US20230102284A1 - Tethering cysteine residues using cyclic disulfides - Google Patents

Tethering cysteine residues using cyclic disulfides Download PDF

Info

Publication number
US20230102284A1
US20230102284A1 US18/057,854 US202218057854A US2023102284A1 US 20230102284 A1 US20230102284 A1 US 20230102284A1 US 202218057854 A US202218057854 A US 202218057854A US 2023102284 A1 US2023102284 A1 US 2023102284A1
Authority
US
United States
Prior art keywords
protein
compound
alkyl
disease
formula
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.)
Pending
Application number
US18/057,854
Inventor
Jeffrey N. Agar
Joseph Salisbury
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.)
Brandeis University
Original Assignee
Brandeis University
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 Brandeis University filed Critical Brandeis University
Priority to US18/057,854 priority Critical patent/US20230102284A1/en
Publication of US20230102284A1 publication Critical patent/US20230102284A1/en
Assigned to BRANDEIS UNIVERSITY reassignment BRANDEIS UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AGAR, JEFFREY, SALISBURY, Joseph
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/095Sulfur, selenium, or tellurium compounds, e.g. thiols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/385Heterocyclic compounds having sulfur as a ring hetero atom having two or more sulfur atoms in the same ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0089Oxidoreductases (1.) acting on superoxide as acceptor (1.15)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1025Acyltransferases (2.3)
    • C12N9/104Aminoacyltransferases (2.3.2)
    • C12N9/1044Protein-glutamine gamma-glutamyltransferase (2.3.2.13), i.e. transglutaminase or factor XIII
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/78Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5)
    • C12N9/80Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5) acting on amide bonds in linear amides (3.5.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y115/00Oxidoreductases acting on superoxide as acceptor (1.15)
    • C12Y115/01Oxidoreductases acting on superoxide as acceptor (1.15) with NAD or NADP as acceptor (1.15.1)
    • C12Y115/01001Superoxide dismutase (1.15.1.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y203/00Acyltransferases (2.3)
    • C12Y203/02Aminoacyltransferases (2.3.2)
    • C12Y203/02013Protein-glutamine gamma-glutamyltransferase (2.3.2.13), i.e. transglutaminase or factor XIII
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • One third of therapeutic molecules form covalent bonds with their targets.
  • These electrophilic drugs generally bond to a nucleophilic amino acid, often serine or cysteine, on a target protein.
  • Aspirin and penicillin (and their many derivatives) acylate serines and numerous drugs form covalent bonds with specific cysteines.
  • These therapeutic agents are effective despite the potential for off-target reactions with hundreds of highly reactive, nucleophilic residues, which are often required for the function of essential proteins.
  • a worst case scenario for reaction with the “wrong” nucleophile is nerve gases (e.g., Sarin, intravenous LD 50 ⁇ 30 ⁇ g/kg), which covalently modify the active site serine of acetylcholine esterase.
  • the antithrombosis factors clopidogrel/PlavixTM, ticlopide/Ticlid, etc. are also prodrugs. Activation by cytochrome P450 enzymes results in the scission of a ring carbon-sulfur bond, creating a sulfhydryl group that can then form a disulfide bond with its target cysteine on the adenosine diphosphate (ADP) chemoreceptor P2Y 12 . In addition to increased specificity for its target, which it permanently inactivates, the active metabolite has improved plasma protein binding characteristics.
  • the thrombosis drugs had their beginnings in functional assays, and inevitably animal studies, because the active metabolite is not produced in most cell-based assays. Both their mechanism of action and target were unknown at the time of discovery.
  • Dacomitinib, afatinib, and neratinib are EGFR kinase inhibitors with a high-affinity, nucleotide-analogue moiety that reversibly binds the ATP-binding pockets of numerous kinases and a second moiety designed to covalently bond with a non-conserved cysteine (present in EGFR but not its homologues).
  • the electrophilic moiety is purposefully a low-reactivity acrylamide to minimize off-target reactions.
  • a related chemical approach used low-reactivity, acrylamide-based, electrophiles to target non-conserved (in humans) and non-catalytic-residue cysteine of the HCV NS3/4A viral protease (HCVP).
  • HCV NS3/4A viral protease HCV NS3/4A viral protease
  • the invention relates to a method comprising the step of contacting a compound of Formula I or a compound of Formula II with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, wherein the first Protein X comprises a first cysteine residue; the second Protein X comprises a second cysteine residue; the compound of Formula I is
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ;
  • n is 0, 1, 2, 3, or 4;
  • R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, —OMs, —OTs, —OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and the compound of Formula II is
  • R′′ is —H, alkyl, or aryl, or both R′′, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo and wherein the first and the second Protein X are selected from Table 1.
  • the invention relates to a method comprising the step of contacting a compound with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, wherein the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% homologous or identical to any one of the Protein X listed in Table 1 (in preferred embodiments, the first Protein X and the second Protein X are derived from the same Protein X); and the compound is a compound of Formula I
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ; n is 0, 1, 2, 3, or 4; and R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl.
  • the invention relates to a method comprising the step of contacting a compound with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, wherein the first Protein X and the second Protein X is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% homologous or identical to any one of the Protein X listed in Table 1 (in preferred embodiments, the first Protein X and the second Protein X are derived from the same Protein X); and the compound is a compound of Formula II
  • R′′ is —H, alkyl, or aryl, or both R′′, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • the invention relates to a method of treating or preventing a Disease Y listed in Table 1 for each Protein X, comprising the step of administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or a compound of Formula II, wherein the compound of Formula I is
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ; n is 0, 1, 2, 3, or 4; and R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and the compound of Formula II is
  • R′′ is —H, alkyl, or aryl, or both R′′, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • the invention relates to a method of treating or preventing a Disease Y listed in Table 1 for each Protein X, comprising the step of administering to a subject in need thereof a therapeutically effective amount of a stabilized analogue of Protein X, wherein said analogue has a tertiary structure and comprises a first monomer of Protein X and a second monomer of the Protein X; wherein the first monomer of Protein X comprises a first cysteine residue; the second monomer of Protein X comprises a second cysteine residue; the first cysteine residue is connected to the second cysteine residue by a connection; and the connection is a connection of Formula III or a connection of Formula IV, wherein Formula III is
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ; n is 0, 1, 2, 3, or 4; and R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and Formula IV is
  • the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% homologous or identical to any one of the Protein X listed in Table 1.
  • the first Protein X and the second Protein X are derived from the same Protein X.
  • the invention relates to a compound of Formula I
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ; n is 0, 1, 2, 3, or 4; and R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl.
  • One aspect of the invention is a stabilized analogue of Protein X, wherein said analogue has a tertiary structure and comprises a first monomer of Protein X and a second monomer of the Protein X; wherein the first monomer of Protein X comprises a first cysteine residue; the second monomer of Protein X comprises a second cysteine residue; the first cysteine residue is connected to the second cysteine residue by a connection; and the connection is a connection of Formula III or Formula IV:
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ; n is 0, 1, 2, 3, or 4; R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and R′′ is —H, alkyl, or aryl, or both R′′, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% homologous or identical to any one of the Protein X listed in Table 1.
  • the first Protein X and the second Protein X are derived from the same Protein X.
  • these stabilized analogues are used to treat or prevent a Disease Y listed in Table 1 for each Protein X by administering to a subject in need thereof a therapeutically effective amount of a stabilized analogue of Protein X.
  • FIG. 1 depicts a) a potential reaction mechanism where a cyclic disulfide reacts with the cysteine of one monomer and the resulting thiolate can then react with, for example, a hydrogen peroxide-modified thiolate on the other monomer (Isaac, et al. Chemical Science 2012); b) side-chains of Cys53's in the dimer interface of DJ-1 (PDB: 3SF8), demonstrating their close spacing (Premkumar, et al. J. Struct. Biol. 2011, 176, 414). The appearance of Cys111's in the dimer interface of SOD1 is very similar.
  • FIG. 2 depicts a preliminary LC-ESI-IonTrap-MS screen of cyclic disulfides that identifies multiple compounds that form covalent SOD1 dimers.
  • FIG. 3 depicts a preliminary LC-ESI-IonTrap-MS screen of cyclic disulfides that identifies compounds that form covalent DJ-1 dimers at Cys53 and increase the thermal stability of DJ-1.
  • NSC72268 was identified as a specific covalent dimerizer of DJ-1.
  • FIG. 4 depicts examples of cyclic disulfide compounds that are able to form covalent dimers of SOD1.
  • FIG. 5 depicts a dithiol that is able to form covalent dimers of SOD1.
  • FIG. 6 depicts examples of cyclic disulfide compounds that are able to form covalent dimers of DJ-1.
  • the covalent attachment of molecules can be used to affect protein structure and function. Covalently attached molecules can be used to inhibit, promote activity, stabilize, and destabilize proteins and peptides.
  • One problem is encoding adequate specificity in covalent binders for the intended target.
  • the invention relates to chemical tools, cyclic disulfides, which target pairs of cysteine residues and significantly enhance the specificity for pairs of cysteine over lone cysteine residues.
  • cyclic disulfides offer a launching point for compound optimization for novel targets. Whereas previous approaches to covalent modification tended to be devoted to inactivation of an enzyme, cyclic disulfides are also amenable to protein (including enzyme) stabilization.
  • the disease that can be treated using the compounds disclosed herein include, but are not limited to neurodegenerative disease (such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD), cancer, autoimmune disease and/or Celiac disease.
  • neurodegenerative disease such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD
  • cancer such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD
  • autoimmune disease such as, for example, autoimmune disease, and/or Celiac disease.
  • Table 1 shows a consolidated list of protein targets (Protein X) that can form the cyclic disulfide connections to form the stabilized analogues as described in herein.
  • Column 1 lists the “Protein X” of each protein target that can be used to derive the stabilized analogues described herein.
  • Column 2 provides the “Accession Number” corresponding to the protein target listed in Column 1 and connects the Protein X to the publicly available information known about each Protein X. Additionally, the Accession Number is unique identifier used in the art that provides sequence information for each protein listed in Column 1.
  • Column 3 describes “Disease Y” for each Protein X. Each Disease Y has been shown to be associated with each Protein X as demonstrated in Column 4 Here, “Exemplified References” provides literature evidence associating each Protein X with Disease Y.
  • Parkinson van der Merwe C1 et al. “Evidence For A Common Biological Pathway Linking Three disease Parkinson's Disease-Causing Genes: Parkin, PINK1 And DJ-1,” Eur J Neurosci. protein 7) 2015 May; 41(9): 1113-25.
  • Choi et al. “Oxidative damage of DJ-1 is linked to sporadic Parkinson and Alzheimer diseases,” J Biol Chem. 2006 Apr. 21; 281(16): 10916-24. Epub 2006 Mar. 3.
  • Amyotrophic Lev et al. “DJ-1 knockout augments disease severity and shortens survival in a mouse lateral model of ALS,” PLoS One. 2015 Mar. 30; 10(3): e0117190.
  • Parkinson's Bandmann et al. “Sequence of the superoxide dismutase 1 (SOD 1) gene in familial Disease Parkinson's disease,” J Neurol Neurosurg Psychiatry. 1995 July; 59(1): 90-91.
  • Alzheimer's Zemlan et al. “Superoxide dismutase activity in Alzheimer's disease: possible Disease mechanism for paired helical filament formation,” Brain Res. 1989 Jan. 2; 476(1): 160-2.
  • Murakami et al. “SOD1 (Copper/Zinc superoxide dismutase) deficiency drives amyloid beta protein oligomerization and memory loss in mouse model of Alzheimer Disease,” J Biol Chem.
  • Liu et al. “Expression, characterization, and detection of human uridine phosphorylase and identification of variant uridine phosphorolytic activity in selected human tumors,” Cancer Res. 1998 Dec. 1; 58(23): 5418-24.
  • Alzheimer's D'Amelio et al. “Caspase-3 triggers early synaptic dysfunction in a mouse model of Disease Alzheimer's disease,” Nature Neuroscience 14, 69-76 (2011). Doi: 10.1038/nn.2709. S100beta NP_006263 Alzheimer's Griffin et al., “Life-long overexpression of S100beta in Down's syndrome: implications Disease for Alzheimer pathogenesis. Neurobiol Aging. 1998 Sepember-October; 19(5): 401-5. Lambert et al., “Evidence for the association of the S100beta gene with low cognitive performance and dementia in the elderly,” Mol Psychiatry. 2007 September; 12(9): 870-80. Epub 2007 Mar. 6.
  • Gerlai et al. “Abnormal exploratory behavior in transgenic mice carrying multiple copies of the human gene for S100 beta,” J Psychiatry Neurosci. 1995 March; 20(2): 105-12.
  • Royston et al. “Overexpression of S100beta in Down's syndrome: correlation with patient age and with beta-amyloid deposition,” Neuropathol Appl Neurobiol. 1999 October; 25(5): 387-93.
  • Phospholipase A2 AAF09020 Alzheimer's Sanchez-Mejia et al., “Phospholipase A2 and arachidonic acid in Alzheimer's disease,” Disease Biochim Biophys Acta. 2010 August; 1801(8): 784-90.
  • Alzheimer's Mohri et al. “Hematopoietic prostaglandin D synthase and DP1 receptor are selectively Disease upregulated in microglia and astrocytes within senile plaques from human patients and in a mouse model of Alzheimer disease,” J Neuropathol Exp Neurol. 2007 June; 66(6): 469-80. Glutathione AAC51280.1 Cancer; CML; Kassogue et al., “Association of glutathione S-transferase (GSTM1 and GSTT1) genes with S-transferase lung cancer chronic myeloid leukemia,” Springerplus. 2015 May 1; 4: 210. doi: 10.1186/ s40064-015-0966-y. eCollection 2015.
  • Paritala et al. “A continuous spectrophotometric assay for adenosine 5′-phosphosulfate reductase activity with sulfite-selective probes,” Anal Biochem. 2013 Sep. 1; 440(1): 32-9. doi: 10.1016/j.ab.2013.05.007. Epub 2013 May 24. Paritala et al., “Design, synthesis and evaluation of Fe—S targeted adenosine 5′-phosphosulfate reductase inhibitors,” Nucleosides Nucleotides Nucleic Acids. 2015; 34(3): 199-220. doi: 10.1080/15257770.2014.978012.
  • stabilized analogues of Cu/Zn-SOD1 or DJ-1 can be used to treat ALS, Parkinson's and/or Alzheimer's Disease.
  • Amyotrophic lateral sclerosis is a progressive neurodegenerative disease caused by death of motor neurons in the brain and spinal cord.
  • the overall median survival from onset of symptoms ranges between 2-3 years for cases with bulbar onset to 3-5 years for cases with limb onset.
  • Lifetime risk of ALS is 1/400 to 1/1000 with a median annual incidence of 1.89 and a median prevalence of 5.2 per 100,000 each year.
  • riluzole riluzole (Rilutek) prolongs median survival by a mere 2-3 months when taken for an eighteen month duration.
  • novel therapeutic strategies for ALS continue to be crucial.
  • ALS familial
  • SOD1 ubiquitously expressed protein
  • ALS-associated variants of SOD1 are found as monomers in ALS patients and a number of modifications, including loss of Cu or Zn, cleavage of the native, intramolecular disulfide, oxidation, glutathionylation, and fALS-associated mutation, predispose the SOD1 dimer to dissociate.
  • SOD1 dimers contain two cysteine residues at the dimer interface whose sulfhydryl groups are approximately nine angstroms apart. These sulfhydryl groups can be targeted by maleimide cross-linkers which lead to strong stabilization of ALS-associated SOD1 dimers.
  • the invention relates to the discovery of molecules that can cross-link SOD1 dimers in order to fully assess small molecule-mediated covalent dimer formation of SOD1 as a therapeutic strategy for ALS.
  • the progressive neurodegenerative disorder PD is characterized by the loss of dopaminergic neurons in the substania nigra pars compacta and ⁇ -synuclein-rich protein deposits known as Lewy bodies.
  • Biochemical and cell culture analysis of PD-linked variants of DJ-1 suggest a number of mechanisms through which structural defects, including loss of stability and dimer formation, may lead to a loss-of-function that is associated with PD pathogenicity, such as reduced ability to prevent ⁇ -synuclein aggregation, deficiency in oxidative stress-dependent RNA-binding activity, reduced ability to act as a neuroprotective transcriptional co-activator, and increased sensitivity to oxidative stress-induced cell death related to mitochondrial defects.
  • DJ-1 In additional to recessive PD-related mutants of DJ-1 being implicated in disease, analysis of DJ-1 in the frontal cortex of patients with sporadic PD and Alzheimer's disease reveal that acidic isoforms of monomeric DJ-1 and basic isoforms of SDS-resistant dimeric DJ-1 selectively accumulate in these diseases, with DJ-1 irreversibly oxidized by carbonylation as well as by methionine oxidation to methionine sulfone. Over-oxidation of DJ-1 has been found to produce structural destabilization similar to PD-related mutations, suggesting that dysfunctional DJ-1 due to aberrant modifications could be a cause of sporadic neurodegenerative cases.
  • DJ-1 protects against degeneration of nigral dopaminergic neurons in PD rat models involving both 6-hydroxydopamine and rotenone treatment. Viral-mediated DJ-1 overexpression in the MPTP mouse model has also proved efficacious in reducing nigral dopamine neuron loss.
  • DJ-1 histone deacetylase inhibitor
  • phenylbutyrate rescues cells from oxidative stress and mutant ⁇ -synuclein toxicity, as well as protects dopaminergic neurons from MPTP-induced neurotoxicity and prevents age-related motor and cognitive decline in mice with diffuse Lewy body disease.
  • enhancement of DJ-1 activity could serve as a therapeutic strategy in a possibly wide variety of PD cases.
  • in silico methods have been used to identify potential small molecule binding sites on DJ-1 and for identifying small molecules capable of interacting with DJ-1 and modulating its oxidation state that have neuroprotective effects in vivo.
  • Cys53s in DJ-1 are more closely spaced than Cys111s in SOD1, hindering previous attempts to covalently dimerize DJ-1 using maleimide crosslinkers.
  • the cyclic disulfides described herein can covalently dimerize any one of the Protein X listed in Table 1 through their respective closely spaced dimer interface cysteines.
  • a cyclic disulfide could undergo thiol-disulfide exchange with the cysteine of a Protein X, leaving a free thiolate to react with the remaining monomer ( FIG. 1 ).
  • Dithiols might also be capable of forming covalent dimers if the thiol groups are properly spaced and appropriately reactive.
  • the invention relates to a method comprising the step of contacting a compound of Formula I or a compound of Formula II with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, thereby cross-linking the first Protein X to the second Protein X, wherein the first Protein X comprises a first cysteine residue; the second Protein X comprises a second cysteine residue; the compound of Formula I is
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ; n is 0, 1, 2, 3, or 4; and R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and the compound of Formula II is
  • R′′ is —H, alkyl, or aryl, or both R′′, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo and wherein Protein X is selected from Table 1.
  • the invention relates to a method comprising the step of contacting a compound with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, thereby cross-linking the first Protein X to the second Protein X, wherein the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical or homologous to one another and wherein the first Protein X and the second Protein X are any one of the Protein X listed in Table 1; and the compound is a compound of Formula I
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ; n is 0, 1, 2, 3, or 4; and
  • R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl.
  • the invention relates to any one of the aforementioned methods, wherein Y is S.
  • the invention relates to any one of the aforementioned methods, wherein Y is S ⁇ O.
  • the invention relates to any one of the aforementioned methods, wherein Y is S( ⁇ O) 2 .
  • the invention relates to any one of the aforementioned methods, wherein n is 1 or 2.
  • the invention relates to any one of the aforementioned methods, wherein n is 1.
  • the invention relates to any one of the aforementioned methods, wherein n is 2.
  • the invention relates to any one of the aforementioned methods, wherein the compound is selected from the group consisting of
  • the invention relates to a method comprising the step of contacting a compound disclosed herein with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, thereby cross-linking the first Protein X to the second Protein X, wherein the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% homologous or identical to one another; the first Protein X and the second Protein X are any one of the Protein X listed in Table 1; and the compound is a compound of Formula II
  • R′′ is —H, alkyl, or aryl, or both R′′, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • the invention relates to any one of the aforementioned methods, wherein the R′′ form a six-membered ring. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the R′′ form an aromatic ring. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the R′′ form a six-membered aromatic ring.
  • the invention relates to any one of the aforementioned methods, wherein the compound is
  • the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X have at least 95% sequence homology or identity to one another.
  • the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X have at least 98% sequence homology or identity to one another.
  • the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X have at least 99% sequence homology or identity to one another.
  • the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X are wild type of any one of the Protein X listed in Table 1.
  • the invention relates to any one of the aforementioned methods, wherein the method is performed in vitro.
  • the cross-linking of the first and second Protein X with a compound of Formula I or Formula II occurs outside of a patient and then the resultant cross-linked, stabilized analogue of Formula III or Formula IV is administered to the patient in a therapeutically effective amount, such as for example, for enzyme replacement therapy.
  • the cross-linking of the first and the second Protein X can occur in vivo.
  • the molecule represented by Formula I or Formula II is administered to a patient in a therapeutically effective amount to cross-link the first and second Protein X in the patient.
  • the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X are cross-linked within a cell.
  • the invention relates to any one of the aforementioned methods, wherein the method is a method of inhibiting the activity of the first Protein X or the second Protein X.
  • the invention relates to any one of the aforementioned methods, wherein the method is a method of increasing the activity of the first Protein X or a second Protein X.
  • the invention relates to any one of the aforementioned methods, wherein the method is a method of stabilizing the first Protein X or the second Protein X.
  • the invention relates to any one of the aforementioned methods, wherein the method is a method of destabilizing the first Protein X or the second Protein X.
  • the invention relates to a method of treating or preventing a Disease Y listed in Table 1 for each Protein X, comprising the step of administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or a compound of Formula II, wherein the compound of Formula I is
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ; n is 0, 1, 2, 3, or 4; and
  • R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and the compound of Formula II is
  • R′′ is —H, alkyl, or aryl, or both R′′, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and Y is S.
  • the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and Y is S ⁇ O.
  • the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and Y is S( ⁇ O) 2 .
  • the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and n is 1 or 2.
  • the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and n is 1.
  • the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and n is 2.
  • the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and the compound is selected from the group consisting of
  • the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula II; and the R′′ form a six-membered ring. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula II; and the R′′ form an aromatic ring. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula II; and the R′′ form a six-membered aromatic ring.
  • the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula II; and the compound is
  • the invention relates to any one of the aforementioned methods, wherein a Disease Y is a neurodegenerative disease (such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD), cancer, autoimmune disease and/or Celiac disease, or any other Disease Y listed in Table 1.
  • a neurodegenerative disease such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD
  • cancer such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD
  • autoimmune disease and/or Celiac disease or any other Disease Y listed in Table 1.
  • the invention relates to a compound of Formula I
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ; n is 0, 1, 2, 3, or 4; and
  • R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl.
  • the invention relates to any one of the aforementioned compounds, wherein Y is S.
  • the invention relates to any one of the aforementioned compounds, wherein Y is S ⁇ O.
  • the invention relates to any one of the aforementioned compounds, wherein Y is S( ⁇ O) 2 .
  • the invention relates to any one of the aforementioned compounds, wherein n is 1 or 2.
  • the invention relates to any one of the aforementioned compounds, wherein n is 1.
  • the invention relates to any one of the aforementioned compounds, wherein n is 2.
  • the invention relates to any one of the aforementioned compounds, wherein the proviso that the compound is not selected from the group consisting of
  • One aspect of the invention is a stabilized analogue of any one of the Protein X listed in Table 1, wherein said analogue has a tertiary structure and comprises a first Protein X monomer and a second Protein X monomer; wherein the first Protein X monomer comprises a first cysteine residue; the second Protein X monomer comprises a second cysteine residue; the first cysteine residue is connected to the second cysteine residue by a connection; and the connection is a connection of Formula III or Formula IV:
  • Y is S, S ⁇ O, or S( ⁇ O) 2 ;
  • n is 0, 1, 2, 3, or 4;
  • R is independently selected from the group consisting of —H, —OH, —NH 2 , —NHR′, —N(R′) 2 , alkyl, -OMs, -OTs, -OTf, and —CO 2 H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and R′′ is —H, alkyl, or aryl, or both R′′, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • the present invention relates to any one of the aforementioned analogues, wherein the tertiary structure is substantially the same as the wild-type of any one of the Protein X listed in Table 1.
  • the present invention relates to any one of the aforementioned analogues, wherein the sequence homology or identity of the first Protein X monomer and the second Protein X monomer is greater than or equal to about 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100.
  • the present invention relates to any one of the aforementioned analogues, wherein the first Protein X monomer and the second Protein X monomer have substantially the same amino acid sequence.
  • the present invention relates to any one of the aforementioned analogues, wherein the first Protein X monomer of said analogue is the wild-type sequence or comprises one, two, three, four, five, six, seven, eight, nine, or ten point mutations as compared to the wild type sequence.
  • the present invention relates to any one of the aforementioned analogues, wherein the second Protein X monomer of said analogue is the wild-type sequence or comprises one, two, three, four, five, six, seven, eight, nine, or ten point mutations as compared to the wild type sequence.
  • the present invention relates to any one of the aforementioned analogues, wherein said analogue retains at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% activity of the wild-type of any one of the Protein X listed in Table 1.
  • the present invention relates to any one of the aforementioned analogues, wherein said analogue retains at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% activity of the wild-type of any one of the Protein X listed in Table 1 up to a temperature of about 75° C.
  • the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization from about 10° C. to about 60° C.
  • the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization from about 20° C. to about 40° C.
  • the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization from about 15° C. to about 25° C.
  • the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization from about 30° C. to about 50° C.
  • the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization about 20° C.
  • the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization about 40° C.
  • analogue refers to a molecule substantially similar in function to any one of the Protein X listed in Table 1 or a fragment thereof.
  • sequence similarity generally refers to the degree of identity or correspondence between different nucleotide sequences of nucleic acid molecules or amino acid sequences of polypeptides that may or may not share a common evolutionary origin (see Reeck et al., supra). Sequence identity can be determined using any of a number of publicly available sequence comparison algorithms, such as BLAST, FASTA, DNA Strider, GCG (Genetics Computer Group, Program Manual for the GCG Package, Version 7, Madison, Wis.), etc.
  • percent (%) amino acid sequence identity or “percent amino acid sequence homology” or “percent (%) identical” as used herein with respect to a reference polypeptide is defined as the percentage of amino acid residues in a candidate polypeptide sequence that are identical with the amino acid residues in the reference polypeptide sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, without considering any conservative substitutions as part of the sequence identity. Alignment for the purpose of determining percent amino acid sequence identity can be achieved by various techniques known in the art, for instance, using publicly available computer software such as ALIGN or Megalign (DNASTAR).
  • an analogue of SOD1 is said to share “substantial homology” with SOD1 if the amino acid sequence of said analogue is at least about 85%, at least about 90%, at least about 95%, or at least about 99% identical to wild-type.
  • the sequences are aligned for optimal comparison purposes.
  • the two sequences are, or are about, of the same length.
  • the percent identity between two sequences can be determined using techniques similar to those described below, with or without allowing gaps. In calculating percent sequence identity, typically exact matches are counted.
  • the determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • a non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, Proc. Natl. Acad. Sci. USA 1990, 87:2264, modified as in Karlin and Altschul, Proc. Natl. Acad. Sci. USA 1993, 90:5873-5877.
  • Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al, J. Mol. Biol. 1990; 215: 403.
  • Gapped BLAST can be utilized as described in Altschul et al, Nucleic Acids Res. 1997, 25:3389.
  • PSI-Blast can be used to perform an iterated search that detects distant relationship between molecules. See Altschul et al. (1997) supra.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • the percent identity between two amino acid sequences is determined using the algorithm of Needleman and Wunsch (J. Mol. Biol. 1970, 48:444-453), which has been incorporated into the GAP program in the GCG software package (Accelrys, Burlington, Mass.; available at accelrys.com on the WorldWideWeb), using either a Blossum 62 matrix or a PAM250 matrix, a gap weight of 16, 14, 12, 10, 8, 6, or 4, and a length weight of 1, 2, 3, 4, 5, or 6.
  • the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package using a NWSgapdna.CMP matrix, a gap weight of 40, 50, 60, 70, or 80, and a length weight of 1, 2, 3, 4, 5, or 6.
  • a particularly preferred set of parameters (and the one that can be used if the practitioner is uncertain about what parameters should be applied to determine if a molecule is a sequence identity or homology limitation of the invention) is using a Blossum 62 scoring matrix with a gap open penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • phrases “pharmaceutically acceptable” is employed herein to refer to those ligands, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals, substantially non-pyrogenic, without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ or portion of the body, to another organ or portion of the body.
  • a pharmaceutically acceptable material, composition or vehicle such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ or portion of the body, to another organ or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, not injurious to the patient, and substantially non-pyrogenic.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum
  • preventing is art-recognized, and when used in relation to Disease Y listed in Table 1 for each Protein X, such as a local recurrence (e.g., pain), a disease such as cancer, a syndrome complex such as heart failure or any other medical condition, is well understood in the art, and includes administration of a composition which reduces the frequency of, or delays the onset of, symptoms of a medical condition in a subject relative to a subject which does not receive the composition.
  • a local recurrence e.g., pain
  • a disease such as cancer
  • a syndrome complex such as heart failure or any other medical condition
  • prevention of cancer includes, for example, reducing the number of detectable cancerous growths in a population of patients receiving a prophylactic treatment relative to an untreated control population, and/or delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population, e.g., by a statistically and/or clinically significant amount.
  • Prevention of an infection includes, for example, reducing the number of diagnoses of the infection in a treated population versus an untreated control population, and/or delaying the onset of symptoms of the infection in a treated population versus an untreated control population.
  • Prevention of pain includes, for example, reducing the magnitude of, or alternatively delaying, pain sensations experienced by subjects in a treated population versus an untreated control population.
  • Prevention of a neurodegenerative disease or disorder includes the reduction and/or slowing down of the appearance of symptoms (e.g., a decrease in cognitive function) associated with the neurodegenerative disease or disorder as compared to subjects in a treated population versus an untreated control population
  • a “therapeutically effective amount” of a compound, e.g., such as a polypeptide or peptide analogue of the present invention, with respect to use in treatment refers to an amount of the polypeptide or peptide analogue of Formula I or Formula II described herein and/or the stabilized analogue of Formula III or Formula IV as described herein in a preparation which, when administered as part of a desired dosage regimen (to a mammal, preferably a human) alleviates a symptom, ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated or the cosmetic purpose, e.g., at a reasonable benefit/risk ratio applicable to any medical treatment.
  • a desired dosage regimen to a mammal, preferably a human
  • prophylactic or therapeutic treatment are art-recognized and include administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • the unwanted condition e.g., disease or other unwanted state of the host animal
  • the term “about” is used to refer to an amount that is approximately, nearly, almost, or in the vicinity of being equal to or is equal to a stated amount, e.g., the state amount plus/minus about 5%, about 4%, about 3%, about 2% or about 1%.
  • patient or “subject” as used herein in reference can encompasses veterinary uses, such as, for example, the testing of a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), rabbit, murine (e.g. a mouse), canine (e.g. a dog), feline (e.g. a cat), equine (e.g. a horse), bovine (e.g., cow) a primate, simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon), an ape (e.g. gorilla, chimpanzee, orangutan, gibbon).
  • a rodent e.g. a guinea pig, a hamster, a rat, a mouse
  • rabbit murine
  • feline e.g. a cat
  • equine e.g. a horse
  • WtSOD1 or wtDJ-1 was incubated with 5-25 mM DTT for approximately 20 minutes and either buffer exchanged using Amicon Ultra-4 centrifugal spin concentrators (MWCo 10K) or using reversed phase chromatography (ZIPTIP, Millipore, Inc). Samples cleaned by ZIPTIPs were also subjected to incubation with 5 mM EDTA. SOD1 samples that were buffer exchanged using Amicon concentrators were exchanged into in HPLC water, whereas ZIPTIP samples were further exchanged after ZIPTIP into PBS, pH 7.4 or HPLC water.
  • DTT-reduced SOD1 or DJ-1 was incubated at a 1:1 (20 ⁇ M:20 ⁇ M or 10 ⁇ M:10 ⁇ M) or 1:3 (20 ⁇ M:60 ⁇ M or 10 ⁇ M:30 ⁇ M) ratio of protein to cross-linker.
  • cross-linkers were used. Cross-linking was achieved by incubating the reaction in either PBS pH 7.4 or water at room temperature for 1 hour. After an hour the reactions were analyzed on a 15% SDS-PAGE gel with a non-cross-linked control, transferred to nitrocellulose membrane and western blotted using a polyclonal antibody to SOD1 or DJ-1. Repeated in triplicate.
  • DTME is a cleavable sulfhydryl-sulfhydryl cross-linking agent. Therefore, a cross-linking reaction containing 1:1 molar ratio of wtSOD1 or wtDJ-1 to DTME was performed at room temperature for one hour. After cross-linking, the reaction was split in half and half of the sample was run in a sample buffer containing DTT (reducing) and the other half in one containing no DTT (non-reducing). These samples along with non-cross-linked controls were then analyzed on a 15% SDS PAGE gel and western blotted as above.
  • MALDI Matrix Assisted Laser Desorption Ionization
  • TOF Time of Flight
  • wtSOD1 or wtDJ-1 was cross-linked as below. After cross-linking, 1 ⁇ L of sample was spotted on a MALDI target containing 1 ⁇ l of matrix, 20 mg/mL sinipic acid, and analyzed on a Bruker Daltonics Microflex. The MALDI was calibrated each time using a high molecular weight protein calibration standard, Protein Calibration Standard I (Bruker Daltonics). The MALDI-TOF was operated in linear mode using a laser power of between 72-90%. MALDI-TOF spectra were of cross-linked and non-cross-linked samples were analyzed using FlexAnalysis software (Bruker Daltonics). Repeated in triplicate.
  • the changes in mass of the covalently linked SOD1 dimers observed suggest both 1,2-dithiolane-4,4-dimethanol and 1-oxo-1,2-dithiane ( FIG. 2 a ) are capable of covalently dimerizing SOD1.
  • 1-oxo-1,2-dithiane was able to cross-link the majority of SOD1 monomers in the sample ( FIG. 2 b ) and the mass of the cross-linked SOD1 dimer corresponds to the loss of one water molecule after the addition of 1-oxo-1,2-dithiane to a SOD1 dimer.
  • 1-oxo-1,2-dithiane (NSC56224) and its analogues have been partially characterized previously for their ability to attack retroviral zinc fingers. Identified compounds can be found in FIG. 4 and FIG. 5 .
  • NSC56224 again being identified as a covalent dimerizer of DJ-1
  • NSC72268 was identified as a specific DJ-1 covalent dimerizer ( FIG. 3 a,b ).
  • Digesting NSC56224-linked DJ-1 with trypsin following by MALDI-TOF-MS confirmed NSC56224 covalently linked DJ-1 dimers at Cys53 ( FIG. 3 c ).
  • Both NSC56224 and NSC72268 were found to increase the denaturation temperature of DJ-1 measured with differential scanning fluorimetry ( FIG. 3 d ) (Niesen et al., 2007), suggesting covalent dimerization increased DJ-1 thermal stability.
  • NSC72268 and NSC56224 are shown in FIG. 6 .

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Described herein are compounds and methods for tethering proteins. For example, dimers of Protein X listed in Table 1 are described, where the dimers are formed by the covalent bonding of a cysteine on the first monomer to a cysteine on the second monomer via a cyclic disulfide linker. The covalently attached dimers exhibit increased stabilization and can be used to treat neurodegenerative diseases (such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD), cancer, autoimmune disease, and/or Celiac disease.

Description

    BACKGROUND OF THE INVENTION
  • Many therapeutic molecules form covalent bonds with cysteine residues on their protein targets. The mechanisms of the majority of these molecules were either elucidated long after development or are not fully understood. Recent successful drug discovery efforts, however, moved to structure-based design. These require both an accurate structural model of the target protein and a high-specificity ligand.
  • One third of therapeutic molecules, including many blockbuster drugs, form covalent bonds with their targets. These electrophilic drugs generally bond to a nucleophilic amino acid, often serine or cysteine, on a target protein. Aspirin and penicillin (and their many derivatives) acylate serines and numerous drugs form covalent bonds with specific cysteines. These therapeutic agents are effective despite the potential for off-target reactions with hundreds of highly reactive, nucleophilic residues, which are often required for the function of essential proteins. A worst case scenario for reaction with the “wrong” nucleophile is nerve gases (e.g., Sarin, intravenous LD50˜30 μg/kg), which covalently modify the active site serine of acetylcholine esterase. On the other hand, comparable toxicity has been harnessed to selectively target cancer cells—bortezomib/Velcade (LD100<250 μg/kg) selectively modifies an active site threonine of the proteasome. Unintended reaction with a highly reactive nucleophile isn't necessarily disastrous—it has led to a drug. The disulfide-containing substance, disulfiram, was intended to treat parasitic infections, but when tested on humans gave severe “hangover” symptoms upon alcohol consumption. Years after its therapeutic use began, this compound, dubbed antabuse, was found to bind the highly reactive active site cysteines of alcohol dehydrogenase. Nevertheless, the paucity of therapeutic suicide inhibitors to most human proteases, which (unlike viral proteases) have numerous homologues with identical off-target catalytic sites, has been attributed to off-target nucleophiles.
  • With effective covalent drugs, off-target binding tends to be offset by selectivity for the target and the enhanced potency inherent to irreversible inhibition. The uncanny specificities of cysteine-binding therapeutics involve elegant and usually serendipitous chemistry. The gastroesophageal reflux disease drugs (GERD, e.g., omeprazole/Prilosec™ lansoprazole/Prevacid™, etc.) use a cyclic sulphenamide to irreversibly bind a cysteine residue of the proton pump of the intestinal lumen. These benzolamide-derivative prodrugs require protonation of a low pKa pyridine nitrogen (pKa<4.5) for activation and sequestration. They are neutral, inactive, and permeable, but are activated upon encountering the pH˜0.8 parietal cell canaliculus, which contains their target (i.e., the proton pump). Here, they accumulate at 1000-fold higher concentrations. While the chemical basis of proton-mediated accumulation of omeprazole was appreciated, if not designed, the elegant sulfur-based chemistry behind activation and binding of a target cysteine was serendipitous.
  • The antithrombosis factors clopidogrel/Plavix™, ticlopide/Ticlid, etc. are also prodrugs. Activation by cytochrome P450 enzymes results in the scission of a ring carbon-sulfur bond, creating a sulfhydryl group that can then form a disulfide bond with its target cysteine on the adenosine diphosphate (ADP) chemoreceptor P2Y12. In addition to increased specificity for its target, which it permanently inactivates, the active metabolite has improved plasma protein binding characteristics. The thrombosis drugs had their beginnings in functional assays, and fortunately animal studies, because the active metabolite is not produced in most cell-based assays. Both their mechanism of action and target were unknown at the time of discovery.
  • More recent compounds employing sulfhydryl moieties were rationally designed. Dacomitinib, afatinib, and neratinib are EGFR kinase inhibitors with a high-affinity, nucleotide-analogue moiety that reversibly binds the ATP-binding pockets of numerous kinases and a second moiety designed to covalently bond with a non-conserved cysteine (present in EGFR but not its homologues). The electrophilic moiety is purposefully a low-reactivity acrylamide to minimize off-target reactions. A related chemical approach used low-reactivity, acrylamide-based, electrophiles to target non-conserved (in humans) and non-catalytic-residue cysteine of the HCV NS3/4A viral protease (HCVP).
  • In sum, all known approaches either minimize the exposure of highly reactive electrophiles (“hiding” a reactive sulfur in disulfides or in rings), or minimize the reactivity of exposed electrophiles (using acrylamide adducts). Unfortunately, however, the specificity of sulphenamides depends upon an acidic environment (pH<4.5) found only in the intestinal lumen, and the specificity of therapeutics employing reactive sulfhydryl groups is poorly understood. A few therapeutic molecules were obtained by rationally attaching low-reactivity electrophiles to high affinity and specificity moieties. Unfortunately, compounds with high affinity and specificity tend to appear in the final stages of a drug development effort making this approach best suited for improving existing specificity.
  • There exists a need for a strategy for conferring specificity to drugs that target cysteine, in general, but pairs of cysteine, in particular.
  • SUMMARY OF THE INVENTION Representative Methods of the Invention
  • In certain embodiments, the invention relates to a method comprising the step of contacting a compound of Formula I or a compound of Formula II with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, wherein the first Protein X comprises a first cysteine residue; the second Protein X comprises a second cysteine residue; the compound of Formula I is
  • Figure US20230102284A1-20230330-C00001
  • Wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4;
  • and R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, —OMs, —OTs, —OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and the compound of Formula II is
  • Figure US20230102284A1-20230330-C00002
  • wherein R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo and wherein the first and the second Protein X are selected from Table 1.
  • In certain embodiments, the invention relates to a method comprising the step of contacting a compound with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, wherein the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% homologous or identical to any one of the Protein X listed in Table 1 (in preferred embodiments, the first Protein X and the second Protein X are derived from the same Protein X); and the compound is a compound of Formula I
  • Figure US20230102284A1-20230330-C00003
  • wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4;
    and R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl.
  • In certain embodiments, the invention relates to a method comprising the step of contacting a compound with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, wherein the first Protein X and the second Protein X is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% homologous or identical to any one of the Protein X listed in Table 1 (in preferred embodiments, the first Protein X and the second Protein X are derived from the same Protein X); and the compound is a compound of Formula II
  • Figure US20230102284A1-20230330-C00004
  • wherein R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • In certain embodiments, the invention relates to a method of treating or preventing a Disease Y listed in Table 1 for each Protein X, comprising the step of administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or a compound of Formula II, wherein the compound of Formula I is
  • Figure US20230102284A1-20230330-C00005
  • wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4; and
    R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and the compound of Formula II is
  • Figure US20230102284A1-20230330-C00006
  • wherein R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • In certain embodiments, the invention relates to a method of treating or preventing a Disease Y listed in Table 1 for each Protein X, comprising the step of administering to a subject in need thereof a therapeutically effective amount of a stabilized analogue of Protein X, wherein said analogue has a tertiary structure and comprises a first monomer of Protein X and a second monomer of the Protein X; wherein the first monomer of Protein X comprises a first cysteine residue; the second monomer of Protein X comprises a second cysteine residue; the first cysteine residue is connected to the second cysteine residue by a connection; and the connection is a connection of Formula III or a connection of Formula IV, wherein Formula III is
  • Figure US20230102284A1-20230330-C00007
  • wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4; and
    R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and Formula IV is
  • Figure US20230102284A1-20230330-C00008
  • wherein R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo. In preferred embodiments, the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% homologous or identical to any one of the Protein X listed in Table 1. In further preferred embodiments, the first Protein X and the second Protein X are derived from the same Protein X.
  • In certain embodiments, the invention relates to a compound of Formula I
  • Figure US20230102284A1-20230330-C00009
  • wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4; and
    R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl.
  • One aspect of the invention is a stabilized analogue of Protein X, wherein said analogue has a tertiary structure and comprises a first monomer of Protein X and a second monomer of the Protein X; wherein the first monomer of Protein X comprises a first cysteine residue; the second monomer of Protein X comprises a second cysteine residue; the first cysteine residue is connected to the second cysteine residue by a connection; and the connection is a connection of Formula III or Formula IV:
  • Figure US20230102284A1-20230330-C00010
  • wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4;
    R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo. In preferred embodiments, the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% homologous or identical to any one of the Protein X listed in Table 1. In further preferred embodiments, the first Protein X and the second Protein X are derived from the same Protein X. In certain embodiments, these stabilized analogues are used to treat or prevent a Disease Y listed in Table 1 for each Protein X by administering to a subject in need thereof a therapeutically effective amount of a stabilized analogue of Protein X.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 depicts a) a potential reaction mechanism where a cyclic disulfide reacts with the cysteine of one monomer and the resulting thiolate can then react with, for example, a hydrogen peroxide-modified thiolate on the other monomer (Isaac, et al. Chemical Science 2012); b) side-chains of Cys53's in the dimer interface of DJ-1 (PDB: 3SF8), demonstrating their close spacing (Premkumar, et al. J. Struct. Biol. 2011, 176, 414). The appearance of Cys111's in the dimer interface of SOD1 is very similar.
  • FIG. 2 depicts a preliminary LC-ESI-IonTrap-MS screen of cyclic disulfides that identifies multiple compounds that form covalent SOD1 dimers. a) Cyclic disulfides identified in our preliminary screen that form covalent-linked SOD1 dimers. b) Deconvoluted LC-MS spectra of SOD1 with no compound (top) and with 1-oxo-1,2-dithiane (bottom).
  • FIG. 3 depicts a preliminary LC-ESI-IonTrap-MS screen of cyclic disulfides that identifies compounds that form covalent DJ-1 dimers at Cys53 and increase the thermal stability of DJ-1. a) NSC72268 was identified as a specific covalent dimerizer of DJ-1. b) Deconvoluted spectra of untreated DJ-1 (top) and NSC72268-treated DJ-1. c) MALDI-TOF-MS spectra showing a detected ion specific for NSC56224-treated DJ-1 corresponding to two trypsin digest fragments of DJ-1 containing Cys53 (underlined) linked by NSC56224. d) NSC56224 and NSC72268 increase the measured denaturation temperature of DJ-1 relative to untreated DJ-1 (N=3, error bars not shown but standard deviation is less than thickness of lines).
  • FIG. 4 depicts examples of cyclic disulfide compounds that are able to form covalent dimers of SOD1.
  • FIG. 5 depicts a dithiol that is able to form covalent dimers of SOD1.
  • FIG. 6 depicts examples of cyclic disulfide compounds that are able to form covalent dimers of DJ-1.
  • DETAILED DESCRIPTION OF THE INVENTION Overview
  • The covalent attachment of molecules can be used to affect protein structure and function. Covalently attached molecules can be used to inhibit, promote activity, stabilize, and destabilize proteins and peptides. One problem is encoding adequate specificity in covalent binders for the intended target. In certain embodiments, the invention relates to chemical tools, cyclic disulfides, which target pairs of cysteine residues and significantly enhance the specificity for pairs of cysteine over lone cysteine residues. In addition to augmenting current approaches to rational design, cyclic disulfides offer a launching point for compound optimization for novel targets. Whereas previous approaches to covalent modification tended to be devoted to inactivation of an enzyme, cyclic disulfides are also amenable to protein (including enzyme) stabilization. We apply cyclic-disulfides to the stabilization of any one of the Protein X listed in Table 1 involved in diseases, such as, for example, Disease Y shown in Table 1. In preferred embodiments, the disease that can be treated using the compounds disclosed herein, include, but are not limited to neurodegenerative disease (such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD), cancer, autoimmune disease and/or Celiac disease.
  • Table 1 shows a consolidated list of protein targets (Protein X) that can form the cyclic disulfide connections to form the stabilized analogues as described in herein. Column 1 lists the “Protein X” of each protein target that can be used to derive the stabilized analogues described herein. Column 2 provides the “Accession Number” corresponding to the protein target listed in Column 1 and connects the Protein X to the publicly available information known about each Protein X. Additionally, the Accession Number is unique identifier used in the art that provides sequence information for each protein listed in Column 1. Column 3 describes “Disease Y” for each Protein X. Each Disease Y has been shown to be associated with each Protein X as demonstrated in Column 4 Here, “Exemplified References” provides literature evidence associating each Protein X with Disease Y.
  • TABLE 1
    Summary Table of Protein Targets and their Associated Diseases
    Accession
    Number
    (Homo)
    Protein X sapiens Disease Y Exemplified References
    DJ-1 BAB71782 Parkinson's Lev N et al., “A DJ-1 Based Peptide Attenuates Dopaminergic Degeneration In Mice
    (Protein Disease Models Of Parkinson's Disease Via Enhancing Nrf2,” PLoS ONE 10(5): e0127549 (2015).
    deglycase, Bonifati et al., “Mutations In The DJ-1 Gene Associated With Autosomal Recessive
    also known Early-Onset Parkinsonism,” Science. 2003 Jan. 10; 299 (5604): 256-9.
    as Parkinson van der Merwe C1 et al., “Evidence For A Common Biological Pathway Linking Three
    disease Parkinson's Disease-Causing Genes: Parkin, PINK1 And DJ-1,” Eur J Neurosci.
    protein 7) 2015 May; 41(9): 1113-25.
    Choi et al., “Oxidative damage of DJ-1 is linked to sporadic Parkinson and Alzheimer
    diseases,” J Biol Chem. 2006 Apr. 21; 281(16): 10916-24. Epub 2006 Mar. 3.
    Amyotrophic Lev et al., “DJ-1 knockout augments disease severity and shortens survival in a mouse
    lateral model of ALS,” PLoS One. 2015 Mar. 30; 10(3): e0117190. doi: 10.1371/
    sclerosis journal.pone.0117190. eCollection 2015.
    (ALS) Knippenberg et al., “Altered expression of DJ-1 and PINK1 in sporadic ALS and in the
    SOD1(G93A) ALS mouse model,” J Neuropath Exp Neurol. 2013 November; 72(11):
    1052-61. doi: 10.1097/NEN.0000000000000004.
    Cancer; Cao et al., “DJ-1 as a human oncogene and potential therapeutic target,” Biochem
    breast Pharmacol. 2015 Feb. 1; 93(3): 241-50. doi: 10.1016/j.bcp.2014.11.012. Epub 2014 Dec. 11.
    cancer; Tanti et al., “SG2NA enhances cancer cell survival by stabilizing DJ-1 and thus activating
    laryngeal Akt,” Biochem Biophys Res Commun. 2015 May 27. pii: S0006-291X(15)00992-4. doi:
    cancer. 10.1016/j.bbrc.2015.05.069
    Kawate et al., “High levels of DJ-1 protein and isoelectric point 6.3 isoform in sera of
    breast cancer patients,” Cancer Sci. 2015 Apr. 13. doi: 10.1111/cas.12673.
    Zhu et al., “DJ-1-induced phosphatase and tensin homologue downregulation is associated
    with proliferative and invasive activity of laryngeal cancer cells,” Mol Med Rep.
    2015 Apr. 15. doi: 10.3892/mmr.2015.3617.
    Ismail et al., “DJ-1 upregulates breast cancer cell invasion by repressing KLF17
    expression,” Br J Cancer. 2014 Mar. 4; 110(5): 1298-306. doi: 10.1038/bjc.2014.40.
    Epub 2014 Feb. 6.
    Alzheimer's Choi et al., “Oxidative damage of DJ-1 is linked to sporadic Parkinson and Alzheimer
    Disease diseases,” J Biol Chem. 2006 Apr. 21; 281(16): 10916-24. Epub 2006 Mar. 3.
    SOD-1 AAB27562.1 Amyotrophic Valentine et al., “Misfolded CuZnSOD and amyotrophic lateral sclerosis,” Proc Natl
    lateral Acad Sci USA. 2003 Apr. 1; 100(7): 3617-3622.
    sclerosis Pasinelli et al., “Amyotrophic lateral sclerosis-associated SOD1 mutant proteins bind
    (ALS) and aggregate with Bcl-2 in spinal cord mitochondria,” Neuron 2004 July; 43(1): 19-30.
    Rosen et al., “Mutations in Cu/Zn superoxide dismutase gene are associated with familial
    amyotrophic lateral sclerosis,” Nature. 1993 March; 362(6415): 59-62.
    Parkinson's Bandmann et al., “Sequence of the superoxide dismutase 1 (SOD 1) gene in familial
    Disease Parkinson's disease,” J Neurol Neurosurg Psychiatry. 1995 July; 59(1): 90-91.
    Alzheimer's Zemlan et al., “Superoxide dismutase activity in Alzheimer's disease: possible
    Disease mechanism for paired helical filament formation,” Brain Res. 1989 Jan. 2; 476(1): 160-2.
    Murakami et al., “SOD1 (Copper/Zinc superoxide dismutase) deficiency drives amyloid
    beta protein oligomerization and memory loss in mouse model of Alzheimer Disease,”
    J Biol Chem. 2011 December; 286(52): 44557-68.
    Transglutaminase BAA14329.1 Celiac disease Frulio et al., “Evaluating diagnostic accuracy of anti-tissue Transglutaminase IgA
    antibodies as first screening for Celiac Disease in very young children,” Clin Chim
    Acta. 2015 Jun. 15; 446: 237-40. doi: 10.1016/j.cca.2015.04.035. Epub 2015 May 2.
    Zamot et al., “Presence of tissue transglutaminase IgA antibody as a celiac disease
    marker in a sample of patients with irritable bowel syndrome,” P R Health Sci J.
    2015 March; 34(1): 38-9.
    Cardoso et al., “Transglutaminase 2 interactions with extracellular matrix proteins as
    probed with celiac disease autoantibodies,” FEBS J. 2015 June; 282(11): 2063-75. doi:
    10.1111/febs.13276. Epub 2015 Apr. 13.
    Webb et al., “Celiac disease can be predicted by high levels of anti-tissue
    transglutaminase antibodies in population-based screening,” J Pediatr Gastroenterol
    Nutr. 2015 June; 60(6): 787-91. doi: 10.1097/MPG.0000000000000688.
    Klock et al., “Role of transglutaminase 2 in Celiac disease pathogenesis,”
    Semin Immunopathol. 2012 July; 34(4): 513-522. Published online 2012 Mar. 22. doi:
    10.1007/s00281-012-0305-0.
    Di Sabtino et al., “The function of tissue transglutaminase in celiac disease,”
    Autoimmun Rev. 2012 August; 11(10): 746-53. doi: 10.1016/j.autrev.2012.01.007.
    Epub 2012 Feb. 3.
    Huntington's Karpuj et al., “Evidence for a role for transglutaminase in Huntington's disease
    Disease and the potential therapeutic implications,” Neurochem Int. 2002 January; 40(1): 31-6.
    Mastroberardino et al., “Type 2 transglutaminase in Huntington's disease: a
    double-edged sword with clinical potential,” J Intern Med. 2010 November; 268(5):
    419-31. doi: 10.1111/j.1365-2796.2010.02275.x.
    Kahlem et al., “Transglutaminase Action Imitates Huntington's Disease: Selective
    Polymerization of Huntingtin Containing Expanded Polyglutamine,” Mol Cell. 1998
    March; 1(4): 595-601.
    Parkinson's Vermes et al., “Elevated concentration of cerebrospinal fluid tissue transglutaminase in
    Disease Parkinson's disease indicating apoptosis,” Mov Disord. 2004 October; 19(10): 1252-4.
    Junn et al., “Tissue transglutaminase-induced aggregation of alpha-synuclein:
    Implications for Lewy body formation in Parkinson's disease and dementia with Lewy
    Bodies,” Proc Natl Acad Sci USA. 2003 Feb. 18; 100(4): 2047-2052. Published online
    2003 Feb. 7. doi: 10.1073/pnas.0438021100.
    Andringa et al., “Tissue transglutaminase catalyzes the formation of alpha-synuclein
    crosslinks in Parkinson's disease,” FASEB J. 2004 May; 18(7): 932-4. Epub 2004 Mar. 4.
    Tau NP_058519.3 Alzheimer's Brunden et al., “Advances in Tau-focused drug discovery for Alzheimer's disease
    (for isoform Disease and related tauopathies,” Nat Rev Drug Discov. 2009 October; 8(10): 783-793. doi:
    1) 10. 1038/nrd2959.
    Mandelkow et al., “Tau in Alzheimer's disease” Trends Cell Biol. 1998 November;
    8(11): 425-7.
    Parkinson's Lei et al., “Tau protein: Relevance to Parkinson's disease,” Int J Biochem Cell
    Disease Biol. 2010 November; 42(11): 1775-8. doi: 10.1016/j.biocel.2010.07.016. Epub
    2010 Aug. 1.
    Wray et al., “A tangled web- Tau and sporadic Parkinson's disease,” Front
    Psychiatry. 2010; 1: 150. Published online 2010 Dec. 27. doi: 10.3389/
    fpsyt.2010.00150
    Irwin et al., “Parkinson's disease dementia: convergence of α-synuclein, tau and
    amyloid-β pathologies,” Nat Rev Neurosci. 2013 September; 14(9): 626-36. doi:
    10.1038/nrn3549. Epub 2013 Jul. 31.
    Epilepsy Holth et al., “Tau loss attenuates neuronal network hyperexcitability in mouse and
    Drosophila genetic models of epilepsy,” J Neurosci. 2013 Jan. 23;
    33(4): 1651-1659. doi: 10.1523/JNEUROSCI.3191-12.2013.
    DeVos et al., “Antisense reduction of Tau in adult mice protects against seizures,”
    J Neurosci. 2013 Jul. 31; 33(31): 12887-12897.
    Progranulin AAH00324 Alzheimer's Perry et al., “Progranulin mutations as risk factors for Alzheimer disease,” JAMA
    (Accession Disease Neurol. 2013 June; 70(6): 774-8. doi: 10.1001/2013.jamaneurol.393.
    Number of D'Alton et al., “Understanding the role of progranulin in Alzheimer's disease,”
    granulin; Nat Med. 2014 October; 20(10): 1099-100. doi: 10.1038/nm.3712.
    Progranulin Antonell et al., “Serum progranulin levels in patients with frontotemporal lobar
    is the degeneration and Alzheimer's disease: detection of GRN mutations in a Spanish cohort,”
    precursor of J Alzheimers Dis. 2012; 31(3): 581-91. doi: 10.3233/JAD-2012-112120.
    granulin) Frontotemporal Antonell et al., “Serum progranulin levels in patients with frontotemporal lobar
    Dementia degeneration and Alzheimer's disease: detection of GRN mutations in a Spanish cohort,”
    J Alzheimers Dis. 2012; 31(3): 581-91. doi: 10.3233/JAD-2012-112120.
    Almeida et al., “Progranulin, a glycoprotein deficient in frontotemporal dementia, is a
    novel substrate of several protein disulfide isomerase family proteins,” PLoS One. 2011;
    6(10): e26454. Published online 2011 Oct. 18. doi: 10,1371/journal.pone.0026454.
    Chiang et al., “Progranulin mutation causes frontotemporal dementia in the Swedish
    Karolinska family,” Alzheimers Dement. 2008 November; 4(6): 414-20. doi:
    10.1016/j.jalz.2008.09.001.
    Cancer; Edelman et al., “GP88 (progranulin): a novel tissue and circulating biomarker for
    small cell non-small cell lung carcinoma,” Hum Pathol. 2014 September; 45(9): 1893-9. doi:
    lung cancer; 10.1016/j.humpath.2014.05.011. Epub 2014 Jun. 5.
    cervical cancer; Wei et al., “Elevated expression of secreted autocrine growth factor progranulin
    gastrointestinal increases cervical cancer growth,” Cell Biochem Biophys. 2015 January; 71(1): 189-93.
    cancer. doi: 10.1007/s12013-014-0183-2.
    Lu et al., “Growth factor progranulin contributes to cervical cancer cell
    proliferation and transformation in vivo and in vitro,,” Gynecol Oncol. 2014
    August; 134(2): 364-71. doi: 10.1016/j.ygyno.2014.05.025. Epub 2014 Jun. 3.
    Hu et al., “Progranulin promotes tumour necrosis factor-induced proliferation of
    suppressive mouse CD4+ Foxp3+ regulatory T cells,”
    Immunology. 2014 June; 142(2): 193-201. doi: 10.1111/imm.12241.
    Demorrow. “Progranulin: a novel regulator of gastrointestinal cancer progression,”
    Transl Gastrointest Cancer. 2013 July; 2(3): 145-151.
    snRNP SM D3 NP_004166 Autoimmune Mahler. “Sm peptides in differentiation of autoimmune diseases,” Adv Clin Chem.
    disease 2011; 54: 109-28.
    Zieve et al., “The anti-Sm immune response in autoimmunity and cell biology,”
    Autoimmun Rev. 2003 September; 2(5): 235-40.
    McClain et al., “Anti-sm autoantibodies in systemic lupus target highly basic
    surface structures of complexed spliceosomal autoantigens,” J Immunol.
    2002 Feb. 15; 168(4): 2054-62.
    Galectin-1 NP_002296.1 Cancer; Thijssen et al., “Galectin expression in cancer diagnosis and prognosis: A systematic
    pancreatic review,” Biochim Biophys Acta. 2015 April; 1855(2): 235-247. doi:
    cancer; bladder 10.1016/j.bbcan.2015.03.003. Epub 2015 Mar. 25.
    cancer; breast Martinez-Bosch et al., “Targeting Galectin-1 in pancreatic cancer: immune surveillance
    cancer. on guard,” Oncoimmunology. 2014 Aug. 3; 3(8): e952201.
    Tang et al., “Apoptosis and anergy of T cell induced by pancreatic stellate
    cells-derived galectin-1 in pancreatic cancer,” Tumour Biol. 2015 Mar. 1.
    [Epub ahead of print].
    Wu et al., “Galectin-1 dysregulation independently predicts disease specific survival
    in bladder urothelial carcinoma,” J Urol. 2015 March; 193(3): 1002-8. doi:
    10.1016/j.juro.2014.09.107. Epub 2014 Oct. 2.
    Martinez-Bosch et al., “Galectin-1 drives pancreatic carcinogenesis through stroma
    remodeling and Hedgehog signaling activation,” Cancer Res. 2014 Jul. 1; 74(13):
    3512-24. doi: 10.1158/0008-5472.CAN- 13-3013. Epub 2014 May 8.
    Dalotto-Moreno et al., “Targeting galectin-1 overcomes breast cancer-associated
    immunosuppression and prevents metastatic disease,” Cancer Res. 2013 Feb. 1; 73(3):
    1107-17. doi: 10.1158/0008- 5472.CAN-12-2418. Epub 2012 Nov. 29.
    Heart disease Al-Salam et al., “Galectin-1 in early acute myocardial infarction,” PLoS One.
    2014 Jan. 31; 9(1): e86994. doi: 10.1371/journal.pone.0086994. eCollection 2014.
    Seropian et al., “Galectin-1 controls cardiac inflammation and ventricular remodeling
    during acute myocardial infarction,” Am J Pathol. 2013 January; 182(1): 29-40. doi:
    10.1016/j.ajpath.2012.09.022. Epub 2012 Nov. 9.
    HIV-1 St-Pierre et al., “Galectin-1-specific inhibitors as a new class of compounds to
    treat HIV-1 infection,” Antimicrob Agents Chemother. 2012 January; 56(1): 154-62.
    doi: 10.1128/AAC.05595-11. Epub 2011 Nov. 7.
    Sato et al., “Glycans, galectins, and HIV-1 infection,” Ann NY Acad Sci. 2012
    April; 1253: 133-48. doi: 10.1111/j.1749-6632.2012.06475.x.
    Mercier et al., “Galectin-1 promotes HIV-1 infectivity in macrophages through
    stabilization of viral adsorption,” Virology. 2008 Feb. 5; 371(1): 121-9. Epub
    2007 Oct. 29.
    Uridine NP_0011285 Cancer; colon Ashour et al., “Effect of administration of 5-(phenylselenenyl)acyclouridine, an
    phosphorylase 70.1 (for cancer inhibitor of uridine phosphorylase, on the anti-tumor efficacy of
    2 isoform b) 5-fluoro-2′-deoxyuridine against murine colon tumor C26- 10,” Biochem Pharmacol.
    2000 Sep. 1; 60(5): 687-92.
    Liu et al., “Expression, characterization, and detection of human uridine
    phosphorylase and identification of variant uridine phosphorolytic activity in selected
    human tumors,” Cancer Res. 1998 Dec. 1; 58(23): 5418-24.
    Pizzorno et al., “Phase I clinical and pharmacological studies of benzylacyclouridine,
    a uridine phosphorylase inhibitor,” Clin Cancer Res. 1998 May; 4(5): 1165-75.
    Estrogen receptor AAC05985 Cancer; prostate Dey et al., “Estrogen Receptor β2 Induces Hypoxia Signature of Gene Expression by
    beta cancer; breast Stabilizing HIF-1α in Prostate Cancer,” PLoS One. 2015 May 26; 10(5): e0128239.
    cancer doi: 10.1371/journal.pone.0128239. eCollection 2015.
    Hamilton et al., “Biologic Roles of Estrogen Receptor- β and Insulin-Like Growth
    Factor-2 in Triple- Negative Breast Cancer,” Biomed Res Int. 2015; 2015: 925703.
    doi: 10.1155/2015/925703. Epub 2015 Mar. 22.
    Gallo et al., “Estrogen receptor beta in cancer: an attractive target for therapy,”
    Curr Pharm Des. 2012; 18(19): 2734-57.
    Caspase-6 AAH04460.1 Alzheimer's LeBlanc. “Caspase-6 as a novel early target in the treatment of Alzheimer's
    Disease disease,” Eur J Neurosci. 2013 June; 37(12): 2005-18. doi: 10.1111/ejn.12250.
    Ramcharitar et al., “Cerebrospinal fluid tau cleaved by caspase-6 reflects brain
    levels and cognition in aging and Alzheimer disease,” J Neuropathol Exp Neurol.
    2013 September; 72(9): 824-32. doi: 10.1097/NEN.0b013e3182a0a39f.
    Albrecht et al., “Caspase-6 activation in familial alzheimer disease brains carrying
    amyloid precursor protein or presenilin i or presenilin II mutations,” J Neuropathol
    Exp Neurol. 2009 December; 68(12): 1282-93. doi: 10.1097/NEN.0b013e3181c1da10.
    Horowitz et al., “Early N-terminal changes and caspase-6 cleavage of tau in Alzheimer's
    disease,” J Neurosci. 2004 Sep. 8; 24(36): 7895-902.
    Caspase-3 CAC88866.1 Parkinson's Hartmann et al., “Caspase-3: A vulnerability factor and final effector in apoptotic
    Disease death of dopaminergic neurons in Parkinson's disease,” Proc Natl Acad Sci USA.
    2000 Mar. 14; 97(6): 2875-80.
    Zawada et al., “Loss of angiotensin II receptor expression in dopamine neurons in
    Parkinson's disease correlates with pathological progression and is accompanied by
    increases in Nox4- and 8-OH guanosine-related nucleic acid oxidation and caspase-3
    activation,” Acta Neuropathol Commun. 2015 Feb. 3; 3: 9. doi: 10.1186/
    s40478-015-0189-z.
    Tatton et al., “Apoptosis in Parkinson s disease: signals for neuronal degradation,”
    Ann Neurol. 2003; 53 Suppl 3: S61-70; discussion S70-2.
    Tatton et al., “Increased caspase 3 and Bax immunoreactivity accompany nuclear
    GAPDH translocation and neuronal apoptosis in Parkinson's disease,” Exp Neurol. 2000
    November; 166(1): 29-43.
    Cancer; breast O'Donovan et al., “Caspase-3 in breast cancer,” Clin Cancer Res. 2003 February;
    cancer 9(2): 738-42. Devarajan et al., “Down-regulation of caspase 3 in breast cancer: a
    possible mechanism for chemoresistance,” Oncogene. 2002 Dec. 12; 21(57): 8843-51.
    Alzheimer's D'Amelio et al., “Caspase-3 triggers early synaptic dysfunction in a mouse model of
    Disease Alzheimer's disease,” Nature Neuroscience 14, 69-76 (2011). Doi: 10.1038/nn.2709.
    S100beta NP_006263 Alzheimer's Griffin et al., “Life-long overexpression of S100beta in Down's syndrome: implications
    Disease for Alzheimer pathogenesis. Neurobiol Aging. 1998 Sepember-October; 19(5): 401-5.
    Lambert et al., “Evidence for the association of the S100beta gene with low cognitive
    performance and dementia in the elderly,” Mol Psychiatry. 2007 September; 12(9):
    870-80. Epub 2007 Mar. 6.
    Yu et al., “S100beta interaction with tau is promoted by zinc and inhibited by
    hyperphosphorylation in Alzheimer's disease,” J Neurosci. 2001 Apr. 1; 21(7): 2240-6.
    Cirillo et al., “S100B inhibitor pentamidine attenuates reactive gliosis and reduces
    neuronal loss in a mouse model of Alzheimer's disease,” BioMed Research International,
    Article ID 508342.
    Down Griffin et al., “Life-long overexpression of s100beta in Down's syndrome: implications
    Syndrome for Alzheimer pathogenesis,” Neurobiol Aging. 1998 September-October; 19(5): 401-5.
    Gerlai et al., “Abnormal exploratory behavior in transgenic mice carrying multiple copies
    of the human gene for S100 beta,” J Psychiatry Neurosci. 1995 March; 20(2): 105-12.
    Royston et al., “Overexpression of S100beta in Down's syndrome: correlation with patient
    age and with beta-amyloid deposition,” Neuropathol Appl Neurobiol. 1999 October; 25(5):
    387-93.
    Phospholipase A2 AAF09020 Alzheimer's Sanchez-Mejia et al., “Phospholipase A2 and arachidonic acid in Alzheimer's disease,”
    Disease Biochim Biophys Acta. 2010 August; 1801(8): 784-90. doi: 10.1016/j.bbalip.2010.05.013.
    Epub 2010 May 27.
    Stephenson et al., “Cytosolic phospholipase A2 (cPLA2) immunoreactivity is elevated in
    Alzheimer's disease brain,” Neurobiol Dis. 1996 February; 3(1): 51-63.
    Fonteh et al., “Alterations in cerebrospinal fluid glycerophospholipids and phospholipase
    A2 activity in Alzheimer's disease,” J Lipid Res. 2013 October; 54(10): 2884-97. doi:
    10.1194/jlr.M037622. Epub 2013 Jul. 18.
    Sanchez-Mejia et al., “Phospholipase A2 reduction ameliorates cognitive deficits in a
    mouse model of Alzheimer's disease,” Nature Neuroscience 11, 1311-1318 (2008)
    Published online: 19 Oct. 2008. doi: 10.1038/nn.2213.
    Epilepsy Farooqui et al., “Inhibitors of brain phospholipase A2 activity: their neuropharmacological
    effects and therapeutic importance for the treatment of neurologic disorders,” Pharmacol
    Rev. 2006 September; 58(3): 591- 620.
    Gattaz et al., “Increased PLA2 activity in the hippocampus of patients with temporal
    lobe epilepsy and psychosis,” J Psychiatr Res. 2011 December; 45(12): 1617-20. doi:
    10.1016/j.jpsychires.2011.07.005. Epub 2011 Aug. 3.
    Schizophrenia Law et al., “The role of phospholipases A2 in schizophrenia,” Mol Psychiatry.
    2006 June; 11(6): 547-56.
    Gattaz et al., “Phospholipase A2 and the hypofrontality of schizophrenia, Prostaglandins,”
    Leukot Essent Fatty Acids, 1996 August; 55(1-2): 109-13.
    Smesny et al., “Phospholipase A2 activity is associated with structural brain changes in
    schizophrenia,” Neuroimage. 2010 Oct. 1; 52(4): 1314-27. doi:
    10.1016/j.neuroimage.2010.05.009. Epub 2010 May 18.
    Hematopoietic AAK07679.1 Duchenne Kamauchi et al., “Hematopoietic prostaglandin D synthase inhibitors for the treatment of
    prostaglandin muscular duchenne muscular dystrophy,” Brain Nerve. 2011 November; 63(11): 1261-9.
    D synthase dystrophy Okinaga et al., “Induction of hematopoietic prostaglandin D synthase in hyalinated necrotic
    (DMD) muscle fibers: its implication in grouped necrosis,” Acta Neuropathol. 2002 October;
    104(4): 377-84. Epub 2002 Jun. 6.
    Alzheimer's Mohri et al., “Hematopoietic prostaglandin D synthase and DP1 receptor are selectively
    Disease upregulated in microglia and astrocytes within senile plaques from human patients and in a
    mouse model of Alzheimer disease,” J Neuropathol Exp Neurol. 2007 June; 66(6): 469-80.
    Glutathione AAC51280.1 Cancer; CML; Kassogue et al., “Association of glutathione S-transferase (GSTM1 and GSTT1) genes with
    S-transferase lung cancer chronic myeloid leukemia,” Springerplus. 2015 May 1; 4: 210. doi: 10.1186/
    s40064-015-0966-y. eCollection 2015.
    Houlston, “Glutathione S-Transferase M1 Status and Lung Cancer Risk,” Cancer
    Epidemiol Biomarkers Prev August 1999 8; 675.
    Townsend et al., “The role of glutathione-S-transferase in anti-cancer drug resistance,”
    Oncogene (2003) 22, 7369-7375. doi: 10.1038/sj.one.1206940.
    Parkinson's Shi et al., “Identification of glutathione S-transferase Pi as a protein involved in
    Disease Parkinson disease progression,” Am J Pathol. 2009 July; 175(1): 54-65.
    Wang et al., “Association between Glutathione S-transferase M1/Glutathione
    S-transferase T1 polymorphisms and Parkinson's disease: a meta-analysis,” J Neurol
    Sci. 2014 Mar. 15; 338(1-2): 65-70. doi: 10.1016/j.jns.2013.12.018. Epub 2013 Dec. 17.
    Alzheimer's Lovell et al., “Decreased glutathione transferase activity in brain and ventricular fluid
    Disease in Alzheimer's disease,” Neurology. 1998 December; 51(6): 1562-6.
    Spalletta et al., “Glutathione S-transferase P1 and T1 gene polymorphisms predict
    longitudinal course and age at onset of Alzheimer disease,” Am J Geriatr Psychiatry.
    2007 October; 15(10): 879-87.
    11-beta- AAC31757 Metabolic Yoon et al., “Discovery of pyridyl sulfonamide 11-beta-hydroxysteroid dehydrogenase type
    hydroxysteroid Disorder 1 (11β- HSD1) inhibitors for the treatment of metabolic disorders,” Bioorg Med Chem
    dehydrogenase Lett. 2014 Nov. 1; 24(21): 5045-9. doi: 10.1016/j.bmcl.2014.09.012. Epub 2014 Sep. 16.
    Anil et al., “A novel 11β-hydroxysteroid dehydrogenase type1 inhibitor CNX-010-49
    improves hyperglycemia, lipid profile and reduces body weight in diet induced obese
    C57B6/J mice with a potential to provide cardio protective benefits,” BMC Pharmacol
    Toxicol. 2014 Aug. 7; 15: 43. doi: 10.1186/2050-6511-15-43.
    Ratziu. “Targeting non-alcoholic fatty liver disease through 11-βHSD1 inhibition,”
    Lancet Diabetes Endocrinol. 2014 May; 2(5): 354-6. doi: 10.1016/
    s2213-8587(14)70028-2. Epub 2014 Feb. 17.
    Wamil et al., “Inhibition of 11 beta-hydroxysteroid dehydrogenase type 1 as a promising
    therapeutic target,” Drug Discovery Today. 2007 July; 12(13-14): 504-20. Epub 2007
    Jun. 27.
    Cancer; colon Moravec et al., “Expression of 11β-hydroxysteroid dehydrogenase type 2 is deregulated in
    cancer; colon carcinoma,” Histol Histopathol. 2014 April; 29(4): 489-96. Epub 2013 Nov. 5.
    osteosarcoma Jiang et al., “Epithelial-specific deletion of 11β-HSD2 hinders Apcmin/+ mouse
    tumorigenesis,” Mol Cancer Res. 2013 September; 11(9): 1040-50. doi:
    10.1158/1541-7786.MCR-13-0084-T. Epub 2013 Jun. 5.
    Patel et al., “Expression of 11β-hydroxysteroid dehydrogenase enzymes in human
    osteosarcoma: potential role in pathogenesis and as targets for treatments,” Endocr Relat
    Cancer. 2012 Jul. 22; 19(4): 589-98. doi: 10.1530/ERC-12-0079. Print 2012 August.
    Adenosine 5′- NP_216908 Tuberculosis Cosconati et al., “Structure-based virtual screening and biological evaluation of
    phosphosulfate Mycobacterium tuberculosis adenosine 5′-phosphosulfate
    reductase reductase inhibitors,” J Med Chem. 2008 Nov. 13; 51(21): 6627-30. doi: 10.1021/
    (Mycobacterium jm800571m. Epub 2008 Oct. 15.
    tuberculosis) Paritala et al., “A continuous spectrophotometric assay for adenosine 5′-phosphosulfate
    reductase activity with sulfite-selective probes,” Anal Biochem. 2013 Sep. 1; 440(1):
    32-9. doi: 10.1016/j.ab.2013.05.007. Epub 2013 May 24.
    Paritala et al., “Design, synthesis and evaluation of Fe—S targeted adenosine
    5′-phosphosulfate reductase inhibitors,” Nucleosides Nucleotides Nucleic Acids.
    2015; 34(3): 199-220. doi: 10.1080/15257770.2014.978012.
  • For example, as described below, stabilized analogues of Cu/Zn-SOD1 or DJ-1 can be used to treat ALS, Parkinson's and/or Alzheimer's Disease.
  • SOD1 and Amyotrophic Lateral Sclerosis (ALS)
  • Amyotrophic lateral sclerosis is a progressive neurodegenerative disease caused by death of motor neurons in the brain and spinal cord. The overall median survival from onset of symptoms ranges between 2-3 years for cases with bulbar onset to 3-5 years for cases with limb onset. Lifetime risk of ALS is 1/400 to 1/1000 with a median annual incidence of 1.89 and a median prevalence of 5.2 per 100,000 each year. There exists no cure for ALS and the only FDA-approved treatment for ALS, riluzole (Rilutek), prolongs median survival by a mere 2-3 months when taken for an eighteen month duration. Thus, novel therapeutic strategies for ALS continue to be crucial. Approximately ten percent of ALS is familial (fALS) and approximately twenty percent of fALS cases are caused by autosomal dominant mutations in the ubiquitously expressed protein SOD1. Over 100 SOD1 mutations have been identified which are linked with fALS and it is thought they confer a toxic gain of function. As the clinical phenotypes of patients with various fALS SOD1-associated mutations are more alike than different, and all appear to cause the death of motor neurons, it has been hypothesized that mutations share common properties and mechanisms of cytotoxicity. In addition to causing twenty percent of fALS, SOD1 may be playing a role in sporadic ALS. Evidence is emerging that a subset of sporadic ALS is characterized by unfolded WT SOD1, and that oxidatively modified SOD1 slows axonal transport to a similar extent to the G93A SOD1 variant. Numerous other reports have also implicated oxidized/misfolded WT SOD1 as being cytotoxic and/or related to sporadic ALS.
  • One prevailing hypothesis for the mechanism of the toxicity of ALS-associated SOD1 variants involves dimer destabilization and dissociation into monomers, which then nucleate the formation of higher-order aggregates. ALS-associated variants of SOD1, such as G85R, are found as monomers in ALS patients and a number of modifications, including loss of Cu or Zn, cleavage of the native, intramolecular disulfide, oxidation, glutathionylation, and fALS-associated mutation, predispose the SOD1 dimer to dissociate. X-ray crystal structures of both A4V, and to a lesser extent I113T, yeast two-hybrid analysis of H46R, A4V, and H48Q, dissociation of G85R, G93R, E100G, and I113T by chaotrophs, and molecular dynamics simulations are all consistent with this hypothesis; mutations and modifications destabilize dimer formation.
  • SOD1 dimers contain two cysteine residues at the dimer interface whose sulfhydryl groups are approximately nine angstroms apart. These sulfhydryl groups can be targeted by maleimide cross-linkers which lead to strong stabilization of ALS-associated SOD1 dimers. Surprisingly, while cross-linking at sulfhydryl groups by the maleimides occurred by predicted maleimide-mediated mechanisms, for the maleimide dithio-bismaleimidoethane (DTME), it was found that stabilization of the SOD1 dimer possibly occurred through both maleimide interaction with the sulfhydryl group of Cys111 on one SOD1 monomer as well as thiol-disulfide exchange between the disulfide spacer of DTME and the sulfhydryl group of the Cys111 on the second SOD1 monomer. Unfortunately, maleimides are highly irritating locally and have an LD50 in mice of 9 mg/kg with renal, hepatic, neurologic and hematologic toxicities as the principal effects of the drug in this species. Therefore, in certain embodiments, the invention relates to the discovery of molecules that can cross-link SOD1 dimers in order to fully assess small molecule-mediated covalent dimer formation of SOD1 as a therapeutic strategy for ALS.
  • DJ-1 and Parkinson's Disease (PD)
  • The progressive neurodegenerative disorder PD is characterized by the loss of dopaminergic neurons in the substania nigra pars compacta and α-synuclein-rich protein deposits known as Lewy bodies. A variety of pharmacological treatment options exist for the early-stage symptoms of PD as the patient becomes functionally impaired. However, as the disease progresses, all of these agents, which primarily treat the symptoms of PD, become ineffective as fewer dopaminergic neurons survive. Thus, as the ability to slow the progression of the disease remains elusive, novel directions in therapeutic development are necessary to further combat PD.
  • While the majority (>90%) of PD cases are idiopathic, mutations in PARK7, encoding the 189-amino acid homodimeric protein DJ-1, are known to be a rare cause of autosomal recessive early-onset Parkinson disease. Some evidence also indicates polymorphisms in PARK7 confer risk in sporadic PD patients. Biochemical and cell culture analysis of PD-linked variants of DJ-1 suggest a number of mechanisms through which structural defects, including loss of stability and dimer formation, may lead to a loss-of-function that is associated with PD pathogenicity, such as reduced ability to prevent α-synuclein aggregation, deficiency in oxidative stress-dependent RNA-binding activity, reduced ability to act as a neuroprotective transcriptional co-activator, and increased sensitivity to oxidative stress-induced cell death related to mitochondrial defects. In additional to recessive PD-related mutants of DJ-1 being implicated in disease, analysis of DJ-1 in the frontal cortex of patients with sporadic PD and Alzheimer's disease reveal that acidic isoforms of monomeric DJ-1 and basic isoforms of SDS-resistant dimeric DJ-1 selectively accumulate in these diseases, with DJ-1 irreversibly oxidized by carbonylation as well as by methionine oxidation to methionine sulfone. Over-oxidation of DJ-1 has been found to produce structural destabilization similar to PD-related mutations, suggesting that dysfunctional DJ-1 due to aberrant modifications could be a cause of sporadic neurodegenerative cases.
  • Just as loss of DJ-1 function appears to contribute to the etiology of PD, evidence suggests that enhancement of DJ-1 function could compensate for other causes of PD. DJ-1 protects against degeneration of nigral dopaminergic neurons in PD rat models involving both 6-hydroxydopamine and rotenone treatment. Viral-mediated DJ-1 overexpression in the MPTP mouse model has also proved efficacious in reducing nigral dopamine neuron loss. Likewise, pharmacological upregulation of DJ-1 with the histone deacetylase inhibitor phenylbutyrate rescues cells from oxidative stress and mutant α-synuclein toxicity, as well as protects dopaminergic neurons from MPTP-induced neurotoxicity and prevents age-related motor and cognitive decline in mice with diffuse Lewy body disease. Thus, enhancement of DJ-1 activity could serve as a therapeutic strategy in a possibly wide variety of PD cases. Previously, in silico methods have been used to identify potential small molecule binding sites on DJ-1 and for identifying small molecules capable of interacting with DJ-1 and modulating its oxidation state that have neuroprotective effects in vivo. However, Cys53s in DJ-1 are more closely spaced than Cys111s in SOD1, hindering previous attempts to covalently dimerize DJ-1 using maleimide crosslinkers.
  • Dithiols and Cyclic Disulfides as Covalent Dimerizers and Therapeutics
  • The cyclic disulfides described herein can covalently dimerize any one of the Protein X listed in Table 1 through their respective closely spaced dimer interface cysteines. Theoretically, a cyclic disulfide could undergo thiol-disulfide exchange with the cysteine of a Protein X, leaving a free thiolate to react with the remaining monomer (FIG. 1 ). Dithiols might also be capable of forming covalent dimers if the thiol groups are properly spaced and appropriately reactive. In agreement with this hypothesis, we present several cyclic disulfides and a dithiol discovered in preliminary screens that are capable of covalently dimerizing any one of the Protein X listed in Table 1.
  • Representative Methods of the Invention
  • In certain embodiments, the invention relates to a method comprising the step of contacting a compound of Formula I or a compound of Formula II with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, thereby cross-linking the first Protein X to the second Protein X, wherein the first Protein X comprises a first cysteine residue; the second Protein X comprises a second cysteine residue; the compound of Formula I is
  • Figure US20230102284A1-20230330-C00011
  • wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4; and
    R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and the compound of Formula II is
  • Figure US20230102284A1-20230330-C00012
  • Wherein R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo and wherein Protein X is selected from Table 1.
  • In certain embodiments, the invention relates to a method comprising the step of contacting a compound with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, thereby cross-linking the first Protein X to the second Protein X, wherein the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical or homologous to one another and wherein the first Protein X and the second Protein X are any one of the Protein X listed in Table 1; and the compound is a compound of Formula I
  • Figure US20230102284A1-20230330-C00013
  • Wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4; and
  • R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein Y is S.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein Y is S═O.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein Y is S(═O)2.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein n is 1 or 2.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein n is 1.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein n is 2.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is selected from the group consisting of
  • Figure US20230102284A1-20230330-C00014
  • In certain embodiments, the invention relates to a method comprising the step of contacting a compound disclosed herein with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, thereby cross-linking the first Protein X to the second Protein X, wherein the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% homologous or identical to one another; the first Protein X and the second Protein X are any one of the Protein X listed in Table 1; and the compound is a compound of Formula II
  • Figure US20230102284A1-20230330-C00015
  • Wherein R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the R″ form a six-membered ring. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the R″ form an aromatic ring. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the R″ form a six-membered aromatic ring.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is
  • Figure US20230102284A1-20230330-C00016
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X have at least 95% sequence homology or identity to one another.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X have at least 98% sequence homology or identity to one another.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X have at least 99% sequence homology or identity to one another.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X are wild type of any one of the Protein X listed in Table 1.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the method is performed in vitro. For example, the cross-linking of the first and second Protein X with a compound of Formula I or Formula II occurs outside of a patient and then the resultant cross-linked, stabilized analogue of Formula III or Formula IV is administered to the patient in a therapeutically effective amount, such as for example, for enzyme replacement therapy. Alternatively, the cross-linking of the first and the second Protein X can occur in vivo. In this situation, the molecule represented by Formula I or Formula II is administered to a patient in a therapeutically effective amount to cross-link the first and second Protein X in the patient. In further embodiments, the invention relates to any one of the aforementioned methods, wherein the first Protein X and the second Protein X are cross-linked within a cell.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the method is a method of inhibiting the activity of the first Protein X or the second Protein X.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the method is a method of increasing the activity of the first Protein X or a second Protein X.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the method is a method of stabilizing the first Protein X or the second Protein X.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the method is a method of destabilizing the first Protein X or the second Protein X.
  • In certain embodiments, the invention relates to a method of treating or preventing a Disease Y listed in Table 1 for each Protein X, comprising the step of administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or a compound of Formula II, wherein the compound of Formula I is
  • Figure US20230102284A1-20230330-C00017
  • Wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4; and
  • R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and the compound of Formula II is
  • Figure US20230102284A1-20230330-C00018
  • wherein R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and Y is S.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and Y is S═O.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and Y is S(═O)2.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and n is 1 or 2.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and n is 1.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and n is 2.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula I; and the compound is selected from the group consisting of
  • Figure US20230102284A1-20230330-C00019
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula II; and the R″ form a six-membered ring. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula II; and the R″ form an aromatic ring. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula II; and the R″ form a six-membered aromatic ring.
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein the compound is a compound of Formula II; and the compound is
  • Figure US20230102284A1-20230330-C00020
  • In certain embodiments, the invention relates to any one of the aforementioned methods, wherein a Disease Y is a neurodegenerative disease (such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD), cancer, autoimmune disease and/or Celiac disease, or any other Disease Y listed in Table 1.
  • Representative Compounds of the Invention
  • In certain embodiments, the invention relates to a compound of Formula I
  • Figure US20230102284A1-20230330-C00021
  • Wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4; and
  • R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl.
  • In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein Y is S.
  • In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein Y is S═O.
  • In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein Y is S(═O)2.
  • In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein n is 1 or 2.
  • In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein n is 1.
  • In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein n is 2.
  • In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein the proviso that the compound is not selected from the group consisting of
  • Figure US20230102284A1-20230330-C00022
  • Representative Analogues of the Invention
  • One aspect of the invention is a stabilized analogue of any one of the Protein X listed in Table 1, wherein said analogue has a tertiary structure and comprises a first Protein X monomer and a second Protein X monomer; wherein the first Protein X monomer comprises a first cysteine residue; the second Protein X monomer comprises a second cysteine residue; the first cysteine residue is connected to the second cysteine residue by a connection; and the connection is a connection of Formula III or Formula IV:
  • Figure US20230102284A1-20230330-C00023
  • Wherein Y is S, S═O, or S(═O)2; n is 0, 1, 2, 3, or 4;
  • R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and R′ is alkyl or aryl; and R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein the tertiary structure is substantially the same as the wild-type of any one of the Protein X listed in Table 1.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein the sequence homology or identity of the first Protein X monomer and the second Protein X monomer is greater than or equal to about 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein the first Protein X monomer and the second Protein X monomer have substantially the same amino acid sequence.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein the first Protein X monomer of said analogue is the wild-type sequence or comprises one, two, three, four, five, six, seven, eight, nine, or ten point mutations as compared to the wild type sequence.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein the second Protein X monomer of said analogue is the wild-type sequence or comprises one, two, three, four, five, six, seven, eight, nine, or ten point mutations as compared to the wild type sequence.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein said analogue retains at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% activity of the wild-type of any one of the Protein X listed in Table 1.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein said analogue retains at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% activity of the wild-type of any one of the Protein X listed in Table 1 up to a temperature of about 75° C.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization from about 10° C. to about 60° C.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization from about 20° C. to about 40° C.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization from about 15° C. to about 25° C.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization from about 30° C. to about 50° C.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization about 20° C.
  • In certain embodiments, the present invention relates to any one of the aforementioned analogues, wherein said analogue is increased in stabilization about 40° C.
  • Definitions
  • The term “analogue” refers to a molecule substantially similar in function to any one of the Protein X listed in Table 1 or a fragment thereof.
  • The terms “percent (%) sequence similarity”, “percent (%) sequence identity”, and the like, generally refer to the degree of identity or correspondence between different nucleotide sequences of nucleic acid molecules or amino acid sequences of polypeptides that may or may not share a common evolutionary origin (see Reeck et al., supra). Sequence identity can be determined using any of a number of publicly available sequence comparison algorithms, such as BLAST, FASTA, DNA Strider, GCG (Genetics Computer Group, Program Manual for the GCG Package, Version 7, Madison, Wis.), etc.
  • Specifically, the terms “percent (%) amino acid sequence identity” or “percent amino acid sequence homology” or “percent (%) identical” as used herein with respect to a reference polypeptide is defined as the percentage of amino acid residues in a candidate polypeptide sequence that are identical with the amino acid residues in the reference polypeptide sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, without considering any conservative substitutions as part of the sequence identity. Alignment for the purpose of determining percent amino acid sequence identity can be achieved by various techniques known in the art, for instance, using publicly available computer software such as ALIGN or Megalign (DNASTAR). Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the peptide sequence being used in the comparison. For example, in the context of the present invention, an analogue of SOD1 is said to share “substantial homology” with SOD1 if the amino acid sequence of said analogue is at least about 85%, at least about 90%, at least about 95%, or at least about 99% identical to wild-type.
  • To determine the percent identity and/or homology between two amino acid sequences or two nucleic acid molecules, the sequences are aligned for optimal comparison purposes. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., percent identity=number of identical positions/total number of positions (e.g., overlapping positions)×100). In one embodiment, the two sequences are, or are about, of the same length. The percent identity between two sequences can be determined using techniques similar to those described below, with or without allowing gaps. In calculating percent sequence identity, typically exact matches are counted.
  • The determination of percent identity between two sequences can be accomplished using a mathematical algorithm. A non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, Proc. Natl. Acad. Sci. USA 1990, 87:2264, modified as in Karlin and Altschul, Proc. Natl. Acad. Sci. USA 1993, 90:5873-5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al, J. Mol. Biol. 1990; 215: 403. BLAST nucleotide searches can be performed with the NBLAST program, score=100, wordlength=12, to obtain nucleotide sequences homologous to sequences of the invention. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3, to obtain amino acid sequences homologous to protein sequences of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al, Nucleic Acids Res. 1997, 25:3389. Alternatively, PSI-Blast can be used to perform an iterated search that detects distant relationship between molecules. See Altschul et al. (1997) supra. When utilizing BLAST, Gapped BLAST, and PSI-Blast programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See ncbi.nlm.nih.gov/BLAST/on the WorldWideWeb.
  • Another non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, CABIOS 1988; 4: 1 1-17. Such an algorithm is incorporated into the ALIGN program (version 2.0), which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used.
  • In a preferred embodiment, the percent identity between two amino acid sequences is determined using the algorithm of Needleman and Wunsch (J. Mol. Biol. 1970, 48:444-453), which has been incorporated into the GAP program in the GCG software package (Accelrys, Burlington, Mass.; available at accelrys.com on the WorldWideWeb), using either a Blossum 62 matrix or a PAM250 matrix, a gap weight of 16, 14, 12, 10, 8, 6, or 4, and a length weight of 1, 2, 3, 4, 5, or 6. In yet another preferred embodiment, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package using a NWSgapdna.CMP matrix, a gap weight of 40, 50, 60, 70, or 80, and a length weight of 1, 2, 3, 4, 5, or 6. A particularly preferred set of parameters (and the one that can be used if the practitioner is uncertain about what parameters should be applied to determine if a molecule is a sequence identity or homology limitation of the invention) is using a Blossum 62 scoring matrix with a gap open penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • Statistical analysis of the properties described herein may be carried out by standard tests, for example, t-tests, ANOVA, or Chi squared tests. Typically, statistical significance will be measured to a level of p=0.05 (5%), more preferably p=0.01, p=0.001, p=0.0001, p=0.000001.
  • The phrase “pharmaceutically acceptable” is employed herein to refer to those ligands, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals, substantially non-pyrogenic, without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • The phrase “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ or portion of the body, to another organ or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, not injurious to the patient, and substantially non-pyrogenic. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations. In certain embodiments, pharmaceutical compositions of the present invention are non-pyrogenic, i.e., do not induce significant temperature elevations when administered to a patient.
  • The term “preventing” is art-recognized, and when used in relation to Disease Y listed in Table 1 for each Protein X, such as a local recurrence (e.g., pain), a disease such as cancer, a syndrome complex such as heart failure or any other medical condition, is well understood in the art, and includes administration of a composition which reduces the frequency of, or delays the onset of, symptoms of a medical condition in a subject relative to a subject which does not receive the composition. Thus, prevention of cancer includes, for example, reducing the number of detectable cancerous growths in a population of patients receiving a prophylactic treatment relative to an untreated control population, and/or delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population, e.g., by a statistically and/or clinically significant amount. Prevention of an infection includes, for example, reducing the number of diagnoses of the infection in a treated population versus an untreated control population, and/or delaying the onset of symptoms of the infection in a treated population versus an untreated control population. Prevention of pain includes, for example, reducing the magnitude of, or alternatively delaying, pain sensations experienced by subjects in a treated population versus an untreated control population. Prevention of a neurodegenerative disease or disorder, for example, includes the reduction and/or slowing down of the appearance of symptoms (e.g., a decrease in cognitive function) associated with the neurodegenerative disease or disorder as compared to subjects in a treated population versus an untreated control population
  • A “therapeutically effective amount” of a compound, e.g., such as a polypeptide or peptide analogue of the present invention, with respect to use in treatment, refers to an amount of the polypeptide or peptide analogue of Formula I or Formula II described herein and/or the stabilized analogue of Formula III or Formula IV as described herein in a preparation which, when administered as part of a desired dosage regimen (to a mammal, preferably a human) alleviates a symptom, ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated or the cosmetic purpose, e.g., at a reasonable benefit/risk ratio applicable to any medical treatment.
  • The terms “prophylactic” or “therapeutic” treatment are art-recognized and include administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • As used herein, the term “about” is used to refer to an amount that is approximately, nearly, almost, or in the vicinity of being equal to or is equal to a stated amount, e.g., the state amount plus/minus about 5%, about 4%, about 3%, about 2% or about 1%.
  • The term “patient” or “subject” as used herein in reference can encompasses veterinary uses, such as, for example, the testing of a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), rabbit, murine (e.g. a mouse), canine (e.g. a dog), feline (e.g. a cat), equine (e.g. a horse), bovine (e.g., cow) a primate, simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon), an ape (e.g. gorilla, chimpanzee, orangutan, gibbon). In preferred embodiments the patient is a human.
  • Other aspects and embodiments of the invention provide the aspects and embodiments described above with the term “comprising” replaced by the term “consisting of” and the aspects and embodiments described above with the term “comprising” replaced by the term “consisting essentially of”.
  • “and/or” where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. For example “A and/or B” is to be taken as specific disclosure of each (i) A, (ii) B and (iii) A and B, just as if each is set out individually.
  • It is to be understood that the application discloses all combinations of any of the above aspects and embodiments described above with each other, unless the context demands otherwise. Similarly, the application discloses all combinations of the preferred and/or optional features either singly or together with any of the other aspects, unless the context demands otherwise.
  • Modifications of the above embodiments, further embodiments and modifications thereof will be apparent to the skilled person on reading this disclosure, and as such these are within the scope of the present invention.
  • All documents and sequence database entries mentioned in this specification are incorporated herein by reference in their entirety for all purposes.
  • EXEMPLIFICATION
  • The invention now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.
  • Example 1—General Materials and Methods Cross-Linking and Western Blots
  • WtSOD1 or wtDJ-1 was incubated with 5-25 mM DTT for approximately 20 minutes and either buffer exchanged using Amicon Ultra-4 centrifugal spin concentrators (MWCo 10K) or using reversed phase chromatography (ZIPTIP, Millipore, Inc). Samples cleaned by ZIPTIPs were also subjected to incubation with 5 mM EDTA. SOD1 samples that were buffer exchanged using Amicon concentrators were exchanged into in HPLC water, whereas ZIPTIP samples were further exchanged after ZIPTIP into PBS, pH 7.4 or HPLC water. DTT-reduced SOD1 or DJ-1 was incubated at a 1:1 (20 μM:20 μM or 10 μM:10 μM) or 1:3 (20 μM:60 μM or 10 μM:30 μM) ratio of protein to cross-linker.
  • A variety of cross-linkers were used. Cross-linking was achieved by incubating the reaction in either PBS pH 7.4 or water at room temperature for 1 hour. After an hour the reactions were analyzed on a 15% SDS-PAGE gel with a non-cross-linked control, transferred to nitrocellulose membrane and western blotted using a polyclonal antibody to SOD1 or DJ-1. Repeated in triplicate.
  • In addition, DTME is a cleavable sulfhydryl-sulfhydryl cross-linking agent. Therefore, a cross-linking reaction containing 1:1 molar ratio of wtSOD1 or wtDJ-1 to DTME was performed at room temperature for one hour. After cross-linking, the reaction was split in half and half of the sample was run in a sample buffer containing DTT (reducing) and the other half in one containing no DTT (non-reducing). These samples along with non-cross-linked controls were then analyzed on a 15% SDS PAGE gel and western blotted as above.
  • Matrix Assisted Laser Desorption Ionization (MALDI)-Time of Flight (TOF)
  • wtSOD1 or wtDJ-1 was cross-linked as below. After cross-linking, 1 μL of sample was spotted on a MALDI target containing 1 μl of matrix, 20 mg/mL sinipic acid, and analyzed on a Bruker Daltonics Microflex. The MALDI was calibrated each time using a high molecular weight protein calibration standard, Protein Calibration Standard I (Bruker Daltonics). The MALDI-TOF was operated in linear mode using a laser power of between 72-90%. MALDI-TOF spectra were of cross-linked and non-cross-linked samples were analyzed using FlexAnalysis software (Bruker Daltonics). Repeated in triplicate.
  • Example 2
  • LC-MS Screen of Cyclic Disulfides with SOD1 Reveals Small Molecules that Covalently Link SOD1 Dimers.
  • Compounds being evaluated were dissolved and incubated with recombinant human WT SOD1. Reactions were analyzed by LC-ESI-IonTrap-MS on a HCT Ultra ion trap (Bruker Daltonics, Billerica, Mass., USA). The resulting data was examined using DataAnalysis 3.4 (Bruker Daltonics Inc., Billerica, Mass., USA). Mass spectra were averaged across the retention times corresponding to when SOD1 was found to be eluting and Maximum Entropy Deconvolution was applied to the resulting average mass spectrum in order to determine the molecular weight of the uncharged species detected. Significant dithiol- and cyclic disulfide-mediated covalent dimer formation has been observed with multiple different compounds. For example, the changes in mass of the covalently linked SOD1 dimers observed suggest both 1,2-dithiolane-4,4-dimethanol and 1-oxo-1,2-dithiane (FIG. 2 a ) are capable of covalently dimerizing SOD1. Of note, 1-oxo-1,2-dithiane was able to cross-link the majority of SOD1 monomers in the sample (FIG. 2 b ) and the mass of the cross-linked SOD1 dimer corresponds to the loss of one water molecule after the addition of 1-oxo-1,2-dithiane to a SOD1 dimer. 1-oxo-1,2-dithiane (NSC56224) and its analogues have been partially characterized previously for their ability to attack retroviral zinc fingers. Identified compounds can be found in FIG. 4 and FIG. 5 .
  • Example 3
  • LC-MS Screen of Cyclic Disulfides with DJ-1 Reveals Molecules that Covalently Link DJ-1 Dimers.
  • Using the same method as described for SOD1, compounds were screened against WT DJ-1. In addition to NSC56224 again being identified as a covalent dimerizer of DJ-1, NSC72268 was identified as a specific DJ-1 covalent dimerizer (FIG. 3 a,b ). Digesting NSC56224-linked DJ-1 with trypsin following by MALDI-TOF-MS confirmed NSC56224 covalently linked DJ-1 dimers at Cys53 (FIG. 3 c ). Both NSC56224 and NSC72268 were found to increase the denaturation temperature of DJ-1 measured with differential scanning fluorimetry (FIG. 3 d ) (Niesen et al., 2007), suggesting covalent dimerization increased DJ-1 thermal stability. NSC72268 and NSC56224 are shown in FIG. 6 .
  • INCORPORATION BY REFERENCE
  • All of the U.S. patents and U.S. published patent applications cited herein are hereby incorporated by reference.
  • EQUIVALENTS
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims (21)

1. A stabilized analogue comprising a first Protein X and a second Protein X cross-linked to one another by a compound of Formula I or a compound of Formula II, wherein
the first Protein X comprises a first cysteine residue and is listed in Table 1;
the second Protein X comprises a second cysteine residue and is listed in Table 1;
the compound of Formula I is
Figure US20230102284A1-20230330-C00024
wherein
Y is S, S═O, or S(═O)2;
n is 0, 1, 2, 3, or 4; and
R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and
R′ is alkyl or aryl; and
the compound of Formula II is
Figure US20230102284A1-20230330-C00025
wherein
R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
2. The stabilized analogue of claim 1, wherein the first Protein X and the second Protein X have at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence homology or identity to any one of the Protein X listed in Table 1.
3. The stabilized analogue of claim 1 wherein the first Protein X and the second Protein X are derived from different Protein X shown in Table 1.
4. The stabilized analogue of claim 1, wherein the first Protein X and the second Protein X are derived from the same Protein X shown in Table 1.
5. The stabilized analogue of claim 4, wherein the first Protein X and the second Protein X are at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% homologous or identical to one another.
6. The stabilized analogue of claim 1, wherein the compound is a compound of Formula I; and the compound is selected from the group consisting of
Figure US20230102284A1-20230330-C00026
7. The stabilized analogue of claim 1, wherein the compound is a compound of Formula II, and the compound is
Figure US20230102284A1-20230330-C00027
8. The stabilized analogue of claim 1, wherein the stabilized analogue is a compound of Formula III or Formula IV:
Figure US20230102284A1-20230330-C00028
wherein
Y is S, S═O, or S(═O)2;
n is 0, 1, 2, 3, or 4;
R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and
R′ is alkyl or aryl; and
R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
9. The stabilized analogue of claim 1, wherein the stabilized analogue has reduced activity as compared to Protein X.
10. The stabilized analogue of claim 1, wherein the stabilized analogue has increased activity as compared to Protein X.
11. A method of treating or preventing Disease Y, comprising administering to a subject in need thereof a therapeutically effective amount of the stabilized analogue of claim 1, wherein Disease Y is a neurodegenerative disease (such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD), cancer, autoimmune disease, Celiac disease, and/or any of the Disease Y listed in Table 1.
12. A method of treating or preventing Disease Y, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or a compound of Formula II, wherein the compound of Formula I is
Figure US20230102284A1-20230330-C00029
wherein
Y is S, S═O, or S(═O)2;
n is 0, 1, 2, 3, or 4; and
R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and
R′ is alkyl or aryl; and
the compound of Formula II is
Figure US20230102284A1-20230330-C00030
wherein
R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo, wherein Disease Y is a neurodegenerative disease (such as, for example, Parkinson's Disease, ALS, Alzheimer's Disease, Huntington's Disease, Epilepsy, Frontotemporal Dementia, and/or DMD), cancer, autoimmune disease, Celiac disease, and/or any of the Disease Y listed in Table 1.
13. The method of claim 12, wherein the compound is a compound of Formula I, and the compound is selected from:
Figure US20230102284A1-20230330-C00031
14. The method of claim 12, wherein the compound is a compound of Formula II, and the compound is
Figure US20230102284A1-20230330-C00032
15. The method of claim 12, wherein the compound of Formula I or the compound of Formula II forms the stabilized analogue of Formula III or Formula IV.
16. A method comprising the step of contacting a compound of Formula I or a compound of Formula II with a first Protein X and a second Protein X under conditions suitable for cross-linking the first Protein X to the second Protein X, thereby cross-linking the first Protein X to the second Protein X, wherein
the first Protein X comprises a first cysteine residue and is listed in Table 1;
the second Protein X comprises a second cysteine residue and is listed in Table 1;
the compound of Formula I is
Figure US20230102284A1-20230330-C00033
wherein
Y is S, S═O, or S(═O)2;
n is 0, 1, 2, 3, or 4; and
R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and
R′ is alkyl or aryl; and
the compound of Formula II is
Figure US20230102284A1-20230330-C00034
wherein
R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
17. The method of claim 16, wherein the first Protein X and the second Protein X have at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence homology or identity to any one of the Protein X listed in Table 1; and the compound is a compound of Formula I
Figure US20230102284A1-20230330-C00035
wherein
Y is S, S═O, or S(═O)2;
n is 0, 1, 2, 3, or 4; and
R is independently selected from the group consisting of —H, —OH, —NH2, —NHR′, —N(R′)2, alkyl, -OMs, -OTs, -OTf, and —CO2H; or any two geminal R groups, taken together, form an imine; or any two vicinal R groups, taken together, form a ring; wherein any alkyl or imine may be substituted with a carbamide, a carboxylate, or a hydroxyl; and
R′ is alkyl or aryl.
18. The method of claim 16, wherein the compound is a compound of Formula I; and the compound is selected from:
Figure US20230102284A1-20230330-C00036
19. The method of claim 16, wherein the first Protein X and the second Protein X have at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence homology or identity to any one of the Protein X listed in Table 1; and the compound is a compound of Formula II
Figure US20230102284A1-20230330-C00037
wherein
R″ is —H, alkyl, or aryl, or both R″, taken together, form a ring; wherein any alkyl, aryl, or ring may be substituted with —OH, alkyl, or halo.
20. The method of claim 16, wherein the compound is a compound of Formula II, and the compound is
Figure US20230102284A1-20230330-C00038
21.-27. (canceled)
US18/057,854 2015-08-18 2022-11-22 Tethering cysteine residues using cyclic disulfides Pending US20230102284A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/057,854 US20230102284A1 (en) 2015-08-18 2022-11-22 Tethering cysteine residues using cyclic disulfides

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201562206316P 2015-08-18 2015-08-18
PCT/US2016/047395 WO2017031226A1 (en) 2015-08-18 2016-08-17 Tethering cysteine residues using cyclic disulfides
US201815753319A 2018-02-18 2018-02-18
US16/885,741 US20200291380A1 (en) 2015-08-18 2020-05-28 Tethering cysteine residues using cyclic disulfides
US18/057,854 US20230102284A1 (en) 2015-08-18 2022-11-22 Tethering cysteine residues using cyclic disulfides

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/885,741 Continuation US20200291380A1 (en) 2015-08-18 2020-05-28 Tethering cysteine residues using cyclic disulfides

Publications (1)

Publication Number Publication Date
US20230102284A1 true US20230102284A1 (en) 2023-03-30

Family

ID=56851696

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/753,319 Active US10711261B2 (en) 2015-08-18 2016-08-17 Tethering cysteine residues using cyclic disulfides
US16/885,741 Abandoned US20200291380A1 (en) 2015-08-18 2020-05-28 Tethering cysteine residues using cyclic disulfides
US18/057,854 Pending US20230102284A1 (en) 2015-08-18 2022-11-22 Tethering cysteine residues using cyclic disulfides

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US15/753,319 Active US10711261B2 (en) 2015-08-18 2016-08-17 Tethering cysteine residues using cyclic disulfides
US16/885,741 Abandoned US20200291380A1 (en) 2015-08-18 2020-05-28 Tethering cysteine residues using cyclic disulfides

Country Status (4)

Country Link
US (3) US10711261B2 (en)
EP (1) EP3337468A1 (en)
CA (1) CA2994298A1 (en)
WO (1) WO2017031226A1 (en)

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69734734D1 (en) 1996-07-05 2005-12-29 Us Gov Health & Human Serv SATURATED 1,2-DITHIACHETEROCYCLIC COMPOUNDS CONTAINING ANTIVIRAL PHARMACEUTICAL PREPARATIONS
IL123887A0 (en) 1997-04-02 1998-10-30 Sankyo Co Dithiolan derivatives their use and pharmaceutical compositions containing the same
GB0619291D0 (en) 2006-09-29 2006-11-08 Ucb Sa Altered antibodies
ES2535222T3 (en) 2009-06-02 2015-05-07 Brandeis University Cross-linking of superoxide dismutase monomers
WO2010147957A2 (en) 2009-06-15 2010-12-23 Encore Health, Llc Dithiol compounds, derivatives, and uses therefor
CA2891261C (en) * 2012-11-15 2023-04-04 Brandeis University Tethering cysteine residues using cyclic disulfides

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Nare, B., "Schistosoma mansoni : role of antioxidant systems in protection of developmental stages against oxidative killing and the effects of oltipraz on glutathione S-transferase", Thesis, McGill University, 1991 (Year: 1991) *
Shen et al., Arch. Biochem. Biophys. 300:137-141, 1993 (Year: 1993) *

Also Published As

Publication number Publication date
EP3337468A1 (en) 2018-06-27
WO2017031226A1 (en) 2017-02-23
CA2994298A1 (en) 2017-02-23
US20180237763A1 (en) 2018-08-23
US10711261B2 (en) 2020-07-14
US20200291380A1 (en) 2020-09-17

Similar Documents

Publication Publication Date Title
US11254923B2 (en) Tethering cysteine residues using cyclic disulfides
Li et al. Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease
Kondapalli et al. PINK1 is activated by mitochondrial membrane potential depolarization and stimulates Parkin E3 ligase activity by phosphorylating Serine 65
Kwok et al. Mitochondrial UCP5 is neuroprotective by preserving mitochondrial membrane potential, ATP levels, and reducing oxidative stress in MPP+ and dopamine toxicity
Yamamoto et al. VDAC1, having a shorter N-terminus than VDAC2 but showing the same migration in an SDS− polyacrylamide gel, is the predominant form expressed in mitochondria of various tissues
McMillan et al. Attenuation of lung fibrosis in mice with a clinically relevant inhibitor of glutathione-S-transferase π
Aukrust et al. SUMOylation of Pancreatic Glucokinase Regulates Its Cellular Stability and Activity*[S]
Kawana et al. Reactive astrocytes and perivascular macrophages express NLRP 3 inflammasome in active demyelinating lesions of multiple sclerosis and necrotic lesions of neuromyelitis optica and cerebral infarction
Finger et al. Proteasomal degradation induced by DPP9‐mediated processing competes with mitochondrial protein import
Wang et al. The nitrated proteome in heart mitochondria of the db/db mouse model: characterization of nitrated tyrosine residues in SCOT
Kang et al. α-Synuclein binds and sequesters PIKE-L into Lewy bodies, triggering dopaminergic cell death via AMPK hyperactivation
Kokotas et al. Biomarkers in primary open angle glaucoma
Kunieda et al. 8-Nitro-cGMP enhances SNARE complex formation through S-guanylation of Cys90 in SNAP25
Evankovich et al. Receptor for advanced glycation end products is targeted by FBXO10 for ubiquitination and degradation
Zheng et al. Cdk5 inhibitory peptide (CIP) inhibits Cdk5/p25 activity induced by high glucose in pancreatic beta cells and recovers insulin secretion from p25 damage
Mozzi et al. SARS-CoV-2 ORF3c impairs mitochondrial respiratory metabolism, oxidative stress, and autophagic flux
US20230102284A1 (en) Tethering cysteine residues using cyclic disulfides
Lee et al. Role of Post-translational Modification of Silent Mating Type Information Regulator 2 Homolog 1 in Cancer and Other Disorders
Shim et al. Oxidative stress modulates the expression pattern of peroxiredoxin-6 in peripheral blood mononuclear cells of asthmatic patients and bronchial epithelial cells
Trinh et al. Parkinson's disease pathology is directly correlated to SIRT3 in human subjects and animal models: Implications for AAV. SIRT3-myc as a disease-modifying therapy
US20230181672A1 (en) Aberrant post-translational modifications (ptms) in methyl- and propionic acidemia and a mutant sirtuin (sirt) to metabolize ptms
WO2024035929A2 (en) Insulin sensitizers for reversal of insulin resistance
US7786090B2 (en) Methods and compositions for treating and preventing neurologic disorders
Veetil Glucosamine induced insulin resistance in primary rat hepatocytes and the role of selenium as an insulin mimetic
Adiyodi Veetil Glucosamine-Induced Insulin Resistance in Primary Rat Hepatocytes and the Role of Selenium as an Insulin Mimetic

Legal Events

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: BRANDEIS UNIVERSITY, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SALISBURY, JOSEPH;AGAR, JEFFREY;REEL/FRAME:063780/0649

Effective date: 20190716

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED