US20160283652A1 - Methods for identifying inhibitors of amyloid protein aggregation - Google Patents

Methods for identifying inhibitors of amyloid protein aggregation Download PDF

Info

Publication number
US20160283652A1
US20160283652A1 US15/155,643 US201615155643A US2016283652A1 US 20160283652 A1 US20160283652 A1 US 20160283652A1 US 201615155643 A US201615155643 A US 201615155643A US 2016283652 A1 US2016283652 A1 US 2016283652A1
Authority
US
United States
Prior art keywords
amyloid
atom
model
amyloid protein
aggregation
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
US15/155,643
Inventor
Christopher J. Barden
Michael D. Carter
Donald F. Weaver
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.)
Treventis Corp
Original Assignee
Treventis Corp
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 Treventis Corp filed Critical Treventis Corp
Priority to US15/155,643 priority Critical patent/US20160283652A1/en
Publication of US20160283652A1 publication Critical patent/US20160283652A1/en
Priority to US16/172,289 priority patent/US11568956B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • G06F19/16
    • C40B30/02
    • G06F19/12
    • 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
    • G16B15/30Drug targeting using structural data; Docking or binding prediction
    • 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
    • G16B35/00ICT specially adapted for in silico combinatorial libraries of nucleic acids, proteins or peptides
    • 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
    • G16C20/60In silico combinatorial chemistry
    • 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
    • G16C20/60In silico combinatorial chemistry
    • G16C20/64Screening of libraries

Definitions

  • This invention relates to methods of identifying compounds that are inhibitors or are likely to be inhibitors of amyloid protein aggregation, and three-dimensional, non-crystallographic, models of amyloid receptors and models of amyloid protein aggregation utilized in the methods.
  • amyloidosis a condition known as amyloidosis
  • amyloidosis a condition known as amyloidosis
  • Certain models of beta-amyloid aggregation that exist in vitro or in vivo are used for high-throughput screening to discover new compounds that modulate amyloid aggregation (Dolphin et al, 2007, ChemMedChem 2:1613-1623; Gazit, 2006, ACS Chem. Biol. 1:417-419).
  • These existing in vitro or in vivo models however require the synthesis and/or procurement of every candidate compound, as well as the synthesis or incubation of the model systems.
  • amyloid diseases utilizes peptides as modulators of disease targets, being amyloid proteins in this case.
  • peptides as modulators of disease targets, being amyloid proteins in this case.
  • amyloid diseases remain invariably fatal using current medical practice.
  • Crystallized amyloid protein means an amyloid protein for which a binding pocket is known.
  • Uncrystallized amyloid protein means an amyloid protein for which a binding pocket is unknown.
  • Multiply anti-amyloid compound means a compound that has activity against both a crystallized amyloid protein and an uncrystallized amyloid protein.
  • Modulating amyloid aggregation means changing the distribution of oligomers such that amyloid deposition is reduced in a subject and encompasses a process of inhibition of aggregation.
  • Model means a molecular model, which is a representation of atoms and their positions with respect to each other, substantially including such features as covalent bonds between highly interacting atoms in a molecule and/or non-covalent interactions between the atoms of two or more molecules.
  • Super-model means a model of models.
  • Computer modeling program means any program that can visualize or otherwise manipulate molecular models on a computer, preferably also featuring molecular mechanics or molecular dynamics routines.
  • Interacts means chemical interactions, especially those that result in covalent bonds being formed between atoms in a molecule and/or non-covalent bonds between the atoms of two or more molecules. If said molecules are composed of amino acid residues, it is commonly said that two residues interact when at least one atom from one residue interacts with at least one atom from the other residue.
  • Non-crystallographic means that the model was not directly obtained by the techniques of crystallography, especially X-ray crystallography.
  • Candidate model means a model of an uncrystallized amyloid protein that is to be evaluated for utility as a pseudo-crystal structure of a crystallized or uncrystallized amyloid protein and/or amyloid aggregation.
  • Halogen means fluorine, chlorine, bromine, or iodine.
  • “Extracting” means the step of operating the computer modeling program such that the relevant subset is highlighted, saved, and/or placed in a different workspace within the program for the purposes of manipulating the subset without reference to the rest of a model.
  • Such a subset may be considered a model in and of itself.
  • Validating means the step of performing docking and/or quantitative structure-activity relationship (QSAR) calculations in a computer modeling program, in order to verify that a given model can generally correctly classify and/or predict the anti-amyloid activity of a set of known positive and negative controls for anti-amyloid activity.
  • QSAR quantitative structure-activity relationship
  • Consing means operating a computer modeling program to instantiate an in silico representation of a given model.
  • “Selecting a list of candidate compounds” means choosing a set of compounds for evaluation in a model. Selection of said set may be reference to commercially available screening libraries such as Hit2Lead or Enamine, by reference to an internal list of available and/or proprietary compounds for in vitro or in silico screening, or by utilizing lead-hopping or de novo design methods.
  • “Iterative docking” means docking each candidate compound in turn into the model using a computer modeling program.
  • “Scoring” means applying formulas to obtain a score from “scoring functions” pertaining to drug-target interactions, the output of which furnishes a measurement of the degree of complementarity of the compound with respect to the pocket.
  • Score cutoff means some value, usually an absolute energy, that can be set higher or lower depending on the level of activity that is deemed to separate active from inactive, or as a reference between more active and less active.
  • “Improving potency” means providing a means to furnish a compound analogous to an active compound that has minor structural differences which produce a better modulating effect with respect to the target.
  • the invention provides methods of utilizing three-dimensional, non-crystallographic, models of amyloid aggregation to identify compounds that are inhibitors or are likely to be inhibitors of amyloid protein aggregation. It also provides three-dimensional, non-crystallographic models, of amyloid receptors and models of amyloid protein aggregation utilized in these methods, as well as methods of producing such models for these methods.
  • FIG. 1 is a depiction of the super-model developed in Example 1, in which the crystallized amyloid protein is a fragment of transthyretin, the multiply anti-amyloid compound is resveratrol, and the uncrystallized amyloid protein is a dimer of A ⁇ 17-42 .
  • FIG. 2 is a stereographic view of the pseudo-crystal structure, a model of amyloid protein aggregation, of Example 1 with resveratrol bound in its pocket.
  • Amyloidosis refers to a variety of conditions in which amyloid proteins are abnormally deposited in organs and/or tissues.
  • amyloidosis occurs principally in the central nervous system, e.g., with aggregation of beta-amyloid protein in Alzheimer's Disease, alpha-synuclein in Parkinson's Disease, huntingtin protein in Huntington's Disease, and prion protein in Creutzfeldt-Jacob and other prion diseases.
  • Other types of amyloidosis are systemic in nature, as, e.g., with aggregation of transthyretin in senile systemic amyloidosis.
  • aggregations of protein were classified as “amyloid” if they displayed apple-green birefringence under polarized light when stained with the dyes Congo red or Thioflavin T (ThT) (Sipe and Cohen, 2000, J. Struct. Biol. 130:88-98). That definition of “amyloid protein” has been expanded in recent years to apply to any fibrous structure which is ordered and insoluble, and which is composed of fibrils of approximately 10 nm in width and up to several microns in length (Zheng, Ma, and Nussinov, 2006, Phys. Biol. 3:P1-P4).
  • amyloidogenic polypeptide is one which, regardless of sequence, can polymerize in a cross- ⁇ sheet conformation in vitro or in vivo to form amyloid protein (Xu, 2007, Amyloid 14:119-31).
  • Amyloidogenic polypeptides include, e.g., amyloid light chain, amyloid associated protein, ⁇ amyloid, transthyretin, ⁇ 2 microglobulin, amylin, prion related protein, and other amyloid proteins.
  • amyloid proteins have commonalities. However, whether certain amyloid proteins are related depends on the amyloid proteins having commonality of both pathology and location. For example, beta-amyloid protein and huntingtin protein are both located in a human brain, but their divergence in pathology is sufficient to consider them unrelated. Beta-amyloid protein, tau protein, and alpha-synuclein protein on the other hand are related, all being located in the human brain and being thought to have overlapping roles in the pathology of neurodegenerative diseases, e.g., Alzheimer's disease and Parkinson's disease.
  • neurodegenerative diseases e.g., Alzheimer's disease and Parkinson's disease.
  • models of amyloid protein aggregation provided by the present invention will have significant utility even with unrelated amyloid proteins, in addition to having utility with the related amyloid proteins (i.e., beta-amyloid protein, tau protein, and alpha-synuclein protein).
  • a lock and key analogy is commonly used to characterize drug-target interactions where a specific “key” (compound/drug) interacts only with its respective molecular “lock” (target/receptor).
  • An appropriate degree of shape and electronic complimentarily between the drug and target must occur to produce drug-target interactions which are necessary for a pharmacological response.
  • the specific location on the “lock” or target is often referred to as the active or catalytic site, and the intuitive shape of the active site is that of a pocket.
  • a “binding pocket,” or more simply “pocket,” is used in the art to refer to that space on the target in which a molecule can be inserted to modulate activity of the target.
  • the three-dimensional shape and electronic properties of the pocket form the basis for rational drug design and for virtual screening techniques (e.g., in silico) utilized in the methods of the present invention, provides means for identifying the pocket for non-peptidic compound insertion and suggests means for modulating amyloid protein aggregation.
  • “Rational drug design” attempts to formulate drug design hypotheses that specify and optimize the physical contacts between a drug and its target, often by aid of a computer.
  • Such “computer-aided drug design” generally depends on high resolution, three-dimensional models of the target, and such models are usually constructed by X-ray crystallography of a crystal form of the target. Crystallographic techniques are the primary means of obtaining three-dimensional structure and binding information in the art. In the absence of a solved, high-resolution (i.e.
  • the virtual screening techniques utilized in the methods of the present invention and the virtual models of the present invention allow, e.g., for the virtual identification of the binding pockets of amyloid proteins, and construction of three-dimensional non-crystallographic models of amyloid receptors and models of amyloid aggregation.
  • the methods of the present invention generally do not require construction of crystal forms of the target by X-ray crystallography, and instead provide or utilize non-crystallographic models (“pseudo-crystal structures”) that could approximate or exceed the utility of traditional crystal structures.
  • the invention provides for candidate models of amyloid receptors and of amyloid protein aggregation to be constructed, evaluated and subsequently validated.
  • the models of the present invention are based, in part, both on the symmetry inherent in many anti-amyloid compounds and on the multi-target activity of such compounds.
  • the models of the present invention may be referred to as “pseudo-crystal structures” or as surrogate models, as the models are not obtained by the technique of crystallography. As stated above and as demonstrated by the examples below, the utility of “pseudo-crystal structures” of the present invention may approach and exceed that of crystal structures.
  • Successful candidate models will include elements directly related to the process of aggregation and will be of sufficient detail to enable the design of compounds for disrupting or otherwise altering the interactions essential to amyloid protein aggregation. Indeed, if a pseudo-crystal structure does not indicate the binding mode of an anti-amyloid compound and/or the chemical interactions essential to aggregation, then it is not a proper model of amyloid protein aggregation and would not be expected to have utility to enable the design of anti-amyloid compounds.
  • Successful candidate models in accordance with the present invention may comprise pseudo-crystal structures of uncrystallized amyloid proteins.
  • Such pseudo-crystal structures generally comprise two parts.
  • the first part generally comprises an amyloid monomer
  • the second part generally comprises an amyloid monomer or an amyloid oligomer.
  • the first part is generally positioned with respect to the second part such that it forms a pocket in conjunction with the second part, such that a candidate compound may be inserted into the pocket, thereby potentially modulating amyloid aggregation.
  • the amyloid monomer comprises one peptide chain from an amyloid oligomer or amyloid protein.
  • the amyloid monomer in the first part and the amyloid monomer (or each monomer in an amyloid oligomer) in the second part may comprise the same sequence or different sequences with respect to the first part.
  • the amyloid oligomer comprises a heterogeneous or homogeneous link comprising an amyloid protein (or amyloidogenic fragments thereof).
  • the amyloid oligomer comprises an amyloid protein (e.g., either beta-amyloid protein, tau protein, alpha-synuclein protein, huntingtin protein, or prion protein) or amyloidogenic fragments thereof; and in other embodiments the amyloid oligomer comprises two or more amyloid proteins (e.g., a combination of beta-amyloid protein and tau protein, a combination of beta-amyloid protein and alpha-synuclein protein, etc.) or amyloidogenic fragments thereof.
  • the amyloid oligomer may also comprise a protein which has not yet shown to be amyloidogenic or a portion of such protein.
  • the amyloid oligomer or amyloidogenic fragment thereof comprises a region comprising approximately residues 17 through 40 of beta-amyloid protein of length 40; approximately residues 17 through 42 of beta-amyloid protein of length 42; approximately residues 1 through 15 of alpha-synuclein protein; approximately residues 25 through 40 or 275 through 280 of tau protein; approximately residues 90 through 110 of the major prion precursor protein; or approximately residues 5 through 20 of the huntingtin protein.
  • the model comprises the pseudo-crystal structure formed by SEQ ID: 1 and SEQ ID: 2 and interacting in the manner depicted stereographically in FIG. 2 , which is a model of amyloid protein aggregation of Example 1, with resveratrol bound in its pocket.
  • FIG. 2 is a model of amyloid protein aggregation of Example 1, with resveratrol bound in its pocket.
  • Three-dimensional features of the model may be appreciated by a person of normal binocular vision by superimposing the images visually.
  • the first part comprises substantially SEQ ID: 1 and the second part comprise substantially SEQ ID: 2, wherein SEQ ID: 1 is
  • the first part is further positioned with respect to the second part such that the amino acid Val at position 8 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2; the amino acid Gly at position 9 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2; the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Gly at position 9 of SEQ ID: 2; the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Ser at position 10 of SEQ ID:2; and the amino acid Lys at position 12 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2.
  • FIG. 2 is a stereographic view of the pseudo-crystal structure, a model of amyloid protein aggregation, of Example 1 with resveratrol bound in its pocket.
  • the model of FIG. 2 features two A ⁇ 17-42 monomers bound to each other near the N terminals in a “double candycane” structure. Each monomer is folded in a loop from residues 23 through 33, which is a conformation well supported by electron microscopy and NMR spectroscopy. The loops are stabilized by intra-loop cationic-anionic interactions between Asp 23 and Lys 28 , and they also show an inter-loop attraction through these same residues. Short-run (up to 100 ns) molecular dynamics calculations on this model indicate that the pocket is stable.
  • a virtual non-crystallographic model of amyloid protein aggregation comprises a pseudo-crystal structure comprising a three-dimensional model of an uncrystallized monomeric amyloid protein and a three-dimensional model of an uncrystallized amyloid protein comprising one or more amyloid peptides; said three-dimensional model of an uncrystallized amyloid protein is positioned with respect to said amyloid monomer model such that it forms a pocket in conjunction with said amyloid monomer model, such that a candidate compound may be inserted into said pocket thereby, e.g., potentially modulating amyloid aggregation.
  • Aggregation is a fundamentally different process from fibrillization. Aggregation occurs among monomers and oligomers of incipient amyloid protein, while fibrillization is directed to the further lengthening of pre-formed amyloid protofibrils and subsequent assembly into fibrils. Modulators of aggregation are preferred to modulators of fibrillization for treatment of amyloid diseases, since soluble oligomers of beta-amyloid protein are thought to be the pathogenic species in Alzheimer's Disease, rather than the mature fibrils thereof (Klyubin et al, 2008, J. Neurosci. 28:4231-4237; Townsend et al, 2006, J. Physiol. 572:477-492).
  • a model of amyloid protein aggregation promulgated in the present application is substantially a model of addition of monomeric amyloid peptide to an amyloid oligomer or an amyloid protein.
  • the monomeric amyloid peptide and the amyloid oligomer (or the amyloid protein) is composed of chains of different amyloid peptides.
  • the monomeric amyloid peptide and the amyloid oligomer or protein are composed of chains of the same amyloid peptide.
  • modulating amyloid aggregation may result in greatly increasing the proportion of amyloid peptide assemblies with size n, where n is between 1 and 10. In other embodiments, modulating amyloid aggregation may result in increasing the proportion of amyloid peptide assemblies with size n, where n is between 1 and 5. In still other embodiments of the invention, modulating amyloid aggregation may result in increasing the proportion of amyloid peptide monomer (i.e. where n is only 1).
  • a virtual non-crystallographic model of amyloid protein aggregation comprises a pseudo-crystal structure comprising an amyloid monomer model and a three-dimensional model of an uncrystallized amyloid protein.
  • the “amyloid protein” in the minimal case may comprise only one amyloidogenic peptide having substantially the same sequence as an amyloid protein or an amyloidogenic fragment thereof.
  • the “amyloid protein” may also comprise a plurality of chains of one amyloid protein or amyloidogenic portions thereof, and does not include other amyloid protein or amyloidogenic portions thereof. In certain embodiments, the plurality of chains are aggregated according, e.g., to a building-up principle.
  • the amyloid monomer model and the three-dimensional model of an uncrystallized amyloid protein are both composed of beta-amyloid protein or an amyloidogenic fragment thereof.
  • the amyloids may comprise substantially the sequence of beta-amyloid 1-42 and/or the sequence of beta-amyloid 1-40 and/or the sequence of the beta-amyloid fragment 25-35 and/or the sequences of beta-amyloid fragment 17-42, which are sequences known to form aggregated beta-amyloid (Hughes et al, 2000, J Biol Chem 275:25109-25115).
  • candidate models of amyloid protein aggregation comprise a pseudo-crystal structure comprising a three-dimensional model of an uncrystallized, monomeric amyloid protein and a three-dimensional model of an uncrystalized amyloid protein comprising one or more amyloid peptides positioned with respect to the monomeric amyloid protein model such that it forms a pocket in conjunction with the monomeric amyloid protein.
  • the monomeric amyloid model is substantially SEQ ID: 1; and the amyloid protein model comprises SEQ ID: 2.
  • three-dimensional, non-crystallographic models, of amyloid aggregation comprise pseudo-crystal structures for amyloid proteins that lack extant crystal structures (e.g., beta-amyloid protein, tau, alpha-synuclein, huntingtin, and prion protein, etc.).
  • pseudo-crystal structures for amyloid proteins that lack extant crystal structures (e.g., beta-amyloid protein, tau, alpha-synuclein, huntingtin, and prion protein, etc.).
  • a model may be recorded in a mathematical form using a combination of atomic information and three-dimensional coordinates (e.g. in the PDB format, or in the proprietary format of a computer modeling program), which are then visualized or otherwise represented virtually, e.g., in a computer modeling program.
  • the model reflects the likely protonation state at physiological pH such that basic moieties of the model are protonated and acidic moieties are deprotonated in a manner consistent with valence considerations.
  • force fields and charges are applied appropriate to the model such that molecular mechanics and/or dynamics can be used; details on these and other computational techniques characteristically found in computer modeling programs are described, e.g., in A. Leach, Molecular Modeling: Principles and Applications, 2 nd edition, Prentice Hall, 2001, herein incorporated by reference.
  • the models of the present invention may, e.g., enable the identification of new chemical classes of modulators of their respective amyloid protein and related amyloid proteins. For example, if a candidate compound has a three-dimensional shape and electronic properties that will allow it to “fit” into the pseudo-crystal structure and interact with the pseudo-crystal structure, it is likely to be a potential modulator of the amyloid protein aggregation.
  • the models of the present invention may also provide a means of improvement in potency for compounds known to have modulating effects on a relevant amyloid protein, as they may suggest structural modification to the compounds known to have modulating effects on the amyloid protein, to allow the compounds to better “fit” and interact with the three-dimensional pseudo-crystal structure. They may also provide means to the de novo design of compounds which may “fit” into the three-dimensional structure.
  • the models may further provide means for the identification and discovery of the compounds which may be useful in modulation of either or both aggregation and fibrilization.
  • the present invention further provide means for constructing surrogate models (i.e., non-crystallographic virtual models) of amyloid protein aggregation which may be used for drug design much as crystal structures are customarily used but at a lower cost and without a need for synthesis of every candidate compound.
  • surrogate models i.e., non-crystallographic virtual models
  • the three-dimensional, non-crystallographic models of amyloid aggregation models in accordance with the present invention may be constructed, e.g., virtually (e.g., in a computer modeling system), depicted pictorially, instantiated in a physical form by way of a modeling kit, or visualized mentally.
  • the virtual modeling afforded by the present invention allows for greatly reduced cost and labor as compared to in vitro or in vivo systems, as it does not require the synthesis and/or procurement of every candidate compound.
  • it allows for the screening and prediction of the anti-amyloid properties of a candidate compound based on its three-dimensional shape and electronic configuration, even before the compounds are synthesized. It disposes of a potentially great number of candidate compounds which will not “fit” into the three-dimensional, non-crystallographic, models of the amyloid aggregation afforded by the present invention, and are therefore are unlikely to have anti-amyloid activity.
  • construction of the non-crystallographic models is performed manually, using a molecule or peptide/biomolecule building routine as included in a computer modeling program. In some embodiments of the invention, construction may proceed automatically through the reading of a previously constructed file or sequence.
  • the invention provides means for constructing virtual non-crystallographic models of amyloid aggregations for proteins for which the three-dimensional structure and binding information (e.g., binding mode and chemical interactions essential for aggregation) are known. In other embodiments, the invention provides means for constructing virtual non-crystallographic models of amyloid aggregations for proteins for which the three-dimensional structure and binding information (e.g., binding mode and chemical interactions essential for aggregation) are unknown.
  • the three-dimensional structure and binding information e.g., binding mode and chemical interactions essential for aggregation
  • the means for constructing virtual non-crystallographic models of amyloid aggregations for proteins for which the three-dimensional structure and binding information are known and the means for constructing virtual non-crystallographic models of amyloid aggregations for proteins for which the three-dimensional structure and binding information are unknown are the same.
  • a pseudo-crystal structure of an uncrystallized amyloid protein can be virtually identified and constructed by manipulating a computer program to orient an uncrystallized amyloid protein and/or oligomer such that a pocket is formed around a multiply anti-amyloid compound already bound in the crystallized amyloid protein.
  • the pocket has a particular three-dimensional shape and electronic properties and represents, e.g., a pseudo-crystal structure of the uncrystallized amyloid protein.
  • the multiply anti-amyloid compound is removed and is not part of the pseudo-crystal structure. In other embodiments, the multiply anti-amyloid compound is part of the pseudo-crystal structure.
  • a binding pocket corresponds to a common conformational region on a variety of amyloid proteins, e.g., the Common Conformational Motif (CCM).
  • CCM Common Conformational Motif
  • the CCM may correspond to a conformational region found in both A ⁇ and tau; or a conformational region found in both A ⁇ and alpha-synuclein; or a conformational region found in both A ⁇ and transthyretin.
  • the conformational region may also be common to A ⁇ , tau and alpha-synuclein (which are all located in the human brain and being thought to have overlapping roles in the pathology of neurodegenerative diseases).
  • the binding pocket may therefore be useful for the identification of candidate compounds which are likely to inhibit the aggregation of several amyloid proteins simultaneously (e.g., A ⁇ and tau; A ⁇ and alpha synuclein; A ⁇ , tau and alpha synuclein, etc.). It should be understood for this reason that the CCM is not strictly sequence-dependent.
  • a method of creating pseudo-crystal structures of uncrystallized amyloid protein receptor comprises (1) applying molecular mechanics and/or dynamics to a three-dimensional super-model comprising a crystallized amyloid protein model, a model of multiply anti-amyloid compound bound in the pocket of the crystallized amyloid protein or oligomer, and a candidate model of an uncrystallized amyloid protein, such that a pocket on the uncrystallized amyloid protein or oligomer is formed around the multiply anti-amyloid compound; (2) extracting said uncrystallized amyloid protein model from said super-model; and (3) validating said uncrystallized amyloid protein model with respect to experimental data so as to confirm its utility as a pseudo-crystal structure.
  • the extracted model comprises the multiply anti-amyloid compound.
  • the extracted model is free of the multiply anti-amyloid compound.
  • Example 1 describes an exemplary method for construction of one pseudo-crystal structure of beta-amyloid protein in accordance with one embodiment of the invention.
  • the uncrystallized amyloid protein is an amyloidogenic fragment of beta-amyloid protein
  • the crystallized amyloid protein is transthyretin protein
  • the multiply anti-amyloid compound is resveratrol.
  • the pseudo-crystal structure of Example 1 has two parts: one monomer of an amyloidogenic beta-amyloid fragment (i.e.
  • amyloid monomer the “amyloid monomer”); and another monomer of an amyloidogenic beta-amyloid fragment, which could have multiple additional peptides attached thereon without disrupting its interaction with the amyloid monomer (as such, this second part is sometimes referred to as the “amyloid oligomer”, as it is only contingently monomeric).
  • the invention provides a method of constructing pseudo-crystal structures of uncrystallized amyloid protein which are models of amyloid protein aggregation.
  • This method's first step is to apply molecular mechanics and/or dynamics to a three-dimensional super-model comprising a crystallized amyloid protein model, a model of multiply anti-amyloid compound bound in the pocket of the crystallized amyloid protein, and a candidate model of an uncrystallized amyloid protein or oligomer, such that a pocket on the uncrystallized amyloid protein is formed around the multiply anti-amyloid compound.
  • the method's second step is to extract said uncrystallized amyloid protein or oligomer model from the super-model.
  • the extracted model comprises the multiply anti-amyloid compound.
  • the extracted model is free of the multiply anti-amyloid compound.
  • the method's third step is to validate said uncrystallized amyloid protein model with respect to experimental data so as to confirm its utility as a pseudo-crystal structure.
  • the crystallized amyloid protein may be transthyretin, islet amyloid polypeptide, beta-2-microglobulin, and insulin protein; and the uncrystallized amyloid protein may be either of beta-amyloid, tau, alpha-synuclein, huntingtin, or prion protein; an amyloidogenic fragment of any of the foregoing, or composed of a heterogeneous combination of such proteins.
  • a multiply anti-amyloid compound may be resveratrol, or a compound having low energy conformations that overlap geometrically and electrostatically with resveratrol.
  • a multiply anti-amyloid compound is a compound selected from the lists of compounds provided in paragraphs [00106] to [0117] of the present application.
  • validation comprises performing docking and/or quantitative structure-activity relationship (QSAR) calculations in a computer modeling program, in order to verify that a given model can generally correctly classify and/or predict the anti-amyloid activity of a set of known positive and negative controls for anti-amyloid activity.
  • QSAR quantitative structure-activity relationship
  • Examples of computer modeling programs include but are not limited to MOE, Sybyl, Cerius 2 , CHARMm, DOCK, AUTODOCK, GLIDE, and FlexX. Manuals of these computer modeling programs are herein incorporated by reference in their entirety.
  • Example 2 describes an exemplary method for validating an exemplary of amyloid protein aggregation.
  • the resulting pseudo-crystal structure can be used as a three-dimensional, non-crystallographic, model of amyloid aggregation in the methods of the present invention.
  • the methods of the invention may be used to identify or improve the potency of candidate compounds that are anti-amyloid with respect to the amyloid protein or fragment thereof depicted in a particular three-dimensional, non-crystallographic, model, or is related to the amyloid protein or fragment thereof that is depicted in the model.
  • the methods of the invention may also be used to identify, or improve the potency of, compounds that are anti-amyloid with respect to an amyloid protein or fragment thereof that is unrelated to the amyloid protein or fragment thereof that is depicted in a particular three-dimensional non-crystallographic model.
  • the methods of the present invention utilize the “virtual” non-crystallographic models to identify new compounds which may be inhibitors or likely to be inhibitors of amyloid protein aggregation, as well as improve existing modulators of amyloid proteins, all without the construction of crystal form of the target by X-ray crystallography.
  • the models are used to predict the potential interactions between a candidate compound and an amyloid protein, e.g., by allowing visualization of the candidate compound fitting into the binding pocket.
  • One empirical determinant for whether two atoms interact is their proximity to each other in a given chemical system. For example, a covalent bond between carbons is approximately 1.5 angstroms in interatomic distance, while a weak non-covalent bond between atoms could be as long as 3 angstroms (although stronger non-covalent bonds may approach 1.5 angstroms in length).
  • the hydrogens in the head group of a protonated Lys residue interacting with the oxygens in the head group of a deprotonated Glu residue is an example of a primarily ionic non-covalent bond.
  • the hydrogen in the head group of a Ser residue interacting with the carbonyl oxygen in the backbone of a different Ser residue, or a hydrogen in the backbone of a Gly residue interacting with the oxygen in the head group of a Ser residue are examples of primarily hydrogen bonds.
  • the virtual visualization provided by the methods of the present invention supply a means of predicting the likelihood of these interactions between a candidate model compound and a particular binding pocket. If the visualization shows that the modeling compound is unlikely to interact with a particular binding pocket on an amyloid protein, it is unlikely that the modeling compound will be useful as an inhibitor, e.g., of the amyloid protein aggregation. To the contrary, if the compound is likely to interact with the binding pocket of the amyloid aggregation, it is likely that the compound may be capable of modulating activity and potentially preventing or minimizing aggregation of this particular amyloid protein.
  • one of the most important intermolecular interactions are those which hold the amyloid oligomer together as well as those which attract the amyloid monomer.
  • the latter said interactions are pivotal to the conformation and eventual incorporation of the amyloid monomer into the amyloid oligomer, a process which is believed to continue repeatedly until reaching the critical size at which fibrillization occurs.
  • the candidate compound may disrupt or otherwise alter the process of aggregation. Whether the candidate compound can do so depends, e.g., on its shape and electronic complementarity, i.e., the ability of moieties on the compound to interact with important residues in the pocket.
  • the visualization of the fitting provided by the present invention will allow the prediction of whether the candidate compound is likely to interact with the important residues of the pocket.
  • Other intermolecular interactions which may be less important to describing aggregation, yet are potentially of importance in achieving the desired accuracy with respect to a given model of amyloid protein aggregation.
  • the presently claimed methods may also have utility in predicting these other intermolecular interactions.
  • regions of amyloid proteins or amyloidogenic fragments thereof that should make excellent models include but are not limited to the following: in beta-amyloid protein of length 40, approximately residues 17 through 40; in beta-amyloid protein of length 42, approximately residues 17 through 42; in alpha-synuclein, approximately residues 1 through 15; in tau protein, approximately residues 25 through 40 or approximately residues 275 through 280; in the major prion precursor protein, approximately residues 90 through 110; and in huntingtin, approximately residues 5 through 20.
  • Chemical interaction is a matter of degree; especially in non-covalent bonds, the type of bonding involved can often be an admixture of one or more types of bonding named above, possibly including other effects such as hydrophobic or van der Waals interactions.
  • Selected examples of chemical interactions which may be predicted utilizing the methods of the present invention include (but are not limited) to non-covalent bonds including, e.g., ionic, dipole-dipole, dipole-induced dipole, and hydrogen bonding.
  • Covalent bonds may also be predicted by the methods of the present invention. Covalent bonds are the most permanent of chemical interactions and are generally not altered by intermolecular interactions, which are governed primarily by non-covalent bonds.
  • the present invention provides method for identification and characterization of a previously unknown binding pocket or a conformational region of an amyloid, and subsequent utilization of the previously unknown pocket/conformational region as a screening tool for identification and optimization of anti-amyloid compounds.
  • the binding pocket is identified in silico, e.g., by using molecular modeling (especially optimization and dynamical techniques) on a “super-model” comprising a model of amyloid protein for which a binding pocket is known (a “crystallized amyloid protein”); a model of a compound (a “multiply anti-amyloid compound”) that has activity against said crystallized amyloid protein, as well as having activity against an amyloid protein for which a binding pocket is unknown (an “uncrystallized amyloid protein”); and a candidate model for said uncrystallized amyloid protein.
  • these methods further allow for the construction of a pseudo-crystal structure of beta-amyloid protein or a receptor having utility in identification and optimization of anti-amyloid compounds.
  • a crystallized amyloid protein may be selected, e.g., from the group consisting of transthyretin, islet amyloid polypeptide, beta-2-microglobulin, and insulin protein.
  • the crystallized amyloid protein is transthyretin protein, or an amyloidogenic fragments thereof.
  • An uncrystalized amyloid protein may be selected, e.g., from the group consisting of beta-amyloid, tau, alpha-synuclein, huntingtin, and prion protein.
  • the uncrystalized amyloid protein is beta-amyloid proteins, and amyloidogenic fragments thereof.
  • a multiply anti-amyloid compound may be any compound which has activity against any crystallized amyloid protein.
  • the multiply anti-amyloid compound is resveratrol or an analog thereof.
  • the crystallized amyloid protein is selected from the group of transthyretin, islet amyloid polypeptide, beta-2-microglobulin, and insulin protein; and the uncrystallized amyloid protein is selected from the group of beta-amyloid, tau, alpha-synuclein, huntingtin, and prion protein.
  • the crystallized amyloid protein is transthyretin protein or an amyloidogenic fragment thereof, and the uncrystallized amyloid protein is beta-amyloid protein or an amyloidogenic fragment thereof, and the multiply anti-amyloid compound is resveratrol.
  • the invention further encompasses and provides a method of identifying compounds that modulate amyloid aggregation.
  • the method generally comprises the steps of constructing a pseudo-crystal structure in a computer modeling program; selecting a list of candidate compounds; constructing said candidate compounds in a computer modeling program; performing an iterative docking and scoring of all candidate compounds, by means of docking each candidate compound into the pocket formed by said model and scoring each candidate compound to reflect its degree of complementarity with respect to said pocket; and identifying compounds that modulate amyloid aggregation by reference to a score cutoff that substantially distinguishes active compounds from inactive compounds.
  • a candidate compound may, e.g., be a compounds of Formulas Ia, Ib, Ic, pharmaceutically acceptable salts, stereo-isomers, polymorphs, metabolites, prod-drugs, analogs and combinations of any of the foregoing:
  • R 1 is selected from the group consisting of H, nitro, carboxylic acid, alkylcarboxylic acid, acetamide connected in either direction, N-(2-ethanol)amine, N-(2-morpholinethyl)amine, amine optionally substituted with one or more alkyl groups, amide optionally substituted with one or more alkyl groups, and alkoxy;
  • R 2 is selected from the group consisting of H, carboxylic acid, alkyl, alkanoyl, alkanesulfonyl, benzenesulfonyl, phenonyl optionally substituted with any one or more of alkoxy, halogen, or alkyl groups, benzyl optionally substituted with any one or more of alkoxy, halogen, or alkyl groups, and amide optionally substituted with any one or more of alkyl or aryl groups;
  • R 3 is selected from the group consisting of H, alkyl, furanylalkyl, thiophenealkyl, alkanoyl, phenyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups, benzyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups, and phenonyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups; and
  • R 4 is selected from the group consisting of H, alkyl, or phenyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups.
  • R 1 is selected from the group consisting of nitro, acetamide connected in either direction, N-(2-ethanol)amine, amino optionally substituted with any one or more alkyl groups, and amide optionally substituted with any one or more alkyl groups;
  • R 2 is selected from the group consisting of carboxylic acid, amide optionally substituted with any one or more of alkyl or aryl groups, and phenonyl optionally substituted with any one or more of alkoxy, alkyl, or aryl groups;
  • R 3 is selected from the group consisting of methyl, phenyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups, benzyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups, and phenonyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups;
  • R 4 is selected from the group consisting of H, alkoxy, and aryl.
  • R 1 is selected from the group consisting of nitro, acetamide connected in either direction, N-(2-ethanol)amine, amino optionally substituted with methyl or dimethyl, amide optionally substituted with methyl, ethyl, dimethyl, or diethyl, and methoxy;
  • R 2 is selected from the group consisting of phenonyl optionally substituted with any one or more of methoxy, alkyl, or halogen, amide optionally substituted with any one or more of methyl, phenyl, benzyl, or dimethyl, and carboxylic acid;
  • R 3 is selected from the group consisting of methyl, phenyl optionally substituted with any one or more of halogen, alkyl, or methoxy, benzyl optionally substituted with any one or more of halogen, alkyl, or methoxy, and phenonyl optionally substituted with any one or more of halogen, alkyl, or methoxy;
  • R 4 is selected from the group
  • R 3 and R 4 may be used in combination to produce a nitro moiety on the phenyl ring, or to create ring systems such as morpholine, quinoline, or isoquinoline.
  • a candidate compound is a compound of Formula Ia, pharmaceutically acceptable salts, stereo-isomers, polymorphs, metabolites, prod-drugs and combinations thereof:
  • R 1 is selected from the group consisting of nitro, amino optionally substituted with methyl or dimethyl, and amide optionally substituted with methyl, dimethyl, ethyl, or diethyl;
  • R 2 is phenonyl optionally substituted with halogen or methoxy
  • R 3 is selected from the group consisting of phenyl optionally substituted with halogen or methoxy and benzyl optionally substituted with halogen or methoxy;
  • R 4 is selected from the group consisting of H, methyl, and phenyl; with the following exception: when R 1 is nitro, R 4 is H, and R 3 is benzyl optionally substituted with fluoro or isopropyl.
  • a candidate compound is a compound of Formula Ic, pharmaceutically acceptable salts, stereo-isomers, polymorphs, metabolites, prod-drugs and combinations thereof:
  • R 1 is selected from the group consisting of nitro, amino optionally substituted with methyl or dimethyl, and amide optionally substituted with methyl, dimethyl, ethyl, or diethyl; the R 2 moiety is connected meta with respect to the phenyl ring;
  • R 2 is carboxylic acid
  • R 3 is selected from the group consisting of phenyl optionally substituted by any one or more of methoxy or halogen and benzyl optionally substituted by any one or more of methoxy or halogen;
  • R 4 is selected from the group consisting of H and methyl.
  • a candidate compound is a compound of Formula II:
  • X is selected from the group consisting of hydrogen, methyl, amine, methoxy, phenyl optionally substituted with up to a total of three methyl and/or methoxy and/or halogen groups, cyclopentane, morpholine, piperidine, N-methylpiperidine, N-ethylpiperidine, (N,N-diethyl)formamide, pyridine, pyrazine, pyrrole, pyrrolidine, furan, thiophene, tetrahydrofuran, pyran, tetrahydroisoquinoline, isoquinoline, quinoline, N-phenylpiperazine optionally substituted with up to a total of three methoxy and/or halogen groups, or N-benzylpiperazine;
  • A is an optional spacer group, attachable in either direction, selected from the group consisting of —NH—, —NHCH 2 —, —NHCH 2 CH 2 —, —NHCH 2 CH 2 O—, and —NHCH 2 (CH 3 );
  • D is selected from the group consisting of methyl, isopropyl, tert-butyl, dimethylamine, morpholine, alcohol, phenyl optionally substituted with up to a total of three methyl and/or ethyl and/or methoxy and/or halogen and/or acetamide and/or ethoxy and/or cyano groups, pyridine, pyrazine, pyrrole, pyrrolidine, furan, thiophene, tetrahydrofuran, and pyran; and
  • Z is an optional spacer group, selected from the group consisting of —CH 2 —, —SO 2 —, —SO 2 CH 2 —, —CH 2 C( ⁇ O)—, —CH 2 CH 2 —, —C( ⁇ O)—, and —C( ⁇ S)NHC( ⁇ O)—.
  • A is absent (thus X is directly connected to the phenyl ring at the position held by A); X is tetrahydroisoquinoline, attached to the phenyl ring by its lone nitrogen; Z is —C( ⁇ O)—; and D is methyl.
  • A is absent; X is morpholine, attached to the phenyl ring by its lone nitrogen; Z is —CH 2 —; and D is methyl.
  • a candidate compound is a compound of Formula III:
  • X is selected from the group consisting of methyl, methylamine, halogen, and phenyl optionally substituted with up to a total of three methyl and/or methoxy and/or halogen groups;
  • A is an optional spacer group, attachable in either direction, selected from the group consisting of —NH—, —N(CH 3 )H—, —O—, —OCH 3 —, —C( ⁇ O)NH—, and —NHCH 2 —;
  • Z is selected from the group consisting of phenyl optionally substituted with up to a total of three methyl and/or ethyl and/or methoxy and/or halogen and/or acetamide and/or ethoxy and/or cyano groups; excepting those compounds that include X as phenyl and A as —NHCH 2 —, the nitrogen in said A being connected to the nitro-containing phenyl ring in said formula and the carbon in said A being connected to said X in said formula.
  • a candidate compound is selected from the group consisting of:
  • a candidate compound is selected from the group consisting of:
  • the candidate compound is represented by one of one of the following structural formulas:
  • the candidate compound has activity against both A ⁇ and A-Syn, as determined by a ThT functional aggregation assay.
  • the candidate compound has activity against both A ⁇ and tau, as determined by a ThT functional aggregation assay.
  • the candidate compound only has activity against A ⁇ , as determined by a ThT functional aggregation assay.
  • the IC50/ ⁇ M for A ⁇ for the candidate compound is from about 0.5 to about 5734, as determined by ThT functional aggregation assay. In certain embodiments, the IC50/ ⁇ M for A ⁇ is from about 0.8 to about 180. In certain embodiments, the IC50/ ⁇ M for A ⁇ is from about 0.8 to about 107. In certain embodiments, the IC50/ ⁇ M for A ⁇ is 0.85, 0.88, 1.47, 1.9, 2.3, 2.29, 3.5, 7.5, 8.4, 9.64, 15.5, 18, 23, 25, 26.5, 54, 66, 84, 90, 92, and 100.
  • the IC50/ ⁇ M for A-syn for A-Syn, as determined by A-Syn functional ThT aggregation assay, for the candidate compound is from about 1 to about 70. In certain embodiment, the IC50/ ⁇ M for A-syn is from about 5 to about 50. In certain embodiment, the IC50/ ⁇ M for A-syn is 4, 5, 10, 15 or 45.
  • the IC50/ ⁇ M for A ⁇ for the candidate compound is 0.85, 9.64, 25, 1.47, 9.23, 27, 26.6, or 3.5; and the IC50/ ⁇ M for A-syn is 6.34, 5, 4, 7, 8, 10, 15, or 45.
  • pseudo-crystal structure utilized in these methods is the one formed by SEQ ID: 1 and SEQ ID: 2 when interacting in the manner depicted in FIG. 2 .
  • a method of identifying compounds that modulate amyloid aggregation comprises the steps of constructing a pseudo-crystal structure in a computer modeling program; selecting a list of candidate compounds; constructing said candidate compounds in a computer modeling program; performing an iterative docking and scoring of all candidate compounds, by means of docking each candidate compound into the pocket formed by said model and scoring each candidate compound to reflect its degree of complementarity with respect to said pocket; and identifying compounds that modulate amyloid aggregation by reference to a score cutoff that substantially distinguishes active compounds from inactive compounds.
  • a score cutoff is usually an absolute energy, that can be set higher or lower depending on the level of activity that is deemed to separate active from inactive, or as a reference between more active and less active.
  • the score cutoff is the value of a known active compound. In other embodiments, the score cutoff is the value that distinguishes a small part (i.e. 1% to 10%) of a set of compounds as being substantially more active than other members of the set.
  • the pseudo-crystal structure comprises a three-dimensional model of uncrystallized, monomeric amyloid protein and a three-dimensional model of uncrystallized amyloid protein or oligomer comprising one or more amyloid peptides, said amyloid protein or oligomer model to be positioned with respect to said amyloid monomer model such that it forms a pocket in conjunction with said amyloid monomer model, such that a compound may be inserted into said pocket thereby modulating amyloid aggregation.
  • amyloid monomer model and the amyloid protein or oligomer model are both composed of beta-amyloid protein or an amyloidogenic fragment thereof.
  • said amyloid monomer model is substantially SEQ ID: 1 and said amyloid oligomer model is substantially SEQ ID: 2.
  • said amyloid monomer model is further positioned with respect to said amyloid protein model such that the amino acid Val at position 8 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2; the amino acid Gly at position 9 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2; the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Gly at position 9 of SEQ ID: 2; the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Ser at position 10 of SEQ ID:2; and the amino acid Lys at position 12 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2.
  • the amyloid monomer model is positioned with respect to the amyloid protein model substantially in the orientation shown in FIG. 2 .
  • Scoring functions are generally applied after docking and may be integral to a given docking method. Examples of scoring functions include DrugScore, London, and Affinity dG. In some embodiments, the scoring function may be a composite of two or more scoring functions.
  • a method of improving potency of a compound known to modulate amyloid aggregation comprising the steps of constructing a pseudo-crystal structure in a computer modeling program; selecting a list of candidate compounds; constructing said candidate compounds in a computer modeling program; performing an iterative docking and scoring of all candidate compounds, by means of docking each candidate compound into the pocket formed by said model and scoring each candidate compound to reflect its degree of complementarity with respect to said pocket; and identifying compounds that modulate amyloid aggregation by reference to a score cutoff that substantially distinguishes more active compounds from less active compounds.
  • the list of candidate compounds includes both the compound known to be active and analogs of said compound, and the score cutoff is that score which belongs to the compound known to be active.
  • the pocket comprises substantially SEQ ID: 1 and amyloid oligomer model has substantially SEQ ID: 2, wherein SEQ ID: 1 is
  • the present invention uses known non-peptidic compounds amyloid and/or amyloidogenic fragments to identify new compounds which may be inhibitors or likely to be inhibitors of amyloid protein aggregation.
  • non-peptidic compounds have shown the ability to inhibit the aggregation of amyloid proteins in vitro, and many such compounds can inhibit the aggregation of beta-amyloid protein as well as other kinds of amyloid (see for example Klabunde et al, 2000, Nat. Struct. Biol. 7:312-321; Green et al, 2003, J. Am. Chem. Soc.
  • amyloid proteins or amyloidogenic fragments thereof have been crystallized, including islet amyloid polypeptide or amylin (Wilizius et al, 2008, Protein Sci. June 12 online); beta-2-microglobulin or light chain (Iwata et al, 2007, J. Biochem. 142:413-419; Schormann et al, 1995, Proc. Natl. Acad. Sci.
  • anti-amyloid compounds compounds that modulate the aggregation of amyloid protein in vitro
  • amyloid diseases e.g., Alzheimer's Disease
  • the present invention uses these known compounds to identify new compounds which may be inhibitors or likely to be inhibitors of amyloid protein aggregation.
  • One compound which may be used in the methods of the present invention is resveratrol, an antioxidant component of red wine and an inhibitor of beta-amyloid aggregation at an effective concentration of 5.6 ⁇ M (Riviere et al, 2007, Bioorg. Med. Chem. 15:1160-1167).
  • the invention is directed to methods of improving potency of a compound known to modulate amyloid aggregation comprising the steps of constructing a pseudo-crystal structure in a computer modeling program; selecting a list of candidate compounds; constructing said candidate compounds in a computer modeling program; performing an iterative docking and scoring of all candidate compounds, by means of docking each candidate compound into the pocket formed by said model and scoring each candidate compound to reflect its degree of complementarity with respect to said pocket; and identifying compounds that modulate amyloid aggregation by reference to a score cutoff that substantially distinguishes more active compounds from less active compounds.
  • the pseudo-crystal structure for this method is the one formed by SEQ ID: 1 and SEQ ID: 2 when interacting in the manner depicted in FIG. 2 .
  • the list of candidate compounds may include, e.g., both the compound known to be active anti-amyloid compounds and analogs of such compounds, and the score cutoff is that score which belongs to the compound known to be active.
  • the known compound is a compound selected from the lists recited in paragraphs [0106] to [0117] of the application.
  • the known compound is resveratrol.
  • the invention also encompasses and provides for use of the compounds identified or improved using the invention's methods as therapies for diseases in which amyloidosis occurs.
  • the disease include (but are not limited to) Alzheimer's disease, Huntington's disease, Parkinson's disease, Creutzfeldt-Jacob disease, amyotrophic lateral sclerosis, and senile systemic amyloidosis.
  • the disease is Alzheimer's disease.
  • the therapeutic compound inhibits aggregation of one or more of the following amyloidogenic proteins: beta-amyloid protein, tau protein, alpha-synuclein protein, huntingtin protein, prion precursor protein; other amyloidogenic protein not named herein, and combinations of any of the foregoing.
  • both the aggregation of alpha-synuclein protein and of tau protein are inhibited. In certain embodiments, both the aggregation of beta-amyloid protein and of tau protein are inhibited. In certain embodiments, both the aggregation of beta-amyloid protein and of alpha-synuclein protein are inhibited. In certain embodiments, only the aggregation of tau protein, beta-amyloid protein, or alpha-synuclein protein is inhibited.
  • the aggregation of beta-amyloid protein, tau protein, and alpha-synuclein protein are all inhibited. In certain embodiments, huntingtin aggregation is inhibited. In certain embodiments, prion protein aggregation is inhibited.
  • the methods comprise administering to a subject a therapeutic compound identified or improved using the invention's methods (including analogs and derivatives of such compounds) and/or pharmaceutically acceptable salts thereof, such that the therapeutic compound inhibits the aggregation of an amyloidogenic protein.
  • the subject in these methods may be, e.g., a vertebrate, a mammal, a human, or a non-human animal.
  • the administration may be, e.g., oral, parenteral, transdermal, intrathecal, or intranasal.
  • the methods comprise using known amyloid and/or amyloidogenic fragments to discover and/or optimize compounds that modulate their respective amyloid protein, and other amyloid proteins, e.g., due to the purported relative universality of amyloid pathogenesis.
  • known amyloid and/or amyloidogenic fragments to discover and/or optimize compounds that modulate their respective amyloid protein, and other amyloid proteins, e.g., due to the purported relative universality of amyloid pathogenesis.
  • transthyretin is known to bind beta-amyloid protein in vivo, probably as aggregated beta-amyloid (Tsuzuki et al, 2000, Neurosci. Lett.
  • transthyretin 281:171-174; Liu and Murphy, 2006, Biochemistry 45:15702-15709
  • several inhibitors of transthyretin are also known to inhibit beta-amyloid protein.
  • the utility of transthyretin crystal structures per se for discovering inhibitors of beta-amyloid protein does not approach what might be expected if a similar crystal structure of beta-amyloid were available.
  • the methods of present invention in certain embodiments provide for the utilization of these compounds (despite the lack of known crystal structure of beta-amyloid) to identify new compounds which may be inhibitors or likely to be inhibitors of simultaneous inhibition of several amyloid proteins.
  • the invention relates to methods of virtually screening of compound library, e.g., in silico screening. More specifically, the invention relates to methods of virtually screening a library or libraries to identify compounds that are likely to inhibit amyloid protein aggregation. Such compounds would be of use as therapies for neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, prion diseases, and systemic amyloidosis.
  • a Common Conformational Motif model was constructed using a computer modeling program in interactive session and validated.
  • resveratrol was drawn and minimized to a root-mean-square gradient of 0.05 using the MMFF94 force field with corresponding partial charges.
  • the PDB structure 1BM7 www.pdb.org
  • the binding site of the other flufenamic acid i.e. the other homodimer
  • the flufenamic acid moiety in the resveratrol-occupied binding site and any solvent molecules were deleted.
  • the 17-42 residue sequence of beta-amyloid protein was then drawn and duplicated, and the two strands of beta-amyloid protein were placed in close, non-overlapping proximity to each other and to resveratrol.
  • the entire structure was then correctly protonated/deprotonated for physiological pH and minimized to a root-mean-square gradient of 0.05 using the MMFF94 force field with corresponding partial charges.
  • the orientation of one strand of beta-amyloid protein with respect to the other was a matter of routine experimentation, as the method furnished both a means of adjusting the structure such that a pocket is formed and a means of validating that the candidate model was useful.
  • Several dozen candidate models were created and extracted. One model was validated as the Common Conformational Motif model and is depicted in FIG. 1 .
  • FIG. 2 shows two A ⁇ 17-42 monomers bound to each other near the N terminals in a “double candycane” structure. Each monomer is folded in a loop from residues 23 through 33. The loops are stabilized by intra-loop cationic-anionic interactions between Asp 23 and Lys 28 , and they also show an inter-loop attraction through these same residues. Short-run (up to 100 ns) molecular dynamics calculations on this model indicate that the pocket is stable.
  • the model of amyloid protein aggregation depicted in FIG. 2 was compared to the structure of transthyretin (“TTR”) depicted in FIG. 1 , a number of similarities were observed.
  • the model of amyloid protein aggregation depicted in FIG. 2 provided a similar expanse of hydrophobic contact by lining up along two flattened sides of what would otherwise be a much smaller area of ⁇ -sheet, rather than forming a pocket across the top of several ⁇ -sheets.
  • Anti-amyloid compounds in the amyloid protein aggregation model of FIG. 2 were stabilized in the pocket by Lys and Ser and by exterior Leu, just as in the structure of TTR. Similarly, the positive charge of Lys was shared between the substrate and a nearby Asp in both structures.
  • model of amyloid protein aggregation depicted in FIG. 2 may be a transient species on the way to a stacked dimer using a “dock-lock” mechanism. It is known that many anti-amyloid compounds are multiply anti-amyloid. As such, it was postulated that the utility of the models of the present invention extends beyond the prevention of beta-amyloid protein aggregation and includes additional species of the amyloid proteins, therefore we hypothesized that it can be considered a common conformational motif, of utility for other amyloids beyond beta-amyloid protein.
  • a burgeoning number of compounds have been shown to be anti-amyloidogenic and/or anti-fibrillogenic.
  • a set of these compounds was evaluated in an in silico docking screen using a collection of 79 compounds for which IC 50 s have been computed for AD, tau, and ⁇ -synuclein inhibition.
  • This diverse set comprised polyphenols, benzothiazoles, phenothizaines, macrolides, prophyrins, steroids, derivatives of Congo red, and other anti-amyloid molecules of note.
  • the best docking scores were predicted for purpurogallin, epigallocatechin, catechin gallate, hinokiflavone, myricetin, and gallocatechin gallate—all of which have IC 50 s below 7 ⁇ M.
  • the worst scores in the test were predicted for rutin (expt IC 50 32 ⁇ M) and chlorogenic acid (>40 ⁇ M). Since each compound was represented by several poses, computing a ranking for the remaining compounds was somewhat ambiguous. Nevertheless, the preliminary result was encouraging enough to consider a more stringent criterion: the quantitative structure-activity relationship (QSAR).
  • the Masuda test set was subjected to a CoMFA (comparative molecular field analysis) in order to demonstrate the validity of the model of amyloid protein aggregation depicted in FIG. 2 .
  • CoMFA is a 3-D QSAR technique generally used to posit a “pseudoreceptor” when none is known. Because the algorithm is highly sensitive to alignment of compounds with respect to each other, it is often difficult to produce statistically significant results, even with structurally similar compounds. However, when statistically significant results are obtained, this can be a great confirmation of a pharmacophore hypothesis.
  • the model of amyloid protein aggregation of FIG. 2 has utility in predicting the anti-amyloid effects of candidate compounds tested in the example, and that these compounds have activity against aggregation of ⁇ amyloid protein. It was also postulated that, given conformational similarities between certain amyloid proteins, the candidate compounds may inhibit aggregation of other amyloid proteins, and that dimers of beta-amyloid protein in general may have significant utility in predicting the anti-amyloid effects of candidate compounds on other amyloids such as tau, alpha-synuclein, huntingtin, and/or prion protein.
  • the top 125 compounds were examined for commercial availability, and six of these compounds, evenly distributed with respect to docking score, were acquired from Hit2Lead for in vitro testing. Three of these compounds were inactive in the ThT assay, one was weakly active, and two were highly active (see U.S. Provisional Application Ser. No. 61/092,845 for initial compound disclosures and subsequent use of the model for improving.
  • the present example therefore confirms, e.g., that the model created in Example 1 has utility for rapid lead discovery.
  • This set of 14,172 compounds was then downloaded in MDL SDfile format, imported into a molecular modeling program, stripped of counterions and assigned physiologically appropriate protonation states, and minimized using a molecular mechanics force field.
  • Alzheimer's Disease and related disorders e.g., Parkinson's Disease
  • exploration of hits in the top 1% included those mentioned above as well as the following, in no particular order of activity: Pradimicin, Oleuropein, Tunicamycin, Hyaluronic acid, Coumermycin, Ouabain, Amikacin, Rifaximin, delavirdine, almitrine, and fluvastatin, lumiracoxib, and fenoterol. It was therefore postulated that these compounds may have utility in treating amyloid diseases.
  • This listing can be transcribed into a text file (Unicode, ASCII, or other standard encoding), saved to disc as with extension .PDB, and loaded into a PDB-aware visualization or modeling program in order to reconstruct the model.
  • a text file Unicode, ASCII, or other standard encoding
  • pseudo-crystal structures described in the application have utility and may be used for virtual (in silico) screening in Alzheimer Disease, and, possibly, related disorders, e.g., Parkinson's Disease.
  • ThT aggregation assays of A ⁇ 1-40 and alpha synuclein were evaluated by ThT aggregation assays of A ⁇ 1-40 and alpha synuclein, as set forth below.
  • the column “A-Syn” below is a functional ThT aggregation assay of alpha synuclein.
  • a ⁇ - ThT Structure (IC 50/ ⁇ M) A-syn >20 >20 >20 >20 4.03 2.7 >20 >20 >20 2.1 1.4 >20 — >20 >20 >20 >100 >20 >20 >100 >100 20-50 — 10-20 51 9.0 11.61 0.85 6.34 2.5 142 25.8 >50 >50 >50 8.82 >50 23.2 >50 >50 INACTIVE 23 >50 8.4 39 177.5 9.64 5 44 25 4 100 41.9 184 152 42 217 1035 631 189 ND 84 196 265 66.2 63 174 18 105 1.47 8 5734 14935 23 544 975 92 1.9 361 21.7 9.23 10 27 15 360 428 539 26.6 45 106.6 148 3.5 ⁇ 7 15.5 7.8 2.29 90 54.4 238.4

Abstract

Methods for identifying compounds that are inhibitors or are likely to be inhibitors of amyloid protein aggregation, as well as three-dimensional, non-crystallographic models (i.e. “pseudo-crystal structures”) of amyloid aggregation utilized in the methods, are described. Means for creating the three-dimensional, non-crystallographic models (i.e. “pseudo-crystal structures”) of amyloid aggregation are also described.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Application No. 61/092,826, filed on Aug. 29, 2008, entitled “Methods for identifying inhibitors of amyloid protein aggregation,” herein incorporated by reference in its entirety for all purposes.
  • This application is related to U.S. Provisional Application Ser. No. 61/092,845, entitled “Methods of treating amyloid disease using analogs of 1-(4-nitrophenyl)piperazine,” filed on Aug. 29, 2008, herein incorporated by reference in its entirety for all purposes.
  • FIELD OF THE INVENTION
  • This invention relates to methods of identifying compounds that are inhibitors or are likely to be inhibitors of amyloid protein aggregation, and three-dimensional, non-crystallographic, models of amyloid receptors and models of amyloid protein aggregation utilized in the methods.
  • BACKGROUND OF THE INVENTION
  • The build-up of amyloid proteins in living tissue, a condition known as amyloidosis, is either the cause or a major factor in the pathology of many so-called “amyloid diseases,” e.g., Alzheimer's, Parkinson's, Huntington's, and prion diseases.
  • Certain models of beta-amyloid aggregation that exist in vitro or in vivo (rather than “virtual” (i.e. in silico)) are used for high-throughput screening to discover new compounds that modulate amyloid aggregation (Dolphin et al, 2007, ChemMedChem 2:1613-1623; Gazit, 2006, ACS Chem. Biol. 1:417-419). These existing in vitro or in vivo models however require the synthesis and/or procurement of every candidate compound, as well as the synthesis or incubation of the model systems. In addition, these existing in vitro and in vivo models do not describe a pocket in which the candidate compounds may be inserted, and as such do not furnish a means for de novo design of compounds or for improving the potency of known modulating compounds “virtually” (i.e. in silico).
  • Most prior work in the field of amyloid modeling focused on the structure and dynamics of aggregation, without regard to the discovery of modulating compounds (see for example Buchete, Tycho, and Hummer, 2005, J. Molec. Biol. 353:804-821, which describes the overall structure of beta-amyloid protofibrils; Luhrs et al, 2005, Proc. Natl. Acad. Sci. USA. 102:17342-17347, which verifies the singular axis of fibril addition). These works also do not describe an amyloid protein pocket for non-peptidic compound insertion nor suggest a means of modulating amyloid protein aggregation.
  • Mastrangelo et al. (2006, J. Molec. Biol. 358:106-119) describes a model of beta-amyloid fibrillization, consisting of two protofibrils of beta-amyloid and a pocket found between them that can be used to insert modulating, peptidic compounds. These compounds are specific modulators of beta-amyloid fibrillization and not of aggregation, which is a fundamentally different process from fibrillization.
  • One treatment of amyloid diseases utilizes peptides as modulators of disease targets, being amyloid proteins in this case. (Findeis et al, 1999, Biochemistry 38:6791-6800; Ghanta et al, 1996, J Biol Chem 271:29525-29528; Tjernberg, 1996, J Biol Chem 271:8545-8548; Kokkoni et al, 2006, Biochemistry 45:9906-9918). However, utilities of the peptidic compounds described in these references as drugs is doubtful, e.g., due to in vivo stability issues.
  • While a number of non-peptidic compound classes have been identified as inhibitors of beta-amyloid build-up, the discovery of further compound classes, as well as the further optimization of known compound classes to improve potency may be haphazard due to the lack of known crystal structures for beta-amyloid protein.
  • The amyloid diseases remain invariably fatal using current medical practice.
  • Accordingly, there exists a need for novel methods of identifying compounds that are inhibitors or are likely to be inhibitors of amyloid protein aggregation, and three-dimensional, non-crystallographic, models of amyloid receptors and models of amyloid protein aggregation which may be utilized in these methods.
  • DEFINITIONS
  • The following terms should have the following meaning whenever used in the present specification, regardless whether the use is singular or plural.
  • “Crystallized amyloid protein” means an amyloid protein for which a binding pocket is known.
  • “Uncrystallized amyloid protein” means an amyloid protein for which a binding pocket is unknown.
  • “Multiply anti-amyloid compound” means a compound that has activity against both a crystallized amyloid protein and an uncrystallized amyloid protein.
  • “Modulating amyloid aggregation” means changing the distribution of oligomers such that amyloid deposition is reduced in a subject and encompasses a process of inhibition of aggregation.
  • “Model” means a molecular model, which is a representation of atoms and their positions with respect to each other, substantially including such features as covalent bonds between highly interacting atoms in a molecule and/or non-covalent interactions between the atoms of two or more molecules.
  • “Super-model” means a model of models.
  • “Computer modeling program” means any program that can visualize or otherwise manipulate molecular models on a computer, preferably also featuring molecular mechanics or molecular dynamics routines.
  • “Interacts” means chemical interactions, especially those that result in covalent bonds being formed between atoms in a molecule and/or non-covalent bonds between the atoms of two or more molecules. If said molecules are composed of amino acid residues, it is commonly said that two residues interact when at least one atom from one residue interacts with at least one atom from the other residue.
  • “Non-crystallographic” means that the model was not directly obtained by the techniques of crystallography, especially X-ray crystallography.
  • “Candidate model” means a model of an uncrystallized amyloid protein that is to be evaluated for utility as a pseudo-crystal structure of a crystallized or uncrystallized amyloid protein and/or amyloid aggregation.
  • “Halogen” means fluorine, chlorine, bromine, or iodine.
  • “Extracting” means the step of operating the computer modeling program such that the relevant subset is highlighted, saved, and/or placed in a different workspace within the program for the purposes of manipulating the subset without reference to the rest of a model.
  • Such a subset may be considered a model in and of itself.
  • “Validating” means the step of performing docking and/or quantitative structure-activity relationship (QSAR) calculations in a computer modeling program, in order to verify that a given model can generally correctly classify and/or predict the anti-amyloid activity of a set of known positive and negative controls for anti-amyloid activity. In preferred embodiments of the invention, the set is as described in the Examples below.
  • “Constructing” means operating a computer modeling program to instantiate an in silico representation of a given model.
  • “Selecting a list of candidate compounds” means choosing a set of compounds for evaluation in a model. Selection of said set may be reference to commercially available screening libraries such as Hit2Lead or Enamine, by reference to an internal list of available and/or proprietary compounds for in vitro or in silico screening, or by utilizing lead-hopping or de novo design methods.
  • “Iterative docking” means docking each candidate compound in turn into the model using a computer modeling program.
  • “Scoring” means applying formulas to obtain a score from “scoring functions” pertaining to drug-target interactions, the output of which furnishes a measurement of the degree of complementarity of the compound with respect to the pocket.
  • “Score cutoff” means some value, usually an absolute energy, that can be set higher or lower depending on the level of activity that is deemed to separate active from inactive, or as a reference between more active and less active.
  • “Improving potency” means providing a means to furnish a compound analogous to an active compound that has minor structural differences which produce a better modulating effect with respect to the target.
  • OBJECTS AND SUMMARY OF THE INVENTION
  • The invention provides methods of utilizing three-dimensional, non-crystallographic, models of amyloid aggregation to identify compounds that are inhibitors or are likely to be inhibitors of amyloid protein aggregation. It also provides three-dimensional, non-crystallographic models, of amyloid receptors and models of amyloid protein aggregation utilized in these methods, as well as methods of producing such models for these methods.
  • It is an object of the invention to provide three-dimensional pseudo-crystal structures of uncrystallized amyloid proteins.
  • It is a further object of the invention to provide virtual three-dimensional models of amyloid aggregation and methods for producing such models.
  • It is also an object of the invention to provide virtual three-dimensional models of amyloid aggregation, indicating pockets in which candidate anti-amyloid models may be placed.
  • It is another object of the invention to provide methods of constructing three-dimensional pseudo-crystal structures or models of uncrystallized amyloid proteins and of validating the models to verify that the structures can classify and predict anti-amyloid activity of candidate compounds.
  • It is a further object of the invention to provide methods of utilizing the three-dimensional pseudo-crystal structures or models to identify compounds that modulate or are likely to modulate amyloid aggregation.
  • It is an additional object of the invention to provide methods of improving the anti-amyloid potency of candidate compounds.
  • It is a further object of the invention to provide methods of predicting potential interactions between candidate compounds and amyloid proteins.
  • It is also an object of the invention to provide methods for identification and characterization of previously unknown binding pockets or conformational regions of amyloid proteins.
  • It is yet another object of the invention to use compound identified or improved using the methods of the invention as therapies for diseases in which amyloidosis occurs.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a depiction of the super-model developed in Example 1, in which the crystallized amyloid protein is a fragment of transthyretin, the multiply anti-amyloid compound is resveratrol, and the uncrystallized amyloid protein is a dimer of Aβ17-42.
  • FIG. 2 is a stereographic view of the pseudo-crystal structure, a model of amyloid protein aggregation, of Example 1 with resveratrol bound in its pocket.
  • DETAILED DESCRIPTION
  • Amyloidosis refers to a variety of conditions in which amyloid proteins are abnormally deposited in organs and/or tissues.
  • Certain types of amyloidosis occur principally in the central nervous system, e.g., with aggregation of beta-amyloid protein in Alzheimer's Disease, alpha-synuclein in Parkinson's Disease, huntingtin protein in Huntington's Disease, and prion protein in Creutzfeldt-Jacob and other prion diseases. Other types of amyloidosis are systemic in nature, as, e.g., with aggregation of transthyretin in senile systemic amyloidosis.
  • Historically, aggregations of protein were classified as “amyloid” if they displayed apple-green birefringence under polarized light when stained with the dyes Congo red or Thioflavin T (ThT) (Sipe and Cohen, 2000, J. Struct. Biol. 130:88-98). That definition of “amyloid protein” has been expanded in recent years to apply to any fibrous structure which is ordered and insoluble, and which is composed of fibrils of approximately 10 nm in width and up to several microns in length (Zheng, Ma, and Nussinov, 2006, Phys. Biol. 3:P1-P4).
  • An “amyloidogenic” polypeptide is one which, regardless of sequence, can polymerize in a cross-β sheet conformation in vitro or in vivo to form amyloid protein (Xu, 2007, Amyloid 14:119-31). “Amyloidogenic” polypeptides include, e.g., amyloid light chain, amyloid associated protein, β amyloid, transthyretin, β2 microglobulin, amylin, prion related protein, and other amyloid proteins.
  • All amyloid proteins have commonalities. However, whether certain amyloid proteins are related depends on the amyloid proteins having commonality of both pathology and location. For example, beta-amyloid protein and huntingtin protein are both located in a human brain, but their divergence in pathology is sufficient to consider them unrelated. Beta-amyloid protein, tau protein, and alpha-synuclein protein on the other hand are related, all being located in the human brain and being thought to have overlapping roles in the pathology of neurodegenerative diseases, e.g., Alzheimer's disease and Parkinson's disease. Nevertheless, given the preponderance of non-peptidic anti-amyloid compounds that inhibit aggregation of more than one amyloid protein, it is reasonable to assume that models of amyloid protein aggregation provided by the present invention will have significant utility even with unrelated amyloid proteins, in addition to having utility with the related amyloid proteins (i.e., beta-amyloid protein, tau protein, and alpha-synuclein protein).
  • A lock and key analogy is commonly used to characterize drug-target interactions where a specific “key” (compound/drug) interacts only with its respective molecular “lock” (target/receptor). An appropriate degree of shape and electronic complimentarily between the drug and target must occur to produce drug-target interactions which are necessary for a pharmacological response. The specific location on the “lock” or target is often referred to as the active or catalytic site, and the intuitive shape of the active site is that of a pocket. Thus a “binding pocket,” or more simply “pocket,” is used in the art to refer to that space on the target in which a molecule can be inserted to modulate activity of the target. The three-dimensional shape and electronic properties of the pocket form the basis for rational drug design and for virtual screening techniques (e.g., in silico) utilized in the methods of the present invention, provides means for identifying the pocket for non-peptidic compound insertion and suggests means for modulating amyloid protein aggregation.
  • “Rational drug design” attempts to formulate drug design hypotheses that specify and optimize the physical contacts between a drug and its target, often by aid of a computer. Such “computer-aided drug design” generally depends on high resolution, three-dimensional models of the target, and such models are usually constructed by X-ray crystallography of a crystal form of the target. Crystallographic techniques are the primary means of obtaining three-dimensional structure and binding information in the art. In the absence of a solved, high-resolution (i.e. below 2 angstroms) crystal structure of a given amyloid protein indicating the binding mode of an anti-amyloid compound and/or the chemical interactions essential to aggregation, rational design of anti-amyloid compounds for said protein was generally considered haphazard and difficult prior to the methods and models of the present invention.
  • The virtual screening techniques utilized in the methods of the present invention and the virtual models of the present invention allow, e.g., for the virtual identification of the binding pockets of amyloid proteins, and construction of three-dimensional non-crystallographic models of amyloid receptors and models of amyloid aggregation. The methods of the present invention generally do not require construction of crystal forms of the target by X-ray crystallography, and instead provide or utilize non-crystallographic models (“pseudo-crystal structures”) that could approximate or exceed the utility of traditional crystal structures.
  • Candidate Models
  • The invention provides for candidate models of amyloid receptors and of amyloid protein aggregation to be constructed, evaluated and subsequently validated. The models of the present invention are based, in part, both on the symmetry inherent in many anti-amyloid compounds and on the multi-target activity of such compounds.
  • The models of the present invention may be referred to as “pseudo-crystal structures” or as surrogate models, as the models are not obtained by the technique of crystallography. As stated above and as demonstrated by the examples below, the utility of “pseudo-crystal structures” of the present invention may approach and exceed that of crystal structures.
  • Successful candidate models (e.g., pseudo-crystal structures) will include elements directly related to the process of aggregation and will be of sufficient detail to enable the design of compounds for disrupting or otherwise altering the interactions essential to amyloid protein aggregation. Indeed, if a pseudo-crystal structure does not indicate the binding mode of an anti-amyloid compound and/or the chemical interactions essential to aggregation, then it is not a proper model of amyloid protein aggregation and would not be expected to have utility to enable the design of anti-amyloid compounds.
  • Successful candidate models in accordance with the present invention may comprise pseudo-crystal structures of uncrystallized amyloid proteins. Such pseudo-crystal structures generally comprise two parts. The first part generally comprises an amyloid monomer, and the second part generally comprises an amyloid monomer or an amyloid oligomer. The first part is generally positioned with respect to the second part such that it forms a pocket in conjunction with the second part, such that a candidate compound may be inserted into the pocket, thereby potentially modulating amyloid aggregation.
  • The amyloid monomer comprises one peptide chain from an amyloid oligomer or amyloid protein. The amyloid monomer in the first part and the amyloid monomer (or each monomer in an amyloid oligomer) in the second part may comprise the same sequence or different sequences with respect to the first part.
  • The amyloid oligomer comprises a heterogeneous or homogeneous link comprising an amyloid protein (or amyloidogenic fragments thereof). For example, in certain embodiments, the amyloid oligomer comprises an amyloid protein (e.g., either beta-amyloid protein, tau protein, alpha-synuclein protein, huntingtin protein, or prion protein) or amyloidogenic fragments thereof; and in other embodiments the amyloid oligomer comprises two or more amyloid proteins (e.g., a combination of beta-amyloid protein and tau protein, a combination of beta-amyloid protein and alpha-synuclein protein, etc.) or amyloidogenic fragments thereof. The amyloid oligomer may also comprise a protein which has not yet shown to be amyloidogenic or a portion of such protein.
  • In certain embodiments, the amyloid oligomer or amyloidogenic fragment thereof comprises a region comprising approximately residues 17 through 40 of beta-amyloid protein of length 40; approximately residues 17 through 42 of beta-amyloid protein of length 42; approximately residues 1 through 15 of alpha-synuclein protein; approximately residues 25 through 40 or 275 through 280 of tau protein; approximately residues 90 through 110 of the major prion precursor protein; or approximately residues 5 through 20 of the huntingtin protein.
  • In certain embodiments, the model comprises the pseudo-crystal structure formed by SEQ ID: 1 and SEQ ID: 2 and interacting in the manner depicted stereographically in FIG. 2, which is a model of amyloid protein aggregation of Example 1, with resveratrol bound in its pocket. Three-dimensional features of the model may be appreciated by a person of normal binocular vision by superimposing the images visually.
  • In certain embodiments, the first part comprises substantially SEQ ID: 1 and the second part comprise substantially SEQ ID: 2, wherein SEQ ID: 1 is
  • Lys Val Phe Phe Ala Glu Asp Val Gly Ser Asn
    1                   6                   11
    Lys Gly Ala Ile Ile Gly Leu Met Val Gly Gly
        Val     16                      21
    Val Ile Ala
            26

    and,
  • SEQ ID: 2 is
  • Lys Val Phe Phe Ala Glu Asp Val Gly Ser Asn
    1                   6                   11
    Lys Gly Ala Ile Ile Gly Leu Met Val Gly Gly
        Val     16                      21
    Val Ile Ala
            26
  • In certain embodiments, the first part is further positioned with respect to the second part such that the amino acid Val at position 8 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2; the amino acid Gly at position 9 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2; the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Gly at position 9 of SEQ ID: 2; the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Ser at position 10 of SEQ ID:2; and the amino acid Lys at position 12 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2.
  • In certain embodiments, the first part is positioned with respect to the second part in the orientation shown in FIG. 2. FIG. 2 is a stereographic view of the pseudo-crystal structure, a model of amyloid protein aggregation, of Example 1 with resveratrol bound in its pocket. The model of FIG. 2 features two Aβ17-42 monomers bound to each other near the N terminals in a “double candycane” structure. Each monomer is folded in a loop from residues 23 through 33, which is a conformation well supported by electron microscopy and NMR spectroscopy. The loops are stabilized by intra-loop cationic-anionic interactions between Asp23 and Lys28, and they also show an inter-loop attraction through these same residues. Short-run (up to 100 ns) molecular dynamics calculations on this model indicate that the pocket is stable.
  • In certain embodiments, a virtual non-crystallographic model of amyloid protein aggregation comprises a pseudo-crystal structure comprising a three-dimensional model of an uncrystallized monomeric amyloid protein and a three-dimensional model of an uncrystallized amyloid protein comprising one or more amyloid peptides; said three-dimensional model of an uncrystallized amyloid protein is positioned with respect to said amyloid monomer model such that it forms a pocket in conjunction with said amyloid monomer model, such that a candidate compound may be inserted into said pocket thereby, e.g., potentially modulating amyloid aggregation.
  • Aggregation is a fundamentally different process from fibrillization. Aggregation occurs among monomers and oligomers of incipient amyloid protein, while fibrillization is directed to the further lengthening of pre-formed amyloid protofibrils and subsequent assembly into fibrils. Modulators of aggregation are preferred to modulators of fibrillization for treatment of amyloid diseases, since soluble oligomers of beta-amyloid protein are thought to be the pathogenic species in Alzheimer's Disease, rather than the mature fibrils thereof (Klyubin et al, 2008, J. Neurosci. 28:4231-4237; Townsend et al, 2006, J. Physiol. 572:477-492).
  • For clarity, it should be understood that a model of amyloid protein aggregation promulgated in the present application is substantially a model of addition of monomeric amyloid peptide to an amyloid oligomer or an amyloid protein. In certain embodiments, the monomeric amyloid peptide and the amyloid oligomer (or the amyloid protein) is composed of chains of different amyloid peptides. In other embodiments, the monomeric amyloid peptide and the amyloid oligomer or protein are composed of chains of the same amyloid peptide.
  • In certain embodiments of the invention, modulating amyloid aggregation may result in greatly increasing the proportion of amyloid peptide assemblies with size n, where n is between 1 and 10. In other embodiments, modulating amyloid aggregation may result in increasing the proportion of amyloid peptide assemblies with size n, where n is between 1 and 5. In still other embodiments of the invention, modulating amyloid aggregation may result in increasing the proportion of amyloid peptide monomer (i.e. where n is only 1).
  • In certain embodiments, a virtual non-crystallographic model of amyloid protein aggregation comprises a pseudo-crystal structure comprising an amyloid monomer model and a three-dimensional model of an uncrystallized amyloid protein. The “amyloid protein” in the minimal case may comprise only one amyloidogenic peptide having substantially the same sequence as an amyloid protein or an amyloidogenic fragment thereof. The “amyloid protein” may also comprise a plurality of chains of one amyloid protein or amyloidogenic portions thereof, and does not include other amyloid protein or amyloidogenic portions thereof. In certain embodiments, the plurality of chains are aggregated according, e.g., to a building-up principle.
  • In certain embodiments, the amyloid monomer model and the three-dimensional model of an uncrystallized amyloid protein are both composed of beta-amyloid protein or an amyloidogenic fragment thereof. For example, the amyloids may comprise substantially the sequence of beta-amyloid 1-42 and/or the sequence of beta-amyloid 1-40 and/or the sequence of the beta-amyloid fragment 25-35 and/or the sequences of beta-amyloid fragment 17-42, which are sequences known to form aggregated beta-amyloid (Hughes et al, 2000, J Biol Chem 275:25109-25115).
  • In certain embodiments, candidate models of amyloid protein aggregation comprise a pseudo-crystal structure comprising a three-dimensional model of an uncrystallized, monomeric amyloid protein and a three-dimensional model of an uncrystalized amyloid protein comprising one or more amyloid peptides positioned with respect to the monomeric amyloid protein model such that it forms a pocket in conjunction with the monomeric amyloid protein. In one embodiment, the monomeric amyloid model is substantially SEQ ID: 1; and the amyloid protein model comprises SEQ ID: 2.
  • In certain embodiments three-dimensional, non-crystallographic models, of amyloid aggregation comprise pseudo-crystal structures for amyloid proteins that lack extant crystal structures (e.g., beta-amyloid protein, tau, alpha-synuclein, huntingtin, and prion protein, etc.).
  • In certain embodiments of the invention, a model may be recorded in a mathematical form using a combination of atomic information and three-dimensional coordinates (e.g. in the PDB format, or in the proprietary format of a computer modeling program), which are then visualized or otherwise represented virtually, e.g., in a computer modeling program. In certain embodiments, the model reflects the likely protonation state at physiological pH such that basic moieties of the model are protonated and acidic moieties are deprotonated in a manner consistent with valence considerations. In further embodiments, force fields and charges are applied appropriate to the model such that molecular mechanics and/or dynamics can be used; details on these and other computational techniques characteristically found in computer modeling programs are described, e.g., in A. Leach, Molecular Modeling: Principles and Applications, 2nd edition, Prentice Hall, 2001, herein incorporated by reference.
  • The models of the present invention may, e.g., enable the identification of new chemical classes of modulators of their respective amyloid protein and related amyloid proteins. For example, if a candidate compound has a three-dimensional shape and electronic properties that will allow it to “fit” into the pseudo-crystal structure and interact with the pseudo-crystal structure, it is likely to be a potential modulator of the amyloid protein aggregation.
  • The models of the present invention may also provide a means of improvement in potency for compounds known to have modulating effects on a relevant amyloid protein, as they may suggest structural modification to the compounds known to have modulating effects on the amyloid protein, to allow the compounds to better “fit” and interact with the three-dimensional pseudo-crystal structure. They may also provide means to the de novo design of compounds which may “fit” into the three-dimensional structure.
  • The models may further provide means for the identification and discovery of the compounds which may be useful in modulation of either or both aggregation and fibrilization.
  • Construction of Models
  • The present invention further provide means for constructing surrogate models (i.e., non-crystallographic virtual models) of amyloid protein aggregation which may be used for drug design much as crystal structures are customarily used but at a lower cost and without a need for synthesis of every candidate compound.
  • For example, the three-dimensional, non-crystallographic models of amyloid aggregation models in accordance with the present invention may be constructed, e.g., virtually (e.g., in a computer modeling system), depicted pictorially, instantiated in a physical form by way of a modeling kit, or visualized mentally. The virtual modeling afforded by the present invention allows for greatly reduced cost and labor as compared to in vitro or in vivo systems, as it does not require the synthesis and/or procurement of every candidate compound. In particular, it allows for the screening and prediction of the anti-amyloid properties of a candidate compound based on its three-dimensional shape and electronic configuration, even before the compounds are synthesized. It disposes of a potentially great number of candidate compounds which will not “fit” into the three-dimensional, non-crystallographic, models of the amyloid aggregation afforded by the present invention, and are therefore are unlikely to have anti-amyloid activity.
  • Generally, construction of the non-crystallographic models is performed manually, using a molecule or peptide/biomolecule building routine as included in a computer modeling program. In some embodiments of the invention, construction may proceed automatically through the reading of a previously constructed file or sequence.
  • In certain embodiments, the invention provides means for constructing virtual non-crystallographic models of amyloid aggregations for proteins for which the three-dimensional structure and binding information (e.g., binding mode and chemical interactions essential for aggregation) are known. In other embodiments, the invention provides means for constructing virtual non-crystallographic models of amyloid aggregations for proteins for which the three-dimensional structure and binding information (e.g., binding mode and chemical interactions essential for aggregation) are unknown. In certain embodiments, the means for constructing virtual non-crystallographic models of amyloid aggregations for proteins for which the three-dimensional structure and binding information are known and the means for constructing virtual non-crystallographic models of amyloid aggregations for proteins for which the three-dimensional structure and binding information are unknown are the same.
  • In certain embodiments, a pseudo-crystal structure of an uncrystallized amyloid protein can be virtually identified and constructed by manipulating a computer program to orient an uncrystallized amyloid protein and/or oligomer such that a pocket is formed around a multiply anti-amyloid compound already bound in the crystallized amyloid protein. The pocket has a particular three-dimensional shape and electronic properties and represents, e.g., a pseudo-crystal structure of the uncrystallized amyloid protein. In certain embodiments, the multiply anti-amyloid compound is removed and is not part of the pseudo-crystal structure. In other embodiments, the multiply anti-amyloid compound is part of the pseudo-crystal structure.
  • In certain embodiments, a binding pocket corresponds to a common conformational region on a variety of amyloid proteins, e.g., the Common Conformational Motif (CCM). For example, the CCM may correspond to a conformational region found in both Aβ and tau; or a conformational region found in both Aβ and alpha-synuclein; or a conformational region found in both Aβ and transthyretin. The conformational region may also be common to Aβ, tau and alpha-synuclein (which are all located in the human brain and being thought to have overlapping roles in the pathology of neurodegenerative diseases). The binding pocket may therefore be useful for the identification of candidate compounds which are likely to inhibit the aggregation of several amyloid proteins simultaneously (e.g., Aβ and tau; Aβ and alpha synuclein; Aβ, tau and alpha synuclein, etc.). It should be understood for this reason that the CCM is not strictly sequence-dependent.
  • In certain embodiments, a method of creating pseudo-crystal structures of uncrystallized amyloid protein receptor comprises (1) applying molecular mechanics and/or dynamics to a three-dimensional super-model comprising a crystallized amyloid protein model, a model of multiply anti-amyloid compound bound in the pocket of the crystallized amyloid protein or oligomer, and a candidate model of an uncrystallized amyloid protein, such that a pocket on the uncrystallized amyloid protein or oligomer is formed around the multiply anti-amyloid compound; (2) extracting said uncrystallized amyloid protein model from said super-model; and (3) validating said uncrystallized amyloid protein model with respect to experimental data so as to confirm its utility as a pseudo-crystal structure. In certain embodiments, the extracted model comprises the multiply anti-amyloid compound. In other embodiments, the extracted model is free of the multiply anti-amyloid compound.
  • Example 1 below describes an exemplary method for construction of one pseudo-crystal structure of beta-amyloid protein in accordance with one embodiment of the invention. In the method of Example 1, the uncrystallized amyloid protein is an amyloidogenic fragment of beta-amyloid protein, the crystallized amyloid protein is transthyretin protein, and the multiply anti-amyloid compound is resveratrol. In particular, the pseudo-crystal structure of Example 1 has two parts: one monomer of an amyloidogenic beta-amyloid fragment (i.e. the “amyloid monomer”); and another monomer of an amyloidogenic beta-amyloid fragment, which could have multiple additional peptides attached thereon without disrupting its interaction with the amyloid monomer (as such, this second part is sometimes referred to as the “amyloid oligomer”, as it is only contingently monomeric).
  • In certain embodiments, the invention provides a method of constructing pseudo-crystal structures of uncrystallized amyloid protein which are models of amyloid protein aggregation. This method's first step is to apply molecular mechanics and/or dynamics to a three-dimensional super-model comprising a crystallized amyloid protein model, a model of multiply anti-amyloid compound bound in the pocket of the crystallized amyloid protein, and a candidate model of an uncrystallized amyloid protein or oligomer, such that a pocket on the uncrystallized amyloid protein is formed around the multiply anti-amyloid compound.
  • The method's second step is to extract said uncrystallized amyloid protein or oligomer model from the super-model. In certain embodiments, the extracted model comprises the multiply anti-amyloid compound. In other embodiments, the extracted model is free of the multiply anti-amyloid compound.
  • The method's third step is to validate said uncrystallized amyloid protein model with respect to experimental data so as to confirm its utility as a pseudo-crystal structure.
  • The crystallized amyloid protein may be transthyretin, islet amyloid polypeptide, beta-2-microglobulin, and insulin protein; and the uncrystallized amyloid protein may be either of beta-amyloid, tau, alpha-synuclein, huntingtin, or prion protein; an amyloidogenic fragment of any of the foregoing, or composed of a heterogeneous combination of such proteins.
  • A multiply anti-amyloid compound may be resveratrol, or a compound having low energy conformations that overlap geometrically and electrostatically with resveratrol. In certain embodiments, a multiply anti-amyloid compound is a compound selected from the lists of compounds provided in paragraphs [00106] to [0117] of the present application.
  • Once a model (i.e., a pseudo-crystal structure) is constructed, it is then extracted and validated with respect to experimental data to verify that it can classify and predict, e.g., the anti-amyloid activity of candidate compounds. In certain embodiments, validation comprises performing docking and/or quantitative structure-activity relationship (QSAR) calculations in a computer modeling program, in order to verify that a given model can generally correctly classify and/or predict the anti-amyloid activity of a set of known positive and negative controls for anti-amyloid activity. Examples of computer modeling programs include but are not limited to MOE, Sybyl, Cerius2, CHARMm, DOCK, AUTODOCK, GLIDE, and FlexX. Manuals of these computer modeling programs are herein incorporated by reference in their entirety.
  • Example 2 below describes an exemplary method for validating an exemplary of amyloid protein aggregation.
  • Once validated, the resulting pseudo-crystal structure can be used as a three-dimensional, non-crystallographic, model of amyloid aggregation in the methods of the present invention.
  • The Methods of the Invention
  • The methods of the invention may be used to identify or improve the potency of candidate compounds that are anti-amyloid with respect to the amyloid protein or fragment thereof depicted in a particular three-dimensional, non-crystallographic, model, or is related to the amyloid protein or fragment thereof that is depicted in the model. The methods of the invention may also be used to identify, or improve the potency of, compounds that are anti-amyloid with respect to an amyloid protein or fragment thereof that is unrelated to the amyloid protein or fragment thereof that is depicted in a particular three-dimensional non-crystallographic model.
  • In certain embodiments, the methods of the present invention utilize the “virtual” non-crystallographic models to identify new compounds which may be inhibitors or likely to be inhibitors of amyloid protein aggregation, as well as improve existing modulators of amyloid proteins, all without the construction of crystal form of the target by X-ray crystallography.
  • In certain embodiments, the models are used to predict the potential interactions between a candidate compound and an amyloid protein, e.g., by allowing visualization of the candidate compound fitting into the binding pocket.
  • One empirical determinant for whether two atoms interact is their proximity to each other in a given chemical system. For example, a covalent bond between carbons is approximately 1.5 angstroms in interatomic distance, while a weak non-covalent bond between atoms could be as long as 3 angstroms (although stronger non-covalent bonds may approach 1.5 angstroms in length). The hydrogens in the head group of a protonated Lys residue interacting with the oxygens in the head group of a deprotonated Glu residue is an example of a primarily ionic non-covalent bond. The hydrogen in the head group of a Ser residue interacting with the carbonyl oxygen in the backbone of a different Ser residue, or a hydrogen in the backbone of a Gly residue interacting with the oxygen in the head group of a Ser residue, are examples of primarily hydrogen bonds. The virtual visualization provided by the methods of the present invention supply a means of predicting the likelihood of these interactions between a candidate model compound and a particular binding pocket. If the visualization shows that the modeling compound is unlikely to interact with a particular binding pocket on an amyloid protein, it is unlikely that the modeling compound will be useful as an inhibitor, e.g., of the amyloid protein aggregation. To the contrary, if the compound is likely to interact with the binding pocket of the amyloid aggregation, it is likely that the compound may be capable of modulating activity and potentially preventing or minimizing aggregation of this particular amyloid protein.
  • In the context of a pseudo-crystal structures described in the present application, one of the most important intermolecular interactions are those which hold the amyloid oligomer together as well as those which attract the amyloid monomer. The latter said interactions are pivotal to the conformation and eventual incorporation of the amyloid monomer into the amyloid oligomer, a process which is believed to continue repeatedly until reaching the critical size at which fibrillization occurs. By inserting into the pocket where these intermolecular interactions occur, the candidate compound may disrupt or otherwise alter the process of aggregation. Whether the candidate compound can do so depends, e.g., on its shape and electronic complementarity, i.e., the ability of moieties on the compound to interact with important residues in the pocket. The visualization of the fitting provided by the present invention will allow the prediction of whether the candidate compound is likely to interact with the important residues of the pocket. Other intermolecular interactions, which may be less important to describing aggregation, yet are potentially of importance in achieving the desired accuracy with respect to a given model of amyloid protein aggregation. The presently claimed methods may also have utility in predicting these other intermolecular interactions.
  • A common interaction within such systems involves ionic, acid-base interactions such as Lys-Glu and Lys-Asp as well as hydrogen-bonding interactions such as Ser-Ser. Therefore, regions of amyloid proteins or amyloidogenic fragments thereof that should make excellent models include but are not limited to the following: in beta-amyloid protein of length 40, approximately residues 17 through 40; in beta-amyloid protein of length 42, approximately residues 17 through 42; in alpha-synuclein, approximately residues 1 through 15; in tau protein, approximately residues 25 through 40 or approximately residues 275 through 280; in the major prion precursor protein, approximately residues 90 through 110; and in huntingtin, approximately residues 5 through 20.
  • Chemical interaction is a matter of degree; especially in non-covalent bonds, the type of bonding involved can often be an admixture of one or more types of bonding named above, possibly including other effects such as hydrophobic or van der Waals interactions.
  • Selected examples of chemical interactions which may be predicted utilizing the methods of the present invention include (but are not limited) to non-covalent bonds including, e.g., ionic, dipole-dipole, dipole-induced dipole, and hydrogen bonding.
  • Covalent bonds may also be predicted by the methods of the present invention. Covalent bonds are the most permanent of chemical interactions and are generally not altered by intermolecular interactions, which are governed primarily by non-covalent bonds.
  • In certain embodiments, the present invention provides method for identification and characterization of a previously unknown binding pocket or a conformational region of an amyloid, and subsequent utilization of the previously unknown pocket/conformational region as a screening tool for identification and optimization of anti-amyloid compounds. In these embodiments, the binding pocket is identified in silico, e.g., by using molecular modeling (especially optimization and dynamical techniques) on a “super-model” comprising a model of amyloid protein for which a binding pocket is known (a “crystallized amyloid protein”); a model of a compound (a “multiply anti-amyloid compound”) that has activity against said crystallized amyloid protein, as well as having activity against an amyloid protein for which a binding pocket is unknown (an “uncrystallized amyloid protein”); and a candidate model for said uncrystallized amyloid protein. In certain embodiments, these methods further allow for the construction of a pseudo-crystal structure of beta-amyloid protein or a receptor having utility in identification and optimization of anti-amyloid compounds.
  • A crystallized amyloid protein may be selected, e.g., from the group consisting of transthyretin, islet amyloid polypeptide, beta-2-microglobulin, and insulin protein. In certain embodiments, the crystallized amyloid protein is transthyretin protein, or an amyloidogenic fragments thereof.
  • An uncrystalized amyloid protein may be selected, e.g., from the group consisting of beta-amyloid, tau, alpha-synuclein, huntingtin, and prion protein. In certain embodiments, the uncrystalized amyloid protein is beta-amyloid proteins, and amyloidogenic fragments thereof.
  • A multiply anti-amyloid compound may be any compound which has activity against any crystallized amyloid protein. In a preferred embodiment, the multiply anti-amyloid compound is resveratrol or an analog thereof.
  • In certain embodiments, the crystallized amyloid protein is selected from the group of transthyretin, islet amyloid polypeptide, beta-2-microglobulin, and insulin protein; and the uncrystallized amyloid protein is selected from the group of beta-amyloid, tau, alpha-synuclein, huntingtin, and prion protein. In certain embodiments, the crystallized amyloid protein is transthyretin protein or an amyloidogenic fragment thereof, and the uncrystallized amyloid protein is beta-amyloid protein or an amyloidogenic fragment thereof, and the multiply anti-amyloid compound is resveratrol.
  • The invention further encompasses and provides a method of identifying compounds that modulate amyloid aggregation. The method generally comprises the steps of constructing a pseudo-crystal structure in a computer modeling program; selecting a list of candidate compounds; constructing said candidate compounds in a computer modeling program; performing an iterative docking and scoring of all candidate compounds, by means of docking each candidate compound into the pocket formed by said model and scoring each candidate compound to reflect its degree of complementarity with respect to said pocket; and identifying compounds that modulate amyloid aggregation by reference to a score cutoff that substantially distinguishes active compounds from inactive compounds.
  • A candidate compound may, e.g., be a compounds of Formulas Ia, Ib, Ic, pharmaceutically acceptable salts, stereo-isomers, polymorphs, metabolites, prod-drugs, analogs and combinations of any of the foregoing:
  • Figure US20160283652A1-20160929-C00001
  • wherein
  • R1 is selected from the group consisting of H, nitro, carboxylic acid, alkylcarboxylic acid, acetamide connected in either direction, N-(2-ethanol)amine, N-(2-morpholinethyl)amine, amine optionally substituted with one or more alkyl groups, amide optionally substituted with one or more alkyl groups, and alkoxy;
  • R2 is selected from the group consisting of H, carboxylic acid, alkyl, alkanoyl, alkanesulfonyl, benzenesulfonyl, phenonyl optionally substituted with any one or more of alkoxy, halogen, or alkyl groups, benzyl optionally substituted with any one or more of alkoxy, halogen, or alkyl groups, and amide optionally substituted with any one or more of alkyl or aryl groups;
  • R3 is selected from the group consisting of H, alkyl, furanylalkyl, thiophenealkyl, alkanoyl, phenyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups, benzyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups, and phenonyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups; and
  • R4 is selected from the group consisting of H, alkyl, or phenyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups.
  • In certain embodiments, R1 is selected from the group consisting of nitro, acetamide connected in either direction, N-(2-ethanol)amine, amino optionally substituted with any one or more alkyl groups, and amide optionally substituted with any one or more alkyl groups; R2 is selected from the group consisting of carboxylic acid, amide optionally substituted with any one or more of alkyl or aryl groups, and phenonyl optionally substituted with any one or more of alkoxy, alkyl, or aryl groups; R3 is selected from the group consisting of methyl, phenyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups, benzyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups, and phenonyl optionally substituted with any one or more halogen, alkyl, or alkoxy groups; and R4 is selected from the group consisting of H, alkoxy, and aryl.
  • In other embodiments, R1 is selected from the group consisting of nitro, acetamide connected in either direction, N-(2-ethanol)amine, amino optionally substituted with methyl or dimethyl, amide optionally substituted with methyl, ethyl, dimethyl, or diethyl, and methoxy; R2 is selected from the group consisting of phenonyl optionally substituted with any one or more of methoxy, alkyl, or halogen, amide optionally substituted with any one or more of methyl, phenyl, benzyl, or dimethyl, and carboxylic acid; R3 is selected from the group consisting of methyl, phenyl optionally substituted with any one or more of halogen, alkyl, or methoxy, benzyl optionally substituted with any one or more of halogen, alkyl, or methoxy, and phenonyl optionally substituted with any one or more of halogen, alkyl, or methoxy; and R4 is selected from the group consisting of H, methyl, and phenyl optionally substituted with any one or more of halogen, alkyl, or alkoxy.
  • The compounds disclosed in Formulas Ia, Ib and Ic should be understood as also accommodating methyl, ethyl, methoxy, fluoro, or chloro groups at any position otherwise occupied by a ring hydrogen. Moreover, R3 and R4 may be used in combination to produce a nitro moiety on the phenyl ring, or to create ring systems such as morpholine, quinoline, or isoquinoline.
  • In certain embodiments, a candidate compound is a compound of Formula Ia, pharmaceutically acceptable salts, stereo-isomers, polymorphs, metabolites, prod-drugs and combinations thereof:
  • Figure US20160283652A1-20160929-C00002
  • wherein the NR3R4 moiety is connected ortho to the R1 moiety on the phenyl ring;
  • R1 is selected from the group consisting of nitro, amino optionally substituted with methyl or dimethyl, and amide optionally substituted with methyl, dimethyl, ethyl, or diethyl;
  • R2 is phenonyl optionally substituted with halogen or methoxy;
  • R3 is selected from the group consisting of phenyl optionally substituted with halogen or methoxy and benzyl optionally substituted with halogen or methoxy; and
  • R4 is selected from the group consisting of H, methyl, and phenyl; with the following exception: when R1 is nitro, R4 is H, and R3 is benzyl optionally substituted with fluoro or isopropyl.
  • In other embodiments, a candidate compound is a compound of Formula Ic, pharmaceutically acceptable salts, stereo-isomers, polymorphs, metabolites, prod-drugs and combinations thereof:
  • Figure US20160283652A1-20160929-C00003
  • wherein the NR3R4 moiety is connected ortho to the R1 moiety on the phenyl ring;
  • R1 is selected from the group consisting of nitro, amino optionally substituted with methyl or dimethyl, and amide optionally substituted with methyl, dimethyl, ethyl, or diethyl; the R2 moiety is connected meta with respect to the phenyl ring;
  • R2 is carboxylic acid;
  • R3 is selected from the group consisting of phenyl optionally substituted by any one or more of methoxy or halogen and benzyl optionally substituted by any one or more of methoxy or halogen; and
  • R4 is selected from the group consisting of H and methyl.
  • In additional embodiments, a candidate compound is a compound of Formula II:
  • Figure US20160283652A1-20160929-C00004
  • wherein
  • X is selected from the group consisting of hydrogen, methyl, amine, methoxy, phenyl optionally substituted with up to a total of three methyl and/or methoxy and/or halogen groups, cyclopentane, morpholine, piperidine, N-methylpiperidine, N-ethylpiperidine, (N,N-diethyl)formamide, pyridine, pyrazine, pyrrole, pyrrolidine, furan, thiophene, tetrahydrofuran, pyran, tetrahydroisoquinoline, isoquinoline, quinoline, N-phenylpiperazine optionally substituted with up to a total of three methoxy and/or halogen groups, or N-benzylpiperazine;
  • A is an optional spacer group, attachable in either direction, selected from the group consisting of —NH—, —NHCH2—, —NHCH2CH2—, —NHCH2CH2O—, and —NHCH2(CH3);
  • D is selected from the group consisting of methyl, isopropyl, tert-butyl, dimethylamine, morpholine, alcohol, phenyl optionally substituted with up to a total of three methyl and/or ethyl and/or methoxy and/or halogen and/or acetamide and/or ethoxy and/or cyano groups, pyridine, pyrazine, pyrrole, pyrrolidine, furan, thiophene, tetrahydrofuran, and pyran; and
  • Z is an optional spacer group, selected from the group consisting of —CH2—, —SO2—, —SO2CH2—, —CH2C(═O)—, —CH2CH2—, —C(═O)—, and —C(═S)NHC(═O)—.
  • In other embodiments, A is absent (thus X is directly connected to the phenyl ring at the position held by A); X is tetrahydroisoquinoline, attached to the phenyl ring by its lone nitrogen; Z is —C(═O)—; and D is methyl. In other embodiments, A is absent; X is morpholine, attached to the phenyl ring by its lone nitrogen; Z is —CH2—; and D is methyl.
  • In certain additional embodiments, a candidate compound is a compound of Formula III:
  • Figure US20160283652A1-20160929-C00005
  • wherein
  • X is selected from the group consisting of methyl, methylamine, halogen, and phenyl optionally substituted with up to a total of three methyl and/or methoxy and/or halogen groups;
  • A is an optional spacer group, attachable in either direction, selected from the group consisting of —NH—, —N(CH3)H—, —O—, —OCH3—, —C(═O)NH—, and —NHCH2—; and
  • Z is selected from the group consisting of phenyl optionally substituted with up to a total of three methyl and/or ethyl and/or methoxy and/or halogen and/or acetamide and/or ethoxy and/or cyano groups; excepting those compounds that include X as phenyl and A as —NHCH2—, the nitrogen in said A being connected to the nitro-containing phenyl ring in said formula and the carbon in said A being connected to said X in said formula.
  • In certain embodiments, a candidate compound is selected from the group consisting of:
    • (4-(4-nitro-3-(phenylamino)phenyl)piperazin-1-yl)(phenyl)methanone;
    • 5-(4-dimethylcarbamylpiperazin-1-yl)-2-nitro-N-phenylbenzenamine;
    • N-methyl-5-(4-benzoylpiperazin-1-yl)-2-nitro-N-phenylbenzenamine;
    • N,N-dimethyl-5-(4-ethylpiperazin-1-yl)-2-nitrobenzenamine;
    • N-(2-(4-benzoylpiperazin-1-yl)-5-nitrophenyl)acetamide;
    • 2-(benzylamino)-N,N-dimethyl-4-(4-benzoylpiperazin-1-yl)benzamide;
    • 2-(benzylamino)-N-ethyl-4-(4-benzoylpiperazin-1-yl)benzamide;
    • 3′-(benzylamino)-4′-nitrophenyl-3-carboxylic acid;
    • 3′-(benzylamnino)-4′-nitro-N-phenylbiphenyl-3-carboxamide;
    • ethyl-1-(3-(benzylamino)-4-nitrophenyl) piperidine-4-carboxylate; N-(2-(4-benzoylpiperazin-1-yl)-5-nitrophenyl)benzenamine;
    • (4-(4-amino-3-(phenylamino)phenyl)piperazin-1-yl)(phenyl)methanone;
    • 1-(3-(benzylamino)-4-nitrophenyl) piperidine-3-carboxylic acid;
    • 4′-nitro-3′-(phenylamino) biphenyl-3-carboxylic acid;
    • N,N-dimethyl-2-(4-benzoylpiperazin-1-yl)-5-nitrobenzenamine;
    • 4′-amino-3′-(phenylamino) biphenyl-3-carboxylic acid;
    • (4-(3-(N-benzyl-N-phenylamino)-4-aminophenyl)piperazin-1-yl)(phenyl)methanone;
    • (4-(3-(N-methyl-N-phenylamino)-4-(dimethylamino)phenyl)piperazin-1-yl)(phenyl)methanone;
    • (4-(4-(dimethylamino)-3-(phenylamino)phenyl)piperazin-1-yl)(phenyl)methanone;
    • (4-(3-(N-methyl-N-phenylamino)-4-aminophenyl)piperazin-1-yl)(phenyl)methanone;
    • (4-(3-(N-methyl-N-phenylamino)-4-(methylamino)phenyl)piperazin-1-yl)(phenyl)methanone;
    • (4-(4-(methylamino)-3-(phenylamino)phenyl)piperazin-1-yl)(phenyl)methanone;
    • 2-(4-(4-benzoylpiperazin-1-yl)-2-(phenylamino)phenylamino)ethanol;
    • N-benzyl-2-(4-benzoylpiperazin-1-yl)-5-nitrobenzenamine; N-(4-(4-benzoylpiperazin-1-yl)-2-(phenylamino)phenyl)acetamide;
    • 4-(4-benzoylpiperazin-1-yl)-N1-(2-morpholinoethyl)-N2-phenylbenzene-1,2-diamine,
    • pharmaceutically acceptable salts, stereo-isomers, polymorphs, metabolites, prod-drugs, analogs and combinations thereof.
  • In other embodiments, a candidate compound is selected from the group consisting of:
    • [4-[4-nitro-3-(tricyclo[3.3.1.13,7]dec-2-ylamino)phenyl]-1-piperazinyl]phenylmethanone,
    • 2-(4-benzoyl-1-piperazinyl)-5-nitrobenzonitrile,
    • [4-[3-[(4-methyl-4H-1,2,4-triazol-3-yl)thio]-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-3-(2-propen-1-ylamino)phenyl]-1-piperazinyl]phenylmethanone,
    • [4-[3-[(2-methylpropyl)amino]-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-3-[[(tetrahydro-2-furanyl)methyl]amino]phenyl]-1-piperazinyl]phenylmethanone,
    • [4-[3-[(2,2-dimethylpropyl)amino]-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-[3-(ethylamino)-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-(2-methyl-4-nitrophenyl)-1-piperazinyl]phenylmethanone,
    • 5-[[2-(4-benzoyl-1-piperazinyl)-5-nitrophenyl]methylene]-2,4,6(1H,3H,5H)-pyrimidinetrione,
    • [4-[2-(2,5-dimethyl-1H-pyrrol-1-yl)-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-2-(1H-pyrrol-1-yl)phenyl]-1-piperazinyl]phenylmethanone,
    • 2-[5-(4-benzoyl-1-piperazinyl)-2-nitrophenyl]-4-methyl-1(2H)phthalazinone,
    • [4-[3-(methylamino)-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-3-[(3-pyridinylmethyl)amino]phenyl]-1-piperazinyl]phenylmethanone,
    • [4-[3-(3,4-dihydro-2(1H)-isoquinolinyl)-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-3-(1-piperidinyl)phenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-2-(trifluoromethyl)phenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]phenylmethanone,
    • [4-[3-(4-morpholinyl)-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-3-[(1-tricyclo[3.3.1.13,7]dec-1-ylethyl)amino]phenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]phenylmethanone,
    • [4-[4-nitro-3-[(2-phenylethyl)amino]phenyl]-1-piperazinyl]phenylmethanone,
    • [4-[3-[(2-furanylmethyl)amino]-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-[3-(3,5-dimethyl-1H-pyrazol-1-yl)-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-[3-(cyclopropylamino)-4-nitrophenyl]-1-piperazinyl]phenylmethanone,
    • [4-(2-chloro-4-nitrophenyl)-1-piperazinyl]phenylmethanone,
    • [4-(2-fluoro-4-nitrophenyl)-1-piperazinyl]phenylmethanone,
    • 1-benzoyl-4-(3-formyl-4-nitrophenyl)piperazine,
    • 1-benzoyl-4-[3-[(2,5-dioxo-4-imidazolidinylidene)methyl]-4-nitrophenyl]piperazine,
    • 1-(3-amino-4-nitrophenyl)-4-benzoylpiperazine, 1-benzoyl-4-(4-nitrophenyl)piperazine,
    • (2,4-dichlorophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (5-chloro-2-methoxyphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl](4-propoxyphenyl)methanone,
    • (3,4-dimethoxyphenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3,4-dimethoxyphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-chloro-4-methylphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-methoxy-3-methylphenyl) [4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-methoxy-3-methylphenyl) [4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-(1-methylethoxy)phenyl][4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-(1-methylethoxy)phenyl][4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [3-(1-methylethoxy)phenyl][4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [3-(1-methylethoxy)phenyl][4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-ethylphenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-ethylphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-ethoxyphenyl) [4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-ethoxyphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3,4-dichlorophenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3,4-dichlorophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-(1-methylethyl)phenyl][4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-iodophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-(1,1-dimethylethyl)phenyl][4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-bromophenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]phenylmethanone,
    • (2-bromophenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-butoxyphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-(1-methylethyl)phenyl][4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-methoxyphenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-methylphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-methylphenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-fluorophenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-fluorophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-methoxyphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-bromophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-(1,1-dimethylethyl)phenyl][4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-ethoxyphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]phenylmethanone,
    • (4-fluorophenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-chlorophenyl) [4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-fluorophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-chlorophenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-methylphenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-[3-[[(4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl](2-fluorophenyl)methanone,
    • (2-chlorophenyl)[4-[3-[[4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl]methanone,
    • (4-bromophenyl)[4-[3-[[(4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl]methanone,
    • (2-chloro-4,5-difluorophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-chlorophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-methylphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-bromophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-[3-[[(4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl](4-methylphenyl)methanone,
    • [4-[3-[[[4-(1-methylethyl)phenyl]methyl]amino]-4-nitrophenyl]-1-piperazinyl](4-methylphenyl)methanone,
    • (3,5-dichloro-4-methoxyphenyl)[4-[3-[[[4-(1-methylethyl)phenyl]methyl]amino]-4-nitrophenyl]-1-piperazinyl]methanone,
    • (2-fluorophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-fluorophenyl)[4-[3-[[[4-(1-methylethyl)phenyl]methyl]amino]-4-nitrophenyl]-1-piperazinyl]methanone,
    • (4-chlorophenyl)[4-[3-[[[4-(1-methylethyl)phenyl]methyl]amino]-4-nitrophenyl]-1-piperazinyl]methanone,
    • [4-[3-[[(4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl](3-methylphenyl)methanone,
    • (3-methylphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-chlorophenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-chlorophenyl)[4-[3-[[(4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl]methanone,
    • (2-chloro-4,5-difluorophenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-chloro-4,5-difluorophenyl)[4-[3-[[(4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl]methanone,
    • (4-bromophenyl)[4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (3-chlorophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-methoxyphenyl) [4-[4-nitro-3-[(1-phenylethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-[3-[[(4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl](4-methoxyphenyl)methanone,
    • (4-methoxyphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • [4-[3-[[(4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl](2-methoxyphenyl)methanone,
    • (2-methoxyphenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (4-bromophenyl)[4-[4-nitro-3-[(phenylmethyl)amino]phenyl]-1-piperazinyl]methanone,
    • (2-bromophenyl)[4-[3-[[(4-chlorophenyl)methyl]amino]-4-nitrophenyl]-1-piperazinyl]methanone,
    • pharmaceutically acceptable salts, stereo-isomers, polymorphs, metabolites, prod-drugs, analogs and combinations of any of the foregoing.
  • In certain embodiments, the candidate compound is represented by one of one of the following structural formulas:
  • Figure US20160283652A1-20160929-C00006
    Figure US20160283652A1-20160929-C00007
    Figure US20160283652A1-20160929-C00008
    Figure US20160283652A1-20160929-C00009
    Figure US20160283652A1-20160929-C00010
    Figure US20160283652A1-20160929-C00011
  • pharmaceutically acceptable salts, stereo-isomers, polymorphs, metabolites, pro-drugs, analogs and mixtures of any of the foregoing.
  • In certain embodiments, the candidate compound has activity against both Aβ and A-Syn, as determined by a ThT functional aggregation assay.
  • In certain embodiments, the candidate compound has activity against both Aβ and tau, as determined by a ThT functional aggregation assay.
  • In certain embodiments, the candidate compound only has activity against Aβ, as determined by a ThT functional aggregation assay.
  • In certain embodiments, the IC50/μM for Aβ for the candidate compound is from about 0.5 to about 5734, as determined by ThT functional aggregation assay. In certain embodiments, the IC50/μM for Aβ is from about 0.8 to about 180. In certain embodiments, the IC50/μM for Aβ is from about 0.8 to about 107. In certain embodiments, the IC50/μM for Aβ is 0.85, 0.88, 1.47, 1.9, 2.3, 2.29, 3.5, 7.5, 8.4, 9.64, 15.5, 18, 23, 25, 26.5, 54, 66, 84, 90, 92, and 100.
  • In certain embodiment, the IC50/μM for A-syn for A-Syn, as determined by A-Syn functional ThT aggregation assay, for the candidate compound is from about 1 to about 70. In certain embodiment, the IC50/μM for A-syn is from about 5 to about 50. In certain embodiment, the IC50/μM for A-syn is 4, 5, 10, 15 or 45.
  • In certain embodiments, the IC50/μM for Aβ for the candidate compound is 0.85, 9.64, 25, 1.47, 9.23, 27, 26.6, or 3.5; and the IC50/μM for A-syn is 6.34, 5, 4, 7, 8, 10, 15, or 45.
  • One example of a pseudo-crystal structure utilized in these methods is the one formed by SEQ ID: 1 and SEQ ID: 2 when interacting in the manner depicted in FIG. 2.
  • In certain embodiments, a method of identifying compounds that modulate amyloid aggregation comprises the steps of constructing a pseudo-crystal structure in a computer modeling program; selecting a list of candidate compounds; constructing said candidate compounds in a computer modeling program; performing an iterative docking and scoring of all candidate compounds, by means of docking each candidate compound into the pocket formed by said model and scoring each candidate compound to reflect its degree of complementarity with respect to said pocket; and identifying compounds that modulate amyloid aggregation by reference to a score cutoff that substantially distinguishes active compounds from inactive compounds. A score cutoff is usually an absolute energy, that can be set higher or lower depending on the level of activity that is deemed to separate active from inactive, or as a reference between more active and less active. In some embodiments, the score cutoff is the value of a known active compound. In other embodiments, the score cutoff is the value that distinguishes a small part (i.e. 1% to 10%) of a set of compounds as being substantially more active than other members of the set.
  • In certain embodiments, the pseudo-crystal structure comprises a three-dimensional model of uncrystallized, monomeric amyloid protein and a three-dimensional model of uncrystallized amyloid protein or oligomer comprising one or more amyloid peptides, said amyloid protein or oligomer model to be positioned with respect to said amyloid monomer model such that it forms a pocket in conjunction with said amyloid monomer model, such that a compound may be inserted into said pocket thereby modulating amyloid aggregation.
  • In certain embodiments, the amyloid monomer model and the amyloid protein or oligomer model are both composed of beta-amyloid protein or an amyloidogenic fragment thereof.
  • In other embodiments, said amyloid monomer model is substantially SEQ ID: 1 and said amyloid oligomer model is substantially SEQ ID: 2. In certain embodiments, said amyloid monomer model is further positioned with respect to said amyloid protein model such that the amino acid Val at position 8 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2; the amino acid Gly at position 9 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2; the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Gly at position 9 of SEQ ID: 2; the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Ser at position 10 of SEQ ID:2; and the amino acid Lys at position 12 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2. In certain embodiments, the amyloid monomer model is positioned with respect to the amyloid protein model substantially in the orientation shown in FIG. 2. In certain embodiments, the candidate compounds are drawn from proprietary compound collections and/or from commercially available screening libraries.
  • The process of docking as implemented in the computer modeling program or programs is described, e.g., in the manuals of said program or programs. Docking may be performed, e.g., interactively or in batch mode. Computer modeling programs especially suited to docking include, but are not restricted to, DOCK, AUTODOCK, GLIDE, and FlexX. The manuals of these programs are herein incorporated by reference in their entirety for all purposes.
  • Scoring functions are generally applied after docking and may be integral to a given docking method. Examples of scoring functions include DrugScore, London, and Affinity dG. In some embodiments, the scoring function may be a composite of two or more scoring functions.
  • In certain embodiments, a method of improving potency of a compound known to modulate amyloid aggregation comprising the steps of constructing a pseudo-crystal structure in a computer modeling program; selecting a list of candidate compounds; constructing said candidate compounds in a computer modeling program; performing an iterative docking and scoring of all candidate compounds, by means of docking each candidate compound into the pocket formed by said model and scoring each candidate compound to reflect its degree of complementarity with respect to said pocket; and identifying compounds that modulate amyloid aggregation by reference to a score cutoff that substantially distinguishes more active compounds from less active compounds. In certain embodiments of the invention, the list of candidate compounds includes both the compound known to be active and analogs of said compound, and the score cutoff is that score which belongs to the compound known to be active. In certain embodiments, the pocket comprises substantially SEQ ID: 1 and amyloid oligomer model has substantially SEQ ID: 2, wherein SEQ ID: 1 is
  • Lys Val Phe Phe Ala Glu Asp Val Gly Ser Asn
    1                   6                   11
    Lys Gly Ala Ile Ile Gly Leu Met Val Gly Gly
        Val     16                      21
    Val Ile Ala
            26

    and,
  • SEQ ID: 2 is
  • Lys Val Phe Phe Ala Glu Asp Val Gly Ser Asn
    1                   6                   11
    Lys Gly Ala Ile Ile Gly Leu Met Val Gly Gly
        Val     16                      21
    Val Ile Ala
            26

    In certain, embodiments the two sequences comprising the pocket are not substantially the same.
  • In certain embodiments, the present invention uses known non-peptidic compounds amyloid and/or amyloidogenic fragments to identify new compounds which may be inhibitors or likely to be inhibitors of amyloid protein aggregation. For example, a wide variety of non-peptidic compounds have shown the ability to inhibit the aggregation of amyloid proteins in vitro, and many such compounds can inhibit the aggregation of beta-amyloid protein as well as other kinds of amyloid (see for example Klabunde et al, 2000, Nat. Struct. Biol. 7:312-321; Green et al, 2003, J. Am. Chem. Soc. 125:13404-13414; Masuda et al, 2006, Biochemistry 45:6085-6094; Ono et al, 2003, J. Neurochem 87:172-181; Tagliavini et al, 2000, J. Mol Biol. 300:1309-1322). In addition, a number of amyloid proteins or amyloidogenic fragments thereof have been crystallized, including islet amyloid polypeptide or amylin (Wilizius et al, 2008, Protein Sci. June 12 online); beta-2-microglobulin or light chain (Iwata et al, 2007, J. Biochem. 142:413-419; Schormann et al, 1995, Proc. Natl. Acad. Sci. USA 92:9490-9494), insulin; and notably transthyretin, which has been crystallized several times in co-crystal with known inhibitors (Hamilton et al, 1993, J. Biol. Chem. 268:2416-2424; Peterson et al, 1998, Proc. Natl. Acad. Sci. USA 95:12956-12960; Klabunde et al, 2000, Nat. Struct. Biol. 7:312-321). Some known compounds have also been shown to have beneficial in vivo effects, including reducing the size of amyloid plaques and delaying mortality in mouse models of amyloid disease (Chen et al, 2000, Nat. Med. 6:797-801; Imbimbo et al, 2007, Pharmacol. Res. 55:318-328). It is postulated that compounds that modulate the aggregation of amyloid protein in vitro (so-called “anti-amyloid compounds”) and have in vivo effects may be beneficial for the treatment of amyloid diseases, e.g., Alzheimer's Disease. In certain embodiments, the present invention uses these known compounds to identify new compounds which may be inhibitors or likely to be inhibitors of amyloid protein aggregation. One compound which may be used in the methods of the present invention (e.g., as a multiply anti-amyloid compound) is resveratrol, an antioxidant component of red wine and an inhibitor of beta-amyloid aggregation at an effective concentration of 5.6 μM (Riviere et al, 2007, Bioorg. Med. Chem. 15:1160-1167).
  • In certain embodiments, the invention is directed to methods of improving potency of a compound known to modulate amyloid aggregation comprising the steps of constructing a pseudo-crystal structure in a computer modeling program; selecting a list of candidate compounds; constructing said candidate compounds in a computer modeling program; performing an iterative docking and scoring of all candidate compounds, by means of docking each candidate compound into the pocket formed by said model and scoring each candidate compound to reflect its degree of complementarity with respect to said pocket; and identifying compounds that modulate amyloid aggregation by reference to a score cutoff that substantially distinguishes more active compounds from less active compounds. In certain embodiments, the pseudo-crystal structure for this method is the one formed by SEQ ID: 1 and SEQ ID: 2 when interacting in the manner depicted in FIG. 2. The list of candidate compounds may include, e.g., both the compound known to be active anti-amyloid compounds and analogs of such compounds, and the score cutoff is that score which belongs to the compound known to be active. In certain embodiments, the known compound is a compound selected from the lists recited in paragraphs [0106] to [0117] of the application. In certain embodiments, the known compound is resveratrol.
  • The invention also encompasses and provides for use of the compounds identified or improved using the invention's methods as therapies for diseases in which amyloidosis occurs. The disease include (but are not limited to) Alzheimer's disease, Huntington's disease, Parkinson's disease, Creutzfeldt-Jacob disease, amyotrophic lateral sclerosis, and senile systemic amyloidosis. In certain embodiments, the disease is Alzheimer's disease. In certain embodiments, the therapeutic compound inhibits aggregation of one or more of the following amyloidogenic proteins: beta-amyloid protein, tau protein, alpha-synuclein protein, huntingtin protein, prion precursor protein; other amyloidogenic protein not named herein, and combinations of any of the foregoing.
  • In certain preferred embodiments, both the aggregation of alpha-synuclein protein and of tau protein are inhibited. In certain embodiments, both the aggregation of beta-amyloid protein and of tau protein are inhibited. In certain embodiments, both the aggregation of beta-amyloid protein and of alpha-synuclein protein are inhibited. In certain embodiments, only the aggregation of tau protein, beta-amyloid protein, or alpha-synuclein protein is inhibited.
  • In certain embodiments, the aggregation of beta-amyloid protein, tau protein, and alpha-synuclein protein are all inhibited. In certain embodiments, huntingtin aggregation is inhibited. In certain embodiments, prion protein aggregation is inhibited.
  • In certain embodiments, the methods comprise administering to a subject a therapeutic compound identified or improved using the invention's methods (including analogs and derivatives of such compounds) and/or pharmaceutically acceptable salts thereof, such that the therapeutic compound inhibits the aggregation of an amyloidogenic protein. The subject in these methods may be, e.g., a vertebrate, a mammal, a human, or a non-human animal. The administration may be, e.g., oral, parenteral, transdermal, intrathecal, or intranasal.
  • In certain embodiments, the methods comprise using known amyloid and/or amyloidogenic fragments to discover and/or optimize compounds that modulate their respective amyloid protein, and other amyloid proteins, e.g., due to the purported relative universality of amyloid pathogenesis. (Kayed et al, 2003, Science 300:486-489; Bucciantini et al, 2002, Nature 416:507-511). For example, transthyretin is known to bind beta-amyloid protein in vivo, probably as aggregated beta-amyloid (Tsuzuki et al, 2000, Neurosci. Lett. 281:171-174; Liu and Murphy, 2006, Biochemistry 45:15702-15709), and several inhibitors of transthyretin are also known to inhibit beta-amyloid protein. However, as such compounds do not inhibit both transthyretin and beta-amyloid protein with equal potency, the utility of transthyretin crystal structures per se for discovering inhibitors of beta-amyloid protein does not approach what might be expected if a similar crystal structure of beta-amyloid were available. The methods of present invention in certain embodiments provide for the utilization of these compounds (despite the lack of known crystal structure of beta-amyloid) to identify new compounds which may be inhibitors or likely to be inhibitors of simultaneous inhibition of several amyloid proteins.
  • In other embodiments, the invention relates to methods of virtually screening of compound library, e.g., in silico screening. More specifically, the invention relates to methods of virtually screening a library or libraries to identify compounds that are likely to inhibit amyloid protein aggregation. Such compounds would be of use as therapies for neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, prion diseases, and systemic amyloidosis.
  • The utility and operation of the embodiments of the invention can be further appreciated by reference to the following non-limiting examples:
  • Example 1 Construction of a Common Conformational Motif Model and Subsequent Extraction of a Model of Amyloid Protein Aggregation
  • A Common Conformational Motif model was constructed using a computer modeling program in interactive session and validated.
  • First, resveratrol was drawn and minimized to a root-mean-square gradient of 0.05 using the MMFF94 force field with corresponding partial charges. The PDB structure 1BM7 (www.pdb.org) of transthyretin bound to flufenamic acid was loaded, and resveratrol was manually placed in a position coincident with one of the two flufenamic acid binding sites. The binding site of the other flufenamic acid (i.e. the other homodimer), the flufenamic acid moiety in the resveratrol-occupied binding site, and any solvent molecules were deleted. The 17-42 residue sequence of beta-amyloid protein was then drawn and duplicated, and the two strands of beta-amyloid protein were placed in close, non-overlapping proximity to each other and to resveratrol. The entire structure was then correctly protonated/deprotonated for physiological pH and minimized to a root-mean-square gradient of 0.05 using the MMFF94 force field with corresponding partial charges. The orientation of one strand of beta-amyloid protein with respect to the other was a matter of routine experimentation, as the method furnished both a means of adjusting the structure such that a pocket is formed and a means of validating that the candidate model was useful. Several dozen candidate models were created and extracted. One model was validated as the Common Conformational Motif model and is depicted in FIG. 1.
  • The model of amyloid protein aggregation was then extracted. The extracted model of amyloid protein aggregation is depicted in FIG. 2. FIG. 2 shows two Aβ17-42 monomers bound to each other near the N terminals in a “double candycane” structure. Each monomer is folded in a loop from residues 23 through 33. The loops are stabilized by intra-loop cationic-anionic interactions between Asp23 and Lys28, and they also show an inter-loop attraction through these same residues. Short-run (up to 100 ns) molecular dynamics calculations on this model indicate that the pocket is stable.
  • When the model of amyloid protein aggregation depicted in FIG. 2 was compared to the structure of transthyretin (“TTR”) depicted in FIG. 1, a number of similarities were observed. The model of amyloid protein aggregation depicted in FIG. 2 provided a similar expanse of hydrophobic contact by lining up along two flattened sides of what would otherwise be a much smaller area of β-sheet, rather than forming a pocket across the top of several β-sheets. Anti-amyloid compounds in the amyloid protein aggregation model of FIG. 2 were stabilized in the pocket by Lys and Ser and by exterior Leu, just as in the structure of TTR. Similarly, the positive charge of Lys was shared between the substrate and a nearby Asp in both structures.
  • It was therefore postulated that a small negatively charged compound might be able to fit between both Lys residues and benefit from the salt bridge interaction, as was reported in TTR. It was further postulated that model of amyloid protein aggregation depicted in FIG. 2 may be a transient species on the way to a stacked dimer using a “dock-lock” mechanism. It is known that many anti-amyloid compounds are multiply anti-amyloid. As such, it was postulated that the utility of the models of the present invention extends beyond the prevention of beta-amyloid protein aggregation and includes additional species of the amyloid proteins, therefore we hypothesized that it can be considered a common conformational motif, of utility for other amyloids beyond beta-amyloid protein.
  • Example 2 Validation of the CCM Model of Amyloid Protein Aggregation of Example 1
  • A burgeoning number of compounds have been shown to be anti-amyloidogenic and/or anti-fibrillogenic. To validate the model of amyloid protein aggregation constructed in Example 1, and in view of its potential extension to other amyloid proteins like α-synuclein, a set of these compounds was evaluated in an in silico docking screen using a collection of 79 compounds for which IC50s have been computed for AD, tau, and α-synuclein inhibition. This diverse set comprised polyphenols, benzothiazoles, phenothizaines, macrolides, prophyrins, steroids, derivatives of Congo red, and other anti-amyloid molecules of note.
  • Among the 39 polyphenols evaluated in the screen, the best docking scores were predicted for purpurogallin, epigallocatechin, catechin gallate, hinokiflavone, myricetin, and gallocatechin gallate—all of which have IC50s below 7 μM. The worst scores in the test were predicted for rutin (expt IC 50 32 μM) and chlorogenic acid (>40 μM). Since each compound was represented by several poses, computing a ranking for the remaining compounds was somewhat ambiguous. Nevertheless, the preliminary result was encouraging enough to consider a more stringent criterion: the quantitative structure-activity relationship (QSAR). The Masuda test set was subjected to a CoMFA (comparative molecular field analysis) in order to demonstrate the validity of the model of amyloid protein aggregation depicted in FIG. 2.
  • CoMFA is a 3-D QSAR technique generally used to posit a “pseudoreceptor” when none is known. Because the algorithm is highly sensitive to alignment of compounds with respect to each other, it is often difficult to produce statistically significant results, even with structurally similar compounds. However, when statistically significant results are obtained, this can be a great confirmation of a pharmacophore hypothesis.
  • Using the three best poses from the docking calculation as the initial test set (3×79=237 total poses), a partial least squares analysis of the fit indicated a cross-validated r2 (i.e. q2) of 0.248. Removing 25 outlying poses improved the leave-one-out q2 to 0.616. A more laborious bootstrapping calculation produced a q2 of 0.619. This result indicated that the CoMFA pseudoreceptor had a good agreement with experiment when the molecules are aligned in accordance with the model of amyloid protein aggregation depicted in FIG. 2. Only eight of the 79 compounds were predicted as false positives:
  • Predicted Expt
    Polyphenols
    Apigenin 1.4 >40
    Baicalein 7.8 4.5
    Catechin 10.6 >40
    Catechin gallate 6.2 5.0
    chlorogenic acid >40 >40
    curcumin 2.0 1.7
    cyanidin 4.8 4.0
    daidzein 2.8 >40
    delphinidin 4.4 3.0
    2,2′-dihydroxybenzophenone >40 >40
    4,4′-dihydroxybenzophenone 1.6 >40
    dopamine chloride 6.7 28.6
    epicatechin 16.1 >40
    epicatechin 3-gallate 2.3 3.0
    epigallocatechin 8.7 7.0
    epigallocatechin gallate 2.5 2.0
    exifone 2.2 0.7
    gallocatechin 12.4 7.0
    gallocatechin gallate 2.1 1.5
    gingerol 19.8 25.0
    gossypetin 1.2 1.3
    hinokiflavone 5.8 5.0
    hypericin 1.5 0.9
    kaempferol 8.3 8.0
    luteolin 3.6 3.0
    myricetin 0.9 0.9
    naringenin 3.2 25.0
    2,3,4,2′,4′-pentahydroxybenzophenone 2.2 2.8
    procyanidin B1 21.6 14.0
    procyanidin B2 >40 >40
    purpurogallin 3.2 0.5
    quercetin 4.3 5.0
    rosmarinic acid 8.7 12.0
    rutin >40 32.0
    taxifolin 1.0 >40
    2,2′,4,4′-tetrahydroxybenzophenone 3.2 >40
    theaflavone 2.8 2.0
    tocopherol >40 >40
    2,3,4-trihydroxybenzophenone 6.4 3.1
    anthracycline
    daunorubicin hydrochloride 1.3 1.4
    benzothiazoles
    2-(4-aminophenyl)-6-methylbenzothiazole 2.4 2.0
    basic blue 41 1.2 1.4
    2-[4-(dimethylamino)phenyl]-6- 2.0 2.0
    methylbenzothiazole
    3,3′-dipropyl thiodicarbocyanine iodine 0.3 0.3
    lignans
    magnelol >40 >40
    sesamin 19.2 >40
    phenothiazines
    acetopromazine maleate salt >40 >40
    azure A 0.5 0.4
    azure C 0.5 0.2
    chlorpromazine hydrochloride >40 >40
    lacmoid 3.8 1.4
    methylene blue 20.0 2.3
    perphenazine >40 >40
    promazine hydrochloride >40 >40
    propionylpromazine >40 >40
    hydrochloride
    quinacrine 8.9 8.4
    quinacrine mustard 1.8 1.2
    polyene macrolides
    amphotericin B 2.0 2.2
    filipin III 14.7 14.6
    porphyrins
    ferric dehydroporphyrin IX 0.2 0.2
    hematin 0.2 0.2
    phthalocyanine tetrasulfonate 3.9 3.2
    rifamycin
    rifampicin 4.0 4.9
    steroids
    taurochenodeoxycholic acid >40 >40
    taurohydroxycholic acid >40 >40
    taurolithocholic acid >40 >40
    taurolithocholic acid 3-sulfate >40 >40
    tauroursodeoxycholic acid >40 >40
    Congo red and derivatives
    Congo red 1.0 0.9
    chlorazol black E 0.2 0.3
    BSB 5.5 6.4
    FSB 3.0 1.9
    Ponceau SS 1.2 1.2
    terpenoids
    asiatic acid >40 >40
    ginkgolide A >40 >40
    ginkgolide B 14.4 11.0
    ginkgolide C >40 >40
    others
    4,5-dianilinophthalimide 5.4 2.9
    methyl yellow 2.2 1.5
  • It was therefore concluded that the model of amyloid protein aggregation of FIG. 2 has utility in predicting the anti-amyloid effects of candidate compounds tested in the example, and that these compounds have activity against aggregation of β amyloid protein. It was also postulated that, given conformational similarities between certain amyloid proteins, the candidate compounds may inhibit aggregation of other amyloid proteins, and that dimers of beta-amyloid protein in general may have significant utility in predicting the anti-amyloid effects of candidate compounds on other amyloids such as tau, alpha-synuclein, huntingtin, and/or prion protein.
  • Example 3 Identification of Compounds that Modulate Amyloid Protein Aggregation and Improvement of Potency Using the Common Conformational Motif Model
  • The Common Conformational Motif model created in Example 1 was used to demonstrate its utility for rapid lead discovery.
  • A simple geometric query against a diverse collection of around 750,000 drug-like compounds was constructed and yielded 1,260 compounds with non-hydrogenic atoms lying within 0.1 Å of each point in the 3-point pharmacophore. Searching in this manner allowed for a quick (<90 min) pre-screening prior to the more computationally laborious docking procedure. Using the parameters for docking as described above, each of the 1,260 compounds were fit into the CCM model (i.e., depicted in FIG. 2).
  • The top 125 compounds were examined for commercial availability, and six of these compounds, evenly distributed with respect to docking score, were acquired from Hit2Lead for in vitro testing. Three of these compounds were inactive in the ThT assay, one was weakly active, and two were highly active (see U.S. Provisional Application Ser. No. 61/092,845 for initial compound disclosures and subsequent use of the model for improving.
  • One-half of compounds (50%) initially tested had some activity, and one-third (33.3%) had significant activity, as compared to the typical hit rate of a high-throughput screen (generally no more than 0.5%) with traditional screening techniques. These results therefore confirmed that the screening technique utilized in this example is highly hit-enriched screening technique and may be more effective, as compared to traditional screening techniques.
  • The present example therefore confirms, e.g., that the model created in Example 1 has utility for rapid lead discovery.
  • It was then postulated that, e.g., because a number of the compounds described in the application were shown to be multiply anti-amyloid compounds, that pseudo-crystal structures described in the application may be useful for identification and optimization of anti-tau, anti-alpha-synuclein, and other anti-amyloid applications, in addition to being useful in rapid lead discovery of compounds that may prevent aggregation of beta-amyloid protein.
  • Example 4 Identification of Known Compounds that Modulate Amyloid Protein Aggregation Using the Model of FIG. 2
  • Further validation of the CCM model of protein aggregation (i.e., the model of FIG. 2) was performed using a screen of the PubChem collection (http://www.ncbi.nlm.nih.gov) using a blank structure query and a limit query of “Pharmacological Action.” The query returned 14,172 compounds, consisting entirely of molecules known to have a pharmacological effect.
  • This set of 14,172 compounds was then downloaded in MDL SDfile format, imported into a molecular modeling program, stripped of counterions and assigned physiologically appropriate protonation states, and minimized using a molecular mechanics force field.
  • Next, an unguided docking study was performed, using docking parameters and binding site identification identical to that above, on the PubChem set. The results for the top 25, in decreasing order of binding score and repeated hits in parenthesis, were as follows: chlorophyllin (2), epigallocatechin gallate (3), coumermycin A1, chlorotetracycline (2), doxycycline (2), hygromycin B, rolitetracycline, desfarrioxamine, epicatechin gallate, transcycline, oligoadenylade, 8-azido-ADP, citrinin, tetrakis(4-N-methylpyridyl)porphyrin, tetracycline, acteoside, actinonin, lactitol, minocycline, and rocephalin.
  • What was striking in these results was that they included a number of known anti-amyloid compounds and compound families. Specifically, porphyrins, polyphenols, and tetracyclines constituted fully 60% of the top scores, which is significantly better than most high-throughput screening campaigns. The list also included known anti-amyloid compounds not considered part of any family, including desferrioxamine and the neuroprotectant acteoside.
  • It was therefore postulated that most of the remaining top twenty five compounds were also likely to have effects against Aβ aggregation, especially given their highly symmetrical structures (coumermycin, hygromycin, oligoadenylate), structural affinity with NSAIDs (citrinin), and/or reported utility in related protein misfolding/aggregation diseases such as Huntington's disease (actinonin) and ALS (rocephilin).
  • Moreover, some compounds which were known for uses other than anti-amyloid have been identified as being anti-amyloid by the screen. It was therefore postulated that these compounds may have utility in treating Alzheimer's Disease and related disorders (e.g., Parkinson's Disease). For example, exploration of hits in the top 1% included those mentioned above as well as the following, in no particular order of activity: Pradimicin, Oleuropein, Tunicamycin, Hyaluronic acid, Coumermycin, Ouabain, Amikacin, Rifaximin, delavirdine, almitrine, and fluvastatin, lumiracoxib, and fenoterol. It was therefore postulated that these compounds may have utility in treating amyloid diseases.
  • In this regard, it is notable that even compounds that do not ordinarily cross the blood-brain barrier are often capable of crossing it in patients with neurologic disorders, and that neurologic disorders of the peripheral nervous system also can benefit from anti-amyloid compounds. As such, it was postulated that the physiochemical parameters of an identified anti-amyloid molecule may not need to fit into Log P, molecular weight, and other ranges associated with blood-brain barrier penetration in order to have utility in treatment of neurologic disorders (e.g., Alzheimer's Disease and/or related disorders).
  • Data Listing
  • The model of FIG. 2 in PDB format.
  • This listing can be transcribed into a text file (Unicode, ASCII, or other standard encoding), saved to disc as with extension .PDB, and loaded into a PDB-aware visualization or modeling program in order to reconstruct the model.
  • HEADER PROTEIN FIBRIL
    REMARK 99 MOE v2007.09 (Chemical Computing Group Inc) Mon Jun 16 15:27:17 2008
    ATOM 1 N LEU E 17 −4.245 −9.375 10.249 1.00 0.00 N1+
    ATOM 2 H1 LEU E 17 −4.274 −8.787 9.368 0.00 0.00 H
    ATOM 3 H2 LEU E 17 −4.396 −8.764 11.070 0.00 0.00 H
    ATOM 4 CA LEU E 17 −5.267 −10.479 10.168 1.00 0.00 C
    ATOM 5 C LEU E 17 −4.618 −11.861 10.006 1.00 0.00 C
    ATOM 6 O LEU E 17 −3.393 −11.958 9.924 1.00 0.00 O
    ATOM 7 CB LEU E 17 −6.281 −10.169 9.008 1.00 0.00 C
    ATOM 8 CG LEU E 17 −7.425 −9.144 9.299 1.00 0.00 C
    ATOM 9 CD1 LEU E 17 −6.943 −7.714 9.627 1.00 0.00 C
    ATOM 10 CD2 LEU E 17 −8.432 −9.109 8.124 1.00 0.00 C
    ATOM 11 H3 LEU E 17 −3.272 −9.775 10.284 1.00 0.00 H
    ATOM 12 HA LEU E 17 −5.791 −10.502 11.115 1.00 0.00 H
    ATOM 13 HB2 LEU E 17 −5.722 −9.849 8.101 1.00 0.00 H
    ATOM 14 HB3 LEU E 17 −6.795 −11.117 8.729 1.00 0.00 H
    ATOM 15 HG LEU E 17 −7.992 −9.514 10.187 1.00 0.00 H
    ATOM 16 HD11 LEU E 17 −7.811 −7.028 9.744 1.00 0.00 H
    ATOM 17 HD12 LEU E 17 −6.372 −7.687 10.576 1.00 0.00 H
    ATOM 18 HD13 LEU E 17 −6.304 −7.337 8.802 1.00 0.00 H
    ATOM 19 HD21 LEU E 17 −9.283 −8.433 8.356 1.00 0.00 H
    ATOM 20 HD22 LEU E 17 −7.942 −8.747 7.195 1.00 0.00 H
    ATOM 21 HD23 LEU E 17 −8.844 −10.122 7.930 1.00 0.00 H
    ATOM 22 N VAL E 18 −5.424 −12.964 9.972 1.00 0.00 N
    ATOM 23 CA VAL E 18 −4.953 −14.332 9.721 1.00 0.00 C
    ATOM 24 C VAL E 18 −4.942 −14.537 8.195 1.00 0.00 C
    ATOM 25 O VAL E 18 −5.993 −14.639 7.554 1.00 0.00 O
    ATOM 26 CB VAL E 18 −5.740 −15.453 10.461 1.00 0.00 C
    ATOM 27 CG1 VAL E 18 −5.106 −16.852 10.206 1.00 0.00 C
    ATOM 28 CG2 VAL E 18 −5.825 −15.184 11.990 1.00 0.00 C
    ATOM 29 HN VAL E 18 −6.412 −12.896 10.080 1.00 0.00 H
    ATOM 30 HA VAL E 18 −3.933 −14.389 10.082 1.00 0.00 H
    ATOM 31 HB VAL E 18 −6.781 −15.475 10.063 1.00 0.00 H
    ATOM 32 HG11 VAL E 18 −5.679 −17.636 10.745 1.00 0.00 H
    ATOM 33 HG12 VAL E 18 −5.126 −17.113 9.127 1.00 0.00 H
    ATOM 34 HG13 VAL E 18 −4.054 −16.887 10.561 1.00 0.00 H
    ATOM 35 HG21 VAL E 18 −6.395 −15.996 12.491 1.00 0.00 H
    ATOM 36 HG22 VAL E 18 −4.811 −15.143 12.443 1.00 0.00 H
    ATOM 37 HG23 VAL E 18 −6.346 −14.228 12.203 1.00 0.00 H
    ATOM 38 N PHE E 19 −3.722 −14.584 7.596 1.00 0.00 N
    ATOM 39 CA PHE E 19 −3.476 −14.796 6.177 1.00 0.00 C
    ATOM 40 C PHE E 19 −3.187 −16.283 5.942 1.00 0.00 C
    ATOM 41 O PHE E 19 −2.095 −16.776 6.243 1.00 0.00 O
    ATOM 42 CB PHE E 19 −2.318 −13.880 5.682 1.00 0.00 C
    ATOM 43 CG PHE E 19 −2.186 −13.850 4.177 1.00 0.00 C
    ATOM 44 CD1 PHE E 19 −2.922 −12.924 3.413 1.00 0.00 C
    ATOM 45 CD2 PHE E 19 −1.293 −14.712 3.515 1.00 0.00 C
    ATOM 46 CE1 PHE E 19 −2.769 −12.862 2.022 1.00 0.00 C
    ATOM 47 CE2 PHE E 19 −1.130 −14.644 2.126 1.00 0.00 C
    ATOM 48 CZ PHE E 19 −1.871 −13.721 1.379 1.00 0.00 C
    ATOM 49 HN PHE E 19 −2.899 −14.472 8.145 1.00 0.00 H
    ATOM 50 HA PHE E 19 −4.374 −14.519 5.636 1.00 0.00 H
    ATOM 51 HB2 PHE E 19 −2.509 −12.835 6.008 1.00 0.00 H
    ATOM 52 HB3 PHE E 19 −1.350 −14.202 6.121 1.00 0.00 H
    ATOM 53 HD1 PHE E 19 −3.604 −12.243 3.901 1.00 0.00 H
    ATOM 54 HD2 PHE E 19 −0.719 −15.434 4.080 1.00 0.00 H
    ATOM 55 HE1 PHE E 19 −3.337 −12.145 1.447 1.00 0.00 H
    ATOM 56 HE2 PHE E 19 −0.430 −15.302 1.632 1.00 0.00 H
    ATOM 57 HZ PHE E 19 −1.746 −13.671 0.307 1.00 0.00 H
    ATOM 58 N PHE E 20 −4.188 −17.030 5.405 1.00 0.00 N
    ATOM 59 CA PHE E 20 −4.165 −18.482 5.235 1.00 0.00 C
    ATOM 60 C PHE E 20 −3.313 −18.915 4.027 1.00 0.00 C
    ATOM 61 O PHE E 20 −3.832 −19.325 2.985 1.00 0.00 O
    ATOM 62 CB PHE E 20 −5.608 −19.088 5.165 1.00 0.00 C
    ATOM 63 CG PHE E 20 −6.384 −18.903 6.453 1.00 0.00 C
    ATOM 64 CD1 PHE E 20 −7.240 −17.798 6.628 1.00 0.00 C
    ATOM 65 CD2 PHE E 20 −6.277 −19.845 7.494 1.00 0.00 C
    ATOM 66 CE1 PHE E 20 −7.970 −17.638 7.813 1.00 0.00 C
    ATOM 67 CE2 PHE E 20 −7.010 −19.689 8.678 1.00 0.00 C
    ATOM 68 CZ PHE E 20 −7.858 −18.586 8.836 1.00 0.00 C
    ATOM 69 HN PHE E 20 −5.048 −16.597 5.154 1.00 0.00 H
    ATOM 70 HA PHE E 20 −3.690 −18.900 6.114 1.00 0.00 H
    ATOM 71 HB2 PHE E 20 −6.180 −18.626 4.331 1.00 0.00 H
    ATOM 72 HB3 PHE E 20 −5.547 −20.180 4.967 1.00 0.00 H
    ATOM 73 HD1 PHE E 20 −7.341 −17.060 5.846 1.00 0.00 H
    ATOM 74 HD2 PHE E 20 −5.629 −20.702 7.382 1.00 0.00 H
    ATOM 75 HE1 PHE E 20 −8.617 −16.782 7.935 1.00 0.00 H
    ATOM 76 HE2 PHE E 20 −6.922 −20.421 9.468 1.00 0.00 H
    ATOM 77 HZ PHE E 20 −8.422 −18.464 9.750 1.00 0.00 H
    ATOM 78 N ALA E 21 −1.962 −18.826 4.174 1.00 0.00 N
    ATOM 79 CA ALA E 21 −0.961 −19.299 3.227 1.00 0.00 C
    ATOM 80 C ALA E 21 0.265 −19.892 3.935 1.00 0.00 C
    ATOM 81 O ALA E 21 0.658 −19.456 5.022 1.00 0.00 O
    ATOM 82 CB ALA E 21 −0.506 −18.178 2.263 1.00 0.00 C
    ATOM 83 HN ALA E 21 −1.593 −18.392 4.999 1.00 0.00 H
    ATOM 84 HA ALA E 21 −1.420 −20.086 2.641 1.00 0.00 H
    ATOM 85 HB1 ALA E 21 0.004 −17.364 2.821 1.00 0.00 H
    ATOM 86 HB2 ALA E 21 0.203 −18.574 1.505 1.00 0.00 H
    ATOM 87 HB3 ALA E 21 −1.381 −17.749 1.731 1.00 0.00 H
    ATOM 88 N GLU E 22 0.889 −20.915 3.294 1.00 0.00 N
    ATOM 89 CA GLU E 22 2.127 −21.571 3.695 1.00 0.00 C
    ATOM 90 C GLU E 22 2.991 −21.853 2.456 1.00 0.00 C
    ATOM 91 O GLU E 22 2.485 −22.046 1.343 1.00 0.00 O
    ATOM 92 CB GLU E 22 1.869 −22.866 4.533 1.00 0.00 C
    ATOM 93 CG GLU E 22 3.110 −23.651 5.058 1.00 0.00 C
    ATOM 94 CD GLU E 22 4.098 −22.807 5.870 1.00 0.00 C
    ATOM 95 OE1 GLU E 22 4.107 −22.938 7.119 1.00 0.00 O1−
    ATOM 96 OE2 GLU E 22 4.857 −22.025 5.232 1.00 0.00 O
    ATOM 97 HN GLU E 22 0.553 −21.237 2.412 1.00 0.00 H
    ATOM 98 HA GLU E 22 2.673 −20.873 4.318 1.00 0.00 H
    ATOM 99 HB2 GLU E 22 1.263 −22.574 5.421 1.00 0.00 H
    ATOM 100 HB3 GLU E 22 1.253 −23.567 3.929 1.00 0.00 H
    ATOM 101 HG2 GLU E 22 2.758 −24.477 5.711 1.00 0.00 H
    ATOM 102 HG3 GLU E 22 3.668 −24.105 4.214 1.00 0.00 H
    ATOM 103 N ASP E 23 4.330 −21.871 2.667 1.00 0.00 N
    ATOM 104 CA ASP E 23 5.370 −22.061 1.567 1.00 0.00 C
    ATOM 105 C ASP E 23 6.435 −22.990 2.332 1.00 0.00 C
    ATOM 106 O ASP E 23 6.210 −23.825 1.448 1.00 0.00 O
    ATOM 107 CB ASP E 23 6.040 −20.683 1.370 1.00 0.00 C
    ATOM 108 CG ASP E 23 6.797 −20.649 0.046 1.00 0.00 C
    ATOM 109 OD1 ASP E 23 7.643 −21.546 −0.204 1.00 0.00 O1−
    ATOM 110 OD2 ASP E 23 6.537 −19.695 −0.736 1.00 0.00 O
    ATOM 111 HN ASP E 23 4.638 −21.817 3.645 1.00 0.00 H
    ATOM 112 HA ASP E 23 4.879 −22.397 0.748 1.00 0.00 H
    ATOM 113 HB2 ASP E 23 5.239 −19.918 1.301 1.00 0.00 H
    ATOM 114 HB3 ASP E 23 6.728 −20.384 2.185 1.00 0.00 H
    ATOM 115 N VAL E 24 7.231 −24.037 2.574 1.00 0.00 N
    ATOM 116 CA VAL E 24 8.168 −24.125 3.703 1.00 0.00 C
    ATOM 117 C VAL E 24 9.537 −24.715 3.298 1.00 0.00 C
    ATOM 118 O VAL E 24 10.575 −24.135 3.628 1.00 0.00 O
    ATOM 119 CB VAL E 24 7.544 −24.791 4.968 1.00 0.00 C
    ATOM 120 CG1 VAL E 24 8.427 −24.572 6.227 1.00 0.00 C
    ATOM 121 CG2 VAL E 24 7.165 −26.285 4.774 1.00 0.00 C
    ATOM 122 HN VAL E 24 7.136 −24.802 1.922 1.00 0.00 H
    ATOM 123 HA VAL E 24 8.382 −23.101 3.985 1.00 0.00 H
    ATOM 124 HB VAL E 24 6.587 −24.245 5.166 1.00 0.00 H
    ATOM 125 HG11 VAL E 24 7.907 −24.960 7.128 1.00 0.00 H
    ATOM 126 HG12 VAL E 24 8.621 −23.490 6.387 1.00 0.00 H
    ATOM 127 HG13 VAL E 24 9.400 −25.099 6.131 1.00 0.00 H
    ATOM 128 HG21 VAL E 24 6.624 −26.658 5.670 1.00 0.00 H
    ATOM 129 HG22 VAL E 24 6.494 −26.404 3.898 1.00 0.00 H
    ATOM 130 HG23 VAL E 24 8.068 −26.914 4.628 1.00 0.00 H
    ATOM 131 N GLY E 25 9.571 −25.782 2.570 1.00 0.00 N
    ATOM 132 CA GLY E 25 10.728 −26.711 2.245 1.00 0.00 C
    ATOM 133 C GLY E 25 11.732 −26.887 3.369 1.00 0.00 C
    ATOM 134 O GLY E 25 11.461 −27.541 4.381 1.00 0.00 O
    ATOM 135 HN GLY E 25 8.703 −26.152 2.117 1.00 0.00 H
    ATOM 136 HA2 GLY E 25 10.344 −27.695 2.018 1.00 0.00 H
    ATOM 137 HA3 GLY E 25 11.215 −26.256 1.394 1.00 0.00 H
    ATOM 138 N SER E 26 12.939 −26.294 3.198 1.00 0.00 N
    ATOM 139 CA SER E 26 13.972 −26.116 4.226 1.00 0.00 C
    ATOM 140 C SER E 26 14.983 −25.119 3.655 1.00 0.00 C
    ATOM 141 O SER E 26 16.115 −25.473 3.305 1.00 0.00 O
    ATOM 142 CB SER E 26 14.553 −27.454 4.655 1.00 0.00 C
    ATOM 143 OG SER E 26 13.746 −28.270 5.393 1.00 0.00 O
    ATOM 144 HN SER E 26 13.143 −25.862 2.316 1.00 0.00 H
    ATOM 145 HA SER E 26 13.512 −25.665 5.095 1.00 0.00 H
    ATOM 146 HB2 SER E 26 14.987 −28.007 3.751 1.00 0.00 H
    ATOM 147 HB3 SER E 26 15.537 −27.247 5.297 1.00 0.00 H
    ATOM 148 HG SER E 26 12.849 −28.037 5.051 1.00 0.00 H
    ATOM 149 N ASN E 27 14.542 −23.833 3.507 1.00 0.00 N
    ATOM 150 CA ASN E 27 15.010 −22.894 2.483 1.00 0.00 C
    ATOM 151 C ASN E 27 14.612 −23.372 1.059 1.00 0.00 C
    ATOM 152 O ASN E 27 14.216 −24.523 0.833 1.00 0.00 O
    ATOM 153 CB ASN E 27 16.529 −22.500 2.633 1.00 0.00 C
    ATOM 154 CH ASN E 27 16.990 −21.234 1.884 1.00 0.00 C
    ATOM 155 OD1 ASN E 27 16.265 −20.531 1.173 1.00 0.00 O
    ATOM 156 ND2 ASN E 27 18.302 −20.924 2.051 1.00 0.00 N
    ATOM 157 HN ASN E 27 13.700 −23.565 3.970 1.00 0.00 H
    ATOM 158 HA ASN E 27 14.436 −21.994 2.667 1.00 0.00 H
    ATOM 159 HB2 ASN E 27 16.730 −22.330 3.713 1.00 0.00 H
    ATOM 160 HB3 ASN E 27 17.156 −23.355 2.302 1.00 0.00 H
    ATOM 161 HD21 ASN E 27 18.883 −21.499 2.621 1.00 0.00 H
    ATOM 162 HD22 ASN E 27 18.654 −20.116 1.585 1.00 0.00 H
    ATOM 163 N LYS E 28 14.695 −22.445 0.075 1.00 0.00 N
    ATOM 164 CA LYS E 28 14.381 −22.629 −1.341 1.00 0.00 C
    ATOM 165 C LYS E 28 15.351 −21.770 −2.154 1.00 0.00 C
    ATOM 166 O LYS E 28 16.222 −22.307 −2.839 1.00 0.00 O
    ATOM 167 CB LYS E 28 12.877 −22.355 −1.687 1.00 0.00 C
    ATOM 168 CG LYS E 28 11.905 −23.328 −0.984 1.00 0.00 C
    ATOM 169 CD LYS E 28 10.407 −23.125 −1.274 1.00 0.00 C
    ATOM 170 CE LYS E 28 9.565 −24.127 −0.470 1.00 0.00 C
    ATOM 171 N2 LYS E 28 8.121 −23.991 −0.712 1.00 0.00 N1+
    ATOM 172 HN LYS E 28 15.075 −21.548 0.336 1.00 0.00 H
    ATOM 173 HA LYS E 28 14.600 −23.660 −1.591 1.00 0.00 H
    ATOM 174 HB2 LYS E 28 12.608 −21.320 −1.392 1.00 0.00 H
    ATOM 175 HB3 LYS E 28 12.733 −22.451 −2.786 1.00 0.00 H
    ATOM 176 HG2 LYS E 28 12.188 −24.371 −1.257 1.00 0.00 H
    ATOM 177 HG3 LYS E 28 12.040 −23.226 0.119 1.00 0.00 H
    ATOM 178 HD2 LYS E 28 10.112 −22.090 −0.985 1.00 0.00 H
    ATOM 179 HD3 LYS E 28 10.218 −23.245 −2.362 1.00 0.00 H
    ATOM 180 HE2 LYS E 28 9.855 −25.173 −0.714 1.00 0.00 H
    ATOM 181 HE3 LYS E 28 9.735 −23.955 0.615 1.00 0.00 H
    ATOM 182 HZ1 LYS E 28 7.642 −24.701 −0.075 1.00 0.00 H
    ATOM 183 HZ2 LYS E 28 7.824 −22.985 −0.460 1.00 0.00 H
    ATOM 184 HZ3 LYS E 28 7.862 −24.209 −1.698 1.00 0.00 H
    ATOM 185 N GLY E 29 15.238 −20.414 −2.069 1.00 0.00 N
    ATOM 186 C GLY E 29 16.221 −19.472 −2.602 1.00 0.00 C
    ATOM 187 C GLY E 29 16.329 −18.178 −1.820 1.00 0.00 C
    ATOM 188 O GLY E 29 16.342 −17.105 −2.425 1.00 0.00 O
    ATOM 189 HN GLY E 29 14.466 −20.019 −1.577 1.00 0.00 H
    ATOM 190 HA2 GLY E 29 17.197 −19.939 −2.578 1.00 0.00 H
    ATOM 191 HA3 GLY E 29 15.907 −19.224 −3.607 1.00 0.00 H
    ATOM 192 N ALA E 30 16.422 −18.252 −0.458 1.00 0.00 N
    ATOM 193 CA ALA E 30 16.545 −17.145 0.489 1.00 0.00 C
    ATOM 194 C ALA E 30 15.354 −16.174 0.492 1.00 0.00 C
    ATOM 195 O ALA E 30 14.532 −16.217 1.409 1.00 0.00 O
    ATOM 196 CB ALA E 30 17.924 −16.432 0.416 1.00 0.00 C
    ATOM 197 HN ALA E 30 16.394 −19.157 −0.010 1.00 0.00 H
    ATOM 198 HA ALA E 30 16.518 −17.620 1.461 1.00 0.00 H
    ATOM 199 HB1 ALA E 30 18.057 −15.934 −0.568 1.00 0.00 H
    ATOM 200 HB2 ALA E 30 18.013 −15.667 1.216 1.00 0.00 H
    ATOM 201 HB3 ALA E 30 18.745 −17.169 0.545 1.00 0.00 H
    ATOM 202 N ILE E 31 15.230 −15.285 −0.535 1.00 0.00 N
    ATOM 203 CA ILE E 31 14.218 −14.224 −0.629 1.00 0.00 C
    ATOM 204 C ILE E 31 12.989 −14.791 −1.375 1.00 0.00 C
    ATOM 205 O ILE E 31 12.672 −14.414 −2.507 1.00 0.00 O
    ATOM 206 CB ILE E 31 14.773 −12.883 −1.203 1.00 0.00 C
    ATOM 207 CG1 ILE E 31 16.143 −12.433 −0.582 1.00 0.00 C
    ATOM 208 CG2 ILE E 31 13.711 −11.745 −1.112 1.00 0.00 C
    ATOM 209 CD1 ILE E 31 16.172 −12.182 0.938 1.00 0.00 C
    ATOM 210 HN ILE E 31 15.848 −15.388 −1.317 1.00 0.00 H
    ATOM 211 HA ILE E 31 13.897 −14.006 0.382 1.00 0.00 H
    ATOM 212 HB ILE E 31 14.972 −13.051 −2.289 1.00 0.00 H
    ATOM 213 HG12 ILE E 31 16.911 −13.200 −0.825 1.00 0.00 H
    ATOM 214 HG13 ILE E 31 16.462 −11.498 −1.097 1.00 0.00 H
    ATOM 215 HG21 ILE E 31 13.430 −11.548 −0.056 1.00 0.00 H
    ATOM 216 HG22 ILE E 31 14.122 −10.808 −1.543 1.00 0.00 H
    ATOM 217 HG23 ILE E 31 12.794 −12.002 −1.681 1.00 0.00 H
    ATOM 218 HD11 ILE E 31 15.913 −13.106 1.499 1.00 0.00 H
    ATOM 219 HD12 ILE E 31 17.189 −11.863 1.253 1.00 0.00 H
    ATOM 220 HD13 ILE E 31 15.456 −11.383 1.222 1.00 0.00 H
    ATOM 221 N ILE E 32 12.276 −15.746 −0.715 1.00 0.00 N
    ATOM 222 CA ILE E 32 11.097 −16.464 −1.206 1.00 0.00 C
    ATOM 223 C ILE E 32 10.373 −17.088 −0.006 1.00 0.00 C
    ATOM 224 O ILE E 32 10.993 −17.721 0.856 1.00 0.00 O
    ATOM 225 CB ILE E 32 11.393 −17.479 −2.356 1.00 0.00 C
    ATOM 226 CG1 ILE E 32 10.127 −18.060 −3.076 1.00 0.00 C
    ATOM 227 CG2 ILE E 32 12.450 −18.553 −1.962 1.00 0.00 C
    ATOM 228 CD1 ILE E 32 9.433 −19.286 −2.452 1.00 0.00 C
    ATOM 229 HN ILE E 32 12.602 −16.048 −0.185 1.00 0.00 H
    ATOM 230 HA ILE E 32 10.449 −15.702 −1.620 1.00 0.00 H
    ATOM 231 HB ILE E 32 11.890 −15.860 −3.146 1.00 0.00 H
    ATOM 232 HG12 ILE E 32 9.378 −17.249 −3.211 1.00 0.00 H
    ATOM 233 HG13 ILE E 32 10.447 −18.361 −4.101 1.00 0.00 H
    ATOM 234 HG21 ILE E 32 13.389 −18.065 −1.627 1.00 0.00 H
    ATOM 235 HG22 ILE E 32 12.064 −19.187 −1.136 1.00 0.00 H
    ATOM 236 HG23 ILE E 32 12.676 −19.197 −2.837 1.00 0.00 H
    ATOM 237 HD11 ILE E 32 10.124 −20.154 −2.407 1.00 0.00 H
    ATOM 238 HD12 ILE E 32 9.064 −19.088 −1.426 1.00 0.00 H
    ATOM 239 HD13 ILE E 32 8.554 −19.585 −3.062 1.00 0.00 H
    ATOM 240 N GLY E 33 9.029 −16.901 0.075 1.00 0.00 N
    ATOM 241 CA GLY E 33 8.210 −17.456 1.144 1.00 0.00 C
    ATOM 242 C GLY E 33 6.829 −16.852 1.184 1.00 0.00 C
    ATOM 243 O GLY E 33 6.600 −15.880 1.908 1.00 0.00 O
    ATOM 244 HN GLY E 33 8.548 −16.391 0.632 1.00 0.00 H
    ATOM 245 HA2 GLY E 33 8.131 −18.520 0.977 1.00 0.00 H
    ATOM 246 HA3 GLY E 33 8.691 −17.222 2.083 1.00 0.00 H
    ATOM 247 N LEU E 34 5.864 −17.435 0.423 1.00 0.00 N
    ATOM 248 CA LEU E 34 4.456 −17.032 0.382 1.00 0.00 C
    ATOM 249 C LEU E 34 3.710 −17.618 1.602 1.00 0.00 C
    ATOM 250 O LEU E 34 2.849 −18.495 1.492 1.00 0.00 O
    ATOM 251 CB LEU E 34 3.807 −17.395 −0.999 1.00 0.00 C
    ATOM 252 CG LEU E 34 2.606 −16.518 −1.483 1.00 0.00 C
    ATOM 253 CD1 LEU E 34 2.248 −16.841 −2.954 1.00 0.00 C
    ATOM 254 CD2 LEU E 34 1.347 −16.601 −0.591 1.00 0.00 C
    ATOM 255 HN LEU E 34 6.101 −18.272 −0.128 1.00 0.00 H
    ATOM 256 HA LEU E 34 4.439 −15.953 0.474 1.00 0.00 H
    ATOM 257 HB2 LEU E 34 4.604 −17.288 −1.771 1.00 0.00 H
    ATOM 258 HB3 LEU E 34 3.517 −18.468 −1.000 1.00 0.00 H
    ATOM 259 HG LEU E 34 2.950 −15.455 −1.466 1.00 0.00 H
    ATOM 260 HD11 LEU E 34 1.898 −17.891 −3.047 1.00 0.00 H
    ATOM 261 HD12 LEU E 34 1.446 −16.167 −3.320 1.00 0.00 H
    ATOM 262 HD13 LEU E 34 3.136 −16.709 −3.503 1.00 0.00 H
    ATOM 263 HD21 LEU E 34 1.569 −16.255 0.440 1.00 0.00 H
    ATOM 264 HD22 LEU E 34 0.532 −15.965 −0.996 1.00 0.00 H
    ATOM 265 HD23 LEU E 34 0.984 −17.650 −0.534 1.00 0.00 H
    ATOM 266 N MEI E 35 4.075 −17.119 2.814 1.00 0.00 N
    ATOM 267 CA MEI E 35 3.624 −17.583 4.123 1.00 0.00 C
    ATOM 268 C MEI E 35 2.523 −16.670 4.714 1.00 0.00 C
    ATOM 269 O MEI E 35 1.822 −15.947 3.995 1.00 0.00 O
    ATOM 270 CB MEI E 35 4.861 −17.768 5.073 1.00 0.00 C
    ATOM 271 CG MEI E 35 5.942 −18.759 4.568 1.00 0.00 C
    ATOM 272 SD MEI E 35 7.383 −18.952 5.668 1.00 0.00 S
    ATOM 273 CE MEI E 35 8.170 −17.332 5.411 1.00 0.00 C
    ATOM 274 HN MEI E 35 4.792 −16.420 2.823 1.00 0.00 H
    ATOM 275 HA MEI E 35 3.166 −18.555 3.990 1.00 0.00 H
    ATOM 276 HB2 MEI E 35 5.330 −16.775 5.244 1.00 0.00 H
    ATOM 277 HB3 MEI E 35 4.519 −18.161 6.055 1.00 0.00 H
    ATOM 278 HG2 MEI E 35 5.456 −19.752 4.443 1.00 0.00 H
    ATOM 279 HG3 MEI E 35 6.290 −18.447 3.561 1.00 0.00 H
    ATOM 280 HE1 MEI E 35 8.373 −17.150 4.334 1.00 0.00 H
    ATOM 281 HE2 MEI E 35 7.526 −16.507 5.783 1.00 0.00 H
    ATOM 282 HE3 MEI E 35 9.138 −17.276 5.953 1.00 0.00 H
    ATOM 283 N VAL E 36 2.330 −16.695 6.067 1.00 0.00 N
    ATOM 284 CA VAL E 36 1.301 −15.972 6.822 1.00 0.00 C
    ATOM 285 C VAL E 36 1.640 −14.462 6.866 1.00 0.00 C
    ATOM 286 O VAL E 36 2.086 −13.905 7.875 1.00 0.00 O
    ATOM 287 C VAL E 36 1.004 −16.594 8.226 1.00 0.00 C
    ATOM 288 CG1 VAL E 36 −0.158 −15.864 8.962 1.00 0.00 C
    ATOM 289 CG2 VAL E 36 0.676 −18.113 8.129 1.00 0.00 C
    ATOM 290 HN VAL E 36 2.894 −17.297 6.623 1.00 0.00 H
    ATOM 291 HA VAL E 36 0.386 −16.0.74 6.251 1.00 0.00 H
    ATOM 292 HB VAL E 36 1.921 −16.486 8.850 1.00 0.00 H
    ATOM 293 HG11 VAL E 36 0.075 −14.792 9.128 1.00 0.00 H
    ATOM 294 HG12 VAL E 36 −1.101 −15.941 8.383 1.00 0.00 H
    ATOM 295 HG13 VAL E 36 −0.326 −16.326 9.958 1.00 0.00 H
    ATOM 296 HG21 VAL E 36 0.481 −18.535 9.137 1.00 0.00 H
    ATOM 297 HG22 VAL E 36 −0.224 −18.282 7.499 1.00 0.00 H
    ATOM 298 HG23 VAL E 36 1.517 −18.684 7.683 1.00 0.00 H
    ATOM 299 N GLY E 37 1.403 −13.749 5.730 1.00 0.00 N
    ATOM 300 CA GLY E 37 1.476 −12.292 5.611 1.00 0.00 C
    ATOM 301 C GLY E 37 0.283 −11.611 6.248 1.00 0.00 C
    ATOM 302 O GLY E 37 −0.559 −11.032 5.560 1.00 0.00 O
    ATOM 303 HN GLY E 37 1.196 −14.271 4.095 1.00 0.00 H
    ATOM 304 HA2 GLY E 37 2.377 −11.951 6.105 1.00 0.00 H
    ATOM 305 HA3 GLY E 37 1.465 −12.057 4.556 1.00 0.00 H
    ATOM 306 N GLY E 37 0.186 −11.673 7.601 1.00 0.00 H
    ATOM 307 CA GLY E 38 −0.986 −11.294 8.387 1.00 0.00 C
    ATOM 308 C GLY E 38 −1.293 −19.816 8.541 1.00 0.00 C
    ATOM 309 O GLY E 38 −1.509 −9.350 9.663 1.00 0.00 O
    ATOM 310 HN GLY E 38 0.930 −12.143 8.086 1.00 0.00 H
    ATOM 311 HA2 GLY E 38 −1.847 −11.747 7.917 1.00 0.00 H
    ATOM 312 HA3 GLY E 38 −0.823 −11.687 9.380 1.00 0.00 H
    ATOM 313 N VAL E 39 −1.365 −9.070 7.407 1.00 0.00 N
    ATOM 314 CA VAL E 39 −1.581 −7.621 7.265 1.00 0.00 C
    ATOM 315 C VAL E 39 −2.828 −7.129 8.043 1.00 0.00 C
    ATOM 316 O VAL E 39 −3.865 −7.803 8.123 1.00 0.00 O
    ATOM 317 CB VAL E 39 −1.591 −7.158 5.770 1.00 0.00 C
    ATOM 318 CG1 VAL E 39 −1.813 −5.628 5.590 1.00 0.00 C
    ATOM 319 CG2 VAL E 39 −0.276 −7.557 5.037 1.00 0.00 C
    ATOM 320 HN VAL E 39 −0.196 −9.555 6.546 1.00 0.00 H
    ATOM 321 HA VAL E 39 −0.718 −7.166 7.733 1.00 0.00 H
    ATOM 322 HB VAL E 39 −2.433 −7.680 5.258 1.00 0.00 H
    ATOM 323 HG11 VAL E 39 −2.801 −5.311 5.981 1.00 0.00 H
    ATOM 324 HD12 VAL E 39 −1.021 −5.044 6.104 1.00 0.00 H
    ATOM 325 HD13 VAL E 39 −1.791 −5.361 4.511 1.00 0.00 H
    ATOM 326 HG21 VAL E 39 −0.312 −7.229 3.976 1.00 0.00 H
    ATOM 327 HG22 VAL E 39 0.607 −7.082 5.516 1.00 0.00 H
    ATOM 328 HG23 VAL E 39 −0.125 −8.655 5.035 1.00 0.00 H
    ATOM 329 N VAL E 40 −2.722 −5.916 8.658 1.00 0.00 N
    ATOM 330 CA VAL E 40 −3.784 −5.198 9.369 1.00 0.00 C
    ATOM 331 C VAL E 40 −4.604 −4.416 8.325 1.00 0.00 C
    ATOM 332 O VAL E 40 −4.082 −3.559 7.606 1.00 0.00 O
    ATOM 333 CB VAL E 40 −3.280 −4.281 10.522 1.00 0.00 C
    ATOM 334 CG1 VAL E 40 −4.454 −3.576 11.262 1.00 0.00 C
    ATOM 335 CG2 VAL E 40 −2.414 −5.071 11.545 1.00 0.00 C
    ATOM 336 HN VAL E 40 −1.866 −5.410 8.597 1.00 0.00 H
    ATOM 337 HA VAL E 40 −4.422 −5.947 9.817 1.00 0.00 H
    ATOM 338 HB VAL E 40 −2.635 −3.486 10.078 1.00 0.00 H
    ATOM 339 HG11 VAL E 40 −4.060 −2.931 12.077 1.00 0.00 H
    ATOM 340 HG12 VAL E 40 −5.031 −2.922 10.575 1.00 0.00 H
    ATOM 341 HG13 VAL E 40 −5.148 −4.316 11.712 1.00 0.00 H
    ATOM 342 HG21 VAL E 40 −2.043 −4.389 12.340 1.00 0.00 H
    ATOM 343 HG22 VAL E 40 −3.003 −5.878 12.029 1.00 0.00 H
    ATOM 344 HG23 VAL E 40 −1.527 −5.526 11.056 1.00 0.00 H
    ATOM 345 N ILE E 41 −5.924 −4.729 8.236 1.00 0.00 N
    ATOM 346 CA ILE E 41 −6.893 −4.168 7.297 1.00 0.00 C
    ATOM 347 C ILE E 41 −7.889 −3.330 8.114 1.00 0.00 C
    ATOM 348 O ILE E 41 −8.504 −3.828 9.064 1.00 0.00 O
    ATOM 349 CB ILE E 41 −7.608 −5.248 6.430 1.00 0.00 C
    ATOM 350 CG1 ILE E 41 −6.621 −6.237 5.713 1.00 0.00 C
    ATOM 351 CG2 ILE E 41 −8.613 −4.614 5.420 1.00 0.00 C
    ATOM 352 CD1 ILE E 41 −5.635 −5.625 4.699 1.00 0.00 C
    ATOM 353 HN ILE E 41 −6.322 −5.394 8.860 1.00 0.00 H
    ATOM 354 HA ILE E 41 −6.357 −3.510 6.624 1.00 0.00 H
    ATOM 355 HB ILE E 41 −8.215 −5.878 7.125 1.00 0.00 H
    ATOM 356 HG12 ILE E 41 −6.032 −6.778 6.487 1.00 0.00 H
    ATOM 357 HG13 ILE E 41 −7.228 −7.008 5.187 1.00 0.00 H
    ATOM 358 HG21 ILE E 41 −9.411 −4.048 5.944 1.00 0.00 H
    ATOM 359 HG22 ILE E 41 −8.092 −3.930 4.717 1.00 0.00 H
    ATOM 360 HG23 ILE E 41 −9.106 −5.412 4.824 1.00 0.00 H
    ATOM 361 HD11 ILE E 41 −4.985 −6.419 4.272 1.00 0.00 H
    ATOM 362 HD12 ILE E 41 −6.178 −5.137 3.863 1.00 0.00 H
    ATOM 363 HD13 ILE E 41 −4.982 −4.870 5.185 1.00 0.00 H
    ATOM 364 N ALA E 42 −8.047 −2.029 7.739 1.00 0.00 N
    ATOM 365 CA ALA E 42 −8.966 −1.076 8.334 1.00 0.00 C
    ATOM 366 C ALA E 42 −10.273 −1.087 7.568 1.00 0.00 C
    ATOM 367 G ALA E 42 −10.303 −0.735 6.660 0.00 0.00 H
    ATOM 368 O ALA E 42 −11.305 −1.536 8.069 1.00 0.00 O
    ATOM 369 CB ALA E 42 −8.369 0.348 8.322 1.00 0.00 C
    ATOM 370 HN ALA E 42 −7.500 −1.661 6.994 1.00 0.00 H
    ATOM 371 HA ALA E 42 −9.170 −1.376 9.356 1.00 0.00 H
    ATOM 372 HB1 ALA E 42 −7.417 0.368 8.894 1.00 0.00 H
    ATOM 373 HB2 ALA E 42 −8.162 0.694 7.287 1.00 0.00 H
    ATOM 374 HB3 ALA E 42 −9.068 1.058 8.801 1.00 0.00 H
    TER 375 ALA E 42 1.00 0.00
    ATOM 376 N LEU E 17 −4.694 −36.140 7.746 1.00 0.00 N1+
    ATOM 377 H1 LEU E 17 −4.458 −36.755 8.545 0.00 0.00 H
    ATOM 378 H2 LEU E 17 −5.616 −36.439 7.314 0.00 0.00 H
    ATOM 379 CA LEU E 17 −4.787 −34.713 8.198 1.00 0.00 C
    ATOM 380 C LEU E 17 −4.985 −33.740 7.028 1.00 0.00 C
    ATOM 381 O LEU E 17 −4.889 −34.132 5.863 1.00 0.00 O
    ATOM 382 CB LEU E 17 −3.484 −34.357 8.997 1.00 0.00 C
    ATOM 383 CG LEU E 17 −3.348 −34.891 10.461 1.00 0.00 C
    ATOM 384 CD1 LEU E 17 −3.351 −36.431 10.610 1.00 0.00 C
    ATOM 385 CD2 LEU E 17 −2.088 −34.291 11.132 1.00 0.00 C
    ATOM 386 H3 LEU E 17 −3.931 −36.221 7.013 1.00 0.00 H
    ATOM 387 HA LEU E 17 −5.656 −34.629 8.839 1.00 0.00 H
    ATOM 388 HB2 LEU E 17 −2.595 −34.674 8.409 1.00 0.00 H
    ATOM 389 HB3 LEU E 17 −3.420 −33.246 9.076 1.00 0.00 H
    ATOM 390 HG LEU E 17 −4.222 −34.505 11.040 1.00 0.00 H
    ATOM 391 HD11 LEU E 17 −3.145 −36.712 11.667 1.00 0.00 H
    ATOM 392 HD12 LEU E 17 −4.339 −36.863 10.355 1.00 0.00 H
    ATOM 393 HD13 LEU E 17 −2.563 −36.894 9.978 1.00 0.00 H
    ATOM 394 HD21 LEU E 17 −2.027 −34.598 12.198 1.00 0.00 H
    ATOM 395 HD22 LEU E 17 −1.165 −34.631 10.616 1.00 0.00 H
    ATOM 396 HD23 LEU E 17 −2.117 −33.181 11.098 1.00 0.00 H
    ATOM 397 N VAL E 18 −5.250 −32.431 7.323 1.00 0.00 N
    ATOM 398 CA VAL E 18 −5.140 −31.322 6.362 1.00 0.00 C
    ATOM 399 C VAL E 18 −3.628 −31.029 6.265 1.00 0.00 C
    ATOM 400 O VAL E 18 −3.062 −30.266 7.053 1.00 0.00 O
    ATOM 401 CB VAL E 18 −6.000 −30.072 6.707 1.00 0.00 C
    ATOM 402 CG1 VAL E 18 −5.834 −28.954 5.638 1.00 0.00 C
    ATOM 403 CG2 VAL E 18 −7.504 −30.432 6.853 1.00 0.00 C
    ATOM 404 HN VAL E 18 −5.442 −32.132 8.254 1.00 0.00 H
    ATOM 405 HA VAL E 18 −5.481 −31.687 5.400 1.00 0.00 H
    ATOM 406 HB VAL E 18 −5.647 −29.662 7.682 1.00 0.00 H
    ATOM 407 HG11 VAL E 18 −6.453 −28.072 5.907 1.00 0.00 H
    ATOM 408 HG12 VAL E 18 −4.779 −28.612 5.576 1.00 0.00 H
    ATOM 409 HG13 VAL E 18 −6.153 −29.308 4.635 1.00 0.00 H
    ATOM 410 HG21 VAL E 18 −8.091 −29.534 7.135 1.00 0.00 H
    ATOM 411 HG22 VAL E 18 −7.914 −30.840 5.913 1.00 0.00 H
    ATOM 412 HG23 VAL E 18 −7.656 −31.191 7.662 1.00 0.00 H
    ATOM 413 N PHE E 19 −2.950 −31.706 5.300 1.00 0.00 N
    ATOM 414 CA PHE E 19 −1.507 −31.863 5.235 1.00 0.00 C
    ATOM 415 C PHE E 19 −0.908 −31.009 4.116 1.00 0.00 C
    ATOM 416 O PHE E 19 −1.194 −31.223 2.934 1.00 0.00 O
    ATOM 417 CB PHE E 19 −1.144 −33.374 5.046 1.00 0.00 C
    ATOM 418 CG PHE E 19 0.246 −33.730 5.530 1.00 0.00 C
    ATOM 419 CD1 PHE E 19 0.416 −34.446 6.732 1.00 0.00 C
    ATOM 420 CD2 PHE E 19 1.389 −33.412 4.774 1.00 0.00 C
    ATOM 421 CE1 PHE E 19 1.690 −34.842 7.157 1.00 0.00 C
    ATOM 422 CE2 PHE E 19 2.665 −33.796 5.201 1.00 0.00 C
    ATOM 423 C2 PHE E 19 2.815 −34.517 6.390 1.00 0.00 C
    ATOM 424 HN PHE E 19 −3.455 −32.243 4.629 1.00 0.00 H
    ATOM 425 HA PHE E 19 −1.099 −31.539 6.186 1.00 0.00 H
    ATOM 426 HB2 PHE E 19 −1.860 −33.993 5.630 1.00 0.00 H
    ATOM 427 HB3 PHE E 19 −1.236 −33.677 3.981 1.00 0.00 H
    ATOM 428 HD1 PHE E 19 −0.444 −34.712 7.329 1.00 0.00 H
    ATOM 429 HD2 PHE E 19 1.294 −32.865 3.849 1.00 0.00 H
    ATOM 430 HE1 PHE E 19 1.808 −35.396 8.078 1.00 0.00 H
    ATOM 431 HE2 PHE E 19 3.534 −33.535 4.610 1.00 0.00 H
    ATOM 432 H2 PHE E 19 3.799 −34.822 6.714 1.00 0.00 H
    ATOM 433 N PHE E 20 −0.039 −30.030 4.488 1.00 0.00 N
    ATOM 434 CA PHE E 20 0.810 −29.269 3.578 1.00 0.00 C
    ATOM 435 C PHE E 20 2.023 −30.138 3.216 1.00 0.00 C
    ATOM 436 O PHE E 20 2.811 −30.498 4.094 1.00 0.00 O
    ATOM 437 CB PHE E 20 1.247 −27.904 4.195 1.00 0.00 C
    ATOM 438 CG PHE E 20 1.908 −27.003 3.173 1.00 0.00 C
    ATOM 439 CD1 PHE E 20 1.129 −26.172 2.345 1.00 0.00 C
    ATOM 440 CD2 PHE E 20 3.310 −26.966 3.043 1.00 0.00 C
    ATOM 441 CE1 PHE E 20 1.735 −25.324 1.410 1.00 0.00 C
    ATOM 442 CE2 PHE E 20 3.916 −26.119 2.106 1.00 0.00 C
    ATOM 443 C2 PHE E 20 3.129 −25.300 1.291 1.00 0.00 C
    ATOM 444 HN PHE E 20 0.114 −29.837 5.453 1.00 0.00 H
    ATOM 445 HA PHE E 20 0.227 −29.072 2.686 1.00 0.00 H
    ATOM 446 HB2 PHE E 20 0.355 −27.366 4.583 1.00 0.00 H
    ATOM 447 HB3 PHE E 20 1.948 −28.061 5.043 1.00 0.00 H
    ATOM 448 HD1 PHE E 20 0.052 −26.176 2.434 1.00 0.00 H
    ATOM 449 HD2 PHE E 20 3.929 −27.588 3.674 1.00 0.00 H
    ATOM 450 HE1 PHE E 20 1.134 −24.674 0.791 1.00 0.00 H
    ATOM 451 HE2 PHE E 20 4.991 −26.076 2.016 1.00 0.00 H
    ATOM 452 H2 PHE E 20 3.601 −24.631 0.585 1.00 0.00 H
    ATOM 453 N ALA E 21 2.164 −30.498 1.906 1.00 0.00 N
    ATOM 454 CA ALA E 21 3.079 −31.491 1.341 1.00 0.00 C
    ATOM 455 C ALA E 21 4.497 −31.514 1.936 1.00 0.00 C
    ATOM 456 O ALA E 21 4.883 −32.512 2.545 1.00 0.00 O
    ATOM 457 CB ALA E 21 3.099 −31.403 −0.206 1.00 0.00 C
    ATOM 458 HN ALA E 21 1.535 −30.112 1.238 1.00 0.00 H
    ATOM 459 HA ALA E 21 2.642 −32.445 1.597 1.00 0.00 H
    ATOM 460 HB1 ALA E 21 3.737 −32.206 −0.634 1.00 0.00 H
    ATOM 461 HB2 ALA E 21 2.074 −31.531 −0.614 1.00 0.00 H
    ATOM 462 HB3 ALA E 21 3.492 −30.422 −0.548 1.00 0.00 H
    ATOM 463 N GLU E 22 5.263 −30.400 1.771 1.00 0.00 N
    ATOM 464 CA GLU E 22 6.475 −30.018 2.485 1.00 0.00 C
    ATOM 465 C GLU E 22 7.570 −31.077 2.664 1.00 0.00 C
    ATOM 466 O GLU E 22 7.654 −31.678 3.738 1.00 0.00 O
    ATOM 467 CB GLU E 22 7.018 −28.644 1.978 1.00 0.00 C
    ATOM 468 CG GLU E 22 7.380 −28.531 0.468 1.00 0.00 C
    ATOM 469 CD GLU E 22 7.607 −27.099 −0.012 1.00 0.00 C
    ATOM 470 OE1 GLU E 22 7.580 −26.133 0.797 1.00 0.00 O1−
    ATOM 471 OE2 GLU E 22 7.823 −26.935 −1.242 1.00 0.00 O
    ATOM 472 HN GLU E 22 4.917 −29.682 1.178 1.00 0.00 H
    ATOM 473 HA GLU E 22 6.131 −29.822 3.493 1.00 0.00 H
    ATOM 474 HB2 GLU E 22 6.226 −27.892 2.186 1.00 0.00 H
    ATOM 475 HB3 GLU E 22 7.902 −28.351 2.586 1.00 0.00 H
    ATOM 476 HG2 GLU E 22 8.310 −29.089 0.241 1.00 0.00 H
    ATOM 477 HG3 GLU E 22 6.560 −28.945 −0.157 1.00 0.00 H
    ATOM 478 N ASP E 23 8.453 −31.311 1.649 1.00 0.00 N
    ATOM 479 CA ASP E 23 9.659 −32.116 1.788 1.00 0.00 C
    ATOM 480 C ASP E 23 10.698 −31.346 2.631 1.00 0.00 C
    ATOM 481 O ASP E 23 11.428 −30.480 2.139 1.00 0.00 O
    ATOM 482 CB ASP E 23 10.222 −32.625 0.425 1.00 0.00 C
    ATOM 483 CG ASP E 23 11.244 −33.725 0.658 1.00 0.00 C
    ATOM 484 OD1 ASP E 23 10.844 −34.910 0.826 1.00 0.00 O1−
    ATOM 485 OD2 ASP E 23 12.465 −33.425 0.700 1.00 0.00 O
    ATOM 486 HN ASP E 23 8.323 −30.871 0.766 1.00 0.00 H
    ATOM 487 HA ASP E 23 9.361 −32.997 2.342 1.00 0.00 H
    ATOM 488 HB2 ASP E 23 9.402 −33.049 −0.193 1.00 0.00 H
    ATOM 489 HB3 ASP E 23 10.701 −31.795 −0.134 1.00 0.00 H
    ATOM 490 N VAL E 24 10.738 −31.665 3.954 1.00 0.00 N
    ATOM 491 CA VAL E 24 11.639 −31.109 4.967 1.00 0.00 C
    ATOM 492 C VAL E 24 13.004 −31.808 4.869 1.00 0.00 C
    ATOM 493 O VAL E 24 13.086 −33.050 4.779 1.00 0.00 O
    ATOM 494 CB VAL E 24 11.046 −31.152 6.410 1.00 0.00 C
    ATOM 495 CG1 VAL E 24 12.015 −30.571 7.480 1.00 0.00 C
    ATOM 496 CG2 VAL E 24 9.688 −30.395 6.481 1.00 0.00 C
    ATOM 497 HN VAL E 24 10.056 −32.300 4.304 1.00 0.00 H
    ATOM 498 HA VAL E 24 11.782 −30.069 4.705 1.00 0.00 H
    ATOM 499 HB VAL E 24 10.854 −32.218 6.675 1.00 0.00 H
    ATOM 500 HG11 VAL E 24 11.547 −30.618 8.486 1.00 0.00 H
    ATOM 501 HG12 VAL E 24 12.961 −31.149 7.527 1.00 0.00 H
    ATOM 502 HG13 VAL E 24 12.257 −29.509 7.262 1.00 0.00 H
    ATOM 503 HG21 VAL E 24 9.816 −29.327 6.201 1.00 0.00 H
    ATOM 504 HG22 VAL E 24 8.938 −30.846 5.799 1.00 0.00 H
    ATOM 505 HG23 VAL E 24 9.272 −3.439 7.510 1.00 0.00 H
    ATOM 506 N GLY E 25 14.111 −31.024 4.883 1.00 0.00 H
    ATOM 507 CA GLY E 25 15.491 −31.477 4.729 1.00 0.00 C
    ATOM 508 C GLY E 25 16.017 −31.220 3.332 1.00 0.00 C
    ATOM 509 O GLY E 25 15.815 −32.033 2.430 1.00 0.00 O
    ATOM 510 HN GLY E 25 13.991 −30.028 5.022 1.00 0.00 H
    ATOM 511 HA2 GLY E 25 16.089 −30.927 5.442 1.00 0.00 H
    ATOM 512 HA3 GLY E 25 15.532 −32.543 4.902 1.00 0.00 H
    ATOM 513 N SER E 26 16.721 −30.075 3.137 1.00 0.00 N
    ATOM 514 CA SER E 26 17.327 −29.655 1.868 1.00 0.00 C
    ATOM 515 C SER E 26 18.644 −30.379 1.577 1.00 0.00 C
    ATOM 516 O SER E 26 19.451 −30.630 2.474 1.00 0.00 O
    ATOM 517 CB SER E 26 17.571 −28.122 1.768 1.00 0.00 C
    ATOM 518 OG SER E 26 16.328 −27.439 1.628 1.00 0.00 O
    ATOM 519 HN SER E 26 16.861 −29.447 3.893 1.00 0.00 H
    ATOM 520 HA SER E 26 16.623 −29.917 1.088 1.00 0.00 H
    ATOM 521 HB2 SER E 26 18.115 −27.758 2.668 1.00 0.00 H
    ATOM 522 HB3 SER E 26 18.180 −27.881 0.869 1.00 0.00 H
    ATOM 523 HG SER E 26 16.363 −26.617 2.166 1.00 0.00 H
    ATOM 524 N ASN E 27 18.871 −30.711 0.274 1.00 0.00 N
    ATOM 525 CA ASN E 27 20.009 −31.447 −0.279 1.00 0.00 C
    ATOM 526 C ASN E 27 20.023 −32.925 0.159 1.00 0.00 C
    ATOM 527 O ASN E 27 20.841 −33.343 0.984 1.00 0.00 O
    ATOM 528 CB ASN E 27 21.374 −30.702 −0.060 1.00 0.00 C
    ATOM 529 CG ASN E 27 22.516 −31.239 −0.934 1.00 0.00 C
    ATOM 530 OD1 ASN E 27 22.885 −30.637 −1.944 1.00 0.00 O
    ATOM 531 ND2 ASN E 27 23.095 −32.400 −0.539 1.00 0.00 N
    ATOM 532 HN ASN E 27 18.204 −30.396 −0.412 1.00 0.00 H
    ATOM 533 HA ASN E 27 19.842 −31.445 −1.348 1.00 0.00 H
    ATOM 534 HB2 ASN E 27 21.243 −29.629 −0.315 1.00 0.00 H
    ATOM 535 HB3 ASN E 27 21.668 −30.750 1.008 1.00 0.00 H
    ATOM 536 HD21 ASN E 27 22.675 −32.914 0.214 1.00 0.00 H
    ATOM 537 HD22 ASN E 27 23.857 −32.7500 −1.077 1.00 0.00 H
    ATOM 538 N LYS E 28 19.102 −33.739 −0.428 1.00 0.00 N
    ATOM 539 CA LYS E 28 18.955 −35.175 −0.209 1.00 0.00 C
    ATOM 540 C LYS E 28 19.695 −35.968 −1.294 1.00 0.00 C
    ATOM 541 O LYS E 28 20.870 −36.290 −1.118 1.00 0.00 O
    ATOM 542 CB LYS E 28 17.450 −35.571 −0.050 1.00 0.00 C
    ATOM 543 CG LYS E 28 16.846 −35.172 1.314 1.00 0.00 C
    ATOM 544 CD LYS E 28 15.304 −35.182 1.346 1.00 0.00 C
    ATOM 545 CE LYS E 28 14.738 −35.040 2.763 1.00 0.00 C
    ATOM 546 N2 LYS E 28 13.306 −34.710 2.730 1.00 0.00 N1+
    ATOM 547 HN LYS E 28 18.436 −33.340 −1.050 1.00 0.00 H
    ATOM 548 HA LYS E 28 19.452 −35.425 0.721 1.00 0.00 H
    ATOM 549 HB2 LYS E 28 16.859 −35.104 −0.868 1.00 0.00 H
    ATOM 550 HB3 LYS E 28 17.340 −36.675 −0.145 1.00 0.00 H
    ATOM 551 HG2 LYS E 28 17.245 −35.869 2.086 1.00 0.00 H
    ATOM 552 HG3 LYS E 28 17.179 −34.143 1.587 1.00 0.00 H
    ATOM 553 HD2 LYS E 28 14.952 −34.334 0.713 1.00 0.00 H
    ATOM 554 HD3 LYS E 28 14.918 −36.122 0.900 1.00 0.00 H
    ATOM 555 HE2 LYS E 28 14.783 −35.978 3.343 1.00 0.00 H
    ATOM 556 HE3 LYS E 28 15.255 −34.220 3.305 1.00 0.00 H
    ATOM 557 H21 LYS E 28 12.695 −35.544 2.088 1.00 0.00 H
    ATOM 558 H22 LYS E 28 13.082 −34.111 3.574 1.00 0.00 H
    ATOM 559 H23 LYS E 28 13.075 −34.124 1.855 1.00 0.00 H
    ATOM 560 N GLY E 29 19.027 −36.300 −2.437 1.00 0.00 N
    ATOM 561 CA GLY E 29 19.565 −37.146 −3.501 1.00 0.00 C
    ATOM 562 C GLY E 29 18.480 −38.474 −3.542 1.00 0.00 C
    ATOM 563 O GLY E 29 18.975 −39.289 −2.625 1.00 0.00 O
    ATOM 564 HN GLY E 29 18.087 −35.992 −2.549 1.00 0.00 H
    ATOM 565 HA2 GLY E 29 19.426 −36.618 −4.434 1.00 0.00 H
    ATOM 566 HA3 GLY E 29 20.606 −37.358 −3.299 1.00 0.00 H
    ATOM 567 N ALA E 30 18.044 −38.713 −4.623 1.00 0.00 N
    ATOM 568 CA ALA E 30 17.195 −39.887 −4.853 1.00 0.00 C
    ATOM 569 C ALA E 30 16.033 −40.028 −3.855 1.00 0.00 C
    ATOM 570 O ALA E 30 14.870 −10.004 −4.264 1.00 0.00 O
    ATOM 571 CB ALA E 30 17.996 −41.209 −5.016 1.00 0.00 C
    ATOM 572 HN ALA E 30 18.002 −38.032 −5.347 1.00 0.00 H
    ATOM 573 HA ALA E 30 15.728 −39.708 −5.812 1.00 0.00 H
    ATOM 574 HB1 ALA E 30 18.515 −41.475 −4.071 1.00 0.00 H
    ATOM 575 HB2 ALA E 30 17.320 −42.045 −5.296 1.00 0.00 H
    ATOM 576 HB3 ALA E 30 18.761 −41.098 −5.813 1.00 0.00 H
    ATOM 577 N ILE E 31 16.329 −40.162 −2.532 1.00 0.00 N
    ATOM 578 CA ILE E 31 15.363 −40.317 −1.441 1.00 0.00 C
    ATOM 579 C ILE E 31 14.825 −38.916 −1.073 1.00 0.00 C
    ATOM 580 O ILE E 31 15.252 −38.282 −0.104 1.00 0.00 O
    ATOM 581 CB ILE E 31 15.912 −41.130 −0.230 1.00 0.00 C
    ATOM 582 CG1 ILE E 31 16.630 −42.468 −0.626 1.00 0.00 C
    ATOM 583 CG2 ILE E 31 14.803 −41.375 0.839 1.00 0.00 C
    ATOM 584 CD1 ILE E 31 15.789 −43.516 −1.380 1.00 0.00 C
    ATOM 585 HN ILE E 31 17.299 −40.107 −2.271 1.00 0.00 H
    ATOM 586 HA ILE E 31 14.533 −40.888 −1.840 1.00 0.00 H
    ATOM 587 HB ILE E 31 16.694 −40.500 0.259 1.00 0.00 H
    ATOM 588 HG12 ILE E 31 17.520 −42.222 −1.248 1.00 0.00 H
    ATOM 589 HG13 ILE E 31 17.021 −42.937 0.304 1.00 0.00 H
    ATOM 590 HG21 ILE E 31 13.963 −41.963 0.413 1.00 0.00 H
    ATOM 591 HG22 ILE E 31 15.222 −41.940 1.698 1.00 0.00 H
    ATOM 592 HG23 ILE E 31 14.402 −40.417 1.230 1.00 0.00 H
    ATOM 593 HD11 ILE E 31 15.426 −43.112 −2.349 1.00 0.00 H
    ATOM 594 HD12 ILE E 31 16.402 −44.418 −1.591 1.00 0.00 H
    ATOM 595 HD13 ILE E 31 14.911 −43.829 −0.776 1.00 0.00 H
    ATOM 596 N ILE E 32 13.866 −38.413 −1.894 1.00 0.00 N
    ATOM 597 CA ILE E 32 13.175 −37.125 −1.794 1.00 0.00 C
    ATOM 598 C ILE E 32 11.673 −37.376 −2.006 1.00 0.00 C
    ATOM 599 O ILE E 32 11.276 −38.069 −2.950 1.00 0.00 O
    ATOM 600 CB ILE E 32 13.798 −36.045 −2.733 1.00 0.00 C
    ATOM 601 CG1 ILE E 32 13.397 −34.568 −2.405 1.00 0.00 C
    ATOM 602 CG2 ILE E 32 13.703 −36.399 −4.249 1.00 0.00 C
    ATOM 603 CD1 ILE E 32 12.021 −34.061 −2.875 1.00 0.00 C
    ATOM 604 HN ILE E 32 13.600 −38.950 −2.697 1.00 0.00 H
    ATOM 605 HA ILE E 32 13.305 −36.780 −0.776 1.00 0.00 H
    ATOM 606 HB ILE E 32 14.892 −36.078 −2.497 1.00 0.00 H
    ATOM 607 HG12 ILE E 32 13.485 −34.405 −1.308 1.00 0.00 H
    ATOM 608 HG13 ILE E 32 14.163 −33.909 −2.875 1.00 0.00 H
    ATOM 609 HG21 ILE E 32 14.149 −37.394 −4.455 1.00 0.00 H
    ATOM 610 HG22 ILE E 32 12.648 −36.415 −4.583 1.00 0.00 H
    ATOM 611 HG23 ILE E 32 14.247 −35.643 −4.853 1.00 0.00 H
    ATOM 612 HD11 ILE E 32 11.908 −34.171 −3.974 1.00 0.00 H
    ATOM 613 HD12 ILE E 32 11.192 −34.601 −2.378 1.00 0.00 H
    ATOM 614 HD13 ILE E 32 11.913 −32.983 −2.623 1.00 0.00 H
    ATOM 615 N GLY E 33 10.798 −36.830 −1.114 1.00 0.00 N
    ATOM 616 CA GLY E 33 9.354 −37.053 −1.143 1.00 0.00 C
    ATOM 617 C GLY E 33 8.801 −37.518 0.184 1.00 0.00 C
    ATOM 618 O GLY E 33 8.339 −38.654 0.302 1.00 0.00 O
    ATOM 619 HN GLY E 33 11.092 −36.185 −0.384 1.00 0.00 H
    ATOM 620 HA2 GLY E 33 8.892 −36.106 −1.383 1.00 0.00 H
    ATOM 621 HA3 GLY E 33 9.122 −37.819 −1.871 1.00 0.00 H
    ATOM 622 N LEU E 34 8.823 −36.622 1.207 1.00 0.00 N
    ATOM 623 CA LEU E 34 8.145 −36.731 2.499 1.00 0.00 C
    ATOM 624 C LEU E 34 6.660 −37.126 2.362 1.00 0.00 C
    ATOM 625 O LEU E 34 5.918 −36.579 1.541 1.00 0.00 O
    ATOM 626 CB LEU E 34 8.344 −34.415 3.327 1.00 0.00 C
    ATOM 627 CG LEU E 34 7.941 −35.418 4.838 1.00 0.00 C
    ATOM 628 CD1 LEU E 34 8.825 −34.451 5.664 1.00 0.00 C
    ATOM 629 CD2 LEU E 34 6.448 −35.085 5.072 1.00 0.00 C
    ATOM 630 HN LEU E 34 9.416 −35.803 1.082 1.00 0.00 H
    ATOM 631 HA LEU E 34 8.659 −37.529 3.017 1.00 0.00 H
    ATOM 632 HB2 LEU E 34 9.434 −35.189 3.272 1.00 0.00 H
    ATOM 633 HB3 LEU E 34 7.822 −34.577 2.813 1.00 0.00 H
    ATOM 634 HG LEU E 34 8.133 −36.439 5.243 1.00 0.00 H
    ATOM 635 HD11 LEU E 34 8.647 −33.401 5.351 1.00 0.00 H
    ATOM 636 HD12 LEU E 34 8.585 −34.534 6.746 1.00 0.00 H
    ATOM 637 HD13 LEU E 34 9.901 −34.588 5.530 1.00 0.00 H
    ATOM 638 HD21 LEU E 34 5.779 −35.811 4.569 1.00 0.00 H
    ATOM 639 HD22 LEU E 34 6.209 −35.100 6.156 1.00 0.00 H
    ATOM 640 HD23 LEU E 34 6.211 −34.073 4.675 1.00 0.00 H
    ATOM 641 N MET E 35 6.217 −38.123 3.183 1.00 0.00 N
    ATOM 642 CA MET E 35 4.910 −38.770 3.124 1.00 0.00 C
    ATOM 643 C MET E 35 3.765 −37.812 3.481 1.00 0.00 C
    ATOM 644 O MET E 35 3.585 −37.420 4.638 1.00 0.00 O
    ATOM 645 C MET E 35 4.833 −40.051 4.011 1.00 0.00 C
    ATOM 646 CG MET E 35 5.798 −41.192 3.604 1.00 0.00 C
    ATOM 647 SD MET E 35 5.642 −42.707 4.610 1.00 0.00 S
    ATOM 648 CE MET E 35 6.391 −42.115 6.158 1.00 0.00 C
    ATOM 649 HN MET E 35 6.827 −38.499 3.871 1.00 0.00 H
    ATOM 650 HA MET E 35 4.779 −39.088 2.097 1.00 0.00 H
    ATOM 651 HB2 MET E 35 5.024 −39.765 5.068 1.00 0.00 H
    ATOM 652 HB3 MET E 35 3.798 −40.460 3.961 1.00 0.00 H
    ATOM 653 HG2 MET E 35 5.593 −41.452 2.541 1.00 0.00 H
    ATOM 654 HG3 MET E 35 6.847 −40.826 3.644 1.00 0.00 H
    ATOM 655 HE1 MET E 35 7.428 −41.752 5.989 1.00 0.00 H
    ATOM 656 HE2 MET E 35 5.802 −41.284 6.601 1.00 0.00 H
    ATOM 657 HE3 MET E 35 6.437 −42.932 6.909 1.00 0.00 H
    ATOM 658 N MET E 35 2.966 −37.423 2.450 1.00 0.00 N
    ATOM 659 CA MET E 35 1.879 −36.446 2.512 1.00 0.00 C
    ATOM 660 C VAL E 36 0.537 −37.124 3.120 1.00 0.00 C
    ATOM 661 O VAL E 36 −0.166 −37.757 2.424 1.00 0.00 O
    ATOM 662 CB VAL E 36 1.582 −35.759 1.142 1.00 0.00 C
    ATOM 663 CG1 VAL E 36 0.490 −34.657 1.256 1.00 0.00 C
    ATOM 664 CG2 VAL E 36 2.873 −35.169 0.501 1.00 0.00 C
    ATOM 665 HN VAL E 36 3.162 −37.755 1.533 1.00 0.00 H
    ATOM 666 HA VAL E 36 2.204 −35.668 3.190 1.00 0.00 H
    ATOM 667 HB VAL E 36 1.193 −36.535 0.443 1.00 0.00 H
    ATOM 668 HG11 VAL E 36 −0.456 −35.076 1.643 1.00 0.00 H
    ATOM 669 HG12 VAL E 36 0.814 −33.845 1.946 1.00 0.00 H
    ATOM 670 HG13 VAL E 36 0.286 −34.209 0.270 1.00 0.00 H
    ATOM 671 HG21 VAL E 36 2.633 −34.629 −0.439 1.00 0.00 H
    ATOM 672 HG22 VAL E 36 3.379 −34.468 1.197 1.00 0.00 H
    ATOM 673 HG23 VAL E 36 3.597 −35.972 0.250 1.00 0.00 H
    ATOM 674 N GLY E 37 0.443 −36.995 4.463 1.00 0.00 N
    ATOM 675 CA GLY E 37 −0.684 −37.578 5.203 1.00 0.00 C
    ATOM 676 C GLY E 37 −2.018 −36.872 5.009 1.00 0.00 C
    ATOM 677 O GLY E 37 −2.704 −36.541 5.983 1.00 0.00 O
    ATOM 678 HN GLY E 37 1.129 −36.501 5.000 1.00 0.00 H
    ATOM 679 HA2 GLY E 37 −0.802 −38.602 4.875 1.00 0.00 H
    ATOM 680 HA3 GLY E 37 −0.432 −37.512 6.252 1.00 0.00 H
    ATOM 681 N GLY E 38 −2.423 −36.650 3.731 1.00 0.00 N
    ATOM 682 CA GLY E 38 −3.565 −35.839 3.306 1.00 0.00 C
    ATOM 683 C GLY E 38 −4.940 −36477 3.363 1.00 0.00 C
    ATOM 684 O GLY E 38 −5.800 −36.135 2.551 1.00 0.00 O
    ATOM 685 HN GLY E 38 −1.826 −37.022 3.007 1.00 0.00 H
    ATOM 686 HA2 GLY E 38 −3.596 −34.961 3.934 1.00 0.00 H
    ATOM 687 HA3 GLY E 38 −3.385 −35.582 2.272 1.00 0.00 H
    ATOM 688 N VAL E 39 −5.190 −37.396 4.335 1.00 0.00 H
    ATOM 689 CA VAL E 39 −6.488 −38.012 4.618 1.00 0.00 C
    ATOM 690 C VAL E 39 −7.270 −37.008 5.483 1.00 0.00 C
    ATOM 691 O VAL E 39 −7.001 −36.850 6.682 1.00 0.00 O
    ATOM 692 CB VAL E 39 −6.406 −39.429 5.261 1.00 0.00 C
    ATOM 693 CG1 VAL E 39 −7.819 −40.027 5.516 1.00 0.00 C
    ATOM 694 CG2 VAL E 39 −5.568 −40.403 4.385 1.00 0.00 C
    ATOM 695 HN VAL E 39 −4.429 −37.670 4.917 1.00 0.00 H
    ATOM 696 HA VAL E 39 −7.000 −38.124 3.670 1.00 0.00 H
    ATOM 697 HB VAL E 39 −5.895 −39.340 6.247 1.00 0.00 H
    ATOM 698 HG11 VAL E 39 −8.406 −39.393 6.213 1.00 0.00 H
    ATOM 699 HG12 VAL E 39 −8.388 −40.127 4.566 1.00 0.00 H
    ATOM 700 HG13 VAL E 39 −7.733 −41.035 5.974 1.00 0.00 H
    ATOM 701 HG21 VAL E 39 −5.510 −41.402 4.867 1.00 0.00 H
    ATOM 702 HG22 VAL E 39 −6.027 −40.529 3.382 1.00 0.00 H
    ATOM 703 HG23 VAL E 39 −4.530 −40.034 4.251 1.00 0.00 H
    ATOM 704 N VAL E 40 −8.241 −36.292 4.858 1.00 0.00 N
    ATOM 705 CA VAL E 40 −9.103 −35.281 5.475 1.00 0.00 C
    ATOM 706 C VAL E 40 −10.416 −35.968 5.885 1.00 0.00 C
    ATOM 707 O VAL E 40 −11.125 −36.545 5.054 1.00 0.00 O
    ATOM 708 CB VAL E 40 −9.351 −34.024 4.589 1.00 0.00 C
    ATOM 709 CG1 VAL E 40 −10.227 −32.964 5.318 1.00 0.00 C
    ATOM 710 CG2 VAL E 40 −8.011 −33.379 4.132 1.00 0.00 C
    ATOM 711 HN VAL E 40 −8.399 −36.424 3.882 1.00 0.00 H
    ATOM 712 HA VAL E 40 −8.501 −34.939 −6.371 1.00 0.00 H
    ATOM 713 HB VAL E 40 −9.899 −34.345 3.673 1.00 0.00 H
    ATOM 714 HG11 VAL E 40 −10.381 −32.079 4.663 1.00 0.00 H
    ATOM 715 HG12 VAL E 40 −11.230 −33.369 5.567 1.00 0.00 H
    ATOM 716 HG13 VAL E 40 −9.743 −32.619 6.256 1.00 0.00 H
    ATOM 717 HG21 VAL E 40 −8.207 −32.485 3.502 1.00 0.00 H
    ATOM 718 HG22 VAL E 40 −7.403 −33.066 5.006 1.00 0.00 H
    ATOM 719 HG23 VAL E 40 −7.410 −34.091 3.527 1.00 0.00 H
    ATOM 720 N ILE E 41 −10.751 −35.908 7.203 1.00 0.00 N
    ATOM 721 CA ILE E 41 −11.966 −36.466 7.800 1.00 0.00 C
    ATOM 722 C ILE E 41 −13.051 −35.375 7.786 1.00 0.00 C
    ATOM 723 O ILE E 41 −12.874 −34.293 8.357 1.00 0.00 O
    ATOM 724 CB ILE E 41 −11.750 −37.057 9.220 1.00 0.00 C
    ATOM 725 CG1 ILE E 41 −10.532 −38.054 9.311 1.00 0.00 C
    ATOM 726 CG2 ILE E 41 −13.054 −37.722 9.772 1.00 0.00 C
    ATOM 727 CD1 ILE E 41 −10.581 −39.309 8.417 1.00 0.00 C
    ATOM 728 HN ILE E 41 −10.162 −35.435 7.484 1.00 0.00 H
    ATOM 729 HA ILE E 41 −12.287 −37.278 7.159 1.00 0.00 H
    ATOM 730 HB ILE E 41 −11.507 −36.219 9.904 1.00 0.00 H
    ATOM 731 HG12 ILE E 41 −9.601 −37.494 9.073 1.00 0.00 H
    ATOM 732 HG13 ILE E 41 −10.437 −38.384 10.370 1.00 0.00 H
    ATOM 733 HG21 ILE E 41 −13.880 −36.983 9.847 1.00 0.00 H
    ATOM 734 HG22 ILE E 41 −13.387 −38.557 9.121 1.00 0.00 H
    ATOM 735 HG23 ILE E 41 −12.874 −38.127 10.790 1.00 0.00 H
    ATOM 736 HD11 ILE E 41 −9.674 −39.929 8.578 1.00 0.00 H
    ATOM 737 HD12 ILE E 41 −11.470 −39.931 8.656 1.00 0.00 H
    ATOM 738 HD13 ILE E 41 −10.623 −39.030 7.343 1.00 0.00 H
    ATOM 739 N ALA E 42 −14.200 −35.665 7.117 1.00 0.00 N
    ATOM 740 CA ALA E 42 −15.376 −34.818 7.035 1.00 0.00 C
    ATOM 741 C ALA E 42 −16.315 −35.048 8.212 1.00 0.00 C
    ATOM 742 H ALA E 42 −16.713 −34.272 8.647 1.00 0.00 H
    ATOM 743 O ALA E 42 −16.591 −35.175 8.635 1.00 0.00 O
    ATOM 744 CB ALA E 42 −16.139 −35.030 5.708 1.00 0.00 C
    ATOM 745 HN ALA E 42 −14.299 −36.539 6.652 1.00 0.00 H
    ATOM 746 HA ALA E 42 −15.047 −33.788 7.079 1.00 0.00 H
    ATOM 747 HB1 ALA E 42 −15.467 −34.837 4.844 1.00 0.00 H
    ATOM 748 HB2 ALA E 42 −16.519 −36.071 5.631 1.00 0.00 H
    ATOM 749 HB3 ALA E 42 −17.002 −34.335 5.635 1.00 0.00 H
    TER 750 ALA 42 1.00 0.00
    HETATM 751 C * 53 9.744 −25.831 −3.543 1.00 0.00 C
    HETATM 752 H * 53 9.721 −26.606 −2.793 1.00 0.00 H
    HETATM 753 C * 53 10.953 −25.485 −4.191 1.00 0.00 C
    HETATM 754 C * 53 12.240 −26.077 −3.862 1.00 0.00 C
    HETATM 755 H * 53 13.086 −25.808 −4.512 1.00 0.00 H
    HETATM 756 C * 53 12.487 −26.907 −2.815 1.00 0.00 C
    HETATM 757 H * 53 11.648 −27.207 −2.169 1.00 0.00 H
    HETATM 758 C * 53 13.793 −27.472 −2.513 1.00 0.00 C
    HETATM 759 C * 53 10.919 −24.274 −5.175 1.00 0.00 C
    HETATM 760 H * 53 11.823 −24.190 −5.694 1.00 0.00 H
    HETATM 761 C * 53 9.724 −23.811 −5.478 1.00 0.00 C
    HETATM 762 O * 53 9.702 −22.789 −6.448 1.00 0.00 O
    HETATM 763 H * 53 10.584 −22.705 −5.812 1.00 0.00 H
    HETATM 764 C * 53 8.547 −24.143 −4.805 1.00 0.00 C
    HETATM 765 H * 53 7.632 −23.614 −5.026 1.00 0.00 H
    HETATM 766 C * 53 8.557 −25.151 −3.839 1.00 0.00 C
    HETATM 767 O * 53 7.372 −25.430 −3.127 1.00 0.00 O
    HETATM 768 H * 53 7.560 −26.138 −2.437 1.00 0.00 H
    HETATM 769 C * 53 15.002 −26.920 −3.000 1.00 0.00 C
    HETATM 770 H * 53 15.006 −26.007 −3.577 1.00 0.00 H
    HETATM 771 C * 53 10.227 −27.545 −2.744 1.00 0.00 C
    HETATM 772 H * 53 17.133 −27.111 −3.140 1.00 0.00 H
    HETATM 773 C * 53 16.268 −28.721 −1.992 1.00 0.00 C
    HETATM 774 O * 53 17.505 −29.359 −1.788 1.00 0.00 O
    HETATM 775 H * 53 18.150 −28.868 −2.305 1.00 0.00 H
    HETATM 776 C * 53 15.089 −29.261 −1.468 1.00 0.00 C
    HETATM 777 H * 53 15.123 −30.135 −0.878 1.00 0.00 H
    HETATM 778 C * 53 13.859 −28.637 −1.721 1.00 0.00 C
    HETATM 779 H * 53 12.960 −29.082 −1.315 1.00 0.00 H
    CONECT 5 22
    CONECT 24 38
    CONECT 40 58
    CONECT 60 78
    CONECT 80 88
    CONECT 90 103
    CONECT 105 115
    CONECT 117 131
    CONECT 133 138
    CONECT 140 149
    CONECT 151 163
    CONECT 165 185
    CONECT 187 192
    CONECT 194 202
    CONECT 204 221
    CONECT 223 240
    CONECT 242 247
    CONECT 249 266
    CONECT 268 283
    CONECT 285 299
    CONECT 301 306
    CONECT 308 313
    CONECT 315 329
    CONECT 331 345
    CONECT 347 364
    CONECT 380 397
    CONECT 399 413
    CONECT 415 433
    CONECT 435 453
    CONECT 455 463
    CONECT 465 478
    CONECT 480 490
    CONECT 492 506
    CONECT 508 513
    CONECT 516 524
    CONECT 526 538
    CONECT 540 560
    CONECT 562 567
    CONECT 269 577
    CONECT 579 596
    CONECT 298 615
    CONECT 617 622
    CONECT 624 641
    CONECT 643 658
    CONECT 660 674
    CONECT 676 681
    CONECT 683 688
    CONECT 690 704
    CONECT 706 720
    CONECT 722 739
    CONECT 751 752 753 766
    CONECT 753 754 759
    CONECT 754 755 756
    CONECT 756 757 758
    CONECT 758 769 778
    CONECT 759 760 761
    CONECT 761 762 764
    CONECT 762 763
    CONECT 764 765 765
    CONECT 766 767
    CONECT 767 768
    CONECT 769 770 771
    CONECT 771 772 773
    CONECT 773 774 776
    CONECT 774 775
    CONECT 776 777 778
    CONECT 778 779
    END
  • The present example is further evidence that pseudo-crystal structures described in the application have utility and may be used for virtual (in silico) screening in Alzheimer Disease, and, possibly, related disorders, e.g., Parkinson's Disease.
  • Example 5
  • Using the methods described in the applications, many compounds in the following Table were identified and predicted to have an anti-amyloid activity at Aβ and/or A-Syn amyloid proteins. Some of these compounds were synthesized and some of the compounds were obtained from commercial sources.
  • The anti-amyloid activity of these compounds was evaluated by ThT aggregation assays of Aβ 1-40 and alpha synuclein, as set forth below. The column “A-Syn” below is a functional ThT aggregation assay of alpha synuclein.
  • Aβ - ThT
    Structure (IC50/μM) A-syn
    Figure US20160283652A1-20160929-C00012
    >20
    Figure US20160283652A1-20160929-C00013
    >20
    Figure US20160283652A1-20160929-C00014
    >20
    Figure US20160283652A1-20160929-C00015
    >20
    Figure US20160283652A1-20160929-C00016
    4.03
    Figure US20160283652A1-20160929-C00017
    2.7
    Figure US20160283652A1-20160929-C00018
    >20
    Figure US20160283652A1-20160929-C00019
    >20
    Figure US20160283652A1-20160929-C00020
    >20
    Figure US20160283652A1-20160929-C00021
    2.1
    Figure US20160283652A1-20160929-C00022
    1.4
    Figure US20160283652A1-20160929-C00023
    >20
    Figure US20160283652A1-20160929-C00024
    Figure US20160283652A1-20160929-C00025
    >20
    Figure US20160283652A1-20160929-C00026
    >20
    Figure US20160283652A1-20160929-C00027
    >20
    Figure US20160283652A1-20160929-C00028
    >20
    Figure US20160283652A1-20160929-C00029
    >100
    Figure US20160283652A1-20160929-C00030
    >20
    Figure US20160283652A1-20160929-C00031
    >20
    Figure US20160283652A1-20160929-C00032
    >100
    Figure US20160283652A1-20160929-C00033
    >100
    Figure US20160283652A1-20160929-C00034
    20-50
    Figure US20160283652A1-20160929-C00035
    Figure US20160283652A1-20160929-C00036
    10-20 51
    Figure US20160283652A1-20160929-C00037
    9.0 11.61
    Figure US20160283652A1-20160929-C00038
    0.85 6.34
    Figure US20160283652A1-20160929-C00039
    2.5 142
    Figure US20160283652A1-20160929-C00040
    25.8
    Figure US20160283652A1-20160929-C00041
    >50
    Figure US20160283652A1-20160929-C00042
    >50
    Figure US20160283652A1-20160929-C00043
    >50
    Figure US20160283652A1-20160929-C00044
    >50
    Figure US20160283652A1-20160929-C00045
    8.82
    Figure US20160283652A1-20160929-C00046
    >50
    Figure US20160283652A1-20160929-C00047
    23.2
    Figure US20160283652A1-20160929-C00048
    >50
    Figure US20160283652A1-20160929-C00049
    >50
    Figure US20160283652A1-20160929-C00050
    INACTIVE
    Figure US20160283652A1-20160929-C00051
    23
    Figure US20160283652A1-20160929-C00052
    >50
    Figure US20160283652A1-20160929-C00053
    8.4
    Figure US20160283652A1-20160929-C00054
    39
    Figure US20160283652A1-20160929-C00055
    177.5
    Figure US20160283652A1-20160929-C00056
    9.64 5
    Figure US20160283652A1-20160929-C00057
    44
    Figure US20160283652A1-20160929-C00058
    25 4
    Figure US20160283652A1-20160929-C00059
    100
    Figure US20160283652A1-20160929-C00060
    41.9
    Figure US20160283652A1-20160929-C00061
    184
    Figure US20160283652A1-20160929-C00062
    152
    Figure US20160283652A1-20160929-C00063
    42
    Figure US20160283652A1-20160929-C00064
    217
    Figure US20160283652A1-20160929-C00065
    1035
    Figure US20160283652A1-20160929-C00066
    631
    Figure US20160283652A1-20160929-C00067
    189
    Figure US20160283652A1-20160929-C00068
    ND
    Figure US20160283652A1-20160929-C00069
    84
    Figure US20160283652A1-20160929-C00070
    196
    Figure US20160283652A1-20160929-C00071
    265
    Figure US20160283652A1-20160929-C00072
    66.2
    Figure US20160283652A1-20160929-C00073
    63
    Figure US20160283652A1-20160929-C00074
    174
    Figure US20160283652A1-20160929-C00075
    18
    Figure US20160283652A1-20160929-C00076
    105
    Figure US20160283652A1-20160929-C00077
    1.47 8
    Figure US20160283652A1-20160929-C00078
    5734
    Figure US20160283652A1-20160929-C00079
    14935
    Figure US20160283652A1-20160929-C00080
    23
    Figure US20160283652A1-20160929-C00081
    544
    Figure US20160283652A1-20160929-C00082
    975
    Figure US20160283652A1-20160929-C00083
    92
    Figure US20160283652A1-20160929-C00084
    1.9
    Figure US20160283652A1-20160929-C00085
    361
    Figure US20160283652A1-20160929-C00086
    21.7
    Figure US20160283652A1-20160929-C00087
    9.23 10
    Figure US20160283652A1-20160929-C00088
    27 15
    Figure US20160283652A1-20160929-C00089
    360
    Figure US20160283652A1-20160929-C00090
    428
    Figure US20160283652A1-20160929-C00091
    539
    Figure US20160283652A1-20160929-C00092
    26.6 45
    Figure US20160283652A1-20160929-C00093
    106.6
    Figure US20160283652A1-20160929-C00094
    148
    Figure US20160283652A1-20160929-C00095
    3.5 ~7
    Figure US20160283652A1-20160929-C00096
    15.5
    Figure US20160283652A1-20160929-C00097
    7.8
    Figure US20160283652A1-20160929-C00098
    2.29
    Figure US20160283652A1-20160929-C00099
    90
    Figure US20160283652A1-20160929-C00100
    54.4
    Figure US20160283652A1-20160929-C00101
    238.4
  • In the preceding specification, the invention has been described with reference to specific exemplary embodiments and examples thereof. It will, however, be evident that various modifications and changes may be made thereto without departing from the broader spirit and scope of the invention as set forth in the claims that follow. The specification and drawings are accordingly to be regarded in an illustrative manner rather than a restrictive sense.

Claims (11)

What is claimed is:
1. A method of creating non-crystallographic models of amyloid protein aggregation comprising:
applying molecular mechanics and/or dynamics to a three-dimensional model comprising a model of crystallized amyloid protein, a model of multiply anti-amyloid compound bound in the pocket of the crystallized amyloid protein, and a candidate model of an uncrystallized amyloid protein, such that a pocket on the uncrystallized amyloid protein is formed around the multiply anti-amyloid compound;
extracting the model of uncrystallized amyloid protein from said model; and
validating said model of uncrystallized amyloid protein with respect to experimental data so as to confirm its utility as a non-crystallographic model of amyloid protein aggregation.
2. A non-crystallographic model of amyloid protein aggregation comprising:
a three-dimensional model of a monomeric amyloid peptide;
and a three-dimensional model of an amyloid protein consisting of one or more amyloid peptides, said amyloid protein model to be positioned with respect to said amyloid peptide model such that it forms a pocket in conjunction with said amyloid peptide model, such that a candidate compound may be inserted into said pocket thereby modulating amyloid aggregation.
3. The non-crystallographic model of amyloid protein aggregation of claim 2, wherein said amyloid peptide model and said amyloid protein model are both composed of beta-amyloid protein or an amyloid-forming fragment thereof.
4. The non-crystallographic model of amyloid protein aggregation of claim 2, wherein said amyloid peptide model is substantially SEQ ID: 1.
5. The non-crystallographic model of amyloid protein aggregation of claim 4, wherein said amyloid protein model is substantially SEQ ID: 2.
6. The non-crystallographic model of amyloid protein aggregation of claim 5, wherein said amyloid peptide model is further positioned with respect to said amyloid protein model such that:
the amino acid Val at position 8 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2;
the amino acid Gly at position 9 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2;
the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Gly at position 9 of SEQ ID: 2;
the amino acid Ser at position 10 of SEQ ID: 1 interacts with the amino acid Ser at position 10 of SEQ ID:2;
and the amino acid Lys at position 12 of SEQ ID: 1 interacts with the amino acid Glu at position 6 of SEQ ID: 2.
7. The non-crystallographic model of amyloid protein aggregation of claim 5, wherein said amyloid peptide model is further positioned with respect to said amyloid protein model substantially in the orientation shown in FIG. 2.
8. A method of identifying compounds that modulate amyloid aggregation comprising the steps of:
constructing the non-crystallographic model of amyloid protein aggregation according to claim 7 in a computer modeling program;
selecting a list of candidate compounds;
constructing said candidate compounds in a computer modeling program;
performing an iterative docking and scoring of all candidate compounds, by means of docking each candidate compound into a pocket formed by said model and scoring each candidate compound to reflect its degree of complementarity with respect to said pocket; and
identifying compounds that modulate amyloid aggregation or better modulate amyloid aggregation by reference to a score cutoff that substantially distinguishes active compounds from inactive compounds, or more active compounds from less active compounds, respectively.
9. A method of improving the potency of a compound known to modulate amyloid aggregation according to claim 8, wherein the list of candidate compounds includes both the compound known to be active and analogs of said compound, and the score cutoff is that score which belongs to the compound known to be active.
10. The non-crystallographic model of amyloid protein aggregation of claim 1, wherein said amyloid peptide model and said amyloid protein model are both composed of alpha-synuclein protein or an amyloid-forming fragment thereof.
11. The non-crystallographic model of amyloid protein aggregation of claim 1, wherein said amyloid peptide model and said amyloid protein model are both composed of tau protein or an amyloid-forming fragment thereof.
US15/155,643 2008-08-29 2016-05-16 Methods for identifying inhibitors of amyloid protein aggregation Abandoned US20160283652A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/155,643 US20160283652A1 (en) 2008-08-29 2016-05-16 Methods for identifying inhibitors of amyloid protein aggregation
US16/172,289 US11568956B2 (en) 2008-08-29 2018-10-26 Methods for identifying inhibitors of amyloid protein aggregation

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US9284508P 2008-08-29 2008-08-29
US9282608P 2008-08-29 2008-08-29
US54985109A 2009-08-28 2009-08-28
US201313872234A 2013-04-29 2013-04-29
US15/155,643 US20160283652A1 (en) 2008-08-29 2016-05-16 Methods for identifying inhibitors of amyloid protein aggregation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US201313872234A Continuation 2008-08-29 2013-04-29

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/172,289 Continuation US11568956B2 (en) 2008-08-29 2018-10-26 Methods for identifying inhibitors of amyloid protein aggregation

Publications (1)

Publication Number Publication Date
US20160283652A1 true US20160283652A1 (en) 2016-09-29

Family

ID=56975445

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/155,643 Abandoned US20160283652A1 (en) 2008-08-29 2016-05-16 Methods for identifying inhibitors of amyloid protein aggregation
US16/172,289 Active 2032-07-21 US11568956B2 (en) 2008-08-29 2018-10-26 Methods for identifying inhibitors of amyloid protein aggregation

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/172,289 Active 2032-07-21 US11568956B2 (en) 2008-08-29 2018-10-26 Methods for identifying inhibitors of amyloid protein aggregation

Country Status (1)

Country Link
US (2) US20160283652A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10464901B2 (en) * 2017-09-26 2019-11-05 Unist(Ulsan National Institute Of Science And Technology) Composition including benzene diamine derivative for preventing or treating degenerative brain diseases

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5981168A (en) 1998-05-15 1999-11-09 The University Of British Columbia Method and composition for modulating amyloidosis
US20070010573A1 (en) 2003-06-23 2007-01-11 Xianqi Kong Methods and compositions for treating amyloid-related diseases
DK1656361T3 (en) 2003-08-11 2008-05-05 Hoffmann La Roche Piperazine with OR-substituted phenyl group and its use as GLYT1 inhibitors
WO2008157425A2 (en) 2007-06-14 2008-12-24 The Regents Of The University Of California Compounds for inhibiting protein aggregation, and methods for making and using them

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10464901B2 (en) * 2017-09-26 2019-11-05 Unist(Ulsan National Institute Of Science And Technology) Composition including benzene diamine derivative for preventing or treating degenerative brain diseases

Also Published As

Publication number Publication date
US11568956B2 (en) 2023-01-31
US20190050526A1 (en) 2019-02-14

Similar Documents

Publication Publication Date Title
Zou et al. Norepinephrine inhibits Alzheimer’s amyloid-β peptide aggregation and destabilizes amyloid-β protofibrils: A molecular dynamics simulation study
Wang et al. Inhibition of amyloid-β aggregation in Alzheimer's disease
Doig et al. Inhibition of protein aggregation and amyloid formation by small molecules
Ruzza et al. Ceftriaxone blocks the polymerization of α-synuclein and exerts neuroprotective effects in vitro
Barale et al. Molecular insights into destabilization of Alzheimer’s Aβ protofibril by arginine containing short peptides: a molecular modeling approach
McLendon et al. Proteotoxicity and cardiac dysfunction
Lemkul et al. Morin inhibits the early stages of amyloid β-peptide aggregation by altering tertiary and quaternary interactions to produce “off-pathway” structures
Richman et al. In vitro and mechanistic studies of an antiamyloidogenic self-assembled cyclic D, L-α-peptide architecture
Lao et al. Molecular dynamics simulations reveal the inhibitory mechanism of dopamine against human islet amyloid polypeptide (hIAPP) aggregation and its destabilization effect on hIAPP protofibrils
US8754034B2 (en) Structure-based design of peptide inhibitors of amyloid fibrillation
Otzen Amyloid fibrils and prefibrillar aggregates: molecular and biological properties
Neddenriep et al. Short peptides as inhibitors of amyloid aggregation
CA2530381C (en) Combination product comprising an antagonist or inverse agonist of histamine receptor h&lt;sb&gt;3 &lt;/sb&gt;and an antipsychotic or antidepressant agent, and use thereof for the preparation of a medicament that prevents the adverse effects of psychotropic drugs
Matthes et al. Resolving the atomistic modes of Anle138b inhibitory action on peptide oligomer formation
Liang et al. Modulation of amyloid-β42 conformation by small molecules through nonspecific binding
Chen et al. A comprehensive insight into the mechanisms of dopamine in disrupting Aβ protofibrils and inhibiting Aβ aggregation
Vijayan et al. Human glutaminyl cyclase: Structure, function, inhibitors and involvement in Alzheimer’s disease
US11568956B2 (en) Methods for identifying inhibitors of amyloid protein aggregation
CN107260726A (en) Composition and method for treating nerve degenerative diseases
US11214540B2 (en) Compositions for treating neurodegenerative diseases
Dong et al. N-Terminus Binding Preference for Either Tanshinone or Analogue in Both Inhibition of Amyloid Aggregation and Disaggregation of Preformed Amyloid Fibrils Toward Introducing a Kind of Novel Anti-Alzheimer Compounds
Ortet et al. Recapitulating the binding affinity of Nrf2 for KEAP1 in a cyclic heptapeptide, guided by NMR, X-ray crystallography, and machine learning
Zhong et al. Amyloid fibril formation of Arctic amyloid-β 1–42 peptide is efficiently inhibited by the BRICHOS domain
US8937091B2 (en) Inhibitor of protein crosslinking and use of the same
Parikh et al. Molecular mechanisms of Alzheimer’s biomarker FDDNP binding to Aβ amyloid fibril

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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