US20130289024A1 - Compositions and methods for treatment of filovirus-mediated diseases - Google Patents

Compositions and methods for treatment of filovirus-mediated diseases Download PDF

Info

Publication number
US20130289024A1
US20130289024A1 US13/928,630 US201313928630A US2013289024A1 US 20130289024 A1 US20130289024 A1 US 20130289024A1 US 201313928630 A US201313928630 A US 201313928630A US 2013289024 A1 US2013289024 A1 US 2013289024A1
Authority
US
United States
Prior art keywords
methyl
dihydro
trifluoromethyl
chloro
methoxy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/928,630
Inventor
Lisa M. Johansen
Joseph Lehár
Benjamin G. Hoffstrom
Gene G. Olinger
Andrea R. Stossel
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Army Medical Research and Materiel Command USAMRMC
Zalicus Inc
Original Assignee
Zalicus Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zalicus Inc filed Critical Zalicus Inc
Priority to US13/928,630 priority Critical patent/US20130289024A1/en
Assigned to Zalicus, Inc. reassignment Zalicus, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HOFFSTROM, BENJAMIN G., LEHAR, JOSEPH, JOHANSEN, LISA M.
Publication of US20130289024A1 publication Critical patent/US20130289024A1/en
Assigned to U.S. ARMY MEDICAL RESEARCH AND MATERIEL COMMAND reassignment U.S. ARMY MEDICAL RESEARCH AND MATERIEL COMMAND ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: STOSSEL, ANDREW R
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals

Definitions

  • the invention relates to the treatment of diseases caused by a filovirus.
  • Filoviruses are particularly deadly viruses that cause severe hemorrhagic fever in humans and non-human primates. Filoviruses are sporadically transmitted to humans from a natural reservoir species thought to be fruit bats, after which the virus can easily spread from the human having the infection to a caregiver through bodily fluids. Infection with a filovirus of the Ebola or Marburg genus causes death in 25% to 90% of human victims, often within 10 days from the first appearance of symptoms.
  • the present invention features compositions, methods, and kits for the treatment of filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus.
  • the invention features a method for treating a patient with a filovirus-mediated disease, e.g., a disease caused by an Ebola virus or Marburg virus.
  • the method includes administering to the patient a first agent selected from the agents of Table 2, or an analog thereof, in an amount that is effective to treat the patient.
  • the first agent is selected from the agents of Table 1. More preferably, the first agent is clomiphene, toremifene, astemizole, bepridil, clomopramine, lomerizine, sertraline, or an analog thereof.
  • the method further includes administering a second agent selected from the agents of Table 1.
  • the first and second agents are both selected from Table 1. More preferably, the first and second agents are selected from the agent pairs of Table 3.
  • the invention also features a method for treating a patient having a filovirus-mediated disease, e.g., a disease caused by Ebola virus or Marburg virus, the method including administering to the patient: a first agent that is an inhibitor of estrogen receptor ⁇ and a second agent that is an inhibitor of histamine receptor 1 in amounts that together are effective to treat the patient.
  • the first agent is preferably clomiphene, tamoxifen, raloxifene, toremifene, diethylstilbestrol, or an analog thereof. More preferably, the first agent is clomiphene, tamoxifen, toremifene, or an analog thereof.
  • the second agent is preferably chlorcyclizine, astemizole, clemastine, terfenadine, chlorphenoxamine, oxatomide, azelastine, methdilazine, homochlorcyclizine, desloratadine, flunarizine, loratadine, doxylamine, or an analog thereof. More preferably, the second agent is astemizole or an analog thereof.
  • the invention also features a method for treating a patient having a filovirus-mediated disease, e.g., a disease caused by Ebola virus or Marburg virus, the method including administering to the patient: a first agent that is an inhibitor of estrogen receptor ⁇ and a second agent that is sertraline or an analog thereof in amounts that together are effective to treat the patient.
  • the first agent is preferably clomiphene, tamoxifen, raloxifene, toremifene, diethylstilbestrol, or an analog thereof. More preferably, the first agent is clomiphene, tamoxifen, toremifene, or an analog thereof.
  • the second agent is preferably sertraline, paroxetine, or UK-416244.
  • the agents When the methods includes administering to the patient a pair of active agents, the agents may be administered within within 28, 21, 14, 10, 7, 5, 4, 3, 2, or 1 days; within 24, 12, 6, 3, 2, or 1 hours; or substantially simultaneously.
  • the methods of the invention may include administering one or more agents to the patient by oral, systemic, parenteral, topical (e.g., ophthalmic, dermatologic), intravenous, inhalational, or intramuscular administration.
  • the patient being treated has not been diagnosed with or does not suffer from breast cancer, osteoporosis, prostate hyperplasia, metabolic syndrome X, male infertility, testosterone deficiency, hypogonadism, non-insulin-dependent diabetes, infertility due to an ovulatory disorder, infertility due to polycystic ovary syndrome, gynaecomastia, or hormone deficiency.
  • any of the above methods e.g., a method including administration of an inhibitor of histamine receptor 1
  • the patient has not been diagnosed with or does not suffer from urticaria, seasonal allergic rhinitis, allergy or excessive itching, or cancer (e.g., cancer of the lung, pancreatic, ovary, breast, prostate, colon, brain, or skin).
  • cancer e.g., cancer of the lung, pancreatic, ovary, breast, prostate, colon, brain, or skin.
  • the patient being treated has not been diagnosed with or does not suffer from a major depressive disorder, an obsessive-compulsive disorder, an anxiety disorder, or a psychotic disorder (e.g., schizophrenia).
  • the patient being treated has not been diagnosed with or does not suffer from migraine, cardiac arrhythmia, hypertension, pneumocystis pneumonia, toxoplasmosis, malaria, organ transplant rejection, fungal infection, hypolipidemia, hypercholesterolemia, chronic myelogenous leukemia, cancer (e.g., cancer of the lung, pancreatic, ovary, breast, prostate, colon, brain, or skin), bacterial infection, schistosomal infection, malaria, HIV, osteoporosis, gynaecomastia.
  • the invention features a composition including two or more agents selected from the agents of Table 1.
  • the two or more agents are present in amounts that, when administered together to a patient with a filovirus-mediated disease (e.g., a disease caused by Ebola virus or Marburg virus), are effective to treat the patient.
  • the first and second agents are selected from the agent pairs of Table 3.
  • the composition consists of active ingredients and excipients, and the active ingredients consist of said two agents selected from the agents of Table 1.
  • the invention also features a composition that includes a first agent that is an inhibitor of estrogen receptor ⁇ and a second agent that is an inhibitor of histamine receptor 1.
  • the first and second agents are present in amounts that, when administered together to a patient with a filovirus-mediated disease (e.g., a disease caused by Ebola virus or Marburg virus), are effective to treat the patient.
  • the first agent is preferably clomiphene, tamoxifen, raloxifene, toremifene, diethylstilbestrol, or an analog thereof. More preferably, the first agent is clomiphene, tamoxifen, toremifene, or an analog thereof.
  • the second agent is preferably chlorcyclizine, astemizole, clemastine, terfenadine, chlorphenoxamine, oxatomide, azelastine, methdilazine, homochlorcyclizine, desloratadine, flunarizine, loratadine, doxylamine, or an analog thereof. More preferably, the second agent is astemizole or an analog thereof.
  • the composition consists of active ingredients and excipients, and the active ingredients consist of the first and second agents
  • the invention also features a composition that includes a first agent that is an inhibitor of estrogen receptor ⁇ and a second agent that is sertraline or an analog thereof.
  • the first and second agents are present in amounts that, when administered together to a patient with a filovirus-mediated disease (e.g., a disease caused by Ebola virus or Marburg virus), are effective to treat the patient.
  • the first agent is preferably clomiphene, tamoxifen, raloxifene, toremifene, diethylstilbestrol, or an analog thereof. More preferably, the first agent is clomiphene, tamoxifen, toremifene, or an analog thereof.
  • the second agent is preferably sertraline, paroxetine, or UK-416244.
  • the composition consists of active ingredients and excipients, and the active ingredients consist of the first and second agents.
  • compositions of the invention may be formulated, for example, for oral, systemic, parenteral, topical (e.g., ophthalmic, dermatologic), intravenous, inhalational, or intramuscular administration.
  • the invention features a kit including an active agent selected from the agents of Table 2 and instructions for administering the agent to a patient having a filovirus-mediated disease, e.g., a disease caused by an Ebola or a Marburg virus.
  • the active agent is selected from the agents of Table 1. More preferably, the active agent is clomiphene, toremifene, astemizole, bepridil, clomopramine, lomerizine, sertraline, or an analog thereof.
  • the invention also features a kit including two or more agents selected from the agents of Table 1 and instructions for administering the agents to a patient having a filovirus-mediated disease, e.g., a disease causes by an Ebola or a Marburg virus.
  • the two agents are selected from the agent pairs of Table 3.
  • the two agents may be included together in a composition or may be formulated separately.
  • kits including a first agent and selected from Table 1 and instructions for administering the first agent with a second agent selected from Table 1 to a patient having a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus.
  • the first and second agents are each clomiphene, toremifene, astemizole, bepridil, clomopramine, lomerizine, sertraline, or an analog thereof.
  • kits that include an agent that is an inhibitor of estrogen receptor ⁇ , an agent that is an inhibitor of histamine receptor 1, and instructions for administering the agents to a patient with a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus.
  • kits that include an agent that is an inhibitor of estrogen receptor ⁇ , an agent that is an inhibitor of histamine receptor 1, and instructions for administering the agents to a patient with a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus.
  • the inhibitor of estrogen receptor ⁇ is preferably selected from clomiphene, tamoxifen, toremifene, and analogs thereof and more preferably from clomiphene, tamoxifen, raloxifene, toremifene, and diethylstilbestrol, and analogs thereof.
  • the inhibitor of histamine receptor 1 is preferably selected from the group consisting of chlorcyclizine, astemizole, clemastine, terfenadine, chlorphenoxamine, oxatomide, azelastine, methdilazine, homochlorcyclizine, desloratadine, flunarizine, loratadine, doxylamine, and analogs thereof, and more preferably from astemizole or an analog thereof.
  • the kit includes the inhibitor of estrogen receptor ⁇ and the inhibitor of histamine receptor, the two agents may be included together in a composition or they may be formulated separately.
  • kits that include an agent that is an inhibitor of estrogen receptor ⁇ , an agent that is sertraline or an analog thereof, and instructions for administering the agents to a patient with a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus.
  • kits that include an agent that is an inhibitor of estrogen receptor ⁇ , an agent that is sertraline or an analog thereof, and instructions for administering the agents to a patient with a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus.
  • the inhibitor of estrogen receptor ⁇ is preferably selected from clomiphene, tamoxifen, toremifene, and analogs thereof and more preferably from clomiphene, tamoxifen, raloxifene, toremifene, and diethylstilbestrol, and analogs thereof.
  • the sertraline or analog thereof is preferably sertraline, paroxetine, or UK-416244.
  • the kit includes the inhibitor of estrogen receptor ⁇ and the sertraline or analog thereof, the two agents may be included together in a composition or they may be formulated separately.
  • compositions, methods, or kits that include an inhibitor of estrogen receptor ⁇ and an inhibitor of histamine receptor 1, these agents are preferably clomiphene and chlorcyclizine, clomiphene and astemizole, clomiphene and clemastine, clomiphene and terfenadine, clomiphene and chlorphenoxamine, clomiphene and oxatomide, clomiphene and azelastine, clomiphene and methdilazine, clomiphene and homochlorcyclizine, clomiphene and desloratadine, clomiphene and flunarizine, clomiphene and loratadine, clomiphene and doxylamine, tamoxifen and chlorcyclizine, tamoxifen and astemizole, tamoxifen and clemastine, tamoxifen and terfenadine, tamoxifen and
  • one or more active agents may be formulated, e.g., for oral, parenteral, systemic, topical, or inhalational administration.
  • Compounds useful in the invention include those described herein in any of their pharmaceutically acceptable forms, including isomers such as diastereomers and enantiomers, salts, solvates, and polymorphs thereof, as well as racemic mixtures.
  • Compounds useful in the invention may also be isotopically labeled compounds.
  • Useful isotopes include hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, (e.g., 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl).
  • Isotopically-labeled compounds can be prepared by synthesizing a compound using a readily available isotopically-labeled reagent in place of a non-isotopically-labeled reagent.
  • Histamine receptor 1 refers to the histamine receptor also known as H 1 receptor and HRH1.
  • an “inhibitor of histamine receptor 1” is meant an agent that antagonizes the signaling of histamine receptor 1 by at least 5%, 10%, 25%, 50%, 60%, 70%, 80%, 90%, or 95%. Inhibition of histamine receptor 1 signaling by an agent can be measured, for example, by reporter assays known in the art.
  • estrogen receptor ⁇ refers to estrogen hormone receptors containing the estrogen receptor polypeptide encoded by the estrogen receptor 1 (ESR1) gene.
  • An estrogen receptor ⁇ protein can include a homodimer of polypeptides encoded by ESR1 or a heterodimer of polypeptides encoded by the ESR1 and estrogen receptor 2 (ESR2) genes.
  • an “estrogen receptor ⁇ inhibitor” is meant an agent that binds to and antagonizes the activity of estrogen receptor ⁇ by at least 5%, 10%, 25%, 50%, 60%, 70%, 80%, 90%, or 95% in at least one cell type of an animal.
  • Certain estrogen receptor ⁇ inhibitors e.g., agents known in the art as “selective estrogen receptor modulators,” may inhibit estrogen receptors in one organ, e.g, the breast, while activating estrogen receptors in another organ, e.g., the liver, bone, or uterus. Such compounds are included in the term “estrogen receptor ⁇ inhibitors.”
  • the % inhibition of estrogen receptor ⁇ activity by an agent can be measured, for example, by a reporter assay known in the art.
  • filovirus is meant a virus belonging to the family Filoviridae.
  • Exemplary filoviruses are Ebola virus and Marburg virus.
  • patient any animal, e.g., a primate, e.g., a human. Any animal can be treated using the methods, compositions, and kits of the invention.
  • To “treat” is meant to administer one or more agents to measurably slow or stop the replication of a virus in vitro or in vivo, to measurably decrease the load of a virus (e.g., any virus described herein including a filovirus virus such as an Ebola virus or Marburg virus) in a cell in vitro or in vivo, or to reduce at least one symptom (e.g., those described herein) associated with having a filovirus-mediated disease in a patient.
  • a virus e.g., any virus described herein including a filovirus virus such as an Ebola virus or Marburg virus
  • at least one symptom e.g., those described herein
  • the slowing in replication or the decrease in viral load is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99%, as determined using a suitable assay (e.g., a replication assay described herein).
  • a suitable assay e.g., a replication assay described herein.
  • treatment is observed by a trained physician as an appreciable or substantial relief of symptoms in a patient with a filovirus-mediated disease.
  • a decrease in viral replication is accomplished by reducing the rate of RNA polymerization, RNA translation, protein processing or modification, or by reducing the activity of a molecule involved in any step of viral replication (e.g., proteins or coded by the genome of the virus or host important for viral replication).
  • an effective amount is meant the amount of a compound, alone or in combination with another therapeutic regimen, required to treat a patient with a viral disease (e.g., any virus described herein including an Ebola virus or Marburg virus) in a clinically relevant manner.
  • a viral disease e.g., any virus described herein including an Ebola virus or Marburg virus
  • a sufficient amount of an active compound used to practice the present invention for therapeutic treatment of conditions caused by a virus varies depending upon the manner of administration, the age, body weight, and general health of the patient.
  • the prescribers will decide the appropriate amount and dosage regimen.
  • the effective amount of an agent may less be than the effective amount if the agent were administered in a non-combinatorial (single-agent) therapy.
  • an effective amount may be an amount of an agent in a combination therapy of the invention that is safe and efficacious in the treatment of a patient having a viral disease over each agent alone as determined and approved by a regulatory authority (such as the U.S. Food and Drug Administration).
  • a treatment exhibits greater efficacy, or is less toxic, safer, more convenient, or less expensive than another treatment with which it is being compared. Efficacy may be measured by a skilled practitioner using any standard method that is appropriate for a given indication.
  • a “low dosage” is meant at least 5% less (e.g., at least 10%, 20%, 50%, 80%, 90%, or even 95%) than the lowest standard recommended dosage of a particular compound formulated for a given route of administration for treatment of any human disease or condition.
  • a low dosage of an agent that inhibits viral replication and that is formulated for administration by intravenous injection will differ from a low dosage of the same agent formulated for oral administration.
  • the number of atoms of a particular type in a substituent group is generally given as a range, e.g., an alkyl group containing from 1 to 4 carbon atoms or C 1-4 alkyl. Reference to such a range is intended to include specific references to groups having each of the integer number of atoms within the specified range.
  • an alkyl group from 1 to 4 carbon atoms includes each of C 1 , C 2 , C 3 , and C 4 .
  • a C 1-12 heteroalkyl for example, includes from 1 to 12 carbon atoms in addition to one or more heteroatoms.
  • Other numbers of atoms and other types of atoms may be indicated in a similar manner.
  • alkyl and the prefix “alk-” are inclusive of both straight chain and branched chain groups and of cyclic groups, i.e., cycloalkyl.
  • Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 12 ring carbon atoms, inclusive.
  • Exemplary cyclic groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl groups.
  • C 1-4 alkyl is meant a branched or unbranched hydrocarbon group having from 1 to 4 carbon atoms.
  • a C 1-4 alkyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • C 1-4 alkyls include, without limitation, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclopropylmethyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, and cyclobutyl.
  • C 2-4 alkenyl is meant a branched or unbranched hydrocarbon group containing one or more double bonds and having from 2 to 4 carbon atoms.
  • a C 2-4 alkenyl may optionally include monocyclic or polycyclic rings, in which each ring desirably has from three to six members.
  • the C 2-4 alkenyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • C 2-4 alkenyls include, without limitation, vinyl, allyl, 2-cyclopropyl-1-ethenyl, 1-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2-methyl-1-propenyl, and 2-methyl-2-propenyl.
  • C 2-4 alkynyl is meant a branched or unbranched hydrocarbon group containing one or more triple bonds and having from 2 to 4 carbon atoms.
  • a C 2-4 alkynyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has five or six members.
  • the C 2-4 alkynyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxy, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • C 2-4 alkynyls include, without limitation, ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, and 3-butynyl.
  • C 2-6 heterocyclyl is meant a stable 5- to 7-membered monocyclic or 7- to 14-membered bicyclic heterocyclic ring which is saturated, partially unsaturated, or unsaturated (aromatic), and which consists of 2 to 6 carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from N, O, and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxy, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • the nitrogen and sulfur heteroatoms may optionally be oxidized.
  • the heterocyclic ring may be covalently attached via any heteroatom or carbon atom which results in a stable structure, e.g., an imidazolinyl ring may be linked at either of the ring-carbon atom positions or at the nitrogen atom.
  • a nitrogen atom in the heterocycle may optionally be quaternized.
  • Heterocycles include, without limitation, 1H-indazole, 2-pyrrolidonyl, 2H,6H-1,5,2-dithiazinyl, 2H-pyrrolyl, 3H-indolyl, 4-piperidonyl, 4aH-carbazole, 4H-quinolizinyl, 6H-1,2,5-thiadiazinyl, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 4aH-carbazolyl, b-carboliny
  • Preferred 5 to 10 membered heterocycles include, but are not limited to, pyridinyl, pyrimidinyl, triazinyl, furanyl, thienyl, thiazolyl, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, tetrazolyl, benzofuranyl, benzothiofuranyl, indolyl, benzimidazolyl, 1H-indazolyl, oxazolidinyl, isoxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, quinolinyl, and isoquinolinyl.
  • Preferred 5 to 6 membered heterocycles include, without limitation, pyridinyl, pyrimidinyl, triazinyl, furanyl, thienyl, thiazolyl, pyrrolyl, piperazinyl, piperidinyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, and tetrazolyl.
  • C 6-12 aryl is meant an aromatic group having a ring system comprised of carbon atoms with conjugated ⁇ electrons (e.g., phenyl).
  • the aryl group has from 6 to 12 carbon atoms.
  • Aryl groups may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has five or six members.
  • the aryl group may be substituted or unsubstituted.
  • substituents include alkyl, hydroxy, alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, fluoroalkyl, carboxyl, hydroxyalkyl, carboxyalkyl, amino, aminoalkyl, monosubstituted amino, disubstituted amino, and quaternary amino groups.
  • C 7-14 alkaryl is meant an alkyl substituted by an aryl group (e.g., benzyl, phenethyl, or 3,4-dichlorophenethyl) having from 7 to 14 carbon atoms.
  • aryl group e.g., benzyl, phenethyl, or 3,4-dichlorophenethyl
  • C 3-10 alkheterocyclyl is meant an alkyl substituted heterocyclic group having from 3 to 10 carbon atoms in addition to one or more heteroatoms (e.g., 3-furanylmethyl, 2-furanylmethyl, 3-tetrahydrofuranylmethyl, or 2-tetrahydrofuranylmethyl).
  • C 1-7 heteroalkyl is meant a branched or unbranched alkyl, alkenyl, or alkynyl group having from 1 to 7 carbon atoms in addition to 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, S, and P.
  • Heteroalkyls include, without limitation, tertiary amines, secondary amines, ethers, thioethers, amides, thioamides, carbamates, thiocarbamates, hydrazones, imines, phosphodiesters, phosphoramidates, sulfonamides, and disulfides.
  • a heteroalkyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has three to six members.
  • the heteroalkyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • Examples of C 1-7 heteroalkyls include, without limitation, methoxymethyl and ethoxyethyl.
  • halide or “halogen” is meant bromine, chlorine, iodine, or fluorine.
  • fluoroalkyl is meant an alkyl group that is substituted with a fluorine atom.
  • perfluoroalkyl is meant an alkyl group consisting of only carbon and fluorine atoms.
  • Carboxyalkyl is meant a chemical moiety with the formula —(R)—COOH, wherein R is selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl.
  • hydroxyalkyl is meant a chemical moiety with the formula —(R)—OH, wherein R is selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl.
  • alkoxy is meant a chemical substituent of the formula —OR, wherein R is selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl.
  • aryloxy is meant a chemical substituent of the formula —OR, wherein R is a C 6-12 aryl group.
  • alkylthio is meant a chemical substituent of the formula —SR, wherein R is selected from C 1-7 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl.
  • arylthio is meant a chemical substituent of the formula —SR, wherein R is a C 6-12 aryl group.
  • quaternary amino is meant a chemical substituent of the formula N(R)(R′)(R′′)(R′′′) + , wherein R, R′, R′′, and R′′′ are each independently an alkyl, alkenyl, alkynyl, or aryl group.
  • R may be an alkyl group linking the quaternary amino nitrogen atom, as a substituent, to another moiety.
  • the nitrogen atom, N is covalently attached to four carbon atoms of alkyl, heteroalkyl, heteroaryl, and/or aryl groups, resulting in a positive charge at the nitrogen atom.
  • compositions of the invention can include a combination pair of any two agents selected from Table 2 or the particular pairs of agents of Table 3.
  • Treatment methods of the invention include administration of a single agent from Table 1 or Table 2 or a pair of agents selected from the agents of Table 1 or from the agent pairs listed in Table 3.
  • functional or structural analogs e.g., those described herein
  • these agents may be employed in the compositions, methods, and kits of the invention.
  • the composition may function by decreasing RNA polymerization, RNA translation, RNA transcription, a decrease in posttranslational protein processing, or a decrease in activity of a protein involved in viral replication (e.g., a protein coded for by the viral genome or a host protein required for viral replication).
  • a protein involved in viral replication e.g., a protein coded for by the viral genome or a host protein required for viral replication.
  • the patient being treated is administered a combination of two agents listed in Table 1 within 7 days of each other in amounts that together are sufficient to treat the patient having a filovirus-mediated disease.
  • An effective amount of one or both of the agents may be a low dosage relative the effective amount of the agent when administered singly.
  • the invention relates to the treatment of diseases caused by viruses of the family Filoviridae (filoviruses).
  • Filoviruses are negative strand RNA viruses that can infect humans and primates.
  • Filoviruses include viruses of the genera Ebolavirus and Marburgvirus.
  • Ebola hemorrhagic fever is a severe, often-fatal disease in humans and has appeared sporadically since its initial recognition in 1976. The disease is caused by infection with an Ebola virus.
  • Ebola-Zaire Five identified subtypes of Ebola virus are Ebola-Zaire, Ebola-Sudan, Ebola-Ivory Coast, and Ebola-Bundibugyo, each of which have caused disease in humans, and Ebola-Reston, which has caused disease in nonhuman primates, but not in humans. Other undiscovered subtypes may exist and are intended to be included in the scope of Ebola virus.
  • Infection with an Ebola or Marburg virus usually causes life-threatening hemorrhagic fever. Symptoms include fever, severe headache, joint and muscle aches, chills, sore throat, weakness, nausea and vomiting, diarrhea, red eyes, raised rash, chest pain and cough, hiccups (Ebola virus), stomach pain, bleeding (from any bodily orifice), and psychological symptoms (confusion, irribility, aggression, or depression). As the illness progresses, jaundice, dilirium, seizures, severe bleeding, organ failure, coma, shock, and death can occur.
  • an estrogen receptor ⁇ inhibitor can be used in the compositions, methods, and kits of the invention.
  • an “estrogen receptor ⁇ inhibitor” is meant a compound that inhibits the activity of an estrogen receptor ⁇ by at least 5%, e.g., greater than 10%, 20%, 40%, 60%, 80%, 90%, or 95%.
  • ER ⁇ inhibitors include clomiphene, tamoxifen, tomerifene, and raloxifene. ER ⁇ inhibitors that may be particularly efficacious are described in more detail below.
  • Clomiphene is described in U.S. Pat. No. 2,914,563 and has the following structure:
  • Structural analogs of clomiphene include olefinic isomers. Structural analogs of clomiphene are also described by the following formula:
  • R 1 , R 2 , and R 3 may be located at any position of the phenyl group and are selected, independently, from H, halogen, C 1-6 alkyl, C 1-6 alkoxy, —OC n H 2n A, and at least one of R 1 , R 2 , and R 3 is —OC n H 2n A, wherein n is 2, 4, 5, or 6;
  • A NR 4 R 5 , wherein each R 4 and R 5 is, independently, an optionally substituted C 1-6 alkyl, or R 4 and R 5 combined to form an optionally substituted cyclic structure.
  • R 1 , R 2 , or R 3 is —OC n H 2n A
  • the substituents is located para to the olefin substituents.
  • Examples of C 1-6 alkyls include, but are not limited to: methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, isoamyl, and n-hexyl.
  • C 1-6 alkoxy groups include, but are not limited to: methoxy, ethoxy, propoxy, isopropoxy, n-butyloxy, iso-butyloxy, sec-butyloxy, tert-butyloxy, n-pentoxy, O-isoamyl, and O-hexyl.
  • rings formed by the combination R 4 and R 5 include, but are not limited to pyrrolidine and piperidine.
  • Tamoxifen is described in U.S. Pat. No. 4,536,516 and has the following structure:
  • Tamoxifen analogs are described by general formula (I) in U.S. Pat. No. 4,806,685, e.g., threo-1-[4-(2,3-epoxypropoxy)-phenyl]-1,2-diphenyl-3,3,3-trifluoro-propane, (E)-1,2-diphenyl-3,3,3-trifluoro-1-[4-(2-[4-methylpiperazino]-ethoxy)-phenyl]-propene, 1-[4-(2-dimethylaminoethoxy)-phenyl]-2-phenyl-3,3,3-trifluoro-1-(4-hydroxy phenyl)propene, (E)1,2-diphenyl-3,3,3-trifluoro-1-[4-(2-[2-hydroxyethylamino]-ethoxy)-phenyl]-propene, (E)-1-[4-(2-azidoethoxy)-phenyl]-1,
  • tamoxifen analogs are described by general formula (I) in U.S. Pat. No. 5,047,431, e.g., (E)-1-[4′-(2-dimethylaminoethoxy)phenyl]-1-(3′-hydroxyphenyl)-2-phenylbut-1-ene, (E)-1-[4′(2-diethylaminoethoxy)phenyl]-1-(3′-hydroxyphenyl)-2-phenylbut-1-ene, (E)-1-(3′-hydroxyphenyl)-1-[4′-(2-methylaminoethoxy)phenyl]-2-phenylbut-1-ene, and (E)-1-[4′-ethylaminoethoxy)phenyl]-1-(3′-hydroxyphenyl)-2-phenylbut-1-ene.
  • tamoxifen analogs are droloxifene, 4-iodotamoxifen, idoxifene, described in U.S. Pat. No. 6,096,874, in U.S. Pat. No. 6,576,645, in U.S. Pat. No. 6,875,775, and by general formula (I) of U.S. Pat. No. 5,807,899.
  • the methods, compositions, and kits of the invention employ an inhibitor of histamine receptor 1.
  • the inhibitor may inhibit the activity of histamine receptor 1 by at least 5%, e.g., greater than 10%, 20%, 40%, 60%, 80%, 90%, or 95%. Either non-sedating or sedating inhibitors may be employed.
  • Exemplary histamine receptor 1 inhibitors include acrivastine, alcaftadine, antazoline, azatadine, AZD-1744, azelastine, bepotastine, bepotastine besilate, betotastine besilate, bilastine, BM-113, carebastine, cetirizine, chlorpheniramine, chlorphenoxamine, clemastine, cyclizine, desloratadine, doxepin, E-4716, ebastine, efletirizine, epinastine, epinastine, fexofenadine, FK-613, GSK-1004723, HSR-609, IOT-101, KA-398, KAA-276, KC-11404, KC-11425, ketotifen, levocetirizine, loratadine, MDL-28163, mianserin, mizolastine, NBI-75043,
  • Analogs of astemizole include, e.g, 1-(4-fluorophenylmethyl)-N- ⁇ 1-[2-(4-methoxyphenyl)ethyl]-4-piperidinyl ⁇ -1H-benzimidazol-2-amine, 4-[2- ⁇ 4-[1-(4-fluorophenylmethyl)-1H-benzimidazol-2-ylamino]-1-piperidinyl ⁇ ethyl]phenol, ⁇ 4-[2- ⁇ 4-[1-(4-fluorophenylmethyl)-1H-benzimidazol-2-ylamino]-1-piperidinyl ⁇ ethyl]phenyl ⁇ benzeneacetate, ⁇ 4-[2- ⁇ 4-[1-(4-fluorophenylmethyl)-1H-benzimidazol-2-ylamino]-1-piperidinyl ⁇ ethyl]phenoxy ⁇ acetonitrile, and compounds described by the formula of claim 1 in U.S
  • Clemastine is a sedating anti-histamine agent often provided as clemastine fumarate.
  • Analogs of clemastine include, e.g., dephenhydramine, meclizine, clobenztropine, nchembio873-comp44 ((1S,5S)-3-[(4-chlorophenyl)-phenylmethoxy]-8-methyl-8-azabicyclo[3.2.1]octane), AHR 209 (3-[(4-chlorophenyl)-phenylmethoxy]-1-propan-2-ylpyrrolidine hydrochloride), chlortropbenzyl, pyroxamine, AHR225 (1-butyl-3-[(4-chlorophenyl)-phenylmethoxy]pyrrolidine), AHR226 (1-tert-butyl-3-[(4-chlorophenyl)-phenylmethoxy]pyrrolidine hydrochloride), AHR211 (3-[(4-
  • phenothiazine is meant a polycyclic heterocycle including two optionally substituted benzene rings fused to one of thiazine and described by the formula:
  • each of R a and R c is, independently, selected from of H, F, Cl, Br, CF 3 , cyano, S—R d , S(O) 2 —R d , C 1 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, and C 1-7 heteroalkyl; each R d is, independently, selected from C 1 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, and C 1-7 heteroalkyl; and R b is selected from C 3-10 alkheterocyclyl and C 1-7 heteroalkyl.
  • compositions, methods, or kits of the invention employ thioridazine, prochlorperazine, piperacetazine, or fluphenazine.
  • a related phenothiazine such as one described below may also be employed.
  • Thioridazine, prochlorperazine (U.S. Pat. No. 2,902,484), piperacetazine (GB Pat. No. 861,807), and fluphenazine (U.S. Pat. No. 3,058,979) have the following structures:
  • phenothiazines which include, without limitation, acepromazine, cyamemazine, fluphenazine, mepazine, methotrimeprazine, methoxypromazine, perazine, pericyazine, perimethazine, perphenazine, pipamazine, pipazethate, piperacetazine, pipotiazine, promethazine, propionylpromazine, propiomazine, sulforidazine, thiazinaminiumsalt, thiethylperazine, thiopropazate, thioridazine, trifluoperazine, trimeprazine, thioproperazine, trifluomeprazine, triflupromazine, chlorpromazine, chlorproethazine, those compounds in PCT application WO02/057244, and those compounds in U.S.
  • phenothiazines include, without limitation, acepromazine, c
  • Phenothiazine compounds can be prepared using, for example, the synthetic techniques described in U.S. Pat. Nos. 2,415,363; 2,519,886; 2,530,451; 2,607,773; 2,645,640; 2,766,235; 2,769,002; 2,784,185; 2,785,160; 2,837,518; 2,860,138; 2,877,224; 2,921,069; 2,957,870; 2,989,529; 3,058,979; 3,075,976; 3,194,733; 3,350,268; 3,875,156; 3,879,551; 3,959,268; 3,966,930; 3,998,820; 4,785,095; 4,514,395; 4,985,559; 5,034,019; 5,157,118; 5,178,784; 5,550,143; 5,595,989; 5,654,323; 5,688,788; 5,693,649; 5,712,292; 5,721,254;
  • compositions, methods, or kits of the invention employ a calcium channel-blocking agent, e.g., lomerizine or bepridil.
  • a calcium channel-blocking agent e.g., lomerizine or bepridil.
  • Lomerizine and analogs of lomerizine e.g., 1-(2,4-dimethoxybenzyl)-4-(4-fluorobenzhydryl)-piperazine, 1-(2,4-dimethoxybenzyl)-4-[bis(4-fluorophenyl)-methyl]piperazine], and 1-(2,3,4-trimethoxybenzyl)-4-(4-fluorobenzhydryl)piperazine, are described by general formula (I) in U.S. Pat. No. 4,663,325.
  • Other analogs include cinnamylpiperazine derivatives described by formula (I) in U.S. Pat. No.
  • 4,703,048, such as 1-[bis(4-fluorophenyl)methyl]-4-(2,3,4-trimethoxycinnamyl)piperazine, 1-benzhydryl-4-(2,3,4-trimethoxycinnamyl)piperazine, 1-[bis(4-fluorophenyl)methyl]-4-(2,4-dimethoxycinnamyl)piperazine, and 1-benzhydryl-4-(2,4-dimethoxycinnamyl)piperazine, diene derivatives of general formula (I) described in U.S. Pat. No.
  • Bepridil and analogs of bepridil are described by general formula (I) of U.S. Pat. No. 3,962,238. Other analogs are described by the general formula of claim 1 in U.S. Pat. No. 4,645,778, e.g., 2-(N-pyrrolidino)-3-isobutoxy-N-(3,4-methylenedioxyphenyl)-N-benzyl-propyl amine, 2-(N-pyrrolidino)-3-isobutoxy-N(2,6-dimethylphenyl)-N-benzyl-propylamine, and 2-(N-pyrrolidino)-3-isobutoxy-N-(3,4-dichlorophenyl)-N-benzyl propylamine; by formula (I) of U.S.
  • Pat. No. 4,999,361 e.g., ethyl 2,2-bis-(4-methoxyphenyl)-4-(1-pyrrolidino)-5-isobutoxyvalerate, 2-phenyl-2-(2-pyridyl)-4-(1-pyrrolidino)-5-isobutoxyvaleronitrile, ethyl 2,2-diphenyl-4-(N,N-diethylamino)-5-isobutoxyvalerate, ethyl 2,2-diphenyl-4-(1-pyrrolidino)-5-benzyloxyvalerate, and ethyl 2,2-diphenyl-4-(1-pyrrolidino)-5-(2-picolyloxy)valerate.
  • ethyl 2,2-bis-(4-methoxyphenyl)-4-(1-pyrrolidino)-5-isobutoxyvalerate 2-phenyl-2-(2-pyridyl)-4-(
  • Analogs of lomerizine and bepridil with calcium-channel blocking activity include clentiazem (U.S. Pat. No. 4,567,175), gallopamil (U.S. Pat. No. 3,261,859), mibefradil (U.S. Pat. No. 4,808,605), prenylamine (U.S. Pat. No. 3,152,173), semotiadil (U.S. Pat. No. 4,786,635), terodiline (U.S. Pat. No. 3,371,014), aranipine (U.S. Pat. No. 4,572,909), bamidipine (U.S. Pat. No.
  • lidoflazine U.S. Pat. No. 3,267,104
  • bencyclane Hungarian Pat. No. 151,865
  • etafenone German Pat. No. 1,265,758
  • perhexyline British Pat. No. 1,025,578
  • Analogs of lomerizine with vasodilating activity may also be employed in certain embodiments of the inventions.
  • Examples are cinnarizine, citicoline (which may be isolated from natural sources as disclosed in Kennedy et al. ( J. Am. Chem. Soc., 77:250 (1955)) or synthesized as disclosed in Kennedy ( J. Biol. Chem., 222:185 (1956)), cyclandelate (see U.S. Pat. No. 3,663,597), ciclonicate (see German Pat. No. 1,910,481), diisopropylamine dichloroacetate (see GB Pat. No. 862,248), eburnamonine (see Hermann et al., J. Am.
  • Selective serotonin reuptake inhibitors may be employed in certain embodiments of the invention.
  • selective serotonin reuptake inhibitor or “SSRI” is meant any member of the class of compounds that (i) inhibit the uptake of serotonin by neurons of the central nervous system, (ii) have an inhibition constant (Ki) of 10 nM or less, and (iii) a selectivity for serotonin over norepinephrine (i.e., the ratio of K i (norepinephrine) over K i (serotonin)) of greater than 100.
  • SSRIs that may be particularly useful are paroxetine, sertraline, and UK-416244.
  • Paroxetine is an SSRI described in U.S. Pat. No. 3,912,743.
  • paroxetine analogs are compounds described by formula IV of U.S. Pat. No. 4,485,109, e.g., 4-phenyl-a, a,1-trimethyl-4-piperidinemethanol; ( ⁇ )-trans-4-(4-fluorophenyl)-3-(4-methoxyphenoxy)methylpiperidine and ( ⁇ )-trans-4-(4-fluorophenyl)-3-[(4-methoxyphenoxy)methyl]-1-methylpiperidine, described in U.S. Pat. No. 4,585,777 and U.S. Pat. No.
  • Pat. No. 5,328,917 e.g., 1-butyl-3-(4-methoxybenzylaminomethyl)-4-phenylpiperidine, 1-butyl-3-(4-trifluoromethylphenylaminomethyl)-4-phenylpiperidine, and ( ⁇ ) trans-1-butyl-3-(2-phenylethylaminomethyl)-4-phenylpiperidine.
  • sertraline or an analog thereof can be used in the compositions, methods, and kits of the invention.
  • Sertraline has the structure:
  • Structural analogs of sertraline include those having the formula:
  • R 1 and R 2 are independently selected from the group consisting of H, optionally substituted C 1-6 alkyl (e.g., CH 3 , (CH 2 )—OH, cyclopropyl, (CH 2 ) x COOH, or CH 2 CHOH(CH 2 ) x , (CH 2 ) x N(CfH 3 ) 2 , where x is 1, 2, 3, 4, or 5), and optionally substituted C 1-7 heteroalkyl (e.g., CH 2 CH 2 N(CH 3 ) 2 ) or R 1 and R 2 together form a C 3-8 cycloalkyl optionally heterocyclic, optionally substituted (e.g., forming a morpholine ring), R 3 , R 4 , R 5 , and R 6 are independently H, Cl, F, Br, OH, or optionally substituted C 1-6 alkyl; X and Y are each selected from the group consisting of H, F, Cl, Br, CF 3 , C 1-6 alkoxy
  • CONHcylopropyl C(S)NH 2 , NHC(S)CH 3 , CONHCH 2 COOCH 3 , CONHCH 2 COOH, CONHCH 2 cyclopropyl, CONHcyclobutyl, NHCOcyclopropyl, NH(CH 3 )COCH 3 , and CH 2 S(O) n R 11 , where n is 0, 1, or 2 and R 11 is phenyl, C 2-6 heterocyclyl, optionally substituted C 1-8 alkyl (e.g., C 4-8 unsubstituted alkyl such as Bu or C 3-8 substituted alkyl).
  • R 1 is CH 3 and R 2 is CH 3 , CH 2 CH 2 OH, cyclopropyl, CH 2 COOH, CH 2 CH 2 NH 2 , CH 2 CH(OH)R 8 , or CH 2 CH(R 8 )NR 9 R 10 , where n is 0, 1, or 2 and R 8 , R 9 , and R 10 are independently H or C 1-6 alkyl.
  • X is H and Y is p-OPh, p-OCF 3 , o-OCH 3 m-OCH 3 , or p-OCH 3 .
  • the sertraline analog has the formula:
  • R 3 , R 4 , R 5 , R 6 , W, X, and Y are as defined above, and R 7 is independently H, NH(CH 2 ) m CH 3 , O(CH 2 ) m CH 3 , OH, O(CH 2 ) m CH 3 , ⁇ O, C 1-6 alkyl (e.g., isopropyl), or C 1-6 alkyoxy, where m is 0, 1, 2, 3, 4, 5, or 6.
  • R 3 , R 4 , R 5 , and R 6 are H; X and Y are each Cl at the 3 and 4 positions of the benzyl ring.
  • Exemplary analogs include:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , X and Y are as defined above, and R 7 is H or C 1-6 optionally substituted alkyl.
  • Other sertraline analogs are described by the formula:
  • R 8 , R 9 , and R 10 are independently H, optionally substituted C 1-6 alkyl (e.g., CH 3 , (CH 2 ) x OH, cyclopropyl, (CH 2 ) x COOH, or CH 2 CHOH(CH 2 ) x , (CH 2 ) x N(CH 3 ) 2 , where x is 1, 2, 3, 4, or 5), and optionally substituted C 1-7 heteroalkyl (e.g., CH 2 CH 2 N(CH 3 ) 2 ).
  • C 1-6 alkyl e.g., CH 3 , (CH 2 ) x OH, cyclopropyl, (CH 2 ) x COOH, or CH 2 CHOH(CH 2 ) x , (CH 2 ) x N(CH 3 ) 2 , where x is 1, 2, 3, 4, or 5
  • C 1-7 heteroalkyl e.g., CH 2 CH 2 N(CH 3 ) 2 ).
  • sertraline analogs are in the cis-isomeric configuration.
  • the term “cis-isomeric” refers to the relative orientation of the NR 1 R 2 and phenyl moieties on the cyclohexene ring (i.e., they are both oriented on the same side of the ring). Because both the 1- and 4-carbons are asymmetrically substituted, each cis-compound has two optically active enantiomeric forms denoted (with reference to the 1-carbon) as the cis-(1R) and cis-(1S) enantiomers. Sertraline analogs are also described in U.S. Pat. No. 4,536,518.
  • Other related compounds include (S,S)—N-desmethylsertraline, rac-cis-N-desmethylsertraline, (1S,4S)-desmethyl sertraline, 1-des (methylamine)-1-oxo-2-(R,S)-hydroxy sertraline, (1R,4R)-desmethyl sertraline, sertraline sulfonamide, sertraline (reverse) methanesulfonamide, 1R,4R sertraline enantiomer, N,N-dimethyl sertraline, nitro sertraline, sertraline aniline, sertraline iodide, sertraline sulfonamide NH 2 , sertraline sulfonamide ethanol, sertraline nitrile, sertraline-CME, dimethyl sertraline reverse sulfonamide, sertraline reverse sulfonamide (CH 2 linker), sertraline B-ring ortho methoxy, sertraline A-ring methyl ester, sertraline A-ring
  • UK-416244 is an SSRI that is phenoxybenzylamine derivative. UK-416244 has the structure:
  • R 1 and R 2 independently, are H, C 1-6 alkyl (e.g., CH 3 ) or substituted heteroalkyl, or (CH 2 ) d (C 3-6 cycloalkyl) where d is 0, 1, 2, or 3; or R 1 and R 2 together with the nitrogen to which they are attached form an azetidine ring;
  • Z or Y is —S(O) n R 3 and the other Z or Y is halogen or —R 3 ;
  • R 3 is independently C 1-4 alkyl optionally substituted with fluorine (e.g., where R 3 is or is not CF 3 ) and n is 0, 1, or 2; or Z and Y are linked so that, together with the interconnecting atoms, Z and Y form a fused 5 to 7-membered carbocyclic or heterocyclic ring which may be saturated, unsaturated, or aromatic, and where when Z and Y form a heterocyclic ring, in addition to carbon atoms, the link
  • R 1 and R 2 are independently H, C 1-6 alkyl (e.g., CH 3 ) or substituted heteroalkyl, (CH 2 ) m (C 3-6 cycloalkyl) where m is 0, 1, 2, or 3, or R 1 and R 2 together with the nitrogen to which they are attached form an azetidine ring; each R 3 is independently H, I, Br, F, Cl, C 1-6 alkyl (e.g., CH 3 ), CF 3 , CN, OCF 3 , C 1-4 alkylthio (e.g., SCH 3 ), C 1-4 alkoxy (e.g., OCH 3 ), aryloxy (e.g., OPh), or CONR 6 R 7 ; n is 1, 2, or 3; and R 4 and R 5 are independently A-X, where A is —CH ⁇ CH— or —(CH 2 ) p — where p is 0, 1, or 2; X is H, F, Cl, Br, I, CONR 6 R
  • n is 1 or 2
  • the R 3 group(s) is/are at positions 3 and/or 4 of the B ring, for example, are CH 3 , SCH 3 , OCH 3 , Br, or CF 3 .
  • R 4 or R 5 can be SO 2 NHPh, SO 2 NHCH 3 , CN, H, Br, CONH 2 , COOH, SO 2 NHCH 2 Ph, SO 2 NHCOCH 3 , CH 2 NHSO 2 CH 3 NH 2 , OR NO 2 , benzyl amide, acylsulfonamide, reverse sulfonamide, NHCH 3 , N(CH 3 ) 2 , SO 2 NH 2 , CH 2 OH, NHSO 2 CH 3 , SO 2 NHCH 2 CCH 2 , CH 2 NH 2 , SO 2 NHBu, and SO 2 NHcyclopropyl.
  • UK-416244 structural analogs are described in U.S. Pat. Nos. 6,448,2
  • R 3 , R 4 , and R 5 are as defined above and Z is CH 2 NR 1 R 2 where R 1 and R 2 are as defined above, C 1-6 alkyl, optionally substituted (e.g., with hydroxyl, NH 2 , NHC 1-6 alkyl).
  • Z is CH 2 CH(CH 3 ) 2 , CH 2 OCH 3 , CH 2 N(CH 3 )CH 2 CH 2 OH, N(CH 3 ) 2 , CH 2 N(CH 3 ) 2 , COOH, CH 2 NHCH 3 , CH 2 OH, CH 2 NHCOCH 3 , or CONHCH 3 .
  • R 1 is H, I, Br, F, Cl, C 1-6 alkyl (e.g., CH 3 ), CF 3 , CN, OCF 3 , C 1-4 alkylthio (e.g., SCH 3 ), C 1-4 alkoxy (e.g., OCH 3 ), aryloxy, or CONR 2 R 3 ;
  • n is 1, 2, or 3;
  • R 2 and R 3 are independently H or C 1-6 alkyl (e.g., (CH 2 ) 3 CH 3 or cyclopropyl), C 6-12 aryl (e.g., phenyl) optionally substituted independently by one or more R 4 , or C 1-6 alkyl-aryl optionally substituted;
  • R 4 is F (preferably up to 3), OH, CO 2 H, C 3-6 cycloalkyl, NH 2 , CONH 2 , C 1-6 alkoxy, C 1-6 alkoxycarbonyl or a 5- or 6-membered heterocyclic ring
  • X is N, O, or S
  • R 1 is H, C 1-6 alkyl or substituted heteroalkyl, (CH 2 ) m (C 3-6 cycloalkyl) where m is 0, 1, 2, or 3.
  • R 1 is H or C 1-6 alkyl (e.g., CH 3 , CH 2 CH 3 ) and R 2 is C 1-6 alkyl substituted with OH, such as CH 2 OH, CH 2 CH 2 OH, CH(OH)CH 3 , CH 2 CH 2 CH 2 OH, CH(CH 2 )CH 2 OH, and CH 2 CH 2 CH 2 CH 2 OH, CH(OH)CH 2 CH 2 CH 3 , CH 2 CH(OH)CH 2 CH 3 , and CH 2 CH 2 CH(OH)CH 3 ) or is CH 2 XR 14 or CH 2 CH 2 XR 14 , where X is N, O, or S, and R 14 is H, C 1-6 alkyl or substituted heteroalkyl, (CH 2 ) q (C 3-6 cycloalkyl) where q is 0, 1, 2, or 3, and where R 3 , R 4 , and R 5 are as defined above.
  • OH such as CH 2 OH, CH 2 CH 2 OH, CH(OH)CH 3
  • the analog has the structure:
  • R 1 is H or C 1-6 alkyl (e.g., CH 3 , CH 2 CH 3 ) and R 2 is C 1-6 alkyl substituted with OH, e.g., CH 2 OH, CH 2 CH 2 OH, CH(OH)CH 3 , CH 2 CH 2 CH 2 OH, CH(CH 2 )CH 2 OH, and CH 2 CH 2 CH 2 CH 2 OH, CH(OH)CH 2 CH 2 CH 3 , CH 2 CH(OH)CH 2 CH 3 , and CH 2 CH 2 CH(OH)CH 3 ).
  • the compound is:
  • SSRIs are considered herein to be analogs of paroxetine, sertraline, and UK-416244 and, thus, may be used in connection with the invention.
  • SSRIs include cericlamine (e.g., cericlamine hydrochloride); citalopram (e.g., citalopram hydrobromide); clovoxamine; cyanodothiepin; dapoxetine; escitalopram (escitalopram oxalate); femoxetine (e.g., femoxetine hydrochloride); fluoxetine (e.g., fluoxetine hydrochloride); fluvoxamine (e.g., fluvoxamine maleate); ifoxetine; indalpine (e.g., indalpine hydrochloride); indeloxazine (e.g., indeloxazine hydrochloride); litoxetine; milnacipran (e.g., min
  • SNRIs selective serotonin norepinephrine reuptake inhibitors
  • venlafaxine duloxetine
  • Pharmacologically active metabolites of any of the foregoing SSRIs and SNRIs can also be used in the methods, compositions, and kits of the invention.
  • Exemplary metabolites are didesmethylcitalopram, desmethylcitalopram, desmethylsertraline, and norfluoxetine.
  • Flupentixol is a thioxanthene anti-psychotic drug. Flupentixol is described in U.S. Pat. No. 3,282,930, and exemplary analogs of flupentixol are described by formula I in U.S. Pat. No.
  • Aripiprazole is an atypical antipsychotic described in U.S. Pat. No. 5,006,528.
  • Exemplary analogs are given by variations of formula I in U.S. Pat. No. 5,006,528, e.g., 7- ⁇ 4-[4-(b 2,3-dichlorophenyl)-1-piperazinyl]-butoxy ⁇ carbostyril, 7- ⁇ 4-[4-(2-ethoxyphenyl)-1-piperazinyl]butoxy ⁇ -3,4-dihydrocarbostyril, and 7- ⁇ 4-[4-]2-ethoxyphenyl)-1-piperazinyl]butoxy ⁇ -carbostyril.
  • Additional exemplary analogs include compounds described in U.S. Pat. No. 4,234,584 and compounds of formula I in U.S. Pat. No. 7,053,092, e.g., 7- ⁇ 4-[4-(2,3-dichlorophenyl)-1-piperazinyl]butoxyl ⁇ -3,4-dihydrocarbostyril; compounds of formula I of U.S. Pat. No.
  • 7,160,888 e.g., 7-[4-(4-naphthalen-1-yl-piperazin-1-yl)-butoxy]-3,4-dihydro-1H-[1,8] napht-hyridin-2-one, 7-[4-(4-napthalen-1-yl-piperazin-1-yl)-butoxy]-3,4-dihydro-1H-[1,8]naphth-hyridin-2-one, and 7-[4-(4-naphthalen-1-yl-piperazin-1-yl)-butoxy]-3,4-dihydro-1H-[1,8]napht-hyridin-2-one.
  • Pimozide is a benzimidazole compound with anti-psychotic activities.
  • Analogs of pimozide may include, e.g., 3-[1-[4,4-bis(4-fluorophenyl)butyl]piperidin-4-yl]-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)butyl]piperidin-4-yl]-6-fluoro-1H-benzimidazol-2-one, 1-[4,4-bis(4-fluorophenyl)butyl]-3-piperidin-4-ylbenzimidazol-2-one, 3-[2-[4-[bis(4-fluorophenyl)methyl]piperidin-1-yl]ethyl]-1H-benzimidazol-2-one, 4-[4,4-bis(4-fluorophenyl)butyl]spiro[1,4-d]azinan-1-ium-1,3
  • Clomipramine is a member of the tricyclic compounds described in U.S. Pat. No. 3,467,650. Analogs of clomipramine may be described by one the formulas (I), (II), (III), or (IV):
  • each X is, independently, H, Cl, F, Br, I, CH 3 , CF 3 , OH, OCH 3 , CH 2 CH 3 , or OCH 2 CH 3 ;
  • Y is CH 2 , O, NH, S(O) 0-2 , (CH 2 ) 3 , (CH) 2 , CH 2 O, CH 2 NH, CHN, or CH 2 S;
  • Z is C or S;
  • A is a branched or unbranched, saturated or monounsaturated hydrocarbon chain having between 3 and 6 carbons, inclusive;
  • each B is, independently, H, Cl, F, Br, I, CX 3 , CH 2 CH 3 , OCX 3 , or OCX 2 CX 3 ;
  • D is CH 2 , O, NH, or S(O) 0-2 .
  • Tricyclic compounds include tricyclic antidepressants such as clomipramine, amoxapine, 8-hydroxyamoxapine, 7-hydroxyamoxapine, loxapine (e.g., loxapine succinate, loxapine hydrochloride), 8-hydroxyloxapine, amitriptyline, doxepin, imipramine, trimipramine, desipramine, nortriptyline, and protriptyline.
  • a tricyclic compound does not need to have antidepressant activities to be within the scope of clomipramine analogs.
  • Maprotiline is a bridged-ring tetracyclic compound with psychotropic activities similar to those of the tricyclic antidepressants.
  • the chemical structure of maprotiline and analogs of maprotiline are given by structural formulae in U.S. Pat. No. 3,399,201. Additional exemplary analogs of maprotiline are described by formula I in U.S. Pat. No.
  • 4,045,560 e.g., 9-(2-morpholinylmethyl)-9,10-dihydro-9,10-methanoanthracene and 9-(4-benzyl-2-morpholinylmethyl)-9,10-dihydro-9,10-methanoanthracene; by formula I of U.S. Pat. No.
  • 4,045,580 e.g., 9-(2-hydroxy-3-methylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene, 9-(2-hydroxy-3-dimethylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene, 2-chloro-9-(2-hydroxy-3-methylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene, and 2-chloro-9-(2-hydroxy-3-dimethylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene; by formula I of U.S. Pat. No.
  • 4,224,344 e.g., ⁇ -methylaminopropyl-9,10-dihydro-9,10-methanoanthracene and ⁇ -dimethylaminopropyl-9,10-dihydro-9,10-methanoanthracene; by formula I of U.S. Pat. No. 4,358,620, e.g., 9-formyl-9,10-dihydro-9,10-methanoanthracene; by formulae I, Ia, and Ib of U.S. Pat. No.
  • 5,266,570 e.g., N-(1-[(9S,10S)-(+)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-ethoxyacetamide, (2R)—N-(1-[2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-methoxypropionamide, (2R)—N-(1-[(9S,10S)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-methoxypropionamide, N-(1-[2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-(2-pyridyl)acetamide, N-(1-[2-
  • Benzatropine (also known as benzatropine mesilate and benztropine mesylate) is a muscarinic receptor antagonist.
  • the preparation of benzotropine is described in U.S. Pat. No. 2,595,405.
  • Exemplary analogs of benztropine are described in U.S. Pat. No. 5,506,359.
  • Vinorelbine is a vinblastine-like anti-mitotic drug and is described in U.S. Pat. No. 4,307,100. Exemplary analogs of vinorelbine are described by formula I of U.S. Pat. No. 4,430,269; by formula I of U.S. Pat. No.
  • 7,238,704 e.g., 12′-bromovinblastine trifluoroacetate, 12′-iodovinblastine, 12′-bromovincristine, 12′-iodovincristine, and 12′-(hexynyl)vinblastine.
  • 12′-bromovinblastine trifluoroacetate e.g., 12′-iodovinblastine, 12′-bromovincristine, 12′-iodovincristine, and 12′-(hexynyl)vinblastine.
  • Yet other vinorelbine analogs are navelbine derivatives described in U.S. Pat. No. 5,220,016.
  • Azacitidine is a pyrimidine analog that interferes with DNA metabolism and has been used as a chemotherapeutic agent.
  • Exemplary analogs of azacitidine are 2′-beta-deoxy-6-azacytidine, 2′-deoxy-6-methyl-5-azacytidine, 2′-deoxy-N4-(2-(4-nitrophenyl)ethoxycarbonyl)-5-azacytidine, 5′-azacytidine 5′-triphosphate, 5,6-dihydro-5-azacytidine, 5-aza-2′-deoxycytidine-5′-monophosphate, 5-aza-2′-deoxycytidine-5′-triphosphate, 6-azacytidine, decitabine, therapeuticrabine, N(4),N(4)-dimethyl-5-azacytidine, N(4)-methyl-5-azacytidine, 2′-beta-deoxy-6-azacytidine, 2′-deoxy-6-methyl-5-azacytidine, 2′-deoxy-N4-(2-(4-nitrophenyl)ethoxycarbonyl)-5-azacyt
  • Dasatinib is a tyrosine kinase inhibitor used for the treatment of certain cancers.
  • Exemplary analogs of dasatinib are described by formula I in U.S. Pat. No. 7,091,223, e.g., N′-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-[[2-(4-morpholinyl)ethyl]amin-o]-4-pyrimidinyl]amino]-5-thiazolecarboxamide, and by formula I of U.S. Pat. No. 7,091,223, e.g., N-(2-chloro-6-methylphenyl)-2-[(4,6-dimethyl-2-pyridinyl)amino]-5-thiazol-ecarboxamide.
  • Simvastatin is an HMG-CoA reductase inhibitor and anti-hypercholesterolemia drug described in European Pat. No. EP0033538 and U.S. Pat. No. 4,444,784.
  • An analog of simvastatin may be a structurally related compound, a compound that inhibits HMG-CoA reductase, or both.
  • Specific examples of simvastatin analogs are lovastatin (GB2046737A), mevastatin (U.S. Pat. No.
  • L669262 (described in EP-00331250 and EP-00408806), NCX6560 (WO/2004/105754), NR-300s, P882222, P882284, PD134965, PD135022 (CAS 122548-95-2), rawsonol (CAS 125111-69-5), RBx-10558 (WO/2004/05250), RP61969, 52467, 52468, SC37111, SC45355, SQ33600 (Sliskovic et al., Drug News and Perspectives, 5:517-533), SR12813 (U.S. Pat. No.
  • Teicoplanin is a complex antiobiotic agent containing several compounds, five major (named teicoplanin A 2 -1 through A 2 -5) and four minor (named teicoplanin R S -1 through R S -4).
  • teicoplanin encompasses each of these major and minor compounds. All teicoplanins share a same glycopeptide core, termed teicoplanin A3-1, featuring a fused ring structure to which two carbohydrates (mannose and N-acetylglucosamine) are attached.
  • the major and minor components also contain a third carbohydrate moiety and differ by the length and conformation of a side chain attached to it.
  • Teicoplanin analogs include compounds structurally related to the major and minor teicoplanin constituents and products of teicoplanin hydrolysis, e.g., L 17054 and L 17392.
  • Other analogs of teicoplanin include L 17046, L 17932 (U.S. Pat. No. 5,594,102), 4,7-decadienoyl-teicoplanin, 4-hydroxydecanoyl-teicoplanin, MDL 62873, aglycone forms of any teicoplanin compound or teicoplanin derivative, compounds of formula I of U.S. Pat. No.
  • demannosyl teichoplanin derivatives e.g., compounds of formula I of U.S. Pat. No. 5,064,811; compounds described in U.S. Pat. No. 5,085,990; de-(acetylglucosaminyl)-di(dehydro)-deoxy teicoplanin derivatives, e.g., compounds of formula I of U.S. Pat. No. 4,789,661; and substituted alkymides of teicoplanin according to formula I of U.S. Pat. No. 5,198,418.
  • Structural analogs of terconazole include any stereochemical isomers thereof. Other structural analogs are described in U.S. Pat. Nos. 3,575,999, 3,936,470, 4,223,036, 4,358,449 (see, for example, Examples I-LXXII), in Belgian Pat. No. 935,579, and in the PCT Publication No. WO00/76316.
  • Q is —CH— or —N—;
  • Ar is optionally substituted phenyl, wherein a substituted phenyl has 1, 2, or 3 substituents that are, independently, halogen, C 1-6 alkyl, or C 1-6 alkoxy;
  • A is —NCS, —NR 2 R 3 , —NHC(X)—(Y) m —R 4 , or
  • Exemplary, non-limiting structural analogs of terconazle are 4-[2-(3-chlorophenyl)-2-(1H-imidazol-1-ylmethyl)-1,3-dioxolan-4-ylmethoxy]-N-ethylbenzenamine, 4-[2-(4-bromophenyl)-2-(1H-imidazol-1-ylmethyl)-1,3-dioxolan-4-ylmethoxy]-N-ethylbenzenamine, N-ethyl-4-[2-(1H-imidazol-1-ylmethyl)-2-(3-methylphenyl)-1,3-dioxolan-4-ylmethoxy]benzenamine, N-ethyl-4-[2-(1H-imidazol-1-ylmethyl)-2-(4-methoxyphenyl)-1,3-dioxolan-4-yl methoxy]benzenamine, 4-[2-(2,4-
  • Hycanthone is a thioxanthenone schistosomicide described in U.S. Pat. Nos. 3,294,803 and 3,312,598. Analogs of hycanthone include other compounds of formula I in U.S. Pat. No.
  • Atovaquone is an anti-microbial napthalene compound described in U.S. Pat. No. 5,053,432.
  • Analogs of atovaquone include compounds described by formula I of U.S. Pat. No. 5,053,432, e.g., 2-hydroxy-3-(4-methoxycyclohexyl)-1,4-naphthoquinone, 2-hydroxy-3-[4-(1-methoxy-1-methylethyl)cyclohexyl]-1,4-naphthoquinone, 2-(4-benzyloxycyclohexyl)-3-hydroxy-1,4-naphthoquinone, 2,4-(3,4-dichlorophenyl)cyclohexyl-3-hydroxy-1,4-naphthoquinone, 2-[4-(3,4-dimethylphenyl)cyclohexyl]-1,4-naphthoquinone, 2-(4-fluorocyclo
  • exemplary analogs include compounds described by formula II in U.S. Pat. No. 5,310,762, e.g., 2-(4-t-butylcyclohexyl)-3-hydroxy-1,4-naphthoquinone; by formula II in U.S. Pat. No. 5,466,711, e.g., 2-acetoxy-3-[trans-4-(4-chlorophenyl)cyclohexyl]-1,4-naphthoquinone; by formula II in U.S. Pat. No.
  • Quinacrine is an acridine derivative described in U.S. Pat. No. 2,113,357.
  • Analogs of quinacrine include, for example, chloroquine, hydroxychloroquine, amodiaquine, mefloquine, primaquine, quinine, chemiochin (4-N-(6-chloro-2-methoxyacridin-9-yl)-1-N,1-N-diethylpentane-1,4-diamine dihydrochloride), atabrine hydrochloride (4-[(6-chloro-2-methoxyacridin-9-yl)azaniumyl]pentyl-diethylazanium dichloride), NSC240788 (2-N-(6-chloro-2-methoxyacridin-9-yl)-1-N,1-N-diethylpropane-1,2-diamine), NSC 8591 (4-[(6-chloro-2-methoxyacridin
  • Efavirenz is a non-nucleoside reverse transcriptase inhibitor described in U.S. Pat. No. 5,519,021.
  • Analogs of efavirenz include compounds described by formula I or II of U.S. Pat. No. 5,519,021, e.g., ( ⁇ )6-chloro-4-cyclopropyl-ethynyl-4-frifluoromethyl-1-4-dihydro-2H-3,1-benzoxazin-2-one, ( ⁇ ) 6-chloro-4-phenylethynyl-4-trifluoromethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/ ⁇ ) 4-(1-chloro-1,1-difluoromethyl)-4-(2-phenylethynyl)-6-chloro-1,4-dihydro-2H-3,1-benzoxazin-2-one, or (+/ ⁇ ) 4-(2-[dimethylamino
  • K-strophanthin (cymarine) is a cardiac glycoside derived from Strophanthus plant species. Analogs of strophanthins include other cardiac glycosides and their aglycone derivatives. Cardiac glycosides are derived from the cyclopentanoperhydro-phenanthrene nucleus of steroid compounds and are characterized by lactone rings joined by an ether linkage to one or more sugar residues. Aglycone derivatives of cardiac glycosides lack the carbohydrates characteristic of the cardiac glycosides.
  • Exemplary analogs of strophanthins include, but are not limited to, G-strophanthin (ouabain), lanatoside A, desacetyllanatoside A, actyl digitoxin, digitoxin, lanatoside c, desacetyllanatoside c, digoxin, strophanthoside, scillaren A, proscillaridin A, uzarin, digitoxose, gitoxin, strophanthidine-3b-digitoxoside, strophanthidin aL-rhamnopyranoside, strophanthidol, oleandrin, acovenoside A, strophanthidine digilanobioside, strophanthidin-D-cymaroside, digitoxigenin-L-rhamnoside digitoxigenin theretoside, and the like.
  • G-strophanthin ouabain
  • lanatoside A desacetyll
  • Aglycones include, but are not limited to, strophanthidin, digitoxigenin, uzarigenin, digoxigenin, digoxigenin 3,12-diacetate, gitoxigenin, gitoxigenin 3-acetate, gitoxigenin 3,16-diacetate, 16-acetyl gitoxigenin, acetyl strophanthidin, ouabagenin, 3-epidigoxigenin, and the like.
  • Mycophenolate mofetil is an immunosuppressant described in U.S. Pat. No. 4,753,935. Analogs of mycophenolate mofetil are described by formula A in U.S. Pat. No. 4,753,935; by formulae I and II in U.S. Pat. No.
  • agents used in any of the combinations described herein may be covalently attached to one another to form a conjugate of formula I.
  • (A) is a Compound A and (B) is Compound B of a pair of agents from e.g., Table 1, and L is a covalent linker that tethers (A) to (B).
  • Conjugates of the invention can be administered to a subject by any route and for the treatment of a filovirus-mediated disease (e.g., those described herein).
  • the conjugates of the invention can be prodrugs, releasing drug (A) and drug (B) upon, for example, cleavage of the conjugate by intracellular and extracellular enzymes (e.g., amidases, esterases, and phosphatases).
  • the conjugates of the invention can also be designed to largely remain intact in vivo, resisting cleavage by intracellular and extracellular enzymes. The degradation of the conjugate in vivo can be controlled by the design of linker (L) and the covalent bonds formed with drug (A) and drug (B) during the synthesis of the conjugate.
  • Conjugates can be prepared using techniques familiar to those skilled in the art.
  • the conjugates can be prepared using the methods disclosed in G. Hermanson, Bioconjugate Techniques , Academic Press, Inc., 1996.
  • the synthesis of conjugates may involve the selective protection and deprotection of alcohols, amines, ketones, sulfhydryls or carboxyl functional groups of drug (A), the linker, and/or drug (B).
  • commonly used protecting groups for amines include carbamates, such as tert-butyl, benzyl, 2,2,2-trichloroethyl, 2-trimethylsilylethyl, 9-fluorenylmethyl, allyl, and m-nitrophenyl.
  • amides such as formamides, acetamides, trifluoroacetamides, sulfonamides, trifluoromethanesulfonyl amides, trimethylsilylethanesulfonamides, and tert-butylsulfonyl amides.
  • protecting groups for carboxyls include esters, such as methyl, ethyl, tert-butyl, 9-fluorenylmethyl, 2-(trimethylsilyl)ethoxy methyl, benzyl, diphenylmethyl, O-nitrobenzyl, ortho-esters, and halo-esters.
  • Examples of commonly used protecting groups for alcohols include ethers, such as methyl, methoxymethyl, methoxyethoxymethyl, methylthiomethyl, benzyloxymethyl, tetrahydropyranyl, ethoxyethyl, benzyl, 2-napthylmethyl, O-nitrobenzyl, P-nitrobenzyl, P-methoxybenzyl, 9-phenylxanthyl, trityl (including methoxy-trityls), and silyl ethers.
  • Examples of commonly used protecting groups for sulfhydryls include many of the same protecting groups used for hydroxyls.
  • sulfhydryls can be protected in a reduced form (e.g., as disulfides) or an oxidized form (e.g., as sulfonic acids, sulfonic esters, or sulfonic amides).
  • Protecting groups can be chosen such that selective conditions (e.g., acidic conditions, basic conditions, catalysis by a nucleophile, catalysis by a lewis acid, or hydrogenation) are required to remove each, exclusive of other protecting groups in a molecule.
  • the conditions required for the addition of protecting groups to amine, alcohol, sulfhydryl, and carboxyl functionalities and the conditions required for their removal are provided in detail in T. W. Green and P. G. M. Wuts, Protective Groups in Organic Synthesis (2 nd Ed.), John Wiley & Sons, 1991 and P. J. Kocienski, Protecting Groups , Georg Thieme Verlag, 1994. Additional synthetic details are provided below.
  • the linker component of the invention is, at its simplest, a bond between drug (A) and drug (B), but typically provides a linear, cyclic, or branched molecular skeleton having pendant groups covalently linking drug (A) to drug (B).
  • linking of drug (A) to drug (B) is achieved by covalent means, involving bond formation with one or more functional groups located on drug (A) and drug (B).
  • functional groups located on drug (A) and drug (B).
  • chemically reactive functional groups include, without limitation, amino, hydroxyl, sulfhydryl, carboxyl, carbonyl, carbohydrate groups, vicinal diols, thioethers, 2-aminoalcohols, 2-aminothiols, guanidinyl, imidazolyl, and phenolic groups.
  • the covalent linking of drug (A) and drug (B) may be effected using a linker that contains reactive moieties capable of reaction with such functional groups present in drug (A) and drug (B).
  • a linker that contains reactive moieties capable of reaction with such functional groups present in drug (A) and drug (B).
  • an amine group of drug (A) may react with a carboxyl group of the linker, or an activated derivative thereof, resulting in the formation of an amide linking the two.
  • N-Maleimide derivatives are also considered selective towards sulfhydryl groups, but may additionally be useful in coupling to amino groups under certain conditions.
  • Reagents such as 2-iminothiolane (Traut et al., Biochemistry 12:3266 (1973)), which introduce a thiol group through conversion of an amino group, may be considered as sulfhydryl reagents if linking occurs through the formation of disulfide bridges.
  • reactive moieties capable of reaction with amino groups include, for example, alkylating and acylating agents.
  • Representative alkylating agents include:
  • N-maleimide derivatives which may react with amino groups either through a Michael type reaction or through acylation by addition to the ring carbonyl group, for example, as described by Smyth et al., J. Am. Chem. Soc. 82:4600 (1960) and Biochem. J. 91:589 (1964);
  • epoxide derivatives such as epichlorohydrin and bisoxiranes, which may react with amino, sulfhydryl, or phenolic hydroxyl groups;
  • Representative amino-reactive acylating agents include:
  • active esters such as nitrophenylesters or N-hydroxysuccinimidyl esters
  • acylazides e.g., wherein the azide group is generated from a preformed hydrazide derivative using sodium nitrite, as described by Wetz et al., Anal. Biochem. 58:347 (1974); and
  • Aldehydes and ketones may be reacted with amines to form Schiff's bases, which may advantageously be stabilized through reductive amination.
  • Alkoxylamino moieties readily react with ketones and aldehydes to produce stable alkoxamines, for example, as described by Webb et al., in Bioconjugate Chem. 1:96 (1990).
  • reactive moieties capable of reaction with carboxyl groups include diazo compounds such as diazoacetate esters and diazoacetamides, which react with high specificity to generate ester groups, for example, as described by Herriot, Adv. Protein Chem. 3:169 (1947).
  • Carboxyl modifying reagents such as carbodiimides, which react through O-acylurea formation followed by amide bond formation, may also be employed.
  • functional groups in drug (A) and/or drug (B) may, if desired, be converted to other functional groups prior to reaction, for example, to confer additional reactivity or selectivity.
  • methods useful for this purpose include conversion of amines to carboxyls using reagents such as dicarboxylic anhydrides; conversion of amines to thiols using reagents such as N-acetylhomocysteine thiolactone, S-acetylmercaptosuccinic anhydride, 2-iminothiolane, or thiol-containing succinimidyl derivatives; conversion of thiols to carboxyls using reagents such as ⁇ -haloacetates; conversion of thiols to amines using reagents such as ethylenimine or 2-bromoethylamine; conversion of carboxyls to amines using reagents such as carbodiimides followed by diamines; and conversion of alcohols to
  • So-called zero-length linkers involving direct covalent joining of a reactive chemical group of drug (A) with a reactive chemical group of drug (B) without introducing additional linking material may, if desired, be used in accordance with the invention.
  • the linker will include two or more reactive moieties, as described above, connected by a spacer element.
  • the presence of such a spacer permits bifunctional linkers to react with specific functional groups within drug (A) and drug (B), resulting in a covalent linkage between the two.
  • the reactive moieties in a linker may be the same (homobifunctional linker) or different (heterobifunctional linker, or, where several dissimilar reactive moieties are present, heteromultifunctional linker), providing a diversity of potential reagents that may bring about covalent attachment between drug (A) and drug (B).
  • Spacer elements in the linker typically consist of linear or branched chains and may include a C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-10 heteroalkyl.
  • linker is described by formula (II):
  • G 1 is a bond between drug (A) and the linker;
  • G 2 is a bond between the linker and drug (B);
  • Z 1 , Z 2 , Z 3 , and Z 4 each, independently, is selected from O, S, and NR 31 ;
  • R 31 is hydrogen, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 2-6 heterocyclyl, C 6-12 aryl, C 7-14 alkaryl, C 3-10 alkheterocyclyl, or C 1-7 heteroalkyl;
  • Y 1 and Y 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl;
  • o, p, s, t, u, and v are each, independently, 0 or 1;
  • R 30 is a C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 2-6 heterocyclyl, C 6-12 ary
  • homobifunctional linkers useful in the preparation of conjugates of the invention include, without limitation, diamines and diols selected from ethylenediamine, propylenediamine and hexamethylenediamine, ethylene glycol, diethylene glycol, propylene glycol, 1,4-butanediol, 1,6-hexanediol, cyclohexanediol, and polycaprolactone diol.
  • compositions, methods, and kits of the invention can include formulation(s) of compound(s) that, upon administration to a subject, result in a concentration of the compound(s) that treats a filovirus-mediated disease.
  • the compound(s) may be contained in any appropriate amount in any suitable carrier substance, and are generally present in an amount of 1-95% by weight of the total weight of the composition.
  • the composition may be provided in a dosage form that is suitable for the oral, parenteral (e.g., intravenously or intramuscularly), rectal, dermatological, cutaneous, nasal, vaginal, inhalant, skin (patch), ocular, intrathecal, or intracranial administration route.
  • the composition may be in the form of, e.g., tablets, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectables, implants, sprays, or aerosols.
  • the pharmaceutical compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy, 20th edition, 2000, ed. A. R. Gennaro, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology , eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York).
  • compositions according to the invention or used in the methods of the invention may be formulated to release the active compound immediately upon administration or at any predetermined time or time period after administration.
  • the latter types of compositions are generally known as controlled release formulations, which include (i) formulations that create substantially constant concentrations of the agent(s) of the invention within the body over an extended period of time; (ii) formulations that after a predetermined lag time create substantially constant concentrations of the agent(s) of the invention within the body over an extended period of time; (iii) formulations that sustain the agent(s) action during a predetermined time period by maintaining a relatively constant, effective level of the agent(s) in the body with concomitant minimization of undesirable side effects associated with fluctuations in the plasma level of the agent(s) (sawtooth kinetic pattern); (iv) formulations that localize action of agent(s), e.g., spatial placement of a controlled release composition adjacent to or in the diseased tissue or organ; (v) formulations that achieve convenience of dosing, e.
  • controlled release is obtained by appropriate selection of various formulation parameters and ingredients, including, e.g., various types of controlled release compositions and coatings.
  • the compound(s) are formulated with appropriate excipients into a pharmaceutical composition that, upon administration, releases the compound(s) in a controlled manner. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, molecular complexes, microspheres, nanoparticles, patches, and liposomes.
  • a first agent is delivered orally, and a second agent is delivered intravenously.
  • the dosage of a compound or a combination of compounds depends on several factors, including: the administration method, the type of disease to be treated, the severity of the infection, whether administration first occurs at an early or late stage of infection, and the age, weight, and health of the patient to be treated.
  • the recommended dosage for the anti-viral agent can be less than or equal to the recommended dose as given in the Physician's Desk Reference, 60 th Edition (2006).
  • the compound(s) in question may be administered orally in the form of tablets, capsules, elixirs or syrups, or rectally in the form of suppositories.
  • Parenteral administration of a compound is suitably performed, for example, in the form of saline solutions or with the compound(s) incorporated into liposomes.
  • a solubilizer such as ethanol can be applied.
  • the correct dosage of a compound can be determined by examining the efficacy of the compound in viral replication assays, as well as its toxicity in humans.
  • An antiviral agent is usually given by the same route of administration that is known to be effective for delivering it as a monotherapy.
  • an agent of Table 2 or Table 3 is dosed in amounts and frequencies equivalent to or less than those that result in its effective monotherapeutic use.
  • the compounds of the invention may be employed in mechanistic assays to determine whether other combinations, or single agents, are as effective as the combinations of the invention in inhibiting a viral disease (e.g., those described herein) using assays generally known in the art.
  • candidate compounds may be tested, alone or in combination (e.g., with an agent that inhibits viral replication, such as those described herein) and applied to cells (e.g., hepatic cells such as HepG2, kidney epithelial cells such as 293T, macrophages such as THP-1, or isolated primary cells). After a suitable time, viral replication or load of these cells is examined. A decrease in viral replication or viral load identifies a candidate compound or combination of agents as an effective agent for treating a viral disease.
  • the agents of the invention are also useful tools in elucidating mechanistic information about the biological pathways involved in viral diseases. Such information can lead to the development of new combinations or single agents for treating, preventing, or reducing a viral disease.
  • Methods known in the art to determine biological pathways can be used to determine the pathway, or network of pathways affected by contacting cells (e.g., hepatic cells) infected with a virus with the compounds of the invention. Such methods can include, analyzing cellular constituents that are expressed or repressed after contact with the compounds of the invention as compared to untreated, positive or negative control compounds, and/or new single agents and combinations, or analyzing some other activity of the cell or virus such as an enzymatic activity, nutrient uptake, and proliferation.
  • Cellular components analyzed can include gene transcripts, and protein expression. Suitable methods can include standard biochemistry techniques, radiolabeling the compounds of the invention (e.g., 14 C or 3 H labeling), and observing the compounds binding to proteins, e.g., using 2D gels, gene expression profiling. Once identified, such compounds can be used in in vivo models (e.g., knockout or transgenic mice) to further validate the tool or develop new agents or strategies to treat viral disease.
  • in vivo models e.g., knockout or transgenic mice
  • Peptides, peptide mimetics, and peptide fragments are suitable for use in the methods of the invention.
  • exemplary inhibitors include compounds that reduce the amount of a target protein or RNA levels (e.g., antisense compounds, dsRNA, ribozymes) and compounds that compete with viral reproduction machinery (e.g., dominant negative proteins or polynucleotides encoding the same).
  • RNA secondary structure folding program such as MFOLD (M. Zuker, D. H. Mathews & D. H. Turner, Algorithms and Thermodynamics for RNA Secondary Structure Prediction: A Practical Guide . In: RNA Biochemistry and Biotechnology , J. Barciszewski & B. F. C.
  • Sub-optimal folds with a free energy value within 5% of the predicted most stable fold of the mRNA are predicted using a window of 200 bases within which a residue can find a complimentary base to form a base pair bond. Open regions that do not form a base pair are summed together with each suboptimal fold and areas that are predicted as open are considered more accessible to the binding to antisense nucleobase oligomers.
  • Other methods for antisense design are described, for example, in U.S. Pat. No. 6,472,521 , Antisense Nucleic Acid Drug Dev. 7:439-444 (1997), Nucleic Acids Res. 28:2597-2604 (2000), and Nucleic Acids Res. 31:4989-4994 (2003).
  • RNA interference employing, e.g., a double stranded RNA (dsRNA) or small interfering RNA (siRNA) directed to the signaling molecule in question (see, e.g., Miyamoto et al., Prog. Cell Cycle Res. 5:349-360 (2003); U.S. Pat. Application Publication No. 20030157030).
  • dsRNA double stranded RNA
  • siRNA small interfering RNA
  • Methods for designing such interfering RNAs are known in the art. For example, software for designing interfering RNA is available from Oligoengine (Seattle, Wash.).
  • Ebolavirus expressing the green fluorescent protein (GFP) has been used to develop a high throughput assay for drug discovery (Towner et al., Virology, 332: 20 (2005)).
  • the genetically engineered virus has the unique property of making infected cells glow fluorescent green.
  • the green signal can be detected by fluorescence microscopy, flow cytometry, or fluorometry, and other types of fluorescence detection systems adapted for high-throughput format.
  • the plated cells are incubated cells at 37° C., 5% CO 2 until they achieve a stationary growth phase.
  • 50 ⁇ l of pre-diluted compounds are added at a 4 ⁇ concentration to each well to achieve the desired final concentration.
  • 50 ⁇ l of GFP-EBOV multiplicity of infection of 0.01 is added to cells. Plates are centrifuged at 2000 RPM, 5 minutes following the addition of EBOV. Cells are incubated an additional 48 hours at 37° C., 5% CO 2 , and the plates are then removed from the incubator and the amount of GPF in each well is determined using a spectrofluorometer from Molecular Devices (excitation: 485 nm, emission: 515 nm, cutoff: 495 nm). Antiviral activity is identified by the inhibition of GFP compared to untreated control cells.
  • a counter screen is run in tandem using uninfected Vero or HepG2 cells. Seed cells on a 96-well plate as described above. Cells are mock infected and treated with compounds the following day. After a 48 hour incubation at 37° C., 5% CO 2 , the amount of ATP is determined using Promega Cell Titer-Glo luminescent Cell viability kit. This assay provides a quantitative measure of the levels of ATP in the cell cultures in each well, where higher levels of ATP correlate with greater cellular viability. Thus, a compound with antiviral activity is expected to inhibit the levels of GPF measured without any or minimal effect on the ATP levels measured by the Cell Titer-Glo assay.
  • IC50 Agents thus identified (IC50) are listed in Table 6.
  • the extent of inhibition is represented as maximum fraction inhibition (Max Effect), where a value of 1.0 is indicative of complete inhibition, and 0 is indicative of no effect. As available, 50% inhibitory concentrations of the agents are provided.
  • the top 30 agents identified in the initial screen as outlined in Table 6 were re-tested for inhibitory activity in both Vero and HepG2 cells. The results of this analysis are in provided in Table 7. These agents effectively inhibited replication of the Ebola replicon at low to sub micromolar concentrations.
  • the screening method described in Example 1 was used to identify combinations of agents that synergistically inhibit viral replication.
  • the magnitude of the synergy score indicates the strength of the synergistic interaction.
  • synergy scores >1.0 indicate a synergistic effect of Agent 1 and Agent 2
  • synergy scores between 0.3 and 1.0 indicate an additive effect of Agent 1 and Agent 2.
  • CI values in the range of 0.5-0.7 are typical for in vitro measurements of current clinical combinations (Greco et al., Pharmacol. Rev., 47:331-85 (1995)).
  • the choice of effect levels for the CI calculations were selected by using an algorithm that finds the observed I data with the largest I data ⁇ I Loewe , excluding those points with I data exceeding the lesser single agent's I max . This exclusion is applied to ensure that the CI reflects the best synergy at levels covered by both single agents.
  • the chosen iso-level is then floored at a 20% inhibition level, beneath which inhibitions often are unreliable, and rounded to the nearest 5%.
  • the synergy scores and CI for the agent pairs identified in our screen are listed in Table 8.
  • a mouse model of Ebola infection developed by Bray et al. ( J. Infect. Dis., 179:S248-58 (1999)) was used to show in vivo efficacy of single agents.
  • the animals were treated with the single agents in Table 9 at the indicated dose and frequency for a total of 10 days. Animals were monitored for survival for a total of 28 days following infection and the mean time to death determined for all cases where there was a sufficient number of non-survivors. In cases where all animals survived, the mean time to death was not determined and is greater than 28 days.

Abstract

The invention features compositions, methods, and kits useful for the treatment of filovirus-mediated diseases, e.g., hemorrhagic fever caused by Ebola virus, in an animal.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. Ser. No. 12/710,203, filed Feb. 22, 2010, which claims benefit from U.S. Provisional Application No. 61/154,279, filed Feb. 20, 2009, which is hereby incorporated by reference.
  • BACKGROUND OF THE INVENTION
  • The invention relates to the treatment of diseases caused by a filovirus.
  • Diseases caused by viruses are major health problems worldwide. Filoviruses are particularly deadly viruses that cause severe hemorrhagic fever in humans and non-human primates. Filoviruses are sporadically transmitted to humans from a natural reservoir species thought to be fruit bats, after which the virus can easily spread from the human having the infection to a caregiver through bodily fluids. Infection with a filovirus of the Ebola or Marburg genus causes death in 25% to 90% of human victims, often within 10 days from the first appearance of symptoms.
  • No known anti-viral therapies are effective for treating filovirus-mediated diseases. Filoviruses thus pose serious unmet health and bioterrorism concerns. Given the lack of safe and efficacious therapies for filovirus-mediated diseases, there is a need for such therapies.
  • SUMMARY OF THE INVENTION
  • Based on the results of our screen identifying compounds and combinations of compounds having inhibitory activity against a model filovirus, the present invention features compositions, methods, and kits for the treatment of filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus.
  • Accordingly, in a first aspect, the invention features a method for treating a patient with a filovirus-mediated disease, e.g., a disease caused by an Ebola virus or Marburg virus. The method includes administering to the patient a first agent selected from the agents of Table 2, or an analog thereof, in an amount that is effective to treat the patient. Preferably, the first agent is selected from the agents of Table 1. More preferably, the first agent is clomiphene, toremifene, astemizole, bepridil, clomopramine, lomerizine, sertraline, or an analog thereof. In another embodiment, the method further includes administering a second agent selected from the agents of Table 1. Preferably, the first and second agents are both selected from Table 1. More preferably, the first and second agents are selected from the agent pairs of Table 3.
  • TABLE 1
    Azacitidine Thioridazine Piperacetazine
    Clomiphene Prochlorperazine Fluphenazine
    Toremifene Sertraline Aripiprazole
    Lomerizine Paroxetine Maprotiline
    Bepridil Clomipramine Dasatinib
    Strophanthin Pimozide Vinorelbine
    Atovaquone Flupentixol Teicoplanin
    Mycophenolate Mofetil Clemastine Hycanthone
    Terconazole Astemizole Quinacrine
    Simvastatin Benztropine Efavirenz
  • TABLE 2
    Cetrimide Bafilomycin A1 Homochlorcyclizine
    Pentamidine 2-Methoxy-antimycin A3 Desloratadine
    Hexachlorophene Oligomycin Flunarizine
    Emetine Antimycin A Loratadine
    Puromycin Rotenone Doxylamine
    Thiostrepton FR122047 Propoxyphene
    Gramicidin Fenoprofen Calcium Benztropine
    Chlorhexidine Perhexiline Maleate Dicyclomine
    Teicoplanin PDMP Piperacetazine
    Cephapirin Licochalcone-A Vanoxerine
    Pyrithione Zinc Tiratricol Pergolide
    Haloprogin CAPE Acetophenazine
    Ciclopirox Amlodipine Bromocriptine
    hycanthone Diphenyleneiodonium Fluphenazine
    Niclosamide Terconazole N-(4-Aminobenzoyl)-L-glutamic acid
    Efavirenz Sulconazole Aripiprazole
    Ritonavir Tioconazole Drotaverine
    Chromomycin A3 Oxiconazole Maprotiline
    Azacitidine Simvastatin CKI7
    (−)-Arctigenin Cerivastatin Triptolide
    Danazol Metergoline NSC 625987
    Bicalutamide Thioridazine Bay 41-2272
    Hydroxyprogesterone Thiethylperazine Alverine
    Equilin Cyproheptadine Tannic Acid
    Clomiphene Prochlorperazine IMD-0354
    Quinestrol Triflupromazine Arbidol
    Tamoxifen Paroxetine Andrographis
    Raloxifene Sertraline Pyrvinium
    Tamoxifen Clomipramine Deguelin
    Toremifene Fenretinide Dasatinib
    PGG Ciclesonide 5-Iodotubercidin
    Diethylstilbestrol Brefeldin A SP 600125
    Tibolone PGG (1,2,3,4,6-b-O-Pentagalloyl glucose) Carbobenzoxy-valinyl-phenylalaninal
    Lomerizine BML-248 Cepharanthine
    Maduramicin Leupeptin Sangivamycin
    Bepridil Tunicamycin PKR inhibitor
    Loperamide MG115 Okadaic Acid
    6-Azauridine MG-132 Sorafenib
    Strophanthin Epoxomicin Sodium Vanadate
    Beta Escin Anisomycin Nocodazole
    Auranofin CGS 15943 Mebendazole
    Calcimycin A23187 Pimozide TN-16
    Edetate Guanethidine Fenbendazole
    Octyl Gallate Flupentixol Podofilox
    Magnesium Sulfate Perphenazine Triclabendazole
    Rescinnamine Trifluoperazine Oxibendazole
    Cilastatin EGFR Inhibitor Vinorelbine
    Bromelain Sunitinib Thapsigargin
    Quinacrine Chlorcyclizine 2,6-Divanillylidenecyclohexanone
    Methdilazine Astemizole 1,5′-Bis(2-Nitrophenoxy)pentane
    L-Asparagine Clemastine Desmethyl Sertraline
    Chondroitin Terfenadine Podophyllum
    Atovaquone Chlorphenoxamine Saponin
    Mycophenolate Mofetil Oxatomide Nonoxynol-9
    Mycophenolic Acid Azelastine Domiphen Bromide
    Flucytosine Clemastine Sodium Bicarbonate
    Carbonyl cyanide 4- Cycloheximide Latrunculin B
    (trifluoromethoxy)phenylhydrazone
  • TABLE 3
    Lomerizine and Vinolrelbine Dasatinib and Lomerizine
    Pimozide and Vinolrelbine Aripiprazole and and Piperacetazine
    Aripiprazole and Vinolrelbine Pimozide and Thioridazine
    Aripiprazole and Dasatinib Aripiprazole and Astemizole
    Clemastine and Sertraline Bepridil and Clomiphene
    Bepridil and Toremifene Clomiphene and Sertraline
    Toremifene and Sertraline Sertraline and Vinorelbine
    Aripiprazole and Paroxetine
  • The invention also features a method for treating a patient having a filovirus-mediated disease, e.g., a disease caused by Ebola virus or Marburg virus, the method including administering to the patient: a first agent that is an inhibitor of estrogen receptor α and a second agent that is an inhibitor of histamine receptor 1 in amounts that together are effective to treat the patient. The first agent is preferably clomiphene, tamoxifen, raloxifene, toremifene, diethylstilbestrol, or an analog thereof. More preferably, the first agent is clomiphene, tamoxifen, toremifene, or an analog thereof. The second agent is preferably chlorcyclizine, astemizole, clemastine, terfenadine, chlorphenoxamine, oxatomide, azelastine, methdilazine, homochlorcyclizine, desloratadine, flunarizine, loratadine, doxylamine, or an analog thereof. More preferably, the second agent is astemizole or an analog thereof.
  • The invention also features a method for treating a patient having a filovirus-mediated disease, e.g., a disease caused by Ebola virus or Marburg virus, the method including administering to the patient: a first agent that is an inhibitor of estrogen receptor α and a second agent that is sertraline or an analog thereof in amounts that together are effective to treat the patient. The first agent is preferably clomiphene, tamoxifen, raloxifene, toremifene, diethylstilbestrol, or an analog thereof. More preferably, the first agent is clomiphene, tamoxifen, toremifene, or an analog thereof. The second agent is preferably sertraline, paroxetine, or UK-416244.
  • When the methods includes administering to the patient a pair of active agents, the agents may be administered within within 28, 21, 14, 10, 7, 5, 4, 3, 2, or 1 days; within 24, 12, 6, 3, 2, or 1 hours; or substantially simultaneously. The methods of the invention may include administering one or more agents to the patient by oral, systemic, parenteral, topical (e.g., ophthalmic, dermatologic), intravenous, inhalational, or intramuscular administration.
  • In certain embodiments of any of the above methods (e.g., a method including administration of an inhibitor of estrogen receptor α), the patient being treated has not been diagnosed with or does not suffer from breast cancer, osteoporosis, prostate hyperplasia, metabolic syndrome X, male infertility, testosterone deficiency, hypogonadism, non-insulin-dependent diabetes, infertility due to an ovulatory disorder, infertility due to polycystic ovary syndrome, gynaecomastia, or hormone deficiency. In certain other embodiments of any of the above methods (e.g., a method including administration of an inhibitor of histamine receptor 1), the patient has not been diagnosed with or does not suffer from urticaria, seasonal allergic rhinitis, allergy or excessive itching, or cancer (e.g., cancer of the lung, pancreatic, ovary, breast, prostate, colon, brain, or skin). In other certain embodiments of any of the methods (e.g., a method including administration of a selective serotonin reuptake inhibitor or a phenothiazine), the patient being treated has not been diagnosed with or does not suffer from a major depressive disorder, an obsessive-compulsive disorder, an anxiety disorder, or a psychotic disorder (e.g., schizophrenia). In yet other certain embodiments of any of the above methods, the patient being treated has not been diagnosed with or does not suffer from migraine, cardiac arrhythmia, hypertension, pneumocystis pneumonia, toxoplasmosis, malaria, organ transplant rejection, fungal infection, hypolipidemia, hypercholesterolemia, chronic myelogenous leukemia, cancer (e.g., cancer of the lung, pancreatic, ovary, breast, prostate, colon, brain, or skin), bacterial infection, schistosomal infection, malaria, HIV, osteoporosis, gynaecomastia.
  • In another aspect, the invention features a composition including two or more agents selected from the agents of Table 1. Preferably, the two or more agents are present in amounts that, when administered together to a patient with a filovirus-mediated disease (e.g., a disease caused by Ebola virus or Marburg virus), are effective to treat the patient. More preferably, the first and second agents are selected from the agent pairs of Table 3. In another embodiment, the composition consists of active ingredients and excipients, and the active ingredients consist of said two agents selected from the agents of Table 1.
  • The invention also features a composition that includes a first agent that is an inhibitor of estrogen receptor α and a second agent that is an inhibitor of histamine receptor 1. Preferably, the first and second agents are present in amounts that, when administered together to a patient with a filovirus-mediated disease (e.g., a disease caused by Ebola virus or Marburg virus), are effective to treat the patient. The first agent is preferably clomiphene, tamoxifen, raloxifene, toremifene, diethylstilbestrol, or an analog thereof. More preferably, the first agent is clomiphene, tamoxifen, toremifene, or an analog thereof. The second agent is preferably chlorcyclizine, astemizole, clemastine, terfenadine, chlorphenoxamine, oxatomide, azelastine, methdilazine, homochlorcyclizine, desloratadine, flunarizine, loratadine, doxylamine, or an analog thereof. More preferably, the second agent is astemizole or an analog thereof. In another embodiment, the composition consists of active ingredients and excipients, and the active ingredients consist of the first and second agents
  • The invention also features a composition that includes a first agent that is an inhibitor of estrogen receptor α and a second agent that is sertraline or an analog thereof. Preferably, the first and second agents are present in amounts that, when administered together to a patient with a filovirus-mediated disease (e.g., a disease caused by Ebola virus or Marburg virus), are effective to treat the patient. The first agent is preferably clomiphene, tamoxifen, raloxifene, toremifene, diethylstilbestrol, or an analog thereof. More preferably, the first agent is clomiphene, tamoxifen, toremifene, or an analog thereof. The second agent is preferably sertraline, paroxetine, or UK-416244. In another embodiment, the composition consists of active ingredients and excipients, and the active ingredients consist of the first and second agents.
  • Any of the compositions of the invention may be formulated, for example, for oral, systemic, parenteral, topical (e.g., ophthalmic, dermatologic), intravenous, inhalational, or intramuscular administration.
  • In another aspect, the invention features a kit including an active agent selected from the agents of Table 2 and instructions for administering the agent to a patient having a filovirus-mediated disease, e.g., a disease caused by an Ebola or a Marburg virus. Preferably, the active agent is selected from the agents of Table 1. More preferably, the active agent is clomiphene, toremifene, astemizole, bepridil, clomopramine, lomerizine, sertraline, or an analog thereof.
  • The invention also features a kit including two or more agents selected from the agents of Table 1 and instructions for administering the agents to a patient having a filovirus-mediated disease, e.g., a disease causes by an Ebola or a Marburg virus. Preferably, the two agents are selected from the agent pairs of Table 3. The two agents may be included together in a composition or may be formulated separately.
  • Related embodiments of the invention are kits including a first agent and selected from Table 1 and instructions for administering the first agent with a second agent selected from Table 1 to a patient having a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus. Preferably, the first and second agents are each clomiphene, toremifene, astemizole, bepridil, clomopramine, lomerizine, sertraline, or an analog thereof.
  • The invention also features kits that include an agent that is an inhibitor of estrogen receptor α, an agent that is an inhibitor of histamine receptor 1, and instructions for administering the agents to a patient with a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus. Related embodiments of the invention feature a kit in which one of the estrogen receptor α and histamine receptor 1 inhibitory agents is included with instructions for administering the other agent of the combination to a patient with a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus. In any of these embodiments, the inhibitor of estrogen receptor α is preferably selected from clomiphene, tamoxifen, toremifene, and analogs thereof and more preferably from clomiphene, tamoxifen, raloxifene, toremifene, and diethylstilbestrol, and analogs thereof. In these embodiments, the inhibitor of histamine receptor 1 is preferably selected from the group consisting of chlorcyclizine, astemizole, clemastine, terfenadine, chlorphenoxamine, oxatomide, azelastine, methdilazine, homochlorcyclizine, desloratadine, flunarizine, loratadine, doxylamine, and analogs thereof, and more preferably from astemizole or an analog thereof. When the kit includes the inhibitor of estrogen receptor α and the inhibitor of histamine receptor, the two agents may be included together in a composition or they may be formulated separately.
  • The invention also features kits that include an agent that is an inhibitor of estrogen receptor α, an agent that is sertraline or an analog thereof, and instructions for administering the agents to a patient with a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus. Related embodiments of the invention feature a kit in which one of the estrogen receptor α inhibitors and sertraline or analog thereof is included with instructions for administering the other agent of the combination to a patient with a filovirus-mediated disease, e.g., one caused by an Ebola virus or Marburg virus. In any of these embodiments, the inhibitor of estrogen receptor α is preferably selected from clomiphene, tamoxifen, toremifene, and analogs thereof and more preferably from clomiphene, tamoxifen, raloxifene, toremifene, and diethylstilbestrol, and analogs thereof. In these embodiments, the sertraline or analog thereof is preferably sertraline, paroxetine, or UK-416244. When the kit includes the inhibitor of estrogen receptor α and the sertraline or analog thereof, the two agents may be included together in a composition or they may be formulated separately.
  • In any of the compositions, methods, or kits that include an inhibitor of estrogen receptor α and an inhibitor of histamine receptor 1, these agents are preferably clomiphene and chlorcyclizine, clomiphene and astemizole, clomiphene and clemastine, clomiphene and terfenadine, clomiphene and chlorphenoxamine, clomiphene and oxatomide, clomiphene and azelastine, clomiphene and methdilazine, clomiphene and homochlorcyclizine, clomiphene and desloratadine, clomiphene and flunarizine, clomiphene and loratadine, clomiphene and doxylamine, tamoxifen and chlorcyclizine, tamoxifen and astemizole, tamoxifen and clemastine, tamoxifen and terfenadine, tamoxifen and chlorphenoxamine, tamoxifen and oxatomide, tamoxifen and azelastine, tamoxifen and methdilazine, tamoxifen and homochlorcyclizine, tamoxifen and desloratadine, tamoxifen and flunarizine, tamoxifen and loratadine, tamoxifen and doxylamine, toremifene and chlorcyclizine, toremifene and astemizole, toremifene and clemastine, toremifene and terfenadine, toremifene and chlorphenoxamine, toremifene and oxatomide, toremifene and azelastine, toremifene and methdilazine, toremifene and homochlorcyclizine, toremifene and desloratadine, toremifene and flunarizine, toremifene and loratadine, toremifene and doxylamine, raloxifene and chlorcyclizine, raloxifene and astemizole, raloxifene and clemastine, raloxifene and terfenadine, raloxifene and chlorphenoxamine, raloxifene and oxatomide, raloxifene and azelastine, raloxifene and methdilazine, raloxifene and homochlorcyclizine, raloxifene and desloratadine, raloxifene and flunarizine, raloxifene and loratadine, raloxifene and doxylamine, diethylstilbestrol and chlorcyclizine, diethylstilbestrol and astemizole, diethylstilbestrol and clemastine, diethylstilbestrol and terfenadine, diethylstilbestrol and chlorphenoxamine, diethylstilbestrol and oxatomide, diethylstilbestrol and azelastine, diethylstilbestrol and methdilazine, diethylstilbestrol and homochlorcyclizine, diethylstilbestrol and desloratadine, diethylstilbestrol and flunarizine, diethylstilbestrol and loratadine, or diethylstilbestrol and doxylamine.
  • In any of the kits of the invention, one or more active agents may be formulated, e.g., for oral, parenteral, systemic, topical, or inhalational administration.
  • Analogs of any of the compounds listed in Tables 1 and 2 may be used in any of the compositions, methods, and kits of the invention.
  • Compounds useful in the invention include those described herein in any of their pharmaceutically acceptable forms, including isomers such as diastereomers and enantiomers, salts, solvates, and polymorphs thereof, as well as racemic mixtures. Compounds useful in the invention may also be isotopically labeled compounds. Useful isotopes include hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, (e.g., 2H, 3H, 13C, 14C, 15N, 18O, 17O, 31P, 32P, 35S, 18F, and 36Cl). Isotopically-labeled compounds can be prepared by synthesizing a compound using a readily available isotopically-labeled reagent in place of a non-isotopically-labeled reagent.
  • By “agent” is meant a compound, e.g., toremifene, or mixture of compounds, e.g., teicoplanin, having a pharmacological activity. The terms “agent,” “compound,” and “drug” are used interchangeably herein.
  • “Histamine receptor 1” refers to the histamine receptor also known as H1 receptor and HRH1.
  • By an “inhibitor of histamine receptor 1,” is meant an agent that antagonizes the signaling of histamine receptor 1 by at least 5%, 10%, 25%, 50%, 60%, 70%, 80%, 90%, or 95%. Inhibition of histamine receptor 1 signaling by an agent can be measured, for example, by reporter assays known in the art.
  • As used herein, the term “estrogen receptor α” (ERα) refers to estrogen hormone receptors containing the estrogen receptor polypeptide encoded by the estrogen receptor 1 (ESR1) gene. An estrogen receptor α protein can include a homodimer of polypeptides encoded by ESR1 or a heterodimer of polypeptides encoded by the ESR1 and estrogen receptor 2 (ESR2) genes.
  • By an “estrogen receptor α inhibitor” is meant an agent that binds to and antagonizes the activity of estrogen receptor α by at least 5%, 10%, 25%, 50%, 60%, 70%, 80%, 90%, or 95% in at least one cell type of an animal. Certain estrogen receptor α inhibitors, e.g., agents known in the art as “selective estrogen receptor modulators,” may inhibit estrogen receptors in one organ, e.g, the breast, while activating estrogen receptors in another organ, e.g., the liver, bone, or uterus. Such compounds are included in the term “estrogen receptor α inhibitors.” The % inhibition of estrogen receptor α activity by an agent can be measured, for example, by a reporter assay known in the art.
  • By a “filovirus” is meant a virus belonging to the family Filoviridae. Exemplary filoviruses are Ebola virus and Marburg virus.
  • By “patient” is meant any animal, e.g., a primate, e.g., a human. Any animal can be treated using the methods, compositions, and kits of the invention.
  • To “treat” is meant to administer one or more agents to measurably slow or stop the replication of a virus in vitro or in vivo, to measurably decrease the load of a virus (e.g., any virus described herein including a filovirus virus such as an Ebola virus or Marburg virus) in a cell in vitro or in vivo, or to reduce at least one symptom (e.g., those described herein) associated with having a filovirus-mediated disease in a patient. Desirably, the slowing in replication or the decrease in viral load is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99%, as determined using a suitable assay (e.g., a replication assay described herein). Alternatively, in certain embodiments, treatment is observed by a trained physician as an appreciable or substantial relief of symptoms in a patient with a filovirus-mediated disease. Typically, a decrease in viral replication is accomplished by reducing the rate of RNA polymerization, RNA translation, protein processing or modification, or by reducing the activity of a molecule involved in any step of viral replication (e.g., proteins or coded by the genome of the virus or host important for viral replication).
  • By “an effective amount” is meant the amount of a compound, alone or in combination with another therapeutic regimen, required to treat a patient with a viral disease (e.g., any virus described herein including an Ebola virus or Marburg virus) in a clinically relevant manner. A sufficient amount of an active compound used to practice the present invention for therapeutic treatment of conditions caused by a virus varies depending upon the manner of administration, the age, body weight, and general health of the patient. Ultimately, the prescribers will decide the appropriate amount and dosage regimen. In a combination therapy of the invention, the effective amount of an agent may less be than the effective amount if the agent were administered in a non-combinatorial (single-agent) therapy. Additionally, an effective amount may be an amount of an agent in a combination therapy of the invention that is safe and efficacious in the treatment of a patient having a viral disease over each agent alone as determined and approved by a regulatory authority (such as the U.S. Food and Drug Administration).
  • By “more effective” is meant that a treatment exhibits greater efficacy, or is less toxic, safer, more convenient, or less expensive than another treatment with which it is being compared. Efficacy may be measured by a skilled practitioner using any standard method that is appropriate for a given indication.
  • By a “low dosage” is meant at least 5% less (e.g., at least 10%, 20%, 50%, 80%, 90%, or even 95%) than the lowest standard recommended dosage of a particular compound formulated for a given route of administration for treatment of any human disease or condition. For example, a low dosage of an agent that inhibits viral replication and that is formulated for administration by intravenous injection will differ from a low dosage of the same agent formulated for oral administration.
  • In the generic descriptions of compounds of this invention, the number of atoms of a particular type in a substituent group is generally given as a range, e.g., an alkyl group containing from 1 to 4 carbon atoms or C1-4 alkyl. Reference to such a range is intended to include specific references to groups having each of the integer number of atoms within the specified range. For example, an alkyl group from 1 to 4 carbon atoms includes each of C1, C2, C3, and C4. A C1-12 heteroalkyl, for example, includes from 1 to 12 carbon atoms in addition to one or more heteroatoms. Other numbers of atoms and other types of atoms may be indicated in a similar manner.
  • As used herein, the terms “alkyl” and the prefix “alk-” are inclusive of both straight chain and branched chain groups and of cyclic groups, i.e., cycloalkyl. Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 12 ring carbon atoms, inclusive. Exemplary cyclic groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl groups.
  • By “C1-4 alkyl” is meant a branched or unbranched hydrocarbon group having from 1 to 4 carbon atoms. A C1-4 alkyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups. C1-4 alkyls include, without limitation, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclopropylmethyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, and cyclobutyl.
  • By “C2-4 alkenyl” is meant a branched or unbranched hydrocarbon group containing one or more double bonds and having from 2 to 4 carbon atoms. A C2-4 alkenyl may optionally include monocyclic or polycyclic rings, in which each ring desirably has from three to six members. The C2-4 alkenyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups. C2-4 alkenyls include, without limitation, vinyl, allyl, 2-cyclopropyl-1-ethenyl, 1-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2-methyl-1-propenyl, and 2-methyl-2-propenyl.
  • By “C2-4 alkynyl” is meant a branched or unbranched hydrocarbon group containing one or more triple bonds and having from 2 to 4 carbon atoms. A C2-4 alkynyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has five or six members. The C2-4 alkynyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxy, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups. C2-4 alkynyls include, without limitation, ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, and 3-butynyl.
  • By “C2-6 heterocyclyl” is meant a stable 5- to 7-membered monocyclic or 7- to 14-membered bicyclic heterocyclic ring which is saturated, partially unsaturated, or unsaturated (aromatic), and which consists of 2 to 6 carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from N, O, and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxy, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups. The nitrogen and sulfur heteroatoms may optionally be oxidized. The heterocyclic ring may be covalently attached via any heteroatom or carbon atom which results in a stable structure, e.g., an imidazolinyl ring may be linked at either of the ring-carbon atom positions or at the nitrogen atom. A nitrogen atom in the heterocycle may optionally be quaternized. Preferably when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. Heterocycles include, without limitation, 1H-indazole, 2-pyrrolidonyl, 2H,6H-1,5,2-dithiazinyl, 2H-pyrrolyl, 3H-indolyl, 4-piperidonyl, 4aH-carbazole, 4H-quinolizinyl, 6H-1,2,5-thiadiazinyl, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 4aH-carbazolyl, b-carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinylperimidinyl, phenanthridinyl, phenanthrolinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, pteridinyl, piperidonyl, 4-piperidonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, carbolinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, and xanthenyl. Preferred 5 to 10 membered heterocycles include, but are not limited to, pyridinyl, pyrimidinyl, triazinyl, furanyl, thienyl, thiazolyl, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, tetrazolyl, benzofuranyl, benzothiofuranyl, indolyl, benzimidazolyl, 1H-indazolyl, oxazolidinyl, isoxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, quinolinyl, and isoquinolinyl. Preferred 5 to 6 membered heterocycles include, without limitation, pyridinyl, pyrimidinyl, triazinyl, furanyl, thienyl, thiazolyl, pyrrolyl, piperazinyl, piperidinyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, and tetrazolyl.
  • By “C6-12 aryl” is meant an aromatic group having a ring system comprised of carbon atoms with conjugated π electrons (e.g., phenyl). The aryl group has from 6 to 12 carbon atoms. Aryl groups may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has five or six members. The aryl group may be substituted or unsubstituted. Exemplary substituents include alkyl, hydroxy, alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, fluoroalkyl, carboxyl, hydroxyalkyl, carboxyalkyl, amino, aminoalkyl, monosubstituted amino, disubstituted amino, and quaternary amino groups.
  • By “C7-14 alkaryl” is meant an alkyl substituted by an aryl group (e.g., benzyl, phenethyl, or 3,4-dichlorophenethyl) having from 7 to 14 carbon atoms.
  • By “C3-10 alkheterocyclyl” is meant an alkyl substituted heterocyclic group having from 3 to 10 carbon atoms in addition to one or more heteroatoms (e.g., 3-furanylmethyl, 2-furanylmethyl, 3-tetrahydrofuranylmethyl, or 2-tetrahydrofuranylmethyl).
  • By “C1-7 heteroalkyl” is meant a branched or unbranched alkyl, alkenyl, or alkynyl group having from 1 to 7 carbon atoms in addition to 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, S, and P. Heteroalkyls include, without limitation, tertiary amines, secondary amines, ethers, thioethers, amides, thioamides, carbamates, thiocarbamates, hydrazones, imines, phosphodiesters, phosphoramidates, sulfonamides, and disulfides. A heteroalkyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has three to six members. The heteroalkyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, hydroxyalkyl, carboxyalkyl, and carboxyl groups. Examples of C1-7 heteroalkyls include, without limitation, methoxymethyl and ethoxyethyl.
  • By “halide” or “halogen” is meant bromine, chlorine, iodine, or fluorine.
  • By “fluoroalkyl” is meant an alkyl group that is substituted with a fluorine atom.
  • By “perfluoroalkyl” is meant an alkyl group consisting of only carbon and fluorine atoms.
  • By “carboxyalkyl” is meant a chemical moiety with the formula —(R)—COOH, wherein R is selected from C1-7 alkyl, C2-7 alkenyl, C2-7 alkynyl, C2-6 heterocyclyl, C6-12 aryl, C7-14 alkaryl, C3-10 alkheterocyclyl, or C1-7 heteroalkyl.
  • By “hydroxyalkyl” is meant a chemical moiety with the formula —(R)—OH, wherein R is selected from C1-7 alkyl, C2-7 alkenyl, C2-7 alkynyl, C2-6 heterocyclyl, C6-12 aryl, C7-14 alkaryl, C3-10 alkheterocyclyl, or C1-7 heteroalkyl.
  • By “alkoxy” is meant a chemical substituent of the formula —OR, wherein R is selected from C1-7 alkyl, C2-7 alkenyl, C2-7 alkynyl, C2-6 heterocyclyl, C6-12 aryl, C7-14 alkaryl, C3-10 alkheterocyclyl, or C1-7 heteroalkyl.
  • By “aryloxy” is meant a chemical substituent of the formula —OR, wherein R is a C6-12 aryl group.
  • By “alkylthio” is meant a chemical substituent of the formula —SR, wherein R is selected from C1-7 alkyl, C2-7 alkenyl, C2-7 alkynyl, C2-6 heterocyclyl, C6-12 aryl, C7-14 alkaryl, C3-10 alkheterocyclyl, or C1-7 heteroalkyl.
  • By “arylthio” is meant a chemical substituent of the formula —SR, wherein R is a C6-12 aryl group.
  • By “quaternary amino” is meant a chemical substituent of the formula N(R)(R′)(R″)(R′″)+, wherein R, R′, R″, and R′″ are each independently an alkyl, alkenyl, alkynyl, or aryl group. R may be an alkyl group linking the quaternary amino nitrogen atom, as a substituent, to another moiety. The nitrogen atom, N, is covalently attached to four carbon atoms of alkyl, heteroalkyl, heteroaryl, and/or aryl groups, resulting in a positive charge at the nitrogen atom.
  • Other features and advantages of the invention will be apparent from the following Detailed Description and the claims.
  • DETAILED DESCRIPTION OF THE INVENTION
  • We have identified compounds that decrease replication of an infectious filovirus engineered with a green fluorescent protein (GFP) in mammalian cells. Accordingly, the present invention provides compositions, methods, and kits useful in the treatment of viral diseases caused by a filovirus. In certain embodiments, the viral disease is caused by an Ebola virus or Marburg virus. Compositions of the invention can include a combination pair of any two agents selected from Table 2 or the particular pairs of agents of Table 3. Treatment methods of the invention include administration of a single agent from Table 1 or Table 2 or a pair of agents selected from the agents of Table 1 or from the agent pairs listed in Table 3. Optionally, functional or structural analogs (e.g., those described herein) of these agents may be employed in the compositions, methods, and kits of the invention. The composition may function by decreasing RNA polymerization, RNA translation, RNA transcription, a decrease in posttranslational protein processing, or a decrease in activity of a protein involved in viral replication (e.g., a protein coded for by the viral genome or a host protein required for viral replication).
  • In one particular example, the patient being treated is administered a combination of two agents listed in Table 1 within 7 days of each other in amounts that together are sufficient to treat the patient having a filovirus-mediated disease. An effective amount of one or both of the agents may be a low dosage relative the effective amount of the agent when administered singly.
  • Filovirus-Mediated Diseases
  • The invention relates to the treatment of diseases caused by viruses of the family Filoviridae (filoviruses). Filoviruses are negative strand RNA viruses that can infect humans and primates. Filoviruses include viruses of the genera Ebolavirus and Marburgvirus. In particular, Ebola hemorrhagic fever is a severe, often-fatal disease in humans and has appeared sporadically since its initial recognition in 1976. The disease is caused by infection with an Ebola virus. Five identified subtypes of Ebola virus are Ebola-Zaire, Ebola-Sudan, Ebola-Ivory Coast, and Ebola-Bundibugyo, each of which have caused disease in humans, and Ebola-Reston, which has caused disease in nonhuman primates, but not in humans. Other undiscovered subtypes may exist and are intended to be included in the scope of Ebola virus.
  • Infection with an Ebola or Marburg virus usually causes life-threatening hemorrhagic fever. Symptoms include fever, severe headache, joint and muscle aches, chills, sore throat, weakness, nausea and vomiting, diarrhea, red eyes, raised rash, chest pain and cough, hiccups (Ebola virus), stomach pain, bleeding (from any bodily orifice), and psychological symptoms (confusion, irribility, aggression, or depression). As the illness progresses, jaundice, dilirium, seizures, severe bleeding, organ failure, coma, shock, and death can occur.
  • Agents of the Invention
  • Certain agents that may be employed in the methods, compositions, and kits of the present invention are discussed in greater detail below. It is understood that an analog of any compound of Table 1 and Table 2 can be used instead of the compound of Table 1 or Table 2 in the methods, compositions, and kits of the present invention.
  • Estrogen Receptor Inhibitors
  • In certain embodiments, an estrogen receptor α inhibitor can be used in the compositions, methods, and kits of the invention. By an “estrogen receptor α inhibitor” is meant a compound that inhibits the activity of an estrogen receptor α by at least 5%, e.g., greater than 10%, 20%, 40%, 60%, 80%, 90%, or 95%. ERα inhibitors include clomiphene, tamoxifen, tomerifene, and raloxifene. ERα inhibitors that may be particularly efficacious are described in more detail below.
  • Clomiphene
  • Clomiphene is described in U.S. Pat. No. 2,914,563 and has the following structure:
  • Figure US20130289024A1-20131031-C00001
  • Structural analogs of clomiphene include olefinic isomers. Structural analogs of clomiphene are also described by the following formula:
  • Figure US20130289024A1-20131031-C00002
  • wherein X is any halogen (e.g., F, Cl, Br, or I), R1, R2, and R3 may be located at any position of the phenyl group and are selected, independently, from H, halogen, C1-6 alkyl, C1-6 alkoxy, —OCnH2nA, and at least one of R1, R2, and R3 is —OCnH2nA, wherein n is 2, 4, 5, or 6; A=NR4R5, wherein each R4 and R5 is, independently, an optionally substituted C1-6 alkyl, or R4 and R5 combined to form an optionally substituted cyclic structure.
  • Desirably, when R1, R2, or R3 is —OCnH2nA, the substituents is located para to the olefin substituents. Examples of C1-6 alkyls include, but are not limited to: methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, isoamyl, and n-hexyl. Examples of C1-6 alkoxy groups include, but are not limited to: methoxy, ethoxy, propoxy, isopropoxy, n-butyloxy, iso-butyloxy, sec-butyloxy, tert-butyloxy, n-pentoxy, O-isoamyl, and O-hexyl. Examples of rings formed by the combination R4 and R5 include, but are not limited to pyrrolidine and piperidine.
  • Other clomiphene analogs are described in U.S. Pat. Nos. 2,914,563 and 5,189,212 and by the general formula of U.S. Pat. No. 5,410,080.
  • Tamoxifen
  • Tamoxifen is described in U.S. Pat. No. 4,536,516 and has the following structure:
  • Figure US20130289024A1-20131031-C00003
  • Tamoxifen analogs are described by general formula (I) in U.S. Pat. No. 4,806,685, e.g., threo-1-[4-(2,3-epoxypropoxy)-phenyl]-1,2-diphenyl-3,3,3-trifluoro-propane, (E)-1,2-diphenyl-3,3,3-trifluoro-1-[4-(2-[4-methylpiperazino]-ethoxy)-phenyl]-propene, 1-[4-(2-dimethylaminoethoxy)-phenyl]-2-phenyl-3,3,3-trifluoro-1-(4-hydroxy phenyl)propene, (E)1,2-diphenyl-3,3,3-trifluoro-1-[4-(2-[2-hydroxyethylamino]-ethoxy)-phenyl]-propene, (E)-1-[4-(2-azidoethoxy)-phenyl]-1,2-diphenyl-3,3,3-trifluoro-propene, and 1-[4-(2-dimethylamino-ethoxy)-phenyl]-3,3,3-trifluoro-1,2-bis-(4-hydroxyphenyl)-propene.
  • Other tamoxifen analogs are described by general formula (I) in U.S. Pat. No. 5,047,431, e.g., (E)-1-[4′-(2-dimethylaminoethoxy)phenyl]-1-(3′-hydroxyphenyl)-2-phenylbut-1-ene, (E)-1-[4′(2-diethylaminoethoxy)phenyl]-1-(3′-hydroxyphenyl)-2-phenylbut-1-ene, (E)-1-(3′-hydroxyphenyl)-1-[4′-(2-methylaminoethoxy)phenyl]-2-phenylbut-1-ene, and (E)-1-[4′-ethylaminoethoxy)phenyl]-1-(3′-hydroxyphenyl)-2-phenylbut-1-ene.
  • Yet other tamoxifen analogs are described by formula (I) in U.S. Pat. No. 5,681,835, e.g., 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-N,N-diethyl acrylamide, 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-N,N-diethyl propionamide, 2-[4-(1,2-diphenyl-but-1-enyl)-phenyl]cyclopropanecarboxylic acid diethylamide, 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-N,N-diethyl-2-methyl-acrylamide, 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-but-2-enoic acid diethylamide, 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-acrylic acid methyl ester, 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-acrylonitrile, 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-acrylic acid tert-butyl ester, 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-acrylic acid, 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-1-morpholin-4-yl-prop-2-en-1-one, 3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]-N-(3-methoxy-propyl)-acrylamide, and N,N-dicyclohexyl-3-[4-(1,2-diphenyl-but-1-enyl)-phenyl]acrylamide.
  • Additional tamoxifen analogs are droloxifene, 4-iodotamoxifen, idoxifene, described in U.S. Pat. No. 6,096,874, in U.S. Pat. No. 6,576,645, in U.S. Pat. No. 6,875,775, and by general formula (I) of U.S. Pat. No. 5,807,899.
  • Toremifene
  • Toremifene is described in U.S. Pat. No. 4,696,949 and has the following structure:
  • Figure US20130289024A1-20131031-C00004
  • Analogs of toremifene analogs are described by several structural formulae described in U.S. Pat. No. 4,696,949, U.S. Pat. No. 4,996,225, and U.S. Pat. No. 5,491,173. Examples are 1-phenyl-1,2-bis(4-hydroxyphenyl)-1-buten-4-ol, 4-bromo-1-phenyl-1,2-bis(4-hydroxyphenyl)-1-butene, 2-phenyl-2,3-bis(4-hydroxyphenyl)tetrahydrofuran, 1,2-diphenyl-1-(4-hydroxyphenyl)-1-penten-5-ol, 2,3-diphenyl-2-(4-hydroxyphenyl)tetrahydropyran, 1,2-diphenyl-1-(4-hydroxyphenyl)-1-penten-5-al, 1,2-diphenyl-1-(4-hydroxyphenyl)-1-buten-4-ol, 2,3-diphenyl-2-(4-hydroxyphenyl)tetrahydrofuran, 1,2-diphenyl-1-[4-[2-(N,N-dimethylamino)ethoxy]phenyl]-1-buten-4-ol, 4-chloro-1,2-diphenyl-1-[4-[2-(N,N-dimethylamino)ethoxy]phenyl]-1-butene, 4-chloro-1,2-diphenyl-1-[4-[2-(1-aziridinyl)ethoxy]phenyl]-1-butene, 4-bromo-1,2-diphenyl-1-[4-[2-(1-pyrrolidinyl)ethoxy]phenyl]-1-butene, 2,3-diphenyl-2-[4-[2-(N,N-diethylamino)ethoxy]phenyl]tetrahydrofuran, 1-phenyl-1,2-bis(4-hydroxyphenyl)butan-4-ol, 4-bromo-1-phenyl-1,2-bis(4-hydroxyphenyl)butane, 1,2-diphenyl-1-(4-hydroxyphenyl)butan-4-ol, 4-chloro-1,2-diphenyl-1-[4-(2-piperidinoethoxy)phenyl]butane, 1,2-diphenyl-1-(4-hydroxyphenyl)butane-1,4-diol, 1-phenyl-1,2-bis(4-hydroxyphenyl)butane-1,4-diol, 1,2-diphenyl-1-(4-methoxyphenyl)-1-buten-4-ol, 4-bromo-1,2-diphenyl-1-[4-[2-(N,N-dimethylamino)ethoxy]phenyl]-1-butene, 4-chloro-1,2-diphenyl-1-(4-hydroxyphenyl)butane, 4-chloro-1,2-diphenyl-1-(4-hydroxyphenyl)-1-butene, and 1,2-diphenyl-1-[4-[2-(N,N-dimethylamino)ethoxy]phenyl]butane-1,4-diol.
  • Additional examples of toremifene analogs are given by formula (I) of U.S. Pat. No. 5,491,173.
  • Inhibitors of Histamine Receptor 1
  • In certain embodiments, the methods, compositions, and kits of the invention employ an inhibitor of histamine receptor 1. The inhibitor may inhibit the activity of histamine receptor 1 by at least 5%, e.g., greater than 10%, 20%, 40%, 60%, 80%, 90%, or 95%. Either non-sedating or sedating inhibitors may be employed. Exemplary histamine receptor 1 inhibitors include acrivastine, alcaftadine, antazoline, azatadine, AZD-1744, azelastine, bepotastine, bepotastine besilate, betotastine besilate, bilastine, BM-113, carebastine, cetirizine, chlorpheniramine, chlorphenoxamine, clemastine, cyclizine, desloratadine, doxepin, E-4716, ebastine, efletirizine, epinastine, epinastine, fexofenadine, FK-613, GSK-1004723, HSR-609, IOT-101, KA-398, KAA-276, KC-11404, KC-11425, ketotifen, levocetirizine, loratadine, MDL-28163, mianserin, mizolastine, NBI-75043, NIP-530, noberastine, olopatadine, oxatomide, periciazine, pheniramine, promethazine, ReN-1869, rupatadine, selenotifen, SUN-1334H, tagorizine, tecastemizole, terfenadine, tranilast, tranilast, triprolidine, UCB-35440, vapitadine, VUF-K-9015, WY-49051, YM-344484, ZCR-2060. Structural analogs of any of the above histamine receptor 1 inhibitors or other inhibitors of histamine receptor 1 may also be employed instead of these.
  • Inhibitors of histamine receptor 1 that may be particularly efficacious is described in more detail below.
  • Astemizole
  • Astemizole is described in U.S. Pat. No. 4,219,559 and has the following structure:
  • Figure US20130289024A1-20131031-C00005
  • Analogs of astemizole include, e.g, 1-(4-fluorophenylmethyl)-N-{1-[2-(4-methoxyphenyl)ethyl]-4-piperidinyl}-1H-benzimidazol-2-amine, 4-[2-{4-[1-(4-fluorophenylmethyl)-1H-benzimidazol-2-ylamino]-1-piperidinyl}ethyl]phenol, {4-[2-{4-[1-(4-fluorophenylmethyl)-1H-benzimidazol-2-ylamino]-1-piperidinyl}ethyl]phenyl}benzeneacetate, {4-[2-{4-[1-(4-fluorophenylmethyl)-1H-benzimidazol-2-ylamino]-1-piperidinyl}ethyl]phenoxy}acetonitrile, and compounds described by the formula of claim 1 in U.S. Pat. No. 4,219,559. Additional analogs are described by formula I in U.S. Pat. No. 4,556,660, e.g., [(4-fluorophenyl)methyl]-N-[1-[2-[(2-pyridinyl)amino]ethyl]-4-piperidinyl]-3H-imidazo[4,5-b]pyridin-2-amine, 2-[[2-[4-[[3-[(4-fluorophenyl)methyl]-3H-imidazo[4,5-b]pyridin-2-yl]amino]-1-piperidinyl]ethyl]amino]-3-pyridinecarboxamide, 1-[(4-fluorophenyl)methyl]-N-[1-[2-[(2-pyrimidinyl)amino]ethyl]-4-piperidinyl]-1H-imidazo[4,5-b]pyridin-2-amine, 1[(4-fluorophenyl)methyl]-N-[1-[2-(2-pyrimidinylamino]ethyl]-4-piperidinyl]1H-imidazo[4,5-c]pyridin-2-amine, 3-(2-pyridinylmethyl)-N-[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]3H-imidazo[4,5-b]pyridin-2-amine (E)-2-butenedioate, 3-(2-furanylmethyl)-N-[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]-3H-imidazo[4,5-b]pyridin-2-amine, 3-[(4-fluorophenyl)methyl]-N-[1-[2-[(2-pyrimidinyl)amino]ethyl]-4-piperidinyl]-3H-imidazo[4,5-c]pyridin-2-amine (E)-2-butenedioate, N-[1-[3-[(5-chloro-2-pyridinyl)amino]propyl]-4-piperidinyl]-1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-amine trihydrochloride monohydrate, 6-chloro-N. sup.4-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-4,5-pyrimidinediamine, 8-chloro-N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-3-phthalazinamine 2-propanolate, 1-(phenylmethyl)-N-[1-[2-[(2-pyrimidinyl)amino]ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, 1-methyl-N-[1-[2-[(2-pyrimidinyl)amino]ethyl]-4-piperidinyl]-1H-benzimidazo 1-2-amine hemihydrate, 1-[(4-methylphenyl)methyl]-N-[1-[2-[(2-pyrimidinyl)amino]ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, 1-[(4-chlorophenyl)methyl]-N-[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, 1-[(4-methoxyphenyl)methyl]-N-[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, N-[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, 1-[(4-fluorophenyl)methyl]-5-methoxy-N-[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, N-[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]-1-(4-thiazolylmethyl)-1H-benzimidazol-2-amine (E)-2-butenedioate, 4-[[2-[[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]amino]-1H-benzimidazo 1-1-yl]methyl]phenol, 1-[(4-fluorophenyl)methyl]-6-methoxy-N-[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, 1-[(4-fluorophenyl)methyl]-N-methyl-N-[1-[2-pyrimidinylamino)ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, N-(phenylmethyl)-N-[1-[2-(2-pyrimidinylamino)ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, N-[1-[2-[(5-bromo-2-pyridinyl)oxy]ethyl]-4-piperidinyl]-1-(2-furanylmethyl)-1H-benzimidazol-2-amine, 1-[(4-fluorophenyl)methyl]-N-[1-[2-[[2-(methylthio)-4-pyrimidinyl]oxy]ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, 1-[(4-fluorophenyl)methyl]-N-[1-[2-[(3-methyl-2-quinoxalinyl)oxy]ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, 1-[(4-fluorophenyl)methyl]-N-[1-[2-(2-pyrimidinyloxy)ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine, N-[1-[2-[(5-bromo-2-pyridinyl)oxy]ethyl]-4-piperidinyl]-1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-amine, 1-(2-furanylmethyl)-N-[1-[2-(2-pyrimidinyloxy)ethyl]-4-piperidinyl]-1H-benz imidazol-2-amine (E)-2-butenedioate, 1-[(4-fluorophenyl)methyl]-N-[1-[2-(2-pyridinylmethoxy)ethyl]-4-piperidinyl]-1H-benzimidazol-2-amine (E)-2-butenedioate, 2-[[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]amino]-6-propyl-4-pyrimidinol, 2-[[2-[[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]amino]-4(1H)-pyrimidinone, 2-[[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]1-piperidinyl]ethyl]amino]-4(1H)-quinazolinone, 2-[[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]amino]-6-(phenylmethyl)-4-(1H)-pyrimidinone, 2-[[2-[4-8 [1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]amino]-6-methyl-4(1H)-pyrimidinone, N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-1-phthalazinamine, N4-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-4,5-pyrimidinediamine, N-[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]-1′-(2-pyridinyl)-[1,4′-bipiperidin]-4-amine (E)-2-butenedioate (2:3) monohydrate, N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-N′-(2-pyridinylmethyl)thiourea, N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-N′-(3-pyridinyl)thiourea, N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-N′-(2-pyridinyl)thiourea, N-(4-amino-3-pyridinyl)-N′-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]thiourea, N-(3-amino-2-pyridinyl)-N′-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidaol-2-yl]amino]-1-piperidinyl]ethyl]thiourea, N-(4-amino-3-pyridinyl)-N′-[2-[4-[[1-(2-furanylmethyl)-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]thiourea, N-(4-amino-3-pyridinyl)-N′-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-imidazo[4,5-b]pyridin-2-yl]amino]-1-piperidinyl]ethyl]thiourea, N-(4-amino-3-pyridinyl)-N′-[2-[4-[[3-(2-pyridinylmethyl)-3H-imidazo[4,5-b]pyridin-2-yl]amino]-1-piperidinyl]ethyl]thiourea, N-(4-amino-3-pyridinyl)-N′-[2-[4-[[3-[(4-fluorophenyl)methyl]-3H-imidazo[4,5-b]pyridin-2-yl]amino]-1-piperidinyl]ethyl]thiourea, N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-2-quinolinecarboxamide (E)-2-butenedioate, 2-chloro-N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-3-pyridinecarboxamide (E)-2-butenedioate (1:2) hemihydrate, and 6-chloro-N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-3-pyridinecarboxamide (E)-2-butenedioate; by formula I of U.S. Pat. No. 4,588,722, e.g., 1-[(4-fluorophenyl)methyl]-N-[1-[2-(4-methoxyphenyl)ethyl]-4-piperidinyl]-1H-imidazo[4,5-b]pyridin-2-amine, 1-[(4-fluorophenyl)methyl]-N-[1-[2-(4-methoxyphenyl)ethyl]-4-piperidinyl]-1H-imidazo[4,5-c]pyridin-2-amine, 3-[(4-fluorophenyl)methyl]-N-[1-[2-(4-methoxyphenyl)ethyl]-4-piperidinyl]-3H-imidazo[4,5-c]pyridin-2-amine (E)-2-butenedioate, 3-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-2,4-(1H,3H)-pyrimidinedione, 3-[2-[4-[[1-(4-thiazolylmethyl)-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one dihydrochloride sesquihydrate, 3-[2-[4-[[3-(2-pyridinylmethyl)-3H-imidazo[4,5-b]pyridin-2-yl]amino]-1-piperidinyl]ethyl]-2H-1benzopyran-2-one trihydrochloride dehydrate, 3-[2-[4-[[1-(2-thienylmethyl)-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one, 3-[2-[4-[[1-(2-pyridinylmethyl)-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one dihydrochloride dehydrate, 3-[2-[4-[[1-(3-pyridinylmethyl)-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one monohydrate, 3-[2-[4-[[1-(2-thienylmethyl)-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one, N-[1-[2:(4-methoxyphenyl)ethyl]-4-piperidinyl]-1-[(2-pyrazinyl)methyl]-1H-benzimidazol-2-amine, 3-[2-[4-[[1-(2-furanylmethyl)-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one monohydrate, 3-[2-[4-[[3-(2-furanylmethyl)-3H-imidazo[4,5-b]pyridin-2-yl]amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one, 3-[2-[4-[[3-[(4-fluorophenyl)methyl]-3H-imidazo[4,5-b]pyridin-2-yl]-amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one monohydrate, N-[1-[2:(4-methoxyphenyl)ethyl]-4-piperidinyl]-1-[(2-pyrazinyl)methyl]-1H-benzimidazol-2-amine, 3-[2-[4-[[1-(2-furanylmethyl)-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one monohydrate, 3-[2-[4-[[3-(2-furanylmethyl)-3H-imidazo[4,5-b]pyridin-2-yl]amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one, 3-[2-[4-[[3-[(4-fluorophenyl)methyl]-3H-imidazo[4,5-b]pyridin-2-yl]-amino]-1-piperidinyl]ethyl]-2H-1-benzopyran-2-one monohydrate, N,N-diethyl-4-[1-(4-fluorophenylmethyl)-1H-benzimidazol-2-ylamino]-[1,4′-bipiperidine]-1′-carboxamide, N″-cyano-N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]-amino]-1-piperidinyl]ethyl-N′,N′-dimethylguanidine, N″-cyano-N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]-amino-1]-1-piperidinyl]ethyl]-N′-[2-(4-morpholinyl)ethyl]guanidine monohydrate, N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-4-morpholinecarbothioamide, N-[2-[4-[[1-(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-N′,N′-dimethylthiourea, N,N-diethyl-N′-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]-amino]-1-piperidinyl]ethyl]thiourea, N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-N′(2-phenylethyl)thiourea (E)-2-butenedioate, N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]hydrazinecarbothioamide monohydrate, 4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-N-methyl-[1,4′-bipiperidine]-1′-carboxamide, N-cyclohexyl-N′-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]thiourea, N-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]-N′ phenylthiourea, N-[2-[4-[1-(4-fluorophenylmethyl)-1H-benzimidazol-2-ylamino]-1-piperidinyl]ethyl]-N′(4-methoxyphenyl)thiourea, N-[4-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1 piperidinyl]ethyl]phenyl]benzenamide monohydrochloride, and N-[4-[2-[4-[[1-[(4-fluorophenyl)methyl]-1H-benzimidazol-2-yl]amino]-1-piperidinyl]ethyl]phenyl]acetamide.
  • Yet other analogs of astemizole are compounds according to formula I of U.S. Pat. No. 4,634,704, formula I of U.S. Pat. No. 4,689,330, formula I of U.S. Pat. No. 4,695,569, formula I of U.S. Pat. No. 4,695,575, formula XVIII of U.S. Pat. No. 4,760,074, formula M-a of U.S. Pat. No. 4,820,822, formula I of U.S. Pat. No. 4,835,161, formula I of U.S. Pat. No. 4,861,785, formula III-a of U.S. Pat. No. 4,888,426, formula I in U.S. Pat. No. 4,897,401, formula I of U.S. Pat. No. 4,908,372, formula I of U.S. Pat. No. 4,943,580, formula I of U.S. Pat. No. 4,988,689, formula I of U.S. Pat. No. 5,006,527, formula I of U.S. Pat. No. 5,008,268, formula I of U.S. Pat. No. 5,025,014, formula I of U.S. Pat. No. 5,041,448, formula I of claim 1 of U.S. Pat. No. 5,071,846, formula III-a of U.S. Reissue Pat. No. 33,833, formula I of U.S. Pat. No. 5,106,857, formula I of U.S. Pat. No. 5,126,339, formula I of U.S. Pat. No. 5,151,424, formula I of U.S. Pat. No. 5,217,980, the formula of claim 1 of U.S. Pat. No. 5,258,380, formula I of U.S. Pat. No. 5,272,150, and exemplary compounds described therein. Yet other analogs of astemizole are described in U.S. Pat. No. 5,278,165, U.S. Pat. No. 5,360,807, U.S. Pat. No. 5,380,731, U.S. Pat. No. 6,130,233 (norastemizole), and U.S. Pat. No. 7,355,051, each of which is incorporated by reference.
  • Clemastine
  • Clemastine is a sedating anti-histamine agent often provided as clemastine fumarate. Analogs of clemastine include, e.g., dephenhydramine, meclizine, clobenztropine, nchembio873-comp44 ((1S,5S)-3-[(4-chlorophenyl)-phenylmethoxy]-8-methyl-8-azabicyclo[3.2.1]octane), AHR 209 (3-[(4-chlorophenyl)-phenylmethoxy]-1-propan-2-ylpyrrolidine hydrochloride), chlortropbenzyl, pyroxamine, AHR225 (1-butyl-3-[(4-chlorophenyl)-phenylmethoxy]pyrrolidine), AHR226 (1-tert-butyl-3-[(4-chlorophenyl)-phenylmethoxy]pyrrolidine hydrochloride), AHR211 (3-[(4-chlorophenyl)-phenylmethoxy]-1-ethylpyrrolidine hydrochloride), AHR 179 (3-[(4-chlorophenyl)-phenylmethoxy]-1-(2-methylpropyl)pyrrolidine hydrochloride), substanz NR (1-[2-[(4-chlorophenyl)-phenylmethoxy]ethyl]pyrrolidine), and NSC63285 (3-[(4-chlorophenyl)-phenylmethoxy]-N,N-diethylpropan-1-amine bromide).
  • Phenothiazines
  • By a “phenothiazine” is meant a polycyclic heterocycle including two optionally substituted benzene rings fused to one of thiazine and described by the formula:
  • Figure US20130289024A1-20131031-C00006
  • wherein each of Ra and Rc is, independently, selected from of H, F, Cl, Br, CF3, cyano, S—Rd, S(O)2—Rd, C1 alkyl, C2-4 alkenyl, C2-4 alkynyl, C2-6 heterocyclyl, C6-12 aryl, C7-14 alkaryl, C3-10 alkheterocyclyl, and C1-7 heteroalkyl; each Rd is, independently, selected from C1 alkyl, C2-4 alkenyl, C2-4 alkynyl, C2-6 heterocyclyl, C6-12 aryl, C7-14 alkaryl, C3-10 alkheterocyclyl, and C1-7 heteroalkyl; and Rb is selected from C3-10 alkheterocyclyl and C1-7 heteroalkyl.
  • Thioridazine, Prochlorperazine, Piperacetazine, and Fluphenazine
  • In certain embodiments, the compositions, methods, or kits of the invention employ thioridazine, prochlorperazine, piperacetazine, or fluphenazine. A related phenothiazine such as one described below may also be employed.
  • Thioridazine, prochlorperazine (U.S. Pat. No. 2,902,484), piperacetazine (GB Pat. No. 861,807), and fluphenazine (U.S. Pat. No. 3,058,979) have the following structures:
  • Figure US20130289024A1-20131031-C00007
  • Analogs of these compounds that may be useful in the present invention include phenothiazines, which include, without limitation, acepromazine, cyamemazine, fluphenazine, mepazine, methotrimeprazine, methoxypromazine, perazine, pericyazine, perimethazine, perphenazine, pipamazine, pipazethate, piperacetazine, pipotiazine, promethazine, propionylpromazine, propiomazine, sulforidazine, thiazinaminiumsalt, thiethylperazine, thiopropazate, thioridazine, trifluoperazine, trimeprazine, thioproperazine, trifluomeprazine, triflupromazine, chlorpromazine, chlorproethazine, those compounds in PCT application WO02/057244, and those compounds in U.S. Pat. Nos. 2,415,363; 2,519,886; 2,530,451; 2,607,773; 2,645,640; 2,766,235; 2,769,002; 2,784,185; 2,785,160; 2,837,518; 2,860,138; 2,877,224; 2,921,069; 2,957,870; 2,989,529; 3,058,979; 3,075,976; 3,194,733; 3,350,268; 3,875,156; 3,879,551; 3,959,268; 3,966,930; 3,998,820; 4,785,095; 4,514,395; 4,985,559; 5,034,019; 5,157,118; 5,178,784; 5,550,143; 5,595,989; 5,654,323; 5,688,788; 5,693,649; 5,712,292; 5,721,254; 5,795,888; 5,597,819; 6,043,239; and 6,569,849, each of which is incorporated herein by reference. Other structurally related phenothiazines having similar antiviral properties are also intended to be encompassed by this group.
  • The structures of several of the above-mentioned phenothiazines are provided below.
  • Figure US20130289024A1-20131031-C00008
    Figure US20130289024A1-20131031-C00009
    Figure US20130289024A1-20131031-C00010
    Figure US20130289024A1-20131031-C00011
    Figure US20130289024A1-20131031-C00012
    Figure US20130289024A1-20131031-C00013
    Figure US20130289024A1-20131031-C00014
  • Phenothiazine compounds can be prepared using, for example, the synthetic techniques described in U.S. Pat. Nos. 2,415,363; 2,519,886; 2,530,451; 2,607,773; 2,645,640; 2,766,235; 2,769,002; 2,784,185; 2,785,160; 2,837,518; 2,860,138; 2,877,224; 2,921,069; 2,957,870; 2,989,529; 3,058,979; 3,075,976; 3,194,733; 3,350,268; 3,875,156; 3,879,551; 3,959,268; 3,966,930; 3,998,820; 4,785,095; 4,514,395; 4,985,559; 5,034,019; 5,157,118; 5,178,784; 5,550,143; 5,595,989; 5,654,323; 5,688,788; 5,693,649; 5,712,292; 5,721,254; 5,795,888; 5,597,819; 6,043,239; and 6,569,849, each of which is incorporated herein by reference.
  • Calcium Channel Blockers
  • In certain embodiments, the compositions, methods, or kits of the invention employ a calcium channel-blocking agent, e.g., lomerizine or bepridil.
  • Lomerizine
  • Lomerizine and analogs of lomerizine, e.g., 1-(2,4-dimethoxybenzyl)-4-(4-fluorobenzhydryl)-piperazine, 1-(2,4-dimethoxybenzyl)-4-[bis(4-fluorophenyl)-methyl]piperazine], and 1-(2,3,4-trimethoxybenzyl)-4-(4-fluorobenzhydryl)piperazine, are described by general formula (I) in U.S. Pat. No. 4,663,325. Other analogs include cinnamylpiperazine derivatives described by formula (I) in U.S. Pat. No. 4,703,048, such as 1-[bis(4-fluorophenyl)methyl]-4-(2,3,4-trimethoxycinnamyl)piperazine, 1-benzhydryl-4-(2,3,4-trimethoxycinnamyl)piperazine, 1-[bis(4-fluorophenyl)methyl]-4-(2,4-dimethoxycinnamyl)piperazine, and 1-benzhydryl-4-(2,4-dimethoxycinnamyl)piperazine, diene derivatives of general formula (I) described in U.S. Pat. No. 4,792,553, e.g., 1-benzhydryl-4-(5-phenyl-2,4-pentadienyl)piperazine, 1-(4,4′-difluorobenzhydryl)-4-(5-phenyl-2,4-pentadienyl)piperazine, 1-benzhydryl-4-(5-(3,4,5-trimethoxyphenyl)-2,4-pentadienyl)piperazine, 1-(4,4′-dichloro(or dibromo)benzhydryl)-4-(5-phenyl-2,4-pentadienyl)piperazine, 1-(4,4′-difluoro(or dichloro or dibromo)-benzhydryl)-4-(5-phenyl-2,4-pentadienyl)piperazine, 1-benzhydryl-4-(5-(3,4,5-triethoxyphenyl)-2,4-pentadienyl)piperazine, 1-benzhydryl-4-(5-(3,5-dimethoxy-4-hydroxy-phenyl)-2,4-pentadienyl)piperazine, 1-(4,4′-difluoro(or dichloro or dibromo)-benzhydryl)-4-(5-(3,5-dimethoxy-4-hydroxyphenyl)-2,4-pentadienyl) piperazine, 1-(4,4′-difluorobenzhydryl)-4-(5-(2,4-dimethoxyphenyl)-2,4-pentadienyl)piperazine, 1-benzhydryl-4-(5-(2,3,4-trimethoxyphenyl)-2,4-pentadienyl)piperazine, and 1-(4,4′-difluorobenzhydryl)-4-(5-(2,3,4-trimethoxyphenyl)-2,4-pentadienyl)piperazine, and indazole replacement bioisosteres of lomerizene described in U.S. Pat. No. 6,391,872.
  • Bepridil
  • Bepridil and analogs of bepridil are described by general formula (I) of U.S. Pat. No. 3,962,238. Other analogs are described by the general formula of claim 1 in U.S. Pat. No. 4,645,778, e.g., 2-(N-pyrrolidino)-3-isobutoxy-N-(3,4-methylenedioxyphenyl)-N-benzyl-propyl amine, 2-(N-pyrrolidino)-3-isobutoxy-N(2,6-dimethylphenyl)-N-benzyl-propylamine, and 2-(N-pyrrolidino)-3-isobutoxy-N-(3,4-dichlorophenyl)-N-benzyl propylamine; by formula (I) of U.S. Pat. No. 4,727,072, e.g., N-(2,6-dichlorophenyl)-beta-[[(1-methylcyclohexyl)methoxy]methyl]-N-(phenyl methyl)-1-pyrrolidineethanamine, beta-[(2,2-dimethylpropoxy)methyl]-N-(3-methoxyphenyl)-N-(phenylmethyl)-1-pyrrolidineethanamine, N-(2-chlorophenyl)-beta-[(2,2-dimethylpropoxy)methyl]-N-(phenylmethyl)-1-pyrrolidineethanamine, N-(2,6-dichlorophenyl)-N-[[4-(dimethylamino)phenyl]methyl]-beta-[(2,2-dimethylpropoxy)methyl]-1-pyrrolidineethanamine, N-(2,6-dichlorophenyl)-N-[(3,4-dimethoxyphenyl)methyl]-beta-[(2,2-dimethylpropoxy)methyl]-1-pyrrolidineethanamine, N-(2,6-dichlorophenyl)-beta-[(2,2-dimethylpropoxy)methyl]-N-(phenylmethyl)-1-pyrrolidineethanamine, N-(2,6-dichlorophenyl)-beta-[(2,2-dimethylpropoxy)methyl]-N-(4-pyridinylmethyl)-1-pyrrolidineethanamine, N-(2,6-dimethylphenyl)-beta-[(2,2-dimethylpropoxy)methyl]-N-(phenylmethyl)-1-pyrrolidineethanamine, beta-[(2,2-dimethylpropoxy)methyl]-N-(phenylmethyl)-N-[3-(trifluoromethyl)phenyl]-1-pyrrolidineethanamine, beta-[(2,2-dimethylpropoxy)methyl]-N-phenyl-N-(phenylmethyl)-1-pyrrolidinee thanamine, N-(cyclohexylmethyl)-N-(2,6-dichlorophenyl)-beta-[(2,2-dimethylpropoxy)methyl]-1-pyrrolidineethanamine, and N-(2,6-dichlorophenyl)-beta-[(1,1-dimethylethoxy)methyl]-N-(phenylmethyl)-1-pyrrolidineethanamine; by the formula of claim 1 or the compound 2-diethylamino-3-isobutoxy-N-phenyl-N-2-furanyl-methyl-propylamine described in U.S. Pat. No. 4,923,889, e.g., 2-(N-pyrrolidino)-3-isobutoxy-N-phenyl-N-(4-methoxy-benzyl)propylamine, 2-diethylamino-3-isobutoxy-N-phenyl-N-2-furanyl-methyl-propylamine, and 2-(N-pyrrolidino)-3-isoamyloxy-N-phenyl-N-(4-methoxy-benzyl)propylamine; by the formula of claim 1 in U.S. Pat. No. 4,927,834, e.g., 2-(N-pyrrolidino)-3-methallyloxy-N-benzyl-N-(3,4-dioxymethylenephenyl)-propylamine, 2-(N-pyrrolidino)-3-methallyloxy-N-phenyl-N-(4-methoxy-benzyl)propylamine, 2-(N-pyrrolidino)-3-methallyloxy-N-(3-methoxy-phenyl)-N-(4-methoxy-benzyl)-propylamine, 2-(N-pyrrolidino)-3-methallyloxy-N-(4-methoxy-phenyl)-N-(4-methoxy-benzyl)-propylamine, and 2-(N-pyrrolidino)-3-isopentenyloxy-N-phenyl-N-benzyl-propylamine; by the general formula of claim 1 in U.S. Pat. No. 4,999,361, e.g., ethyl 2,2-bis-(4-methoxyphenyl)-4-(1-pyrrolidino)-5-isobutoxyvalerate, 2-phenyl-2-(2-pyridyl)-4-(1-pyrrolidino)-5-isobutoxyvaleronitrile, ethyl 2,2-diphenyl-4-(N,N-diethylamino)-5-isobutoxyvalerate, ethyl 2,2-diphenyl-4-(1-pyrrolidino)-5-benzyloxyvalerate, and ethyl 2,2-diphenyl-4-(1-pyrrolidino)-5-(2-picolyloxy)valerate.
  • Analogs of lomerizine and bepridil with calcium-channel blocking activity include clentiazem (U.S. Pat. No. 4,567,175), gallopamil (U.S. Pat. No. 3,261,859), mibefradil (U.S. Pat. No. 4,808,605), prenylamine (U.S. Pat. No. 3,152,173), semotiadil (U.S. Pat. No. 4,786,635), terodiline (U.S. Pat. No. 3,371,014), aranipine (U.S. Pat. No. 4,572,909), bamidipine (U.S. Pat. No. 4,220,649), benidipine (EP Patent Application Publication No. 106,275), cilnidipine (U.S. Pat. No. 4,672,068), efonidipine (U.S. Pat. No. 4,885,284), elgodipine (U.S. Pat. No. 4,962,592), lacidipine (U.S. Pat. No. 4,801,599), manidipine (U.S. Pat. No. 4,892,875), nifendipine (U.S. Pat. No. 3,485,847), nilvadipine (U.S. Pat. No. 4,338,322), flunarizine (U.S. Pat. No. 3,773,939) lidoflazine (U.S. Pat. No. 3,267,104), bencyclane (Hungarian Pat. No. 151,865), etafenone (German Pat. No. 1,265,758), and perhexyline (British Pat. No. 1,025,578).
  • Analogs of lomerizine with vasodilating activity may also be employed in certain embodiments of the inventions. Examples are cinnarizine, citicoline (which may be isolated from natural sources as disclosed in Kennedy et al. (J. Am. Chem. Soc., 77:250 (1955)) or synthesized as disclosed in Kennedy (J. Biol. Chem., 222:185 (1956)), cyclandelate (see U.S. Pat. No. 3,663,597), ciclonicate (see German Pat. No. 1,910,481), diisopropylamine dichloroacetate (see GB Pat. No. 862,248), eburnamonine (see Hermann et al., J. Am. Chem. Soc., 101:1540 (1979)), fenoxedil (see U.S. Pat. No. 3,818,021), flunarizine (see U.S. Pat. No. 3,773,939), nafronyl (see U.S. Pat. No. 3,334,096), nicametate (Blicke et al., J. Am. Chem. Soc. 64:1722 (1942)), nimodipine (see U.S. Pat. No. 3,799,934), papaverine (see Goldberg, Chem. Prod. Chem. News 17:371 (1954)); tinofedrine (see U.S. Pat. No. 3,563,997), and viquidil (see U.S. Pat. No. 2,500,444).
  • Selective Serotonin Reuptake Inhibitors
  • Selective serotonin reuptake inhibitors may be employed in certain embodiments of the invention. By “selective serotonin reuptake inhibitor” or “SSRI” is meant any member of the class of compounds that (i) inhibit the uptake of serotonin by neurons of the central nervous system, (ii) have an inhibition constant (Ki) of 10 nM or less, and (iii) a selectivity for serotonin over norepinephrine (i.e., the ratio of Ki(norepinephrine) over Ki(serotonin)) of greater than 100. SSRIs that may be particularly useful are paroxetine, sertraline, and UK-416244.
  • Paroxetine
  • Paroxetine is an SSRI described in U.S. Pat. No. 3,912,743. Non-limiting examples of paroxetine analogs are compounds described by formula IV of U.S. Pat. No. 4,485,109, e.g., 4-phenyl-a, a,1-trimethyl-4-piperidinemethanol; (−)-trans-4-(4-fluorophenyl)-3-(4-methoxyphenoxy)methylpiperidine and (−)-trans-4-(4-fluorophenyl)-3-[(4-methoxyphenoxy)methyl]-1-methylpiperidine, described in U.S. Pat. No. 4,585,777 and U.S. Pat. No. 4,593,036, respectively; compounds described by formula I of U.S. Pat. No. 4,985,446, e.g., (−)-trans-44-4-fluorophenyl)-3-(3,4-methylenedioxyphenoxymethyl)-1-(2-methoxymethyl)-piperidine hydrochloride, (−)-trans-1-ethyl-4-(4-fluorophenyl)-3-(4-methoxyphenoxy-methyl)-piperidine hydrochloride, trans-1-ethyl-4-(4-fluorophenyl)-3-(4-t-butylphenoxymethyl)-piperidine hydrochloride, and trans 3-(4-benzyloxyphenoxymethyl)-4-(4-fluorophenyl)-1-methylpiperidine; compounds of formula I in U.S. Pat. No. 5,019,582, e.g., (−)-trans-44-4-fluorocyclopentyl)-3-(3,4-methylenedioxyphenoxymethyl)-1-p entylpiperidine hydrochloride, (+)-trans-4-phenyl-3-(3,4-methylenedioxyphenoxymethyl)-1-pentylpiperidine hydrochloride, (−)-trans-4-(−4-fluorophenyl)-3-(4-methoxyphenoxymethyl)-1-pentylpiperidine hydrochloride, (−)-trans-4(−4-fluorophenyl)-3-(5,6,7,8-tetrahydro-2-naphthoxymethyl)-1-pentylpiperidine hydrochloride, (−)-trans-4-(4-fluorophenyl)-3(3,4-methylenedioxyphenoxymethyl)-1-pentylpiperidine, and (−)-trans-4-(4-fluorophenyl)-3-(3,4-methylenedioxyphenoxymethyl)-1-pentylpiperidinee hydrochloride; compounds of formula I of U.S. Pat. No. 5,158,961, e.g., (−)-trans-4-(4-methoxyphenyl)-3-(3,4-methylenedioxyphenoxymethyl)-1-pentyl piperidine oxalate, (+)-trans-4-(4-methoxyphenyl)-3-(3,4-methylenedioxyphenoxymethyl)-1-pentyl-piperidine oxalate, (+/−)-trans-3-(3,4-methylenedioxyphenoxymethyl)-1-pentyl-4-(3-trifluoromethylphenyl)piperidine hydrochloride, (+)-trans-3-(2-bromo-4,5-methylenedioxyphenoxymethyl)-4-(4-fluorophenyl)-1-pentylpiperidine hydrochloride, and (+/−)-trans-3-(3,4-methylenedioxyphenoxymethyl)-1-pentyl-4(4-pentyloxyphenyl)piperidine hydrochloride; and compounds of formula I of U.S. Pat. No. 5,328,917, e.g., 1-butyl-3-(4-methoxybenzylaminomethyl)-4-phenylpiperidine, 1-butyl-3-(4-trifluoromethylphenylaminomethyl)-4-phenylpiperidine, and (−) trans-1-butyl-3-(2-phenylethylaminomethyl)-4-phenylpiperidine.
  • Sertraline
  • In certain embodiments, sertraline or an analog thereof can be used in the compositions, methods, and kits of the invention. Sertraline has the structure:
  • Figure US20130289024A1-20131031-C00015
  • Structural analogs of sertraline include those having the formula:
  • Figure US20130289024A1-20131031-C00016
  • where R1 and R2 are independently selected from the group consisting of H, optionally substituted C1-6 alkyl (e.g., CH3, (CH2)—OH, cyclopropyl, (CH2)xCOOH, or CH2CHOH(CH2)x, (CH2)xN(CfH3)2, where x is 1, 2, 3, 4, or 5), and optionally substituted C1-7 heteroalkyl (e.g., CH2CH2N(CH3)2) or R1 and R2 together form a C3-8 cycloalkyl optionally heterocyclic, optionally substituted (e.g., forming a morpholine ring), R3, R4, R5, and R6 are independently H, Cl, F, Br, OH, or optionally substituted C1-6 alkyl; X and Y are each selected from the group consisting of H, F, Cl, Br, CF3, C1-6 alkoxy (e.g., OPh and OCH3), and cyano; and W is selected from the group consisting of H, F, Cl, Br, CF3, C1-3 alkoxy, COOH, CH2CH2OH, NHCOH, NHCOCH3, CH2S(O)nCH3, CH2NH2, CONH2, CH2OH, NHCOPh, CH2NHS(O)nCH3, NHS(O)nPh, N(CH3)2, S(O)nNH2, NHCOBu, NHS(O)nCH3, NHCOcyclopentyl, CN, NHS(O)ncyclopropyl, NH2, NO2, I, SO2N(CH3)2, SO2NHMe, SO2NHCH2CH2OH, CO2Me, NHSO2Bu, CONHCH3, CH2NHCOCH3, CONHPh,
  • Figure US20130289024A1-20131031-C00017
  • CONHcylopropyl, C(S)NH2, NHC(S)CH3, CONHCH2COOCH3, CONHCH2COOH, CONHCH2cyclopropyl, CONHcyclobutyl, NHCOcyclopropyl, NH(CH3)COCH3, and CH2S(O)nR11, where n is 0, 1, or 2 and R11 is phenyl, C2-6 heterocyclyl, optionally substituted C1-8 alkyl (e.g., C4-8 unsubstituted alkyl such as Bu or C3-8 substituted alkyl). In certain embodiments, R1 is CH3 and R2 is CH3, CH2CH2OH, cyclopropyl, CH2COOH, CH2CH2NH2, CH2CH(OH)R8, or CH2CH(R8)NR9R10, where n is 0, 1, or 2 and R8, R9, and R10 are independently H or C1-6 alkyl. In certain embodiments, X is H and Y is p-OPh, p-OCF3, o-OCH3 m-OCH3, or p-OCH3. In certain embodiments of the above structure, the sertraline analog has the formula:
  • Figure US20130289024A1-20131031-C00018
  • Other sertraline analogs have the formula:
  • Figure US20130289024A1-20131031-C00019
  • where R3, R4, R5, R6, W, X, and Y are as defined above, and R7 is independently H, NH(CH2)mCH3, O(CH2)mCH3, OH, O(CH2)mCH3, ═O, C1-6 alkyl (e.g., isopropyl), or C1-6 alkyoxy, where m is 0, 1, 2, 3, 4, 5, or 6. In certain embodiments, R3, R4, R5, and R6 are H; X and Y are each Cl at the 3 and 4 positions of the benzyl ring. Exemplary analogs include:
  • Figure US20130289024A1-20131031-C00020
  • Other sertraline analogs have the formula:
  • Figure US20130289024A1-20131031-C00021
  • where R1, R2, R3, R4, R5, R6, X and Y are as defined above, and R7 is H or C1-6 optionally substituted alkyl. Other sertraline analogs are described by the formula:
  • Figure US20130289024A1-20131031-C00022
  • wherein R8, R9, and R10 are independently H, optionally substituted C1-6 alkyl (e.g., CH3, (CH2)xOH, cyclopropyl, (CH2)xCOOH, or CH2CHOH(CH2)x, (CH2)xN(CH3)2, where x is 1, 2, 3, 4, or 5), and optionally substituted C1-7 heteroalkyl (e.g., CH2CH2N(CH3)2).
  • In certain embodiments, sertraline analogs are in the cis-isomeric configuration. The term “cis-isomeric” refers to the relative orientation of the NR1R2 and phenyl moieties on the cyclohexene ring (i.e., they are both oriented on the same side of the ring). Because both the 1- and 4-carbons are asymmetrically substituted, each cis-compound has two optically active enantiomeric forms denoted (with reference to the 1-carbon) as the cis-(1R) and cis-(1S) enantiomers. Sertraline analogs are also described in U.S. Pat. No. 4,536,518. Other related compounds include (S,S)—N-desmethylsertraline, rac-cis-N-desmethylsertraline, (1S,4S)-desmethyl sertraline, 1-des (methylamine)-1-oxo-2-(R,S)-hydroxy sertraline, (1R,4R)-desmethyl sertraline, sertraline sulfonamide, sertraline (reverse) methanesulfonamide, 1R,4R sertraline enantiomer, N,N-dimethyl sertraline, nitro sertraline, sertraline aniline, sertraline iodide, sertraline sulfonamide NH2, sertraline sulfonamide ethanol, sertraline nitrile, sertraline-CME, dimethyl sertraline reverse sulfonamide, sertraline reverse sulfonamide (CH2 linker), sertraline B-ring ortho methoxy, sertraline A-ring methyl ester, sertraline A-ring ethanol, sertraline N,N-dimethylsulfonamide, sertraline A-ring carboxylic acid, sertraline B-ring para-phenoxy, sertraline B-ring para-trifluoromethane, N,N-dimethyl sertraline B-Ring para-trifluoromethane, sertraline A-ring methyl sulfoxide (CH2 linker), sertraline A-ring carboxamide, sertraline A-ring reverse carboxamide, Sertraline A-ring methanamine, sertraline A-ring sulfonylmethane (CH2 linker), sertraline (reverse) methane sulfonamide, sertraline A-ring thiophene, reduced sulfur sertraline A-ring methyl sulfoxide (CH2 linker), and heterocyclic substituted stertraline (reverse) methanesulfonamide. Structures of these analogs are shown in Table 4.
  • TABLE 4
    Sertraline analogs.
    Figure US20130289024A1-20131031-C00023
    (1R,4S) Sertraline Hydrochloride
    Figure US20130289024A1-20131031-C00024
    (1S,4R) Sertraline Hydrochloride
    Figure US20130289024A1-20131031-C00025
    Sertraline B-Ring Para-Phenoxy
    Figure US20130289024A1-20131031-C00026
    Sertraline B-Ring Ortho-Methoxy
    Figure US20130289024A1-20131031-C00027
    1R,4R Sertraline Enantiomer
    Figure US20130289024A1-20131031-C00028
    Sertraline Sulfonamide
    Figure US20130289024A1-20131031-C00029
    Nitro Sertraline
    Figure US20130289024A1-20131031-C00030
    Sertraline Aniline
    Figure US20130289024A1-20131031-C00031
    Sertraline Reverse Sulfonamide (CH2 linker)
    Figure US20130289024A1-20131031-C00032
    (1R,4R)-Desmethyl Sertraline
    Figure US20130289024A1-20131031-C00033
    Sertraline A-Ring Methyl Ester
    Figure US20130289024A1-20131031-C00034
    rac-cis-N-Desmethyl Sertraline, Hydrochloride
    Figure US20130289024A1-20131031-C00035
    Dimethyl Sertraline Reverse Sulfonamide
    Figure US20130289024A1-20131031-C00036
    Sertraline N,N-Dimethylsulfonamide
    Figure US20130289024A1-20131031-C00037
    Sertraline A-Ring Ethanol
    Figure US20130289024A1-20131031-C00038
    Sertraline-CME
    Figure US20130289024A1-20131031-C00039
    (1S,4S)-Desmethyl Sertraline, Hydrochloride
    Figure US20130289024A1-20131031-C00040
    Sertraline Iodide
    Figure US20130289024A1-20131031-C00041
    1-Des(methylamine)-1-oxo-2-(R,S)-hydroxy Sertraline
    Figure US20130289024A1-20131031-C00042
    Sertraline Nitrile
    Figure US20130289024A1-20131031-C00043
    N,N-Dimethyl Sertraline B-Ring Para-Trifluoromethane
    Figure US20130289024A1-20131031-C00044
    Sertraline Sulfonamide NH2
    Figure US20130289024A1-20131031-C00045
    Sertraline (Reverse) Methanesulfonamide
    Figure US20130289024A1-20131031-C00046
    Sertraline A-Ring Carboxylic Acid
    Figure US20130289024A1-20131031-C00047
    Sertraline Sulfonamide Ethanol
    Figure US20130289024A1-20131031-C00048
    Sertraline B-Ring Para-Trifluoromethane
    Figure US20130289024A1-20131031-C00049
    N,N-Dimethyl Sertraline
    Figure US20130289024A1-20131031-C00050
    Sertraline A-ring Methyl Sulfoxide (CH2 Linker)
    Figure US20130289024A1-20131031-C00051
    Sertraline A-ring carboxamide
    Figure US20130289024A1-20131031-C00052
    Sertraline A-ring reverse carboxamide
    Figure US20130289024A1-20131031-C00053
    Sertraline A-Ring methanamine
    Figure US20130289024A1-20131031-C00054
    Sertraline A-Ring Sulfonylmethane (CH2-Linker)
    Figure US20130289024A1-20131031-C00055
    Sertraline (Reverse) Methanesulfonamide
    Figure US20130289024A1-20131031-C00056
    Sertraline A-ring Thiophene
    Figure US20130289024A1-20131031-C00057
    Sertraline A-ring Methylsulfide
    Figure US20130289024A1-20131031-C00058
    Sertraline A-ring Methylimidazole Reverse Sulfonamide
    Figure US20130289024A1-20131031-C00059
    Sertraline A-ring reverse formamide
    Figure US20130289024A1-20131031-C00060
    N,N-Dimethyl Sertraline A-ring Reverse benzamide
    Figure US20130289024A1-20131031-C00061
    Sertraline A-ring Methylcarboxamide
    Figure US20130289024A1-20131031-C00062
    Sertraline A-ring Reverse Carobxamide (CH2 linker)
    Figure US20130289024A1-20131031-C00063
    Sertraline A-ring Benzamide
    Figure US20130289024A1-20131031-C00064
    Sertraline A-ring Pyridine Carboxamide
    Figure US20130289024A1-20131031-C00065
    Setraline cyclopropyl carboxamide
    Figure US20130289024A1-20131031-C00066
    Sertraline A-Ring Thiocarboxamide
    Figure US20130289024A1-20131031-C00067
    Sertraline A-Ring Reverse Thiocarboxamide
    Figure US20130289024A1-20131031-C00068
    N-methyl,N—CH2CH2N(CH3)2 Sertraline
    Figure US20130289024A1-20131031-C00069
    Sertraline B-ring 2-thiophene
    Figure US20130289024A1-20131031-C00070
    Sertraline without B-ring
    Figure US20130289024A1-20131031-C00071
    N-ethanol Sertraline
    Figure US20130289024A1-20131031-C00072
    N-cyclopropyl Sertraline
    Figure US20130289024A1-20131031-C00073
    Sertraline B-Ring p-methoxy
    Figure US20130289024A1-20131031-C00074
    N,N-dimethyl Sertraline A-ring Carboxylic Acid
    Figure US20130289024A1-20131031-C00075
    Sertraline B-ring m-Methoxy
    Figure US20130289024A1-20131031-C00076
    N,N-Dimethyl Sertraline A-Ring Carboxamide
    Figure US20130289024A1-20131031-C00077
    4S-Sertraline Ketone
    Figure US20130289024A1-20131031-C00078
    Sertraline A-Ring Butane Reverse Sulfonamide
    Figure US20130289024A1-20131031-C00079
    Sertraline A-Ring Reverse Pentanamide
    Figure US20130289024A1-20131031-C00080
    Alcohol Sertraline
    Figure US20130289024A1-20131031-C00081
    Sertraline A-ring Cycloproprane Reverse Sulfonamide
    Figure US20130289024A1-20131031-C00082
    Sertraline A-ring Benzene Reverse Sulfonamide
    Figure US20130289024A1-20131031-C00083
    Sertraline A-Ring Reverse Benzamide
    Figure US20130289024A1-20131031-C00084
    Sertraline A-Ring N,N-Dimethylamine
    Figure US20130289024A1-20131031-C00085
    Methoxy 4S-Sertraline
    Figure US20130289024A1-20131031-C00086
    Sertraline A-Ring Reverse cyclopetanecarboxamide
    Figure US20130289024A1-20131031-C00087
    Sertraline A-Ring Methyl sulfide
    Figure US20130289024A1-20131031-C00088
    Sertraline A-Ring Phenylsulfone (CH2 linker)
    Figure US20130289024A1-20131031-C00089
    Sertraline A-Ring Methyltriazolesulfone (CH2 linker)
    Figure US20130289024A1-20131031-C00090
    Sertraline A_Ring Methyltriazolesulfoxide (CH2 linker)
    Figure US20130289024A1-20131031-C00091
    Isopropyl Sertraline
    Figure US20130289024A1-20131031-C00092
    Isopropyl Sertraline reverse carboxamide
    Figure US20130289024A1-20131031-C00093
    Isopropyl Sertraline carboxamide
    Figure US20130289024A1-20131031-C00094
    Isopropyl Sertraline methanamine
    Figure US20130289024A1-20131031-C00095
    N-ethanol Sertraline A-ring reverse carboxamide
    Figure US20130289024A1-20131031-C00096
    N-ethanol Sertraline A-ring sulfoxide (CH2 linker)
    Figure US20130289024A1-20131031-C00097
    N-(N,N-dimethyl)ethyl Sertraline
    Figure US20130289024A1-20131031-C00098
    N-morpholine Sertraline
    Figure US20130289024A1-20131031-C00099
    Sertraline cyclopropyl (CH2 linker) carboxamide
    Figure US20130289024A1-20131031-C00100
    Sertraline cyclobutyl carboxamide
    Figure US20130289024A1-20131031-C00101
    N,N dimethyl Sertraline cyclopropyl carboxamide
    Figure US20130289024A1-20131031-C00102
    N,N-dimethyl Sertraline phenylcarboxamide
    Figure US20130289024A1-20131031-C00103
    Sertraline reverse cyclopropyl carboxamide
    Figure US20130289024A1-20131031-C00104
    Sertraline reverse phenylcarboxamide
    Figure US20130289024A1-20131031-C00105
    Sertraline A-Ring Methyl Acetate Carboxamide
    Figure US20130289024A1-20131031-C00106
    Sertraline A-Ring Acetic Acid Carboxamide
    Figure US20130289024A1-20131031-C00107
    Sertraline A-Ring Cyclopropyl Carboxamide (CH2 linker)
    Figure US20130289024A1-20131031-C00108
    Sertraline A-Ring Cyclobutyl Carboxamide
    Figure US20130289024A1-20131031-C00109
    No-N Sertraline
    Figure US20130289024A1-20131031-C00110
    N,N Dimethyl Sertraline A-Ring Reverse Carboxamide (CH2 linker)
    Figure US20130289024A1-20131031-C00111
    Sertraline A-ring N-methyl reverse carboxamide
  • Particularly useful are the following compounds, in either the (1S)-enantiomeric or (1S)(1R) racemic forms, and their pharmaceutically acceptable salts: cis-N-methyl-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydro-1-naphthalenamine; cis-N-methyl-4-(4-bromophenyl)-1,2,3,4-tetrahydro-1-naphthalenamine; cis-N-methyl-4-(4-chlorophenyl)-1,2,3,4-tetrahydro-1-naphthalenamine; cis-N-methyl-4-(3-trifluoromethyl-phenyl)-1,2,3,4-tetrahydro-1-naphthalenamine; cis-N-methyl-4-(3-trifluoromethyl-4-chlorophenyl)-1,2,3,4-tetrahydro-1-naphthalenamine; cis-N,N-dimethyl-4-(4-chlorophenyl)-1,2,3,4-tetrahydro-1-naphthalenamine; cis-N,N-dimethyl-4-(3-trifluoromethyl-phenyl)-1,2,3,4-tetrahydro-1-naphthalenamine; and cis-N-methyl-4-(4-chlorophenyl)-7-chloro-1,2,3,4-tetrahydro-1-naphthalenamine. Of interest also is the (1R)-enantiomer of cis-N-methyl-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydro-1-naphthalenamine.
  • UK-416244
  • UK-416244 is an SSRI that is phenoxybenzylamine derivative. UK-416244 has the structure:
  • Figure US20130289024A1-20131031-C00112
  • Structural analogs of UK-416244 are compounds having the formula:
  • Figure US20130289024A1-20131031-C00113
  • where R1 and R2, independently, are H, C1-6 alkyl (e.g., CH3) or substituted heteroalkyl, or (CH2)d(C3-6 cycloalkyl) where d is 0, 1, 2, or 3; or R1 and R2 together with the nitrogen to which they are attached form an azetidine ring; Z or Y is —S(O)nR3 and the other Z or Y is halogen or —R3; where R3 is independently C1-4 alkyl optionally substituted with fluorine (e.g., where R3 is or is not CF3) and n is 0, 1, or 2; or Z and Y are linked so that, together with the interconnecting atoms, Z and Y form a fused 5 to 7-membered carbocyclic or heterocyclic ring which may be saturated, unsaturated, or aromatic, and where when Z and Y form a heterocyclic ring, in addition to carbon atoms, the linkage contains one or two heteroatoms independently selected from O, S, and N; (e.g., with the proviso that when R5 is F and R2 is methyl then the fused ring is not 1,3-dioxolane and Z and Y together do not form a fused phenyl ring); R4 and R5 are, independently, A-X, where A is —CH═CH— or —(CH2)p— where p is 0, 1, or 2; X is H, F, Cl, Br, I, NH2, OH, CONR6R7, SO2NR6R7, SO2NHC(═O)R6, C1-4 alkoxy, NR8SO2R9, NO2, NR6R11 (e.g., N(CH3)2, CN, CO2R10 (e.g., COOH), CHO, SR10, S(O)R9 or SO2R10; R6, R7, R8 and R10 independently are H, C1-6 alkyl (e.g., CH3, (CH2)3CH3 or cyclopropyl), C6-12 aryl (e.g., phenyl) optionally substituted independently by one or more R12, or C1-6 alkyl-aryl optionally substituted (e.g., CH2Ph); R9 is C1-6 alkyl optionally substituted independently by one or more R12; R11 is H, C1-6 alkyl optionally substituted independently by one or more R12, C(O)R6, CO2R9, C(O)NHR6, or SO2NR6R7; R12 is F (preferably up to 3), OH, CO2H, C3-6 cycloalkyl, NH2, CONH2, C1-6 alkoxy, C1-6 alkoxycarbonyl, or a 5- or 6-membered heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, S, and O optionally substituted independently by one or more R13; or R6 and R7, together with the nitrogen to which they are attached, form a 4-, 5-, or 6-membered heterocyclic ring optionally substituted independently by one or more R13; or a 5- or 6-membered heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, S, and O optionally substituted independently by one or more R13; where R13 is hydroxy, C1-4 alkoxy, F, C1-6 alkyl, haloalkyl, haloalkoxy, —NH2, —NH(C1-6 alkyl), or —N(C1-6 alkyl)2-;
  • or compounds having the formula:
  • Figure US20130289024A1-20131031-C00114
  • where R1 and R2 are independently H, C1-6 alkyl (e.g., CH3) or substituted heteroalkyl, (CH2)m(C3-6 cycloalkyl) where m is 0, 1, 2, or 3, or R1 and R2 together with the nitrogen to which they are attached form an azetidine ring; each R3 is independently H, I, Br, F, Cl, C1-6 alkyl (e.g., CH3), CF3, CN, OCF3, C1-4 alkylthio (e.g., SCH3), C1-4 alkoxy (e.g., OCH3), aryloxy (e.g., OPh), or CONR6R7; n is 1, 2, or 3; and R4 and R5 are independently A-X, where A is —CH═CH— or —(CH2)p— where p is 0, 1, or 2; X is H, F, Cl, Br, I, CONR6R7, SO2NR6R7, SO2NHC(═O)R6, OH, C1-4 alkoxy, NR8SO2R9, NO2, NR6R11, CN, CO2R10 (e.g., COOH), CHO, SR10, S(O)R9, or SO2R10; R6, R7, R8, and R10 are independently H or C1-6 alkyl (e.g., (CH2)3CH3 or cyclopropyl), C6-12 aryl (e.g., phenyl) optionally substituted independently by one or more R12, or C1-6 alkyl-aryl optionally substituted; R9 is C1-6 alkyl optionally substituted independently by one or more R12; R11 is H, C1-6 alkyl optionally substituted independently by one or more R12, C(O)R6, CO2R9, C(O)NHR6, or SO2NR6R7; R12 is F (preferably up to 3), OH, CO2H, C3-6 cycloalkyl, NH2, CONH2, C1-6 alkoxy, C1-6 alkoxycarbonyl or a 5- or 6-membered heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, S, and O optionally substituted independently by one or more R13; or R6 and R7, together with the nitrogen to which they are attached, form a 4-, 5-, or 6-membered heterocyclic ring optionally substituted independently by one or more R13; or a 5- or 6-membered heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, S, and O optionally substituted independently by one or more R13; where R13 is hydroxy, C1-4 alkoxy, F, C1-6 alkyl, haloalkyl, haloalkoxy, —NH2, —NH(C1-6 alkyl) or —N(C1-6 alkyl)2 (e.g., where when R1 and R2 are methyl, R4 and R5 are hydrogen and n is 1, R3 is not a —SMe group para to the ether linkage linking rings A and B). In certain embodiments, n is 1 or 2, and the R3 group(s) is/are at positions 3 and/or 4 of the B ring, for example, are CH3, SCH3, OCH3, Br, or CF3. For either of the above structures, R4 or R5 can be SO2NHPh, SO2NHCH3, CN, H, Br, CONH2, COOH, SO2NHCH2Ph, SO2NHCOCH3, CH2NHSO2CH3 NH2, OR NO2, benzyl amide, acylsulfonamide, reverse sulfonamide, NHCH3, N(CH3)2, SO2NH2, CH2OH, NHSO2CH3, SO2NHCH2CCH2, CH2NH2, SO2NHBu, and SO2NHcyclopropyl. UK-416244 structural analogs are described in U.S. Pat. Nos. 6,448,293 and 6,610,747. UK-416244 analogs are described below.
  • Other analogs of UK-416244 can be described by the formula:
  • Figure US20130289024A1-20131031-C00115
  • where are R3, R4, and R5 are as defined above and Z is CH2NR1R2 where R1 and R2 are as defined above, C1-6 alkyl, optionally substituted (e.g., with hydroxyl, NH2, NHC1-6 alkyl). In certain embodiments, Z is CH2CH(CH3)2, CH2OCH3, CH2N(CH3)CH2CH2OH, N(CH3)2, CH2N(CH3)2, COOH, CH2NHCH3, CH2OH, CH2NHCOCH3, or CONHCH3.
  • Other UK-416244 analogs are described by the formula.
  • Figure US20130289024A1-20131031-C00116
  • where R1 is H, I, Br, F, Cl, C1-6 alkyl (e.g., CH3), CF3, CN, OCF3, C1-4 alkylthio (e.g., SCH3), C1-4 alkoxy (e.g., OCH3), aryloxy, or CONR2R3; n is 1, 2, or 3; R2 and R3 are independently H or C1-6 alkyl (e.g., (CH2)3CH3 or cyclopropyl), C6-12 aryl (e.g., phenyl) optionally substituted independently by one or more R4, or C1-6 alkyl-aryl optionally substituted; R4 is F (preferably up to 3), OH, CO2H, C3-6 cycloalkyl, NH2, CONH2, C1-6 alkoxy, C1-6 alkoxycarbonyl or a 5- or 6-membered heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, S, and O optionally substituted independently by one or more R5; or R2 and R3, together with the nitrogen to which they are attached, form a 4-, 5-, or 6-membered heterocyclic ring optionally substituted independently by one or more R5; or a 5- or 6-membered heterocyclic ring containing 1, 2, or 3 heteroatoms selected from N, S, and O optionally substituted independently by one or more R5; where R5 is hydroxy, C1-4 alkoxy, F, C1-6 alkyl, haloalkyl, haloalkoxy, —NH2, —NH(C1-6 alkyl) or —N(C1-6 alkyl)2. In certain embodiments, where R1 is Br, OMe, NO2, CO2Me, or CN. R1 may be at the ortho, meta, or para position)
  • Still other UK-416244 analogs are described by the formula:
  • Figure US20130289024A1-20131031-C00117
  • where X is N, O, or S, and R1 is H, C1-6 alkyl or substituted heteroalkyl, (CH2)m(C3-6 cycloalkyl) where m is 0, 1, 2, or 3.
  • Additional analogs have the structure:
  • Figure US20130289024A1-20131031-C00118
  • where R1 is H or C1-6 alkyl (e.g., CH3, CH2CH3) and R2 is C1-6 alkyl substituted with OH, such as CH2OH, CH2CH2OH, CH(OH)CH3, CH2CH2CH2OH, CH(CH2)CH2OH, and CH2CH2CH2CH2OH, CH(OH)CH2CH2CH3, CH2CH(OH)CH2CH3, and CH2CH2CH(OH)CH3) or is CH2XR14 or CH2CH2XR14, where X is N, O, or S, and R14 is H, C1-6 alkyl or substituted heteroalkyl, (CH2)q(C3-6 cycloalkyl) where q is 0, 1, 2, or 3, and where R3, R4, and R5 are as defined above.
  • In certain embodiments, the analog has the structure:
  • Figure US20130289024A1-20131031-C00119
  • where R1 is H or C1-6 alkyl (e.g., CH3, CH2CH3) and R2 is C1-6 alkyl substituted with OH, e.g., CH2OH, CH2CH2OH, CH(OH)CH3, CH2CH2CH2OH, CH(CH2)CH2OH, and CH2CH2CH2CH2OH, CH(OH)CH2CH2CH3, CH2CH(OH)CH2CH3, and CH2CH2CH(OH)CH3). In particular embodiments, the compound is:
  • Figure US20130289024A1-20131031-C00120
  • UK-416244 analogs include those of Table 5:
  • TABLE 5
    UK-416244 analogs
    Figure US20130289024A1-20131031-C00121
    Compound 1
    Figure US20130289024A1-20131031-C00122
    Compound 2
    Figure US20130289024A1-20131031-C00123
    Compound 3
    Figure US20130289024A1-20131031-C00124
    Compound 4
    Figure US20130289024A1-20131031-C00125
    Compound 5
    Figure US20130289024A1-20131031-C00126
    Compound 6
    Figure US20130289024A1-20131031-C00127
    Compound 7
    Figure US20130289024A1-20131031-C00128
    Compound 8
    Figure US20130289024A1-20131031-C00129
    Compound 9
    Figure US20130289024A1-20131031-C00130
    Compound 10
    Figure US20130289024A1-20131031-C00131
    Compound 11
    Figure US20130289024A1-20131031-C00132
    Compound 12
    Figure US20130289024A1-20131031-C00133
    Compound 13
    Figure US20130289024A1-20131031-C00134
    Compound 14
    Figure US20130289024A1-20131031-C00135
    Compound 15
    Figure US20130289024A1-20131031-C00136
    Compound 16
    Figure US20130289024A1-20131031-C00137
    Compound 17
    Figure US20130289024A1-20131031-C00138
    Compound 18
    Figure US20130289024A1-20131031-C00139
    Compound 19
    Figure US20130289024A1-20131031-C00140
    Compound 20
    Figure US20130289024A1-20131031-C00141
    Compound 21
    Figure US20130289024A1-20131031-C00142
    Compound 22
    Figure US20130289024A1-20131031-C00143
    Compound 23
    Figure US20130289024A1-20131031-C00144
    Compound 24
    Figure US20130289024A1-20131031-C00145
    Compound 25
    Figure US20130289024A1-20131031-C00146
    Compound 26
    Figure US20130289024A1-20131031-C00147
    Compound 27
    Figure US20130289024A1-20131031-C00148
    Compound 28
    Figure US20130289024A1-20131031-C00149
    Compound 29
    Figure US20130289024A1-20131031-C00150
    Compound 30
    Figure US20130289024A1-20131031-C00151
    Compound 31
    Figure US20130289024A1-20131031-C00152
    Compound 32
    Figure US20130289024A1-20131031-C00153
    Compound 33
    Figure US20130289024A1-20131031-C00154
    Compound 34
    Figure US20130289024A1-20131031-C00155
    Compound 35
    Figure US20130289024A1-20131031-C00156
    Compound 36
    Figure US20130289024A1-20131031-C00157
    Compound 37
    Figure US20130289024A1-20131031-C00158
    Compound 38
    Figure US20130289024A1-20131031-C00159
    Compound 39
    Figure US20130289024A1-20131031-C00160
    Compound 40
    Figure US20130289024A1-20131031-C00161
    Compound 41
    Figure US20130289024A1-20131031-C00162
    Compound 42
    Figure US20130289024A1-20131031-C00163
    Compound 43
    Figure US20130289024A1-20131031-C00164
    Compound 44
    Figure US20130289024A1-20131031-C00165
    Compound 45
    Figure US20130289024A1-20131031-C00166
    Compound 46
    Figure US20130289024A1-20131031-C00167
    Compound 47
    Figure US20130289024A1-20131031-C00168
    Compound 48
    Figure US20130289024A1-20131031-C00169
    Compound 49
    Figure US20130289024A1-20131031-C00170
    Compound 50
    Figure US20130289024A1-20131031-C00171
    Compound 51
    Figure US20130289024A1-20131031-C00172
    Compound 52
    Figure US20130289024A1-20131031-C00173
    Compound 53
    Figure US20130289024A1-20131031-C00174
    Compound 54
    Figure US20130289024A1-20131031-C00175
    Compound 55
    Figure US20130289024A1-20131031-C00176
    Compound 56
  • Sertraline, UK-416244, and analogs thereof are considered herein to be equivalents in the methods, compositions, and kits of the invention.
  • SSRIs are considered herein to be analogs of paroxetine, sertraline, and UK-416244 and, thus, may be used in connection with the invention. SSRIs include cericlamine (e.g., cericlamine hydrochloride); citalopram (e.g., citalopram hydrobromide); clovoxamine; cyanodothiepin; dapoxetine; escitalopram (escitalopram oxalate); femoxetine (e.g., femoxetine hydrochloride); fluoxetine (e.g., fluoxetine hydrochloride); fluvoxamine (e.g., fluvoxamine maleate); ifoxetine; indalpine (e.g., indalpine hydrochloride); indeloxazine (e.g., indeloxazine hydrochloride); litoxetine; milnacipran (e.g., minlacipran hydrochloride); 6-nitroquipazine; tametraline hydrochloride; viqualine; and zimeldine (e.g., zimeldine hydrochloride). SNRIs (selective serotonin norepinephrine reuptake inhibitors), which include venlafaxine, duloxetine, and 4-(2-fluorophenyl)-6-methyl-2-piperazinothieno[2,3-d]pyrimidine, are also considered herein to be analogs of paroxetine, sertraline, and UK-416244 and, thus, may be used in connection with the invention. Pharmacologically active metabolites of any of the foregoing SSRIs and SNRIs can also be used in the methods, compositions, and kits of the invention. Exemplary metabolites are didesmethylcitalopram, desmethylcitalopram, desmethylsertraline, and norfluoxetine.
  • Other Compounds
  • Flupentixol
  • Flupentixol is a thioxanthene anti-psychotic drug. Flupentixol is described in U.S. Pat. No. 3,282,930, and exemplary analogs of flupentixol are described by formula I in U.S. Pat. No. 3,951,961, e.g., 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperazin-1-yl)-1-propenyl)thioxanthene, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperazin-1-yl)-1-propenyl)thioxanthene, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperazin-1-yl)-1-propenyl)thioxanthene dihydrochloride, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperidine) 1-propenyl)thioxanthene, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperidine) 1-propenyl)thioxanthene, and 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperidine) 1-propenyl)thioxanthene sulphate; by formula I of U.S. Pat. No. 4,022,896, e.g., 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperazinl-yl)-1-propen yl)thioxanthene, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperidine) 1-propenyl)thioxanthene, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperazinl-yl)-1-propenyl)thioxanthene, and 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperidine) 1-propenyl)thioxanthene; by formula I of U.S. Pat. No. 4,042,695, e.g., 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)-piperazine-1-yl)propyl)-thiaxanthene, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)-piperazine-1-yl)propyl)-thiaxanthene dihydrochloride, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)-piperazine-1-yl)propylidene)-thiaxanthene, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperazine-1-yl)propyl-thiaxanthene dihydrochloride, 2-trifluoromethyl-6-fluoro-9-(3-(piperazine-1-yl)propyl)thiaxanthene dioxalate salt, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperazine-1-yl)propyl)-thiaxanthene dihydrochloride, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)piperazine-1-yl)propyl)-thiaxanthene, 2-trifluoromethyl-6-fluoro-9-(3-(4-methyl-1-piperazinyl)propyl)thiaxanthene, 2-trifluoromethyl-6-fluoro-9-(3-(4-(2-hydroxyethyl)-piperazine-1-yl)propyl)-thiaxanthene palmitic acid ester, and 2-dimethylsulfamoyl-6-fluoro-9-(3-(4-methyl-piperazinyl)propyl-thiaxanthene; and by formula I of U.S. Pat. No. 4,044,024. Yet other examples are trifluoromethyl-6-fluoro-9-(3-dimethylaminopropylidene)-thiaxanthene and 2-trifluoromethyl-6-fluoro-9-(2-propenylidene)-thioxanthene.
  • Aripiprazole
  • Aripiprazole is an atypical antipsychotic described in U.S. Pat. No. 5,006,528. Exemplary analogs are given by variations of formula I in U.S. Pat. No. 5,006,528, e.g., 7-{4-[4-(b 2,3-dichlorophenyl)-1-piperazinyl]-butoxy}carbostyril, 7-{4-[4-(2-ethoxyphenyl)-1-piperazinyl]butoxy}-3,4-dihydrocarbostyril, and 7-{4-[4-]2-ethoxyphenyl)-1-piperazinyl]butoxy}-carbostyril. Additional exemplary analogs include compounds described in U.S. Pat. No. 4,234,584 and compounds of formula I in U.S. Pat. No. 7,053,092, e.g., 7-{4-[4-(2,3-dichlorophenyl)-1-piperazinyl]butoxyl}-3,4-dihydrocarbostyril; compounds of formula I of U.S. Pat. No. 7,160,888, e.g., 7-[4-(4-naphthalen-1-yl-piperazin-1-yl)-butoxy]-3,4-dihydro-1H-[1,8] napht-hyridin-2-one, 7-[4-(4-napthalen-1-yl-piperazin-1-yl)-butoxy]-3,4-dihydro-1H-[1,8]naphth-hyridin-2-one, and 7-[4-(4-naphthalen-1-yl-piperazin-1-yl)-butoxy]-3,4-dihydro-1H-[1,8]napht-hyridin-2-one.
  • Pimozide
  • Pimozide is a benzimidazole compound with anti-psychotic activities. Analogs of pimozide may include, e.g., 3-[1-[4,4-bis(4-fluorophenyl)butyl]piperidin-4-yl]-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)butyl]piperidin-4-yl]-6-fluoro-1H-benzimidazol-2-one, 1-[4,4-bis(4-fluorophenyl)butyl]-3-piperidin-4-ylbenzimidazol-2-one, 3-[2-[4-[bis(4-fluorophenyl)methyl]piperidin-1-yl]ethyl]-1H-benzimidazol-2-one, 4-[4,4-bis(4-fluorophenyl)butyl]spiro[1,4-d]azinan-1-ium-1,3′-1H-benzimidazol-3-ium]-2′-one, 3-[1-[4,4-bis(4-fluorophenyl)butyl]piperidin-4-yl]-6-iodo-1H-benzimidazol-2-one, 3-[3-[4-[bis(4-fluorophenyl)methyl]piperazin-1-yl]-2-methylpropyl]-1H-benzimidazol-2-one, 3-[3-[4-[bis(4-fluorophenyl)methyl]piperazin-1-yl]propyl]-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)butyl]-1-oxidopiperidin-1-ium-4-yl]-1H-benzimidazol-2-one, 3-[2-[4-[(4-fluoroanilino)-(4-fluorophenyl)methyl]piperidin-1-yl]ethyl]-1H-benzimidazol-2-one, 3-[2-[4-[bis(4-fluorophenyl)methyl]piperazin-1-yl]ethyl]-1H-benzimidazol-2-one, 3-[3-[4-[(4-fluorophenyl)-phenylmethyl]piperazin-1-yl]propyl]-1H-benzimidazol-2-one, 3-[4-[4-[bis(4-fluorophenyl)methyl]piperazin-1-yl]butyl]-1H-benzimidazol-2-one, 3-[(3R,6S)-6-(4-fluorophenyl)-1-(phenylmethyl)piperidin-3-yl]-1H-benzimidazol-2-one, 3-[(3R,6R)-6-(4-fluorophenyl)-1-(phenylmethyl)piperidin-3-yl]-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)butyl]piperidin-4-yl]-6-methyl-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)cyclohex-2-en-1-yl]piperidin-4-yl]-6-fluoro-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)cyclohex-2-en-1-yl]piperidin-4-yl]-1H-benzimidazol-2-one, 3-[5-[4-[bis(4-fluorophenyl)methyl]piperazin-1-yl]pentyl]-1H-benzimidazol-2-one, 3-[6-[4-[bis(4-fluorophenyl)methyl]piperazin-1-yl]hexyl]-1H-benzimidazol-2-one, 3-[4-[4-[(4-fluorophenyl)-phenylmethyl]piperazin-1-yl]butyl]-1H-benzimidazol-2-one, 3-[2-[4-[(4-fluorophenyl)-phenylmethyl]piperazin-1-yl]ethyl]-1H-benzimidazol-2-one, 3-[4,4-bis(4-fluorophenyl)butyl]-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)cyclohexyl]piperidin-4-yl]-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)cyclohex-2-en-1-yl]piperidin-4-yl]-6-fluoro-1H-benzimidazol-2-one hydrochloride, 3-[1-[4,4-bis(4-fluorophenyl)cyclohex-2-en-1-yl]piperidin-4-yl]-1H-benzimidazol-2-one hydrochloride, 3-[1-[3,3-bis(4-fluorophenyl)cyclopentyl]piperidin-4-yl]-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)cyclohexyl]piperidin-4-yl]-6-fluoro-1H-benzimidazol-2-one, 3-[1-[4,4-bis(2,4-difluorophenyl)cyclohex-2-en-1-yl]piperidin-4-yl]-6-fluoro-1H-benzimidazol-2-one, 3-[1-[3-[2-(4-fluorophenyl)anilino]propyl]piperidin-4-yl]-1H-benzimidazol-2-one, 3-[3-[4-[[2-(2-fluorophenyl)phenyl]methyl]piperazin-1-yl]propyl]-1H-benzimidazol-2-one, 1-[1-[4,4-bis(4-fluorophenyl)butyl]piperidin-4-yl]-3-(2,3-ditritiopropyl)benzimidazol-2-one, 3-[3-[4-[(4-fluorophenyl)methyl]piperidin-1-yl]propyl]-1H-benzimidazol-2-one, 3-[2-[4-[di(phenyl)methyl]piperidin-1-yl]ethyl]-1H-benzimidazol-2-one,-[4-[8-fluoro-5-(4-fluorophenyl)-3,4,4a,9b-tetrahydro-1H-pyrido[4,3-b]indol-2-yl]butyl]-1H-benzimidazol-2-one, 3-[1-[4,4-bis(4-fluorophenyl)butyl]-3-methylpiperidin-4-yl]-6-(trifluoromethyl)-1H-benzimidazol-2-one, and 3-[1-[4,4-bis(4-fluorophenyl)butyl]-2-methylpiperidin-4-yl]-6-(trifluoromethyl)-1H-benzimidazol-2-one.
  • Clomipramine
  • Clomipramine is a member of the tricyclic compounds described in U.S. Pat. No. 3,467,650. Analogs of clomipramine may be described by one the formulas (I), (II), (III), or (IV):
  • Figure US20130289024A1-20131031-C00177
  • wherein each X is, independently, H, Cl, F, Br, I, CH3, CF3, OH, OCH3, CH2CH3, or OCH2CH3; Y is CH2, O, NH, S(O)0-2, (CH2)3, (CH)2, CH2O, CH2NH, CHN, or CH2S; Z is C or S; A is a branched or unbranched, saturated or monounsaturated hydrocarbon chain having between 3 and 6 carbons, inclusive; each B is, independently, H, Cl, F, Br, I, CX3, CH2CH3, OCX3, or OCX2CX3; and D is CH2, O, NH, or S(O)0-2. In preferred embodiments, each X is, independently, H, Cl, or F; Y is (CH2)2, Z is C; A is (CH2)3; and each B is, independently, H, Cl, or F. Tricyclic compounds include tricyclic antidepressants such as clomipramine, amoxapine, 8-hydroxyamoxapine, 7-hydroxyamoxapine, loxapine (e.g., loxapine succinate, loxapine hydrochloride), 8-hydroxyloxapine, amitriptyline, doxepin, imipramine, trimipramine, desipramine, nortriptyline, and protriptyline. A tricyclic compound does not need to have antidepressant activities to be within the scope of clomipramine analogs.
  • Maprotiline
  • Maprotiline is a bridged-ring tetracyclic compound with psychotropic activities similar to those of the tricyclic antidepressants. The chemical structure of maprotiline and analogs of maprotiline are given by structural formulae in U.S. Pat. No. 3,399,201. Additional exemplary analogs of maprotiline are described by formula I in U.S. Pat. No. 4,017,542, e.g., 9-(2-hydroxy-3-methylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene, 9-(2-hydroxy-3-dimethylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene, 2-chloro-9-(2-hydroxy-3-methylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene, 2-chloro-9-(2-hydroxy-3-dimethylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene; by formula I of U.S. Pat. No. 4,045,560, e.g., 9-(2-morpholinylmethyl)-9,10-dihydro-9,10-methanoanthracene and 9-(4-benzyl-2-morpholinylmethyl)-9,10-dihydro-9,10-methanoanthracene; by formula I of U.S. Pat. No. 4,045,580, e.g., 9-(2-hydroxy-3-methylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene, 9-(2-hydroxy-3-dimethylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene, 2-chloro-9-(2-hydroxy-3-methylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene, and 2-chloro-9-(2-hydroxy-3-dimethylamino-propyl)-9,10-dihydro-9,10-ethano-anthracene; by formula I of U.S. Pat. No. 4,224,344, e.g., γ-methylaminopropyl-9,10-dihydro-9,10-methanoanthracene and γ-dimethylaminopropyl-9,10-dihydro-9,10-methanoanthracene; by formula I of U.S. Pat. No. 4,358,620, e.g., 9-formyl-9,10-dihydro-9,10-methanoanthracene; by formulae I, Ia, and Ib of U.S. Pat. No. 5,266,570, e.g., N-(1-[(9S,10S)-(+)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-ethoxyacetamide, (2R)—N-(1-[2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-methoxypropionamide, (2R)—N-(1-[(9S,10S)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-methoxypropionamide, N-(1-[2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-(2-pyridyl)acetamide, N-(1-[2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-ethoxypropionamide, N-(1-[2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2-(2,2,2-trifluoroethoxy)acetamide, N-(1-[2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)-2,2-dimethylpropionamide, 2-pyridylmethyl-N-(1-[2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-piperidyl)carbamate, N-(1[(9S,10S)-2-chlor-9,10-dihydro-9,10-methoanthracen-9-yl-methyl]-4-(piperidyl)-2,2-diethoxyacetamide hydrochloride, N-(1-[(9S,10S)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-(piperidyl)-2-(2,2,2-trifluoroethoxy)acetamide, N-(1-[(9S,10S)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl]-4-(piperidyl)-2-methoxy-2-methylpropionamide, and N-(1-[2-chloro-9,10-dihydro-9,10-methanoanthracen-9-yl-methyl]-4-(piperidyl)-2-(4-tetrahydropyranyloxy)acetamide; by formula I of U.S. Pat. No. 5,399,568, e.g., R-1-[1-((9S,10S)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl)-4-piperidyl]-1-(3-pyridyl)methanol, S-1-[1-((9S,10S)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl)-4-piperidyl]-1-(3-pyridyl)methanol, (R,S)-1-[1-(9RS,10RS)-(2-chloro-9,10-dihydro-9,10-methanoanthracen-9-yl-methyl)-4-piperidyl]-1-(3-pyridyl)methanol, 1-((9S,10S)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl)-4-(2-hydroxy-3-pyridylmethyl)piperidine, 1-((9RS,10RS)-2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl)-4-(2-hydroxy-3-pyridylmethyl)piperidine; by formula I of U.S. Pat. No. 5,455,246, e.g., 1-(2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl)-4-(trans-2,6-dimethyl-4-morpholinyl)piperidine, 1-(2-chloro-9,10-dihydro-9,10-methanoanthracen-9-ylmethyl)-4-(4-morpholinyl)piperidine dihydrochloride, and 2-[1-(9S,10S-2-chloro-9,10-methanoanthracen-9-yl-methyl)-4-piperidylamino]pyrimidine; by formula 1 or 1′ of U.S. Pat. No. 5,512,575, e.g., (−)-1-(9,10-dihydro-9,10-methanoanthracen-9-ylmethyl)-4-(2-ethylsulfinyl-3-pyridyl)piperidin-4-ol and (−)-1-(9,10-dihydro-9,10-methanoanthracen-9-yl-methyl)-4-(2-ethylsulfinyl-3-pyridyl)piperidin-4-ol; by formula I of U.S. Pat. No. 5,550,136; and by formulae I or I′ of U.S. Pat. No. 5,681,840.
  • Benzatropine
  • Benzatropine (also known as benzatropine mesilate and benztropine mesylate) is a muscarinic receptor antagonist. The preparation of benzotropine is described in U.S. Pat. No. 2,595,405. Exemplary analogs of benztropine are described in U.S. Pat. No. 5,506,359. Other examples are 3′-chloro-3-(diphenylmethoxy)tropane, 3′-chlorobenztropine, 4′,4″-dichloro-3-(diphenylmethoxy)tropane, 4′-chloro-3-(diphenylmethoxy)tropane, 4′-chlorobenztropine, metoclophen, N-(n-butyl)-(bis-fluorophenylorophenyl)methoxytropane, N-allyl-(bisfluorophenyl)methoxytropane, N-methyl-3-(bis(4′-fluorophenyl)methoxy)tropane, 3′-chloro-3-(diphenylmethoxy)tropane, 3′-chlorobenztropine, 4′,4″-dichloro-3-(diphenylmethoxy)tropane, 4′-chloro-3-(diphenylmethoxy)tropane, and 4′-chlorobenztropine.
  • Vinorelbine
  • Vinorelbine is a vinblastine-like anti-mitotic drug and is described in U.S. Pat. No. 4,307,100. Exemplary analogs of vinorelbine are described by formula I of U.S. Pat. No. 4,430,269; by formula I of U.S. Pat. No. 5,100,881, e.g., diethyl N-(4-O-deacetyl-5′-noranhydro-23-vinblastinoyl)-1-amino-2-methylpropylphosphonate, (+)-[diethyl N—(Na-deformyl-4-O-deacetyl-23-vincristinoyl)-1-amino-2-methylpropylphosphonate, (+)-[diethyl N-(4-O-deacetyl-23-vincristinoyl)-1-amino-2-methylpropylphosphonate], (−)-[diethyl N-(4-O-deacetyl-23-vincristinoyl)-1-amino-2-methylpropylphosphonate], and (+)-[diethyl N-(4-O-deacetyl-23-vincristinoyl)-1-aminoethylpropylphosphonate]; by formula I in U.S. Pat. No. 5,620,985, e.g., 19′,19′-difluoro 15′,20′-dihydrovinorelbine, 19′,19′-difluoro 15′,20′-dihydrovinorelbine, and 20′-deoxy 19′,19′-difluorovinblastine; by formula I in U.S. Pat. No. 7,235,564, e.g., 11′-bromovinorelbine, 11′-iodovinorelbine, 11′-vinylvinorelbine, 11′-(3-oxohex-1-enyl)vinorelbine, 11′-(2-tert-butoxycarbonylvinyl)vinorelbine, and 11′-(methoxycarbonylmethylsulfanyl)vinorelbine trifluoroacetate; and by formula I of U.S. Pat. No. 7,238,704, e.g., 12′-bromovinblastine trifluoroacetate, 12′-iodovinblastine, 12′-bromovincristine, 12′-iodovincristine, and 12′-(hexynyl)vinblastine. Yet other vinorelbine analogs are navelbine derivatives described in U.S. Pat. No. 5,220,016.
  • Azacitidine
  • Azacitidine is a pyrimidine analog that interferes with DNA metabolism and has been used as a chemotherapeutic agent.
  • Exemplary analogs of azacitidine are 2′-beta-deoxy-6-azacytidine, 2′-deoxy-6-methyl-5-azacytidine, 2′-deoxy-N4-(2-(4-nitrophenyl)ethoxycarbonyl)-5-azacytidine, 5′-azacytidine 5′-triphosphate, 5,6-dihydro-5-azacytidine, 5-aza-2′-deoxycytidine-5′-monophosphate, 5-aza-2′-deoxycytidine-5′-triphosphate, 6-azacytidine, decitabine, fazarabine, N(4),N(4)-dimethyl-5-azacytidine, N(4)-methyl-5-azacytidine, 2′-beta-deoxy-6-azacytidine, 2′-deoxy-6-methyl-5-azacytidine, 2′-deoxy-N4-(2-(4-nitrophenyl)ethoxycarbonyl)-5-azacytidine, 5′-azacytidine 5′-triphosphate, 5,6-dihydro-5-azacytidine, 5-aza-2′-deoxycytidine-5′-monophosphate, and 5-aza-2′-deoxycytidine-5′-triphosphate. Additional analogs of azacitidine are described in U.S. Pat. No. 4,788,181.
  • Dasatinib
  • Dasatinib is a tyrosine kinase inhibitor used for the treatment of certain cancers. Exemplary analogs of dasatinib are described by formula I in U.S. Pat. No. 7,091,223, e.g., N′-(2-chloro-6-methylphenyl)-2-[[2-methyl-6-[[2-(4-morpholinyl)ethyl]amin-o]-4-pyrimidinyl]amino]-5-thiazolecarboxamide, and by formula I of U.S. Pat. No. 7,091,223, e.g., N-(2-chloro-6-methylphenyl)-2-[(4,6-dimethyl-2-pyridinyl)amino]-5-thiazol-ecarboxamide.
  • Simvastatin
  • Simvastatin is an HMG-CoA reductase inhibitor and anti-hypercholesterolemia drug described in European Pat. No. EP0033538 and U.S. Pat. No. 4,444,784. An analog of simvastatin may be a structurally related compound, a compound that inhibits HMG-CoA reductase, or both. Specific examples of simvastatin analogs are lovastatin (GB2046737A), mevastatin (U.S. Pat. No. 3,983,140), pravastatin, monacolin M, monacolin X, fluvastatin (WO/1984/00213)1, atorvastatin, carvastatin (WO/1989/08094), cerivastatin, rosuvastatin, fluindostatin, velostatin, acitemate (101197-99-3), acitretin (CAS 55079-83-9), compactin, dihydrocompactin, rivastatin, dalvastatin (CAS 132100-55-1), itavastatin (U.S. Pat. No. 5,011,930), advicor (WO99/06035), BAY102987, BAY X 2678, BB476, bervastatin (CAS 132017-01-7), BMS-644950 (described in Ahmad et al., J. Med. Chem. 51:2722-2733, 2008), BMS-180431 (described in U.S. Pat. No. 4,824,959), BMY21950, BMY22089, colestolone (described in Green et al., Biochem. J. 135:63-71, 1973), CP-83101 (CAS 120360-17-0), crilvastatin (CAS 120551-59-9), DMP565 (CAS 199480-80-3), glenvastatin (described in EP-00307342), FR901512 (described in Hatori et al., J. Antibiot. 57: 390-393, 2004), L659699 (described in U.S. Pat. No. 4,988,697), L669262 (described in EP-00331250 and EP-00408806), NCX6560 (WO/2004/105754), NR-300s, P882222, P882284, PD134965, PD135022 (CAS 122548-95-2), rawsonol (CAS 125111-69-5), RBx-10558 (WO/2004/05250), RP61969, 52467, 52468, SC37111, SC45355, SQ33600 (Sliskovic et al., Drug News and Perspectives, 5:517-533), SR12813 (U.S. Pat. No. 5,043,330; WO/2002/95652), SR45023A, tocotrienols (described in Parker et al., J. Biol. Chem. 268:11230-11238, 1993) U20685, U88156, and U-9888 (CAS 190783-55-2), as well as pharmaceutically acceptable salts thereof (e.g., simvastatin sodium, lovastatin sodium, fluvastatin sodium, etc.). In addition, HMG CoA-reductase inhibitors are described in Procopiou, et al. (J. Med. Chem. 36: 3658-3665, 1993) and Chan et al. (J. Med. Chem. 36: 3646-3657, 1993).
  • Yet other simvastatin analogs are described in U.S. Pat. Nos. 3,983,140; 4,231,938; 4,282,155; 4,293,496; 4,294,926; 4,319,039; 4,343,814; 4,346,227; 4,351,844; 4,361,515; 4,376,863; 4,444,784; 4,448,784; 4,448,979; 4,450,171; 4,503,072; 4,517,373; 4,661,483; 4,668,699; 4,681,893; 4,719,229; 4,738,982; 4,739,073; 4,766,145; 4,782,084; 4,804,770; 4,824,959; 4,841,074; 4,847,306; 4,857,546; 4,857,547; 4,940,727; 4,946,864; 5,001,148; 5,006,530; 5,075,311; 5,112,857; 5,116,870; 5,120,848; 5,166,364; 5,173,487; 5,177,080; 5,273,995; 5,276,021; 5,369,123; 5,385,932; 5,502,199; 5,763,414; 5,877,208; and 6,541,511; U.S. Pat. Application Publication Nos. 2002/0013334 A1; 2002/0028826 A1; 2002/0061901 A1; and 2002/0094977 A1; and PCT publications WO/2001/96311 and WO/1996/08248.
  • Teicoplanin
  • Teicoplanin is a complex antiobiotic agent containing several compounds, five major (named teicoplanin A2-1 through A2-5) and four minor (named teicoplanin RS-1 through RS-4). Thus, as used herein, the term teicoplanin encompasses each of these major and minor compounds. All teicoplanins share a same glycopeptide core, termed teicoplanin A3-1, featuring a fused ring structure to which two carbohydrates (mannose and N-acetylglucosamine) are attached. The major and minor components also contain a third carbohydrate moiety and differ by the length and conformation of a side chain attached to it. Teicoplanin analogs include compounds structurally related to the major and minor teicoplanin constituents and products of teicoplanin hydrolysis, e.g., L 17054 and L 17392. Other analogs of teicoplanin include L 17046, L 17932 (U.S. Pat. No. 5,594,102), 4,7-decadienoyl-teicoplanin, 4-hydroxydecanoyl-teicoplanin, MDL 62873, aglycone forms of any teicoplanin compound or teicoplanin derivative, compounds of formula I of U.S. Pat. No. 4,661,470, demannosyl teichoplanin derivatives, e.g., compounds of formula I of U.S. Pat. No. 5,064,811; compounds described in U.S. Pat. No. 5,085,990; de-(acetylglucosaminyl)-di(dehydro)-deoxy teicoplanin derivatives, e.g., compounds of formula I of U.S. Pat. No. 4,789,661; and substituted alkymides of teicoplanin according to formula I of U.S. Pat. No. 5,198,418.
  • Terconazole
  • Terconazole is described in U.S. Pat. No. 4,144,346 and has the following structure:
  • Figure US20130289024A1-20131031-C00178
  • Structural analogs of terconazole include any stereochemical isomers thereof. Other structural analogs are described in U.S. Pat. Nos. 3,575,999, 3,936,470, 4,223,036, 4,358,449 (see, for example, Examples I-LXXII), in Belgian Pat. No. 935,579, and in the PCT Publication No. WO00/76316.
  • Structural analogs of terconazole can also be described by the following formula:
  • Figure US20130289024A1-20131031-C00179
  • Wherein Q is —CH— or —N—; Ar is optionally substituted phenyl, wherein a substituted phenyl has 1, 2, or 3 substituents that are, independently, halogen, C1-6 alkyl, or C1-6 alkoxy; A is —NCS, —NR2R3, —NHC(X)—(Y)m—R4, or
  • Figure US20130289024A1-20131031-C00180
  • wherein each R2 and R3 is, independently, H or C1-6 alkyl; X is O or W; Y is O or NH; m is 0 or 1; R4 is H, optionally substituted C1-6 alkyl, or optionally substituted phenyl, wherein a substituted C1-6 alkyl, or substituted phenyl has 1 or 2 substituents that are each, independently, halogen, C1-6 alkyl, or C1-6 alkoxy; R5 is a bond, —CH2—, —O—, —S—, or —NR6—, where R6 is H or optionally substituted C1-6 alkyl; and R is H or NO2.
  • Exemplary, non-limiting structural analogs of terconazle are 4-[2-(3-chlorophenyl)-2-(1H-imidazol-1-ylmethyl)-1,3-dioxolan-4-ylmethoxy]-N-ethylbenzenamine, 4-[2-(4-bromophenyl)-2-(1H-imidazol-1-ylmethyl)-1,3-dioxolan-4-ylmethoxy]-N-ethylbenzenamine, N-ethyl-4-[2-(1H-imidazol-1-ylmethyl)-2-(3-methylphenyl)-1,3-dioxolan-4-ylmethoxy]benzenamine, N-ethyl-4-[2-(1H-imidazol-1-ylmethyl)-2-(4-methoxyphenyl)-1,3-dioxolan-4-yl methoxy]benzenamine, 4-[2-(2,4-dichlorophenyl)-2-(1H-1,2,4-triazol-1-ylmethyl)-1,3-dioxolan-4-yl methoxy]-N-ethylbenzenamine, N-{4-[2-(3-chlorophenyl)-2-(1H-imidazol-1-ylmethyl)-1,3-dioxolan-4-ylmethoxy]phenyl}acetamide. N-{4-[2-(4-bromophenyl)-2-(1H-imidazol-1-ylmethyl)-1,3-dioxolan-4-ylmethoxy]phenyl}benzamide ethyl {4-[2-(1H-imidazol-1-ylmethyl)-2-(3-methylphenyl)-1,3-dioxolan-4-ylmethoxy]phenyl}carbamate, N-{4-[2-(1H-imidazol-1-ylmethyl)-2-(4-methoxyphenyl)-1,3-dioxolan-4-ylmethoxy]phenyl}-4-fluorobenzamide, N-{4-[2-(2,4-dichlorophenyl)-2-(1H-1,2,4-triazol-1-ylmethyl)-1,3 dioxolan-4-ylmethoxy]phenyl}acetamide, 1-{2-(3-chlorophenyl)-4-[4-(1-pyrrolidinyl)phenoxymethyl]-1,3-dioxolan-2-yl methyl}-1H-imidazole, 1-{2-(4-bromophenyl)-4-[4-(1-piperidinyl)phenoxymethyl]-1,3-dioxolan-2-ylmethyl}-1H-imidazole, 1-{2-(3-methylphenyl)-4-[4-(1-pyrrolidinyl)phenoxymethyl]-1,3-dioxolan-2-yl methyl}-1H-imidazole, 1-{2-(4-methoxyphenyl)-4-[4-(1-piperidinyl)phenoxymethyl]-1,3-dioxolan-2-yl methyl}-1H-imidazole, 1-{2-(2,4-dichlorophenyl)-4-[4-(1-pyrrolidinyl)phenoxymethyl]-1,3-dioxolan-2-ylmethyl}-1H-1,2,4-triazole, 1-{2-(2,4-dichlorophenyl)-4-[4-(1-piperidinyl)phenoxymethyl]-1,3-dioxolan-2-ylmethyl}-1H-1,2,4-triazole, 4-{4-[2-(3-chlorophenyl)-2-(1H-imidazol-1-ylmethyl)-1,3-dioxolan-4-ylmethoxy]phenyl}morpholine, 4-{4-[2-(1H-imidazol-1-ylmethyl)-2-(3-methylphenyl)-1,3-dioxolan-4-ylmethoxy]phenyl}morpholine, 4-{4-[2-(1H-imidazol-1-ylmethyl)-2-(4-methoxyphenyl)-1,3-dioxolan-4-ylmethoxy]phenyl}morpholine, and 4-{4-[2-(2,4-dichlorophenyl)-2-(1H-1,2,4-triazol-1-ylmethyl)-1,3-dioxolan-4-ylmethoxy]phenyl}morpholine.
  • Hycanthone
  • Hycanthone is a thioxanthenone schistosomicide described in U.S. Pat. Nos. 3,294,803 and 3,312,598. Analogs of hycanthone include other compounds of formula I in U.S. Pat. No. 3,312,598, lucanthone, methixene, prothixene, quantacure QTX, teflutixol, thiothixene, WIN 33377, chlorprothixene, clopenthixol, doxantrazole, flupenthixol, hycanthone methanesulfonate, hycanthone N-methylcarbamate, hycanthone sulfamate, IA 4, IA 4 N-oxide, 2-(beta-diethylaminoethylamino)-3,4-cyclohexenothia-xanthone, 2-chlorothioxanthen-9-one, 3-carboxy-thioxanthone-10,10-dioxide, 4-(beta-diethylaminoethylamino)-1,2-cyclohexenothiaxanthone, 7-oxo-7-thiomethoxyxanthone-2-carboxylic acid, BW 616U76, and 4-(bis(2′-chloroethyl)amino)propylamino-1,2-cyclohexenothioxanthone.
  • Atovaquone
  • Atovaquone is an anti-microbial napthalene compound described in U.S. Pat. No. 5,053,432. Analogs of atovaquone include compounds described by formula I of U.S. Pat. No. 5,053,432, e.g., 2-hydroxy-3-(4-methoxycyclohexyl)-1,4-naphthoquinone, 2-hydroxy-3-[4-(1-methoxy-1-methylethyl)cyclohexyl]-1,4-naphthoquinone, 2-(4-benzyloxycyclohexyl)-3-hydroxy-1,4-naphthoquinone, 2,4-(3,4-dichlorophenyl)cyclohexyl-3-hydroxy-1,4-naphthoquinone, 2-[4-(3,4-dimethylphenyl)cyclohexyl]-1,4-naphthoquinone, 2-(4-fluorocyclohexyl)-3-hydroxy-1,4-naphthoquinone, 2-hydroxy-3-(4-trifluoromethylcyclohexyl)-1,4-naphthoquinone, 2-(4-n-butoxycyclohexyl)-3-hydroxy-1,4-naphthoquinone, 2-(4-t-butoxycyclohexyl)-3-hydroxy-1,4-naphthoquinone, 2-(4,4-dimethylcyclohexyl)-3-hydroxy-1,4-naphthoquinone, 2-(4,4-diethylcyclohexyl)-3-hydroxy-1,4-naphthoquinone, 2-(4,4-diphenylcyclohexyl)-3-hydroxy-1,4-naphthoquinone, 2-(4,4-di-n-propylcyclohexyl)-3-hydroxy-1,4-naphthoquinone, and 2-[4-(4-chlorophenoxymethyl)cyclohexyl]-3-hydroxy-1,4-naphthoquinone.
  • Other exemplary analogs include compounds described by formula II in U.S. Pat. No. 5,310,762, e.g., 2-(4-t-butylcyclohexyl)-3-hydroxy-1,4-naphthoquinone; by formula II in U.S. Pat. No. 5,466,711, e.g., 2-acetoxy-3-[trans-4-(4-chlorophenyl)cyclohexyl]-1,4-naphthoquinone; by formula II in U.S. Pat. No. 5,567,738, e.g., 2-(4-(4-chlorophenyl)cyclohexyl)-3-hydroxy-1,4-Naphthoquinone; and by formula I, II, or III in U.S. Pat. No. 5,684,035, e.g., 5,8-dihydroxy-1,4-naphthoquinone.
  • Quinacrine
  • Quinacrine is an acridine derivative described in U.S. Pat. No. 2,113,357. Analogs of quinacrine include, for example, chloroquine, hydroxychloroquine, amodiaquine, mefloquine, primaquine, quinine, chemiochin (4-N-(6-chloro-2-methoxyacridin-9-yl)-1-N,1-N-diethylpentane-1,4-diamine dihydrochloride), atabrine hydrochloride (4-[(6-chloro-2-methoxyacridin-9-yl)azaniumyl]pentyl-diethylazanium dichloride), NSC240788 (2-N-(6-chloro-2-methoxyacridin-9-yl)-1-N,1-N-diethylpropane-1,2-diamine), NSC 8591 (4-[(6-chloro-2-methoxyacridin-9-yl)azaniumyl]butyl-diethylazanium dichloride), NSC56618 (N′-(6-chloro-2-methoxyacridin-9-yl)-N,N-diethylheptane-1,7-diamine), NSC56619 (N′-(6-chloro-2-methoxyacridin-9-yl)-N,N-diethyloctane-1,8-diamine), AIDS185224 (N-(3-aminopropyl)-N′-(6-chloro-2-methoxyacridin-9-yl)-N-methylpropane-1,3-diamine), NSC353 (N′-(6-chloro-2-methoxyacridin-9-yl)-N,N-diethylpropane-1,3-diamine hydrochloride), NSC 8591 (N′-(6-chloro-2-methoxyacridin-9-yl)-N,N-diethylbutane-1,4-diamine hydrochloride), (WIN 501 (N-(6-chloro-2-methoxyacridin-9-yl)-N′,N′-dimethylbutane-1,4-diamine), NSC353 (N′-(6-chloro-2-methoxyacridin-9-yl)-N′,N′-diethylpropane-1,3-diamine), AIDS185219 (N-(6-chloro-2-methoxyacridin-9-yl)-N′,N′-dimethylpropane-1,3-diamine), LS-14291 (9[(E)-4-[(6-chloro-2-methoxyacridin-9-yl)azaniumyl]pent-2-enyl]-diethylazanium dichloride), (LS-14292 ([(Z)-4-[(6-chloro-2-methoxyacridin-9-yl)azaniumyl]pent-2-enyl]-diethylazanium dichloride), (S)-quinacrine, N′-(6-chloro-2-methoxyacridin-9-yl)-N,N-di(propan-2-yl)propane-1,3-diamine, (E)-4-N-(6-chloro-2-methoxyacridin-9-yl)-1-N,1-N-diethylpent-2-ene-1,4-diamine, 4-N-(6-chloro-2-methoxyacridin-9-yl)-1-N,1-N-diethylpentane-1,4-diamine hydrochloride, 5-N-(6-chloro-2-methoxyacridin-9-yl)-1-N,1-N-diethylhexane-1,5-diamine, 4-N-(6-chloro-2-methoxyacridin-9-yl)-1-N,1-N-diethylpentane-1,4-diamine chloride, 8-N-(6-chloro-2-methoxyacridin-9-yl)-1-N,1-N-diethylnonane-1,8-diamine, N′-(6-chloro-2-methoxyacridin-9-yl)-N,N-dipropylpropane-1,3-diamine, N′-(6-chloro-2-methoxyacridin-9-yl)-N,N-diethylpentane-1,5-diamine, and 4-N-(6-chloro-2-methoxyacridin-9-yl)pentane-1,4-diamine. Additional quinacrine analogs are described in Macfarlane and Manzel (J. Immunol. 160:1122-1131, 1998), Breslow et al. (J. Am. chem. Soc. 67:1472-1475, 1945), and Bloom et al. (J. Am. chem. Soc. 67:2206-2208, 1945).
  • Efavirenz
  • Efavirenz is a non-nucleoside reverse transcriptase inhibitor described in U.S. Pat. No. 5,519,021. Analogs of efavirenz include compounds described by formula I or II of U.S. Pat. No. 5,519,021, e.g., (−)6-chloro-4-cyclopropyl-ethynyl-4-frifluoromethyl-1-4-dihydro-2H-3,1-benzoxazin-2-one, (−) 6-chloro-4-phenylethynyl-4-trifluoromethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−) 4-(1-chloro-1,1-difluoromethyl)-4-(2-phenylethynyl)-6-chloro-1,4-dihydro-2H-3,1-benzoxazin-2-one, or (+/−) 4-(2-[dimethylaminomethyl]ethynyl)-4-trifluoromethyl-6-chloro-1,4-dihydro-2H-3,1-benzoxazin-2-one, or a pharmaceutically acceptable salt thereof; by formula I of U.S. Pat. No. 5,874,430, e.g., (+/−)-6-chloro-4-(cyclopropylethynyl)-8-hydroxy-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (−)-6-chloro-4-(cyclopropylethynyl)-8-hydroxy-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(cyclopropylethynyl)-8-fluoro-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-cyclopropylethynyl-4-isopropyl-6-methyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-isopropylethynyl-4-trifluoromethyl-6-methyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-acetyl-4-cyclopropylethynyl-4-trifluoromethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-(3-methyl)-1-buten-1-yl-4-trifluoromethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-isopropylethynyl-4-trifluoromethyl-5,6-difluoro-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-cyclopropylethynyl-6-chloro-4-trifluoromethyl-7-aza-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(2-methoxyethoxy)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-propylamino-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-(1-butynyl)-6-methoxy-4-trifluoromethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-(1′-hydroxy)-cyclopropylethynyl-4-trifluoromethyl-6-chloro-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-isopropylethynyl-4-trifluoromethyl-5-fluoro-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(1-deuterocycloprop-1-ylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-isopropylethynyl-4-trifluoromethyl-5-fluoro-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(cyclopropylethynyl)-8-methoxy-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(cyclopropylethynyl)-7-hydroxy-4-(trifluoromethyl)-1,4-di hydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(1-butynyl)-8-fluoro-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(isopentyl)-8-fluoro-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-(1-pentynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-(1-butynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-(1-propynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-pentyl-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-isopentyl-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-butyl-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-methylenedioxy-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-methylenedioxy-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-methylenedioxy-4-(1-pentynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-methylenedioxy-4-(1-butynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-methylenedioxy-4-(2-pentynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-methylenedioxy-4-(2-butynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-methylenedioxy-4-(isopentyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(1-pentynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(1-propynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(2-pentynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(isopentyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-butyl-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(phenylethyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(cyclopropylethynyl)-8-fluoro-4-(trifluoromethyl)-1,4-di hydro-2H-3,1-benzoxazin-2-one, (+/−)-6-dimethylamino-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-dimethylamino-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-dimethylamino-4-pentyl-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-dimethylamino-4-isopentyl-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-acetyl-4-(1-butynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methyl-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methyl-4-(1-butynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6,8-dichloro-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6,8-dichloro-4-(phenylethyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6,8-trifluoro-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6,8-trifluoro-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6,8-trifluoro-4-(1-pentynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6,8-trifluoro-4-(1-butynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,8-difluoro-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,8-difluoro-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,8-difluoro-4-(1-pentynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,8-difluoro-4-(1-butynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-isopropyl-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-isopropyl-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-isopropyl-4-(phenylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-isopropyl-4-pentyl-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-isopropyl-4-(isopentyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-isopropyl-4-(phenylethyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-trifluoromethoxy-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-trifluoromethoxy-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-trifluoromethoxy-4-(phenylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-trifluoromethoxy-4-pentyl-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-trifluoromethoxy-4-(isopentyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-trifluoromethoxy-4-(phenylethyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-(phenylethyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-(phenylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-(pentyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-(isopentyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-phenyl-4-(cyclopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-phenyl-4-(isopropylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-phenyl-4-(1-pentynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-phenyl-4-(4-methylpentynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-phenyl-4-(1-butynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-phenyl-4-(isopentyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(cyclopropylethynyl)-4-isopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(isopropylethynyl)-4-isopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methyl-4-(isopropylethynyl)-4-cyclopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methyl-4-(isopropylethynyl)-4-isopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methyl-4-(isopropylethynyl)-4-ethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methyl-4-(1-butynyl)-4-ethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6,7-dichloro-4-(isopropylethynyl)-4-cyclopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6,7-dichloro-4-(isopropylethynyl)-4-isopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-7-chloro-4-(cyclopropylethynyl)-4-cyclopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-7-chloro-4-(isopropylethynyl)-4-cyclopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-7-chloro-4-(4-methylpentynyl)-4-cyclopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-7-chloro-4-(cyclopropylethynyl)-4-isopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-7-chloro-4-(isopropylethynyl)-4-isopropyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(cyclopropylethynyl)-4-(trifluoromethyl)-8-aza-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(isopropylethynyl)-4-(trifluoromethyl)-8-aza-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-44-phenylethyl)-4-(trifluoromethyl)-8-aza-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-methoxy-4-(cyclopropylethynyl)-4-(trifluoromethyl)-7-aza-1,4-dihydro-2H-3,1-benzoxazin-2-one, and (+/−)-4-(cyclopropylethynyl)-4-(trifluoromethyl)-6-aza-1,4-dihydro-2H-3,1-benzoxazin-2-one; by formula II, Ia, or Ib of U.S. Pat. No. 6,090,821; by formula I of U.S. Pat. No. 6,124,302, e.g., (+/−)-6-chloro-4-cyclopropylethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-6-chloro-4-(2-pyridyl)ethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-6-chloro-4-phenylethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-4-cyclopropylethynyl-6-methoxy-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-6-methoxy-4-(2-pyridyl)ethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-6-methoxy-4-phenylethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-4-cyclopropylethynyl-5,6-difluoro-4-trifluoromethyl-3,4-dihydro-2(1H-quinazolinone, (+/−)-5,6-difluoro-4-(2-pyridyl)ethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-5,6-difluoro-4-phenylethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-4-cyclopropylethynyl-6-fluoro-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-6-fluoro-4-(2-pyridyl)ethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-6-fluoro-4-phenylethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-6-fluoro-4-(2′-2-pyridyl)ethyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+/−)-6-fluoro-4-phenylethyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (−)-6-chloro-4-cyclopropylethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+)-6-chloro-4-cyclopropylethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+)-4-cyclopropylethynyl-5,6-difluoro-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (−)-4-cyclopropylethynyl-5,6-difluoro-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, (+)-E-4-cyclopropylethynyl-5,6-difluoro-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone, and (−)-6-chloro-4-E-cyclopropylethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinone; by formula I of U.S. Pat. No. 6,127,375, e.g., (+/−)-4-cyclopropylethynyl-5,6-difluoro-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinthione, (+/−)-6-chloro-4-cyclopropylethynyl-4-trifluoromethyl-3,4-dihydro-2(1H)-quinazolinthione, (+/−)-4-cyclopropylethynyl-5,6-difluoro-4-trifluoromethyl-3,4-dihydro-2-methyl(1H)-quinazolinthione, and (+/−)-6-chloro-4-cyclopropylethynyl-4-trifluoromethyl-3,4-dihydro-2-methyl(1H)-quinazolinthione; by formula I of U.S. Pat. No. 6,140,320, e.g., 5-(1-butynyl)-7-chloro-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-5-(1-butynyl)-7-chloro-1,5-dihydro-3-phenyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, 7-chloro-1,5-dihydro-5-(isopropylethynyl)-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, (+)-(5S)-7-chloro-1,5-dihydro-5-(isopropylethynyl)-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, trans-7-chloro-5-(2-cyclopropylethenyl)-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-7-chloro-5-(2cyclopropylethenyl)-1,5-dihydro-3-methyl-5-(trifluozomethyl)-4,1-benzoxazepin-2(3H)-one, 1,5-dihydro-7-fluoro-5-isopropylethynyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, 1,5-dihydro-7-fluoro-5-(3-methylbutyl)-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-7-chloro-1,5-dihydro-5-(2-furan-2-ylethenyl)-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, trans-7-chloro-1,5-dihydro-5-(2-furan-2-yl)ethenyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-chloro-1,5-dihydro-5-(2-furanyl)ethynyl-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, 5-butyl-7-chloro-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one 4-isopropylethynyl-4-trifluoromethyl-5,6-difluoro-1,4-dihydro-2H-3,1-benzoxazepin-2-one, rel-(3S,5S)-7-chloro-5-cyclopropylethynyl-1,5-dihydro-3-propyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3R,5S)-7-chloro-5-cyclopropylethynyl-1,5-dihydro-3-propyl-5-(trifluoro methyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-chloro-5-cycloproplethynyl-1,5-dihydro-3-isopropyl-5-(trifluoromethyl-)-4,1-benzoxazepin-2(3H)-one, 7-chloro-5-phenylethynyl-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2 (3H)-one, rel-(3S,5S)-7-chloro-5-isopropylethynyl-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, 7-chloro-5-cyclopropylethynyl-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, 7-chloro-5-isopropylethynyl-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, trans-7-chloro-5-(2-isopropylethenyl)-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, 7-methoxy-5-(3-methylbutyl)-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-chloro-5-cyclopropylethynyl-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3R,5S)-7-chloro-5-cyclopropylethynyl-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, 7-chloro-5-(3-pyridylethynyl)-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxaze pin-2(3H)-one, trans-7-chloro-5-(3-pyrid-3-ylethenyl)-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, trans-7-fluoro-5-(2-isopropylethenyl)-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, trans-6,7-difluoro-5-(2-isopropylethenyl)-1,5-dihydro-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-chloro-5-cyclopropylethynyl-1,5-dihydro-3-methyl-5-(trifluoro methyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-7-chloro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-7-chloro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-propyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-chloro-5-(3-furanylethynyl)-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-chloro-5-(3-furanylethynyl)-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-6,7-difluoro-5-cyclopropylethynyl-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-6,7-difluoro-5-cyclopropylethynyl-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-6,7-difluoro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, (+)-(3S,5S)-7-chloro-5-cyclopropylethynyl-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, (3S)-7-chloro-5-cyclopropylethynyl)-1,5-dihydro-5-(trifluoromethyl)-4,1-benzocazepin-2(3H)-one, rel-(3S,5S)-trans-7-chloro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, (+)-(3S,5S)-trans-7-chloro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, (+)-(3S,5S)-trans-7-chloro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-chloro-5-(2-cyclopropylethynyl)-1,5-dihydro-3-ethenyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-7-chloro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-ethenyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-chloro-5-(2-cyclopropylethynyl)-1,5-dihydro-3-cyclopropyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-7-chloro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-cyclopropyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-6,7-difluoro-5-(2-cyclopropylethynyl)-1,5-dihydro-3-ethenyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-6,7-difluoro-5-(2-cyclopropylethynyl)-1,5-dihydro-3-cyclopropyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-fluoro-5-(2-cyclopropylethynyl)-1,5-dihydro-3-ethenyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-7-fluoro-5-(2-cyclopropylethynyl)-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2 (3H)-one, rel-(3S,5S)-7-fluoro-5-(2-cyclopropylethynyl)-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-7-fluoro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-7-fluoro-5-(2-cyclopropylethenyl)-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-6,7-methylenedioxy-5-(2-cyclopropylethynyl)-1,5-dihydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-6,7-methylenedioxy-5-2-cyclopropylethynyl)-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, rel-(3S,5S)-trans-6,7-methylenedioxy-5-(2-cyclopropylethenyl)-1,5-dilydro-3-methyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one, and rel-(3S,5S)-trans-6,7-methylenedioxy-5-(2-cyclopropylethenyl)-1,5-dihydro-3-ethyl-5-(trifluoromethyl)-4,1-benzoxazepin-2(3H)-one; by formula I or II of U.S. Pat. No. 6,303,780, e.g., (+/−)-6-chloro4-(cyclopropylethynyl)-8-hydroxy-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one; (−)-6-chloro-4-(cyclopropylethynyl)-8-hydroxy-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(cyclopropylethynyl)-8-fluoro-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-cyclopropylethynyl-4-isopropyl-6-methyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-isopropylethynyl-4-trifluoromethyl-6-methyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-acetyl-4-cyclopropylethynyl-4-trifluoromethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-5,6-difluoro-4-(3-methyl)-1-buten-1-yl-4-trifluoromethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-isopropylethynyl-4-trifluoromethyl-5,6-difluoro-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-cyclopropylethynyl-6-chloro-4-trifluoromethyl-7-aza-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(2-methoxyethoxy)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-propylamino-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(2-(furan-2-yl)ethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-(1-butynyl)-6-methoxy-4-trifluoromethyl-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-(1′-hydroxy)-cyclopropylethynyl-4-trifluoromethyl-6-chloro-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-4-isopropylethynyl-4-trifluoromethyl-5-fluoro-1,4-dihydro-2H-3,1-benzoxazin-2-one, (+/−)-6-chloro-4-(1-deuterocycloprop-1-ylethynyl)-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, and (+/−)-4-isopropylethynyl-4-trifluoromethyl-5-fluoro-1,4-dihydro-2H-3,1-benzoxazin-2-one; by formula I of U.S. Pat. No. 6,492,515, e.g., (+/−)-6-chloro-4-(cyclopropylethynyl)-8-hydroxy-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, (−)-6-chloro-4-(cyclopropylethynyl)-8-hydroxy-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one, and (+/−)-6-chloro-4-(cyclopropylethynyl)-8-fluoro-4-(trifluoromethyl)-1,4-dihydro-2H-3,1-benzoxazin-2-one; by formula I of U.S. Pat. No. 6,593,337, e.g., 7-chloro-5-(cyclopropylmethoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(benzyloxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(cyclobutylmethoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(ethoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(hydroxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(n-propoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(i-propoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(butyl)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(methoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5(S)-(cyclopropylmethoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5(R)-(cyclopropylmethoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(2-cyclopropylethyl)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1, 8]naphthyridine, 7-chloro-5-(2,2,2-trifluoroethoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(propargoxy)-5,10-dihydro-5 (trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(ethyl)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(cyclopropylmethoxy)-2-methyl-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(n-butyl)-2-methyl-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(2-cyclopropylethyl)-2-methyl-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(cyclopropylamino)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(i-propylamino)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(N,N-dimethylaminoethoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(N-morpholinylethylamino)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-((1-methylcyclopropyl)methoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(3,3,3-trifluoroprop-1-oxy)-5,10-dihydro-5-(trifluoromethyl)benz o[b][1,8]naphthyridine, 7-chloro-5-(cyclopropylmethylamino)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(methylamino)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(ethylamino)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, (S)-7-chloro-5-(cyclopropylethyl)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, (R)-7-chloro-5-(cyclopropylethyl)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-fluoro-5-(cyclopropylmethoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-fluoro-5-(cyclopropylethoxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-fluoro-5-(allyloxy)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(phenylamino)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(cyclopropylmethoxy)-2-methyl-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(n-butyl)-2-methyl-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]naphthyridine, 7-chloro-5-(cyclopropylethyl)-2-methyl-5,10-dihydro-5-(trifluoromethyl)benz o[b][1,8]naphthyridine, 7-chloro-5-cyclopropylmethoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 5-Allyloxy-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine-5-carbonitrile, 7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-ol, 5-cyclopropylmethoxy-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-5-prop-2-ynyloxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-5-(1-methyl-cyclopropylmethoxy)-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-5-(2-cyclopropy-ethoxy)-5-trifluoromethyl-5,10-dihydro-benzo[b][1, 8]naphthyridine 1-oxide, (7-chloro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-yl)-isopropyl-amine, (7-chloro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-yl)-cyclobutylmethyl-amine, 7-chloro-5-(2-cyclopropyl-ethyl)-5-trifluoromethyl-5,10-dihydro-benzo[b][1, 8]naphthyridine 1-oxide, 5-cyclobutylmethoxy-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (7-fluoro-1-oxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-yl)-isopropyl-amine, 5-cyclobutylmethoxy-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-2-ol, 7-chloro-5-(pyridin-2-ylmethoxy)-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 5-butyl-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 7-chloro-1-oxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-ol, 7-chloro-5-cyclopropylmethoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-5-pyridin-2-ylmethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-fluoro-5-pyridin-2-ylmethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 5-cyclopropylmethoxy-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-5-pyridin-2-ylmethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 3,7-dichloro-5-cyclopropylmethoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 3,7-dichloro-5-cyclopropylmethoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 3,7-dichloro-5-pentyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 5-(2-cyclopropyl-ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1, 8]naphthyridine, 5-(2-cyclopropyl-ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1, 8]naphthyridine 1-oxide, 3,7-dichloro-5-cyclopropylmethoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 5-(2-cyclopropyl-ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1, 8]naphthyridine 1-oxide, 3-chloro-5-cyclopropylmethoxy-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo [b][1,8]naphthyridine, 3-chloro-5-cyclopropylmethoxy-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-5-isobutoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 5-butyl-7-chloro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (S) 3-chloro-5-cyclopropylmethoxy-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (7-chloro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-yl)-methanol, 7-fluoro-5-isobutoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-fluoro-5-isopropoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, methanesulfonic acid 7-chloro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-ylmethyl ester, 7-chloro-5-isopropoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 3-bromo-5-cyclopropylmethoxy-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 5-butyl-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 5-diisopropoxymethyl-7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 7-fluoro-5-isopropoxymethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-5-isobutyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-5-propoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (S) 7-fluoro-5-isobutoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (R) 7-fluoro-5-isobutoxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (7-chloro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-yl)-acetaldehyde, 7-chloro-5-(2,2-diisopropoxy-ethyl)-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 7-chloro-5-(2-isopropoxy-ethyl)-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 2-(7-chloro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-yl)-ethanol, 7-chloro-5-(2-isopropoxy-ethyl)-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (R) 7-fluoro-5-(2-isopropoxy-ethyl)-5-trifluoromethyl-5,10-dihydro-benzo[b][1, 8]naphthyridine 1-oxide, (7-fluoro-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-yl)-acetic acid tert-butyl ester, (7-fluoro-1-oxy-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridin-5-yl)-acetic acid tert-butyl ester, 7-chloro-5-cyclopropylmethoxy-5-(1,1-difluoro-ethyl)-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 5-cyclopropylmethoxy-5-(1,1-difluoro-ethyl)-7-fluoro-5,10-dihydro-benzo[b][1,8]naphthyridine, 5-cyclopropylmethoxy-5-(1,1-difluoro-ethyl)-7-fluoro-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-5-(1,1-difluoro-ethyl)-5-isobutoxy-5,10-dihydro-benzo[b][1,8]naphthyridine, 7-chloro-5-(1,1-difluoro-ethyl)-5-isobutoxy-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (R) 7-chloro-5-cyclopropylmethoxy-5-(1,1-difluoro-ethyl)-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (S) 7-chloro-5-cyclopropylmethoxy-5-(1,1-difluoro-ethyl)-5,10-dihydro-benzo[b][1, 8]naphthyridine 1-oxide, 7-chloro-5-difluoromethyl-5-isopropoxymethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 7-chloro-5-difluoromethyl-5-isopropoxymethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 7-chloro-1,5-dihydro-5-(N-ethylaminomethyl)-5-(trifluoromethyl)benzo[b][1,8]napthyridine, 7-chloro-5,10-dihydro-5-(N-isopropylaminomethyl)-5-(trifluoromethyl)benzo[b][1,8]napthyridine, 7-chloro-5,10-dihydro-5-(N-isopropyl-N-ethylaminomethyl)-5-(trifluoromethyl)benzo[b][1,8]napthyridine, 7-chloro-5-(N,N-diethylaminomethyl)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]napthyridine, 5-(acetamidomethyl)-7-chloro-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]napthyridine, 5,10-dihydro-7-fluoro-5-(N-methylsulfonylmethyl)-5-(trifluoromethyl) benzo[b][1,8]napthyridine, 5,10-dihydro-7-fluoro-5-(isopropylamidomethyl)-5-(trifluoromethyl)benzo[b][1,8]napthyridine, 5,10-dihydro-7-fluoro-5-(isopropylguanadinomethyl)-5-(trifluormethyl)benzo[b][1,8]napthyridine, 5,10-dihydro-7-fluoro-5-(N-isopropylmethyl)-5-(trifluoromethyl)benzo[b][1,8]napthyridine-1-(N-oxide), 5-(N,N-diethylaminomethyl)-5,10-dihydro-7-fluoro-5-(trifluoromethyl)benzo[b][1,8]napthyridine-1-(N-oxide), 5,10-dihydro-5-(N,N-dimethylaminomethyl)-7-fluoro-5-(trifluoromethyl)benzo[b][1,8]napthyridine-1-(N-oxide), 7-chloro-5,10-dihydro-5-(N-isopropylaminomethyl)-5-(trifluoromethyl)benzo[b][1,8]napthyridine-1-(N-oxide), 7-chloro-5-(N,N-diethylaminomethyl)-5,10-dihydro-5-(trifluoromethyl)benzo[b][1,8]napthyridine-1-(N-oxide), and 7-chloro-5,10-dihydro-5-(N,N-dimethylaminomethyl)-5-(trifluoromethyl) benzo[b][1,8]napthyridine-1-(N-oxide); by formula I of U.S. Pat. No. 6,596,729, e.g., 7-fluoro-2-methyl-5-[(6-methyl-2-pyridinyl)methyl]-5-(trifluoromethyl)-5,10-dihydrobenzo [b]-1,7-naphthyridin-1(2H)-one, 5-(2-cyclopropylethynyl)-7-fluoro-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-propyl-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-butyl-7-fluoro-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(4-fluorophenylmethyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(2-pyridylmethyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(isopropyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(3-pyridylmethyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(4-pyridylmethyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(3-propynyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(2-pyridylethynyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(2-(2-pyridyl)ethyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 3-chloro-7-fluoro-5-propyl-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(3-propenyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-cyclopropylethyl)-7-fluoro-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(ethynyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(2-ethoxyethyl)-5-(trifluoromethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-butyl-7-chloro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(2-pyridylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(2-cyclopropylethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1 (2H)-one, 7-chloro-5-cyclopropylethynyl-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(N-cyclopropylaminomethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-hydroxymethyl-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-3-methyl-5-(2-pyridylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(2-cyclopropylethyl)-3-methyl-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(n-propoxymethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(i-propoxymethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(2-methoxyethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(i-propylaminomethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(N-methyl-N-1-propylaminomethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(cyclopropylaminomethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(n-propylaminomethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(cyclobutylaminomethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(i-butylaminomethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(i-propoxymethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-cyano-5-(n-butyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-cyano-5-(i-propoxymethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(cyclopropylsulfanylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(cyclopropanesulfinylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(t-butylsulfinylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(methylsulfanylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(ethylsulfanylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(i-propylsulfanylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-fluoro-5-(i-propylsulfanylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(t-butylsulfanylmethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(cyclopropylmethoxymethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(cyclobutoxymethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(cyclobutoxymethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(cyclopropylmethoxymethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-3-methyl-5-(i-propoxymethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-3-methyl-5-(n-butyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-cyano-3-methyl-5-(n-butyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-2-methyl-5-(i-propoxymethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 3,7-dichloro-5-(n-butyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 4,7-dichloro-5-(n-butyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(ethoxyethyl)-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(n-butyl)-5-methyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(i-propoxymethyl)-5-methyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(n-butyl)-5-cyano-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(n-butyl)-5-(hydroxymethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(n-butyl)-5-difluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(i-propoxymethyl)-5-difluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(n-butyl)-5-(1,1-difluoroethyl)-7-fluoro-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(n-butyl)-5-(1,1-difluoroethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-cyano-5-(n-butyl)-5-(1,1-difluoroethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 7-chloro-5-(ethoxymethyl)-5-(1,1-difluoroethyl)-5,10-dihydrobenzo[b]-1,7-naphthyridin-1 (2H)-one, 5-(allyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1 (2H)-one, 5-(2-methyl-1-propenyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(1-propynyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1 (2H)-one, 5-(cyanomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(ethylamino)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(dimethylamino)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(methylamino)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-ethoxyethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(i-propylamino)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(diethylamino)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(cyclopropylamino)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(pentyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(i-butyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(vinyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(imidazolylethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(pyrazolylethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(1,2,4-triazolylethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(i-propylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(i-propoxymethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(methylethylamino)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(i-propylethylamino)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(pyrrolidinyl)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(methoxy)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(i-propoxymethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(3-pentanylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(dimethoxymethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(i-butylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(cyclopropylmethylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(allylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-((R)-sec-butylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-((S)-sec-butylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(diethoxymethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 3-chloro-5-(propyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(butyl)-7-fluoro-2-methyl-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(i-propoxy)ethyl)-7-fluoro-2-methyl-5-trifluoromethyl-5,10-dihydrobenz o[b]-1,7-naphthyridin-1(2H)-one, 5-(i-propylaminomethyl)-7-fluoro-2-methyl-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(i-propoxymethyl)-7-fluoro-2-methyl-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-ethoxyethyl)-7-fluoro-2-methyl-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(sec-butylaminomethyl)-7-fluoro-2-methyl-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(cyclopentylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(cyclobutylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(dimethylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(pyrrolidinylmethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(cyclopropylaminomethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(dimethoxy)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(diethoxy)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, 5-(2-(1,3-dioxolanyl)methyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one, and 5-(2-(methoxy)ethyl)-7-fluoro-5-trifluoromethyl-5,10-dihydrobenzo[b]-1,7-naphthyridin-1(2H)-one; by formula I of U.S. Pat. No. 6,825,210, e.g., 3,7-dichloro-5-isopropoxymethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 3,7-dichloro-5-isopropoxymethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 3-chloro-7-fluoro-5-isopropoxymethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, 3-chloro-7-fluoro-5-isopropoxymethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, 3-chloro-5-isopropoxymethyl-5-trifluoromethyl-4-a,5,10,10a-tetrahydro-benzo[b][1,8]naphthyridine-7-carbonitrile, 3-chloro-5-isopropoxymethyl-1-oxy-5-trifluoromethyl-4-a,5,10,10a-tetrahydro-benzo[b][1,8]naphthyridine-7-carbonitrile, 3-bromo-7-cyano-5-trifluoromethyl-5-isopropoxymethyl-5,10-dihydrobenzo[b][1,8]naphthyridine, 3-bromo-7-cyano-5-trifluoromethyl-5-isopropoxymethyl-5,10-dihydrobenzo[b][1,8]naphthyridine-1-N-oxide, 3,7-dicyano-5-trifluoromethyl-5-isopropoxymethyl-5,10-dihydrobenzo[b][1,8]naphthyridine, and 3,7-dicyano-5-trifluoromethyl-5-isopropoxymethyl-5,10-dihydrobenzo[b][1,8]naphthyridine-1-N-oxide, (R)3,7-dichloro-5-isopropoxymethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine, (R)3,7-dichloro-5-isopropoxymethyl-5-trifluoromethyl-5,10-dihydro-benzo[b][1,8]naphthyridine 1-oxide, (R)3-chloro-7-fluoro-5-isopropoxymethyl-5-trifluoromethyl-5,10-dihydro-benz o[b][1,8]naphthyridine, (R)3-chloro-7-fluoro-5-isopropoxymethyl-5-trifluoromethyl-5,10-dihydro-benz o[b][1,8]naphthyridine 1-oxide, (R)3-chloro-5-isopropoxymethyl-5-trifluoromethyl-4-a,5,10,10a-tetrahydro-benzo[b][1,8]naphthyridine-7-carbonitrile, (R)3-chloro-5-isopropoxymethyl-1-oxy-5-trifluoromethyl-4-a,5,10,10a-tetrahydro-benzo[b][1,8]naphthyridine-7-carbonitrile, (R)3-bromo-7-cyano-5-trifluoromethyl-5-isopropoxymethyl-5,10-dihydrobenzo[b][1,8]naphthyridine, (R)3-bromo-7-cyano-5-trifluoromethyl-5-isopropoxymethyl-5,10-dihydrobenzo[b][1,8]naphthyridine-1-N-oxide, (R)3,7-dicyano-5-trifluoromethyl-5-isopropoxymethyl-5,10-dihydrobenzo[b][1, 8]naphthyridine, and (R)3,7-dicyano-5-trifluoromethyl-5-isopropoxymethyl-5,10-dihydrobenzo[b][1,8]naphthyridine-1-N-oxide; and by formula I of U.S. Pat. No. 6,844,340, e.g., b-chloro-10-(isopropoxmethyl)-10-(trifluoromethyl)-5,10-dihydropyrimido [5,4-b]quinolin-4 (3H)-one, 8-cyano-10-(isopropoxymethyl)-10-(trifluoromethyl)-5,10-dihydropyrimido[5,4-b]quinolin-4(3H)-one, 8-cyano-10-(isopropoxymethyl)-2-methyl-10-(trifluoromethyl)-5,10-dihydropyrimido[5,4-b]quinolin-4(3H)-one, 8-cyano-10-(2-cyclopropylethyl)-10-(trifluoromethyl)-5,10-dihydropyrimido-[5,4-b]quinolin-4(3H)-one, and (R)-8-cyano-10-(isopropoxymethyl)-10-(trifluoromethyl)-5,10-dihydropyrimido-[5,4-b]quinolin-4(3H)-one. Additional analogs include phosphonate derivatives of efavirenz, e.g, compounds described in U.S. Pat. No. 7,462,608.
  • K-Strophanthin
  • K-strophanthin (cymarine) is a cardiac glycoside derived from Strophanthus plant species. Analogs of strophanthins include other cardiac glycosides and their aglycone derivatives. Cardiac glycosides are derived from the cyclopentanoperhydro-phenanthrene nucleus of steroid compounds and are characterized by lactone rings joined by an ether linkage to one or more sugar residues. Aglycone derivatives of cardiac glycosides lack the carbohydrates characteristic of the cardiac glycosides. Exemplary analogs of strophanthins include, but are not limited to, G-strophanthin (ouabain), lanatoside A, desacetyllanatoside A, actyl digitoxin, digitoxin, lanatoside c, desacetyllanatoside c, digoxin, strophanthoside, scillaren A, proscillaridin A, uzarin, digitoxose, gitoxin, strophanthidine-3b-digitoxoside, strophanthidin aL-rhamnopyranoside, strophanthidol, oleandrin, acovenoside A, strophanthidine digilanobioside, strophanthidin-D-cymaroside, digitoxigenin-L-rhamnoside digitoxigenin theretoside, and the like. Aglycones include, but are not limited to, strophanthidin, digitoxigenin, uzarigenin, digoxigenin, digoxigenin 3,12-diacetate, gitoxigenin, gitoxigenin 3-acetate, gitoxigenin 3,16-diacetate, 16-acetyl gitoxigenin, acetyl strophanthidin, ouabagenin, 3-epidigoxigenin, and the like.
  • Mycophenolate Mofetil
  • Mycophenolate mofetil is an immunosuppressant described in U.S. Pat. No. 4,753,935. Analogs of mycophenolate mofetil are described by formula A in U.S. Pat. No. 4,753,935; by formulae I and II in U.S. Pat. No. 4,861,776, e.g., 2-(pyrrolidin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(isoxazolidin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(thiazolidin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(2-oxazin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(4-thiazin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(piperazin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(4-methylpiperazin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(3-methyl-1,3-perhydrodiazin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(perhydroazepin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(perhydro-2-oxazepin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(perhydro-4-thiazepin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(1,2-perhydrodiazepin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(3-ethyl-1,3-perhydrodiazepin-1-yl)ethyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 3-(pyrrolidin-1-yl)propyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 3-(piperidin-1-yl)propyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 3-(morpholin-1-yl)propyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 3-(perhydroazepin-1-yl)propyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 4-(pyrazolidin-1-yl)butyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 4-(piperidin-1-yl)butyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 4-(morpholin-1-yl)butyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 4-(4-thiazin-1-yl)butyl E-6-(1,3-dihdydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 4-(perhydro-2-oxazepin-1-yl)butyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 4-(4-t-butyl-1,4-perhydrodiazepin-1-yl)butyl E-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, morpholinoethyl (E)-6-(1,3-dihydro-4-heptanoyloxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuran yl)-4-methyl-4-hexenoate, morpholinoethyl (E)-6-{1,3-dihydro-4-{N-(4-carbomethoxyphenyl) carbamoyloxy}-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl}-4-methyl-4-hexenoate, morpholinoethyl (E)-6-{1,3-dihydro-4-(N,N-diethylcarbamoyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl}-4-methyl-4-hexenoate, morpholinoethyl (E)-6-{1,3-dihydro-4-(N-methyl-N-isobutylcarbamoyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl}-4-methyl-4-hexenoate, morpholinoethyl (E)-6-{1,3-dihydro-4-{N-(4-carboethoxyphenyl)-N-ethylcarbamoyloxy}-6-methoxy-7-methyl-3-oxo-5-iso-benzofuranyl}-4-methyl-4-hexenoate, morpholinoethyl (E)-6-(1,3-dihydro-4-thiopropionoyloxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, morpholinoethyl (E)-6-(1,3-dihydro-4-thiopivaloyloxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, morpholinoethyl (E)-6-(1,3-dihydro-4-thioheptanoyloxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, morpholinoethyl (E)-6-(1,3-dihydro-4-thiobenzoyloxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, morpholinoethyl (E)-6-{1,3-dihydro-4-[N-(4-carboethoxyphenyl)thiocarbamoyloxy]-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl}-4-methyl-4-hexenoate, morpholinoethyl (E)-6-[1,3-dihydro-4-(N,N-diethylthiocarbamoyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, morpholinoethyl (E)-6-[1,3-dihydro-4-(N-methyl-N-isobutylthiocarbamoyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, morpholinoethyl (E)-6-{1,3-dihydro-4-[N-(4-carboethoxyphenyl)-N-ethylthiocarbamoyloxy]-6-methoxy-7-methyl-3-oxo-5-iso-benzofuranyl}-4-methyl-4-hexenoate, morpholinoethyl (E)-6-[1,3-dihydro-4-(carboethoxy-carbonyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, morpholinoethyl (E)-6-[1,3-dihydro-4-(carbomethoxyethanoyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, morpholinoethyl (E)-6-[1,3-dihydro-4-(carboethoxyethanoyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, morpholinoethyl (E)-6-[1,3-dihydro-4-(3-carbomethoxypropionyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, morpholinoethyl (E)-6-[1,3-dihydro-4-(3-carboethoxypropionyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, morpholinoethyl (E)-6-[1,3-dihydro-4-(4-carbomethoxybutanoyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, morpholinoethyl (E)-6-[1,3-dihydro-4-(5-carbomethoxypentanoyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, 2-(thiazolidin-1-yl)ethyl E-6-[1,3-dihydro-4-(2-methylpropionyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl E-6-(1,3-dihydro-4-pivaloyloxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 3-(morpholin-1-yl)propyl E-6-(1,3-dihydro-4-adamantoyloxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 4-(perhydro-2-oxazepin-1-yl)butyl E-6-(1,3-dihydro-4-benzoyloxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 2-(4-thiazin-1-yl)ethyl E-6-{1,3-dihydro-4-[N-(4-carbomethoxyphenyl) thiocarbamoyloxy]-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl}-4-methyl-4-hexenoate, 3-(morpholin-1-yl)propyl (E)-6-(1,3-dihydro-4-carboethoxycarbonyloxy-6-methoxy-7-methyl-3-oxo-5-iso benzofuranyl)-4-methyl-4-hexenoate, 3-(morpholin-1-yl)propyl (E)-6-(1,3-dihydro-4-carbomethoxyethanoyloxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate, 3-(morpholin-1-yl)propyl (E)-6-(1,3-dihydro-4-carboethoxyethanoyloxy-6-methoxy-7-methyl-3-oxo-5-iso benzofuranyl)-4-methyl-4-hexenoate, and 3-(morpholin-1-yl)propyl (E)-6-[1,3-dihydro-4-(3-carbomethoxypropionyloxy)-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl]-4-methyl-4-hexenoate; by formula I in U.S. Pat. No. 5,380,879, e.g., methyl (E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, methyl (E)-6-(1,3-dihydro-4-trifluoromethylsulfonyloxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, methyl (E)-6-(1,3-dihydro-4-cyano-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 2-(morpholin-4-yl)ethyl E-6-[1,3-dihydro-4-(3,3-dimethylureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(morpholin-4-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(morpholin-4-yl)ethyl (E)-6-(1,3-dihydro-4-ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 2-(morpholin-4-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(morpholin-4-yl)ethyl (E)-6-[1,3-dihydro-4-(3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(morpholin-4-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-4-3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(morpholin-4-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diphenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(morpholin-4-yl)ethyl (E)-6-[1,3-dihydro-4-(3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate; and 2-(morpholin-4-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-4-3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate.2-(pyrrolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-dimethylureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-dimethylureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-dimethylureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl)propyl (E)-6-[1,3-dihydro-4-(3,3-dimethylureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3,3-dimethylureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-dimethylureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-dimethylureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl)propyl (E)-6-[1,3-dihydro-4-(3-methyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3-methyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-(1,3-dihydro-4-ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-(1,3-dihydro-4-ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-(1,3-dihydro-4-ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 3-(morpholin-4-yl)propyl (E)-6-(1,3-dihydro-4-ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-(1,3-dihydro-4-ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-(1,3-dihydro-4-ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-(1,3-dihydro-4-ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl)propyl (E)-6-[1,3-dihydro-4-(3,3-diethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3,3-diethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl)propyl (E)-6-[1,3-dihydro-4-(3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-[13-dihydro-4-(3-methyl-3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl)propyl (E)-6-[13-dihydro-4-(3-methyl-3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3-methyl-3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-3-ethyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diphenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diphenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diphenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl)propyl (E)-6-[1,3-dihydro-4-(3,3-diphenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3,3-diphenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diphenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3,3-diphenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl)propyl (E)-6-[1,3-dihydro-4-(3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl) propyl (E)-6-[1,3-dihydro-4-(3-methyl-3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3-methyl-3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-methyl-3-phenyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-butyl-3-propyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-butyl-3-propyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3-butyl-3-propyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl) propyl (E)-6-[1,3-dihydro-4-(3-butyl-3-propyl)ureido-6-methoxy-7-methyl-3oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3-butyl-3-propyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-butyl-3-propyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-butyl-3-propyl)ureido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(pyrrolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-(2-chlorophenyl)ureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(piperidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-3-(2-chlorophenyl)ureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(thiazolidin-3-yl)ethyl (E)-6-[1,3-dihydro-4-(3-(2-chlorophenyl)ureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 3-(morpholin-4-yl)propyl (E)-6-[1,3-dihydro-4-(3-(2-chlorophenyl)ureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 4-(morpholin-4-yl)butyl (E)-6-[1,3-dihydro-4-(3-(2-chlorophenyl)ureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, 2-(imidazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-(2-chlorophenyl)ureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate; and methyl E-6-[1,3-dihydro-4-(trifluoroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, methyl (E)-6-[1,3-dihydro-4-(difluoroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, methyl (E)-6-[1,3-dihydro-4-(fluoroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, methyl (E)-6-[1,3-dihydro-4-(trichloroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate; and methyl (E)-6-[1,3-dihydro-4-(fluorodichloroacetyl)-amino-6-methoxy-7-methyl-3-oxo isobenzofuran-5-yl]-4-methyl-4-hexenoate. (E)-6-[1,3-dihydro-4-(difluoroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoic acid, (E)-6-[1,3-dihydro-4-(fluoroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoic acid, (E)-6-[1,3-dihydro-4-(trichloroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoic acid, (E)-6-[1,3-dihydro-4-(fluorodichloroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoic acid. 2-(2-methyl-1,2-pyrazolidin-1-yl)ethyl (E)-6-[1,3-dihydro-4-(3-(2-chlorophenyl)ureido)-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate. (E)-6-[1,3-dihydro-4-(trifluoroacetyl) amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoic acid, (E)-6-[1,3-dihydro-4-acetamido-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoic acid, methyl (E)-6-[1,3-dihydro-4-(difluoroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, methyl (E)-6-[1,3-dihydro-4-(fluoroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, methyl (E)-6-[1,3-dihydro-4-(trichloroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, methyl (E)-6-[1,3-dihydro-4-(fluorodichloroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]-4-methyl-4-hexenoate, ethyl (E)-6-(1,3-dihydro-4-methylamino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, n-propyl (E)-6-(1,3-dihydro-4-methylamino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, isopropyl (E)-6-(1,3-dihydro-4-methylamino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, t-butyl (E)-6-(1,3-dihydro-4-methylamino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 2-trifluoromethylphenyl (E)-6-(1,3-dihydro-4-methylamino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, 2-chloro-3,4-dimethoxyphenyl (E)-6-(1,3-dihydro-4-methylamino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate, and ethyl (E)-6-(1,3-dihydro-4-ethylamino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoate; formula I in U.S. Pat. No. 5,441,953, e.g., E)-6-(1,3-dihydro-4-amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoic acid and E-6-(1,3-dihydro-4-(trifluoroacetyl)amino-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-hexenoic acid; formula I in U.S. Pat. No. 5,444,072, e.g., E) 6-(1,3-dihydro-4-hydroxy-6,7-dimethyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoic acid, (E) 6-(1,3-dihydro-6-ethyl-4-hydroxy-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoic acid, (E) 6-(6-cyclopropyl-1,3-dihydro-4-hydroxy-7-methyl-3oxoisobenzofuran-5-yl)-4-methyl-4-hexenoic acid, (E) 6-(1,3-dihydro-4-hydroxy-7-methyl-3-oxo-6-vinylisobenzofuran-5-yl)-4-methyl-4-hexenoic acid, (E) 6-(1,3-dihydro-6-ethyl-4-hydroxy-7-methyl-3-oxoisobenzofuran-5-yl)-3,4-dimethyl-4-hexenoic acid, and (E)-2-[2-[2-[1,3-dihydro-6-ethyl 4-hydroxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-1-yl]acetic acid; by formula I in U.S. Pat. No. 5,493,030, e.g., (+) (E) 6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-3,4-dimethyl-4-hexenoic acid, (E) 6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2-(S),4-dimethyl-4-hexenoic acid, (+) (E) 6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2,3,4-trimethyl-4-hexenoic acid, (E) 6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2-(S)-ethyl-4-methyl-4-hexenoic acid, 2-{2-[2-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)ethylidene]cyclohex-1-(S)-yl}acetic acid, 2-{2-[2-[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-1-(S)-yl}acetic acid, 2-{2-[2-(4-hydroxy-1,3-dihydro-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)ethylidene]cyclohex-1-(S)-yl}-2-(S)-methylacetic acid, 2-{2-[2-[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-1-(S)-yl}-2-(S)-ethylacetic acid, (−)-2-{2-[2o[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-1-(S)-yl}-2-methylacetic acid, including the 2-(R)-methylacetic acid and 2-(S)-methylacetic acid isomers, (−)-2-{4-[2-[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]tetrahydropyran-3-yl}acetic acid, (E) 2-[3-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-1-methylprop-1-en-1-yl]tetrahydropyran-1-carboxylic acid, (E) 2-(S)-{4-[2-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)ethylidene]-tetrahydropyran-3-(S)-yl}propionic acid, (E) 2-[3-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-1-methylprop-1-en-1-yl]cyclopentane-1-carboxylic acid, and (E) 2-[3-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-1-methylprop-1-en-1-yl]cyclohexane-1-carboxylic acid, most particularly: (E) 6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2-(S),4-dimethyl-4-hexenoic acid, (+) (E) 6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3 oxoisobenzofuran-5-yl)-2,3,4-trimethyl-4-hexenoic acid (having a melting point of 146.degree.-148.degree. C. when recrystallized from hexane/methylene chloride), (E) 2-(S)-{4-[2-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-isobenzofuran-5-yl)ethylidene]-tetrahydropyran-3-(S)-yl}propionic acid, and (−)-2-{2-[2-[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-1(S)-yl}-2-methylacetic acid, including the 2-(R)-methylacetic acid and 2-(S)-methylacetic acid isomers; by formula I in U.S. Pat. No. 5,538,969, e.g., (E)-6-(4-amino-1,3-dihydro-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2(S), 4-dimethyl-4-hexenoic acid, (E)-6-(4-amino-1,3-dihydro-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-3(S), 4-dimethyl-4-hexenoic acid, (E)-2-[2-[2-[4-amino-1,3-dihydro-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-1(S)-yl]acetic acid, and (E)-2-{4-[2-(4-amino-1,3-dihydro-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)ethylidene]tetrahydropyran-3(S)-yl}acetic acid; by formula I of U.S. Pat. No. 5,554,612, e.g., (E)-6-(4-amino-1,3-dihydro-6-ethyl-7-methyl-3-oxoisobenzofuran-5-yl)-4-methyl-4-hexenoic acid, (E)-6-(4-amino-1,3-dihydro-6-ethyl-7-methyl-3-oxoisobenzofuran-5-yl)-3,4-di methyl-4-hexenoic acid, and (E)-2-{2-[2-(4-amino-1,3-dihydro-6-ethyl-7-methyl-3-oxoisobenzofuran-5-yl)-ethylidene]-cyclopent-1-yl}acetic acid; and by formula I of U.S. Pat. No. 5,633,279, e.g., (+) (E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-3,4-dimethyl-4-hexenoic acid; (E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2(S), 4-dimethyl-4-hexenoic acid, (+) (E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2,3,4-trimethyl-4-hexenoic acid, (E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2(S)-ethyl-4-methyl-4-hexenoic acid, 2-{2-[2-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)ethylidene]cyclohex-1(S)-yl}acetic acid, 2-{2-[2-[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-1(S)-yl}acetic acid, 2-{2-[2-(4-hydroxy-1,3-dihydro-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)ethylidene]cyclohex-1(S)-yl}-2(S)-methylacetic acid, 2-{2-[2-[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-[(5)-yl}-2(S)-ethylacetic acid, (−) 2-{2-[2-[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-1(S)-yl}-2-methylacetic acid, including the 2(R)-methylacetic acid and 2(S)-methylacetic acid isomers, (−) 2-{4-[2-[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]tetrahydropyran-3-yl}acetic acid, (E)-2-[3-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-1-methylprop-1-en-1-yl]tetrahydropyran-1-carboxylic acid, (E)-2(S)-{4-[2-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)ethylidene]-tetrahydropyran-3(S)-yl}propionic acid, (E)-2-[3-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-1-methylprop-1-en-1-yl]cyclopentane-1-carboxylic acid, and (E)-2-[3-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-1-methylprop-1-en-1-yl]cyclohexane-1-carboxylic acid, (E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2(S), 4-dimethyl-4-hexenoic acid, (+) (E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl)-2,3,4-trimethyl-4-hexenoic acid, (E)-2(S)-{4-[2-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-isobenzofuran-5-yl)ethylidene]-tetrahydropyran-3(S)-yl}propionic acid, and (−) 2-{2-[2-[1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxoisobenzofuran-5-yl]ethylidene]cyclopent-1(S)-yl}-2-methylacetic acid, including the 2(R)-methylacetic acid and 2(S)-methylacetic acid isomers.
  • Conjugates
  • If desired, the agents used in any of the combinations described herein may be covalently attached to one another to form a conjugate of formula I.

  • (A)-(L)-(B)  (I)
  • In formula I, (A) is a Compound A and (B) is Compound B of a pair of agents from e.g., Table 1, and L is a covalent linker that tethers (A) to (B). Conjugates of the invention can be administered to a subject by any route and for the treatment of a filovirus-mediated disease (e.g., those described herein).
  • The conjugates of the invention can be prodrugs, releasing drug (A) and drug (B) upon, for example, cleavage of the conjugate by intracellular and extracellular enzymes (e.g., amidases, esterases, and phosphatases). The conjugates of the invention can also be designed to largely remain intact in vivo, resisting cleavage by intracellular and extracellular enzymes. The degradation of the conjugate in vivo can be controlled by the design of linker (L) and the covalent bonds formed with drug (A) and drug (B) during the synthesis of the conjugate.
  • Conjugates can be prepared using techniques familiar to those skilled in the art. For example, the conjugates can be prepared using the methods disclosed in G. Hermanson, Bioconjugate Techniques, Academic Press, Inc., 1996. The synthesis of conjugates may involve the selective protection and deprotection of alcohols, amines, ketones, sulfhydryls or carboxyl functional groups of drug (A), the linker, and/or drug (B). For example, commonly used protecting groups for amines include carbamates, such as tert-butyl, benzyl, 2,2,2-trichloroethyl, 2-trimethylsilylethyl, 9-fluorenylmethyl, allyl, and m-nitrophenyl. Other commonly used protecting groups for amines include amides, such as formamides, acetamides, trifluoroacetamides, sulfonamides, trifluoromethanesulfonyl amides, trimethylsilylethanesulfonamides, and tert-butylsulfonyl amides. Examples of commonly used protecting groups for carboxyls include esters, such as methyl, ethyl, tert-butyl, 9-fluorenylmethyl, 2-(trimethylsilyl)ethoxy methyl, benzyl, diphenylmethyl, O-nitrobenzyl, ortho-esters, and halo-esters. Examples of commonly used protecting groups for alcohols include ethers, such as methyl, methoxymethyl, methoxyethoxymethyl, methylthiomethyl, benzyloxymethyl, tetrahydropyranyl, ethoxyethyl, benzyl, 2-napthylmethyl, O-nitrobenzyl, P-nitrobenzyl, P-methoxybenzyl, 9-phenylxanthyl, trityl (including methoxy-trityls), and silyl ethers. Examples of commonly used protecting groups for sulfhydryls include many of the same protecting groups used for hydroxyls. In addition, sulfhydryls can be protected in a reduced form (e.g., as disulfides) or an oxidized form (e.g., as sulfonic acids, sulfonic esters, or sulfonic amides). Protecting groups can be chosen such that selective conditions (e.g., acidic conditions, basic conditions, catalysis by a nucleophile, catalysis by a lewis acid, or hydrogenation) are required to remove each, exclusive of other protecting groups in a molecule. The conditions required for the addition of protecting groups to amine, alcohol, sulfhydryl, and carboxyl functionalities and the conditions required for their removal are provided in detail in T. W. Green and P. G. M. Wuts, Protective Groups in Organic Synthesis (2nd Ed.), John Wiley & Sons, 1991 and P. J. Kocienski, Protecting Groups, Georg Thieme Verlag, 1994. Additional synthetic details are provided below.
  • Linkers
  • The linker component of the invention is, at its simplest, a bond between drug (A) and drug (B), but typically provides a linear, cyclic, or branched molecular skeleton having pendant groups covalently linking drug (A) to drug (B).
  • Thus, linking of drug (A) to drug (B) is achieved by covalent means, involving bond formation with one or more functional groups located on drug (A) and drug (B). Examples of chemically reactive functional groups which may be employed for this purpose include, without limitation, amino, hydroxyl, sulfhydryl, carboxyl, carbonyl, carbohydrate groups, vicinal diols, thioethers, 2-aminoalcohols, 2-aminothiols, guanidinyl, imidazolyl, and phenolic groups.
  • The covalent linking of drug (A) and drug (B) may be effected using a linker that contains reactive moieties capable of reaction with such functional groups present in drug (A) and drug (B). For example, an amine group of drug (A) may react with a carboxyl group of the linker, or an activated derivative thereof, resulting in the formation of an amide linking the two.
  • Examples of moieties capable of reaction with sulfhydryl groups include a-haloacetyl compounds of the type XCH2CO— (where X=Br, Cl, or I), which show particular reactivity for sulfhydryl groups, but which can also be used to modify imidazolyl, thioether, phenol, and amino groups as described by Gurd, Methods Enzymol. 11:532 (1967). N-Maleimide derivatives are also considered selective towards sulfhydryl groups, but may additionally be useful in coupling to amino groups under certain conditions. Reagents such as 2-iminothiolane (Traut et al., Biochemistry 12:3266 (1973)), which introduce a thiol group through conversion of an amino group, may be considered as sulfhydryl reagents if linking occurs through the formation of disulfide bridges.
  • Examples of reactive moieties capable of reaction with amino groups include, for example, alkylating and acylating agents. Representative alkylating agents include:
  • (i) α-haloacetyl compounds, which show specificity towards amino groups in the absence of reactive thiol groups and are of the type XCH2CO— (where X=Br, Cl, or I), for example, as described by Wong Biochemistry 24:5337 (1979);
  • (ii) N-maleimide derivatives, which may react with amino groups either through a Michael type reaction or through acylation by addition to the ring carbonyl group, for example, as described by Smyth et al., J. Am. Chem. Soc. 82:4600 (1960) and Biochem. J. 91:589 (1964);
  • (iii) aryl halides such as reactive nitrohaloaromatic compounds;
  • (iv) alkyl halides, as described, for example, by McKenzie et al., J. Protein Chem. 7:581 (1988);
  • (v) aldehydes and ketones capable of Schiff's base formation with amino groups, the adducts formed usually being stabilized through reduction to give a stable amine;
  • (vi) epoxide derivatives such as epichlorohydrin and bisoxiranes, which may react with amino, sulfhydryl, or phenolic hydroxyl groups;
  • (vii) chlorine-containing derivatives of s-triazines, which are very reactive towards nucleophiles such as amino, sufhydryl, and hydroxyl groups;
  • (viii) aziridines based on s-triazine compounds detailed above, e.g., as described by Ross, J. Adv. Cancer Res. 2:1 (1954), which react with nucleophiles such as amino groups by ring opening;
  • (ix) squaric acid diethyl esters as described by Tietze, Chem. Ber. 124:1215 (1991); and
  • (x) α-haloalkyl ethers, which are more reactive alkylating agents than normal alkyl halides because of the activation caused by the ether oxygen atom, as described by Benneche et al., Eur. J. Med. Chem. 28:463 (1993).
  • Representative amino-reactive acylating agents include:
  • (i) isocyanates and isothiocyanates, particularly aromatic derivatives, which form stable urea and thiourea derivatives respectively;
  • (ii) sulfonyl chlorides, which have been described by Herzig et al., Biopolymers 2:349 (1964);
  • (iii) acid halides;
  • (iv) active esters such as nitrophenylesters or N-hydroxysuccinimidyl esters;
  • (v) acid anhydrides such as mixed, symmetrical, or N-carboxyanhydrides;
  • (vi) other useful reagents for amide bond formation, for example, as described by M. Bodansky, Principles of Peptide Synthesis, Springer-Verlag, 1984;
  • (vii) acylazides, e.g., wherein the azide group is generated from a preformed hydrazide derivative using sodium nitrite, as described by Wetz et al., Anal. Biochem. 58:347 (1974); and
  • (viii) imidoesters, which form stable amidines on reaction with amino groups, for example, as described by Hunter and Ludwig, J. Am. Chem. Soc. 84:3491 (1962).
  • Aldehydes and ketones may be reacted with amines to form Schiff's bases, which may advantageously be stabilized through reductive amination. Alkoxylamino moieties readily react with ketones and aldehydes to produce stable alkoxamines, for example, as described by Webb et al., in Bioconjugate Chem. 1:96 (1990).
  • Examples of reactive moieties capable of reaction with carboxyl groups include diazo compounds such as diazoacetate esters and diazoacetamides, which react with high specificity to generate ester groups, for example, as described by Herriot, Adv. Protein Chem. 3:169 (1947). Carboxyl modifying reagents such as carbodiimides, which react through O-acylurea formation followed by amide bond formation, may also be employed.
  • It will be appreciated that functional groups in drug (A) and/or drug (B) may, if desired, be converted to other functional groups prior to reaction, for example, to confer additional reactivity or selectivity. Examples of methods useful for this purpose include conversion of amines to carboxyls using reagents such as dicarboxylic anhydrides; conversion of amines to thiols using reagents such as N-acetylhomocysteine thiolactone, S-acetylmercaptosuccinic anhydride, 2-iminothiolane, or thiol-containing succinimidyl derivatives; conversion of thiols to carboxyls using reagents such as α-haloacetates; conversion of thiols to amines using reagents such as ethylenimine or 2-bromoethylamine; conversion of carboxyls to amines using reagents such as carbodiimides followed by diamines; and conversion of alcohols to thiols using reagents such as tosyl chloride followed by transesterification with thioacetate and hydrolysis to the thiol with sodium acetate.
  • So-called zero-length linkers, involving direct covalent joining of a reactive chemical group of drug (A) with a reactive chemical group of drug (B) without introducing additional linking material may, if desired, be used in accordance with the invention.
  • More commonly, however, the linker will include two or more reactive moieties, as described above, connected by a spacer element. The presence of such a spacer permits bifunctional linkers to react with specific functional groups within drug (A) and drug (B), resulting in a covalent linkage between the two. The reactive moieties in a linker may be the same (homobifunctional linker) or different (heterobifunctional linker, or, where several dissimilar reactive moieties are present, heteromultifunctional linker), providing a diversity of potential reagents that may bring about covalent attachment between drug (A) and drug (B).
  • Spacer elements in the linker typically consist of linear or branched chains and may include a C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, C2-6 heterocyclyl, C6-12 aryl, C7-14 alkaryl, C3-10 alkheterocyclyl, or C1-10 heteroalkyl.
  • In some instances, the linker is described by formula (II):

  • G1-(Z1)o—(Y1)s—(Z2)s—(R30)—(Z3)t—(Y2)v—(Z4)p-G2  (II)
  • In formula (II), G1 is a bond between drug (A) and the linker; G2 is a bond between the linker and drug (B); Z1, Z2, Z3, and Z4 each, independently, is selected from O, S, and NR31; R31 is hydrogen, C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C2-6 heterocyclyl, C6-12 aryl, C7-14 alkaryl, C3-10 alkheterocyclyl, or C1-7 heteroalkyl; Y1 and Y2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; o, p, s, t, u, and v are each, independently, 0 or 1; and R30 is a C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, C2-6 heterocyclyl, C6-12 aryl, C7-14 alkaryl, C3-10 alkheterocyclyl, or C1-10 heteroalkyl, or a chemical bond linking G1-(Z1)o—(Y1)u—(Z2)s— to —(Z3)t—(Y2)v—(Z4)p-G2.
  • Examples of homobifunctional linkers useful in the preparation of conjugates of the invention include, without limitation, diamines and diols selected from ethylenediamine, propylenediamine and hexamethylenediamine, ethylene glycol, diethylene glycol, propylene glycol, 1,4-butanediol, 1,6-hexanediol, cyclohexanediol, and polycaprolactone diol.
  • Formulation of Pharmaceutical Compositions
  • The compositions, methods, and kits of the invention can include formulation(s) of compound(s) that, upon administration to a subject, result in a concentration of the compound(s) that treats a filovirus-mediated disease. The compound(s) may be contained in any appropriate amount in any suitable carrier substance, and are generally present in an amount of 1-95% by weight of the total weight of the composition. The composition may be provided in a dosage form that is suitable for the oral, parenteral (e.g., intravenously or intramuscularly), rectal, dermatological, cutaneous, nasal, vaginal, inhalant, skin (patch), ocular, intrathecal, or intracranial administration route. Thus, the composition may be in the form of, e.g., tablets, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectables, implants, sprays, or aerosols. The pharmaceutical compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy, 20th edition, 2000, ed. A. R. Gennaro, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York).
  • Pharmaceutical compositions according to the invention or used in the methods of the invention may be formulated to release the active compound immediately upon administration or at any predetermined time or time period after administration. The latter types of compositions are generally known as controlled release formulations, which include (i) formulations that create substantially constant concentrations of the agent(s) of the invention within the body over an extended period of time; (ii) formulations that after a predetermined lag time create substantially constant concentrations of the agent(s) of the invention within the body over an extended period of time; (iii) formulations that sustain the agent(s) action during a predetermined time period by maintaining a relatively constant, effective level of the agent(s) in the body with concomitant minimization of undesirable side effects associated with fluctuations in the plasma level of the agent(s) (sawtooth kinetic pattern); (iv) formulations that localize action of agent(s), e.g., spatial placement of a controlled release composition adjacent to or in the diseased tissue or organ; (v) formulations that achieve convenience of dosing, e.g., administering the composition once per week or once every two weeks; and (vi) formulations that target the action of the agent(s) by using carriers or chemical derivatives to deliver the combination to a particular target cell type. Administration of compound(s) in the form of a controlled release formulation is especially preferred for compounds having a narrow absorption window in the gastro-intestinal tract or a relatively short biological half-life.
  • Any of a number of strategies can be pursued in order to obtain controlled release in which the rate of release outweighs the rate of metabolism of the compound in question. In one example, controlled release is obtained by appropriate selection of various formulation parameters and ingredients, including, e.g., various types of controlled release compositions and coatings. Thus, the compound(s) are formulated with appropriate excipients into a pharmaceutical composition that, upon administration, releases the compound(s) in a controlled manner. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, molecular complexes, microspheres, nanoparticles, patches, and liposomes.
  • Delivery of Compound(s)
  • It is not intended that administration of compounds be limited to a single formulation and delivery method for all compounds of a combination. The combination can be administered using separate formulations and/or delivery methods for each compound of the combination using, for example, any of the above-described formulations and methods. In one example, a first agent is delivered orally, and a second agent is delivered intravenously.
  • Dosages
  • The dosage of a compound or a combination of compounds depends on several factors, including: the administration method, the type of disease to be treated, the severity of the infection, whether administration first occurs at an early or late stage of infection, and the age, weight, and health of the patient to be treated.
  • For combinations that include a synergistic pair of agents identified herein (e.g., a pair of Table 4), the recommended dosage for the anti-viral agent can be less than or equal to the recommended dose as given in the Physician's Desk Reference, 60th Edition (2006).
  • As described above, the compound(s) in question may be administered orally in the form of tablets, capsules, elixirs or syrups, or rectally in the form of suppositories. Parenteral administration of a compound is suitably performed, for example, in the form of saline solutions or with the compound(s) incorporated into liposomes. In cases where the compound in itself is not sufficiently soluble to be dissolved, a solubilizer such as ethanol can be applied. The correct dosage of a compound can be determined by examining the efficacy of the compound in viral replication assays, as well as its toxicity in humans.
  • An antiviral agent is usually given by the same route of administration that is known to be effective for delivering it as a monotherapy. When used in combination therapy according to the methods of this invention, an agent of Table 2 or Table 3 is dosed in amounts and frequencies equivalent to or less than those that result in its effective monotherapeutic use.
  • Additional Applications
  • If desired, the compounds of the invention may be employed in mechanistic assays to determine whether other combinations, or single agents, are as effective as the combinations of the invention in inhibiting a viral disease (e.g., those described herein) using assays generally known in the art. For example, candidate compounds may be tested, alone or in combination (e.g., with an agent that inhibits viral replication, such as those described herein) and applied to cells (e.g., hepatic cells such as HepG2, kidney epithelial cells such as 293T, macrophages such as THP-1, or isolated primary cells). After a suitable time, viral replication or load of these cells is examined. A decrease in viral replication or viral load identifies a candidate compound or combination of agents as an effective agent for treating a viral disease.
  • The agents of the invention are also useful tools in elucidating mechanistic information about the biological pathways involved in viral diseases. Such information can lead to the development of new combinations or single agents for treating, preventing, or reducing a viral disease. Methods known in the art to determine biological pathways can be used to determine the pathway, or network of pathways affected by contacting cells (e.g., hepatic cells) infected with a virus with the compounds of the invention. Such methods can include, analyzing cellular constituents that are expressed or repressed after contact with the compounds of the invention as compared to untreated, positive or negative control compounds, and/or new single agents and combinations, or analyzing some other activity of the cell or virus such as an enzymatic activity, nutrient uptake, and proliferation. Cellular components analyzed can include gene transcripts, and protein expression. Suitable methods can include standard biochemistry techniques, radiolabeling the compounds of the invention (e.g., 14C or 3H labeling), and observing the compounds binding to proteins, e.g., using 2D gels, gene expression profiling. Once identified, such compounds can be used in in vivo models (e.g., knockout or transgenic mice) to further validate the tool or develop new agents or strategies to treat viral disease.
  • Exemplary Candidate Compounds
  • Peptide Moieties
  • Peptides, peptide mimetics, and peptide fragments (whether natural, synthetic or chemically modified) are suitable for use in the methods of the invention. Exemplary inhibitors include compounds that reduce the amount of a target protein or RNA levels (e.g., antisense compounds, dsRNA, ribozymes) and compounds that compete with viral reproduction machinery (e.g., dominant negative proteins or polynucleotides encoding the same).
  • Antisense Compounds
  • The biological activity of any protein that increases viral replication, viral RNA or DNA replication, viral RNA translation, viral protein processing or activity, or viral packaging can be reduced through the use of an antisense compound directed to RNA encoding the target protein. Antisense compounds can be identified using standard techniques. For example, accessible regions of the target the mRNA of the target enzyme can be predicted using an RNA secondary structure folding program such as MFOLD (M. Zuker, D. H. Mathews & D. H. Turner, Algorithms and Thermodynamics for RNA Secondary Structure Prediction: A Practical Guide. In: RNA Biochemistry and Biotechnology, J. Barciszewski & B. F. C. Clark, eds., NATO ASI Series, Kluwer Academic Publishers, (1999)). Sub-optimal folds with a free energy value within 5% of the predicted most stable fold of the mRNA are predicted using a window of 200 bases within which a residue can find a complimentary base to form a base pair bond. Open regions that do not form a base pair are summed together with each suboptimal fold and areas that are predicted as open are considered more accessible to the binding to antisense nucleobase oligomers. Other methods for antisense design are described, for example, in U.S. Pat. No. 6,472,521, Antisense Nucleic Acid Drug Dev. 7:439-444 (1997), Nucleic Acids Res. 28:2597-2604 (2000), and Nucleic Acids Res. 31:4989-4994 (2003).
  • RNA Interference
  • The biological activity of a molecule involved in a viral infection or viral replication can be reduced through the use of RNA interference (RNAi), employing, e.g., a double stranded RNA (dsRNA) or small interfering RNA (siRNA) directed to the signaling molecule in question (see, e.g., Miyamoto et al., Prog. Cell Cycle Res. 5:349-360 (2003); U.S. Pat. Application Publication No. 20030157030). Methods for designing such interfering RNAs are known in the art. For example, software for designing interfering RNA is available from Oligoengine (Seattle, Wash.).
  • Dominant Negative Proteins
  • One skilled in the art would know how to make dominant negative proteins to the molecules involved in a viral infection or viral replication. Such dominant negative proteins are described, for example, in Gupta et al., J. Exp. Med., 186:473-478 (1997); Maegawa et al., J. Biol. Chem. 274:30236-30243 (1999); and Woodford-Thomas et al., J. Cell Biol. 117:401-414 (1992).
  • EXAMPLES
  • The following examples are intended to illustrate rather than limit the invention.
  • Example 1 Identification of single therapeutic agents using a genetically engineered fully Infectious Ebola Virus
  • A genetically engineered Ebolavirus (EBOV) expressing the green fluorescent protein (GFP) has been used to develop a high throughput assay for drug discovery (Towner et al., Virology, 332: 20 (2005)). The genetically engineered virus has the unique property of making infected cells glow fluorescent green. The green signal can be detected by fluorescence microscopy, flow cytometry, or fluorometry, and other types of fluorescence detection systems adapted for high-throughput format. In order to perform the assay, seed Vero or HepG2 cells on a 96-well plate at 40,000 cells/well in a total volume of 100 μL/well. The plated cells are incubated cells at 37° C., 5% CO2 until they achieve a stationary growth phase. Next 50 μl of pre-diluted compounds are added at a 4× concentration to each well to achieve the desired final concentration. Finally, 50 μl of GFP-EBOV (multiplicity of infection of 0.01) is added to cells. Plates are centrifuged at 2000 RPM, 5 minutes following the addition of EBOV. Cells are incubated an additional 48 hours at 37° C., 5% CO2, and the plates are then removed from the incubator and the amount of GPF in each well is determined using a spectrofluorometer from Molecular Devices (excitation: 485 nm, emission: 515 nm, cutoff: 495 nm). Antiviral activity is identified by the inhibition of GFP compared to untreated control cells.
  • To confirm that a decrease in GFP activity correlates with inhibition of Ebola replication and not an increase in cell death, a counter screen is run in tandem using uninfected Vero or HepG2 cells. Seed cells on a 96-well plate as described above. Cells are mock infected and treated with compounds the following day. After a 48 hour incubation at 37° C., 5% CO2, the amount of ATP is determined using Promega Cell Titer-Glo luminescent Cell viability kit. This assay provides a quantitative measure of the levels of ATP in the cell cultures in each well, where higher levels of ATP correlate with greater cellular viability. Thus, a compound with antiviral activity is expected to inhibit the levels of GPF measured without any or minimal effect on the ATP levels measured by the Cell Titer-Glo assay.
  • The assay described above was used to identify agents that inhibit the replication of Ebola virus in Vero cells. Agents thus identified (IC50) are listed in Table 6. The extent of inhibition is represented as maximum fraction inhibition (Max Effect), where a value of 1.0 is indicative of complete inhibition, and 0 is indicative of no effect. As available, 50% inhibitory concentrations of the agents are provided.
  • TABLE 6
    Max IC50
    Agent Effect (μM) Previously described activity
    Cetrimide 0.692 5.6 Antibiotic
    Pentamidine 0.422 10.5 Antiparasitic
    Hexachlorophene 0.713 3.2 Bacterial dihydrolipoamide dehydrogenase
    inhibitor
    Emetine dihydrochloride hydrate 0.893 0.3 Bacteria ribosome binder
    Puromycin dihydrochloride 0.835 6.6 Bacteria ribosome binder
    Thiostrepton 0.454 Bacteria ribosome binder
    Gramicidin 0.678 1.7 Bacterial membrane disrupter
    Chlorhexidine Diacetate 0.543 6.1 Bacterial wall disrupter
    Teicoplanin 0.930 5.6 Bacterial penicillin binding protein inhibitor
    Cephapirin Sodium 0.662 9.8 Bacterial penicillin binding protein inhibitor
    Pyrithione Zinc 0.767 2.4 Antifungal
    Haloprogin 0.588 2.9 Antifungal
    Ciclopirox Olamine 0.431 Fungal Na—K channel blocker
    Hycanthone 0.597 10.4 Antiparasitic, anthelmintic
    Niclosamide 0.842 2.6 Antiparasitic, inhibits parasite metabolism
    Efavirenz 0.516 7.8 HIV non-nucleoside reverse transcriptase
    inhibitor
    Ritonavir 0.485 HIV protease inhibitor
    Chromomycin A3 0.629 0.4 DNA damaging agent, DNA minor groove
    binder
    Azacitidine 0.595 5.0 DNA methyltransferase inhibitor
    (−)-Arctigenin 0.411 DNA_function, topoisomerase 2 inhibitor
    Danazol 0.546 12.9 Androgen receptor antagonist
    Bicalutamide 0.464 Androgen receptor antagonist
    Hydroxyprogesterone Caproate 0.719 3.7 Estrogen receptor antagonist
    Equilin 0.424 Estrogen receptor antagonist
    Clomiphene Citrate 0.912 3.0 Estrogen receptor antagonist, antifungal
    squalene epoxidase inhibitor
    Quinestrol 0.549 7.2 Estrogen receptor 1 agonist
    Tamoxifen 0.900 2.3 Estrogen receptor 1 agonist
    Raloxifene Hydrochloride 0.691 2.4 Estrogen receptor 1 agonist
    Tamoxifen Citrate 0.900 1.7 Estrogen receptor 1 agonist
    Toremifene Citrate 0.900 >1 Estrogen receptor 1 inhibitor
    Toremifene 0.461 2.7 Estrogen receptor 1 inhibitor
    Diethylstilbestrol 0.436 Estrogen receptor 1 inhibitor
    Tibolone 1.119 1.2 Nuclear GC glucocorticoid receptor
    activator
    Lomerizine Dihydrochloride 0.665 10.1 Ca channel blocker
    Maduramicin NH4 0.452 Ca ionophore
    Bepridil Hydrochloride 0.720 3.1 Ca-A1A channel blocker
    Monohydrate
    Loperamide 0.716 5.5 Ca-A1A channel blocker
    Loperamide Hydrochloride 0.418 Ca-A1A channel blocker
    K-Strophanthin 0.900 <0.5 H channel blocker
    Carbonyl cyanide 4- 0.752 4.5 Ion_transport, H ionophore
    (trifluoromethoxy)phenylhydrazone
    Beta Escin 0.878 6.3 Ion_transport, Na channel modulator
    Auranofin 0.020 <1 Gold compound
    Calcimycin A23187 0.789 1.1 Ions, Cation Ionophore
    Edetate Calcium Disodium 0.841 6.6 Ions, Ca chelator
    Octyl Gallate 0.439 Ions, Ga
    Magnesium Sulfate 0.719 9.2 Ions, Mg
    Rescinnamine 0.512 Angiotensin-Converting Enzyme inhibitor
    Cilastatin Sodium 0.912 8.8 Dipeptidase 1 inhibitor
    Bromelain 0.769 4.0 Prostaglandin E2 antagonist
    Quinacrine Dihydrochloride 0.779 9.5 Phospholipase A2 receptor inhibitor
    PGG (1,2,3,4,6-b-O-Pentagalloyl 0.487 3.2 Metabolism, unknown herbal
    glucose)
    L-Asparagine 0.771 7.2 Metabolism, amino acid
    Chondroitin Sulfate 0.535 11.4 Sietary supplement
    Atovaquone 0.900 <1 Dihydroorotate dehydrogenase inhibitor
    Mycophenolate Mofetil 0.900 <1 Inosine monophosphate dehydrogenase
    inhibitor
    Mycophenolic Acid 0.800 <1 Inosine monophosphate dehydrogenase
    inhibitor
    Flucytosine 0.793 4.4 Thymidylate synthase inhibitor
    6-Azauridine 0.800 <1 Pyrimidine biowynthesis inhibitor
    Bafilomycin A1 0.880 0.019 Vacuolar-type H+ pump or ATPase
    inhibitor
    2-Methoxy-antimycin A3 0.791 1.2 Mitochondrial ATP synthase B chain
    inhibitor, cytochrome B inhibitor
    Oligomycin 0.426 Mitochondrial ATP synthase B chain
    inhibitor
    Antimycin A 0.850 <0.04 Mitochondrial ATP synthase B chain
    inhibitor, cytochrome b inhibitor
    Rotenone 0.700 <2 Mitochondrial ATP synthase B
    inhibitor; Tubulin destablizer
    FR122047 0.688 3.7 Cyclooxygenase-1 inhibitor
    Fenoprofen Calcium 0.454 Cyclooxygenase 1 and 2 inhibtor
    Perhexiline Maleate 0.890 4.1 Carnitine palmitoyltransferase 1A/Carnitine
    palmitoyltransferase 2 inhibitor
    PDMP Hydrochloride 0.390 UDP-glucose ceramide glucosyltransferase
    inhibitor
    Licochalcone-A 0.522 4.7 Metabolism, antioxidant
    Tiratricol 0.478 Metabolism, antioxidant
    CAPE 0.368 Metabolism, antioxidant
    Amlodipine Besylate 0.528 13.2 Redox metabolism, Carbonic anhydrase I
    inhibitor; Ca channel blocker
    Diphenyleneiodonium chloride 0.800 <1.6 Redox metabolism, iNOS inhibitor
    Terconazole 0.785 7.7 Sterol metabolism, fungal ERG11/CYP51
    inhibitor
    Sulconazole Nitrate 0.904 4.1 Sterol metabolism, fungal ERG11/CYP51
    inhibitor inhib
    Tioconazole 0.729 8.5 Sterol metabolism, fungal ERG11/CYP51
    inhibitor
    Oxiconazole Nitrate 0.411 Fungal ERG11/CYP51 inhib
    Simvastatin 0.690 2.0 HMG-CoA reductase (or 3-hydroxy-3-
    methyl-glutaryl-CoA reductase/fung HMG2
    inhib
    Cerivastatin Sodium 0.443 0.1 HMG-CoA reductase (or 3-hydroxy-3-
    methyl-glutaryl-CoA reductase/fung HMG2
    inhib
    Metergoline 0.577 6.1 Serotonin receptor inhibitor
    Thioridazine Hydrochloride 0.915 4.3 5-Hydroxytryptamine(serotonin) receptor
    2A/adrenergic, alpha-1A receptor inhibitor,
    dopamine receptor antagonist/
    ADRA1A inhib; DRD antag
    Thiethylperazine Maleate 0.832 4.4 5-Hydroxytryptamine(serotonin) receptor
    2A/adrenergic, alpha-1A receptor inhibitor,
    dopamine receptor antagonist/
    ADRA1A inhib; DRD antag
    Cyproheptadine Hydrochloride 0.493 5-Hydroxytryptamine(serotonin) receptor
    2A/histamine H(1) receptor inhibitor
    Prochlorperazine Edisylate 0.890 5.2 5-Hydroxytryptamine(serotonin) receptor
    2A/histamine H(1) receptor inhibitor;
    dopamine D2 receptor agonist
    Triflupromazine Hydrochloride 0.541 12.1 5-Hydroxytryptamine (serotonin) receptor
    2B/cholinergic receptor inhibitor;
    dopamine receptor antagonist
    Paroxetine Hydrochloride 0.820 9.3 Solute carrier family 6 (neurotransmitter
    Hemihydrate transporter, serotonin),
    member 4 inhibibitor
    Sertraline Hydrochloride 0.418 Solute carrier family 6 (neurotransmitter
    transporter, serotonin),
    member 4 inhibibitor
    Clomipramine Hydrochloride 0.585 6.3 Solute carrier family 6 (neurotransmitter
    transporter, serotonin),
    member 4/member 2 inhibitor; glutathione
    transferase inhibitor
    Fenretinide 0.426 Nuclear, retinoic acid receptor binder
    Ciclesonide 0.512 16.0 Nuclear receptor subfamily 3, group C,
    member 1 (glucocorticoid receptor)
    activator
    Brefeldin A 0.900 <1.6 ADP-ribosylation factor 1 binder
    Carbobenzoxy-valinyl- 0.607 2.9 Calpain inhibitor
    phenylalaninal
    BML-248 0.554 0.2 Cysteine Protease inhibitor
    Leupeptin HCl 0.865 3.2 Cysteine Protease inhibitor
    Tunicamycin 0.800 <0.28 P-MurNAc penapeptide synthase;
    glycosyltransferase inhibitor
    MG115 0.693 0.9 Proteasome 26S inhibitor
    MG-132 0.900 <0.6 Proteasome 26S inhibitor
    Epoxomicin 0.466 Proteasome inhibitor
    Cycloheximide 0.900 <1.6 Ribosome ribosomal peptidyl transferase
    inhibitor
    Anisomycin 0.900 <1.6 Ribosome ribosomal peptidyl transferase
    inhibitor
    CGS 15943 0.449 Adenosine receptor inhibitor
    Pimozide 0.685 4.8 Receptor_Dopamine receptor
    antagonist; Delta-type opioid receptor
    binder; Calcium channel alpha-1G blocker
    Guanethidine Monosulfate 0.793 8.7 Receptor_adrenergic, Adrenergic receptor
    antagonist; solute carrier family 6
    (neurotransmitter transporter, serotonin),
    member 4 inhibitor
    Flupentixol Dihydrochloride 0.660 3.8 Adrenergic receptor 1A antagonist
    Perphenazine 0.793 5.8 Adrenergic receptor 1A inhibitor;
    Dopaminergic 2A receptor antagonist
    Trifluoperazine Hydrochloride 0.955 5.0 Adrenergic receptor 1A inhibitor;
    Dopaminergic antagonist
    EGFR Inhibitor 0.659 0.6 Epidermal growth factor receptor inhibitor
    Sunitinib Malate 0.788 4.4 Vascular endothelial growth factor recptor 2
    inhibitor
    Chlorcyclizine Hydrochloride 0.456 Histamine receptor inhibitor
    Astemizole 0.835 4.3 Histamine receptor 1 inhibitor
    Clemastine Fumarate 0.850 4.0 Histamine receptor 1 inhibitor
    Terfenadine 0.914 6.1 Histamine receptor 1 inhibitor
    Chlorphenoxamine Hydrochloride 0.789 5.0 Histamine receptor 1 inhibitor
    Oxatomide 0.622 2.1 Histamine receptor 1 inhibitor
    Azelastine 0.648 121.6 Histamine receptor 1 inhibitor
    Clemastine 0.525 2.9 Histamine receptor 1 inhibitor
    Methdilazine Hydrochloride 0.519 7.1 Histamine receptor 1 inhibitor
    Homochlorcyclizine 0.508 8.7 Histamine receptor 1 inhibitor
    dihydrochloride
    Desloratadine 0.430 Histamine receptor 1 inhibitor
    Flunarizine Hydrochloride 0.402 Histamine receptor 1 inhibitor
    Loratadine 0.371 Histamine receptor 1 inhibitor
    Doxylamine Succinate 0.731 5.2 Receptor_histamine, histamine receptor
    H1/cholinergic receptor 1 inhibitor
    Propoxyphene Hydrochloride 0.422 Receptor_neural, opiate receptor binder
    Benztropine Mesylate 0.633 3.7 Receptor_neural, cholinergic receptor 1
    inhibitor
    Dicyclomine Hydrochloride 0.596 5.7 Receptor_neural, cholinergic receptor 1
    inhibitor
    Piperacetazine 0.641 6.6 Dopamine receptor antagonist
    Vanoxerin 0.564 5.8 Dopamine receptor antagonist
    Pergolide Mesylate 0.383 Dopamine receptor 1 and 2 agonist
    Acetophenazine Maleate 0.478 Dopamine receptor 1 and 2 antagonist
    Bromocriptine Mesylate 0.422 dopamine receptor 2 agonist
    Fluphenazine Hydrochloride 0.988 4.2 Dopamine receptor 2 antagonist
    N-(4-Aminobenzoyl)-L-glutamic 0.855 9.7 GABA receptor A inhibitor
    acid
    Aripiprazole 0.807 7.5 Histamine receptor inhibitor; dopaminergic
    receptor 2 antagonist
    Drotaverine Hydrochloride 0.367 Opioid receptor, mu 1/K1 antagonist
    Maprotiline Hydrochloride 0.795 7.6 Solute carrier family 6 (neurotransmitter
    transporter, noradrenalin),
    member/histamine receptor H1 inhibitor;
    adrenergic receptor 1A antagonist
    CKI7 0.565 10.7 Signal, casein kinase inhibitor
    Triptolide 0.850 <0.1 DEVD cleaving caspase activator
    NSC 625987 0.418 CDK4-CycD1 inhibitor
    Bay 41-2272 0.900 <0.5 Guanylate cyclase activator
    Alverine Citrate 0.495 Phosphodiesterase inhibitor
    Tannic Acid 0.543 CXL12 inhibitor
    IMD-0354 0.661 3.1 IKKA inhibitor
    Arbidol 0.592 6.0 Immunostimulator
    Andrographis 0.523 12.9 Immunostimulator
    Pyrvinium Pamoate 0.800 3.4 AKT kinase antagonist
    Deguelin 0.418 0.5 AKT kinase antagonist
    Dasatinib 0.428 BCR-ABL1/Src inhibitor
    5-Iodotubercidin 0.369 ERK2/ADK/CK1/CK2/IRK inhibitor
    SP 600125 0.365 JNK kinase inhibitor
    PMA 0.800 <0.18 PKC kinase activator
    Cepharanthine 0.652 5.6 PKC kinase inhibitor
    Sangivamycin Hydrate 0.900 <0.2 PKC kinase inhibitor
    PKR inhibitor 0.434 PKR kinase inhibitor
    Okadaic Acid 0.488 Protein phosphatase 1/protein phosphatase
    2A inhibitor
    Sorafenib Tosylate 0.819 3.9 RAF kinase inhibitor
    Sodium Vanadate 0.525 13.0 Tyrosine phosphatase inhibitor
    Latrunculin B 0.621 0.4 Actin polymerization antagonist
    Nocodazole 0.800 0.5 Tubulin destablizer
    Mebendazole 0.500 0.7 Tubulin destablizer
    TN-16 0.800 1.8 Tubulin destablizer
    Fenbendazole 0.500 5.8 Tubulin polymerization inhibitor
    Podofilox 0.800 1.8 Tubulin polymerization inhibitor
    Triclabendazole 0.552 9.2 Tubulin polymerization stabilizer
    Oxibendazole 0.472 Tubulin polymerization stabilizer
    Vinorelbine Tartrate Hydrate 0.472 3.2 Tubulin beta 2 destabilizer
    Thapsigargin 0.700 <0.3 Sarcoplasmic/endoplasmic reticulum
    calcium ATPase inhibitor
    2,6-Divanillyridenecyclohexanone 0.932 6.4 Unknown_aliment, choleretic
    1,5′-Bis(2-Nitrophenoxy)pentane 0.531 13.0 Unknown analog, pentamidine
    (1S,4S)-Desmethyl Sertraline, 0.688 1.3 Unknown analog, sertraline metabolite
    Hydrochloride
    Podophyllum 0.800 <1.6 Unknown, herbal
    Saponin 0.558 4.8 Antiseptic
    Nonoxynol-9 0.535 8.7 Antiseptic
    Domiphen Bromide 0.715 7.5 Unknown, antiseptic
    Sodium Bicarbonate 0.389 Unknown
  • The top 30 agents identified in the initial screen as outlined in Table 6 were re-tested for inhibitory activity in both Vero and HepG2 cells. The results of this analysis are in provided in Table 7. These agents effectively inhibited replication of the Ebola replicon at low to sub micromolar concentrations.
  • TABLE 7
    Max Effect IC50 (μM) Max Effect IC50 (μM)
    Agent Name Vero Vero HepG2 HepG2
    Azacitidine 0.746 8.9 0.899 10
    Clomiphene Citrate 0.964 2.4 0.934 0.76
    Toremifene Citrate 0.935 0.16 0.949 0.026
    Lomerizine 0.899 11 0.918 2.1
    Dihydrochloride
    Bepridil 0.970 5.1 0.912 3.2
    Hydrochloride
    Monohydrate
    Strophanthin 0.742 0.035 0.867 0.021
    Atovaquone 0.739 0.44 0.231 NA
    Mycophenolate 0.438 NA 0.886 0.29
    Mofetil
    Terconazole 0.965 8.2 0.918 2.4
    Simvastatin 0.557 20 0.353 NA
    Thioridazine 0.954 6.2 0.934 2.1
    Hydrochloride
    Prochlorperazine 0.963 5.9 0.947 3.6
    Edisylate
    Sertraline 0.962 3.1 0.950 1.4
    Hydrochloride
    Paroxetine 0.960 7.4 0.920 1.4
    Hydrochloride
    Hemihydrate
    Clomipramine 0.961 11.3 0.897 2.6
    Hydrochloride
    Pimozide 0.968 3.1 0.910 1.7
    Flupentixol 0.959 5.7 0.744 1.6
    Dihydrochloride
    Clemastine 0.958 5.4 0.933 0.65
    Fumarate
    Astemizole 0.965 6.2 0.914 1.4
    Benztropine 0.899 8.0 0.943 2.6
    Mesylate
    Piperacetazine 0.962 12 0.954 3.3
    Fluphenazine 0.968 5.5 0.920 3.0
    Hydrochloride
    Aripiprazole 0.530 20 0.947 3.8
    Maprotiline 0.986 9.6 0.925 2.9
    Hydrochloride
    Dasatinib 0.642 16 0.941 4.2
    Vinorelbine 0.256 NA 0.788 0.86
    Tartrate Hydrate
    Teicoplanin 0.846 7.3 0.770 2.4
    Hycanthone 0.968 11 0.962 6.0
    Quinacrine 0.923 5.7 0.862 1.0
    Dihydrochloride
    Efavirenz 0.639 10 0.860 14
  • Example 2 Identification of Synergistic Combination Therapies Using the Ebola Replicon Assay
  • The screening method described in Example 1 was used to identify combinations of agents that synergistically inhibit viral replication. For each combination of agents, a synergy score was calculated by the formula S=log fX log fY ΣIdata (Idata−ILoewe), summed over all non-single-agent concentration pairs, and where log fX,Y are the natural logarithm of the dilution factors used for each single agent. This effectively calculates a volume between the measured and Loewe additive response surfaces, weighted towards high inhibition and corrected for varying dilution factors. In general, the magnitude of the synergy score indicates the strength of the synergistic interaction. For the Ebola screening described in Example 2, synergy scores >1.0 indicate a synergistic effect of Agent 1 and Agent 2 and synergy scores between 0.3 and 1.0 indicate an additive effect of Agent 1 and Agent 2.
  • Potency shifting was scored using a combination index (CI) (Chou et al., Adv. Enzyme Regul., 22:27-55 (1984)). For a chosen iso-effect level Icut, CII=(CX/ECX)I+(CY/ECY)I, where (CX/ECX)I for a particular data point is the ratio of the X compound's measured concentration to its effective concentration at the chosen inhibition level. The CI is a rough estimate of how much less drug was needed in combination relative to the single agent doses required to achieve the chosen effect level. CI values around 1.0 are dose additive interactions. CI values <1 are synergistic. CI values in the range of 0.5-0.7 are typical for in vitro measurements of current clinical combinations (Greco et al., Pharmacol. Rev., 47:331-85 (1995)). The choice of effect levels for the CI calculations were selected by using an algorithm that finds the observed Idata with the largest Idata−ILoewe, excluding those points with Idata exceeding the lesser single agent's Imax. This exclusion is applied to ensure that the CI reflects the best synergy at levels covered by both single agents. The chosen iso-level is then floored at a 20% inhibition level, beneath which inhibitions often are unreliable, and rounded to the nearest 5%. The synergy scores and CI for the agent pairs identified in our screen are listed in Table 8.
  • TABLE 8
    Synergy
    Agent A Agent B Score CI (Std. Error)
    Lomerizine Vinorelbine 2.42  0.209 (0.007)
    Pimozide Vinorelbine 2.24 0.236 (0.01)
    Aripiprazole Vinorelbine 3.55  0.210 (0.005)
    Aripiprazole Dasatinib 4.08 0.298 (0.05)
    Dasatinib Lomerizine 2.85 0.400 (0.09)
    Aripiprazole Piperacetazine 4.43 0.257 (0.04)
    Pimozide Thioridazine 1.04 0.769 (0.1) 
    Aripiprazole Astemizole 0.34 0.911 (0.02)
    Clemastine Sertraline 1.07 0.578 (0.04)
    Bepridil Clomiphene 0.385  1.05 (0.02)
    Bepridil Toremifene 1.18 0.864 (0.06)
    Clomiphene Sertraline 1.77 0.537 (0.02)
    Toremifene Sertraline 1.11 0.684 (0.05)
    Sertraline Vinorelbine 1.24 0.506 (0.05)
    Aripiprazole Paroxetine 0.994 0.869 (0.03)
  • Example 3 In Vivo Efficacy of Single Agents in a Mouse Model of Ebola Infection
  • A mouse model of Ebola infection developed by Bray et al. (J. Infect. Dis., 179:S248-58 (1999)) was used to show in vivo efficacy of single agents. C57BL/6 mice (n=10 mice per group) were challenged with 100 pfu of a mouse adapted Ebola virus by IP injection. One hour after challenge, the animals were treated with the single agents in Table 9 at the indicated dose and frequency for a total of 10 days. Animals were monitored for survival for a total of 28 days following infection and the mean time to death determined for all cases where there was a sufficient number of non-survivors. In cases where all animals survived, the mean time to death was not determined and is greater than 28 days.
  • TABLE 9
    Dosing Survival
    Compound Study Dose frequency day 28 MTD
    Clomiphene 1 60 mg/kg QID 90% 12.00
    citrate
    Clomiphene 2 60 mg/kg QID 100% 
    citrate
    Clomiphene 3 60 mg/kg QID male 60% 9.83
    citrate
    Clomiphene 3 60 mg/kg QID female 70% 9.00
    citrate
    Toremifene 1 60 mg/kg QID 50% 9.80
    Toremifene 2 60 mg/kg QID 60% 10.25
    Toremifene 2 60 mg/kg SID 60% 8.75
    Toremifene 3 60 mg/kg SID 80% 8.00
    Toremifene 3 60 mg/kg BID 10% 10.22
    Toremifene 3 80 mg/kg SID 60% 8.75
    Bepridil 1 12 mg/kg BID 100% 
    Bepridil + 1 2 12 mg/kg BID 100% 
    BID+
    Bepridil 2 12 mg/kg QID 60% 8.25
    Bepridil 2 12 mg/kg SID 100% 
    Bepridil 2 12 mg/kg BID 100% 
    Bepridil + 1 3 12 mg/kg BID 90% 7.00
    BID+
    Bepridil + 2 3 12 mg/kg BID 30% 7.14
    BID+
    Bepridil + 3 3 12 mg/kg BID 10% 7.00
    BID+
    Bepridil + 4 3 12 mg/kg BID 30% 7.43
    BID+
    Clomipramine 1 45 mg/kg SID 80% 8.83
    Clomipramine 2 45 mg/kg BID 50% 9.40
    Lomerizine 1 22 mg/kg SID 30% 8.14
    Sertraline 1 12 mg/kg BID 20% 8.63
    QID—every other day
    SID—once daily
    BID—twice daily
    “+” Dosing x day after viral infection
    “/” mice compared to female/male control animals
  • Other Embodiments
  • All publications, patent applications, and patents mentioned in this specification are herein incorporated by reference.
  • Various modifications and variations of the described compositions, methods, and kits of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific embodiments, it will be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in the fields of molecular biology, medicine, immunology, pharmacology, virology, or related fields are intended to be within the scope of the invention.

Claims (18)

What is claimed is:
1. A method for treating a patient having a filovirus-mediated disease, said method comprising administering to said patient an agent selected from the agents listed in Table 1 in an amount that is effective to treat said patient.
2. The method of claim 1, wherein said filovirus is Ebola virus or Marburg virus.
3. The method of claim 1, said method further comprising administering a second agent selected from the agents listed in Table 1.
4. The method of claim 3, wherein the first agent is aripiprazole.
5. The method of claim 4, wherein the second agent is piperacetazine.
6. The method of claim 3, wherein said agents are administered within twenty-four hours of each other.
7. The method of claim 3, wherein said agents are administered within 1 hour of each other or substantially simultaneously.
8. The method of claim 1, wherein said agent is administered orally, parenterally, systemically, topically, or inhalationally.
9. The method of any of claims 6-8, wherein said first agent is aripiprazole and said second agent is piperacetazine.
10. A composition comprising two or more agents selected from the group consisting of the agents of Table 1.
11. The composition of claim 10, wherein said two or more agents are present in amounts that, when administered together to a patient with a filovirus-mediated disease, are effective to treat said patient.
12. The composition of claim 11, wherein said filovirus is Ebola virus or Marburg virus.
13. The composition of claim 10, wherein said two or more agents are aripiprazole and piperacetazine.
14. A kit comprising:
(a) an active agent selected from Table 1 or an analog thereof; and
(b) instructions for administering said agent to a patient having a filovirus-mediated disease.
15. The kit of claim 14, wherein said filovirus is Ebola virus or Marburg virus.
16. The kit of claim 14, further comprising a second active agent selected from Table 1 or an analog thereof.
17. The kit of claim 16, wherein said first active agent is aripiprazole and said second active agent is piperacetazine.
18. The kit of claim 17, wherein said first active agent and said second active agent are formulated separately.
US13/928,630 2009-02-20 2013-06-27 Compositions and methods for treatment of filovirus-mediated diseases Abandoned US20130289024A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/928,630 US20130289024A1 (en) 2009-02-20 2013-06-27 Compositions and methods for treatment of filovirus-mediated diseases

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US15427909P 2009-02-20 2009-02-20
US12/710,203 US8475804B2 (en) 2009-02-20 2010-02-22 Compositions and methods for treatment of filovirus-mediated diseases
US13/928,630 US20130289024A1 (en) 2009-02-20 2013-06-27 Compositions and methods for treatment of filovirus-mediated diseases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/710,203 Continuation US8475804B2 (en) 2009-02-20 2010-02-22 Compositions and methods for treatment of filovirus-mediated diseases

Publications (1)

Publication Number Publication Date
US20130289024A1 true US20130289024A1 (en) 2013-10-31

Family

ID=43527613

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/710,203 Expired - Fee Related US8475804B2 (en) 2009-02-20 2010-02-22 Compositions and methods for treatment of filovirus-mediated diseases
US13/928,630 Abandoned US20130289024A1 (en) 2009-02-20 2013-06-27 Compositions and methods for treatment of filovirus-mediated diseases

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/710,203 Expired - Fee Related US8475804B2 (en) 2009-02-20 2010-02-22 Compositions and methods for treatment of filovirus-mediated diseases

Country Status (1)

Country Link
US (2) US8475804B2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015200205A1 (en) 2014-06-24 2015-12-30 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
WO2021242683A3 (en) * 2020-05-27 2022-01-06 University Of Washington Inhibition of arenaviruses by combinations of approved therapeutic drugs
WO2022250971A1 (en) * 2021-05-28 2022-12-01 Arizona Board Of Regents On Behalf Of The University Of Arizona Small molecule inhibitors of autophagy and histone deactylases and uses thereof
US11613517B2 (en) 2018-03-02 2023-03-28 Oregon Health & Science University Amide prodrugs of small molecule nuclear receptor modulators
US11667606B2 (en) 2019-03-01 2023-06-06 Autobahn Therapeutics, Inc. Thyromimetics
US11827596B2 (en) 2018-12-12 2023-11-28 Autobahn Therapeutics, Inc. Thyromimetics

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012118599A1 (en) * 2011-02-28 2012-09-07 Emory University C-abl tyrosine kinase inhibitors useful for inhibiting filovirus replication
US9056284B2 (en) * 2012-08-10 2015-06-16 The United States Of America, As Represented By The Secretary Of The Interior Chlorine resistant amides, polyamides, and membranes made from the same
CN103353530A (en) * 2012-12-12 2013-10-16 河南省农业科学院 Test strip for rapidly detecting maduramicin, and test card
WO2016063269A1 (en) 2014-10-20 2016-04-28 Prendergast Patrick T Use of antagonists to the nuclear steroid receptor alone or in combination as direct antiviral agents to inhibit alphavirus, togaviridae, arenaviridae, filoviridae, bunyaviridae, flaviviridae and rhabdoviridae
CN105616404A (en) * 2014-10-27 2016-06-01 中国医学科学院药物研究所 Application of arbidol in resisting filo virus infection
US10576077B2 (en) 2015-03-23 2020-03-03 Southwest Research Institute Pharmaceutical salt forms of Cepharanthine and Tetrandrine
US20180353568A1 (en) * 2015-04-22 2018-12-13 Sun Yat-Sen University Use of teicoplanin against ebola virus
AU2016301188A1 (en) 2015-08-06 2018-02-15 Chimerix, Inc. Pyrrolopyrimidine nucleosides and analogs thereof useful as antiviral agents
JP2019527207A (en) * 2016-06-28 2019-09-26 アシスタンス ピュブリーク−オピト ド パリAssistance Publique−Hopitaux De Paris Hicanton derivatives and primaquine derivatives for use in the prevention and / or treatment of disorders associated with gammaherpesviruses
US11617753B2 (en) * 2016-11-10 2023-04-04 Oyagen, Inc. Methods of treating and inhibiting Ebola virus infection
EP3381905A1 (en) * 2017-03-30 2018-10-03 Institut National de la Sante et de la Recherche Medicale (INSERM) Novel antiviral compounds
US11111264B2 (en) 2017-09-21 2021-09-07 Chimerix, Inc. Morphic forms of 4-amino-7-(3,4-dihydroxy-5-(hydroxymethyl)tetrahydrofuran-2-yl)-2-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide and uses thereof
CN107789344A (en) * 2017-10-26 2018-03-13 中国食品药品检定研究院 A kind of inhibitor of Ebola virus
WO2019183729A1 (en) * 2018-03-28 2019-10-03 The Governors Of The University Of Alberta Treatment of infectious disease
CN110938599B (en) * 2019-12-30 2020-09-29 马旭艳 Culture method of neural stem cells and application thereof
CN115611927A (en) * 2020-01-06 2023-01-17 刘力 New compound of vinca antitumor drug
WO2021158248A1 (en) 2020-02-04 2021-08-12 Oyagen, Inc. Method for treating coronavirus infections
KR102185945B1 (en) * 2020-04-17 2020-12-03 한국화학연구원 Antiviral composition
WO2022173841A1 (en) * 2021-02-09 2022-08-18 University Of Virginia Patent Foundation Methods of modeling in vivo efficacy of drug combinations for treatment of viral infections
CN114177176B (en) * 2021-12-04 2022-07-12 中国农业科学院兰州兽医研究所 Application of compound cyproheptadine hydrochloride in preparation of drug for preventing or treating African swine fever

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060039890A1 (en) * 2004-08-20 2006-02-23 Renshaw Perry F Treatment of psychological and cognitive disorders using a cholesterol -lowering agent in combination with an antidepressant
US20080161324A1 (en) * 2006-09-14 2008-07-03 Johansen Lisa M Compositions and methods for treatment of viral diseases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060039890A1 (en) * 2004-08-20 2006-02-23 Renshaw Perry F Treatment of psychological and cognitive disorders using a cholesterol -lowering agent in combination with an antidepressant
US20080161324A1 (en) * 2006-09-14 2008-07-03 Johansen Lisa M Compositions and methods for treatment of viral diseases

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015200205A1 (en) 2014-06-24 2015-12-30 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
EP3160475A4 (en) * 2014-06-24 2018-02-21 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US10052342B2 (en) 2014-06-24 2018-08-21 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US10780105B2 (en) 2014-06-24 2020-09-22 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US11590155B2 (en) 2014-06-24 2023-02-28 Janssen Pharmaceutica Nv Substituted nucleosides, nucleotides and analogs thereof
US11613517B2 (en) 2018-03-02 2023-03-28 Oregon Health & Science University Amide prodrugs of small molecule nuclear receptor modulators
US11827596B2 (en) 2018-12-12 2023-11-28 Autobahn Therapeutics, Inc. Thyromimetics
US11667606B2 (en) 2019-03-01 2023-06-06 Autobahn Therapeutics, Inc. Thyromimetics
WO2021242683A3 (en) * 2020-05-27 2022-01-06 University Of Washington Inhibition of arenaviruses by combinations of approved therapeutic drugs
WO2022250971A1 (en) * 2021-05-28 2022-12-01 Arizona Board Of Regents On Behalf Of The University Of Arizona Small molecule inhibitors of autophagy and histone deactylases and uses thereof

Also Published As

Publication number Publication date
US8475804B2 (en) 2013-07-02
US20110028564A1 (en) 2011-02-03

Similar Documents

Publication Publication Date Title
US8475804B2 (en) Compositions and methods for treatment of filovirus-mediated diseases
US20200206188A1 (en) Compositions and methods to improve the therapeutic benefit of indirubin and analogs thereof, including meisoindigo
JP6280546B2 (en) Treating tyrosine kinase inhibitor resistant malignancies in patients with genetic polymorphisms or dysregulation or mutation of AHI1 using dianhydrogalactitol, diacetyldianhydrogalactitol, dibromodulucitol, or analogs or derivatives thereof How to
US20080161324A1 (en) Compositions and methods for treatment of viral diseases
US20100009970A1 (en) Compositions and methods for treatment of viral diseases
US8802702B2 (en) Compounds for reducing drug resistance and uses thereof
US20160158186A1 (en) Use of dianhydrogalactitol and analogs and derivatives thereof to treat recurrent malignant glioma or progressive secondary brain tumor
US20060194769A1 (en) Small molecules that reduce fungal growth
KR20140022750A (en) Compositions and methods to improve the therapeutic benefit of suboptimally administered chemical compounds including substituted hexitols such as dianhydrogalactitol and diacetyldianhydrogalactitol
TW200848063A (en) Methods and compositions for the treatment of neurodegenerative disorders
US20160045502A1 (en) Therapeutic benefit of suboptimally administered chemical compounds
US20110172251A1 (en) Method of treating mental disorders using of D4 and 5-HT2A antagonists, inverse agonists or partial agonists
US7855195B2 (en) Method of treating mental disorders using D4 and 5-HT2A antagonists, inverse agonists or partial agonists
CN105792823B (en) The pharmaceutical composition for treating malignant tumour
PT1621198E (en) Substituted carbostyril derivatives as 5-ht 1a receptor subtype agonists
AU2017208341A1 (en) Compositions and methods to improve the therapeutic benefit of suboptimally administered chemical compounds including substituted hexitols such as dibromodulcitol
EP3061451B1 (en) Kinase inhibitors for the treatment of cancer
EP1547650A1 (en) Use of D4 and 5-HT2A antagonists, inverse agonists or partial agonists
ES2655115T3 (en) Procedures for administration of an EGFR inhibitor
US20230277524A1 (en) Combination therapy for treatment of viral infections
WO2013028866A1 (en) Therapeutic compounds and methods

Legal Events

Date Code Title Description
AS Assignment

Owner name: ZALICUS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JOHANSEN, LISA M.;LEHAR, JOSEPH;HOFFSTROM, BENJAMIN G.;SIGNING DATES FROM 20110325 TO 20110401;REEL/FRAME:031036/0383

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: U.S. ARMY MEDICAL RESEARCH AND MATERIEL COMMAND, M

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:STOSSEL, ANDREW R;REEL/FRAME:035566/0240

Effective date: 20141007