US20020156092A1 - Diazabicyclo[4.3.0]nonanes, and methods of use thereof - Google Patents

Diazabicyclo[4.3.0]nonanes, and methods of use thereof Download PDF

Info

Publication number
US20020156092A1
US20020156092A1 US10/021,277 US2127701A US2002156092A1 US 20020156092 A1 US20020156092 A1 US 20020156092A1 US 2127701 A US2127701 A US 2127701A US 2002156092 A1 US2002156092 A1 US 2002156092A1
Authority
US
United States
Prior art keywords
compound
occurrence
independently
aryl
heteroaryl
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.)
Granted
Application number
US10/021,277
Other versions
US6476046B1 (en
Inventor
Xinhe Wu
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.)
Sunovion Pharmaceuticals Inc
Original Assignee
Sepracor 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 Sepracor Inc filed Critical Sepracor Inc
Priority to US10/021,277 priority Critical patent/US6476046B1/en
Priority to AU2002225790A priority patent/AU2002225790A1/en
Priority to PCT/US2001/044990 priority patent/WO2002046187A1/en
Assigned to SEPRACOR INC. reassignment SEPRACOR INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WU, XINHE
Publication of US20020156092A1 publication Critical patent/US20020156092A1/en
Application granted granted Critical
Publication of US6476046B1 publication Critical patent/US6476046B1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • Pain is an unpleasant sensation varying in severity in a local part of the body or several parts of the body resulting from injury, disease, or emotional disorder. Pain can be classified according to its duration. Acute pain, which lasts less than one month, usually has a readily identifiable cause and signals tissue damage. In addition, acute pain syndromes can be episodic, for example recurrent discomfort from arthritis. Chronic pain can be defined as pain that persists more than one month beyond the usual course of an acute illness or injury, or pain that recurs at intervals over months or years, or pain that is associated with a chronic pathologic process. In contrast to acute pain, chronic pain loses its adaptive biologic function. Depression is common, and abnormal illness behavior often compounds the patient's impairment.
  • the nonsteriodal anti-inflammatories class includes drugs such as aspirin, ibuprofen, diclofenac, acetaminophen, celecoxib, and rofecoxib.
  • the opioid class includes morphine, oxycodone, fentanyl, and pentazocine.
  • Adjuvant analgesics include various antidepressants, anticonvulsants, neuroleptics, and corticosteroids.
  • Opioids are the major class of analgesics used in the management of moderate to severe pain because of their effectiveness, ease of titration, and favorable risk-to-benefit ratio. Opioids produce analgesia by binding to specific receptors both within and outside the CNS. Opioid analgesics are classified as full agonists, partial agonists, or mixed agonist-antagonists, depending on the receptors to which they bind and their intrinsic activities at each receptor.
  • ⁇ -, ⁇ -, and ⁇ -opioid receptors Three subclasses of opioid receptor have been identified in humans, namely the ⁇ -, ⁇ -, and ⁇ -opioid receptors.
  • Analgesia is thought to involve activation of ⁇ and/or ⁇ receptors. Notwithstanding their low selectivity for ⁇ over ⁇ receptors, it is likely that morphine and morphine-like opioid agonists produce analgesia primarily through interaction with ⁇ receptors; selective agonists of ⁇ receptors in humans produce analgesia, because rather than the euphoria associated with morphine and congeners, these compounds often produce dysphoria and psychotomimetic effects. The consequences of activating ⁇ receptors in humans remain unclear.
  • opioids can be very effective in pain management, they do cause several side effects, such as respiratory depression, constipation, physical dependence, tolerance, withdraw. These unwanted effects can severely limit their use.
  • Opioids are known to produce respiratory depression that is proportional to their analgesia. This respiratory depression can be life threatening. This results in a narrow range between the effective dose and a dose that produces respiratory depression. Because of this narrow therapeutic index, patients receiving opioid therapy must be closely monitored for signs of respiratory failure.
  • Opioids can also cause constipation in individuals receiving them. This side effect can be severe and may require prolonged hospitalization, or even surgical intervention.
  • full agonists include morphine, hydromorphone, meperidine, methadone, levorphanol, and fentanyl. These opioids are classified as full agonists because there is not a ceiling to their analgesic efficacy, nor will they reverse or antagonize the effects of other opioids within this class when given simultaneously. Side effects include respiratory depression, constipation, nausea, urinary retention, confusion, and sedation. Morphine is the most commonly used opioid for moderate to severe pain because of its availability in a wide variety of dosage forms, its well-characterized pharmacokinetics and pharmacodynamics, and its relatively low cost.
  • Meperidine may be useful for brief courses (e.g., a few days) to treat acute pain and to manage rigors (shivering) induced by medication, but it generally should be avoided in patients with cancer because of its short duration of action (2.5 to 3.5 hours) and its toxic metabolite, normeperidine. This metabolite accumulates, particularly when renal function is impaired, and causes CNS stimulation, which may lead to dysphoria, agitation, and seizures; meperidine, therefore, should not be used if continued opioid use is anticipated.
  • the known opioids have been very effective in pain management. However, they have restricted use because of several potentially severe side effects. Therefore, there is a current need for pharmaceutical agents that retain the analgesic properties of the known opioid, but that have reduced side effect profiles.
  • One aspect of the present invention relates to a compound represented by A:
  • Z represents NR, NC(O)R, NS(O) 2 R, O, S, SO, or SO 2 ;
  • m is independently for each occurrence 0, 1, 2, 3, or 4;
  • y is 0, 1, or 2;
  • R represents H, alkyl, cycloalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl;
  • R 1 represents H, alkyl, cycloalkyl, aryl, or heteroaryl
  • R when a constituent of Z, and R 1 may be connected through a covalent bond
  • R 4 represents independently for each occurrence H, alkyl, cycloalkyl, alkenyl, aryl, heteroaryl, acyl, or sulfonyl;
  • R 5 and R 6 are selected independently for each occurrence from the group consisting of H, alkyl, CH 2 ZR, aryl, heteroaryl, F, OR, and OC(O)R;
  • a covalent bond may connect R 4 and an instance of R 5 or R 6 ;
  • any geminal or vicinal pairs of R 5 and R 6 may be connected through a covalent bond
  • the stereochemical configuration at any stereocenter of a compound represented by A is R, S, or a mixture of these configurations.
  • a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R.
  • a compound of the present invention is represented by A and the attendant definitions, wherein m is independently for each occurrence 0, 1, or 2.
  • a compound of the present invention is represented by A and the attendant definitions, wherein y is 1.
  • a compound of the present invention is represented by A and the attendant definitions, wherein R 1 represents aryl or heteroaryl.
  • a compound of the present invention is represented by A and the attendant definitions, wherein R 4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein R 4 represents independently for each occurrence aryl. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein R 5 represents independently for each occurrence H. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein R 6 represents independently for each occurrence H.
  • a compound of the present invention is represented by A and the attendant definitions, wherein R 5 represents independently for each occurrence H; and R 6 represents independently for each occurrence H.
  • a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; and m is independently for each occurrence 0, 1, or 2.
  • a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; and y is 1.
  • a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; and y is 1.
  • a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; and R 1 represents aryl or heteroaryl.
  • a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; R 1 represents aryl or heteroaryl; and R 4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl.
  • a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; R 1 represents aryl or heteroaryl; and R 4 represents independently for each occurrence aryl.
  • a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; R 1 represents aryl or heteroaryl; R 4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl; R 1 represents independently for each occurrence H; and R 6 represents independently for each occurrence H.
  • a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; R 1 represents aryl or heteroaryl; R 4 represents independently for each occurrence aryl; R 1 represents independently for each occurrence H; and R 6 represents independently for each occurrence H.
  • a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a single stereoisomer.
  • a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a ligand for a cellular receptor.
  • a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a ligand for a G-protein-coupled receptor.
  • a compound of the present invention is represented by A and the attendant definitions, wherein said compound is an agonist of a G-protein-coupled receptor.
  • a compound of the present invention is represented by A and the attendant definitions, wherein said compound is an antagonist of a G-protein-coupled receptor.
  • a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a ligand for an opioid receptor.
  • a compound of the present invention is represented by A and the attendant definitions, wherein said compound is an agonist of an opioid receptor.
  • a compound of the present invention is represented by A and the attendant definitions, wherein said compound is an antagonist of an opioid receptor.
  • a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a ligand which is selective for a subclass of opioid receptor.
  • a formulation of the present invention comprises a pharmaceutically acceptable excipient selected from the group consisting of cyclodextrins, liposomes, micelle forming agents, and polymeric carriers.
  • Another aspect of the present invention relates to a method of treating pain, drug addiction, or tinnitus in a mammal, comprising the step of administering to a mammal suffering from pain, drug addiction, or tinnitus a therapeutically effective amount of a compound represented by A and the attendant definitions.
  • said mammal is a primate, equine, canine or feline.
  • said mammal is a human.
  • said compound is administered orally.
  • said said compound is administered intravenously.
  • said said compound is administered sublingually.
  • said compound is administered ocularly.
  • Pain is an unpleasant sensation varying in severity in a local part of the body or several parts of the body resulting from injury, disease, or emotional disorder. Pain can be classified according to its duration. Acute pain, which lasts less than one month, usually has a readily identifiable cause (e.g., hip fracture) and signals tissue damage. The associated effect is often anxiety, and the concomitant physiologic findings are those of sympathetic stimulation (e.g., tachycardia, tachypnea, diaphoresis). In addition, acute pain syndromes can be episodic, for example recurrent discomfort from arthritis.
  • Chronic pain can be defined as pain that persists more than one month beyond the usual course of an acute illness or injury, or pain that recurs at intervals over months or years, or pain that is associated with a chronic pathologic process. In contrast to acute pain, chronic pain loses its adaptive biologic function. Depression is common, and abnormal illness behavior often compounds the patient's impairment. Chronic pain can be divided broadly into that which is inferred to be predominantly somatogenic and that which is inferred to be predominantly psychogenic. A similar classification based on inferred pathophysiology designates chronic pain as nociceptive (commensurate with ongoing activation of pain-sensitive nerve fibers), neuropathic (due to aberrant somatosensory processing in afferent neural pathways), or psychogenic.
  • Nociceptive pain can be somatic or visceral. Most chronic pain in the elderly is nociceptive and somatic; arthritis, cancer pain, and myofascial pain are most common. Relief is likely with removal of the peripheral cause (e.g., reducing periarticular inflammation), and analgesic drugs are often effective.
  • a common subtype of neuropathic pain is presumably sustained by mechanisms that involve disturbances in the peripheral nerve or nerve root; neuroma formation after axonal injury and nerve compression are the two major processes.
  • Another subtype of neuropathic pain is related to the reorganization of nociceptive information processing by the CNS; it persists without ongoing activation of pain-sensitive fibers.
  • This type of pain known collectively as the deafferentation syndromes, includes postherpetic neuralgia, central pain (which can result from a lesion at any level of the CNS), phantom limb pain, and others.
  • a third subtype of neuropathic pain can be ameliorated by interruption of sympathetic nerves to the painful area; the prototypic disorder is reflex sympathetic dystrophy.
  • the precise mechanisms involved in these disorders are conjectural, but all can produce an unfamiliar pain, often described as burning and stabbing.
  • this type of pain responds poorly to analgesics.
  • idiopathic pain syndrome that is best described by the generic diagnosis chronic nonmalignant pain syndrome, a term denoting pain and disability disproportionate to an identifiable somatic cause and usually related to a more pervasive set of abnormal illness behaviors. Some of these patients may be labeled by the more formal psychiatric diagnosis of somatoform pain disorder.
  • chronic pain may be part of a medical illness (e.g., cancer or arthritis).
  • a mixture of pathophysiologic mechanisms may be involved; e.g., tumor invasion of nerve and bone may cause neuropathic and somatic nociceptive pains, respectively, and psychological factors may be prominent.
  • the nonsteriodal anti-inflammatories class includes drugs such as aspirin, ibuprofen, diclofenac, acetaminophen, and rofecoxib.
  • the opioid class includes morphine, oxycodone, fentanyl, and pentazocine.
  • Adjuvant analgesics include various antidepressants, anticonvulsants, neuroleptics, and corticosteroids.
  • opioid are usually most efficacious for treating moderate to severe pain. Although opioids can be very effective in pain management, they do cause several side effects, such as respiratory depression, constipation, physical dependence, tolerance, withdraw. These unwanted effects can severely limit their use. Therefore, there is a current need for pharmaceutical agents that retain the analgesic properties of the known opioid, but have reduced side effect profiles for the treatment of pain.
  • Opioids specifically ligands for the ⁇ -opioid receptor
  • Opioids are the major class of analgesics used in the management of moderate to severe pain because of their effectiveness, ease of titration, and favorable risk-to-benefit ratio.
  • the opioids currently available have several unwanted side-effects, such as respiratory depression and constipation.
  • these agents may lead to tolerance and dependence.
  • Research into the development of new, selective ligands for opioid receptors holds the promise of yielding potent analgesics that lack the side effects of morphine and its congeners.
  • Applicants herein disclose novel analgesics, including selective ligands for opioid receptors.
  • new compounds described herein promise to have agonistic, antagonistic, and hybrid effects on opioid and other cellular receptors. Additionally, new compounds reported herein may possess analgesic properties free from respiratory depression and the potential for physical dependence associated with ⁇ -opioid receptor ligands, such as morphine and fentanyl. Moreover, new compounds reported herein may possess properties for the treatment of physical or psychological additions, psychiatric disorders, and neurological pathologies, such as tinnitus.
  • the ⁇ -opioid receptor is a member of a family of cell surface proteins that permit intracellular transduction of extracellular signals.
  • Cell surface proteins provide eukaryotic and prokaryotic cells a means to detect extracellular signals and transduce such signals intracellularly in a manner that ultimately results in a cellular response or a concerted tissue or organ response.
  • Cell surface proteins by intracellularly transmitting information regarding the extracellular environment via specific intracellular pathways induce an appropriate response to a particular stimulus. The response may be immediate and transient, slow and sustained, or some mixture thereof.
  • eukaryotic cells are extraordinarly sensitive to their environment.
  • Extracellular signal molecules such as growth hormones, vasodilators and neurotransmitters exert their effects, at least in part, via interaction with cell surface proteins. For example, some extracellular signal molecules cause changes in transcription of target gene via changes in the levels of secondary messengers, such as cAMP. Other signals, indirectly alter gene expression by activating the expression of genes, such as immediate-early genes that encode regulatory proteins, which in turn activate expression of other genes that encode transcriptional regulatory proteins. For example, neuron gene expression is modulated by numerous extracellular signals, including neurotransmitters and membrane electrical activity.
  • Transsynaptic signals cause rapid responses in neurons that occur over a period of time ranging from milleseconds, such as the opening of ligandgated channels, to seconds and minutes, such as second messenger-mediated events.
  • Genes in neural cells that are responsive to transsynaptic stimulation and membrane electrical activity include genes, called immediate early genes, whose transcription is activated rapidly, within minutes, and transiently (see, e.g., Sheng et al. (1990) Neuron 4: 477-485), and genes whose expression requires protein synthesis and whose expression is induced or altered over the course of hours.
  • Cell surface receptors and ion channels are among the cell surface proteins that respond to extracellular signals and initiate the events that lead to this varied gene expression and response. Ion channels and cell surface-localized receptors are ubiquitous and physiologically important cell surface membrane proteins. They play a central role in regulating intracellular levels of various ions and chemicals, many of which are important for cell viability and function.
  • Cell surface-localized receptors are membrane spanning proteins that bind extracellular signalling molecules or changes in the extracellular environment and transmit the signal via signal transduction pathways to effect a cellular response.
  • Cell surface receptors bind circulating signal polypeptides, such as neurotransmitters, growth factors and hormones, as the initiating step in the induction of numerous intracellular pathways.
  • Receptors are classified on the basis of the particular type of pathway that is induced.
  • HBGF heparin binding growth factor
  • G protein coupled receptors and G proteins include those that bind growth factors and have intrinsic tyrosine kinase activity, such as the heparin binding growth factor (HBGF) receptors, and those that couple to effector proteins through guanine nucleotide binding regulatory proteins, which are referred to as G protein coupled receptors and G proteins, respectively.
  • HBGF heparin binding growth factor
  • G protein transmembrane signaling pathways consist of three proteins: receptors, G proteins and effectors.
  • G proteins which are the intermediaries in transmembrane signaling pathways, are heterodimers and consist of alpha, beta and gamma subunits. Among the members of a family of G proteins the alpha subunits differ. Functions of G proteins are regulated by the cyclic association of GTP with the alpha subunit followed by hydrolysis of GTP to GDP and dissociation of GDP.
  • G protein coupled receptors are a diverse class of receptors that mediate signal transduction by binding to G proteins.
  • Signal transduction is initiated via ligand binding to the cell membrane receptor, which stimulates binding of the receptor to the G protein.
  • the receptor G protein interaction releases GDP, which is specifically bound to the G protein, and permits the binding of GTP, which activates the G protein.
  • Activated G protein dissociates from the receptor and activates the effector protein, which regulates the intracellular levels of specific second messengers. Examples of such effector proteins include adenyl cyclase, guanyl cyclase, phospholipase C, and others.
  • G protein-coupled receptors which are glycoproteins, are known to share certain structural similarities and homologies (see, e-g., Gilman, A. G., Ann. Rev. Biochem.56: 615-649 (1987), Strader, C. D. et al. The FASEB Journal 3: 1825-1832 (1989), Kobilka, B. K., et al. Nature 329:75-79 (1985) and Young et al. Cell 45: 711-719 (1986)).
  • G protein-coupled receptors that have been identified and cloned are the substance P receptor, the angiotensin receptor, the alpha - and beta -adrenergic receptors and the serotonin receptors.
  • G protein-coupled receptors share a conserved structural motif.
  • the general and common structural features of the G protein-coupled receptors are the existence of seven hydrophobic stretches of about 20-25 amino acids each surrounded by eight hydrophilic regions of variable length. It has been postulated that each of the seven hydrophobic regions forms a transmembrane alpha helix and the intervening hydrophilic regions form alternately intracellularly and extracellularly exposed loops.
  • the third cytosolic loop between transmembrane domains five and six is the intracellular domain responsible for the interaction with G proteins.
  • G protein-coupled receptors are known to be inducible. This inducibility was originally described in lower eukaryotes.
  • the cAMP receptor of the cellular slime mold, Dictyostelium is induced during differentiation (Klein et al., Science 241: 1467-1472 (1988).
  • cAMP induces high level expression of its G protein-coupled receptor.
  • This receptor transduces the signal to induce the expression of the other genes involved in chemotaxis, which permits multicellular aggregates to align, organize and form stalks (see, Firtel, R. A., et al. Cell 58: 235-239 (1989) and Devreotes, P., Science 245: 1054-1058 (1989)).
  • CNS central nervous system of an organism.
  • cell surface proteins includes molecules that occur on the surface of cells, interact with the extracellular environment, and transmit or transduce information regarding the environment intracellularly.
  • extracellular signals includes a molecule or a change in the environment that is transduced intracellularly via cell surface proteins that interact, directly or indirectly, with the signal.
  • An extracellular signal is any compound or substance that in some manner specifically alters the activity of a cell surface protein. Examples of such signals include, but are not limited to, molecules such as acetylcholine, growth factors, hormones and other mitogenic substances, such as phorbol mistric acetate (PMA), that bind to cell surface receptors and ion channels and modulate the activity of such receptors and channels.
  • Extracellular signals also includes as yet unidentified substances that modulate the activity of a cell surface protein and thereby affect intracellular finctions and that are potential pharmacological agents that may be used to treat specific diseases by modulating the activity of specific cell surface receptors.
  • ED 50 means the dose of a drug which produces 50% of its maximum response or effect. Alternatively, the dose which produces a predetermined response in 50% of test subjects or preparations.
  • LD 50 means the dose of a drug which is lethal in 50% of test subjects.
  • therapeutic index refers to the therapeutic index of a drug defined as LD 50 /ED 50 .
  • SAR structure-activity relationship
  • agonist refers to a compound that mimics the action of natural transmitter or, when the natural transmitter is not known, causes changes at the receptor complex in the absence of other receptor ligands.
  • inverse agonist and “negative antagonist” refer to compounds that are selective ligands for an inactive form of a cellular receptor which exists as an equilibrating mixture of active and inactive forms.
  • antagonist refers to a compound that binds to a receptor site, but does not cause any physiological changes unless another receptor ligand is present.
  • competitive antagonist refers to a compound that binds to a receptor site; its effects can be overcome by increased concentration of the agonist.
  • partial agonist refers to a compound that binds to a receptor site but does not produce the maximal effect regardless of its concentration.
  • ligand refers to a compound that binds at the receptor site.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are boron, nitrogen, oxygen, phosphorus, sulfur and selenium.
  • electron-withdrawing group is recognized in the art, and denotes the tendency of a substituent to attract valence electrons from neighboring atoms, i.e., the substituent is electronegative with respect to neighboring atoms.
  • Hammett sigma
  • Exemplary electron-withdrawing groups include nitro, acyl, formyl, sulfonyl, trifluoromethyl, cyano, chloride, and the like.
  • Exemplary electron-donating groups include amino, methoxy, and the like.
  • alkyl refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups.
  • a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C 1 -C 30 for straight chain, C 3 -C 30 for branched chain), and more preferably 20 or fewer.
  • preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure.
  • lower alkyl as used herein means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six carbon atoms in its backbone structure. Likewise, “lower alkenyl” and “lower alkynyl” have similar chain lengths. Preferred alkyl groups are lower alkyls. In preferred embodiments, a substituent designated herein as alkyl is a lower alkyl.
  • aralkyl refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).
  • alkenyl and alkynyl refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • aryl as used herein includes 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • aryl groups having heteroatoms in the ring structure may also be referred to as “aryl heterocycles” or “heteroaromatics.”
  • the aromatic ring can be substituted at one or more ring positions with such substituents as described above, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, —CF 3 , —CN, or the like.
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls.
  • heterocyclyl or “heterocyclic group” refer to 3- to 10-membered ring structures, more preferably 3- to 7-membered rings, whose ring structures include one to four heteroatoms. Heterocycles can also be polycycles.
  • Heterocyclyl groups include, for example, azetidine, azepine, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxathiin, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, pheno
  • the heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, —CF 3 , —CN, or the like.
  • substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxy
  • polycyclyl or “polycyclic group” refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are “fused rings”. Rings that are joined through non-adjacent atoms are termed “bridged” rings.
  • Each of the rings of the polycycle can be substituted with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, —CF 3 , —CN, or the like.
  • substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, si
  • carrier refers to an aromatic or non-aromatic ring in which each atom of the ring is carbon.
  • nitro means —NO 2 ;
  • halogen designates —F, —Cl, —Br or —I;
  • sulfhydryl means —SH;
  • hydroxyl means —OH; and
  • sulfonyl means —SO 2 —.
  • amine and “amino” are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that can be represented by the general formula:
  • R 9 , R 10 and R′ 10 each independently represent a group permitted by the rules of valence.
  • acylamino is art-recognized and refers to a moiety that can be represented by the general formula:
  • R 9 is as defined above
  • R′ 11 represents a hydrogen, an alkyl, an alkenyl or —(CH 2 ) m -R 8 , where m and R 8 are as defined above.
  • amido is art recognized as an amino-substituted carbonyl and includes a moiety that can be represented by the general formula:
  • R 9 , R 10 are as defined above.
  • Preferred embodiments of the amide will not include imides which may be unstable.
  • alkylthio refers to an alkyl group, as defined above, having a sulfur radical attached thereto.
  • the “alkylthio” moiety is represented by one of —S-alkyl, —S-alkenyl, —S-alkynyl, and —S—(CH 2 ) m -R 8 , wherein m and R 8 are defined above.
  • Representative alkylthio groups include methylthio, ethyl thio, and the like.
  • carbonyl is art recognized and includes such moieties as can be represented by the general formula:
  • X is a bond or represents an oxygen or a sulfur
  • R 11 represents a hydrogen, an alkyl, an alkenyl, —(CH 2 ) m -R 8 or a pharmaceutically acceptable salt
  • R′ 11 represents a hydrogen, an alkyl, an alkenyl or —(CH 2 ) m -R 8 , where m and R 8 are as defined above.
  • X is an oxygen and R 11 or R′ 11 is not hydrogen
  • the formula represents an “ester”.
  • X is an oxygen
  • R 11 is as defined above, the moiety is referred to herein as a carboxyl group, and particularly when R 11 is a hydrogen, the formula represents a “carboxylic acid”.
  • alkoxyl refers to an alkyl group, as defined above, having an oxygen radical attached thereto.
  • Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like.
  • An “ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as can be represented by one of —O-alkyl, —O-alkenyl, —O-alkynyl, —O-(CH 2 ) m -R 8 , where m and R 8 are described above.
  • R 41 is an electron pair, hydrogen, alkyl, cycloalkyl, or aryl.
  • triflyl, tosyl, mesyl, and nonaflyl are art-recognized and refer to trifluoromethanesulfonyl, p-toluenesulfonyl, methanesulfonyl, and nonafluorobutanesulfonyl groups, respectively.
  • triflate, tosylate, mesylate, and nonaflate are art-recognized and refer to trifluoromethanesulfonate ester, p-toluenesulfonate ester, methanesulfonate ester, and nonafluorobutanesulfonate ester functional groups and molecules that contain said groups, respectively.
  • sulfonyl refers to a moiety that can be represented by the general formula:
  • R 44 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl.
  • sulfoxido refers to a moiety that can be represented by the general formula:
  • R 44 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aralkyl, or aryl.
  • a “selenoalkyl” refers to an alkyl group having a substituted seleno group attached thereto.
  • Exemplary “selenoethers” which may be substituted on the alkyl are selected from one of —Se-alkyl, —Se-alkenyl, —Se-alkynyl, and —Se—(CH 2 ) m -R 7 , m and R 7 being defined above.
  • Analogous substitutions can be made to alkenyl and alkynyl groups to produce, for example, aminoalkenyls, aminoalkynyls, amidoalkenyls, amidoalkynyls, iminoalkenyls, iminoalkynyls, thioalkenyls, thioalkynyls, carbonyl-substituted alkenyls or alkynyls.
  • each expression e.g. alkyl, m, n, etc., when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.
  • substitution or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
  • the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • Illustrative substituents include, for example, those described herein above.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This invention is not intended to be limited in any manner by the permissible substituents of organic compounds.
  • protecting group means temporary substituents which protect a potentially reactive functional group from undesired chemical transformations.
  • protecting groups include esters of carboxylic acids, silyl ethers of alcohols, and acetals and ketals of aldehydes and ketones, respectively.
  • the field of protecting group chemistry has been reviewed (Greene, T. W.; Wuts, P. G. M. Protective Groups in Organic Synthesis, 2 nd ed.; Wiley: New York, 1991).
  • Certain compounds of the present invention may exist in particular geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
  • a particular enantiomer of a compound of the present invention may be prepared by asymmetric synthesis, it may be isolated using chiral chromatography methods, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers.
  • diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.
  • Contemplated equivalents of the compounds described above include compounds which otherwise correspond thereto, and which have the same general properties thereof (e.g., functioning as analgesics), wherein one or more simple variations of substituents are made which do not adversely affect the efficacy of the compound in binding to opioid receptors.
  • the compounds of the present invention may be prepared by the methods illustrated in the general reaction schemes as, for example, described below, or by modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants which are in themselves known, but are not mentioned here.
  • the compounds of the present invention are represented by A:
  • Z represents NR, NC(O)R, NS(O) 2 R, O, S, SO, or SO 2 ;
  • m is independently for each occurrence 0, 1, 2, 3, or 4;
  • y is 0, 1, or 2;
  • R represents H, alkyl, cycloalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl;
  • R 1 represents H, alkyl, cycloalkyl, aryl, or heteroaryl
  • R when a constituent of Z, and R 1 may be connected through a covalent bond
  • R 4 represents independently for each occurrence H, alkyl, cycloalkyl, alkenyl, aryl, heteroaryl, acyl, or sulfonyl;
  • R 5 and R 6 are selected independently for each occurrence from the group consisting of H, alkyl, CH 2 ZR, aryl, heteroaryl, F, OR, and OC(O)R;
  • a covalent bond may connect R 4 and an instance of R 5 or R 6 ;
  • any geminal or vicinal pairs of R 5 and R 6 may be connected through a covalent bond
  • the stereochemical configuration at any stereocenter of a compound represented by A is R, S, or a mixture of these configurations.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein m is independently for each occurrence 0, 1, or 2.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein y is 1.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein R 1 represents aryl or heteroaryl.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein R 4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein R 4 represents independently for each occurrence aryl.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein R 5 represents independently for each occurrence H.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein R 6 represents independently for each occurrence H.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein R 5 represents independently for each occurrence H; and R 6 represents independently for each occurrence H.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; and m is independently for each occurrence 0, 1, or 2.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; and y is 1.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; and y is 1.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; and R 1 represents aryl or heteroaryl.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; R 1 represents aryl or heteroaryl; and R 4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; R 1 represents aryl or heteroaryl; and R 4 represents independently for each occurrence aryl.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; R 1 represents aryl or heteroaryl; R 4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl; R 5 represents independently for each occurrence H; and R 6 represents independently for each occurrence H.
  • the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O) 2 R; m is independently for each occurrence 0, 1, or 2; y is 1; R 1 represents aryl or heteroaryl; R 4 represents independently for each occurrence aryl; R 5 represents independently for each occurrence H; and R 6 represents independently for each occurrence H.
  • certain compounds according to structure A have IC 50 values less than 10 ⁇ M against at least one subclass of opioid receptor, more preferably less than 5 ⁇ M, and most preferably less than 1 ⁇ M.
  • the present invention relates to a compound represented by A and the attendant defintions, wherein said compound is a single stereoisomer.
  • the present invention relates to a formulation, comprising a compound represented by A and the attendant defintions; and a pharmaceutically acceptable excipient.
  • the present invention relates to a method of treating pain, drug addiction, or tinnitus in a mammal, comprising the step of administering to a mammal with pain, drug addiction, or tinnitus an effective amount of a formulation comprising a compound represented by A and the attendant defintions; and a pharmaceutically acceptable excipient.
  • said mammal is a primate, equine, canine or feline.
  • said mammal is a human.
  • said formulation is administered orally.
  • said formulation is administered intravenously.
  • said formulation is administered sublingually.
  • said formulation is administered ocularly.
  • the present invention relates to ligands for G-protein-coupled or opioid receptors, wherein the ligands are represented by any of generalized structures A and the attendant definitions.
  • the ligands of the present invention are antagonists, agonists, partial agonists or inverse agonists of one or more G-protein-coupled or opioid receptor.
  • the ligands of the present invention preferably exert their effect on the receptors at a concentration less than about 10 micromolar, more preferably at a concentration less than about 1 micromolar, and most preferably at a concentration less than 100 nanomolar.
  • the ligands of the present invention bind to multiple families of G-protein-coupled receptors.
  • the ligands of the present invention bind selectively to a single family of G-protein-coupled or opioid receptors. In other embodiments, the ligands of the present invention bind selectively to a subtype of receptor within a family of G-protein-coupled or opioid receptors.
  • the selectivity of a ligand for a specific family or subtype of receptor renders that ligand an effective therapeutic agent for an acute or chronic ailment, disease or malady.
  • the selectivity of a ligand for a specific family or subtype of receptor consists of a binding affinity for that family or subtype of receptor at least a factor of ten greater than its binding affinity for other families or subtypes of G-protein-coupled or opioid receptors.
  • the selectivity of a ligand for a specific family or subtype of receptor consists of a binding affinity for that family or subtype of receptor at least a factor of one hundred greater than its binding affinity for other families or subtypes of G-protein-coupled or opioid receptors. In certain embodiments, the selectivity of a ligand for a specific family or subtype of receptor consists of a binding affinity for that family or subtype of receptor at least a factor of one thousand greater than its binding affinity for other families or subtypes of G-protein-coupled or opioid receptors.
  • the present invention contemplates pharmaceutical formulations (see below) of the ligands of the present invention.
  • the pharmaceutical formulations will comprise ligands of the present invention that effect only a specific family or subtype of G-protein-coupled or opioid receptor, and thereby have a therapeutic effect on an acute or chronic ailment, disease or malady that is at least in part due to biochemical or physiological processes associated with the receptor(s).
  • the pharmaceutical formulations will comprise ligands of the present invention that effect only a subtype of receptor, and thereby have a therapeutic effect on an acute or chronic ailment, disease or malady that is at least in part due to biochemical or physiological processes associated with the specific subtype of receptor.
  • the Background of the Invention teaches examples of acute or chronic ailments, diseases or maladies that are caused or exacerbated by biochemical or physiological processes associated with specific G-protein-coupled or opioid receptors.
  • One of ordinary skill in the art will be able to accumulate, by reference to the scientific literature, a more comprehensive list of acute or chronic ailments, diseases or maladies that are caused or exacerbated by biochemical or physiological processes associated with specific G-protein-coupled or opioid receptors.
  • the present invention contemplates pharmaceutical formulations of ligands of the present invention that will be of medicinal value against the aforementioned acute or chronic ailments, diseases or maladies.
  • Assaying processes are well known in the art in which a reagent is added to a sample, and measurements of the sample and reagent are made to identify sample attributes stimulated by the reagent.
  • one such assay process concerns determining in a chromogenic assay the amount of an enzyme present in a biological sample or solution.
  • Such assays are based on the development of a colored product in the reaction solution. The reaction develops as the enzyme catalyzes the conversion of a colorless chromogenic substrate to a colored product.
  • Another assay useful in the present invention concerns determining the ability of a ligand to bind to a biological receptor utilizing a technique well known in the art referred to as a radioligand binding assay.
  • This assay accurately determines the specific binding of a radioligand to a targeted receptor through the delineation of its total and nonspecific binding components.
  • Total binding is defined as the amount of radioligand that remains following the rapid separation of the radioligand bound in a receptor preparation (cell homogenates or recombinate receptors) from that which is unbound.
  • the nonspecific binding component is defined as the amount of radioligand that remains following separation of the reaction mixture consisting of receptor, radioligand and an excess of unlabeled ligand.
  • radioligand binding assay for ⁇ -opioid receptor, see Wang, J. B. et al. FEBS Letters 1994, 338, 217.
  • Assays useful in the present invention concern determining the activity of receptors the activation of which initiates subsequent intracellular events in which intracellular stores of calcium ions are released for use as a second messenger.
  • Activation of some G-protein-coupled receptors stimulates the formation of inositol triphosphate (IP3, a G-protein-coupled receptor second messenger) through phospholipase C-mediated hydrolysis of phosphatidylinositol, Berridge and Irvine (1984). Nature 312:315-21.
  • IP3 in turn stimulates the release of intracellular calcium ion stores.
  • G-protein-coupled receptor function A change in cytoplasmic calcium ion levels caused by release of calcium ions from intracellular stores is used to determine G-protein-coupled receptor function. This is another type of indirect assay.
  • G-protein-coupled receptors are muscarinic acetylcholine receptors (mAChR), adrenergic receptors, sigma receptors, serotonin receptors, dopamine receptors, angiotensin receptors, adenosine receptors, bradykinin receptors, metabotropic excitatory amino acid receptors and the like.
  • Cells expressing such G-protein-coupled receptors may exhibit increased cytoplasmic calcium levels as a result of contribution from both intracellular stores and via activation of ion channels, in which case it may be desirable although not necessary to conduct such assays in calcium-free buffer, optionally supplemented with a chelating agent such as EGTA, to distinguish fluorescence response resulting from calcium release from internal stores.
  • Another type of indirect assay involves determining the activity of receptors which, when activated, result in a change in the level of intracellular cyclic nucleotides, e.g., cAMP, cGMP. For example, activation of some dopamine, serotonin, metabotropic glutamate receptors and muscarinic acetylcholine receptors results in a decrease in the cAMP or cGMP levels of the cytoplasm.
  • cyclic nucleotide-gated ion channels e.g., rod photoreceptor cell channels and olfactory neuron channels [see, Altenhofen, W. et al. (1991) Proc. Natl. Acad. Sci U.S.A. 88:9868-9872 and Dhallan et al. (1990) Nature 347:184-187] that are permeable to cations upon activation by binding of cAMP or cGMP.
  • a change in cytoplasmic ion levels caused by a change in the amount of cyclic nucleotide activation of photo-receptor or olfactory neuron channels is used to determine function of receptors that cause a change in cAMP or cGMP levels when activated.
  • activation of the receptor results in a decrease in cyclic nucleotide levels, it may be preferable to expose the cells to agents that increase intracellular cyclic nucleotide levels, e.g., forskolin, prior to adding a receptor-activating compound to the cells in the assay.
  • Cell for this type of assay can be made by co-transfection of a host cell with DNA encoding a cyclic nucleotide-gated ion channel and a DNA encoding a receptor (e.g., certain metabotropic glutamate receptors, muscarinic acetylcholine receptors, dopamine receptors, serotonin receptors and the like, which, when activated, causes a change in cyclic nucleotide levels in the cytoplasm.
  • a receptor e.g., certain metabotropic glutamate receptors, muscarinic acetylcholine receptors, dopamine receptors, serotonin receptors and the like, which, when activated, causes a change in cyclic nucleotide levels in the cytoplasm.
  • Any cell expressing a receptor protein which is capable, upon activation, of directly increasing the intracellular concentration of calcium, such as by opening gated calcium channels, or indirectly affecting the concentration of intracellular calcium as by causing initiation of a reaction which utilizes Ca ⁇ 2+> as a second messenger (e.g., G-protein-coupled receptors), may form the basis of an assay.
  • Cells endogenously expressing such receptors or ion channels and cells which may be transfected with a suitable vector encoding one or more such cell surface proteins are known to those of skill in the art or may be identified by those of skill in the art.
  • HEK human embryonic kidney
  • CRL1573 Chinese hamster ovary (CHO) cells (ATCC Nos. CRL9618, CCL61, CRL9096), PC12 cells (ATCC No. CRL1721) and COS-7 cells (ATCC No. CRL1651).
  • Preferred cells for heterologous cell surface protein expression are those that can be readily and efficiently transfected.
  • Preferred cells include HEK 293 cells, such as those described in U.S. Pat. No. 5,024,939.
  • Any compound which is known to activate ion channels or receptors of interest may be used to initiate an assay. Choosing an appropriate ion channel- or receptor-activating reagent depending on the ion channel or receptor of interest is within the skill of the art.
  • Direct depolarization of the cell membrane to determine calcium channel activity may be accomplished by adding a potassium salt solution having a concentration of potassium ions such that the final concentration of potassium ions in the cell-containing well is in the range of about 50-150 mM (e.g., 50 mM KCl).
  • ligands are known which have affinity for and activate such receptors.
  • nicotinic acetyloholine receptors are known to be activated by nicotine or acetylcholine; similarly, muscarinic and acetylcholine receptors may be activated by addition of muscarine or carbamylcholine.
  • Agonist assays may be carried out on cells known to possess ion channels and/or receptors to determine what effect, if any, a compound has on activation or potentiation of ion channels or receptors of interest. Agonist assays also may be carried out using a reagent known to possess ion channel- or receptor-activating capacity to determine whether a cell expresses the respective finctional ion channel or receptor of interest.
  • a functional receptor or ion channel with agonist typically activates a transient reaction; and prolonged exposure to an agonist may desensitize the receptor or ion channel to subsequent activation.
  • assays for determining ion channel or receptor function should be initiated by addition of agonist (i.e., in a reagent solution used to initiate the reaction).
  • the potency of a compound having agonist activity is determined by the detected change in some observable in the cells (typically an increase, although activation of certain receptors causes a decrease) as compared to the level of the observable in either the same cell, or substantially identical cell, which is treated substantially identically except that reagent lacking the agonist (i.e., control) is added to the well.
  • an agonist assay is performed to test whether or not a cell expresses the functional receptor or ion channel of interest
  • known agonist is added to test-cell-containing wells and to wells containing control cells (substantially identical cell that lacks the specific receptors or ion channels) and the levels of observable are compared.
  • cells lacking the ion channel and/or receptor of interest should exhibit substantially no increase in observable in response to the known agonist.
  • a substantially identical cell may be derived from the same cells from which recombinant cells are prepared but which have not been modified by introduction of heterologous DNA. Alternatively, it may be a cell in which the specific receptors or ion channels are removed. Any statistically or otherwise significant difference in the level of observable indicates that the test compound has in some manner altered the activity of the specific receptor or ion channel or that the test cell possesses the specific functional receptor or ion channel.
  • individual wells contain a distinct cell type, or distinct recombinant cell line expressing a homogeneous population of a receptor or ion channel of interest, so that the compound having unidentified activity may be screened to determine whether it possesses modulatory activity with respect to one or more of a variety of functional ion channels or receptors. It is also contemplated that each of the individual wells may contain the same cell type so that multiple compounds (obtained from different reagent sources in the apparatus or contained within different wells) can be screened and compared for modulating activity with respect to one particular receptor or ion channel type.
  • Antagonist assays may be carried out by incubating cells having finctional ion channels and/or receptors in the presence and absence of one or more compounds, added to the solution bathing the cells in the respective wells of the microtiter plate for an amount of time sufficient (to the extent that the compound has affinity for the ion channel and/or receptor of interest) for the compound(s) to bind to the receptors and/or ion channels, then activating the ion channels or receptors by addition of known agonist, and measuring the level of observable in the cells as compared to the level of observable in either the same cell, or substantially identical cell, in the absence of the putative antagonist.
  • the assays are thus useful for rapidly screening compounds to identify those that modulate any receptor or ion channel in a cell.
  • assays can be used to test functional ligand-receptor or ligand-ion channel interactions for cell receptors including ligand-gated ion channels, voltage-gated ion channels, G-protein-coupled receptors and growth factor receptors.
  • assays may encompass measuring a detectable change of a solution as a consequence of a cellular event which allows a compound, capable of differential characteristics, to change its characteristics in response to the cellular event.
  • various assays may be performed. For example, assays for determining the capacity of a compound to induce cell injury or cell death may be carried out by loading the cells with a pH-sensitive fluorescent indicator such as BCECF (Molecular Probes, Inc., Eugene, Oreg. 97402, Catalog #B1150) and measuring cell injury or cell death as a function of changing fluorescence over time.
  • BCECF pH-sensitive fluorescent indicator
  • the function of receptors whose activation results in a change in the cyclic nucleotide levels of the cytoplasm may be directly determined in assays of cells that express such receptors and that have been injected with a fluorescent compound that changes fluorescence upon binding cAMP.
  • the fluorescent compound comprises cAMP-dependent-protein kinase in which the catalytic and regulatory subunits are each labelled with a different fluorescent-dye [Adams et al. (1991) Nature 349:694-697].
  • cAMP binds to the regulatory subunits, the fluorescence emission spectrum changes; this change can be used as an indication of a change in cAMP concentration.
  • the function of certain neurotransmitter transporters which are present at the synaptic cleft at the junction between two neurons may be determined by the development of fluorescence in the cytoplasm of such neurons when conjugates of an amine acid and fluorescent indicator (wherein the fluorescent indicator of the conjugate is an acetoxymethyl ester derivative e.g., 5-(aminoacetamido)fluorescein; Molecular Probes, Catalog #A1363) are transported by the neurotransmitter transporter into the cytoplasm of the cell where the ester group is cleaved by esterase activity and the conjugate becomes fluorescent.
  • the fluorescent indicator of the conjugate is an acetoxymethyl ester derivative e.g., 5-(aminoacetamido)fluorescein; Molecular Probes, Catalog #A1363
  • a reporter gene construct is inserted into an eukaryotic cell to produce a recombinant cell which has present on its surface a cell surface protein of a specific type.
  • the cell surface receptor may be endogenously expressed or it may be expressed from a heterologous gene that has been introduced into the cell.
  • Methods for introducing heterologous DNA into eukaryotic cells are-well known in the art and any such method may be used.
  • DNA encoding various cell surface proteins is known to those of skill in the art or it may be cloned by any method known to those of skill in the art.
  • the recombinant cell is contacted with a test compound and the level of reporter gene expression is measured.
  • the contacting may be effected in any vehicle and the testing may be by any means using any protocols, such as serial dilution, for assessing specific molecular interactions known to those of skill in the art.
  • the level of gene expression is measured.
  • the amount of time to effect such interactions may be empirically determined, such as by running a time course and measuring the level of transcription as a finction of time.
  • the amount of transcription may be measured using any method known to those of skill in the art to be suitable.
  • specific mRNA expression may be detected using Northern blots or specific protein product may be identified by a characteristic stain.
  • the amount of transcription is then compared to the amount of transcription in either the same cell in the absence of the test. compound or it may be compared with the amount of transcription in a substantially identical cell that lacks the specific receptors.
  • a substantially identical cell may be derived from the same cells from which the recombinant cell was prepared but which had not been modified by introduction of heterologous DNA. Alternatively, it may be a cell in which the specific receptors are removed. Any statistically or otherwise significant difference in the amount of transcription indicates that the test compound has in some manner altered the activity of the specific receptor.
  • the assay may be repeated and modified by the introduction of a step in which the recombinant cell is first tested for the ability of a known agonist or activator of the specific receptor to activate transcription if the transcription is induced, the test compound is then assayed for its ability to inhibit, block or otherwise affect the activity of the agonist.
  • the transcription based assay is useful for identifying compounds that interact with any cell surface protein whose activity ultimately alters gene expression.
  • the assays can be used to test functional ligand-receptor or ligand-ion channel interactions for a number of categories of cell surface-localized receptors, including: ligand-gated ion channels and voltage-gated ion channels, and G protein-coupled receptors.
  • Any transfectable cell that can express the desired cell surface protein in a manner such the protein finctions to intracellularly transduce an extracellular signal may be used.
  • the cells may be selected such that they endogenously express the cell surface protein or may be genetically engineered to do so. Many such cells are known to those of skill in the art. Such cells include, but are not limited to Ltk ⁇ > cells, PC12 cells and COS-7 cells.
  • any cell surface protein that is known to those of skill in the art or that may be identified by those of skill in the art may used in the assay.
  • the cell surface protein may endogenously expressed on the selected cell or it may be expressed from cloned DNA.
  • Exemplary cell surface proteins include, but are not limited to, cell surface receptors and ion channels.
  • Cell surface receptors include, but are not limited to, muscarinic receptors (e.g., human M2 (GenBank accession #M16404); rat M3 (GenBank accession #M16407); human M4 (GenBank accession #M16405); human M5 (Bonner et al.
  • Neuronal nicotinic acetylcholine receptors e.g., the alpha 2, alpha 3 and beta 2 subtypes disclosed in U.S. Ser. No. 504,455 (filed Apr. 3, 1990), hereby expressly incorporated by reference herein in its entirety
  • the rat alpha 2 subunit Wanga et al. (1988) Science 240:330-334
  • the rat alpha 3 subunit Boulter et al. (1986) Nature 319:368-374
  • the rat alpha 4 subunit Goldman et al. (1987) cell 48:965-973
  • the rat alpha 5 subunit Boulter et al.
  • adrenergic receptors e.g., human beta 1 (Frielle et al. (1987) Proc. Natl. Acad. Sci. 84.:7920-7924); human alpha 2 (Kobilka et al. (1987) Science 238:650-656); hamster beta 2 (Dixon et al. (1986) Nature 321:75-79); and the like); dopamine receptors (e.g., human D2 (Stormann et al. (1990) Molec. Pharm.37:1-6); rat (Bunzow et al.
  • NGF receptors e.g., human NGF receptors (Johnson et al. (1986) Cell 47:545-554); and the like); serotonin receptors (e.g., human 5HT1a (Kobilka et al. (1987) Nature 329:75-79); rat 5HT2 (Julius et al. (1990) PNAS 87:928- 932); rat 5HT1c (Julius et al. (1988) Science 241:558-564); and the like).
  • NGF receptors e.g., human NGF receptors (Johnson et al. (1986) Cell 47:545-554); and the like
  • serotonin receptors e.g., human 5HT1a (Kobilka et al. (1987) Nature 329:75-79); rat 5HT2 (Julius et al. (1990) PNAS 87:928- 932); rat 5HT1c (Julius e
  • Reporter gene constructs are prepared by operatively linking a reporter gene with at least one transcriptional regulatory element. If only one transcriptional regulatory element is included it must be a regulatable promoter, At least one of the selected transcriptional regulatory elements must be indirectly or directly regulated by the activity of the selected cell-surface receptor whereby activity of the receptor can be monitored via transcription of the reporter genes.
  • the construct may contain additional transcriptional regulatory elements, such as a FIRE sequence, or other sequence, that is not necessarily regulated by the cell surface protein, but is selected for its ability to reduce background level transcription or to amplify the transduced signal and to thereby increase the sensitivity and reliability of the assay.
  • additional transcriptional regulatory elements such as a FIRE sequence, or other sequence, that is not necessarily regulated by the cell surface protein, but is selected for its ability to reduce background level transcription or to amplify the transduced signal and to thereby increase the sensitivity and reliability of the assay.
  • a reporter gene includes any gene that expresses a detectable gene product, which may be RNA or protein. Preferred reporter genes are those that are readily detectable. The reporter gene may also be included in the construct in the form of a fusion gene with a gene that includes desired transcriptional regulatory sequences or exhibits other desirable properties.
  • reporter genes include, but are not limited to CAT (chloramphenicol acetyl transferase) (Alton and Vapnek (1979), Nature 282: 864-869) luciferase, and other enzyme detection systems, such as beta-galactosidase; firefly luciferase (deWet et al. (1987), Mol. Cell. Biol. 7:725-737); bacterial luciferase (Engebrecht and Silverman (1984), PNAS 1: 4154-4158; Baldwin et al. (1984), Biochemistry 23: 3663-3667); alkaline phosphatase (Toh et al. (1989) Eur. J. Biochem. 182: 231-238, Hall et al. (1983) J. Mol. Appl. Gen. 2: 101).
  • CAT chloramphenicol acetyl transferase
  • Transcriptional control elements include, but are not limited to, promoters, enhancers, and repressor and activator binding sites, Suitable transcriptional regulatory elements may be derived from the transcriptional regulatory regions of genes whose expression is rapidly induced, generally within minutes, of contact between the cell surface protein and the effector protein that modulates the activity of the cell surface protein. Examples of such genes include, but are not limited to, the immediate early genes (see, Sheng et al. (1990) Neuron 4: 477-485), such as c-fos, Immediate early genes are genes that are rapidly induced upon binding of a ligand to a cell surface protein.
  • the transcriptional control elements that are preferred for use in the gene constructs include transcriptional control elements from immediate early genes, elements derived from other genes that exhibit some or all of the characteristics of the immediate early genes, or synthetic elements that are constructed such that genes in operative linkage therewith exhibit such characteristics.
  • the characteristics of preferred genes from which the transcriptional control elements are derived include, but are not limited to, low or undetectable expression in quiescent cells, rapid induction at the transcriptional level within minutes of extracellular simulation, induction that is transient and independent of new protein synthesis, subsequent shut-off of transcription requires new protein synthesis, and mRNAs transcribed from these genes have a short half-life. It is not necessary for all of these properties to be present.
  • the “tail flick” test can be performed by applying a noxious thermal stimulus to the rat's tail and determining the time until the nociceptive tail flick occurs. Analgesia is demonstrated by an increase in time to occurrence of a tail flick response.
  • the “hot plate” test is similarly performed, except that the noxious thermal stimulus is applied to the rat's paws.
  • An experimental procedure, well known in the art, useful in the present invention to assess the ability of compounds to cause respiratory depression is to monitor blood gases.
  • This method employees measuring the partial pressures of oxygen and carbon dioxide in blood samples taken from animals following compound administration. A decrease in the partial pressures of oxygen and an increase in the partial pressure of carbon dioxide may be indicative of respiratory depression.
  • Tolerance can be defmed as a condition characterized by unresponsiveness or decreased responsiveness following prolonged or multiple exposure to a compound compared to the responsiveness demonstrated upon initial exposure.
  • test compounds to cause physical dependence were accessed by giving animals escalating doses of test compounds for five days. After the final dose the animals were given naloxone, an opioid antagonist and observed for behavioral signs of dependence, such as vertical jumping.
  • the present invention provides pharmaceutically acceptable compositions which comprise a therapeutically-effective amount of one or more of the compounds described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents.
  • the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarect
  • terapéuticaally-effective amount means that amount of a compound, material, or composition comprising a compound of the present invention which is effective for producing some desired therapeutic effect in at least a sub-population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment.
  • phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically-acceptable carrier means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
  • a pharmaceutically-acceptable material such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydrox
  • certain embodiments of the present compounds may contain a basic functional group, such as amino or alkylamino, and are, thus, capable of forming pharmaceutically-acceptable salts with pharmaceutically-acceptable acids.
  • pharmaceutically-acceptable salts refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds of the present invention. These salts can be prepared in situ in the administration vehicle or the dosage form manufacturing process, or by separately reacting a purified compound of the invention in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed during subsequent purification.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like.
  • sulfate bisulfate
  • phosphate nitrate
  • acetate valerate
  • oleate palmitate
  • stearate laurate
  • benzoate lactate
  • phosphate tosylate
  • citrate maleate
  • fumarate succinate
  • tartrate napthylate
  • mesylate glucoheptonate
  • lactobionate lactobionate
  • laurylsulphonate salts and the like See, for example,
  • the pharmaceutically acceptable salts of the subject compounds include the conventional nontoxic salts or quaternary ammonium salts of the compounds, e.g., from non-toxic organic or inorganic acids.
  • such conventional nontoxic salts include those derived from inorganic acids such as hydrochloride, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, palmitic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicyclic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isothionic, and the like.
  • the compounds of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically-acceptable salts with pharmaceutically-acceptable bases.
  • pharmaceutically-acceptable salts in these instances refers to the relatively non-toxic, inorganic and organic base addition salts of compounds of the present invention. These salts can likewise be prepared in situ in the administration vehicle or the dosage form manufacturing process, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with ammonia, or with a pharmaceutically-acceptable organic primary, secondary or tertiary amine.
  • a suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with ammonia, or with a pharmaceutically-acceptable organic primary, secondary or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. (See, for example, Berge et al., supra)
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), le
  • Formulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
  • a formulation of the present invention comprises an excipient selected from the group consisting of cyclodextrins, liposomes, micelle forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and polyanhydrides; and a compound of the present invention.
  • an aforementioned formulation renders orally bioavailable a compound of the present invention.
  • Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient.
  • a compound of the present invention may also be administered as a bolus, electuary or paste.
  • the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-shelled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharnaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be formulated for rapid release, e.g., freeze-dried.
  • compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions which can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • Suspensions in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
  • suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
  • Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body.
  • dosage forms can be made by dissolving or dispersing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.
  • Ophthalmic formulations are also contemplated as being within the scope of this invention.
  • compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
  • biodegradable polymers such as polylactide-polyglycolide.
  • Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
  • the compounds of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • the preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given in forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administrations are preferred.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticulare, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • systemic administration means the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracistemally and topically, as by powders, ointments or drops, including buccally and sublingually.
  • the compounds of the present invention which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.
  • compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Generally, intravenous, intracerebroventricular and subcutaneous doses of the compounds of this invention for a patient, when used for the indicated analgesic effects, will range from about 0.0001 to about 100 mg per kilogram of body weight per day.
  • the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • composition While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation (composition).
  • the compounds according to the invention may be formulated for administration in any convenient way for use in human or veterinary medicine, by analogy with other pharmaceuticals.
  • treatment is intended to encompass also prophylaxis, therapy and cure.
  • the patient receiving this treatment is any animal in need, including primates, in particular humans, and other mammals such as equines, cattle, swine and sheep; and poultry and pets in general.
  • the compound of the invention can be administered as such or in admixtures with pharmaceutically acceptable carriers and can also be administered in conjunction with antimicrobial agents such as penicillins, cephalosporins, aminoglycosides and glycopeptides.
  • Conjunctive therapy thus includes sequential, simultaneous and separate administration of the active compound in a way that the therapeutical effects of the first administered one is not entirely disappeared when the subsequent is administered.
  • the addition of the active compound of the invention to animal feed is preferably accomplished by preparing an appropriate feed premix containing the active compound in an effective amount and incorporating the premix into the complete ration.
  • an intermediate concentrate or feed supplement containing the active ingredient can be blended into the feed.
  • feed premixes and complete rations can be prepared and administered are described in reference books (such as “Applied Animal Nutrition”, W. H. Freedman and CO., San Francisco, U.S.A., 1969 or “Livestock Feeds and Feeding” O and B books, Corvallis, Oreg., U.S.A., 1977).
  • microemulsification technology to improve bioavailability of some lipophilic (water insoluble) pharmaceutical agents.
  • examples include Trimetrine (Dordunoo, S. K., et al., Drug Development and Industrial Pharmacy, 17(12), 1685-1713, 1991 and REV 5901 (Sheen, P. C., et al., J Pharm Sci 80(7), 712-714, 1991).
  • microemulsification provides enhanced bioavailability by preferentially directing absorption to the lymphatic system instead of the circulatory system, which thereby bypasses the liver, and prevents destruction of the compounds in the hepatobiliary circulation.
  • the formulations contain micelles formed from a compound of the present invention and at least one amphiphilic carrier, in which the micelles have an average diameter of less than about 100 nm. More preferred embodiments provide micelles having an average diameter less than about 50 nm, and even more preferred embodiments provide micelles having an average diameter less than about 30 nm, or even less than about 20 nm.
  • amphiphilic carriers While all suitable amphiphilic carriers are contemplated, the presently preferred carriers are generally those that have Generally-Recognized-as-Safe (GRAS) status, and that can both solubilize the compound of the present invention and microemulsify it at a later stage when the solution comes into a contact with a complex water phase (such as one found in human gastro-intestinal tract).
  • GRAS Generally-Recognized-as-Safe
  • amphiphilic ingredients that satisfy these requirements have HLB (hydrophilic to lipophilic balance) values of 2-20, and their structures contain straight chain aliphatic radicals in the range of C-6 to C-20. Examples are polyethylene-glycolized fatty glycerides and polyethylene glycols.
  • Particularly preferred amphiphilic carriers are saturated and monounsaturated polyethyleneglycolyzed fatty acid glycerides, such as those obtained from fully or partially hydrogenated various vegetable oils.
  • oils may advantageously consist of tri-. di- and mono-fatty acid glycerides and di- and mono-polyethyleneglycol esters of the corresponding fatty acids, with a particularly preferred fatty acid composition including capric acid 4-10, capric acid 3-9, lauric acid 40-50, myristic acid 14-24, palmitic acid 4-14 and stearic acid 5-15%.
  • amphiphilic carriers includes partially esterified sorbitan and/or sorbitol, with saturated or mono-unsaturated fatty acids (SPAN-series) or corresponding ethoxylated analogs (TWEEN-series).
  • SPAN-series saturated or mono-unsaturated fatty acids
  • TWEEN-series corresponding ethoxylated analogs
  • amphiphilic carriers are particularly contemplated, including Gelucire-series, Labrafil, Labrasol, or Lauroglycol (all manufactured and distributed by Gattefosse Corporation, Saint Priest, France), PEG-mono-oleate, PEG-di- oleate, PEG-mono-laurate and di-laurate, Lecithin, Polysorbate 80, etc (produced and distributed by a number of companies in USA and worldwide).
  • Hydrophilic polymers suitable for use in the present invention are those which are readily water-soluble, can be covalently attached to a vesicle-forning lipid, and which are tolerated in vivo without toxic effects (i.e., are biocompatible).
  • Suitable polymers include polyethylene glycol (PEG), polylactic (also termed polylactide), polyglycolic acid (also termed polyglycolide), a polylactic-polyglycolic acid copolymer, and polyvinyl alcohol.
  • PEG polyethylene glycol
  • polylactic also termed polylactide
  • polyglycolic acid also termed polyglycolide
  • a polylactic-polyglycolic acid copolymer a polyvinyl alcohol.
  • Preferred polymers are those having a molecular weight of from about 100 or 120 daltons up to about 5,000 or 10,000 daltons, and more preferably from about 300 daltons to about 5,000 daltons.
  • the polymer is polyethyleneglycol having a molecular weight of from about 100 to about 5,000 daltons, and more preferably having a molecular weight of from about 300 to about 5,000 daltons.
  • the polymer is polyethyleneglycol of 750 daltons (PEG(750)).
  • Polymers may also be defined by the number of monomers therein; a preferred embodiment of the present invention utilizes polymers of at least about three monomers, such PEG polymers consisting of three monomers (approximately 150 daltons).
  • hydrophilic polymers which may be suitable for use in the present invention include polyvinylpyrrolidone, polymethoxazoline, polyethyloxazoline, polyhydroxypropyl methacrylamide, polymethacrylamide, polydimethylacrylamide, and derivatized celluloses such as hydroxymethylcellulose or hydroxyethylcellulose.
  • a formulation of the present invention comprises a biocompatible polymer selected from the group consisting of polyamides, polycarbonates, polyalkylenes, polymers of acrylic and methacrylic esters, polyvinyl polymers, polyglycolides, polysiloxanes, polyurethanes and co-polymers thereof, celluloses, polypropylene, polyethylenes, polystyrene, polymers of lactic acid and glycolic acid, polyanhydrides, poly(ortho)esters, poly(butic acid), poly(valeric acid), poly(lactide-co-caprolactone), polysaccharides, proteins, polyhyaluronic acids, polycyanoacrylates, and blends, mixtures, or copolymers thereof.
  • a biocompatible polymer selected from the group consisting of polyamides, polycarbonates, polyalkylenes, polymers of acrylic and methacrylic esters, polyvinyl polymers, polyglycolides, polysiloxanes, polyurethanes and
  • Cyclodextrins are cyclic oligosaccharides, consisting of 6, 7 or 8 glucose units, designated by the Greek letter alpha., .beta. or gamma., respectively. Cyclodextrins with fewer than six glucose units are not known to exist. The glucose units are linked by alpha-1,4-glucosidic bonds. As a consequence of the chair conformation of the sugar units, all secondary hydroxyl groups (at C-2, C-3) are located on one side of the ring, while all the primary hydroxyl groups at C-6 are situated on the other side. As a result, the external faces are hydrophilic, making the cyclodextrins water-soluble.
  • the cavities of the cyclodextrins are hydrophobic, since they are lined by the hydrogen of atoms C-3 and C-5, and by ether-like oxygens.
  • These matrices allow complexation with a variety of relatively hydrophobic compounds, including, for instance, steroid compounds such as 17.beta.-estradiol (see, e.g., van Uden et al. Plant Cell Tiss. Org. Cult. 38:1-3-113 (1994)).
  • the complexation takes place by Van der Waals interactions and by hydrogen bond formation.
  • the physico-chemical properties of the cyclodextrin derivatives depend strongly on the kind and the degree of substitution. For example, their solubility in water ranges from insoluble (e.g., triacetyl-beta-cyclodextrin) to 147% soluble (w/v) (G-2-beta-cyclodextrin). In addition, they are soluble in many organic solvents. The properties of the cyclodextrins enable the control over solubility of various formulation components by increasing or decreasing their solubility.
  • cyclodextrin derivatives with anionic properties carboxylic acids, phosphorous acids, phosphinous acids, phosphonic acids, phosphoric acids, thiophosphonic acids, thiosulphinic acids, and sulfonic acids have been appended to the parent cyclodextrin [see, Parmeter (III), supra]. Furthermore, sulfoalkyl ether cyclodextrin derivatives have been described by Stella, et al. (U.S. Pat. No. 5,134,127).
  • Liposomes consist of at least one lipid bilayer membrane enclosing an aqueous internal compartment. Liposomes may be characterized by membrane type and by size. Small unilamellar vesicles (SUVs) have a single membrane and typically range between 0.02 and 0.05 ⁇ m in diameter; large unilamellar vesicles (LUVS) are typically larger than 0.05 ⁇ m Oligolamellar large vesicles and multilamellar vesicles have multiple, usually concentric, membrane layers and are typically larger than 0.1 ⁇ m. Liposomes with several nonconcentric membranes, i.e., several smaller vesicles contained within a larger vesicle, are termed multivesicular vesicles.
  • SUVs Small unilamellar vesicles
  • LUVS large unilamellar vesicles
  • Oligolamellar large vesicles and multilamellar vesicles have multiple, usually concentric
  • One aspect of the present invention relates to formulations comprising liposomes containing a compound of the present invention, where the liposome membrane is formulated to provide a liposome with increased carrying capacity.
  • the compound of the present invention may be contained within, or adsorbed onto, the liposome bilayer of the liposome.
  • the compound of the present invention may be aggregated with a lipid surfactant and carried within the liposome's internal space; in these cases, the liposome membrane is formulated to resist the disruptive effects of the active agent-surfactant aggregate.
  • the lipid bilayer of a liposome contains lipids derivatized with polyethylene glycol (PEG), such that the PEG chains extend from the inner surface of the lipid bilayer into the interior space encapsulated by the liposome, and extend from the exterior of the lipid bilayer into the surrounding environment.
  • PEG polyethylene glycol
  • Active agents contained within liposomes of the present invention are in solubilized form. Aggregates of surfactant and active agent (such as emulsions or micelles containing the active agent of interest) may be entrapped within the interior space of liposomes according to the present invention.
  • a surfactant acts to disperse and solubilize the active agent, and may be selected from any suitable aliphatic, cycloaliphatic or aromatic surfactant, including but not limited to biocompatible lysophosphatidylcholines (LPCs) of varying chain lengths (for example, from about C.sub.14 to about C.sub.20).
  • Polymer-derivatized lipids such as PEG-lipids may also be utilized for micelle formation as they will act to inhibit micelle/membrane fusion, and as the addition of a polymer to surfactant molecules decreases the CMC of the surfactant and aids in micelle formation.
  • Liposomes according to the present invention may be prepared by any of a variety of techniques that are known in the art. See, e.g., U.S. Pat. No. 4,235,871; Published PCT applications WO 96/14057; New RRC, Liposomes: A practical approach, IRL Press, Oxford (1990), pages 33-104; Lasic DD, Liposomes from physics to applications, Elsevier Science Publishers BV, Amsterdam, 1993.
  • liposomes of the present invention may be prepared by diffusing a lipid derivatized with a hydrophilic polymer into preformed liposomes, such as by exposing preformed liposomes to micelles composed of lipid-grafted polymers, at lipid concentrations corresponding to the final mole percent of derivatized lipid which is desired in the liposome.
  • Liposomes containing a hydrophilic polymer can also be formed by homogenization, lipid-field hydration, or extrusion techniques, as are known in the art.
  • the active agent is first dispersed by sonication in a lysophosphatidylcholine or other low CMC surfactant (including polymer grafted lipids) that readily solubilizes hydrophobic molecules.
  • a lysophosphatidylcholine or other low CMC surfactant including polymer grafted lipids
  • the resulting micellar suspension of active agent is then used to rehydrate a dried lipid sample that contains a suitable mole percent of polymer-grafted lipid, or cholesterol.
  • the lipid and active agent suspension is then formed into liposomes using extrusion techniques as are known in the art, and the resulting liposomes separated from the unencapsulated solution by standard column separation.
  • the liposomes are prepared to have substantially homogeneous sizes in a selected size range.
  • One effective sizing method involves extruding an aqueous suspension of the liposomes through a series of polycarbonate membranes having a selected uniform pore size; the pore size of the membrane will correspond roughly with the largest sizes of liposomes produced by extrusion through that membrane. See e.g., U.S. Pat. No. 4,737,323 (Apr. 12, 1988).
  • the release characteristics of a formulation of the present invention depend on the encapsulating material, the concentration of encapsulated drug, and the presence of release modifiers.
  • release can be manipulated to be pH dependent, for example, using a pH sensitive coating that releases only at a low pH, as in the stomach, or a higher pH, as in the intestine.
  • An enteric coating can be used to prevent release from occurring until after passage through the stomach.
  • Multiple coatings or mixtures of cyanamide encapsulated in different materials can be used to obtain an initial release in the stomach, followed by later release in the intestine.
  • Release can also be manipulated by inclusion of salts or pore forming agents, which can increase water uptake or release of drug by diffusion from the capsule.
  • Excipients which modify the solubility of the drug can also be used to control the release rate.
  • Agents which enhance degradation of the matrix or release from the matrix can also be incorporated. They can be added to the drug, added as a separate phase (i.e., as particulates), or can be co-dissolved in the polymer phase depending on the compound. In all cases the amount should be between 0.1 and thirty percent (w/w polymer).
  • Types of degradation enhancers include inorganic salts such as ammonium sulfate and ammonium chloride, organic acids such as citric acid, benzoic acid, and ascorbic acid, inorganic bases such as sodium carbonate, potassium carbonate, calcium carbonate, zinc carbonate, and zinc hydroxide, and organic bases such as protamine sulfate, spermine, choline, ethanolamine, diethanolamine, and triethanolamine and surfactants such as Tween.RTM. and Pluronic.RTM..
  • Pore forming agents which add microstructure to the matrices i.e., water soluble compounds such as inorganic salts and sugars
  • the range should be between one and thirty percent (w/w polymer).
  • Uptake can also be manipulated by altering residence time of the particles in the gut. This can be achieved, for example, by coating the particle with, or selecting as the encapsulating material, a mucosal adhesive polymer.
  • a mucosal adhesive polymer examples include most polymers with free carboxyl groups, such as chitosan, celluloses, and especially polyacrylates (as used herein, polyacrylates refers to polymers including acrylate groups and modified acrylate groups such as cyanoacrylates and methacrylates).
  • a combinatorial library for the purposes of the present invention is a mixture of chemically related compounds which may be screened together for a desired property; said libraries may be in solution or covalently linked to a solid support.
  • the preparation of many related compounds in a single reaction greatly reduces and simplifies the number of screening processes which need to be carried out. Screening for the appropriate biological, pharmaceutical, agrochemical or physical property may be done by conventional methods.
  • the substrate aryl groups used in a combinatorial approach can be diverse in terms of the core aryl moiety, e.g., a variegation in terms of the ring structure, and/or can be varied with respect to the other substituents.
  • a library of substituted diversomers can be synthesized using the subject reactions adapted to the techniques described in the Still et al. PCT publication WO 94/08051, e.g., being linked to a polymer bead by a hydrolyzable or photolyzable group, e.g., located at one of the positions of substrate.
  • the library is synthesized on a set of beads, each bead including a set of tags identifying the particular diversomer on that bead.
  • the beads can be dispersed on the surface of a permeable membrane, and the diversomers released from the beads by lysis of the bead linker.
  • the diversomer from each bead will diffuse across the membrane to an assay zone, where it will interact with an enzyme assay.
  • MS mass spectrometry
  • a compound selected from a combinatorial library can be irradiated in a MALDI step in order to release the diversomer from the matrix, and ionize the diversomer for MS analysis.
  • the libraries of the subject method can take the multipin library format.
  • Geysen and co-workers (Geysen et al. (1984) PNAS 81:3998-4002) introduced a method for generating compound libraries by a parallel synthesis on polyacrylic acid-grated polyethylene pins arrayed in the microtitre plate format.
  • the Geysen technique can be used to synthesize and screen thousands of compounds per week using the multipin method, and the tethered compounds may be reused in many assays.
  • Appropriate linker moieties can also been appended to the pins so that the compounds may be cleaved from the supports after synthesis for assessment of purity and further evaluation (c.f., Bray et al. (1990) Tetrahedron Lett 31:5811-5814; Valerio et al. (1991) Anal Biochem 197:168-177; Bray et al. (1991) Tetrahedron Lett 32:6163-6166).
  • a variegated library of compounds can be provided on a set of beads utilizing the strategy of divide-couple-recombine (see, e.g., Houghten (1985) PNAS 82:5131-5135; and U.S. Pat. Nos. 4,631,211; 5,440,016; 5,480,971).
  • the beads are divided into separate groups equal to the number of different substituents to be added at a particular position in the library, the different substituents coupled in separate reactions, and the beads recombined into one pool for the next iteration.
  • the divide-couple-recombine strategy can be carried out using an analogous approach to the so-called “tea bag” method first developed by Houghten, where compound synthesis occurs on resin sealed inside porous polypropylene bags (Houghten et al. (1986) PNAS 82:5131-5135). Substituents are coupled to the compound-bearing resins by placing the bags in appropriate reaction solutions, while all common steps such as resin washing and deprotection are performed simultaneously in one reaction vessel. At the end of the synthesis, each bag contains a single compound.
  • a scheme of combinatorial synthesis in which the identity of a compound is given by its locations on a synthesis substrate is termed a spatially-addressable synthesis.
  • the combinatorial process is carried out by controlling the addition of a chemical reagent to specific locations on a solid support (Dower et al. (1991) Annu Rep Med Chem 26:271-280; Fodor, S.P.A. (1991) Science 251:767; Pirrung et al. (1992) U.S. Pat. No. 5,143,854; Jacobs et al. (1994) Trends Biotechnol 12:19-26).
  • the spatial resolution of photolithography affords miniaturization. This technique can be carried out through the use protection/deprotection reactions with photolabile protecting groups.
  • a synthesis substrate is prepared for coupling through the covalent attachment of photolabile nitroveratryloxycarbonyl (NVOC) protected amino linkers or other photolabile linkers.
  • Light is used to selectively activate a specified region of the synthesis support for coupling. Removal of the photolabile protecting groups by light (deprotection) results in activation of selected areas. After activation, the first of a set of amino acid analogs, each bearing a photolabile protecting group on the amino terminus, is exposed to the entire surface. Coupling only occurs in regions that were addressed by light in the preceding step.
  • the reaction is stopped, the plates washed, and the substrate is again illuminated through a second mask, activating a different region for reaction with a second protected building block.
  • the pattern of masks and the sequence of reactants define the products and their locations. Since this process utilizes photolithography techniques, the number of compounds that can be synthesized is limited only by the number of synthesis sites that can be addressed with appropriate resolution. The position of each compound is precisely known; hence, its interactions with other molecules can be directly assessed.
  • the subject method utilizes a compound library provided with an encoded tagging system.
  • a recent improvement in the identification of active compounds from combinatorial libraries employs chemical indexing systems using tags that uniquely encode the reaction steps a given bead has undergone and, by inference, the structure it carries.
  • this approach mimics phage display libraries, where activity derives from expressed peptides, but the structures of the active peptides are deduced from the corresponding genomic DNA sequence.
  • the first encoding of synthetic combinatorial libraries employed DNA as the code.
  • a variety of other forms of encoding have been reported, including encoding with sequenceable bio- oligomers (e.g., oligonucleotides and peptides), and binary encoding with additional non- sequenceable tags.
  • a combinatorial library of nominally 7 7 ( 823,543) peptides composed of all combinations of Arg, Gln, Phe, Lys, Val, D-Val and Thr (three-letter amino acid code), each of which was encoded by a specific dinucleotide (TA, TC, CT, AT, TT, CA and AC, respectively), was prepared by a series of alternating rounds of peptide and oligonucleotide synthesis on solid support.
  • the amine linking functionality on the bead was specifically differentiated toward peptide or oligonucleotide synthesis by simultaneously preincubating the beads with reagents that generate protected OH groups for oligonucleotide synthesis and protected NH2 groups for peptide synthesis (here, in a ratio of 1:20).
  • the tags each consisted of 69-mers, 14 units of which carried the code.
  • the bead-bound library was incubated with a fluorescently labeled antibody, and beads containing bound antibody that fluoresced strongly were harvested by fluorescence-activated cell sorting (FACS).
  • FACS fluorescence-activated cell sorting
  • compound libraries can be derived for use in the subject method, where the oligonucleotide sequence of the tag identifies the sequential combinatorial reactions that a particular bead underwent, and therefore provides the identity of the compound on the bead.
  • oligonucleotide tags permits extremelyly sensitive tag analysis. Even so, the method requires careful choice of orthogonal sets of protecting groups required for alternating co-synthesis of the tag and the library member. Furthermore, the chemical lability of the tag, particularly the phosphate and sugar anomeric linkages, may limit the choice of reagents and conditions that can be employed for the synthesis of non- oligomeric libraries. In preferred embodiments, the libraries employ linkers permitting selective detachment of the test compound library member for assay.
  • Peptides have also been employed as tagging molecules for combinatorial libraries.
  • Two exemplary approaches are described in the art, both of which employ branched linkers to solid phase upon which coding and ligand strands are alternately elaborated.
  • orthogonality in synthesis is achieved by employing acid-labile protection for the coding strand and base-labile protection for the compound strand.
  • branched linkers are employed so that the coding unit and the test compound can both be attached to the same functional group on the resin.
  • a cleavable linker can be placed between the branch point and the bead so that cleavage releases a molecule containing both code and the compound (Ptek et al. (1991) Tetrahedron Lett 32:3891- 3894).
  • the cleavable linker can be placed so that the test compound can be selectively separated from the bead, leaving the code behind. This last construct is particularly valuable because it permits screening of the test compound without potential interference of the coding groups. Examples in the art of independent cleavage and sequencing of peptide library members and their corresponding tags has confirmed that the tags can accurately predict the peptide structure.
  • An alternative form of encoding the test compound library employs a set of non-sequencable electrophoric tagging molecules that are used as a binary code (Ohlmeyer et al. (1993) PNAS 90:10922-10926).
  • Exemplary tags are haloaromatic alkyl ethers that are detectable as their trimethylsilyl ethers at less than femtomolar levels by electron capture gas chromatography (ECGC). Variations in the length of the alkyl chain, as well as the nature and position of the aromatic halide substituents, permit the synthesis of at least 40 such tags, which in principle can encode 240 (e.g., upwards of 1012) different molecules.
  • Both libraries were constructed using an orthogonal attachment strategy in which the library member was linked to the solid support by a photolabile linker and the tags were attached through a linker cleavable only by vigorous oxidation. Because the library members can be repetitively partially photoeluted from the solid support, library members can be utilized in multiple assays. Successive photoelution also permits a very high throughput iterative screening strategy: first, multiple beads are placed in 96-well microtiter plates; second, compounds are partially detached and transferred to assay plates; third, a metal binding assay identifies the active wells; fourth, the corresponding beads are rearrayed singly into new microtiter plates; fifth, single active compounds are identified; and sixth, the structures are decoded.

Abstract

One aspect of the present invention relates to diazabicyclo[4.3.0]nonanes. A second aspect of the present invention relates to the use of the diazabicyclo[4.3.0]nonanes as ligands for various cellular receptors, including opiate receptors. An additional aspect of the present invention relates to the use of the diazabicyclo[4.3.0]nonanes as analgesics.

Description

    RELATED APPLICATIONS
  • This application claims the benefit of priority to U.S. Provisional Patent Application serial No. 60/251,108, filed Dec. 4, 2000.[0001]
  • BACKGROUND OF THE INVENTION
  • Pain is an unpleasant sensation varying in severity in a local part of the body or several parts of the body resulting from injury, disease, or emotional disorder. Pain can be classified according to its duration. Acute pain, which lasts less than one month, usually has a readily identifiable cause and signals tissue damage. In addition, acute pain syndromes can be episodic, for example recurrent discomfort from arthritis. Chronic pain can be defined as pain that persists more than one month beyond the usual course of an acute illness or injury, or pain that recurs at intervals over months or years, or pain that is associated with a chronic pathologic process. In contrast to acute pain, chronic pain loses its adaptive biologic function. Depression is common, and abnormal illness behavior often compounds the patient's impairment. [0002]
  • Millions of people suffer from chronic or intractable pain. Persistent pain varies in etiology and presentation. In some cases, symptoms and signs may be evident within a few weeks to a few months after the occurrence of an injury or the onset of disease, e.g. cancer or AIDS. Like many illnesses that at one time were not well understood, pain and its many manifestations may be poorly treated and seriously underestimated. Inappropriately treated pain seriously compromises the patient's quality of life, causing emotional suffering and increasing the risk of lost livelihood and disrupted social integration. Severe chronic pain affects both the pediatric and adult population, and often leads to mood disorders, including depression and, in rare cases, suicide. [0003]
  • In the last several years, health policy-makers, health professionals, regulators, and the public have become increasingly interested in the provision of better pain therapies. This interest is evidenced, in part, by the U.S. Department of Health and Human Services' dissemination of Clinical Practice Guidelines for the management of acute pain and cancer pain. There is currently no nationally accepted consensus for the treatment of chronic pain not due to cancer, yet the economic and social costs of chronic pain are substantial, with estimates ranging in the tens of billions of dollars annually. [0004]
  • Three general classes of drugs are currently available for pain management, nonsteriodal anti-inflammatories, opioids, and adjuvant analgesics. The nonsteriodal anti-inflammatories class includes drugs such as aspirin, ibuprofen, diclofenac, acetaminophen, celecoxib, and rofecoxib. The opioid class includes morphine, oxycodone, fentanyl, and pentazocine. Adjuvant analgesics include various antidepressants, anticonvulsants, neuroleptics, and corticosteroids. [0005]
  • Opioids are the major class of analgesics used in the management of moderate to severe pain because of their effectiveness, ease of titration, and favorable risk-to-benefit ratio. Opioids produce analgesia by binding to specific receptors both within and outside the CNS. Opioid analgesics are classified as full agonists, partial agonists, or mixed agonist-antagonists, depending on the receptors to which they bind and their intrinsic activities at each receptor. [0006]
  • Three subclasses of opioid receptor have been identified in humans, namely the δ-, κ-, and μ-opioid receptors. Analgesia is thought to involve activation of μ and/or κ receptors. Notwithstanding their low selectivity for μ over κ receptors, it is likely that morphine and morphine-like opioid agonists produce analgesia primarily through interaction with μ receptors; selective agonists of κ receptors in humans produce analgesia, because rather than the euphoria associated with morphine and congeners, these compounds often produce dysphoria and psychotomimetic effects. The consequences of activating δ receptors in humans remain unclear. [0007]
  • Although opioids can be very effective in pain management, they do cause several side effects, such as respiratory depression, constipation, physical dependence, tolerance, withdraw. These unwanted effects can severely limit their use. [0008]
  • Opioids are known to produce respiratory depression that is proportional to their analgesia. This respiratory depression can be life threatening. This results in a narrow range between the effective dose and a dose that produces respiratory depression. Because of this narrow therapeutic index, patients receiving opioid therapy must be closely monitored for signs of respiratory failure. [0009]
  • Opioids can also cause constipation in individuals receiving them. This side effect can be severe and may require prolonged hospitalization, or even surgical intervention. [0010]
  • Commonly used full agonists include morphine, hydromorphone, meperidine, methadone, levorphanol, and fentanyl. These opioids are classified as full agonists because there is not a ceiling to their analgesic efficacy, nor will they reverse or antagonize the effects of other opioids within this class when given simultaneously. Side effects include respiratory depression, constipation, nausea, urinary retention, confusion, and sedation. Morphine is the most commonly used opioid for moderate to severe pain because of its availability in a wide variety of dosage forms, its well-characterized pharmacokinetics and pharmacodynamics, and its relatively low cost. Meperidine may be useful for brief courses (e.g., a few days) to treat acute pain and to manage rigors (shivering) induced by medication, but it generally should be avoided in patients with cancer because of its short duration of action (2.5 to 3.5 hours) and its toxic metabolite, normeperidine. This metabolite accumulates, particularly when renal function is impaired, and causes CNS stimulation, which may lead to dysphoria, agitation, and seizures; meperidine, therefore, should not be used if continued opioid use is anticipated. [0011]
  • The development of physical dependence with repeated use is a characteristic feature of the opioid drugs, and the possibility of developing drug dependence is one of the major limitations of their clinical use. Almost all opioid users rapidly develop drug dependency which can lead to apathy, weight loss, loss of sex drive, anxiety, insomnia, and drug cravings. Although physical dependence is common, addiction and abuse are not common in pain patients who are treated appropriately with opioid drugs. [0012]
  • Historically, the development of analgesic tolerance was believed to limit the ability to use opioids efficaciously on a long-term basis for pain management. Tolerance, or decreasing pain relief with the same dosage over time, has not proven to be a prevalent limitation to long-term opioid use. Experience with treating cancer pain has shown that what initially appears to be tolerance is usually progression of the disease. Furthermore, for most opioids, there does not appear to be an arbitrary upper dosage limit, as was once thought. [0013]
  • Cessation of opioid administration may result in a withdrawal syndrome. Symptoms of withdrawal are often the opposite of the effects achieved by the drug; withdrawal from morphine, however, results in complex symptoms that may seem unrelated to its effects. Misunderstanding of addiction and mislabeling of patients as addicts result in unnecessary withholding of opioid medications. Addiction is a compulsive disorder in which an individual becomes preoccupied with obtaining and using a substance, the continued use of which results in a decreased quality of life. Studies indicate that the de novo development of addiction is low when opioids are used for the relief of pain. Furthermore, even opioid addicts can benefit from the carefully supervised, judicious use of opioids for the treatment of pain due to cancer, surgery, or recurrent painful illnesses such as sickle cell disease. [0014]
  • The known opioids have been very effective in pain management. However, they have restricted use because of several potentially severe side effects. Therefore, there is a current need for pharmaceutical agents that retain the analgesic properties of the known opioid, but that have reduced side effect profiles. [0015]
  • SUMMARY OF THE INVENTION
  • One aspect of the present invention relates to a compound represented by A: [0016]
    Figure US20020156092A1-20021024-C00001
  • wherein [0017]
  • Z represents NR, NC(O)R, NS(O)[0018] 2R, O, S, SO, or SO2;
  • m is independently for each occurrence 0, 1, 2, 3, or 4; [0019]
  • y is 0, 1, or 2; [0020]
  • R represents H, alkyl, cycloalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl; [0021]
  • R[0022] 1 represents H, alkyl, cycloalkyl, aryl, or heteroaryl;
  • R, when a constituent of Z, and R[0023] 1 may be connected through a covalent bond;
  • R[0024] 4 represents independently for each occurrence H, alkyl, cycloalkyl, alkenyl, aryl, heteroaryl, acyl, or sulfonyl;
  • R[0025] 5 and R6 are selected independently for each occurrence from the group consisting of H, alkyl, CH2ZR, aryl, heteroaryl, F, OR, and OC(O)R;
  • a covalent bond may connect R[0026] 4 and an instance of R5 or R6;
  • any geminal or vicinal pairs of R[0027] 5 and R6 may be connected through a covalent bond; and
  • the stereochemical configuration at any stereocenter of a compound represented by A is R, S, or a mixture of these configurations. [0028]
  • In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0029] 2R. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein m is independently for each occurrence 0, 1, or 2. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein y is 1. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein R1 represents aryl or heteroaryl. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein R4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein R4 represents independently for each occurrence aryl. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein R5 represents independently for each occurrence H. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein R6 represents independently for each occurrence H.
  • In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein R[0030] 5 represents independently for each occurrence H; and R6 represents independently for each occurrence H. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)2R; and m is independently for each occurrence 0, 1, or 2. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)2R; and y is 1. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; and y is 1. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; and R1 represents aryl or heteroaryl. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; and R4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; and R4 represents independently for each occurrence aryl. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; R4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl; R1 represents independently for each occurrence H; and R6 represents independently for each occurrence H. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; R4 represents independently for each occurrence aryl; R1 represents independently for each occurrence H; and R6 represents independently for each occurrence H.
  • In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a single stereoisomer. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a ligand for a cellular receptor. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a ligand for a G-protein-coupled receptor. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein said compound is an agonist of a G-protein-coupled receptor. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein said compound is an antagonist of a G-protein-coupled receptor. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a ligand for an opioid receptor. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein said compound is an agonist of an opioid receptor. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein said compound is an antagonist of an opioid receptor. In certain embodiments, a compound of the present invention is represented by A and the attendant definitions, wherein said compound is a ligand which is selective for a subclass of opioid receptor. [0031]
  • Another aspect of the present invention relates to a formulation, comprising a compound represented by A and the attendant definitions; and a pharmaceutically acceptable excipient. In certain embodiments, a formulation of the present invention comprises a pharmaceutically acceptable excipient selected from the group consisting of cyclodextrins, liposomes, micelle forming agents, and polymeric carriers. [0032]
  • Another aspect of the present invention relates to a method of treating pain, drug addiction, or tinnitus in a mammal, comprising the step of administering to a mammal suffering from pain, drug addiction, or tinnitus a therapeutically effective amount of a compound represented by A and the attendant definitions. In certain embodiments of the method of the present invention, said mammal is a primate, equine, canine or feline. In certain embodiments of the method of the present invention, said mammal is a human. In certain embodiments of the method of the present invention, said compound is administered orally. In certain embodiments of the method of the present invention, said said compound is administered intravenously. In certain embodiments of the method of the present invention, said said compound is administered sublingually. In certain embodiments of the method of the present invention, said compound is administered ocularly. [0033]
  • DETAILED DESCRIPTION OF THE INVENTION
  • Pain is an unpleasant sensation varying in severity in a local part of the body or several parts of the body resulting from injury, disease, or emotional disorder. Pain can be classified according to its duration. Acute pain, which lasts less than one month, usually has a readily identifiable cause (e.g., hip fracture) and signals tissue damage. The associated effect is often anxiety, and the concomitant physiologic findings are those of sympathetic stimulation (e.g., tachycardia, tachypnea, diaphoresis). In addition, acute pain syndromes can be episodic, for example recurrent discomfort from arthritis. [0034]
  • Chronic pain can be defined as pain that persists more than one month beyond the usual course of an acute illness or injury, or pain that recurs at intervals over months or years, or pain that is associated with a chronic pathologic process. In contrast to acute pain, chronic pain loses its adaptive biologic function. Depression is common, and abnormal illness behavior often compounds the patient's impairment. Chronic pain can be divided broadly into that which is inferred to be predominantly somatogenic and that which is inferred to be predominantly psychogenic. A similar classification based on inferred pathophysiology designates chronic pain as nociceptive (commensurate with ongoing activation of pain-sensitive nerve fibers), neuropathic (due to aberrant somatosensory processing in afferent neural pathways), or psychogenic. [0035]
  • Nociceptive pain can be somatic or visceral. Most chronic pain in the elderly is nociceptive and somatic; arthritis, cancer pain, and myofascial pain are most common. Relief is likely with removal of the peripheral cause (e.g., reducing periarticular inflammation), and analgesic drugs are often effective. [0036]
  • A common subtype of neuropathic pain, known collectively as peripheral neuropathic pain, is presumably sustained by mechanisms that involve disturbances in the peripheral nerve or nerve root; neuroma formation after axonal injury and nerve compression are the two major processes. Another subtype of neuropathic pain is related to the reorganization of nociceptive information processing by the CNS; it persists without ongoing activation of pain-sensitive fibers. This type of pain, known collectively as the deafferentation syndromes, includes postherpetic neuralgia, central pain (which can result from a lesion at any level of the CNS), phantom limb pain, and others. A third subtype of neuropathic pain, often called sympathetically maintained pain, can be ameliorated by interruption of sympathetic nerves to the painful area; the prototypic disorder is reflex sympathetic dystrophy. The precise mechanisms involved in these disorders are conjectural, but all can produce an unfamiliar pain, often described as burning and stabbing. Currently, this type of pain responds poorly to analgesics. [0037]
  • Some patients have persistent pain without either nociceptive foci or evidence of a neuropathic mechanism for the pain. Many others have nociceptive lesions that do not sufficiently explain the degree of pain and disability. Psychopathologic processes account for these complaints in some patients. If no evidence for a psychological cause is found, the pain is referred to as idiopathic. Many patients have an idiopathic pain syndrome that is best described by the generic diagnosis chronic nonmalignant pain syndrome, a term denoting pain and disability disproportionate to an identifiable somatic cause and usually related to a more pervasive set of abnormal illness behaviors. Some of these patients may be labeled by the more formal psychiatric diagnosis of somatoform pain disorder. Others have complaints that constitute a specific pain diagnosis, most commonly the failed low back syndrome or atypical facial pain. Still others have significant organic lesions (e.g., lumbar arachnoiditis) but also have a clear psychological contribution associated with excessive disability. Diagnosis may be difficult, but the relative contributions of both organic and psychological components of the pain can be defined. [0038]
  • Another clinically useful classification of chronic pain is broadly syndromic. For example, chronic pain may be part of a medical illness (e.g., cancer or arthritis). A mixture of pathophysiologic mechanisms may be involved; e.g., tumor invasion of nerve and bone may cause neuropathic and somatic nociceptive pains, respectively, and psychological factors may be prominent. [0039]
  • Three general classes of drugs are currently available for pain management, nonsteriodal anti-inflammatories, opioids, and adjuvant analgesics. The nonsteriodal anti-inflammatories class includes drugs such as aspirin, ibuprofen, diclofenac, acetaminophen, and rofecoxib. The opioid class includes morphine, oxycodone, fentanyl, and pentazocine. Adjuvant analgesics include various antidepressants, anticonvulsants, neuroleptics, and corticosteroids. [0040]
  • Of the three classes of pharmaceutical agents used for pain management, opioid are usually most efficacious for treating moderate to severe pain. Although opioids can be very effective in pain management, they do cause several side effects, such as respiratory depression, constipation, physical dependence, tolerance, withdraw. These unwanted effects can severely limit their use. Therefore, there is a current need for pharmaceutical agents that retain the analgesic properties of the known opioid, but have reduced side effect profiles for the treatment of pain. [0041]
  • Opioids, specifically ligands for the μ-opioid receptor, are the major class of analgesics used in the management of moderate to severe pain because of their effectiveness, ease of titration, and favorable risk-to-benefit ratio. Unfortunately, the opioids currently available have several unwanted side-effects, such as respiratory depression and constipation. In addition, these agents may lead to tolerance and dependence. Research into the development of new, selective ligands for opioid receptors holds the promise of yielding potent analgesics that lack the side effects of morphine and its congeners. Applicants herein disclose novel analgesics, including selective ligands for opioid receptors. Individual compounds described herein promise to have agonistic, antagonistic, and hybrid effects on opioid and other cellular receptors. Additionally, new compounds reported herein may possess analgesic properties free from respiratory depression and the potential for physical dependence associated with μ-opioid receptor ligands, such as morphine and fentanyl. Moreover, new compounds reported herein may possess properties for the treatment of physical or psychological additions, psychiatric disorders, and neurological pathologies, such as tinnitus. [0042]
  • The μ-opioid receptor is a member of a family of cell surface proteins that permit intracellular transduction of extracellular signals. Cell surface proteins provide eukaryotic and prokaryotic cells a means to detect extracellular signals and transduce such signals intracellularly in a manner that ultimately results in a cellular response or a concerted tissue or organ response. Cell surface proteins, by intracellularly transmitting information regarding the extracellular environment via specific intracellular pathways induce an appropriate response to a particular stimulus. The response may be immediate and transient, slow and sustained, or some mixture thereof. By virtue of an array of varied membrane surface proteins, eukaryotic cells are exquisitely sensitive to their environment. [0043]
  • Extracellular signal molecules, such as growth hormones, vasodilators and neurotransmitters, exert their effects, at least in part, via interaction with cell surface proteins. For example, some extracellular signal molecules cause changes in transcription of target gene via changes in the levels of secondary messengers, such as cAMP. Other signals, indirectly alter gene expression by activating the expression of genes, such as immediate-early genes that encode regulatory proteins, which in turn activate expression of other genes that encode transcriptional regulatory proteins. For example, neuron gene expression is modulated by numerous extracellular signals, including neurotransmitters and membrane electrical activity. Transsynaptic signals cause rapid responses in neurons that occur over a period of time ranging from milleseconds, such as the opening of ligandgated channels, to seconds and minutes, such as second messenger-mediated events. Genes in neural cells that are responsive to transsynaptic stimulation and membrane electrical activity, include genes, called immediate early genes, whose transcription is activated rapidly, within minutes, and transiently (see, e.g., Sheng et al. (1990) Neuron 4: 477-485), and genes whose expression requires protein synthesis and whose expression is induced or altered over the course of hours. [0044]
  • Cell surface receptors and ion channels are among the cell surface proteins that respond to extracellular signals and initiate the events that lead to this varied gene expression and response. Ion channels and cell surface-localized receptors are ubiquitous and physiologically important cell surface membrane proteins. They play a central role in regulating intracellular levels of various ions and chemicals, many of which are important for cell viability and function. [0045]
  • Cell surface-localized receptors are membrane spanning proteins that bind extracellular signalling molecules or changes in the extracellular environment and transmit the signal via signal transduction pathways to effect a cellular response. Cell surface receptors bind circulating signal polypeptides, such as neurotransmitters, growth factors and hormones, as the initiating step in the induction of numerous intracellular pathways. Receptors are classified on the basis of the particular type of pathway that is induced. Included among these classes of receptors are those that bind growth factors and have intrinsic tyrosine kinase activity, such as the heparin binding growth factor (HBGF) receptors, and those that couple to effector proteins through guanine nucleotide binding regulatory proteins, which are referred to as G protein coupled receptors and G proteins, respectively. [0046]
  • The G protein transmembrane signaling pathways consist of three proteins: receptors, G proteins and effectors. G proteins, which are the intermediaries in transmembrane signaling pathways, are heterodimers and consist of alpha, beta and gamma subunits. Among the members of a family of G proteins the alpha subunits differ. Functions of G proteins are regulated by the cyclic association of GTP with the alpha subunit followed by hydrolysis of GTP to GDP and dissociation of GDP. [0047]
  • G protein coupled receptors are a diverse class of receptors that mediate signal transduction by binding to G proteins. Signal transduction is initiated via ligand binding to the cell membrane receptor, which stimulates binding of the receptor to the G protein. The receptor G protein interaction releases GDP, which is specifically bound to the G protein, and permits the binding of GTP, which activates the G protein. Activated G protein dissociates from the receptor and activates the effector protein, which regulates the intracellular levels of specific second messengers. Examples of such effector proteins include adenyl cyclase, guanyl cyclase, phospholipase C, and others. [0048]
  • G protein-coupled receptors, which are glycoproteins, are known to share certain structural similarities and homologies (see, e-g., Gilman, A. G., Ann. Rev. Biochem.56: 615-649 (1987), Strader, C. D. et al. The FASEB Journal 3: 1825-1832 (1989), Kobilka, B. K., et al. Nature 329:75-79 (1985) and Young et al. Cell 45: 711-719 (1986)). Among the G protein-coupled receptors that have been identified and cloned are the substance P receptor, the angiotensin receptor, the alpha - and beta -adrenergic receptors and the serotonin receptors. G protein-coupled receptors share a conserved structural motif. The general and common structural features of the G protein-coupled receptors are the existence of seven hydrophobic stretches of about 20-25 amino acids each surrounded by eight hydrophilic regions of variable length. It has been postulated that each of the seven hydrophobic regions forms a transmembrane alpha helix and the intervening hydrophilic regions form alternately intracellularly and extracellularly exposed loops. The third cytosolic loop between transmembrane domains five and six is the intracellular domain responsible for the interaction with G proteins. [0049]
  • G protein-coupled receptors are known to be inducible. This inducibility was originally described in lower eukaryotes. For example, the cAMP receptor of the cellular slime mold, Dictyostelium, is induced during differentiation (Klein et al., Science 241: 1467-1472 (1988). During the Dictyostelium discoideum differentiation pathway, cAMP, induces high level expression of its G protein-coupled receptor. This receptor transduces the signal to induce the expression of the other genes involved in chemotaxis, which permits multicellular aggregates to align, organize and form stalks (see, Firtel, R. A., et al. Cell 58: 235-239 (1989) and Devreotes, P., Science 245: 1054-1058 (1989)). [0050]
  • Definitions [0051]
  • For convenience, certain terms employed in the specification, examples, and appended claims are collected here. [0052]
  • The abbreviation “CNS” refers to the central nervous system of an organism. [0053]
  • The term “cell surface proteins” includes molecules that occur on the surface of cells, interact with the extracellular environment, and transmit or transduce information regarding the environment intracellularly. [0054]
  • The term “extracellular signals” includes a molecule or a change in the environment that is transduced intracellularly via cell surface proteins that interact, directly or indirectly, with the signal. An extracellular signal is any compound or substance that in some manner specifically alters the activity of a cell surface protein. Examples of such signals include, but are not limited to, molecules such as acetylcholine, growth factors, hormones and other mitogenic substances, such as phorbol mistric acetate (PMA), that bind to cell surface receptors and ion channels and modulate the activity of such receptors and channels. Extracellular signals also includes as yet unidentified substances that modulate the activity of a cell surface protein and thereby affect intracellular finctions and that are potential pharmacological agents that may be used to treat specific diseases by modulating the activity of specific cell surface receptors. [0055]
  • The term “ED[0056] 50” means the dose of a drug which produces 50% of its maximum response or effect. Alternatively, the dose which produces a predetermined response in 50% of test subjects or preparations.
  • The term “LD[0057] 50” means the dose of a drug which is lethal in 50% of test subjects.
  • The term “therapeutic index” refers to the therapeutic index of a drug defined as LD[0058] 50/ED50.
  • The term “structure-activity relationship (SAR)” refers to the way in which altering the molecular structure of drugs alters their interaction with a receptor, enzyme, etc. [0059]
  • The term “agonist” refers to a compound that mimics the action of natural transmitter or, when the natural transmitter is not known, causes changes at the receptor complex in the absence of other receptor ligands. [0060]
  • The terms “inverse agonist” and “negative antagonist” refer to compounds that are selective ligands for an inactive form of a cellular receptor which exists as an equilibrating mixture of active and inactive forms. [0061]
  • The term “antagonist” refers to a compound that binds to a receptor site, but does not cause any physiological changes unless another receptor ligand is present. [0062]
  • The term “competitive antagonist” refers to a compound that binds to a receptor site; its effects can be overcome by increased concentration of the agonist. [0063]
  • The term “partial agonist” refers to a compound that binds to a receptor site but does not produce the maximal effect regardless of its concentration. [0064]
  • The term “ligand” refers to a compound that binds at the receptor site. [0065]
  • The term “heteroatom” as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are boron, nitrogen, oxygen, phosphorus, sulfur and selenium. [0066]
  • The term “electron-withdrawing group” is recognized in the art, and denotes the tendency of a substituent to attract valence electrons from neighboring atoms, i.e., the substituent is electronegative with respect to neighboring atoms. A quantification of the level of electron-withdrawing capability is given by the Hammett sigma (σ) constant. This well known constant is described in many references, for instance, J. March, [0067] Advanced Organic Chemistry, McGraw Hill Book Company, New York, (1977 edition) pp. 251-259. The Hammett constant values are generally negative for electron donating groups (σ[P]=−0.66 for NH2) and positive for electron withdrawing groups (σ[P]=0.78 for a nitro group), σ[P] indicating para substitution. Exemplary electron-withdrawing groups include nitro, acyl, formyl, sulfonyl, trifluoromethyl, cyano, chloride, and the like. Exemplary electron-donating groups include amino, methoxy, and the like.
  • The term “alkyl” refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. In preferred embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C[0068] 1-C30 for straight chain, C3-C30 for branched chain), and more preferably 20 or fewer. Likewise, preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure.
  • Unless the number of carbons is otherwise specified, “lower alkyl” as used herein means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six carbon atoms in its backbone structure. Likewise, “lower alkenyl” and “lower alkynyl” have similar chain lengths. Preferred alkyl groups are lower alkyls. In preferred embodiments, a substituent designated herein as alkyl is a lower alkyl. [0069]
  • The term “aralkyl”, as used herein, refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group). [0070]
  • The terms “alkenyl” and “alkynyl” refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively. [0071]
  • The term “aryl” as used herein includes 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as “aryl heterocycles” or “heteroaromatics.” The aromatic ring can be substituted at one or more ring positions with such substituents as described above, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, —CF[0072] 3, —CN, or the like. The term “aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls.
  • The terms ortho, meta and para apply to 1,2-, 1,3- and 1,4-disubstituted benzenes, respectively. For example, the names 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous. [0073]
  • The terms “heterocyclyl” or “heterocyclic group” refer to 3- to 10-membered ring structures, more preferably 3- to 7-membered rings, whose ring structures include one to four heteroatoms. Heterocycles can also be polycycles. Heterocyclyl groups include, for example, azetidine, azepine, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxathiin, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine, oxolane, thiolane, oxazole, piperidine, piperazine, morpholine, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like. The heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, —CF[0074] 3, —CN, or the like.
  • The terms “polycyclyl” or “polycyclic group” refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are “fused rings”. Rings that are joined through non-adjacent atoms are termed “bridged” rings. Each of the rings of the polycycle can be substituted with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, —CF[0075] 3, —CN, or the like.
  • The term “carbocycle”, as used herein, refers to an aromatic or non-aromatic ring in which each atom of the ring is carbon. [0076]
  • As used herein, the term “nitro” means —NO[0077] 2; the term “halogen” designates —F, —Cl, —Br or —I; the term “sulfhydryl” means —SH; the term “hydroxyl” means —OH; and the term “sulfonyl” means —SO2—.
  • The terms “amine” and “amino” are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that can be represented by the general formula: [0078]
    Figure US20020156092A1-20021024-C00002
  • wherein R[0079] 9, R10 and R′10 each independently represent a group permitted by the rules of valence.
  • The term “acylamino” is art-recognized and refers to a moiety that can be represented by the general formula: [0080]
    Figure US20020156092A1-20021024-C00003
  • wherein R[0081] 9 is as defined above, and R′11 represents a hydrogen, an alkyl, an alkenyl or —(CH2)m-R8, where m and R8 are as defined above.
  • The term “amido” is art recognized as an amino-substituted carbonyl and includes a moiety that can be represented by the general formula: [0082]
    Figure US20020156092A1-20021024-C00004
  • wherein R[0083] 9, R10 are as defined above. Preferred embodiments of the amide will not include imides which may be unstable.
  • The term “alkylthio” refers to an alkyl group, as defined above, having a sulfur radical attached thereto. In preferred embodiments, the “alkylthio” moiety is represented by one of —S-alkyl, —S-alkenyl, —S-alkynyl, and —S—(CH[0084] 2)m-R8, wherein m and R8 are defined above. Representative alkylthio groups include methylthio, ethyl thio, and the like.
  • The term “carbonyl” is art recognized and includes such moieties as can be represented by the general formula: [0085]
    Figure US20020156092A1-20021024-C00005
  • wherein X is a bond or represents an oxygen or a sulfur, and R[0086] 11 represents a hydrogen, an alkyl, an alkenyl, —(CH2)m-R8 or a pharmaceutically acceptable salt, R′11 represents a hydrogen, an alkyl, an alkenyl or —(CH2)m-R8, where m and R8 are as defined above. Where X is an oxygen and R11 or R′11 is not hydrogen, the formula represents an “ester”. Where X is an oxygen, and R11 is as defined above, the moiety is referred to herein as a carboxyl group, and particularly when R11 is a hydrogen, the formula represents a “carboxylic acid”. Where X is an oxygen, and R′11 is hydrogen, the formula represents a “formate”. In general, where the oxygen atom of the above formula is replaced by sulfur, the formula represents a “thiolcarbonyl” group. Where X is a sulfur and R11 or R′11 is not hydrogen, the formula represents a “thiolester.” Where X is a sulfur and R11 is hydrogen, the formula represents a “thiolcarboxylic acid.” Where X is a sulfur and R11′ is hydrogen, the formula represents a “thiolformate.” On the other hand, where X is a bond, and R11 is not hydrogen, the above formula represents a “ketone” group. Where X is a bond, and R11 is hydrogen, the above formula represents an “aldehyde” group.
  • The terms “alkoxyl” or “alkoxy” as used herein refers to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like. An “ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as can be represented by one of —O-alkyl, —O-alkenyl, —O-alkynyl, —O-(CH[0087] 2)m-R8, where m and R8 are described above.
  • The term “sulfonate” is art recognized and includes a moiety that can be represented by the general formula: [0088]
    Figure US20020156092A1-20021024-C00006
  • in which R[0089] 41 is an electron pair, hydrogen, alkyl, cycloalkyl, or aryl.
  • The terms triflyl, tosyl, mesyl, and nonaflyl are art-recognized and refer to trifluoromethanesulfonyl, p-toluenesulfonyl, methanesulfonyl, and nonafluorobutanesulfonyl groups, respectively. The terms triflate, tosylate, mesylate, and nonaflate are art-recognized and refer to trifluoromethanesulfonate ester, p-toluenesulfonate ester, methanesulfonate ester, and nonafluorobutanesulfonate ester functional groups and molecules that contain said groups, respectively. [0090]
  • The abbreviations Me, Et, Ph, Tf, Nf, Ts, Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, nonafluorobutanesulfonyl, p-toluenesulfonyl and methanesulfonyl, respectively. A more comprehensive list of the abbreviations utilized by organic chemists of ordinary skill in the art appears in the first issue of each volume of the [0091] Journal of Organic Chemistry; this list is typically presented in a table entitled Standard List of Abbreviations. The abbreviations contained in said list, and all abbreviations utilized by organic chemists of ordinary skill in the art are hereby incorporated by reference.
  • The term “sulfate” is art recognized and includes a moiety that can be represented by the general formula: [0092]
    Figure US20020156092A1-20021024-C00007
  • in which R[0093] 41 is as defined above.
  • The term “sulfonylamino” is art recognized and includes a moiety that can be represented by the general formula: [0094]
    Figure US20020156092A1-20021024-C00008
  • The term “sulfamoyl” is art-recognized and includes a moiety that can be represented by the general formula: [0095]
    Figure US20020156092A1-20021024-C00009
  • The term “sulfonyl”, as used herein, refers to a moiety that can be represented by the general formula: [0096]
    Figure US20020156092A1-20021024-C00010
  • in which R[0097] 44 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl.
  • The term “sulfoxido” as used herein, refers to a moiety that can be represented by the general formula: [0098]
    Figure US20020156092A1-20021024-C00011
  • in which R[0099] 44 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aralkyl, or aryl.
  • A “selenoalkyl” refers to an alkyl group having a substituted seleno group attached thereto. Exemplary “selenoethers” which may be substituted on the alkyl are selected from one of —Se-alkyl, —Se-alkenyl, —Se-alkynyl, and —Se—(CH[0100] 2)m-R7, m and R7 being defined above.
  • Analogous substitutions can be made to alkenyl and alkynyl groups to produce, for example, aminoalkenyls, aminoalkynyls, amidoalkenyls, amidoalkynyls, iminoalkenyls, iminoalkynyls, thioalkenyls, thioalkynyls, carbonyl-substituted alkenyls or alkynyls. [0101]
  • As used herein, the definition of each expression, e.g. alkyl, m, n, etc., when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure. [0102]
  • It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. [0103]
  • As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described herein above. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This invention is not intended to be limited in any manner by the permissible substituents of organic compounds. [0104]
  • The phrase “protecting group” as used herein means temporary substituents which protect a potentially reactive functional group from undesired chemical transformations. Examples of such protecting groups include esters of carboxylic acids, silyl ethers of alcohols, and acetals and ketals of aldehydes and ketones, respectively. The field of protecting group chemistry has been reviewed (Greene, T. W.; Wuts, P. G. M. [0105] Protective Groups in Organic Synthesis, 2nd ed.; Wiley: New York, 1991).
  • Certain compounds of the present invention may exist in particular geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention. [0106]
  • If, for instance, a particular enantiomer of a compound of the present invention is desired, it may be prepared by asymmetric synthesis, it may be isolated using chiral chromatography methods, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers. Alternatively, where the molecule contains a basic functional group, such as amino, or an acidic fimctional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers. [0107]
  • Contemplated equivalents of the compounds described above include compounds which otherwise correspond thereto, and which have the same general properties thereof (e.g., functioning as analgesics), wherein one or more simple variations of substituents are made which do not adversely affect the efficacy of the compound in binding to opioid receptors. In general, the compounds of the present invention may be prepared by the methods illustrated in the general reaction schemes as, for example, described below, or by modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants which are in themselves known, but are not mentioned here. [0108]
  • For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 67th Ed., 1986-87, inside cover. [0109]
  • Compounds of the Invention [0110]
  • In certain embodiments, the compounds of the present invention are represented by A: [0111]
    Figure US20020156092A1-20021024-C00012
  • wherein [0112]
  • Z represents NR, NC(O)R, NS(O)[0113] 2R, O, S, SO, or SO2;
  • m is independently for each occurrence 0, 1, 2, 3, or 4; [0114]
  • y is 0, 1, or 2; [0115]
  • R represents H, alkyl, cycloalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl; [0116]
  • R[0117] 1 represents H, alkyl, cycloalkyl, aryl, or heteroaryl;
  • R, when a constituent of Z, and R[0118] 1 may be connected through a covalent bond;
  • R[0119] 4 represents independently for each occurrence H, alkyl, cycloalkyl, alkenyl, aryl, heteroaryl, acyl, or sulfonyl;
  • R[0120] 5 and R6 are selected independently for each occurrence from the group consisting of H, alkyl, CH2ZR, aryl, heteroaryl, F, OR, and OC(O)R;
  • a covalent bond may connect R[0121] 4 and an instance of R5 or R6;
  • any geminal or vicinal pairs of R[0122] 5 and R6 may be connected through a covalent bond; and
  • the stereochemical configuration at any stereocenter of a compound represented by A is R, S, or a mixture of these configurations. [0123]
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0124] 2R.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein m is independently for each occurrence 0, 1, or 2. [0125]
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein y is 1. [0126]
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein R[0127] 1 represents aryl or heteroaryl.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein R[0128] 4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein R[0129] 4 represents independently for each occurrence aryl.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein R[0130] 5 represents independently for each occurrence H.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein R[0131] 6 represents independently for each occurrence H.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein R[0132] 5 represents independently for each occurrence H; and R6 represents independently for each occurrence H.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0133] 2R; and m is independently for each occurrence 0, 1, or 2.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0134] 2R; and y is 1.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0135] 2R; m is independently for each occurrence 0, 1, or 2; and y is 1.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0136] 2R; m is independently for each occurrence 0, 1, or 2; y is 1; and R1 represents aryl or heteroaryl.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0137] 2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; and R4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0138] 2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; and R4 represents independently for each occurrence aryl.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0139] 2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; R4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl; R5 represents independently for each occurrence H; and R6 represents independently for each occurrence H.
  • In certain embodiments, the compounds of the present invention are represented by A and the attendant definitions, wherein Z is NR, NC(O)R, or NS(O)[0140] 2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; R4 represents independently for each occurrence aryl; R5 represents independently for each occurrence H; and R6 represents independently for each occurrence H.
  • In assays based on opioid receptors from mammalian brain, certain compounds according to structure A have IC[0141] 50 values less than 10 μM against at least one subclass of opioid receptor, more preferably less than 5 μM, and most preferably less than 1 μM.
  • In certain embodiments, the present invention relates to a compound represented by A and the attendant defintions, wherein said compound is a single stereoisomer. [0142]
  • In certain embodiments, the present invention relates to a formulation, comprising a compound represented by A and the attendant defintions; and a pharmaceutically acceptable excipient. [0143]
  • In certain embodiments, the present invention relates to a method of treating pain, drug addiction, or tinnitus in a mammal, comprising the step of administering to a mammal with pain, drug addiction, or tinnitus an effective amount of a formulation comprising a compound represented by A and the attendant defintions; and a pharmaceutically acceptable excipient. In certain embodiments of this method, said mammal is a primate, equine, canine or feline. In certain embodiments of this method, said mammal is a human. In certain embodiments of this method, said formulation is administered orally. In certain embodiments of this method, said formulation is administered intravenously. In certain embodiments of this method, said formulation is administered sublingually. In certain embodiments of this method, said formulation is administered ocularly. [0144]
  • In certain embodiments, the present invention relates to ligands for G-protein-coupled or opioid receptors, wherein the ligands are represented by any of generalized structures A and the attendant definitions. Preferably the ligands of the present invention are antagonists, agonists, partial agonists or inverse agonists of one or more G-protein-coupled or opioid receptor. In any event, the ligands of the present invention preferably exert their effect on the receptors at a concentration less than about 10 micromolar, more preferably at a concentration less than about 1 micromolar, and most preferably at a concentration less than 100 nanomolar. In other embodiments, the ligands of the present invention bind to multiple families of G-protein-coupled receptors. In certain embodiments, the ligands of the present invention bind selectively to a single family of G-protein-coupled or opioid receptors. In other embodiments, the ligands of the present invention bind selectively to a subtype of receptor within a family of G-protein-coupled or opioid receptors. [0145]
  • In certain embodiments, the selectivity of a ligand for a specific family or subtype of receptor renders that ligand an effective therapeutic agent for an acute or chronic ailment, disease or malady. In certain embodiments, the selectivity of a ligand for a specific family or subtype of receptor consists of a binding affinity for that family or subtype of receptor at least a factor of ten greater than its binding affinity for other families or subtypes of G-protein-coupled or opioid receptors. In certain embodiments, the selectivity of a ligand for a specific family or subtype of receptor consists of a binding affinity for that family or subtype of receptor at least a factor of one hundred greater than its binding affinity for other families or subtypes of G-protein-coupled or opioid receptors. In certain embodiments, the selectivity of a ligand for a specific family or subtype of receptor consists of a binding affinity for that family or subtype of receptor at least a factor of one thousand greater than its binding affinity for other families or subtypes of G-protein-coupled or opioid receptors. [0146]
  • The present invention contemplates pharmaceutical formulations (see below) of the ligands of the present invention. In certain embodiments, the pharmaceutical formulations will comprise ligands of the present invention that effect only a specific family or subtype of G-protein-coupled or opioid receptor, and thereby have a therapeutic effect on an acute or chronic ailment, disease or malady that is at least in part due to biochemical or physiological processes associated with the receptor(s). In certain embodiments, the pharmaceutical formulations will comprise ligands of the present invention that effect only a subtype of receptor, and thereby have a therapeutic effect on an acute or chronic ailment, disease or malady that is at least in part due to biochemical or physiological processes associated with the specific subtype of receptor. The Background of the Invention (see above) teaches examples of acute or chronic ailments, diseases or maladies that are caused or exacerbated by biochemical or physiological processes associated with specific G-protein-coupled or opioid receptors. One of ordinary skill in the art will be able to accumulate, by reference to the scientific literature, a more comprehensive list of acute or chronic ailments, diseases or maladies that are caused or exacerbated by biochemical or physiological processes associated with specific G-protein-coupled or opioid receptors. The present invention contemplates pharmaceutical formulations of ligands of the present invention that will be of medicinal value against the aforementioned acute or chronic ailments, diseases or maladies. [0147]
  • Biochemical Activity at Cellular Receptors, and Assays to Detect That Activity [0148]
  • Assaying processes are well known in the art in which a reagent is added to a sample, and measurements of the sample and reagent are made to identify sample attributes stimulated by the reagent. For example, one such assay process concerns determining in a chromogenic assay the amount of an enzyme present in a biological sample or solution. Such assays are based on the development of a colored product in the reaction solution. The reaction develops as the enzyme catalyzes the conversion of a colorless chromogenic substrate to a colored product. [0149]
  • Another assay useful in the present invention concerns determining the ability of a ligand to bind to a biological receptor utilizing a technique well known in the art referred to as a radioligand binding assay. This assay accurately determines the specific binding of a radioligand to a targeted receptor through the delineation of its total and nonspecific binding components. Total binding is defined as the amount of radioligand that remains following the rapid separation of the radioligand bound in a receptor preparation (cell homogenates or recombinate receptors) from that which is unbound. The nonspecific binding component is defined as the amount of radioligand that remains following separation of the reaction mixture consisting of receptor, radioligand and an excess of unlabeled ligand. Under this condition, the only radioligand that remains represents that which is bound to components other that receptor. The specific radioligand bound is determined by subtracting the nonspecific from total radioactivity bound. For a specific example of radioligand binding assay for μ-opioid receptor, see Wang, J. B. et al. [0150] FEBS Letters 1994, 338, 217.
  • Assays useful in the present invention concern determining the activity of receptors the activation of which initiates subsequent intracellular events in which intracellular stores of calcium ions are released for use as a second messenger. Activation of some G-protein-coupled receptors stimulates the formation of inositol triphosphate (IP3, a G-protein-coupled receptor second messenger) through phospholipase C-mediated hydrolysis of phosphatidylinositol, Berridge and Irvine (1984). Nature 312:315-21. IP3 in turn stimulates the release of intracellular calcium ion stores. [0151]
  • A change in cytoplasmic calcium ion levels caused by release of calcium ions from intracellular stores is used to determine G-protein-coupled receptor function. This is another type of indirect assay. Among G-protein-coupled receptors are muscarinic acetylcholine receptors (mAChR), adrenergic receptors, sigma receptors, serotonin receptors, dopamine receptors, angiotensin receptors, adenosine receptors, bradykinin receptors, metabotropic excitatory amino acid receptors and the like. Cells expressing such G-protein-coupled receptors may exhibit increased cytoplasmic calcium levels as a result of contribution from both intracellular stores and via activation of ion channels, in which case it may be desirable although not necessary to conduct such assays in calcium-free buffer, optionally supplemented with a chelating agent such as EGTA, to distinguish fluorescence response resulting from calcium release from internal stores. Another type of indirect assay involves determining the activity of receptors which, when activated, result in a change in the level of intracellular cyclic nucleotides, e.g., cAMP, cGMP. For example, activation of some dopamine, serotonin, metabotropic glutamate receptors and muscarinic acetylcholine receptors results in a decrease in the cAMP or cGMP levels of the cytoplasm. [0152]
  • Furthermore, there are cyclic nucleotide-gated ion channels, e.g., rod photoreceptor cell channels and olfactory neuron channels [see, Altenhofen, W. et al. (1991) Proc. Natl. Acad. Sci U.S.A. 88:9868-9872 and Dhallan et al. (1990) Nature 347:184-187] that are permeable to cations upon activation by binding of cAMP or cGMP. A change in cytoplasmic ion levels caused by a change in the amount of cyclic nucleotide activation of photo-receptor or olfactory neuron channels is used to determine function of receptors that cause a change in cAMP or cGMP levels when activated. In cases where activation of the receptor results in a decrease in cyclic nucleotide levels, it may be preferable to expose the cells to agents that increase intracellular cyclic nucleotide levels, e.g., forskolin, prior to adding a receptor-activating compound to the cells in the assay. Cell for this type of assay can be made by co-transfection of a host cell with DNA encoding a cyclic nucleotide-gated ion channel and a DNA encoding a receptor (e.g., certain metabotropic glutamate receptors, muscarinic acetylcholine receptors, dopamine receptors, serotonin receptors and the like, which, when activated, causes a change in cyclic nucleotide levels in the cytoplasm. [0153]
  • Any cell expressing a receptor protein which is capable, upon activation, of directly increasing the intracellular concentration of calcium, such as by opening gated calcium channels, or indirectly affecting the concentration of intracellular calcium as by causing initiation of a reaction which utilizes Ca<2+> as a second messenger (e.g., G-protein-coupled receptors), may form the basis of an assay. Cells endogenously expressing such receptors or ion channels and cells which may be transfected with a suitable vector encoding one or more such cell surface proteins are known to those of skill in the art or may be identified by those of skill in the art. Although essentially any cell which expresses endogenous ion channel and/or receptor activity may be used, it is preferred to use cells transformed or transfected with heterologous DNAs encoding such ion channels and/or receptors so as to express predominantly a single type of ion channel or receptor. Many cells that may be genetically engineered to express a heterologous cell surface protein are known. Such cells include, but are not limited to, baby hamster kidney (BHK) cells (ATCC No. CCL10), mouse L cells (ATCC No. CCLI.3), DG44 cells [see, Chasin (1986) Cell. Molec. Genet. 12:555] human embryonic kidney (HEK) cells (ATCC No. CRL1573), Chinese hamster ovary (CHO) cells (ATCC Nos. CRL9618, CCL61, CRL9096), PC12 cells (ATCC No. CRL1721) and COS-7 cells (ATCC No. CRL1651). Preferred cells for heterologous cell surface protein expression are those that can be readily and efficiently transfected. Preferred cells include HEK 293 cells, such as those described in U.S. Pat. No. 5,024,939. [0154]
  • Any compound which is known to activate ion channels or receptors of interest may be used to initiate an assay. Choosing an appropriate ion channel- or receptor-activating reagent depending on the ion channel or receptor of interest is within the skill of the art. Direct depolarization of the cell membrane to determine calcium channel activity may be accomplished by adding a potassium salt solution having a concentration of potassium ions such that the final concentration of potassium ions in the cell-containing well is in the range of about 50-150 mM (e.g., 50 mM KCl). With respect to ligand-gated receptors and ligand-gated ion channels, ligands are known which have affinity for and activate such receptors. For example, nicotinic acetyloholine receptors are known to be activated by nicotine or acetylcholine; similarly, muscarinic and acetylcholine receptors may be activated by addition of muscarine or carbamylcholine. [0155]
  • Agonist assays may be carried out on cells known to possess ion channels and/or receptors to determine what effect, if any, a compound has on activation or potentiation of ion channels or receptors of interest. Agonist assays also may be carried out using a reagent known to possess ion channel- or receptor-activating capacity to determine whether a cell expresses the respective finctional ion channel or receptor of interest. [0156]
  • Contacting a functional receptor or ion channel with agonist typically activates a transient reaction; and prolonged exposure to an agonist may desensitize the receptor or ion channel to subsequent activation. Thus, in general, assays for determining ion channel or receptor function should be initiated by addition of agonist (i.e., in a reagent solution used to initiate the reaction). The potency of a compound having agonist activity is determined by the detected change in some observable in the cells (typically an increase, although activation of certain receptors causes a decrease) as compared to the level of the observable in either the same cell, or substantially identical cell, which is treated substantially identically except that reagent lacking the agonist (i.e., control) is added to the well. Where an agonist assay is performed to test whether or not a cell expresses the functional receptor or ion channel of interest, known agonist is added to test-cell-containing wells and to wells containing control cells (substantially identical cell that lacks the specific receptors or ion channels) and the levels of observable are compared. Depending on the assay, cells lacking the ion channel and/or receptor of interest should exhibit substantially no increase in observable in response to the known agonist. A substantially identical cell may be derived from the same cells from which recombinant cells are prepared but which have not been modified by introduction of heterologous DNA. Alternatively, it may be a cell in which the specific receptors or ion channels are removed. Any statistically or otherwise significant difference in the level of observable indicates that the test compound has in some manner altered the activity of the specific receptor or ion channel or that the test cell possesses the specific functional receptor or ion channel. [0157]
  • In an example of drug screening assays for identifying compounds which have the ability to modulate ion channels or receptors of interest, individual wells (or duplicate wells, etc.) contain a distinct cell type, or distinct recombinant cell line expressing a homogeneous population of a receptor or ion channel of interest, so that the compound having unidentified activity may be screened to determine whether it possesses modulatory activity with respect to one or more of a variety of functional ion channels or receptors. It is also contemplated that each of the individual wells may contain the same cell type so that multiple compounds (obtained from different reagent sources in the apparatus or contained within different wells) can be screened and compared for modulating activity with respect to one particular receptor or ion channel type. [0158]
  • Antagonist assays, including drug screening assays, may be carried out by incubating cells having finctional ion channels and/or receptors in the presence and absence of one or more compounds, added to the solution bathing the cells in the respective wells of the microtiter plate for an amount of time sufficient (to the extent that the compound has affinity for the ion channel and/or receptor of interest) for the compound(s) to bind to the receptors and/or ion channels, then activating the ion channels or receptors by addition of known agonist, and measuring the level of observable in the cells as compared to the level of observable in either the same cell, or substantially identical cell, in the absence of the putative antagonist. [0159]
  • The assays are thus useful for rapidly screening compounds to identify those that modulate any receptor or ion channel in a cell. In particular, assays can be used to test functional ligand-receptor or ligand-ion channel interactions for cell receptors including ligand-gated ion channels, voltage-gated ion channels, G-protein-coupled receptors and growth factor receptors. [0160]
  • Those of ordinary skill in the art will recognize that assays may encompass measuring a detectable change of a solution as a consequence of a cellular event which allows a compound, capable of differential characteristics, to change its characteristics in response to the cellular event. By selecting a particular compound which is capable of differential characteristics upon the occurrence of a cellular event, various assays may be performed. For example, assays for determining the capacity of a compound to induce cell injury or cell death may be carried out by loading the cells with a pH-sensitive fluorescent indicator such as BCECF (Molecular Probes, Inc., Eugene, Oreg. 97402, Catalog #B1150) and measuring cell injury or cell death as a function of changing fluorescence over time. [0161]
  • In a further example of useful assays, the function of receptors whose activation results in a change in the cyclic nucleotide levels of the cytoplasm may be directly determined in assays of cells that express such receptors and that have been injected with a fluorescent compound that changes fluorescence upon binding cAMP. The fluorescent compound comprises cAMP-dependent-protein kinase in which the catalytic and regulatory subunits are each labelled with a different fluorescent-dye [Adams et al. (1991) Nature 349:694-697]. When cAMP binds to the regulatory subunits, the fluorescence emission spectrum changes; this change can be used as an indication of a change in cAMP concentration. [0162]
  • The function of certain neurotransmitter transporters which are present at the synaptic cleft at the junction between two neurons may be determined by the development of fluorescence in the cytoplasm of such neurons when conjugates of an amine acid and fluorescent indicator (wherein the fluorescent indicator of the conjugate is an acetoxymethyl ester derivative e.g., 5-(aminoacetamido)fluorescein; Molecular Probes, Catalog #A1363) are transported by the neurotransmitter transporter into the cytoplasm of the cell where the ester group is cleaved by esterase activity and the conjugate becomes fluorescent. [0163]
  • In practicing an assay of this type, a reporter gene construct is inserted into an eukaryotic cell to produce a recombinant cell which has present on its surface a cell surface protein of a specific type. The cell surface receptor may be endogenously expressed or it may be expressed from a heterologous gene that has been introduced into the cell. Methods for introducing heterologous DNA into eukaryotic cells are-well known in the art and any such method may be used. In addition, DNA encoding various cell surface proteins is known to those of skill in the art or it may be cloned by any method known to those of skill in the art. [0164]
  • The recombinant cell is contacted with a test compound and the level of reporter gene expression is measured. The contacting may be effected in any vehicle and the testing may be by any means using any protocols, such as serial dilution, for assessing specific molecular interactions known to those of skill in the art. After contacting the recombinant cell for a sufficient time to effect any interactions, the level of gene expression is measured. The amount of time to effect such interactions may be empirically determined, such as by running a time course and measuring the level of transcription as a finction of time. The amount of transcription may be measured using any method known to those of skill in the art to be suitable. For example, specific mRNA expression may be detected using Northern blots or specific protein product may be identified by a characteristic stain. The amount of transcription is then compared to the amount of transcription in either the same cell in the absence of the test. compound or it may be compared with the amount of transcription in a substantially identical cell that lacks the specific receptors. A substantially identical cell may be derived from the same cells from which the recombinant cell was prepared but which had not been modified by introduction of heterologous DNA. Alternatively, it may be a cell in which the specific receptors are removed. Any statistically or otherwise significant difference in the amount of transcription indicates that the test compound has in some manner altered the activity of the specific receptor. [0165]
  • If the test compound does not appear to enhance, activate or induce the activity of the cell surface protein, the assay may be repeated and modified by the introduction of a step in which the recombinant cell is first tested for the ability of a known agonist or activator of the specific receptor to activate transcription if the transcription is induced, the test compound is then assayed for its ability to inhibit, block or otherwise affect the activity of the agonist. [0166]
  • The transcription based assay is useful for identifying compounds that interact with any cell surface protein whose activity ultimately alters gene expression. In particular, the assays can be used to test functional ligand-receptor or ligand-ion channel interactions for a number of categories of cell surface-localized receptors, including: ligand-gated ion channels and voltage-gated ion channels, and G protein-coupled receptors. [0167]
  • Any transfectable cell that can express the desired cell surface protein in a manner such the protein finctions to intracellularly transduce an extracellular signal may be used. The cells may be selected such that they endogenously express the cell surface protein or may be genetically engineered to do so. Many such cells are known to those of skill in the art. Such cells include, but are not limited to Ltk<−> cells, PC12 cells and COS-7 cells. [0168]
  • The preparation of cells which express a receptor or ion channel and a reporter gene expression construct, and which are useful for testing compounds to assess their activities, is exemplified in the Examples provided herewith by reference to mammalian Ltk<−> and COS-7 cell lines, which express the Type I human muscarinic (HM1) receptor and which are transformed with either a c-fos promoter-CAT reporter gene expression construct or a c-fos promoter-luciferase reporter gene expression construct. [0169]
  • Any cell surface protein that is known to those of skill in the art or that may be identified by those of skill in the art may used in the assay. The cell surface protein may endogenously expressed on the selected cell or it may be expressed from cloned DNA. Exemplary cell surface proteins include, but are not limited to, cell surface receptors and ion channels. Cell surface receptors include, but are not limited to, muscarinic receptors (e.g.,, human M2 (GenBank accession #M16404); rat M3 (GenBank accession #M16407); human M4 (GenBank accession #M16405); human M5 (Bonner et al. (1988) Neuron 1:403-410); and the like); neuronal nicotinic acetylcholine receptors (e.g., the alpha 2, alpha 3 and beta 2 subtypes disclosed in U.S. Ser. No. 504,455 (filed Apr. 3, 1990), hereby expressly incorporated by reference herein in its entirety); the rat alpha 2 subunit (Wada et al. (1988) Science 240:330-334); the rat alpha 3 subunit (Boulter et al. (1986) Nature 319:368-374); the rat alpha 4 subunit (Goldman et al. (1987) cell 48:965-973); the rat alpha 5 subunit (Boulter et al. (1990) J. Biol. Chem. 265:4472-4482); the rat beta 2 subunit (Deneris et al. (1988) Neuron 1:45-54); the rat beta 3 subunit (Deneris et al. (1989) J. Biol. Chem. 264: 6268-6272); the rat beta 4 subunit (Duvoisin et al. (1989) Neuron 3:487-496); combinations of the rat alpha subunits, beta subunits and alpha and beta subunits; GABA receptors (e.g., the bovine alpha 1 and beta 1 subunits (Schofield et al. (1987) Nature 328:221-227); the bovine alpha 2 and alpha 3 subunits (Levitan et al. (1988) Nature 335:76-79); the gamma -subunit (Pritchett et al. (1989) Nature 338:582-585); the beta 2 and beta 3 subunits (Ymer et alo (1989) EMBO J. 8:1665-1670); the delta subunit (Shivers, B.D. (1989) Neuron 3:327-337); and the like); glutamate receptors (e.g., receptor isolated from rat brain (Hollmann et al. (1989) Nature 342:643-648); and the like); adrenergic receptors (e.g., human beta 1 (Frielle et al. (1987) Proc. Natl. Acad. Sci. 84.:7920-7924); human alpha 2 (Kobilka et al. (1987) Science 238:650-656); hamster beta 2 (Dixon et al. (1986) Nature 321:75-79); and the like); dopamine receptors (e.g., human D2 (Stormann et al. (1990) Molec. Pharm.37:1-6); rat (Bunzow et al. (1988) Nature 336:783-787); and the like); NGF receptors (e.g., human NGF receptors (Johnson et al. (1986) Cell 47:545-554); and the like); serotonin receptors (e.g., human 5HT1a (Kobilka et al. (1987) Nature 329:75-79); rat 5HT2 (Julius et al. (1990) PNAS 87:928- 932); rat 5HT1c (Julius et al. (1988) Science 241:558-564); and the like). [0170]
  • Reporter gene constructs are prepared by operatively linking a reporter gene with at least one transcriptional regulatory element. If only one transcriptional regulatory element is included it must be a regulatable promoter, At least one of the selected transcriptional regulatory elements must be indirectly or directly regulated by the activity of the selected cell-surface receptor whereby activity of the receptor can be monitored via transcription of the reporter genes. [0171]
  • The construct may contain additional transcriptional regulatory elements, such as a FIRE sequence, or other sequence, that is not necessarily regulated by the cell surface protein, but is selected for its ability to reduce background level transcription or to amplify the transduced signal and to thereby increase the sensitivity and reliability of the assay. [0172]
  • Many reporter genes and transcriptional regulatory elements are known to those of skill in the art and others may be identified or synthesized by methods known to those of skill in the art. [0173]
  • A reporter gene includes any gene that expresses a detectable gene product, which may be RNA or protein. Preferred reporter genes are those that are readily detectable. The reporter gene may also be included in the construct in the form of a fusion gene with a gene that includes desired transcriptional regulatory sequences or exhibits other desirable properties. [0174]
  • Examples of reporter genes include, but are not limited to CAT (chloramphenicol acetyl transferase) (Alton and Vapnek (1979), Nature 282: 864-869) luciferase, and other enzyme detection systems, such as beta-galactosidase; firefly luciferase (deWet et al. (1987), Mol. Cell. Biol. 7:725-737); bacterial luciferase (Engebrecht and Silverman (1984), PNAS 1: 4154-4158; Baldwin et al. (1984), Biochemistry 23: 3663-3667); alkaline phosphatase (Toh et al. (1989) Eur. J. Biochem. 182: 231-238, Hall et al. (1983) J. Mol. Appl. Gen. 2: 101). [0175]
  • Transcriptional control elements include, but are not limited to, promoters, enhancers, and repressor and activator binding sites, Suitable transcriptional regulatory elements may be derived from the transcriptional regulatory regions of genes whose expression is rapidly induced, generally within minutes, of contact between the cell surface protein and the effector protein that modulates the activity of the cell surface protein. Examples of such genes include, but are not limited to, the immediate early genes (see, Sheng et al. (1990) Neuron 4: 477-485), such as c-fos, Immediate early genes are genes that are rapidly induced upon binding of a ligand to a cell surface protein. The transcriptional control elements that are preferred for use in the gene constructs include transcriptional control elements from immediate early genes, elements derived from other genes that exhibit some or all of the characteristics of the immediate early genes, or synthetic elements that are constructed such that genes in operative linkage therewith exhibit such characteristics. The characteristics of preferred genes from which the transcriptional control elements are derived include, but are not limited to, low or undetectable expression in quiescent cells, rapid induction at the transcriptional level within minutes of extracellular simulation, induction that is transient and independent of new protein synthesis, subsequent shut-off of transcription requires new protein synthesis, and mRNAs transcribed from these genes have a short half-life. It is not necessary for all of these properties to be present. [0176]
  • In vivo Activity Assays [0177]
  • Various experimental procedures, well known in the art, are useful in the present invention to assess the analgesic effect of compounds, such as the “tail flick” and “hot plate” tests. The “tail flick” test can be performed by applying a noxious thermal stimulus to the rat's tail and determining the time until the nociceptive tail flick occurs. Analgesia is demonstrated by an increase in time to occurrence of a tail flick response. The “hot plate” test is similarly performed, except that the noxious thermal stimulus is applied to the rat's paws. [0178]
  • An experimental procedure, well known in the art, useful in the present invention to assess the ability of compounds to cause respiratory depression is to monitor blood gases. This method employees measuring the partial pressures of oxygen and carbon dioxide in blood samples taken from animals following compound administration. A decrease in the partial pressures of oxygen and an increase in the partial pressure of carbon dioxide may be indicative of respiratory depression. [0179]
  • An experimental procedure, well known in the art, useful in the present invention to assess the ability of compounds to cause inhibition of gastrointestinal motility is the “charcoal meal test”. This method measures the propulsion of intestinal contents following administration of test compounds. A decrease in the propulsion of intestinal contents may be indicative of inhibition of gastrointestinal motility. [0180]
  • Various experimental procedures, well known in the art, are useful in the present invention to assess the ability of compounds to cause tolerance. Tolerance can be defmed as a condition characterized by unresponsiveness or decreased responsiveness following prolonged or multiple exposure to a compound compared to the responsiveness demonstrated upon initial exposure. [0181]
  • Various experimental procedures, well known in the art, are useful in the present invention to assess the ability of compounds to cause physical dependence. In the present invention, the ability of test compounds to cause physical dependence was accessed by giving animals escalating doses of test compounds for five days. After the final dose the animals were given naloxone, an opioid antagonist and observed for behavioral signs of dependence, such as vertical jumping. [0182]
  • Pharmaceutical Compositions [0183]
  • In another aspect, the present invention provides pharmaceutically acceptable compositions which comprise a therapeutically-effective amount of one or more of the compounds described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. As described in detail below, the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; or (8) nasally. [0184]
  • The phrase “therapeutically-effective amount” as used herein means that amount of a compound, material, or composition comprising a compound of the present invention which is effective for producing some desired therapeutic effect in at least a sub-population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment. [0185]
  • The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. [0186]
  • The phrase “pharmaceutically-acceptable carrier” as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) pH buffered solutions; (21) polyesters, polycarbonates and/or polyanhydrides; and (22) other non-toxic compatible substances employed in pharmaceutical formulations. [0187]
  • As set out above, certain embodiments of the present compounds may contain a basic functional group, such as amino or alkylamino, and are, thus, capable of forming pharmaceutically-acceptable salts with pharmaceutically-acceptable acids. The term “pharmaceutically-acceptable salts” in this respect, refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds of the present invention. These salts can be prepared in situ in the administration vehicle or the dosage form manufacturing process, or by separately reacting a purified compound of the invention in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed during subsequent purification. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like. (See, for example, Berge et al. (1977) “Pharmaceutical Salts”, [0188] J. Pharm. Sci. 66:1-19)
  • The pharmaceutically acceptable salts of the subject compounds include the conventional nontoxic salts or quaternary ammonium salts of the compounds, e.g., from non-toxic organic or inorganic acids. For example, such conventional nontoxic salts include those derived from inorganic acids such as hydrochloride, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, palmitic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicyclic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isothionic, and the like. [0189]
  • In other cases, the compounds of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically-acceptable salts with pharmaceutically-acceptable bases. The term “pharmaceutically-acceptable salts” in these instances refers to the relatively non-toxic, inorganic and organic base addition salts of compounds of the present invention. These salts can likewise be prepared in situ in the administration vehicle or the dosage form manufacturing process, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with ammonia, or with a pharmaceutically-acceptable organic primary, secondary or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. (See, for example, Berge et al., supra) [0190]
  • Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions. [0191]
  • Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. [0192]
  • Formulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent. [0193]
  • In certain embodiments, a formulation of the present invention comprises an excipient selected from the group consisting of cyclodextrins, liposomes, micelle forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and polyanhydrides; and a compound of the present invention. In certain embodiments, an aforementioned formulation renders orally bioavailable a compound of the present invention. [0194]
  • Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product. [0195]
  • Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A compound of the present invention may also be administered as a bolus, electuary or paste. [0196]
  • In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol, glycerol monostearate, and non-ionic surfactants; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-shelled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like. [0197]
  • A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. [0198]
  • The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharnaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be formulated for rapid release, e.g., freeze-dried. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients. [0199]
  • Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. [0200]
  • Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents. [0201]
  • Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof. [0202]
  • Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound. [0203]
  • Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate. [0204]
  • Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required. [0205]
  • The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. [0206]
  • Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. [0207]
  • Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel. [0208]
  • Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. [0209]
  • Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents. [0210]
  • Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. [0211]
  • These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin. [0212]
  • In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. [0213]
  • Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. [0214]
  • When the compounds of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier. [0215]
  • The preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given in forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administrations are preferred. [0216]
  • The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticulare, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion. [0217]
  • The phrases “systemic administration,” “administered systemically,” “peripheral administration” and “administered peripherally” as used herein mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration. [0218]
  • These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracistemally and topically, as by powders, ointments or drops, including buccally and sublingually. [0219]
  • Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art. [0220]
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. [0221]
  • The selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. [0222]
  • A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. [0223]
  • In general, a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Generally, intravenous, intracerebroventricular and subcutaneous doses of the compounds of this invention for a patient, when used for the indicated analgesic effects, will range from about 0.0001 to about 100 mg per kilogram of body weight per day. [0224]
  • If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. [0225]
  • While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation (composition). [0226]
  • The compounds according to the invention may be formulated for administration in any convenient way for use in human or veterinary medicine, by analogy with other pharmaceuticals. [0227]
  • The term “treatment” is intended to encompass also prophylaxis, therapy and cure. [0228]
  • The patient receiving this treatment is any animal in need, including primates, in particular humans, and other mammals such as equines, cattle, swine and sheep; and poultry and pets in general. [0229]
  • The compound of the invention can be administered as such or in admixtures with pharmaceutically acceptable carriers and can also be administered in conjunction with antimicrobial agents such as penicillins, cephalosporins, aminoglycosides and glycopeptides. Conjunctive therapy, thus includes sequential, simultaneous and separate administration of the active compound in a way that the therapeutical effects of the first administered one is not entirely disappeared when the subsequent is administered. [0230]
  • The addition of the active compound of the invention to animal feed is preferably accomplished by preparing an appropriate feed premix containing the active compound in an effective amount and incorporating the premix into the complete ration. [0231]
  • Alternatively, an intermediate concentrate or feed supplement containing the active ingredient can be blended into the feed. The way in which such feed premixes and complete rations can be prepared and administered are described in reference books (such as “Applied Animal Nutrition”, W. H. Freedman and CO., San Francisco, U.S.A., 1969 or “Livestock Feeds and Feeding” O and B books, Corvallis, Oreg., U.S.A., 1977). [0232]
  • Micelles [0233]
  • Recently, the pharmaceutical industry introduced microemulsification technology to improve bioavailability of some lipophilic (water insoluble) pharmaceutical agents. Examples include Trimetrine (Dordunoo, S. K., et al., Drug Development and Industrial Pharmacy, 17(12), 1685-1713, 1991 and REV 5901 (Sheen, P. C., et al., J Pharm Sci 80(7), 712-714, 1991). Among other things, microemulsification provides enhanced bioavailability by preferentially directing absorption to the lymphatic system instead of the circulatory system, which thereby bypasses the liver, and prevents destruction of the compounds in the hepatobiliary circulation. [0234]
  • In one aspect of invention, the formulations contain micelles formed from a compound of the present invention and at least one amphiphilic carrier, in which the micelles have an average diameter of less than about 100 nm. More preferred embodiments provide micelles having an average diameter less than about 50 nm, and even more preferred embodiments provide micelles having an average diameter less than about 30 nm, or even less than about 20 nm. [0235]
  • While all suitable amphiphilic carriers are contemplated, the presently preferred carriers are generally those that have Generally-Recognized-as-Safe (GRAS) status, and that can both solubilize the compound of the present invention and microemulsify it at a later stage when the solution comes into a contact with a complex water phase (such as one found in human gastro-intestinal tract). Usually, amphiphilic ingredients that satisfy these requirements have HLB (hydrophilic to lipophilic balance) values of 2-20, and their structures contain straight chain aliphatic radicals in the range of C-6 to C-20. Examples are polyethylene-glycolized fatty glycerides and polyethylene glycols. [0236]
  • Particularly preferred amphiphilic carriers are saturated and monounsaturated polyethyleneglycolyzed fatty acid glycerides, such as those obtained from fully or partially hydrogenated various vegetable oils. Such oils may advantageously consist of tri-. di- and mono-fatty acid glycerides and di- and mono-polyethyleneglycol esters of the corresponding fatty acids, with a particularly preferred fatty acid composition including capric acid 4-10, capric acid 3-9, lauric acid 40-50, myristic acid 14-24, palmitic acid 4-14 and stearic acid 5-15%. Another useful class of amphiphilic carriers includes partially esterified sorbitan and/or sorbitol, with saturated or mono-unsaturated fatty acids (SPAN-series) or corresponding ethoxylated analogs (TWEEN-series). [0237]
  • Commercially available amphiphilic carriers are particularly contemplated, including Gelucire-series, Labrafil, Labrasol, or Lauroglycol (all manufactured and distributed by Gattefosse Corporation, Saint Priest, France), PEG-mono-oleate, PEG-di- oleate, PEG-mono-laurate and di-laurate, Lecithin, Polysorbate 80, etc (produced and distributed by a number of companies in USA and worldwide). [0238]
  • Polymers [0239]
  • Hydrophilic polymers suitable for use in the present invention are those which are readily water-soluble, can be covalently attached to a vesicle-forning lipid, and which are tolerated in vivo without toxic effects (i.e., are biocompatible). Suitable polymers include polyethylene glycol (PEG), polylactic (also termed polylactide), polyglycolic acid (also termed polyglycolide), a polylactic-polyglycolic acid copolymer, and polyvinyl alcohol. Preferred polymers are those having a molecular weight of from about 100 or 120 daltons up to about 5,000 or 10,000 daltons, and more preferably from about 300 daltons to about 5,000 daltons. In a particularly preferred embodiment, the polymer is polyethyleneglycol having a molecular weight of from about 100 to about 5,000 daltons, and more preferably having a molecular weight of from about 300 to about 5,000 daltons. In a particularly preferred embodiment, the polymer is polyethyleneglycol of 750 daltons (PEG(750)). Polymers may also be defined by the number of monomers therein; a preferred embodiment of the present invention utilizes polymers of at least about three monomers, such PEG polymers consisting of three monomers (approximately 150 daltons). [0240]
  • Other hydrophilic polymers which may be suitable for use in the present invention include polyvinylpyrrolidone, polymethoxazoline, polyethyloxazoline, polyhydroxypropyl methacrylamide, polymethacrylamide, polydimethylacrylamide, and derivatized celluloses such as hydroxymethylcellulose or hydroxyethylcellulose. [0241]
  • In certain embodiments, a formulation of the present invention comprises a biocompatible polymer selected from the group consisting of polyamides, polycarbonates, polyalkylenes, polymers of acrylic and methacrylic esters, polyvinyl polymers, polyglycolides, polysiloxanes, polyurethanes and co-polymers thereof, celluloses, polypropylene, polyethylenes, polystyrene, polymers of lactic acid and glycolic acid, polyanhydrides, poly(ortho)esters, poly(butic acid), poly(valeric acid), poly(lactide-co-caprolactone), polysaccharides, proteins, polyhyaluronic acids, polycyanoacrylates, and blends, mixtures, or copolymers thereof. [0242]
  • Cyclodextrins [0243]
  • Cyclodextrins are cyclic oligosaccharides, consisting of 6, 7 or 8 glucose units, designated by the Greek letter alpha., .beta. or gamma., respectively. Cyclodextrins with fewer than six glucose units are not known to exist. The glucose units are linked by alpha-1,4-glucosidic bonds. As a consequence of the chair conformation of the sugar units, all secondary hydroxyl groups (at C-2, C-3) are located on one side of the ring, while all the primary hydroxyl groups at C-6 are situated on the other side. As a result, the external faces are hydrophilic, making the cyclodextrins water-soluble. In contrast, the cavities of the cyclodextrins are hydrophobic, since they are lined by the hydrogen of atoms C-3 and C-5, and by ether-like oxygens. These matrices allow complexation with a variety of relatively hydrophobic compounds, including, for instance, steroid compounds such as 17.beta.-estradiol (see, e.g., van Uden et al. Plant Cell Tiss. Org. Cult. 38:1-3-113 (1994)). The complexation takes place by Van der Waals interactions and by hydrogen bond formation. For a general review of the chemistry of cyclodextrins, see, Wenz, Agnew. Chem. Int. Ed. Engl., 33:803-822 (1994). [0244]
  • The physico-chemical properties of the cyclodextrin derivatives depend strongly on the kind and the degree of substitution. For example, their solubility in water ranges from insoluble (e.g., triacetyl-beta-cyclodextrin) to 147% soluble (w/v) (G-2-beta-cyclodextrin). In addition, they are soluble in many organic solvents. The properties of the cyclodextrins enable the control over solubility of various formulation components by increasing or decreasing their solubility. [0245]
  • Numerous cyclodextrins and methods for their preparation have been described. For example, Parmeter (I), et al. (U.S. Pat. No. 3,453,259) and Gramera, et al. (U.S. Pat. No. 3,459,731) described electroneutral cyclodextrins. Other derivatives include cyclodextrins with cationic properties [Parmeter (II), U.S. Pat. No. 3,453,257], insoluble crosslinked cyclodextrins (Solms, U.S. Pat. No. 3,420,788), and cyclodextrins with anionic properties [Parmeter (III), U.S. Pat. No. 3,426,011]. Among the cyclodextrin derivatives with anionic properties, carboxylic acids, phosphorous acids, phosphinous acids, phosphonic acids, phosphoric acids, thiophosphonic acids, thiosulphinic acids, and sulfonic acids have been appended to the parent cyclodextrin [see, Parmeter (III), supra]. Furthermore, sulfoalkyl ether cyclodextrin derivatives have been described by Stella, et al. (U.S. Pat. No. 5,134,127). [0246]
  • Liposomes [0247]
  • Liposomes consist of at least one lipid bilayer membrane enclosing an aqueous internal compartment. Liposomes may be characterized by membrane type and by size. Small unilamellar vesicles (SUVs) have a single membrane and typically range between 0.02 and 0.05 μm in diameter; large unilamellar vesicles (LUVS) are typically larger than 0.05 μm Oligolamellar large vesicles and multilamellar vesicles have multiple, usually concentric, membrane layers and are typically larger than 0.1 μm. Liposomes with several nonconcentric membranes, i.e., several smaller vesicles contained within a larger vesicle, are termed multivesicular vesicles. [0248]
  • One aspect of the present invention relates to formulations comprising liposomes containing a compound of the present invention, where the liposome membrane is formulated to provide a liposome with increased carrying capacity. Alternatively or in addition, the compound of the present invention may be contained within, or adsorbed onto, the liposome bilayer of the liposome. The compound of the present invention may be aggregated with a lipid surfactant and carried within the liposome's internal space; in these cases, the liposome membrane is formulated to resist the disruptive effects of the active agent-surfactant aggregate. [0249]
  • According to one embodiment of the present invention, the lipid bilayer of a liposome contains lipids derivatized with polyethylene glycol (PEG), such that the PEG chains extend from the inner surface of the lipid bilayer into the interior space encapsulated by the liposome, and extend from the exterior of the lipid bilayer into the surrounding environment. [0250]
  • Active agents contained within liposomes of the present invention are in solubilized form. Aggregates of surfactant and active agent (such as emulsions or micelles containing the active agent of interest) may be entrapped within the interior space of liposomes according to the present invention. A surfactant acts to disperse and solubilize the active agent, and may be selected from any suitable aliphatic, cycloaliphatic or aromatic surfactant, including but not limited to biocompatible lysophosphatidylcholines (LPCs) of varying chain lengths (for example, from about C.sub.14 to about C.sub.20). Polymer-derivatized lipids such as PEG-lipids may also be utilized for micelle formation as they will act to inhibit micelle/membrane fusion, and as the addition of a polymer to surfactant molecules decreases the CMC of the surfactant and aids in micelle formation. Preferred are surfactants with CMCs in the micromolar range; higher CMC surfactants may be utilized to prepare micelles entrapped within liposomes of the present invention, however, micelle surfactant monomers could affect liposome bilayer stability and would be a factor in designing a liposome of a desired stability. [0251]
  • Liposomes according to the present invention may be prepared by any of a variety of techniques that are known in the art. See, e.g., U.S. Pat. No. 4,235,871; Published PCT applications WO 96/14057; New RRC, Liposomes: A practical approach, IRL Press, Oxford (1990), pages 33-104; Lasic DD, Liposomes from physics to applications, Elsevier Science Publishers BV, Amsterdam, 1993. [0252]
  • For example, liposomes of the present invention may be prepared by diffusing a lipid derivatized with a hydrophilic polymer into preformed liposomes, such as by exposing preformed liposomes to micelles composed of lipid-grafted polymers, at lipid concentrations corresponding to the final mole percent of derivatized lipid which is desired in the liposome. Liposomes containing a hydrophilic polymer can also be formed by homogenization, lipid-field hydration, or extrusion techniques, as are known in the art. [0253]
  • In another exemplary formulation procedure, the active agent is first dispersed by sonication in a lysophosphatidylcholine or other low CMC surfactant (including polymer grafted lipids) that readily solubilizes hydrophobic molecules. The resulting micellar suspension of active agent is then used to rehydrate a dried lipid sample that contains a suitable mole percent of polymer-grafted lipid, or cholesterol. The lipid and active agent suspension is then formed into liposomes using extrusion techniques as are known in the art, and the resulting liposomes separated from the unencapsulated solution by standard column separation. [0254]
  • In one aspect of the present invention, the liposomes are prepared to have substantially homogeneous sizes in a selected size range. One effective sizing method involves extruding an aqueous suspension of the liposomes through a series of polycarbonate membranes having a selected uniform pore size; the pore size of the membrane will correspond roughly with the largest sizes of liposomes produced by extrusion through that membrane. See e.g., U.S. Pat. No. 4,737,323 (Apr. 12, 1988). [0255]
  • Release Modifiers [0256]
  • The release characteristics of a formulation of the present invention depend on the encapsulating material, the concentration of encapsulated drug, and the presence of release modifiers. For example, release can be manipulated to be pH dependent, for example, using a pH sensitive coating that releases only at a low pH, as in the stomach, or a higher pH, as in the intestine. An enteric coating can be used to prevent release from occurring until after passage through the stomach. Multiple coatings or mixtures of cyanamide encapsulated in different materials can be used to obtain an initial release in the stomach, followed by later release in the intestine. Release can also be manipulated by inclusion of salts or pore forming agents, which can increase water uptake or release of drug by diffusion from the capsule. Excipients which modify the solubility of the drug can also be used to control the release rate. Agents which enhance degradation of the matrix or release from the matrix can also be incorporated. They can be added to the drug, added as a separate phase (i.e., as particulates), or can be co-dissolved in the polymer phase depending on the compound. In all cases the amount should be between 0.1 and thirty percent (w/w polymer). Types of degradation enhancers include inorganic salts such as ammonium sulfate and ammonium chloride, organic acids such as citric acid, benzoic acid, and ascorbic acid, inorganic bases such as sodium carbonate, potassium carbonate, calcium carbonate, zinc carbonate, and zinc hydroxide, and organic bases such as protamine sulfate, spermine, choline, ethanolamine, diethanolamine, and triethanolamine and surfactants such as Tween.RTM. and Pluronic.RTM.. Pore forming agents which add microstructure to the matrices (i.e., water soluble compounds such as inorganic salts and sugars) are added as particulates. The range should be between one and thirty percent (w/w polymer). [0257]
  • Uptake can also be manipulated by altering residence time of the particles in the gut. This can be achieved, for example, by coating the particle with, or selecting as the encapsulating material, a mucosal adhesive polymer. Examples include most polymers with free carboxyl groups, such as chitosan, celluloses, and especially polyacrylates (as used herein, polyacrylates refers to polymers including acrylate groups and modified acrylate groups such as cyanoacrylates and methacrylates). [0258]
  • Combinatorial Libraries [0259]
  • The subject reactions readily lend themselves to the creation of combinatorial libraries of compounds for the screening of pharmaceutical, agrochemical or other biological or medically-related activity or material-related qualities. A combinatorial library for the purposes of the present invention is a mixture of chemically related compounds which may be screened together for a desired property; said libraries may be in solution or covalently linked to a solid support. The preparation of many related compounds in a single reaction greatly reduces and simplifies the number of screening processes which need to be carried out. Screening for the appropriate biological, pharmaceutical, agrochemical or physical property may be done by conventional methods. [0260]
  • Diversity in a library can be created at a variety of different levels. For instance, the substrate aryl groups used in a combinatorial approach can be diverse in terms of the core aryl moiety, e.g., a variegation in terms of the ring structure, and/or can be varied with respect to the other substituents. [0261]
  • A variety of techniques are available in the art for generating combinatorial libraries of small organic molecules. See, for example, Blondelle et al. (1995) [0262] Trends Anal. Chem. 14:83; the Affymax U.S. Pat. Nos. 5,359,115 and 5,362,899: the Ellman U.S. Pat. No. 5,288,514: the Still et al. PCT publication WO 94/08051; Chen et al. (1994) JACS 116:2661: Kerr et al. (1993) JACS 115:252; PCT publications WO92/10092, WO93/09668 and WO91/07087; and the Lerner et al. PCT publication WO93/20242). Accordingly, a variety of libraries on the order of about 16 to 1,000,000 or more diversomers can be synthesized and screened for a particular activity or property.
  • In an exemplary embodiment, a library of substituted diversomers can be synthesized using the subject reactions adapted to the techniques described in the Still et al. PCT publication WO 94/08051, e.g., being linked to a polymer bead by a hydrolyzable or photolyzable group, e.g., located at one of the positions of substrate. According to the Still et al. technique, the library is synthesized on a set of beads, each bead including a set of tags identifying the particular diversomer on that bead. In one embodiment, which is particularly suitable for discovering enzyme inhibitors, the beads can be dispersed on the surface of a permeable membrane, and the diversomers released from the beads by lysis of the bead linker. The diversomer from each bead will diffuse across the membrane to an assay zone, where it will interact with an enzyme assay. Detailed descriptions of a number of combinatorial methodologies are provided below. [0263]
  • A) Direct Characterization [0264]
  • A growing trend in the field of combinatorial chemistry is to exploit the sensitivity of techniques such as mass spectrometry (MS), e.g., which can be used to characterize sub-femtomolar amounts of a compound, and to directly determine the chemical constitution of a compound selected from a combinatorial library. For instance, where the library is provided on an insoluble support matrix, discrete populations of compounds can be first released from the support and characterized by MS. In other embodiments, as part of the MS sample preparation technique, such MS techniques as MALDI can be used to release a compound from the matrix, particularly where a labile bond is used originally to tether the compound to the matrix. For instance, a bead selected from a library can be irradiated in a MALDI step in order to release the diversomer from the matrix, and ionize the diversomer for MS analysis. [0265]
  • B) Multipin Synthesis [0266]
  • The libraries of the subject method can take the multipin library format. Briefly, Geysen and co-workers (Geysen et al. (1984) [0267] PNAS 81:3998-4002) introduced a method for generating compound libraries by a parallel synthesis on polyacrylic acid-grated polyethylene pins arrayed in the microtitre plate format. The Geysen technique can be used to synthesize and screen thousands of compounds per week using the multipin method, and the tethered compounds may be reused in many assays. Appropriate linker moieties can also been appended to the pins so that the compounds may be cleaved from the supports after synthesis for assessment of purity and further evaluation (c.f., Bray et al. (1990) Tetrahedron Lett 31:5811-5814; Valerio et al. (1991) Anal Biochem 197:168-177; Bray et al. (1991) Tetrahedron Lett 32:6163-6166).
  • C) Divide-Couple-Recombine [0268]
  • In yet another embodiment, a variegated library of compounds can be provided on a set of beads utilizing the strategy of divide-couple-recombine (see, e.g., Houghten (1985) PNAS 82:5131-5135; and U.S. Pat. Nos. 4,631,211; 5,440,016; 5,480,971). Briefly, as the name implies, at each synthesis step where degeneracy is introduced into the library, the beads are divided into separate groups equal to the number of different substituents to be added at a particular position in the library, the different substituents coupled in separate reactions, and the beads recombined into one pool for the next iteration. [0269]
  • In one embodiment, the divide-couple-recombine strategy can be carried out using an analogous approach to the so-called “tea bag” method first developed by Houghten, where compound synthesis occurs on resin sealed inside porous polypropylene bags (Houghten et al. (1986) [0270] PNAS 82:5131-5135). Substituents are coupled to the compound-bearing resins by placing the bags in appropriate reaction solutions, while all common steps such as resin washing and deprotection are performed simultaneously in one reaction vessel. At the end of the synthesis, each bag contains a single compound.
  • D) Combinatorial Libraries by Light-Directed, Spatially Addressable Parallel Chemical Synthesis [0271]
  • A scheme of combinatorial synthesis in which the identity of a compound is given by its locations on a synthesis substrate is termed a spatially-addressable synthesis. In one embodiment, the combinatorial process is carried out by controlling the addition of a chemical reagent to specific locations on a solid support (Dower et al. (1991) [0272] Annu Rep Med Chem 26:271-280; Fodor, S.P.A. (1991) Science 251:767; Pirrung et al. (1992) U.S. Pat. No. 5,143,854; Jacobs et al. (1994) Trends Biotechnol 12:19-26). The spatial resolution of photolithography affords miniaturization. This technique can be carried out through the use protection/deprotection reactions with photolabile protecting groups.
  • The key points of this technology are illustrated in Gallop et al. (1994) [0273] J Med Chem 37:1233-1251. A synthesis substrate is prepared for coupling through the covalent attachment of photolabile nitroveratryloxycarbonyl (NVOC) protected amino linkers or other photolabile linkers. Light is used to selectively activate a specified region of the synthesis support for coupling. Removal of the photolabile protecting groups by light (deprotection) results in activation of selected areas. After activation, the first of a set of amino acid analogs, each bearing a photolabile protecting group on the amino terminus, is exposed to the entire surface. Coupling only occurs in regions that were addressed by light in the preceding step. The reaction is stopped, the plates washed, and the substrate is again illuminated through a second mask, activating a different region for reaction with a second protected building block. The pattern of masks and the sequence of reactants define the products and their locations. Since this process utilizes photolithography techniques, the number of compounds that can be synthesized is limited only by the number of synthesis sites that can be addressed with appropriate resolution. The position of each compound is precisely known; hence, its interactions with other molecules can be directly assessed.
  • In a light-directed chemical synthesis, the products depend on the pattern of illumination and on the order of addition of reactants. By varying the lithographic patterns, many different sets of test compounds can be synthesized simultaneously; this characteristic leads to the generation of many different masking strategies. [0274]
  • E) Encoded Combinatorial Libraries [0275]
  • In yet another embodiment, the subject method utilizes a compound library provided with an encoded tagging system. A recent improvement in the identification of active compounds from combinatorial libraries employs chemical indexing systems using tags that uniquely encode the reaction steps a given bead has undergone and, by inference, the structure it carries. Conceptually, this approach mimics phage display libraries, where activity derives from expressed peptides, but the structures of the active peptides are deduced from the corresponding genomic DNA sequence. The first encoding of synthetic combinatorial libraries employed DNA as the code. A variety of other forms of encoding have been reported, including encoding with sequenceable bio- oligomers (e.g., oligonucleotides and peptides), and binary encoding with additional non- sequenceable tags. [0276]
  • 1) Tagging with sequenceable bio-oligomers [0277]
  • The principle of using oligonucleotides to encode combinatorial synthetic libraries was described in 1992 (Brenner et al. (1992) PNAS 89:5381-5383), and an example of such a library appeared the following year (Needles et al. (1993) [0278] PNAS 90:10700-10704). A combinatorial library of nominally 77 (=823,543) peptides composed of all combinations of Arg, Gln, Phe, Lys, Val, D-Val and Thr (three-letter amino acid code), each of which was encoded by a specific dinucleotide (TA, TC, CT, AT, TT, CA and AC, respectively), was prepared by a series of alternating rounds of peptide and oligonucleotide synthesis on solid support. In this work, the amine linking functionality on the bead was specifically differentiated toward peptide or oligonucleotide synthesis by simultaneously preincubating the beads with reagents that generate protected OH groups for oligonucleotide synthesis and protected NH2 groups for peptide synthesis (here, in a ratio of 1:20). When complete, the tags each consisted of 69-mers, 14 units of which carried the code. The bead-bound library was incubated with a fluorescently labeled antibody, and beads containing bound antibody that fluoresced strongly were harvested by fluorescence-activated cell sorting (FACS). The DNA tags were amplified by PCR and sequenced, and the predicted peptides were synthesized. Following such techniques, compound libraries can be derived for use in the subject method, where the oligonucleotide sequence of the tag identifies the sequential combinatorial reactions that a particular bead underwent, and therefore provides the identity of the compound on the bead.
  • The use of oligonucleotide tags permits exquisitely sensitive tag analysis. Even so, the method requires careful choice of orthogonal sets of protecting groups required for alternating co-synthesis of the tag and the library member. Furthermore, the chemical lability of the tag, particularly the phosphate and sugar anomeric linkages, may limit the choice of reagents and conditions that can be employed for the synthesis of non- oligomeric libraries. In preferred embodiments, the libraries employ linkers permitting selective detachment of the test compound library member for assay. [0279]
  • Peptides have also been employed as tagging molecules for combinatorial libraries. Two exemplary approaches are described in the art, both of which employ branched linkers to solid phase upon which coding and ligand strands are alternately elaborated. In the first approach (Kerr J M et al. (1993) [0280] J Am Chem Soc 115:2529-2531), orthogonality in synthesis is achieved by employing acid-labile protection for the coding strand and base-labile protection for the compound strand.
  • In an alternative approach (Nikolaiev et al. (1993) [0281] Pept Res 6:161-170), branched linkers are employed so that the coding unit and the test compound can both be attached to the same functional group on the resin. In one embodiment, a cleavable linker can be placed between the branch point and the bead so that cleavage releases a molecule containing both code and the compound (Ptek et al. (1991) Tetrahedron Lett 32:3891- 3894). In another embodiment, the cleavable linker can be placed so that the test compound can be selectively separated from the bead, leaving the code behind. This last construct is particularly valuable because it permits screening of the test compound without potential interference of the coding groups. Examples in the art of independent cleavage and sequencing of peptide library members and their corresponding tags has confirmed that the tags can accurately predict the peptide structure.
  • 2) Non-sequenceable Tagging: Binary Encoding [0282]
  • An alternative form of encoding the test compound library employs a set of non-sequencable electrophoric tagging molecules that are used as a binary code (Ohlmeyer et al. (1993) [0283] PNAS 90:10922-10926). Exemplary tags are haloaromatic alkyl ethers that are detectable as their trimethylsilyl ethers at less than femtomolar levels by electron capture gas chromatography (ECGC). Variations in the length of the alkyl chain, as well as the nature and position of the aromatic halide substituents, permit the synthesis of at least 40 such tags, which in principle can encode 240 (e.g., upwards of 1012) different molecules. In the original report (Ohlmeyer et al., supra) the tags were bound to about 1% of the available amine groups of a peptide library via a photocleavable o- nitrobenzyl linker. This approach is convenient when preparing combinatorial libraries of peptide-like or other amine-containing molecules. A more versatile system has, however, been developed that permits encoding of essentially any combinatorial library. Here, the compound would be attached to the solid support via the photocleavable linker and the tag is attached through a catechol ether linker via carbene insertion into the bead matrix (Nestler et al. (1994) J Org Chem 59:4723-4724). This orthogonal attachment strategy permits the selective detachment of library members for assay in solution and subsequent decoding by ECGC after oxidative detachment of the tag sets.
  • Although several amide-linked libraries in the art employ binary encoding with the electrophoric tags attached to amine groups, attaching these tags directly to the bead matrix provides far greater versatility in the structures that can be prepared in encoded combinatorial libraries. Attached in this way, the tags and their linker are nearly as unreactive as the bead matrix itself. Two binary-encoded combinatorial libraries have been reported where the electrophoric tags are attached directly to the solid phase (Ohlmeyer et al. (1995) [0284] PNAS 92:6027-6031) and provide guidance for generating the subject compound library. Both libraries were constructed using an orthogonal attachment strategy in which the library member was linked to the solid support by a photolabile linker and the tags were attached through a linker cleavable only by vigorous oxidation. Because the library members can be repetitively partially photoeluted from the solid support, library members can be utilized in multiple assays. Successive photoelution also permits a very high throughput iterative screening strategy: first, multiple beads are placed in 96-well microtiter plates; second, compounds are partially detached and transferred to assay plates; third, a metal binding assay identifies the active wells; fourth, the corresponding beads are rearrayed singly into new microtiter plates; fifth, single active compounds are identified; and sixth, the structures are decoded.
  • EXEMPLIFICATION
  • The invention now being generally described, it will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.[0285]
  • EXAMPLE 1
  • [0286]
    Figure US20020156092A1-20021024-C00013
  • The mixture of 3-pyridylcarbinol (15.39 g, 141 mmol), (2-bromo-ethyl)-benzene (31.17 g, 168.1 mmol) in CH[0287] 3CN (100 mL) was heated at 90° C. for 12 hr. After cool down to room temperature, the white solid was filtered and washed with cool acetone to give 1 as a white solid (39 g, 94% yield). LRMS [calculated for C14H16NO+, (M-Br)+] 214, found 214.
  • EXAMPLE 2
  • [0288]
    Figure US20020156092A1-20021024-C00014
  • To a solution of pyridium salt 1 (25 g, 85 mmol) in MeOH (300 mL) at 0° C. was added NaBH[0289] 4 (4.82 g, 127.6 mmol) slowly in several portions. After stirring overnight, the mixture was concentrated under vacuum. The residue was dissolved in 100 mL of H2O, extracted with EtOAc (3×100 mL). The combined organic solution was dried with Na2SO4, filtered and evaporated. Chromatography (silica gel, 2% MeOH in CH2Cl2) provided 2 as a yellow oil (17.5 g, 95% yield). 1H-NMR (300 MHz, CDCl3) δ7.40-7.20 (m, 5H), 5.8 (br s, 1H), 4.10 (s, 2H), 3.15 (m, 2H), 2.90 (m, 2H), 2.75 (m, 2H), 2.65 (m, 2H), 2.30 (m, 2H) ppm; 13C-NMR (75 MHz, CDCl3) δ 140.20, 136.59, 128.98, 128.76, 126.45, 121.04, 65.00, 60.60, 52.99, 50.29, 50.20, 33.58, 25.64 ppm; LRMS [calculated for C14H20NO2 (M+1)+] 218, found 218.
  • EXAMPLE 3
  • [0290]
    Figure US20020156092A1-20021024-C00015
  • To a solution of oxalyl chloride (7.0 mL, 34.10 mmol) in CH[0291] 2Cl2 (64 mL) at −55° C. was added a solution of DMSO (9.6 mL) in CH2Cl2 (32 mL) in 30 min. After addition, the mixture was stirred about 5 min at −55° C. Then, a solution of alcohol 2 (7.4 g, 34.10 mmol) in CH2Cl2 (732 mL) was added dropwise in 30 min. After addition, the mixture was stirred for 15 min followed by the addition of Et3N (45 mL). The mixture was stirred for 5 min and then allowed to warm up to room temperature. H20 (100 mL) was added. The two layers were separated. Aqueous layer was extracted with CH2Cl2 (2×100 mL). The combined organic solution was washed with brine (2×100 mL), dried with Na2SO4, filtered and evaporated to afford 3 as a light yellow liquid (6.9 g, 95% yield). IR (film) 3027, 2923, 2806, 1680, 1648, 1604, 1455, 1415, 1122, 967 cm−1; LRMS [calculated for C14H18NO, (M+1)+] 216, found 216.
  • EXAMPLE 4
  • [0292]
    Figure US20020156092A1-20021024-C00016
  • To a solution of 3 (3.17 g, 14.76 mmol) in CH[0293] 2Cl2 (50 mL) at 0° C. was added TFA (0.12 mL, 1.5 mmol). Then, benzyl-methoxy-trimethylsilanylmethyl-amine (4.2 g, 17.7 mmol) was added dropwise in 30 min. The mixture was stirred for 24 hr at room temperature. The reaction was quenched with aq. NaHCO3 (50 mL). Aqueous layer was separated, extracted with CH2Cl2 (2×30 mL). The combined organic solution was dried with Na2SO4, filtered and evaporated in vacuum. Chromatography (silica gel, 1-2% MeOH in CH2Cl2) provided 4 as a yellow oil. IR (film) 3079, 3059, 3027, 2915, 2806, 2726, 1725, 1600, 1499, 1455, 1351, 1130, 1097 cm−1; LRMS [calculated for C23H29N2O, (M+1)+] 349, found 349.
  • EXAMPLE 5
  • [0294]
    Figure US20020156092A1-20021024-C00017
  • To a solution of arecoline (5.27 g, 34.0 mmol) in CH[0295] 2Cl2 (34 mL) at 0° C. was added TFA (0.26 mL, 3.4 mmol). Then, benzyl-methoxy-trimethylsilanylmethyl-amine (9.67 g, 40.8 mmol) was added dropwise in 30 min. The mixture was stirred for 24 hr at room temperature. The reaction was quenched with aq. NaHCO3 (50 mL). Aqueous layer was separated, extracted with CH2Cl2 (2×30 mL). The combined organic solution was dried with Na2SO4, filtered and evaporated in vacuum. Chromatography (basic aluminum oxide, 10-30% i-PrOH in hexane with 0.2% Et2NH) provided pure 5 as a yellow oil (4.0 g, 41% yield). More 5 can be obtained from the mixed fractions. 1H-NMR (300 MHz, CDCl3) δ7.40-7.10 (m, 5H), 3.75-3.6 (m, 5H), 2.90-2.60 (m, 6H), 2.40 (m, 1H), 2.30 (m, 1H), 2.20 (s, 3H), 2.10 (m, 1H), 1.90 (m, 1H), 1.60 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 165.91, 137.36, 128.93, 53.21, 51.31, 50.72, 45.66, 26.60 ppm; IR (film) 2940, 2788, 1734, 1452, 1257, 1194, 1149, 1068 cm−1; LRMS [calculated for C17H25N2O2, (M+1)+] 289, found 289.
  • EXAMPLE 6
  • [0296]
    Figure US20020156092A1-20021024-C00018
  • To a solution of arecoline (6.94 g, 44.7 mmol) in 1,2-dichloroethane (40 mL) at 0° C. was added 1-chloroethyl chloroformate (7.3 mL, 67.12 mmol). The mixture was stirred at 0° C. for 30 min and then refluxed at 90° C. for 4 hr. After removal of solvent under vacuum, the residue was refluxed in methanol (40 mL) for additional 4 hr. Solvent was removed under vacuum. The residue was dissolved in CH[0297] 2Cl2 (40 mL), neutralized with sat. K2CO3 (20 mL). The two layers were separated. Aqueous layer was extracted with CH2Cl2 (30 mL). The combined organic solution was dried (Na2SO4) and evaporated to give 6 (3.77 g, 60% yield). 1H-NMR (300 MHz, CDCl3) δ 6.90 (m, 1H), 3.60 (m, 2H), 3.40 (s, 3H), 2.80 (m, 2H), 2.10 (m, 2H), 1.60 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 166.57, 138.47, 130.46, 51.55, 44.21, 42.02, 26.36 ppm.
  • EXAMPLE 7
  • [0298]
    Figure US20020156092A1-20021024-C00019
  • To a mixture of 6 (3.77 g, 26.7 mmol) in THF-H[0299] 2O (20 mL, 1:1) at 0° C. was added K2CO3 (5.5 g, 40.1 mmol) and (BOC)2O (7.0 g, 32.0 mmol). Th mixture was stirred for 4-5 hr. The two layers were separated. Aqueous layer was extracted with EtOAc (2×10 mL). The combined organic solution was dried (Na2SO4) and evaporated to give 7 (4.89 g, 76% yield). 1H-NMR (300 MHz, CDCl3) δ 7.10 (m, 1H), 4.15 (m, 2H), 165.71, 154.71, 137.96, 128.37, 79.88, 51.66, 42.67, 40.00, 38.73, 28.46, 25.58 ppm.
  • Example 8
  • [0300]
    Figure US20020156092A1-20021024-C00020
  • To a solution of 7 (4.53 g, 18.8 mmol) in CH[0301] 2Cl2 (20 mL) at 0° C. was added TFA (0.072 mL, 0.94 mmol). Then, benzyl-methoxy-trimethylsilanylmethyl-amine (6.2 g, 24.4 mmol) was added dropwise in 30 min. The mixture was stirred for 12 hr at room temperature. The reaction was quenched with aq. NaHCO3 (20 mL). Aqueous layer was separated, extracted with CH2Cl2 (2×30 mL). The combined organic solution was dried with Na2SO4 and evaporated in vacuum. Chromatography (basic aluminum oxide, 20-50% EtOAc in hexane) provided pure 8 as a yellow oil. LRMS [calculated for C21H31N2O4, (M+1)+] 375, found 375.
  • EXAMPLE 9
  • [0302]
    Figure US20020156092A1-20021024-C00021
  • To a solution of 4 (1.25 g, 3.59 mmol) in 5 % HOAc in MeOH (10 mL) was added aniline (0.72 g, 7.90 mmol). The mixture was stirred at 0° C. for 10 min and NaCNBH[0303] 3 (676 mg, 10.77 mmol) was added slowly. The mixture was stirred at room temperature for 36 hrs. The reaction was quenched by addition of aq. NaHCO3 (20 mL), extracted with EtOAc (2×20 mL). The combined organic solutions were dried with Na2SO4, filtered and evaporated. The crude product was purified by silica gel chromatography to give 9 as a light yellow oil. 1H-NMR (300 MHz, CDCl3) δ 7.40-7.10 (m, 12H), 6.70 (m, 1H), 6.50 (d, 2H), 5.60 (br, 1H, NH), 3.79 (dd, 2H), 3.30 (d, 1H), 3.18 (d, 1H), 2.95-2.40 (m, 10H), 2.40-2.00 (m, 4H), 1.65 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 149.42, 140.49, 139.72, 129.43, 128.98, 128.85, 128.76, 128.58, 127.22, 126.32, 63.84, 60.92, 60.71, 60.54, 57.88, 54.44, 49.89, 43.65, 39.55, 34.09, 25.89 ppm; IR (film) 3284, 3083, 3063, 3023, 2915, 2806, 1604, 1516, 1495, 1455, 1319, 1258, 1134 cm−1; LRMS [calculated for C29H36N3, (M+1)+] 426, found 426.
  • EXAMPLE 10
  • [0304]
    Figure US20020156092A1-20021024-C00022
  • To a solution of 4 (147 mg, 0.422 mmol) in 5 % HOAc in MeOH (2 mL) was added 2-fluorophenyl amine (93 mg, 0.844 mmol). The mixture was stirred at 0° C. for 10 min and NaCNBH[0305] 3 (265 mg, 4.22 mmol) was added slowly. The mixture was stirred at room temperature for 24 hrs. The reaction was quenched by addition of aq. NaHCO3 (2 mL), extracted with EtOAc (2×3 mL). The combined organic solutions were dried with NaSO4, filtered and evaporated. The crude product was purified preparative TLC (alumina plate, 20% EtOAc in hexane) to give 10 as a light yellow oil. 1H-NMR (300 MHz, CDCl3) δ 7.40-7.20 (m, 1OH), 7.00 (m, 2H), 6.70 (m, 2H), 6.12 (br, 1H, NH), 3.79 (dd, 2H), 3.30 (dd, 1H), 3.18 (d, 1H), 2.95-2.50 (m, 10H), 2.38-2.02 (m, 4H), 1.62 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 140.79, 139.82, 128.98, 128.80, 128.61, 128.52, 127.13, 126.20, 124.82, 115.63, 115.53, 114.51, 114.27, 111.39, 111.37, 63.70, 60.92, 60.22, 57.13, 54.12, 49.98, 43.62, 39.66, 34.22, 25.93 ppm; IR (film) 3256, 3059, 3031, 2927, 2810, 1620, 1528, 1495, 1339, 1298, 1194, 1134, 1029 cm−1; LRMS [calculated for C29H35N3, (M+1)+] 444, found 444.
  • EXAMPLE 11
  • [0306]
    Figure US20020156092A1-20021024-C00023
  • To a solution of 4 (340 mg, 0.97 mmol) in THF (2 mL) at 0° C. was added phenylmagnesiumbromide (3.0 M, 1.30 mL, 3.90 mmol). The mixture was then heated at 60° C. overnight. The reaction was quenched by addition of aq. NaHCO[0307] 3 (10 mL), extracted with EtOAc (3×10 mL). The combined organic solution was dried with NaSO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided 11 (58 mg), 12 (28 mg). The absolute stereochemistry of 11 and 12 were assigned randomly.
  • 11: [0308] 1H-NMR (300 MHz, CDCl3) δ 7.40-7.20 (m, 15H), 4.95 (s, 1H), 3.70-3.50 (AB, 2H), 3.05-2.90 (m, 4H), 3.90-3.65 (m, 4H), 2.60-2.50 (m, 2H), 2.50-2.20 (m, 4H), 1.60 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 141.40, 140.27, 139.87, 1128.96, 128.79, 128.52, 1218.47, 127.89, 127.56, 127.06, 126.49, 81.73, 61.93, 60.59, 60.52, 60.24, 56.59, 49.62, 47.10, 33.89, 33.30, 25.74 ppm; IR (film) 3168, 3079, 3063, 2935, 2814, 1608, 1495, 1475, 1451, 1375, 1347, 1130, 1102, 1073 cm−1; LRMS [calculated for C29H35N2O, (M+1)+] 427, found 427.
  • 12: [0309] 1H-NMR (300 MHz, CDCl3) δ 7.40-7.20 (m, 15H), 4.71 (s, 1H), 3.80-3.60 (m, 2H), 3.05-2.90 (m, 4H), 3.90-3.65 (m, 4H), 3.10-2.60 (m, 10H), 2.50-2.20 (m, 3H), 2.00 (d, 1H), 1.70 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 143.87, 140.45, 139.81, 129.34, 129.05, 128.86, 128.62, 128.47, 127.98, 127.20, 127.04, 126.62, 83.42, 62.50, 60.86, 60.74, 57.36, 56.28, 49.61, 47.38, 41.29, 33.90, 26.35 ppm; IR (film) 3236, 3083, 3023, 2947, 2919, 1604, 1499, 1451, 1375, 1130, 1106, 1029 cm−1; LRMS [calculated for C29H35N2O, (M+1)+] 427, found 427.
  • EXAMPLE 12
  • [0310]
    Figure US20020156092A1-20021024-C00024
  • To a solution of 4 (333 mg, 0.96 mmol) in THF (2 mL) at 0° C. was added MeLi (1.40 M, 4.2 mL, 5.9 mmol). The mixture was stirred at room temperature overnight. The reaction was quenched by addition of aq. NaHCO[0311] 3 (10 mL), extracted with EtOAc (3×10 mL). The combined organic solution was dried with Na2SO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided 13 as a brown oil (210 mg, 60% yield). 13C-NMR (75 MHz, CDCl3) δ 140.18, 139.33, 128.91, 128.77, 128.57, 127.28, 126.47, 75.02, 62.81, 61.16, 60.82, 60.46, 57.46, 50.32, 46.86, 33.85, 33.36, 25.32, 18.19 ppm; IR (film) 3500-3100, 3086, 3064, 2928, 2854, 1603, 1504, 1458, 1368, 1345, 1314, 1133, 1110, 1029 cm−1; LRMS [calculated for C24H33N2O, (M+1)+] 365, found 365.
  • EXAMPLE 13
  • [0312]
    Figure US20020156092A1-20021024-C00025
  • To a solution of 4 (805 mg, 2.31 mmol) in THF (5 mL) at 0° C. was added 3-methoxyphenylmagnesiumbromide (1.0 M, 6.90 mL, 6.90 mmol). The mixture was then heated at 70° C. overnight. The reaction was quenched by addition of aq. NaHCO[0313] 3 (10 mL), extracted with EtOAc (3×10 mL). The combined organic solution was dried with Na2SO4, filtered and evaporated. PTLC (alumina, 20% EtOAc in hexane) provided 14. LRMS [calculated for C30H37N2O2, (M+1)+] 457, found 457.
  • EXAMPLE 14
  • [0314]
    Figure US20020156092A1-20021024-C00026
  • To a solution of 15 (1.56 g, 4.14 mmol) in THF (10 mL) at −78° C. was added LiAlH[0315] 4 (0.20 g, 5.4 mmol) in one portion. The mixture was stirred under N2 and allowed to warm to room temperature. After 20 min, the reaction was quenched by slow addition of 2 mL of H2O. After stirring for about 30 min, the precipitated gray solid was filtered through Celite and rinsed several times with EtOAc. The solution was dried with Na2SO4, filtered and evaporated in vacuum to give pure 16 as a light yellow oil. ). 1H-NMR (300 MHz, CDCl3) δ 7.40-7.10 (m, 10H), 5.30 (broad, 1H), 3.90-3.60 (m, 4H), 3.00-2.00 (m, 14H), 1.70 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 140.32, 139.74, 128.98, 128.87, 128.81, 128.60, 127.24, 126.50, 73.61, 61.55, 61.05, 60.77, 60.59, 57.61, 50.26, 44.56, 39.48, 33.91, 25.96 ppm; LRMS [calculated for C23H31N20, (M+1)+] 351, found 351.
  • EXAMPLE 15
  • [0316]
    Figure US20020156092A1-20021024-C00027
  • To a solution of alcohol 16 (1.18 g, 3.37 mmol), i-Pr[0317] 2NEt (1.76 mL, 10.11 mmol) in CH2Cl2 (10 mL) at 0° C. was added methanesulfonyl chloride (0.39 mL, 5.06 mmol). The mixture was stirred for 0° C. to room temperature for 12 hr. Aqueous NaHCO3 (10 mL) was added. The two layers were separated. Aqueous layer was extracted with CH2Cl2 (2×10 mL). The combined organic solution was dried with NASO4 and evaporated. Chromatography (silica gel, 2% MeOH in CH2Cl2) provided 17 a light yellow oil (1.30 g, 90% yield). Rf=0.35 (silica gel plate, 10% MeOH in CH2Cl2); LRMS [calculated for C24H33N2O3S (M+1)+] 429, found 429.
  • EXAMPLE 16
  • [0318]
    Figure US20020156092A1-20021024-C00028
  • To a solution of 17 (652 mg, 1.52 mmol) in DMF (5 mL) was added phenol (171 mg, 1.82 mmol) and K[0319] 2CO3 (629 mg, 4.56 mmol). The mixture was heated under N2 at 90° C. overnight. After cool down to room temperature, the reaction mixture was diluted with H2O (20 mL) and extracted with EtOAc (2×30 mL). The combined organic solution was dried with Na2SO4 and evaporated. Chromatography (silica gel, 20% EtOAc in hexane) provided 18 as a light yellow oil (400mg, 62% yield). 1H-NMR (300 MHz, CDCl3) δ 7.60-7.20 (m, 13H), 7.10 (m, 2H), 4.25 (m, 1H), 3.90 (m, 2H), 3.05-2.60 (m, 10H), 2.50 (m, 2H), 2.33 (m, 1H), 1.95 (m, 1H), 1.70 (m, 1H), 1.42 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 159.80, 141.15, 140.16, 129.74, 129.15, 128.92, 128.60, 127.15, 126.17, 120.91, 72.30, 61.81, 61.26, 60.86, 56.84, 56.61, 51.08, 45.31, 37.35, 33.95, 25.42 ppm; LRMS [calculated for C29H35N2O (M+1)+] 427, found 427.
  • EXAMPLE 17
  • [0320]
    Figure US20020156092A1-20021024-C00029
  • To a solution of 5 (2.4 g, 8.33 mmol) in THF (10 mL) at −78° C. was added LiAlH[0321] 4 (0.31 g, 8.33 mmol) in one portion. The mixture was stirred under N2 and allowed to warm to room temperature. After 20 min, the reaction was quenched by slow addition of 2 mL of H2O. After stirring for about 30 min, the precipitated gray solid was filtered through Celite and rinsed several times with EtOAc. The solution was dried with Na2SO4, filtered and evaporated in vacuum to give pure 19 as a light yellow oil (2.02 g, 93% yield). 1H-NMR (300 MHz, CDCl3) δ 7.40-7.10 (m, 5H), 5.40 (br s, 1H), 3.60-3.40 (m, 4H), 2.70-2.50 (m, 3H), 2.43-2.28 (m, 3H), 2.20 (m, 1H), 2.10-1.80 (m, 6H), 1.40 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 139.66, 128.70, 128.37, 126.99, 70.32, 61.54, 60.98, 60.77, 57.04, 52.28, 46.71, 45.02, 37.61, 25.55 ppm; IR (film) 3500-3100, 2917, 2794, 1451, 1026 cm−1; LRMS [calculated for C16H25N2O, (M+1)+] 261, found 261.
  • EXAMPLE 18
  • [0322]
    Figure US20020156092A1-20021024-C00030
  • To a solution of oxalyl chloride (0.8 mL, 9.04 mmol) in CH[0323] 2C12 (20 mL) at −55° C. was added a solution of DMSO (1.30 mL, 16.6 mmol) in CH2Cl2 (3 mL) in 10 min. After addition, the mixture was stirred about 5 min at −55° C. Then, a solution of alcohol 19 (1.96 g, 7.54 mmol) in CH2Cl2 (7 mL) was added dropwise in 5 min. After addition, the mixture was stirred for 15 min followed by the addition of Et3N (5.3 mL). The mixture was stirred for 5 min and then allowed to warm up to room temperature. H2O (35 mL) was added. The two layers were separated. Aqueous layer was extracted with CH2Cl2 (2×15 mL). The combined organic solution was washed with brine, dried with Na2SO4, filtered and evaporated to afford 20 as a light yellow liquid (1.70 g, 87.4% yield). 1H-NMR (300 MHz, CDCl3) δ 9.7 (s, 1H), 7.30 (m, 5H), 3.76 (dd, 2H), 2.90-2.60 (m, 6H), 2.40 (m, 203.71, 139.55, 128.61, 128.48, 127.14, 60.45, 57.92, 57.05, 56.51, 56.06, 52.30, 46.96, 35.67, 25.96 ppm; IR (neat) 3026, 2936, 2792, 1722, 1452, 1146, 1028 cm−1; LRMS [calculated for C16H23N2O, (M+1)+] 259, found 259.
  • EXAMPLE 19
  • [0324]
    Figure US20020156092A1-20021024-C00031
  • To a solution of aldehyde 20 (1.60 g, 6.20 mmol) in 5 % HOAc in MeOH (10 mL) was added aniline (1.15 g, 12.40 mmol). The mixture was stirred at 0° C. for 10 min and NaCNBH[0325] 3 (1.60 g, 24.8 mmol) was added slowly. The mixture was stirred at room temperature for 12 hrs. The reaction was quenched by addition of aq. NaHCO3 (20 mL), extracted with EtOAc (2×20 mL). The combined organic solutions were dried with Na2SO4, filtered and evaporated. The crude product was purified by silica gel chromatography (20% EtOAc in hexane, then 30% i-PrOH in hexane) to give 21 as a light yellow liquid (1.20 g, 58% yield). ). 1H-NMR (300 MHz, CDCl3) δ 7.40-7.20 (m, 7H), 6.70 (m, 3H), 5.40 (br, 1H), 3.80 (dd, 2H), 3.30 (d, 1H), 3.18 (d, 1H), 2.95-2.79 (m, 2H), 2.70-2.59 (m, 4H), 2.40 (d, 1H), 2.30 (s, 3H), 2.2 (m, 2H), 2.10 (m, 1H), 1.60 (m, 1H) ppm; 13C-NMR (75 MHz, CDCl3) δ 149.44, 139.53, 129.47, 128.84, 128.59, 127.26, 116.78, 112.54, 63.34, 61.89, 60.95, 57.64, 53.77, 52.11, 46.81, 43.90, 38.68, 25.84 ppm; LRMS [calculated for C22H30N3, (M+1)+] 336, found 336.
  • EXAMPLE 20
  • [0326]
    Figure US20020156092A1-20021024-C00032
  • To a solution of 21 (1.15 g, 3.34 mmol), i-Pr[0327] 2NEt (1.30 mL, 10.3 mmol) in CH2Cl2 (10 mL) at 0° C. was added propionyl chloride (0.5 mL, 5.14 mmol). The mixture was stirred for 3 hrs at room temperature. Aqueous NaHCO3 (10 mL) was added. The two layers were separated. Aqueous layer was extracted with CH2Cl2 (2×10 mL). The combined organic solution was dried with Na2SO4, filtered and evaporated to give 22 as a light yellow oil (1.21 g, 90% yield). LRMS [calculated for C25H34N3O (M+1)+] 392, found 392.
  • EXAMPLE 21
  • [0328]
    Figure US20020156092A1-20021024-C00033
  • To a solution of 9 (17.2 mg, 0.04 mmol), i-Pr[0329] 2NEt (0.02 mL, 0.12 mmol) in CH2Cl2 (0.1 mL) at 0° C. was added propionyl chloride (0.004 mL, 0.048 mmol). The mixture was stirred for 3 hrs at 0° C. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided pure 23 (18.3 mg, 90% yield). 1H-NMR (300MHz, CDCl3) δ 7.45-7.10 (m, 15H), 4.21-4.00 (AB, 2H), 3.80-3.60 (AB, 2H), 2.80-2.00 (m, 15H), 1.80 (m, 1H), 1.50 (m, 1H), 1.30 (m, 1H), 1.10 (t, 3H) ppm; 13C-NMR (75 MHz, CDCl3) δ 175.04, 144.97, 141.07, 140.25, 129.71, 129.02, 128.72, 128.54, 128.40, 127.61, 126.90, 126.12, 61.56, 61.02, 60.87, 58.39, 56.16, 54.23, 50.50, 46.56, 38.60, 33.75, 28.18, 24.80, 10.05 ppm; IR (film) 3059, 3035, 2979, 2935, 2810, 1668, 1596, 1499, 1459, 1399, 1254, 1150, 1085, 1033 cm−1; LRMS [calculated for C32H40N3O (M+1)+] 482, found 482.
  • EXAMPLE 22
  • [0330]
    Figure US20020156092A1-20021024-C00034
  • To a solution of 9 (10 mg, 0.023 mmol), i-Pr2NEt (0.025 mL, 0.12 mmol) in CH[0331] 2Cl2 (0.1 mL) at 0° C. was added cyclobutanecarbonyl chloride (0.005 mL, 0.047 mmol). The mixture was stirred for 3 hrs at 0° C. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided pure 24. Rf=0.36 (aluminum oxide IB-F plate (J. T. Baker), 20% EtOAc in hexane); LRMS [calculated for C34H42N3O (M+1)+] 508, found 508.
  • EXAMPLE 23
  • [0332]
    Figure US20020156092A1-20021024-C00035
  • To a solution of 9 (10 mg, 0.023 mmol), i-Pr[0333] 2NEt (0.02 mL, 0.57 mmol) in CH2Cl2 (0.1 mL) at 0° C. was added methanesulfonyl chloride (0.004 mL, 0.051 mmol). The mixture was stirred for 3 hrs at 0° C. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided pure 25. LRMS [calculated for C30H38N3OS (M+1)+] 504, found 504.
  • EXAMPLE 24
  • [0334]
    Figure US20020156092A1-20021024-C00036
  • To a solution of 10 (10 mg, 0.022 mmol), i-Pr[0335] 2NEt (0.02 mL, 0.12 mmol) in CH2Cl2 (0.1 mL) at 0° C. was added propionyl chloride (0.004 mL, 0.048 mmol). The mixture was stirred for 3 hrs at 0° C. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided pure 26 (18.3 mg, 90% yield). Rf=0.18 (aluminum oxide IB-F plate (J. T. Baker), 20% EtOAc in hexane); IR (film) 3083, 3067, 3023, 2935, 2923, 2806, 1676, 1608, 1488, 1455, 1391, 1258, 1154, 1110, 1037, 913 cm−1; LRMS [calculated for C32H39N3O (M+1)+] 500, found 500.
  • EXAMPLE 25
  • [0336]
    Figure US20020156092A1-20021024-C00037
  • To a solution of 10 (10 mg, 0.022 mmol), i-Pr[0337] 2NEt (0.02 mL, 0.12 mmol) in CH2Cl2 (0.1 mL) at 0° C. was added cyclobutanecarbonyl chloride (0.004 mL). The mixture was stirred for 3 hrs at 0° C. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided pure 27. Rf=0.33 (aluminum oxide IB-F plate (J. T. Baker), 20% EtOAc in hexane); IR (film) 3067, 3023, 2939, 2802, 1668, 1608, 1499, 1455, 1395, 1266, 1134, 1106 cm−1; LRMS [calculated for C34H41FN3O (M+1)+] 526, found 526.
  • EXAMPLE 26
  • [0338]
    Figure US20020156092A1-20021024-C00038
  • To a solution of 10 (10 mg, 0.022 mmol), i-Pr[0339] 2NEt (0.02 mL, 0.12 mmol) in CH2Cl2 (0.1 mL) at 0° C. was added methanesulfonyl chloride (0.004 mL). The mixture was stirred for 3 hrs at 0° C. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with NaSO4, filtered and evaporated. PTLC (aluminua plate, 20% EtOAc in hexane) provided pure 28. Rf=0.11 (aluminum oxide IB-F plate (J. T. Baker), 20% EtOAc in hexane); LRMS [calculated for C30H37FN3O2S (M+1)+] 522, found 522.
  • EXAMPLE 27
  • [0340]
    Figure US20020156092A1-20021024-C00039
  • To a solution of alcohol 13 (20 mg, 0.055 mmol), i-Pr[0341] 2NEt (0.029 mL, 0.16 mmol) in CH2Cl2 (0.2 mL) at 0° C. was added methanesulfonyl chloride (0.0064 mL). The mixture was stirred for 3 hrs at 0° C. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided 29. LRMS [calculated for C25H35N2O3S (M+1)+] 443, found 443.
  • EXAMPLE 28
  • [0342]
    Figure US20020156092A1-20021024-C00040
  • To a solution of 29 (115 mg) in toluene (2 mL) was added aniline (0.077 mL). The mixture was heated at 110° C overnight. After removal of solvent, the crude product was purified by PTLC (alumina plate, 20% EtOAc in hexane) to provide the 30 (65 mg, 57% yield). R[0343] f=0.78 (aluminum oxide IB-F plate (J. T. Baker), 20% EtOAc in hexane); LRMS [calculated for C30H38N3 (M+1)+] 440, found 440.
  • EXAMPLE 29
  • [0344]
    Figure US20020156092A1-20021024-C00041
  • To a solution of 30 (5 mg, 0.011 mmol), i-Pr[0345] 2NEt (0.02 mL) in CH2Cl2 (0.1 mL) at 0° C. was added propionyl chloride (0.01 mL). The mixture was stirred for 12 hrs at room temperature. The mixture was diluted with CH2Cl2 (1 ML), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided pure 31. Rf=0.35 (aluminum oxide IB-F plate (J. T. Baker), 20% EtOAc in hexane); IR (film) 3063, 3023, 2935, 2806, 2862, 1660, 1600, 1499, 1451, 1379, 1355, 1250, 1158, 1110, 1081, 1029, cm−1; LRMS [calculated for C33H42N3O (M+1)+] 496, found 496.
  • EXAMPLE 30
  • [0346]
    Figure US20020156092A1-20021024-C00042
  • To a solution of 30 (5 mg, 0.011 mmol), i-Pr[0347] 2NEt (0.05 mL) in CH2Cl2 (0.1 mL) at 0° C. was added cyclobutanecarbonyl chloride (0.01 mL). The mixture was stirred for 12 hrs at room temperature. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. The two diastereomers 32/33 were separated by PTLC (alumina plate 20% EtOAc in hexane). 32: Rf=0.44, (aluminum oxide IB-F plate (J. T. Baker), 20% EtOAc in hexane); IR (film) 3063, 3027, 2943, 2802, 2866, 1656, 1600, 1495, 1451, 1367, 1266, 1238, 1114, 1081, 1029 cm−1; LRMS [calculated for C35H44N3O (M+1)+] 522, found 522. 33: Rf=0.31, (aluminum oxide IB-F plate (J. T. Baker), 20% EtOAc in hexane); IR (film) 3067, 3031, 2947, 2870, 2810, 1656, 1600, 1499, 1451, 1395, 1371, 1154, 1106, 1029 cm−1; LRMS [calculated for C35H44N3O (M+1)+] 522, found 522. The stereochemistry of 32 and 33 was assigned randomly.
  • EXAMPLE 31
  • [0348]
    Figure US20020156092A1-20021024-C00043
  • To a solution of 30 (15 mg, 0.034 mmol), i-Pr[0349] 2NEt (0.05 mL) in CH2Cl2 (0.1 mL) at 0° C. was added methanesulfonyl chloride (0.01 mL). The mixture was stirred at room temperature overnight. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (alumina plate, 20% EtOAc in hexane) provided pure 34. Rf=0.15, (aluminum oxide IB-F plate (J. T. Baker), 20% EtOAc in hexane); LRMS [calculated for C31H40N3O2S (M+1)+] 518, found 518.
  • EXAMPLE 32
  • [0350]
    Figure US20020156092A1-20021024-C00044
  • To a solution of 23 (52 mg, 0.11 mmol) in 1,2-dichloroethane (2 mL) at 0° C. was added 1-chloroethyl chloroformate (0.012 mL, 0.11 mmol). The mixture was stirred at 0° C. for 20 min and then refluxed at 98° C. for 1.5 hr. After removal of solvent under vacuum, the residue was refluxed in methanol (1 mL) for additional 2-3 hr. Solvent was removed under vacuum. The residue was dissolved in CH[0351] 2Cl2 (4 mL), neutralized with sat. K2CO3 (4 mL). The two layers were separated. Aqueous layer was extracted with CH2Cl2 (3 mL). The combined organic solution was dried (NaSO4) and evaporated to give 35. LRMS [calculated for C25H34N3O (M+1)+] 392, found 392.
  • EXAMPLE 33
  • [0352]
    Figure US20020156092A1-20021024-C00045
  • The mixture of 35 (19.6 mg, 0.05 mmol), (2-bromoethyl)-benzene (0.08 ml, 0.055 mmol), CH[0353] 3CN (0.25 mL), H2O (0.25 mL), K2CO3 (0.02 g, 5.6 mmol) was stirred at 70° C. for 12 hrs. After cool down to room temperature, the mixture was diluted with EtOAc (2 mL) and washed with brine (2 mL), dried with NASO4 and evaporated. The crude product was purified by PTLC (alumina plate, 20% EtOAc in hexane) to give 36. 1H-NMR (300 MHz, CDCl3) 7.40-7.10 (m, 15H), 4.15 (m, 2H), 2.80 (m, 7H), 2.55 (m, 6H), 2.35 (m, 1H), 2.18 (m, 4H), 1,80 (m, 1H), 1.50 (m, 1H), 1.10 (t, 3H) ppm; 13C-NMR (75 MHz, CDCl3) d 175.08, 144.97, 141.04, 140.90, 129.72, 129.00, 128.54, 127.62, 126.18, 126.12, 61.72, 60.80, 59.26, 58.19, 56.37, 54.11, 50.33, 46.43, 38.41, 35.97, 33.72, 28.19, 24.46, 10.03 ppm; IR (film) 3059, 3023, 2933, 2797, 1665, 1600, 1494, 1393, 1257, 1147, 1031, 915 cm−1; LRMS [calculated for C33H42N3O (M+1)+] 496, found 496.
  • EXAMPLE 34
  • [0354]
    Figure US20020156092A1-20021024-C00046
  • The two enantiomers 37 and 38 were obtained by chiral HPLC separation: ChiralPak AD column, 14% EtOH in hexane, 4.0 mL/min, 254 nm; Rt[0355] 1=5.4 min (assumed 37), Rt2=6.92 min (assumed 38).
  • EXAMPLE 35
  • [0356]
    Figure US20020156092A1-20021024-C00047
  • The mixture of 35 (22.6 mg, 0.058 mmol), (2-bromoethyl)-benzene (0.011 ml, 0.07 mmol), CH[0357] 3CN (0.25 mL), H2O (0.25 mL), K2CO3 (0.02 g, 5.6 mmol) was stirred at 70° C. for 12 hrs. After cool down to room temperature, the mixture was diluted with EtOAc (2 mL) and washed with brine (2 mL), dried with Na2SO4 and evaporated. The crude product was purified by PTLC (alumina plate, 20% EtOAc in hexane) to give 39. IR (film) 3063, 3027, 2923, 2854, 2806, 1668, 1596, 1499, 1459, 1403, 1258, 1158, 1033 cm−1; LRMS [calculated for C33H48N3O (M+1)+] 502, found 502.
  • EXAMPLE 36
  • [0358]
    Figure US20020156092A1-20021024-C00048
  • To a solution of 35 (7 mg, 0.018 mmol), i-Pr[0359] 2NEt (0.05 mL) in CH2Cl2 (0.2 mL) at 0° C. was added (CH3CO)2O (0.01 mL). The mixture was stirred at room temperature overnight. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (aluminua plate, 20% EtOAc in hexane) provided pure 40. LRMS [calculated for C27H36N3O2, (M+1)+] 434, found 434.
  • EXAMPLE 37
  • [0360]
    Figure US20020156092A1-20021024-C00049
  • To a solution of 35 (7 mg, 0.018 mmol), i-Pr[0361] 2NEt (0.05 mL) in CH2Cl2 (0.2 mL) at 0° C. was added CH3SO2Cl (0.01 mL). The mixture was stirred at room temperature overnight. The mixture was diluted with CH2Cl2 (1 mL), washed with aqueous NaHCO3 (1 mL), dried with Na2SO4, filtered and evaporated. PTLC (alumina plate 20% EtOAc in hexane) provided pure 41. LRMS [calculated for C26H36N3O2S, (M+1)+] 470, found 470.
  • EXAMPLE 38
  • [0362]
    Figure US20020156092A1-20021024-C00050
  • To a sample of 35 (10 mg) was added 0.5 mL of formic acid (88% purity) and 0.75 mL of formaldehyde (37% wt. in water). The mixture was heated at 78° C. overnight. The mixture was neutralized with saturated aqueous NaHCO[0363] 3, and extracted with CH2Cl2. The combined organics were dried with Na2SO4, and purified by PTLC (alumina oxide plate, 20% EtOAc in hexane) to provide pure 42. LRMS [calculated for C26H35N3O, (M+1)+] 406, found 406.
  • EXAMPLE 39 Radioligand Binding Assays (IC50, μM)
  • The radiobinding assays for human opiate receptor μ, κ and δ are conducted according the procedures outlined by Maguire et al. ([0364] Eur. Pharmacol. 1992, 213, 219). The results are presented below.
    μ κ δ
    Compound (IC50, μM) (IC50, μM) (IC50, μM)
    11 <1 <5
    12 <1 <5
    14 <1
    23 <0.1 <5 <1
    25 <1 <10
    31 <0.01 <5 <1
    32 <0.1 <5 <1
    33 <1 <5 <5
    36 <0.01 <5 <1
    37 <0.01 <1 <1
    38 <0.1 <5 <1
    40 <1
    41 <1
    42 <1
  • EXAMPLE 40 In vivo Analgesia Experiments
  • The tail flick model was conducted according to D'Amour F. E. and Smith D. L. [0365] J. Pharmacol. Exp. Ther. 1941, 72, 74. The results are presented below.
    ED50(mg/kg)
    Compound Tail flick Hot plate
    37 0.05 0.06
    38 0.18 152
  • Incorporation By Reference
  • All of the patents and publications cited herein are hereby incorporated by reference. [0366]
  • Equivalents
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims. [0367]

Claims (36)

We claim:
1. A compound represented by A:
Figure US20020156092A1-20021024-C00051
wherein
Z represents NR, NC(O)R, NS(O)2R, O, S, SO, or SO2;
m is independently for each occurrence 0, 1, 2, 3, or 4;
y is 0, 1, or 2;
R represents H, alkyl, cycloalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl;
R1 represents H, alkyl, cycloalkyl, aryl, or heteroaryl;
R, when a constituent of Z, and R1 may be connected through a covalent bond;
R4 represents independently for each occurrence H, alkyl, cycloalkyl, alkenyl, aryl, heteroaryl, acyl, or sulfonyl;
R5 and R6 are selected independently for each occurrence from the group consisting of H, alkyl, CH2ZR, aryl, heteroaryl, F, OR, and OC(O)R;
a covalent bond may connect R4 and an instance of R5 or R6;
any geminal or vicinal pairs of R5 and R6 may be connected through a covalent bond; and
the stereochemical configuration at any stereocenter of a compound represented by A is R, S, or a mixture of these configurations.
2. The compound of claim 1, wherein Z is NR, NC(O)R, or NS(O)2R.
3. The compound of claim 1, wherein m is independently for each occurrence 0, 1, or 2.
4. The compound of claim 1, wherein y is 1.
5. The compound of claim 1, wherein R1 represents aryl or heteroaryl.
6. The compound of claim 1, wherein R4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl.
7. The compound of claim 1, wherein R4 represents independently for each occurrence aryl.
8. The compound of claim 1, wherein R5 represents independently for each occurrence H.
9. The compound of claim 1, wherein R6 represents independently for each occurrence H.
10. The compound of claim 1, wherein R5 represents independently for each occurrence H; and R6 represents independently for each occurrence H.
11. The compound of claim 1, wherein Z is NR, NC(O)R, or NS(O)2R; and m is independently for each occurrence 0, 1, or 2.
12. The compound of claim 1, wherein Z is NR, NC(O)R, or NS(O)2R; and y is 1.
13. The compound of claim 1, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; and y is 1.
14. The compound of claim 1, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; and R1 represents aryl or heteroaryl.
15. The compound of claim 1, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; and R4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl.
16. The compound of claim 1, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; and R4 represents independently for each occurrence aryl.
17. The compound of claim 1, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; R4 represents independently for each occurrence cycloalkyl, aryl, heteroaryl, acyl, or sulfonyl; R5 represents independently for each occurrence H; and R6 represents independently for each occurrence H.
18. The compound of claim 1, wherein Z is NR, NC(O)R, or NS(O)2R; m is independently for each occurrence 0, 1, or 2; y is 1; R1 represents aryl or heteroaryl; R4 represents independently for each occurrence aryl; R5 represents independently for each occurrence H; and R6 represents independently for each occurrence H.
19. The compound of claim 1, wherein said compound is a single stereoisomer.
20. The compound of claim 1, wherein said compound is a ligand for a cellular receptor.
21. The compound of claim 1, wherein said compound is a ligand for a G-protein-coupled receptor.
22. The compound of claim 1, wherein said compound is an agonist of a G-protein-coupled receptor.
23. The compound of claim 1, wherein said compound is an antagonist of a G-protein-coupled receptor.
24. The compound of claim 1, wherein said compound is a ligand for an opioid receptor.
25. The compound of claim 1, wherein said compound is an agonist of an opioid receptor.
26. The compound of claim 1, wherein said compound is an antagonist of an opioid receptor.
27. The compound of claim 1, wherein said compound is a ligand which is selective for a subclass of opioid receptor.
28. A formulation, comprising a compound of claim 1; and a pharmaceutically acceptable excipient.
29. The formulation of claim 28, wherein said pharmaceutically acceptable excipient is selected from the group consisting of cyclodextrins, liposomes, micelle forming agents, and polymeric carriers.
30. A method of treating pain, drug addiction, or tinnitus in a mammal, comprising the step of:
administering to a mammal suffering from pain, drug addiction, or tinnitus a therapeutically effective amount of a compound of claim 1.
31. The method of claim 30, wherein said mammal is a primate, equine, canine or feline.
32. The method claim 30, wherein said mammal is a human.
33. The method of claim 30, wherein said compound is administered orally.
34. The method of claim 30, wherein said compound is administered intravenously.
35. The method of claim 30, wherein said compound is administered sublingually.
36. The method of claim 30, wherein said compound is administered ocularly.
US10/021,277 2000-12-04 2001-10-30 Diazabicyclo[4.3.0]nonanes, and methods of use thereof Expired - Lifetime US6476046B1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/021,277 US6476046B1 (en) 2000-12-04 2001-10-30 Diazabicyclo[4.3.0]nonanes, and methods of use thereof
AU2002225790A AU2002225790A1 (en) 2000-12-04 2001-11-30 Analgesic diazabicyclo(4.3.0)nonanes
PCT/US2001/044990 WO2002046187A1 (en) 2000-12-04 2001-11-30 Analgesic diazabicyclo[4.3.0]nonanes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US25110800P 2000-12-04 2000-12-04
US10/021,277 US6476046B1 (en) 2000-12-04 2001-10-30 Diazabicyclo[4.3.0]nonanes, and methods of use thereof

Publications (2)

Publication Number Publication Date
US20020156092A1 true US20020156092A1 (en) 2002-10-24
US6476046B1 US6476046B1 (en) 2002-11-05

Family

ID=26694502

Family Applications (1)

Application Number Title Priority Date Filing Date
US10/021,277 Expired - Lifetime US6476046B1 (en) 2000-12-04 2001-10-30 Diazabicyclo[4.3.0]nonanes, and methods of use thereof

Country Status (3)

Country Link
US (1) US6476046B1 (en)
AU (1) AU2002225790A1 (en)
WO (1) WO2002046187A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8182836B2 (en) 2003-04-08 2012-05-22 Elite Laboratories, Inc. Abuse-resistant oral dosage forms and method of use thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6997202B2 (en) * 2002-12-17 2006-02-14 Advanced Technology Materials, Inc. Gas storage and dispensing system for variable conductance dispensing of gas at constant flow rate
TW200606161A (en) * 2004-05-28 2006-02-16 Glaxo Group Ltd Muscarinic acetylcholine receptor antagonists
JP7185633B2 (en) 2017-02-17 2022-12-07 トレベナ・インコーポレイテッド Delta-Opioid Receptor Modulating Compounds Containing 7-Membered Azaheterocycles, Methods of Use and Preparation Thereof
KR20190129867A (en) 2017-02-17 2019-11-20 트레베나, 인코포레이티드. 5-membered aza-heterocycle-containing delta-opioid receptor modulating compounds, and methods of using and preparing the same
CN107383033A (en) * 2017-06-29 2017-11-24 上海药明康德新药开发有限公司 The synthetic method of cis 5 tertbutyloxycarbonyl tetrahydrofuran simultaneously [3,4 c] pyrroles's 3A carboxylate methyl esters

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK124089A (en) 1988-03-16 1989-09-17 Zambeletti Spa L HETEROCYCLIC DERIVATIVES
GB9127041D0 (en) 1991-12-20 1992-02-19 Fujisawa Pharmaceutical Co New use
US6004956A (en) 1992-01-10 1999-12-21 Bayer Aktiengesellschaft Enantiomerically pure 2-oxa-5,8-dizaabicyclo[4.3.0] nonanes and process for their preparation
DE4217846C1 (en) 1992-05-29 1993-12-23 Basf Ag Process for the preparation of 5-dichloroacetyl-3,3,6-trimethyl-9-oxo-1,5-diazabicyclo [4,3,0] nonane
JPH07502758A (en) 1992-10-23 1995-03-23 チバ−ガイギー アクチエンゲゼルシャフト 3-Aryluracil derivatives and their use for controlling weeds
IL117200A0 (en) 1995-02-21 1996-06-18 Schering Ag Diethylenetriamine-pentaacetic acid monoamide derivatives pharmaceutical compostions containing the same and processes for the preparation thereof
NZ322553A (en) 1995-11-23 1998-12-23 British Biotech Pharm Metalloproteinase inhibitors
FR2757158B1 (en) 1996-12-18 1999-04-02 Lipha NOVEL 4- (1-PIPERAZINYL) BENZOIC ACID DERIVATIVES, PROCESS FOR THEIR PREPARATION AND THERAPEUTIC APPLICATIONS
IL137166A0 (en) 1998-02-04 2001-07-24 Banyu Pharma Co Ltd N-acyl cyclic amine derivatives
AR014975A1 (en) 1998-04-08 2001-04-11 Nps Pharma Inc CALCILITICAL COMPOUNDS, A PHARMACEUTICAL COMPOSITION THAT UNDERSTANDS THEM, AND THE USE OF THEM FOR THE MANUFACTURE OF A MEDICINAL PRODUCT
DE19821039A1 (en) 1998-05-11 1999-11-18 Bayer Ag Production of (S,S)-8-benzyl-2,8-diazabicyclo(4.3.0)nonane useful as intermediate for antibiotic moxifloxacin
AU777760B2 (en) 1999-05-25 2004-10-28 Sepracor, Inc. Heterocyclic analgesic compounds and methods of use thereof
DE19927412A1 (en) 1999-06-16 2000-12-21 Bayer Ag Process for the enantiomeric enrichment of cis-8-benzyl-7,9-dioxo-2,8-diazabicyclo [4.3.0] nonane

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8182836B2 (en) 2003-04-08 2012-05-22 Elite Laboratories, Inc. Abuse-resistant oral dosage forms and method of use thereof
US8425933B2 (en) 2003-04-08 2013-04-23 Elite Laboratories, Inc. Abuse-resistant oral dosage forms and method of use thereof
US8703186B2 (en) 2003-04-08 2014-04-22 Elite Laboratories, Inc. Abuse-resistant oral dosage forms and method of use thereof

Also Published As

Publication number Publication date
US6476046B1 (en) 2002-11-05
WO2002046187A1 (en) 2002-06-13
AU2002225790A1 (en) 2002-06-18

Similar Documents

Publication Publication Date Title
US7812035B2 (en) 4-substituted piperidines, and methods of use thereof
AU777760B2 (en) Heterocyclic analgesic compounds and methods of use thereof
US20070225331A1 (en) 4,4-Disubstituted piperidines, and methods of use thereof
US6645980B1 (en) Heterocyclic analgesic compounds and methods of use thereof
US7511141B2 (en) Piperidine-piperazine ligands for neurotransmitter receptors
US6476050B2 (en) 3-substituted piperidines comprising urea functionality, and methods of use thereof
US6635661B2 (en) Heterocyclic analgesic compounds and methods of use thereof
US6703383B2 (en) Antipsychotic sulfonamide-heterocycles, and methods of use thereof
US7446115B2 (en) Peptidomimetic ligands for cellular receptors and ion channels
US6677332B1 (en) Heterocyclic analgesic compounds and methods of use thereof
US20030073681A1 (en) 2-substituted piperidines that are ligands for monoamine receptors and transporters
US7361666B2 (en) Heterocyclic analgesic compounds and methods of use thereof
US6476046B1 (en) Diazabicyclo[4.3.0]nonanes, and methods of use thereof
US20080234247A1 (en) Heterocyclic analgesic compounds and methods of use thereof
US7030122B2 (en) 1,4-disubstituted piperazine ligands for neurotransmitter receptors
WO2002083071A2 (en) Heterocyclic analgesic compounds and methods of use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: SEPRACOR INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WU, XINHE;REEL/FRAME:012722/0129

Effective date: 20020301

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12