chemistry, supra). Included in the definition are any bicyclic groups where any of the above heteroaryl rings are fused to a benzene ring. The following ring systems are examples of the heteroaryl (whether substituted or unsubstituted) group denoted by the term "heteroaryl": thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, tetrazolyi, thiatriazolyl, oxatriazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, thiazinyl, oxazinyl, triazinyl, thiadiazinyl, oxadiazinyl, dithiazinyl, dioxazinyl, oxathiazinyl, tetrazinyl, thiatriazinyl, oxatriazinyl, dithiadiazinyl, imidazolinyl, dihydropyrimidyl, tetrahydropyrimidyi, tetrazolo[l ,5-b]pyridazinyl and purinyl, as well as benzo-fused derivatives, for example, benzoxazolyl, benzofuryl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl, benzoimidazolyl, and indolyl. Particular "heteroaryls" include: 1 ,3-thiazol-2-yl, 4-(carboxymethyl)-5-methyl, l ,3-thiazoI-2-yl, 4-(carboxymethyl)-5- methyl-l ,3-thiazol-2-yl sodium salt, l,2,4-thiadiazol-5-yl, 3-mefhyl-l ,2,4-thiadiazol-5-yl, 1 ,3,4- triazol-5-yl, 2-methyl- l,3,4-triazol-5-yl, 2-hydroxy-l,3,4-triazol-5-yl. 2-carboxy-4-methyl- 1,3,4- triazol-5-yl sodium salt, 2-carboxy-4-methyl-l,3,4-triazol-5-yl, l,3-oxazol-2-yI, 1 ,3,4-oxadiazol- 5-yl, 2-methyl-l ,3,4-oxadiazol-5-yl, 2-(hydroxymethyl)-l,3,4-oxadiazoI-5-yl, l,2,4-oxadiazol-5- yl, l,3,4-thiadiazoi-5-yl, 2-thiol- l,3,4-thiadiazol-5-yl, 2-(methylthio)-l,3,4-thiadiazol-5-yl, 2- amino-l ,3,4-thiadiazol-5-yl, lH-tetrazol-5-yl, 1 -methyl- l H-tetrazol-5-yl, 1-(1-
(dimethylamino)eth-2-yl)-l H-tetrazol-5-yI, l-(carboxymethyl)-lH-tetrazoI-5-yl, 1-
(carboxymethyI)-lH-tetrazol-5-yl sodium salt, l~(methylsulfonic acid)-lH-tetrazoI-5-yl, 1- (methylsulfonic acid)-IH-tetrazol-5-yl sodium salt, 2-methyl-lH-tetrazol-5-y], l,2,3-triazol-5-yl, l-methyM ,2,3-triazol-5-yl, 2-methyl- l ,2,3-triazol-5-yl, 4-methyl-l,2,3-triazoI-5-yl, ρyrid-2-yl N-oxide, 6-methoxy-2-(n-oxide)-pyridaz-3-yi, 6-hydroxypyridaz-3-yl, l-methylpyrid-2-yl, 1- methylpyrid-4-yl, 2-hydroxypyrimid-4~yl, l ,4,5,6-tetrahydro-5, 6-dioxo-4-methyl-as-triazin-3-yI, 1 ,4,5,6-tetrahydro-4-(formylmethyl)-5, 6-dioxo-as~triazin-3-yl, 2,5-dihydro-5-oxo-6-hydroxy- astriazin-3-yl, 2,5-dihydro-5-oxo-ό-hydroxy-as-triazin-3-yl sodium salt, 2,5-dihydro-5-oxo-6- hydroxy-2-methyl-astriazin-3-y] sodium salt, 2,5-dihydro-5-oxo-ό-hydroxy-2-methyl-as-triazin- 3-yl, 2,5-dihydro-5-oxo-6-methoxy-2-methyl-as-triazin-3-yl, 2,5-dihydro-5-oxo-as-triazin-3-yl, 2,5-dihydro-5-oxo-2-methyl-as-triazin-3-yi, 2,5-dihydro-5-oxo-2, 6-dimethyl-as-triazin-3-yl, tetrazolo[ l ,5-b]pyridazin-6-y! and 8-aminotetrazolof l,5-b]-pyridazin-6-yl. An alternative group of "heteroaryl" includes: 4-(carboxymethyI)-5-methyl-l, 3-thiazoi-2-yl, 4-(carboxymefhyl)-5- methyl-I ,3-thiazol-2-yl sodium salt, l ,3,4-triazol-5-yl, 2-methyl-l ,3,4-triazol-5-yl, I H-tetrazol- 5-yl, 1 -methyl- lH-tetrazol-5-yl, l-(l-(dimethy]amino)eth-2-yl)-lH-tetrazol-5-yi, 1- (carboxymethyl)-lH-tetrazol-5-yl, l-(carboxymethyl)-lH-tetrazol-5-yl sodium salt, 1- (methylsulfonic acid)-lH- tetrazol-5-yl, l-(methylsulfonic acid)-lH-tetrazol-5-yl sodium salt, l,2,3-triazol-5-yl, 1 ,4,5,6-tetrahydro-5,6-dioxo-4-methyϊ-as-triazin-3-yl, 1,4,5, 6-tetrahydro-4-(2- formylmethyl)-5, 6-dioxo~as-triazin-3~yl, 2,5-dihydro-5-oxo-6-hydroxy-2-methyl-as-triazin-3-yl sodium salt, 2,5-dihydro-5-oxo~6-hydroxy-2-methy]-as-triazin-3-yl, tetrazolo[l ,5-b]pyridazin-6- yl, and 8-aminotetrazolo[l ,5-b]pyridazin-6-yl.
[0041] A "linker" is a bond or linking group whereby two chemical moieties, such as, monomers of an active compound, are directly covalently linked to one another or are indirectly linked via a third chemical moiety to form a homo- or heterodimer. The compounds set forth herein may include a single linker linking the two chemical moieties, or more than one linker linking the two chemical moieties at one or more position independently on each of the two chemical moieties. A "linker" (L, Lj or L2) may be a single or double covalent bond or a branched or unbranched, substituted or unsubstituted, hydrocarbon chain of 1 to about 100 atoms, typically, 1 to about 20 atoms, having a molecular weight up to about 500 MW. For example, a linker can be a bond, alkylene, alkenylene, alkynylene, cycloalkylene, cycloalkylalkyiene, heterocycloalkylene, heterocycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, or heteroarylalkylene, or an optionally-substituted alkylene, alkenylene, alkynyiene cycloalkylene, cycloalkylalkyiene, heterocycloalkylene, heterocycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, or heteroarylalkylene, of 2 to 12 atoms where one or more carbon atoms can be replaced with N, O, or S or an amino, substituted amino, oxygen atom, sulfide (-S-), sulfoxide (-SO-), sulfone (-SO2-), or disulfide (- SS-) group. Illustrative linkers and linking groups are described in U.S. Patent Publication No. 20050197403, as well as in U.S. Patent Application Serial Number 1 1/363,387, filed February 27, 2006, both of which are incorporated herein by reference as though fully set forth. For example, particular "linkers" include, but are not limited to, -CH2CH2-, -CH2CHiCH2-, - CH-CH-, 1 ,4-phcnyl, 2,5-thiopheny], -CH(OH)CH(OH)-, -CH2CH-O-CHCH2-, and - CH2OCOCCH2-.
[0042] The term "homodimer" as used herein refers to a compound composed of two covalently bound monomeric units of a chemical moiety wherein the monomelic units are identical.
{0043] The term "heterodimer" as used herein refers to a compound composed of two covalently bound monomeric units of a chemical moiety wherein the monomeric units are different. For example, one monomeric unit of a heterodimer may include a substituent that is different from the other monomeric unit at one or more position.
JΘ044] "Inhibitor" means a compound which reduces or prevents a particular interaction or reaction. For example, the binding of IAP proteins to caspase proteins reduces or prevents the inhibition of apoptosis by an IAP protein.
[0045] "Pharmaceutically acceptable salts" include both acid and base addition salts. "Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid, and the like. Organic acids may be selected from aliphatic, cycloaliphatic, aromatic, arylaliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids, such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, p-toiuenesulfonic acid, salicyclic acid, and the like.
[0046] The present invention is generally directed to Smac peptidomimetics (herein referred to as "'Srnac mimetics" or "a Smac mimetic") and the uses of Smac mimetics. One embodiment of the invention is a therapeutic composition including a Smac mimetic. In another embodiment, Smac mimetics act as chemopotentiating or chemotherapeutic agents. The term "chemopotentiating agent" as used herein refers to an agent that acts to increase the sensitivity of an organism, tissue, or cell to a chemical compound, or treatment namely "chemotherapeutic agents" or "chemo drugs" or radiation treatment. Therefore, a further embodiment of the invention is the therapeutic composition of a Smac mimetic, which acts as a chemopotentiating agent, and a biological agent, chemotherapeutic agent or radiation. Another embodiment of the invention is a method of inhibiting tumor growth in vivo by administering a Smac mimetic. Yet another embodiment is a method of inhibiting tumor growth in vivo by administering a Smac mimetic and a biologic agent, chemotherapeutic agent or radiation. Still another embodiment of the invention is a method of treating an individual, such as, for example, patient with cancer, by administering Smac mimetics of the present invention alone, or in combination with, a biological agent, chemotherapeutic agent or radiation.
(0047] In various embodiments of the invention, in situ cells or pathogenic cells, in an individual, may be treated with a Smac mimetic or a Smac mimietic in combination with a secondary agent, such as, a biological agent, chemotherapeutic agent or radiation, In such embodiments, the contacting step is affected by administering a pharmaceutical composition including a therapeutically effective amount of the Smac mimetic, wherein the individual may be subject to concurrent or antecedent radiation or chemotherapy for treatment of a neoproliferative pathology. Pathogenic cells may be of a tumor such as, but not limited to, bladder cancer, breast cancer, prostate cancer, lung cancer, pancreatic cancer, gastric cancer, colon cancer, ovarian cancer, renal cancer, hepatoma, melanoma, lymphoma, sarcoma, and combinations thereof.
JΘ048] In addition to apoptosis defects found in tumors, defects in the ability to eliminate seif-reactive cells of the immune system due to apoptosis resistance may be considered to play a key role in the pathogenesis of autoimmune diseases. Autoimmune diseases are characterized in that the cells of the immune system produce antibodies against its own organs and molecules or directly attack tissues resulting in the destruction of these tissues. Failure of these self-reactive cells to undergo apoptosis leads to the manifestation of the disease. Defects in apoptosis regulation have been identified in autoimmune diseases such as systemic lupus erthematosus, or rheumatoid arthritis,
(0049J In some embodiments of the invention, pathogenic cells may be those cells effected by an autoimmune disease or any disease whose symptoms include production of cells that are resistant to apoptosis. In at least one embodiment, affected cells are resistant to apoptosis due to the expression or overexpression of members of the Bcl-2 family of caspases. Examples of such autoimmune diseases include, but are not limited to, collagen diseases, such as, rheumatoid arthritis, systemic lupus erythematosus. Sharp's syndrome, CREST syndrome, calcinosis, Raynaud's syndrome, esophageal dysmotility, telangiectasia, dermatomyositis, vasculitis (Morbus Wegener's), and Sjogren's syndrome; renal diseases, such as, Goodpasture's syndrome, rapidly-progressing glomerulonephritis, and membrano-proliferative glomerulonephritis type II; endocrine diseases, such as, type-I diabetes, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), autoimmune parathyroidism, pernicious anemia, gonad insufficiency, idiopathic Morbus Addison's, hyperthyreosis, Hashimoto's thyroiditis, and primary myxedema; skin diseases, such as, pemphigus vulgaris, bullous pemphigoid, herpes gestationis, epidermolysis bullosa, and erythema multiforme major; liver diseases, such as, primary biliary cirrhosis, autoimmune cholangitis, autoimmune hepatitis type-1, autoimmune hepatitis type-2, primary sclerosing cholangitis; neuronal diseases, such as, multiple sclerosis, myasthenia gravis, myasthenic Lambert-Eaton syndrome, acquired neuromyotony, Guillain-Barre syndrome (Mϋller-Fischer syndrome), stiff-man syndrome, cerebellar degeneration, ataxia, opsoklonus, sensoric neuropathy, and achalasia; blood diseases, such as, autoimmune hemolytic anemia and idiopathic thrombocytopenic purpura (Morbus Werlhof); and infectious diseases with associated autoimmune reactions, such as, AIDS, Malaria, and Chagas disease.
[0050] It has been demonstrated in accordance with the present invention that the IAP- binding peptides or mimetics, thereof, are capable of potentiating apoptosis of cells. The mimetics described herein are suitably small, and since structural features in relation to the IAP binding groove are well-characterized, a wide variety of mimetic compounds may be synthesized. Mimetics of the core IAP-binding portions are preferred. Added advantages of compounds of this size include improved solubility in aqueous solution and ease of delivery to selected targets in vivo.
[0051 j The following compounds are illustrative of IAP-binding compounds that may be prepared as dimers and dimers of these IAP-binding compounds. Thus, various embodiments of the invention include these dimers. Such dimers can be prepared using any synthetic technique available to persons of ordinary skill in the art, such as, for example, the dimeric Smac peptidomimetics disclosed in U.S. Patent Application Serial Number 1 1/363,387, filed 2/27/2006, which provides guidance on preparation of the dimers of the instant invention. [0052] Various embodiments of the invention also include homodimers and heterodimer of monomeric units of general formula (1):
Figure imgf000039_0001
wherein: each R1 is, independently, H; CYtYalkyl;
Figure imgf000039_0002
C,-C4-alkynyl or C3-CiO- cycloalkyl which are unsubstituted or substituted; each R2 is, independently, II; CrC4-alkyl; Ci-C4-alkenyI; C,-C4-alkynyl or C3-C10- cycioalkyl which are unsubstituted or substituted; each R3 is, independently, H; -CF3; -C2H5; C|-C4-alkyl; C|-C4-alkenyl; Ci-d-alkynyl; - CH2-Z or any R2 and R3 together form a heterocyclic ring; each Z is, independently, II; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F Or -CH2OH; each R4 is, independently, C]-Ci6 straight or branched alkyl; Ci-Ci6-alkenyl; Ci-C16- alkynyl; C3-C]o-cycloalkyl; -(CH2)].6-Zι ; -(CH2)o-ό~aryl; and -(Cϊl2)0-6-het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each Zi is, independently, -N(R10)-C(0)-C|.io-aIkyl; -N(R,0)-C(O)-(CH2)1-6-C3.r cycloalkyl; -N(R,0)-C(O)-(CH2Wphenyl; -N(Ri0)-C(O)-(CH2)1 -6-het; -C(O)-N(Rn)(R12); - C(0)-0-C,.,o-alkyl: -C(O)-O-(CH2), ^C3-7-cycloalkyl; -C(O)-O-(CH2)0-6-phenyl; -C(O)-O- (CH2), -6-het; -O-C(O)-CM0-alkyl; -O-C(OMCH2)ι-6-C3.7-cycloalkyl; -0-C(0)-(CH2)o^-phenyI; - 0-C(O)-(CH2), -6-het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1-4 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom; each R10 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each Rn and R12 is, independently, H; Cu-alkyl; C3.7-cycIoalkyl; -(CH2)I-6-C3-7- cycloalkyl; (CH2)o-6-ρhenyl; wherein alkyl, cycloaikyi, and phenyl are unsubstituted or substituted; or Rn and R12 together with the nitrogen form het; each R5 is, independently, H; C|.jO-alkyl; aryl; phenyl; C3,7-cycloalkyl; -(CH2) 1-0-C3.7- cycloalkyl; -CM 0-alkyl-aryl; -(CH2)c-6-C>7-cydoalkyi-(CΗ2)o^-phenyI; -(CH2)o-4-CH[(CH2)M- phenyij2; indanyi; -C(O)-CM0-alkyl; -C(O)-(CH2)1-ή-C3-7-cyc]oalkyl; -C(O)-(CH2)0.6-phenyl; - (CH2)0-6-C(0)-phenyl; -(CHi)o-ό-het; -C(O)-(CH2) ι-6-het; or R5 is a residue of an amino acid, wherein the alkyl, cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; each U is, independently, as shown in structure (II):
Figure imgf000040_0001
wherein: each n is, independently, O to 5; each X is, independently, -CH or N; each Ra and Rj, is, independently, an O, S, or N atom or Co-s-alkyl wherein one or more of the carbon atoms in the alkyl chain are optionally replaced by a heteroatom selected from O, S, or N, and where each alkyl is, independently, either unsubstituted or substituted; each Rd is, independently, selected from: R6-Q-(Rf)P(RgV and ATrD-Ar2; each Rc is, independently, H or any R^ and R4 together form a cycloalkyl or het; where if Rj and R0 form a cycloalkyl or het, Rs is attached to the formed ring at a C or N atom; each p and q is, independently, O or 1 ; each Rt is, independently, Ci-s-alkyl or alkylidene, and each R- is either unsubstituted or substituted; each Q is, independently, N, O, S, S(O), or S(O)2; each Ari and Ar2 is, independently, substituted or unsubstituted aryl or het; each Rf and Rg is, independently, H; -Cno-alkyl; Cno-alkylaryl; -OH; -O-Ci.io-alkyl; - (CHa)O-O-C3.7-cycloalkyl; -0-(CH2)o-ό-aryl; phenyl; aryl; phenyl-phenyl;
Figure imgf000040_0002
-O-(CH2)|. 6-het; -OR13; -C(O)-R13; -C(O)-N(R13)(R14); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2-R13; -S(O)2- NR13R14; -NR13-S(O)2-R14; -S-CM0-alkyl; aryl-CM-alkyl; or het-CM~alkyl wherein alkyl, cycloaϊkyl, het, and aryl are unsubstituted or substituted;
Figure imgf000041_0001
-SO2-C 1.2-alkylphenyl; -O-Ci-4-alkyh or any Rg and Rf together form a ring selected from het or aryl; each D is, independently, -CO-; -C(O)-C ]_7-alkylene or arylene; -CF2-; -0-; -S(O)r where r is 0-2; 1 ,3-dioxalane; or C|.7-alkyl-OH; where alkyl, alkylene, or arylene are unsubstituted or substituted with one or more halogens, OH, -O-C[.6-alkyl, -S-Ci-6-alkyl; or -CF3; or each D is, independently. N(Rh) wherein each Rh is, independently, H; unsubstituted or substituted Ci-7-alkyl; aryl; unsubstituted or substituted -O-(C[-7-cycloalkyl); -C(O)-C Mo-alky!; - C(0)-C0-1o-alky]-aryI; -C-O-C M0-alkyl; -C-O-Co-io-alkyl-aryl; -SO2-C, -10-alkyl; or -SO2-(C0-Kr aikylaryl); each R^, R7, R8, and Rg is, independently, H, -C|.10-alkyl; -Q-io-alkoxy; aryl-C 1.io- alkoxy; -OH; -O-CM0-alkyl; -(CH2)0-6-C3.7-cycloalkyl; -0-(CH2)0-6-aryl; phenyl; -(CH2) ,^-het; - O-(CH2)!-6-het; -OR13; -C(O)-R,3; -C(O)-N(R13)(R14); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2-
R13; -S(O)2-NR13R14; or -NR13-S(O)2-R14; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; and any R^, R7, R5, and R9 optionally together form a ring system; each R13 and Rf4 is, independently, H; C1- 10-alkyl; -(CH2)0-6-C3.7-cycloalkyl; -(CH2V6- (CH)o.,-(aryl)ι-2; -C(O)-C1-10-aIkyl; -C(O)-(CH2) ^-C^-cycIoalkyl; -C(O)-O-(CH2)0-6-aiyl; - C(0)-(CH2)o.6-0-fluorenyl; -C(O)-NH-(CH2)0.6-aryl; -C(O)-(C H2 Waryl; -C(0)-(CH2)o-6-het; - C(S)-CM 0-alkyl; -C(SHCH2)^-C3-7-cycloalkyl; -C(S)-O-(CH2)0.6-aryl; -C(SHCH2)0-6-O- fluorenyl; -C(S)-NH-(CH2)o^-aiyl; -C(S)-(CH2)o-6-aryl; or -C(S)-(CH2) 1 -6-het; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; or any Rt3 and R14 together with a nitrogen atom form het; wherein alkyl substituents of R;3 and R14 are unsubstituted or substituted and when substituted are substituted by one or more substituents selected from Ci-io-alkyl, halogen, OH,- O-Cι-6-alkyl, -S-C|.6-alkyl, and -CF3; and substituted phenyl or aryl of Ri3 and Ru are substituted by one or more substituents selected from halogen, hydroxyl, C^-alkyl, Cι-4-alkoxy, nitro, -CN, -0-C(O)-C M-alkyl, and -C(O)-O-C M-aryI; and pharmaceutically acceptable salts and hydrates thereof.
[0053] In some embodiments, compounds of the invention are of general formula (III):
Figure imgf000042_0001
wherein:
R1 and Rf are each, independently, H; C]-C4-alkyl; C]-C4-alkenyl; C|-C4-alkynyl; or C3- C 1o-cycloalkyl which are unsubstituted or substituted; R2 and R2 " are each, independently, H; C|-C4-alkyl; Ci-C4-aIkenyl; C|-C4-alkynyl; or C3- Cιo-cycloalkyl which are unsubstituted or substituted; R3 and R3' are each, independently, H; -CF3; -C2H5; C3-C4-a!kyl; C|-C4-alkenyl; CrC4- alkynyl; or -CH2-Z or R2 and R3 together or independently with R2' and R3' form a heterocyclic ring; each Z is, independently. H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; or ^CH2OH;
R4 and R4' are each, independently, C1-C16 straight or branched alkyl; CrC -alkenyl; Q- Ci6-alkynyl; or C3-Cιo-cycloalkyl;
Figure imgf000042_0002
; -(CH2)0-6-aryl; or -{CH2)0-6-het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each Z1 is, independently, -N(R!0)-C(O)-CM0-alkyl; -N(R So)-C(O)-(CH2)! ^-C3-7- cycloalkyl; -N(R,o)-C(0)-(CH2)0*-phenyl; -N(R,0)-C(O)-(CH2)ι^het; -C(O)-N(R1 1)(R12); - C(O)-O-CM0-alkyl; -C(O>O-(CH2)1-6-C3-7-cycloalkyl; -C(O)-O-(CH2)0-6-phenyl; -C(O)-O- (CH2)t-6-het; -0-C(O)-C i-10-alkyl; -O-C(O)-(CH2)^-C3.7-cycloalkyl; -O-C(O)-(CH2)(w-phenyl; or -0-C(O)-(CH2) i^-het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1-4 heteroatoms selected from N, O, and S. or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S. which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom; each R1o is, independently, H; -CH3; -CF3; -CH2OH; or -ClI2Cl; each Rj 1 and Rj2 is, independently, H; C^-alkyl; C3_7-cycloalkyl; -(CH2) s-6-C3.7- cycloalkyl; or (CH2)o^-phenyl; wherein alky!, cycloalkyl, and phenyl are unsubstituted or substituted; or Rn and R12 together with a nitrogen form het; R5 and IV are each, independently, H; C|.io-alkyl; aryl; phenyl; C3-7-cycIoalkyi; -(CH2)I- 6-C3.7-cycloa]kyl; -Cι-fo-alkyl-aryl; -(CH2)α^-C3-7-cycloalkyl-(CH2)o.<j-phenyl; -(CH2)<M- CHf(CH2), -»-phenylJ2; indanyl; -C(O)-C M 0-alkyl; -C(O)-(CH2)ι.6-C3.7-cycIoalkyl; -C(O)-(CH2)*- 6-phenyl; -(CH2)o-6-C(0)-ρhenyl; -(CH2)0-6-het; or -C(O)-(CH2) ι-6-het; wherein the alkyl, cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; or R5 and R5' are, independently, an amino acid residue;
U and U' are each as shown in structure (II):
Figure imgf000043_0001
wherein: each X is, independently, -CH or N; each Ra and Rb is, independently, an O, S, or N atom or Co-g-alkyl wherein one or more of the carbon atoms in the alkyl chain are optionally replaced by a heteroatom selected from O, S, or N, and where each alkyl is, independently, either unsubstituted or substituted; each Rd is, independently, selected from:
Figure imgf000043_0002
ArrD-Ar2; each Rc is, independently, H or any R0 and Rd together form a cycloalkyl or het; where if R(i and R0 form a cycloalkyl or het, R5 is attached to the formed ring at a C or N atom; each p and q is, independently, O or 1 ; each R6 is, independently, C^-alkyl or alkylidene, and each Re is either unsubstituted or substituted; each Q is, independently, N, O, S7 S(O), or S(O)2; each An and Ar2 is, independently, substituted or unsubstituted aryl or het; each Rr and Rg is, independently, H; -C 1.io-alkyl; C|.ιo-alkylaryl; -OH; -O-C].|0-alkyl; - (CH2)o-6-C3.7-cycloalkyl; -O-(CH2)0-6-aryI; phenyl; aryl; phenyl-phenyl;
Figure imgf000043_0003
-O-(CH2)ι. 6-het; -OR13; -C(O)-R13; -C(O)-N(RB)(R14); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2-R13; -S(O)2- NRJ3RH; -NRJ3-S(O)2-RH; -S-CVio-alkyl; aryl-C^-alkyl; or het-CM-alkyl wherein alkyl, cycioalkyl, het, and aryl are unsubstituted or substituted; -SO2-C ]-2-alkyl; -SO2-C 1-2-alkyiphenyI; or -O-CM-alkyl; or any Rg and R[ together form a ring selected from het or aryl; each D is, independently, -CO-; -C(O)-C 1.7-alkylene or arylene; -CF2-; -0-; -S(O)1- where r is 0-2; 1 ,3-dioxalane; or C].7-alkyl-OH; where alkyl, alkylene, or arylene are unsubstituted or substituted with one or more halogens, OH, -O-C|-6-alkyl, -S-Ci-6-alkyl; or -CF3; or each D is, independently, N(Rh) wherein each Rh is, independently, H; unsubstituted or substituted C1.7-alk.yl; aryl; unsubstituted or substituted -O-(C|.7-cycloalkyl); -C(O)-C Mo-alkyl; - C(0)-Co-ιo-alkyl-aiyI; -C-O-C M 0-alky!; -C-O-Co-io-alkyl-aryl; or -SO2-CM0-alkyl; or -SO2-(C0. 10-alkylaryl); each R6, R7, Rg, and R9 is, independently, H, -C 1.io-alkyl; -C|.,o-alkoxy; aryl-C 1.io- alkoxy; -OH; -O-CM0-alkyl; -(CH2)0-6-C3.7-cycloaIkyl; -0-(CH2)0-6-aiyl; phenyl; -(CH2) 1 -6-het; - O-(CH2)i-6-het; -0R!3; -C(O)-R13; -C(O)-N(R13)(R14); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2- R13; -S(O)2-NR]3R14; or -NR13-S(O)2-R14; wherein each alkyl, cycioalkyl, and aryl is unsubstituted or substituted; and any R6, R7, R8, and R9 optionally together form a ring system; each R13 and R]4 is, independently, H; C^o-alkyl; -(CH2)o-6-C3-7-cycloalkyl; -(CH2)0-6- (CH)0-ι-(aryl)1-2; -C(O)-CM0-alkyI; -C(O)-(CH2)i-6-C3-7-cycloalkyl; -C(0)-0-(CH2)o^-aryl; - C(O)-(CH2)0.6-O-fluorenyl; -C(O)-NH-(CH2)0-6-aryl; -C(O)-(CH2)0-6-aryl; -C(O)-(CH2)0.6-het; - C(S)-C, -lo-alkyl; -C(S)-(CH2),.6-C3.7-cycloalkyl; -C(S)-O-(CH2)0-6-aryI; -C(S)-(CH2WO- fluorenyl; -C(S)-NH-(CH2)o.6-aryl; -C(S)-(CH2)0.6-aτyl; or -C(S)-(CH2)1-6-het; wherein each alkyl, cycioalkyl, and aryl is unsubstituted or substituted; or any R13 and R14 together with a nitrogen atom form het; wherein alkyl subsituents of Rj3 and Rj4 are unsubstituted or substituted and when substituted are substituted by one or more substituents selected from Cμio-alkyl, halogen, OH,- O-Ci^-alkyl, -S-C^-alkyl, and -CF3; and substituted phenyl or aryl of R13 and R]4 are substituted by one or more substituents selected from halogen, hydroxyl, CM-alkyl, C^-alkoxy, nitro, -CN, -0-C(O)-C M-alkyl, and -C(O)-O-CM-aryl;
L is one or more linkers covalently linking one or more of the positions R4, R5, or U, with R4 1, R5', or U'; and pharmaceutically acceptable salts and hydrates thereof.
[0054] In certain embodiments, the compounds of the invention are of a formula selected from compounds of formula (IV):
Figure imgf000045_0001
wherein
R1,
Figure imgf000045_0002
R3, R3'. R4, Rt', R5, R5', U, and U' are defined as described above; and
L1 and L2, independently, link position R4 with R4' and R5 with R5'; or pharmaceutical iy acceptable salts thereof; compounds of formula (V):
Figure imgf000045_0003
wherein
R1, R1', R2, R2', R3, R3', R4- R4', R5, R5', U, and U' are defined as described above; and L links position R4 with R4'; or pharmaceutically acceptable salts thereof; and a compound of formula (VI):
Figure imgf000046_0001
wherein
R1, R1', R2, R2', R3, R3', R45 R4'. Rs, Rs', V, and U" are defined as described above; and L links position R5 with R5'; or pharmaceutically acceptable salts thereof.
[0055] Other embodiments of the invention includes compounds having a homodimcr or heterodimer with monomeric units of general formula (XlII):
wherein: each Rf is, independently, H; each R2 is, independently, H or Ci-C4~alkyl which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH3, -SCH3, -CN, -SCN, and nitro; each R3 is, independently, H; -CF3; -C2F5; -CH2-Z or any R2 and R3 together with the nitrogen form a C3-C6 heteroaliphatic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; Or -CH2OH; each R4 is. independently, C)-Ci6 straight chain alkyl; C3-C 1O branched chain alkyl; - (CH2)o-6-C3-C7-cycloalkyl; -(CH2)^-Z1 ; -(CH2)0.6-phenyI; or -{CH2)0-6-het; wherein each alkyl. cycloalkyl, and phenyl are unsubstituted or substituted; each Z, is, independently, -N(R9)-C(O)-C|.i 0-alkyl; -N(R9)-C(O)-(CH2)!-6-C3-7- cycloaikyl; -N(R9)-C(0)-(CH2)o^-phenyl; -N(R9)-C(O)-(CH2)!.6-het; -C(O)-N(Rj0XR11); -C(O)- O-Ci-io-alkyϊ; -C(O)-O-(CH2), ^C3.7-cycloaIkyl; -C(0)-0-(CH2)0-6-phenyl; -C(O)-O-(CH2),^- hct; -O-C(O)-CM0-alkyi; -O-C(O)-(CH2)ι^-C3.7-cycloalkyl; -O-C(O)-(CH2y6-phenyl; or -O- C(O)-(CHa) i H5-het; wherein each alkyl, cycloalkyl, and phenyl are unsubstitυted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, O, and S, or an 8- 12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted, and when substituted is substituted on a carbon atom by halogen, hydroxyl, C^-alkyl, CM-aϊkoxy, nitro, -0-C(O)-Ci- 4-alkyl, -€(O)-O-CM-alkyl, or on a nitrogen by CM-alkyl, -0-C(O)-C M-alkyl, or -C(O)-O-C M- alkyl; each R9 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each R1o and Rn is, independently, H; C M-alkyl; C3-7-cycloalkyl; -(CH2)^-C3.?- cycloalkyl; or (CH2)0-6-phenyl; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; or any R1o and R1 j together with a nitrogen form het; each X is, independently, CH or N; each R5 is, independently H; C(-1o-alkyl; aryl; phenyl; C3.7-cycloaIkyl; -(CH2) i-6-C3.7- cycioalkyl; -CM 0-aϊkyi-aryl; -(CH2)o^C3-7-cycloalkyl-(CH2Wphenyl; -(CH2)0-4-CH[(CH2)M- phenyl]2; -(CH2)0-6-CH(phenyl)2; -C(O)-CM0-alkyl;
Figure imgf000047_0001
-C(O)- (CH2)o-6-phenyl; -(CH2)0-6-C(0)-phenyl; -(CH2)!-6-het; -C(O)-(CH2)i-6-het; or R5 an amino acid residue, wherein the alkyl, cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; each R63 is, independently, H, methyl, ethyl, -CF3, -CH2OH or -CH2Cl; or each any R5 and R6a together with a nitrogen form a het; each Ra and R8 is independently, cis relative to acyl substituents at the one position of the ring and are each, independently, H, -CM0-alkyl; -OH; -O-Ci-io-alkyl; -(CH2)0-6-C3.7-cycloalkyl; -0-(CH2)o.6-aryl: phenyl; -(CH2),.6-het; -O-(CH2),.6-het; -N(R12)(R13); -S-R12; -S(O)-R12; -S(O)2-
R12; or -S(O)2-NRJ2R13; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; each R|2 and R!3 is, independently, H; CM0-alkyl; -(CH2)0-6-Cv7-cycloalkyl; -(CH2V6- (CH)0-ι-(aryl)|.2; -C(O>CM 0-alkyl; -C(OHCH2), -6-C3.7-cycloalkyl; -C(O)-O-(CH2)0-6-aryl; - C(0>(CH2)o-6-0-fluorenyI; -C(O>NH-(CH2)0-6-aryI; -C(OHCH2)0-6-aryI; or -C(OHCH2) ,^-het; wherein each alkyl cycloalkyl, or aryl is unsubstituted or substituted; or any R)2 and R13, together with a nitrogen atom form het; each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; each n is 0, 1 , or 2; and wherein substituted alkyl is substituted by one or more substituents selected from a double bond, halogen, OH7-O-C ι-6-alkyl, -S-Ci-e-alkyl, and -CF3; substituted cycloalkyl is substituted by one or more substituents selected from a double bond, C|.6-alky]s halogen, OH7-O-C ,^-alky I, -S-C|.6-alkyl, and -CF3; and substituted phenyl or aryl is substituted by one or more substituents selected from halogen, hydroxy, C-M-alkyl, CM-aϊkoxy, nitro, -CN. -0-C(O)-C M-alky 1, and -C(O)-O-CM- aryl; and pharmaceutically acceptable salts and hydrates thereof.
(0Θ56] Still other embodiments of the invention include compounds of formula (XIV):
Figure imgf000048_0001
wherein:
Figure imgf000048_0002
R2 and R2' are each, independently, H or C]-C4-alkyl which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH3, -SCH3, -CN, - SCN, and nitro; R3 and R3' are each, independently, H; -CF3; -C2F5; or -CH2-Z or R2 and R3 or R2' and R3' together with a nitrogen form a C3-C6 heteroaliphatic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; or -CH2OH;
R4 and RV are each, independently, Cf-C16 straight chain alkyl; C3-CiO branched chain alkyl; -(CHiJo-ό-Cj-CT-cycloalkyl; -(CH2)I -6-Zi; -(CH2)o-ό-phenyl; or -(CH2)o-ό-het; wherein each alkyl, cycloaikyl, and phenyl is unsubstituted or substituted; each Z, is, independently, -N(R9)-C(O)-CM()-alkyl; -N(Rg)-C(O)-(CH2)S-6-C3.?- cycloalkyl; -N(R9)-C(0)-(CH2)o-6-phenyl; -N(R9)-C(O)-(CH2)i-6-het; -C(O)-N(R10)(R] 1); -C(O)- O-C 1.io-alkyl; -C(O)-O-(CH2),.6-C3.7-cycloalkyl; -C(O)-O-(CH2 Wphenyϊ; -C(O)-O-(CH2)i-6- het; -0-C(O)-C ,.,0-alkyl; -0-C(O)-(CH2) i-*,-C3.7-cycloalkyl; -0-C(0)-(CH2)o^-phenyi; or -O- C(O)-(CH2)).6-het; wherein each alkyl, cycioalkyl, and phenyl is unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S. which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon atom by halogen, hydroxyl, Cι-4-alkyl, C^-alkoxy, nitro, -0-C(O)-C M-alkyl, -C(O)-O-C M-alkyl, or on a nitrogen by CM-alkyl, -0-C(O)-C M-alkyl, or -C(O)-O-C M-alkyl; each R9 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each R1o and Rn is, independently, H; C M-alkyl; C3-7-cycloalkyl; -(CH2)I-O-C3-7- cycloalkyl; or (CH2)o-6-phenyl; wherein each alkyl, cycioalkyl, and phenyl is unsubstituted or substituted; or any R1o and Rn together with a nitrogen form het; X and X' are each, independently, CH or N;
R5 and Rs" are each, independently, 11; d-io-alkyl; aryl; phenyl; C3-7-cycloalkyl; -(CH2)I- 6-C3-7-cycloalkyl; -C).|0-aikyl-aryl; -(CH2)o-6-C3-7-cyc]oalkyl-(CH2)o.6-phenyl; -(CH2)o-4- CH[(CH2)M-phenyl]2; -(CH2)o-6-CH(pheny!)2; -C(O)-C,.,0-alkyl; -C(O)-(CH2)1-6-C3-7-cycloalkyl; -C(O)-(CH2)0-6-phenyl; -(CH2)0.6-C(O)-phenyi; -(CH2)t^-het; or -C(O)-(CH2) ,^-het; or R5 is an amino acid residue, wherein each alkyl, cycioalkyl, phenyl, and aryl substituent is unsubstituted or substituted;
R63 and R63' are each, independently, H, methyl, ethyl, -CF3, -CH2OH ,or -CH2Cl; or R5 and R^ are, independently, or together with R5' and R63' together with a nitrogen are het;
R1J, Rg, Ra\ and R«' are cis relative to an acyl substituent at position one of a ring to which they are attached and are each, independently, H, -CMo-alkyl; -OH; -O-Ci.|0-alkyl; - (CH2)o.6-C3.7-cycloalkyJ; -0-(CH2Waryl; phenyl; -(CII2)ι-6-het; -O-(CH2)].6-het; -N(R12)(R13); - S-R!2, -S(O)-RI2; -S(O)2-RI2; or -S(O)2-NR12R]3; wherein each alkyl, cycioalkyl, and aryl is unsubstituted or substituted;
Rj2 and R!3 are each, independently, H; CYio-alkyl; -(CH2)o^-C3.7-cycioaikyl; -(CH2)o-ό- (CH)o-r(aryl)ι.2; -C(O)-CM0-alkyl; -C(O)-(CH2)N6-C3-7-cycloalkyl; -C(O)-O-(CH2)0^-aryl; - C(O)-(CH2 Vό-O-fluorenyl; -C(O)-NH-(CH2)0.6-aryl; -C(OHCH2)o^-aryI; or -C(O)-(CH2)i.6-het; wherein each alky!, cycioalky], and aryl is unsubstituted or substituted; or R|2 and R13 together with a nitrogen atom form het; each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; n and n" are each, independently, 0, 1, or 2; wherein each substituted alkyl is substituted by one or more substituents selected from a double bond,
Figure imgf000050_0001
and -CF3; each substituted cycloalkyl is substituted by one or more substituents selected from a double bond, Ci-6-alkyl, halogen, OH,-O-C|-6-alkyl, -S-Ci.<;-alkyl, and -CF3; and each substituted phenyl or aryl is substituted by one or more substituents selected from halogen, hydroxy, CM-alkyl, CM-alkoxy, nitro, -CN, -0-C(O)-C M-alkyl, and -C(O)-O-Ci-4- aryl; and
L is one or more linkers covalently linking one or more of the positions R4, R5, R8, with R4', R5', R8'; and pharmaceutically acceptable salts and hydrates thereof.
|0057) In some embodiments, the compounds of compounds of the invention may be selected from compounds of formula (XV):
Figure imgf000050_0002
wherein
R1, R1 ', R2, R2 ', R3, R3', R4, R4', R5, R5 ", R*a, R6a', R8, Ra', R8, R8', X, X', n, and n' are defined as described above; and
L| and L2, independently link position R4 with R4 1 and R5 with R5'; or apharmaceutically acceptable salts and hydrates thereof; compounds of formula (XVl):
Figure imgf000051_0001
wherein
R1, R 1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a', Ra, Ra', R8, R8', X, X', n. and n' are defined as described above; and
L1 and L2, independently link position R4 with R4 and R8 with R8'; or pharmaceutically acceptable salts and hydrates thereof; compounds of formula (XVII):
Figure imgf000051_0002
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a', Ra, Ra', R8, R8', X, X', n, and n' are defined as described above; and
L links position R4 with R4'; or pharmaceutically acceptable salts and hydrates thereof; compounds of formula (XVIII):
Figure imgf000052_0001
(XVIII) wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a' , Ra, Ra', R8, R8', X, X', n, and n' are defined as described above; and
L links position R5 with R5 "; or pharmaceutically acceptable salts and hydrates thereof; and compounds of formula (XIX):
Figure imgf000052_0002
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a' , Ra, Ra', R8, R8', X, X', n, and n' are defined as described above; and
L links position R8 with R8'; or pharmaceutically acceptable salts and hydrates thereof. [0058] Still other embodiments of the invention include compouds of formulae (VIl), (VlIl). (IX), (X), (Xl) and (XII):
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
[0059] Mimetic, specifically, peptidomimetic design strategies are readily available in the art and can be easily adapted for use in the present invention (see, e.g., Ripka & Rich, Curr. Op. Chem. Biol. 2, 441-452, 1998; Hruby et al., Curr. Op. Chem. Biol. 1 , 1 14-1 19, 1997; Hruby & Balse, Curr. Med, Chem. 9, 945-970, 2000). One class of mimetic mimics a backbone that is partially or completely non-peptide, but mimics the peptide backbone atom-for-atom and comprises side groups that likewise mimic the functionality of the side groups of the native amino acid residues. Several types of chemical bonds, e.g. ester, thioester, thioamide, retroamide, reduced carbonyl, dimethylene, and ketomethylene bonds, are known in the art to be generally useful substitutes for peptide bonds in the construction of protease-resistant peptidomimetics. Another class of peptidomimetics comprises a small non-peptide molecule that binds to another peptide or protein, but which is not necessarily a structural mimetic of the native peptide. Yet another class of peptidomimetics has arisen from combinatorial chemistry and the generation of massive chemical libraries. These generally comprise novei templates which, though structurally unrelated to the native peptide, possess necessary functional groups positioned on a non-peptide scaffold to serve as "topographical" mimetics of the original peptide (Ripka & Rich, 1998, supra).
(0060] In one embodiment, the Smac mimetics of the invention are modified to produce peptide mimetics by replacement of one or more naturally occurring side chains of the 20 genetically encoded amino acids, or D-amino acids, with other side chains, for instance with groups, such as, alkyl, lower alkyl, cyclic 4-, 5-, 6-, to 7-membered alkyl, amide, amide lower alkyl, amide di-(lower alkyl), lower alkoxy, hydroxy, carboxy, and the lower ester derivatives thereof, and with 4-, 5-, 6-, to 7-membered heterocycles. For example, proline analogs can be made in which the ring size of the proline residue is changed from 5 members to 4, 6, or 7 members. Cyclic groups can be saturated or unsaturated, and if unsaturated, can be aromatic or non-aromatic. Heterocyclic groups can contain one or more nitrogen, oxygen, and/or sulphur heteroatoms. Examples of such groups include furazanyl, imidazolidinyl, imidazolyl, imidazolinyl, isothiazolyl, isoxazolyl, morpholinyl (e.g. morpholino), oxazolyl, piperazinyl (e.g. 1 -piperazinyl), piperidyl (e.g. 1-piperidyl, piperidϊno), pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolidinyl (e.g. 1-pyrrolidinyl), pyrrolinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, thiomorpholiny! (e.g. thiomorpholino), and triazolyl. These heterocyclic groups can be substituted or unsubstituted. Where a group is substituted, the substituent can be alkyi, alkoxy, halogen, oxygen, or substituted or unsubstituted phenyl. Peptidomimetics may also have amino acid residues that have been chemically modified by phosphorylation, sulfonation, biotinylation, or the addition or removal of other moieties.
[0061] Pharmaceutical compositions The subject compositions encompass pharmaceutical compositions including a therapeutically effective amount of a Smac mimetic in dosage form and a pharmaceutically acceptable carrier, wherein the Smac mimetic inhibits the activity of an Inhibitor of Apoptosis protein (1ΛP), thus promoting apoptosis. In another embodiment, the compositions include a therapeutically effective amount of a Smac mimetic in dosage form and a pharmaceutically acceptable carrier in combination with a chemotherapeutic and/or radiotherapy, wherein the Smac mimetic inhibits the activity of an IAP, thus promoting apoptosis and enhancing the effectiveness of the chemotherapeutic and/or radiotherapy.
[0062] In an embodiment of the invention, a therapeutic composition for promoting apoptosis can be a therapeutically effective amount of a Smac mimetic which binds to at least one IAP. In one embodiment, the IAP can be XIAP, In another embodiment, the IAP can be ML-IAP, and in yet another embodiment, the IAP can be cIAP-1 or cIAP-2. fn further embodiments, the IAP can be multiple IAPs.
[0063] Embodiments of the invention also include methods for treating a patient having a condition characterized by inhibited apoptosis, wherein administration of a therapeutically effective amount of a Smac mimetic is delivered to the patient, and the Smac mimetic binds to at least one IAP, In one embodiment, the IAP can be XIAP. In another embodiment the IAP can be ML-IAP, and in still another embodiment, the IAP can be cIAP-1 or cIAP-2. In further embodiments, the IAP can be multiple IAPs.
|0064] In one embodiment of the invention, an additional chemotherapeutic agent (infra) or radiation may be administered prior to, along with, or following administration of the Smac mimetic. Exemplary chemotherapeutic agent, include, but are not limited to, alkylating agents, antimetabolites, anti-tumor antibiotics, taxanes, hormonal agents, monoclonal antibodies, glucocorticoids, mitotic inhibitors, topoisomerase I inhibitors, topoisomerase II inhibitors, immunomodulating agents, cellular growth factors, cytokines, and nonsteroidal antiinflammatory compounds.
[0065] In various embodiments, the Smac mimetics of the invention may be combined with a pharmaceutically acceptable carrier or excipient, and in some embodiments, the Smac mimetics of the invention may be combined with an additional chemotherapeutic agent and a pharmaceutically acceptable carrier or excipient. The term "pharmaceutically-acceptable carrier" as used herein means one or more compatible solid or liquid fillers, diluents or encapsulating substances which are suitable for administration into a human. The term "carrier" or "excipient" denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions are also capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
[0066} The delivery systems of the invention are designed to include time-released, delayed release or sustained release delivery systems such that the delivering of the Smac mimetic occurs prior to, and with sufficient time, to cause sensitization of the site to be treated. A Smac mimetic may be used in conjunction with radiation and/or additional anti-cancer chemical agents (infra). Such systems can avoid repeated administrations of the Smac mimetic compound, increasing convenience to the subject and the physician, and may be particularly suitable for certain compositions of the present invention.
[0067] Many types of release delivery systems are available and known to those of ordinary skill in the art. They include, but are not limited to, polymer base systems, such as, poly(lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides. Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Pat. No. 5,075,109. Delivery systems also include non-polymer systems including: lipids including sterols, such as cholesterol, cholesterol esters and fatty acids or neutral fats, such as mono-, di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like. Specific examples include, but are not limited to: (a) erosional systems in which the active compound is contained in a form within a matrix such as those described in U.S. Pat. Nos. 4,452,775, 4,667,014, 4,748,034, and 5,239,660 and (b) diffusional systems in which an active component permeates at a controlled rate from a polymer, such as described in U.S. Pat. Nos. 3,832,253, and 3,854,480. In addition, pump-based hardware delivery systems can be used, some of which are adapted for implantation.
|0068] Use of a long-term sustained release implant may be desirable. Long-term release is used herein, and means that the implant is constructed and arranged to deliver therapeutic levels of the active ingredient for at least about 30 days, and preferably about 60 days. Long-term sustained release implants are well-known to those of ordinary skill in the art and include some of the release systems described above.
{0069) The pharmaceutical compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier that constitutes one or more accessory ingredients. In general, the compositions may be prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
[0070] Compositions suitable for parenteral administration conveniently include a sterile aqueous preparation of a Smac mimetic which is preferably isotonic with the blood of the recipient. This aqueous preparation may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may aiso be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1 ,3-butanediol. Among the acceptable vehicles and soivents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. In addition, fatty acids, such as oleic acid, may be used in the preparation of injectables. Carrier formulation suitable for oral, subcutaneous, intravenous, intramuscular, etc. administrations can be found, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, PA which is incorporated herein in its entirety by reference thereto.
[0071] Administration of Smac peptidomimetics The Smac peptidomimetics of the invention may be administered in effective amounts. An effective amount is that amount of a preparation that alone, or together with further doses, produces the desired response. This may involve only slowing the progression of the disease temporarily, although it may involve halting the progression of the disease permanently or delaying the onset of or preventing the disease or condition from occurring. This can be monitored by routine methods known and practiced in the art. Generally, doses of active compounds may be from about 0.01 mg/kg per day to about 1000 mg/kg per day, and in some embodiments, the dosage may be from 50-500 mg/kg. In various embodiments, the compounds of the invention may be administered intravenously, intramuscularly, or intradermally, and in one or several administrations per day. The administration of the Smac peptidomimetic can occur simultaneous with, subsequent to, or prior to chemotherapy or radiation.
|0072] In general, clinical trials will determine specific ranges for optimal therapeutic effect for each therapeutic agent and each administrative protocol, and administration to specific patients will be adjusted to within effective and safe ranges depending on the patient condition and responsiveness to initial administrations. However, the ultimate administration protocol will be regulated according to the judgment of the attending clinician considering such factors as age, condition and size of the patient, the potency of the Smac mimetic administered, the duration of the treatment and the severity of the disease being treated. For example, a dosage regimen of the Smac mimetic to reduce tumor growth can be oral administration of from about 1 mg to about 2000 mg/day, preferably about 1 to about 1000 mg/day, more preferably about 50 to about 600 mg/day, in two to four divided doses. Intermittent therapy (e.g., one week out of three weeks or three out of four weeks) may also be used.
[0073] In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that the patient's tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of compounds. Generally, a maximum dose is used, that is, the highest safe dose according to sound medical judgment. Those of ordinary skill in the art will understand, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reason.
|0074] Embodiments of the invention also include a method of treating a patient with cancer or an autoimmune disease by promoting apoptosis wherein administration of a therapeutically effective amount of a Smac mimetic and the Smac mimetic binds to at least one IAP. In one embodiment, the IAP can be XIAP. In another embodiment, the IAP can be ML- IAP, and in still another embodiment, the IAP can be cIAP-1 or cIAP-2. In further embodiments, the IAP can be multiple IAPs. The method may further include concurrent administration of a chemotherapeutic agent including, but not limited to, alkylating agents, antimetabolites, anti-tumor antibiotics, taxanes, hormonal agents, monoclonal antibodies, glucocorticoids, mitotic inhibitors, topoisomerase I inhibitors, topoisomerase II inhibitors, immunomodulating agents, cellular growth factors, cytokines, and nonsteroidal antiinflammatory compounds.
[0075} A variety of administration routes are available. The particular mode selected will depend, of course, upon the particular chemotherapeutic drug selected, the severity of the condition being treated, and the dosage required for therapeutic efficacy. The methods of the invention may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of active compounds without causing clinically unacceptable adverse effects. Such modes of administration include, but are not limited to, oral, rectal, topical, nasal, intradermal, inhalation, intra-peritoneal, or parenteral routes. The term "parenteral" includes subcutaneous, intravenous, intramuscular, or infusion. Intravenous or intramuscular routes are particularly suitable for purposes of the present invention. [0076] In one aspect of the invention, a Smac mimetic as described herein, with or without additional biological or chemotherapeutic agents or radiotherapy, does not adversely affect normal tissues, while sensitizing tumor cells to the additional chemotherapeutic/radiation protocols. While not wishing to be bound by theory, because the induced apoptosis is tumor specific, marked and adverse side effects such as inappropriate vasodilation or shock may be minimized. Preferably, the composition or method may be designed to allow sensitization of the cell or tumor to the chemotherapeutic or radiation therapy by administering at least a portion of the Smac mimetic prior to chemotherapeutic or radiation therapy. The radiation therapy, and/or inclusion of chemotherapeutic agents, may be included as part of the therapeutic regimen to further potentiate the tumor cell killing by the Smac mimetic.
[0077] Combination Therapy A combination of a Smac mimetic and a chemotherapeutic/anti-neoplastic agent and/or radiation therapy of any type may be used in embodiments of the invention and may provide a more effective approach to destroying tumor cells. Smac mimetics generally interact with IAPs, such as XlAP, cIAP-1, cIAP-2, ML-IAP, etc., and block the IAP mediated inhibition of apoptosis while chemotherapeutics/anti neoplastic agents and/or radiation therapy kills actively dividing cells by activating the intrinsic apoptotic pathway leading to apoptosis and cell death. As is described in more detail below, embodiments of the invention provide combinations of a Smac mimetic and chemotherapeutic/anti-neoplastic agents and/or radiation that may provide synergistic action against unwanted cell proliferation. This synergistic action between a Smac mimetic and a chemotherapeutic/anti-neoplastic agent and/or radiation therapy can improve the efficiency of the chemotherapeutic/anti-neoplastic agent and/or radiation therapy. This may allow for an increase in the effectiveness of current chemotherapeutic/anti-neoplastic agents or radiation treatment allowing the dose of the chemotherapeutic/anti-neoplastic agent to be lowered, thereby providing both a more effective dosing schedule, as well as a more tolerable dose of chemotherapeutic/anti-neoplastic agent and/or radiation therapy.
[0078] Additional chemotherapeutic agents Suitable chemotherapeutic agents include, but are not limited to the chemotherapeutic agents described in "Modern Pharmacology with Clinical Applications," Sixth Edition, Craig & Stitzel, Chpt. 56, pgs, 639-656 (2004), hereby incorporated by reference. This reference describes chemotherapeutic drugs including alkylating agents, antimetabolites, anti-tumor antibiotics, plant-derived products such as taxanes, enzymes, hormonal agents such as glucocorticoids, miscellaneous agents such as cisplatin, monoclonal antibodies, immunomodulating agents such as interferons, and cellular growth factors. Other suitable classifications for chemotherapeutic agents include mitotic inhibitors and nonsteroidal antiestrogenic analogs. Other suitable chemotherapeutic agents include toposiomerase ϊ and II inhibitors and kinase inhibitors.
[0079J Specific examples of suitable biological and chemotherapeutic agents include, but are not limited to, cisplatin, carmustine (BCNU), 5-flourouracil (5-FU), cytarabine (Ara-C), gemcitabine, methotrexate, daunorubicin, doxorubicin, dexamethasone, topotecan, etoposide, paciitaxel, vincristine, tamoxifen, TNF-alpha, TRAIL, interferon (in both its alpha and beta forms), thalidomide, and melphalan. Other specific examples of suitable chemotherapeutic agents include nitrogen mustards such as cyclophosphamide, alkyl sulfonates, nitrosoureas, ethylenimines, triazenes, folate antagonists, purine analogs, pyrimidine analogs, anthracyclines, bleomycins, mitomycins, dactinomycins, plicamycin, vinca alkaloids, epipodophyllotoxins, taxanes, glucocorticoids, L-asparaginase, estrogens, androgens, progestins, luteinizing hormones, octreotide actetate, hydroxyurea, procarbazine, mitotane, hexamethylmeϊamme, carboplatin, mitoxantrone, monoclonal antibodies, levamisole, interferons, interleukins, filgrastim, and sargramostim. Chemotherapeutic compositions also comprise other members, i.e., other than TRAIL of the TNF superfamily of compounds.
[0080] For example, in one embodiment of the invention, the therapeutic compounds of the present invention may be administered with TRAIL or other chemical or biological agents which bind to and activate the TRAIL receptor(s). Many cancer cell types are sensitive to TRAIL-induced apoptosis, while most normal cells appear to be resistant to TRAIL-induced apoptosis. TRAΪL-resistant cells may arise by a variety of different mechanisms including loss of the receptor, presence of decoy receptors, or overexpression of FLIP which competes for zymogen caspase-8 binding during DISC formation. Smac mimetics appear to increase tumor cell sensitivity to TRAIL leading to enhanced apoptosis, the clinical correlations of which are expected to be increased apoptotic activity in TRAIL resistant tumors, improved clinical response, increased response duration, and ultimately, enhanced patient survival rate. In support of this, reduction in XIAP levels by in vitro antisense treatment has been shown to cause sensitization of resistant melanoma cells and renal carcinoma cells to TRAIL (Chawla-Sarkar, et al., 2004). The Smac mimetics disclosed herein may bind to IAPs and inhibit their interaction with caspases, therein potentiating TRAIL-induced apoptosis,
[0081] Another embodiment of the invention provides Smac mimetics that act synergisticaliy with a topoismerase inhibitor to potentiate their apoptotic inducing effect. Topoisomerase inhibitors inhibit DNA replication and promote DNA damage by inhibiting the enzymes that are required in the DNA repair process thereby promoting apoptosis. Therefore, export of Smac from the mitochondria into the eel! cytosol is provoked by the DNA damage caused by topoisomerase inhibitors. Topoisomerase inhibitors, such as those of the Type I class, including camptothecin, topotecan, SN-38 (irinotecan active metabolite), and those of the Type II class including etoposide, show potent synergy with the Smac mimetics of the invention in a multi-resistant glioblastoma cell line (T98G), breast cancer line (MDA-MB-231), and ovarian cancer line (OVCAR-3) among others. Exemplary topoisomerase inhibiting agents that may be used in embodiments of the invention include, but are not limited to irinotecan, topotecan, etoposide, amsacrine, exatecan, gimatecan, aclacinomycin A, camptothecin, daunorubicin, doxorubicin, ellipticine, epirubicin, and mitaxantrone.
{0082] In still another embodiment of the invention, a platinum containing compound may be used as chemotherapeutic/anti-neoplastic agent in combination with a Smac mimetic. Exemplary platinum containing compounds that may synergize with a Smac mimetic include, but are not limited to, cisplatin, carboplatin, and oxaliplatin.
|0083] In yet another embodiment of the invention, taxanes may be used as the chemotherapeutic /anti-neoplastic agent that synergizes with a compound according to the invention. Taxanes may act as, for example, anti-mitotic, mitotic inhibitors or microtubule polymerization agents and include, but are not limited to, docetaxel and paclitaxel. Taxanes are characterized as compounds that promote assembly of microtubules by inhibiting tubulin depolymerization, thereby blocking cell cycle progression. Microtubules are highly dynamic cellular polymers made of alpha-beta-tubulin and associated proteins that play key roles during mitosis by participating in the organization and function of the spindle, assuring the integrity of the segregated DNA. Therefore, microtubules represent an effective target for cancer therapy, and taxanes may effectively attack this target by causing, for example, centrosomal impairment, induction of abnormal spindles, and suppression of spindle microtubule dynamics. [0084] Another class of agents that may be utilized in embodiments of the invention includes microtubule poisons which, in contrast to taxanes, inhibit tubulin polymerization. These compounds include, but are not limited to vinca alkaloids, colchicine, and cryptophycines.
I0085| In a further embodiment, any agent that activates the intrinsic apoptotic pathway and/or causes the release of Smac or cytochrome c from the mitochondria has the potential to act synergistically with a Smac mimetic and may be used in combination with the compounds of embodiments of the invention.
[0086] Radiotherapy protocols Additionally, in several embodiments of the invention, Smac mimetic therapy may be used in connection with chemo-radiation or other radiation treatment protocols used to inhibit tumor cell growth.
[0087] Radiation therapy (or radiotherapy) is the medical use of ionizing radiation as part of cancer treatment to control malignant cells and is suitable for use in embodiments of the present invention. Although radiotherapy is often used as part of curative, primary, therapy, it is occasionally used as a palliative treatment where cure is not possible and the aim is for symptomatic relief. Radiotherapy is commonly used for the treatment of tumors, and it is common to combine radiotherapy with surgery and/or chemotherapy. The most common tumors treated with radiotherapy are breast cancer, prostate cancer, rectal cancer, head and neck cancers, gynecological tumors, bladder cancer, and lymphoma. Radiation therapy is commonly applied just to the localized area involved with the tumor. Often the radiation fields include the draining lymph nodes. It is possible, but uncommon, to give radiotherapy to the whole body or entire skin surface. Radiation therapy is usually given daily for up to 35-38 fractions (a daily dose is a fraction). These small frequent doses allow healthy cells time to grow back, repairing damage inflicted by the radiation. Three main divisions of radiotherapy are external beam radiotherapy, or teletherapy, brachytherapy or sealed source radiotherapy and unsealed source radiotherapy, which are all suitable examples of treatment protocol in the present invention. The differences relate to the position of the radiation source: external is outside the body, while sealed and unsealed source radiotherapy has radioactive material delivered internally. Brachytherapy sealed sources are usually extracted later, while unsealed sources are injected into the body. Administration of a Smac mimetic may occur prior to and/or concurrently with the treatment protocol. EXAMPLE
[0088] Annex in V/Propidium Iodide Stain ins Annexin V-fluorescein isothiocyanate staining shows the ability of dimeric Smac mimetics to induce apoptosis. Cells are briefly exposed to various concentrations of dimeric Smac mimetics for 18-24 hours and removed from the assay plate by trypsinization. Cells are pelleted and resuspended in assay buffer (supplied by manufacturer). Annexin V and propidium iodide are added to the cell preparations and incubated for 1 hour in the dark at room temperature. Following the incubation, additional buffer (200 μl) is added to each tube, and the samples are analyzed by flow cytometry. In the presence of Smac mimetics apoptosis is strongly promoted as assessed by Annexin WPI staining and analyzed by flow cytometry. The amplification in the number of apoptotic cells by IAP antagonists as compared to control was dose dependent and due to the induction of apoptosis and not via increasing the proportion of necrotic cells,
[0089] Biological and chemotherapeutics/anti-neoplastic agents and radiation induce apoptosis by activating the extrinsic or intrinsic apoptotic pathways. Since Smac mimetics relieve inhibitors of apoptotic proteins (IAPs) and thus, remove the block in apoptosis, the combination of chemotherapeutics/anti-neoplastic agents and radiation with Smac mimetics should work synergistically to facilitate apoptosis.
[0090] The relevance of this potent synergy is that it makes possible the use of the dimeric Smac mimetics, which are IAP antagonists, to improve the efficacy of conventional chemotherapeutic agents, such as, marketed platinum containing compounds (cisplatin and carboplatin). This may be accomplished by lowering the required dose of the poorly tolerated platinum containing compounds and/or by improving the response rate at the marketed dose.
[0091 j The present invention is not limited to the embodiments described and exemplified above, but is capable of variation and modification within the scope of the appended claims.

Claims

J. CLAIMS
What is claimed is:
1. A compound comprising a homodϊmer or heterodimer having monomeric units of formula (I):
Figure imgf000066_0001
wherein: each Rt is, independently, H; C|-C4-alkyl; Ci-C4-alkenyl; C[-C4-alkynyl or C3-C10- cycloalkyl which are unsubstituted or substituted; each R2 is, independently, H; Ci-C4-alkyl; C|-C4-alkenyI; CrCf-alkynyl or C3-CJO- cycloalkyl which are unsubstituted or substituted; each R3 is, independently, H; -CF3; -C2H5;
Figure imgf000066_0002
Ci-Cj-aikenyl; C|-C4-alkynyI; - CH2-Z or any R2 and R3 together form a heterocyclic ring; each Z is, independently, H; -OH; F; CI; -CH3; -CF3; -CH2Cl; -CH2F or -CH2OH; each R4 is, independently, C I -CI6 straight or branched alkyl; Ci-Ciό-alkenyl; Ci-C 16- alkynyl; C3-C|0-cycloalkyl; -(CH2)i.6-Zj; -(CH2)o-6-aryl; and -(CH2)0-6-het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each Zi is, independently, -N(R10)-C(O)-CM 0-alkyi; -N(R1O)-C(O)-(CH2)^-C3-T- cycloalkyl; -N(R!0)-C(O)-(CH2)0-6-phenyl;
Figure imgf000066_0003
-C(O)-N(Rn)(R12); - C(O)-O-CM 0-alkyl; -C(O)-O-(CH2)^-C3.7-cydoaikyl; -C(O)-O-(CH2)0-6-phenyl; -C(O)-O- (CH2)i-6-het; -0-C(O)-C M ϋ-alkyl; -O-C(O)-(CH2)^-C3-7-cycloalky]; -O-C(O)-(CH2Wphenyl; - 0-C(O)-(CH2) ι-6-het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1 -4 heteroatoms selected from N. O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom; each R1o is, independently, H; -CH3; -CF3; -CH2OH; or -CH3CI; each Rn and R12 is, independently, H; C^-allcyl; C3-7-cycloalkyl; -(CH2)t-6-C3-7- cycloaJkyl; (CH2)0-6-phenyl; wherein alkyl, cycloalkyl, and phenyl are unsubstitυted or substituted; or R1 i and R12 together with the nitrogen form het; each R5 is, independently, H: CVio-alkyl; aryl; phenyl; C3-7-cycloaIkyl; -(CH2)I-6-C3-7- cycloalkyi; -C,.ιo-aJkyl-aryl;
Figure imgf000067_0001
phenyl]2; indanyl; -C(O)-C M0-alkyl; -C(O)-(CH2)] ^-C3.7-cycloalkyI; -C(O)-(CH2)0^-phenyl; - (CHi)0-6-C(0)-phenyl; -(CH2)o-ό-het; -C(O)-(CH2) i^-hel; or R5 is a residue of an amino acid, wherein the alkyl, cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; each U is, independently, as shown in structure (II):
Figure imgf000067_0002
wherein: each n is, independently, 0 to 5; each X is. independently, -CH or N; each Ra and Rb is. independently, an O, S, or N atom or C0.g-alkyl wherein one or more of the carbon atoms in the alkyl chain are optionally replaced by a heteroatom selected from O, S, or N, and where each alkyl is, independently, either unsubstituted or substituted; each Rd is, independently, selected from:
Figure imgf000067_0003
Ar1-D-Ar2; each Rc is, independently, II or any Rc and R^ together form a cycloalkyl or het; where if R<j and Rc form a cycioalkyl or het, R5 is attached to the formed ring at a C or N atom; each p and q is, independently, O or 1 ; each Re is, independently, Ct-g-alkyl or aikylϊdene, and each R^ is either unsubstituted or substituted; each Q is, independently, N, O, S, S(O), or S(O)2; each Ar1 and Ar2 is, independently, substituted or unsubstituted aryl or het; each Rf and Rg is, independently, H; -CM0-alkyl; Ci-io-alkylaryl; -OH; -O-C 1.io-alkyl; - (CH2)o.6-C3.7-cycloalkyl; -0-(CH2)o-ό-aryI; phenyl; aryl; phenyl-phenyl; -(CH2),-6-het; -0-(CH2),. 6-het; -OR13; -C(O)-R13; -C(O)-N(Rn)(R14); -N(R13)(RH); -S-R13; -S(O)-R13; -S(O)2-R 13; -S(O)2- NRnR14; -NR13-S(O)2-R14; -S-C1-10-alkyl; aryl-C 1-4-alkyl; or het-C 1-4-alkyl wherein alkyl, cycloalkyl, het, and aryl are unsubstituted or substituted; -SO2-C 1-2-alkyl; -SO2-C 1-2-alkyiphenyl; -O-C1-4-alkyl; or any Rg and R1 together form a ring selected from het or aryl; each D is, independently, -CO-; -C(O)-C 1-7-alkylene or arylene; -CF2-; -O- ; -S(O)1- where r is 0-2; 1 ,3-dioxalane; or C1-7-alkyl-OH; where alky!, alkylene, or arylene are unsubstituted or substituted with one or more halogens, OH, -O-C 1-6-alkyl, -S-C1-6-alkyl; or -CF3; or each D is, independently, N(R11) wherein each Rh is, independently, H; unsubstituted or substituted C 1-7-alkyl; aryl; unsubstituted or substituted -O-(C1-7-cycloalkyl); -C(O)-C1-10-alkyl; - C(O)-C0-10-alkyl-aryl; -C-O-C1-10-alkyl; -C-O-C0- 10-alkyl-aryl; -SO2-C1-10-alkyl; or -SO2-(C0-10- alkylaryl); each R6, R7, R8, and R9 is, independently, H, -C1-10-alkyl; -C1-10-alkoxy; aryl -C1-10- alkoxy; -OH; -O-C1-10-alkyl; -(CH2)(0-6-C3-7-cycIoalkyl; -0-(CH2)0-6-aryl; phenyl; -(CH2)1-6-het; - O-(CH2)1 -6-het; -OR13; -C(O)-R13; -C(O)-N(R13)(R14); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2-
R13; -S(O)2-NR13R14; or -NR13-S(O)2-R14; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; and any R6, R7, R8, and R9 optionally together form a ring system; each R13 and R14 is, independently, H; C1-10-alkyl; -(CH2)0-6-C3-7-cycloalkyI; -(CH2)0-6,- (CH)(M-(aryl)1-2; -C(O)-C1-10-alkyl; -C(O)-(CH2)1-6-C3-7-cycloalkyl; -C(0)-0-(CH2)0-6 -aryl; - C(0)-(CH2)0-6-O-fluorenyl; -C(O)-NlI-(CH2)0-6 aryl; -C(O)-(CH2)0-6-aryl; -C(O)-(CH2)0-6-het; - C(S)-C1-10-alkyl; -C(S)-(CH2)1-6-C3.7-cycloalkyl; -C(S)-O-(CH2)0-6-aryl; -C(S)-(CH2)0-6-O- fluorenyl; -C(S)-NH-(CH2)0-6-aryl; -C(S)-(CH2)0-6-aryl; or -C(S)-(CH2) 1-6-het; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; or any R13 and R14 together with a nitrogen atom form het; wherein alkyl substituents of R13 and R14 are unsubstituted or substituted and when substituted are substituted by one or more substituents selected from C1-10-alkyl, halogen, OH,- O-C 1-6-aIkyl, -S-C 1-6-alkyl, and -CF3; and substituted phenyl or aryl of R13 and R14 are substituted by one or more substituents selected from halogen, hydroxyl, C1-4-alkyl, C1-4-alkoxy, nitro, -CN, -0-C(O)-C 1-4-alkyl, and -C(O)-O-C 1-4-aryI; or a pharmaceutically acceptable salt or hydrate thereof.
2. The compound of claim 1 , having the formula (III):
Figure imgf000069_0001
wherein:
R1 and Rj' are each, independently, H; Ci-Gi-alkyl; C)-C4-alkenyl; C|-C4-alkynyl; or C3- Cio-cycloaikyl which are unsubstituted or substituted; R2 and R2' are each, independently, H; Ci-C4-alkyl; C|-C4-alkenyl; Ci-C4-alkynyl; or C3- Cio-cycloalkyl which are unsubstituted or substituted; R3 and R3' are each, independently, H; -CF3; -C2H5; C|-C4-alkyl; C|-C4-alkenyI; CrC4- alkynyl; or -CH2-Z or R2 and R3 together or independently with R2' and R3' form a heterocyclic ring; each Z is, independently. H; -OH; F: Cl; -CH3; -CF3; -CH2CI; -CH2F; Or -CH2OH; R4 and R4' are each, independently, C 1-Ci6 straight or branched alky]; Cι-C)6-alkenyl; C\- C 16-alkynyl; or C3-Cio-cycloaIkyl; -(CH2)M-Zi;
Figure imgf000069_0002
or -(CH2)0-6-het; wherein alky], cycloalkyl, and phenyl are unsubstituted or substituted; each Z1 is, independently, -N(R,0)-C(0)-Ci.ιo-alkyl; -N(R!0)-C(OHCH2)1-6-C3.7- cycloalkyl; -N(R10)-C(O)-(CH2)0-6-phenyl; -N(R,o)-C(OMCH2),.6-het; -C(O)-N(Rn)(R12); - C(O)-O-C i.to-alkyl; -C(O)-O-(CH2)i.6-C3-7-cycloalky]; -C(O)-O-(CH2)0-6-phenyl; -C(O)-O- (CH2)]-6-het; -O-C(O)-CM0-alkyl; -0-C(O)-(CH2) ι^C3-7-cycloalkyl; -0-C(0)-(CH2)o.6-phenyl; or -0-C(O)-(CH2) i-6-het; wherein alky], cycloalkyl, and phenyl are unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1 -4 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom; each R10 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each Rn and R;2 is, independently, H; C^-alkyl; C3.7-cycloalkyl; -(CIb)[^-C3-7- cycloalkyl; or (CH2)o-ό-phenyl; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; or RH and Rt2 together with a nitrogen form het; R5 and R5" are each, independently, H; Ci_,o-aikyi; aryl; phenyl; C3.7~cycioaik.yl; -(CH2)!- 6-C3.7-cycloalkyl; -C 1.io-aJkyl-aryl; -(CH2)0-6-C3-7-cycloalkyI-(CH2)o^-phenyI; -(CH2)0-4- CH[(CH2)M-phenylJ2; indanyl; -C(O)-CM0-aIkyI; -C(O)-(CH2)i-6-C3.7-cycloaIky]; -C(OHCH2)0. 6-phenyl; -(CH2)0-6-C(0)-phenyl; -(CH2 Whet; or -C(O)-(CI I2) 1 -6-het; wherein the alkyl, cycloaikyl, phenyl, and aryl substituents are unsubstituted or substituted; or Rs and R5' are, independently, an amino acid residue;
U and U' are each as shown in structure (II):
Figure imgf000070_0001
wherein: each X is, independently, -CH or N; each Ra and Rb is, independently, an O, S, or N atom or Co-s-aϊkyl wherein one or more of the carbon atoms in the alkyl chain are optionally replaced by a heteroatom selected from O, S, or N, and where each alkyl is, independently, either unsubstituted or substituted; each R<j is, independently, selected from:
Figure imgf000070_0002
An-D-Ar2; each Rc is, independently, II or any R^ and Rj together form a cycloaikyl or het; where if Rd and Rc form a cycloalkyl or het. Rs is attached to the formed ring at a C or N atom; each p and q is, independently, O or 1 ; each Re is, independently, d-s-alkyl or alkylidene, and each R6 is either unsubstituted or substituted; each Q is, independently, N, O, S. S(O), or S(O)2; each Ar1 and Ar2 is, independently, substituted or unsubstituted aryl or het; each Rf and Rg is, independently, H; -C|.to-alkyl; C 1.jo-alkylaryj; -OH; -O-Ci.jo-alkyl; - (CH2)0-6-C3.7-cycloa3kyi; -O-(CH2Waryl; phenyl; aryl; phenyl-phenyl; -(CH2), .6-het; -0-(CH2),. 6-het; -OR13; -C(O)-R13; -C(O)-N(R13)(RN); -N(R13)(R14); -S-Rn; -S(O)-R13; -S(O)2-R13; -S(O)2- NR13R14; -NRB-S(O)2-R54; -S-CM0-alkyl; aryl-CM-aIkyl; or het-CM-alkyl wherein alkyl, cycloalkyl, het, and ary! are unsubstituted or substituted; -SO2-C 1.2-all.yl; -SU2-C 1,2-alkylphenyl; or -0-CM-aikyl; or any Rg and Rf together form a ring selected from het or aryl; each D is. independently, -CO-: -C(O)-C ι-7-alkylene or aryiene; -CF2-; -O-; -S(O)r where r is 0-2; 1,3-dioxalane; or C|_7-alkyl-OH; where alkyl, aikylene, or aryiene are unsubstituted or substituted with one or more halogens, OH,
Figure imgf000071_0001
or -CF3; or each D is, independently, N(Rh) wherein each Rh is, independently, II; unsubstituted or substituted Ci-7-alkyl; aryl; unsubstituted or substituted -O-(Ci-7-cycloalkyl); -C(0)-C[.io-alkyl; - C(0)-C0-io-aIkyl-aryl; -C-O-C1. to-alkyl; -C-O-C0.10-alkyl-aryϊ; or -SO2-C, -!0-aIkyl; or -SO2-(C0. 10-alkylaryl); each R6, R7, Rg, and Ry is, independently. H, -C 1.io-alkyl; -C 1.io-alkoxy; aryl-Ci-io- alkoxy; -OH; -O-C,-1(ralkyl; -(CH2)0^C3.7-cycloalkyl; -0-(CH2)0-6-aryl; phenyl; -(CH2)i-6-het; - O-(CH2)w-het; -0R]3; -C(O)-R13; -C(O)-N(Rn)(R14); -N(R13)(R14); -S-R13; -S(O)-R13; -S(O)2- RB; -S(O)2-NR13R14; or -NRB-S(O)2-RI4; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; and any R6, R7, Rg. and R9 optionally together form a ring system; each R13 and R)4 is, independently, H; Cμio-alkyl; -(CH2)0-6-C3,7-cycloalkyl; -(CH2)0-6- (CH)o-ι-(aryl),.2; -C(O)-C M0-alkyl; -C(O)-(CH2),^-C3-7-cycIoalkyl; -C(O)-O-(CH2)0-6-aryl; - C(O)-(CH2)0-6-O-fluorenyl; -C(O)-NH-(CH2)0-6-aryI; -C(0)-(CH2)0-6-aryl; -C(O)-(CH2)0-6-het; - C(S)-CM0-alkyl; -C(S)-(CH2) ,.<,-C3.7-cycloalkyl; -C(S)-O-(CH2 Waryl; -C(S)-(CH2)0-6-O- fluorenyl; -C(S)-NH-(CH2)o.6-aryl; -C(S)-(CH2)0^aryl; or -C(S)-(CH2) 1-6-het; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; or any R13 and R14 together with a nitrogen atom form het; wherein alkyl subsituents of R13 and R]4 are unsubstituted or substituted and when substituted are substituted by one or more substituents selected from Cno-alkyl, halogen, OH,- O-Ci-6-alkyl, -S-C|-6-alkyl, and -CF3; and substituted phenyl or aryl of R13 and R14 are substituted by one or more substituents selected from halogen, hydroxyl, C1-4-alkyl, C 1_4-alkoxy, nitro, -CN, -0-C(O)-C M-alkyl, and -C(O)-O-CM-aryl;
L is one or more linkers covalently linking one or more of the positions R4, R5, or U, with R4', R5", or U'; or a pharmaceutically acceptable salt or hydrate thereof.
3. The composition of claim 2, wherein L covalently links two identical monomeric units or L covalently links two non-identical monomeric units.
4. The compound of claim 2, wherein L is selected from alkylene, alkenylene, alkynylene, cycloalkylene, cycloalkylalkylene, aryl, arylalkyiene, arylalkylalkylene, and heterocycloalkylene, heterocycloalkylalkylene, hcteroaryl and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O1 or S, optionally-substituted alkylene, alkenylene, alkynylene cycloalkylene, cycloalkyialkylene, heterocycloalkylene, heterocycloaikyialkylene, aryl, arylaikylene, arylalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, amino, substituted amino, oxygen atom, sulfide, sulfoxide, sulfone, and disulfide.
5. The compound of claim 2, wherein L is selected from -CH2CH2-, - CH2CH2CH2-, -CH-CH-, 1,4-phenyl, 2,5-thiophenyl. -CH(OH)CH(OH)-, -CH2CH-O-CHCH2-, and -CH2C≡CC≡CCH2-.
6. The compound of claim 2, having a formula selected from a compound of formula (IV):
Figure imgf000072_0001
wherein
R1, R]', R2, R2', R3, R3', R4, R4', R5, R5', U, and U' are defined as in claim 2; and
L1 and L2, independently, link position R4 with R4' and R5 with R5'; or a pharmaceutically acceptable salt thereof; a compound of formula (V):
Figure imgf000073_0001
wherein
R1, R1', R2, R2 ', R3, R3', R4, R4 ', Rs- IV, U, and U' are defined as in claim 2; and
L links position R4 with R4'; or a pharmaceutically acceptable salt thereof; and a compound of formula (VI):
Figure imgf000073_0002
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, Rs', U, and U' are defined as in claim 2; and L links position R5 with R5'; or a pharmaceutically acceptable salt thereof.
7 . A compound comprising a homodimer or heterodimer having moπomeric units of general formula (XIII):
Figure imgf000074_0001
wherein: each Rt is, independently, H; each R2 is, independently, H or C|-C4-alkyl which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH3, -SCH3, -CN, -SCN, and nitro; each R3 is, independently, H; -CF3; -C2F5; -CH2-Z or any R2 and R3 together with the nitrogen form a C3-C0 heteroaliphatic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; or -CH2OH; each R4 is, independently, Ci-Ci6 straight chain alkyl; C3-C10 branched chain alkyl; - (CH2)o-6-C3-C7-cycIoalky]; -(CH2)^-Z,; ~(CH2)0.6-phenyl; or -(CH2)0-6-het; wherein each alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each Z, is, independently, -N(R9)-C(O)-CM0-alkyl; -N(R9)-C(O)-(CH2)1-6-C3^- cycloalkyl; -N(R9)-C(O)-(CH2)0^-phenyl; ™N(R9)-C(O)-(CH2)i-6-het; -C(O)-N(R,0)(Rπ); -C(O)- 0-CMo-alkyl; -C(O)-O-(CH2)w-C3-7-cycIoalkyl; -C(0)-0-(CH2)o-6-ρhenyl; -C(O)-O-(CH2) i ^- het; -0-C(O)-C I-, o-alkyl; -0-C(O)-(CH2) ,.6-C3.7-cycloalkyl; -0-C(0)-(CH2Wphenyl; or -O- C(O)-(CH2) ι-6-het; wherein each alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted, and when substituted is substituted on a carbon atom by halogen, hydroxyl, C]-4-alkyl, Ci-4-alkoxy, nitro, -0-C(O)-Ci- 4-alkyl, -C(O)-O-C M-aIkyl, or on a nitrogen by CM-alkyl, -0-C(O)-C M-alkyl, or -C(O)-O-CM- alkyl; each R9 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each R1o and Rn is, independently, H; C^-alkyl; C3_7-cycloalkyl; -(CH2)I-O-C3-7- cycloalkyl; or (CH2)o-<j-phenyl; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; or any R1O and Rt j together with a nitrogen form het; each X is, independently, CH or N: each R5 is, independently H; Ci-io-alkyl; aryl; phenyl; C3-7~cycloalkyl; -(CH2)I-O-C3.?- cycloaikyϊ; -CM0-alkyl-aryI; -(CH2)0-6-C3.7-cyc!oalkyKCH2)o-6-phenyl; -(CH2)o.4-CH[(CH2)M- phenylj2; -(CH2)0.6-CH(phenyl)2; -C(O)-C M 0-alkyl; -C(O)-(CH2) 1 -6-C3.7-cycloalky); -C(O)- (CH2)0-6-phenyi; -(CH2)0^-C(O)-phenyl; -(ClbWhet; -C(O)-(CH2)i-6-het; or R5 an amino acid residue, wherein the alkyl, cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; each R^a is, independently, H, methyl, ethyl, -CF3, -CH2OH or -CH2Cl; or each any R5 and R^a together with a nitrogen form a het; each R3 and Rg is independently, cis relative to acyl substituents at the one position of the ring and are each, independently, H, -C 1.io-alkyl; -OH; -O-Cs-io-alkyl; -(CH2)0-6-C3-7-cycloalkyl; -0-(CH2)o-6-aryl; phenyl; -(CH2), -6-het; ~O-(CH2)!-6-het; -N(R12)(R13); -S-R12; -S(O)-R13; -S(O)2- R 12; or -S(O)2-NRI2RI 3; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; each R12 and R]3 is, independently, H; Ci-io-alkyl; -(CH2)o-ό-C3-7-cycloalkyl; -(CH2Ve- (CH)o-f-(aryl),.2; -C(O)-CM0-alkyl; ~C(O)-(CH2),-6-C3,7-cycloalkyl; -C(O)-O-(CH2)0.6-aryI; - C(0)-(CH2)o-6-0-fluorenyl; -C(O)-NH-(CH2)0.6-aiyl; -C(0)-(CH2)o-6-aryl; or -C(O)-(CH3)!-6-het; wherein each alkyl cycloalkyl, or aryl is unsubstituted or substituted; or any R]2 and R13, together with a nitrogen atom form het; each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; each n is O, 1, or 2; and wherein substituted alkyl is substituted by one or more substituents selected from a double bond, halogen, OH,-O-C^-alkyl, -S-C|.6-aikyl, and -CF3; substituted cycloalkyl is substituted by one or more substituents selected from a double bond, C-6-alkyl, halogen, OH7-O-C, -6-alkyl, -S-C(-6-alkyl, and -CF3; and substituted phenyl or aryl is substituted by one or more substituents selected from halogen, hydroxy, C^-alkyl, Cs-4-alkoxy, nitro, -CN, -0-C(O)-C 1-4-alkyj, and -C(O)-O-CM- aryl; or a pharmaceutically acceptable salt or hydrate thereof. 8, The compound of claim 7, having the formula (XlV):
Figure imgf000076_0001
wherein:
R[ and R| ' are each H; R2 and R2' are each, independently, H or C|-C4-alkyl which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH3, -SCH3, -CN, - SCN, and nitro; R3 and R3' are each, independently, H; -CFy, -C2F5; or -CH2-Z or R2 and R3 or R2 ' and R3' together with a nitrogen form a C3-C6 heteroaliphatic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; Or -CH2OH;
R4 and R4' are each, independently, C1 -CJ6 straight chain alkyl; C3-C10 branched chain alkyl; -(CH2)O-S-Cs-C 7-cycIoalkyl;
Figure imgf000076_0002
-(CH2)0-6-phenyl; or ™{CH2)o-ό-het; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; each Zj is, independently, -N(R9)-C(O)-Ci.i0-alkyI; -N(R9J-C(O)-(CH2)^-C3.?- cycloaikyl; -N(R9)-C(0)-(CH2)0-6-phenyI; ~N(R9)-C(O)-(CH2)!-6-het; -C(O)-N(R10)(R, ,); -C(O)- 0-Ci.ιo-alkyl; -C(0)-0-(CH2)i.6-C3.7-cycloaIkyl; -C(0)-0-(CH2)o-6-phenyi; -C(O)-O-(CH2),^- het; -0-C(0)-C|.ιo-alkyI;
Figure imgf000076_0003
-0-C(0)-(CH2)0-6-phenyl; or -O- C(O)-(CH2) 1 -6-het; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon atom by halogen, hydroxyl, C|^-alkyl, C^-alkoxy, nitro, -0-C(O)-CM-alkyl, -C(O)-O-C M-alkyl, or on a nitrogen by CM-alkyl, -0-C(O)-C M-aIkyl, or -C(O)-O-C M-alkyl; each R9 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each Rfo and Rn is, independently, H; C^-alky.; C3.7-cycloaIk.yi; -(CH2)i^-C3-7- cycloalkyl; or (CH2)o^-phenyI; wherein each alkyt, cycloalkyl, and phenyl is unsυbstituted or substituted; or any Rjo and Rn together with a nitrogen form het;
X and X' are each, independently, CH or N;
R5 and R5' are each, independently, H; C 1,ιo-alkyl; aryl; phenyl; C^^-cycloalkyl; -(CH2)I- 6-C3-7-cycloalkyl; -C 1.io-alkyl-aryl; -(CH2)0-6-C3-7-cycloalkyl-(CH2)ϋ-6-phenyl; -(CH2)o-4- CH[(CH2)M-phenyl]2; -(CH2)o^-CH(phenyl)2; -C(O)-C M0-alkyl; -C(O)-(CH2), _6-C3.7-cycloalkyl; -C(0)-(CH2)o-6-phenyl; -(CH2)0-6-C(O)-phenyl; -(CH2), or R5 is an amino acid residue, wherein each alkyl, cycloalkyl, phenyl, and aryl substituent is unsubstituted or substituted;
R6a and R63' are each, independently, H, methyl, ethyl, -CF3, -CH2OH ,or -CH2Cl; or
R5 and R6a are, independently, or together with R5' and R^3' together with a nitrogen are het;
Ra, Rg, Ra', and Rg' are cis relative to an acyl substituent at position one of a ring to which they are attached and are each, independently, H, -C 1.io-alkyl; -OH; -O-C|-10-alkyl; - (CH2)o-6-C3.7-cycloalkyl; -0-(CH2Wary]; phenyl; -(CH2),.6-het; -O-(CH2) ,.6-het; -N(R12)(R13); - S-R12; -S(O)-Rs2; -S(O)2-R12; or -S(O)2-NR12R13; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted;
R12 and R13 are each, independently, H; Ci-io-alkyl; -(CH2)tH,-C3.7-cycloalkyl; -(CH2)O-6- (CH)0-r(aryl)1 -2; -C(O)-CM0-aIkyl; -C(O)-(CH2)i*-C3.7-cycloaIkyl; ~C(0)-0-(CH2)0-6-aryl; - C(0)-(CH2)o-6-0-fluorenyl; -C(O)-NH-(CH2)0.6-aiyl; -C(OHCH2)0.6-aryl; or -C(O)-(CH2) ,.6-het; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; or R12 and R!3 together with a nitrogen atom form het; each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; n and n' are each, independently, O, 1 , or 2; wherein each substituted alkyl is substituted by one or more substituents selected from a double bond, halogen,
Figure imgf000077_0002
and -CF3; each substituted cycloalkyl is substituted by one or more substituents selected from a double bond, C|.6-alkyl, halogen. OH1-O-C w-alkyl, -S-C^-alkyl, and -CF3; and each substituted phenyl or aryi is substituted by one or more substituents selected from halogen, hydroxy, C1-4-alkyl, C,-4-alkoxy, nitro, -CN, -0-C(O)-C M-alkyl, and -C(O)-O-CM- aryl; and
L is one or more linkers covalently linking one or more of the positions R4, R5, Rg, with R4\ R5', RS';or a pharmaceutically acceptable salt or hydrate thereof.
9. The compound of claim 8, wherein L covalently links two identical monomeric units or L covalently links two non-identical monomelic units.
10. The compound of claim 8, wherein L is selected from alkylene, alkenylene, alkynylene, cycloalkylene, cycloalkyiaikyiene, aryl, arylalkyiene, arylalkylalkylene, and heterocycloalkylene, heterocycloalkylalkylene, heteroaryl and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, optionally-substituted alkylene, alkenylene, alkynylene cycloalkylene, cycloalkyiaikyiene, heterocycioalkylene, heterocycloalkylalkylene, aryl, arylalkyiene, arylalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, amino, substituted amino, oxygen atom, sulfide, sulfoxide, sulfone, and disulfide.
1 1. The compound of claim 8, wherein L is selected from -CH2CH2-, - CH2CH2CH2-, -CH=CH-, 1,4-phenyl, 2,5-thiophenyl, -CH(OH)CH(OH)-, -CH2CH-O-CHCH2-, and -CH2C≡CC≡CCH2-,
12. The compound of claim 8, having a formula selected from a compound of formula (XV):
Figure imgf000078_0001
wherein
R1, Rs', R2, R2', R3, R3', R4, R4', R5, R5'. R6a, R6a', Ra, Ra', R8, R8', X, X*, n, and n' are defined as in claim 8; and
L1 and L2, independently link position R4 with R4' and R5 with R5'; or a pharmaceutically acceptable salt or hydrate thereof; a compound of formula (XVl):
Figure imgf000079_0001
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a', Ra, Ra', R8, R8', X, X', n, and n' are defined as in claim 8; and
L1 and L2, independently link position R4 with R4' and R8 with R8'; or a pharmaceutically acceptable salt or hydrate thereof; a compound of formula (XVH):
Figure imgf000079_0002
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a' , Ra, Ra', R8, R8', X= X'. n, and n' are defined as in claim 8; and
L links position R4 with R4'; or a pharmaceutically acceptable salt or hydrate thereof; a compound of formula (XVlIl):
Figure imgf000080_0001
wherein
R1, R1', R2, R2', R3, R3', R4, R4', R5, R5', R6a, R6a' , Ra, Ra', R8, R8', X, X', n, and n' are defined as in claim 8; and
L links position R5 with R5'; o; r a pharmaceutically acceptable sait or hydrate thereof; and a compound of formula (XlX):
Figure imgf000080_0002
wherein
R1, R1 ', R2, R2', R3, R3', R4, R4', R5, R5' , R6a, R6a'. Ra, Ra', R8, R8', X, X', n, and n' are defined as in claim 8; and
L links position R8 with R8'; or a pharmaceutically acceptable salt or hydrate thereof.
13, The composition of claim 1, having a formula selected from formula (VII):
Figure imgf000081_0001
formula (IX):
formula (X):
Figure imgf000082_0001
formula (Xl):
Figure imgf000083_0001
formula (XII)
Figure imgf000083_0002
14 A pharmaceutical composition comprising a compound of formula (III):
Figure imgf000083_0003
wherein
R1 and Rj ' are each, independently, H, C|-C4-alkyl, C|-C4-alkenyl, Ci-CYalkynyi, or C3- Cio-cycloalkyl which are unsubstituted or substituted, R2 and R2' are each, independently, H, Ci-Cj-alkyl. Ci-C 1-afkenyl; Ci-C4-alkynyl, or C3- Cio'Cycloalkyi which are unsubstituted or substituted. R3 and R3' are each, independently, H; -CF3; -C2H5; C|-C,-alkyl; Ci-Q-aJkenyl; C1-Cj- alkynyl; or -QW1-Z or R2 and R3 together or independently with R2 7 and R3 * form a heterocyclic ring; each Z is, independently, H; -OH; F; Cl; -CH3; -CF3; -CH2Cl; -CH2F; or -CH2OH;
R4 and R4' are each, independently,
Figure imgf000084_0001
C,- Ci6-alkynyl; or C3-C,0-cycloalkyl; -(CH2)^6-Zi ; -(CH2V6-aryl; or -(CH2)o.6-het; wherein alkyl, cycloalky], and phenyl are unsubstituted or substituted; each Zi is, independently, -N(Rιo)-C(0)-CM O-alkyl;
Figure imgf000084_0002
cycloaϊkyl; -N(Rιo)~C(0)-(CH2)o-6-ρhenyl; -N(R1o)-C(0)-(CH2),.6-het; -C(O)-N(R1 IXR12); - C(O)-O-C,- l 0-alkyI; -C(O)-O-(CH2)i.6-C3-7-cycloalky]; -C(0)-0-(CH2)o-6-phenyl; -C(O)-O- (CH2)i.6-het; -0-C(O)-C M0-aIkyl; -0-C(O)-(CH2),.6-C3.7-cycloalkyI; -O-C(O)-(CH2)0-6-phenyl; or -0-C(O)-(CH2), ^-het; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1-4 heteroatoms selected from N, O, and S, or an 8-12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon or nitrogen atom; each R1o is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each Rn and R12 is, independently, H; CM-alkyl; C3_7-cycloaikyl;
Figure imgf000084_0003
cycloalkyl; or (CH2)o-^-phenyl; wherein alkyl, cycloalkyl, and phenyl are unsubstituted or substituted; or Rn and Rj2 together with a nitrogen form het;
R5 and R5' are each, independently, H; CM0-alkyl; aryl; phenyl; C3-7-cycloalkyl; -(CH2)f. 6-C3-7-cycioalkyl; -CM0-alkyl-aryl; -(CH2WC3-7-cycloalkyl-(CH2)0-6-phenyl; -(CH2)o-4- CH[(CH2)M-phenyl]2; indanyl; -C(O)-CM 0-a]kyl; -C(O)-(CH2), ^-C3.7-cycloalkyl; -C(O)-(CH2)0. 6-phenyl; -(CH2)ϋ-<)-C(O)-phenyi; -(CH2)o-ό-het; or -C(O)-(CH3) s^-het; wherein the alky], cycloalkyl, phenyl, and aryl substituents are unsubstituted or substituted; or R5 and R5' are, independently, an amino acid residue;
U and U' are each as shown in structure (11):
Figure imgf000085_0001
wherein: each X is, independently, -CH or N; each Ra and Rt, is, independently, an O, S, or N atom or Co-8-alkyl wherein one or more of the carbon atoms in the alkyl chain are optionally replaced by a heteroatom selected from O, S, or N, and where each alkyl is, independently, either unsubstituted or substituted; each Rd is, independently, selected from: R41-Q-(Rf)P(Rg)1,; and Ar]-D-Ar2; each Rc is, independently, H or any R0 and Rd together form a cycloalkyl or het; where if Rd and R6 form a cycloalkyl or het, R5 is attached to the formed ring at a C or N atom; each p and q is, independently, O or 1 ; each Rc- is, independently, d.s-alkyl or alkylidene, and each Re is either unsubstituted or substituted; each Q is, independently, N, O, S, S(O), or S(O)2; each Art and Ar2 is, independently, substituted or unsubstituted aryl or het; each Rf and Rg is, independently, H; -Ci-to-alkyl; d-io-alkylaryl; -OH; -O-Cμio-alkyl; - (CH2)o-6-C3-7-cycloalky]; -O-(CH2)0-6-aryl; phenyl; aryl; phenyl-phenyl; -(CH2) i-e-het; -O-(CH2)i- 6-het; -OR13; -C(O)-R13; -C(O)-N(R13)(R14); -N(R13)(RH); -S-R13; -S(O)-R13; -S(O)2-R13; -S(O)2- NR13RI4; -NR13-S(O)2-Ru; -S-CM 0-alkyl; aryl-CM-alkyl; or het-CM-alkyl wherein alkyl, cycloalkyl, het, and aryl are unsubstituted or substituted; -Sθ2-Ci-2-aJkyl; -SO2-C 1.2-alkylphenyl; or -O-C|-4-alkyl; or any Re and Rf together form a ring selected from het or aryl; each D is, independently, -CO-; -C(O)-C 1.7-alkyIene or arylene; -CF2-; -0-; -S(0)r where r is 0-2; i,3-dioxalane; or C|-7-alkyl-OH; where alkyl, alkylene, or arylene are unsubstituted or substituted with one or more halogens, OH, -O-Ci^-alkyl, -S-Cι-6-alkyl; or -CF3; or each D is, independently, N(Rj1) wherein each Rh is, independently, H; unsubstituted or substituted Ci-7-alky!; aryl; unsubstituted or substituted -O-(C1_7-cycioalkyI); -C(O)-C no-alkyl; - C(0)-Co-io-alkyi-aryl; -C-O-C M0~alkyI; -C-O-C0. ,0-alkyl-aryl; or -SO2-C M0-alkyl; or -SO2-(C0. lo-alkylaryl); each R6, R?, Rs, and R9 is, independently, H, -Ci.[0-alkyl; -Cμio-alkoxy; aryl-C 1-io- alkoxy: -OH; -O-CM0-alkyI:
Figure imgf000086_0001
- O-(CH2),-6-het; -OR13; -C(O)-R13; -C(O)-N(R13)(R14); -N(R13)(Ru); -S-R13; -S(O)-R13; -S(O)2-
R13; -S(O)2-NR[3R14; or -NR13-S(O)2-R14; wherein each alkyl, cycioalkyl, and aryl is unsubstituted or substituted; and any RO, R7. Rg, and R9 optionally together form a ring system; each R13 and Ru is, independently, H; CMo-alkyl; -(CH2)o-6-C3-7-cycloalkyl; -(CH2)0-6- (CH)o.r(aryI),-2; -C(O)-CM 0-alkyl; -C(O)-(CH2)i-6-C3-7-cycloalkyl; -C(0)-0-(CH2)o-6-aryl; - C(O)-(CH2 WO-fluorenyl; -C(O)-NH-(CH2)0-6-aryl; -C(O)-(Cl I2)0-6-aryl; -C(O)-(CH2)0-6-het; - C(S)-C,.,0-aIkyl; -C(S)-(CH2),^-C3-7-cycloalkyl; -C(S)-O-(CH2)0-6-aryl; -C(S)-(CH2)0-6-O- fluorenyl; -C(S)-NH-(CH2)0-6-aryI; -C(S)-(CH2)0^-aryl; or -C(S)-(CH2), ^-het; wherein each alkyl, cycioalkyl, and aryl is unsubstituted or substituted; or any R 13 and R14 together with a nitrogen atom form het; wherein alkyl subsituents of R13 and Ru are unsubstituted or substituted and when substituted are substituted by one or more substituents selected from Ci-io-alkyl, halogen, OH,- O-Ci-6-alkyl, -S-C!-6-alkyl, and -CF3; and substituted phenyl or aryl of R13 and R[4 are substituted by one or more substituents selected from halogen, hydroxyl,
Figure imgf000086_0002
nitro, -CN, -O-C(O)-CM-alkyl, and -C(O)-O-C 1-4-aryl;
L is one or more linkers covalently linking one or more of the positions R4, R5, or U, with R4', R5', or LT; or a pharmaceutically acceptable salt or hydrate thereof; and a pharmaceutically acceptable excipient or carrier; or a compound of formula:
Figure imgf000086_0003
wherein:
Figure imgf000086_0004
Ri and R2' are each, independently, H or Ci-Cr alkyl which is unsubstituted or substituted by one or more substituents selected from halogen, -OH, -SH, -OCH3, -SCH3. -CN, - SCN, and nitro; R3 and R3' are each, independently, H; -CF3; -C2F5; or -CH2-Z or R2 and R3 or R2' and R3' together with a nitrogen form a C3-C6 heteroaϋphatic ring; each Z is, independently, H; -OH; F; CI; -CH3; -CF3; -CH2Cl; -CH2F; Or -CH2OH; R4 and R4' are each, independently, Ci-Ci6 straight chain alkyl; C3-C(O branched chain alkyl; -(CH2)ϋ.6-C3-C7-cycloalkyl;
Figure imgf000087_0001
-(CH2Wphenyl; or -(CH2)0-6-het; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; each Z1 is, independently, ~N(R9)-C<O)-C M (raikyl; -N(R9)-C(O)-(CH2),.6-C3.7- cycloalkyl; -N(R9)-C(0)-(CH2)o^phenyl; -N(R<0-C(O)-(CH2)i.6-het; -C(O)-N(R10)(Rn); -C(O)- O-Ci-10-alkyl; -C(0)-O-(CH2)1 -6-C3-7-cycϊoalkyl; -C(O)-O-(CH2)0.6-phenyl; -C(O)-O-(CH2) ,-6- het; -0-C(0)-CMo-aIkyI; -O-C(OHCH2)i.6-C3.7-cycloaIkyl; -O-C(O)-(CH2Wphenyl; or -O- C(O)-{CH2)i-6-het; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; each het is, independently, a 5-7 member heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, O, and S, or an 8- 12 member fused ring system including at least one 5-7 member heterocyclic ring containing 1 , 2, or 3 heteroatoms selected from N, O, and S, which heterocyclic ring or fused ring system is unsubstituted or substituted on a carbon atom by halogen, hydroxyl, Ci^-alkyl, C^-alkoxy, nitro, -0-C(O)-C 1-4-alky], -C(O)-O-C 1 -4-alkyl, or on a nitrogen by C-M-alkyl, -O-C(O)-C1-4-alkyl, or -C(O)-O-C M-alkyl; each R9 is, independently, H; -CH3; -CF3; -CH2OH; or -CH2Cl; each Rjo and Rn is, independently, H; Ci-4-alkyl; C3-7-cycloalkyl; -(CH2)I-O-C3-7- cycloalkyl; or (CH2)o.6-phenyl; wherein each alkyl, cycloalkyl, and phenyl is unsubstituted or substituted; or any R1o and R) , together with a nitrogen form het;
X and X' are each, independently, CH or N;
R5 and R5' are each, independently, H; CM0-aIkyl; aryl; phenyl; C^-cycloalkyl; -(CH2) 1- 6-C3-7-cycloalkyI; -CM0-alkyl-aryl; -(CH2)o.6-C3-7-cycloaIky]-(CH2)0.6-ρhenyI; -(CH2)(M- CH[(CH2)M-phenylj2; -(CH2WCH(phenyl)2; -C(O)-C,.|0-alkyl; -C(O)-(CH2)i.6-C3-7-cyc]oaIkyl; -C(O)-(CH2Wphenyl; -(CH2)0-6-C(O)-pheny]; -(CH2) i-6-het; or -C(O)-(CH2)Mi-het; or R5 is an amino acid residue, wherein each alkyl, cycloalkyi, phenyl, and aryl substituent is unsubstituted or substituted; R6a and R63' are each, independently, H, methyl, ethyl, -CF3, -CH2OH ,Or -CH2Cl; or
R5 and R6a are, independently, or together with R5' and R^3' together with a nitrogen are het;
Ra, Rg, Ra\ and Rg' are cis relative to an acyl substituent at position one of a ring to which they are attached and are each, independently, H, -C 1,io-alkyl; -OH; -O-Cμjo-alkyl; - (CH2)0-6-C3.7-cycloaIkyl; -O-(CH3)0-6-aryl; phenyl; -(CH2), ^-het; -O-(CH2)t.6-het; -N(R12)(R13); - S-R!2; -S(O)-R13; -S(O)2-R12; or -S(O)2-NRJ2R13; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted;
Rj2 and Rj3 are each, independently, H; Ci-tϋ-alkyl; -(CH2)o-6-C3-7-cycIoalkyl; -(CH2)O-O- (CH)0-i-(aryl),.2; -C(O)-CM 0-alkyl; -C(O)-(CH2),.6-C3-7-cycloalkyl; -C(O)-O-(CH2 Waryl; - C(0)-(CH2)0-6-0-fluorenyl; -C(O)-NH-(CH2)0-6-aryl; -C(O)-(CH2)0^-aryl; or ~C(O)-(CH2)i-6-het; wherein each alkyl, cycloalkyl, and aryl is unsubstituted or substituted; or R12 and Rt3 together with a nitrogen atom form het; each aryl is, independently, an unsubstituted or substituted phenyl or naphthyl; n and n' are each, independently, O. 1 , or 2; wherein each substituted alkyl is substituted by one or more substituents selected from a double bond, halogen, OH5-O-C i-6-alkyl,
Figure imgf000088_0001
and -CF3; each substituted cycloalkyl is substituted by one or more substituents selected from a double bond, C1-(,-alky], halogen, OH,-O-C]^-alkyl, -S-Ci-6-alkyl, and -CF3; and each substituted phenyl or aryl is substituted by one or more substituents selected from halogen, hydroxy, CM-alkyϊ, Cu-alkoxy, nitro, -CN, -0-C(O)-Ci -4-alkyl, and -C(O)-O-CM- aryi; and
L is one or more linkers covalently linking one or more of the positions R4, R5, Rg, with R4', R5', Rs';or a pharmaceutically acceptable salt or hydrate thereof; and a pharmaceutically acceptable excipient or carrier.
15. The pharmaceutical composition of claim 14, wherein L and L' are selected from aikylene, alkenylene, alkynylene, cycloaikylene, cycloalkylalkylene, aryl, arylalkylene, arylalkylalkyiene, heterocycloalkylene. heterocycloalkylaikylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, optionally-substituted aikylene, alkenylene, alkynylene cycloaikylene. cycloalkylalkylene, heterocycloalkylene, heterocycloalkylalkylene, aryl, arylalkylene, arylalkylalkylene, heteroaryl, and heteroarylalkylene where one or more carbon atoms are optionally replaced with N, O, or S, amino, substituted amino, oxygen atom, sulfide, sulfoxide, sulfone, and disulfide.
16. The compound of claim 14, wherein L and L' are selected from -CH2CH2- . -CH2CH2CH2-. -CH-CH-, 1 ,4-phenyϊ, 2,5-thiophenyl, -CH(OH)CH(OH)-, -CH2CH-O-CHCH2- , and -CH2C≡CC≡CCH2-.
17. The pharmaceutical composition of claim 14, further comprising a second therapeutic agent.
18. The pharmaceutical composition of claim 17, wherein said compound and said second therapeutic agent provide therapy to an individual.
19. The pharmaceutical composition of claim 17, wherein said second therapeutic agent is selected from a chemotherapeutic agent, radiation, and a combination thereof.
20. The pharmaceutical composition of claim 18, wherein said chemotherapeutic is selected from an alkylating agent, a plant alkaloid, an antitumor antibiotic, an antimetabolite, a topoisomerase inhibitor, and a combination thereof.
PCT/US2007/074181 2006-07-24 2007-07-24 Dimeric iap inhibitors WO2008014236A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/374,731 US20100056495A1 (en) 2006-07-24 2007-07-24 Dimeric iap inhibitors
US13/431,745 US20120184530A1 (en) 2006-07-24 2012-03-27 Dimeric IAP Inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US82016906P 2006-07-24 2006-07-24
US60/820,169 2006-07-24

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/431,745 Continuation US20120184530A1 (en) 2006-07-24 2012-03-27 Dimeric IAP Inhibitors

Publications (1)

Publication Number Publication Date
WO2008014236A1 true WO2008014236A1 (en) 2008-01-31

Family

ID=38617462

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/074181 WO2008014236A1 (en) 2006-07-24 2007-07-24 Dimeric iap inhibitors

Country Status (2)

Country Link
US (2) US20100056495A1 (en)
WO (1) WO2008014236A1 (en)

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7547724B2 (en) 2005-10-25 2009-06-16 Aegera Therpeutics, Inc. IAP BIR domain binding compounds
US7579320B2 (en) 2006-03-16 2009-08-25 Aegera Therapeutics, Inc. IAP BIR domain binding compounds
WO2009140447A1 (en) * 2008-05-16 2009-11-19 Novartis Ag Immunomodulation by iap inhibitors
US7772177B2 (en) 2005-05-18 2010-08-10 Aegera Therapeutics, Inc. BIR domain binding compounds
WO2010142994A1 (en) * 2009-06-12 2010-12-16 Astrazeneca Ab 2, 3-dihydro-1h-indene compounds and their use to treat cancer
WO2011104266A1 (en) * 2010-02-25 2011-09-01 Novartis Ag Dimeric iap inhibitors
US8163792B2 (en) 2006-05-16 2012-04-24 Pharmascience Inc. IAP BIR domain binding compounds
US8541424B2 (en) 2008-12-23 2013-09-24 Abbott Laboratories Anti-viral compounds
US8546405B2 (en) 2008-12-23 2013-10-01 Abbott Laboratories Anti-viral compounds
JP2013545781A (en) * 2010-12-13 2013-12-26 ノバルティス アーゲー Dimer IAP inhibitor
US8686026B2 (en) 2010-06-10 2014-04-01 Abbvie Inc. Solid compositions
US8691938B2 (en) 2009-06-11 2014-04-08 Abbvie Inc. Anti-viral compounds
US8716454B2 (en) 2009-06-11 2014-05-06 Abbvie Inc. Solid compositions
US8937150B2 (en) 2009-06-11 2015-01-20 Abbvie Inc. Anti-viral compounds
US9034832B2 (en) 2011-12-29 2015-05-19 Abbvie Inc. Solid compositions
US9278922B2 (en) 2009-04-15 2016-03-08 Abbvie Inc. Anti-viral compounds
US9284350B2 (en) 2010-02-12 2016-03-15 Pharmascience Inc. IAP BIR domain binding compounds
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
US9394279B2 (en) 2009-06-11 2016-07-19 Abbvie Inc. Anti-viral compounds
US9988376B2 (en) 2013-07-03 2018-06-05 Glaxosmithkline Intellectual Property Development Limited Benzothiophene derivatives as estrogen receptor inhibitors
US9993514B2 (en) 2013-07-03 2018-06-12 Glaxosmithkline Intellectual Property Development Limited Compounds
WO2018119357A1 (en) 2016-12-24 2018-06-28 Arvinas, Inc. Compounds and methods for the targeted degradation of enhancer of zeste homolog 2 polypeptide
US10034912B2 (en) 2009-07-02 2018-07-31 TetraLogic Birinapant UK Ltd. SMAC Mimetic
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
WO2019099926A1 (en) 2017-11-17 2019-05-23 Arvinas, Inc. Compounds and methods for the targeted degradation of interleukin-1 receptor-associated kinase 4 polypeptides
WO2019195609A2 (en) 2018-04-04 2019-10-10 Arvinas Operations, Inc. Modulators of proteolysis and associated methods of use
US10441654B2 (en) 2014-01-24 2019-10-15 Children's Hospital Of Eastern Ontario Research Institute Inc. SMC combination therapy for the treatment of cancer
WO2020023851A1 (en) 2018-07-26 2020-01-30 Yale University Bifunctional substitued pyrimidines as modulators of fak proteolyse
WO2020041331A1 (en) 2018-08-20 2020-02-27 Arvinas Operations, Inc. Proteolysis targeting chimeric (protac) compound with e3 ubiquitin ligase binding activity and targeting alpha-synuclein protein for treating neurodegenerative diseases
WO2020051564A1 (en) 2018-09-07 2020-03-12 Arvinas Operations, Inc. Polycyclic compounds and methods for the targeted degradation of rapidly accelerated fibrosarcoma polypeptides
US10604506B2 (en) 2017-01-26 2020-03-31 Arvinas Operations, Inc. Modulators of estrogen receptor proteolysis and associated methods of use
US10647698B2 (en) 2016-12-01 2020-05-12 Arvinas Operations, Inc. Tetrahydronaphthalene and tetrahydroisoquinoline derivatives as estrogen receptor degraders
US10710986B2 (en) 2018-02-13 2020-07-14 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10723717B2 (en) 2016-12-23 2020-07-28 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of rapidly accelerated fibrosarcoma polypeptides
US10772962B2 (en) 2015-08-19 2020-09-15 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of bromodomain-containing proteins
US10774071B2 (en) 2018-07-13 2020-09-15 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10806737B2 (en) 2016-12-23 2020-10-20 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of fetal liver kinase polypeptides
US10899735B2 (en) 2018-04-19 2021-01-26 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10946017B2 (en) 2015-06-05 2021-03-16 Arvinas Operations, Inc. Tank-binding kinase-1 PROTACs and associated methods of use
US10994015B2 (en) 2016-12-23 2021-05-04 Arvinas Operations, Inc. EGFR proteolysis targeting chimeric molecules and associated methods of use
US11028088B2 (en) 2018-03-10 2021-06-08 Yale University Modulators of BTK proteolysis and methods of use
WO2021148396A1 (en) 2020-01-20 2021-07-29 Astrazeneca Ab Epidermal growth factor receptor tyrosine kinase inhibitors for the treatment of cancer
US11173211B2 (en) 2016-12-23 2021-11-16 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of rapidly accelerated Fibrosarcoma polypeptides
US11236085B2 (en) 2018-10-24 2022-02-01 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11267803B2 (en) 2016-06-21 2022-03-08 Orion Ophthalmology LLC Carbocyclic prolinamide derivatives
US11352351B2 (en) 2015-01-20 2022-06-07 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of androgen receptor
US11377439B2 (en) 2016-06-21 2022-07-05 Orion Ophthalmology LLC Heterocyclic prolinamide derivatives
US11427548B2 (en) 2015-01-20 2022-08-30 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of androgen receptor
US11458123B2 (en) 2016-11-01 2022-10-04 Arvinas Operations, Inc. Tau-protein targeting PROTACs and associated methods of use
US11484534B2 (en) 2013-03-14 2022-11-01 Abbvie Inc. Methods for treating HCV
US11912699B2 (en) 2019-07-17 2024-02-27 Arvinas Operations, Inc. Tau-protein targeting compounds and associated
US11957759B1 (en) 2022-09-07 2024-04-16 Arvinas Operations, Inc. Rapidly accelerated fibrosarcoma (RAF) degrading compounds and associated methods of use

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005084317A2 (en) * 2004-03-01 2005-09-15 Board Of Regents, The University Of Texas System Dimeric small molecule potentiators of apoptosis
WO2006020060A2 (en) * 2004-07-15 2006-02-23 Tetralogic Pharmaceuticals Corporation Iap binding compounds
WO2006122408A1 (en) * 2005-05-18 2006-11-23 Aegera Therapeutics Inc. Bir domain binding compounds
WO2007048224A1 (en) * 2005-10-25 2007-05-03 Aegera Therapeutics Inc. Iap bir domain binding compounds

Family Cites Families (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3854480A (en) * 1969-04-01 1974-12-17 Alza Corp Drug-delivery system
US3832253A (en) * 1973-03-21 1974-08-27 Baxter Laboratories Inc Method of making an inflatable balloon catheter
DE2714880A1 (en) * 1977-04-02 1978-10-26 Hoechst Ag CEPHEMDER DERIVATIVES AND PROCESS FOR THEIR PRODUCTION
US4667014A (en) * 1983-03-07 1987-05-19 Syntex (U.S.A.) Inc. Nonapeptide and decapeptide analogs of LHRH, useful as LHRH antagonists
US4452775A (en) * 1982-12-03 1984-06-05 Syntex (U.S.A.) Inc. Cholesterol matrix delivery system for sustained release of macromolecules
CA1200416A (en) * 1983-05-13 1986-02-11 Societe Des Produits Nestle S.A. Food process
US5075109A (en) * 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
US5208007A (en) * 1988-11-22 1993-05-04 Board Of Regents Of The University Of Oklahoma Isotopic tracer composition and method for making and using same
JPH04167172A (en) * 1990-10-31 1992-06-15 Nec Corp Vector processor
WO1994003501A1 (en) * 1992-08-05 1994-02-17 Meito Sangyo Kabushiki Kaisha Small-diameter composite composed of water-soluble carboxypolysaccharide and magnetic iron oxide
US6187557B1 (en) * 1995-08-08 2001-02-13 Tularik Inc. c-IAP1 and c-IAP2: inhibitors of apoptosis
US5786173A (en) * 1996-03-19 1998-07-28 Idun Pharmaceuticals, Inc. MCH4 and MCH5, apoptotic protease, nucleic acids encoding and methods of use
US6133437A (en) * 1997-02-13 2000-10-17 Apoptogen, Inc. Modulation of IAPs for the treatment of proliferative diseases
US5961955A (en) * 1997-06-03 1999-10-05 Coulter Pharmaceutical, Inc. Radioprotectant for peptides labeled with radioisotope
ES2156567B1 (en) * 1999-09-15 2001-12-16 Castillo Juan Luis Bellvis DYNAMIC FOOTBALL.
US6514964B1 (en) * 1999-09-27 2003-02-04 Amgen Inc. Fused cycloheptane and fused azacycloheptane compounds and their methods of use
US6110691A (en) * 2000-01-06 2000-08-29 Board Of Regents, The University Of Texas System Activators of caspases
IL137226A0 (en) * 2000-07-09 2001-07-24 Nirosoft Ind Ltd Method for modifying membrane rejection characteristics
US6608026B1 (en) * 2000-08-23 2003-08-19 Board Of Regents, The University Of Texas System Apoptotic compounds
EP1315811A2 (en) * 2000-08-24 2003-06-04 Thomas Jefferson University An iap binding peptide or polypeptide and methods of using the same
US6992063B2 (en) * 2000-09-29 2006-01-31 The Trustees Of Princeton University Compositions and method for regulating apoptosis
US6561897B1 (en) * 2000-10-17 2003-05-13 Shuffle Master, Inc. Casino poker game table that implements play of a casino table poker game
WO2002060914A2 (en) * 2001-02-01 2002-08-08 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Identification of small rnas and orfs form e. coli as mediators of cell and intercell regulation
US20020160975A1 (en) * 2001-02-08 2002-10-31 Thomas Jefferson University Conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO for mediating apoptosis
CN101029080A (en) * 2001-11-21 2007-09-05 伯纳姆研究院 Methods and compositions for derepression of IAP-inhibited caspase
US20060258581A1 (en) * 2001-11-21 2006-11-16 Reed John C Methods and composition for derepressions of IAP-inhibited caspase
ES2289293T3 (en) * 2002-01-09 2008-02-01 Faron Pharmaceuticals Oy COMMON VASCULAR ENDOTELIAL AND ENDOTELIAL RECEIVER-1 AND CLEAR-1 ENDOTELIAL AND ITS USES.
JP4541882B2 (en) * 2002-07-02 2010-09-08 ノバルティス アーゲー Peptide inhibitor for binding of SMAC protein to apoptotic protein inhibitor (IAP)
JP4142357B2 (en) * 2002-07-04 2008-09-03 株式会社神戸製鋼所 Waste high pressure fluid treatment method and apparatus
IL152271A (en) * 2002-10-13 2006-04-10 Meir Hefetz Microneedles structures and production methods
US6965040B1 (en) * 2002-11-04 2005-11-15 Xiaolian Gao Photogenerated reagents
JP4173422B2 (en) * 2002-12-04 2008-10-29 日本電信電話株式会社 Quantum key distribution method and quantum key distribution system
CA2424708A1 (en) * 2003-04-08 2004-10-08 Lee A. Danisch A method and apparatus for sensing impact between a vehicle and an object
US7200353B2 (en) * 2003-09-01 2007-04-03 Seiko Epson Corporation Image forming apparatus with intermediate transfer member
US20050084317A1 (en) * 2003-10-17 2005-04-21 Adriana Kliegman Soap dispensing cleaning device
US20050097791A1 (en) * 2003-11-10 2005-05-12 Guadagno Lisa M. Keepsake system
WO2005060777A1 (en) * 2003-12-12 2005-07-07 Lonnie Dale Tiegs An attachment for an item of protective sports gear
CA2558615C (en) * 2004-03-23 2013-10-29 Genentech, Inc. Azabicyclo-octane inhibitors of iap
DK2253614T3 (en) * 2004-04-07 2013-01-07 Novartis Ag IAP inhibitors
CN101035802A (en) * 2004-07-02 2007-09-12 健泰科生物技术公司 Inhibitors of IAP
US20060010118A1 (en) * 2004-07-09 2006-01-12 Juergen Sattler System and method for role-based spreadsheet data integration
KR100559590B1 (en) * 2004-07-15 2006-03-10 동부아남반도체 주식회사 Method for forming device isolation layer of semiconductor device
JP5153053B2 (en) * 2004-07-22 2013-02-27 Sabicイノベーティブプラスチックスジャパン合同会社 Flame retardant antistatic polyester resin composition
US20060069063A1 (en) * 2004-09-27 2006-03-30 Yng-Jiin Wang Crosslinked polygalacturonic acid used for postsurgical tissue adhesion prevention
US7205864B2 (en) * 2004-11-02 2007-04-17 Nextg Networks, Inc. Distributed matrix switch
US8734341B2 (en) * 2004-12-20 2014-05-27 Ipventure, Inc. Method and apparatus to sense hydration level of a person
EP1836201B2 (en) * 2004-12-20 2013-09-04 Genentech, Inc. Pyrrolidine inhibitors of iap
KR100591773B1 (en) * 2004-12-20 2006-06-26 삼성전자주식회사 Nonvolatile semiconductor memory device and voltage generating circuit for the same
EP1851200B1 (en) * 2005-02-25 2014-01-15 Tetralogic Pharmaceuticals Corporation Dimeric iap inhibitors
US20070003535A1 (en) * 2005-03-17 2007-01-04 Reed John C Methods and compositions for derepression of IAP-inhibited caspase
JP4353129B2 (en) * 2005-04-19 2009-10-28 ソニー株式会社 Disk drive device and electronic device
WO2006133147A2 (en) * 2005-06-08 2006-12-14 Novartis Ag Organic compounds
WO2007021825A2 (en) * 2005-08-09 2007-02-22 Tetralogic Pharmaceuticals Corporation Treatment of proliferative disorders
KR20080087781A (en) * 2005-09-21 2008-10-01 퀵커, 인크. Device-aware content delivery
US20070106192A1 (en) * 2005-09-23 2007-05-10 Axiom Worldwide, Inc. System and method for treating the spine with light therapy
US7972474B2 (en) * 2005-12-13 2011-07-05 Kimberly-Clark Worldwide, Inc. Tissue products having enhanced cross-machine directional properties
MX2008008191A (en) * 2005-12-20 2008-11-04 Novartis Ag Combination of an iap-inhibitor and a taxane7.
US8202902B2 (en) * 2006-05-05 2012-06-19 The Regents Of The University Of Michigan Bivalent SMAC mimetics and the uses thereof
US8163792B2 (en) * 2006-05-16 2012-04-24 Pharmascience Inc. IAP BIR domain binding compounds
US7771739B2 (en) * 2006-06-30 2010-08-10 Abbott Cardiovascular Systems Inc. Implantable medical devices comprising semi-crystalline poly(ester-amide)
PE20110224A1 (en) * 2006-08-02 2011-04-05 Novartis Ag PROCEDURE FOR THE SYNTHESIS OF A PEPTIDOMIMETIC OF Smac INHIBITOR OF IAP, AND INTERMEDIARY COMPOUNDS FOR THE SYNTHESIS OF THE SAME
KR20090082221A (en) * 2006-10-19 2009-07-29 노파르티스 아게 Organic Compounds
CN101541325A (en) * 2006-11-28 2009-09-23 诺瓦提斯公司 Use of IAP inhibitors for the treatment of acute myeloid leukemia
ATE509629T1 (en) * 2006-11-28 2011-06-15 Novartis Ag COMBINATION OF IAP INHIBITORS AND FLT3 INHIBITORS
US20080134679A1 (en) * 2006-12-11 2008-06-12 Cavanaugh David B Solar powered turbine driven generator system
KR20090094461A (en) * 2006-12-19 2009-09-07 제넨테크, 인크. Imidazopyridine inhibitors of iap
BRPI0810178A2 (en) * 2007-04-12 2014-09-23 Joyant Pharmaceuticals Inc SMAC MIME DIMERS AND THERMALS USEFUL AS ANTICCAN AGENTS

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005084317A2 (en) * 2004-03-01 2005-09-15 Board Of Regents, The University Of Texas System Dimeric small molecule potentiators of apoptosis
WO2006020060A2 (en) * 2004-07-15 2006-02-23 Tetralogic Pharmaceuticals Corporation Iap binding compounds
WO2006122408A1 (en) * 2005-05-18 2006-11-23 Aegera Therapeutics Inc. Bir domain binding compounds
WO2007048224A1 (en) * 2005-10-25 2007-05-03 Aegera Therapeutics Inc. Iap bir domain binding compounds

Cited By (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8575113B2 (en) 2005-05-18 2013-11-05 Pharmascience Inc. BIR domain binding compounds
US7772177B2 (en) 2005-05-18 2010-08-10 Aegera Therapeutics, Inc. BIR domain binding compounds
US7589118B2 (en) 2005-10-25 2009-09-15 Aegera Therapeutics, Inc. IAP BIR domain binding compounds
US7795298B2 (en) 2005-10-25 2010-09-14 Aegera Therapeutics, Inc. IAP BIR domain binding compounds
US7547724B2 (en) 2005-10-25 2009-06-16 Aegera Therpeutics, Inc. IAP BIR domain binding compounds
US8063095B2 (en) 2005-10-25 2011-11-22 Pharmascience Inc. IAP BIR domain binding compounds
US7579320B2 (en) 2006-03-16 2009-08-25 Aegera Therapeutics, Inc. IAP BIR domain binding compounds
US9365614B2 (en) 2006-03-16 2016-06-14 Pharmascience Inc. IAP BIR domain binding compounds
US7645741B2 (en) 2006-03-16 2010-01-12 Aegera Therapeutics, Inc. IAP BIR domain binding compounds
US8765681B2 (en) 2006-03-16 2014-07-01 Pharmascience Inc. IAP BIR domain binding compounds
US8648094B2 (en) 2006-05-16 2014-02-11 Pharmascience, Inc. IAP BIR domain binding compounds
US8163792B2 (en) 2006-05-16 2012-04-24 Pharmascience Inc. IAP BIR domain binding compounds
US11382905B2 (en) 2008-05-16 2022-07-12 Novartis Ag Immunomodulation by IAP inhibitors
EP2698158A1 (en) * 2008-05-16 2014-02-19 Novartis AG Immunomodulation by IAP Inhibitors
EP3701947A1 (en) * 2008-05-16 2020-09-02 Novartis AG Immunomodulation by iap inhibitors
US10786491B2 (en) 2008-05-16 2020-09-29 Novartis Ag Immunomodulation by IAP inhibitors
US9750729B2 (en) 2008-05-16 2017-09-05 Dana-Farber Cancer Institute, Inc. Immunomodulation by IAP inhibitors
WO2009140447A1 (en) * 2008-05-16 2009-11-19 Novartis Ag Immunomodulation by iap inhibitors
JP2016094429A (en) * 2008-05-16 2016-05-26 ノバルティス アーゲー Immunomodulation by iap inhibitors
JP2011524339A (en) * 2008-05-16 2011-09-01 ノバルティス アーゲー Immune modulation by IAP inhibitors
US9163017B2 (en) 2008-12-23 2015-10-20 Abbvie Inc. Anti-viral compounds
US9249138B2 (en) 2008-12-23 2016-02-02 Abbvie Inc. Anti-viral compounds
US8541424B2 (en) 2008-12-23 2013-09-24 Abbott Laboratories Anti-viral compounds
US8546405B2 (en) 2008-12-23 2013-10-01 Abbott Laboratories Anti-viral compounds
US9278922B2 (en) 2009-04-15 2016-03-08 Abbvie Inc. Anti-viral compounds
US8937150B2 (en) 2009-06-11 2015-01-20 Abbvie Inc. Anti-viral compounds
US9394279B2 (en) 2009-06-11 2016-07-19 Abbvie Inc. Anti-viral compounds
US10039754B2 (en) 2009-06-11 2018-08-07 Abbvie Inc. Anti-viral compounds
US10028937B2 (en) 2009-06-11 2018-07-24 Abbvie Inc. Anti-viral compounds
US9586978B2 (en) 2009-06-11 2017-03-07 Abbvie Inc. Anti-viral compounds
US8921514B2 (en) 2009-06-11 2014-12-30 Abbvie Inc. Anti-viral compounds
US8716454B2 (en) 2009-06-11 2014-05-06 Abbvie Inc. Solid compositions
US8691938B2 (en) 2009-06-11 2014-04-08 Abbvie Inc. Anti-viral compounds
CN102459238A (en) * 2009-06-12 2012-05-16 阿斯利康(瑞典)有限公司 2, 3-dihydro-1h-indene compounds and their use to treat cancer
WO2010142994A1 (en) * 2009-06-12 2010-12-16 Astrazeneca Ab 2, 3-dihydro-1h-indene compounds and their use to treat cancer
US10314881B2 (en) 2009-07-02 2019-06-11 Medivir Ab SMAC mimetic
US11351221B2 (en) 2009-07-02 2022-06-07 Medivir Ab SMAC mimetic
US10596220B2 (en) 2009-07-02 2020-03-24 Medivir Ab SMAC mimetic
US11951147B2 (en) 2009-07-02 2024-04-09 Medivir Ab SMAC mimetic
US10034912B2 (en) 2009-07-02 2018-07-31 TetraLogic Birinapant UK Ltd. SMAC Mimetic
US9284350B2 (en) 2010-02-12 2016-03-15 Pharmascience Inc. IAP BIR domain binding compounds
WO2011104266A1 (en) * 2010-02-25 2011-09-01 Novartis Ag Dimeric iap inhibitors
CN102834389A (en) * 2010-02-25 2012-12-19 诺瓦提斯公司 Dimeric iap inhibitors
JP2013520466A (en) * 2010-02-25 2013-06-06 ノバルティス アーゲー Dimer IAP inhibitor
US8445440B2 (en) 2010-02-25 2013-05-21 Novartis Ag Dimeric IAP inhibitors
US8686026B2 (en) 2010-06-10 2014-04-01 Abbvie Inc. Solid compositions
JP2013545781A (en) * 2010-12-13 2013-12-26 ノバルティス アーゲー Dimer IAP inhibitor
US10201541B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US9034832B2 (en) 2011-12-29 2015-05-19 Abbvie Inc. Solid compositions
US11484534B2 (en) 2013-03-14 2022-11-01 Abbvie Inc. Methods for treating HCV
US9988376B2 (en) 2013-07-03 2018-06-05 Glaxosmithkline Intellectual Property Development Limited Benzothiophene derivatives as estrogen receptor inhibitors
US9993514B2 (en) 2013-07-03 2018-06-12 Glaxosmithkline Intellectual Property Development Limited Compounds
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
US10105365B2 (en) 2014-01-03 2018-10-23 Abbvie Inc. Solid antiviral dosage forms
US9744170B2 (en) 2014-01-03 2017-08-29 Abbvie Inc. Solid antiviral dosage forms
US10441654B2 (en) 2014-01-24 2019-10-15 Children's Hospital Of Eastern Ontario Research Institute Inc. SMC combination therapy for the treatment of cancer
US11427548B2 (en) 2015-01-20 2022-08-30 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of androgen receptor
US11352351B2 (en) 2015-01-20 2022-06-07 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of androgen receptor
US10946017B2 (en) 2015-06-05 2021-03-16 Arvinas Operations, Inc. Tank-binding kinase-1 PROTACs and associated methods of use
US11554171B2 (en) 2015-08-19 2023-01-17 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of bromodomain-containing proteins
US10772962B2 (en) 2015-08-19 2020-09-15 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of bromodomain-containing proteins
US11267803B2 (en) 2016-06-21 2022-03-08 Orion Ophthalmology LLC Carbocyclic prolinamide derivatives
US11377439B2 (en) 2016-06-21 2022-07-05 Orion Ophthalmology LLC Heterocyclic prolinamide derivatives
US11866422B2 (en) 2016-06-21 2024-01-09 Orion Ophthalmology LLC Carbocyclic prolinamide derivatives
US11458123B2 (en) 2016-11-01 2022-10-04 Arvinas Operations, Inc. Tau-protein targeting PROTACs and associated methods of use
US11597720B2 (en) 2016-12-01 2023-03-07 Arvinas Operations, Inc. Tetrahydronaphthalene and tetrahydroisoquinoline derivatives as estrogen receptor degraders
EP3689868A1 (en) 2016-12-01 2020-08-05 Arvinas Operations, Inc. Tetrahydronaphthalene and tetrahydroisoquinoline derivatives as estrogen receptor degraders
US10899742B1 (en) 2016-12-01 2021-01-26 Arvinas Operations, Inc. Tetrahydronaphthalene and tetrahydroisoquinoline derivatives as estrogen receptor degraders
US10647698B2 (en) 2016-12-01 2020-05-12 Arvinas Operations, Inc. Tetrahydronaphthalene and tetrahydroisoquinoline derivatives as estrogen receptor degraders
US11104666B2 (en) 2016-12-01 2021-08-31 Arvinas Operations, Inc. Tetrahydronaphthalene and tetrahydroisoquinoline derivatives as estrogen receptor degraders
US10723717B2 (en) 2016-12-23 2020-07-28 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of rapidly accelerated fibrosarcoma polypeptides
US11173211B2 (en) 2016-12-23 2021-11-16 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of rapidly accelerated Fibrosarcoma polypeptides
US10994015B2 (en) 2016-12-23 2021-05-04 Arvinas Operations, Inc. EGFR proteolysis targeting chimeric molecules and associated methods of use
US10806737B2 (en) 2016-12-23 2020-10-20 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of fetal liver kinase polypeptides
US11857519B2 (en) 2016-12-24 2024-01-02 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of enhancer of zeste homolog 2 polypeptide
US11191741B2 (en) 2016-12-24 2021-12-07 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of enhancer of zeste homolog 2 polypeptide
WO2018119357A1 (en) 2016-12-24 2018-06-28 Arvinas, Inc. Compounds and methods for the targeted degradation of enhancer of zeste homolog 2 polypeptide
US11384063B2 (en) 2017-01-26 2022-07-12 Arvinas Operations, Inc. Modulators of estrogen receptor proteolysis and associated methods of use
US10604506B2 (en) 2017-01-26 2020-03-31 Arvinas Operations, Inc. Modulators of estrogen receptor proteolysis and associated methods of use
WO2019099926A1 (en) 2017-11-17 2019-05-23 Arvinas, Inc. Compounds and methods for the targeted degradation of interleukin-1 receptor-associated kinase 4 polypeptides
US11065231B2 (en) 2017-11-17 2021-07-20 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of interleukin-1 receptor- associated kinase 4 polypeptides
US10710986B2 (en) 2018-02-13 2020-07-14 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11555029B2 (en) 2018-02-13 2023-01-17 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11028088B2 (en) 2018-03-10 2021-06-08 Yale University Modulators of BTK proteolysis and methods of use
WO2019195609A2 (en) 2018-04-04 2019-10-10 Arvinas Operations, Inc. Modulators of proteolysis and associated methods of use
US11161841B2 (en) 2018-04-04 2021-11-02 Arvinas Operations, Inc. Modulators of proteolysis and associated methods of use
US10899735B2 (en) 2018-04-19 2021-01-26 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10774071B2 (en) 2018-07-13 2020-09-15 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
WO2020023851A1 (en) 2018-07-26 2020-01-30 Yale University Bifunctional substitued pyrimidines as modulators of fak proteolyse
WO2020041331A1 (en) 2018-08-20 2020-02-27 Arvinas Operations, Inc. Proteolysis targeting chimeric (protac) compound with e3 ubiquitin ligase binding activity and targeting alpha-synuclein protein for treating neurodegenerative diseases
US11707452B2 (en) 2018-08-20 2023-07-25 Arvinas Operations, Inc. Modulators of alpha-synuclein proteolysis and associated methods of use
WO2020051564A1 (en) 2018-09-07 2020-03-12 Arvinas Operations, Inc. Polycyclic compounds and methods for the targeted degradation of rapidly accelerated fibrosarcoma polypeptides
US11236085B2 (en) 2018-10-24 2022-02-01 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11912699B2 (en) 2019-07-17 2024-02-27 Arvinas Operations, Inc. Tau-protein targeting compounds and associated
WO2021148396A1 (en) 2020-01-20 2021-07-29 Astrazeneca Ab Epidermal growth factor receptor tyrosine kinase inhibitors for the treatment of cancer
US11957759B1 (en) 2022-09-07 2024-04-16 Arvinas Operations, Inc. Rapidly accelerated fibrosarcoma (RAF) degrading compounds and associated methods of use

Also Published As

Publication number Publication date
US20100056495A1 (en) 2010-03-04
US20120184530A1 (en) 2012-07-19

Similar Documents

Publication Publication Date Title
WO2008014236A1 (en) Dimeric iap inhibitors
US20100143499A1 (en) Dimeric iap inhibitors
US9920093B2 (en) Dimeric IAP inhibitors
EP2049563B1 (en) Dimeric iap antagonists
US20100113326A1 (en) Dimeric iap inhibitors
US20100144650A1 (en) Dimeric iap inhibitors
US8143426B2 (en) IAP inhibitors

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: 07813261

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 07813261

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 12374731

Country of ref document: US