WO2012155066A2 - Spiro-oxindole mdm2 antagonists - Google Patents

Spiro-oxindole mdm2 antagonists Download PDF

Info

Publication number
WO2012155066A2
WO2012155066A2 PCT/US2012/037570 US2012037570W WO2012155066A2 WO 2012155066 A2 WO2012155066 A2 WO 2012155066A2 US 2012037570 W US2012037570 W US 2012037570W WO 2012155066 A2 WO2012155066 A2 WO 2012155066A2
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
compound
pharmaceutically acceptable
group
acceptable salt
Prior art date
Application number
PCT/US2012/037570
Other languages
French (fr)
Other versions
WO2012155066A3 (en
Inventor
Shaomeng Wang
Wei Sun
Angelo AGUILAR
Carlos Garcia-Echeverria
Original Assignee
Shaomeng Wang
Wei Sun
Aguilar Angelo
Carlos Garcia-Echeverria
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CN201280033358.3A priority Critical patent/CN103717605B/en
Priority to MX2013013167A priority patent/MX346375B/en
Priority to KR1020137032975A priority patent/KR101688268B1/en
Priority to NZ617580A priority patent/NZ617580B2/en
Priority to ES12782304.5T priority patent/ES2624808T3/en
Priority to SG2013083118A priority patent/SG194873A1/en
Priority to CA2835422A priority patent/CA2835422C/en
Priority to AU2012253339A priority patent/AU2012253339B2/en
Application filed by Shaomeng Wang, Wei Sun, Aguilar Angelo, Carlos Garcia-Echeverria filed Critical Shaomeng Wang
Priority to BR112013028983A priority patent/BR112013028983A2/en
Priority to EP12782304.5A priority patent/EP2707372B1/en
Priority to JP2014510503A priority patent/JP2014513699A/en
Publication of WO2012155066A2 publication Critical patent/WO2012155066A2/en
Publication of WO2012155066A3 publication Critical patent/WO2012155066A3/en
Priority to IL229387A priority patent/IL229387A/en

Links

Classifications

    • 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
    • C07D487/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D471/12Heterocyclic 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 three hetero rings
    • C07D471/20Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/20Spiro-condensed systems

Definitions

  • the aggressive cancer cell phenotype is the result of a variety of genetic and epigenetic alterations leading to deregulation of intracellular signaling pathways (Ponder, Nature 411:336 (2001)). Cancer cells typically fail to execute an apoptotic program, and lack of appropriate apoptosis due to defects in the normal apoptosis machinery is considered a hallmark of cancer (Lowe et al, Carcinogenesis 27:485 (2000)). The inability of cancer cells to execute an apoptotic program due to defects in the normal apoptotic machinery is often associated with an increase in resistance to chemotherapy, radiation, or immunotherapy-induced apoptosis.
  • MDM2 and p53 are part of an auto- regulatory feed-back loop (Wu et al, Genes Dev. 7: 1 126 (1993)). MDM2 is transcriptionally activated by p53 and MDM2, in turn, inhibits p53 activity by at least three mechanisms (Wu et al, Genes Dev. 7: 1126 (1993). First, MDM2 protein directly binds to the p53 transactivation domain and thereby inhibits p53- mediated transactivation.
  • MDM2 protein contains a nuclear export signal sequence, and upon binding to p53, induces the nuclear export of p53, preventing p53 from binding to the targeted D As.
  • MDM2 protein is an E3 ubiquitin ligase and upon binding to p53 is able to promote p53 degradation.
  • the present disclosure provides spiro-oxindoles having
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
  • R 2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
  • E is selected from the group consisting of -OR 26a and -NR 26b R 26c ;
  • R 26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
  • R 26b is R 4 ;
  • R 26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, -S0 2 R 5b , and R 5 ; or
  • R 26b and R 26c taken together form a 4- to 9-membered optionally substituted heterocyclo
  • R 4 and R 5 has the meanings as described below in connection with Formula II;
  • R 5b is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, and optionally substituted heteroaryl;
  • X is selected from the group consisting of O, S, and NR;
  • Y is selected from the group consisting of O, S, and NR ;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • the present disclosure provides spiro-oxindoles having
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
  • R 2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl;
  • R is selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
  • R 5 is selected from the group consisting of
  • each R 6a and R 6b is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
  • R 7 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
  • R 8a and R 8b are each independently selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl; or
  • R 8a and R 8b taken together with the carbon that they are attached form a 3- to 8-membered optionally substituted cycloalkyl
  • W 1 is selected from the group consisting of -OR 9a and -NR 9b R 9c ;
  • R 9a is hydrogen
  • R 9b is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S0 2 R 9d , and -CONR 9e R 9f ;
  • R 9c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 9b and R 9c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R 9d is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
  • R 9e and R 9f are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or R 9e and R 9f taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • W 2 is selected from the group consisting of -OR 10 and -NR l la R l lb ;
  • R 10 is hydrogen
  • R 9a and R 10 are hydrogen and the other is a metabolically cleavable group
  • R l la is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S0 2 R l lc , and -CONR lld R l le ;
  • R l lb is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R l la and R l lb taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R l lc is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
  • R l ld and R l le are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
  • R l ld and R l le taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • n 1, 2, 3, 4, or 5;
  • each R 12a , R 12b , R 12c and R 12d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
  • R 13 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl
  • R 14 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
  • Z is selected from the group consisting of -OR 15 and -NR 16a R 16b ; or
  • Z and R 14 taken together form a carbonyl, i.e., a C 0, group.
  • R 15 is selected from the group consisting of hydrogen and metabolically cleavable group
  • R lba is selected from the group consisting of -S0 2 R and -CONR ⁇ R 106
  • R 16b is selected from the group consisting of hydrogen and optionally substituted alkyl
  • R 16c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 16d and R 16e are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 16d and R 16e taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo
  • o 1, 2, or 3;
  • p 0, 1, 2, or 3;
  • each R 17a , R 17b , R 17c and R 17d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
  • R 18 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl
  • R 19 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
  • R 20 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
  • R 21a and R 21b are each hydrogen;
  • R 21a and R 21b are hydrogen and the other is metabolically cleavable group
  • q 0, 1, 2, or 3;
  • r is 1, 2, or 3;
  • each R 22a , R 22b , R 22c , and R 22d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
  • R 23 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl
  • R 24 is selected from the group consisting of -S0 2 R 24a and -CONR 24b R 24c ;
  • R a is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 24b and R 24c are each independently selected from the group consisting of hydrogen, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 24b and R 24c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo
  • s and t are each independently 1, 2, or 3;
  • X is selected from the group consisting of O, S, and NR;
  • Y is selected from the group consisting of O, S, and NR ;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • the present disclosure provides compounds having
  • Formula I that inhibit the interaction between p53 or p53 -related proteins and MDM2 or MDM2 -related proteins.
  • the present disclosure provides methods to induce senescence, cell cycle arrest and/or apoptosis in cells containing functional p53 or p53 -related proteins, comprising contacting the cell with a compound of Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof,
  • the present disclosure provides methods of treating, ameliorating, or preventing a hyperproliferative disease, e.g., cancer, e.g., adrenal cortical cancer, advanced cancer, anal cancer, aplastic anemia, bile duct cancer, bladder cancer, bone cancer, bone metastasis, brain/CNS tumors in adults, brain/CNS tumors in children, breast cancer, breast cancer in men, cancer in children, cancer of unknown primary, Castleman disease, cervical cancer, colon/rectum cancer, endometrial cancer, esophagus cancer, Ewing family of tumors, eye cancer, gallbladder cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumor (GIST), gestational trophoblastic disease, Hodgkin disease, Kaposi sarcoma, kidney cancer, laryngeal and hypopharyngeal cancer, leukemia - acute lymphocytic (ALL) in adults, leukemia - acute myeloid (AML), leukemia
  • the present disclosure provides methods of treating, ameliorating, or preventing a hyperproliferative disease, e.g., cancer, in a patient comprising administering to the patient a compound of Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, in combination with one or more additional therapeutic agents, e.g., one or more additional anticancer agents.
  • a hyperproliferative disease e.g., cancer
  • additional therapeutic agents e.g., one or more additional anticancer agents.
  • compositions comprising a compound of Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, for treating, ameliorating, or preventing a hyperproliferative disease, e.g., cancer, in a patient.
  • a hyperproliferative disease e.g., cancer
  • kits comprising a compound of Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, for treating, ameliorating, or preventing a hyperproliferative disease, e.g., cancer, in a patient.
  • a hyperproliferative disease e.g., cancer
  • Fig. 1 is a line graph showing cell growth inhibition of MDM2 inhibitors in the SJSA-1 cell line as determined using the WST-based assay. SJSA-1 cells were treated with each compound for 4 days.
  • Fig. 2 is a line graph showing cell growth inhibition of MDM2 inhibitors in the RS4; 1 1 leukemia cell line as determined using the WST-based assay. RS4; 1 1 cells were treated with each compound for 4 days.
  • Fig. 3 is a line graph showing the antitumor activity of Compound
  • Example Nos. 22 and 24 in the SJSA-1 xenograft tumor model Mice bearing SJSA-1 tumors (one tumor per mouse) were treated with Compound Example
  • Fig. 4 is a line graph showing the stability of MDM2 inhibitors in a 1 : 1 methanol/water solution.
  • the Y-axis represents the percent amount of the compound measured by UPLC.
  • the X-axis represents the number of days the sample has been in the methanol/water solution.
  • Fig. 5 is a line graph showing the stability of MDM2 inhibitors in a 1 : 1 acetonitrile/water solution.
  • the Y-axis represents the percent amount of the stable isomer measured by UPLC.
  • the X-axis represents the number of days the sample has been in the acetonitrile/water solution.
  • Fig. 6 is a line graph showing the stability of Compound Example Nos. 10 and 1 1 in a 1 : 1 methanol/water solution with 10% TFA added.
  • the Y-axis represents the percent amount of the compound measured by UPLC.
  • the X-axis represents the number of hours the sample has been in solution.
  • compounds having Formulae I-XXVIII inhibit the interaction between p53 or p53-related proteins and MDM2 or MDM2 -related proteins. By inhibiting the negative effect of MDM2 or MDM2-related proteins on p53 or p53 -related proteins, these compounds sensitize cells to inducers of apoptosis and/or cell cycle arrest. In one embodiment, compounds having Formulae I-XXVIII induce apoptosis and/or cell cycle arrest.
  • the methods comprise contacting the cells with one or more compounds having Formulae I-XXVIII alone or in combination with additional agent(s), e.g., an inducer of apoptosis or a cell cycle disrupter.
  • additional agent(s) e.g., an inducer of apoptosis.
  • Such disorders include those characterized by a dysregulation of apoptosis and those characterized by the proliferation of cells expressing functional p53 or p53-related proteins.
  • methods of protecting normal (e.g., non-hyperproliferative) cells in an animal from the toxic side effects of chemotherapeutic agents and treatments are provided. This method comprises administering to the animal one or more compounds having Formulae I-XXVIII.
  • Formulae I-XXVIII for use in the manufacture of a medicament for treating a hyperproliferative disease such as cancer.
  • anticancer agent refers to any therapeutic agent (e.g., chemotherapeutic compound and/or molecular therapeutic compound), antisense therapy, radiation therapy, or surgical intervention, used in the treatment of hyperproliferative diseases such as cancer (e.g., in mammals, e.g.., in humans).
  • therapeutic agent e.g., chemotherapeutic compound and/or molecular therapeutic compound
  • antisense therapy e.g., radiation therapy, or surgical intervention
  • prodrug refers to a pharmacologically inactive derivative of a parent "drug” molecule that requires biotransformation (e.g., either spontaneous or enzymatic) within the target physiological system to release, or to convert (e.g., enzymatically, physiologically, mechanically, electromagnetically) the prodrug into the active drug.
  • Prodrugs are designed to overcome problems associated with stability, water solubility, toxicity, lack of specificity, or limited bioavailability.
  • Exemplary prodrugs comprise an active drug molecule itself and a chemical masking group (e.g., a group that reversibly suppresses the activity of the drug).
  • prodrugs are variations or derivatives of compounds that have groups cleavable under metabolic conditions.
  • Prodrugs can be readily prepared from the parent compounds using methods known in the art, such as those described in A Textbook of Drug Design and Development, Krogsgaard-Larsen and H. Bundgaard (eds.), Gordon & Breach, 1991, particularly Chapter 5: "Design and Applications of Prodrugs”; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; Prodrugs: Topical and Ocular Drug Delivery, K. B. Sloan (ed.), Marcel Dekker, 1998; Methods in Enzymology, K. Widder et al. (eds.), Vol.
  • prodrugs become pharmaceutically active in vivo or in vitro when they undergo solvolysis under physiological conditions or undergo enzymatic degradation or other biochemical transformation (e.g., phosphorylation, hydrogenation, dehydrogenation, glycosylation).
  • Prodrugs often offer advantages of water solubility, tissue compatibility, or delayed release in the mammalian organism. (See e.g., Bundgard, Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam (1985); and Silverman, The Organic Chemistry of Drug Design and Drug Action, pp. 352-401, Academic Press, San Diego, CA (1992)).
  • Common prodrugs include acid derivatives such as esters prepared by reaction of parent acids with a suitable alcohol (e.g., a lower alkanol) or esters prepared by reaction of parent alcohol with a suitable carboxylic acid, (e.g., an amino acid), amides prepared by reaction of the parent acid compound with an amine, basic groups reacted to form an acylated base derivative (e.g., a lower alkylamide), or phosphorus-containing derivatives, e.g., phosphate, phosphonate, and phosphoramidate esters, including cyclic phosphate, phosphonate, and phosphoramidate, see, e.g., US 2007/0249564 Al.
  • a suitable alcohol e.g., a lower alkanol
  • amides prepared by reaction of the parent acid compound with an amine basic groups reacted to form an acyl
  • metabolically cleavable group refers to groups which can be cleaved from the parent molecule by metabolic processes and be substituted with hydrogen. Certain compounds containing metabolically cleavable groups may be prodrugs, i.e., they are pharmacologically inactive. Certain other compounds containing metabolically cleavable groups may be antagonists of the interaction between p53 and MDM2. In such cases, these compounds may have more, less, or equivalent activity of the parent molecule.
  • metabolically cleavable groups include those derived from amino acids (see, e.g., US 2006/0241017 Al ; US 2006/0287244 Al; and WO 2005/046575 A2) or phosphorus-containing compounds (see, e.g., U.S. 2007/0249564 Al) as illustrated in Scheme 1.
  • salt refers to any salt (e.g., obtained by reaction with an acid or a base) of a compound provided herein that is physiologically tolerated in the target animal (e.g., a mammal). Salts of the compounds of provided herein may be derived from inorganic or organic acids and bases.
  • acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, sulfonic, naphthalene-2-sulfonic, benzenesulfonic acid, and the like.
  • Other acids such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds provided herein and their pharmaceutically acceptable acid addition salts.
  • bases include, but are not limited to, alkali metal (e.g., sodium) hydroxides, alkaline earth metal (e.g., magnesium) hydroxides, ammonia, and compounds of formula NW , wherein W is C 1-4 alkyl, and the like.
  • alkali metal e.g., sodium
  • alkaline earth metal e.g., magnesium
  • W is C 1-4 alkyl
  • salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, chloride, bromide, iodide, 2-hydroxyethanesulfonate, lactate, maleate, mesylate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate,
  • salts include anions of the compounds provided herein compounded with a suitable cation such as Na + , NH 4 + , and NW 4 + (wherein W is a Ci-4 alkyl group), and the like.
  • a suitable cation such as Na + , NH 4 + , and NW 4 + (wherein W is a Ci-4 alkyl group), and the like.
  • salts of the compounds provided herein are contemplated as being pharmaceutically acceptable.
  • salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
  • solvate refers to the physical association of a compound provided herein with one or more solvent molecules, whether organic or inorganic. This physical association often includes hydrogen bonding. In certain instances, the solvate is capable of isolation, for example, when one or more solvate molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate” encompasses both solution-phase and isolable solvates. Exemplary solvates include hydrates, ethanolates, and methanolates.
  • the term "monovalent pharmaceutically acceptable cation" as used herein refers to inorganic cations such as, but not limited to, alkaline metal ions, e.g., Na + and K + , as well as organic cations such as, but not limited to, ammonium and substituted ammonium ions, e.g., NH 4 , NHMe 3 , NH 2 Me 2 , NHMe 3 and NMe 4 + .
  • divalent pharmaceutically acceptable cation refers to inorganic cations such as, but not limited to, alkaline earth metal cations, e.g., Ca 2+ and Mg 2+ .
  • terapéuticaally effective amount refers to that amount of the therapeutic agent (including the compounds and compositions of matter provided herein) sufficient to result in amelioration of one or more symptoms of a disorder, or prevent advancement of a disorder, or cause regression of the disorder.
  • a therapeutically effective amount can refer to the amount of a therapeutic agent that decreases the rate of tumor growth, decreases tumor mass, decreases the number of metastases, increases time to tumor progression, increase tumor cell apoptosis, or increases survival time by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%.
  • sensitize and “sensitizing,” as used herein, refer to making, through the administration of a first therapeutic agent (e.g., a compound provided herein), an animal or a cell within an animal more susceptible, or more responsive, to the biological effects (e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell division, cell growth, proliferation, invasion, angiogenesis, necrosis, or apoptosis) of a second therapeutic agent.
  • a first therapeutic agent e.g., a compound provided herein
  • the biological effects e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell division, cell growth, proliferation, invasion, angiogenesis, necrosis, or apoptosis
  • the sensitizing effect of a first agent on a target cell can be measured as the difference in the intended biological effect (e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell growth, proliferation, invasion, angiogenesis, or apoptosis) observed upon the administration of a second agent with and without administration of the first agent.
  • the intended biological effect e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell growth, proliferation, invasion, angiogenesis, or apoptosis
  • the response of the sensitized cell can be increased by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 150%, at least about 200%, at least about 250%, at least 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500% over the response in the absence of the first agent.
  • the term "dysregulation of apoptosis,” as used herein, refers to any aberration in the ability of (e.g., predisposition) a cell to undergo cell death via apoptosis.
  • Dysregulation of apoptosis is associated with or induced by a variety of conditions, non-limiting examples of which include, autoimmune disorders (e.g., systemic lupus erythematosus, rheumatoid arthritis, graft-versus-host disease, myasthenia gravis, or Sj5gren's syndrome), chronic inflammatory conditions (e.g., psoriasis, asthma or Crohn's disease), hyperproliferative disorders (e.g., tumors, B cell lymphomas, or T cell lymphomas), viral infections (e.g., herpes, papilloma, or HIV), and other conditions such as osteoarthritis and atherosclerosis. It should be noted that when the dysregulation is induced by or
  • the term "functional p53,” as used herein, refers to wild-type p53 expressed at normal, high, or low levels and mutant or allelic variants of p53 that retain(s) at least about 5% of the activity of wild-type p53, e.g., at least about 10%, about 20%, about 30%, about 40%, about 50%, or more of wild-type activity.
  • p53 -related protein refers to proteins that have at least 25% sequence homology with p53, have tumor suppressor activity, and are inhibited by interaction with MDM2 or MDM2 -related proteins.
  • p53-related proteins include, but are not limited to, p63 and p73.
  • MDM2 -related protein refers to proteins that have at least 25% sequence homology with MDM2, and interact with and inhibit p53 or p53-related proteins. Examples of MDM2-related proteins include, but are not limited to, MDMX.
  • Senescence refers to the phenomenon whereby non-cancerous diploid cells lose the ability to divide, and characterized in part by telomeric dysfunction or shortening.
  • hyperproliferative disease refers to any condition in which a localized population of proliferating cells in an animal is not governed by the usual limitations of normal growth.
  • hyperproliferative disorders include tumors, neoplasms, lymphomas, leukemias and the like.
  • a neoplasm is said to be benign if it does not undergo invasion or metastasis and malignant if it does either of these.
  • a "metastatic" cell means that the cell can invade neighboring body structures.
  • Hyperplasia is a form of cell proliferation involving an increase in cell number in a tissue or organ without significant alteration in structure or function.
  • Metaplasia is a form of controlled cell growth in which one type of fully differentiated cell substitutes for another type of differentiated cell.
  • autoimmune disorder refers to any condition in which an organism produces antibodies or immune cells which recognize the organism's own molecules, cells or tissues.
  • Non-limiting examples of autoimmune disorders include autoimmune hemolytic anemia, autoimmune hepatitis, Berger's disease or IgA nephropathy, celiac sprue, chronic fatigue syndrome, Crohn's disease, dermatomyositis, fibromyalgia, graft versus host disease, Grave's disease, Hashimoto's thyroiditis, idiopathic thrombocytopenia purpura, lichen planus, multiple sclerosis, myasthenia gravis, psoriasis, rheumatic fever, rheumatic arthritis, scleroderma, Sj5gren's syndrome, systemic lupus erythematosus, type 1 diabetes, ulcerative colitis, vitiligo, and the like.
  • neoplastic disease refers to any abnormal growth of cells being either benign (non-cancerous) or malignant (cancerous).
  • normal cell refers to a cell that is not undergoing abnormal growth or division. Normal cells are non-cancerous and are not part of any hyperproliferative disease or disorder.
  • anti-neoplastic agent refers to any compound that retards the proliferation, growth, or spread of a targeted (e.g., malignant) neoplasm.
  • prevention refers to a decrease in the occurrence of pathological cells (e.g., hyperproliferative or neoplastic cells) in an animal.
  • the prevention may be complete, e.g., the total absence of pathological cells in a subject.
  • the prevention may also be partial, such that the occurrence of pathological cells in a subject is less than that which would have occurred without treatment with one or more compounds provided herein.
  • apoptosis-modulating agents refers to agents which are involved in modulating (e.g., inhibiting, decreasing, increasing, promoting) apoptosis.
  • apoptosis-modulating agents include proteins which comprise a death domain such as, but not limited to, Fas/CD95, TRAMP, TNF RI, DRl, DR2, DR3, DR4, DR5, DR6, FADD, and RIP.
  • apoptosis-modulating agents include, but are not limited to, TNFa, Fas ligand, antibodies to Fas/CD95 and other TNF family receptors, TRAIL (also known as Apo2 Ligand or Apo2L/TRAIL), antibodies to TRAIL-Rl or TRAIL-R2, Bcl-2, p53, BAX, BAD, Akt, CAD, PI3 kinase, PPl, and caspase proteins.
  • Modulating agents broadly include agonists and antagonists of TNF family receptors and TNF family ligands.
  • Apoptosis-modulating agents may be soluble or membrane bound (e.g. ligand or receptor).
  • Apoptosis-modulating agents include those which are inducers of apoptosis, such as TNF or a TNF-related ligand, particularly a TRAMP ligand, a Fas/CD95 ligand, a TNFR-1 ligand, or TRAIL.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable vehicle” encompasses any of the standard pharmaceutical carriers, solvents, surfactants, or vehicles. Suitable pharmaceutically acceptable vehicles include aqueous vehicles and nonaqueous vehicles. Standard pharmaceutical carriers and their formulations are described in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, PA, 19th ed. 1995.
  • Pulsatile dosing refer to intermittent (i.e., not continuous) administration of compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof, to a patient.
  • Pulsatile dose administration regimens useful in the present disclosure encompass any discontinuous administration regimen that provides a therapeutically effective amount of compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof, to a patient in need thereof.
  • Pulsatile dosing regimens can use equivalent, lower, or higher doses of compounds having Formulae I-XXVIII than would be used in continuous dosing regimens.
  • Advantages of pulsatile dose administration of compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof include, but are not limited to, improved safety, decreased toxicity, increased exposure, increased efficacy, and increased patient compliance. These advantages may be realized when compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof, are administered as a single agent or are administered in combination with one or more additional anticancer agents.
  • compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof are scheduled to be administered to the patient, administration can occur in a single or in divided doses, e.g., once-a-day, twice-a-day, three times a day, four times a day or more.
  • compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof are administered once (QD) or twice (BID) on the day it is schedule to be administered
  • alkyl refers to a straight-chain or branched saturated aliphatic hydrocarbon having from one to eighteen carbons or the number of carbons designated (e.g., Ci-Cis means 1 to 18 carbons).
  • the alkyl is a Ci-Cio alkyl.
  • the alkyl is a C1-C6 alkyl.
  • the alkyl is a C1-C4 alkyl.
  • the alkyl is a C1-C3 alkyl.
  • the alkyl is a C2-C1 0 alkyl.
  • the alkyl is a C3-C1 0 alkyl. In another embodiment, the alkyl is a C3-C6 alkyl.
  • Exemplary alkyl groups include methyl, ethyl, w-propyl, isopropyl, w-butyl, sec-butyl, isobutyl, tert-butyl, w-pentyl, isopentyl, neopentyl, w-hexyl, isohexyl, w-heptyl, 4,4-dimethylpentyl, w-octyl, 2,2,4-trimethylpentyl, nonyl, decyl and the like.
  • alkyl as used herein by itself or part of another group means that the alkyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from hydroxy (i.e. , -OH), nitro (i.e. , -N0 2 ), cyano (i.e.
  • R c is hydrogen, optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl;
  • R d is optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl;
  • R e is hydrogen, optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl;
  • R f is hydrogen, optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl;
  • the optionally substituted alkyl is substituted with two substituents. In another embodiment, the optionally substituted alkyl is substituted with one substituent. In another embodiment, the substituents are selected from hydroxyl (i.e. , a hydroxyalkyl, e.g. , a monohydroxyalkyl or dihydroxyalkyl), optionally substituted cycloalkyl (i.e. , a (cycloalkyl)alkyl), optionally substituted heterocyclo (i.e., a (heterocyclo)alkyl), -CO2H, or amino (i.e. , an aminoalkyl).
  • hydroxyl i.e. , a hydroxyalkyl, e.g. , a monohydroxyalkyl or dihydroxyalkyl
  • optionally substituted cycloalkyl i.e. , a (cycloalkyl)alkyl
  • optionally substituted heterocyclo i.e., a (he
  • Exemplary optionally substituted alkyl groups include -CH2OCH3, -CH2CH2NH2, -CH 2 CH 2 NH(CH 3 ), -CH2CH2CN, -CH2CO2H, -CH2CONH2, -CH2SO2CH 3 , -CH2CH2SO2CH 3 , -C(CH 3 ) 2 C0 2 H, hydroxymethyl, hydroxyethyl, hydroxypropyl, and the like.
  • alkylenyl as used herein by itself or part of another group refers to a divalent alkyl radical containing one, two, three, four, or more joined methylene groups.
  • exemplary alkylenyl groups include -((3 ⁇ 4)-, -(CH 2 ) 2 -, - (CH 2 ) 3 -, -(CH 2 ) 4 -, and the like.
  • optionally substituted alkylenyl as used herein by itself or part of another group means the alkylenyl as defined above is either unsubstituted or substituted with one, two, three, or four substituents independently selected from the group consisting of optionally substituted Ci-Ce alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl.
  • the optionally substituted Ci-Ce alkyl is methyl.
  • the optionally substituted aryl is a phenyl optionally substituted with one or two halo groups.
  • Exemplary optionally substituted alkylenyl groups include -CH(CH 3 )-, -C(CH 3 ) 2 -, -CH 2 CH(CH 3 )-, -CH 2 CH(CH 3 )CH 2 -, -CH 2 CH(Ph)CH 2 -, -CH(CH 3 )CH(CH 3 )-, and the like.
  • haloalkyl as used herein by itself or part of another group refers to an alkyl as defined above having one to six halo substituents. In one embodiment, the haloalkyl has one, two or three halo substituents. Exemplary haloalkyl groups include trifluoromethyl, -CH 2 CH 2 F and the like.
  • hydroxyalkyl as used herein by itself or part of another group refers to an alkyl as defined above having exactly one hydroxy substituent.
  • exemplary hydroxyalkyl groups include hydroxymethyl, hydroxyethyl, hydroxypropyl, and the like.
  • dihydroxyalkyl as used herein by itself or part of another group refers to alkyl as defined above having exactly two hydroxyl substituents.
  • exemplary dihydroxyalkyl groups include -CH 2 CH 2 CCH 3 (OH)CH 2 OH, -CH 2 CH 2 CH(OH)CH(CH 3 )OH, -CH 2 (OH)CH 2 OH, -CH 2 CH(CH 2 OH) 2 , -CH 2 CH 2 CH(OH)C(CH 3 ) 2 OH, -CH 2 CH 2 CCH 3 (OH)CH(CH 3 )OH, and the like, including stereoisomers thereof.
  • hydroxycycloalkyl as used herein by itself or part of another group refers to an optionally substituted cycloalkyl as defined below having a least one, e.g. , one or two hydroxy substituents.
  • exemplary hydroxycycloalkyl groups include:
  • optionally substituted (cycloalkyl)alkyl refers to an optionally substituted alkyl as defined above having an optionally substituted cycloalkyl (as defined below) substituent.
  • exemplary optionally substituted (cycloalkyl)alkyl groups include:
  • heterocycloalkyl as used herein by itself or part of another group refers to an alkyl as defined above having an optionally substituted heterocyclo (as defined below) substituent.
  • aralkyl refers to an optionally substituted alkyl as defined above having one, two or three optionally substituted aryl substituents. In one embodiment, the aralkyl has two optionally substituted aryl substituents. In another embodiment, the aralkyl has one optionally substituted aryl substituent. In another embodiment, the aralkyl is an aryl(Ci-C4 alkyl). In another embodiment, the aryl(Ci-C4 alkyl) has two optionally substituted aryl substituents. In another embodiment, the aryl(Ci-C 4 alkyl) has one optionally substituted aryl substituent.
  • Exemplary aralkyl groups include, for example, benzyl, phenylethyl, (4-fluorophenyl)ethyl, phenylpropyl, diphenylmethyl (i.e. , Ph 2 CH-), diphenylethyl (Ph 2 CHCH 2 -) and the like.
  • cycloalkyl refers to saturated and partially unsaturated (containing one or two double bonds) cyclic hydrocarbon groups containing one to three rings having from three to twelve carbon atoms (i.e. , C3-C12 cycloalkyl) or the number of carbons designated.
  • the cycloalkyl has one ring.
  • the cycloalkyl is a C3-C6 cycloalkyl.
  • Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, norbornyl, decalin, adamantyl and the like.
  • cycloalkyl as used herein by itself or part of another group means the cycloalkyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, -C0 2 R c , -COR c , -S0 2 R d , -N(R e )COR f , -N(R e )S0 2 R
  • optionally substituted cycloalkyl also means the cycloalkyl as defined above may be fused to an optionally substituted aryl.
  • the optionally substituted cycloalkyl is substituted with two substituents.
  • the optionally substituted cycloalkyl is substituted with one substituent.
  • the substituents are selected from hydroxy (i.e., a hydroxycycloalkyl, e.g., a monohydroxycycloalkyl or dihydroxycycloalkyl) or -CO 2 H.
  • exemplary optionally substituted cycloalkyl groups include:
  • alkenyl as used herein by itself or part of another group refers to an alkyl group as defined above containing one, two or three carbon-to-carbon double bonds. In one embodiment, the alkenyl has one carbon-to-carbon double bond.
  • optionally substituted alkenyl as used herein by itself or part of another group means the alkenyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido.
  • cycloalkenyl as used herein by itself or part of another group refers to a cycloalkyl group as defined above containing one, two or three carbon- to-carbon double bonds. In one embodiment, the cycloalkenyl has one carbon-to- carbon double bond.
  • exemplary cycloalkenyl groups include cyclopentene, cyclohexene and the like.
  • cycloalkenyl as used herein by itself or part of another group means the cycloalkenyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido.
  • alkynyl as used herein by itself or part of another group refers to an alkyl group as defined above containing one to three carbon-to-carbon triple bonds. In one embodiment, the alkynyl has one carbon-to-carbon triple bond.
  • exemplary alkynyl groups include -C ⁇ CH, -C ⁇ CCH 3 , -CH 2 C ⁇ CH, - CH 2 CH 2 C ⁇ CH and -CH 2 CH 2 C ⁇ CCH 3 .
  • alkynyl as used herein by itself or part of another group means the alkynyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido.
  • exemplary optionally substituted alkenyl groups include -C ⁇ CPh, -CH 2 C ⁇ CPh and the like.
  • aryl as used herein by itself or part of another group refers to monocyclic and bicyclic aromatic ring systems having from six to fourteen carbon atoms (i.e., C6-C14 aryl) such as phenyl (abbreviated as Ph), 1-naphthyl and 2-naphthyl and the like.
  • aryl as used herein by itself or part of another group means the aryl as defined above is either unsubstituted or substituted with one to five substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, -CONHS0 2 Me, -C0 2 R c , -COR c , -S0 2 R d , - N(R e )COR f , -N(R e )S0 2 R s
  • the optionally substituted aryl is an optionally substituted phenyl. In one embodiment, the optionally substituted phenyl has four substituents. In another embodiment, the optionally substituted phenyl has three substituents. In another embodiment, the optionally substituted phenyl has two substituents. In another embodiment, the optionally substituted phenyl has one substituent.
  • Exemplary substituted aryl groups include 2-methylphenyl, 2- methoxyphenyl, 2-fluorophenyl, 2-chlorophenyl, 2-bromophenyl, 3- methylphenyl, 3 -methoxyphenyl, 3 -fluorophenyl, 3-chlorophenyl, 4- methylphenyl, 4-ethylphenyl, 4-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 2,6-di- fluorophenyl, 2,6-di-chlorophenyl, 2-methyl, 3 -methoxyphenyl, 2-ethyl, 3- methoxyphenyl, 3,4-di-methoxyphenyl, 3,5-di-fluorophenyl 3,5-di-methylphenyl and 3,5-dimethoxy, 4-methylphenyl, 2-fluoro-3-chlorophenyl, 3-chloro-4- fluorophenyl, 4-C0 2 H-
  • heteroaryl refers to monocyclic and bicyclic aromatic ring systems having from five to fourteen ring atoms (i.e., 5- to 14-membered heteroaryl) and one, two, three, or four heteroatoms independently selected from the group consisting of oxygen, nitrogen and sulfur.
  • the heteroaryl has three heteroatoms.
  • the heteroaryl has two heteroatoms.
  • the heteroaryl has one heteroatom.
  • the heteroaryl is a 5-membered heteroaryl.
  • the heteroaryl is a 6-membered heteroaryl.
  • the heteroaryl is a 6-membered heteroaryl having one or two nitrogen atoms.
  • Exemplary heteroaryl groups include
  • heteroaryl is meant to include possible N-oxides.
  • Exemplary N-oxides include pyridyl N-oxide and the like.
  • heteroaryl as used herein by itself or part of another group means the heteroaryl as defined above is either unsubstituted or substituted with one to four substituents, typically one or two substituents, independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, -C0 2 R c , -COR c , -S0 2 R d , -N(R e )COR f , -N(R e )S0 2 R
  • the optionally substituted heteroaryl has one substituent.
  • the substituent is an optionally substituted aryl, aralkyl, or optionally substituted alkyl.
  • the substituent is an optionally substituted phenyl. Any available carbon or nitrogen atom may be substituted.
  • Exemplary optionally substituted heteroaryl groups include
  • heterocyclo refers to saturated and partially unsaturated (containing one or two double bonds) cyclic groups containing one to three rings having from three to fourteen ring members (i.e., 3- to 14-membered heterocyclo) and at least one oxygen, sulfur, including sulfoxide and sulfone, and/or nitrogen atom.
  • the heterocyclo group is chosen from a 5- or 6-membered cyclic group containing one ring and one or two oxygen and/or nitrogen atoms.
  • the heterocyclo group is a 6-membered cyclic group containing one ring and one sulfur atom, including sulfoxide and sulfone.
  • the heterocyclo can be optionally linked to the rest of the molecule through a carbon or nitrogen atom.
  • Exemplary heterocyclo groups include:
  • heterocyclo as used herein by itself or part of another group means the heterocyclo as defined above is either unsubstituted or substituted with one to four substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, -C0 2 R c , -COR c , -S0 2 d , -N(R e )COR f , -N(R e )S0 2 R s or -N(R e
  • An optionally substituted heterocyclo may be fused to an aryl group to provide an optionally substituted aryl as described above.
  • alkoxy refers to a haloalkyl, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl or optionally substituted alkynyl attached to a terminal oxygen atom.
  • aryloxy as used herein by itself or part of another group refers to an optionally substituted aryl attached to a terminal oxygen atom.
  • exemplary aryloxy groups include phenoxy and the like.
  • aralkyloxy refers to an aralkyl attached to a terminal oxygen atom.
  • exemplary aralkyloxy groups include benzyloxy and the like.
  • alkylthio refers to a haloalkyl, aralkyl, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl or optionally substituted alkynyl attached to a terminal sulfur atom.
  • alkyl groups include -SCH 3 and the like.
  • halo or halogen as used herein by itself or part of another group refers to fluoro, chloro, bromo or iodo. In one embodiment, the halo is fluoro or chloro.
  • amino refers to a radical of formula -NR a R b wherein R a and R b are independently hydrogen, haloalkyl, aralkyl, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl or optionally substituted heteroaryl; or R a and R b taken together with the nitrogen atom to which they are attached form a four to seven membered optionally substituted heterocyclo.
  • exemplary amino groups include -NH 2 , -N(H)CH 3 , -N(CH 3 ) 2 ,
  • carboxamido as used herein by itself or part of another group refers to a radical of formula -CO-amino.
  • exemplary carboxamido groups include -CONH 2 , -CON(H)CH 3 , -CON(H)Ph, -CON(H)CH 2 CH 2 Ph, - CON(CH 3 ) 2 , CON(H)CHPh 2 and the like.
  • sulfonamido as used herein by itself or part of another group refers to a radical of formula -S0 2 -amino.
  • exemplary sulfonamido groups include -S0 2 NH 2 , -S0 2 N(H)CH 3 , -S0 2 N(H)Ph and the like.
  • Certain of the compounds of the present disclosure may exist as stereoisomers, i.e., isomers that differ only in the spatial arrangement of atoms, including optical isomers and conformational isomers (or conformers).
  • the disclosure includes all stereoisomers, both as pure individual stereoisomer preparations and enriched preparations of each, and both the racemic mixtures of such stereoisomers as well as the individual diastereomers and enantiomers that may be separated according to methods that are well known to those of skill in the art.
  • substantially free of as used herein means that the compound comprises less than about 25% of other stereoisomers, e.g., diastereomers and/or enantiomers, as established using conventional analytical methods routinely used by those of skill in the art.
  • the amount of other stereoisomers is less than about 24%, less than about 23%, less than about 22%, less than about 21%, less than about 20%, less than about 19%, less than about 18%, less than about 17%, less than about 16%, less than about 15%, less than about 14%, less than about 13%, less than about 12%, less than about 1 1%, less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, less than about 1%, or less than about 0.5%.
  • Stereoisomerically enriched compounds that contain about 95% or more of a desired stereoisomer, for example, about 96% or more, about 97% or more, about 98% or more, or about 99% or more are referred to herein as "substantially pure stereoisomers.”
  • Stereoisomerically enriched compounds that contain about 99% or more of a desired stereoisomer are referred to herein as "pure" stereoisomers.”
  • the purity of any stereoisomerically enriched compound can be determined using conventional analytical methods such as, for example, normal phase HPLC, reverse phase HPLC, chiral HPLC, and X H and 13 C NMR.
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
  • R 2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
  • E is selected from the group consisting of -OR 26a and -NR 26b R 26c ;
  • R 26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
  • R 26b is R 4 ;
  • R 26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, -S0 2 R 5b , and R 5 ;
  • R 4 and R 5 have the meanings as described below for Formula II; or R 26b and R 26c taken together form a 4- to 9-membered optionally substituted heterocyclo;
  • R 5b is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, and optionally substituted heteroaryl;
  • X is selected from the group consisting of O, S, and NR;
  • Y is selected from the group consisting of O, S, and NR ;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
  • R 2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
  • E is selected from the group consisting of -OR 26a and -NR 26b R 26c ;
  • R 26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
  • R 26b is selected from the group consisting of hydrogen and optionally substituted alkyl
  • R 26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, and -S0 2 R 5b ; or R and R c taken together form a 4- to 9-membered optionally substituted heterocyclo;
  • R 5b is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, and optionally substituted heteroaryl;
  • X is selected from the group consisting of O, S, and NR;
  • Y is selected from the group consisting of O, S, and NR ;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
  • R 2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
  • R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
  • E is selected from the group consisting of -OR 26a and -NR 26b R 26c ;
  • R 26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
  • R 26b is selected from the group consisting of hydrogen and optionally substituted alkyl
  • R 26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, and -S0 2 R 5b ; wherein R 4 and R 5 have the meanings as described below for Formula II; or R 26b and R 26c taken together form a 4- to 9-membered optionally substituted heterocyclo;
  • R 5b is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, and optionally substituted heteroaryl;
  • X is selected from the group consisting of O, S, and NR;
  • Y is selected from the group consisting of O, S, and NR ;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
  • R 2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl
  • R 4 is selected from the group consisting of hydrogen and optionally substituted alkyl
  • R 5 is selected from the group consisting of:
  • each R 6a and R 6b is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
  • R 7 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
  • R 8a and R 8b are each independently selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl; or
  • R 8a and R 8b taken together with the carbon that they are attached form a 3- to 8-membered optionally substituted cycloalkyl
  • W 1 is selected from the group consisting of -OR 9a and -NR 9b R 9c ;
  • R 9a is hydrogen
  • R 9b is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S0 2 R 9d , and -CONR 9e R 9f ;
  • R 9c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or R 9b and R 9c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R 9d is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
  • R 9e and R 9f are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
  • R 9e and R 9f taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • W 2 is selected from the group consisting of -OR 10 and -NR l la R l lb ;
  • R 10 is hydrogen
  • R 9a and R 10 are hydrogen and the other is a metabolically cleavable group
  • R l la is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S0 2 R l lc , and -CONR l ld R l le ;
  • R l lb is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R l la and R l lb taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R l lc is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
  • R l ld and R l le are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
  • n 1, 2, 3, 4, or 5;
  • each R 12a , R 12b , R 12c and R 12d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
  • R 13 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
  • R is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
  • Z is selected from the group consisting of -OR 15 and -NR 16a R 16b ; or
  • Z and R 14 taken together form a carbonyl, i.e. , a C 0, group.
  • R 15 is selected from the group consisting of hydrogen and metabolically cleavable group
  • R 16a is selected from the group consisting of -S0 2 R 16c and -CONR 16d R 16e ;
  • R 16b is selected from the group consisting of hydrogen and optionally substituted alkyl;
  • R 16c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 16d and R 16e are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 16d and R 16e taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo
  • o is 1 , 2, or 3 ;
  • p 0, 1 , 2, or 3 ;
  • each R 17a , R 17b , R 17c and R 17d is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
  • R 18 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl
  • R 19 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
  • R 20 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
  • R 21a and R 21b are each hydrogen;
  • R 21a and R 21b are hydrogen and the other is metabolically cleavable group
  • q 0, 1 , 2, or 3 ;
  • each R a , R , R c , and R is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
  • R 23 is selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl
  • R 24 is selected from the group consisting of -S0 2 R 24a and -CONR 24b R 24c ;
  • R 24a is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 24b and R 24c are each independently selected from the group consisting of hydrogen, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 24b and R 24c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo
  • s and t are each independently 1 , 2, or 3;
  • X is selected from the group consisting of O, S, and NR ;
  • Y is selected from the group consisting of O, S, and NR ;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • the compound of Formula I or II is provided as a mixture of stereoisomers, e.g., a mixture of diastereomers and/or enantiomers, e.g., a racemic mixture.
  • the compound of Formula I or II is provided as a single stereoisomer.
  • R la , R lb , R lc , R ld , R 2 , R 3a , R 3b , E, X, and Y have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • E is NR 4 R 5 , and R la , R lb , R lc , R ld , R 2 , R 3a , R 3b , R 4 , R 5 , X, and Y have the meanings as described above for Formula II, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R la , R lb , R lc , R ld , R 2 , R 3a , R 3b , E, X, and Y have the meanings as described above for Formula I, or tautomer thereof, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • E is -NR 4 R 5 , and R la , R lb , R lc , R ld , R 2 , R 3a , R 3b , R 4 , R 5 , X, and Y have the meanings as described above for Formula II, or tautomer thereof, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R 3a and R 3b of Formula I are taken together form a 3- to 9-membered optionally substituted cycloalkyl, wherein R la , R lb , R lc , R ld , R 2 , E, X, and Y have the meanings as described above for Formula I, u is 0, 1, 2, 3, 4, 5, or 6, v is 0, 1,
  • each R is independently halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula V are provided wherein E is NR 4 R 5 , and R la , R lb , R lc , R ld , R 2 , R 4 , R 5 , X, and Y have the meanings as described above for Formula II, u is 0, 1, 2, 3, 4, 5, or 6, v is 0, 1, 2, 3, or 4, and each R is independently halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido, or a pharmaceutically acceptable salt, solvate, or pro
  • compounds of Formula V are provided wherein u is 1, 2, 3, 4, 5, or 6, each R is independently (Ci-C4)alkyl and v is 0, 1, or 2, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula V are provided wherein v is 0 and u is 0, 1, 2, 3, 4, 5, or 6, , i.e., R 3a and R 3b of Formula I are taken together form a 3 - to 9-membered unsubstituted cycloalkyl.
  • R la , R 1D , R lc , R ia , R z , R Ja , R 3b , E, X, and Y have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae VI-XXI are provided, wherein E is NR 4 R 5 , and R la , R lb , R lc , R ld , R 2 , R 3a , R 3b , R 4 , R 5 , X, and Y have the meanings as described above for Formula II, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae VI-XXI are provided substantially free of one or more other stereoisomers. In another embodiment, compounds of any one of Formulae VI-XXI are substantially pure stereoisomers. In another embodiment, compounds of any one of Formulae VI-XXI are pure stereoisomers.
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, fluoro, and chloro;
  • R la and R ld are hydrogen;
  • R lb is selected from the group consisting of hydrogen and fluoro; and
  • R lc is selected from the group consisting of fluoro and chloro;
  • R 2 is optionally substituted phenyl
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, and optionally substituted cycloalkyl;
  • R 3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, and optionally substituted cycloalkyl;
  • R 3a and R 3b taken together form an optionally substituted 3- to 9-membered cycloalkyl
  • Y is NH
  • X and Y are NH;
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR 4 R 5 , R 5 is R5-1; R 6a and R 6b are hydrogen; R 7 is d-C 4 alkyl; R 8a and R 8b are hydrogen; W is -OR 10 , R 9 and R 10 are hydrogen; and n is 2.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR 4 R 5 ; R 5 is R5-1; R 6a and R 6b are hydrogen; R 7 is d-C 4 alkyl; R 8a and R 8b are hydrogen; W is -NR l la R llb , R 9 is hydrogen; and n is 2.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR 4 R 5 ; R 5 is R5-1; R 6a and R 6b are hydrogen; R 7 is d-C 4 alkyl; R 8a and R 8b are hydrogen; W is -OR 10 , one of R 9 and R 10 is hydrogen and the other is a metabolically cleavable group; and n is 2.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR 4 R 5 ; R 5 is R5-2; R 12a , R 12b , R 12c , and R 12d are each hydrogen; R 13 is hydrogen; Z is -OR 15 and R 15 is hydrogen; o is 1 or 2; and p is 1 or 2.
  • any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR 4 R 5 ; R 5 is R5-2; R 12a , R 12b , R 12c , and R 12d are each hydrogen; R 13 is hydrogen; Z is -NR 16a R 16b ; o is 1 or 2; and p is 1 or 2.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR 4 R 5 ; R 5 is R5-2; R 12a , R 12b , R 12c , and R 12d are each hydrogen; R 13 is hydrogen; Z is -OR 15 and R 15 a metabolically cleavable group; o is 1 or 2; and p is 1 or 2.
  • R 18 , R 19 , and R 20 are hydrogen; R 21a and R 21b are hydrogen; and q and r are 1.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR 4 R 5 ; R 5 is R5-3; R 17a , R 17b , R 17c , and R 17d are each hydrogen; R 18 , R 19 , and R 20 are hydrogen; one of R 21a and R 21b is hydrogen and the other is a metabolically cleavable group; and q and r are 1.
  • R 2 is an optionally substituted aryl having the Formula R2-1 :
  • R 25a , R 25 , R 25c , R 25d , and R 25e are each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, amino, cyano, alkoxy, optionally substituted alkyl, haloalkyl, optionally substituted aryl, and optionally substituted heteroaryl.
  • R 25a is selected from the group consisting of hydrogen and fluoro
  • R 25b is chloro
  • R 25c is selected from the group consisting of hydrogen and fluoro
  • R 25d and R 25e are hydrogen.
  • stereoisomers e.g., enantiomers, thereof, wherein:
  • R 7 is optionally substituted C 1 -C4 alkyl
  • R 9a and R 10 are each hydrogen;
  • R 9a and R 10 are hydrogen and the other is a metabolically cleavable group
  • R 9b is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S0 2 R 9d , and -CONR 9e R 9f ;
  • R 9c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 9b and R 9c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R 9d is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
  • R 9e and R 9f are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or R 9e and R 9f taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R l la is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S0 2 R l lc , and -CONR lld R l le ;
  • R l lb is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R l la and R l lb taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R l lc is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
  • R l ld and R l le are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or R l ld and R l le taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R 14 is selected from the group consisting of hydrogen, C 1 -C 4 alkyl, or C3-C6 cycloalkyl;
  • R 15 is hydrogen or a metabolically cleavable group
  • R 16a is selected from the group consisting of -S0 2 R 16c and -CONR 16d R 16e ;
  • R 16b is selected from the group consisting of hydrogen and optionally substituted alkyl
  • R 16c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 16d and R 16e are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 16d and R 16e taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo
  • R 19 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
  • R is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
  • R 21a and R 21b are each hydrogen;
  • R 21a and R 21b are hydrogen and the other is metabolically cleavable group
  • R 24 is selected from the group consisting of -S0 2 R 24a and -CONR 24b R 24c ;
  • R 24a is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 24b and R 24c are each independently selected from the group consisting of hydrogen, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl, or
  • R 24b and R 24c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR 4 R 5 , R 4 is hydrogen, and R 5 is selected from the group consisting of R5-5, R5-6, R5-10, R5-11, R5-12, R5-13, and R5-14.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR 4 R 5 , R 4 is hydrogen, and R 5 is selected from the group consisting of R5-10 and R5-12 and R 14 is hydrogen or methyl and R 15 is hydrogen.
  • R 7 is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl;
  • R 8a and R 8b are each independently selected from the group consisting of hydrogen, methyl, ethyl, propyl, isopropyl, and cyclopropyl.
  • R 7 is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl;
  • R 8a and R 8b are each independently is selected from the group consisting of hydrogen, methyl, ethyl, propyl, isopropyl, and cyclopropyl;
  • R 9d is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl; and R l lc is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl.
  • R 7 is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl;
  • R 8a and R 8b are each independently is selected from the group consisting of hydrogen, methyl, ethyl, propyl, isopropyl, and cyclopropyl;
  • R 9e is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl;
  • R l ld is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl.
  • R is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl;
  • R 19 and R 20 are each independently is selected from the group consisting of hydrogen, methyl, ethyl, propyl, isopropyl, and cyclopropyl.
  • R 14 is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl; and R c is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl.
  • R is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl;
  • R 16d is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl.
  • R 14 is selected from the group consisting of hydrogen and C1-C4 alkyl; and R 15 is hydrogen or a metabolically cleavable group.
  • the metabolically cleavable group at R 15 is selected from the group consisting of:
  • each R 28a and R 28b is independently selected from the group consisting of hydrogen, optionally substituted alkyl, and aralkyl;
  • R 29a and R 29b are each selected from the group consisting of hydrogen and optionally substituted alkyl
  • v 1, 2, 3, or 4;
  • R 30a and R 30b are each selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, optionally substituted aryl, and monovalent pharmaceutically acceptable cation; or
  • R 30a and R 30b represent a divalent pharmaceutically acceptable cation or an optionally substituted alkylenyl.
  • the metabolically cleavable group at R 15 is the residue of a natural or unnatural amino acid.
  • the metabolically cleavable group at R 15 is the residue of glycine, isoleucine alanine, leucine, asparagine, lysine, aspartic acid, methionine, cysteine, phenylalanine, glutamic acid, threonine, glutamine, tryptophan, valine, proline, serine, tyrosine, arginine, and histidine.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided wherein X is NH and Y is NH.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided wherein X is O and Y is NH.
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R 3a and R 3b are the same or different Ci-Cio alkyl.
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R 3a and R 3b are the same or different C2-C1 0 alkyl.
  • R 3a and R 3b are the same or different C1-C6 alkyl, e.g., methyl, ethyl, M-propyl, isopropyl, w-butyl, sec-butyl, isobutyl, tert-butyl, w-pentyl, isopentyl, neopentyl, w-hexyl, isohexyl.
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R 3a and R 3b are the same or different C2-C6 alkyl.
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R 3a and R 3b are the same or different C1-C4 alkyl.
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R 3a and R 3b are the same or different C1-C3 alkyl.
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R 3a and R 3b are the same or different, and are selected from the group consisting of methyl, ethyl, propyl, isopropyl, butyl, and neopentyl.
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R 3a and R 3b are the same or different, and are selected from the group consisting of ethyl, propyl, isopropyl, butyl, and neopentyl.
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R 3a and R 3b are the same, e.g., R 3a is methyl and R 3b is methyl, R 3a is ethyl and R 3b is ethyl, R 3a is propyl and R 3b is propyl, etc. If R 3a and R 3b are the same, the carbon atom to which R 3a and R 3b are attached is not an asymmetric center.
  • Formula VI and X; Formula VII and XIII; Formula VIII and IX; Formula XI and XII; Formula XIV and XVI; Formula XV and XXI; Formula XVII and XX; and Formula XVIII and XIX represent equivalent isomers.
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R 3a and R 3b are different, e.g., R 3a is methyl and R 3b is ethyl, R 3a is methyl and R 3b is neopentyl, R 3a is ethyl and R 3b is propyl, etc..
  • VI-XXI or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R 3a and R 3b are taken together to form an optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring.
  • the optional substituent is a (Ci-C4)alkyl.
  • the cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring is unsubstituted.
  • compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R la and R ld are hydrogen, R lb and R lc are each independently selected from the group consisting of hydrogen, chloro, and fluoro, and R 2 is R2-1 wherein R 25a , R 25b , R 25c , R 25d , and R 25e are each independently selected from the group consisting of hydrogen, chloro, and fluoro.
  • E is NR 4 R 5 ;
  • R la , R lb , R lc , and R ld are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
  • R 2 is:
  • R 25a , R 25b , R 25c , R 25d , and R 25e are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
  • R 3a is Ci-C 6 alkyl
  • R 3b is C1-C4 alkyl
  • R 3a and R 3b are taken together to form an optionally substituted 3- to 7-membered cycloalkyl
  • R 4 is selected from the group consisting of hydrogen and optionally substituted C1-C4 alkyl
  • R 5 is selected from the group consisting of: wherein:
  • R 14 is selected from the group consisting of hydrogen and optionally substituted C 1 -C 4 alkyl
  • X is selected from the group consisting of O, S, and NR;
  • Y is selected from the group consisting of O, S, and NR ;
  • R is selected from the group consisting of hydrogen and optionally substituted C 1 -C 4 alkyl;
  • R is selected from the group consisting of hydrogen and optionally substituted C 1-C4 alkyl
  • E is NR 4 R 5 ;
  • R la is hydrogen
  • R lb , R lc , and R ld are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
  • R 2 is:
  • R 25a , R 25b , R 25c , R 25d , and R 2Se are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
  • R 3a and R 3b are methyl
  • R 4 is hydrogen
  • R 5 is selected from the group consisting of:
  • X and Y are NH
  • compounds having Formula VI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein the compounds are substantially pure stereoisomers.
  • compounds having Formula XVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein the compounds are substantially pure stereoisomers.
  • compounds having Formula XVI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein the compounds are substantially pure stereoisomers.
  • E is NR 4 R 5 ;
  • R 4 is hydrogen
  • X and Y are NH
  • R 3a and R 3b are methyl or ethyl, or
  • R 3a and R 3b taken together form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring, each optionally substituted by one of more C1-C4 alkyl groups; and R 5 is selected from the group consisting of:
  • compounds of any one of Formulae VI-XXI are provided, wherein E is -OR 26a and R la , R lb , R lc , R ld , R 2 , R 3a , R 3b , R 26a , X, and Y have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R 26a is hydrogen.
  • compounds of any one of Formulae VI-XXI are provided, wherein E is -OR 26a ; R la , R lb , R lc , R ld , R 2 , R 26a , X, and Y have the meanings as described above for Formula I; and R 3a and R 3b taken together form a 4- to 7-membered optionally substituted cycloalkyl or 4- to 7-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R 26a is hydrogen.
  • R 3a and R 3b taken together form a cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or optionally substituted piperidinyl group.
  • compounds of any one of Formulae VI-XXI are provided, wherein E is -NR 26b R 26c ; R la , R lb , R lc , R ld , R 2 , R 3a , R 3b ' R 26b , R 26c , X, and Y have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae VI-XXI are provided, wherein E is -NR 26b R 26c ; R la , R lb , R lc , R ld , R 2 , R 26b , R 26c , X, and Y have the meanings as described above for Formula I; and R 3a and R 3b taken together form a 4- to 7-membered optionally substituted cycloalkyl or 4- to 7-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae VI-XXI are provided, wherein E is -NR 26b R 26c ; R la , R lb , R lc , R ld , R 2 , R 26b , R 26c , X, and Y have the meanings as described above for Formula I; and R 3a and R 3b taken together form a 4- to 7-membered optionally substituted cycloalkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae VI-XXI are provided, wherein E is -NR 26b R 26c , and R la , R lb , R lc , R ld , R 2 , R 26b , R 26c , X, and Y have the meanings as described above for Formula I, and R 3a and R 3b taken together form a cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae VI-XXI are provided, wherein E is -NR 26b R 26c , and R la , R lb , R lc , R ld , R 2 , R 26b , R 26c , X, and Y have the meanings as described above for Formula I, and R 3a and R 3b taken together form a 4- to 7-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formula VI-XXI are provided, wherein E is -NR 26b R 26c , and R la , R lb , R lc , R ld , R 2 , R 26b , R 26c , X, and Y have the meanings as described above for Formula I, and R 3a and R 3b taken together form a tetrahydropyranyl or optionally substituted piperidinyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae VI-XXI are provided, wherein E is -NR 26b R 26c , and R la , R lb , R lc , R ld , R 2 , R 3a , R 3b , X, and Y have the meanings as described above for Formula I, R 26b is hydrogen, and R 26c is optionally substituted cycloalkyl. In another embodiment, R 26c is hydroxycycloalkyl. In another embodiment, R 26c is cycloalkyl substituted with -C0 2 H.
  • compounds of any one of Formulae VI-XXI are provided, wherein E is -NR 26b R 26c , and R la , R lb , R lc , R ld , R 2 , R 3a , R 3b , X, and Y have the meanings as described above for Formula I, R 26b is hydrogen, and R 26c is optionally substituted aryl. In another embodiment R 26c is phenyl substituted with -C0 2 H.
  • R la , R lb , R lc , R ld , R 2 , E, X, and Y have the meanings as described above for Formula I;
  • A is selected from the group consisting of CR 27a R 27b , O, S, SO, S0 2 , and
  • R a is selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, sulfonamido, and -CO 2 R 31 ;
  • R 27b is selected from the group consisting of hydrogen and optionally substituted alkyl
  • R 27a and R 27b taken together form a 3- to 6-membered optionally substituted cycloalkyl or 3- to 6-membered optionally substituted heterocyclo;
  • R 28 is selected from the group consisting of hydrogen, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, carboxamido, sulfonamido, amino, -C0 2 R 32a , -COR 32a ,
  • R 32a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 32b is selected from the group consisting of optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 32c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl
  • R 32d is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl
  • R 32e is selected from the group consisting of optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 32f is selected from the group consisting of hydrogen, -CN, optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • B is -(CR 29a R 29b ) w -;
  • D is -(CR 30a R 30b ) x -;
  • each R 29a , R 29b , R 30a , and R 30b is independently selected from the group consisting of hydrogen, halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido and sulfonamido;
  • R 31 is selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted aryl;
  • w 0, 1, 2, 3, 4, 5, 6, 7, or 8;
  • x 0, 1, 2, 3, 4, 5, 6, 7, or 8;
  • compounds of Formula XXII are provided as a single stereoisomer, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, and each R 29a , R 29b , R 30a , and R 30b is independently hydrogen or (Ci-C4)alkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, each R 29a , R 29b , R 30a , and R 30b is independently hydrogen or (Ci-C4)alkyl, and A is CR a R , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, each R 29a , R 29b , R 30a , and R 30b is independently hydrogen or (Ci-C 4 )alkyl, A is CR 27a R 27b , and R 27a and R 27b are independently selected from the group consisting of hydrogen and alkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, each R 29a , R 29b , R 30a , and R 30b is independently hydrogen or (Ci-C 4 )alkyl, A is CR 27a R 27b , and R 27a and R 27b are hydrogen, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1 , 2, or 3, each R 29a , R 29b , R 30a , and R 0b is independently hydrogen or (Ci-C 4 )alkyl, and A is O, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, each R 29a , R 29b , R 30a , and R 30b is independently hydrogen or (Ci-C 4 )alkyl, and A is S0 2 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein w is 1 , 2, or 3, x is 1, 2, or 3, each R 9a , R 9b , R 30a , and R 0b is independently hydrogen or (Ci-C 4 )alkyl, and A is NR 28 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, R 29a , R 29b , R 0a , and R 30b are hydrogen, and A is NR 28 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein E is -OR 26a , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXII are provided wherein E is -NR 26b R 26c , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R a , R , R c , R , R , E, X, and Y have the meanings as described above for Formula I, A has the meaning as described above for Formula XXII, and z is 0, 1, or 2, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of Formula XXIV are provided:
  • R la , R lb , R lc , R ld , R 2 , E, X, and Y have the meanings as described above for Formula I
  • A has the meaning as described above for Formula XXII
  • z has the meaning as described above for Formula XXIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R la , R lb , R lc , R ld , R 2 , E, X, and Y have the meanings as described above for Formula I
  • A has the meaning as described above for Formula XXII
  • z has the meaning as described above for Formula XXIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae XXIII-XXV are provided, wherein E is -NR 26b R 26c , R 26b is hydrogen, and R 26c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and aralkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R 26c is a hydroxyalkyl, e.g., a monohydroxyalkyl or dihydroxyalkyl.
  • R 26c is an optionally substituted (cycloalkyl)alkyl.
  • R 26c is a (heterocyclo)alkyl. In another embodiment, R 26c is optionally substituted cycloalkyl. In another embodiment, R 26c c is optionally substituted phenyl. [0184] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided wherein A is CR 27a R 27b , O, or NR 28 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae XXIII-XXV are provided wherein A is CHR 27a , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae XXIII-XXV are provided wherein A is CHR 27a and R 27a is selected from the group consisting of hydrogen, hydroxy, amino, alkoxy, optionally substituted alkyl, haloalkyl, substituted heteroaryl, carboxamido, sulfonamido, and -CO 2 H, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae XXIII-XXV are provided wherein A is CH 2 or NR 28 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae XXIII-XXV are provided wherein A is CH 2 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae XXIII-XXV are provided wherein A is NR 28 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae XXIII-XXV are provided wherein A is CH 2 and z is 1, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • compounds of any one of Formulae XXIII-XXV are provided wherein A is CH 2 , z is 1, E is -NR 26b R 26c , R 26b is hydrogen, and R 26c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and aralkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R 26c is optionally substituted cycloalkyl, e.g., a hydroxycycloalkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R 26c is optionally substituted aryl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R c is optionally substituted heteroaryl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R la , R lb , R lc , R ld , R 2 , and R 26c have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R la , R lb , R lc , R ld , R 2 , and R 26c have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R la , R lb , R lc , R ld , R 2 , and R 26c have the meanings as described above for
  • Formula I or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, fluoro, and chloro;
  • R 2 is optionally substituted phenyl
  • R c is selected from the group consisting of optionally substituted (Ci-C 4 ) alkyl, optionally substituted (C 4 -C 8 ) cycloalkyl, optionally substituted phenyl, optionally substituted heteroaryl, and aralkyl;
  • R 26c is optionally substituted alkyl.
  • R 26c is hydroxyalkyl.
  • R 26c is dihydroxyalkyl.
  • R 26c is (heterocyclo)alkyl.
  • R 26c is optionally substituted cycloalkyl.
  • R 26c is hydroxycycloalkyl.
  • R 26 is cycloalkyl substituted with -CO 2 H.
  • R 26c is selected from the group consisting of:
  • R c is optionally substituted phenyl.
  • R 26c is phenyl substituted with one or two of the following groups: halo, cyano, alkyl, haloalkyl, alkoxy, carboxamido, sulfonamido, -C0 2 R c , or -S0 2 R d , wherein R c and R d are as defined above in connection with optionally substituted alkyl.
  • R 26c is phenyl substituted with -C0 2 H.
  • R 26c is selected from the group consisting of:
  • R c is optionally substituted heteroaryl.
  • the optionally substituted heteroaryl is an optionally substituted furyl, thienyl, pyridyl, pyrimidyl, benzimidazolyl, benzthiazolyl, or indolyl,
  • R 26c is heteroaryl substituted with one or two of the following groups: halo, cyano, hydroxy, alkyl, haloalkyl, alkoxy, carboxamido, sulfonamido, -CONHS0 2 Me, -C0 2 R c , -COR c , -S0 2 R d , -N(R e )COR f , -N(R e )S0 2 R s or -N(R e
  • R 26c is heteroaryl substituted with one or two of the following groups: halo, cyano, alkyl, haloalkyl, alkoxy, carboxamido, sulfonamido, -C0 2 R c , or -S0 2 R d , wherein R c and R d are as defined above in connection with optionally substituted alkyl.
  • R 26c is heteroaryl substituted with -C0 2 H.
  • R 26c is selected from the group consisting of:
  • R 26c is selected from the group consisting of:
  • R 26c is selected from the group consisting of:
  • compounds of Formulae XXVII or XXVIII are provided substantially free of one or more other stereoisomers.
  • compounds of any one of Formulae XXVII or XXVIII are substantially pure stereoisomers.
  • compounds of any one of Formulae XXVII or XXVIII are pure stereoisomers.
  • the disclosure provides a method of preparing a compound having Formula
  • R la , R lb , R lc , R ld , R 2 , R 3a , R 3b , and E have the meanings as described above for Formula I, and X and Y are NH.
  • E is -OR 26a
  • E is -NR R c .
  • R > 26 ⁇ b ⁇ is hydrogen, and R 26c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl.
  • R 3a and R 3b are taken together form an unsubstituted 4- to 8-membered cycloalkyl.
  • Formula XVI comprises all Formula VI:
  • Formula XVI comprises dissolving a compound having Formula VI in a solvent or a mixture of solvents.
  • Formula XVI comprises:
  • Formula XVI comprises:
  • the method of preparing a compound having Formula XVI comprises:
  • the solvent is selected from the group consisting of acetonitrile, methanol, ethyl acetate, and water, or a mixture thereof.
  • the isomerization is carried out at a pH of less than 7, e.g., at a pH of about 6, about 5, about 4, about 3, about 2, or about 1. In one embodiment, the isomerization is carried out at a pH of about 7. In one embodiment, the isomerization is carried out at a pH of greater than 7, e.g., at a pH of about 8, about 9, about 10, about 1 1, about 12, or about 13.
  • the isomerization is carried out in the presence of an acid, e.g., trifluoroacetic acid or acetic acid.
  • an acid e.g., trifluoroacetic acid or acetic acid.
  • the isomerization is carried out in the presence of a base, e.g., NaHC0 3 .
  • isomerization is carried out at a temperature of about 20°C to about 100°C, e.g., at a temperature of about 20°C to about 70°C, e.g., at a temperature of about 45°C to about 65°C. In one embodiment the isomerization is carried out at about room temperature, e.g., at about 20°C.
  • the isomerization is carried out above room temperature, e.g., at about 25°C, at about 30°C, about 35°C, about 40°C, about 45°C, about 50°C, about 55°C, about 60°C, about 65°C, about 70°C, about 75°C, about 80°C, about 85°C, about 90°C, about 95°C, or about 100°C.
  • the isomerization is carried about for a period of time between about 0.5 hours and about 2 weeks, e.g., for about 1 hour, about 3 hours, about 6 hours, about 12 hours, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 1 week.
  • the period of time needed for isomerization to occur may depend on a variety of factors including the chemical structure of Formula VI, the solvent(s), the temperature, and/or the pH.
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
  • R 2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl
  • R 4 is selected from the group consisting of hydrogen and optionally substituted Ci-C 6 alkyl
  • R 5 is selected from the group consisting of:
  • each R 6a and R 6b is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
  • R 7 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
  • R 8a and R 8b are each independently selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl; or
  • R 8a and R 8b taken together with the carbon that they are attached form a 3- to 8-membered optionally substituted cycloalkyl
  • W 1 is selected from the group consisting of -OR 9a and -NR 9b R 9c ;
  • R 9a is hydrogen
  • R 9b is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S0 2 R 9d , and -CONR 9e R 9f ;
  • R 9c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 9b and R 9c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R 9d is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
  • R 9e and R 9f are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
  • R 9e and R 9f taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • W 2 is selected from the group consisting of -OR 10 and -NR l la R l lb ;
  • R 10 is hydrogen
  • R 9a and R 10 are hydrogen and the other is a metabolically cleavable group
  • R l la is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S0 2 R l lc , and -CONR lld R l le ;
  • R l lb is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R l la and R l lb taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • R l lc is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
  • R l ld and R l le are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
  • R l ld and R l le taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
  • n 1, 2, 3, 4, or 5;
  • each R 12a , R 12b , R 12c and R 12d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
  • R 13 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl
  • R 14 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
  • Z is selected from the group consisting of -OR 15 and -NR 16a R 16b ; or
  • Z and R 14 taken together form a carbonyl, i.e., a C 0, group.
  • R 15 is selected from the group consisting of hydrogen and metabolically cleavable group
  • R 16a is selected from the group consisting of -S0 2 R 16c and -CONR 16d R 16e ;
  • R 16b is selected from the group consisting of hydrogen and optionally substituted alkyl
  • R 16c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R and R e are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 16d and R 16e taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo
  • o 1, 2, or 3;
  • p 0, 1, 2, or 3;
  • each R 17a , R 17b , R 17c and R 17d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
  • R 18 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl
  • R 19 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
  • R 20 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
  • R 21a and R 21b are each hydrogen;
  • R 21a and R 21b are hydrogen and the other is metabolically cleavable group
  • q 0, 1, 2, or 3;
  • r is 1, 2, or 3;
  • each R 22a , R 22b , R 22c , and R 22d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
  • R 23 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl
  • R 24 is selected from the group consisting of -S0 2 R 24a and -CONR 24b R 24c ;
  • R 24a is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 24b and R 24c are each independently selected from the group consisting of hydrogen, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 24b and R 24c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo; s and t are each independently 1, 2, or 3;
  • X is selected from the group consisting of O, S, and NR;
  • Y is selected from the group consisting of O, S, and NR ;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R la , R lb , R lc , and R ld are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
  • R 2 is optionally substituted aryl
  • R 3a is halo or d-do alkyl
  • R 3b is halo or Ci-Cio alkyl; or R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl;
  • R 4 is hydrogen
  • X and Y are NH
  • R la and R ld are each hydrogen
  • R lb is selected from the group consisting of hydrogen and fluoro; and R lc is selected from the group consisting of fluoro and chloro, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • R 2 is an optionally substituted aryl having Formula R2-1:
  • R 25a , R 25 , R 25c , R 25d , and R 25e are each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, amino, cyano, alkoxy, alkyl, or haloalkyl,
  • R 25a is selected from the group consisting of hydrogen and fluoro
  • R 25b is chloro
  • R 25c is selected from the group consisting of hydrogen and fluoro
  • R 25d and R 25e are each hydrogen
  • R 14 is hydrogen or C1-C4 alkyl
  • R 15 is hydrogen
  • a pharmaceutical composition comprising the compound of any one of particular embodiments I- VIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and a pharmaceutically acceptable carrier.
  • XV A method of treating a patient comprising administering to the patient a therapeutically effective amount of the compound of any one of particular embodiments I- VIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein the patient has a hyperproliferative disease.
  • XVI A method of treating a patient comprising administering to the patient a therapeutically effective amount of the pharmaceutical composition of particular embodiment XIV, wherein the patient has a hyperproliferative disease.
  • XVII The method of particular embodiments XV or XVI, wherein the hyperproliferative disease is cancer.
  • XVIII The method of particular embodiments XV or XVI, wherein cells of the hyperproliferative disease express functional p53.
  • XX The method of particular embodiment XIX, wherein the anticancer agent is a chemotherapeutic agent.
  • XXI The method of particular embodiment XIX, wherein the anticancer agent is radiation therapy.
  • XXII A method of treating a patient, wherein the patient has a hyperproliferative disorder and is being treated with an anticancer agent, comprising administering to the patient a compound of any one of particular embodiments I-XIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • XXIII The method of particular embodiment XXII, wherein the patient is experiencing side-effects of the anticancer agent treatment selected from the group consisting of mucositis, stomatitis, xerostoma, alopecia, and gastrointestinal disorder.
  • XXrV The method of particular embodiment XXII, wherein cells of the hyperproliferative disorder express functional p53.
  • XXV A kit comprising a compound of any one of particular embodiments I-XIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and instructions for administering the compound to a patient having a hyperproliferative disease.
  • XXVI The kit of particular embodiment XXV, wherein the hyperproliferative disease is cancer.
  • XXVII The kit of particular embodiment XXVI, further comprising one or more anticancer agents.
  • XXVIII The kit of particular embodiment XXVII, wherein the instructions direct co-administration of the compound together with the one or more anticancer agents.
  • R la , R lb , R lc , and R ld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
  • R 2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
  • E is selected from the group consisting of -OR 26a and -NR 26b R 26c ;
  • R 26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
  • R 26b is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl
  • R 26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, and -S0 2 R 5b ; or R and R c taken together form a 4- to 9-membered optionally substituted heterocyclo;
  • X is selected from the group consisting of O, S, and NR;
  • Y is selected from the group consisting of O, S, and NR ;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
  • E is -NR 26b R 26c ;
  • R 2 is optionally substituted aryl
  • R 3a is halo or Ci-Cio alkyl
  • R 3b is halo or Ci-Cio alkyl
  • R 3a and R 3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3- to 9-membered optionally substituted heterocyclo;
  • R 26b is hydrogen
  • X and Y are NH
  • XXXrV The compound of any one of particular embodiments XXIX-XXXIII, wherein R 3a and R 3b are each independently Ci-C w alkyl.
  • R 3a and R 3b taken together form a 6-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt thereof.
  • R 3a and R 3b taken together form an optionally substituted piperidine or a tetrahydropyran.
  • R 3a and R 3b taken together form a 4- to 8-membered optionally substituted cycloalkyl, or a pharmaceutically acceptable salt thereof.
  • XXXIX The compound of particular embodiment XXXVIII, wherein R 3a and R 3b are taken together to form an unsubstituted cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring, or a pharmaceutically acceptable salt thereof.
  • R la and R ld are each hydrogen
  • R lb is selected from the group consisting of hydrogen and fluoro; and R lc is selected from the group consisting of fluoro and chloro; or a pharmaceutically acceptable salt thereof.
  • XLI The compound of any one of particular embodiments XXIX -XL, wherein R 2 is optionally substituted aryl having Formula R2-1 :
  • R 25a , R 25 , R 25c , R 25d , and R 25e are each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, amino, cyano, alkoxy, alkyl, or haloalkyl,
  • XLII The compound of any one of particular embodiments XXIX -XL, wherein R 2 is optionally substituted pyridyl, or a pharmaceutically acceptable salt thereof.
  • XLVI The compound of any one of particular embodiments XXIX-XLV, wherein R 26c is optionally substituted alkyl, or a pharmaceutically acceptable salt thereof.
  • XLVII The compound of particular embodiment XLVI, wherein the optionally substituted alkyl is substituted with an optionally substituted cycloalkyl, or a pharmaceutically acceptable salt thereof.
  • XL VIII The compound of particular embodiment XLVI, wherein the optionally substituted alkyl is substituted with an optionally substituted heteroaryl, or a pharmaceutically acceptable salt thereof.
  • LVII The compound of particular embodiment LVI, wherein the optionally substituted phenyl is substituted with at least one -CO 2 H, or a pharmaceutically acceptable salt thereof.
  • LVIII The compound of any one of particular embodiments XXIX-XLV, wherein R 26c is optionally substituted heteroaryl, or a pharmaceutically acceptable salt thereof.
  • LX A compound selected from the group consisting of:
  • LXI A compound selected from the group consisting of:
  • LXIII The compound of particular embodiment LXII, wherein R 26a hydrogen, or a pharmaceutically acceptable salt thereof.
  • LXIV. A pharmaceutical composition comprising the compound of any one of particular embodiments XXIX-LXI, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • LXV A method of treating a patient comprising administering to the patient a therapeutically effective amount of the compound of any one of particular embodiments XXIX-LXI, or a pharmaceutically acceptable salt thereof, wherein the patient has a hyperproliferative disease.
  • LXVI The method of particular embodiment LXV, wherein the hyperproliferative disease is cancer.
  • LXVII The method of particular embodiment LXVI, wherein the cancer is selected from the group consisting of melanoma, lung cancer, sarcoma, colon cancer, prostate cancer, choriocarcinoma, breast cancer, retinoblastoma, stomach carcinoma, acute myeloid leukemia, lymphoma, multiple myeloma, and leukemia.
  • LXVIII The method of particular embodiment LXVII, wherein the cancer is selected from the group consisting of liposarcoma and melanoma.
  • LXIX A kit comprising a compound of any one of particular embodiments XXIX-LXI, or a pharmaceutically acceptable salt, thereof, and instructions for administering the compound to a patient having a hyperproliferative disease.
  • LXX The kit of particular embodiment LXIX, wherein the hyperproliferative disease is cancer.
  • LXXII The method of particular embodiment LXXI, wherein E is -OR'
  • LXXIII The method of particular embodiment LXXI, wherein E is
  • Compounds of Formula III can be separated by chiral resolution methods well known in the art, e.g., chiral column chromatography, to give compounds of Formulae VI-XXI.
  • Suitable chiral columns for use in chiral resolutions include, for example, Daicel CHIRALCEL ® OD-H, Daicel CHIRAKPAK ® AD-H and Regis Technologies ULMO chiral columns. Other chiral resolution methods are also possible.
  • Compounds of Formulae VI-XXI can also be prepared by asymmetric synthetic methods.
  • compounds of Formula VI wherein Y is NH and E is NR 26b R 26c , can be synthesized by using a asymmetric 1,3-dipolar cycloaddition as the key step as previously described (See U.S. Patent Nos. 7,759,383 B2 and 7,737, 174 B2, and Ding et al, J. Am. Chem. Soc. 727: 10130-10131 (2005)).
  • Compounds of Formula VI can undergo isomerization in solution to give compounds of Formula XVI.
  • VI to Formula XVI may involve formation of the imine intermediate shown in Scheme 5.
  • Compounds of Formula XVI may be less likely to isomerize, i.e., they may be chemically more stable, than compounds of Formula VI.
  • isolation and purification of compounds of Formula XVI may be improved when R 3a and R 3a are the same, e.g., R 3a and R 3b are methyl, R 3a and R 3b taken together form a cyclohexyl ring, because the 2' position of the pyrrolidine ring is not an asymmetric center, and the number of possible isomerization products of Formula VI is reduced.
  • General methods to prepare compounds provided in the present disclosure are shown in Schemes 4-8.
  • the present disclosure contemplates that exposure of animals suffering from cancer to therapeutically effective amounts of drug(s) (e.g., small molecules) that increase the function(s) of p53 and p53-related proteins (e.g.. p63, p73) inhibits the growth of cancer cells or supporting cells.
  • drug(s) e.g., small molecules
  • the compounds provided herein inhibit the interaction between p53 or p53 -related proteins and MDM2 or MDM2 -related proteins (e.g., MDMX). Inhibiting the interaction between p53 or p53 -related proteins and MDM2 or MDM2 -related proteins inhibits the growth of cancer cells or supporting cells and/or renders such cells as a population more susceptible to the cell death-inducing activity of cancer therapeutic drugs or radiation therapies.
  • the inhibitors provided herein prolong the half-life of p53 by interfering with the p53-MDM2 interaction that would normally promote degradation of p53.
  • the compounds provided herein satisfy an unmet need for the treatment of multiple cancer types, either when administered as monotherapy to induce senescence, cell growth inhibition, apoptosis and/or cell cycle arrest in cancer cells, or when administered in a temporal relationship with additional agent(s), such as other cell death-inducing or cell cycle disrupting cancer therapeutic drugs or radiation therapies (combination therapies), so as to render a greater proportion of the cancer cells or supportive cells susceptible to executing the apoptosis program compared to the corresponding proportion of cells in an animal treated only with the cancer therapeutic drug or radiation therapy alone.
  • treatment of patients with a therapeutically effective amount of one or more compounds having Formulae I-XXVIII and one or more anticancer agents produces a greater anti-tumor activity and clinical benefit in such patients compared to those treated with the compound or anticancer drugs/radiation alone.
  • the compounds provided herein can lower the apoptotic threshold of cells that express p53 or p53-related protein, the proportion of cells that successfully execute the apoptosis program in response to the apoptosis inducing activity of anticancer drugs/radiation will be increased when used in combination with one or more of the compounds provided herein.
  • compounds having Formulae I-XXVIII can be used to allow administration of a lower, and therefore less toxic and more tolerable, dose of an anticancer drug and/or radiation to produce the same tumor response/clinical benefit as the conventional dose of the anticancer drug/radiation alone. Since the doses for approved anticancer drugs and radiation treatments are known, the compounds, compositions, and methods provided herein can be used with one or more approved anticancer drugs and/or radiation treatment.
  • compounds having Formulae I-XXVIII may act, at least in part, by stimulating the pro-apoptotic and/or cell cycle-inhibiting activities of p53 and p53 -related proteins, the exposure of cancer cells and supporting cells to therapeutically effective amounts of these compounds can be temporally linked to coincide with the attempts of cells to execute the apoptosis program in response to the anticancer drug or radiation therapy.
  • administering the compounds or pharmaceutical compositions provided herein in combination with other known anticancer drugs provides especially efficacious therapeutic practices.
  • the inhibitors of the interaction between p53 or p53- related proteins and MDM2 and MDM2-related proteins having Formulae I-XXVIII may protect normal (e.g., non-hyperproliferative) cells from the toxic effects of certain chemotherapeutic agents and radiation, possibly through the ability of the inhibitors to induce cell cycle arrest of normal cells.
  • the inhibitors provided herein may cause cell cycle arrest in cells comprising wild-type or functional p53 (and/or wild-type or functional p53- related proteins) while having no or less effect on cancer cells comprising mutated, deleted, or otherwise non- or less functional p53 (and/or mutated, deleted, or otherwise non-or less functional p53-related proteins).
  • This differential protective effect may allow for more effective treatment of cancer by allowing the use of higher doses or longer treatments of chemotherapeutic agents or treatments without increasing the toxic side effects of such treatment when administered in combination with inhibitors provided herein.
  • Compounds having Formulae I-XXVIII can also be used to provide chemoprotection of normal cells through the induction of cell cycle arrest prior to treatment with chemotherapeutic agents.
  • methods of rendering a normal cell resistant to chemotherapeutic agents or treatments comprises contacting the cell with one or more compounds having Formulae I-XXVIII are provided.
  • methods of protecting normal cells in an animal having a hyperproliferative disease from the toxic side effects of chemotherapeutic agents or treatments comprises administering to the animal a compound having Formulae I-XXVIII are provided.
  • methods for the treatment, amelioration, or prevention of disorders, side effects, or conditions caused by the administration of chemotherapeutic agents to normal cells comprising administering to an animal undergoing chemotherapy a compound having Formulae I-XXVIII.
  • disorders and conditions caused by chemotherapy include, without limitation, mucositis, stomatitis, xerostomia, gastrointestinal disorders, and alopecia.
  • Compounds having Formulae I-XXVIII are useful for the treatment, amelioration, or prevention of disorders, such as those responsive to induction of apoptotic cell death, e.g., disorders characterized by dysregulation of apoptosis, including hyperproliferative diseases such as cancer.
  • these compounds can be used to treat, ameliorate, or prevent cancer that is characterized by resistance to cancer therapies (e.g., those cancer cells which are chemoresistant, radiation resistant, hormone resistant, and the like).
  • these compounds can be used to treat hyperproliferative diseases characterized by expression of functional p53 or p53 -related proteins.
  • these compounds can be used to protect normal (e.g., non-hyperproliferative) cells from the toxic side effects of chemotherapeutic agents and treatments by the induction of cell cycle arrest in those cells.
  • compounds having Formulae I-XXVIII induce cell cycle arrest and/or apoptosis and also potentiate the induction of cell cycle arrest and/or apoptosis either alone or in response to additional apoptosis induction signals. Therefore, it is contemplated that these compounds sensitize cells to induction of cell cycle arrest and/or apoptosis, including cells that are resistant to such inducing stimuli. By inhibiting the interaction between p53 or p53 -related proteins and MDM2 or MDM2-realted proteins, these compounds can be used to induce apoptosis in any disorder that can be treated, ameliorated, or prevented by the induction of apoptosis.
  • compounds having Formulae I-XXVIII can be used to induce apoptosis in cells comprising functional p53 or p53-related proteins.
  • the disclosure pertains to modulating apoptosis with compounds having Formulae I-XXVIII in combination with one or more additional apoptosis-modulating agents, e.g., anticancer agents.
  • apoptosis-modulating agents include, but are not limited to, Fas/CD95, TRAMP, TNF RI, DR1, DR2, DR3, DR4, DR5, DR6, FADD, RIP, TNFa, Fas ligand, TRAIL, antibodies to TRAIL-R1 or TRAIL-R2, Bcl-2, p53, BAX, BAD, Akt, CAD, PI3 kinase, PP1, and caspase proteins. Other agents involved in the initiation, decision and degradation phase of apoptosis are also included. Examples of apoptosis-modulating agents include agents, the activity, presence, or change in concentration of which, can modulate apoptosis in a subject.
  • Apoptosis-modulating agents include those which are inducers of apoptosis, such as TNF or a TNF-related ligand, particularly a TRAMP ligand, a Fas/CD95 ligand, a TNFR-1 ligand, or TRAIL.
  • the compounds, compositions, and methods provided herein are used to treat diseased cells, tissues, organs, or pathological conditions and/or disease states in an animal (e.g., a mammalian patient including, but not limited to, humans and veterinary animals).
  • an animal e.g., a mammalian patient including, but not limited to, humans and veterinary animals.
  • various diseases and pathologies are amenable to treatment or prophylaxis using the present methods and compositions.
  • a non-limiting exemplary list of these diseases and conditions includes, but is not limited to, breast cancer, prostate cancer, lymphoma, skin cancer, pancreatic cancer, colon cancer, melanoma, malignant melanoma, ovarian cancer, brain cancer, primary brain carcinoma, head-neck cancer, glioma, glioblastoma, liver cancer, bladder cancer, non-small cell lung cancer, head or neck carcinoma, breast carcinoma, ovarian carcinoma, lung carcinoma, small-cell lung carcinoma, Wilms' tumor, cervical carcinoma, testicular carcinoma, bladder carcinoma, pancreatic carcinoma, stomach carcinoma, colon carcinoma, prostatic carcinoma, genitourinary carcinoma, thyroid carcinoma, esophageal carcinoma, myeloma, multiple myeloma, adrenal carcinoma, renal cell carcinoma, endometrial carcinoma, adrenal cortex carcinoma, malignant pancreatic insulinoma, malignant carcinoid carcinoma, choriocarcinoma, mycosis fungoides, malignant hypercalcemia, cervical hyperplasia, leuk
  • the compounds, compositions, and methods provided herein are used to treat cancers that express functional or wild type p53 or p53-related proteins. In one embodiment, the compounds, compositions, and methods provided herein are used to treat cancers that express elevated levels of MDM2 or MDM2-related proteins.
  • the compounds, compositions, and methods provided herein can be used to treat a patient having a sarcoma, including, for example, liposarcoma, malignant fibrous histiocytoma, osteosarcoma, and rhabdomyosarcoma.
  • the compounds, compositions, and methods provided herein can be used to treat a patient having a soft tissue tumor, including, for example, Ewing's sarcoma, leiomyosarcoma, lipoma, and malignant Schwannomas.
  • the compounds, compositions, and methods provided herein can be used to treat a patient having lung, breast, liver, or colon cancer.
  • the compounds, compositions, and methods provided herein can be used to treat a patient having B-cell chronic lymphocytic leukemia and acute myeloid leukemia.
  • the compounds, compositions, and methods provided here can be used to treat a patient having melanoma, lung cancer, sarcoma, colon cancer, prostate cancer, choriocarcinoma, breast cancer, retinoblastoma, stomach carcinoma, acute myeloid leukemia, lymphoma, multiple myeloma, or leukemia.
  • the compounds, compositions, and methods provided here can be used to treat a patient having liposarcoma or melanoma.
  • infections suitable for treatment with the compounds, compositions, and methods provided herein include, but are not limited to, infections caused by viruses, bacteria, fungi, mycoplasma, prions, and the like.
  • a further aspect of the present disclosure is to provide the use of a compound having any one of Formulae I-XXVIII for the manufacture of a medicament for treating a hyperproliferative disease such as cancer.
  • the medicament is to be administered with one or more additional agents.
  • a further aspect of the present disclosure is to provide a compound having any one of Formulae I-XXVIII, or a pharmaceutical composition comprising a compound having any one of Formulae I-XXVIII, for use in treating a hyperproliferative disease such as cancer.
  • methods are provided for administering an effective amount of a compound having Formulae I-XXVIII in combination with at least one additional therapeutic agent (including, but not limited to, chemotherapeutic antineoplastics, apoptosis-modulating agents, antimicrobials, antivirals, antifungals, and anti-inflammatory agents) and/or therapeutic technique (e.g., surgical intervention, and/or radiotherapies).
  • the additional therapeutic agent(s) is an anticancer agent.
  • a number of suitable therapeutic or anticancer agents are contemplated for use in the methods provided herein. Indeed, the methods provided herein can include but are not limited to, administration of numerous therapeutic agents such as: agents that induce apoptosis; polynucleotides (e.g., anti-sense, ribozymes, siRNA); polypeptides (e.g., enzymes and antibodies); biological mimetics (e.g., gossypol or BH3 mimetics); agents that bind (e.g., oligomerize or complex) with a Bcl-2 family protein such as Bax; alkaloids; alkylating agents; antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal or polyclonal antibodies (e.g., antibodies conjugated with anticancer drugs, toxins, defensins), toxins; radionuclides; biological response modifiers (e.g., interferons (e.g., IFN-a) and inter
  • anticancer agents comprise agents that induce or stimulate apoptosis.
  • Agents that induce or stimulate apoptosis include, for example, agents that interact with or modify DNA, such as by intercalating, cross-linking, alkylating, or otherwise damaging or chemically modifying DNA.
  • Agents that induce apoptosis include, but are not limited to, radiation (e.g., X- rays, gamma rays, UV); tumor necrosis factor (TNF)-related factors (e.g., TNF family receptor proteins, TNF family ligands, TRAIL, antibodies to TRAIL-Rl or TRAIL-R2); kinase inhibitors (e.g., epidermal growth factor receptor (EGFR) kinase inhibitor.
  • radiation e.g., X- rays, gamma rays, UV
  • TNF tumor necrosis factor
  • TRAIL TNF family receptor proteins
  • TRAIL TRAIL
  • TRAIL antibodies to TRAIL-Rl or TRAIL-R2
  • kinase inhibitors e.g., epidermal growth factor receptor (EGFR) kinase inhibitor.
  • vascular growth factor receptor (VGFR) kinase inhibitor vascular growth factor receptor (VGFR) kinase inhibitor, fibroblast growth factor receptor (FGFR) kinase inhibitor, platelet-derived growth factor receptor (PDGFR) kinase inhibitor, and Bcr-Abl kinase inhibitors (such as GLEEVEC)); antisense molecules; antibodies (e.g., HERCEPTIN, RITUXAN, ZEVALIN, and AVASTIN); anti-estrogens (e.g., raloxifene and tamoxifen); anti-androgens (e.g., flutamide, bicalutamide, finasteride, aminoglutethamide, ketoconazole, and corticosteroids); cyclooxygenase 2 (COX-2) inhibitors (e.g., celecoxib, meloxicam, NS-398, and non-steroidal anti-inflammatory drugs (NSAIDs)); antiinflammatory drugs (e.
  • compositions and methods provided herein include one or more compounds provided herein and at least one anti-hyperproliferative or anticancer agent, e.g., alkylating agents, antimetabolites, and natural products (e.g., herbs and other plant and/or animal derived compounds).
  • anti-hyperproliferative or anticancer agent e.g., alkylating agents, antimetabolites, and natural products (e.g., herbs and other plant and/or animal derived compounds).
  • Alkylating agents suitable for use in the present compositions and methods include, but are not limited to: 1) nitrogen mustards (e.g., mechlorethamine, cyclophosphamide, ifosfamide, melphalan (L-sarcolysin); and chlorambucil); 2) ethylenimines and methylmelamines (e.g., hexamethylmelamine and thiotepa); 3) alkyl sulfonates (e.g., busulfan); 4) nitrosoureas (e.g., carmustine (BCNU); lomustine (CCNU); semustine (methyl- CCNU); and streptozocin (streptozotocin)); and 5) triazenes (e.g., dacarbazine (DTIC; dimethyltriazenoimid-azolecarboxamide).
  • nitrogen mustards e.g., mechlorethamine, cyclophosphamide
  • antimetabolites suitable for use in the present compositions and methods include, but are not limited to: 1) folic acid analogs (e.g., methotrexate (amethopterin)); 2) pyrimidine analogs (e.g., fluorouracil (5- fluorouracil; 5-FU), floxuridine (fluorode-oxyuridine; FudR), and cytarabine (cytosine arabinoside)); and 3) purine analogs (e.g., mercaptopurine (6- mercaptopurine; 6-MP), thioguanine (6-thioguanine; TG), and pentostatin (2'- deoxycoformycin)).
  • folic acid analogs e.g., methotrexate (amethopterin)
  • pyrimidine analogs e.g., fluorouracil (5- fluorouracil; 5-FU), floxuridine (fluorode-oxyuridine; FudR), and cytara
  • chemotherapeutic agents suitable for use in the present compositions and methods include, but are not limited to: 1) vinca alkaloids (e.g., vinblastine (VLB), vincristine); 2) epipodophyllotoxins (e.g., etoposide and teniposide); 3) antibiotics (e.g., dactinomycin (actinomycin D), daunorubicin (daunomycin; rubidomycin), doxorubicin, bleomycin, plicamycin (mithramycin), and mitomycin (mitomycin C)); 4) enzymes (e.g., L- asparaginase); 5) biological response modifiers (e.g., interferon-alfa); 6) platinum coordinating complexes (e.g., cisplatin (cis-DDP) and carboplatin); 7) anthracenediones (e.g., mitoxantrone); 8) substituted
  • any anticancer agent that is routinely used in a cancer therapy context finds use in the compositions and methods of the present disclosure.
  • the U.S. Food and Drug Administration maintains a formulary of oncolytic agents approved for use in the United States. International counterpart agencies to the U.S.F.D.A. maintain similar formularies.
  • Table 1 provides a list of exemplary antineoplastic agents approved for use in the U.S. Those skilled in the art will appreciate that the "product labels" required on all U.S. approved chemotherapeutics describe approved indications, dosing information, toxicity data, and the like, for the exemplary agents.
  • Anticancer agents further include compounds which have been identified to have anticancer activity. Examples include, but are not limited to, 3-AP, 12-0- tetradecanoylphorbol- 13 -acetate, 17AAG, 852A, ABI-007, ABR-217620, ABT- 751, ADI-PEG 20, AE-941, AG-013736, AGROIOO, alanosine, AMG 706, antibody G250, antineoplastons, AP23573, apaziquone, APC8015, atiprimod, ATN-161, atrasenten, azacitidine, BB-10901, BCX-1777, bevacizumab, BGOOOOl, bicalutamide, BMS 247550, bortezomib, bryostatin-1, buserelin, calcitriol, CCI-779, CDB-2914, cefixime, cetuximab, CG0070, cilengitide, clofarabine, comb
  • anticancer agents and other therapeutic agents those skilled in the art are referred to any number of instructive manuals including, but not limited to, the Physician's Desk Reference and to Goodman and Gilman's "Pharmaceutical Basis of Therapeutics" tenth edition, Eds. Hardman et ah, 2002.
  • the methods provided herein comprise administering one or more compounds having Formulae I-XXVIII in combination with radiation therapy.
  • the methods provided herein are not limited by the types, amounts, or delivery and administration systems used to deliver the therapeutic dose of radiation to an animal.
  • the animal may receive photon radiotherapy, particle beam radiation therapy, other types of radiotherapies, and combinations thereof.
  • the radiation is delivered to the animal using a linear accelerator.
  • the radiation is delivered using a gamma knife.
  • the source of radiation can be external or internal to the animal.
  • External radiation therapy is most common and involves directing a beam of high-energy radiation to a tumor site through the skin using, for instance, a linear accelerator. While the beam of radiation is localized to the tumor site, it is nearly impossible to avoid exposure of normal, healthy tissue. However, external radiation is usually well tolerated by animals.
  • Internal radiation therapy involves implanting a radiation-emitting source, such as beads, wires, pellets, capsules, particles, and the like, inside the body at or near the tumor site including the use of delivery systems that specifically target cancer cells (e.g., using particles attached to cancer cell binding ligands). Such implants can be removed following treatment, or left in the body inactive.
  • Types of internal radiation therapy include, but are not limited to, brachytherapy, interstitial irradiation, intracavity irradiation, radioimmunotherapy, and the like.
  • the animal may optionally receive radiosensitizers (e.g., metronidazole, misonidazole, intra-arterial Budr, intravenous iododeoxyuridine (IudR), nitroimidazole, 5-substituted-4-nitroimidazoles, 2H-isoindolediones, [[(2- bromoethyl)-amino]methyl]-nitro- 1 H-imidazole- 1 -ethanol, nitroaniline derivatives, DNA-affinic hypoxia selective cytotoxins, halogenated DNA ligand, 1,2,4 benzotriazine oxides, 2-nitroimidazole derivatives, fluorine-containing nitroazole derivatives, benzamide, nicotinamide, acri
  • any type of radiation can be administered to an animal, so long as the dose of radiation is tolerated by the animal without unacceptable negative side- effects.
  • Suitable types of radiotherapy include, for example, ionizing (electromagnetic) radiotherapy (e.g., X-rays or gamma rays) or particle beam radiation therapy (e.g., high linear energy radiation).
  • Ionizing radiation is defined as radiation comprising particles or photons that have sufficient energy to produce ionization, i.e., gain or loss of electrons (as described in, for example, U.S. 5,770,581 incorporated herein by reference in its entirety).
  • the effects of radiation can be at least partially controlled by the clinician.
  • the dose of radiation is fractionated for maximal target cell exposure and reduced toxicity.
  • the total dose of radiation administered to an animal is about .01 Gray (Gy) to about 100 Gy.
  • about 10 Gy to about 65 Gy e.g., about 15 Gy, 20 Gy, 25 Gy, 30 Gy, 35 Gy, 40 Gy, 45 Gy, 50 Gy, 55 Gy, or 60 Gy
  • a complete dose of radiation can be administered over the course of one day
  • the total dose is ideally fractionated and administered over several days.
  • radiotherapy is administered over the course of at least about 3 days, e.g., at least 5, 7, 10, 14, 17, 21, 25, 28, 32, 35, 38, 42, 46, 52, or 56 days (about 1-8 weeks).
  • a daily dose of radiation will comprise approximately 1-5 Gy (e.g., about 1 Gy, 1.5 Gy, 1.8 Gy, 2 Gy, 2.5 Gy, 2.8 Gy, 3 Gy, 3.2 Gy, 3.5 Gy, 3.8 Gy, 4 Gy, 4.2 Gy, or 4.5 Gy), or 1-2 Gy (e.g., 1.5-2 Gy).
  • the daily dose of radiation should be sufficient to induce destruction of the targeted cells.
  • radiation is not administered every day, thereby allowing the animal to rest and the effects of the therapy to be realized.
  • radiation desirably is administered on 5 consecutive days, and not administered on 2 days, for each week of treatment, thereby allowing 2 days of rest per week.
  • radiation can be administered 1 day/week, 2 days/week, 3 days/week, 4 days/week, 5 days/week, 6 days/week, or all 7 days/week, depending on the animal's responsiveness and any potential side effects.
  • Radiation therapy can be initiated at any time in the therapeutic period. In one embodiment, radiation is initiated in week 1 or week 2, and is administered for the remaining duration of the therapeutic period. For example, radiation is administered in weeks 1-6 or in weeks 2-6 of a therapeutic period comprising 6 weeks for treating, for instance, a solid tumor. Alternatively, radiation is administered in weeks 1-5 or weeks 2-5 of a therapeutic period comprising 5 weeks.
  • These exemplary radiotherapy administration schedules are not intended, however, to limit the methods provided herein.
  • Antimicrobial therapeutic agents may also be used as therapeutic agents in combination with the compounds having Formulae I-XXVIII. Any agent that can kill, inhibit, or otherwise attenuate the function of microbial organisms may be used, as well as any agent contemplated to have such activities.
  • Antimicrobial agents include, but are not limited to, natural and synthetic antibiotics, antibodies, inhibitory proteins (e.g., defensins), antisense nucleic acids, membrane disruptive agents and the like, used alone or in combination. Indeed, any type of antibiotic may be used including, but not limited to, antibacterial agents, antiviral agents, antifungal agents, and the like.
  • one or more compounds having Formulae I-XXVIII are administered to an animal in need thereof.
  • one or more compounds having Formulae I-XXVIII and one or more additional therapeutic agents or anticancer agents are administered to an animal in need thereof under one or more of the following conditions: at different periodicities, at different durations, at different concentrations, by different administration routes, etc.
  • the compound having Formulae I-XXVIII is administered prior to the therapeutic or anticancer agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks prior to the administration of the therapeutic or anticancer agent.
  • the compound having Formulae I-XXVIII is administered after the therapeutic or anticancer agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks after the administration of the anticancer agent.
  • the compound having Formulae I-XXVIII and the therapeutic or anticancer agent are administered concurrently but on different schedules, e.g., the compound is administered daily while the therapeutic or anticancer agent is administered once a week, once every two weeks, once every three weeks, or once every four weeks.
  • the compound is administered once a week while the therapeutic or anticancer agent is administered daily, once a week, once every two weeks, once every three weeks, or once every four weeks.
  • a method of treating, preventing, or ameliorating cancer in a patient comprises pulsatile administration to the patient a therapeutically effective amount of a compound having Formulae I-XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the pharmaceutical compositions provided herein comprise one or more compounds having Formulae I-XXVIII in an amount which is effective to achieve its intended purpose. While individual needs vary, determination of optimal ranges of effective amounts of each component is within the skill of the art.
  • the compounds may be administered to mammals, e.g. humans, orally at a dose of 0.0025 to 50 mg/kg, or an equivalent amount of the pharmaceutically acceptable salt thereof, per day of the body weight of the mammal being treated for disorders responsive to induction of apoptosis. In one embodiment, about 0.01 to about 25 mg/kg is orally administered to treat, ameliorate, or prevent such disorders.
  • the dose is generally about one-half of the oral dose.
  • a suitable intramuscular dose would be about 0.0025 to about 25 mg kg, or from about 0.01 to about 5 mg/kg.
  • the unit oral dose may comprise from about 0.01 to about 1000 mg, for example, about 0.1 to about 100 mg of the compound.
  • the unit dose may be administered one or more times daily as one or more tablets or capsules each containing from about 0.1 to about 10 mg, conveniently about 0.25 to 50 mg of the compound or its solvates.
  • the compound may be present at a concentration of about 0.01 to 100 mg per gram of carrier. In a one embodiment, the compound is present at a concentration of about 0.07-1.0 mg/ml, for example, about 0.1-0.5 mg/ml, and in one embodiment, about 0.4 mg/ml.
  • compounds having Formulae I-XXVIII may be administered as part of a pharmaceutical preparation or composition.
  • the pharmaceutical composition comprises one or more pharmaceutically acceptable carriers, excipients, and/or auxiliaries.
  • the one or more carriers, excipients, and auxiliaries facilitate processing of the compound having Formulae I-XXVIII into a preparation which can be used pharmaceutically.
  • compositions particularly those compositions which can be administered orally or topically and which can be used for one type of administration, such as tablets, dragees, slow release lozenges and capsules, mouth rinses and mouth washes, gels, liquid suspensions, hair rinses, hair gels, shampoos and also preparations which can be administered rectally, such as suppositories, as well as suitable solutions for administration by intravenous infusion, injection, topically or orally, contain from about 0.01 to 99 percent, in one embodiment from about 0.25 to 75 percent of active compound(s), together with the one or more carriers, excipients, and/or auxiliaries.
  • compositions provided herein may be administered to any patient which may experience the beneficial effects of compounds having Formulae I-XXVIII.
  • mammals e.g., humans, although the methods and compositions provided herein are not intended to be so limited.
  • Other patients include veterinary animals (cows, sheep, pigs, horses, dogs, cats and the like).
  • Compounds having Formulae I-XXVIII and pharmaceutical compositions thereof may be administered by any means that achieve their intended purpose.
  • administration may be by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, intranasal or topical routes.
  • administration may be by the oral route.
  • the dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
  • compositions and preparations provided herein are manufactured by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes.
  • pharmaceutical compositions for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone.
  • fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose,
  • disintegrating agents may be added such as the above-mentioned starches and also carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate.
  • Auxiliaries can be suitable flow-regulating agents and lubricants. Suitable auxiliaries include, for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol.
  • Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices.
  • concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethyl-cellulose phthalate, are used.
  • Dye stuffs or pigments may be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.
  • Other pharmaceutical preparations which can be used orally include push- fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol.
  • the push-fit capsules can contain the active compounds in the form of granules which may be mixed with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds are in one embodiment dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin.
  • suitable liquids such as fatty oils, or liquid paraffin.
  • stabilizers may be added.
  • Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of one or more of the active compounds with a suppository base.
  • Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons.
  • gelatin rectal capsules which consist of a combination of the active compounds with a base.
  • Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
  • Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water- soluble salts and alkaline solutions.
  • suspensions of the active compounds as appropriate oily injection suspensions may be administered.
  • Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides or polyethylene glycol-400.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran.
  • the suspension may also contain stabilizers.
  • the topical compositions provided herein are formulated in one embodiment as oils, creams, lotions, ointments and the like by choice of appropriate carriers.
  • Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C 12 ).
  • the carriers may be those in which the active ingredient is soluble.
  • Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired.
  • transdermal penetration enhancers can be employed in these topical formulations. Examples of such enhancers can be found in U.S. Pat. Nos. 3,989,816 and 4,444,762.
  • Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil such as almond oil with warm soft paraffin and allowing the mixture to cool.
  • a vegetable oil such as almond oil
  • a typical example of such an ointment is one which includes about 30% almond oil and about 70% white soft paraffin by weight.
  • Lotions may be conveniently prepared by dissolving the active ingredient, in a suitable high molecular weight alcohol such as propylene glycol or polyethylene glycol.

Abstract

Provided herein are compounds, compositions, and methods in the field of medicinal chemistry. The compounds and compositions provided herein relate to spiro oxindoles which function as antagonists of the interaction between p53 and MDM2, and their use as therapeutics for the treatment of cancer and other diseases.

Description

SPIRO-OXINDOLE MDM2 ANTAGONISTS
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to pending U.S. Provisional Patent
Application No. 61/484,986, filed May 11, 201 1, the contents of which are incorporated by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] This invention was made with government support under CA121279 awarded by the National Institutes of Health. The government has certain rights in the invention
BACKGROUND
[0003] The aggressive cancer cell phenotype is the result of a variety of genetic and epigenetic alterations leading to deregulation of intracellular signaling pathways (Ponder, Nature 411:336 (2001)). Cancer cells typically fail to execute an apoptotic program, and lack of appropriate apoptosis due to defects in the normal apoptosis machinery is considered a hallmark of cancer (Lowe et al, Carcinogenesis 27:485 (2000)). The inability of cancer cells to execute an apoptotic program due to defects in the normal apoptotic machinery is often associated with an increase in resistance to chemotherapy, radiation, or immunotherapy-induced apoptosis. Primary or acquired resistance of human cancer of different origins to current treatment protocols due to apoptosis defects is a major problem in current cancer therapy (Lowe et al, Carcinogenesis 27:485 (2000); Nicholson, Mature 407:810 (2000)). Accordingly, current and future efforts towards designing and developing new molecular target-specific anticancer therapies to improve survival and quality of life of cancer patients must include strategies that specifically target cancer cell resistance to apoptosis.
[0004] The p53 tumor suppressor plays a central role in controlling cell cycle progression, senescence, and apoptosis (Vogelstein et al, Nature 408:301 (2000); Goberdhan, Cancer Cell 7:505 (2005)). MDM2 and p53 are part of an auto- regulatory feed-back loop (Wu et al, Genes Dev. 7: 1 126 (1993)). MDM2 is transcriptionally activated by p53 and MDM2, in turn, inhibits p53 activity by at least three mechanisms (Wu et al, Genes Dev. 7: 1126 (1993). First, MDM2 protein directly binds to the p53 transactivation domain and thereby inhibits p53- mediated transactivation. Second, MDM2 protein contains a nuclear export signal sequence, and upon binding to p53, induces the nuclear export of p53, preventing p53 from binding to the targeted D As. Third, MDM2 protein is an E3 ubiquitin ligase and upon binding to p53 is able to promote p53 degradation.
[0005] Although high-affinity peptide-based inhibitors of MDM2 have been successfully designed in the past (Garcia-Echeverria et al, Med. Chem. 43:3205 (2000)), these inhibitors are not suitable therapeutic molecules because of their poor cell permeability and in vivo bioavailability. Despite intensive efforts by the pharmaceutical industry, high throughput screening strategies have had very limited success in identifying potent, non-peptide small molecule inhibitors. Accordingly, there is a need for non-peptide, drug-like, small molecule inhibitors of the p53-MDM2 interaction. The structural basis of the interaction p53 and MDM2 has been established by x-ray crystallography (Kussie et al, Science 274:948 (1996)). Spiro-oxindole-based antagonists of the p53-MDM2 interaction are described in U.S. Patent Nos. 7,759,383 B2 and 7,737, 174 B2, U.S. Patent Appl. Pub. No. 2011/0112052 A2, and U.S. Appl. No. 13/294,315.
SUMMARY OF THE INVENTION
[0006] In one aspect, the present disclosure provides spiro-oxindoles having
Formula I:
Figure imgf000003_0001
wherein:
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
R2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
R3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
E is selected from the group consisting of -OR26a and -NR26bR26c;
R26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
R26b is R4;
R26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, -S02R5b, and R5; or
R26b and R26c taken together form a 4- to 9-membered optionally substituted heterocyclo;
wherein R4 and R5 has the meanings as described below in connection with Formula II;
R5b is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, and optionally substituted heteroaryl;
X is selected from the group consisting of O, S, and NR;
Y is selected from the group consisting of O, S, and NR ; R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
represents a single or a double bond,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
In another aspect, the present disclosure provides spiro-oxindoles having
Formula II:
Figure imgf000005_0001
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
R2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
R3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl; R is selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
R5 is selected from the group consisting of
Figure imgf000006_0001
R5-1 R5-2 R5-3 R5-4 wherein:
each R6a and R6b is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
R7 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
R8a and R8b are each independently selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl; or
R8a and R8b taken together with the carbon that they are attached form a 3- to 8-membered optionally substituted cycloalkyl;
W1 is selected from the group consisting of -OR9a and -NR9bR9c;
R9a is hydrogen;
R9b is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S02R9d, and -CONR9eR9f;
R9c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R9b and R9c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
R9d is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
R9e and R9f are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or R9e and R9f taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
W2 is selected from the group consisting of -OR10 and -NRl laRl lb;
with the proviso that when W1 is -OR9a and W2 is -OR10 then at least one of R7, R8a, and R8b is other than hydrogen;
R10 is hydrogen; or
one of R9a and R10 is hydrogen and the other is a metabolically cleavable group;
Rl la is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S02Rl lc, and -CONRlldRl le;
Rl lb is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
Rl la and Rl lb taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
Rl lc is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
Rl ld and Rl le are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
Rl ld and Rl le taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
n is 1, 2, 3, 4, or 5;
each R12a, R12b, R12c and R12d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R13 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R14 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
Z is selected from the group consisting of -OR15 and -NR16aR16b; or
Z and R14 taken together form a carbonyl, i.e., a C=0, group.
R15 is selected from the group consisting of hydrogen and metabolically cleavable group; Rlba is selected from the group consisting of -S02R and -CONR^R106; R16b is selected from the group consisting of hydrogen and optionally substituted alkyl;
R16c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R16d and R16e are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R16d and R16e taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo;
o is 1, 2, or 3;
p is 0, 1, 2, or 3;
each R17a, R17b, R17c and R17d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R18 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R19 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
R20 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
R21a and R21b are each hydrogen; or
one of R21a and R21b is hydrogen and the other is metabolically cleavable group;
q is 0, 1, 2, or 3;
r is 1, 2, or 3;
each R22a, R22b, R22c, and R22d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R23 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R24 is selected from the group consisting of -S02R24a and -CONR24bR24c; R a is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R24b and R24c are each independently selected from the group consisting of hydrogen, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R24b and R24c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo;
s and t are each independently 1, 2, or 3;
X is selected from the group consisting of O, S, and NR;
Y is selected from the group consisting of O, S, and NR ;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
represents a single or a double bond,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0008] In another aspect, the present disclosure provides compounds having
Formula I that inhibit the interaction between p53 or p53 -related proteins and MDM2 or MDM2 -related proteins.
[0009] In another aspect, the present disclosure provides methods to induce senescence, cell cycle arrest and/or apoptosis in cells containing functional p53 or p53 -related proteins, comprising contacting the cell with a compound of Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof,
[0010] In another aspect, the present disclosure provides methods of treating, ameliorating, or preventing a hyperproliferative disease, e.g., cancer, e.g., adrenal cortical cancer, advanced cancer, anal cancer, aplastic anemia, bile duct cancer, bladder cancer, bone cancer, bone metastasis, brain/CNS tumors in adults, brain/CNS tumors in children, breast cancer, breast cancer in men, cancer in children, cancer of unknown primary, Castleman disease, cervical cancer, colon/rectum cancer, endometrial cancer, esophagus cancer, Ewing family of tumors, eye cancer, gallbladder cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumor (GIST), gestational trophoblastic disease, Hodgkin disease, Kaposi sarcoma, kidney cancer, laryngeal and hypopharyngeal cancer, leukemia - acute lymphocytic (ALL) in adults, leukemia - acute myeloid (AML), leukemia - chronic lymphocytic (CLL), leukemia - chronic myeloid (CML), leukemia - chronic myelomonocytic (CMML), leukemia in children, liver cancer, lung cancer - non-small cell, lung cancer - small cell, lung carcinoid tumor, lymphoma of the skin, malignant mesothelioma, multiple myeloma, myelodysplasia syndrome, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, non-Hodgkin lymphoma in children, oral cavity and oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, pituitary tumors, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma - adult soft tissue cancer, skin cancer - basal and squamous cell, skin cancer - melanoma, small intestine cancer, stomach cancer, testicular cancer, thymus cancer, thyroid cancer, uterine sarcoma, vaginal cancer, vulvar cancer, Waldenstrom macroglobulinemia, or Wilms Tumor, in a patient comprising administering to the patient a compound of Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0011] In another aspect, the present disclosure provides methods of treating, ameliorating, or preventing a hyperproliferative disease, e.g., cancer, in a patient comprising administering to the patient a compound of Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, in combination with one or more additional therapeutic agents, e.g., one or more additional anticancer agents.
[0012] In another aspect, the present disclosure provides pharmaceutical compositions comprising a compound of Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, for treating, ameliorating, or preventing a hyperproliferative disease, e.g., cancer, in a patient.
[0013] In another aspect, the present disclosure provides kits comprising a compound of Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, for treating, ameliorating, or preventing a hyperproliferative disease, e.g., cancer, in a patient. BRIEF DESCRIPTION OF DRAWINGS
[0014] Fig. 1 is a line graph showing cell growth inhibition of MDM2 inhibitors in the SJSA-1 cell line as determined using the WST-based assay. SJSA-1 cells were treated with each compound for 4 days.
[0015] Fig. 2 is a line graph showing cell growth inhibition of MDM2 inhibitors in the RS4; 1 1 leukemia cell line as determined using the WST-based assay. RS4; 1 1 cells were treated with each compound for 4 days.
[0016] Fig. 3 is a line graph showing the antitumor activity of Compound
Example Nos. 22 and 24 in the SJSA-1 xenograft tumor model. Mice bearing SJSA-1 tumors (one tumor per mouse) were treated with Compound Example
Nos. 22 and 24 daily for 2 weeks via oral gavage at 100 mg/kg qD.
[0017] Fig. 4 is a line graph showing the stability of MDM2 inhibitors in a 1 : 1 methanol/water solution. The Y-axis represents the percent amount of the compound measured by UPLC. The X-axis represents the number of days the sample has been in the methanol/water solution.
[0018] Fig. 5 is a line graph showing the stability of MDM2 inhibitors in a 1 : 1 acetonitrile/water solution. The Y-axis represents the percent amount of the stable isomer measured by UPLC. The X-axis represents the number of days the sample has been in the acetonitrile/water solution.
[0019] Fig. 6 is a line graph showing the stability of Compound Example Nos. 10 and 1 1 in a 1 : 1 methanol/water solution with 10% TFA added. The Y-axis represents the percent amount of the compound measured by UPLC. The X-axis represents the number of hours the sample has been in solution.
DETAILED DESCRIPTION OF THE INVENTION [0020] Provided herein are compounds having Formulae I-XXVIII. These compounds inhibit the interaction between p53 or p53-related proteins and MDM2 or MDM2 -related proteins. By inhibiting the negative effect of MDM2 or MDM2-related proteins on p53 or p53 -related proteins, these compounds sensitize cells to inducers of apoptosis and/or cell cycle arrest. In one embodiment, compounds having Formulae I-XXVIII induce apoptosis and/or cell cycle arrest. Therefore, also provided herein are methods of sensitizing cells to inducers of apoptosis and/or cell cycle arrest and to methods of inducing apoptosis and/or cell cycle arrest in cells. In one embodiment, the methods comprise contacting the cells with one or more compounds having Formulae I-XXVIII alone or in combination with additional agent(s), e.g., an inducer of apoptosis or a cell cycle disrupter.
[0021] Also provided herein are methods of treating, ameliorating, or preventing disorders in an patient, comprising administering to the patient one or more compounds having Formulae I-XXVIII alone or in combination with additional agent(s), e.g., an inducer of apoptosis. Such disorders include those characterized by a dysregulation of apoptosis and those characterized by the proliferation of cells expressing functional p53 or p53-related proteins. In another embodiment, methods of protecting normal (e.g., non-hyperproliferative) cells in an animal from the toxic side effects of chemotherapeutic agents and treatments are provided. This method comprises administering to the animal one or more compounds having Formulae I-XXVIII.
[0022] Also provided herein are compounds having any one of
Formulae I-XXVIII for use in the manufacture of a medicament for treating a hyperproliferative disease such as cancer.
[0023] Also provided herein are compounds having any one of Formulae I-XXVIII, or a pharmaceutical composition comprising a compound having any one of Formulae I-XXVIII, for use in treating a hyperproliferative disease such as cancer.
Definitions
[0024] The term "anticancer agent" as used herein, refers to any therapeutic agent (e.g., chemotherapeutic compound and/or molecular therapeutic compound), antisense therapy, radiation therapy, or surgical intervention, used in the treatment of hyperproliferative diseases such as cancer (e.g., in mammals, e.g.., in humans).
[0025] The term "prodrug" as used herein, refers to a pharmacologically inactive derivative of a parent "drug" molecule that requires biotransformation (e.g., either spontaneous or enzymatic) within the target physiological system to release, or to convert (e.g., enzymatically, physiologically, mechanically, electromagnetically) the prodrug into the active drug. Prodrugs are designed to overcome problems associated with stability, water solubility, toxicity, lack of specificity, or limited bioavailability. Exemplary prodrugs comprise an active drug molecule itself and a chemical masking group (e.g., a group that reversibly suppresses the activity of the drug). Some prodrugs are variations or derivatives of compounds that have groups cleavable under metabolic conditions. Prodrugs can be readily prepared from the parent compounds using methods known in the art, such as those described in A Textbook of Drug Design and Development, Krogsgaard-Larsen and H. Bundgaard (eds.), Gordon & Breach, 1991, particularly Chapter 5: "Design and Applications of Prodrugs"; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; Prodrugs: Topical and Ocular Drug Delivery, K. B. Sloan (ed.), Marcel Dekker, 1998; Methods in Enzymology, K. Widder et al. (eds.), Vol. 42, Academic Press, 1985, particularly pp. 309-396; Burger's Medicinal Chemistry and Drug Discovery, 5th Ed., M. Wolff (ed.), John Wiley & Sons, 1995, particularly Vol. 1 and pp. 172-178 and pp. 949-982; Pro- Drugs as Novel Delivery Systems, T. Higuchi and V. Stella (eds.), Am. Chem. Soc, 1975; and Bioreversible Carriers in Drug Design, E. B. Roche (ed.), Elsevier, 1987.
Exemplary prodrugs become pharmaceutically active in vivo or in vitro when they undergo solvolysis under physiological conditions or undergo enzymatic degradation or other biochemical transformation (e.g., phosphorylation, hydrogenation, dehydrogenation, glycosylation). Prodrugs often offer advantages of water solubility, tissue compatibility, or delayed release in the mammalian organism. (See e.g., Bundgard, Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam (1985); and Silverman, The Organic Chemistry of Drug Design and Drug Action, pp. 352-401, Academic Press, San Diego, CA (1992)). Common prodrugs include acid derivatives such as esters prepared by reaction of parent acids with a suitable alcohol (e.g., a lower alkanol) or esters prepared by reaction of parent alcohol with a suitable carboxylic acid, (e.g., an amino acid), amides prepared by reaction of the parent acid compound with an amine, basic groups reacted to form an acylated base derivative (e.g., a lower alkylamide), or phosphorus-containing derivatives, e.g., phosphate, phosphonate, and phosphoramidate esters, including cyclic phosphate, phosphonate, and phosphoramidate, see, e.g., US 2007/0249564 Al.
[0027] The term "metabolically cleavable group" as used herein, refers to groups which can be cleaved from the parent molecule by metabolic processes and be substituted with hydrogen. Certain compounds containing metabolically cleavable groups may be prodrugs, i.e., they are pharmacologically inactive. Certain other compounds containing metabolically cleavable groups may be antagonists of the interaction between p53 and MDM2. In such cases, these compounds may have more, less, or equivalent activity of the parent molecule. Examples of metabolically cleavable groups include those derived from amino acids (see, e.g., US 2006/0241017 Al ; US 2006/0287244 Al; and WO 2005/046575 A2) or phosphorus-containing compounds (see, e.g., U.S. 2007/0249564 Al) as illustrated in Scheme 1.
Scheme 1
Figure imgf000014_0001
parent amino acid amino acid ester parent drug drug
Figure imgf000014_0002
Parent phosphite phosphate ester parent drug U[ uy
[0028] The term "pharmaceutically acceptable salt" as used herein, refers to any salt (e.g., obtained by reaction with an acid or a base) of a compound provided herein that is physiologically tolerated in the target animal (e.g., a mammal). Salts of the compounds of provided herein may be derived from inorganic or organic acids and bases. Examples of acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, sulfonic, naphthalene-2-sulfonic, benzenesulfonic acid, and the like. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds provided herein and their pharmaceutically acceptable acid addition salts.
[0029] Examples of bases include, but are not limited to, alkali metal (e.g., sodium) hydroxides, alkaline earth metal (e.g., magnesium) hydroxides, ammonia, and compounds of formula NW , wherein W is C1-4 alkyl, and the like.
[0030] Examples of salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, chloride, bromide, iodide, 2-hydroxyethanesulfonate, lactate, maleate, mesylate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, undecanoate, and the like. Other examples of salts include anions of the compounds provided herein compounded with a suitable cation such as Na+, NH4 +, and NW4 + (wherein W is a Ci-4 alkyl group), and the like. For therapeutic use, salts of the compounds provided herein are contemplated as being pharmaceutically acceptable. However, salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
[0031] The term "solvate" as used herein, refers to the physical association of a compound provided herein with one or more solvent molecules, whether organic or inorganic. This physical association often includes hydrogen bonding. In certain instances, the solvate is capable of isolation, for example, when one or more solvate molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolable solvates. Exemplary solvates include hydrates, ethanolates, and methanolates.
[0032] The term "monovalent pharmaceutically acceptable cation" as used herein refers to inorganic cations such as, but not limited to, alkaline metal ions, e.g., Na+ and K+, as well as organic cations such as, but not limited to, ammonium and substituted ammonium ions, e.g., NH4 , NHMe3 , NH2Me2 , NHMe3 and NMe4 +.
[0033] The term "divalent pharmaceutically acceptable cation" as used herein refers to inorganic cations such as, but not limited to, alkaline earth metal cations, e.g., Ca2+ and Mg2+.
[0034] Examples of monovalent and divalent pharmaceutically acceptable cations are discussed, e.g., in Berge et al. J. Pharm. Set, 66: \-\9 (1997).
[0035] The term "therapeutically effective amount," as used herein, refers to that amount of the therapeutic agent (including the compounds and compositions of matter provided herein) sufficient to result in amelioration of one or more symptoms of a disorder, or prevent advancement of a disorder, or cause regression of the disorder. For example, with respect to the treatment of cancer, in one embodiment, a therapeutically effective amount can refer to the amount of a therapeutic agent that decreases the rate of tumor growth, decreases tumor mass, decreases the number of metastases, increases time to tumor progression, increase tumor cell apoptosis, or increases survival time by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%.
[0036] The terms "sensitize" and "sensitizing," as used herein, refer to making, through the administration of a first therapeutic agent (e.g., a compound provided herein), an animal or a cell within an animal more susceptible, or more responsive, to the biological effects (e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell division, cell growth, proliferation, invasion, angiogenesis, necrosis, or apoptosis) of a second therapeutic agent. The sensitizing effect of a first agent on a target cell can be measured as the difference in the intended biological effect (e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell growth, proliferation, invasion, angiogenesis, or apoptosis) observed upon the administration of a second agent with and without administration of the first agent. The response of the sensitized cell can be increased by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 150%, at least about 200%, at least about 250%, at least 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500% over the response in the absence of the first agent.
[0037] The term "dysregulation of apoptosis," as used herein, refers to any aberration in the ability of (e.g., predisposition) a cell to undergo cell death via apoptosis. Dysregulation of apoptosis is associated with or induced by a variety of conditions, non-limiting examples of which include, autoimmune disorders (e.g., systemic lupus erythematosus, rheumatoid arthritis, graft-versus-host disease, myasthenia gravis, or Sj5gren's syndrome), chronic inflammatory conditions (e.g., psoriasis, asthma or Crohn's disease), hyperproliferative disorders (e.g., tumors, B cell lymphomas, or T cell lymphomas), viral infections (e.g., herpes, papilloma, or HIV), and other conditions such as osteoarthritis and atherosclerosis. It should be noted that when the dysregulation is induced by or associated with a viral infection, the viral infection may or may not be detectable at the time dysregulation occurs or is observed. That is, viral-induced dysregulation can occur even after the disappearance of symptoms of viral infection.
[0038] The term "functional p53," as used herein, refers to wild-type p53 expressed at normal, high, or low levels and mutant or allelic variants of p53 that retain(s) at least about 5% of the activity of wild-type p53, e.g., at least about 10%, about 20%, about 30%, about 40%, about 50%, or more of wild-type activity.
[0039] The term "p53 -related protein," as used herein, refers to proteins that have at least 25% sequence homology with p53, have tumor suppressor activity, and are inhibited by interaction with MDM2 or MDM2 -related proteins. Examples of p53-related proteins include, but are not limited to, p63 and p73.
[0040] The term "MDM2 -related protein," as used herein, refers to proteins that have at least 25% sequence homology with MDM2, and interact with and inhibit p53 or p53-related proteins. Examples of MDM2-related proteins include, but are not limited to, MDMX. [0041] The term "senescence" as used herein, refers to the phenomenon whereby non-cancerous diploid cells lose the ability to divide, and characterized in part by telomeric dysfunction or shortening.
[0042] The term "hyperproliferative disease," as used herein, refers to any condition in which a localized population of proliferating cells in an animal is not governed by the usual limitations of normal growth. Examples of hyperproliferative disorders include tumors, neoplasms, lymphomas, leukemias and the like. A neoplasm is said to be benign if it does not undergo invasion or metastasis and malignant if it does either of these. A "metastatic" cell means that the cell can invade neighboring body structures. Hyperplasia is a form of cell proliferation involving an increase in cell number in a tissue or organ without significant alteration in structure or function. Metaplasia is a form of controlled cell growth in which one type of fully differentiated cell substitutes for another type of differentiated cell.
[0043] The pathological growth of activated lymphoid cells often results in an autoimmune disorder or a chronic inflammatory condition. As used herein, the term "autoimmune disorder" refers to any condition in which an organism produces antibodies or immune cells which recognize the organism's own molecules, cells or tissues. Non-limiting examples of autoimmune disorders include autoimmune hemolytic anemia, autoimmune hepatitis, Berger's disease or IgA nephropathy, celiac sprue, chronic fatigue syndrome, Crohn's disease, dermatomyositis, fibromyalgia, graft versus host disease, Grave's disease, Hashimoto's thyroiditis, idiopathic thrombocytopenia purpura, lichen planus, multiple sclerosis, myasthenia gravis, psoriasis, rheumatic fever, rheumatic arthritis, scleroderma, Sj5gren's syndrome, systemic lupus erythematosus, type 1 diabetes, ulcerative colitis, vitiligo, and the like.
[0044] The term "neoplastic disease," as used herein, refers to any abnormal growth of cells being either benign (non-cancerous) or malignant (cancerous).
[0045] The term "normal cell," as used herein, refers to a cell that is not undergoing abnormal growth or division. Normal cells are non-cancerous and are not part of any hyperproliferative disease or disorder. [0046] The term "anti-neoplastic agent," as used herein, refers to any compound that retards the proliferation, growth, or spread of a targeted (e.g., malignant) neoplasm.
[0047] The terms "prevent," "preventing," and "prevention," as used herein, refer to a decrease in the occurrence of pathological cells (e.g., hyperproliferative or neoplastic cells) in an animal. The prevention may be complete, e.g., the total absence of pathological cells in a subject. The prevention may also be partial, such that the occurrence of pathological cells in a subject is less than that which would have occurred without treatment with one or more compounds provided herein.
[0048] The term "apoptosis-modulating agents," as used herein, refers to agents which are involved in modulating (e.g., inhibiting, decreasing, increasing, promoting) apoptosis. Examples of apoptosis-modulating agents include proteins which comprise a death domain such as, but not limited to, Fas/CD95, TRAMP, TNF RI, DRl, DR2, DR3, DR4, DR5, DR6, FADD, and RIP. Other examples of apoptosis-modulating agents include, but are not limited to, TNFa, Fas ligand, antibodies to Fas/CD95 and other TNF family receptors, TRAIL (also known as Apo2 Ligand or Apo2L/TRAIL), antibodies to TRAIL-Rl or TRAIL-R2, Bcl-2, p53, BAX, BAD, Akt, CAD, PI3 kinase, PPl, and caspase proteins. Modulating agents broadly include agonists and antagonists of TNF family receptors and TNF family ligands. Apoptosis-modulating agents may be soluble or membrane bound (e.g. ligand or receptor). Apoptosis-modulating agents include those which are inducers of apoptosis, such as TNF or a TNF-related ligand, particularly a TRAMP ligand, a Fas/CD95 ligand, a TNFR-1 ligand, or TRAIL.
[0049] The term "pharmaceutically acceptable carrier" or "pharmaceutically acceptable vehicle" encompasses any of the standard pharmaceutical carriers, solvents, surfactants, or vehicles. Suitable pharmaceutically acceptable vehicles include aqueous vehicles and nonaqueous vehicles. Standard pharmaceutical carriers and their formulations are described in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, PA, 19th ed. 1995.
[0050] The terms "pulsatile administration," "pulsatile dose administration" or
"pulsatile dosing" as used herein, refer to intermittent (i.e., not continuous) administration of compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof, to a patient. Pulsatile dose administration regimens useful in the present disclosure encompass any discontinuous administration regimen that provides a therapeutically effective amount of compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof, to a patient in need thereof. Pulsatile dosing regimens can use equivalent, lower, or higher doses of compounds having Formulae I-XXVIII than would be used in continuous dosing regimens. Advantages of pulsatile dose administration of compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof, include, but are not limited to, improved safety, decreased toxicity, increased exposure, increased efficacy, and increased patient compliance. These advantages may be realized when compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof, are administered as a single agent or are administered in combination with one or more additional anticancer agents. On the day that compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof, are scheduled to be administered to the patient, administration can occur in a single or in divided doses, e.g., once-a-day, twice-a-day, three times a day, four times a day or more. In one embodiment, compounds having Formulae I-XXVIII, or pharmaceutically acceptable salts, solvates, or prodrugs thereof, are administered once (QD) or twice (BID) on the day it is schedule to be administered
The term "alkyl" as used herein by itself or part of another group refers to a straight-chain or branched saturated aliphatic hydrocarbon having from one to eighteen carbons or the number of carbons designated (e.g., Ci-Cis means 1 to 18 carbons). In one embodiment, the alkyl is a Ci-Cio alkyl. In another embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. In another embodiment, the alkyl is a C1-C3 alkyl. In another embodiment, the alkyl is a C2-C10 alkyl. In another embodiment, the alkyl is a C3-C10 alkyl. In another embodiment, the alkyl is a C3-C6 alkyl. Exemplary alkyl groups include methyl, ethyl, w-propyl, isopropyl, w-butyl, sec-butyl, isobutyl, tert-butyl, w-pentyl, isopentyl, neopentyl, w-hexyl, isohexyl, w-heptyl, 4,4-dimethylpentyl, w-octyl, 2,2,4-trimethylpentyl, nonyl, decyl and the like. [0052] The term "optionally substituted alkyl" as used herein by itself or part of another group means that the alkyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from hydroxy (i.e. , -OH), nitro (i.e. , -N02), cyano (i.e. , -CN), amino, optionally substituted cycloalkyl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, -C02Rc, -CORc, -S02Rd, -N(Re)CORf, -N(Re)S02Rs or -N(Re)C=N(Rh)-amino, wherein Rc is hydrogen, optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl; Rd is optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl; Re is hydrogen, optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl; Rf is hydrogen, optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl; Rs is optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl; and Rh is hydrogen, -CN, optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl. In one embodiment, the optionally substituted alkyl is substituted with two substituents. In another embodiment, the optionally substituted alkyl is substituted with one substituent. In another embodiment, the substituents are selected from hydroxyl (i.e. , a hydroxyalkyl, e.g. , a monohydroxyalkyl or dihydroxyalkyl), optionally substituted cycloalkyl (i.e. , a (cycloalkyl)alkyl), optionally substituted heterocyclo (i.e., a (heterocyclo)alkyl), -CO2H, or amino (i.e. , an aminoalkyl). Exemplary optionally substituted alkyl groups include -CH2OCH3, -CH2CH2NH2, -CH2CH2NH(CH3), -CH2CH2CN, -CH2CO2H, -CH2CONH2, -CH2SO2CH3, -CH2CH2SO2CH3, -C(CH3)2C02H, hydroxymethyl, hydroxyethyl, hydroxypropyl, and the like.
[0053] The term "alkylenyl" as used herein by itself or part of another group refers to a divalent alkyl radical containing one, two, three, four, or more joined methylene groups. Exemplary alkylenyl groups include -((¾)-, -(CH2)2-, - (CH2)3-, -(CH2)4-, and the like.
[0054] The term "optionally substituted alkylenyl" as used herein by itself or part of another group means the alkylenyl as defined above is either unsubstituted or substituted with one, two, three, or four substituents independently selected from the group consisting of optionally substituted Ci-Ce alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl. In one embodiment, the optionally substituted Ci-Ce alkyl is methyl. In one embodiment, the optionally substituted aryl is a phenyl optionally substituted with one or two halo groups. Exemplary optionally substituted alkylenyl groups include -CH(CH3)-, -C(CH3)2-, -CH2CH(CH3)-, -CH2CH(CH3)CH2-, -CH2CH(Ph)CH2-, -CH(CH3)CH(CH3)-, and the like.
[0055] The term "haloalkyl" as used herein by itself or part of another group refers to an alkyl as defined above having one to six halo substituents. In one embodiment, the haloalkyl has one, two or three halo substituents. Exemplary haloalkyl groups include trifluoromethyl, -CH2CH2F and the like.
[0056] The term "monohydroxyalkyl" as used herein by itself or part of another group refers to an alkyl as defined above having exactly one hydroxy substituent. Exemplary hydroxyalkyl groups include hydroxymethyl, hydroxyethyl, hydroxypropyl, and the like.
[0057] The term "dihydroxyalkyl" as used herein by itself or part of another group refers to alkyl as defined above having exactly two hydroxyl substituents. Exemplary dihydroxyalkyl groups include -CH2CH2CCH3(OH)CH2OH, -CH2CH2CH(OH)CH(CH3)OH, -CH2(OH)CH2OH, -CH2CH(CH2OH)2, -CH2CH2CH(OH)C(CH3)2OH, -CH2CH2CCH3(OH)CH(CH3)OH, and the like, including stereoisomers thereof.
[0058] The term "hydroxycycloalkyl" as used herein by itself or part of another group refers to an optionally substituted cycloalkyl as defined below having a least one, e.g. , one or two hydroxy substituents. Exemplary hydroxycycloalkyl groups include:
Figure imgf000022_0001
Figure imgf000022_0002
and the like, including stereoisomers thereof. [0059] The term "optionally substituted (cycloalkyl)alkyl" as used herein by itself or part of another group refers to an optionally substituted alkyl as defined above having an optionally substituted cycloalkyl (as defined below) substituent. Exemplary optionally substituted (cycloalkyl)alkyl groups include:
Figure imgf000023_0001
and the like, including stereoisomers thereof.
[0060] The term "(heterocyclo)alkyl" as used herein by itself or part of another group refers to an alkyl as defined above having an optionally substituted heterocyclo (as defined below) substituent.
[0061] The term "aralkyl" as used herein by itself or part of another group refers to an optionally substituted alkyl as defined above having one, two or three optionally substituted aryl substituents. In one embodiment, the aralkyl has two optionally substituted aryl substituents. In another embodiment, the aralkyl has one optionally substituted aryl substituent. In another embodiment, the aralkyl is an aryl(Ci-C4 alkyl). In another embodiment, the aryl(Ci-C4 alkyl) has two optionally substituted aryl substituents. In another embodiment, the aryl(Ci-C4 alkyl) has one optionally substituted aryl substituent. Exemplary aralkyl groups include, for example, benzyl, phenylethyl, (4-fluorophenyl)ethyl, phenylpropyl, diphenylmethyl (i.e. , Ph2CH-), diphenylethyl (Ph2CHCH2-) and the like.
[0062] The term "cycloalkyl" as used herein by itself or part of another group refers to saturated and partially unsaturated (containing one or two double bonds) cyclic hydrocarbon groups containing one to three rings having from three to twelve carbon atoms (i.e. , C3-C12 cycloalkyl) or the number of carbons designated. In one embodiment, the cycloalkyl has one ring. In another embodiment, the cycloalkyl is a C3-C6 cycloalkyl. Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, norbornyl, decalin, adamantyl and the like. The term "optionally substituted cycloalkyl" as used herein by itself or part of another group means the cycloalkyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, -C02Rc, -CORc, -S02Rd, -N(Re)CORf, -N(Re)S02Rs or -N(Re)C=N(Rh)-amino, wherein Rc, Rd, Re, Rf, Rs, and Rh are as defined above in connection with optionally substituted alkyl. The term "optionally substituted cycloalkyl" also means the cycloalkyl as defined above may be fused to an optionally substituted aryl. In one embodiment, the optionally substituted cycloalkyl is substituted with two substituents. In another embodiment, the optionally substituted cycloalkyl is substituted with one substituent. In another embodiment, the substituents are selected from hydroxy (i.e., a hydroxycycloalkyl, e.g., a monohydroxycycloalkyl or dihydroxycycloalkyl) or -CO2H. Exemplary optionally substituted cycloalkyl groups include:
Figure imgf000024_0001
and the like.
[0064] The term "alkenyl" as used herein by itself or part of another group refers to an alkyl group as defined above containing one, two or three carbon-to-carbon double bonds. In one embodiment, the alkenyl has one carbon-to-carbon double bond. Exemplary alkenyl groups include -CH=CH2, -CH2CH=CH2, - CH2CH2CH=CH2, -CH2CH2CH=CHCH3 and the like.
[0065] The term "optionally substituted alkenyl" as used herein by itself or part of another group means the alkenyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido. Exemplary optionally substituted alkenyl groups include -CH=CHPh, -CH2CH=CHPh and the like.
[0066] The term "cycloalkenyl" as used herein by itself or part of another group refers to a cycloalkyl group as defined above containing one, two or three carbon- to-carbon double bonds. In one embodiment, the cycloalkenyl has one carbon-to- carbon double bond. Exemplary cycloalkenyl groups include cyclopentene, cyclohexene and the like.
[0067] The term "optionally substituted cycloalkenyl" as used herein by itself or part of another group means the cycloalkenyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido.
[0068] The term "alkynyl" as used herein by itself or part of another group refers to an alkyl group as defined above containing one to three carbon-to-carbon triple bonds. In one embodiment, the alkynyl has one carbon-to-carbon triple bond. Exemplary alkynyl groups include -C≡CH, -C≡CCH3, -CH2C≡CH, - CH2CH2C≡CH and -CH2CH2C≡CCH3.
[0069] The term "optionally substituted alkynyl" as used herein by itself or part of another group means the alkynyl as defined above is either unsubstituted or substituted with one, two or three substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido. Exemplary optionally substituted alkenyl groups include -C≡CPh, -CH2C≡CPh and the like.
[0070] The term "aryl" as used herein by itself or part of another group refers to monocyclic and bicyclic aromatic ring systems having from six to fourteen carbon atoms (i.e., C6-C14 aryl) such as phenyl (abbreviated as Ph), 1-naphthyl and 2-naphthyl and the like.
[0071] The term "optionally substituted aryl" as used herein by itself or part of another group means the aryl as defined above is either unsubstituted or substituted with one to five substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, -CONHS02Me, -C02Rc, -CORc, -S02Rd, - N(Re)CORf, -N(Re)S02Rs or -N(Re)C=N(Rh)-amino, wherein Rc, Rd, Re, Rf, Rs, and Rh are as defined above in connection with optionally substituted alkyl. In one embodiment, the optionally substituted aryl is an optionally substituted phenyl. In one embodiment, the optionally substituted phenyl has four substituents. In another embodiment, the optionally substituted phenyl has three substituents. In another embodiment, the optionally substituted phenyl has two substituents. In another embodiment, the optionally substituted phenyl has one substituent. Exemplary substituted aryl groups include 2-methylphenyl, 2- methoxyphenyl, 2-fluorophenyl, 2-chlorophenyl, 2-bromophenyl, 3- methylphenyl, 3 -methoxyphenyl, 3 -fluorophenyl, 3-chlorophenyl, 4- methylphenyl, 4-ethylphenyl, 4-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 2,6-di- fluorophenyl, 2,6-di-chlorophenyl, 2-methyl, 3 -methoxyphenyl, 2-ethyl, 3- methoxyphenyl, 3,4-di-methoxyphenyl, 3,5-di-fluorophenyl 3,5-di-methylphenyl and 3,5-dimethoxy, 4-methylphenyl, 2-fluoro-3-chlorophenyl, 3-chloro-4- fluorophenyl, 4-C02H-phenyl and the like. The term optionally substituted aryl is meant to include groups having fused optionally substituted cycloalkyl and fused optionally substituted heterocyclo rings. Examples include
Figure imgf000027_0001
and the like.
[0072] The term "heteroaryl" as used herein by itself or part of another group refers to monocyclic and bicyclic aromatic ring systems having from five to fourteen ring atoms (i.e., 5- to 14-membered heteroaryl) and one, two, three, or four heteroatoms independently selected from the group consisting of oxygen, nitrogen and sulfur. In one embodiment, the heteroaryl has three heteroatoms. In one embodiment, the heteroaryl has two heteroatoms. In one embodiment, the heteroaryl has one heteroatom. In one embodiment, the heteroaryl is a 5-membered heteroaryl. In another embodiment, the heteroaryl is a 6-membered heteroaryl. In another embodiment, the heteroaryl is a 6-membered heteroaryl having one or two nitrogen atoms. Exemplary heteroaryl groups include
1- pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl,
2- oxazolyl, 4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl,
2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, purinyl, 2-benzimidazolyl, 4-benzimidazolyl, 5-benzimidazolyl, 2-benzthiazolyl, 4-benzthiazolyl, 5-benzthiazolyl, 5-indolyl, 3-indazolyl, 4-indazolyl, 5-indazolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 2-quinolyl 3-quinolyl, 6-quinolyl and the like. The term heteroaryl is meant to include possible N-oxides. Exemplary N-oxides include pyridyl N-oxide and the like.
[0073] The term "optionally substituted heteroaryl" as used herein by itself or part of another group means the heteroaryl as defined above is either unsubstituted or substituted with one to four substituents, typically one or two substituents, independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, -C02Rc, -CORc, -S02Rd, -N(Re)CORf, -N(Re)S02Rs or
-N(Re)C=N(Rh)-amino, wherein Rc, Rd, Re, Rf, Rs, and Rh are as defined above in connection with optionally substituted alkyl. In one embodiment, the optionally substituted heteroaryl has one substituent. In another embodiment, the substituent is an optionally substituted aryl, aralkyl, or optionally substituted alkyl. In another embodiment, the substituent is an optionally substituted phenyl. Any available carbon or nitrogen atom may be substituted. Exemplary optionally substituted heteroaryl groups include
Figure imgf000028_0001
and the like.
The term "heterocyclo" as used herein by itself or part of another group refers to saturated and partially unsaturated (containing one or two double bonds) cyclic groups containing one to three rings having from three to fourteen ring members (i.e., 3- to 14-membered heterocyclo) and at least one oxygen, sulfur, including sulfoxide and sulfone, and/or nitrogen atom. In one embodiment, the heterocyclo group is chosen from a 5- or 6-membered cyclic group containing one ring and one or two oxygen and/or nitrogen atoms. In one embodiment, the heterocyclo group is a 6-membered cyclic group containing one ring and one sulfur atom, including sulfoxide and sulfone. The heterocyclo can be optionally linked to the rest of the molecule through a carbon or nitrogen atom. Exemplary heterocyclo groups include:
Figure imgf000029_0001
and the like.
[0075] The term "optionally substituted heterocyclo" as used herein by itself or part of another group means the heterocyclo as defined above is either unsubstituted or substituted with one to four substituents independently selected from halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, -C02Rc, -CORc, -S02 d, -N(Re)CORf, -N(Re)S02Rs or -N(Re)C=N(Rh)-amino, wherein Rc, Rd, Re, Rf, Rs, and Rh are as defined above in connection with optionally substituted alkyl. Substitution may occur on any available carbon or nitrogen atom. Exemplary substituted heterocyclo groups include
Figure imgf000029_0002
and the like. An optionally substituted heterocyclo may be fused to an aryl group to provide an optionally substituted aryl as described above.
[0076] The term "alkoxy" as used herein by itself or part of another group refers to a haloalkyl, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl or optionally substituted alkynyl attached to a terminal oxygen atom. Exemplary alkoxy groups include methoxy, tert-butoxy, -OCH2CH=CH2, -OCH2CH2OH, -OC(CH3)2C02H, and the like.
[0077] The term "aryloxy" as used herein by itself or part of another group refers to an optionally substituted aryl attached to a terminal oxygen atom. Exemplary aryloxy groups include phenoxy and the like.
[0078] The term "aralkyloxy" as used herein by itself or part of another group refers to an aralkyl attached to a terminal oxygen atom. Exemplary aralkyloxy groups include benzyloxy and the like. [0079] The term "alkylthio" as used herein by itself or part of another group refers to a haloalkyl, aralkyl, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl or optionally substituted alkynyl attached to a terminal sulfur atom. Exemplary alkyl groups include -SCH3 and the like.
[0080] The term "halo" or "halogen" as used herein by itself or part of another group refers to fluoro, chloro, bromo or iodo. In one embodiment, the halo is fluoro or chloro.
[0081] The term "amino" as used herein by itself or part of another group refers to a radical of formula -NRaRb wherein Ra and Rb are independently hydrogen, haloalkyl, aralkyl, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl or optionally substituted heteroaryl; or Ra and Rb taken together with the nitrogen atom to which they are attached form a four to seven membered optionally substituted heterocyclo. Exemplary amino groups include -NH2, -N(H)CH3, -N(CH3)2,
-N(H)CH2CH3, -N(CH2CH3), -N(H)CH2Ph and the like.
[0082] The term "carboxamido" as used herein by itself or part of another group refers to a radical of formula -CO-amino. Exemplary carboxamido groups include -CONH2, -CON(H)CH3, -CON(H)Ph, -CON(H)CH2CH2Ph, - CON(CH3)2, CON(H)CHPh2 and the like.
[0083] The term "sulfonamido" as used herein by itself or part of another group refers to a radical of formula -S02-amino. Exemplary sulfonamido groups include -S02NH2, -S02N(H)CH3, -S02N(H)Ph and the like.
[0084] The term "about," as used herein, includes the recited number ± 10%.
Thus, "about 10" means 9 to 1 1.
[0085] Certain of the compounds of the present disclosure may exist as stereoisomers, i.e., isomers that differ only in the spatial arrangement of atoms, including optical isomers and conformational isomers (or conformers). The disclosure includes all stereoisomers, both as pure individual stereoisomer preparations and enriched preparations of each, and both the racemic mixtures of such stereoisomers as well as the individual diastereomers and enantiomers that may be separated according to methods that are well known to those of skill in the art. [0086] The term "substantially free of as used herein means that the compound comprises less than about 25% of other stereoisomers, e.g., diastereomers and/or enantiomers, as established using conventional analytical methods routinely used by those of skill in the art. In one embodiment, the amount of other stereoisomers is less than about 24%, less than about 23%, less than about 22%, less than about 21%, less than about 20%, less than about 19%, less than about 18%, less than about 17%, less than about 16%, less than about 15%, less than about 14%, less than about 13%, less than about 12%, less than about 1 1%, less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, less than about 1%, or less than about 0.5%.
[0087] Stereoisomerically enriched compounds that contain about 95% or more of a desired stereoisomer, for example, about 96% or more, about 97% or more, about 98% or more, or about 99% or more are referred to herein as "substantially pure stereoisomers."
[0088] Stereoisomerically enriched compounds that contain about 99% or more of a desired stereoisomer are referred to herein as "pure" stereoisomers." The purity of any stereoisomerically enriched compound can be determined using conventional analytical methods such as, for example, normal phase HPLC, reverse phase HPLC, chiral HPLC, and XH and 13C NMR.
Compounds
[0089] In one embodiment, compounds of Formula I are provided:
Figure imgf000031_0001
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
R2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
R3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
E is selected from the group consisting of -OR26a and -NR26bR26c;
R26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
R26b is R4;
R26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, -S02R5b, and R5;
wherein R4 and R5 have the meanings as described below for Formula II; or R26b and R26c taken together form a 4- to 9-membered optionally substituted heterocyclo;
R5b is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, and optionally substituted heteroaryl;
X is selected from the group consisting of O, S, and NR;
Y is selected from the group consisting of O, S, and NR ;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
represents a single or a double bond,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
In another embodiment, compounds of Formula I are provided, wherein:
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
R2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
R3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
E is selected from the group consisting of -OR26a and -NR26bR26c;
R26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
R26b is selected from the group consisting of hydrogen and optionally substituted alkyl;
R26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, and -S02R5b; or R and R c taken together form a 4- to 9-membered optionally substituted heterocyclo;
R5b is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, and optionally substituted heteroaryl;
X is selected from the group consisting of O, S, and NR;
Y is selected from the group consisting of O, S, and NR ;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
represents a single or a double bond,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
In another embodiment, compounds of Formula I are provided, wherein:
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
R2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
E is selected from the group consisting of -OR26a and -NR26bR26c;
R26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
R26b is selected from the group consisting of hydrogen and optionally substituted alkyl;
R26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, and -S02R5b; wherein R4 and R5 have the meanings as described below for Formula II; or R26b and R26c taken together form a 4- to 9-membered optionally substituted heterocyclo;
R5b is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, and optionally substituted heteroaryl;
X is selected from the group consisting of O, S, and NR;
Y is selected from the group consisting of O, S, and NR ;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
represents a single or a double bond,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
In another embodiment, compounds of Formula II are provided:
Figure imgf000035_0001
wherein:
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
R2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; R is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl;
R4 is selected from the group consisting of hydrogen and optionally substituted alkyl;
R5 is selected from the group consisting of:
Figure imgf000036_0001
R5-1 R5-2 R5-3 R5-4 wherein:
each R6a and R6b is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
R7 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
R8a and R8b are each independently selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl; or
R8a and R8b taken together with the carbon that they are attached form a 3- to 8-membered optionally substituted cycloalkyl;
W1 is selected from the group consisting of -OR9a and -NR9bR9c;
R9a is hydrogen;
R9b is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S02R9d, and -CONR9eR9f;
R9c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or R9b and R9c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
R9d is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
R9e and R9f are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
R9e and R9f taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
W2 is selected from the group consisting of -OR10 and -NRl laRl lb;
with the proviso that when W1 is -OR9a and W2 is -OR10 then at least one of R7, R8a, and R8b is other than hydrogen;
R10 is hydrogen; or
one of R9a and R10 is hydrogen and the other is a metabolically cleavable group;
Rl la is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S02Rl lc, and -CONRl ldRl le;
Rl lb is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
Rl la and Rl lb taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
Rl lc is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
Rl ld and Rl le are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
Rl ld and Rl le together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
n is 1, 2, 3, 4, or 5;
each R12a, R12b, R12c and R12d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R13 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl; R is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
Z is selected from the group consisting of -OR15 and -NR16aR16b; or
Z and R14 taken together form a carbonyl, i.e. , a C=0, group.
R15 is selected from the group consisting of hydrogen and metabolically cleavable group;
R16a is selected from the group consisting of -S02R16c and -CONR16dR16e; R16b is selected from the group consisting of hydrogen and optionally substituted alkyl;
R16c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R16d and R16e are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R16d and R16e taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo;
o is 1 , 2, or 3 ;
p is 0, 1 , 2, or 3 ;
each R17a, R17b, R17c and R17d is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
R18 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R19 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
R20 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
R21a and R21b are each hydrogen; or
one of R21a and R21b is hydrogen and the other is metabolically cleavable group;
q is 0, 1 , 2, or 3 ;
r is 1 , 2, or 3; each R a, R , R c, and R is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
R23 is selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
R24 is selected from the group consisting of -S02R24a and -CONR24bR24c;
R24a is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R24b and R24c are each independently selected from the group consisting of hydrogen, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R24b and R24c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo;
s and t are each independently 1 , 2, or 3;
X is selected from the group consisting of O, S, and NR ;
Y is selected from the group consisting of O, S, and NR ;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
represents a single or a double bond,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0093] In another embodiment, the compound of Formula I or II is provided as a mixture of stereoisomers, e.g., a mixture of diastereomers and/or enantiomers, e.g., a racemic mixture. In another embodiment, the compound of Formula I or II is provided as a single stereoisomer.
[0094] In another embodiment, compounds of Formula III are provided:
Figure imgf000039_0001
wherein Rla, Rlb, Rlc, Rld, R2, R3a, R3b, E, X, and Y have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0095] In another embodiment, compounds of Formula III are provided wherein
E is NR4R5, and Rla, Rlb, Rlc, Rld, R2, R3a, R3b, R4, R5, X, and Y have the meanings as described above for Formula II, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0096] In another embodiment, compounds of Formula IV are provided:
Figure imgf000040_0001
IV
wherein Rla, Rlb, Rlc, Rld, R2, R3a, R3b, E, X, and Y have the meanings as described above for Formula I, or tautomer thereof, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0097] In another embodiment, compounds of Formula IV are provided wherein
E is -NR4R5, and Rla, Rlb, Rlc, Rld, R2, R3a, R3b, R4, R5, X, and Y have the meanings as described above for Formula II, or tautomer thereof, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0098] In another embodiment, compounds of Formula V are provided:
Figure imgf000040_0002
1. e. , R3a and R3b of Formula I are taken together form a 3- to 9-membered optionally substituted cycloalkyl, wherein Rla, Rlb, Rlc, Rld, R2, E, X, and Y have the meanings as described above for Formula I, u is 0, 1, 2, 3, 4, 5, or 6, v is 0, 1,
2, 3, or 4, and each R is independently halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0099] In another embodiment, compounds of Formula V are provided wherein E is NR4R5, and Rla, Rlb, Rlc, Rld, R2, R4, R5, X, and Y have the meanings as described above for Formula II, u is 0, 1, 2, 3, 4, 5, or 6, v is 0, 1, 2, 3, or 4, and each R is independently halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido or sulfonamido, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[00100] In another embodiment, compounds of Formula V are provided wherein u is 1, 2, 3, 4, 5, or 6, each R is independently (Ci-C4)alkyl and v is 0, 1, or 2, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[00101] In another embodiment compounds of Formula V are provided wherein v is 0 and u is 0, 1, 2, 3, 4, 5, or 6, , i.e., R3a and R3b of Formula I are taken together form a 3 - to 9-membered unsubstituted cycloalkyl.
[0100] In another embodiment compounds of Formula V are provided wherein v is 0 and u is 2, 3, or 4, i.e., R3a and R3b of Formula I are taken together form a 5-, 6-, or 7-membered unsubstituted cycloalkyl.
[0101] In another embodiment, compounds of any one of Formulae VI-XXI are provided:
Figure imgf000042_0001
VI VII VIII IX
Figure imgf000042_0002
wherein Rla, R1D, Rlc, Ria, Rz, RJa, R3b, E, X, and Y have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0102] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is NR4R5, and Rla, Rlb, Rlc, Rld, R2, R3a, R3b, R4, R5, X, and Y have the meanings as described above for Formula II, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0103] In another embodiment, compounds of any one of Formulae VI-XXI are provided substantially free of one or more other stereoisomers. In another embodiment, compounds of any one of Formulae VI-XXI are substantially pure stereoisomers. In another embodiment, compounds of any one of Formulae VI-XXI are pure stereoisomers.
[0104] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided wherein:
a) Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, fluoro, and chloro;
b) Rla and Rld are hydrogen; Rlb is selected from the group consisting of hydrogen and fluoro; and Rlc is selected from the group consisting of fluoro and chloro;
c) R2 is optionally substituted phenyl;
d) R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, and optionally substituted cycloalkyl;
e) R3b is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, and optionally substituted cycloalkyl;
f) R3a and R3b taken together form an optionally substituted 3- to 9-membered cycloalkyl;
g) E is -NR4R5 and R4 is hydrogen;
h) X is NH;
i) X is O;
j) X is S;
k) Y is O;
1) Y is S;
m) Y is NH; or
n) X and Y are NH;
or any combination thereof.
[0105] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R5 is R5-1; R6a and R6b are hydrogen; R7 is d-C4 alkyl; R8a and R8b are hydrogen; W is -OR10, R9 and R10 are hydrogen; and n is 2. [0106] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5; R5 is R5-1; R6a and R6b are hydrogen; R7 is d-C4 alkyl; R8a and R8b are hydrogen; W is -NRl laRllb, R9 is hydrogen; and n is 2.
[0107] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5; R5 is R5-1; R6a and R6b are hydrogen; R7 is d-C4 alkyl; R8a and R8b are hydrogen; W is -OR10, one of R9 and R10 is hydrogen and the other is a metabolically cleavable group; and n is 2.
[0108] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5; R5 is R5-2; R12a, R12b, R12c, and R12d are each hydrogen; R13 is hydrogen; Z is -OR15 and R15 is hydrogen; o is 1 or 2; and p is 1 or 2.
[0109] In another embodiment, of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5; R5 is R5-2; R12a, R12b, R12c, and R12d are each hydrogen; R13 is hydrogen; Z is -NR16aR16b; o is 1 or 2; and p is 1 or 2.
[0110] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5; R5 is R5-2; R12a, R12b, R12c, and R12d are each hydrogen; R13 is hydrogen; Z is -OR15 and R15 a metabolically cleavable group; o is 1 or 2; and p is 1 or 2.
[0111] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5; R5 is R5-3; R17a, R17b, R17c, and R17d are each hydrogen;
R18, R19, and R20 are hydrogen; R21a and R21b are hydrogen; and q and r are 1.
[0112] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5; R5 is R5-3; R17a, R17b, R17c, and R17d are each hydrogen; R18, R19, and R20 are hydrogen; one of R21a and R21b is hydrogen and the other is a metabolically cleavable group; and q and r are 1. [0113] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R2 is an optionally substituted aryl having the Formula R2-1 :
Figure imgf000045_0001
and R25a, R25 , R25c, R25d, and R25e are each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, amino, cyano, alkoxy, optionally substituted alkyl, haloalkyl, optionally substituted aryl, and optionally substituted heteroaryl. In one embodiment, R25a is selected from the group consisting of hydrogen and fluoro; R25b is chloro; R25c is selected from the group consisting of hydrogen and fluoro; and R25d and R25e are hydrogen.
[0114] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R2 is an optionally substituted pyridyl.
[0115] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of:
Figure imgf000046_0001
Figure imgf000046_0002
Figure imgf000046_0003
including stereoisomers, e.g., enantiomers, thereof, wherein:
R7 is optionally substituted C1-C4 alkyl;
R9a and R10 are each hydrogen; or
one of R9a and R10 is hydrogen and the other is a metabolically cleavable group;
R9b is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S02R9d, and -CONR9eR9f;
R9c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R9b and R9c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
R9d is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
R9e and R9f are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or R9e and R9f taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
Rl la is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S02Rl lc, and -CONRlldRl le;
Rl lb is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
Rl la and Rl lb taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
Rl lc is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
Rl ld and Rl le are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or Rl ld and Rl le taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
R14 is selected from the group consisting of hydrogen, C1-C4 alkyl, or C3-C6 cycloalkyl;
R15 is hydrogen or a metabolically cleavable group;
R16a is selected from the group consisting of -S02R16c and -CONR16dR16e;
R16b is selected from the group consisting of hydrogen and optionally substituted alkyl;
R16c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R16d and R16e are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R16d and R16e taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo;
R19 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl; R is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
R21a and R21b are each hydrogen; or
one of R21a and R21b is hydrogen and the other is metabolically cleavable group;
R24 is selected from the group consisting of -S02R24a and -CONR24bR24c;
R24a is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; and
R24b and R24c are each independently selected from the group consisting of hydrogen, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl, or
R24b and R24c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo.
[0116] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of R5-5, R5-6, R5-10, R5-11, R5-12, R5-13, and R5-14.
[0117] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of R5-10 and R5-12 and R14 is hydrogen or methyl and R15 is hydrogen.
[0118] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of:
Figure imgf000048_0001
wherein:
R7 is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl; and
R8a and R8b are each independently selected from the group consisting of hydrogen, methyl, ethyl, propyl, isopropyl, and cyclopropyl.
In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of:
Figure imgf000049_0001
R7 is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl;
R8a and R8b are each independently is selected from the group consisting of hydrogen, methyl, ethyl, propyl, isopropyl, and cyclopropyl;
R9d is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl; and Rl lc is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl.
[0120] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of:
Figure imgf000050_0001
wherein:
R7 is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl;
R8a and R8b are each independently is selected from the group consisting of hydrogen, methyl, ethyl, propyl, isopropyl, and cyclopropyl;
R9e is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl; and
Rl ld is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl.
[0121] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of:
Figure imgf000051_0001
wherein:
R is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl; and
R19 and R20 are each independently is selected from the group consisting of hydrogen, methyl, ethyl, propyl, isopropyl, and cyclopropyl.
[0122] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of:
R16c
Figure imgf000052_0001
wherein:
R14 is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl; and R c is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl.
In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of:
Figure imgf000053_0001
l¾HCONHR16d NHCONHR16d
Figure imgf000053_0002
wherein: R is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and cyclopropyl; and
R16d is selected from the group consisting of methyl, trifluoromethyl, ethyl, propyl, isopropyl, and cyclopropyl.
[0124] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of:
Figure imgf000054_0001
Figure imgf000054_0002
[0125] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided wherein E is -NR4R5, R4 is hydrogen, and R5 is selected from the group consisting of:
Figure imgf000055_0001
wherein:
R14 is selected from the group consisting of hydrogen and C1-C4 alkyl; and R15 is hydrogen or a metabolically cleavable group.
[0126] In one embodiment, the metabolically cleavable group at R15 is selected from the group consisting of:
Figure imgf000055_0002
wherein:
each R28a and R28b is independently selected from the group consisting of hydrogen, optionally substituted alkyl, and aralkyl;
R29a and R29b are each selected from the group consisting of hydrogen and optionally substituted alkyl;
v is 1, 2, 3, or 4; and
R30a and R30b are each selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, optionally substituted aryl, and monovalent pharmaceutically acceptable cation; or
taken together R30a and R30b represent a divalent pharmaceutically acceptable cation or an optionally substituted alkylenyl.
[0127] In another embodiment, the metabolically cleavable group at R15 is the residue of a natural or unnatural amino acid. In another embodiment, the metabolically cleavable group at R15 is the residue of glycine, isoleucine alanine, leucine, asparagine, lysine, aspartic acid, methionine, cysteine, phenylalanine, glutamic acid, threonine, glutamine, tryptophan, valine, proline, serine, tyrosine, arginine, and histidine.
[0128] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided wherein X is NH and Y is NH. [0129] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided wherein X is O and Y is NH.
[0130] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided wherein X is S and Y is NH.
[0131] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R3a and R3b are the same or different Ci-Cio alkyl.
[0132] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R3a and R3b are the same or different C2-C10 alkyl.
[0133] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R3a and R3b are the same or different C1-C6 alkyl, e.g., methyl, ethyl, M-propyl, isopropyl, w-butyl, sec-butyl, isobutyl, tert-butyl, w-pentyl, isopentyl, neopentyl, w-hexyl, isohexyl.
[0134] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R3a and R3b are the same or different C2-C6 alkyl.
[0135] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R3a and R3b are the same or different C1-C4 alkyl.
[0136] In another embodiment, compounds of any one of Formulae I-IV or VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R3a and R3b are the same or different C2-C4 alkyl.
[0137] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein R3a and R3b are the same or different C1-C3 alkyl.
[0138] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R3a and R3b are the same or different halo. [0139] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R3a and R3b are the same or different, and are selected from the group consisting of methyl, ethyl, propyl, isopropyl, butyl, and neopentyl.
[0140] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R3a and R3b are the same or different, and are selected from the group consisting of ethyl, propyl, isopropyl, butyl, and neopentyl.
[0141] In another embodiment, compounds of any one of Formulae I-IV or VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R3a and R3b are fluoro.
[0142] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R3a and R3b are the same, e.g., R3a is methyl and R3b is methyl, R3a is ethyl and R3b is ethyl, R3a is propyl and R3b is propyl, etc. If R3a and R3b are the same, the carbon atom to which R3a and R3b are attached is not an asymmetric center. Under these circumstances, Formula VI and X; Formula VII and XIII; Formula VIII and IX; Formula XI and XII; Formula XIV and XVI; Formula XV and XXI; Formula XVII and XX; and Formula XVIII and XIX represent equivalent isomers.
[0143] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R3a and R3b are different, e.g., R3a is methyl and R3b is ethyl, R3a is methyl and R3b is neopentyl, R3a is ethyl and R3b is propyl, etc..
[0144] In another embodiment, compounds of any one of Formulae I-IV or
VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R3a and R3b are taken together to form an optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring. In one embodiment, the optional substituent is a (Ci-C4)alkyl. In another embodiment, the cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring is unsubstituted.
[0145] In another embodiment, compounds of any one of Formulae I-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein Rla and Rld are hydrogen, Rlb and Rlc are each independently selected from the group consisting of hydrogen, chloro, and fluoro, and R2 is R2-1 wherein R25a, R25b, R25c, R25d, and R25e are each independently selected from the group consisting of hydrogen, chloro, and fluoro.
In another embodiment, compounds having any one of Formulae VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein:
E is NR4R5;
Rla, Rlb, Rlc, and Rld are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
R2 is:
Figure imgf000058_0001
wherein:
R25a, R25b, R25c, R25d, and R25e are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
R3a is Ci-C6 alkyl;
R3b is C1-C4 alkyl; or
R3a and R3b are taken together to form an optionally substituted 3- to 7-membered cycloalkyl;
R4 is selected from the group consisting of hydrogen and optionally substituted C1-C4 alkyl;
R5 is selected from the group consisting of:
Figure imgf000058_0002
wherein:
R14 is selected from the group consisting of hydrogen and optionally substituted C1-C4 alkyl;
X is selected from the group consisting of O, S, and NR;
Y is selected from the group consisting of O, S, and NR ; R is selected from the group consisting of hydrogen and optionally substituted C1-C4 alkyl; and
R is selected from the group consisting of hydrogen and optionally substituted C 1-C4 alkyl,
wherein the compounds are substantially free of one or more other stereoisomers.
In another embodiment, compounds having any one of Formulae VI-XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein:
E is NR4R5;
Rla is hydrogen;
Rlb, Rlc, and Rld are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
R2 is:
wherein:
R25a, R25b, R25c, R25d, and R2Se are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
R3a and R3b are methyl;
or R,a and R3b taken together form a cyclopentyl, cyclohexyl, or cycloheptyl ring;
R4 is hydrogen;
R5 is selected from the group consisting of:
Figure imgf000059_0002
X and Y are NH,
wherein the compounds are substantially free of one or more other stereoisomers.
[0148] In another embodiment, compounds having any one of Formulae VI -XXI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein E is NR4R5 , R4 is hydrogen, and R5 is selected from the group consisting of:
Figure imgf000060_0001
wherein the compound is substantially free of one or more other stereoisomers.
[0149] In another embodiment, compounds having Formula VI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein the compounds are substantially pure stereoisomers.
[0150] In another embodiment, compounds having Formula XVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein the compounds are substantially pure stereoisomers.
[0151] In another embodiment, compounds having Formula XVI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein the compounds are substantially pure stereoisomers.
[0152] In another embodiment, compounds having Formula XVI, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, are provided, wherein:
E is NR4R5;
R4 is hydrogen;
X and Y are NH;
R3a and R3b are methyl or ethyl, or
R3a and R3b taken together form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring, each optionally substituted by one of more C1-C4 alkyl groups; and R5 is selected from the group consisting of:
Figure imgf000061_0001
wherein the compounds are substantially pure stereoisomers.
[0153] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is -OR26a and Rla, Rlb, Rlc, Rld, R2, R3a, R3b, R26a, X, and Y have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In another embodiment, R26a is hydrogen.
[0154] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is -OR26a; Rla, Rlb, Rlc, Rld, R2, R26a, X, and Y have the meanings as described above for Formula I; and R3a and R3b taken together form a 4- to 7-membered optionally substituted cycloalkyl or 4- to 7-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In another embodiment, R26a is hydrogen. In another embodiment, R3a and R3b taken together form a cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or optionally substituted piperidinyl group.
[0155] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is -NR26bR26c; Rla, Rlb, Rlc, Rld, R2, R3a, R3b' R26b, R26c, X, and Y have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0156] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is -NR26bR26c; Rla, Rlb, Rlc, Rld, R2, R26b, R26c, X, and Y have the meanings as described above for Formula I; and R3a and R3b taken together form a 4- to 7-membered optionally substituted cycloalkyl or 4- to 7-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0157] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is -NR26bR26c; Rla, Rlb, Rlc, Rld, R2, R26b, R26c, X, and Y have the meanings as described above for Formula I; and R3a and R3b taken together form a 4- to 7-membered optionally substituted cycloalkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0158] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is -NR26bR26c, and Rla, Rlb, Rlc, Rld, R2, R26b, R26c, X, and Y have the meanings as described above for Formula I, and R3a and R3b taken together form a cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0159] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is -NR26bR26c, and Rla, Rlb, Rlc, Rld, R2, R26b, R26c, X, and Y have the meanings as described above for Formula I, and R3a and R3b taken together form a 4- to 7-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0160] In another embodiment, compounds of any one of Formula VI-XXI are provided, wherein E is -NR26bR26c, and Rla, Rlb, Rlc, Rld, R2, R26b, R26c, X, and Y have the meanings as described above for Formula I, and R3a and R3b taken together form a tetrahydropyranyl or optionally substituted piperidinyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0161] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is -NR26bR26c, and Rla, Rlb, Rlc, Rld, R2, R3a, R3b, X, and Y have the meanings as described above for Formula I, R26b is hydrogen, and R26c is optionally substituted cycloalkyl. In another embodiment, R26c is hydroxycycloalkyl. In another embodiment, R26c is cycloalkyl substituted with -C02H.
[0162] In another embodiment, compounds of any one of Formulae VI-XXI are provided, wherein E is -NR26bR26c, and Rla, Rlb, Rlc, Rld, R2, R3a, R3b, X, and Y have the meanings as described above for Formula I, R26b is hydrogen, and R26c is optionally substituted aryl. In another embodiment R26c is phenyl substituted with -C02H.
[0163] In another embodiment, compounds of Formula XXII are provided:
Figure imgf000063_0001
wherein:
Rla, Rlb, Rlc, Rld, R2, E, X, and Y have the meanings as described above for Formula I;
A is selected from the group consisting of CR27aR27b, O, S, SO, S02, and
28.
NR
R a is selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, sulfonamido, and -CO2R31;
R27b is selected from the group consisting of hydrogen and optionally substituted alkyl; or
R27a and R27b taken together form a 3- to 6-membered optionally substituted cycloalkyl or 3- to 6-membered optionally substituted heterocyclo;
R28 is selected from the group consisting of hydrogen, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, carboxamido, sulfonamido, amino, -C02R32a, -COR32a,
-S02R32b, -N(R32c)COR32d, -N(R32c)S02R32e and -N(R32c)C=N(R32f)-amino;
R32a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R32b is selected from the group consisting of optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R32c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl; R32d is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R32e is selected from the group consisting of optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl; and
R32f is selected from the group consisting of hydrogen, -CN, optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl;
B is -(CR29aR29b )w-;
D is -(CR30aR30b)x-;
each R29a, R29b, R30a, and R30b is independently selected from the group consisting of hydrogen, halo, nitro, cyano, hydroxy, amino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido and sulfonamido;
R31 is selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted aryl;
w is 0, 1, 2, 3, 4, 5, 6, 7, or 8; and
x is 0, 1, 2, 3, 4, 5, 6, 7, or 8;
with the proviso that the sum of w plus x is 1, 2, 3, 4, 5, 6, 7 or 8, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0164] In another embodiment, compounds of Formula XXII are provided as a single stereoisomer, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0165] In another embodiment, compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, and each R29a, R29b, R30a, and R30b is independently hydrogen or (Ci-C4)alkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0166] In another embodiment, compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, each R29a, R29b, R30a, and R30b is independently hydrogen or (Ci-C4)alkyl, and A is CR aR , or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0167] In another embodiment, compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, each R29a, R29b, R30a, and R30b is independently hydrogen or (Ci-C4)alkyl, A is CR27aR27b, and R27a and R27b are independently selected from the group consisting of hydrogen and alkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0168] In another embodiment, compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, each R29a, R29b, R30a, and R30b is independently hydrogen or (Ci-C4)alkyl, A is CR27aR27b, and R27a and R27b are hydrogen, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0169] In another embodiment, compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1 , 2, or 3, each R29a, R29b, R30a, and R 0b is independently hydrogen or (Ci-C4)alkyl, and A is O, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0170] In another embodiment, compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, each R29a, R29b, R30a, and R30b is independently hydrogen or (Ci-C4)alkyl, and A is S02, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0171] In another embodiment, compounds of Formula XXII are provided wherein w is 1 , 2, or 3, x is 1, 2, or 3, each R 9a, R 9b, R30a, and R 0b is independently hydrogen or (Ci-C4)alkyl, and A is NR28, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0172] In another embodiment, compounds of Formula XXII are provided wherein w is 1, 2, or 3, x is 1, 2, or 3, R29a, R29b, R 0a, and R30b are hydrogen, and A is NR28, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0173] In another embodiment, compounds of Formula XXII are provided wherein w is 1 , 2, or 3, x is 1 , 2, or 3, R29a, R29b, R 0a, and R30b are hydrogen, A is NR28, and R28 is selected from the group consisting of hydrogen, optionally substituted alkyl, carboxamido, sulfonamido, amino, -C02R , -COR , -S02R32b, -N(R32c)COR32d, -N(R32c)S02R32e and -N(R32c)C=N(R32f)-amino, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. [0174] In another embodiment, compounds of Formula XXII are provided wherein w and x are 1, each R29a, R29b, R30a, and R30b is hydrogen, A is NR28, and R28 is selected from the group consisting of hydrogen, optionally substituted alkyl, carboxamido, sulfonamido, -C02R32a, -COR32a, -S02R32b, -N(R32c)COR32d, -N(R32c)S02R32e and -N(R32c)C=N(R32f)-amino, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0175] In another embodiment, compounds of Formula XXII are provided wherein w and x are 2, each R29a, R29b, R30a, and R30b is hydrogen, A is NR28, and R28 is selected from the group consisting of hydrogen, optionally substituted alkyl, carboxamido, sulfonamido, -C02R32a, -COR32a, -S02R32b, -N(R32c)COR32d, -N(R32c)S02R32e and -N(R32c)C=N(R32f)-amino, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0176] In another embodiment, compounds of Formula XXII are provided wherein w and x are 3, each R29a, R29b, R30a, and R30b is hydrogen, A is NR28, and R28 is selected from the group consisting of hydrogen, optionally substituted alkyl, carboxamido, sulfonamido, -C02R32a, -COR32a, -S02R32b, -N(R32c)COR32d, -N(R32c)S02R32e and -N(R32c)C=N(R32f)-amino, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0177] In another embodiment, compounds of Formula XXII are provided wherein E is -OR26a, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0178] In another embodiment, compounds of Formula XXII are provided wherein E is -NR26bR26c, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0179] In another embodiment, compounds of Formula XXIII are provided:
Figure imgf000066_0001
, 1a r> lb π lc π Id r> 2
wherein R a, R , R c, R , R , E, X, and Y have the meanings as described above for Formula I, A has the meaning as described above for Formula XXII, and z is 0, 1, or 2, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. [0180] In another embodiment, compounds of Formula XXIV are provided:
Figure imgf000067_0001
wherein Rla, Rlb, Rlc, Rld, R2, E, X, and Y have the meanings as described above for Formula I, A has the meaning as described above for Formula XXII, and z has the meaning as described above for Formula XXIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0181] In another embodiment, compounds of Formula XXV are provided:
Figure imgf000067_0002
wherein Rla, Rlb, Rlc, Rld, R2, E, X, and Y have the meanings as described above for Formula I, A has the meaning as described above for Formula XXII, and z has the meaning as described above for Formula XXIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0182] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided, wherein E is -OR26a and R26a is hydrogen, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0183] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided, wherein E is -NR26bR26c, R26b is hydrogen, and R26c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and aralkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In another embodiment, R26c is a hydroxyalkyl, e.g., a monohydroxyalkyl or dihydroxyalkyl. In another embodiment, R26c is an optionally substituted (cycloalkyl)alkyl. In another embodiment, R26c is a (heterocyclo)alkyl. In another embodiment, R26c is optionally substituted cycloalkyl. In another embodiment, R 26cc is optionally substituted phenyl. [0184] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided wherein A is CR27aR27b, O, or NR28, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0185] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided wherein A is CHR27a, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0186] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided wherein A is CHR27a and R27a is selected from the group consisting of hydrogen, hydroxy, amino, alkoxy, optionally substituted alkyl, haloalkyl, substituted heteroaryl, carboxamido, sulfonamido, and -CO2H, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0187] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided wherein A is CH2 or NR28, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0188] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided wherein A is CH2, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0189] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided wherein A is NR28, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0190] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided wherein A is CH2 and z is 1, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0191] In another embodiment, compounds of any one of Formulae XXIII-XXV are provided wherein A is CH2, z is 1, E is -NR26bR26c, R26b is hydrogen, and R26c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and aralkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In another embodiment, R26c is optionally substituted cycloalkyl, e.g., a hydroxycycloalkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In another embodiment, R26c is optionally substituted aryl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In another embodiment, R c is optionally substituted heteroaryl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0192] In another embodiment, compounds of Formula XXVI are provided:
Figure imgf000069_0001
XXVI
wherein Rla, Rlb, Rlc, Rld, R2, and R26c have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0193] In another embodiment, compounds of Formula XXVII are provided:
Figure imgf000069_0002
XXVII
wherein Rla, Rlb, Rlc, Rld, R2, and R26c have the meanings as described above for Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0194] In another embodiment, compounds of Formula XXVIII are provided:
Figure imgf000069_0003
XXVIII
wherein Rla, Rlb, Rlc, Rld, R2, and R26c have the meanings as described above for
Formula I, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0195] In another embodiment, compounds of any one of Formulae
XXVI-XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein:
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, fluoro, and chloro;
R2 is optionally substituted phenyl; and R c is selected from the group consisting of optionally substituted (Ci-C4) alkyl, optionally substituted (C4-C8) cycloalkyl, optionally substituted phenyl, optionally substituted heteroaryl, and aralkyl;
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0196] In another embodiment, compounds of any one of Formulae I or
III -XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R26c is optionally substituted alkyl. In another embodiment, R26c is hydroxyalkyl. In another embodiment, R26c is dihydroxyalkyl. In another embodiment, R26c is (heterocyclo)alkyl.
[0197] In another embodiment, compounds of any one of Formulae I or
III -XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R26c is optionally substituted cycloalkyl. In another embodiment, R26c is hydroxycycloalkyl. In another embodiment, R26 is cycloalkyl substituted with -CO2H.
[0198] In another embodiment, compounds of any one of Formulae I or
III -XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R26c is selected from the group consisting of:
Figure imgf000070_0001
[0199] In another embodiment, compounds of any one of Formulae I or
III -XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R26c is optionally substituted aryl. In another embodiment,
R c is optionally substituted phenyl. In another embodiment, R c is phenyl substituted with one or two of the following groups: halo, cyano, hydroxy, alkyl, haloalkyl, alkoxy, carboxamido, sulfonamido, -CONHS02Me, -C02Rc, -CORc, -S02Rd, -N(Re)CORf, -N(Re)S02Rs or -N(Re)C=N(Rh)-amino, wherein Rc, Rd, Re, Rf, Rg, and Rh are as defined above in connection with optionally substituted alkyl. In another embodiment, R26c is phenyl substituted with one or two of the following groups: halo, cyano, alkyl, haloalkyl, alkoxy, carboxamido, sulfonamido, -C02Rc, or -S02Rd, wherein Rc and Rd are as defined above in connection with optionally substituted alkyl. In another embodiment, R26c is phenyl substituted with -C02H.
In another embodiment, compounds of any one of Formulae I or III-XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R26c is selected from the group consisting of:
Figure imgf000072_0001
In another embodiment, compounds of any one of Formulae I or III -XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R c is optionally substituted heteroaryl. In another embodiment, the optionally substituted heteroaryl is an optionally substituted furyl, thienyl, pyridyl, pyrimidyl, benzimidazolyl, benzthiazolyl, or indolyl, In another embodiment, R26c is heteroaryl substituted with one or two of the following groups: halo, cyano, hydroxy, alkyl, haloalkyl, alkoxy, carboxamido, sulfonamido, -CONHS02Me, -C02Rc, -CORc, -S02Rd, -N(Re)CORf, -N(Re)S02Rs or -N(Re)C=N(Rh)-amino, wherein Rc, Rd, Re, Rf, Rs, and Rh are as defined above in connection with optionally substituted alkyl. In another embodiment, R26c is heteroaryl substituted with one or two of the following groups: halo, cyano, alkyl, haloalkyl, alkoxy, carboxamido, sulfonamido, -C02Rc, or -S02Rd, wherein Rc and Rd are as defined above in connection with optionally substituted alkyl. In another embodiment, R26c is heteroaryl substituted with -C02H.
[0202] In another embodiment, compounds of any one of Formulae I or
III -XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R26c is selected from the group consisting of:
Figure imgf000073_0001
[0203] In another embodiment, compounds of any one of Formulae I or
III -XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R26c is aralkyl.
[0204] In another embodiment, compounds of any one of Formulae I or
III -XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof are provided, wherein R26c is selected from the group consisting of:
Figure imgf000074_0001
[0205] In another embodiment, compounds of any one of Formulae I or
III-XXVIII are provided, wherein R26c is selected from the group consisting of:
Figure imgf000074_0002
[0206] In another embodiment, compounds of Formulae XXVII or XXVIII are provided substantially free of one or more other stereoisomers. In another embodiment, compounds of any one of Formulae XXVII or XXVIII are substantially pure stereoisomers. In another embodiment, compounds of any one of Formulae XXVII or XXVIII are pure stereoisomers.
[0207] In another embodiment, compounds of Formula I are provided having the structure: 74
Figure imgf000075_0001
Figure imgf000076_0001
or pharmaceutically acceptable salts, solvates, or prodrugs thereof.
In another embodiment, compounds of Formula I are provided having structure:
Figure imgf000077_0001
Figure imgf000077_0002
or pharmaceutically acceptable salts, solvates, or prodrugs thereof.
In another embodiment, compounds of Formula I are provided having the structure:
Figure imgf000077_0003
77
Figure imgf000078_0001

Figure imgf000079_0001

Figure imgf000080_0001
Figure imgf000081_0001
or pharmaceutically acceptable salts, solvates, or prodrugs thereof.
[0211] In another embodiment, compounds of Formula I are provided having structure:
Figure imgf000081_0002
or pharmaceutically acceptable salts, solvates, or prodrugs thereof.
[0212] In another embodiment, the disclosure provides a method of preparing a compound having Formula
Figure imgf000081_0003
wherein Rla, Rlb, Rlc, Rld, R2, R3a, R3b, and E have the meanings as described above for Formula I, and X and Y are NH. In another embodiment, E is -OR 26a
In another embodiment, E is -NR R c. In another embodiment, R > 26 ~b~ is hydrogen, and R26c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl. In another embodiment, R3a and R3b are taken together form an unsubstituted 4- to 8-membered cycloalkyl.
[0213] In another embodiment, the method of preparing a compound having
Formula XVI comprises all Formula VI:
Figure imgf000082_0001
to isomerize to a compound having Formula XVI.
[0214] In another embodiment, the method of preparing a compound having
Formula XVI comprises dissolving a compound having Formula VI in a solvent or a mixture of solvents.
[0215] In another embodiment, the method of preparing a compound having
Formula XVI comprises:
[0216] a) dissolving a compound having Formula VI in a solvent or a mixture of solvents; and
[0217] b) allowing the compound having Formula VI to isomerize to a compound having Formula XVI.
[0218] In another embodiment, the method of preparing a compound having
Formula XVI comprises:
a) allowing the compound having Formula VI to isomerize to a compound having Formula XVI; and
b) isolating the compound having Formula XVI substantially free from the compound having Formula VI, and one or more other stereoisomers.
In another embodiment, the method of preparing a compound having Formula XVI comprises:
a) dissolving a compound having Formula VI in a solvent or a mixture of solvents;
b) allowing the compound having Formula VI to isomerize to a compound having Formula XVI; and
c) isolating the compound having Formula XVI substantially free from the compound having Formula VI, and one or more other stereoisomers. [0219] In another embodiment, the solvent is selected from the group consisting of acetonitrile, methanol, ethyl acetate, and water, or a mixture thereof.
[0220] In another embodiment, the isomerization is carried out at a pH of less than 7, e.g., at a pH of about 6, about 5, about 4, about 3, about 2, or about 1. In one embodiment, the isomerization is carried out at a pH of about 7. In one embodiment, the isomerization is carried out at a pH of greater than 7, e.g., at a pH of about 8, about 9, about 10, about 1 1, about 12, or about 13.
[0221] In another embodiment, the isomerization is carried out in the presence of an acid, e.g., trifluoroacetic acid or acetic acid.
[0222] In one embodiment, the isomerization is carried out in the presence of a base, e.g., NaHC03.
[0223] In another embodiment, isomerization is carried out at a temperature of about 20°C to about 100°C, e.g., at a temperature of about 20°C to about 70°C, e.g., at a temperature of about 45°C to about 65°C. In one embodiment the isomerization is carried out at about room temperature, e.g., at about 20°C. In one embodiment the isomerization is carried out above room temperature, e.g., at about 25°C, at about 30°C, about 35°C, about 40°C, about 45°C, about 50°C, about 55°C, about 60°C, about 65°C, about 70°C, about 75°C, about 80°C, about 85°C, about 90°C, about 95°C, or about 100°C.
[0224] In another embodiment, the isomerization is carried about for a period of time between about 0.5 hours and about 2 weeks, e.g., for about 1 hour, about 3 hours, about 6 hours, about 12 hours, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 1 week. The period of time needed for isomerization to occur may depend on a variety of factors including the chemical structure of Formula VI, the solvent(s), the temperature, and/or the pH.
[0225] In certain aspects, the disclosure provides the following particular embodiments:
[0226] I. A compound having Formula II:
Figure imgf000084_0001
wherein:
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
R2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl;
R4 is selected from the group consisting of hydrogen and optionally substituted Ci-C6 alkyl;
R5 is selected from the group consisting of:
Figure imgf000084_0002
R5-1 R5-2 R5-3 R5-4 wherein:
each R6a and R6b is independently selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
R7 is selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl;
R8a and R8b are each independently selected from the group consisting of hydrogen, optionally substituted Ci-Ce alkyl, and optionally substituted cycloalkyl; or
R8a and R8b taken together with the carbon that they are attached form a 3- to 8-membered optionally substituted cycloalkyl;
W1 is selected from the group consisting of -OR9a and -NR9bR9c;
R9a is hydrogen;
R9b is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S02R9d, and -CONR9eR9f;
R9c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R9b and R9c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
R9d is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
R9e and R9f are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
R9e and R9f taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
W2 is selected from the group consisting of -OR10 and -NRl laRl lb;
with the proviso that when W1 is -OR9a and W2 is -OR10 then at least one of R7, R8a, and R8b is other than hydrogen;
R10 is hydrogen; or
one of R9a and R10 is hydrogen and the other is a metabolically cleavable group; Rl la is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -S02Rl lc, and -CONRlldRl le;
Rl lb is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
Rl la and Rl lb taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
Rl lc is selected from the group consisting of optionally substituted alkyl and optionally substituted cycloalkyl;
Rl ld and Rl le are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and optionally substituted cycloalkyl; or
Rl ld and Rl le taken together with the nitrogen atom to which they are attached form a 4- to 8-membered optionally substituted heterocyclo;
n is 1, 2, 3, 4, or 5;
each R12a, R12b, R12c and R12d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R13 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R14 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
Z is selected from the group consisting of -OR15 and -NR16aR16b; or
Z and R14 taken together form a carbonyl, i.e., a C=0, group.
R15 is selected from the group consisting of hydrogen and metabolically cleavable group;
R16a is selected from the group consisting of -S02R16c and -CONR16dR16e;
R16b is selected from the group consisting of hydrogen and optionally substituted alkyl;
R16c is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; R and R e are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R16d and R16e taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo;
o is 1, 2, or 3;
p is 0, 1, 2, or 3;
each R17a, R17b, R17c and R17d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R18 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R19 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
R20 is selected from the group consisting of hydrogen, optionally substituted C1-C6 alkyl, and optionally substituted cycloalkyl;
R21a and R21b are each hydrogen; or
one of R21a and R21b is hydrogen and the other is metabolically cleavable group;
q is 0, 1, 2, or 3;
r is 1, 2, or 3;
each R22a, R22b, R22c, and R22d is independently selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R23 is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R24 is selected from the group consisting of -S02R24a and -CONR24bR24c;
R24a is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R24b and R24c are each independently selected from the group consisting of hydrogen, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R24b and R24c taken together with the nitrogen atom to which they are attached form a 4- to 8-membered heterocyclo; s and t are each independently 1, 2, or 3;
X is selected from the group consisting of O, S, and NR;
Y is selected from the group consisting of O, S, and NR ;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
represents a single or a double bond,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0227] II. The compound of articular embodiment I having the formula:
Figure imgf000088_0001
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
III. The compound of particular embodiment II having formula:
Figure imgf000088_0002
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0229] rv. The compound of any one of particular embodiments I-III, wherein:
Rla, Rlb, Rlc, and Rld are each independently selected from the group consisting of hydrogen, fluoro, and chloro;
R2 is optionally substituted aryl;
R3a is halo or d-do alkyl;
R3b is halo or Ci-Cio alkyl; or R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl;
R4 is hydrogen; and
X and Y are NH,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0230] V. The compound of particular embodiment rv, wherein:
Rla and Rld are each hydrogen;
Rlb is selected from the group consisting of hydrogen and fluoro; and Rlc is selected from the group consisting of fluoro and chloro, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0231] VI. The compound of any one of particular embodiments I-V, wherein
R2 is an optionally substituted aryl having Formula R2-1:
Figure imgf000089_0001
wherein R25a, R25 , R25c, R25d, and R25e are each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, amino, cyano, alkoxy, alkyl, or haloalkyl,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0232] VII. The compound of particular embodiment VI, wherein:
R25a is selected from the group consisting of hydrogen and fluoro;
R25b is chloro;
R25c is selected from the group consisting of hydrogen and fluoro; and
R25d and R25e are each hydrogen,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0233] VIII. The compound of any one of particular embodiment I- VII, wherein R5 is selected from the group consisting of:
Figure imgf000090_0001
R5-7
R5-5 R5-6
Figure imgf000090_0002
R5-17
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
IX. The compound of particular embodiment VIII, wherein R5 selected from the group consisting of:
Figure imgf000090_0003
R5-10 R5-12
wherein:
R14 is hydrogen or C1-C4 alkyl; and
R15 is hydrogen,
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0235] X. The compound of any one of particular embodiments I-IX, wherein R3a and R3b are each independently Ci-Ce alkyl, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0236] XL The compound of any one of particular embodiments I-IX, wherein R3a and R3b are taken together to form an optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0237] XII. The compound of any one of particular embodiments I-XI, wherein R is selected from the roup consisting of:
Figure imgf000091_0001
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0238] XIII. A compound selected from the roup consisting of:
Figure imgf000091_0002
or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0239] XrV. A pharmaceutical composition comprising the compound of any one of particular embodiments I- VIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and a pharmaceutically acceptable carrier.
[0240] XV. A method of treating a patient comprising administering to the patient a therapeutically effective amount of the compound of any one of particular embodiments I- VIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein the patient has a hyperproliferative disease.
[0241] XVI. A method of treating a patient comprising administering to the patient a therapeutically effective amount of the pharmaceutical composition of particular embodiment XIV, wherein the patient has a hyperproliferative disease. [0242] XVII. The method of particular embodiments XV or XVI, wherein the hyperproliferative disease is cancer.
[0243] XVIII. The method of particular embodiments XV or XVI, wherein cells of the hyperproliferative disease express functional p53.
[0244] XIX. The method of particular embodiment XVII, further comprising administering to the patient one or more anticancer agents.
[0245] XX. The method of particular embodiment XIX, wherein the anticancer agent is a chemotherapeutic agent.
[0246] XXI. The method of particular embodiment XIX, wherein the anticancer agent is radiation therapy.
[0247] XXII. A method of treating a patient, wherein the patient has a hyperproliferative disorder and is being treated with an anticancer agent, comprising administering to the patient a compound of any one of particular embodiments I-XIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0248] XXIII. The method of particular embodiment XXII, wherein the patient is experiencing side-effects of the anticancer agent treatment selected from the group consisting of mucositis, stomatitis, xerostoma, alopecia, and gastrointestinal disorder.
[0249] XXrV. The method of particular embodiment XXII, wherein cells of the hyperproliferative disorder express functional p53.
[0250] XXV. A kit comprising a compound of any one of particular embodiments I-XIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and instructions for administering the compound to a patient having a hyperproliferative disease.
[0251] XXVI. The kit of particular embodiment XXV, wherein the hyperproliferative disease is cancer.
[0252] XXVII. The kit of particular embodiment XXVI, further comprising one or more anticancer agents.
[0253] XXVIII. The kit of particular embodiment XXVII, wherein the instructions direct co-administration of the compound together with the one or more anticancer agents.
[0254] XXIX. A compound having Formula I:
Figure imgf000093_0001
wherein:
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
R2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3 - to 9-membered optionally substituted heterocyclo;
E is selected from the group consisting of -OR26a and -NR26bR26c;
R26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
R26b is selected from the group consisting of hydrogen and optionally substituted C1-C6 alkyl;
R26c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, and -S02R5b; or R and R c taken together form a 4- to 9-membered optionally substituted heterocyclo;
X is selected from the group consisting of O, S, and NR;
Y is selected from the group consisting of O, S, and NR ;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
represents a single or a double bond,
or a pharmaceutically acceptable salt thereof.
XXX. The compound of particular embodiment XXIX having Formula III:
Figure imgf000094_0001
or a pharmaceutically acceptable salt thereof.
[0256] XXXI. The compound of particular embodiment XXX having
Formula VI:
Figure imgf000094_0002
or a pharmaceutically acceptable salt thereof.
[0257] XXXII. The compound of particular embodiment XXX having
Formula XVI:
Figure imgf000095_0001
or a pharmaceutically acceptable salt thereof.
[0258] XXXIII. The compound of any one of particular embodiments
XXIX-XXXII, wherein:
E is -NR26bR26c;
R2 is optionally substituted aryl;
R3a is halo or Ci-Cio alkyl;
R3b is halo or Ci-Cio alkyl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3- to 9-membered optionally substituted heterocyclo;
R26b is hydrogen; and
X and Y are NH,
or a pharmaceutically acceptable salt thereof.
[0259] XXXrV. The compound of any one of particular embodiments XXIX-XXXIII, wherein R3a and R3b are each independently Ci-Cw alkyl.
[0260] XXXV. The compound of any one of particular embodiments
XXIX-XXXIII, wherein R3a and R3b taken together form a 4- to 8-membered optionally substituted cycloalkyl or a 4- to 8-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt thereof.
[0261] XXXVI. The compound of particular embodiment XXXV, wherein
R3a and R3b taken together form a 6-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt thereof.
[0262] XXXVII. The compound of particular embodiment XXXVI, wherein
R3a and R3b taken together form an optionally substituted piperidine or a tetrahydropyran.
[0263] XXXVIII. The compound of particular embodiment XXXV, wherein
R3a and R3b taken together form a 4- to 8-membered optionally substituted cycloalkyl, or a pharmaceutically acceptable salt thereof. [0264] XXXIX. The compound of particular embodiment XXXVIII, wherein R3a and R3b are taken together to form an unsubstituted cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring, or a pharmaceutically acceptable salt thereof.
[0265] XL. The compound of any one of particular embodiments
XXIX-XXXIX, wherein:
Rla and Rld are each hydrogen;
Rlb is selected from the group consisting of hydrogen and fluoro; and Rlc is selected from the group consisting of fluoro and chloro; or a pharmaceutically acceptable salt thereof.
[0266] XLI. The compound of any one of particular embodiments XXIX -XL, wherein R2 is optionally substituted aryl having Formula R2-1 :
Figure imgf000096_0001
wherein R25a, R25 , R25c, R25d, and R25e are each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, amino, cyano, alkoxy, alkyl, or haloalkyl,
or a pharmaceutically acceptable salt thereof.
[0267] XLII. The compound of any one of particular embodiments XXIX -XL, wherein R2 is optionally substituted pyridyl, or a pharmaceutically acceptable salt thereof.
[0268] XLIII. The compound of particular embodiment XXIX having
Formula XXVI
Figure imgf000096_0002
or a pharmaceutically acceptable salt thereof.
[0269] XLIV. The compound of particular embodiment XLIII having
Formula XXVII:
Figure imgf000097_0001
XXVII
or a pharmaceutically acceptable salt thereof.
[0270] XLV. The compound of particular embodiment XLIII having
Formula XXVIII:
Figure imgf000097_0002
XXVIII,
or a pharmaceutically acceptable salt thereof.
[0271] XLVI. The compound of any one of particular embodiments XXIX-XLV, wherein R26c is optionally substituted alkyl, or a pharmaceutically acceptable salt thereof.
[0272] XLVII.The compound of particular embodiment XLVI, wherein the optionally substituted alkyl is substituted with an optionally substituted cycloalkyl, or a pharmaceutically acceptable salt thereof.
[0273] XL VIII. The compound of particular embodiment XLVI, wherein the optionally substituted alkyl is substituted with an optionally substituted heteroaryl, or a pharmaceutically acceptable salt thereof.
[0274] XLIX. The compound of any one of claims XXIX-XLV, wherein R26c is aralkyl, or a pharmaceutically acceptable salt thereof.
[0275] L. The compound of any one of particular embodiments XXIX-XLV, wherein R26c is optionally substituted cycloalkyl, or a pharmaceutically acceptable salt thereof.
[0276] LI. The compound particular embodiment L, wherein R26c is hydroxycycloalkyl, or a pharmaceutically acceptable salt thereof. [0277] LII. The compound of particular embodiment L, wherein the optionally substituted cycloalkyl is substituted with at least one -CO2H, or a pharmaceutically acceptable salt thereof.
[0278] LIII. The compound of particular embodiment L, wherein R26c is selected from the roup consisting of:
or a pharmaceutically acceptable salt thereof.
[0279] LIV. The compound of any one of particular embodiment XXIX-XLV, wherein R26c is optionally substituted heterocyclo, or a pharmaceutically acceptable salt thereof.
[0280] LV. The compound of any one of particular embodiment XXIX-XLV, wherein R26c is optionally substituted aryl, or a pharmaceutically acceptable salt thereof.
[0281] LVI. The compound of particular embodiment LV, wherein R26c is optionally substituted phenyl, or a pharmaceutically acceptable salt thereof.
[0282] LVII. The compound of particular embodiment LVI, wherein the optionally substituted phenyl is substituted with at least one -CO2H, or a pharmaceutically acceptable salt thereof. [0283] LVIII. The compound of any one of particular embodiments XXIX-XLV, wherein R26c is optionally substituted heteroaryl, or a pharmaceutically acceptable salt thereof.
[0284] LIX. The compound of particular embodiment LVIII, wherein R26c is optionally substituted pyridyl, or a pharmaceutically acceptable salt thereof.
[0285] LX. A compound selected from the group consisting of:
Figure imgf000099_0001
99
Figure imgf000100_0001
100
Figure imgf000101_0001
Figure imgf000102_0001
or a pharmaceutically acceptable salt thereof.
[0286] LXI. A compound selected from the group consisting of:
Figure imgf000102_0002
or a pharmaceutically acceptable salt thereof.
[0287] LXII. The compound of particular embodiment XXIX, wherein E
-OR26a, or a pharmaceutically acceptable salt thereof.
[0288] LXIII. The compound of particular embodiment LXII, wherein R26a hydrogen, or a pharmaceutically acceptable salt thereof. [0289] LXIV. A pharmaceutical composition comprising the compound of any one of particular embodiments XXIX-LXI, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
[0290] LXV. A method of treating a patient comprising administering to the patient a therapeutically effective amount of the compound of any one of particular embodiments XXIX-LXI, or a pharmaceutically acceptable salt thereof, wherein the patient has a hyperproliferative disease.
[0291] LXVI. The method of particular embodiment LXV, wherein the hyperproliferative disease is cancer.
[0292] LXVII.The method of particular embodiment LXVI, wherein the cancer is selected from the group consisting of melanoma, lung cancer, sarcoma, colon cancer, prostate cancer, choriocarcinoma, breast cancer, retinoblastoma, stomach carcinoma, acute myeloid leukemia, lymphoma, multiple myeloma, and leukemia.
[0293] LXVIII. The method of particular embodiment LXVII, wherein the cancer is selected from the group consisting of liposarcoma and melanoma.
[0294] LXIX. A kit comprising a compound of any one of particular embodiments XXIX-LXI, or a pharmaceutically acceptable salt, thereof, and instructions for administering the compound to a patient having a hyperproliferative disease.
[0295] LXX. The kit of particular embodiment LXIX, wherein the hyperproliferative disease is cancer.
[0296] LXXI. A method o d having Formula XVI:
Figure imgf000103_0001
XVI,
comprising:
a) allowing a compound of particular embodiment XXXI to isomerize; and
b) isolating the compound having Formula XVI substantially free from the compound of claim 3, wherein X and Y are NH.
> 26a
[0297] LXXII.The method of particular embodiment LXXI, wherein E is -OR'
[0298] LXXIII. The method of particular embodiment LXXI, wherein E is
-NR26bR26c.
[0299] LXXIV. The method of any one of particular embodiments
LXXI-LXXIII, wherein R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl.
[0300] The compounds provided herein will be better understood in connection with the following synthetic schemes which illustrate the methods by which the compounds provided herein may be prepared. Starting materials can be obtained from commercial sources or prepared by well-established literature methods known to those of ordinary skill in the art. It will be readily apparent to one of ordinary skill in the art that the compounds defined above can be synthesized by substitution of the appropriate reagents and agents in the syntheses shown below.
[0301] Compounds of Formula III wherein Y is NH and E is -NR26cR26c can be synthesized as described in Schemes 2 and 3.
Scheme 2
Figure imgf000104_0001
Figure imgf000104_0002
Formula III (wherein Y is NH and E is NR26bR26c) Scheme 3
Figure imgf000105_0001
Formula III
(wherein Y is NH and
E is NR26bR26c)
Compounds of Formula III can be separated by chiral resolution methods well known in the art, e.g., chiral column chromatography, to give compounds of Formulae VI-XXI. Suitable chiral columns for use in chiral resolutions include, for example, Daicel CHIRALCEL® OD-H, Daicel CHIRAKPAK® AD-H and Regis Technologies ULMO chiral columns. Other chiral resolution methods are also possible. Compounds of Formulae VI-XXI can also be prepared by asymmetric synthetic methods. For example, compounds of Formula VI, wherein Y is NH and E is NR26bR26c, can be synthesized by using a asymmetric 1,3-dipolar cycloaddition as the key step as previously described (See U.S. Patent Nos. 7,759,383 B2 and 7,737, 174 B2, and Ding et al, J. Am. Chem. Soc. 727: 10130-10131 (2005)). Compounds of Formula VI can undergo isomerization in solution to give compounds of Formula XVI. Scheme 4
Figure imgf000106_0001
Formula XVI
(wherein Y is NH)
Without intending to be bound by theory, the isomerization of Formula
VI to Formula XVI may involve formation of the imine intermediate shown in Scheme 5. Compounds of Formula XVI may be less likely to isomerize, i.e., they may be chemically more stable, than compounds of Formula VI. In addition, isolation and purification of compounds of Formula XVI may be improved when R3a and R3a are the same, e.g., R3a and R3b are methyl, R3a and R3b taken together form a cyclohexyl ring, because the 2' position of the pyrrolidine ring is not an asymmetric center, and the number of possible isomerization products of Formula VI is reduced. General methods to prepare compounds provided in the present disclosure are shown in Schemes 4-8.
Figure imgf000107_0001
Scheme 6
Figure imgf000107_0002
Formula XVI Formula XVI (Eis-OH) (E is-NR26 R26c) Scheme 7
Figure imgf000108_0001
Formula XXVII Formula XXVIII
Scheme 8
Figure imgf000108_0002
Methods
The present disclosure contemplates that exposure of animals suffering from cancer to therapeutically effective amounts of drug(s) (e.g., small molecules) that increase the function(s) of p53 and p53-related proteins (e.g.. p63, p73) inhibits the growth of cancer cells or supporting cells. The compounds provided herein inhibit the interaction between p53 or p53 -related proteins and MDM2 or MDM2 -related proteins (e.g., MDMX). Inhibiting the interaction between p53 or p53 -related proteins and MDM2 or MDM2 -related proteins inhibits the growth of cancer cells or supporting cells and/or renders such cells as a population more susceptible to the cell death-inducing activity of cancer therapeutic drugs or radiation therapies. In one embodiment, the inhibitors provided herein prolong the half-life of p53 by interfering with the p53-MDM2 interaction that would normally promote degradation of p53. The compounds provided herein satisfy an unmet need for the treatment of multiple cancer types, either when administered as monotherapy to induce senescence, cell growth inhibition, apoptosis and/or cell cycle arrest in cancer cells, or when administered in a temporal relationship with additional agent(s), such as other cell death-inducing or cell cycle disrupting cancer therapeutic drugs or radiation therapies (combination therapies), so as to render a greater proportion of the cancer cells or supportive cells susceptible to executing the apoptosis program compared to the corresponding proportion of cells in an animal treated only with the cancer therapeutic drug or radiation therapy alone.
In one embodiment, treatment of patients with a therapeutically effective amount of one or more compounds having Formulae I-XXVIII and one or more anticancer agents produces a greater anti-tumor activity and clinical benefit in such patients compared to those treated with the compound or anticancer drugs/radiation alone. Put another way, because the compounds provided herein can lower the apoptotic threshold of cells that express p53 or p53-related protein, the proportion of cells that successfully execute the apoptosis program in response to the apoptosis inducing activity of anticancer drugs/radiation will be increased when used in combination with one or more of the compounds provided herein. Alternatively, compounds having Formulae I-XXVIII can be used to allow administration of a lower, and therefore less toxic and more tolerable, dose of an anticancer drug and/or radiation to produce the same tumor response/clinical benefit as the conventional dose of the anticancer drug/radiation alone. Since the doses for approved anticancer drugs and radiation treatments are known, the compounds, compositions, and methods provided herein can be used with one or more approved anticancer drugs and/or radiation treatment. Also, since compounds having Formulae I-XXVIII may act, at least in part, by stimulating the pro-apoptotic and/or cell cycle-inhibiting activities of p53 and p53 -related proteins, the exposure of cancer cells and supporting cells to therapeutically effective amounts of these compounds can be temporally linked to coincide with the attempts of cells to execute the apoptosis program in response to the anticancer drug or radiation therapy. Thus, in one embodiment, administering the compounds or pharmaceutical compositions provided herein in combination with other known anticancer drugs provides especially efficacious therapeutic practices.
In one embodiment, the inhibitors of the interaction between p53 or p53- related proteins and MDM2 and MDM2-related proteins having Formulae I-XXVIII may protect normal (e.g., non-hyperproliferative) cells from the toxic effects of certain chemotherapeutic agents and radiation, possibly through the ability of the inhibitors to induce cell cycle arrest of normal cells. For example, the inhibitors provided herein may cause cell cycle arrest in cells comprising wild-type or functional p53 (and/or wild-type or functional p53- related proteins) while having no or less effect on cancer cells comprising mutated, deleted, or otherwise non- or less functional p53 (and/or mutated, deleted, or otherwise non-or less functional p53-related proteins). This differential protective effect may allow for more effective treatment of cancer by allowing the use of higher doses or longer treatments of chemotherapeutic agents or treatments without increasing the toxic side effects of such treatment when administered in combination with inhibitors provided herein.
Also provided herein are methods of using compounds having Formulae I-XXVIII for sensitizing cells to additional agent(s), such as inducers of senescence, apoptosis and/or cell cycle arrest. Compounds having Formulae I-XXVIII can also be used to provide chemoprotection of normal cells through the induction of cell cycle arrest prior to treatment with chemotherapeutic agents. In one embodiment, methods of rendering a normal cell resistant to chemotherapeutic agents or treatments comprises contacting the cell with one or more compounds having Formulae I-XXVIII are provided. In another embodiment, methods of protecting normal cells in an animal having a hyperproliferative disease from the toxic side effects of chemotherapeutic agents or treatments, comprises administering to the animal a compound having Formulae I-XXVIII are provided. Also provided herein are methods for the treatment, amelioration, or prevention of disorders, side effects, or conditions caused by the administration of chemotherapeutic agents to normal cells comprising administering to an animal undergoing chemotherapy a compound having Formulae I-XXVIII. Examples of such disorders and conditions caused by chemotherapy include, without limitation, mucositis, stomatitis, xerostomia, gastrointestinal disorders, and alopecia.
[0308] Compounds having Formulae I-XXVIII are useful for the treatment, amelioration, or prevention of disorders, such as those responsive to induction of apoptotic cell death, e.g., disorders characterized by dysregulation of apoptosis, including hyperproliferative diseases such as cancer. In one embodiment, these compounds can be used to treat, ameliorate, or prevent cancer that is characterized by resistance to cancer therapies (e.g., those cancer cells which are chemoresistant, radiation resistant, hormone resistant, and the like). In another embodiment, these compounds can be used to treat hyperproliferative diseases characterized by expression of functional p53 or p53 -related proteins. In another embodiment, these compounds can be used to protect normal (e.g., non-hyperproliferative) cells from the toxic side effects of chemotherapeutic agents and treatments by the induction of cell cycle arrest in those cells.
[0309] In one embodiment, compounds having Formulae I-XXVIII induce cell cycle arrest and/or apoptosis and also potentiate the induction of cell cycle arrest and/or apoptosis either alone or in response to additional apoptosis induction signals. Therefore, it is contemplated that these compounds sensitize cells to induction of cell cycle arrest and/or apoptosis, including cells that are resistant to such inducing stimuli. By inhibiting the interaction between p53 or p53 -related proteins and MDM2 or MDM2-realted proteins, these compounds can be used to induce apoptosis in any disorder that can be treated, ameliorated, or prevented by the induction of apoptosis. In one embodiment, compounds having Formulae I-XXVIII can be used to induce apoptosis in cells comprising functional p53 or p53-related proteins. [0310] In another embodiment, the disclosure pertains to modulating apoptosis with compounds having Formulae I-XXVIII in combination with one or more additional apoptosis-modulating agents, e.g., anticancer agents. Examples of apoptosis-modulating agents include, but are not limited to, Fas/CD95, TRAMP, TNF RI, DR1, DR2, DR3, DR4, DR5, DR6, FADD, RIP, TNFa, Fas ligand, TRAIL, antibodies to TRAIL-R1 or TRAIL-R2, Bcl-2, p53, BAX, BAD, Akt, CAD, PI3 kinase, PP1, and caspase proteins. Other agents involved in the initiation, decision and degradation phase of apoptosis are also included. Examples of apoptosis-modulating agents include agents, the activity, presence, or change in concentration of which, can modulate apoptosis in a subject. Apoptosis-modulating agents include those which are inducers of apoptosis, such as TNF or a TNF-related ligand, particularly a TRAMP ligand, a Fas/CD95 ligand, a TNFR-1 ligand, or TRAIL.
[0311] In one embodiment, the compounds, compositions, and methods provided herein are used to treat diseased cells, tissues, organs, or pathological conditions and/or disease states in an animal (e.g., a mammalian patient including, but not limited to, humans and veterinary animals). In this regard, various diseases and pathologies are amenable to treatment or prophylaxis using the present methods and compositions. A non-limiting exemplary list of these diseases and conditions includes, but is not limited to, breast cancer, prostate cancer, lymphoma, skin cancer, pancreatic cancer, colon cancer, melanoma, malignant melanoma, ovarian cancer, brain cancer, primary brain carcinoma, head-neck cancer, glioma, glioblastoma, liver cancer, bladder cancer, non-small cell lung cancer, head or neck carcinoma, breast carcinoma, ovarian carcinoma, lung carcinoma, small-cell lung carcinoma, Wilms' tumor, cervical carcinoma, testicular carcinoma, bladder carcinoma, pancreatic carcinoma, stomach carcinoma, colon carcinoma, prostatic carcinoma, genitourinary carcinoma, thyroid carcinoma, esophageal carcinoma, myeloma, multiple myeloma, adrenal carcinoma, renal cell carcinoma, endometrial carcinoma, adrenal cortex carcinoma, malignant pancreatic insulinoma, malignant carcinoid carcinoma, choriocarcinoma, mycosis fungoides, malignant hypercalcemia, cervical hyperplasia, leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia (CLL) including B-CLL, acute myelogenous leukemia, chronic myelogenous leukemia, chronic granulocytic leukemia, acute granulocytic leukemia, hairy cell leukemia, neuroblastoma, sarcoma such as liposarcoma malignant fibrous histiocytoma, osteosarcoma, Ewing's sarcoma, leiomyosarcoma, and rhabdomyosarcoma, Kaposi's sarcoma, polycythemia vera, essential thrombocytosis, Hodgkin's disease, non-Hodgkin's lymphoma, soft-tissue sarcomas such as lipoma, and malignant Schwannoma, osteogenic sarcoma, primary macroglobulinemia, and retinoblastoma, and the like, T and B cell mediated autoimmune diseases; inflammatory diseases; infections; hyperproliferative diseases; AIDS; degenerative conditions, vascular diseases, and the like. In one embodiment, the cancer cells being treated are metastatic. In another embodiment, the cancer cells being treated are resistant to other anticancer agents.
In one embodiment, the compounds, compositions, and methods provided herein are used to treat cancers that express functional or wild type p53 or p53-related proteins. In one embodiment, the compounds, compositions, and methods provided herein are used to treat cancers that express elevated levels of MDM2 or MDM2-related proteins.
In one embodiment, the compounds, compositions, and methods provided herein can be used to treat a patient having a sarcoma, including, for example, liposarcoma, malignant fibrous histiocytoma, osteosarcoma, and rhabdomyosarcoma. In another embodiment, the compounds, compositions, and methods provided herein can be used to treat a patient having a soft tissue tumor, including, for example, Ewing's sarcoma, leiomyosarcoma, lipoma, and malignant Schwannomas. In another embodiment, the compounds, compositions, and methods provided herein can be used to treat a patient having lung, breast, liver, or colon cancer. In another embodiment, the compounds, compositions, and methods provided herein can be used to treat a patient having B-cell chronic lymphocytic leukemia and acute myeloid leukemia.
In one embodiment, the compounds, compositions, and methods provided here can be used to treat a patient having melanoma, lung cancer, sarcoma, colon cancer, prostate cancer, choriocarcinoma, breast cancer, retinoblastoma, stomach carcinoma, acute myeloid leukemia, lymphoma, multiple myeloma, or leukemia.
In one embodiment, the compounds, compositions, and methods provided here can be used to treat a patient having liposarcoma or melanoma. [0316] In one embodiment, infections suitable for treatment with the compounds, compositions, and methods provided herein include, but are not limited to, infections caused by viruses, bacteria, fungi, mycoplasma, prions, and the like.
[0317] A further aspect of the present disclosure is to provide the use of a compound having any one of Formulae I-XXVIII for the manufacture of a medicament for treating a hyperproliferative disease such as cancer. In one embodiment, the medicament is to be administered with one or more additional agents.
[0318] A further aspect of the present disclosure is to provide a compound having any one of Formulae I-XXVIII, or a pharmaceutical composition comprising a compound having any one of Formulae I-XXVIII, for use in treating a hyperproliferative disease such as cancer.
[0319] In one embodiment, methods are provided for administering an effective amount of a compound having Formulae I-XXVIII in combination with at least one additional therapeutic agent (including, but not limited to, chemotherapeutic antineoplastics, apoptosis-modulating agents, antimicrobials, antivirals, antifungals, and anti-inflammatory agents) and/or therapeutic technique (e.g., surgical intervention, and/or radiotherapies). In one embodiment, the additional therapeutic agent(s) is an anticancer agent.
[0320] A number of suitable therapeutic or anticancer agents are contemplated for use in the methods provided herein. Indeed, the methods provided herein can include but are not limited to, administration of numerous therapeutic agents such as: agents that induce apoptosis; polynucleotides (e.g., anti-sense, ribozymes, siRNA); polypeptides (e.g., enzymes and antibodies); biological mimetics (e.g., gossypol or BH3 mimetics); agents that bind (e.g., oligomerize or complex) with a Bcl-2 family protein such as Bax; alkaloids; alkylating agents; antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal or polyclonal antibodies (e.g., antibodies conjugated with anticancer drugs, toxins, defensins), toxins; radionuclides; biological response modifiers (e.g., interferons (e.g., IFN-a) and interleukins (e.g., IL-2)); adoptive immunotherapy agents; hematopoietic growth factors; agents that induce tumor cell differentiation (e.g., all-trans-retinoic acid); gene therapy reagents (e.g., antisense therapy reagents and nucleotides); tumor vaccines; angiogenesis inhibitors; proteosome inhibitors: NF-KB modulators; anti-CDK compounds; HDAC inhibitors; and the like. Numerous other examples of therapeutic agents such as chemotherapeutic compounds and anticancer therapies suitable for co-administration with the disclosed compounds are known to those skilled in the art.
In one embodiment, anticancer agents comprise agents that induce or stimulate apoptosis. Agents that induce or stimulate apoptosis include, for example, agents that interact with or modify DNA, such as by intercalating, cross-linking, alkylating, or otherwise damaging or chemically modifying DNA. Agents that induce apoptosis include, but are not limited to, radiation (e.g., X- rays, gamma rays, UV); tumor necrosis factor (TNF)-related factors (e.g., TNF family receptor proteins, TNF family ligands, TRAIL, antibodies to TRAIL-Rl or TRAIL-R2); kinase inhibitors (e.g., epidermal growth factor receptor (EGFR) kinase inhibitor. Additional anticancer agents include: vascular growth factor receptor (VGFR) kinase inhibitor, fibroblast growth factor receptor (FGFR) kinase inhibitor, platelet-derived growth factor receptor (PDGFR) kinase inhibitor, and Bcr-Abl kinase inhibitors (such as GLEEVEC)); antisense molecules; antibodies (e.g., HERCEPTIN, RITUXAN, ZEVALIN, and AVASTIN); anti-estrogens (e.g., raloxifene and tamoxifen); anti-androgens (e.g., flutamide, bicalutamide, finasteride, aminoglutethamide, ketoconazole, and corticosteroids); cyclooxygenase 2 (COX-2) inhibitors (e.g., celecoxib, meloxicam, NS-398, and non-steroidal anti-inflammatory drugs (NSAIDs)); antiinflammatory drugs (e.g., butazolidin, DECADRON, DELTASONE, dexamethasone, dexamethasone intensol, DEXONE, HEXADROL, hydroxychloroquine, METICORTEN, ORADEXON, ORASONE, oxyphenbutazone, PEDIAPRED, phenylbutazone, PLAQUENIL, prednisolone, prednisone, PRELONE, and TANDEARIL); and cancer chemotherapeutic drugs (e.g., irinotecan (CAMPTOSAR), CPT-1 1, fludarabine (FLUDARA), dacarbazine (DTIC), dexamethasone, mitoxantrone, MYLOTARG, VP- 16, cisplatin, carboplatin, oxaliplatin, 5-FU, doxorubicin, gemcitabine, bortezomib, gefitinib, bevacizumab, TAXOTERE or TAXOL); cellular signaling molecules; ceramides and cytokines; staurosporine, and the like.
In one embodiment, the compositions and methods provided herein include one or more compounds provided herein and at least one anti-hyperproliferative or anticancer agent, e.g., alkylating agents, antimetabolites, and natural products (e.g., herbs and other plant and/or animal derived compounds).
[0323] Alkylating agents suitable for use in the present compositions and methods include, but are not limited to: 1) nitrogen mustards (e.g., mechlorethamine, cyclophosphamide, ifosfamide, melphalan (L-sarcolysin); and chlorambucil); 2) ethylenimines and methylmelamines (e.g., hexamethylmelamine and thiotepa); 3) alkyl sulfonates (e.g., busulfan); 4) nitrosoureas (e.g., carmustine (BCNU); lomustine (CCNU); semustine (methyl- CCNU); and streptozocin (streptozotocin)); and 5) triazenes (e.g., dacarbazine (DTIC; dimethyltriazenoimid-azolecarboxamide).
[0324] In one embodiment, antimetabolites suitable for use in the present compositions and methods include, but are not limited to: 1) folic acid analogs (e.g., methotrexate (amethopterin)); 2) pyrimidine analogs (e.g., fluorouracil (5- fluorouracil; 5-FU), floxuridine (fluorode-oxyuridine; FudR), and cytarabine (cytosine arabinoside)); and 3) purine analogs (e.g., mercaptopurine (6- mercaptopurine; 6-MP), thioguanine (6-thioguanine; TG), and pentostatin (2'- deoxycoformycin)).
[0325] In one embodiment, chemotherapeutic agents suitable for use in the present compositions and methods include, but are not limited to: 1) vinca alkaloids (e.g., vinblastine (VLB), vincristine); 2) epipodophyllotoxins (e.g., etoposide and teniposide); 3) antibiotics (e.g., dactinomycin (actinomycin D), daunorubicin (daunomycin; rubidomycin), doxorubicin, bleomycin, plicamycin (mithramycin), and mitomycin (mitomycin C)); 4) enzymes (e.g., L- asparaginase); 5) biological response modifiers (e.g., interferon-alfa); 6) platinum coordinating complexes (e.g., cisplatin (cis-DDP) and carboplatin); 7) anthracenediones (e.g., mitoxantrone); 8) substituted ureas (e.g., hydroxyurea); 9) methylhydrazine derivatives (e.g., procarbazine (N-methylhydrazine; MIH)); 10) adrenocortical suppressants (e.g., mitotane (ο,ρ'-DDD) and aminoglutethimide); 1 1) adrenocorticosteroids (e.g., prednisone); 12) progestins (e.g., hydroxyprogesterone caproate, medroxyprogesterone acetate, and megestrol acetate); 13) estrogens (e.g., diethylstilbestrol and ethinyl estradiol); 14) antiestrogens (e.g., tamoxifen); 15) androgens (e.g., testosterone propionate and fluoxymesterone); 16) antiandrogens (e.g. , flutamide): and 17) gonadotropin- releasing hormone analogs (e.g. , leuprolide).
] Any anticancer agent that is routinely used in a cancer therapy context finds use in the compositions and methods of the present disclosure. For example, the U.S. Food and Drug Administration maintains a formulary of oncolytic agents approved for use in the United States. International counterpart agencies to the U.S.F.D.A. maintain similar formularies. Table 1 provides a list of exemplary antineoplastic agents approved for use in the U.S. Those skilled in the art will appreciate that the "product labels" required on all U.S. approved chemotherapeutics describe approved indications, dosing information, toxicity data, and the like, for the exemplary agents.
Table 1
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
monohydrate)
Anticancer agents further include compounds which have been identified to have anticancer activity. Examples include, but are not limited to, 3-AP, 12-0- tetradecanoylphorbol- 13 -acetate, 17AAG, 852A, ABI-007, ABR-217620, ABT- 751, ADI-PEG 20, AE-941, AG-013736, AGROIOO, alanosine, AMG 706, antibody G250, antineoplastons, AP23573, apaziquone, APC8015, atiprimod, ATN-161, atrasenten, azacitidine, BB-10901, BCX-1777, bevacizumab, BGOOOOl, bicalutamide, BMS 247550, bortezomib, bryostatin-1, buserelin, calcitriol, CCI-779, CDB-2914, cefixime, cetuximab, CG0070, cilengitide, clofarabine, combretastatin A4 phosphate, CP-675,206, CP-724,714, CpG 7909, curcumin, decitabine, DENSPM, doxercalciferol, E7070, E7389, ecteinascidin 743, efaproxiral, eflornithine, EKB-569, enzastaurin, erlotinib, exisulind, fenretinide, flavopiridol, fludarabine, flutamide, fotemustine, FR901228, G17DT, galiximab, gefitinib, genistein, glufosfamide, GTI-2040, histrelin, HKI-272, homoharringtonine, HSPPC-96, hul4.18-interleukin-2 fusion protein, HuMax- CD4, iloprost, imiquimod, infliximab, interleukin-12, IP 1-504, irofulven, ixabepilone, lapatinib, lenalidomide, lestaurtinib, leuprolide, LMB-9 immunotoxin, lonafarnib, luniliximab, mafosfamide, MB07133, MDX-010, MLN2704, monoclonal antibody 3F8, monoclonal antibody J591, motexafin, MS-275, MVA-MUC 1 -IL2, nilutamide, nitrocamptothecin, nolatrexed dihydrochloride, nolvadex, NS-9, 06-benzylguanine, oblimersen sodium, O YX-015, oregovomab, OSI-774, panitumumab, paraplatin, PD-0325901, pemetrexed, PHY906, pioglitazone, pirfenidone, pixantrone, PS-341, PSC 833, PXD101, pyrazoloacridine, Rl 15777, RADOOl, ranpirnase, rebeccamycin analogue, rhuAngiostatin protein, rhuMab 2C4, rosiglitazone, rubitecan, S-l, S- 8184, satraplatin, SB-, 15992, SGN-0010, SGN-40, sorafenib, SR31747A, ST1571, SU011248, suberoylanilide hydroxamic acid, suramin, talabostat, talampanel, tariquidar, temsirolimus, TGFa-PE38 immunotoxin, thalidomide, thymalfasin, tipifarnib, tirapazamine, TLK286, trabectedin, trimetrexate glucuronate, TroVax, UCN-1, valproic acid, vinflunine, VNP40101M, volociximab, vorinostat, VX-680, ZD 1839, ZD6474, zileuton, and zosuquidar trihydrochloride.
For a more detailed description of anticancer agents and other therapeutic agents, those skilled in the art are referred to any number of instructive manuals including, but not limited to, the Physician's Desk Reference and to Goodman and Gilman's "Pharmaceutical Basis of Therapeutics" tenth edition, Eds. Hardman et ah, 2002.
In one embodiment, the methods provided herein comprise administering one or more compounds having Formulae I-XXVIII in combination with radiation therapy. The methods provided herein are not limited by the types, amounts, or delivery and administration systems used to deliver the therapeutic dose of radiation to an animal. For example, the animal may receive photon radiotherapy, particle beam radiation therapy, other types of radiotherapies, and combinations thereof. In one embodiment, the radiation is delivered to the animal using a linear accelerator. In another embodiment, the radiation is delivered using a gamma knife.
The source of radiation can be external or internal to the animal. External radiation therapy is most common and involves directing a beam of high-energy radiation to a tumor site through the skin using, for instance, a linear accelerator. While the beam of radiation is localized to the tumor site, it is nearly impossible to avoid exposure of normal, healthy tissue. However, external radiation is usually well tolerated by animals. Internal radiation therapy involves implanting a radiation-emitting source, such as beads, wires, pellets, capsules, particles, and the like, inside the body at or near the tumor site including the use of delivery systems that specifically target cancer cells (e.g., using particles attached to cancer cell binding ligands). Such implants can be removed following treatment, or left in the body inactive. Types of internal radiation therapy include, but are not limited to, brachytherapy, interstitial irradiation, intracavity irradiation, radioimmunotherapy, and the like. [0331] The animal may optionally receive radiosensitizers (e.g., metronidazole, misonidazole, intra-arterial Budr, intravenous iododeoxyuridine (IudR), nitroimidazole, 5-substituted-4-nitroimidazoles, 2H-isoindolediones, [[(2- bromoethyl)-amino]methyl]-nitro- 1 H-imidazole- 1 -ethanol, nitroaniline derivatives, DNA-affinic hypoxia selective cytotoxins, halogenated DNA ligand, 1,2,4 benzotriazine oxides, 2-nitroimidazole derivatives, fluorine-containing nitroazole derivatives, benzamide, nicotinamide, acridine-intercalator, 5- thiotretrazole derivative, 3-nitro-l,2,4-triazole, 4,5-dinitroimidazole derivative, hydroxylated texaphrins, cisplatin, mitomycin, tiripazamine, nitrosourea, mercaptopurine, methotrexate, fluorouracil, bleomycin, vincristine, carboplatin, epirubicin, doxorubicin, cyclophosphamide, vindesine, etoposide, paclitaxel, heat (hyperthermia), and the like), radioprotectors (e.g., cysteamine, aminoalkyl dihydrogen phosphorothioates, amifostine (WR 2721), IL-1, IL-6, and the like). Radiosensitizers enhance the killing of tumor cells. Radioprotectors protect healthy tissue from the harmful effects of radiation.
[0332] Any type of radiation can be administered to an animal, so long as the dose of radiation is tolerated by the animal without unacceptable negative side- effects. Suitable types of radiotherapy include, for example, ionizing (electromagnetic) radiotherapy (e.g., X-rays or gamma rays) or particle beam radiation therapy (e.g., high linear energy radiation). Ionizing radiation is defined as radiation comprising particles or photons that have sufficient energy to produce ionization, i.e., gain or loss of electrons (as described in, for example, U.S. 5,770,581 incorporated herein by reference in its entirety). The effects of radiation can be at least partially controlled by the clinician. In one embodiment, the dose of radiation is fractionated for maximal target cell exposure and reduced toxicity.
[0333] In one embodiment, the total dose of radiation administered to an animal is about .01 Gray (Gy) to about 100 Gy. In another embodiment, about 10 Gy to about 65 Gy (e.g., about 15 Gy, 20 Gy, 25 Gy, 30 Gy, 35 Gy, 40 Gy, 45 Gy, 50 Gy, 55 Gy, or 60 Gy) are administered over the course of treatment. While in some embodiments a complete dose of radiation can be administered over the course of one day, the total dose is ideally fractionated and administered over several days. Desirably, radiotherapy is administered over the course of at least about 3 days, e.g., at least 5, 7, 10, 14, 17, 21, 25, 28, 32, 35, 38, 42, 46, 52, or 56 days (about 1-8 weeks). Accordingly, a daily dose of radiation will comprise approximately 1-5 Gy (e.g., about 1 Gy, 1.5 Gy, 1.8 Gy, 2 Gy, 2.5 Gy, 2.8 Gy, 3 Gy, 3.2 Gy, 3.5 Gy, 3.8 Gy, 4 Gy, 4.2 Gy, or 4.5 Gy), or 1-2 Gy (e.g., 1.5-2 Gy). The daily dose of radiation should be sufficient to induce destruction of the targeted cells. If stretched over a period, in one embodiment, radiation is not administered every day, thereby allowing the animal to rest and the effects of the therapy to be realized. For example, radiation desirably is administered on 5 consecutive days, and not administered on 2 days, for each week of treatment, thereby allowing 2 days of rest per week. However, radiation can be administered 1 day/week, 2 days/week, 3 days/week, 4 days/week, 5 days/week, 6 days/week, or all 7 days/week, depending on the animal's responsiveness and any potential side effects. Radiation therapy can be initiated at any time in the therapeutic period. In one embodiment, radiation is initiated in week 1 or week 2, and is administered for the remaining duration of the therapeutic period. For example, radiation is administered in weeks 1-6 or in weeks 2-6 of a therapeutic period comprising 6 weeks for treating, for instance, a solid tumor. Alternatively, radiation is administered in weeks 1-5 or weeks 2-5 of a therapeutic period comprising 5 weeks. These exemplary radiotherapy administration schedules are not intended, however, to limit the methods provided herein.
[0334] Antimicrobial therapeutic agents may also be used as therapeutic agents in combination with the compounds having Formulae I-XXVIII. Any agent that can kill, inhibit, or otherwise attenuate the function of microbial organisms may be used, as well as any agent contemplated to have such activities. Antimicrobial agents include, but are not limited to, natural and synthetic antibiotics, antibodies, inhibitory proteins (e.g., defensins), antisense nucleic acids, membrane disruptive agents and the like, used alone or in combination. Indeed, any type of antibiotic may be used including, but not limited to, antibacterial agents, antiviral agents, antifungal agents, and the like.
[0335] In one embodiment of the methods provided herein, one or more compounds having Formulae I-XXVIII are administered to an animal in need thereof. In another embodiment of the methods provided herein, one or more compounds having Formulae I-XXVIII and one or more additional therapeutic agents or anticancer agents are administered to an animal in need thereof under one or more of the following conditions: at different periodicities, at different durations, at different concentrations, by different administration routes, etc. In one embodiment, the compound having Formulae I-XXVIII is administered prior to the therapeutic or anticancer agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks prior to the administration of the therapeutic or anticancer agent. In another embodiment, the compound having Formulae I-XXVIII is administered after the therapeutic or anticancer agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks after the administration of the anticancer agent. In another embodiment, the compound having Formulae I-XXVIII and the therapeutic or anticancer agent are administered concurrently but on different schedules, e.g., the compound is administered daily while the therapeutic or anticancer agent is administered once a week, once every two weeks, once every three weeks, or once every four weeks. In another embodiment, the compound is administered once a week while the therapeutic or anticancer agent is administered daily, once a week, once every two weeks, once every three weeks, or once every four weeks.
In one embodiment, a method of treating, preventing, or ameliorating cancer in a patient is provided, wherein the method comprises pulsatile administration to the patient a therapeutically effective amount of a compound having Formulae I-XXVIII, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
In one embodiment, the pharmaceutical compositions provided herein comprise one or more compounds having Formulae I-XXVIII in an amount which is effective to achieve its intended purpose. While individual needs vary, determination of optimal ranges of effective amounts of each component is within the skill of the art. Typically, the compounds may be administered to mammals, e.g. humans, orally at a dose of 0.0025 to 50 mg/kg, or an equivalent amount of the pharmaceutically acceptable salt thereof, per day of the body weight of the mammal being treated for disorders responsive to induction of apoptosis. In one embodiment, about 0.01 to about 25 mg/kg is orally administered to treat, ameliorate, or prevent such disorders. For intramuscular injection, the dose is generally about one-half of the oral dose. For example, a suitable intramuscular dose would be about 0.0025 to about 25 mg kg, or from about 0.01 to about 5 mg/kg.
[0338] The unit oral dose may comprise from about 0.01 to about 1000 mg, for example, about 0.1 to about 100 mg of the compound. The unit dose may be administered one or more times daily as one or more tablets or capsules each containing from about 0.1 to about 10 mg, conveniently about 0.25 to 50 mg of the compound or its solvates.
[0339] In a topical formulation, the compound may be present at a concentration of about 0.01 to 100 mg per gram of carrier. In a one embodiment, the compound is present at a concentration of about 0.07-1.0 mg/ml, for example, about 0.1-0.5 mg/ml, and in one embodiment, about 0.4 mg/ml.
[0340] In addition to administering the compound as a raw chemical, compounds having Formulae I-XXVIII may be administered as part of a pharmaceutical preparation or composition. In one embodiment, the pharmaceutical composition comprises one or more pharmaceutically acceptable carriers, excipients, and/or auxiliaries. In another embodiment, the one or more carriers, excipients, and auxiliaries facilitate processing of the compound having Formulae I-XXVIII into a preparation which can be used pharmaceutically. The compositions, particularly those compositions which can be administered orally or topically and which can be used for one type of administration, such as tablets, dragees, slow release lozenges and capsules, mouth rinses and mouth washes, gels, liquid suspensions, hair rinses, hair gels, shampoos and also preparations which can be administered rectally, such as suppositories, as well as suitable solutions for administration by intravenous infusion, injection, topically or orally, contain from about 0.01 to 99 percent, in one embodiment from about 0.25 to 75 percent of active compound(s), together with the one or more carriers, excipients, and/or auxiliaries.
[0341] The pharmaceutical compositions provided herein may be administered to any patient which may experience the beneficial effects of compounds having Formulae I-XXVIII. Foremost among such patients are mammals, e.g., humans, although the methods and compositions provided herein are not intended to be so limited. Other patients include veterinary animals (cows, sheep, pigs, horses, dogs, cats and the like). [0342] Compounds having Formulae I-XXVIII and pharmaceutical compositions thereof may be administered by any means that achieve their intended purpose. For example, administration may be by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, intranasal or topical routes. Alternatively, or concurrently, administration may be by the oral route. The dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
[0343] The pharmaceutical compositions and preparations provided herein are manufactured by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes. Thus, pharmaceutical compositions for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.
[0344] Suitable excipients are, in particular, fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone. If desired, disintegrating agents may be added such as the above-mentioned starches and also carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate. Auxiliaries can be suitable flow-regulating agents and lubricants. Suitable auxiliaries include, for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol. Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices. For this purpose, concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. In order to produce coatings resistant to gastric juices, solutions of suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethyl-cellulose phthalate, are used. Dye stuffs or pigments may be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.
[0345] Other pharmaceutical preparations which can be used orally include push- fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. The push-fit capsules can contain the active compounds in the form of granules which may be mixed with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds are in one embodiment dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin. In addition, stabilizers may be added.
[0346] Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of one or more of the active compounds with a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons. In addition, it is also possible to use gelatin rectal capsules which consist of a combination of the active compounds with a base. Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
[0347] Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water- soluble salts and alkaline solutions. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides or polyethylene glycol-400. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers.
[0348] The topical compositions provided herein are formulated in one embodiment as oils, creams, lotions, ointments and the like by choice of appropriate carriers. Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C12). The carriers may be those in which the active ingredient is soluble. Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired. Additionally, transdermal penetration enhancers can be employed in these topical formulations. Examples of such enhancers can be found in U.S. Pat. Nos. 3,989,816 and 4,444,762.
Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil such as almond oil with warm soft paraffin and allowing the mixture to cool. A typical example of such an ointment is one which includes about 30% almond oil and about 70% white soft paraffin by weight. Lotions may be conveniently prepared by dissolving the active ingredient, in a suitable high molecular weight alcohol such as propylene glycol or polyethylene glycol.
The following examples are illustrative, but not limiting, of the compounds, compositions, and methods provided herein. Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered in clinical therapy and which are obvious to those skilled in the art are within the spirit and scope of the methods, compounds, and compositions provided herein.
EXAMPLE 1
Synthesis of Compound Example No. 3
Scheme 7
Figure imgf000130_0001
3-Chloro-2-fluorobenzaldehyde (6.24 g, 39.4 mmol) was added to a solution of piperidine (3.88 mL, 39.4 mmol) and 6-chlorooxindole (6.0 g, 35.8 mmol) dissolved in methanol (100 mL). After stirring overnight, the resulting solid was filtered and washed with methanol and hexanes to give 10.6 g of (E)-6- chloro-3-(3-chloro-2-fluorobenzylidene)indolin-2-one (3) as a green solid. Scheme 8
Figure imgf000131_0001
5a = CH2 6a = CH2
5b = CF2 6b = CF2
5c = O 6c = O
5d = NBoc 6d = NBoc
5e = NMe 6e = NMe
5f = NAc 6f = NAc
[0352] Compound 3 (19.5 mmol), (5R,6S)-5,6-diphenylmorpholin-2-one (4)
(23.4 mmol), and ketone 5 (39 mmol) were dissolved in THF (7.5 mL) and toluene (75 mL) and refluxed for 3 hours. After cooling to room temperature, the reaction was filtered. The solution was concentrated and purified by column chromatography to give the product (30-50% yield) as a solid.
Scheme 9
Figure imgf000131_0002
6a
Figure imgf000131_0003
Compound Example No. 3
[0353] Trans-4-aminocyclohexanol (1.8 g, 15.6 mmol) was added to a solution of intermediate 6a (1.0 g, 1.56 mmol), dissolved in THF (30 mL), and refluxed overnight. The solvent was removed and the crude product was purified by column chromatography to give 0.568 g of intermediate 7 as an off white solid. Intermediate 7 (0.568 g, 0.75 mmol) was dissolved in acetonitrile (5 mL). CAN (823 mg, 1.50 mmol) and water (5 mL) were added. After 15 minutes, the reaction was quenched with saturated sodium bicarbonate, extracted with ethyl acetate, dried over sodium sulfate, and filtered through celite. The solvent was removed and the crude product was purified by column chromatography to give 340 mg of the product 8 as a solid. The solid was dissolved in 1 : 1 methanol/water with 10% TFA and aged (for isomerization) in this solution for 1- 2 days. The solvent was removed. The resulting oil was re-dissolved in 3 : 1 methanol/water, purified by preparative HPLC, and lyophilized to give Compound Example No. 3 (as the TFA salt) as a white powder. lH NMR (300 MHz, CD3OD) δ ppm 8.19 (d, J = 7.5 Hz, 1H), 7.64 (t, J = 6.6 Hz, 1H), 7.48 (dd, J = 2.1, 8.2 Hz, 1H), 7.39 (t, J = 7.6 Hz, 1H), 7.16 (t, J = 8.1 Hz, 1H), 7.10 (dd, J = 1.9, 8.2 Hz, 1H), 6.79 (d, J = 1.8 Hz, 1H), 5.09 (d, J = 11.1 Hz, 1H), 4.78 (d, J = 1 1.1 Hz, 1H), 3.70-3.55 (m, 1H), 3.48-3.35 (m, 1H), 2.84 (d, J = 8.0 Hz, 1H), 2.19 (d, J = 13.4 Hz, 1H), 2.05-1.82 (m, 5H), 1.82-1.65 (m, 3H), 1.64-1.41 (m, 2H), 1.40-1.08 (m, 5H), 1.01-0.83 (m, 1H); ESI-MS m/z 560.9 (M+H)+.
EXAMPLE 2
Synthesis of Compound Example No. 22
Scheme 10
Figure imgf000132_0001
12 [0354] Concentrated sulfuric acid (2 rnL) was added to a solution of intermediate
6a (3.65 g, 5.69 mmol) dissolved in methanol (50 mL), and the resulting solution was heated to 50°C for 5 hours. The methanol was removed by evaporation, and the resulting concentrate was cooled to 0°C and slowly neutralized with a solution of saturated sodium bicarbonate. The aqueous solution was extracted with ethyl acetate and the organic layer was dried over sodium sulfate, filtered, and concentrated. The crude product was purified by column chromatography to give 2.20 g (57% yield) of intermediate 11.
[0355] Intermediate 11 (2.20 g, 3.25 mmol) was dissolved in acetonitrile (25 mL) and THF (5 mL), CAN (3.56 g, 6.50 mmol), and water (25 mL) were added.
After 15 minutes, the reaction was quenched with saturated sodium bicarbonate, extracted with ethyl acetate, dried over sodium sulfate, and filtered through celite. The solvent was removed and the crude product was purified by column chromatography to give 1.43 g (92% yield) of the methyl ester intermediate 12 as a solid.
[0356] The methyl ester intermediate 12 (1.43 g, 3.0 mmol) was dissolved in
THF (20 mL) and LiOH.H20 (377 mg, 9.0 mmol) was added, followed by water (20 mL). After 2 hours, the reaction was quenched with water and saturated ammonium chloride, and the aqueous layer was extracted with ethyl acetate. The ethyl acetate solution was dried over sodium sulfate, filtered, and concentrated to produce the carboxylic acid intermediate 13 as an off white solid. The acid was used without further purification. ESI-MS m/z 463.17 (M+H)+.
Scheme 1 1
Figure imgf000134_0001
13 14
Figure imgf000134_0002
Compound Example No. 22
[0357] CDI (525 mg, 3.24 mmol), DIEA (0.941 mL, 5.4 mmol), and DMAP
(catalytic) were added to a solution of carboxylic acid intermediate 13 (500 mg, 1.08 mmol) dissolved in 1,2-dichloroethane, and the resulting solution was heated to 50°C. After 30 minutes, methyl 4-aminobenzoate 14 (816 mg, 5.4 mmol) was added to the reaction and the reaction was heated to reflux. After heating overnight, the solvent was removed and the crude product was purified by column chromatography to give 265 mg (41% yield) of intermediate 15 as a white solid.
[0358] The resulting methyl ester intermediate 15 (265 mg, 0.44 mmol) was dissolved in THF (10 mL) then LiOH.H20 (56 mg, 1.33 mmol), NaOH (53 mg, 1.33 mmol), and H20 were added. After 2 hours, 3 mL of TFA was added, stirred briefly, and the solvent was evaporated. The resulting oil was re-dissolved in methanol and water, purified by preparative HPLC, and lyophilized to give
235 mg of Compound Example No. 22 (as the TFA salt) as a white solid. ¾ NMR (300 MHz, CD3OD) δ ppm 7.98 ( d, J = 8.5 Hz, 2H), 7.72 (t, J = 7.1 Hz, 1H), 7.65 (d, J = 8.5 Hz, 2H), 7.54 (d, J = 8.2 Hz, 1H), 7.37 (t, J = 7.5 Hz, 1H), 7.18 (t, J = 8.0 Hz, 1H), 7.1 1 (d, J = 8.0 Hz, 1H), 6.69 (s, 1H), 5.32 (d, J = 10.9 Hz, 1H), 4.97 (d, J = 10.9 Hz, 1H), 2.89 (d, J = 9.9 Hz, 1H), 2.19 (d, J = 14.0 Hz, 1H), 2.08-1.85 (m, 3H), 1.78 (d, J = 11.8 Hz, 2H), 1.54 (q, J
1.32-1.10 (m, 2H); ESI-MS m/z 582.17 (M+H)+.
EXAMPLE 3
Synthesis of Compound Example No. 23
Scheme 12
Figure imgf000135_0001
1 J l b Compound Example No. 23 EDCI (19 mg, 0.097 mmol), HOBt (13 mg, 0.097 mmol), and DIEA
(0.034 mL, 0.195 mmol) were added to a solution of carboxylic acid intermediate 13 (30 mg, 0.065 mmol) dissolved in DCM. After 10 minutes, 4-aminotetrahydro-2H-thiopyran- 1,1 -dioxide hydrochloride (24 mg, 0.13 mmol) and a catalytic amount of DMAP were added. After stirring overnight, the solvent was removed and the crude product was purified by column chromatography to give 15 mg of Compound Example No. 23 as a white solid. The solid was further purified by preparative HPLC, and lyophilized to give 10 mg of Compound Example No. 23 (as the TFA salt) as a white powder. XH NMR
(300 MHz, CD3OD) δ ppm 7.64 (t, J = 7.1 Hz, 1H), 7.49 (dd, J = 2.1, 8.1 Hz, 1H), 7.41 (t, J = 7.5 Hz, 1H), 7.18 (t, J = 9.9 Hz, 1H), 7.11 (dd, J = 1.6, 8.2 Hz, 1H), 6.79 (d, J = 1.6 Hz, 1H), 5.10 (d, J = 11.1 Hz, 1H), 4.80 (d, J = 11.2 Hz, 1H), 4.10-3.94 (m, 1H), 3.27-2.91 (m, 3H), 2.88-2.74 (m, 2H), 2.35-1.64 (m, 10H), 1.52 (q, J = 13.9 Hz, 1H), 1.31-1.11 (m, 2H); ESI-MS m/z 594.50 (M+H)+.
EXAMPLE 4
Synthesis of Compound Example 24 Scheme 13
Figure imgf000136_0001
13 17
Figure imgf000136_0002
4
[0360] EDCI (19 mg, 0.097 mmol), HOBt (13 mg, 0.097 mmol), and DIEA
(0.034 mL, 0.195 mmol) were added to a solution of carboxylic acid intermediate 13 (30 mg, 0.065 mmol) dissolved in DCM. After 10 minutes, methyl trans-4- aminocyclohexanecarboxylate hydrochloride (25 mg, 0.13 mmol) and a catalytic amount of DMAP were added. After stirring overnight, the solvent was removed and the crude product was purified by column chromatography to give the 30 mg of intermediate 18 as a white solid.
[0361] Methyl ester intermediate 18 (30 mg, 0.05 mmol) was dissolved in THF
(1 mL) then LiOH.H20 (6.2 mg, 0.15 mmol) and H20 were added. After 2 hours, 0.5 mL of TFA was added, and the solvent was evaporated. The oil was re- dissolved in 3 : 1 methanol/water with 10% TFA, purified by preparative HPLC, and lyophilized to give 20 mg of Compound Example No. 24 (as the TFA salt) as a white powder. XH NMR (300 MHz, CD3OD) δ ppm 7.64 (t, J = 7.1 Hz, 1H),
7.49 (d, J = 8.1 Hz, 1H), 7.39 (t, J = 7.5 Hz, 1H), 7.17 (t, J = 8.1 Hz, 1H), 7.1 1 (d, J = 8.1 Hz, 1H), 6.79 (s, 1H), 5.08 (d, J = 11.1 Hz, 1H), 4.79 (d, J = 11.1 Hz, 1H), 3.72-3.54 (m, 1H), 2.84 (d, J = 8.4 Hz, 1H), 2.25-2.07 (m, 2H), 2.04-1.84 (m, 6H), 1.77 (d, J = 12.0 Hz, 2H), 1.63 (d, J = 13.0 Hz, 1H), 1.56-1.34 (m, 3H), 1.32-1.11 (m, 3H), 0.99-0.82 (m, 1H) ESI-MS m/z 588.33 (M+H)+. EXAMPLE 5
[0362] The following compounds were prepare using methodology described in
Examples 1-4. Unless otherwise indicated, each Compound Example was purified by reverse phase HPLC and isolated as the TFA salt. Unless otherwise indicated, all lH NMR chemical shifts reported herein are denoted by the delta
(δ) scale.
[0363] Compound Exampl
Figure imgf000137_0001
[0364] XH NMR (300 MHz, MeOH-d4): 8.14 (d, J = 7.6 Hz, 1H), 7.62 (t, J = 7.6
Hz, 1H), 7.51 (dd, J = 1.9, 8.1 Hz, 1H), 7.40 (t, J = 7.4 Hz, 1H), 7.17 (dd, J = 1.9, 8.2 Hz, 1H), 6.80 (d, J = 1.8 Hz, 1H), 5.07 (d, J = 11.2 Hz, 1H), 4.70-4.90 (m, 1H), 3.51-3.72 (M, 1H), 3.35-3.48 (m, 1H), 1.93 (s, 3H), 1.85-1.95 (m, 1H), 1.85-1.95 (m, 1H), 1.78 (d, J = 12.3 Hz, 1H), 1.59 (d, J = 12.3 Hz, 1H), 1.41 (s, 3H), 1.10-1.35 (m, 4H), 0.85-1.05 (m, 1H); ESI-MS m/z 520.3 (M+H)+, 542.1 (M+Na)+.
[0365] Compound Exampl
Figure imgf000137_0002
[0366] XH NMR (300 MHz, MeOH-d4): 8.24 (d, J = 7.7 Hz, 1H), 7.45-7.62 (m,
2H), 7.39 (t, J = 6.9 Hz, 1H), 7.16 (t, J = 8.1 Hz, 1H), 7.10 (dd, J = 1.9, 8.2 Hz, 1H), 6.81 (d, J = 1.8 Hz, 1H), 5.07 (d, J = 11.1 Hz, 1H), 4.69 (d, J = 11.1 Hz,
1H), 3.51-3.70 (m, 1H), 3.35-3.48 (m, 1H), 2.65-2.82 (m, 1H), 2.38-2.54 (m, 1H), 2.1 1-2.24 (m, 1H), 1.95-2.11 (m, 1H), 1.84-1.95 (m, 2H), 1.78-1.74 (m, 2H), 1.43-1.78 (m, 4H), 1.10-1.40 (m, 3H), 0.83-1.05 (m, 1H); ESI-MS m/z 546.7 (M+H)+. [0367] Compound Exampl
Figure imgf000138_0001
[0368] XH NMR (300 MHz, MeOH-d4): 8.25 (d, J = 7.5 Hz, IH), 7.50-7.66 (m,
2H), 7.38 (t, J = 7.4 Hz, IH), 7.15 (t, J = 8.0 Hz, IH), 7.08 (d, J = 8.2 Hz, IH), 6.79 (s, IH), 5.10 (d, J = 11.2 Hz, IH), 4.80-4.88 (m, IH), 3.53-3.67 (m, IH),
3.35-3.47 (m, IH), 3.05 (dd, J = 8.3, 15.3 Hz, IH), 2.41 (dd, J = 8.8, 14.4 Hz, 2H), 1.89 (d, J = 10.2 Hz, 2H), 1.65-1.84 (m, 4H), 1.40-1.65 (m, 5H), 1.14-1.38 (m, 4H), 1.00-1.12 (m, IH), 0.82-1.00 (m, IH); ESI-MS m/z 574.6 (M+H)+, 596.1 (M+Na)+.
[0369] Compound Exampl
Figure imgf000138_0002
[0370] XH NMR (300 MHz, MeOH-d4): 8.25 (d, J = 7.3 Hz, IH), 7.28-7.42 (m,
2H), 6.91-7.12 (m, 2H), 6.77 (d, J = 1.8 Hz, IH), 4.80-4.97 (m, IH), 4.40 (d, J = 12.0 Hz, IH), 4.04 (d, J = 12.4 Hz, IH), 3.84 (d, J = 12.6 Hz, IH), 3.51-3.68 (m, IH), 3.36-3.51 (m, IH), 1.91 (d, J = 9.3 Hz, 2H), 1.74 (d, J = 12.7 Hz, IH), 1.49 (d, J = 12.3 Hz, IH), 1.14-1.36 (m, 3H), 0.86-1.05 (m, IH); ESI-MS m/z 492.60 (M+H)+.
[0371] Compound Examp
Figure imgf000138_0003
[0372] ESI-MS m/z 563.67 (M+H) [0373] Compound Examp
Figure imgf000139_0001
[0374] The title compound was prepared as described in EXAMPLE 1 using intermediate 6b and 2-morpholinoethanamine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 7.64 (t, J = 7.1 Hz, IH), 7.49 (dd, J = 2.2, 8.2 Hz, IH), 7.33 (t, J = 8.0 Hz, IH), 7.16-7.07 (m, 2H), 6.77 (d, J = 1.7 Hz, IH), 5.02 (d, J = 10.2 Hz, IH), 4.82 (d, J = 10.3 Hz, IH), 4.12-3.38 (m, 11H), 2.75-1.73 (m, 7H), 1.50-1.31 (m, IH); ESI-MS m/z 61 1.25 (M+H)+.
[0375] Compound Examp
Figure imgf000139_0002
[0376] The title compound was prepared as described in Example 1 using intermediate 6b and 2-morpholinoethanamine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 7.33 (t, J = 7.3 Hz, IH), 7.26 (t, J = 7.5 Hz, IH), 7.05-6.84 (m, 3H), 6.82 (s, IH), 4.68 (d, J = 9.2 Hz, IH), 4.56 (d, J = 9.2 Hz, IH), 4.16-3.01 (m, 11H), 2.48-1.63 (m, 7H), 1.31 (dt, J = 3.8, 14.0 Hz, IH); ESI-MS m/z 61 1.25 (M+H)+.
[0377] Compound Example No. 9:
Figure imgf000139_0003
[0378] The title compound was prepared as described in EXAMPLE 1 using intermediate 6b. XH NMR (300 MHz, CD3OD) δ ppm 8.12 (d, J = 8.1 Hz, IH), 7.62 (t, J = 7.2 Hz, IH), 7.49 (dd, J = 2.3, 8.2 Hz, IH), 7.33 (t, J =8.3 Hz, IH), 7.16-7.05 (m, 2H), 6.78 (d, J = 1.9 Hz, 1H), 4.77 (d, J = 10.3 Hz, 1H), 3.70-3.41 (m, 2H), 2.74-1.64 (m, 1 1H), 1.48-1.21 (m, 4H), 1.18-1.02 (m, 1H); ESI-MS m/z 596.75 (M+H)+.
[0379] Compound Examp
Figure imgf000140_0001
[0380] The title compound was prepared as described in Example 1 using intermediate 6c. XH NMR (300 MHz, CD3OD) δ ppm 8.02 (d, J = 8.0 Hz, 1H), 7.31-7.15 (m, 3H), 7.01-6.91 (m, 2H), 6.82 (d, J = 1.9 Hz, 1H), 4.70 (d, J = 10.4 Hz, 1H), 4.59 (d, J = 10.4 Hz, 1H), 4.12-3.80 (m, 3H), 3.76-3.48 (m, 3H), 2.25- 0.94 (m, 12H); ESI-MS m/z 562.92 (M+H)+.
[0381] Compound Examp
Figure imgf000140_0002
[0382] The title compound was prepared as described in EXAMPLE 1 using intermediate 6c. XH NMR (300 MHz, CD3OD) δ ppm 8.19 (d, J = 7.9 Hz, 1H), 7.63 (ddd, J = 1.5, 6.5, 7.9 Hz, 1H), 7.51 (dd, J = 2.3, 8.2 Hz, 1H), 7.37 (t, J = 8.3
Hz, 1H), 7.19-7.07 (m, 2H), 6.80 (d, J = 1.9 Hz, 1H), 5.02 (d, J = 10.8 Hz, 1H), 4.74 (d, J = 10.8 Hz, 1H), 4.1 1-3.93 (m, 2H), 3.87 (dd, J = 3.9, 12.4 Hz, 1H), 3.69-3.55 (m, 2H), 3.50-3.38 (m, 1H), 2.62 (d, J = 13.2 Hz, 1H), 2.26-2.12 (m, 1H), 2.04-1.73 (m, 4H), 1.70-1.17 (m, 5H), 1.08 (ddd, J = 3.5, 12.7, 24.0 Hz, 1H); ESI-MS m/z 562.67 (M+H)+. [0383] Compound Examp
Figure imgf000141_0001
[0384] The title compound was prepared as described in Example 1 using intermediate 6c, and 2-morpholinoethanamine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 7.64 (t, J = 7.9 Hz, 1H), 7.49
(dd, J = 2.6, 8.2 Hz, 1H), 7.33 (t, J = 8.3 Hz, 1H), 7.15-7.07 (m, 2H), 6.78 (d, J = 1.9 Hz, 1H), 5.00 (d, J = 10.3 Hz, 1H), 4.77 (d, J = 10.3 Hz, 1H), 4.12-3.57 (m, 14H), 3.36-3.19 (m, 2H), 2.37 (d, J = 10.3 Hz, 1H), 2.17-2.04 (m, 1H), 1.96-1.80 (m, 1H), 1.50-1.34 (m, 1H); ESI-MS m/z 577.75 (M+H)+.
[0385] Compound Exampl
Figure imgf000141_0002
[0386] The title compound was prepared as described in EXAMPLE 1 using intermediate 6d and methyl amine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 8.10 (s, 1H), 7.27-7.15 (m, 3H), 7.00-6.89 (m, 2H), 6.79 (d, J = 1.6 Hz, 1H), 4.66 (d, J = 10.2 Hz, 1H), 4.60 (d, J = 10.1 Hz, 1H), 3.60 (t, J = 1 1.8 Hz, 1H), 3.39-3.15 (m, 3H), 2.76 (s, 3H), 2.33-2.02 (m, 2H), 1.71 (d, J = 14.2 Hz, 1H), 1.53-1.33 (m, 1H); ESI-MS m/z 477.17 (M+H)+.
[0387] Compound Example No. 14:
Figure imgf000141_0003
[0388] The title compound was prepared as described in EXAMPLE 1 using intermediate 6d and methyl amine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 8.24 (s, 1H), 7.64 (t, J = 7.2 Hz, 1H), 7.49 (dd, J = 2.2, 8.1 Hz, 1H), 7.27 (t, J = 7.3 Hz, 1H), 7.13-7.03 (m, 2H), 6.68 (s, 1H), 4.79 (d, J = 9.6 Hz, 1H), 4.64 (d, J = 9.6 Hz, 1H), 3.70 (t, J = 13.1 Hz, 1H), 3.44-3.18
(m, 3H), 2.77 (d, J = 4.3 Hz, 3H), 2.39 (d, J = 14.5 Hz, 1H), 2.10-1.88 (m, 2H), 1.50-1.26 (m, 1H); ESI-MS m/z 477.17 (M+H)+.
[0389] Compound Examp
Figure imgf000142_0001
[0390] The title compound was prepared as described in EXAMPLE 1 using intermediate 6a and 2-morpholinoethanamine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 7.65 (t, J = 7.1 Hz, 1H), 7.47 (dd, J = 2.3, 8.1 Hz, 1H), 7.35 (t, J = 7.5 Hz, 1H), 7.17-7.05 (m, 2H), 6.77 (s, 1H), 5.02 (d, J = 10.3 Hz, 1H), 4.80 (d, J = 10.5 Hz, 1H), 3.94-3.80 (m, 4H), 3.61 (t, J = 6.2 Hz, 2H), 3.29-3.17 (m, 5H), 2.56 (d, J = 12.5 Hz, 1H), 2.11-1.48 (m, 7H),
1.29-1.02 (m, 2H); ESI-MS m/z 575.25 (M+H)+.
[0391] Compound Exampl
Figure imgf000142_0002
[0392] The title compound was prepared as described in EXAMPLE 1 using intermediate 6e and methyl amine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 8.12 (d, J = 5.4 Hz, 1H), 7.29 (d, J = 8.2 Hz, 1H), 7.26-7.13 (m, 2H), 6.99 (dd, J = 1.9, 8.1 Hz, 1H), 6.92 (t, J = 7.5 Hz, 1H), 6.79 (d, J = 1.9 Hz, 1H), 4.67 (d, J = 9.9 Hz, 1H), 4.61 (d, J = 9.9 Hz, 1H), 3.67 (dt, J = 3.8, 13.0 Hz, 1H), 3.56-3.41 (m, 2H), 3.26-3.13 (m, 1H), 2.87 (s, 3H), 2.77 (d, J = 3.7 Hz, 3H), 2.36-2.10 (m, 2H), 1.77-1.63 (m, 1H), 1.54-1.38 (m, 1H); ESI- MS m/z 491.42 (M+H)+.
[0393] Compound Exampl
Figure imgf000143_0001
[0394] The title compound was prepared as described in EXAMPLE 1 using intermediate 6e and methyl amine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 8.25 (d, J = 5.8 Hz, 1H), 7.62 ( t, J = 7.3 Hz, 1H), 7.47 (dd, J = 2.1, 8.2 Hz, 1H), 7.25 (t, J = 7.5 Hz, 1H), 7.12-7.01 (m, 2H), 6.77 (d, J = 1.6 Hz, 1H), 4.76 (d, J = 9.5 Hz, 1H), 4.62 (d, J = 9.5 Hz, 1H), 3.75 (t, J = 12.5 Hz, 1H), 3.52-3.40 (m, 2H), 3.24-3.12 (m, 1H), 2.86 (s, 3H), 2.76 (d, J = 4.0 Hz, 3H), 2.40 (d, J = 14.4 Hz, 1H), 2.12-1.86 (m, 2H), 1.54-1.34 (m, 1H); ESI-MS m/z 491.08 (M+H)+-
[0395] Compound Example No. 18:
Figure imgf000143_0002
[0396] The title compound was prepared as described in EXAMPLE 1 using intermediate 6f and methyl amine (2 equivalents) at room temperature. ESI-MS m/z 519.17 (M+H)+-
[0397] Compound Example No. 19:
Figure imgf000143_0003
[0398] The title compound was prepared as described in EXAMPLE 1 using intermediate 6f and methyl amine (2 equivalents) at room temperature. ESI-MS m/z 519.17 (M+H)+.
[0399] Compound Example
Figure imgf000144_0001
[0400] The title compound was prepared as described in EXAMPLE 1 using intermediate 6a and 2-(methylsulfonyl)ethanamine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 7.63 (t, J = 7.9 Hz, 1H), 7.50 (dd, J = 2.7, 8.2 Hz, 1H), 7.36 (t, J = 8.3 Hz, 1H), 7.19-7.07 (m, 2H), 6.77 (d, J = 1.9 Hz, 1H), 5.13 (d, J = 10.7 Hz, 1H), 3.84-3.51 (m, 2H), 3.25 (t, J = 6.3 Hz, 2H), 2.90 (s, 3H), 2.75 (d, J = 10.6 Hz, 1H), 2.13 (d, J = 14.6 Hz, 1H), 2.01-1.67 (m, 5H), 1.64-1.42 (m, 1H), 1.31-1.10 (m, 2H); ESI-MS m/z 568.25 (M+H)+
[0401] Compound Example
Figure imgf000144_0002
[0402] The title compound was prepared as described in EXAMPLE 1 using intermediate 6a and methyl amine (2 equivalents) at room temperature. XH NMR (300 MHz, CD3OD) δ ppm 8.29 (s, 1H), 7.63 (t, J = 7.2 Hz, 1H), 7.51 (dd, J = 2.4, 8.2 Hz, 1H), 7.39 (t, J = 7.6 Hz, 1H), 7.21-7.07 (m, 2H), 6.77 (d, J =1.5 Hz, 1H), 5.13 (d, J = 10.9 Hz, 1H), 4.83 (d, J = 1 1.0 Hz, 1H), 2.83 (d, J = 8.3 Hz, 1H), 2.73 (s, 3H), 2.17 (d, J = 15.3 Hz, 1H), 2.04-1.68 (m, 5H), 1.52 (q, J = 14.6 Hz, 1H), 1.31-1.09 (m, 2H); ESI-MS m/z 476.25 (M+H)+. [0403] Compound Example No. 25:
Figure imgf000145_0001
[0404] The title compound was prepared using l-(2-
(methylsulfonyl)ethyl)piperidin-4-amine hydrochloride. XH NMR (300 MHz, CD3OD) δ ppm 7.64 (t, J = 7.1 Hz, 1H), 7.47 (dd, J = 1.8, 8.2 Hz, 1H), 7.37 (t, J = 7.8 Hz, 1H), 7.15 (t, 8.1 Hz, 1H), 7.09 (dd, J = 1.5, 8.3 Hz, 1H), 6.78 (d, J = 1.4 Hz, 1H), 5.04 (d, J = 10.1 Hz, 1H), 4.79 (d, J = 10.8 Hz, 1H), 4.05-3.86 (m, 1H), 3.73-3.41 (m, 6H), 3.25-3.10 (m, 2H), 3.08 (s, 3H), 2.79-2.64 (m, 1H), 2.15 (t, J = 15.6 Hz, 2H), 2.01-1.44 (m, 9H), 1.32-1.08 (m, 2H); ESI-MS m/z 651.83 (M+H)+.
[0405] Compound Example
Figure imgf000145_0002
[0406] Intermediate 13 (see EXAMPLE 3) was dissolved in 3 : 1 methanol/water, treated with 10% TFA, and purified by preparative HPLC to give the title compound as the TFA salt. XH NMR (300 MHz, CD3OD) δ ppm 7.64 (t, J = 7.1
Hz, 1H), 7.56 (dd, J = 2.1, 8.3 Hz, 1H), 7.35 (t, J = 7.4 Hz, 1H), 7.18-7.08 (m, 2H), 6.77 (d, J = 1.4 Hz, 1H), 5.17 (d, J = 10.6 Hz, 1H), 2.64-2.49 (m, 1H), 2.14 (d, J = 13.6 Hz, 1H), 2.02-1.84 (m, 3H), 1.84-1.48 (m, 3H), 1.32-1.10 (m, 2H); ESI-MS m/z 463.17 (M+H)+. [0407] Compound Example No. 27:
Figure imgf000146_0001
[0408] The title compound was prepared using methyl 4-aminobutanoate hydrochloride. XH NMR (300 MHz, CD3OD) δ ppm 7.64 (t, J = 7.1 Hz, 1H), 7.51 (dd, J = 2.2, 8.3 Hz, 1H), 7.39 (t, J = 7.5 Hz, 1H), 7.17 (t, J = 8.1 Hz, 1H),
7.1 1 (dd, J = 1.6, 8.2 Hz, 1H), 6.78 (d, J = 1.6 Hz, 1H), 5.12 (d, J = 1 1.0 Hz, 1H), 4.80 (d, J = 1 1.1 Hz, 1H), 3.21-3.04 (m, 1H), 2.81 (d, J = 7.4 Hz, 1H), 2.17 (d, J = 12.0 Hz, 1H), 2.06 (t, J = 7.4 Hz, 2H), 2.01-1.84 (m, 3H), 1.84-1.40 (m, 5H), 1.32-1.12 (m, 2H); ESI-MS m/z 548.42 (M+H)+.
[0409] Compound Example No. 28:
Figure imgf000146_0002
[0410] The title compound was prepared using methyl trans-4-
(aminomethyl)cyclohexanecarboxylate hydrochloride. XH NMR (300 MHz, CD3OD) δ ppm 8.36-8.26 (m, 1H), 7.67 (t, J = 6.8 Hz, 1H), 7.52 (d, J = 8.2 Hz, 1H), 7.42 (t, J = 7.5 Hz, 1H), 7.20 (t, d = 8.0 Hz, 1H), 7.12 (dd, J = 1.4, 8.2 Hz, 1H), 6.78 (d, J = 1.5 Hz, 1H), 5.14 (d, J = 11.1 Hz, 1H), 4.78 (d, J = 11.3 Hz, 1H), 3.48-3.34 (m, 1H), 2.90-2.64 (m, 2H), 2.19 (d, J = 1 1.3 Hz, 1H), 2.09-1.70 (m, 8H), 1.61-1.1 1 (m, 8H), 0.79-0.59 (m, 2H); ESI-MS m/z 602.58 (M+H)+.
[0411] Compound Example No. 29:
Figure imgf000147_0001
[0412] The title compound was prepared using methyl 4-(aminomethyl)benzoate hydrochloride. XH NMR (300 MHz, CD3OD) δ ppm 7.87 (d, J = 8.2 Hz, 2H), 7.66 (t, J = 7.0 Hz, 1H), 7.50 (dd, J = 2.3, 8.3 Hz, 1H), 7.40 (t, J = 7.6 Hz, 1H), 7.22-7.08 (m, 2H), 7.04 (d, J = 8.2 Hz, 2H), 6.78 (d, J = 1.6 Hz, 1H), 5.20 (d,
J = 11.2 Hz, 1H), 4.80 (d, J = 11.1 Hz, 1H), 4.66 (d, J = 15.3 Hz, 1H), 4.20 (d, J = 15.3 Hz, 1H), 2.83 (d, J = 10.0 Hz, 1H), 2.20 (d, J = 15.8 Hz, 1H), 2.04-1.85 (m, 3H), 1.77 (d, J = 11.9 Hz, 2H), 1.52 (q, J = 13.7 Hz, 1H), 1.33-1.10 (m, 2H); ESI-MS m/z 596.33 (M+H)+.
[0413] Compound Example No. 30:
Figure imgf000147_0002
[0414] The title compound was prepared using methanesulfonamide. XH NMR
(300 MHz, CD3OD) δ ppm 7.61 (t, J = 7.5 Hz, 1H), 7.53 (dd, J = 2.1, 8.2 Hz, 1H), 7.33 (t, J = 8.0 Hz, 1H), 7.17-7.06 (m, 2H), 6.76 (d, J =1.7 Hz, 1H), 4.98 (d, J = 10.4 Hz, 1H), 3.09 (s, 3H), 2.60 (d, J = 13.7 Hz, 1H), 2.09 (d, J = 16.1 Hz, 1H), 2.01-1.43 (m, 6H), 1.34-1.06 (m, 2H); ESI-MS m/z 540.08 (M+H)+. [0415] Compound Example
Figure imgf000148_0001
[0416] The title compound was prepared using methyl 4-amino-3- methoxybenzoate. XH NMR (300 MHz, CD3OD) δ ppm 8.26 (d, J = 8.6 Hz, 1H), 7.79 (t, J = 7.1 Hz, 1H), 7.66 (dd, J = 1.4, 8.4 Hz, 1H), 7.60 (s, 1H), 7.51 (dd,
J = 2.2, 8.3 Hz, 1H), 7.40 (t, J = 8.2 Hz, 1H), 7.21 (t, J = 8.6 Hz, 1H), 7.10 (dd, J = 1.8, 8.1 Hz, 1H), 6.79 (d, J = 1.7 Hz, 1H), 5.49-5.23 (m, 1H), 3.80 (s, 3H), 2.68-2.47 (m, 1H), 2.21-1.52 (m, 7H), 1.35-1.07 (m, 2H); ESI-MS m/z 612.17 (M+H)+.
[0417] Compound Example
Figure imgf000148_0002
[0418] The title compound was prepared using (lH-tetrazol-5-yl)methanamine hydrochloride. ¾ NMR (300 MHz, CD3OD) δ ppm 7.63 (t, J = 7.2 Hz, 1H), 7.51 (dd, J = 2.3, 8.1 Hz, 1H), 7.36 (t, J = 7.6 Hz, 1H), 7.19-7.06 (m, 2H), 6.77 (d, J = 1.4 Hz, 1H), 5.19 (d, J = 10.7 Hz, 1H), 4.67 (q, J = 16.1 Hz, 2H), 2.77 (d, J = 11.4 Hz, 1H), 2.15 (d, J = 12.5 Hz, 1H), 2.03-1.81 (m, 3H), 1.76 (d, J = 13.3 Hz, 2H), 1.67-1.09 (m, 3H); ESI-MS m/z 544.25 (M+H)+. [0419] Compound Example
Figure imgf000149_0001
[0420] The title compound was prepared using (ls,3s)-3-(tert- butyldimethylsilyloxy)-3-methylcyclobutanamine. XH NMR (300 MHz, CD3OD) δ ppm 8.62 (d, J = 6.9 Hz, 1H), 7.64 (t, J = 6.8 Hz, 1H), 7.49 (dd, J = 2.4, 8.2 Hz,
1H), 7.39 (t, J = 7.1 Hz, 1H), 7.17 (t, J = 8.0 Hz, 1H), 7.10 (dd, J = 1.8, 8.2 Hz, 1H), 6.78 (d, J = 1.7 Hz, 1H), 5.10 (d, J = 11.0 Hz, 1H), 4.80 (d, J = 11.1 Hz, 1H), 3.98-3.76 (m, 1H), 2.84 (d, J = 9.8 Hz, 1H), 2.45-2.23 (m, 2H), 2.17 (d, J = 13.5 Hz, 1H), 2.05-1.82 (m, 4H), 1.82-1.61 (m, 3H), 1.61-1.34 (m, 1H), 1.30 (s, 3H), 1.27-1.07 (m, 2H); ESI-MS m/z 546.67 (M+H)+.
[0421] Compound Example
Figure imgf000149_0002
[0422] XH NMR (300 MHz, CD3OD) δ ppm 8.15 (d, J = 5.3 Hz, 1H), 7.67 (d,
J = 2.2 Hz, 1H), 7.49 (d, J = 8.2 Hz, 1H), 7.13 (dd, J = 2.4, 8.9 Hz, 1H), 7.06 (dd, J = 1.6, 8.3 Hz, 1H), 6.76 (d, J = 1.5 Hz, 1H), 6.68 (d, J = 8.9 Hz, 1H), 5.25 (d, J
= 11.3 Hz, 1H), 4.96 (d, J = 11.4 Hz, 1H), 2.85 (d, J = 8.5 Hz, 1H), 2.69 (s, 3H), 2.19 (d, J = 12.2 Hz, 1H), 2.03-1.83 (m, 3H), 1.77 (d, J = 15.3 Hz, 2H), 1.62-1.40 (m, 7H), 1.40-1.08 (m, 2H); ESI-MS m/z 560.58 (M+H)+. [0423] Compound Example
Figure imgf000150_0001
[0424] XH NMR (300 MHz, CD3OD) δ ppm 7.93 (dd, J = 2.2, 18.7 Hz, 2H), 7.47
(d, J = 8.2 Hz, IH), 7.12 (d, J = 8.1 Hz, IH), 6.80 (d, J = 1.3 Hz, IH), 4.91 (s, IH), 2.79-2.62 (m, 4H), 2.13 (d, J = 14.4 Hz, IH), 2.01-1.83 (m, 3H), 1.83-1.68
(m, 2H), 1.66-1.40 (m, 7H), 1.32-1.08 (m, 2H); ESI-MS m/z 561.33 (M+H)+.
[0425] Compound Example
Figure imgf000150_0002
[0426] The title compound was prepared using methyl 5-aminopicolinate.
¾ NMR (300 MHz, CD3OD) δ ppm 8.80 (s, IH), 8.27 (d, J = 8.8 Hz, IH), 8.15
(d, J = 8.7 Hz, IH), 7.71 (t, J = 7.2 Hz, IH), 7.55 (d, J = 8.7 Hz, IH), 7.37 (t, J = 7.1 Hz, IH), 7.18 (t, J = 7.9 Hz, IH), 7.12 (d, J = 8.1 Hz, IH), 6.80 (s, IH), 5.30 (d, J = 11.1 Hz, IH), 5.00 (d, J = 10.8 Hz, IH), 2.90-2.75 (m, IH), 2.16 (d, J = 16.9 Hz, IH), 2.06-1.84 (m, 3H), 1.78 (d, J = 13.2 Hz, 2H), 1.66-1.42 (m, IH), 1.32-1.1 1 (m, 2H); ESI-MS m/z 583.96 (M+H)+. [0427] Compound Example No. 37
Figure imgf000151_0001
[0428] The title compound was prepared using methyl 2-(4-aminopiperidin-l- yl)acetate hydrochloride. ¾ NMR (300 MHz, CD3OD) δ ppm 7.66 (t, J = 7.1 Hz, 1H), 7.48 (dd, J = 2.1, 8.2 Hz, 1H), 7.39 (t, J = 7.3 Hz, 1H), 7.17 (t, J = 8.0 Hz,
1H), 7.10 (dd, J = 1.6, 8.1 Hz, 1H), 6.79 (d, J = 1.7 Hz, 1H), 5.12 (d, J = 1 1.0 Hz, 1H), 4.80 (d, J = 10.9 Hz, 1H), 4.05-3.89 (m, 3H), 3.79-3.39 (m, 2H), 3.29-3.04 (m, 2H), 2.79 (d, J = 9.4 Hz, 1H), 2.17 (d, J = 9.4 Hz, 2H), 2.03-1.42 (m, 8H), 1.42-1.33 (m, 2H), 1.31-1.10 (m, 2H); ESI-MS m/z 603.67 (M+H)+.
[0429] Compound Example
Figure imgf000151_0002
[0430] The title compound was prepared using tert-butyl 3-aminoazetidine-l- carboxylate. The Boc protecting group was removed by treating the compound with TFA in DCM. The free amine was then treated with AcOH, EDCI, HOBt, DIEA, and DMAP (catalytic) in DCM. After stirring overnight, the solvent was removed and the product was purified by column chromatography then preparative HPLC to give Compound Example No. 38 (as the TFA salt) as a white powder. XH NMR (300 MHz, CD3OD) δ ppm 7.64 (t, J = 7.2 Hz, 1H), 7.50 (dd, J = 1.8, 8.2 Hz, 1H), 7.40 (t, J = 7.7 Hz, 1H), 7.17 (t, J = 8.1 Hz, 1H), 7.1 1 (dd, J = 1.7, 8.2 Hz, 1H), 6.79 (s, 1H), 5.10 (d, J = 10.6 Hz, 1H), 4.64-4.36 (m, 2H), 4.29-4.11 (m, 1H), 4.09-3.56 (m, 2H), 2.79 (d, J = 9.8 Hz, 1H), 2.16 (d, J = 14.9 Hz, 1H), 2.01-1.68 (m, 8H), 1.64-1.37 (m, 1H), 1.34-1.11 (m, 2H); ESI- MS m/z 560.08 (M+H)+.
[0431] Compound Example No. 39
°VOH
Figure imgf000152_0001
[0432] The title compound was prepared using methyl trans-3- aminocyclobutanecarboxylate hydrochloride. XH NMR (300 MHz, CD3OD) δ ppm 7.65 (t, J = 7.0 Hz, 1H), 7.49 (dd, J = 1.7, 8.0 Hz, 1H), 7.40 (t, J = 7.3 Hz, 1H), 7.18 (t, J = 8.0 Hz, 1H), 7.1 1 (d, J = 8.3 Hz, 1H), 6.79 (s, 1H), 5.06 (d, J = 10.8, 1H), 4.79 (d, J = 10.8 Hz, 1H), 4.47 (p, J = 8.1 Hz, 1H), 2.96-2.71 (m, 2H), 2.63-2.38 (m, 2H), 2.29-2.08 (m, 2H), 2.05-1.83 (m, 4H), 1.76 (d, J = 16.2 Hz, 2H), 1.62-1.39 (m, 1H), 1.33-1.11 (m, 2H); ESI-MS m/z 560.50 (M+H)+.
[0433] Compound Example
Figure imgf000152_0002
[0434] XH NMR (300 MHz, CD3OD) δ ppm 7.67 (t, J = 6.61 Hz, 1H), 7.49 (dd, J
= 2.0, 8.1 Hz, 1H), 7.37 (t, J = 7.5 Hz, 1H), 7.16 (t, J = 7.9 Hz, 1H), 7.10 (dd, J =
I.7, 8.3 Hz, 1H), 6.79 (d, J = 1.7 Hz, 1H), 5.20 (d, J = 1 1.3 Hz, 1H), 4.76 (d, J =
I I .2 Hz, 1H), 3.89-3.75 (m, 1H), 2.84 (d, J = 9.1 Hz, 1H), 2.41-2.29 (m, 1H), 2.20 (d, J = 15.3 Hz, 1H), 2.03-1.12 (m, 16H); ESI-MS m/z 588.50 (M+H)+. [0435] Compound Example No. 41
Figure imgf000153_0001
[0436] XH NMR (300 MHz, CD3OD) δ ppm 8.18 (d, J = 7.9 Hz, IH), 7.66 (t,
J = 6.8 Hz, IH), 7.48 (dd, J = 2.3, 8.2 Hz, IH), 7.38 (t, J = 7.7 Hz, IH), 7.17 (t, J = 8.1 Hz, IH), 7.10 (dd, J = 1.8, 8.2 Hz, IH), 6.79 (d, J = 1.8 Hz, IH), 5.14 (d,
J = 10.9 Hz, IH), 4.78 (d, J = 11.1 Hz, IH), 3.83-3.65 (m, 2H), 2.83 (d, J = 9.9 Hz, IH), 2.18 (d, J = 13.0 Hz, IH), 2.03-1.10 (m, 17H); ESI-MS m/z 560.83 (M+H)+.
[0437] Compound Example No. 42
Figure imgf000153_0002
[0438] XH NMR (300 MHz, CD3OD) δ ppm 8.45 (d, J = 2.1 Hz, IH), 8.24 (d,
J = 1.7 Hz, IH), 7.89 (s, IH), 7.60 (d, J = 8.2 Hz, IH), 7.14 (dd, J = 1.8, 8.2 Hz, IH), 6.78 (d, J = 1.8 Hz, IH), 5.10 (d, J = 10.9 Hz, IH), 4.47 (d, J = 10.9 Hz, IH), 3.73-3.57 (m, IH), 3.50-3.36 (m, IH), 2.83 (d, J = 12.5 Hz, IH), 2.17 (d, J = 14.3 Hz, IH), 2.03-1.70 (m, 8H), 1.70-1.13 (m, 7H), 1.08-0.88 (m, IH); ESI- MS m/z 543.75 (M+H)+. [0439] Compound Exampl
Figure imgf000154_0001
[0440] XH NMR (300 MHz, CD3OD) δ ppm 8.14 (s, IH), 7.84-7.67 (m, 3H), 7.55
(dd, J = 1.9, 8.2 Hz, IH), 7.48-7.34 (m, 2H), 7.19 (t, J = 8.0 Hz, IH), 7.12 (dd, J = 1.6, 8.2 Hz, IH), 6.79 (d, J = 1.6 Hz, IH), 5.29 (d, J = 11.0 Hz, IH), 4.97 (d, J = 10.7 Hz, IH), 2.96-2.84 (m, IH), 2.18 (d, J = 14.0 Hz, IH), 2.08-1.85 (m, 3H), 1.78 (d, J = 12.2 Hz, 2H), 1.63-1.42 (m, IH), 1.35-1.13 (m, 3H); ESI-MS m/z 582.58 (M+H)+.
[0441] Compound Example 44
Figure imgf000154_0002
[0442] ESI-MS m/z 596.42 (M+H) .
[0443] Compound Example No. 45
Figure imgf000154_0003
[0444] XH NMR (300 MHz, CD3OD) δ ppm 7.98 (d, J = 8.6 Hz, 2H), 7.72 (t,
J = 7.2 Hz, IH), 7.64 (d, J = 8.7 Hz, 2H), 7.54 (dd, J = 2.4, 8.1 Hz, IH), 7.38 (t, J = 7.6 Hz, IH), 7.19 (t, J = 8.0 Hz, IH), 7.12 (dd, J = 1.6, 8.2 Hz, IH), 6.80 (d, J = 1.7 Hz, IH), 5.34 (d, J = 10.7 Hz, IH), 4.97 (d, J = 10.9 Hz, IH), 2.91 (d, J = 7.1 Hz, IH), 2.20 (d, J = 14.8 Hz, IH), 2.06-1.86 (m, 3H), 1.78 (d, J = 12.3
Hz, 2H), 1.64-1.42 (m, IH), 1.34-1.14 (m, 2H); ESI-MS m/z 582.50 (M+H)+.
[0445] Compound Example
Figure imgf000155_0001
[0446] XH NMR (300 MHz, CD3OD) δ ppm 7.90 (t, J = 8.4 Hz, IH), 7.74-7.61 (m, 2H), 7.55 (dd, J = 2.5, 8.2 Hz, IH), 7.38 (t, J = 7.2 Hz, IH), 7.26 (dd, J = 1.7,
8.6 Hz, IH), 7.19 (t, J = 8.2 Hz, IH), 7.12 (dd, J = 1.8, 8.2 Hz, IH), 6.80 (d, J = 1.7 Hz, IH), 5.31 (d, J = 10.8 Hz, IH), 4.97 (d, J = 10.8 Hz, IH), 2.94-2.84 (m, IH), 2.20 (d, J = 15.7 Hz, IH), 2.06-1.85 (m, 3H), 1.79 (d, J = 10.9 Hz, 2H), 1.65-1.43 (m, IH), 1.34-1.1 1 (m, 2H); ESI-MS m/z 600.42 (M+H)+.
[0447] Compound Example
Figure imgf000155_0002
[0448] XH NMR (300MHz, CD3OD) δ ppm 8.42-8.31 (m, IH), 8.21 (t, J = 8.0
Hz, IH), 7.91-7.79 (m, IH), 7.79-7.67 (m, 2H), 7.52 (dd, J = 2.4, 8.2 Hz, IH), 7.37 (t, J = 7.3 Hz, IH), 7.25-7.06 (m, 2H), 6.79 (d, J = 1.7 Hz, IH), 5.41 (d, J = 9.4 Hz, IH), 2.81-2.67 (m, IH), 2.14 (d, J = 14.7 Hz, IH), 2.00-1.84 (m, 3H), 1.84-1.70 (m, 2H), 1.68-1.47 (m, 1H), 1.41-1.09 (m, 2H); ESI-MS m/z 600.83 (M+H)+.
EXAMPLE 6
Fluorescence-polarization MDM2 binding assay
[0449] The binding affinity of the MDM2 inhibitors disclosed herein was determined using a fluorescence polarization-based (FP-based) binding assay using a recombinant human His-tagged MDM2 protein (residues 1-118) and a fluorescently tagged p53 -based peptide.
[0450] The design of the fluorescence probe was based upon a previously reported high-affinity p53 -based peptidomimetic compound called PMDM6-F (Garcia-Echeverria et al, J. Med. Chem. 43: 3205-3208 (2000)). The Kd value of PMDM6-F with the recombinant MDM2 protein was determined from the saturation curve. MDM2 protein was serially double diluted in a Dynex 96-well, black, round-bottom plate, and the PMDM6-F peptide was added at InM concentration. The assay was performed in the buffer: 100 mM potassium phosphate, pH 7.5; 100 μg/mL bovine gamma globulin; 0.02% sodium azide, 0.01% Triton X-100) and the polarization values were measured after 3 h of incubation using an ULTRA READER (Tecan U.S. Inc., Research Triangle Park, NC). The IC50 value was obtained by fitting the mP values in a sigmoidal dose- response curve (variable slope) with a non-linear regression, and was determined to be 1.40 nM ± 0.25. The Kd value was calculated using the equation: ¾ value = IC50 - LO/2. LO/2 is the concentration of the free ligand (PMDM6-F). Since PMDM6-F was used at a final concentration of InM, LO/2 was 0.5 nM.
[0451] Dose-dependent, competitive binding experiments were performed with serial dilutions of a tested compound in DMSO. A 5 sample of the tested compound and pre-incubated MDM2 protein (10 nM) and PMDM6-F peptide (1 nM) in the assay buffer (100 mM potassium phosphate, pH 7.5; 100 μg/mL bovine gamma globulin; 0.02% sodium azide, 0.01% Triton X-100), were added in a Dynex 96-well, black, round-bottom plate to produce a final volume of 125 μϊ^. For each assay, the controls included the MDM2 protein and PMDM6-F (equivalent to 0% inhibition), PMDM6-F peptide alone (equivalent to 100% inhibition). The polarization values were measured after 3 h of incubation. The IC50 values, i.e., the inhibitor concentration at which 50% of bound peptide is displaced, were determined from a plot using nonlinear least-squares analysis. Curve fitting was performed using GRAPHPAD PRISM software (GraphPad Software, Inc., San Diego, CA). The results of this assay are present in Table 2.
EXAMPLE 7
Cell growth assay
[0452] Isogenic HCT-1 16 colon cancer cell lines were a kind gift from Prof. Bert
Vogelstein (Johns Hopkins, Baltimore, MD) and were maintained in McCoy's 5A medium containing 10% FBS. The SJSA-1 cell lines were obtained from ATCC,
(Manassas, VA) and were maintained in RPMI-1640 medium containing 10% FBS.
[0453] Cells were seeded in 96-well flat bottom cell culture plates at a density of
2-3 x lO3 cells/well with compounds and incubated for 4 days. The rate of cell growth inhibition after treatment with increasing concentrations of the tested compounds was determined by WST-8 (2-(2-methoxy-4-nitrophenyl)-3-(4- nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium monosodium salt (Dojindo Molecular Technologies Inc., Gaithersburg, Maryland). WST-8 was added at a final concentration of 10% to each well, and then the plates were incubated at 37°C for 2-3 hrs. The absorbance of the samples was measured at 450 nm using a TECAN ULTRA Reader. The concentration of the compounds that inhibited cell growth by 50% (IC50) was calculated by comparing absorbance in the untreated cells and the cells treated with the compounds using the GraphPad Prism software (GraphPad Software, La Jolla, CA 92037, USA). The results of this assay are presented in Table 2 and Fig. 1 and Fig. 2.
Table 2
Figure imgf000157_0001
Figure imgf000158_0001
EXAMPLE 8
In vivo efficacy studies using SJSA-xenograft models SJSA-1 (osteosarcoma) tumor cells were harvested with Trypsin (0.05%)- EDTA (0.53 mM) (GIBCO™, Invitrogen Corp.), growth medium was added, and the cells were placed on ice. A cell sample was mixed 1 : 1 with Trypan Blue (GIBCO™, Invitrogen Corp.) and counted on a hemocytometer to determine the number of live/dead cells. Cells were washed once with IX PBS (GIBCO™, Invitrogen Corp.) and resuspended in PBS. For Matrigel injections, after washing in PBS, cells are resuspended in an ice cold mixture of 1 : 1 PBS and Matrigel (BD Biosciences, Invitrogen Corp.) for a final Matrigel protein concentration of 5 mg/ml. SJSA-1 tumors were inoculated into C.B-17 SCID mice at 5 x 106 cells in
0.1ml with Matrigel. Cells were injected s.c. into the flank region of each mouse using a 27 gauge needle.
[0455] The size of tumors growing in the mice was measured in two dimensions using calipers. Tumor volume (mm3) = (AxB2)/2 where A and B are the tumor length and width (in mm), respectively. During treatment, tumor volume and body weight was measured three times a week. After the treatment was stopped, tumor volume and body weight was measured at least once a week. Mice were kept for an additional 60 days for further observation of tumor growth and toxicity. The anti-tumor activity of Compound Example Nos. 22 and 24 are shown in Fig. 3.
[0456] Suitable vehicles for in vivo administration of the compounds provided herein include, without limitation, 10% PEG 400:3% Cremophor:87% PBS; 98% PEG 200:2% polysorbate 80; 98% PEG 200:2% TPGS; and 0.5% polysorbate 80:0.6% methyl cellulose:98.9% water.
EXAMPLE 9
Stability of Compounds in Solution
[0457] The stability of Compound Example Nos. 3, 22, 24, and 39, and
MI-77301 (See U.S. Patent Appl. Pub. No. 2011/01 12052 A2) were determined in 1 : 1 MeOH:H20 (Fig. 4) and 1 : 1 CH3CN:H20 (Fig. 5) using ultra performance liquid chromatography. In both solvent systems, Compound Example Nos. 3, 22, 24, and 39 were more stable than MI-77301.
[0458] The stability of Compound Example Nos. 10 and 1 1 were determined in
1 : 1 MeOFLFLO with 10% TFA added using ultra performance liquid chromatography (Fig. 6). Compound Example No. 10 is converted into
Compound Example No. 1 1 under these conditions.
[0459] Having now fully described the compounds, compositions, and methods provided herein, it will be understood by those of skill in the art that the same can be performed within a wide and equivalent range of conditions, formulations, and other parameters without affecting the scope of the compounds, methods, and compositions provided herein or any embodiment thereof. All patents, patent applications, and publications cited herein are fully incorporated by reference herein in their entirety.

Claims

What Is Claimed Is:
Figure imgf000161_0001
wherein:
Rla, Rlb, Rlc, and Rld are independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, nitro, cyano, alkoxy, aryloxy, optionally substituted alkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, optionally substituted heteroaryl, carboxamido, and sulfonamido;
R2 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl;
R3a is selected from the group consisting of halo, optionally substituted alkyl, optionally substituted (cycloalkyl)alkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3- to 9-membered optionally substituted heterocyclo;
E is selected from the group consisting of -OR26a and -NR26bR26c;
R26a is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, and optionally substituted aryl;
R26b is selected from the group consisting of hydrogen and optionally substituted
Ci-C6 alkyl; R c is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, aralkyl, and -S02R5b; or
R26b and R26c taken together form a 4- to 9-membered optionally substituted heterocyclo;
X is selected from the group consisting of O, S, and NR;
Y is selected from the group consisting of O, S, and NR ;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl;
R is selected from the group consisting of hydrogen, optionally substituted alkyl, aralkyl, and optionally substituted cycloalkyl; and
represents a single or a double bond,
or a pharmaceutically acceptable salt thereof. 2. The compound of claim 1 having Formula III:
Figure imgf000162_0001
or a pharmaceutically acceptable salt thereof. The compound of claim 2 having Formula VI:
Figure imgf000162_0002
or a pharmaceutically acceptable salt thereof. The compound of claim 2 having Formula XVI
Figure imgf000163_0001
or a pharmaceutically acceptable salt thereof.
5. The compound of any one of claims 1-4, wherein:
E is -NR26bR26c;
R2 is optionally substituted aryl;
R3a is halo or d-do alkyl;
R3b is halo or Ci-Cio alkyl; or
R3a and R3b taken together form a 3- to 9-membered optionally substituted cycloalkyl or a 3- to 9-membered optionally substituted heterocyclo;
R26b is hydrogen; and
X and Y are NH,
or a pharmaceutically acceptable salt thereof. 6. The compound of any one of claims 1-5, wherein R3a and R3b are each independently Ci-Cio alkyl, or a pharmaceutically acceptable salt thereof.
7. The compound of any one of claims 1-5, wherein R3a and R3b taken together form a 4- to 8-membered optionally substituted cycloalkyl or a 4- to 8-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt thereof.
8. The compound of claim 7, wherein R3a and R3b taken together form a 6-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt thereof.
9. The compound of claim 8, wherein R3a and R3b taken together form an optionally substituted piperidine or a tetrahydropyra, or a pharmaceutically acceptable salt thereof.
10. The compound of claim 7, wherein R3a and R3b taken together form a 4- to 8-membered optionally substituted cycloalkyl, or a pharmaceutically acceptable salt thereof.
11. The compound of claim 10, wherein R3a and R3b are taken together to form an unsubstituted cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring, or a pharmaceutically acceptable salt thereof.
12. The compound of any one of claims 1-11, wherein:
Rla and Rld are each hydrogen;
R is selected from the group consisting of hydrogen and fluoro; and
Rlc is selected from the group consisting of fluoro and chloro,
or a pharmaceutically acceptable salt thereof.
13. The compound of any one of claims 1-12, wherein R2 is optionally substituted aryl having Formula R2-1 :
Figure imgf000164_0001
wherein R a, R , R c, R , and R e are each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, amino, cyano, alkoxy, alkyl, or haloalkyl, or a pharmaceutically acceptable salt thereof.
14. The compound of any one of claims 1-12, wherein R2 is optionally substituted pyridyl, or a pharmaceutically acceptable salt thereof.
15. The compound of claim 1 having Formula XXVI
Figure imgf000164_0002
XXVI, or a pharmaceutically acceptable salt thereof.
The compound of clai
Figure imgf000165_0001
XXVII,
or a pharmaceutically acceptable salt thereof.
The compound of clai
Figure imgf000165_0002
or a pharmaceutically acceptable salt thereof.
18. The compound of any one of claims 1-17, wherein R26c is optionally substituted alkyl, or a pharmaceutically acceptable salt thereof. 19. The compound of claim 18, wherein the optionally substituted alkyl is substituted with an optionally substituted cycloalkyl, or a pharmaceutically acceptable salt thereof.
20. The compound of claim 18, wherein the optionally substituted alkyl is substituted with an optionally substituted heteroaryl, or a pharmaceutically acceptable salt thereof.
21. The compound of any one of claims 1-17, wherein R26c is aralkyl, or a pharmaceutically acceptable salt thereof.
22. The compound of any one of claims 1-17, wherein R c is optionally substituted cycloalkyl, or a pharmaceutically acceptable salt thereof.
23. The compound claim 22, wherein R c is hydroxycycloalkyl, pharmaceutically acceptable salt thereof.
24. The compound of claim 22, wherein the optionally substituted cycloalkyl is substituted with at least one -CO2H, or a pharmaceutically acceptable salt thereof. 25. The compound of claim 22, wherein c is selected from the group consisting of:
Figure imgf000166_0001
larmaceutically acceptable salt thereof.
26. The compound of any one of claims 1-17 wherein R26c is optionally substituted heterocyclo, or a pharmaceutically acceptable salt thereof.
27. The compound of any one of claims 1-17, wherein R26c is optionally substituted aryl, or a pharmaceutically acceptable salt thereof.
28. The compound of claim 27, wherein R c is optionally substituted phenyl, or a pharmaceutically acceptable salt thereof.
29. The compound of claim 28, wherein the optionally substituted phenyl is substituted with at least one -CO2H, or a pharmaceutically acceptable salt thereof.
30. The compound of any one of claims 1-17, wherein R26c is optionally substituted heteroaryl, or a pharmaceutically acceptable salt thereof. 31. The compound of claim 30, wherein R26c is optionally substituted pyridyl, or a pharmaceutically acceptable salt thereof.
A compound selected from the group consisting of:
Figure imgf000167_0001
167
Figure imgf000168_0001
168
Figure imgf000169_0001
Figure imgf000170_0001
or a pharmaceutically acceptable salt thereof. A compound selected from the group consisting of:
Figure imgf000171_0001
or a pharmaceutically acceptable salt thereof.
34. The compound of claim 1, wherein E is -OR26a, or a pharmaceutically acceptable salt thereof.
35. The compound of claim 34, wherein R26a is hydrogen, or a pharmaceutically acceptable salt thereof.
36. A pharmaceutical composition comprising the compound of any one of claims 1-33, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
37. A method of treating a patient comprising administering to the patient a therapeutically effective amount of the compound of any one of claims 1-33, or a pharmaceutically acceptable salt thereof, wherein the patient has a hyperproliferative disease.
38. The method of claim 37, wherein the hyperproliferative disease is cancer.
39. The method of claim 38, wherein the cancer is selected from the group consisting of melanoma, lung cancer, sarcoma, colon cancer, prostate cancer, choriocarcinoma, breast cancer, retinoblastoma, stomach carcinoma, acute myeloid leukemia, lymph multiple myeloma, and leukemia.
40. The method of claim 39, wherein the cancer is selected from the group consisting of liposarcoma and melanoma.
41. A kit comprising a compound of any one of claims 1-33, or a pharmaceutically acceptable salt, thereof, and instructions for administering the compound to a patient having a hyperproliferative disease.
42. The kit of claim 41, wherein the hyperproliferative disease is cancer.
A method of preparing a compound having Formula XVI:
Figure imgf000172_0001
a) allowing a compound of claim 3 to isomerize; and
b) isolating the compound having Formula XVI substantially free from the compound of claim 3,
wherein X and Y are NH.
44. The method of claim 43, wherein E is -OR'
45. The method of claim 43, wherein E is -NR R' 46. The method of any one of claims 43-45, wherein R3a and R3b taken together form a 3 - to 9-membered optionally substituted cycloalkyl.
PCT/US2012/037570 2011-05-11 2012-05-11 Spiro-oxindole mdm2 antagonists WO2012155066A2 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
CA2835422A CA2835422C (en) 2011-05-11 2012-05-11 Spiro-oxindole mdm2 antagonists
KR1020137032975A KR101688268B1 (en) 2011-05-11 2012-05-11 Spiro-oxindole mdm2 antagonists
NZ617580A NZ617580B2 (en) 2011-05-11 2012-05-11 Spiro-oxindole mdm2 antagonists
ES12782304.5T ES2624808T3 (en) 2011-05-11 2012-05-11 MDM2 spirooxindol antagonists
SG2013083118A SG194873A1 (en) 2011-05-11 2012-05-11 Spiro-oxindole mdm2 antagonists
CN201280033358.3A CN103717605B (en) 2011-05-11 2012-05-11 Spiral shell-hydroxyindole MDM2 antagonist
AU2012253339A AU2012253339B2 (en) 2011-05-11 2012-05-11 Spiro-oxindole MDM2 antagonists
MX2013013167A MX346375B (en) 2011-05-11 2012-05-11 Spiro-oxindole mdm2 antagonists.
BR112013028983A BR112013028983A2 (en) 2011-05-11 2012-05-11 mdm2 antagonists spiro-oxindole
EP12782304.5A EP2707372B1 (en) 2011-05-11 2012-05-11 Spiro-oxindole mdm2 antagonists
JP2014510503A JP2014513699A (en) 2011-05-11 2012-05-11 Spiro-oxindole MDM2 antagonist
IL229387A IL229387A (en) 2011-05-11 2013-11-11 Spiro-oxindole mdm2 antagonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161484986P 2011-05-11 2011-05-11
US61/484,986 2011-05-11

Publications (2)

Publication Number Publication Date
WO2012155066A2 true WO2012155066A2 (en) 2012-11-15
WO2012155066A3 WO2012155066A3 (en) 2013-03-28

Family

ID=47140036

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/037570 WO2012155066A2 (en) 2011-05-11 2012-05-11 Spiro-oxindole mdm2 antagonists

Country Status (13)

Country Link
US (1) US8629141B2 (en)
EP (1) EP2707372B1 (en)
JP (2) JP2014513699A (en)
KR (1) KR101688268B1 (en)
CN (1) CN103717605B (en)
AU (1) AU2012253339B2 (en)
BR (1) BR112013028983A2 (en)
CA (1) CA2835422C (en)
ES (1) ES2624808T3 (en)
IL (1) IL229387A (en)
MX (1) MX346375B (en)
SG (1) SG194873A1 (en)
WO (1) WO2012155066A2 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8629133B2 (en) 2011-03-10 2014-01-14 Daiichi Sankyo Company, Limited Dispiropyrrolidine derivatives
WO2014038606A1 (en) * 2012-09-06 2014-03-13 第一三共株式会社 Crystal of dispiropyrrolidine derivative
WO2015033974A1 (en) * 2013-09-04 2015-03-12 第一三共株式会社 Method for producing spirooxindole derivative
WO2015189799A1 (en) 2014-06-12 2015-12-17 Adamed Sp. Z O.O. Compounds comprising 1,1',2,5'-tetrahydrospiro[indole-3,2'-pyrrole]-2,5'-dione system as inhibitors p53-mdm2 protein-protein interaction
WO2016001376A1 (en) 2014-07-03 2016-01-07 Boehringer Ingelheim International Gmbh New spiro[3h-indole-3,2´-pyrrolidin]-2(1h)-one compounds and derivatives as mdm2-p53 inhibitors
JP2017510610A (en) * 2014-04-11 2017-04-13 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Spiro [3H-indole-3,2'-pyrrolidin] -2 (1H) -one derivatives and their use as MDM2-p53 inhibitors
WO2017060431A1 (en) 2015-10-09 2017-04-13 Boehringer Ingelheim International Gmbh Spiro[3h-indole-3,2´-pyrrolidin]-2(1h)-one compounds and derivatives as mdm2-p53 inhibitors
JP2017511360A (en) * 2014-04-17 2017-04-20 ザ リージェンツ オブ ザ ユニバーシティー オブ ミシガン MDM2 inhibitor and therapeutic method using the same
WO2017201449A1 (en) 2016-05-20 2017-11-23 Genentech, Inc. Protac antibody conjugates and methods of use
WO2018027477A1 (en) 2016-08-08 2018-02-15 肖飞 Spirocyclic indolone polyethylene glycol carbonate compound, composition, preparation method and use thereof
CN108864113A (en) * 2018-08-03 2018-11-23 南方科技大学 A kind of bis- target spot inhibitor of MDM2-HDAC, pharmaceutical composition and its preparation and use
EP3511334A1 (en) * 2018-01-16 2019-07-17 Adamed sp. z o.o. 1,2,3',5'-tetrahydro-2'h-spiro[indole-3,1'-pyrrolo[3,4-c]pyrrole]-2,3'-dione compounds as therapeutic agents activating tp53
US10716790B2 (en) 2015-02-20 2020-07-21 Daiichi Sankyo Company, Limited Method for treating cancer by combined use
WO2021018032A1 (en) * 2019-07-26 2021-02-04 Ascentage Pharma (Suzhou) Co., Ltd. Pharmaceutical composition of mdm2 inhibitor and use thereof for preventing and/or treating disease
RU2743432C2 (en) * 2016-04-06 2021-02-18 Дзе Риджентс Оф Дзе Юниверсити Оф Мичиган Mdm2 protein destructors
WO2021175192A1 (en) * 2020-03-02 2021-09-10 Ascentage Pharma (Suzhou) Co., Ltd. Treatment methods and biomarkers for mdm2 inhibitors
WO2022120118A1 (en) 2020-12-03 2022-06-09 Baylor College Of Medicine Novel ripk1 kinase targeting protacs and methods of use thereof
WO2023056069A1 (en) 2021-09-30 2023-04-06 Angiex, Inc. Degrader-antibody conjugates and methods of using same
US11827627B2 (en) 2021-06-04 2023-11-28 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
US11834441B2 (en) 2019-12-06 2023-12-05 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ600430A (en) 2009-11-12 2014-06-27 Univ Michigan Spiro-oxindole mdm2 antagonists
EA201390682A1 (en) 2010-11-12 2014-01-30 Дзе Риджентс Оф Дзе Юниверсити Оф Мичиган SPIROOXINDAL ANTAGONISTS MDM2
JP2014513699A (en) 2011-05-11 2014-06-05 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン Spiro-oxindole MDM2 antagonist
WO2016028391A2 (en) 2014-08-18 2016-02-25 Hudson Biopharma Inc. Spiropyrrolidines as mdm2 inhibitors
JP6637652B2 (en) * 2014-11-21 2020-01-29 株式会社バイオシンクタンク Method for selecting antitumor agent and siRNA contained in the antitumor agent
TWI750129B (en) * 2015-08-03 2021-12-21 瑞士商諾華公司 Gdf-15 as a haematological toxicity biomarker
CA3020281A1 (en) 2016-04-06 2017-10-12 The Regents Of The University Of Michigan Monofunctional intermediates for ligand-dependent target protein degradation
EP3558302A4 (en) 2016-12-21 2020-07-15 Board of Regents of the University of Nebraska Dimers of covalent nfkb inhibitors
US9822128B1 (en) 2017-06-01 2017-11-21 King Saud University Substituted spirooxindoles
KR101857408B1 (en) * 2018-02-28 2018-05-14 경북대학교 산학협력단 Composition for Preventing or Treating Hair Loss
JP2022504541A (en) 2018-10-08 2022-01-13 ザ リージェンツ オブ ザ ユニバーシティー オブ ミシガン Small MDM2 Protein Degrader
EP3656760A1 (en) 2018-11-21 2020-05-27 Evonik Operations GmbH Storage stable form of 3-methylthiopropionaldehyde
EP4121043A1 (en) 2020-03-19 2023-01-25 Kymera Therapeutics, Inc. Mdm2 degraders and uses thereof

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3989816A (en) 1975-06-19 1976-11-02 Nelson Research & Development Company Vehicle composition containing 1-substituted azacycloheptan-2-ones
US4444762A (en) 1980-04-04 1984-04-24 Nelson Research & Development Company Vehicle composition containing 1-substituted azacyclopentan-2-ones
US5770581A (en) 1990-12-20 1998-06-23 Arch Development Corp. Gene transcription and ionizing radiation: methods and compositions
WO2005046575A2 (en) 2003-07-29 2005-05-26 Signature R & D Holdings, Lcc Amino acid prodrugs
WO2006091646A2 (en) 2005-02-22 2006-08-31 The Regents Of The University Of Michigan Small molecule inhibitors of mdm2 and uses thereof
US20060211757A1 (en) 2005-02-22 2006-09-21 Regents Of The University Of Michigan Small molecule inhibitors of MDM2 and the uses thereof
US20060241017A1 (en) 2003-07-29 2006-10-26 Chandran V R Novel compounds with high therapeutic index
US20060287244A1 (en) 2003-07-29 2006-12-21 Chandran V R L-Threonine derivatives of high therapeutic index
US20070249564A1 (en) 1999-03-05 2007-10-25 Metabasis Therapeutics, Inc. Novel phosphorus-containing prodrugs
WO2008036168A2 (en) 2006-08-30 2008-03-27 The Regents Of The University Of Michigan New small molecule inhibitors of mdm2 and the uses thereof
US20080125430A1 (en) 2006-08-30 2008-05-29 The Regents Of The University Of Michigan New small molecule inhibitors of mdm2 and the uses thereof
US20100317661A1 (en) 2006-08-30 2010-12-16 The Regents Of The University Of Michigan Small molecule inhibitors of mdm2 and the uses thereof
US20110112052A1 (en) 2009-11-12 2011-05-12 The Regents Of The University Of Michigan Spiro-oxindole mdm2 antagonists
WO2012121361A1 (en) 2011-03-10 2012-09-13 第一三共株式会社 Dispiropyrrolidine derivative

Family Cites Families (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3219661A (en) 1962-12-14 1965-11-23 Warner Lambert Pharmaceutical Spirooxindole and spirodehydroindole alkaloids and process therefor
GB1056537A (en) 1963-07-16 1967-01-25 Smith Kline French Lab Improvements in or relating to crystalline alkaloids of mitragyna citiata and compositions thereof
RU2084449C1 (en) 1994-03-02 1997-07-20 Всероссийский научный центр по безопасности биологически активных веществ 1-benzyl-2-oxotryptamine hydrochloride and its derivatives showing hepatoprotective activity
US5773455A (en) 1996-06-28 1998-06-30 Biomeasure, Incorporated Inhibitors of prenyl transferases
US7083983B2 (en) 1996-07-05 2006-08-01 Cancer Research Campaign Technology Limited Inhibitors of the interaction between P53 and MDM2
ATE467418T1 (en) 1997-01-20 2010-05-15 Immodal Pharmaka Gmbh METHOD AND SUBSTANCES FOR RELEASING A GROWTH FACTOR FROM ENDOTHELIAL CELLS, AND GROWTH FACTOR RELEASED AFTER THE METHOD AND THE USE THEREOF
US6114540A (en) 1997-09-08 2000-09-05 Arqule, Inc. Spiro[pyrrolidine-2,3'-oxindole] compounds and methods of use
AU5301499A (en) 1998-08-20 2000-03-14 Chugai Seiyaku Kabushiki Kaisha Method for screening candidate compounds for drug against tumor
US6831155B2 (en) 1999-12-08 2004-12-14 President And Fellows Of Harvard College Inhibition of p53 degradation
CN1182083C (en) 2001-10-08 2004-12-29 廖宜芳 Complex fertilizer for killing insect and bacteria and its production method
JP4477351B2 (en) 2001-12-18 2010-06-09 エフ.ホフマン−ラ ロシュ アーゲー Cis-2,4,5-triphenyl-imidazolines and their use in the treatment of tumors
EP1463501B1 (en) 2001-12-18 2008-06-11 F. Hoffmann-La Roche AG Cis-imidazolines as mdm2 inhibitors
JP4023184B2 (en) 2002-03-11 2007-12-19 昭和電工株式会社 Luminescent particles, production method thereof and use thereof
WO2003106384A2 (en) 2002-06-01 2003-12-24 Johns Hopkins University Novel boronic chalcone derivatives and uses thereof
AU2003291269A1 (en) 2002-11-08 2004-06-03 Irm Llc Methods and compositions for modulating p53 transcription factor
US20050227932A1 (en) 2002-11-13 2005-10-13 Tianbao Lu Combinational therapy involving a small molecule inhibitor of the MDM2: p53 interaction
CA2514374A1 (en) 2003-02-13 2004-09-02 Government Of The United States Of America, As Represented By The Secret Ary, Department Of Health And Human Services Deazaflavin compounds and methods of use thereof
US6916833B2 (en) 2003-03-13 2005-07-12 Hoffmann-La Roche Inc. Substituted piperidines
US7425638B2 (en) 2003-06-17 2008-09-16 Hoffmann-La Roche Inc. Cis-imidazolines
US7132421B2 (en) 2003-06-17 2006-11-07 Hoffmann-La Roche Inc. CIS-imidazoles
US7829742B2 (en) 2003-12-22 2010-11-09 Johns Hopkins University Boronic acid aryl analogs
WO2005097820A1 (en) 2004-04-06 2005-10-20 Korea Research Institute Of Bioscience And Biotechnology Peptides for inhibiting mdm2 function
TWI350168B (en) 2004-05-07 2011-10-11 Incyte Corp Amido compounds and their use as pharmaceuticals
ATE414693T1 (en) 2004-05-18 2008-12-15 Hoffmann La Roche NOVEL CIS-IMIDAZOLINES
GB0419481D0 (en) 2004-09-02 2004-10-06 Cancer Rec Tech Ltd Isoindolin-1-one derivatives
JP5156378B2 (en) 2004-09-22 2013-03-06 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Inhibitors of the interaction between MDM2 and p53
UA90145C2 (en) 2005-05-24 2010-04-12 Лаборатуар Сероно С.А. Tricyclic spiro derivatives as crth2 modulators
US7576082B2 (en) 2005-06-24 2009-08-18 Hoffman-La Roche Inc. Oxindole derivatives
US7495007B2 (en) 2006-03-13 2009-02-24 Hoffmann-La Roche Inc. Spiroindolinone derivatives
JP5385125B2 (en) 2006-03-22 2014-01-08 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Inhibitors of the interaction between MDM2 and p53
ATE467413T1 (en) 2006-09-21 2010-05-15 Hoffmann La Roche OXINDOL DERIVATIVES AS ANTICANCER AGENTS
US7638548B2 (en) 2006-11-09 2009-12-29 Hoffmann-La Roche Inc. Spiroindolinone derivatives
KR20090090383A (en) 2006-12-14 2009-08-25 다이이찌 산쿄 가부시키가이샤 Imidazothiazole derivatives
WO2008106507A2 (en) 2007-02-27 2008-09-04 University Of South Florida Mdm2/mdmx inhibitor peptide
AR065860A1 (en) 2007-03-29 2009-07-08 Novartis Ag 3-IMIDAZOLIL-INDOLES FOR THE TREATMENT OF PROLIFERATIVE DISEASES
US7625895B2 (en) 2007-04-12 2009-12-01 Hoffmann-Le Roche Inc. Diphenyl-dihydro-imidazopyridinones
WO2008130614A2 (en) 2007-04-20 2008-10-30 University Of Pittsburg-Of The Commonwealth System Of Higher Education Selective and dual-action p53/mdm2/mdm4 antagonists
US7553833B2 (en) 2007-05-17 2009-06-30 Hoffmann-La Roche Inc. 3,3-spiroindolinone derivatives
US7834179B2 (en) 2007-05-23 2010-11-16 Hoffmann-La Roche Inc. Spiroindolinone derivatives
CA2699707C (en) 2007-09-21 2016-05-17 Janssen Pharmaceutica Nv Inhibitors of the interaction between mdm2 and p53
CZ301561B6 (en) 2007-12-04 2010-04-14 Výzkumný ústav živocišné výroby, v. v. i. Pharmaceutical composition to prevent and suppress coccidia
US8134001B2 (en) 2007-12-14 2012-03-13 Hoffmann-La Roche Inc. Spiroindolinone derivatives
US7776875B2 (en) 2007-12-19 2010-08-17 Hoffman-La Roche Inc. Spiroindolinone derivatives
US7723372B2 (en) 2008-03-19 2010-05-25 Hoffman-La Roche Inc. Spiroindolinone derivatives
WO2009151069A1 (en) 2008-06-12 2009-12-17 第一三共株式会社 Imidazothiazole derivative having 4,7-diazaspiro[2.5]octane ring structure
GB0811643D0 (en) 2008-06-25 2008-07-30 Cancer Rec Tech Ltd New therapeutic agents
US7928233B2 (en) 2009-02-10 2011-04-19 Hoffmann-La Roche Inc. Spiroindolinone pyridine derivatives
US8076482B2 (en) 2009-04-23 2011-12-13 Hoffmann-La Roche Inc. 3,3′-spiroindolinone derivatives
US9039375B2 (en) * 2009-09-01 2015-05-26 General Electric Company Non-axisymmetric airfoil platform shaping
FR2949789B1 (en) * 2009-09-04 2011-12-09 Centre Nat Rech Scient IDENTIFICATION OF CELLULAR TROPISM OF VIRUSES
US8088815B2 (en) 2009-12-02 2012-01-03 Hoffman-La Roche Inc. Spiroindolinone pyrrolidines
US8288431B2 (en) 2010-02-17 2012-10-16 Hoffmann-La Roche Inc. Substituted spiroindolinones
WO2011106650A2 (en) 2010-02-27 2011-09-01 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Novel p53-mdm2/p53-mdm4 antagonists to treat proliferative disease
MX2012011600A (en) 2010-04-09 2012-11-30 Univ Michigan Biomarkers for mdm2 inhibitors for use in treating disease.
US8217044B2 (en) 2010-04-28 2012-07-10 Hoffmann-La Roche Inc. Spiroindolinone pyrrolidines
US20120046306A1 (en) 2010-08-18 2012-02-23 David Joseph Bartkovitz Substituted Heteroaryl Spiropyrrolidine MDM2 Antagonists
US20120071499A1 (en) 2010-09-20 2012-03-22 Xin-Jie Chu Substituted Spiro[3H-Indole-3,6'(5'H)-[1H]Pyrrolo[1,2c]Imidazole-1',2(1H,2'H)-diones
EA201390682A1 (en) 2010-11-12 2014-01-30 Дзе Риджентс Оф Дзе Юниверсити Оф Мичиган SPIROOXINDAL ANTAGONISTS MDM2
KR101853791B1 (en) * 2011-04-05 2018-05-03 삼성전자 주식회사 A method for access control of inter PLMN Handover to a Closed Subscriber Group cell
JP2014513699A (en) 2011-05-11 2014-06-05 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン Spiro-oxindole MDM2 antagonist

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3989816A (en) 1975-06-19 1976-11-02 Nelson Research & Development Company Vehicle composition containing 1-substituted azacycloheptan-2-ones
US4444762A (en) 1980-04-04 1984-04-24 Nelson Research & Development Company Vehicle composition containing 1-substituted azacyclopentan-2-ones
US5770581A (en) 1990-12-20 1998-06-23 Arch Development Corp. Gene transcription and ionizing radiation: methods and compositions
US20070249564A1 (en) 1999-03-05 2007-10-25 Metabasis Therapeutics, Inc. Novel phosphorus-containing prodrugs
US20060287244A1 (en) 2003-07-29 2006-12-21 Chandran V R L-Threonine derivatives of high therapeutic index
WO2005046575A2 (en) 2003-07-29 2005-05-26 Signature R & D Holdings, Lcc Amino acid prodrugs
US20060241017A1 (en) 2003-07-29 2006-10-26 Chandran V R Novel compounds with high therapeutic index
WO2006091646A2 (en) 2005-02-22 2006-08-31 The Regents Of The University Of Michigan Small molecule inhibitors of mdm2 and uses thereof
US20060211757A1 (en) 2005-02-22 2006-09-21 Regents Of The University Of Michigan Small molecule inhibitors of MDM2 and the uses thereof
US7759383B2 (en) 2005-02-22 2010-07-20 The Regents Of The University Of Michigan Small molecule inhibitors of MDM2 and the uses thereof
WO2008036168A2 (en) 2006-08-30 2008-03-27 The Regents Of The University Of Michigan New small molecule inhibitors of mdm2 and the uses thereof
US20080125430A1 (en) 2006-08-30 2008-05-29 The Regents Of The University Of Michigan New small molecule inhibitors of mdm2 and the uses thereof
US7737174B2 (en) 2006-08-30 2010-06-15 The Regents Of The University Of Michigan Indole inhibitors of MDM2 and the uses thereof
US20100317661A1 (en) 2006-08-30 2010-12-16 The Regents Of The University Of Michigan Small molecule inhibitors of mdm2 and the uses thereof
US20110112052A1 (en) 2009-11-12 2011-05-12 The Regents Of The University Of Michigan Spiro-oxindole mdm2 antagonists
WO2012121361A1 (en) 2011-03-10 2012-09-13 第一三共株式会社 Dispiropyrrolidine derivative

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
"A Textbook of Drug Design and Development", 1991, GORDON & BREACH, article "Design and Applications of Prodrugs"
"Burger's Medicinal Chemistry and Drug Discovery", vol. 1, 1995, JOHN WILEY & SONS, pages: 172 - 178,949-
"Design of Prodrugs", 1985, ELSEVIER
"Methods in Enzymology", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING CO., EASTON, PA
BERGE ET AL., J. PHARM. SCI., vol. 66, 1997, pages 1 - 19
BUNDGARD: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
DING ET AL., J. AM. CHEM. SOC., vol. 127, 2005, pages 10130 - 10131
DING ET AL., J. MED. CHEM., vol. 49, 2006, pages 3432 - 3435
DING ET AL., JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 127, no. 29, 2005, pages 10130 - 10131
E. B. ROCHE: "Bioreversible Carriers in Drug Design", 1987, ELSEVIER
GARCIA-ECHEVERRIA ET AL., J. MED. CHEM., vol. 43, 2000, pages 3205 - 3208
GARCIA-ECHEVERRIA ET AL., MED. CHEM., vol. 43, 2000, pages 3205
GOBERDHAN, CANCER CELL, vol. 7, 2005, pages 505
K. B. SLOAN: "Prodrugs: Topical and Ocular Drug Delivery", 1998, MARCEL DEKKER
KUSSIE ET AL., SCIENCE, vol. 274, 1996, pages 948
LOWE ET AL., CARCINOGENESIS, vol. 27, 2000, pages 485
NICHOLSON, NATURE, vol. 407, 2000, pages 810
PONDER, NATURE, vol. 411, 2001, pages 336
See also references of EP2707372A4
SILVERMAN: "The Organic Chemistry of Drug Design and Drug Action", 1992, ACADEMIC PRESS, pages: 352 - 401
T. HIGUCHI AND V. STELLA: "ProDrugs as Novel Delivery Systems", 1975, AM. CHEM. SOC.
VOGELSTEIN ET AL., NATURE, vol. 408, 2000, pages 307
WU ET AL., GENES DEV., vol. 7, 1993, pages 1126

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8629133B2 (en) 2011-03-10 2014-01-14 Daiichi Sankyo Company, Limited Dispiropyrrolidine derivatives
AU2012226890B2 (en) * 2011-03-10 2016-10-06 Daiichi Sankyo Company, Limited Dispiropyrrolidine derivative
US9718830B2 (en) 2012-09-06 2017-08-01 Daiichi Sankyo Company, Limited Crystals of dispiropyrrolidine derivatives
WO2014038606A1 (en) * 2012-09-06 2014-03-13 第一三共株式会社 Crystal of dispiropyrrolidine derivative
US10030030B2 (en) 2012-09-06 2018-07-24 Daiichi Sankyo Company, Limited Crystals of dispiropyrrolidine derivatives
US10023578B2 (en) 2012-09-06 2018-07-17 Daiichi Sankyo Company, Limited Crystals of dispiropyrrolidine derivatives
US9884871B2 (en) 2012-09-06 2018-02-06 Daiichi Sankyo Company, Limited Crystals of dispiropyrrolidine derivatives
US9359368B2 (en) 2012-09-06 2016-06-07 Daiichi Sankyo Company, Limited Crystals of dispiropyrrolidine derivatives
US9540386B2 (en) 2012-09-06 2017-01-10 Daiichi Sankyo Company, Limited Crystals of dispiropyrrolidine derivatives
US9745315B2 (en) 2012-09-06 2017-08-29 Daiichi Sankyo Company, Limited Crystals of dispiropyrrolidine derivatives
US9718831B2 (en) 2012-09-06 2017-08-01 Daiichi Sankyo Company, Limited Crystals of dispiropyrrolidine derivatives
US10851111B2 (en) 2013-09-04 2020-12-01 Daiichi Sankyo Company, Limited Method for producing a spirooxindole derivative
US10030028B2 (en) * 2013-09-04 2018-07-24 Daiichi Sankyo Company, Limited Method for producing a spirooxindole derivative
JPWO2015033974A1 (en) * 2013-09-04 2017-03-02 第一三共株式会社 Process for producing spirooxindole derivatives
CN105555789A (en) * 2013-09-04 2016-05-04 第一三共株式会社 Method for producing spirooxindole derivative
US11884678B2 (en) 2013-09-04 2024-01-30 Daiichi Sankyo Company, Limited Method for producing a spirooxindole derivative
CN105555789B (en) * 2013-09-04 2018-08-10 第一三共株式会社 The preparation method of spiral shell oxygroup indole derivatives
WO2015033974A1 (en) * 2013-09-04 2015-03-12 第一三共株式会社 Method for producing spirooxindole derivative
JP2017510610A (en) * 2014-04-11 2017-04-13 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Spiro [3H-indole-3,2'-pyrrolidin] -2 (1H) -one derivatives and their use as MDM2-p53 inhibitors
JP2020100662A (en) * 2014-04-17 2020-07-02 ザ リージェンツ オブ ザ ユニバーシティー オブ ミシガン MDM2 inhibitor and treatment method using the same
JP2017511360A (en) * 2014-04-17 2017-04-20 ザ リージェンツ オブ ザ ユニバーシティー オブ ミシガン MDM2 inhibitor and therapeutic method using the same
WO2015189799A1 (en) 2014-06-12 2015-12-17 Adamed Sp. Z O.O. Compounds comprising 1,1',2,5'-tetrahydrospiro[indole-3,2'-pyrrole]-2,5'-dione system as inhibitors p53-mdm2 protein-protein interaction
EA030564B1 (en) * 2014-06-12 2018-08-31 Адамед Сп. З О.О. COMPOUNDS COMPRISING 1,1',2,5'-TETRAHYDROSPIRO[INDOLE-3,2'-PYRROLE]-2,5'-DIONE SYSTEM AS INHIBITORS p53-Mdm2 PROTEIN-PROTEIN INTERACTION
WO2016001376A1 (en) 2014-07-03 2016-01-07 Boehringer Ingelheim International Gmbh New spiro[3h-indole-3,2´-pyrrolidin]-2(1h)-one compounds and derivatives as mdm2-p53 inhibitors
US10716790B2 (en) 2015-02-20 2020-07-21 Daiichi Sankyo Company, Limited Method for treating cancer by combined use
WO2017060431A1 (en) 2015-10-09 2017-04-13 Boehringer Ingelheim International Gmbh Spiro[3h-indole-3,2´-pyrrolidin]-2(1h)-one compounds and derivatives as mdm2-p53 inhibitors
US11192898B2 (en) 2016-04-06 2021-12-07 The Regents Of The University Of Michigan MDM2 protein degraders
RU2743432C2 (en) * 2016-04-06 2021-02-18 Дзе Риджентс Оф Дзе Юниверсити Оф Мичиган Mdm2 protein destructors
WO2017201449A1 (en) 2016-05-20 2017-11-23 Genentech, Inc. Protac antibody conjugates and methods of use
WO2018027477A1 (en) 2016-08-08 2018-02-15 肖飞 Spirocyclic indolone polyethylene glycol carbonate compound, composition, preparation method and use thereof
WO2019141549A1 (en) * 2018-01-16 2019-07-25 Adamed Pharma S.A. 1,2,3',5'-tetrahydro-2'h-spiro[indole-3,1'-pyrrolo[3,4-c]pyrrole]-2,3'-dione compounds as therapeutic agents activating tp53
CN111566110A (en) * 2018-01-16 2020-08-21 阿达梅德制药公司 1,2,3',5' -tetrahydro-2 ' H-spiro [ indole-3, 1' -pyrrolo [3,4-C ] pyrrole ] -2,3' -dione compounds as therapeutic agents for activating TP53
EP3511334A1 (en) * 2018-01-16 2019-07-17 Adamed sp. z o.o. 1,2,3',5'-tetrahydro-2'h-spiro[indole-3,1'-pyrrolo[3,4-c]pyrrole]-2,3'-dione compounds as therapeutic agents activating tp53
AU2019209114B2 (en) * 2018-01-16 2022-12-08 Adamed Pharma S.A. 1,2,3',5'-tetrahydro-2'H-spiro(indole-3,1'-pyrrolo(3,4-c)pyrrole)-2,3'-dione compounds as therapeutic agents activating TP53
CN108864113B (en) * 2018-08-03 2021-08-13 南方科技大学 MDM2-HDAC double-target inhibitor, pharmaceutical composition, preparation and application thereof
CN108864113A (en) * 2018-08-03 2018-11-23 南方科技大学 A kind of bis- target spot inhibitor of MDM2-HDAC, pharmaceutical composition and its preparation and use
WO2021018032A1 (en) * 2019-07-26 2021-02-04 Ascentage Pharma (Suzhou) Co., Ltd. Pharmaceutical composition of mdm2 inhibitor and use thereof for preventing and/or treating disease
US11834441B2 (en) 2019-12-06 2023-12-05 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
US11919887B2 (en) 2019-12-06 2024-03-05 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
WO2021175192A1 (en) * 2020-03-02 2021-09-10 Ascentage Pharma (Suzhou) Co., Ltd. Treatment methods and biomarkers for mdm2 inhibitors
WO2022120118A1 (en) 2020-12-03 2022-06-09 Baylor College Of Medicine Novel ripk1 kinase targeting protacs and methods of use thereof
US11827627B2 (en) 2021-06-04 2023-11-28 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
WO2023056069A1 (en) 2021-09-30 2023-04-06 Angiex, Inc. Degrader-antibody conjugates and methods of using same

Also Published As

Publication number Publication date
JP2014513699A (en) 2014-06-05
CN103717605A (en) 2014-04-09
EP2707372A2 (en) 2014-03-19
AU2012253339B2 (en) 2016-03-31
US20120289494A1 (en) 2012-11-15
BR112013028983A2 (en) 2017-02-07
NZ617580A (en) 2015-07-31
KR101688268B1 (en) 2016-12-20
JP6251301B2 (en) 2017-12-20
JP2016106111A (en) 2016-06-16
AU2012253339A1 (en) 2014-01-09
MX2013013167A (en) 2014-07-09
ES2624808T3 (en) 2017-07-17
KR20140079341A (en) 2014-06-26
EP2707372B1 (en) 2016-12-21
WO2012155066A3 (en) 2013-03-28
CA2835422C (en) 2016-10-11
IL229387A (en) 2017-03-30
IL229387A0 (en) 2014-01-30
CA2835422A1 (en) 2012-11-15
SG194873A1 (en) 2013-12-30
US8629141B2 (en) 2014-01-14
CN103717605B (en) 2016-05-18
MX346375B (en) 2017-03-16
EP2707372A4 (en) 2014-10-22

Similar Documents

Publication Publication Date Title
JP6251301B2 (en) Spiro-oxindole MDM2 antagonist
JP2020100662A (en) MDM2 inhibitor and treatment method using the same
US9302120B2 (en) Spiro-oxindole MDM2 antagonists
US8877796B2 (en) Spiro-oxindole MDM2 antagonists
EP2063887A2 (en) New small molecule inhibitors of mdm2 and the uses thereof
WO2020112846A1 (en) Small molecule modulators of sigma-1 and sigma-2 receptors and uses thereof
WO2020123670A1 (en) Small molecule inhibitors of the androgen receptor activity and/or expression and uses thereof
OA16409A (en) Spiro-oxindole MDM2 antagonists.

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: 12782304

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2835422

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2014510503

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/013167

Country of ref document: MX

REEP Request for entry into the european phase

Ref document number: 2012782304

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2012782304

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20137032975

Country of ref document: KR

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013028983

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2012253339

Country of ref document: AU

Date of ref document: 20120511

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112013028983

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20131111