US20040009890A1 - Broad spectrum inhibitors - Google Patents

Broad spectrum inhibitors Download PDF

Info

Publication number
US20040009890A1
US20040009890A1 US10/337,699 US33769903A US2004009890A1 US 20040009890 A1 US20040009890 A1 US 20040009890A1 US 33769903 A US33769903 A US 33769903A US 2004009890 A1 US2004009890 A1 US 2004009890A1
Authority
US
United States
Prior art keywords
compound
inhibitors
atoms
target protein
inhibitor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/337,699
Inventor
John Erickson
Michael Eissenstat
Abelardo Silva
Sergei Gulnik
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.)
Sequoia Pharmaceuticals Inc
Original Assignee
Erickson John W.
Michael Eissenstat
Abelardo Silva
Sergei Gulnik
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 Erickson John W., Michael Eissenstat, Abelardo Silva, Sergei Gulnik filed Critical Erickson John W.
Priority to US10/337,699 priority Critical patent/US20040009890A1/en
Publication of US20040009890A1 publication Critical patent/US20040009890A1/en
Assigned to SEQUOIA PHARMACEUTICALS reassignment SEQUOIA PHARMACEUTICALS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EISSENSTAT, MICHAEL, ERICKSON, JOHN, GULNIK, SERGEI, SILVA, ABEIARDO
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/88Lyases (4.)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D275/00Heterocyclic compounds containing 1,2-thiazole or hydrogenated 1,2-thiazole rings
    • C07D275/02Heterocyclic compounds containing 1,2-thiazole or hydrogenated 1,2-thiazole rings not condensed with other rings
    • C07D275/03Heterocyclic compounds containing 1,2-thiazole or hydrogenated 1,2-thiazole rings not condensed with other rings with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/18Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/20Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/503Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from viruses
    • C12N9/506Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from viruses derived from RNA viruses
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/20Allele or variant detection, e.g. single nucleotide polymorphism [SNP] detection
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/50Molecular design, e.g. of drugs
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2299/00Coordinates from 3D structures of peptides, e.g. proteins or enzymes
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B15/00ICT specially adapted for analysing two-dimensional or three-dimensional molecular structures, e.g. structural or functional relations or structure alignment
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B15/00ICT specially adapted for analysing two-dimensional or three-dimensional molecular structures, e.g. structural or functional relations or structure alignment
    • G16B15/20Protein or domain folding
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A90/00Technologies having an indirect contribution to adaptation to climate change
    • Y02A90/10Information and communication technologies [ICT] supporting adaptation to climate change, e.g. for weather forecasting or climate simulation

Definitions

  • the present invention relates to the field of inhibitors and methods for identifying or designing broad spectrum therapeutics for use in the treatment of infectious diseases and cancers, particularly where drug resistance is, or could reasonably predicted to be, an obstacle to successful long term therapy.
  • Drug resistance is a major obstacle to the successful treatment of many cancers and infections, both bacterial and viral.
  • increased resistance of bacterial infections to antibiotic treatment has been extensively documented and has now become a generally recognized medical problem, particularly with nosocomial infections. See, for example, Jones et al., Diagn. Microbiol. Infect. Dis. 31:379 (1998); Murray, Adv. Intern. Med. 42:339 (1997); and Nakae, Microbiologia 13:273 (1997).
  • PIs HIV protease inhibitors
  • NRTIs and NNRTIs nucleoside and non-nucleoside HIV reverse transcriptase inhibitors
  • FIs HIV fusion inhibitors
  • PIs The particular sequence and pattern of mutations selected by PIs is believed to be somewhat drug-specific and often patient-specific, but high level resistance is typified by multiple mutations in the protease gene which give rise to cross-resistance to all of the PIs.
  • Inhibitors with the ability to inhibit mdrHIV strains, and to slow the emergence of drug resistant strains in wild type HIV infections are defined as “resistance-repellent” inhibitors.
  • drugs may be designed which have similar activity against both the wild type and mutant forms of their target. Such drugs minimize the probability of a mutant population emerging by reducing the selective pressure introduced by the drug when used to treat wild type infections. Such drugs also can be used to treat mutant infections and can be used for salvage therapy.
  • the NIH and CDC have identified a number of High Priority pathogens based on their likelihood of causing widespread contagious disease and/or death to the general population.
  • Research on methods of protection against potential agents of bioterrorism has been a priority for several years at the NIH.
  • a recent analysis suggested the existence of ongoing offensive biological weapons programs in at least 13 countries (Inglesby, T. V., et al., JAMA, 287:2236, (2002)).
  • Penicillin, doxycycline and ciprofloxacin are currently approved by the FDA for the treatment of inhalation anthrax infections. However, it was advised that antibiotic resistance to penicillin- and tetracycline-class antibiotics should be assumed following a terrorist attack (Inglesby, T. V., et al., JAMA, 281:1735-45 (1999)). Moreover, in vitro selection of a B. anthracis strain that is resistant to ofloxacin (a fluoroquinilone closely related to ciprofloxacin) has been reported (Choe, C. H., et al., Antimicrob. Agents.
  • Another group A pathogen, Y. pestis is the causative agent of plague. If 50 kg of Y. pestis were released as an aerosol over a city of 5 million, pneumonic plague would afflict an estimated 150,000 individuals and result in 36,000 deaths (Inglesby, T. V., et al., JAMA, 283: 2281, (2000)). Streptomycin, tetracycline and doxycycline are the FDA-approved treatment for plague. Wide spread use of these antibiotics in the US raises concerns about possible resistance. A US-licensed, formaldehyde-killed whole bacilli vaccine was discontinued by its manufacturers in 1999 and is no longer available.
  • C. jejuni and V. cholerae are category B pathogens which can present a significant threat to the safety of food and water supplies.
  • C. jejuni infections are one of the most commonly identified causes of acute bacterial gastroenteritis worldwide and area frequent cause of Traveler's diarrhea (Allos, B. M., Clin Infect Dis, 32:1201 (2001)).
  • the CDC estimates that 2.4 million cases of Campylobacter infection occur in the United States each year, affecting almost 1% of the entire population. In the past few years, a rapidly increasing proportion of Campylobacter strains all over the world have been found to be fluoroquinolone-resistant.
  • V. cholerae a causative agent of cholera, is responsible for 120,000 deaths annually (Faruque, S. M., et al., Microbiol Mol Biol Rev, 62:1301 (1998)) and is characterized by a rapidly changing pattern of antibiotic resistance.
  • TB is one of the most common and devastating infectious diseases known to man. An estimated one third of the global population is infected with Mycobacteria tuberculosis. Eight million people develop an active infection and 2 million victims die yearly (Dye, C., et al., JAMA, 282:677 (1999.)).
  • a combination of four drugs is recommended for TB treatment: isoniazid, rifampicin, pyrazinamide and ethambutole.
  • the treatment course lasts 6 months.
  • Such a multidrug combination together with the lengthy duration of treatment is prone to side-effects and adherence problems, which in turn can often lead to the development of drug resistance.
  • drugs may be designed which have similar activity against both the wild type and variant forms of their target. Such drugs should minimize the probability of the emergence of mutant populations by reducing the selective pressure introduced by the drug when used to treat wild type infections. Such drugs also can be used to treat mutant infections and can be used for salvage therapy.
  • drugs may be designed which have similar activity against various isotypes of a homologous target. Such drugs can be used to treat multiple species of pathogenic microorganisms since they will be active against the target of each species.
  • drugs can be designed that combine the properties and the uses of both of the above approaches.
  • the invention features a method for the structure-based design of a drug that can act as an inhibitor of at least two different biological entities, the method comprising the steps of: (a) providing at least one structure of a wild type target protein or an inhibitor-wild type target protein complex; (b) providing at least one structure of a variant target protein or an inhibitor-variant target protein complex; (c) comparing at least one structure from step (a) with at least one structure from step (b) to determine whether there exists a common three-dimensionally conserved substructure comprising the atomic coordinates of the structurally conserved atoms of the inhibitors and structurally conserved atoms of the target proteins; and (d) if a conserved substructure exists, using the atomic coordinates of the conserved substructure to select a compound having atoms matching those of the structurally conserved atoms of the inhibitors, wherein the selection of the compound is performed using computer modeling.
  • the invention also features a method for the structure-based drug design of a broad spectrum compound, the method comprising the steps of: (a) providing at least one structure of a wild type target protein or an inhibitor-wild type target protein complex; (b) providing at least one structure of a variant target protein or an inhibitor-variant target protein complex; (c) comparing at least one structure from step (a) with at least one structure from step (b) to determine whether there exists a common three-dimensionally conserved substructure comprising the atomic coordinates of the structurally conserved atoms of the target proteins or a common three-dimensionally conserved substructure comprising the atomic coordinates of the structurally conserved atoms of the inhibitors and structurally conserved atoms of the target proteins; and (d) if a conserved substructure exists, using the atomic coordinates of the conserved substructure to select a compound having atoms matching those of the structurally conserved atoms of the inhibitors or to design a compound that binds to the target protein, wherein the
  • the above method further comprises the steps of: (e) comparing at least one structure from step (a) with at least one structure from step (b) to determine whether there exists a three-dimensionally non-conserved substructure comprising the atomic coordinates of the structurally non-conserved atoms of the inhibitors and structurally non-conserved atoms of the target proteins; and (f) if a non-conserved substructure exists, using the atomic coordinates of the non-conserved substructure to reject a compound having atoms matching those of the structurally non-conserved atoms of the inhibitors, wherein the rejection of the compound is performed in conjunction with computer modeling.
  • step c At least two, four, six, or eight structures from step b can be used in step c.
  • the methods can be applied using several structures, including at least two, four, six, or eight variant forms of the target protein.
  • the inhibitors used in the inhibitor-wild type target protein complex and the inhibitor-variant target protein complex can be the same or different.
  • the inhibitors can be selected from competitive or noncompetitive inhibitors.
  • the inhibitors can be selected from reversible, or irreversible inhibitors.
  • the variant target protein can be a homologous protein or a mutant protein.
  • the structures can be selected from crystal structures, NMR structures, computer models, any acceptable experimental, theoretical or computational method of deriving a three-dimensional representation of a structure, or a combination thereof.
  • Target proteins for use in the present invention include any therapeutically relevant protein.
  • the target protein can be a viral, bacterial, protozoan, or fungal protein.
  • the target protein is one that is expressed in a neoplasm.
  • the target protein can be an enzyme, a receptor, a structural protein, a component of a macromolecular complex, a component of a metabolic pathway, or an assembly of biological molecules.
  • the target protein is necessary for the survival of the replicating biological entity.
  • the target protein can be an enzyme selected from the group consisting of reverse transcriptases, proteases, DNA and RNA polymerases, methylases, oxidases, esterases, acyl transferases, helicases, topoisomerases, and kinases.
  • the target protein can be a component of a metabolic pathway, such as the shikimate pathway. Desirable target proteins include HIV protease or 3-dehydroquinate dehydratase, among others.
  • suitable inhibitors for use in the methods of the invention include those selected from the group consisting of indinavir, nelfinavir, ritonavir, saquinavir, amprenavir, lopinavir, and UIC-94003.
  • a broad spectrum protease inhibitor can be designed using the susbstructure of structurally conserved atoms described by the atomic coordinates in Table 8, which includes the structurally conserved atoms of the inhibitor and structurally conserved atoms of the protease.
  • a broad spectrum protease inhibitor can also be designed using the structurally conserved atoms of the inhibitor alone. These are described by the atomic coordinates in Table 8.
  • a broad spectrum 3-dehydroquinate dehydratase inhibitor can be designed using the susbstructure of structurally conserved atoms described by the atomic coordinates in Table 12, which includes the structurally conserved atoms of the 3-dehydroquinate dehydratase.
  • a broad spectrum 3-dehydroquinate dehydratase inhibitor can also be designed using the structurally conserved atoms of the inhibitor alone. These are described by the atomic coordinates in Table 12.
  • the invention also features compounds having a chemical structure selected using any of the methods above.
  • Such compounds are broad spectrum inhibitors and have broad spectrum activity against replicating biological entities expressing a particular target protein.
  • the target protein is expressed by a microbe or a neoplasm, the compound will have broad spectrum activity against the microbe or neoplasm, respectively.
  • the invention features a compound having broad spectrum activity against HIV protease wherein the compound has a chemical structure selected using the methods above, including those methods utilizing the atomic coordinates of Table 8.
  • the invention features a compound having broad spectrum activity against 3-dehydroquinate dehydratase wherein the compound has a chemical structure selected using the methods above, including those methods utilizing the atomic coordinates of Table 12.
  • the compounds of the invention exclude bis-THF compounds (e.g., analogs of compounds 1 and 3) as described in J. Med. Chem. 39:3278-3290 (1996) (compounds 49-52 and 58-60), Bioorg. Med. Chem. Lett. 8:979-982 (1998), WO99/65870, U.S. Pat. No. 6,319,946, WO02/08657, WO02/092595, WO99/67417, EP00/9917, and WO00/76961; and also exclude fused ring THF structures as described in Bioorg. Med. Chem. Lett. 8:687-690 (1998) and U.S. Pat. No. 5,990,155.
  • broad spectrum activity can be measured by the ratio of the inhibitory concentrations of the broad spectrum inhibitor for the variant and wild type biological entities (IC 50, variant /IC 50, wild type ).
  • IC 50, variant /IC 50, wild type ratio for a broad spectrum inhibitor is less than 100, 80, 60, 40, 30, 20, 10, 8, 6, or, most desirably, less than 3.
  • a broad spectrum inhibitor can be active against several different mutant biological entities. Desirably, the inhibitor will have broad spectrum activity against at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 mutant biological entities.
  • a broad spectrum inhibitor can also be active against different organisms or neoplastic cell types expressing homologous target proteins that possess sufficient structural similarity. Desirably, the inhibitor will have broad spectrum activity against at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 different organisms or neoplastic cell types expressing homologous target proteins.
  • the invention also features a pharmaceutical composition that includes a broad spectrum inhibitor described herein in any pharmaceutically acceptable form, including isomers such as diastereomers and enantiomers, salts, solvates, and polymorphs thereof.
  • the composition can include an inhibitor of the invention along with a pharmaceutically acceptable carrier or diluent.
  • the invention also features methods of treating disease in a patient in need thereof, which includes the administration of a pharmaceutical composition of the invention to the patient in an amount sufficient to treat the disease.
  • the pharmaceutical composition includes any broad spectrum inhibitor described herein. Such broad spectrum inhibitors have broad spectrum activity against replicating biological entities expressing a particular target protein. Thus, if the target protein is expressed by a microbe or a neoplasm, the disease to be treated will be a microbial infection or neoplasm, respectively.
  • the invention features a method of treating an HIV infection in a patient in need thereof, the method including the step of administering to the patient a pharmaceutical composition including a broad spectrum protease inhibitor described herein in amounts effective to treat the HIV infection.
  • the invention features a method of treating a bacterial infection in a patient in need thereof, the method including the step of administering to the patient a pharmaceutical composition including a broad spectrum 3-dehydroquinate dehydratase inhibitor described herein in amounts effective to treat the bacterial infection.
  • the bacterial infection to be treated using the above method can be caused by a bacterium selected from the group consisting of C. jejuni, V. cholerae, Y. pestis, B. anthracis, P. putidas, and M. tuberculosis .
  • this method can be used to treat infections by any microbe the utilizes 3-dehydroquinate dehydratase.
  • the invention also features the use of a pharmaceutical composition described herein in the manufacture of a medicament for the treatment of a disease.
  • the pharmaceutical composition includes any broad spectrum inhibitor described herein. Such broad spectrum inhibitors and have broad spectrum activity against replicating biological entities expressing a particular target protein.
  • the target protein is HIV protease or 3-dehydroquinate dehydratase
  • the disease to be treated will be an HIV infection or bacterial infection, respectively.
  • replicating biological entity includes, for example, bacteria, fungi, yeasts, viruses, protozoa, prions and neoplasms
  • Neoplasms include, for example, carcinomas of the bladder, breast, colon, kidney, liver, lung, head and neck, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, or skin; a hematopoietic tumor of lymphoid lineage; a hematopoietic tumor of myeloid lineage; a tumor of mesenchymal origin; a tumor of the central or peripheral nervous system; melanoma; seminoma; teratocarcinoma; osteosarcoma; thyroid follicular cancer; and Kaposi's sarcoma.
  • Hematopoietic tumors of lymphoid lineage can be leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma.
  • wild type target protein is meant a protein obtained from a replicating biological entity that has not been subjected to drug selection pressure, and could include polymorphisms or isoforms thereof.
  • a replicating biological entity that expresses wild type target protein is referred to herein as a wild type biological entity.
  • variant target protein is meant a mutant target protein or a homologous target protein.
  • a replicating biological entity that expresses variant target protein is referred to herein as a variant biological entity.
  • mutant target protein is meant a target protein that contains one or more amino acid substitutions with respect to the wild type target protein, including proteins from the same organism that have evolved under drug selection pressure. In general, mutant target proteins will have one or more amino acid substitutions and should be readily identified as related to the cognate wild type protein using standard sequence comparison methods.
  • a replicating biological entity that expresses mutant target protein is referred to herein as a mutant biological entity.
  • homologous target protein is meant a variant target protein that is expressed in a different species or neoplastic cell type than the wild type target protein, but has the same, or similar, function.
  • structurally conserved target substructure and by “structurally” or “three-dimensionally conserved substructure” as applied to target proteins, is meant the regions of the target protein structure which are not significantly affected by amino acid mutations or substitutions. Such regions can be defined using standard methods of comparative analysis of three-dimensional structures of proteins, such as superposition analysis, for example. In the case of HIV protease, these regions were identified using a pair wise superposition analysis of wild type and mutant protease structures complexed with inhibitors. The superposition of structures can be performed using the iterative procedure described herein.
  • DHQase these regions were identified using a pair wise superposition analysis of wild type and homologous DHQase structures from different bacterial species with and without inhibitors. It is apparent that the overall compositions of structurally conserved target substructures will likely differ for different, non-homologous target proteins, especially when the frequency of amino acid substitutions in high. However, a quantitative definition can be derived from the superposition analysis, which provides both the identities and the positions of the atoms that comprise these substructures.
  • the regions that comprise structurally conserved target substructures contain atoms whose superimposed pairs have three-dimensional atomic coordinates that match to within a distance of 1 ⁇ , 0.6 ⁇ , 0.4 ⁇ , or 0.2 ⁇ .
  • broad spectrum inhibitor is meant a compound having broad spectrum activity, i.e., an inhibitor that is active against two different-biological entities, e.g., both a wild type biological entity and one or more variants of that biological entity.
  • broad spectrum activity can be described by the inhibitor's action against a particular target protein (e.g., broad spectrum activity against protease) or a particular target organism (e.g., broad spectrum activity against HIV).
  • target protein e.g., broad spectrum activity against protease
  • target organism e.g., broad spectrum activity against HIV
  • Broad spectrum inhibitors will have medically insignificant interactions with non-conserved regions. Broad spectrum inhibitors can be useful for the treatment and/or prevention of infectious diseases caused by multiple infectious agents, as well as for decreasing the development of drug-resistance by these organisms.
  • treating refers to administering a pharmaceutical composition for prophylactic and/or therapeutic purposes.
  • To “prevent disease” refers to prophylactic treatment of a patient who is not yet ill, but who is susceptible to, or otherwise at risk of, a particular disease.
  • To “treat disease” or use for “therapeutic treatment” refers to administering treatment to a patient already suffering from a disease to ameliorate the disease and improve the patient's condition.
  • treating is the administration to a patient either for therapeutic or prophylactic purposes.
  • microbial infection refers to the invasion of the host patient by pathogenic microbes (e.g., bacteria, fungi, yeasts, viruses, protozoa). This includes the excessive growth of microbes that are normally present in or on the body of a patient. More generally, a microbial infection can be any situation in which the presence of a microbial population(s) is damaging to a host patient. Thus, a patient is “suffering” from a microbial infection when excessive numbers of a microbial population are present in or on a patient's body, or when-the presence of a microbial population(s) is damaging the cells or other tissue of a patient.
  • pathogenic microbes e.g., bacteria, fungi, yeasts, viruses, protozoa.
  • microbes includes, for example, bacteria, fungi, yeasts, viruses and protozoa.
  • administration or “administering” refers to a method of giving a dosage of a pharmaceutical composition to a patient, where the method is, e.g., topical, oral, intravenous, intraperitoneal, or intramuscular.
  • the preferred method of administration can vary depending on various factors, e.g., the components of the pharmaceutical composition, site of the potential or actual disease and severity of disease.
  • patient includes humans, cattle, pigs, sheep, horses, dogs, and cats, and also includes other vertebrate, most preferably, mammalian species.
  • atomic coordinates are provided, or otherwise referred to, these coordinates define a three dimensional structure. That such a structure may be defined by more than one different coordinate system, e.g., by translation or rotation of the coordinates, does not change the relative positions of the atoms in the structure. Accordingly, any reference to atomic coordinates herein is intended to include any equivalent three dimensional structure defined by the coordinates.
  • computer modeling is meant the use of a computer to visualize or compute a compound, a portion of a compound, a target protein, a portion of a target protein, a complex between a compound and a target protein, or a portion of a complex between a compound and a target protein.
  • FIG. 1 is a table depicting the structures of compounds 1-7, gt33, and qxa.
  • FIG. 2 illustrates the amino acid alignment of type II DHQases. Fully conserved residues are framed. Catalytically important amino acids are marked by stars. Arrows denote amino acids that make hydrogen bonds and ionic interactions in the structure of M. tuberculosis DHQase complexed with the inhibitor, 3-dehydroquinic acid oxime.
  • FIG. 3 illustrates the key interactions of the substrate-based inhibitor, DHQO, with the active site residues for the Type II DHQase from M. tuberculosis.
  • the methods of the invention entail the design of compounds having a particular structure. The methods rely upon the use of structural information to arrive at these compounds.
  • the structural data define a three dimensional array-of the important contact atoms in an inhibitor that bind to the target protein in a fashion that results in broad spectrum activity against biological entities expressing variants of the target protein.
  • Atomic structural coordinates can be selected from crystal structures, NMR structures, computer models, any acceptable experimental, theoretical or computational method of deriving a three-dimensional representation of a structure, or a combination thereof.
  • Atomic coordinates for use in the methods of the invention can be obtained from publicly available sources, e.g. from the Protein Data Bank, or obtained using known experimental or computational methods.
  • Atomic structural coordinates for use in the methods of the invention include crystal structures of HIV protease/inhibitor complexes derived from wild type and drug-resistant mutant proteases, and of DHQase and DHQase inhibitor complexes derived from two or more bacterial species, among others.
  • the methods of the invention are applied using the coordinates of wild type HIV protease complexed with amprenavir, wild type HIV protease complexed with UIC-94003, and V82F/184V mutant HIV protease complexed with UIC-94003.
  • the methods of the invention are applied using the coordinates of wild type DHQase from M.
  • the coordinates of other representative structures of HIV protease and DHQase should be useful for performing the methods of the present invention.
  • conserved substructures can be identified for target proteins, for target protein-inhibitor complexes, and/or for inhibitors, depending on the nature of the structures that are used in the comparative superposition analysis.
  • at least one structure of a wild type target protein is compared to at least one structure of a mutant or homologous target protein to determine whether a common three-dimensionally conserved substructure is present among the wild type protein and the mutant or homologous proteins, respectively.
  • at least one structure of an inhibitor-wild type target protein complex and at least one structure of an inhibitor-mutant target protein complex are compared to determine whether a common three-dimensionally conserved substructure is present among the mutant and wild type complexes.
  • At least one structure of an inhibitor-wild type target protein complex and at least one structure of a mutant or homologous target protein without inhibitor are compared to determine whether a common three-dimensionally conserved substructure is present among the respective mutant or homologous protein and the wild type complexes. Variations of the approached described above can also be used. In each case, such a comparison can be made by means of (a) an overall superposition of the atoms of the protein structures; and, where feasible, (b) a study of the distances from atoms of the inhibitors to atoms of the protein. This analysis requires three-dimensional atomic coordinates of the protein structures and of the bound inhibitor.
  • the superposition of the protein structures can be performed in a two step process: 1) the distance between all pairs of corresponding Cá atoms (Cá atom of residue number 1 in one protein to Cá atom of residue number 1 in the second protein; Cá atom of residue number 2 in one protein to Cá atom of residue number 2 in the second protein; and so on) of the polypeptide chains is minimized by means of a least-square algorithm; 2) the superposition is refined by minimizing, in an iterative process, the distances between corresponding Cá atoms that are closer than a given distance (0.25 A for example), thus eliminating regions of the structures having large conformational differences to compute the superposition parameters. Furthermore, where a partial structure is provided (e.g., from NMR data) the available coordinates are superimposed.
  • the conserved substructure identifies the relevant portion of the target protein that is the active site, or binding region, defined by that part of the target protein interacting with inhibitor. Important interactions between the target protein and inhibitor are identified by mapping the contacts between the two. Structurally conserved regions of the target protein not near the binding site are generally not relevant to the design of the broad spectrum inhibitor. Accordingly, the selection of the meaningful substructure is identified using the above mentioned contacts.
  • the coordinates of the conserved inhibitor substructure are used to design an inhibitor having atoms matching those of the three-dimensionally structurally conserved atoms of the inhibitors.
  • the result is an inhibitor for which IC 50, variant and IC50, wild type are similar, minimizing the selective pressure introduced by the drug.
  • the methods of the invention can employ computer-based methods for designing broad spectrum inhibitors. These computer-based methods fall into two broad classes: database methods and de novo design methods.
  • database methods the compound of interest is compared to all compounds present in a database of chemical structures and compounds whose structure is in some way similar to the compound of interest are identified.
  • the structures in the database are based on either experimental data, generated by NMR or x-ray crystallography, or modeled three-dimensional structures based on two-dimensional (i.e., sequence) data.
  • de novo design methods models of compounds whose structure is in some way similar to the compound of interest are generated by a computer program using information derived from known structures, e.g., data generated by x-ray crystallography and/or theoretical rules.
  • Such design methods can build a compound having a desired structure in either an atom-by-atom manner or by assembling stored small molecular fragments.
  • the success of both database and de novo methods in identifying compounds having the desired activity depends on the identification of the functionally relevant portion of the compound of interest.
  • the functionally relevant portion of the compound, the pharmacophore is defined by the structurally conserved substructure.
  • a pharmacophore then is an arrangement of structural features and functional groups important for obtaining an inhibitor having broad spectrum activity.
  • Programs suitable for generating predicted three-dimensional structures from two-dimensional data include: Concord (Tripos Associated, St. Louis, Mo.), 3-D Builder (Chemical Design Ltd., Oxford, U.K.), Catalyst (Bio-CAD Corp., Mountain View, Calif.), and Daylight (Abbott Laboratories, Abbott Park, Ill.).
  • Programs suitable for searching three-dimensional databases to identify molecules bearing a desired pharmacophore include: MACCS-3D and ISIS/3D (Molecular Design Ltd., San Leandro, Calif.), ChemDBS-3D (Chemical Design Ltd., Oxford, U.K.), and Sybyl/3DB Unity (Tripos Associates, St. Louis, Mo.).
  • Programs suitable for pharmacophore selection and design include: DISCO (Abbott Laboratories, Abbott Park, Ill.), Catalyst (Bio-CAD Corp., Mountain View, Calif.), and ChemDBS-3D (Chemical Design Ltd., Oxford, U.K.).
  • De novo design programs include Ludi (Biosym Technologies Inc., San Diego, Calif.) and Aladdin (Daylight Chemical Information Systems, Irvine Calif.).
  • One skilled in the art may use one of several methods to screen chemical entities for their ability to match the conserved substructure. This process may begin by visual inspection of, for example, the active site on the computer screen based on the atomic coordinates for the target protein. Docking may be accomplished using software such as Quanta and Sybyl, followed by energy minimization and molecular dynamics with standard molecular mechanics forcefields, such as CHARMM and AMBER.
  • Specialized computer programs may also assist in the process of selecting chemical entities. These include:
  • GRID (Goodford, P. J., “A Computational Procedure for Determining Energetically Favorable Binding Sites on Biologically Important Macromolecules,” J. Med. Chem., 28:849 (1985)). GRID is available from Oxford University, Oxford, UK.
  • MCSS (Miranker, A. and M. Karplus, “Functionality Maps of Binding Sites: A Multiple Copy Simultaneous Search Method.” Proteins: Structure, Function, and Genetics, 11:29 (1991)). MCSS is available from Molecular Simulations, Burlington, Mass.
  • DOCK (Kuntz, L. D. et al., “A Geometric Approach to Macromolecule-Ligand Interactions,” J. Mol. Biol., 161:269 (1982)). DOCK is available from University of California, San Francisco, Calif.
  • the conserved atoms of the inhibitor can be selected for assembly into a single inhibitor. Assembly may be proceed by visual inspection of the relationship of the fragments to each other on the three-dimensional image displayed on a computer screen in relation to the structure coordinates of the target protein. This may be followed by manual model building using software such as Quanta or Sybyl.
  • CAVEAT Bartlett, P. A. et al, “CAVEAT: A Program to Facilitate the Structure-Derived Design of Biologically Active Molecules”. In “Molecular Recognition in Chemical and Biological Problems,” Special Pub., Royal Chem. Soc., 78:182 (1989)). CAVEAT is available from the University of Calif., Berkeley, Calif.
  • 3D Database systems such as MACCS-3D (MDL Information Systems, San Leandro, Calif.). This area is reviewed in Martin, Y. C., “3D Database Searching in Drug Design,” J. Med. Chem., 35:2145 (1992)).
  • substitutions may then be made in some of its atoms or side groups in order to improve or modify its binding properties.
  • initial substitutions are conservative, i.e., the replacement group will have approximately the same size, shape, hydrophobicity and charge as the original group. It should, of course, be understood that components known in the art to alter conformation should be avoided.
  • inhibitors designed using the methods of the invention can be tested for broad spectrum activity using any of the to in vitro and/or in vivo methods described below, among others.
  • Broad spectrum inhibitors match the pharmacophore defined by the structurally conserved substructure.
  • the pharmacophore is the arrangement of structural features and functional groups important for obtaining an inhibitor having broad spectrum activity.
  • This pharmacophore is derived using structural data for known inhibitors complexed to a target protein. Accordingly, broad spectrum inhibitors will often be structurally related to known compounds lacking broad spectrum activity, but useful in the design of broad spectrum inhibitors using the methods disclosed herein. These known inhibitors serve as lead compounds for both the design and synthesis of a broad spectrum inhibitor. Using the synthetic methods for making the lead compounds and standard synthetic methods as described by, for example, J. March, Advanced Organic Chemistry: Reactions, Mechanisms and Structure,” John Wiley & Sons, Inc., 1992; T. W.
  • the lead compounds bear varied functional groups which are present in the pharmacophore, including hydrogen-bond donors, hydrogen-bond acceptors, ionic moieties, polar moieties, hydrophobic moieties, aromatic centers, and electron-donors and acceptors. These are linked by a structural scaffold which imparts the appropriate a three dimension arrangement of the functional groups.
  • One hydrogen-bond donor may be substituted by another.
  • a good hydrogen bond donor has an H atom bonded to a very electronegative atom (e.g., O—H or N—H).
  • examples of hydrogen-bond donors include alcohols, carboxylic acids, oximes, and amides, among others.
  • one hydrogen-bond acceptor may be substituted by another.
  • a good hydrogen bond acceptor has an electronegative element with lone pairs (e.g., O, N, or F).
  • hydrogen bond acceptors include water, halogen atoms, alcohols, amines, carbonyls, ethers, and amides, among others. It may also be desirable to alter the distance between functional groups in a lead compound. This is achieved by employing synthetic methods analogous to those used to prepare the lead compound, but replacing the scaffold with a structurally related scaffold that provides the desired distance (e.g., a scaffold that incorporates more or fewer atoms linking the relevant functional groups). In some instances it may also be desirable to alter the stereochemistry in a lead compound. This can be accomplished by employing racemic starting materials, or by employing reaction conditions that result in racemization of the relevant chiral center, followed by separation of the enantiomeric or diastereomeric mixture.
  • Inhibitors designed using the methods disclosed herein may be further assayed, using standard in vitro models or animal models, to evaluate therapeutic activity and toxicity. These assays are described in the literature and are familiar to those skilled in the art. These include but are not limited to assays for monitoring or measuring efficacy against HIV, bacteria, and neoplasms.
  • IC 50 IC 50 's of a broad spectrum inhibitor described herein.
  • the IC 50 value is determined by plotting percent activity versus inhibitor concentration in the assay and identifying the concentration at which 50% of the activity (e.g., growth, enzymatic activity, protein production, etc.) remains.
  • Inhibitors can be tested for antimicrobial activity against a panel of organisms according to standard procedures described by the National Committee for Clinical Laboratory Standards (NCCLS document 7-A3, Vol. 13, No. 25, 1993/NCCLS document M27-P, Vol. 12, No. 25, 1992).
  • Inhibitors can be dissolved (0.1 ⁇ g/ml-500 ⁇ g/ml) in microbial growth media, diluted, and added to wells of a microtiter plate containing bacteria or fungal cells in a final volume of an appropriate media (Mueller-Hinton Broth; Haemophilus Test Media; Mueller-Hinton Broth+5% Sheep Blood; or RPMI 1690). Typically, the plates are incubated overnight at an appropriate temperature (30° C. to 37° C.) and optical densities (measure of cell growth) are measured using a commercial plate reader.
  • IC 50 's for broad spectrum protease inhibitors can be measured against wild type HIV and clinically isolated mutant HIV isolates, utilizing the PHA-PBMC exposed to HIV-1 (50 TCID 50 dose/X0 6 PBMC) as target cells and using the inhibition of p24 Gag protein production as an endpoint.
  • the amounts of p24 antigen produced by the cells can be determined on day 7 in culture using a commercially available radioimmunoassay kit. Drug concentrations resulting in 50% inhibition (IC 50 's) of p24 antigen production can be determined by comparison with the p24 production level in drug-free control cell cultures.
  • the invention features a method of identifying a compound having broad spectrum activity.
  • Broad spectrum inhibitors of the present invention may be administered by any appropriate route for treatment or prevention of a disease or condition associated with a bacterial infection, viral infection, or neoplastic disorder, among others. These may be administered to humans, domestic pets, livestock, or other animals with a pharmaceutically acceptable diluent, carrier, or excipient, in unit dosage form. Administration may be topical, parenteral, intravenous, intra-arterial, subcutaneous, intramuscular, intracranial, intraorbital, ophthalmic, intraventricular, intracapsular, intraspinal, intracistemal, intraperitoneal, intranasal, aerosol, by suppositories, or oral administration.
  • Therapeutic formulations may be in the form of liquid solutions or suspensions; for oral administration, formulations may be in the form of tablets or capsules; and for intranasal formulations, in the form of powders, nasal drops, or aerosols.
  • Formulations for parenteral administration may, for example, contain excipients, sterile water, or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated napthalenes.
  • Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds.
  • Nanoparticulate formulations may be used to control the biodistribution of the compounds.
  • Other potentially useful parenteral delivery systems include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
  • Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel.
  • concentration of the broad spectrum inhibitor in the formulation will vary depending upon a number of factors, including the dosage of the drug to be administered, and the route of administration.
  • the broad spectrum inhibitor may be optionally administered as a pharmaceutically acceptable salt, such as a non-toxic acid addition salts or metal complexes that are commonly used in the pharmaceutical industry.
  • acid addition salts include organic acids such as acetic, lactic, pamoic, maleic, citric, malic, ascorbic, succinic, benzoic, palmitic, suberic, salicylic, tartaric, methanesulfonic, toluenesulfonic, or trifluoroacetic acids or the like; polymeric acids such as tannic acid, carboxymethyl cellulose, or the like; and inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid phosphoric acid, or the like.
  • Metal complexes include zinc, iron, and calcium, among others.
  • the broad spectrum inhibitor has (i) a narrow therapeutic index (e.g., the difference between the plasma concentration leading to harmful side effects or toxic reactions and the plasma concentration leading to a therapeutic effect is small; generally, the therapeutic index, TI, is defined as the ratio of median lethal dose (LD 50 ) to median effective dose (ED 50 )); (ii) a narrow absorption window in the gastro-intestinal tract; or (iii) a short biological half-life, so that frequent dosing during a day is required in order to sustain the plasma level at a therapeutic level.
  • a narrow therapeutic index e.g., the difference between the plasma concentration leading to harmful side effects or toxic reactions and the plasma concentration leading to a therapeutic effect is small
  • the therapeutic index, TI is defined as the ratio of median lethal dose (LD 50 ) to median effective dose (ED 50 )
  • LD 50 median lethal dose
  • ED 50 median effective dose
  • a narrow absorption window in the gastro-intestinal tract or (iii) a short biological half-life,
  • controlled release can be obtained by the appropriate selection of formulation parameters and ingredients, including, e.g., appropriate controlled release compositions and coatings. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, nanoparticles, patches, and liposomes.
  • Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients.
  • excipients may be, for example, inert diluents or fillers (e.g., sucrose and sorbitol), lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc).
  • Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium.
  • compositions of broad spectrum inhibitor described herein include isomers such as diastereomers and enantiomers, mixtures of isomers, including racemic mixtures, salts, solvates, and polymorphs thereof.
  • the formulations can be administered to human patients in therapeutically effective amounts.
  • the broad spectrum inhibitor is an antimicrobial drug
  • an amount is administered which prevents, stabilizes, eliminates, or reduces a microbial infection.
  • Typical dose ranges are from about 0.01 ⁇ g/kg to about 2 mg/kg of body weight per day.
  • the exemplary dosage of drug to be administered is likely to depend on such variables as the type and extent of the disorder, the overall health status of the particular patient, the formulation of the compound excipients, and its route of administration. Standard clinical trials maybe used to optimize the dose and dosing frequency for any particular broad spectrum inhibitor.
  • This example illustrates the method by which experimentally-determined crystal structures of the same inhibitor in complex with wild type and mutant species of HIV protease can be compared and analyzed for the existence of a three-dimensionally conserved substructure.
  • the structures of wild type HIV-1 protease and a mutant, V82F/184V, HIV-1 protease, both in complexes with the inhibitor shown in FIG. 1 were determined using conventional x-ray crystallography techniques.
  • the structures were analyzed by means of (a) an overall superposition of the atoms of the protein structures; and, (b) a study of the distances from atoms of the inhibitors to atoms of the protein. This analysis requires three dimensional atomic coordinates of the protein structures and of the bound inhibitor.
  • the superposition of the protein structures was performed in a two step process: 1) the distance between all pairs of corresponding Cá atoms (Cá atom of residue number 1 in one protein to Cá atom of residue number 1 in the second protein; Cá atom of residue number 2 in one protein to Cá atom of residue number 2 in the second protein; and so on) of the polypeptide chains is minimized by means of a least-square algorithm; 2) the superposition is refined by minimizing, in an iterative process, the distances between corresponding Cá atoms that are closer than a given distance (0.25 ⁇ in this example), thus eliminating regions of the structures having large conformational differences to compute the superposition parameters.
  • Table 4 shows that the 184V, V82F mutations induce structural changes relative to the wild type structure in some parts of the enzyme, but that other regions are less affected.
  • the regions of the protein structure which are not significantly affected by the amino acid mutations are defined as structurally conserved regions.
  • the mutations result in localized structural changes in the backbone of HIV protease over a wide range, from 0.038-0.774 ⁇ .
  • Table 5 shows that the atoms of the inhibitor interact with the same atoms of the two different proteins, in this case the wild type and V82F/184V mutant HIV proteases. From Table 5, it can be seen that the atoms of the enzymes with which the inhibitor interacts belong to the structurally conserved regions.
  • the effects of mutations on the protein-inhibitor interactions can be quantified in terms of the distances between interacting pairs of atoms from the inhibitor and from atoms of the three-dimensionally conserved substructure of the protein. These distances are similar in the wild type and in the mutant complexes; the average of their differences is only 0.07 ⁇ . The range of the differences is 0.02-0.36 ⁇ .
  • This example illustrates the method by which experimentally-determined crystal structures of two different inhibitors in complexes with wild type HIV protease can be compared and analyzed for the existence of a three-dimensionally conserved substructure.
  • the structures of wild type HIV-1 protease in complexes with inhibitor 1 and with Amprenavir (inhibitor 2) were analyzed by means of (a) an overall superposition of the protein structures; and (b) a study of the distances from atoms of the inhibitors to atoms of the protein.
  • the superposition of the protein structures is performed in a two step process: 1) the distance between all pairs of corresponding Cá atoms (Cá atom of residue number 1 in one protein to Cá atom of residue number 1 in the second protein; Cá atom of residue number 2 in one protein to Cá atom of residue number 2 in the second protein; and so on) of the polypeptide chains is minimized by means of a least-square algorithm; 2) the superposition is refined by minimizing, in an iterative process, the distances between corresponding Cá atoms that are closer than a given distance (0.25 ⁇ in this example), thus eliminating regions of the structures having large conformational differences to compute the superposition parameters.
  • Inhibitors 1 and 2 have similar structural elements, in particular their core, i.e. groups at the P1-P1′ 60 positions. However, 2 has a THF group while 1 has a bis-THF group at the P2′ position.
  • the P2 groups are identical except for the substitution of an ether oxygen atom in 1 as compared to an amine nitrogen atom at the same position in 2.
  • Table 7 shows that 1 forms more interactions with the atoms of the protein that were previously identified as belonging to the structurally conserved substructure than does compound 2. For example, the O7 oxygen atom in compound 1, that forms an interaction with N29 nitrogen of the protease, has no counterpart in compound 2.
  • the O6 oxygen atom of 2 forms longer (and presumably weaker) hydrogen bonds with both N30 (3.50 ⁇ ) and N29 (3.51 ⁇ ).
  • the O6 oxygen of compound 1 forms a shorter (and presumably stronger) hydrogen bond with N29 (3.19 ⁇ ).
  • Table 7 where both compounds 1 and 2 form interactions with atoms in the structurally conserved substructure of HIV protease, the distances between interacting atoms are consistently shorter for compound 1, indicative of presumably stronger binding interactions.
  • Examples 1 and 2 were used to identify a three dimensionally-conserved substructure of HIV protease that is involved in the binding of HIV protease inhibitors and, in particular, to identify atoms of these substructural elements that are involved in forming interactions with atoms of HIV protease inhibitors.
  • This substructure is defined by the set of atomic coordinates (in orthogonal coordinates) provided in Table 8 and any equivalent set derived by applying arbitrary rotations and translations to the set of atomic coordinates in Table 8.
  • the values of the coordinates (X,Y,Z) of the atoms defining the substructure are affected by a standard error ⁇ .
  • protease inhibitor that contains atoms that can make favorable interactions with the atoms of the substructure may exhibit broad spectrum activity.
  • Compounds 1 and 3 contain a Bis-THF group at the P2 position that contains two atoms, in particular, hydrogen bond acceptor oxygen atoms, that can form hydrogen bonds with the two hydrogen atoms attached to the backbone amide nitrogen atoms on the protein at residues 29 and 30.
  • Compound 2 is similar to 1 except that 2 contains a THF group at P2 with only a single hydrogen bond acceptor oxygen atom. All three compounds differ in the P2′ substituent.
  • Compounds 1 and 3 both are unaffected by the two active site mutations, V82F and 184V, and Ki values for wild type and mutant enzymes are similar for both compounds.
  • compound 2 which contains only a single hydrogen bond acceptor atom in the P2 substitutent, is dramatically affected by the active site mutations, which demonstrate high level resistance to 2.
  • compounds 1 and 3 which closely resemble each other as well as compound 2, exhibit broad spectrum activity in that they are equally effective against wild type and mutant HIV strains that exhibit high level multidrug resistance towards compounds 2, 4, 5, 6, and 7.
  • the broad spectrum activity of compound 1 was completely unexpected and contrasts with the common and typical loss of antiviral potency experienced with compounds like 2, 4, 5, 6, 7, and indeed most other HIV protease inhibitors represented as similar or different structures that have been reported.
  • This example illustrates the method by which experimentally-determined crystal structures of two different target proteins, DHQases, from two different bacterial species can be compared and analyzed for the existence of a three-dimensionally conserved substructure even in the absence of readily discernible or statistically significant sequence similarity.
  • DHQases from different bacterial species typically exhibit less than 30% sequence identity (FIG. 2).
  • a schematic map showing the key interactions of the substrate-based inhibitor, DHQO, with the active site residues for the Type II DHQase from M. tuberculosis is provided in FIG. 3.
  • the superposition of the protein structures was performed in a two step process: 1) the distance between all pairs of corresponding Cá atoms (Cá atom of residue number 1 in one protein to Cá atom of residue number 1 in the second protein; Cá atom of residue number 2 in one protein to Cá atom of residue number 2 in the second protein; and so on) of the polypeptide chains is minimized by means of a least-square algorithm; 2) the superposition is refined by minimizing, in an iterative process, the distances between corresponding Cá atoms that are closer than a given distance (0.4 ⁇ in this example), thus eliminating regions of the structures having large conformational differences to compute the superposition parameters.
  • tuberculosis wt gt33 Molecule 1 Molecule 2 distance [ ⁇ ] CA MET 2 CA GLU 2 1.078 CA ALA 3 CA LEU 3 1.504 CA THR 4 CA ILE 4 1.800 CA LEU 5 CA VAL 5 1.283 CA LEU 6 CA ASN 6 0.911 CA VAL 7 CA VAL 7 0.715 CA LEU 8 CA ILE 8 0.298 CA HIS 9 CA ASN 9 0.211 CA GLY 10 CA GLY 10 0.591 CA PRO 11 CA PRO 11 0.599 CA ASN 12 CA ASN 12 0.487 CA LEU 13 CA LEU 13 0.428 CA ASN 14 CA GLY 14 0.229 CA LEU 15 CA ARG 15 0.685 CA LEU 16 CA LEU 16 0.541 CA GLY 17 CA GLY 17 1.693 CA THR 18 CA ARG 18 2.287 CA ARG 19 CA ARG 19 2.956 CA GLN 20 CA GLN 20 3.475 CA PRO 21 CA PRO 21 3.390 CA GLY 22 CA ALA 22 4.037 CA
  • Table 9 shows that the two structures are remarkably similar overall despite their low level sequence identity. However, the structures exhibit very large deviations in some regions, and are highly conserved in others. In particular, this analysis reveals that regions of the enzyme are minimally affected by the large number of amino acid sequence substitutions. The regions of the protein structure which are not significantly affected by the amino acid substitutions are defined as structurally conserved regions. In the present example, the substitutions result in localized structural changes in the backbone of DHQase over a wide range, from 0.095-4.165 ⁇ .
  • Examples 1-3 were applied to the DHQase data to identify a three dimensionally-conserved substructure of DHQase that is involved in the binding of DHQase inhibitors, in particular, to identify the relevant target substructure for developing broad spectrum inhibitors.
  • This substructure is defined by the set of atomic coordinates (in orthogonal coordinates) provided in Table 12 and any equivalent set derived by applying arbitrary rotations and translations to the set of atomic coordinates in Table 12.
  • the values of the coordinates (X,Y,Z) of the atoms defining the substructure are affected by a standard error ⁇ .
  • (X,Y,Z) values for each atom are those defined in the intervals (X ⁇ , X+ ⁇ ) for coordinate X, (Y ⁇ , Y+ ⁇ ) for coordinate Y, and (Z ⁇ , Z+ ⁇ ) for coordinate Z.
  • (X,Y,Z) values for each atom are those defined in the intervals (X ⁇ , X+ ⁇ ) for coordinate X, (Y ⁇ , Y+ ⁇ ) for coordinate Y, and (Z ⁇ , Z+ ⁇ ) for coordinate Z.

Abstract

The invention features a method of designing broad spectrum inhibitors using structural data, compositions having broad spectrum activity, and methods for treating disease using those compositions.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The application claims benefit of U.S. Provisional Application No. 0/344,788, filed Jan. 7, 2002, and No. 60/383,575, filed May 29, 2002, each of which is hereby incorporated by reference.[0001]
  • BACKGROUND OF THE INVENTION
  • The present invention relates to the field of inhibitors and methods for identifying or designing broad spectrum therapeutics for use in the treatment of infectious diseases and cancers, particularly where drug resistance is, or could reasonably predicted to be, an obstacle to successful long term therapy. [0002]
  • The development of drug resistance is one of the most common causes of drug failure in the treatment of diseases involving replicating biological entities (i.e., cancer and infectious diseases). Drug resistance often results from a reduction in drug-binding affinity and can be quantified by the ratio of drug binding affinity (Kd) for variant and wild type target proteins. Administration of a drug introduces a selective pressure upon the replicating biological entity. The result is the emergence of drug resistant strains. [0003]
  • Drug resistance is a major obstacle to the successful treatment of many cancers and infections, both bacterial and viral. For example, increased resistance of bacterial infections to antibiotic treatment has been extensively documented and has now become a generally recognized medical problem, particularly with nosocomial infections. See, for example, Jones et al., [0004] Diagn. Microbiol. Infect. Dis. 31:379 (1998); Murray, Adv. Intern. Med. 42:339 (1997); and Nakae, Microbiologia 13:273 (1997).
  • Drug resistance has complicated the treatment for HIV as new mutant strains of HIV have emerged that are resistant to multiple, structurally diverse, experimental and chemotherapeutic antiretrovirals, including HIV protease inhibitors (PIs), nucleoside and non-nucleoside HIV reverse transcriptase inhibitors (NRTIs and NNRTIs), and HIV fusion inhibitors (FIs). [0005]
  • More than 60 million people have been infected by HIV in the last two decades, and 20 million people have died from HIV/AIDS. While the development of highly active antiretrovirals to treat HIV/AIDS has led to significant reductions in the mortality and morbidity of AIDS, the rapid emergence and spread of drug-resistant mutant strains of HIV is rendering current drugs ineffective, and is the major cause of treatment failure. Recent estimates are that nearly 50% of drug-experienced patients in North America harbor HIV that is resistant to one or more of the 16 FDA-approved antiretroviral agents used in multi-drug ‘cocktails’ (Ref. dont have this ref). Moreover, it has been estimated that drug-resistant HIV accounts for up to 12% of new infections (Little et al., [0006] N. Engl. J. Med., 347:385 (2002)).
  • Accordingly, drug resistant HIV strains represent distinct infectious entities from a therapeutic viewpoint, and pose new challenges for drug design as well as drug treatment of existing infections. Substitutions have been documented in over 45 of the 99 amino acids of the HIV protease monomer in response to protease inhibitor treatment (Mellors, et al., [0007] International Antiviral News, 3:8 (1995); Eastman, et al., J. Virol., 72:5154 (1998); Kozal, et al., Nat. Med., 2:753 (1996)). The particular sequence and pattern of mutations selected by PIs is believed to be somewhat drug-specific and often patient-specific, but high level resistance is typified by multiple mutations in the protease gene which give rise to cross-resistance to all of the PIs.
  • In view of the foregoing problems, there exists a need for inhibitors against drug resistant and mdrHIV strains. Further, there exists a need for inhibitors against drug resistant and multi-drug resistant HIV proteases (mdrPR). Further still, there exists a need for inhibitors of HIV that can prevent or slow the emergence of drug resistant and mdrHIV strains in infected individuals. [0008]
  • Inhibitors with the ability to inhibit mdrHIV strains, and to slow the emergence of drug resistant strains in wild type HIV infections, are defined as “resistance-repellent” inhibitors. [0009]
  • There also exists a need for robust methods that can be used to design “resistance-repellent” inhibitors. [0010]
  • More generally, there is a need for therapeutic regimens that minimize the likelihood that resistance will occur in a disease involving a replicating biological entity. In one approach, drugs may be designed which have similar activity against both the wild type and mutant forms of their target. Such drugs minimize the probability of a mutant population emerging by reducing the selective pressure introduced by the drug when used to treat wild type infections. Such drugs also can be used to treat mutant infections and can be used for salvage therapy. [0011]
  • There is also an urgent need to develop potent, broad-spectrum, and mechanistically-novel antimicrobials suitable for tackling the growing problem of antibiotic-resistant bacteria strains, and for treating and/or preventing outbreaks of infectious diseases, including diseases caused by bioterrorism agents like anthrax, plague, cholera, gastroenteritis, multidrug-resistant tuberculosis (MDR TB). The recent anthrax attack of 2001 underscored the reality of large-scale aerosol bioweapons attack by terrorist groups. It also revealed that there is an urgent and pressing need to discover and develop novel and potent antimicrobials that can be used therapeutically and prophylactically for biodefense against new bioattacks. The NIH and CDC have identified a number of High Priority pathogens based on their likelihood of causing widespread contagious disease and/or death to the general population. Research on methods of protection against potential agents of bioterrorism has been a priority for several years at the NIH. A recent analysis suggested the existence of ongoing offensive biological weapons programs in at least 13 countries (Inglesby, T. V., et al., [0012] JAMA, 287:2236, (2002)).
  • The widespread use of antibiotics in human medical as well as in agricultural applications has promoted the emergence and spread of drug resistant bacteria that are no longer sensitive to existing drugs. The ease with which drug resistant microorganisms can be selected in a simple laboratory setting is a further concern when contemplating pharmaceutical-based strategies for biodefense. There is an urgent need to discover and develop novel therapeutic agents to combat pathogens that are likely to be used in a bioterrorist scenario. [0013]
  • A list of selected agents rated by likelihood to cause the greatest harm in a bioterrorist attack has been compiled by the CDC and NIAID (Lane, H. C., et al., [0014] Nat Med., 7:1271 (2001)). B. anthracis; the bacterium that causes anthrax, is one of the most serious of the group A pathogens. Dissemination of B. anthracis spores via the US Postal Service in 2001 established the feasibility of large-scale aerosol bioweapons attack. It has been estimated that between 130,000 and 3 million deaths would follow the release of 100 kg of B. anthracis, a lethality matching that of a hydrogen bomb (Inglesby, T. V., et al., JAMA, 287:2236, (2002)). Penicillin, doxycycline and ciprofloxacin are currently approved by the FDA for the treatment of inhalation anthrax infections. However, it was advised that antibiotic resistance to penicillin- and tetracycline-class antibiotics should be assumed following a terrorist attack (Inglesby, T. V., et al., JAMA, 281:1735-45 (1999)). Moreover, in vitro selection of a B. anthracis strain that is resistant to ofloxacin (a fluoroquinilone closely related to ciprofloxacin) has been reported (Choe, C. H., et al., Antimicrob. Agents. Chemother., 44:1766 (2000)). Following the anthrax attacks of 2001, the CDC advocated the use of a combination of 2-3 antibiotics. As a post-exposure prophylaxis, 60 days of treatment with ciprofloxacin is currently recommended. Strict compliance to this drug regimen is complicated by moderate to severe gastrointestinal tract intolerance.
  • Another group A pathogen, [0015] Y. pestis, is the causative agent of plague. If 50 kg of Y. pestis were released as an aerosol over a city of 5 million, pneumonic plague would afflict an estimated 150,000 individuals and result in 36,000 deaths (Inglesby, T. V., et al., JAMA, 283: 2281, (2000)). Streptomycin, tetracycline and doxycycline are the FDA-approved treatment for plague. Wide spread use of these antibiotics in the US raises concerns about possible resistance. A US-licensed, formaldehyde-killed whole bacilli vaccine was discontinued by its manufacturers in 1999 and is no longer available.
  • [0016] C. jejuni and V. cholerae are category B pathogens which can present a significant threat to the safety of food and water supplies. C. jejuni infections are one of the most commonly identified causes of acute bacterial gastroenteritis worldwide and area frequent cause of Traveler's diarrhea (Allos, B. M., Clin Infect Dis, 32:1201 (2001)). Currently, the CDC estimates that 2.4 million cases of Campylobacter infection occur in the United States each year, affecting almost 1% of the entire population. In the past few years, a rapidly increasing proportion of Campylobacter strains all over the world have been found to be fluoroquinolone-resistant. High rates of resistance make tetracycline, amoxicillin, ampicillin, metronidazole, and cephalasporins poor choices for the treatment of C. jejuni infections. All Campylobacter species are inherently resistant to vancomycin, rifampin, and trimethoprim. V. cholerae, a causative agent of cholera, is responsible for 120,000 deaths annually (Faruque, S. M., et al., Microbiol Mol Biol Rev, 62:1301 (1998)) and is characterized by a rapidly changing pattern of antibiotic resistance.
  • TB is one of the most common and devastating infectious diseases known to man. An estimated one third of the global population is infected with [0017] Mycobacteria tuberculosis. Eight million people develop an active infection and 2 million victims die yearly (Dye, C., et al., JAMA, 282:677 (1999.)). Currently, a combination of four drugs is recommended for TB treatment: isoniazid, rifampicin, pyrazinamide and ethambutole. The treatment course lasts 6 months. Such a multidrug combination together with the lengthy duration of treatment is prone to side-effects and adherence problems, which in turn can often lead to the development of drug resistance. The current drugs used to treat TB infections were introduced into clinical practice more than 30 years ago, in the absence of any knowledge of molecular mechanism. There is an urgent need to identify novel, effective, non-toxic and specific drugs that can shorten the duration of treatment, reduce side-effects, combat latent infection and reduce the spread of MDR TB strains. In addition, it is important to recognize the need for mechanistically novel drugs, i.e., antimicrobial agents that target biochemical pathways distinct from those of existing TB drugs, in order to be effective against MDR TB strains.
  • In summary, there is a clear need for the discovery of novel, non-toxic, broad spectrum antibiotics that can be used to (1) treat drug-resistant bacterial infections, and (2) protect civilians and military personnel in case of bioterrorist attacks. In one approach, drugs may be designed which have similar activity against both the wild type and variant forms of their target. Such drugs should minimize the probability of the emergence of mutant populations by reducing the selective pressure introduced by the drug when used to treat wild type infections. Such drugs also can be used to treat mutant infections and can be used for salvage therapy. In another approach, drugs may be designed which have similar activity against various isotypes of a homologous target. Such drugs can be used to treat multiple species of pathogenic microorganisms since they will be active against the target of each species. In a third approach, drugs can be designed that combine the properties and the uses of both of the above approaches. [0018]
  • There also exists a need for robust methods that can be used to design such broad spectrum antibiotics. [0019]
  • SUMMARY OF THE INVENTION
  • In a first aspect the invention features a method for the structure-based design of a drug that can act as an inhibitor of at least two different biological entities, the method comprising the steps of: (a) providing at least one structure of a wild type target protein or an inhibitor-wild type target protein complex; (b) providing at least one structure of a variant target protein or an inhibitor-variant target protein complex; (c) comparing at least one structure from step (a) with at least one structure from step (b) to determine whether there exists a common three-dimensionally conserved substructure comprising the atomic coordinates of the structurally conserved atoms of the inhibitors and structurally conserved atoms of the target proteins; and (d) if a conserved substructure exists, using the atomic coordinates of the conserved substructure to select a compound having atoms matching those of the structurally conserved atoms of the inhibitors, wherein the selection of the compound is performed using computer modeling. [0020]
  • The invention also features a method for the structure-based drug design of a broad spectrum compound, the method comprising the steps of: (a) providing at least one structure of a wild type target protein or an inhibitor-wild type target protein complex; (b) providing at least one structure of a variant target protein or an inhibitor-variant target protein complex; (c) comparing at least one structure from step (a) with at least one structure from step (b) to determine whether there exists a common three-dimensionally conserved substructure comprising the atomic coordinates of the structurally conserved atoms of the target proteins or a common three-dimensionally conserved substructure comprising the atomic coordinates of the structurally conserved atoms of the inhibitors and structurally conserved atoms of the target proteins; and (d) if a conserved substructure exists, using the atomic coordinates of the conserved substructure to select a compound having atoms matching those of the structurally conserved atoms of the inhibitors or to design a compound that binds to the target protein, wherein the selection of the compound is performed using computer modeling. [0021]
  • Desirably, the above method further comprises the steps of: (e) comparing at least one structure from step (a) with at least one structure from step (b) to determine whether there exists a three-dimensionally non-conserved substructure comprising the atomic coordinates of the structurally non-conserved atoms of the inhibitors and structurally non-conserved atoms of the target proteins; and (f) if a non-conserved substructure exists, using the atomic coordinates of the non-conserved substructure to reject a compound having atoms matching those of the structurally non-conserved atoms of the inhibitors, wherein the rejection of the compound is performed in conjunction with computer modeling. [0022]
  • In any of the above methods, at least two, four, six, or eight structures from step b can be used in step c. The methods can be applied using several structures, including at least two, four, six, or eight variant forms of the target protein. [0023]
  • The inhibitors used in the inhibitor-wild type target protein complex and the inhibitor-variant target protein complex can be the same or different. The inhibitors can be selected from competitive or noncompetitive inhibitors. Furthermore, the inhibitors can be selected from reversible, or irreversible inhibitors. [0024]
  • In any of the above methods, the variant target protein can be a homologous protein or a mutant protein. [0025]
  • In any of the above methods, the structures can be selected from crystal structures, NMR structures, computer models, any acceptable experimental, theoretical or computational method of deriving a three-dimensional representation of a structure, or a combination thereof. [0026]
  • Target proteins for use in the present invention include any therapeutically relevant protein. The target protein can be a viral, bacterial, protozoan, or fungal protein. In some instances, the target protein is one that is expressed in a neoplasm. [0027]
  • Preferably, the target protein can be an enzyme, a receptor, a structural protein, a component of a macromolecular complex, a component of a metabolic pathway, or an assembly of biological molecules. Desirably, the target protein is necessary for the survival of the replicating biological entity. For example, the target protein can be an enzyme selected from the group consisting of reverse transcriptases, proteases, DNA and RNA polymerases, methylases, oxidases, esterases, acyl transferases, helicases, topoisomerases, and kinases. The target protein can be a component of a metabolic pathway, such as the shikimate pathway. Desirable target proteins include HIV protease or 3-dehydroquinate dehydratase, among others. [0028]
  • Where the target protein is HIV protease, suitable inhibitors for use in the methods of the invention include those selected from the group consisting of indinavir, nelfinavir, ritonavir, saquinavir, amprenavir, lopinavir, and UIC-94003. [0029]
  • A broad spectrum protease inhibitor can be designed using the susbstructure of structurally conserved atoms described by the atomic coordinates in Table 8, which includes the structurally conserved atoms of the inhibitor and structurally conserved atoms of the protease. A broad spectrum protease inhibitor can also be designed using the structurally conserved atoms of the inhibitor alone. These are described by the atomic coordinates in Table 8. [0030]
  • A broad spectrum 3-dehydroquinate dehydratase inhibitor can be designed using the susbstructure of structurally conserved atoms described by the atomic coordinates in Table 12, which includes the structurally conserved atoms of the 3-dehydroquinate dehydratase. A broad spectrum 3-dehydroquinate dehydratase inhibitor can also be designed using the structurally conserved atoms of the inhibitor alone. These are described by the atomic coordinates in Table 12. [0031]
  • The invention also features compounds having a chemical structure selected using any of the methods above. Such compounds are broad spectrum inhibitors and have broad spectrum activity against replicating biological entities expressing a particular target protein. Thus, if the target protein is expressed by a microbe or a neoplasm, the compound will have broad spectrum activity against the microbe or neoplasm, respectively. [0032]
  • The invention features a compound having broad spectrum activity against HIV protease wherein the compound has a chemical structure selected using the methods above, including those methods utilizing the atomic coordinates of Table 8. [0033]
  • The invention features a compound having broad spectrum activity against 3-dehydroquinate dehydratase wherein the compound has a chemical structure selected using the methods above, including those methods utilizing the atomic coordinates of Table 12. [0034]
  • The compounds of the invention exclude bis-THF compounds (e.g., analogs of [0035] compounds 1 and 3) as described in J. Med. Chem. 39:3278-3290 (1996) (compounds 49-52 and 58-60), Bioorg. Med. Chem. Lett. 8:979-982 (1998), WO99/65870, U.S. Pat. No. 6,319,946, WO02/08657, WO02/092595, WO99/67417, EP00/9917, and WO00/76961; and also exclude fused ring THF structures as described in Bioorg. Med. Chem. Lett. 8:687-690 (1998) and U.S. Pat. No. 5,990,155.
  • For any of the broad spectrum inhibitors of the invention, broad spectrum activity can be measured by the ratio of the inhibitory concentrations of the broad spectrum inhibitor for the variant and wild type biological entities (IC[0036] 50, variant/IC50, wild type). Desirably, the IC50, variant/IC50, wild type ratio for a broad spectrum inhibitor is less than 100, 80, 60, 40, 30, 20, 10, 8, 6, or, most desirably, less than 3.
  • A broad spectrum inhibitor can be active against several different mutant biological entities. Desirably, the inhibitor will have broad spectrum activity against at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 mutant biological entities. [0037]
  • A broad spectrum inhibitor can also be active against different organisms or neoplastic cell types expressing homologous target proteins that possess sufficient structural similarity. Desirably, the inhibitor will have broad spectrum activity against at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 different organisms or neoplastic cell types expressing homologous target proteins. [0038]
  • The invention also features a pharmaceutical composition that includes a broad spectrum inhibitor described herein in any pharmaceutically acceptable form, including isomers such as diastereomers and enantiomers, salts, solvates, and polymorphs thereof. The composition can include an inhibitor of the invention along with a pharmaceutically acceptable carrier or diluent. [0039]
  • The invention also features methods of treating disease in a patient in need thereof, which includes the administration of a pharmaceutical composition of the invention to the patient in an amount sufficient to treat the disease. The pharmaceutical composition includes any broad spectrum inhibitor described herein. Such broad spectrum inhibitors have broad spectrum activity against replicating biological entities expressing a particular target protein. Thus, if the target protein is expressed by a microbe or a neoplasm, the disease to be treated will be a microbial infection or neoplasm, respectively. [0040]
  • The invention features a method of treating an HIV infection in a patient in need thereof, the method including the step of administering to the patient a pharmaceutical composition including a broad spectrum protease inhibitor described herein in amounts effective to treat the HIV infection. [0041]
  • The invention features a method of treating a bacterial infection in a patient in need thereof, the method including the step of administering to the patient a pharmaceutical composition including a broad spectrum 3-dehydroquinate dehydratase inhibitor described herein in amounts effective to treat the bacterial infection. The bacterial infection to be treated using the above method can be caused by a bacterium selected from the group consisting of [0042] C. jejuni, V. cholerae, Y. pestis, B. anthracis, P. putidas, and M. tuberculosis. Furthermore, this method can be used to treat infections by any microbe the utilizes 3-dehydroquinate dehydratase.
  • The invention also features the use of a pharmaceutical composition described herein in the manufacture of a medicament for the treatment of a disease. The pharmaceutical composition includes any broad spectrum inhibitor described herein. Such broad spectrum inhibitors and have broad spectrum activity against replicating biological entities expressing a particular target protein. Thus, if the target protein is HIV protease or 3-dehydroquinate dehydratase, the disease to be treated will be an HIV infection or bacterial infection, respectively. [0043]
  • The term “replicating biological entity” includes, for example, bacteria, fungi, yeasts, viruses, protozoa, prions and neoplasms [0044]
  • Neoplasms include, for example, carcinomas of the bladder, breast, colon, kidney, liver, lung, head and neck, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, or skin; a hematopoietic tumor of lymphoid lineage; a hematopoietic tumor of myeloid lineage; a tumor of mesenchymal origin; a tumor of the central or peripheral nervous system; melanoma; seminoma; teratocarcinoma; osteosarcoma; thyroid follicular cancer; and Kaposi's sarcoma. Hematopoietic tumors of lymphoid lineage can be leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma. [0045]
  • By “wild type target protein” is meant a protein obtained from a replicating biological entity that has not been subjected to drug selection pressure, and could include polymorphisms or isoforms thereof. A replicating biological entity that expresses wild type target protein is referred to herein as a wild type biological entity. [0046]
  • By “variant target protein” is meant a mutant target protein or a homologous target protein. A replicating biological entity that expresses variant target protein is referred to herein as a variant biological entity. [0047]
  • By “mutant target protein” is meant a target protein that contains one or more amino acid substitutions with respect to the wild type target protein, including proteins from the same organism that have evolved under drug selection pressure. In general, mutant target proteins will have one or more amino acid substitutions and should be readily identified as related to the cognate wild type protein using standard sequence comparison methods. A replicating biological entity that expresses mutant target protein is referred to herein as a mutant biological entity. [0048]
  • By “homologous target protein” is meant a variant target protein that is expressed in a different species or neoplastic cell type than the wild type target protein, but has the same, or similar, function. [0049]
  • By “structurally conserved target substructure”, and by “structurally” or “three-dimensionally conserved substructure” as applied to target proteins, is meant the regions of the target protein structure which are not significantly affected by amino acid mutations or substitutions. Such regions can be defined using standard methods of comparative analysis of three-dimensional structures of proteins, such as superposition analysis, for example. In the case of HIV protease, these regions were identified using a pair wise superposition analysis of wild type and mutant protease structures complexed with inhibitors. The superposition of structures can be performed using the iterative procedure described herein. In the case of DHQase, these regions were identified using a pair wise superposition analysis of wild type and homologous DHQase structures from different bacterial species with and without inhibitors. It is apparent that the overall compositions of structurally conserved target substructures will likely differ for different, non-homologous target proteins, especially when the frequency of amino acid substitutions in high. However, a quantitative definition can be derived from the superposition analysis, which provides both the identities and the positions of the atoms that comprise these substructures. The regions that comprise structurally conserved target substructures contain atoms whose superimposed pairs have three-dimensional atomic coordinates that match to within a distance of 1 Å, 0.6 Å, 0.4 Å, or 0.2 Å. [0050]
  • By “broad spectrum inhibitor” is meant a compound having broad spectrum activity, i.e., an inhibitor that is active against two different-biological entities, e.g., both a wild type biological entity and one or more variants of that biological entity. Thus, broad spectrum activity can be described by the inhibitor's action against a particular target protein (e.g., broad spectrum activity against protease) or a particular target organism (e.g., broad spectrum activity against HIV). Broad spectrum inhibitors will have medically insignificant interactions with non-conserved regions. Broad spectrum inhibitors can be useful for the treatment and/or prevention of infectious diseases caused by multiple infectious agents, as well as for decreasing the development of drug-resistance by these organisms. [0051]
  • As used herein, the term “treating” refers to administering a pharmaceutical composition for prophylactic and/or therapeutic purposes. To “prevent disease” refers to prophylactic treatment of a patient who is not yet ill, but who is susceptible to, or otherwise at risk of, a particular disease. To “treat disease” or use for “therapeutic treatment” refers to administering treatment to a patient already suffering from a disease to ameliorate the disease and improve the patient's condition. Thus, in the claims and embodiments, treating is the administration to a patient either for therapeutic or prophylactic purposes. [0052]
  • The term “microbial infection” refers to the invasion of the host patient by pathogenic microbes (e.g., bacteria, fungi, yeasts, viruses, protozoa). This includes the excessive growth of microbes that are normally present in or on the body of a patient. More generally, a microbial infection can be any situation in which the presence of a microbial population(s) is damaging to a host patient. Thus, a patient is “suffering” from a microbial infection when excessive numbers of a microbial population are present in or on a patient's body, or when-the presence of a microbial population(s) is damaging the cells or other tissue of a patient. [0053]
  • The term “microbes” includes, for example, bacteria, fungi, yeasts, viruses and protozoa. The term “administration” or “administering” refers to a method of giving a dosage of a pharmaceutical composition to a patient, where the method is, e.g., topical, oral, intravenous, intraperitoneal, or intramuscular. The preferred method of administration can vary depending on various factors, e.g., the components of the pharmaceutical composition, site of the potential or actual disease and severity of disease. [0054]
  • The term “patient” includes humans, cattle, pigs, sheep, horses, dogs, and cats, and also includes other vertebrate, most preferably, mammalian species. [0055]
  • Where “atomic coordinates” are provided, or otherwise referred to, these coordinates define a three dimensional structure. That such a structure may be defined by more than one different coordinate system, e.g., by translation or rotation of the coordinates, does not change the relative positions of the atoms in the structure. Accordingly, any reference to atomic coordinates herein is intended to include any equivalent three dimensional structure defined by the coordinates. [0056]
  • By “computer modeling” is meant the use of a computer to visualize or compute a compound, a portion of a compound, a target protein, a portion of a target protein, a complex between a compound and a target protein, or a portion of a complex between a compound and a target protein. [0057]
  • Other features and advantages of the invention will be apparent from the following detailed description and from the claims.[0058]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a table depicting the structures of compounds 1-7, gt33, and qxa. [0059]
  • FIG. 2 illustrates the amino acid alignment of type II DHQases. Fully conserved residues are framed. Catalytically important amino acids are marked by stars. Arrows denote amino acids that make hydrogen bonds and ionic interactions in the structure of [0060] M. tuberculosis DHQase complexed with the inhibitor, 3-dehydroquinic acid oxime.
  • FIG. 3 illustrates the key interactions of the substrate-based inhibitor, DHQO, with the active site residues for the Type II DHQase from [0061] M. tuberculosis.
  • DETAILED DESCRIPTION
  • We have discovered that the comparative analysis of the structures of complexes of inhibitors bound to wild type and variant forms of a target protein can be used to design compounds that are broad spectrum inhibitors. [0062]
  • The methods of the invention entail the design of compounds having a particular structure. The methods rely upon the use of structural information to arrive at these compounds. The structural data define a three dimensional array-of the important contact atoms in an inhibitor that bind to the target protein in a fashion that results in broad spectrum activity against biological entities expressing variants of the target protein. [0063]
  • Inhibitor-Target Protein Structures [0064]
  • Atomic structural coordinates can be selected from crystal structures, NMR structures, computer models, any acceptable experimental, theoretical or computational method of deriving a three-dimensional representation of a structure, or a combination thereof. Atomic coordinates for use in the methods of the invention can be obtained from publicly available sources, e.g. from the Protein Data Bank, or obtained using known experimental or computational methods. [0065]
  • Atomic structural coordinates for use in the methods of the invention include crystal structures of HIV protease/inhibitor complexes derived from wild type and drug-resistant mutant proteases, and of DHQase and DHQase inhibitor complexes derived from two or more bacterial species, among others. In examples 1-3, the methods of the invention are applied using the coordinates of wild type HIV protease complexed with amprenavir, wild type HIV protease complexed with UIC-94003, and V82F/184V mutant HIV protease complexed with UIC-94003. In example 4, the methods of the invention are applied using the coordinates of wild type DHQase from [0066] M. tuberculosis and from Pseudomonas putidas. a complex between a compound and a target protein. The coordinates of other representative structures of HIV protease and DHQase should be useful for performing the methods of the present invention.
  • Conserved Substructures [0067]
  • Conserved substructures can be identified for target proteins, for target protein-inhibitor complexes, and/or for inhibitors, depending on the nature of the structures that are used in the comparative superposition analysis. In one approach, at least one structure of a wild type target protein is compared to at least one structure of a mutant or homologous target protein to determine whether a common three-dimensionally conserved substructure is present among the wild type protein and the mutant or homologous proteins, respectively. In another approach, at least one structure of an inhibitor-wild type target protein complex and at least one structure of an inhibitor-mutant target protein complex are compared to determine whether a common three-dimensionally conserved substructure is present among the mutant and wild type complexes. In a third approach, at least one structure of an inhibitor-wild type target protein complex and at least one structure of a mutant or homologous target protein without inhibitor are compared to determine whether a common three-dimensionally conserved substructure is present among the respective mutant or homologous protein and the wild type complexes. Variations of the approached described above can also be used. In each case, such a comparison can be made by means of (a) an overall superposition of the atoms of the protein structures; and, where feasible, (b) a study of the distances from atoms of the inhibitors to atoms of the protein. This analysis requires three-dimensional atomic coordinates of the protein structures and of the bound inhibitor. [0068]
  • The superposition of the protein structures can be performed in a two step process: 1) the distance between all pairs of corresponding Cá atoms (Cá atom of [0069] residue number 1 in one protein to Cá atom of residue number 1 in the second protein; Cá atom of residue number 2 in one protein to Cá atom of residue number 2 in the second protein; and so on) of the polypeptide chains is minimized by means of a least-square algorithm; 2) the superposition is refined by minimizing, in an iterative process, the distances between corresponding Cá atoms that are closer than a given distance (0.25 A for example), thus eliminating regions of the structures having large conformational differences to compute the superposition parameters. Furthermore, where a partial structure is provided (e.g., from NMR data) the available coordinates are superimposed.
  • The conserved substructure identifies the relevant portion of the target protein that is the active site, or binding region, defined by that part of the target protein interacting with inhibitor. Important interactions between the target protein and inhibitor are identified by mapping the contacts between the two. Structurally conserved regions of the target protein not near the binding site are generally not relevant to the design of the broad spectrum inhibitor. Accordingly, the selection of the meaningful substructure is identified using the above mentioned contacts. [0070]
  • Design of a Broad Spectrum Inhibitor [0071]
  • The coordinates of the conserved inhibitor substructure are used to design an inhibitor having atoms matching those of the three-dimensionally structurally conserved atoms of the inhibitors. The result is an inhibitor for which IC[0072] 50, variant and IC50, wild type are similar, minimizing the selective pressure introduced by the drug.
  • The methods of the invention can employ computer-based methods for designing broad spectrum inhibitors. These computer-based methods fall into two broad classes: database methods and de novo design methods. In database methods the compound of interest is compared to all compounds present in a database of chemical structures and compounds whose structure is in some way similar to the compound of interest are identified. The structures in the database are based on either experimental data, generated by NMR or x-ray crystallography, or modeled three-dimensional structures based on two-dimensional (i.e., sequence) data. In de novo design methods, models of compounds whose structure is in some way similar to the compound of interest are generated by a computer program using information derived from known structures, e.g., data generated by x-ray crystallography and/or theoretical rules. Such design methods can build a compound having a desired structure in either an atom-by-atom manner or by assembling stored small molecular fragments. [0073]
  • The success of both database and de novo methods in identifying compounds having the desired activity depends on the identification of the functionally relevant portion of the compound of interest. The functionally relevant portion of the compound, the pharmacophore, is defined by the structurally conserved substructure. A pharmacophore then is an arrangement of structural features and functional groups important for obtaining an inhibitor having broad spectrum activity. [0074]
  • Not all identified compounds having the desired pharmacophore will act as broad spectrum inhibitors. The actual activity can be finally determined only by measuring the activity of the compound in relevant biological assays. However, the methods of the invention are extremely valuable because they can be used to greatly reduce the number of compounds which must be tested to identify those likely to exhibit broad spectrum activity. [0075]
  • Programs suitable for generating predicted three-dimensional structures from two-dimensional data include: Concord (Tripos Associated, St. Louis, Mo.), 3-D Builder (Chemical Design Ltd., Oxford, U.K.), Catalyst (Bio-CAD Corp., Mountain View, Calif.), and Daylight (Abbott Laboratories, Abbott Park, Ill.). [0076]
  • Programs suitable for searching three-dimensional databases to identify molecules bearing a desired pharmacophore include: MACCS-3D and ISIS/3D (Molecular Design Ltd., San Leandro, Calif.), ChemDBS-3D (Chemical Design Ltd., Oxford, U.K.), and Sybyl/3DB Unity (Tripos Associates, St. Louis, Mo.). [0077]
  • Programs suitable for pharmacophore selection and design include: DISCO (Abbott Laboratories, Abbott Park, Ill.), Catalyst (Bio-CAD Corp., Mountain View, Calif.), and ChemDBS-3D (Chemical Design Ltd., Oxford, U.K.). [0078]
  • Databases of chemical structures are available from Cambridge Crystallographic Data Centre (Cambridge, U.K.) and Chemical Abstracts Service (Columbus, Ohio). [0079]
  • De novo design programs include Ludi (Biosym Technologies Inc., San Diego, Calif.) and Aladdin (Daylight Chemical Information Systems, Irvine Calif.). [0080]
  • One skilled in the art may use one of several methods to screen chemical entities for their ability to match the conserved substructure. This process may begin by visual inspection of, for example, the active site on the computer screen based on the atomic coordinates for the target protein. Docking may be accomplished using software such as Quanta and Sybyl, followed by energy minimization and molecular dynamics with standard molecular mechanics forcefields, such as CHARMM and AMBER. [0081]
  • Specialized computer programs may also assist in the process of selecting chemical entities. These include: [0082]
  • 1. GRID (Goodford, P. J., “A Computational Procedure for Determining Energetically Favorable Binding Sites on Biologically Important Macromolecules,” [0083] J. Med. Chem., 28:849 (1985)). GRID is available from Oxford University, Oxford, UK.
  • 2. MCSS (Miranker, A. and M. Karplus, “Functionality Maps of Binding Sites: A Multiple Copy Simultaneous Search Method.” [0084] Proteins: Structure, Function, and Genetics, 11:29 (1991)). MCSS is available from Molecular Simulations, Burlington, Mass.
  • 3. AUTODOCK (Goodsell, D. S. and A. J. Olsen, “Automated Docking of Substrates to Proteins by Simulated Annealing,” Proteins: Structure, Function, and Genetics, 8:195 (1990)). AUTODOCK is available from Scripps Research Institute, La Jolla, Calif. [0085]
  • 4. DOCK (Kuntz, L. D. et al., “A Geometric Approach to Macromolecule-Ligand Interactions,” [0086] J. Mol. Biol., 161:269 (1982)). DOCK is available from University of California, San Francisco, Calif.
  • Once the conserved substructure for the inhibitor has been identified, the conserved atoms of the inhibitor can be selected for assembly into a single inhibitor. Assembly may be proceed by visual inspection of the relationship of the fragments to each other on the three-dimensional image displayed on a computer screen in relation to the structure coordinates of the target protein. This may be followed by manual model building using software such as Quanta or Sybyl. [0087]
  • Useful programs to aid one of skill in the art in assembly of the individual chemical entities or fragments include: [0088]
  • 1. CAVEAT (Bartlett, P. A. et al, “CAVEAT: A Program to Facilitate the Structure-Derived Design of Biologically Active Molecules”. In “Molecular Recognition in Chemical and Biological Problems,” [0089] Special Pub., Royal Chem. Soc., 78:182 (1989)). CAVEAT is available from the University of Calif., Berkeley, Calif.
  • 2. 3D Database systems such as MACCS-3D (MDL Information Systems, San Leandro, Calif.). This area is reviewed in Martin, Y. C., “3D Database Searching in Drug Design,” [0090] J. Med. Chem., 35:2145 (1992)).
  • 3. HOOK (available from Molecular Simulations, Burlington, Mass.). [0091]
  • Other molecular modeling techniques may also be employed in accordance with this invention. See, e.g., Cohen, N. C. et al., “Molecular Modeling Software and Methods for Medicinal Chemistry,” [0092] J. Med. Chem., 33:883 (1990). See also, Navia, M. A. and M. A. Murcko, “The Use of Structural Information in Drug Design,” Current Opinions in Structural Biology, 2:202 (1992).
  • Once a broad spectrum inhibitor has been optimally designed, as described above, substitutions may then be made in some of its atoms or side groups in order to improve or modify its binding properties. Generally, initial substitutions are conservative, i.e., the replacement group will have approximately the same size, shape, hydrophobicity and charge as the original group. It should, of course, be understood that components known in the art to alter conformation should be avoided. [0093]
  • In general, inhibitors designed using the methods of the invention can be tested for broad spectrum activity using any of the to in vitro and/or in vivo methods described below, among others. [0094]
  • Broad Spectrum Inhibitors [0095]
  • Broad spectrum inhibitors match the pharmacophore defined by the structurally conserved substructure. The pharmacophore is the arrangement of structural features and functional groups important for obtaining an inhibitor having broad spectrum activity. This pharmacophore is derived using structural data for known inhibitors complexed to a target protein. Accordingly, broad spectrum inhibitors will often be structurally related to known compounds lacking broad spectrum activity, but useful in the design of broad spectrum inhibitors using the methods disclosed herein. These known inhibitors serve as lead compounds for both the design and synthesis of a broad spectrum inhibitor. Using the synthetic methods for making the lead compounds and standard synthetic methods as described by, for example, J. March, Advanced Organic Chemistry: Reactions, Mechanisms and Structure,” John Wiley & Sons, Inc., 1992; T. W. Green and P. G. M. Wuts, “Protective Groups in Organic Synthesis” (2[0096] nd Ed.), John Wiley & Sons, 1991; and P. J. Kocienski, “Protecting Groups,” Georg Thieme Verlag, 1994, one can synthesize the broad spectrum inhibitors described herein.
  • Typically the lead compounds bear varied functional groups which are present in the pharmacophore, including hydrogen-bond donors, hydrogen-bond acceptors, ionic moieties, polar moieties, hydrophobic moieties, aromatic centers, and electron-donors and acceptors. These are linked by a structural scaffold which imparts the appropriate a three dimension arrangement of the functional groups. [0097]
  • Numerous modifications of the lead compound can be made using techniques known in the art. These include changing a functional group by replacing it with another moiety of the same group. For example, one hydrogen-bond donor may be substituted by another. A good hydrogen bond donor has an H atom bonded to a very electronegative atom (e.g., O—H or N—H). Examples of hydrogen-bond donors include alcohols, carboxylic acids, oximes, and amides, among others. Similarly, one hydrogen-bond acceptor may be substituted by another. A good hydrogen bond acceptor has an electronegative element with lone pairs (e.g., O, N, or F). Examples of hydrogen bond acceptors include water, halogen atoms, alcohols, amines, carbonyls, ethers, and amides, among others. It may also be desirable to alter the distance between functional groups in a lead compound. This is achieved by employing synthetic methods analogous to those used to prepare the lead compound, but replacing the scaffold with a structurally related scaffold that provides the desired distance (e.g., a scaffold that incorporates more or fewer atoms linking the relevant functional groups). In some instances it may also be desirable to alter the stereochemistry in a lead compound. This can be accomplished by employing racemic starting materials, or by employing reaction conditions that result in racemization of the relevant chiral center, followed by separation of the enantiomeric or diastereomeric mixture. [0098]
  • Assays [0099]
  • Inhibitors designed using the methods disclosed herein may be further assayed, using standard in vitro models or animal models, to evaluate therapeutic activity and toxicity. These assays are described in the literature and are familiar to those skilled in the art. These include but are not limited to assays for monitoring or measuring efficacy against HIV, bacteria, and neoplasms. [0100]
  • One skilled in the art will be familiar with methods of measuring the IC[0101] 50's of a broad spectrum inhibitor described herein. The IC50 value is determined by plotting percent activity versus inhibitor concentration in the assay and identifying the concentration at which 50% of the activity (e.g., growth, enzymatic activity, protein production, etc.) remains. Inhibitors can be tested for antimicrobial activity against a panel of organisms according to standard procedures described by the National Committee for Clinical Laboratory Standards (NCCLS document 7-A3, Vol. 13, No. 25, 1993/NCCLS document M27-P, Vol. 12, No. 25, 1992). Inhibitors can be dissolved (0.1 μg/ml-500 μg/ml) in microbial growth media, diluted, and added to wells of a microtiter plate containing bacteria or fungal cells in a final volume of an appropriate media (Mueller-Hinton Broth; Haemophilus Test Media; Mueller-Hinton Broth+5% Sheep Blood; or RPMI 1690). Typically, the plates are incubated overnight at an appropriate temperature (30° C. to 37° C.) and optical densities (measure of cell growth) are measured using a commercial plate reader.
  • IC[0102] 50 's for broad spectrum protease inhibitors can be measured against wild type HIV and clinically isolated mutant HIV isolates, utilizing the PHA-PBMC exposed to HIV-1 (50 TCID50 dose/X06 PBMC) as target cells and using the inhibition of p24 Gag protein production as an endpoint. The amounts of p24 antigen produced by the cells can be determined on day 7 in culture using a commercially available radioimmunoassay kit. Drug concentrations resulting in 50% inhibition (IC50's) of p24 antigen production can be determined by comparison with the p24 production level in drug-free control cell cultures.
  • Therapy [0103]
  • The invention features a method of identifying a compound having broad spectrum activity. Broad spectrum inhibitors of the present invention may be administered by any appropriate route for treatment or prevention of a disease or condition associated with a bacterial infection, viral infection, or neoplastic disorder, among others. These may be administered to humans, domestic pets, livestock, or other animals with a pharmaceutically acceptable diluent, carrier, or excipient, in unit dosage form. Administration may be topical, parenteral, intravenous, intra-arterial, subcutaneous, intramuscular, intracranial, intraorbital, ophthalmic, intraventricular, intracapsular, intraspinal, intracistemal, intraperitoneal, intranasal, aerosol, by suppositories, or oral administration. [0104]
  • Therapeutic formulations may be in the form of liquid solutions or suspensions; for oral administration, formulations may be in the form of tablets or capsules; and for intranasal formulations, in the form of powders, nasal drops, or aerosols. [0105]
  • Methods well known in the art for making formulations are found, for example, in “Remington: The Science and Practice of Pharmacy” (20th ed., ed. A. R. Gennaro AR., 2000, Lippincott Williams & Wilkins). Formulations for parenteral administration may, for example, contain excipients, sterile water, or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated napthalenes. Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds. Nanoparticulate formulations (e.g., biodegradable nanoparticles, solid lipid nanoparticles, liposomes) may be used to control the biodistribution of the compounds. Other potentially useful parenteral delivery systems include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel. The concentration of the broad spectrum inhibitor in the formulation will vary depending upon a number of factors, including the dosage of the drug to be administered, and the route of administration. [0106]
  • The broad spectrum inhibitor may be optionally administered as a pharmaceutically acceptable salt, such as a non-toxic acid addition salts or metal complexes that are commonly used in the pharmaceutical industry. Examples of acid addition salts include organic acids such as acetic, lactic, pamoic, maleic, citric, malic, ascorbic, succinic, benzoic, palmitic, suberic, salicylic, tartaric, methanesulfonic, toluenesulfonic, or trifluoroacetic acids or the like; polymeric acids such as tannic acid, carboxymethyl cellulose, or the like; and inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid phosphoric acid, or the like. Metal complexes include zinc, iron, and calcium, among others. [0107]
  • Administration of compounds in controlled release formulations is useful where the broad spectrum inhibitor has (i) a narrow therapeutic index (e.g., the difference between the plasma concentration leading to harmful side effects or toxic reactions and the plasma concentration leading to a therapeutic effect is small; generally, the therapeutic index, TI, is defined as the ratio of median lethal dose (LD[0108] 50) to median effective dose (ED50)); (ii) a narrow absorption window in the gastro-intestinal tract; or (iii) a short biological half-life, so that frequent dosing during a day is required in order to sustain the plasma level at a therapeutic level.
  • Many strategies can be pursued to obtain controlled release in which the rate of release outweighs the rate of metabolism of the broad spectrum inhibitor. For example, controlled release can be obtained by the appropriate selection of formulation parameters and ingredients, including, e.g., appropriate controlled release compositions and coatings. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, nanoparticles, patches, and liposomes. [0109]
  • Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose and sorbitol), lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). [0110]
  • Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium. [0111]
  • Pharmaceutical formulations of broad spectrum inhibitor described herein include isomers such as diastereomers and enantiomers, mixtures of isomers, including racemic mixtures, salts, solvates, and polymorphs thereof. [0112]
  • The formulations can be administered to human patients in therapeutically effective amounts. For example, when the broad spectrum inhibitor is an antimicrobial drug, an amount is administered which prevents, stabilizes, eliminates, or reduces a microbial infection. Typical dose ranges are from about 0.01 μg/kg to about 2 mg/kg of body weight per day. The exemplary dosage of drug to be administered is likely to depend on such variables as the type and extent of the disorder, the overall health status of the particular patient, the formulation of the compound excipients, and its route of administration. Standard clinical trials maybe used to optimize the dose and dosing frequency for any particular broad spectrum inhibitor. [0113]
  • The following examples are meant to illustrate, but in no way limit, the claimed invention. [0114]
  • EXAMPLE I
  • This example illustrates the method by which experimentally-determined crystal structures of the same inhibitor in complex with wild type and mutant species of HIV protease can be compared and analyzed for the existence of a three-dimensionally conserved substructure. [0115]
  • The structures of wild type HIV-1 protease and a mutant, V82F/184V, HIV-1 protease, both in complexes with the inhibitor shown in FIG. 1 were determined using conventional x-ray crystallography techniques. The structures were analyzed by means of (a) an overall superposition of the atoms of the protein structures; and, (b) a study of the distances from atoms of the inhibitors to atoms of the protein. This analysis requires three dimensional atomic coordinates of the protein structures and of the bound inhibitor. [0116]
  • The superposition of the protein structures was performed in a two step process: 1) the distance between all pairs of corresponding Cá atoms (Cá atom of [0117] residue number 1 in one protein to Cá atom of residue number 1 in the second protein; Cá atom of residue number 2 in one protein to Cá atom of residue number 2 in the second protein; and so on) of the polypeptide chains is minimized by means of a least-square algorithm; 2) the superposition is refined by minimizing, in an iterative process, the distances between corresponding Cá atoms that are closer than a given distance (0.25 Å in this example), thus eliminating regions of the structures having large conformational differences to compute the superposition parameters. The distances between equivalenced Cá atoms after the minimization procedure are shown in Table 4.
    TABLE 4
    Distances between equivalent Cá atoms
    Molecule 1: HIV-1 PR wt: 1
    Molecule 2: HIV-1 PR V82F/I84V mutant: 1
    Molecule 1 Molecule 2 distance [Å]
    CA PRO 1 CA PRO 1 0.455
    CA GLN 2 CA GLN 2 0.434
    CA ILE 3 CA ILE 3 0.418
    CA THR 4 CA THR 4 0.317
    CA LEU 5 CA LEU 5 0.172
    CA TRP 6 CA TRP 6 0.228
    CA GLN 7 CA GLN 7 0.364
    CA ARG 8 CA ARG 8 0.166
    CA PRO 9 CA PRO 9 0.057
    CA LEU 10 CA LEU 10 0.183
    CA VAL 11 CA VAL 11 0.194
    CA THR 12 CA THR 12 0.168
    CA ILE 13 CA ILE 13 0.146
    CA LYS 14 CA LYS 14 0.229
    CA ILE 15 CA ILE 15 0.266
    CA GLY 16 CA GLY 16 0.662
    CA GLY 17 CA GLY 17 0.491
    CA GLN 18 CA GLN 18 0.267
    CA LEU 19 CA LEU 19 0.112
    CA LYS 20 CA LYS 20 0.128
    CA GLU 21 CA GLU 21 0.190
    CA ALA 22 CA ALA 22 0.169
    CA LEU 23 CA LEU 23 0.218
    CA LEU 24 CA LEU 24 0.233
    CA ASP 25 CA ASP 25 0.160
    CA THR 26 CA THR 26 0.200
    CA GLY 27 CA GLY 27 0.303
    CA ALA 28 CA ALA 28 0.169
    CA ASP 29 CA ASP 29 0.150
    CA ASP 30 CA ASP 30 0.038
    CA THR 31 CA THR 31 0.047
    CA VAL 32 CA VAL 32 0.173
    CA LEU 33 CA LEU 33 0.194
    CA GLU 34 CA GLU 34 0.310
    CA GLU 35 CA GLU 35 0.260
    CA MET 36 CA MET 36 0.136
    CA SER 37 CA SER 37 0.494
    CA LEU 38 CA LEU 38 0.607
    CA PRO 39 CA PRO 39 0.094
    CA GLY 40 CA GLY 40 0.774
    CA ARG 41 CA ARG 41 0.448
    CA TRP 42 CA TRP 42 0.204
    CA LYS 43 CA LYS 43 0.596
    CA PRO 44 CA PRO 44 0.625
    CA LYS 45 CA LYS 45 0.541
    CA MET 46 CA MET 46 0.643
    CA ILE 47 CA ILE 47 0.361
    CA GLY 48 CA GLY 48 0.240
    CA GLY 49 CA GLY 49 0.182
    CA ILE 50 CA ILE 50 0.110
    CA GLY 51 CA GLY 51 0.243
    CA GLY 52 CA GLY 52 0.200
    CA PHE 53 CA PHE 53 0.119
    CA ILE 54 CA ILE 54 0.255
    CA LYS 55 CA LYS 55 0.295
    CA VAL 56 CA VAL 56 0.108
    CA ARG 57 CA ARG 57 0.129
    CA GLN 58 CA GLN 58 0.074
    CA TYR 59 CA TYR 59 0.372
    CA ASP 60 CA ASP 60 0.496
    CA GLN 61 CA GLN 61 0.780
    CA ILE 62 CA ILE 62 0.406
    CA LEU 63 CA LEU 63 0.211
    CA ILE 64 CA ILE 64 0.260
    CA GLU 65 CA GLU 65 0.193
    CA ILE 66 CA ILE 66 0.181
    CA CYS 67 CA CYS 67 0.518
    CA GLY 68 CA GLY 68 0.641
    CA HIS 69 CA HIS 69 0.319
    CA LYS 70 CA LYS 70 0.179
    CA ALA 71 CA ALA 71 0.265
    CA ILE 72 CA ILE 72 0.350
    CA GLY 73 CA GLY 73 0.253
    CA THR 74 CA THR 74 0.301
    CA VAL 75 CA VAL 75 0.187
    CA LEU 76 CA LEU 76 0.186
    CA VAL 77 CA VAL 77 0.070
    CA GLY 78 CA GLY 78 0.306
    CA PRO 79 CA PRO 79 0.047
    CA THR 80 CA THR 80 0.470
    CA PRO 81 CA PRO 81 0.404
    CA VAL 82 CA PHE 82 0.556
    CA ASN 83 CA ASN 83 0.146
    CA ILE 84 CA VAL 84 0.196
    CA ILE 85 CA ILE 85 0.163
    CA GLY 86 CA GLY 86 0.224
    CA ARG 87 CA ARG 87 0.127
    CA ASN 88 CA ASN 88 0.048
    CA LEU 89 CA LEU 89 0.081
    CA LEU 90 CA LEU 90 0.197
    CA THR 91 CA THR 91 0.226
    CA GLN 92 CA GLN 92 0.176
    CA ILE 93 CA ILE 93 0.151
    CA GLY 94 CA GLY 94 0.338
    CA CYS 95 CA CYS 95 0.233
    CA THR 96 CA THR 96 0.305
    CA LEU 97 CA LEU 97 0.089
    CA ASN 98 CA ASN 98 0.260
    CA PHE 99 CA PHE 99 0.250
    CA PRO 101 CA PRO 101 0.227
    CA GLN 102 CA GLN 102 0.108
    CA ILE 103 CA ILE 103 0.206
    CA THR 104 CA THR 104 0.169
    CA LEU 105 CA LEU 105 0.125
    CA TRP 106 CA TRP 106 0.363
    CA GLN 107 CA GLN 107 0.296
    CA ARG 108 CA ARG 108 0.400
    CA PRO 109 CA PRO 109 0.173
    CA LEU 110 CA LEU 110 0.182
    CA VAL 111 CA VAL 111 0.085
    CA THR 112 CA THR 112 0.123
    CA ILE 113 CA ILE 113 0.107
    CA LYS 114 CA LYS 114 0.368
    CA ILE 115 CA ILE 115 0.226
    CA GLY 116 CA GLY 116 0.638
    CA GLY 117 CA GLY 117 0.516
    CA GLN 118 CA GLN 118 0.414
    CA LEU 119 CA LEU 119 0.102
    CA LYS 120 CA LYS 120 0.191
    CA GLU 121 CA GLU 121 0.206
    CA ALA 122 CA ALA 122 0.197
    CA LEU 123 CA LEU 123 0.231
    CA LEU 124 CA LEU 124 0.145
    CA ASP 125 CA ASP 125 0.235
    CA THR 126 CA THR 126 0.311
    CA GLY 127 CA GLY 127 0.200
    CA ALA 128 CA ALA 128 0.102
    CA ASP 129 CA ASP 129 0.143
    CA ASP 130 CA ASP 130 0.261
    CA THR 131 CA THR 131 0.172
    CA VAL 132 CA VAL 132 0.232
    CA LEU 133 CA LEU 133 0.103
    CA GLU 134 CA GLU 134 0.175
    CA GLU 135 CA GLU 135 0.190
    CA MET 136 CA MET 136 0.220
    CA SER 137 CA SER 137 0.739
    CA LEU 138 CA LEU 138 0.277
    CA PRO 139 CA PRO 139 0.325
    CA GLY 140 CA GLY 140 0.390
    CA ARG 141 CA ARG 141 0.174
    CA TRP 142 CA TRP 142 0.168
    CA LYS 143 CA LYS 143 0.304
    CA PRO 144 CA PRO 144 0.194
    CA LYS 145 CA LYS 145 0.456
    CA MET 146 CA MET 146 0.362
    CA ILE 147 CA ILE 147 0.178
    CA GLY 148 CA GLY 148 0.390
    CA GLY 149 CA GLY 149 0.434
    CA ILE 150 CA ILE 150 0.050
    CA GLY 151 CA GLY 151 0.199
    CA GLY 152 CA GLY 152 0.152
    CA PHE 153 CA PHE 153 0.455
    CA ILE 154 CA ILE 154 0.198
    CA LYS 155 CA LYS 155 0.470
    CA VAL 156 CA VAL 156 0.590
    CA ARG 157 CA ARG 157 0.607
    CA GLN 158 CA GLN 158 0.465
    CA TYR 159 CA TYR 159 0.301
    CA ASP 160 CA ASP 160 0.294
    CA GLN 161 CA GLN 161 0.308
    CA ILE 162 CA ILE 162 0.274
    CA LEU 163 CA LEU 163 0.235
    CA ILE 164 CA ILE 164 0.367
    CA GLU 165 CA GLU 165 0.410
    CA ILE 166 CA ILE 166 0.201
    CA CYS 167 CA CYS 167 0.409
    CA GLY 168 CA GLY 168 0.406
    CA HIS 169 CA HIS 169 0.410
    CA LYS 170 CA LYS 170 0.282
    CA ALA 171 CA ALA 171 0.273
    CA ILE 172 CA ILE 172 0.317
    CA GLY 173 CA GLY 173 0.563
    CA THR 174 CA THR 174 0.129
    CA VAL 175 CA VAL 175 0.237
    CA LEU 176 CA LEU 176 0.155
    CA VAL 177 CA VAL 177 0.240
    CA GLY 178 CA GLY 178 0.386
    CA PRO 179 CA PRO 179 0.340
    CA THR 180 CA THR 180 0.335
    CA PRO 181 CA PRO 181 0.446
    CA VAL 182 CA PHE 182 0.343
    CA ASN 183 CA ASN 183 0.205
    CA ILE 184 CA VAL 184 0.262
    CA ILE 185 CA ILE 185 0.096
    CA GLY 186 CA GLY 186 0.118
    CA ARG 187 CA ARG 187 0.202
    CA ASN 188 CA ASN 188 0.073
    CA LEU 189 CA LEU 189 0.108
    CA LEU 190 CA LEU 190 0.127
    CA THR 191 CA THR 191 0.177
    CA GLN 192 CA GLN 192 0.175
    CA ILE 193 CA ILE 193 0.241
    CA GLY 194 CA GLY 194 0.118
    CA CYS 195 CA CYS 195 0.375
    CA THR 196 CA THR 196 0.437
    CA LEU 197 CA LEU 197 0.167
    CA ASN 198 CA ASN 198 0.178
  • Table 4 shows that the 184V, V82F mutations induce structural changes relative to the wild type structure in some parts of the enzyme, but that other regions are less affected. The regions of the protein structure which are not significantly affected by the amino acid mutations are defined as structurally conserved regions. In the present example, the mutations result in localized structural changes in the backbone of HIV protease over a wide range, from 0.038-0.774 Å. [0118]
  • The distances between the strongly interacting atoms of the inhibitor to atoms of the wild type and mutant protein, that is hydrogen-bond donors and acceptors, were computed and they are displayed in Table 5. [0119]
    TABLE 5
    Distances between atoms of the inhibitor and atoms of
    the protein
    HIV PR wt: 1 V82F/I84V: 1
    O2-Wat301 2.92 2.89
    N1-027 3.36 3.46
    O6-N30 3.30 3.61
    06-N29 3.19 3.55
    O7-N29 2.84 2.87
    O7-OD1 29 3.42 3.54
    O7-O1 3.31 3.19
    O3-OD 25 (out) 2.50 2.94
    O3-OD 25 (in) 2.65 2.67
    O3-OD125 (out) 3.27 3.21
    O3-OD125 (in) 2.80 2.67
    O5-Wat301 2.70 2.79
    O8-N130 3.16 2.96
  • Table 5 shows that the atoms of the inhibitor interact with the same atoms of the two different proteins, in this case the wild type and V82F/184V mutant HIV proteases. From Table 5, it can be seen that the atoms of the enzymes with which the inhibitor interacts belong to the structurally conserved regions. The effects of mutations on the protein-inhibitor interactions can be quantified in terms of the distances between interacting pairs of atoms from the inhibitor and from atoms of the three-dimensionally conserved substructure of the protein. These distances are similar in the wild type and in the mutant complexes; the average of their differences is only 0.07 Å. The range of the differences is 0.02-0.36 Å. [0120]
  • EXAMPLE 2
  • This example illustrates the method by which experimentally-determined crystal structures of two different inhibitors in complexes with wild type HIV protease can be compared and analyzed for the existence of a three-dimensionally conserved substructure. The structures of wild type HIV-1 protease in complexes with [0121] inhibitor 1 and with Amprenavir (inhibitor 2) were analyzed by means of (a) an overall superposition of the protein structures; and (b) a study of the distances from atoms of the inhibitors to atoms of the protein.
  • The superposition of the protein structures is performed in a two step process: 1) the distance between all pairs of corresponding Cá atoms (Cá atom of [0122] residue number 1 in one protein to Cá atom of residue number 1 in the second protein; Cá atom of residue number 2 in one protein to Cá atom of residue number 2 in the second protein; and so on) of the polypeptide chains is minimized by means of a least-square algorithm; 2) the superposition is refined by minimizing, in an iterative process, the distances between corresponding Cá atoms that are closer than a given distance (0.25 Å in this example), thus eliminating regions of the structures having large conformational differences to compute the superposition parameters. The distances between equivalenced Cá atoms after the minimization procedure are shown in Table 6.
    TABLE 6
    Distances between equivalent Cá atoms
    Molecule 1: HIV-1 PR wt: 1
    Molecule 2: HIV-1 PR wt: 2
    Molecule 1 Molecule 2 distance [Å]
    CA PRO 1 CA PRO 1 0.200
    CA GLN 2 CA GLN 2 0.320
    CA ILE 3 CA ILE 3 0.147
    CA THR 4 CA THR 4 0.405
    CA LEU 5 CA LEU 5 0.225
    CA TRP 6 CA TRP 6 0.296
    CA GLN 7 CA GLN 7 0.317
    CA ARG 8 CA ARG 8 0.154
    CA PRO 9 CA PRO 9 0.143
    CA LEU 10 CA LEU 10 0.259
    CA VAL 11 CA VAL 11 0.275
    CA THR 12 CA THR 12 0.307
    CA ILE 13 CA ILE 13 0.207
    CA LYS 14 CA LYS 14 0.273
    CA ILE 15 CA ILE 15 0.434
    CA GLY 16 CA GLY 16 0.469
    CA GLY 17 CA GLY 17 0.414
    CA GLN 18 CA GLN 18 0.319
    CA LEU 19 CA LEU 19 0.161
    CA LYS 20 CA LYS 20 0.155
    CA GLU 21 CA GLU 21 0.196
    CA ALA 22 CA ALA 22 0.338
    CA LEU 23 CA LEU 23 0.246
    CA LEU 24 CA LEU 24 0.292
    CA ASP 25 CA ASP 25 0.142
    CA THR 26 CA THR 26 0.109
    CA GLY 27 CA GLY 27 0.176
    CA ALA 28 CA ALA 28 0.193
    CA ASP 29 CA ASP 29 0.087
    CA ASP 30 CA ASP 30 0.118
    CA THR 31 CA THR 31 0.111
    CA VAL 32 CA VAL 32 0.087
    CA LEU 33 CA LEU 33 0.306
    CA GLU 34 CA GLU 34 0.333
    CA GLU 35 CA GLU 35 0.399
    CA MET 36 CA MET 36 0.296
    CA SER 37 CA SER 37 0.454
    CA LEU 38 CA LEU 38 0.451
    CA PRO 39 CA PRO 39 0.397
    CA GLY 40 CA GLY 40 0.444
    CA ARG 41 CA ARG 41 0.535
    CA TRP 42 CA TRP 42 0.346
    CA LYS 43 CA LYS 43 0.442
    CA PRO 44 CA PRO 44 0.548
    CA LYS 45 CA LYS 45 0.307
    CA MET 46 CA MET 46 0.320
    CA ILE 47 CA ILE 47 0.403
    CA GLY 48 CA GLY 48 0.237
    CA GLY 49 CA GLY 49 0.280
    CA ILE 50 CA ILE 50 0.206
    CA GLY 51 CA GLY 51 0.368
    CA GLY 52 CA GLY 52 0.315
    CA PHE 53 CA PHE 53 0.378
    CA ILE 54 CA ILE 54 0.180
    CA LYS 55 CA LYS 55 0.149
    CA VAL 56 CA VAL 56 0.302
    CA ARG 57 CA ARG 57 0.098
    CA GLN 58 CA GLN 58 0.219
    CA TYR 59 CA TYR 59 0.279
    CA ASP 60 CA ASP 60 0.385
    CA GLN 61 CA GLN 61 0.431
    CA ILE 62 CA ILE 62 0.343
    CA LEU 63 CA LEU 63 0.473
    CA ILE 64 CA ILE 64 0.344
    CA GLU 65 CA GLU 65 0.456
    CA ILE 66 CA ILE 66 0.481
    CA CYS 67 CA CYS 67 0.920
    CA GLY 68 CA CLY 68 0.999
    CA HIS 69 CA HIS 69 0.295
    CA LYS 70 CA LYS 70 0.406
    CA ALA 71 CA ALA 71 0.446
    CA ILE 72 CA ILE 72 0.374
    CA GLY 73 CA GLY 73 0.259
    CA THR 74 CA THR 74 0.276
    CA VAL 75 CA VAL 75 0.165
    CA LEU 76 CA LEU 76 0.220
    CA VAL 77 CA VAL 77 0.202
    CA GLY 78 CA GLY 78 0.231
    CA PRO 79 CA PRO 79 0.131
    CA THR 80 CA THR 80 0.374
    CA PRO 81 CA PRO 81 0.472
    CA VAL 82 CA VAL 82 0.554
    CA ASN 83 CA ASN 83 0.149
    CA ILE 84 CA ILE 84 0.261
    CA ILE 85 CA ILE 85 0.223
    CA GLY 86 CA GLY 86 0.130
    CA ARG 87 CA ARG 87 0.165
    CA ASN 88 CA ASN 88 0.103
    CA LEU 89 CA LEU 89 0.072
    CA LEU 90 CA LEU 90 0.076
    CA THR 91 CA THR 91 0.114
    CA GLN 92 CA GLN 92 0.115
    CA ILE 93 CA ILE 93 0.204
    CA GLY 94 CA GLY 94 0.220
    CA CYS 95 CA CYS 95 0.068
    CA THR 96 CA THR 96 0.185
    CA LEU 97 CA LEU 97 0.095
    CA ASN 98 CA ASN 98 0.311
    CA PHE 99 CA PHE 99 0.216
    CA PRO 101 CA PRO 101 0.455
    CA GLN 102 CA GLN 102 0.121
    CA ILE 103 CA ILE 103 0.120
    CA THR 104 CA THR 104 0.109
    CA LEU 105 CA LEU 105 0.128
    CA TRP 106 CA TRP 106 0.205
    CA GLN 107 CA GLN 107 0.229
    CA ARG 108 CA ARG 108 0.211
    CA PRO 109 CA PRO 109 0.195
    CA LEU 110 CA LEU 110 0.135
    CA VAL 111 CA VAL 111 0.086
    CA THR 112 CA THR 112 0.166
    CA ILE 113 CA ILE 113 0.199
    CA LYS 114 CA LYS 114 0.333
    CA ILE 115 CA ILE 115 0.356
    CA GLY 116 CA GLY 116 0.671
    CA GLY 117 CA GLY 117 0.709
    CA GLN 118 CA GLN 118 0.370
    CA LEU 119 CA LEU 119 0.258
    CA LYS 120 CA LYS 120 0.156
    CA GLU 121 CA GLU 121 0.250
    CA ALA 122 CA ALA 122 0.276
    CA LEU 123 CA LEU 123 0.103
    CA LEU 124 CA LEU 124 0.112
    CA ASP 125 CA ASP 125 0.078
    CA THR 126 CA THR 126 0.057
    CA GLY 127 CA GLY 127 0.121
    CA ALA 128 CA ALA 128 0.098
    CA ASP 129 CA ASP 129 0.190
    CA ASP 130 CA ASP 130 0.302
    CA THR 131 CA THR 131 0.073
    CA VAL 132 CA VAL 132 0.178
    CA LEU 133 CA LEU 133 0.147
    CA GLU 134 CA GLU 134 0.239
    CA GLU 135 CA GLU 135 0.101
    CA MET 136 CA MET 136 0.235
    CA SER 137 CA SER 137 0.391
    CA LEU 138 CA LEU 138 0.364
    CA PRO 139 CA PRO 139 0.532
    CA GLY 140 CA GLY 140 0.213
    CA ARG 141 CA ARG 141 0.448
    CA TRP 142 CA TRP 142 0.133
    CA LYS 143 CA LYS 143 0.195
    CA PRO 144 CA PRO 144 0.082
    CA LYS 145 CA LYS 145 0.359
    CA MET 146 CA MET 146 0.306
    CA ILE 147 CA ILE 147 0.076
    CA GLY 148 CA GLY 148 0.214
    CA GLY 149 CA GLY 149 0.205
    CA ILE 150 CA ILE 150 0.163
    CA GLY 151 CA GLY 151 0.287
    CA GLY 152 CA GLY 152 0.318
    CA PHE 153 CA PHE 153 0.125
    CA ILE 154 CA ILE 154 0.189
    CA LYS 155 CA LYS 155 0.384
    CA VAL 156 CA VAL 156 0.510
    CA ARG 157 CA ARG 157 0.405
    CA GLN 158 CA GLN 158 0.139
    CA TYR 159 CA TYR 159 0.361
    CA ASP 160 CA ASP 160 0.252
    CA GLN 161 CA GLN 161 0.414
    CA ILE 162 CA ILE 162 0.337
    CA LEU 163 CA LEU 163 0.202
    CA ILE 164 CA ILE 164 0.359
    CA GLU 165 CA GLU 165 0.463
    CA ILE 166 CA ILE 166 0.347
    CA CYS 167 CA CYS 167 0.256
    CA GLY 168 CA GLY 168 0.471
    CA HIS 169 CA HIS 169 0.658
    CA LYS 170 CA LYS 170 0.489
    CA ALA 171 CA ALA 171 0.445
    CA ILE 172 CA ILE 172 0.396
    CA GLY 173 CA GLY 173 0.523
    CA THR 174 CA THR 174 0.130
    CA VAL 175 CA VAL 175 0.156
    CA LEU 176 CA LEU 176 0.077
    CA VAL 177 CA VAL 177 0.129
    CA GLY 178 CA GLY 178 0.276
    CA PRO 179 CA PRO 179 0.272
    CA THR 180 CA THR 180 0.580
    CA PRO 181 CA PRO 181 0.436
    CA VAL 182 CA VAL 182 0.328
    CA ASN 183 CA ASN 183 0.180
    CA ILE 184 CA ILE 184 0.151
    CA ILE 185 CA ILE 185 0.104
    CA GLY 186 CA GLY 186 0.059
    CA ARG 187 CA ARG 187 0.058
    CA ASN 188 CA ASN 188 0.183
    CA LEU 189 CA LEU 189 0.164
    CA LEU 190 CA LEU 190 0.051
    CA THR 191 CA THR 191 0.216
    CA GLW 192 CA GLN 192 0.162
    CA ILE 193 CA ILE 193 0.158
    CA GLY 194 CA GLY 194 0.047
    CA CYS 195 CA CYS 195 0.050
    CA THR 196 CA THR 196 0.200
    CA LEU 197 CA LEU 197 0.165
    CA ASN 198 CA ASN 198 0.074
  • The distances between the atoms of the [0123] inhibitors 1 and 2 to atoms of the protein, that is, hydrogen-bond donors and acceptors, were computed and are shown in Table 7.
    TABLE 7
    Distances between atoms of inhibitors and atoms of
    the proteins
    Wt: 1 complex Wt: 2 complex
    O2-Wat301 2.92 3.02
    N1-027 3.36 3.58
    O6-N30 3.30 3.50
    06-N29 3.19 3.51
    O7-N29 2.84
    O7-OD1 29 3.42
    O7-O1 3.31
    O3-OD 25 (out) A 2.50 2.80
    O3-OD 25 (in) A 2.65 2.66
    O3-OD 25 (out) B 3.27 3.07
    O3-OD 25 (in) B 2.80 2.68
    O5-Wat301 2.70 2.77
    O8-N 30 3.16
    N3-N 30 3.17
    N3-OD2 30 3.15
  • Inhibitors 1 (FIG. 1) and 2 (Amprenavir) have similar structural elements, in particular their core, i.e. groups at the P1-[0124] P1′ 60 positions. However, 2 has a THF group while 1 has a bis-THF group at the P2′ position. The P2 groups are identical except for the substitution of an ether oxygen atom in 1 as compared to an amine nitrogen atom at the same position in 2. Table 7 shows that 1 forms more interactions with the atoms of the protein that were previously identified as belonging to the structurally conserved substructure than does compound 2. For example, the O7 oxygen atom in compound 1, that forms an interaction with N29 nitrogen of the protease, has no counterpart in compound 2. Instead, the O6 oxygen atom of 2 forms longer (and presumably weaker) hydrogen bonds with both N30 (3.50 Å) and N29 (3.51 Å). In contrast, the O6 oxygen of compound 1 forms a shorter (and presumably stronger) hydrogen bond with N29 (3.19 Å). Additionally, as can be seen in Table 7, where both compounds 1 and 2 form interactions with atoms in the structurally conserved substructure of HIV protease, the distances between interacting atoms are consistently shorter for compound 1, indicative of presumably stronger binding interactions.
  • Examples 1 and 2 were used to identify a three dimensionally-conserved substructure of HIV protease that is involved in the binding of HIV protease inhibitors and, in particular, to identify atoms of these substructural elements that are involved in forming interactions with atoms of HIV protease inhibitors. This substructure is defined by the set of atomic coordinates (in orthogonal coordinates) provided in Table 8 and any equivalent set derived by applying arbitrary rotations and translations to the set of atomic coordinates in Table 8. The values of the coordinates (X,Y,Z) of the atoms defining the substructure are affected by a standard error σ. Therefore (X,Y,Z) values for each atom are those defined in the intervals (X−σ, X+σ) for coordinate X, (Y−σ, Y+σ) for coordinate Y, and (Z−σ, Z+σ) for coordinate Z. [0125]
    TABLE 8
    Three dimensionally-conserved substructure of HIV protease
    Atom X [Å] Y [Å] Z [Å] σ [Å] Description
    Substructure of the protein atoms
    Oxygen −7.9 13.6 27.4 0.5 Oxygen atom of water molecule
    coordinated to main chain amide
    nitrogen atoms of amino acid
    Gly 49 and Gly 149
    O27 −13.8 17.7 30.4 0.5 Main Chain carbonyl oxygen
    atom of amino acid Gly 27
    N29 −13.4 18.2 34.5 0.5 Main chain amide nitrogen
    atom of amino acid Asp 29
    N30 −11.9 18.6 36.7 0.5 Main chain amide nitrogen
    atom of amino acid Asp 30
    OD1 25 −11.3 21.2 28.7 0.5 Carboxylate oxygen atom of
    aminoacid Asp 25
    OD2 25 −9.4 20.4 29.3 0.5 Carboxylate oxygen atom of
    aminoacid Asp 25
    OD1 125 −12.7 20.3 26.4 0.5 Carboxylate oxygen atom of
    aminoacid Asp 125
    OD2 125 −12.7 20.3 26.4 0.5 Carboxylate oxygen atom of
    aminoacid Asp 125
    N129 −8.9 20.5 20.7 0.5 Main chain amide nitrogen atom
    of amino acid Asp 129
    N130 −10.1 19.5 18.6 0.5 Main chain amide nitrogen atom
    of amino acid Asp 130
    Substructure of the inhibitor atoms
    Hydrogen −8.8 17.5 25.7 0.5 Interacting with main chain
    Bond carbonyl oxygen atom of amino
    donor acid Gly 27
    Atom
    Hydrogen −8.5 15.3 25.1 0.5 Interacting with Oxygen atom of
    Bond water molecule coordinated to
    acceptor main chain amide nitrogen
    Atom atoms of amino acid Gly 49 and
    Gly 149
    Hydrogen −10.4 19.1 27.4 0.5 Interacting with carboxylate
    Bond oxygen atoms of aminoacids
    donor- Asp 25 and Asp 125
    acceptor
    Atom
    Hydrogen −8.9 14.0 29.8 0.5 Interacting with Oxygen atom of
    Bond water molecule coordinated to
    acceptor main chain amide nitrogen
    Atom atoms of amino acid Gly 49 and
    Gly 149
    Hydrogen −8.6 17.3 20.7 0.5 Main chain amide nitrogen atom
    Bond of amino acid Asp 30
    acceptor
    Atom
    Hydrogen −6.9 18.7 21.4 0.5 Interacting with main chain
    Bond amide nitrogen atom of amino
    acceptor acid Asp 29
    Atom
    O8 −10.7 15.8 35.8 0.5 Interacting with main chain
    amide nitrogen atom of amino
    acid Asp 130
  • EXAMPLE 3
  • The following example demonstrates that a protease inhibitor that contains atoms that can make favorable interactions with the atoms of the substructure may exhibit broad spectrum activity. [0126]
  • Compounds 1 and 3 contain a Bis-THF group at the P2 position that contains two atoms, in particular, hydrogen bond acceptor oxygen atoms, that can form hydrogen bonds with the two hydrogen atoms attached to the backbone amide nitrogen atoms on the protein at [0127] residues 29 and 30. Compound 2 is similar to 1 except that 2 contains a THF group at P2 with only a single hydrogen bond acceptor oxygen atom. All three compounds differ in the P2′ substituent. Compounds 1 and 3 both are unaffected by the two active site mutations, V82F and 184V, and Ki values for wild type and mutant enzymes are similar for both compounds. In contrast, compound 2, which contains only a single hydrogen bond acceptor atom in the P2 substitutent, is dramatically affected by the active site mutations, which demonstrate high level resistance to 2.
  • The antiviral activity of [0128] compounds 1 and 3 against HIV derived from patient isolates that contain multiple mutations are equivalent to their activity against wild type HIV strains. In contrast, compound 2 is much less effective against the same mutant viruses. None of the patients from whom virus was isolated had ever been exposed to any of the compounds tested herein. Nonetheless, compound 2 exhibited cross resistance to these virus strains that is typically seen with all clinically useful HIV protease inhibitors −4 (Saquinavir), 5 (Ritonavir), 6 (Indinavir) and 7 (Nelfinavir). Compounds 2, 4, 5, 6, and 7 have very different chemical structures, but nonetheless behave as a single class with respect to their antiviral behavior against wild type and multidrug resistant HIV strains. All compounds are dramatically less potent against the multidrug resistant strains of HIV.
  • In sharp contrast, compounds 1 and 3, which closely resemble each other as well as [0129] compound 2, exhibit broad spectrum activity in that they are equally effective against wild type and mutant HIV strains that exhibit high level multidrug resistance towards compounds 2, 4, 5, 6, and 7. The broad spectrum activity of compound 1 was completely unexpected and contrasts with the common and typical loss of antiviral potency experienced with compounds like 2, 4, 5, 6, 7, and indeed most other HIV protease inhibitors represented as similar or different structures that have been reported.
  • The development and application of the 3D motif method described above successfully revealed the presence of a unique, three dimensionally-conserved substructure of HIV protease that is useful in the design of broad spectrum inhibitors. Based on this method, [0130] compound 3 was predicted, on the basis of comparative molecular modeling using the coordinates of the complexes of compound 1 with wild type and V82F/184V mutant HIV proteases, to be able to make the same key interaction as compound 1 and thereby to exhibit broad spectrum activity. Based on these data, it is feasible to design protease inhibitors that are predicted to have broad spectrum activity, and are predicted to be useful for the treatment of both wild type (first line therapy) and drug resistant (salvage therapy) HIV infections.
  • EXAMPLE 4
  • This example illustrates the method by which experimentally-determined crystal structures of two different target proteins, DHQases, from two different bacterial species can be compared and analyzed for the existence of a three-dimensionally conserved substructure even in the absence of readily discernible or statistically significant sequence similarity. DHQases from different bacterial species typically exhibit less than 30% sequence identity (FIG. 2). A schematic map showing the key interactions of the substrate-based inhibitor, DHQO, with the active site residues for the Type II DHQase from [0131] M. tuberculosis is provided in FIG. 3.
  • The structures of wild type DHQase from [0132] M. tuberculosis and a homologous DHQase from Pseudomonas putidas were determined using conventional x-ray crystallography techniques. The structures were analyzed by means of (a) an overall superposition of the atoms of the protein structures. This analysis requires three dimensional atomic coordinates of the protein structures.
  • The superposition of the protein structures was performed in a two step process: 1) the distance between all pairs of corresponding Cá atoms (Cá atom of [0133] residue number 1 in one protein to Cá atom of residue number 1 in the second protein; Cá atom of residue number 2 in one protein to Cá atom of residue number 2 in the second protein; and so on) of the polypeptide chains is minimized by means of a least-square algorithm; 2) the superposition is refined by minimizing, in an iterative process, the distances between corresponding Cá atoms that are closer than a given distance (0.4 Å in this example), thus eliminating regions of the structures having large conformational differences to compute the superposition parameters. The distances between equivalenced Cá atoms after the minimization procedure are shown in Table 9.
    TABLE 9
    Distances between equivalent Cá atoms
    Molecule 1: DHQase P. putida wt: qxa
    Molecule 2: DHQase M. tuberculosis wt: gt33
    Molecule 1 Molecule 2 distance [Å]
    CA MET 2 CA GLU 2 1.078
    CA ALA 3 CA LEU 3 1.504
    CA THR 4 CA ILE 4 1.800
    CA LEU 5 CA VAL 5 1.283
    CA LEU 6 CA ASN 6 0.911
    CA VAL 7 CA VAL 7 0.715
    CA LEU 8 CA ILE 8 0.298
    CA HIS 9 CA ASN 9 0.211
    CA GLY 10 CA GLY 10 0.591
    CA PRO 11 CA PRO 11 0.599
    CA ASN 12 CA ASN 12 0.487
    CA LEU 13 CA LEU 13 0.428
    CA ASN 14 CA GLY 14 0.229
    CA LEU 15 CA ARG 15 0.685
    CA LEU 16 CA LEU 16 0.541
    CA GLY 17 CA GLY 17 1.693
    CA THR 18 CA ARG 18 2.287
    CA ARG 19 CA ARG 19 2.956
    CA GLN 20 CA GLN 20 3.475
    CA PRO 21 CA PRO 21 3.390
    CA GLY 22 CA ALA 22 4.037
    CA THR 23 CA VAL 23 3.770
    CA TYR 24 CA TYR 24 2.521
    CA GLY 25 CA GLY 25 1.170
    CA SER 26 CA GLY 26 1.642
    CA THR 27 CA THR 27 1.454
    CA THR 28 CA THR 28 1.532
    CA LEU 29 CA HIS 29 1.471
    CA GLY 30 CA ASP 30 1.632
    CA GLN 31 CA GLU 31 1.966
    CA ILE 32 CA LEU 32 1.586
    CA ASN 33 CA VAL 33 1.875
    CA GLN 34 CA ALA 34 2.230
    CA ASP 35 CA LEU 35 2.343
    CA LEU 36 CA ILE 36 1.927
    CA GLU 37 CA GLU 37 2.284
    CA ARG 38 CA ARG 38 2.980
    CA ARG 39 CA GLU 39 2.917
    CA ALA 40 CA ALA 40 2.719
    CA ARG 41 CA ALA 41 3.367
    CA GLU 42 CA GLU 42 3.534
    CA ALA 43 CA LEU 43 3.281
    CA GLY 44 CA GLY 44 3.161
    CA HIS 45 CA LEU 45 2.899
    CA HIS 46 CA LYS 46 1.844
    CA LEU 47 CA ALA 47 1.599
    CA LEU 48 CA VAL 48 1.201
    CA HIS 49 CA VAL 49 2.053
    CA LEU 50 CA ARG 50 1.045
    CA GLN 51 CA GLN 51 0.266
    CA SER 52 CA SER 52 0.300
    CA ASN 53 CA ASP 53 0.282
    CA ALA 54 CA SER 54 0.348
    CA GLU 55 CA GLU 55 0.326
    CA TYR 56 CA ALA 56 0.238
    CA GLU 57 CA GLN 57 0.380
    CA LEU 58 CA LEU 58 0.455
    CA ILE 59 CA LEU 59 0.413
    CA ASP 60 CA ASP 60 0.984
    CA ARG 61 CA TRP 61 1.452
    CA ILE 62 CA ILE 62 1.338
    CA HIS 63 CA HIS 63 1.310
    CA ALA 64 CA GLN 64 2.327
    CA ALA 65 CA ALA 65 2.526
    CA ARG 66 CA ALA 66 3.063
    CA ASP 67 CA ASP 67 3.449
    CA GLU 68 CA
    CA GLY 69 CA ALA 68 2.318
    CA VAL 70 CA ALA 69 1.691
    CA ASP 71 CA GLU 70 0.812
    CA PHE 72 CA PRO 71 0.515
    CA ILE 73 CA VAL 72 0.561
    CA ILE 74 CA ILE 73 0.547
    CA LEU 75 CA LEU 74 0.380
    CA ASN 76 CA ASN 75 0.277
    CA PRO 77 CA ALA 76 0.369
    CA ALA 78 CA GLY 77 0.952
    CA ALA 79 CA GLY 78 0.421
    CA PHE 80 CA LEU 79 0.714
    CA THR 81 CA THR 80 0.575
    CA HIS 82 CA HIS 81 0.142
    CA THR 83 CA THR 82 0.222
    CA SER 84 CA SER 83 0.741
    CA VAL 85 CA VAL 84 0.719
    CA ALA 86 CA ALA 85 0.415
    CA LEU 87 CA LEU 86 0.667
    CA ARG 88 CA ARG 87 0.660
    CA ASP 89 CA ASP 88 0.426
    CA ALA 90 CA ALA 89 0.697
    CA LEU 91 CA CYS 90 1.233
    CA LEU 92 CA ALA 91 1.319
    CA ALA 93 CA GLU 92 2.852
    CA VAL 94 CA LEU 93 4.165
    CA SER 95 CA SER 94 3.605
    CA ILE 96 CA ALA 95 3.840
    CA PRO 97 CA PRO 96 2.414
    CA PHE 98 CA LEU 97 0.314
    CA ILE 99 CA ILE 98 0.251
    CA GLU 100 CA GLU 99 0.095
    CA VAL 101 CA VAL 100 0.131
    CA HIS 102 CA HIS 101 0.318
    CA ILE 103 CA ILE 102 0.117
    CA SER 104 CA SER 103 0.229
    CA ASN 105 CA ASN 104 0.203
    CA VAL 106 CA VAL 105 0.193
    CA HIS 107 CA HIS 106 0.499
    CA LYS 108 CA ALA 107 0.498
    CA ARG 109 CA ARG 108 0.292
    CA GLU 110 CA GLU 109 0.333
    CA PRO 111 CA GLU 110 0.377
    CA PHE 112 CA PHE 111 0.651
    CA ARG 113 CA ARG 112 0.611
    CA ARG 114 CA ARG 113 0.469
    CA HIS 115 CA HIS 114 0.467
    CA SER 116 CA SER 115 0.293
    CA TYR 117 CA TYR 116 0.483
    CA PHE 118 CA LEU 117 0.468
    CA SER 119 CA SER 118 0.367
    CA ASP 120 CA PRO 119 0.676
    CA VAL 121 CA ILE 120 0.445
    CA ALA 122 CA ALA 121 0.334
    CA VAL 123 CA THR 122 0.405
    CA GLY 124 CA GLY 123 0.372
    CA VAL 125 CA VAL 124 0.375
    CA ILE 126 CA ILE 125 0.250
    CA CYS 127 CA VAL 126 0.328
    CA GLY 128 CA GLY 127 0.332
    CA LEU 129 CA LEU 128 0.473
    CA GLY 130 CA GLY 129 0.272
    CA ALA 131 CA ILE 130 0.551
    CA THR 132 CA GLN 131 0.564
    CA GLY 133 CA GLY 132 0.289
    CA TYR 134 CA TYR 133 0.276
    CA ARG 135 CA LEU 134 0.476
    CA LEU 136 CA LEU 135 0.556
    CA ALA 137 CA ALA 136 0.677
    CA LEU 138 CA LEU 137 0.703
    CA GLU 139 CA ARG 138 0.861
    CA SER 140 CA TYR 139 0.876
    CA ALA 141 CA LEU 140 1.330
    CA LEU 142 CA ALA 141 1.529
    CA GLU 143 CA GLU 142 1.492
    CA GLN 144 CA HIS 143 1.738
    CA LEU 145 CA VAL 144 3.487
  • Table 9 shows that the two structures are remarkably similar overall despite their low level sequence identity. However, the structures exhibit very large deviations in some regions, and are highly conserved in others. In particular, this analysis reveals that regions of the enzyme are minimally affected by the large number of amino acid sequence substitutions. The regions of the protein structure which are not significantly affected by the amino acid substitutions are defined as structurally conserved regions. In the present example, the substitutions result in localized structural changes in the backbone of DHQase over a wide range, from 0.095-4.165 Å. [0134]
  • The distances between the strongly interacting atoms of the inhibitor to atoms of the homologous DHQase proteins, that is [0135] P. putida wt: qxa and M. tuberculosis wt: gt33 complexes, were computed and they are displayed in Tables 10 and 11, respectively.
    TABLE 10
    Distances between atoms of the
    inhibitor and atoms of the protein
    DHQase P. putida wt: gxa
    distance [Å]
    C6-PRO11 (O) 3.31
    C6-ASN12 (CB) 3.10
    N7-TYR24 (OH) 2.45
    O12-ASN76 (HD2) 2.96
    O13-HIS102 (CB) 3.38
    O12-SER104 (N) 3.35
    C6-ASN12 (CB) 3.10
  • [0136]
    TABLE 11
    Distances between atoms of the
    inhibitor and atoms of the protein
    DHQase M. tuberculosis wt: gt33
    distance [Å]
    N14-PRO11 (O) 3.35
    O15-PRO11 (O) 3.01
    O15-LEU 13 (CG) 3.36
    O15-ARG 19 (NH1) 3.26
    O15-ARG 19 (NH2) 3.32
    O7-ASN 75 (OD1) 2.50
    O13-ASN 75 (ND2) 2.98
    N14-GLY 77 (CA) 3.01
    O9-HIS 81 (NE2) 2.82
    O7-HIS 101 (ND1) 3.25
    O11-ILE 102 (N) 3.33
    O13-ILE 102 (N) 2.77
    O11-SER 103 (N) 2.96
    O11-SER 103 (OG) 2.68
    O9-ARG 112 (NH2) 3.09
    O10-ARG 112 (NH2) 3.02
  • The methods of Examples 1-3 were applied to the DHQase data to identify a three dimensionally-conserved substructure of DHQase that is involved in the binding of DHQase inhibitors, in particular, to identify the relevant target substructure for developing broad spectrum inhibitors. This substructure is defined by the set of atomic coordinates (in orthogonal coordinates) provided in Table 12 and any equivalent set derived by applying arbitrary rotations and translations to the set of atomic coordinates in Table 12. The values of the coordinates (X,Y,Z) of the atoms defining the substructure are affected by a standard error σ. Therefore (X,Y,Z) values for each atom are those defined in the intervals (X−σ, X+σ) for coordinate X, (Y−σ, Y+σ) for coordinate Y, and (Z−σ, Z+σ) for coordinate Z. [0137]
    TABLE 12
    Three dimensionally-conserved substructure of DHQase, M. tuberculosis
    Atom X [Å] Y [Å] Z [Å] σ [Å] Description
    Substructure of the protein atoms
    OD1 26.265 68.912 21.219 0.5 Side chain carbonyl
    ASN75 oxygen atom of amino
    acid ASN 75
    ND2 27.336 66.960 20.951 0.5 Side chain nitrogen
    ASN 75 atom of amino acid
    ASN 75
    NE2 28.343 76.425 22.111 0.5 Side chain nitrogen
    HIS 81 atom of amino acid
    HIS 81
    ND1 28.079 70.604 23.662 0.5 Side chain nitrogen
    HIS 101 atom of amino acid
    HIS 101
    N ILE 31.227 67.167 22.168 0.5 Main chain amide
    102 nitrogen atom of
    atom acid ILE 102
    N SER 33.754 68.315 21.558 0.5 Main chain amide
    103 nitrogen atom of
    amino acid Ser 103
    OG SER 33.946 71.059 20.735 0.5 Side chain hydroxyl
    103 oxygen atom of amino
    acid SER
    103
    Substructure of the inhibitor
    Hydrogen 29.600 68.554 20.298 0.5 Interacting with main
    bond chain nitrogen atom of
    acceptor ILE 102 and side chain
    atom nitrogen atom of ASN
    75
    Hydrogen 28.031 70.739 20.422 0.5 Interacting with side
    bond chain oxygen atom of
    donor- ASN75 and side chain
    acceptor nitrogen atom of
    atom HIS 101
    Hydrogen 29.664 74.658 20.493 0.5 Interacting side chain
    bond nitrogen atom of HIS
    donor- 81
    acceptor
    atom
    Hydrogen 31.451 69.835 20.531 0.5 Interacting with main
    bond chain nitrogen atom
    acceptor and side chain oxygen
    atom atom of SER 103
  • Other Embodiments
  • All publications and patent applications, and patents mentioned in this specification are herein incorporated by reference. [0138]
  • While the invention has been described in connection with specific embodiments, it will be understood that it is capable of further modifications. Therefore, this application is intended to cover any variations, uses, or adaptations of the invention that follow, in general, the principles of the invention, including departures from the present disclosure that come within known or customary practice within the art.[0139]

Claims (51)

Other embodiments are within the claims. What we claim is:
1. A method for the structure-based design of a drug that can act as an inhibitor of at least two different biological entities, said method comprising the steps of:
(a) providing at least one structure of a wild type target protein or an inhibitor-wild type target protein complex;
(b) providing at least one structure of a variant target protein or an inhibitor-variant target protein complex;
(c) comparing at least one structure from step (a) with at least one structure from step (b) to determine whether there exists a common three-dimensionally conserved substructure comprising the atomic coordinates of the structurally conserved atoms of the inhibitors and structurally conserved atoms of the target proteins; and
(d) if a conserved substructure exists, using said atomic coordinates of said conserved substructure to select a compound having atoms matching those of said structurally conserved atoms of the inhibitors, wherein the selection of said compound is performed using computer modeling.
2. A method for the structure-based drug design of a broad spectrum inhibitor, said method comprising the steps of:
(a) providing at least one structure of a wild type target protein or an inhibitor-wild type target protein complex;
(b) providing at least one structure of a variant target protein or an inhibitor-variant target protein complex;
(c) comparing at least one structure from step (a) with at least one structure from step (b) to determine whether there exists a common three-dimensionally conserved substructure comprising the atomic coordinates of the structurally conserved atoms the target proteins or a common three-dimensionally conserved substructure comprising the atomic coordinates of the structurally conserved atoms of the inhibitors and structurally conserved atoms of the target proteins; and
(d) if a conserved substructure exists, using said atomic coordinates of said conserved substructure to select a compound having atoms matching those of said structurally conserved atoms of the inhibitors or to design a compound that binds to said target protein, wherein the selection of said compound is performed using computer modeling.
3. The method of claim 1, further comprising the steps of:
(e) comparing at least one structure from step (a) with at least one structure from step (b) to determine whether there exists a three-dimensionally non-conserved substructure comprising the atomic coordinates of the structurally non-conserved atoms of the inhibitors and structurally non-conserved atoms of the target proteins; and
(f) if a non-conserved substructure exists, using said atomic coordinates of said non-conserved substructure to reject a compound having atoms matching those of said structurally non-conserved atoms of the inhibitors, wherein the rejection of said compound is performed in conjunction with computer modeling.
4. The method of claim 1, wherein at least two structures from step b are used in step c.
5. The method of claim 4, wherein at least four structures from step b are used in step c.
6. The method of claim 4, wherein said target proteins comprise at least two variant forms.
7. The method of claim 6, wherein said target proteins comprise at least four variant forms
8. The method of claim 1, wherein the inhibitors in said inhibitor-wild type target protein complex and said inhibitor-variant target protein complex are the same.
9. The method of claim 1, wherein the inhibitors in said inhibitor-wild type target protein complex and said inhibitor-variant target protein complex are different.
10. The method of claim 1, wherein said inhibitors are competitive inhibitors.
11. The method of claim 1, wherein said inhibitors are noncompetitive inhibitors.
12. The method of claim 1, wherein said inhibitors are reversible inhibitors.
13. The method of claim 1, wherein said inhibitors are irreversible inhibitors.
14. The method of claim 1, wherein said variant target protein is a homologous target protein.
15. The method of claim 1, wherein said variant target protein is a mutant target protein.
16. The method of claim 1, wherein at least one of said structures is a crystal structure.
17. The method of claim 1, wherein at least one of said structures is an nmr structure.
18. The method of claim 1, wherein at least one of said structures is derived using computational methods.
19. The method of claim 1, wherein said target protein is expressed in a microbe and said microbe is selected from the group consisting of viruses, bacteria, protozoa, or fungi.
20. The method of claim 1, wherein said target protein is expressed in a neoplasm.
21. The method of claims 19 or 20, wherein said target protein is selected from the group consisting of an enzyme, a receptor, a structural protein, a component of a macromolecular complex, a component of a metabolic pathway, or an assembly of biological molecules.
22. The method of claim 21, wherein said enzyme is selected from the group consisting of reverse transcriptases, proteases, DNA and RNA polymerases, methylases, oxidases, hydratases, esterases, acyl transferases, helicases, topoisomerases, and kinases.
23. The method of claim 22, wherein said enzyme is HIV protease.
24. The method of claim 23, wherein said inhibitors are selected from the group consisting of indinavir, nelfinavir, ritonavir, saquinavir, amprenavir, lopinavir, and UIC-94003.
25. The method of claim 23, wherein said structurally conserved atoms of the inhibitor and structurally conserved atoms of the protease have the atomic structural coordinates as provided in Table 8.
26. The method of claim 22, wherein said enzyme is 3-dehydroquinate dehydratase.
27. The method of claim 26, wherein said structurally conserved atoms of the 3-dehydroquinate dehydratase have the atomic structural coordinates as provided in Table 12.
28. A compound having a chemical structure selected using the method of claim 19, wherein said compound has broad spectrum activity against wild type and variant microbes.
29. A compound having a chemical structure selected using the method of claim 20, wherein said compound has broad spectrum activity against wild type and variant neoplasms.
30. The compound of claims 28 or 29, wherein said compound has an IC50, variant/IC50, wild type ratio of less than 20.
31. The compound of claim 30, wherein said IC50, variant/IC50, wildtype ratio is less than 6.
32. The compound of claims 28 or 29, wherein said compound has broad spectrum activity against at least 3 mutant biological entities.
33. The compound of claim 28, wherein said compound has broad spectrum activity against at least 3 different organisms expressing homologous target proteins.
34. A pharmaceutical composition comprising a compound of claim 28 and a pharmaceutically acceptable carrier or diluent.
35. A pharmaceutical composition comprising a compound of claim 29 and a pharmaceutically acceptable carrier or diluent.
36. A compound having a chemical structure selected using the method of any one of claims 23-25, wherein said compound has broad spectrum activity against HIV protease.
37. The compound of claim 36, wherein said compound has an IC50, variant/IC50, wild type ratio of less than 10.
38. The compound of claim 37, wherein said IC50, variant/IC50, wild type ratio is less than 6.
39. The compound of claim 36, wherein said compound has broad spectrum activity against at least 3 mutant biological entities.
40. A pharmaceutical composition comprising a compound of claim 36 and a pharmaceutically acceptable carrier or diluent.
41. A compound having a chemical structure selected using the method of claims 26 or 27, wherein said compound has broad spectrum activity against 3-dehydroquinate dehydratase.
42. The compound of claim 41, wherein said compound has an IC50, variant/IC50, wild type ratio of less than 20.
43. The compound of claim 42, wherein said IC50, variant/IC50, wild type ratio is less than 10.
44. The compound of claim 43, wherein said compound has broad spectrum activity against at least 3 mutant biological entities.
45. The compound of claim 41, wherein said compound has broad spectrum activity against at least 3 different organisms expressing homologous target proteins.
46. A pharmaceutical composition comprising a compound of claim 41 and a pharmaceutically acceptable carrier or diluent.
47. A method of treating a microbial infection in a patient, said method comprising the step of administering to said patient a pharmaceutical composition of claim 34 in an amount effective to prevent or treat said infection.
48. A method of treating a neoplasm in a patient in need thereof, said method comprising the step of administering to said patient a pharmaceutical composition of claim 35 in amounts effective to treat said neoplasm.
49. A method of treating an HIV infection in a patient in need thereof, said method comprising the step of administering to said patient a pharmaceutical composition of claim 40 in amounts effective to treat said infection.
50. A method of treating a bacterial infection in a patient in need thereof, said method comprising the step of administering to said patient a pharmaceutical composition of claim 46 in amounts effective to treat said infection.
51. The method of claim 50, wherein said bacterial infection is caused by a bacterium selected from the group consisting of C jejuni, V. cholerae, Y pestis, B. anthracis, P. putidas, and M. tuberculosis.
US10/337,699 2002-01-07 2003-01-07 Broad spectrum inhibitors Abandoned US20040009890A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/337,699 US20040009890A1 (en) 2002-01-07 2003-01-07 Broad spectrum inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US34478802P 2002-01-07 2002-01-07
US38357502P 2002-05-29 2002-05-29
US10/337,699 US20040009890A1 (en) 2002-01-07 2003-01-07 Broad spectrum inhibitors

Publications (1)

Publication Number Publication Date
US20040009890A1 true US20040009890A1 (en) 2004-01-15

Family

ID=26994100

Family Applications (5)

Application Number Title Priority Date Filing Date
US10/337,349 Expired - Fee Related US7109230B2 (en) 2002-01-07 2003-01-07 Resistance-repellent retroviral protease inhibitors
US10/337,699 Abandoned US20040009890A1 (en) 2002-01-07 2003-01-07 Broad spectrum inhibitors
US11/490,155 Abandoned US20060293286A1 (en) 2002-01-07 2006-07-21 Resistant-repellent retroviral protease inhibitors
US11/490,111 Abandoned US20060258737A1 (en) 2002-01-07 2006-07-21 Resistance-repellent retroviral protease inhibitors
US12/504,277 Abandoned US20100009935A1 (en) 2002-01-07 2009-07-16 Resistance-repellent retroviral protease inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/337,349 Expired - Fee Related US7109230B2 (en) 2002-01-07 2003-01-07 Resistance-repellent retroviral protease inhibitors

Family Applications After (3)

Application Number Title Priority Date Filing Date
US11/490,155 Abandoned US20060293286A1 (en) 2002-01-07 2006-07-21 Resistant-repellent retroviral protease inhibitors
US11/490,111 Abandoned US20060258737A1 (en) 2002-01-07 2006-07-21 Resistance-repellent retroviral protease inhibitors
US12/504,277 Abandoned US20100009935A1 (en) 2002-01-07 2009-07-16 Resistance-repellent retroviral protease inhibitors

Country Status (5)

Country Link
US (5) US7109230B2 (en)
EP (2) EP1483254A4 (en)
AU (2) AU2003202914A1 (en)
CA (2) CA2473231A1 (en)
WO (2) WO2003064406A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060035310A1 (en) * 2004-08-13 2006-02-16 Millipore Corporation Media for recovery of microorganism in the presence of antibiotics
US20060286574A1 (en) * 2005-04-05 2006-12-21 The Scripps Research Institute & Achaogen, Inc. Compositions and methods for enhancing drug sensitivity and treating drug resistant infections and diseases
US20080015834A1 (en) * 2006-07-13 2008-01-17 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Methods and systems for molecular inhibition
US20080014572A1 (en) * 2006-07-13 2008-01-17 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Methods and systems for molecular inhibition
US20080015833A1 (en) * 2006-07-13 2008-01-17 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Methods and systems for molecular inhibition of protein misfolding
US20090082344A1 (en) * 2006-07-13 2009-03-26 Searete Llc Methods and systems for treating disease
US20090136518A1 (en) * 2003-11-19 2009-05-28 The Scripps Research Institute Compositions and methods to reduce mutagenesis
US20100185419A1 (en) * 2008-09-05 2010-07-22 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
US9556426B2 (en) 2009-09-16 2017-01-31 Celgene Avilomics Research, Inc. Protein kinase conjugates and inhibitors
US11542492B2 (en) 2009-12-30 2023-01-03 Celgene Car Llc Ligand-directed covalent modification of protein

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003202914A1 (en) * 2002-01-07 2003-07-24 Sequoia Pharmaceuticals Broad spectrum inhibitors
WO2005087728A1 (en) * 2004-03-11 2005-09-22 Sequoia Pharmaceuticals, Inc. Resistance-repellent retroviral protease inhibitors
EP2422780A1 (en) * 2004-05-07 2012-02-29 Sequoia Pharmaceuticals, Inc. Resistance-repellent retroviral protease inhibitors
PT1856125E (en) 2005-02-25 2009-11-06 Tibotec Pharm Ltd Protease inhibitor precursor synthesis
EP2120562A4 (en) * 2006-11-21 2010-01-13 Purdue Research Foundation Method and compositions for treating hiv infections
KR20090101458A (en) * 2006-12-15 2009-09-28 수미카 켐텍스 가부시키가이샤 Aqueous emulsion of olefin copolymer
WO2008077070A2 (en) * 2006-12-18 2008-06-26 University Of Massachusetts Crystal structures of hiv-1 protease inhibitors bound to hiv-1 protease
AU2007348171B2 (en) 2007-02-28 2012-09-06 Polyphor Ltd. Template-fixed peptidomimetics
EP2134336B1 (en) 2007-03-16 2013-12-11 Sequoia Pharmaceuticals Benzofuran-derived hiv protease inhibitors
WO2009091941A1 (en) * 2008-01-17 2009-07-23 Purdue Research Foundation Small molecule inhibitors of hiv proteases
WO2010002994A1 (en) 2008-07-01 2010-01-07 Purdue Research Foundation Nonpeptide hiv-1 protease inhibitors
US8501961B2 (en) * 2008-07-09 2013-08-06 Purdue Research Foundation HIV protease inhibitors and methods for using
US8580294B2 (en) 2010-10-19 2013-11-12 International Partnership For Microbicides Platinum-catalyzed intravaginal rings
US20140114581A1 (en) * 2011-02-28 2014-04-24 Carnegie Mellon University Using game theory in identifying compounds that bind to targets
US10137031B2 (en) 2013-11-14 2018-11-27 International Partnership For Microbicides, Inc. Combination therapy intravaginal rings
CN114127855A (en) 2018-12-10 2022-03-01 H·J·格勒顿 Method for determining personalized medicament

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5990155A (en) * 1995-04-19 1999-11-23 Vertex Pharmaceuticals Incorporated Oxygenated-heterocycle containing sulfonamide inhibitors of aspartyl protease
US6316603B1 (en) * 1998-09-08 2001-11-13 Agouron Pharmaceuticals, Inc. Modifications of the VEGF receptor-2 protein and methods of use
US6319946B1 (en) * 1999-02-12 2001-11-20 Vertex Pharmaceuticals Incorporated Inhibitors of aspartyl protease
US6333186B1 (en) * 1999-01-08 2001-12-25 Bristol-Myers Squibb Company Modified forms of Hepatitis C NS3 protease for facilitating inhibitor screening and structural studies of protease: inhibitor complexes
US6403312B1 (en) * 1998-10-16 2002-06-11 Xencor Protein design automatic for protein libraries
US6462173B1 (en) * 1997-09-08 2002-10-08 Max-Planck-Gesellschaft Zur Forderung Der Inhibitors of phosphoserine and phosphothreonine-proline-specific isomerases
US6485925B1 (en) * 1998-04-01 2002-11-26 The United States Of America As Represented By The Department Of Health And Human Services Anthrax lethal factor is a MAPK kinase protease
US20030171423A1 (en) * 2002-01-07 2003-09-11 Erickson John W. Resistance-repellent retroviral protease inhibitors

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5475027A (en) * 1990-11-19 1995-12-12 G.D. Searle & Co. Retroviral protease inhibitors
WO1993008184A1 (en) * 1991-10-23 1993-04-29 Merck & Co., Inc. Hiv protease inhibitors
ATE218541T1 (en) * 1992-08-25 2002-06-15 Searle & Co HYDROXYETHYLAMINOSULFONAMIDES USABLE AS INHIBITORS OF RETROVIRAL PROTEASES
US5968942A (en) * 1992-08-25 1999-10-19 G. D. Searle & Co. α- and β-amino acid hydroxyethylamino sulfonamides useful as retroviral protease inhibitors
US5783701A (en) * 1992-09-08 1998-07-21 Vertex Pharmaceuticals, Incorporated Sulfonamide inhibitors of aspartyl protease
IS2334B (en) * 1992-09-08 2008-02-15 Vertex Pharmaceuticals Inc., (A Massachusetts Corporation) Aspartyl protease inhibitor of a new class of sulfonamides
US5728718A (en) * 1994-12-20 1998-03-17 The United States Of America As Represented By The Department Of Health And Human Services 2,5-diamino-3,4-disubstituted-1,6-diphenylhexane isosteres comprising benzamide, sulfonamide and anthranilamide subunits and methods of using same
US6187943B1 (en) * 1996-10-24 2001-02-13 Mitsubishi Rayon Co., Ltd. Process for preparing carbocyclic or heterocyclic nitriles by vapor phase ammoxidation
EP1086076B1 (en) * 1998-06-19 2004-12-22 Vertex Pharmaceuticals Incorporated Sulfonamide inhibitors of aspartyl protease
WO1999067254A2 (en) * 1998-06-23 1999-12-29 The United States Of America Represented By The Secretary, Department Of Health And Human Services Multi-drug resistant retroviral protease inhibitors and use thereof
PT2336134T (en) 1998-06-23 2016-10-19 The Board Of Trustees Of The Univ Of Illionis Fitness assay and methods for reducing resistance of hiv to therapy
AP1459A (en) * 1999-10-06 2005-08-22 Tibotec Pharm Ltd Hexahydrofuro [2,3-b] furan-3- YL-N-{3-[(1,3- benzodioxol-5- ylsufonyl) (isobutyl) amino]-1-benzyl-2-hydroxypropyl} carbamate as retroviral protease inhibitor.
AU1760001A (en) * 1999-11-10 2001-06-06 Quest Diagnostics Investments Incorporated Use of computationally derived protein structures of genetic polymorphisms in pharmacogenomics and clinical applications
HUP0302015A2 (en) * 2000-05-25 2003-10-28 F. Hoffmann-La Roche Ag. Substituted 1-aminoalkyl-lactams and their use as muscarinic receptor antagonists
ES2275866T3 (en) 2001-02-14 2007-06-16 Tibotec Pharmaceuticals Ltd. 2- (AMINOSUSTITUDED) -BENZOTIAZOL-SULFONAMIDES OF SPACIOUS INHIBITING SPIRITS OF THE HIV PROTEASE.
NZ528954A (en) 2001-04-09 2005-04-29 Tibotec Pharm Ltd Broadspectrum 2-(substituted-amino)-benzoxazole sulfonamide HIV protease inhibitors
ATE429431T1 (en) 2001-05-11 2009-05-15 Tibotec Pharm Ltd 2-AMINO-BENZOXAZOLE SUPHONAMIDES AS BROAD-SPECTR HIV PROTEASE INHIBITORS
US7087373B2 (en) * 2001-06-05 2006-08-08 Tibotec Pharmaceuticals Ltd. Methods for determining plasma free drug concentration by direct measurement of binding affinity of protease inhibitors to plasma proteins
US7285566B2 (en) * 2002-01-07 2007-10-23 Erickson John W Resistance-repellent retroviral protease inhibitors
US7157489B2 (en) * 2002-03-12 2007-01-02 The Board Of Trustees Of The University Of Illinois HIV protease inhibitors
MXPA05001792A (en) * 2002-08-14 2005-04-25 Tibotec Pharm Ltd Broadspectrum substituted oxindole sulfonamide hiv protease inhibitors.
WO2005087728A1 (en) * 2004-03-11 2005-09-22 Sequoia Pharmaceuticals, Inc. Resistance-repellent retroviral protease inhibitors
EP2422780A1 (en) * 2004-05-07 2012-02-29 Sequoia Pharmaceuticals, Inc. Resistance-repellent retroviral protease inhibitors

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5990155A (en) * 1995-04-19 1999-11-23 Vertex Pharmaceuticals Incorporated Oxygenated-heterocycle containing sulfonamide inhibitors of aspartyl protease
US6462173B1 (en) * 1997-09-08 2002-10-08 Max-Planck-Gesellschaft Zur Forderung Der Inhibitors of phosphoserine and phosphothreonine-proline-specific isomerases
US6485925B1 (en) * 1998-04-01 2002-11-26 The United States Of America As Represented By The Department Of Health And Human Services Anthrax lethal factor is a MAPK kinase protease
US6316603B1 (en) * 1998-09-08 2001-11-13 Agouron Pharmaceuticals, Inc. Modifications of the VEGF receptor-2 protein and methods of use
US6403312B1 (en) * 1998-10-16 2002-06-11 Xencor Protein design automatic for protein libraries
US6333186B1 (en) * 1999-01-08 2001-12-25 Bristol-Myers Squibb Company Modified forms of Hepatitis C NS3 protease for facilitating inhibitor screening and structural studies of protease: inhibitor complexes
US6319946B1 (en) * 1999-02-12 2001-11-20 Vertex Pharmaceuticals Incorporated Inhibitors of aspartyl protease
US20030171423A1 (en) * 2002-01-07 2003-09-11 Erickson John W. Resistance-repellent retroviral protease inhibitors

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090136518A1 (en) * 2003-11-19 2009-05-28 The Scripps Research Institute Compositions and methods to reduce mutagenesis
US20060035310A1 (en) * 2004-08-13 2006-02-16 Millipore Corporation Media for recovery of microorganism in the presence of antibiotics
US20100075368A1 (en) * 2004-08-13 2010-03-25 Millipore Corporation Media for recovery of microorganism in the presence of antibiotics
US20060286574A1 (en) * 2005-04-05 2006-12-21 The Scripps Research Institute & Achaogen, Inc. Compositions and methods for enhancing drug sensitivity and treating drug resistant infections and diseases
US20080015833A1 (en) * 2006-07-13 2008-01-17 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Methods and systems for molecular inhibition of protein misfolding
US20090055138A1 (en) * 2006-07-13 2009-02-26 Searete Llc Methods and systems for molecular inhibition
US20090081641A1 (en) * 2006-07-13 2009-03-26 Searete Llc Methods and systems for treating disease
US20090082344A1 (en) * 2006-07-13 2009-03-26 Searete Llc Methods and systems for treating disease
US20090082974A1 (en) * 2006-07-13 2009-03-26 Searete Llc Methods and systems for treating disease
US20090083018A1 (en) * 2006-07-13 2009-03-26 Searete Llc, Methods and systems for molecular inhibition of protein misfolding
US20080014572A1 (en) * 2006-07-13 2008-01-17 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Methods and systems for molecular inhibition
US20080015834A1 (en) * 2006-07-13 2008-01-17 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Methods and systems for molecular inhibition
US20090024364A1 (en) * 2006-08-18 2009-01-22 Searete Llc, Methods and systems for molecular inhibition of protein misfolding
US20100185419A1 (en) * 2008-09-05 2010-07-22 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
US9556426B2 (en) 2009-09-16 2017-01-31 Celgene Avilomics Research, Inc. Protein kinase conjugates and inhibitors
US10662195B2 (en) 2009-09-16 2020-05-26 Celgene Car Llc Protein kinase conjugates and inhibitors
US11542492B2 (en) 2009-12-30 2023-01-03 Celgene Car Llc Ligand-directed covalent modification of protein

Also Published As

Publication number Publication date
EP1472536A4 (en) 2007-02-14
US7109230B2 (en) 2006-09-19
EP1472536A2 (en) 2004-11-03
AU2003202914A1 (en) 2003-07-24
US20060293286A1 (en) 2006-12-28
CA2472580A1 (en) 2003-07-17
US20030171423A1 (en) 2003-09-11
CA2473231A1 (en) 2003-08-07
EP1483254A1 (en) 2004-12-08
EP1483254A4 (en) 2005-06-08
AU2008258152A1 (en) 2009-01-08
WO2003064406A1 (en) 2003-08-07
WO2003057173A3 (en) 2003-11-20
US20060258737A1 (en) 2006-11-16
WO2003057173A2 (en) 2003-07-17
US20100009935A1 (en) 2010-01-14

Similar Documents

Publication Publication Date Title
US20040009890A1 (en) Broad spectrum inhibitors
Narayanan et al. Identification of SARS-CoV-2 inhibitors targeting Mpro and PLpro using in-cell-protease assay
Agnello et al. A structural view on medicinal chemistry strategies against drug resistance
US8088891B2 (en) Bacterial ATP synthase binding domain
Zhang et al. Recent development of leucyl-tRNA synthetase inhibitors as antimicrobial agents
US20240033265A2 (en) Treatments for mutations in acute myeloid leukemia
Uengwetwanit et al. Identification of novel SARS-CoV-2 RNA dependent RNA polymerase (RdRp) inhibitors: From in silico screening to experimentally validated inhibitory activity
Toulouse et al. Dual-target inhibitors of the folate pathway inhibit intrinsically trimethoprim-resistant DfrB dihydrofolate reductases
SAMANT et al. Comparative docking analysis of rational drugs against COVID-19 main protease
Xie et al. Targeting aminoacyl tRNA synthetases for antimalarial drug development
Shakour et al. 3D-QSAR studies of 1, 2, 4-oxadiazole derivatives as sortase A inhibitors
Mukherjee et al. Proteomics advances towards developing SARS-CoV-2 therapeutics using in silico drug repurposing approaches
WO2011139636A1 (en) Small molecule inhibitors of functions of the hiv-1 matrix protein
Guerra et al. New targets for antimalarial drug discovery
CN107296807A (en) Application of the MALT1 targeted inhibitions thing in MALT1 dependent tumors medicines are prepared
Luo et al. Specifically targeting Mtb cell-wall and TMM Transporter: The development of MmpL3 inhibitors
Cheng et al. Structure-guided Design of Halofuginone Derivatives as ATP-aided inhibitors against bacterial Prolyl-tRNA Synthetase
Nurcahyaningtyas et al. Identification of potential Indonesian marine invertebrate bioactive compounds as TMPRSS2 and SARS-CoV-2 Omicron spike protein inhibitors through computational screening
Ndagi et al. A consequence of drug targeting of aminoacyl‐tRNA synthetases in Mycobacterium tuberculosis
Keon et al. Cryo-EM of the yeast VO complex reveals distinct binding sites for macrolide V-ATPase inhibitors
GUPTA et al. Anticancer drugs as potential inhibitors of AcrAB-TolC of multidrug resistant Escherichia coli: an in silico molecular modeling and docking study
Hussain et al. Computational Drug Repurposing Resources and Approaches for Discovering Novel Antifungal Drugs against Candida albicans N-Myristoyl Transferase.
Wadhwa Molecular docking study of novel covid-19 protease with current clinical management agents
US20120142682A1 (en) Antivirulence compounds inhibiting bacterial mono-adp-ribosyltransferase toxins
Mammen et al. Computational and experimental validation of methotrexate as Staphylococcal-DHFR inhibitor

Legal Events

Date Code Title Description
AS Assignment

Owner name: SEQUOIA PHARMACEUTICALS, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ERICKSON, JOHN;EISSENSTAT, MICHAEL;SILVA, ABEIARDO;AND OTHERS;REEL/FRAME:015194/0793

Effective date: 20040923

STCB Information on status: application discontinuation

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