WO2019137922A1 - Modulateurs de c1q, en particulier de l'interaction de l'apoe avec c1q, et utilisations des modulateurs dans le traitement de maladies neuronales et de l'inflammation - Google Patents

Modulateurs de c1q, en particulier de l'interaction de l'apoe avec c1q, et utilisations des modulateurs dans le traitement de maladies neuronales et de l'inflammation Download PDF

Info

Publication number
WO2019137922A1
WO2019137922A1 PCT/EP2019/050354 EP2019050354W WO2019137922A1 WO 2019137922 A1 WO2019137922 A1 WO 2019137922A1 EP 2019050354 W EP2019050354 W EP 2019050354W WO 2019137922 A1 WO2019137922 A1 WO 2019137922A1
Authority
WO
WIPO (PCT)
Prior art keywords
clq
apoe
binding
variant
compound
Prior art date
Application number
PCT/EP2019/050354
Other languages
English (en)
Inventor
Christine Skerka
Peter Zipfel
Susanne ACKERMANN
Original Assignee
Leibniz-Institut Für Naturstoff-Forschung Und Infektionsbiologie
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 Leibniz-Institut Für Naturstoff-Forschung Und Infektionsbiologie filed Critical Leibniz-Institut Für Naturstoff-Forschung Und Infektionsbiologie
Priority to EP19701776.7A priority Critical patent/EP3737472A1/fr
Publication of WO2019137922A1 publication Critical patent/WO2019137922A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/472Complement proteins, e.g. anaphylatoxin, C3a, C5a
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/775Apolipopeptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/775Apolipopeptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/70Mechanisms involved in disease identification
    • G01N2800/7095Inflammation

Definitions

  • Modulators of clq in particular of the interaction of ApoE with clq, and uses of the modulators in the therapy of neuronal diseases and inflammation
  • the present invention relates to modulators of complex 1 protein q (Clq) that preferably mimic and/or stabilize the interaction of inhibitor apolipoprotein-E (ApoE) with Clq, and their uses in the prevention or treatment of Clq-related diseases or conditions, such Alzheimer's disease (AD), atherosclerosis, and ageing.
  • Clq complex 1 protein q
  • ApoE inhibitor apolipoprotein-E
  • Clq is the first subcomponent of the Cl complex of the classical pathway of complement activation, the complement cascade (CCC).
  • CCC complement cascade
  • Several functions have been assigned to Clq, which include antibody-dependent and independent immune functions, and are considered to be mediated by Clq receptors present on the effector cell surface. Patients suffering from Lupus erythematosus often have deficient expression of Clq. Clq may also play a central role in the aging of cells, as Clq activates canonical Wnt signaling and promotes aging-related phenotypes.
  • ApoE Human Apolipoprotein-E
  • AD Alzheimer's disease
  • LOAD late onset AD
  • atherosclerosis is the most common cause of death worldwide.
  • the human isoforms of ApoE i.e. ApoE2, ApoE3, and ApoE4 differ by amino acid residues 112 and 158 located outside of the N-terminal receptor- and C-terminal lipid binding domains, respectively, yielding proteins with distinct impacts on tissue homeostasis.
  • the presence of two copies of the E4 allele increases risk by ⁇ l2-fold whereas E2 allele is associated with a ⁇ twofold decreased risk for AD.
  • Roumenina L., et al. disclose low molecular weight disulphate compounds that bind to the globular (gClq) domain.
  • Betulin disulphate (B2S) and 9,9-bis(4'-hydroxyphenyl)fluorene disulphate (F2S) inhibit the interaction of Clq and its recombinant globular modules with target molecules IgGl, C-reactive protein (CRP) and long pentraxin 3 (PTX3).
  • Hajishengallis G, et al. disclose a view of new and previously unanticipated functions of complement and how these affect immunity and disease pathogenesis. They mention neuro inflammation, epilepsy, AD, Ischemia-reperfusion injury, AMD, osteoarthritis, Gaucher‘s disease, and cancer.
  • Haskard DO et al. (in: The role of complement in atherosclerosis. Curr Opin Lipidol. 2008 Oct;l9(5):478-82) disclose that Clq has been found to play a protective role in early lesion formation in LDL receptor deficient mice, and Crry-Ig and soluble Cl inhibitor have both been shown to have therapeutic effects in models of vascular injury in ApoE deficient mice.
  • AD Alzheimer’s disease
  • complement activation by the classic and lectin pathway may be protective by removing apoptotic cells and cell debris from atherosclerotic plaques
  • activation of the complement cascade by the alternative pathway and beyond the C3 convertase with formation of anaphylatoxins and the terminal complement complex may be proatherogenic and may play a role in plaque destabilization leading to its rupture and the onset of acute cardiovascular events.
  • US 2015-337030 discloses methods for the treatment and/or prevention of a neurodegenerative disorder, such as non-familial late-onset Alzheimer's disease (LOAD), by using an inhibitor of APOE4, such as an antibody inhibitor, or by using an excess of APOE3 protein.
  • a neurodegenerative disorder such as non-familial late-onset Alzheimer's disease (LOAD)
  • LOAD non-familial late-onset Alzheimer's disease
  • APOE4 such as an antibody inhibitor
  • new targets for the therapy of diseases or conditions such as Alzheimer's disease (AD), atherosclerosis, and ageing are desired, in particular in the context of cellular functions mediated directly or indirectly by the complement cascade. It is therefore an object of the present invention, to provide these new targets and to employ these targets in the development of new and effective therapies.
  • Other objects and aspects of the present invention will become apparent to the person of skill upon reading the following description of the invention.
  • a compound for use in the treatment or prevention of a disease or condition in a mammalian subject wherein the compound is a modulator of the expression, function, stability of Clq or a variant thereof, and in particular mimics and/or stabilizes the interaction of ApoE with Clq or variants thereof, and wherein said disease or condition is selected from neuronal diseases, cardiovascular diseases, kidney diseases, and ageing.
  • the treatment of the present invention is preferably a method for treating a disease in a subject, comprising a step of administering to the subject a therapeutically effective amount of a modulator of the expression, function, stability of Clq or a variant thereof, and in particular mimics and/or stabilizes the interaction of ApoE with Clq or variants thereof.
  • the inventors explored the roles of ApoE in the aging choroid plexus (ChP) and the aorta of mammals, in particular mice.
  • the data demonstrate that ApoE is a classical complement cascade (CCC) inhibitor by binding to the Cl complex, and that the CCC-ApoE axis impacts neuronal diseases, cardiovascular diseases and ageing, such as AD and atherosclerosis.
  • CCC complement cascade
  • Clq provides a new target for the identification of modulators that mimic and/or stabilize the function of Clq in the context of the ApoE-Clq interaction.
  • modulators should prove beneficial for the treatment or prevention of a disease or condition in a mammalian subject, such as a neuronal disease, cardiovascular disease kidney disease, and ageing, such as AD and atherosclerosis.
  • Other diseases to be treated and/or prevented are IgA nephropathy, vasculitis, SLE nephritis, AMD, trauma, sepsis, ARDS, and SIRS.
  • US 2009-117098 discloses a method of treating glaucoma or elevated intraocular pressure comprising administering a pharmaceutically effective amount of a composition comprising a Complement Clq inhibitor. No specific inhibitor or the function of ApoE binding is mentioned.
  • WO 2005-002513 generally discloses the use of agents that bind and/or inhibit classical complement C 1 subcomponent, Clq, as well as methods of their use. Particularly disclosed is a monoclonal antibody.
  • Clq-protein or“protein of Clq” as used in context of the herein disclosed invention shall preferably pertain to the protein (such as a full-length protein, fusion protein or partial protein) consisting of the subunit chains Clqa, Clqb, and Clqc, comprising the sequence of the Complement Clq subcomponent subunit A (UniProtKB - P02745 for human, SEQ ID NO. 1, UniProtKB - P98086 for mouse, SEQ ID NO. 2); Complement Clq subcomponent subunit B (P02746 for human SEQ ID NO. 3, P14106 for mouse SEQ ID NO. 4); and Complement Clq subcomponent subunit C (P02747 for human SEQ ID NO. 5, Q02105 for mouse SEQ ID NO. 6).
  • the terms shall also refer to the individual or combined one, two or three subunits, as long as they are suitable to maintain Clq function.
  • the terms shall also refer to a protein comprising the amino acid sequence according to SEQ ID NO: 1 to 6 with any protein modifications.
  • Such protein modifications preferably do not alter the amino acid sequence of the polypeptide chain, but constitute a functional group, which is conjugated to the basic amino acid polymer chain.
  • Protein modifications in context of the invention may be selected from a conjugation of additional amino acid sequences to the amino acid chain, such as ubiquitination, sumolation, neddylation, or similar small protein conjugates.
  • Other protein modifications include, but are not limited to, glycosylation, methylation, lipid- conjugation, or other natural or artificial post-translational modifications known to the skilled person.
  • a variant of Clq is, in some embodiments, a protein comprising an amino acid sequence having at least 60%, 70%, 80%, 90%, preferably at least 80% such as at least 90% sequence identity to SEQ ID NO: 1 to 6, and most preferably at least 95% (such as at least 98%) sequence identity to SEQ ID NO: 1 to 6.
  • the variant of Clq comprises an amino acid sequence with at least 80% sequence identity to SEQ ID NO: 1 to 6.
  • the terms“identical” or percent“identity”, when used anywhere herein in the context of two or more nucleic acid or protein/polypeptide sequences, refer to two or more sequences or subsequences that are the same or have (or have at least) a specified percentage of amino acid residues or nucleotides that are the same (i.e., at, or at least, about 60% identity, preferably at, or at least, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93% or 94%, identity, and more preferably at, or at least, about 95%, 96%, 97%, 98%, 99%, or higher identity over a specified region - preferably over their full length sequences - , when compared and aligned for maximum correspondence over the comparison window or designated region) as measured using a sequence comparison algorithms, or by manual alignment and visual inspection (see, e.g., NCBI web site).
  • the percentage identity can be determined by the Blast searches supported at the Human Olfactory Data Explorer (eg, https://genome.weizmann.ac.il/cgi-bin/horde/blastHorde.pl); in particular for amino acid identity, those using BLASTP 2.2.28+ with the following parameters: Matrix: BLOSUM62; Gap Penalties: Existence: 11, Extension: 1; Neighboring words threshold: 11; Window for multiple hits: 40.
  • “Stabilizing” in the context of the present invention with respect to binding of ApoE to Clq shall mean an increase, preferably a substantial increase, of the binding affinity and/or the half-life of the complex of the two proteins or fragments thereof. Detecting and measuring of a stabilizing (with or without other molecules involved) are known in the art, and reviewed, for example, in Andrei SA, et al, Stabilization of protein-protein interactions in drug discovery, Expert Opin Drug Discov. 2017 Sep;l2(9):925-940.
  • a variant of Clq is, in certain embodiments, a functional variant of Clq protein.
  • the variant of Clq is selected from the group consisting of an ortho log or paralog of Clq, and a functional fragment of a Clq protein.
  • the compound for use according to the present invention wherein the variant of Clq is selected from the group consisting of an ortho log or paralog of Clq, and a functional fragment of a Clq protein.
  • ortholog refers to homologs in different species that evolved from a common ancestral gene by speciation. Typically, orthologs retain the same, essentially the same or similar function despite differences in their primary structure (mutations).
  • paralog refers to homologs in the same species that evolved by genetic duplication of a common ancestral gene. In many cases, paralogs exhibit related but not always similar function.
  • splice variant refers to a related protein expressed from the same genomic locus as a parent protein, but having a different amino acid sequence based on a different exon composition due to differential splicing of the transcribed R A.
  • the term“subject” or“patient” preferably refers to a mammal, such as a mouse, rat, guinea pig, rabbit, cat, dog, monkey, or preferably a human, for example a human patient.
  • the subject of the invention may be at danger of suffering or is suffering from neuronal diseases, cardiovascular diseases and ageing, such as AD and atherosclerosis.
  • a more detailed description of medical indications relevant in context of the invention is provided herein below.
  • the compound/modulator for use according to the present invention wherein said compound is an inhibitor or antagonist of said expression, function, stability of Clq or a variant thereof, and/or mimics and/or stabilizes the interaction of ApoE with Clq or variants thereof.
  • the term“modulator” in context of the present invention shall include inhibitors/antagonists.
  • a preferred embodiment of the invention pertains to inhibitors/antagonists as modulators of the expression, function, stability of Clq or a variant thereof.
  • Preferred modulators are, in context of the invention, inhibitors or antagonists that inhibit (e.g., impair or interfere with) Clq or its variant’s expression, function and/or stability, in particularly specifically and/or selectively, in neuronal cells and tissues showing a pathological aberration, senescence, impaired growth or survival.
  • the modulator for use according to the present invention is an antigen binding construct, such as, for example, an antibody, antibody-like molecule or other antigen binding derivative, or an antigen binding fragment thereof), that binds and preferably specifically Clq or a variant thereof, and thereby mimics the preferably specific interaction of Clq with ApoE.
  • an antigen binding construct such as, for example, an antibody, antibody-like molecule or other antigen binding derivative, or an antigen binding fragment thereof
  • the modulator for use according to the present invention is a nucleic acid, such as, for example an anti-sense nucleotide molecule such as a siRNA or shRNA molecule that binds to a nucleic acid that encodes or regulates the expression of: (i) Clq or a variant thereof, or (ii) a gene that controls the expression, function and/or stability of Clq or a variant thereof.
  • an anti-sense nucleotide molecule such as a siRNA or shRNA molecule that binds to a nucleic acid that encodes or regulates the expression of: (i) Clq or a variant thereof, or (ii) a gene that controls the expression, function and/or stability of Clq or a variant thereof.
  • modulators in particular inhibitors/antagonists
  • function and/or stability of Clq or a variant thereof and in particular that mimic and/or stabilizes the preferably specific interaction of Clq with ApoE are in certain embodiments the following specific molecules and/or molecular classes.
  • a nucleic acid such as a DNA or RNA, for example an antisense or inhibitory DNA or RNA, a ribozyme, an RNA or DNA aptamer, RNAi, siRNA, shRNA and the like, including variants or
  • CRISPR/Cas9 mediated gene editing approaches are known to the skilled artisan and for example reviewed in Wiles MV et al.:“CRISPR-Cas9-mediated genome editing and guide RNA design.”, (Mamm Genome. 2015 0ct;26(9-l0):50l-l0) or in Savic N and Schwank G: “Advances in therapeutic CRISPR/Cas9 genome editing.” (Transl Res. 2016 Feb; 168:15-21).
  • said compound is selected from the group consisting of a peptide library, a combinatory library, a cell extract, in particular a plant cell extract, a "small molecular drug", an antisense oligonucleotide, an siRNA, an mRNA, and an antibody or fragment thereof specifically mimicking and/or stabilizing the binding of ApoE to Clq.
  • An anti-sense nucleotide molecule can, by virtue of it comprising an anti-sense nucleotide sequence, bind to a target nucleic acid molecule (e.g. based on sequence complementarity) within a cell and modulate the level of expression (transcription and/or translation) of Clq (or of a variant of Clq), or it may modulate expression of another gene (e.g. ApoE) that controls the expression, function and/or stability of Clq (or the variant thereof).
  • a target nucleic acid molecule e.g. based on sequence complementarity
  • an isolated antigen binding construct according to the present invention that is capable of specifically binding to Clq or a variant thereof, wherein said antigen binding construct mimics, preferably specifically mimics, and/or stabilizes the interaction of ApoE or variants with Clq or variants thereof.
  • Preferred antigen binding constructs are antibodies and antibody-like constructs.
  • antibody includes, but is not limited to, genetically engineered or otherwise modified forms of immunoglobulins, such as intrabodies, chimeric antibodies, fully human antibodies, humanized antibodies, antibody fragments, and heteroconjugate antibodies (e.g., bispecific antibodies, diabodies, triabodies, tetrabodies, etc.).
  • antibody includes cys-diabodies and minibodies.
  • antibody or “antibody like constructs” is also envisioned as, bi-specific antibodies, diabodies, scFv fragments, chimeric antibody receptor (CAR) constructs, diabody and/or minibody embodiments, unless explicitly denoted otherwise.
  • a preferred embodiment of the invention pertains to a monoclonal antibody as an (isolated) antigen binding construct.
  • An antibody of the invention may be an IgG type antibody, for example having any of the IgG isotypes.
  • the antigen binding construct such as an antibody
  • the antigen binding construct is non-natural and/or is not a product of nature.
  • the antigen binding construct may be a non-natural antigen binding construct, such as a synthetic, modified or recombinant antigen binding construct.
  • the antigen binding construct may be first generated following non natural immunization of a (species of) mammal; such as by immunization with an antigen to which such (species of) mammal is not exposed in nature, and hence will not have naturally raised antibodies against.
  • an antigen binding construct refers to a polypeptide that is purified from proteins or polypeptides or other contaminants that would interfere with its therapeutic, diagnostic, prophylactic, research or other use.
  • An antigen binding construct according to the invention may be a recombinant, synthetic or modified (non natural) antigen binding construct.
  • nucleic acid or cells refers to a nucleic acid or cells that is/are purified from DNA, R A, proteins or polypeptides or other contaminants (such as other cells) that would interfere with its therapeutic, diagnostic, prophylactic, research or other use, or it refers to a recombinant, synthetic or modified (non-natural) nucleic acid.
  • a “recombinant” protein/polypeptide or nucleic acid is one made using recombinant techniques. Methods and techniques for the production of recombinant nucleic acids and proteins are well known in the art.
  • A“functional variant” of ApoE or Clq is a variant of, such as a fragment of, the protein of ApoE or Clq that provides, possesses and/or maintains one or more of the herein described functions/activities of the non- variant protein of ApoE or Clq, in particular mimics and/or stabilizes the specific binding of the two proteins to each other.
  • Another aspect of the invention relates to a monoclonal antibody, or a binding fragment thereof, binding to and preferably mimicking and/or stabilizing, ApoE binding to Clq, or vice versa, or the variants of these proteins. Therefore, the present invention relates to the use of Clq as a novel target for the generation of modulating, such as stabilizing, antibodies directed against the respective protein that mimic the binding of ApoE, and thereby modulate the function of these binding partners, specifically Clq.
  • the generation of such antibodies is as such a standard procedure for the skilled artisan.
  • An antibody of the present invention may be a mouse, rat, rabbit, horse, goat, antibody, or a humanized or chimeric antibody.
  • a method for identifying a compound suitable for the prevention or treatment of a disease characterized by an undesired function of Clq or a variant thereof, the method comprising the steps of a) contacting at least one of Clq or a variant thereof, a Clq binding fragment of ApoE or a variant thereof, and an ApoE binding fragment of Clq or a variant thereof and/or a cell expressing Clq or a variant thereof with at least one candidate compound that potentially modulates and preferably mimics and/or stabilizes the interaction of ApoE with Clq in a cell, and b) identifying a binding of said candidate compound to Clq.
  • ApoE-protein or“protein of ApoE” as used in context of the herein disclosed invention shall pertain to a protein (such as a full-length protein, fusion protein or partial protein) comprising the sequence of the apo lipoprotein E protein (UniProtKB - E7ERP7 for human, SEQ ID NO. 7, UniProtKB - Q6GTX3-1 for mouse, SEQ ID NO. 8) including the variant amino acids that make up the isoforms, in particular isoform 3 (ApoE3), e.g. amino acid changes at positions 112 and/or 158, respectively (cysl l2, argl58) in the human protein, There are no variants in mouse ApoE.
  • the terms shall also refer to a protein comprising the amino acid sequence according to SEQ ID NO: 7 or 8 with any protein modifications.
  • Such protein modifications preferably do not alter the amino acid sequence of the polypeptide chain, but constitute a functional group, which is conjugated to the basic amino acid polymer chain.
  • Protein modifications in context of the invention may be selected from a conjugation of additional amino acid sequences to the amino acid chain, such as ubiquitination, sumolation, neddylation, or similar small protein conjugates.
  • Other protein modifications include, but are not limited to, glycosylation, methylation, lipid- conjugation, or other natural or artificial post-translational modifications known to the skilled person.
  • a preferred variant of ApoE is a Clq binding fragment, in particular a functional fragment, which also constitutes a compound according to the present invention, i.e. mimics the function of the full-length ApoE regarding the ApoE/Clq interaction in vivo.
  • a preferred method uses an assay comprising ApoE binding to Clq as a control, in order to identify suitable candidate compounds.
  • said assay is a competitive assay, wherein a modulation is selected from a decrease of the (preferably specific) binding of ApoE or a fragment or variant thereof to Clq or a fragment or variant thereof as measured in the presence of said candidate compound when compared to a control.
  • a modulation is selected from a decrease of the (preferably specific) binding of ApoE or a fragment or variant thereof to Clq or a fragment or variant thereof as measured in the presence of said candidate compound when compared to a control.
  • said assay is a combined assay, wherein a modulation is selected from an increase of the (preferably specific) binding of ApoE or a fragment or variant thereof to Clq or a fragment or variant thereof as measured in the presence of said candidate compound when compared to a control. This will allow to select compounds that specifically stabilize/promote the ApoE binding/function on Clq.
  • the reduction (or enhancement) of expression, function and/or stability of Clq or the Clq (or of the variant thereof) and/or the binding of ApoE to Clq is, preferably, identified by reference to a control method, for example one practiced in the absence of any candidate compound, or with compound having a known effect on such expression, function and/or stability (such as a positive or negative control).
  • said cell can be selected from the group of neuronal or vascular cells, recombinant host cells suitably expressing ApoE and Clq or binding fragments or variants thereof, yeast cells, and recombinant bacterial cells. Further preferred is a method according to the present invention, wherein said ApoE fragment binds to the Clq stalk, and comprises for example the amino acids 139-152 of the human ApoE polypeptide.
  • the method according to the present invention as described herein is thus suitable for the identification of compounds that can interact with and/or mimic the binding of ApoE to Clq, and/or activities of Clq, and thus to identify, for example, inhibitors, competitors or modulators of the Clq function and downstream complement amplification loop, in particular, inhibitors, competitors or modulators or mimics and/or stabilizes the binding of ApoE.
  • Preferred are compounds that mimic and/or stabilize the binding of ApoE to Clq.
  • Another aspect is directed at compounds that modulate the expression of Clq in a cell/in cells.
  • Another aspect is directed at a method according to the present invention, further comprising testing said compound(s) as identified for its activity to specifically bind to the Clq stalk, for example the amino acids 139-152 of the human ApoE polypeptide.
  • Respective assays are known to the person of skill, and can be taken from the respective literature.
  • Another aspect is directed at a method according to the present invention, further comprising testing said compound(s) as identified for its activity to revert neuronal diseases, inflammation, AD, kidney diseases, atherosclerosis or the effects of ageing.
  • Respective assays are known to the person of skill, and can be taken from the respective literature.
  • contacting in the present invention means any interaction between the potentially binding substance(s) (candidate compound) with Clq, whereby any of the two components can be independently of each other in a liquid phase, for example in solution, or in suspension or can be bound to a solid phase, for example, in the form of an essentially planar surface or in the form of particles, pearls or the like.
  • a multitude of different potentially binding substances are immobilized on a solid surface like, for example, on a compound library chip and Clq (or a functional part thereof) is subsequently contacted with such a chip.
  • Measuring of binding of the compound to Clq can be carried out either by measuring a marker that can be attached either to the protein or to the potentially interacting compound.
  • Suitable markers are known to someone of skill in the art and comprise, for example, fluorescence markers.
  • the candidate compound is selected from a polypeptide, peptide, glycoprotein, a peptidomimetic, an antigen binding construct (for example, an antibody, antibody-like molecule or other antigen binding derivative, or an or antigen binding fragment thereof), a nucleic acid such as a DNA or RNA, for example an antisense or inhibitory DNA or RNA, a ribozyme, an RNA or DNA aptamer, RNAi, siRNA, shRNA and the like, including variants or derivatives thereof such as a peptide nucleic acid (PNA), a genetic construct for targeted gene editing, such as a CRISPR/Cas9 construct and/or a guide nucleic acid (gRNA or gDNA) and/or tracrRNA.
  • a nucleic acid such as a DNA or RNA, for example an antisense or inhibitory DNA or RNA, a ribozyme, an RNA or DNA aptamer, RNAi, siRNA, shRNA and the like, including
  • the thus selected binding compound is then in a preferred embodiment modified in a further step.
  • Modification can be effected by a variety of methods known in the art, which include without limitation the introduction of novel side chains or the exchange of functional groups like, for example, introduction of halogens, in particular F, Cl or Br, the introduction of lower alkyl groups, preferably having one to five carbon atoms like, for example, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n- pentyl or isopentyl groups, lower alkenyl groups, preferably having two to five carbon atoms, lower alkynyl groups, preferably having two to five carbon atoms or through the introduction of, for example, a group selected from the group consisting of NH 2 , N0 2 , OH, SH, NH, CN, aryl, heteroaryl, COH or COOH group.
  • the thus modified binding substances are then individually tested with a method of the present invention, i.e. they are contacted with Clq (or a functional part thereof) and subsequently binding of the modified compounds to the Clq (or a functional part thereof) polypeptide is measured.
  • both the binding per se can be measured and/or the effect of the function of the Clq (or a functional part thereof) like, e.g. the binding to ApoE and/or the enzymatic activity of the polypeptide can be measured.
  • the steps of selecting the binding compound, modifying the binding compound, contacting the binding compound with Clq (or a functional part thereof) polypeptide and measuring the binding of the modified compounds to the protein can be repeated a third or any given number of times as required.
  • the above described method is also termed “directed evolution” since it involves a multitude of steps including modification and selection, whereby binding compounds are selected in an "evolutionary” process optimizing its capabilities with respect to a particular property, e.g. its binding activity, its ability to activate or modulate the activity of the Clq polypeptide, and/or its function as an ApoE mimicking and/or stabilizing compound.
  • Another aspect of the present invention relates to a method for manufacturing a pharmaceutical composition for treating or preventing neuronal diseases, cardiovascular diseases and ageing, comprising the steps of: performing a screening method according to the present invention, and formulating said compound as screened and identified into a pharmaceutical composition.
  • the interacting compound identified as outlined above which may or may not have gone through additional rounds of modification and selection, is admixed with suitable auxiliary substances and/or additives.
  • suitable auxiliary substances and/or additives comprise pharmacological acceptable substances, which increase the stability, solubility, biocompatibility, or biological half- life of the interacting compound or comprise substances or materials, which have to be included for certain routs of application like, for example, intravenous solution, sprays, band-aids or pills.
  • Carriers, excipients and strategies to formulate a pharmaceutical composition for example to be administered systemically or topically, by any conventional route, in particular enterally, e.g. orally, e.g. in the form of tablets or capsules, parenterally, e.g. in the form of injectable solutions or suspensions, topically, e.g. in the form of lotions, gels, ointments or creams, or in nasal or a suppository form are well known to the person of skill and described in the respective literature.
  • Another aspect of the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the compound for use according to the present invention and a pharmaceutically acceptable carrier, stabilizer and/or excipient.
  • Another aspect of the present invention pertains to a pharmaceutical composition for use in the prevention or treatment of neuronal diseases, cardiovascular diseases and ageing, in particular AD or atherosclerosis.
  • the pharmaceutical composition of the invention comprises a compound of the invention (i.e., a modulator of ApoE binding to Clq, or of a variant thereof as described herein), and a pharmaceutical acceptable carrier and/or excipient.
  • a pharmaceutical acceptable carrier and/or excipient is intended to include any and all solvents, solubilizers, fillers, stabilizers, binders, absorbents, bases, buffering agents, lubricants, controlled release vehicles, diluents, emulsifying agents, humectants, dispersion media, coatings, antibacterial or antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well-known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary agents can also be incorporated into the compositions.
  • the pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intrathecal, intra-arterial, intravenous, intradermal, subcutaneous, oral, transdermal (topical) and transmucosal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application, as well as comprising a compound of the invention as herein can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin; propylene glycol or other synthetic solvents; anti bacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfate; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • the pharmaceutical composition is formulated for sustained or controlled release of a compound of the invention (e.g., a compound mimicking and/or stabilizing the interaction of ApoE with Clq or a variant thereof).
  • a compound of the invention e.g., a compound mimicking and/or stabilizing the interaction of ApoE with Clq or a variant thereof.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polygly colic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • the materials can also be obtained commercially (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art.
  • Dosage unit form as used herein includes physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED 50 .
  • Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the dosage may vary depending upon the dosage form employed and the route of ad- ministration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • the pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • an “effective amount” is an amount of the compound(s) or the pharmaceutical composition as described herein that reduces on the expression and/or abundance of Clq, or mimics and/or stabilizes the binding of ApoE to Clq, such as specifically mimics and/or stabilizes the binding of ApoE to Clq.
  • the object of the present invention is solved by providing a screening tool for an agent for treating or preventing neuronal diseases, cardiovascular diseases and ageing, in particular a screening tool for screening for a compound that modulates the expression, the biological activity and/or the interaction of Clq, and preferably mimics and/or stabilizes the interaction of ApoE with Clq, comprising an isolated cell expressing Clq and/or expressing an ApoE binding fragment thereof, wherein said cell optionally expresses ApoE and/or an Clq binding fragment thereof.
  • the cell can be a prokaryotic or eukaryotic cell, and the expression construct can be present extrachromosomally or integrated into the chromosome.
  • the polypeptides can be expressed in the form of a fusion protein, for example together with an enzymatically active moiety as reporter-construct, in order to be able to detect the expression product.
  • Preferred host cells are derived from cells selected from the skeletal muscle, liver, adipose tissue, heart, pancreas, kidney, breast tissue, ovarian tissue, and/or hypothalamus.
  • a screening tool according to the present invention wherein said cell is selected from the group of neuronal cells, recombinant host cells expressing Clq and/or an ApoE binding fragment thereof, yeast cells, and recombinant bacterial cells, wherein said recombinant cell optionally expresses ApoE or the Clq binding fragment thereof.
  • the object of the present invention is solved by providing a screening tool for an agent for treating or preventing neuronal diseases, cardiovascular diseases and ageing, in particular a screening tool for screening for a compound that modulates the expression, the biological activity and/or the interaction of Clq in a cell, and preferably mimics and/or stabilizes the interaction of ApoE with Clq in a cell, wherein said cell as above is part of a non- human transgenic mammal, which preferably overexpresses ApoE and/or Clq optionally as a genetic reporter- construct.
  • transgenic mouse, rat, pig, goat or sheep wherein the reporter- construct is preferably expressed in cells selected from the nerves, vasculature, skeletal muscle, liver, adipose tissue, heart, pancreas, kidney, and/or hypothalamus of said animal.
  • Methods to produce these non-human transgenic mammal overexpressing ApoE and/or Clq and/or carrying a ApoE and/or Clq genetic reporter-construct are well known to the person of skill in the art.
  • Preferred is a tool, wherein said Clq binding fragment comprises the amino acids 139-152 of ApoE to bind to the Clq stalk.
  • compounds can be identified that modulate the expression of Clq in a cell.
  • the expression of Clq can be monitored using a genetic reporter-construct for Clq (order to analyze the translation efficiency and/or stability of the Clq polypeptide), for an example a fusion protein comprising a detectable fusion member (such as an enzymatic or fluorophoric group, or GFP as described herein), or the amount of mRNA as present in a cell can be measured, for example, by Northern blot.
  • the expression can also be analyzed and monitored by using chip-analysis or rtPCR.
  • Preferred compounds that modulate the expression of Clq in a cell are selected from specific antisense oligonucleotides, siRNAs, mR As or other preferably mutated nucleic acids encoding Clq.
  • These genetic elements can be used in order to provide/maintain the loss -of- function (e.g. by the truncations as identified) of Clq, or the ApoE binding thereof, in said cell.
  • Another preferred embodiment is the transfer of said genetic elements using gene therapy.
  • encompassed are viral constructs for the introduction of said genetic elements into said cells.
  • the "naked" nucleic acid can be introduced into the cell(s), e.g. by using particle-mediated technologies. Respective methods are well described in the literature and known to the person of skill.
  • the screening tool according to the present invention as described herein, wherein said ApoE and/or Clq and/or the variants or fragments thereof are labeled.
  • Fabels and methods for labeling are known to the person of skill, and can be enzymatic labels, dyes, fluorophores, and/or radioactive labels.
  • the present invention relates to the use of the tools according to the present invention as described herein for screening for a compound that modulates the expression, the biological activity and/or mimics and/or stabilizes the interaction of ApoE with Clq in a cell.
  • the inventors' data indicate that the interaction of ApoE with Clq can be used as a functional target for a drug (modulator) in order to prevent, treat and/or slow down the course of neuronal diseases, cardiovascular diseases, kidney disease, and ageing, in particular AD in mammals/humans.
  • a construct that mimics and/or stabilizes ApoE binding to Clq can be used as a drug as well.
  • another aspect of the present invention relates to a method for treating or preventing neuronal diseases, cardiovascular diseases and ageing in a patient, comprising administering to said patient an effective amount of a pharmaceutical composition according to the invention as described herein.
  • the attending physician will base a treatment on the compound as identified, and optionally also on other individual patient data (clinical data, family history, DNA, etc.), and a treatment can also be performed based on the combination of these factors.
  • This method of the present invention for example involves integrating individual diagnostic data with patient clinical information and general healthcare statistics to enable, for example, the application of personalized medicine to the patient. Significant information about drug effectiveness, drug interactions, and other patient status conditions can be used, too.
  • the neuronal disease or cardiovascular disease to be treated is AD or atherosclerosis.
  • an active agent (modulator) is administered in form of a pharmaceutical composition, such as an antibody, nucleotide or a compound mimicking the ApoE inhibition of Clq.
  • Treating is meant to include, e.g., preventing, treating, reducing the symptoms of, or curing the disease or condition, i.e. AD, inflammation, atherosclerosis, or ageing.
  • prevention and“preventing” as used herein are used according to their ordinary and plain meaning to mean“acting before”.
  • those terms refer to administration or application of an agent, drug, or remedy to a subject or performance of a procedure or modality on a subject for the purpose of blocking the onset of a disease or health-related condition.
  • a subject that does not yet meet the clinical criteria or exhibit the symptoms of glaucoma, but does exhibit increases in intraocular pressure may be administered a composition of the present invention to prevent or delay the onset of glaucoma or perhaps reduce the severity of the condition.
  • the present invention demonstrates in mammals (mice) that ApoE attenuates ChP inflammation and atherosclerosis by inhibiting activation of the CCC through binding to Clq.
  • the inventors present a model in which ApoE binds with high-affinity to Clq which results in a decrease in local accumulation of proinflammatory anaphylatoxin C5a and alleviation from C5b-triggered injury.
  • the data also indicate that Clq- ApoE complexes may act as control nodes in human AD and atherosclerosis (see, e.g., Fig. 3).
  • Each of the currently known vital functions of the ChP requires rigid regulation to avoid brain injury and dysfunction (13-17).
  • the data are also consistent with the hypothesis that the diseased ChP is a gateway to allow circulating leukocytes to enter the brain parenchyma, and that blood-bom innate immune cells may promote AD plaque formation in the neocortex or atherosclerosis in peripheral arteries.
  • Clq- ApoE module impacts inflammation in prototypic inflammatory diseases: Clq- ApoE module formation required local activation of na ' ive Clq by any number of inflammatory stimuli including MDA-LDL, oxLDL, Igs, and Ab fibrils; the frequency of the Clq- ApoE module was markedly reduced; the binding affinity of Clq to ApoE was lO-lOOOfold higher than those of other long-established binding partners of Clq or of ApoE; and the three human iso forms of ApoE and its murine counterpart inhibited the CCC activated by MDA-LDL, oxLDL, and Ab fibrils but not by native LDL or soluble Ab.
  • the inventors conclude that the Clq- ApoE module also fine-tunes the activity of the CCC in common inflammatory diseases (Fig. 3).
  • Inflammatory mediators such as neuritic Ab plaques and hyperlipidemia incite unrelenting activation of the innate immune system of which both Clq and ApoE are ancient and central components.
  • the chronicity of unresolvable inflammation implies ongoing activation of the CCC for extended periods of time and the involvement of adaptive immunity, consistent with a significant lymphocyte component in ChP infiltrates and atherosclerotic plaques in ApoE-/- mice.
  • the inventors data also extend beyond the identification and role of the Clq- ApoE module in prototypic human inflammatory diseases. All chronic inflammatory diseases are associated with activation of one or more complement pathways and that both Clq and ApoE are induced in response to multiple types of acute and chronic stress-inducing types of tissue injury (1, 3-6, 20, 30, 31, 33). The inventors also consider the body of data in line with recent research in which complement constituents have been demonstrated to function as major regulators of basic cellular processes including inflammasome activation and skewing of T cell immunity (32).
  • the BBB is a well-recognized critical gateway to allow circulating leukocytes to enter the brain to promote neuritic AD plaque formation; and endothelial cell injury of large arteries may play similar roles to promote atherosclerotic lesion buildup (3-6, 8-11, 13- 17, 29-33).
  • each of the two building blocks of the module, Clq and ApoE may be subject to stress-dependent regulations which may not necessarily occur in parallel but may be dictated by independent events.
  • stress-dependent regulations may not necessarily occur in parallel but may be dictated by independent events.
  • proteins are ancient molecules having evolved from unicellular organisms they acquired functional domains that may have emerged from independent evolutionary pressures.
  • the ultimate function of the module in AD and atherosclerosis appears to regulate inflammation putting the immune system in a driver seat position.
  • the immune system itself is subject to systemic and local controls independent of the module itself including immune senescence, nature of injury, and response of the inflicted tissue parenchyma, and many more.
  • the Clq-ApoE module may have two faces in disease acquiring a dichotomic nature.
  • the data as produced for the present invention further indicate that the activity of the classical complement cascade (CCC) initiating molecule, i.e. Clq, exists in three states in vitro and in vivo:
  • CCC classical complement cascade
  • Clq is activated on specific surfaces like modified self surfaces (apoptotic cells) for opsonization of dead cells and enhancing phagocytosis without inducing inflammation.
  • Clq is regulated and the Clq-ApoE complex forms.
  • the Clq molecule as found in human blood or as produced by cells can potentially always activate the classical complement pathway either in response to infections or on specific surfaces of self-cells. Infectious microbes are clear activator surfaces, and no regulation is wanted until the microbe is removed. This is different in homeostasis. On self-surfaces opsonisation (complement activation on level of C3 for opsonization) is desired, but no activation on the C5 level (inflammation). This is the reason why Cl requires regulation. In cases of no clearance of apoptotic cells or accumulation of lipids or plaques, the regulation is overwhelmed, and C5 convertases form, leading to inflammation. The inventors performed co-immunoprecipitation experiments (see below) and the in vitro and data strengthens the previous conclusions regarding the role of ApoE in directly regulating the classical complement pathway by binding to activated Clq.
  • ApoE inhibits CCC activation but not the alternative pathway.
  • ApoE isoforms ApoE2, ApoE3, or ApoE4 were incubated in normal human serum (NHS), which was activated either via CCC buffer (left) (1% in GVB++) or alternative pathway buffer (right) (20% in MgEGTA); and lysis of sheep or rabbit erythrocytes by TCC was followed by measuring released hemoglobin at 415 nm.
  • AmoE was incubated with NHS in GVB++ buffer or Mg-EGTA buffer or with Clq-deficient serum in GVB++ to activate the alternative and lectin pathways. Survival of E. coli was analyzed counting colony forming units. Survival of E. coli in normal serum was set as 10%.
  • ApoE isoforms inhibit the CCC at the level of TCC and C4b.
  • ApoE isoforms in NHS were added to IgM-coated microtiter plates and TCC or C4b deposition was determined by specific antibodies, respectively
  • Binding of Cl, Clq, Cls, and Clr to ApoE iso forms was determined by bio layer interferometry
  • e The binding affinities of ApoE iso forms and Cls to Clq were determined by biolayer interferometry.
  • ApoE proteins and Cls were biotinylated, immobilized on streptavidin-coated sensors, and Clq binding was determined by measuring changes of optical thickness on the sensor
  • the ApoE-Clq interaction is dependent on Ca2+.
  • Figure 3 OL-ApoE-Clq complexes are involved in atherosclerosis.
  • FIG. 4 Example of pharmaceutical construct (“Affl”) according to the invention. AFF1 inhibits complement activation
  • A Schematic composition of AFF1.
  • the ApoE binding region that binds to Clq is fused to the regulatory region of factor H (SCR1 to SCR4), the dimerization region of FHR1 (SCR1 to SCR2) and a Histidine tag for purification.
  • B Recombinant expression of AFF1 in Pichia pastoris and purification by nickel chelate chromatography revealed a protein with a mobility of 40 kDa (lanes 2,3 and 4 silver staining). Detection of the FHR1 part by monoclonal antibody JHD confirms presence of FHR1 (SCR1 and SCR2)(lanes 5-7, Western blot).
  • C Binding of AFF1 to Clq.
  • AFF1 was coated to a microtiter surface and binding of increasing amounts of Clq (10 to 200 nM) were followed by ELISA. A representative assay with three replicates is shown.
  • D AFF1 inhibits C3b deposition via the alternative pathway in normal human serum (NHS).
  • AFF1 100 or 200 nM was added to 20 %NHS EGTA buffer, which inhibits the classical pathway but not eh alternative one.
  • the serum with AFF1 was added to an IgM coated microtiter plate and subsequent deposition of C3b was determined by ELISA.
  • Factor H shows also inhibition of the alternative pathway. Heat inactivation of serum (iNHS) inhibits complement activation.
  • a representative assay with triplicates is shown.
  • FIG. 5 Co-immunoprecipitation of Clq-ApoE complexes, (a) Anti Clq antisera precipitate Clq-ApoE complexes composed of purified proteins with activated Clq but not with inactivated Clq from normal human serum (NHS) (left and middle panels) (b) Anti- ApoE antisera precipitate Clq-ApoE complexes but not complexes from NHS in which Clq is present in its inactive form.
  • Anti Clq monoclonal antisera (Invitrogen, clone 9A7; in left and middle panels) or anti ApoE monoclonal antisera (Invitrogen, clone 1H4) were immobilized on protein G-coupled Dynabeads (Complement Technology).
  • Clq-deficient serum (dNHS, Complement Technology, 2%), NHS (2%), NHS with added with activated purified Clq (Complement Technology) (NHS+Clq), or NHS with purified ApoE (NHS+ApoE), and activated purified Clq with purified ApoE (Merck) (Clq + ApoE) (lanes 2-5 in a and b) were incubated with antisera-coated Dynabeads, then eluting fractions were examined for Clq and ApoE precipitate; purified Clq was added directly to the first lane of each gel as size marker for WB (Clq in lane 1).
  • NHS or anti-Clq with NHS were incubated with no pre-coated Dynabeads (NHS in lane 6 in a, or anti Clq plus purified ApoE in b, respectively) were used as controls. Proteins were eluted with glycine buffer (pH 2.7), separated by SDS-PAGE and immunoblotted using goat anti-Clq antiserum (Complement Technology) or goat anti ApoE antiserum (Merck) and rabbit anti goat secondary antibody (Dako).
  • FIG. 6 Clq-ApoE complexes are formed on apoptotic cells in vitro.
  • Apoptotic THP-l cells ATCC® TIB-202TM, UV light for 2h
  • dNHS Clq-depeleted serum
  • Cells were incubated in NHS or Clq-depeleted serum (dNHS) (each 1%) (Complement Technology, Texas, USA) for 30 minutes.
  • Cells were blocked and treated with rabbit anti ApoE antiserum (25 pg/ml; Acris Antibodies, Herford, Germany) and mouse anti-Clq antiserum (25 pg/ml; Thermo Fisher Scientific, Massachusetts, USA).
  • FIG. 7 Ca 2+ -dependent binding of ApoE to Clq.
  • Plasma-purified Clq was coated on a sensor chip (CM5) and plasma-derived ApoE (62-1000 nM) was injected into the fluid phase (75 mM NaCl, 5 mM HEPES, 1 mM CaCl 2 ). Real-time binding of ApoE to Clq was followed using biosensor analyses
  • Binding of ApoE to Clq is reduced in a dose-dependent manner with increasing amounts of EGTA (0.1-3 mM).
  • Figure 8 Interaction of ApoE with Clq stalks as assayed by EM.
  • ApoE molecules were labeled with 6 nm-sized nanogold particles and incubated with commercial purified activated Clq.
  • Three examples of Clq-ApoE complexes are displayed.
  • Typical Clq molecules with its stalk and 6 globular heads and gold- ApoE binding to the stalk of Clq are shown in enlarged insets.
  • Complexes (white arrows) are schematically encircled in the boxes below to outline the globular heads of Clq surrounding gold-labeled ApoE. Bars represent 40 nm.
  • Figure 9 shows that Clq-ApoE complexes form in the atherosclerotic plaques.
  • SEQ ID NO. 7 (ApoE human):
  • SEQ ID NO. 8 (ApoE mouse):
  • the ChP is the major intracranial neuroimmuno logical interface which produces the cerebrospinal fluid (CSF), forms the blood-CSF barrier, exchanges signals between the brain and the circulation, and is the principal gateway for blood-bom leukocytes to infiltrate the central nervous system in inflammatory and degenerative brain diseases (12-17).
  • CSF cerebrospinal fluid
  • Complement components C2, C3, C3b, C4, C4b, Clq, Cls, Factor H, Factor I, and C4BP as well as all primary antibodies were purchased from Complement Technology.
  • ApoE peptides were generated by Peptide 2.0: ApoE 30-40 LGRFWDYLRWV (SEQ ID No. 9); ApoE 75-85 YKSELEEQLTPV (SEQ ID No. 10); ApoE 139-152 SHLRKLRKRLLRDA (SEQ ID No. 11); ApoE 210-232 WGERLRARMEEMGSRTRDRLDEV (SEQ ID No. 12).
  • LDL and malone dialdehyde- modified LDL were from Cell Biolabs, copper oxidized LDL (oxLDL) from Thermo Fisher (L34357), poA from Athens Research & Technology, vitronectin (Vnt) from Coming. Ab and Ab fibrils from GenSript and HRP-coupled polyclonal antibody against goat and rabbit were from DakoCytomatio. Recombinant EfB was expressed as described (Koch, T. K. et al. Staphylococcus aureus proteins Sbi and Efb recruit human plasmin to degrade complement C3 and C3b. PLoS One 7, e47638 (2012).
  • Hemolytic assays were performed according to standard complement assays using sheep or rabbit erythrocytes that are sensitive to the complement terminal complex generated in human serum. Lysis of the erythrocytes by this thermal complex indicates complement activation and free hemoglobin is measured by absorbance.
  • Gram negative bacteria are sensitive for the terminal complement complex and become lysed. Similar to erythrocytes the terminal complement complex forms pores into the bacterial cell wall which kills the microbe. Efficiency of complement activation via the three pathways is tested by different buffers and by survival of the microbes upon plating the treated E. coli on agar plates. Different amounts of ApoE (0.1 - 1 mM) together with 0.2% NHS were pre-incubated for 10 mins at 37° C before adding E. coli pET200/D-TOPO (1000 cells per sample). After an incubation of 30 min at 37° C cells were plated to a LB-agar plate and cultivated o.n. at 37° C before counting the colony forming units.
  • the effect of ApoE on classical complement pathway C4b and C5b-9 deposition was analyzed by standard ELISA.
  • the classical pathway was activated by coating either IgM (2 pg/ml) or MDA-LDL (1 pg/ml) to a microtiter plate.
  • ApoE (0.25 - 2 mM) or Vnt (0.5 mM) were pre-incubated with 1% NHS in GVB++ buffer for 15 mins at 37° C and added to the coated plates for 1 h at 37° C. Complement activation with NHS alone was set as 100%.
  • Cofactor activity of ApoE was measured by incubating C4b (10 pg/ml) with Factor I (FI) (5 pg/ml), C4BP (20 pg/ml), ApoE3 (10 pg/ml) or combinations thereof (FI together with C4BP and FI together with increasing amounts of ApoE3 (0.1 - 100 pg/ml)) for 30 mins at 37° C.
  • C4b cleavage was analyzed by Western Blotting.
  • Coated proteins were incubated for 1 h at 37° C with C3, C3b, and Clq (0.1 mM) or Clq (0.02- 5.33 nM) or NHS (0.075 - 10%) in GVB++ buffer.
  • Calcium-dependent binding of Clq to ApoE was determined by diluting Clq in GVB++ buffer or in PBS and adding increasing amounts of EGTA (3 - 12 pM) (Sigma) with a fixed concentration of Clq (2 or 5 nM) to immobilized ApoE proteins. Binding force was analyzed incubating Clq or the LDLR (0.1 mM) with or w/o NaCl (0.5 M) or SDS (1%) on immobilized ApoE3.
  • Bio layer interferometry (BLItzTM, ForteBio) was used to examine binding of recombinant ApoE iso forms to Cl, C2, C4, Cl complex components Clq, Cls, Clr, and to MBL and the binding of Clq to ApoA.
  • Biotinylated proteins ligands
  • Streptavidin-coated biosensors VWR were hydrated for 10 mins in PBS with Calcium and Magnesium (PBS++, Lonza) before loading 20 pg/ml biotinylated ApoE2, 3 and 4 or ApoA for 120 sec.
  • MST Initial fluorescent analysis
  • Recombinant ApoE2, ApoE3 and ApoE4, plasma- purified ApoE3 or Cls were loaded for 120 sec via biotin onto the sensor. After 30 sec baseline, Clq (0.047 nM - 45.65 nM) or MBL (45.45 nM) was associated for 240 sec followed by a 240 sec dissociation step. Affinity values were generated by BLItzTM software analysis as an advanced kinetics experiment using MBL as reference value. Initial fluorescent analysis (NanoTemper) was used to determine the KD of the binding between Clq and ApoE 139-152.
  • Alexa 647-labeled ApoE 136-152 (10 nM) or ApoE 30-40 (30 nM) were incubated with different amounts of Clq (0.073 - 1196.8 nM) in PBS++ for 30' in the dark. After 10 mins centrifugation samples were transferred into standard capillaries and initial fluorescence was measured using a Monolith NT.l l5Pico (LED power 60%, MST power 20%). To test whether the observed fluorescence changes are due to a binding event, an SD-test was performed: The three samples with the highest and the lowest Clq concentration were centrifuged for 10' at l5000xg before removing the supernatant and adding SD-mix (4% SDS, 40 mM DTT). After an incubation step of 5 ' at 95°C, samples were transferred to capillaries and initial fluorescence was measured. The initial fluorescent analysis v2.0.2 was used to determine the KD.
  • coli remained viable upon ApoE-supplemented NHS challenge, but were killed when complement was activated via the lectin- or alternative pathways (Fig. 1 b), indicates that ApoE inhibits CCC activation, but not the alternative or lectin pathways. All three ApoE iso forms inhibited deposition of C4b and the terminal complement complex (TCC) by ⁇ 80% (Fig. 1 c) indicating that ApoEs act at a site of CCC initiation. Malondialdehyde- modified low-density- lipoprotein (MDA-LDL) binds to Clq and activates the CCC.
  • MDA-LDL Malondialdehyde- modified low-density- lipoprotein
  • Oxidized-LDL (oxLDL) was reported to activate the CCC21; and the inventors found that purified Clq bound MDA-LDL and oxidized-LDL (oxLDL) but not native LDL. Furthermore, ApoE inhibited the CCC and reduced C4b when the CCC was activated by oxLDL. Moreover amyloid fibrils but not soluble amyloid trigger C3b indicating complement activation.
  • the complement system is the central part of the human innate immune system and controls infections and many physiological reactions.
  • the cascade system is spontaneously activated in response to microbial surfaces as well as modified self surfaces and the balanced action of activators and inhibitors directs the newly formed effector components to target surfaces.
  • a number of soluble and membrane bound complement regulators are required to protect intact self-cells and tissue from the damaging effects of complement activation (34-36). Dysregulation of these inhibitors as well as over-activation of the complement system because of mutations/deletion in the corresponding genes have been shown to be associated with a number of different diseases such as atypical Hemolytic Uremic Syndrome, C3-Glomerulopathy or Age related Macular Degeneration (AMD) (34-37).
  • DAMPS danger associated molecular patterns
  • IgM circulating natural antibodies
  • subsequent complement activation recruits a number of immune cells that can clear but can also harm the neighbouring tissue and induce sterile inflammation.
  • the proposed inhibitor construct is composed of an ApoE-derived peptide that recruits the inhibitor to the activated Clq, the site of complement activation. Other regulatory functions may be added to such a construct as schematically indicated.
  • the inventors immobilized anti-Clq antisera on protein G-coated beads and incubated these beads with: 1. Serum from a normal human Clq-sufficient donor (normal human serum; NHS); or 2. Clq-deficient serum (dNHS); or 3. Purified Clq in its activated form with NHS (Clq+NHS); or 4. Purified Clq and purified ApoE (Clq+ApoE). Beads were washed and eluted proteins were separated by electrophoresis and immunoblots for Clq or ApoE proteins were generated.
  • Both Clq and ApoE can be precipitated by anti-Clq antibody, when beads were incubated with purified activated Clq with NHS (Clq+NHS) or purified activated Clq and purified ApoE (Clq+ApoE) indicating the formation of the Clq- ApoE complex under these experimental conditions ( Figure 5).
  • NHS Clq+NHS
  • ApoE purified activated Clq and purified ApoE
  • the inventors used cultured human apoptotic cells which provide a binding surface for the inactive Clq in NHS to initiate conversion to its activated form by changing its conformation:
  • the activated form of Clq in turn activates complement by enhancing opsonization with C3b/iC3b and its uptake by macrophages via CR3 (see also Gaboriaud C. et al, The crystal structure of the globular head of complement protein Clq provides a basis for its versatile recognition properties. J Biol Chem. 2003. 2l;278(47):46974-82; Major et al, Calcium- dependent conformational flexibility of a CUB domain controls activation of the complement serine protease Clr.
  • the inventors conclude that one ApoE molecule bound to one Clq molecule, more than 75% ApoE-gold is bound, there is a 1 :1 interaction of the binding partners, ApoE exclusively binds to the stalk but never to the tulip structures or globular heads of the molecule, gold-labeled peptide ApoEi39_i 52 has the same binding characteristics when compared to the full length ApoE protein, and that two different experimental methods show similar results.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Urology & Nephrology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Neurology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Neurosurgery (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Food Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Analytical Chemistry (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Cardiology (AREA)

Abstract

La présente invention concerne des modulateurs du complexe 1 de protéine q (C1q) qui imitent et/ou stabilisent de préférence l'interaction de l'apolipoprotéine E inhibitrice (ApoE) avec C1q, ainsi que leurs utilisations dans la prévention ou le traitement de maladies ou d'états liés à C1q, tels que la maladie d'Alzheimer (AD), l'athérosclérose, les maladies rénales et le vieillissement.
PCT/EP2019/050354 2018-01-09 2019-01-08 Modulateurs de c1q, en particulier de l'interaction de l'apoe avec c1q, et utilisations des modulateurs dans le traitement de maladies neuronales et de l'inflammation WO2019137922A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP19701776.7A EP3737472A1 (fr) 2018-01-09 2019-01-08 Modulateurs de c1q, en particulier de l'interaction de l'apoe avec c1q, et utilisations des modulateurs dans le traitement de maladies neuronales et de l'inflammation

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DE102018100377 2018-01-09
DE102018100377.3 2018-01-09

Publications (1)

Publication Number Publication Date
WO2019137922A1 true WO2019137922A1 (fr) 2019-07-18

Family

ID=65234522

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2019/050354 WO2019137922A1 (fr) 2018-01-09 2019-01-08 Modulateurs de c1q, en particulier de l'interaction de l'apoe avec c1q, et utilisations des modulateurs dans le traitement de maladies neuronales et de l'inflammation

Country Status (2)

Country Link
EP (1) EP3737472A1 (fr)
WO (1) WO2019137922A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022177724A1 (fr) * 2021-02-17 2022-08-25 University Of Rochester Procédés de traitement, de diagnostic et de prédiction du pronostic d'une sepsie

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005002513A2 (fr) 2003-06-16 2005-01-13 The Brigham And Women's Hospital, Inc. Inhibiteurs du complement c1q et leurs methodes d'utilisation
WO2008132723A2 (fr) * 2007-04-26 2008-11-06 Quark Pharmaceuticals, Inc. Délivrance thérapeutique de molécules d'acide nucléique inhibitrices dans le système respiratoire
US20090117098A1 (en) 2007-11-07 2009-05-07 Alcon Research, Ltd. Complement C1Q Inhibitors For The Prevention And Treatment Of Glaucoma
KR20090061727A (ko) * 2007-12-12 2009-06-17 숙명여자대학교산학협력단 인간 씨티알피1 유전자의 에스아이알엔에이, 및 이를포함하는 고혈압 및 심혈관계 질환 치료용 약학조성물
EP2333074A1 (fr) * 2009-12-14 2011-06-15 Robert Steinfeld Substances et procédés pour le traitement de maladies liées au stockage lysosomal
WO2015006504A1 (fr) * 2013-07-09 2015-01-15 Annexon, Inc. Anticorps anti-facteur du complément c1q et utilisations de ceux-ci
US20150337030A1 (en) 2012-05-31 2015-11-26 The Trustees Of Columbia University In The City Of New York Methods to treat alzheimer's disease using apoe inhibitors
WO2017196874A1 (fr) * 2016-05-09 2017-11-16 Annexon, Inc. Compositions et méthodes pour traiter l'amyotrophie spinale progressive
WO2017196432A1 (fr) * 2016-05-12 2017-11-16 La Jolla Institute For Allergy And Immunology Compositions et méthodes de diagnostic et de traitement d'une maladie neurodégénérative

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005002513A2 (fr) 2003-06-16 2005-01-13 The Brigham And Women's Hospital, Inc. Inhibiteurs du complement c1q et leurs methodes d'utilisation
WO2008132723A2 (fr) * 2007-04-26 2008-11-06 Quark Pharmaceuticals, Inc. Délivrance thérapeutique de molécules d'acide nucléique inhibitrices dans le système respiratoire
US20090117098A1 (en) 2007-11-07 2009-05-07 Alcon Research, Ltd. Complement C1Q Inhibitors For The Prevention And Treatment Of Glaucoma
KR20090061727A (ko) * 2007-12-12 2009-06-17 숙명여자대학교산학협력단 인간 씨티알피1 유전자의 에스아이알엔에이, 및 이를포함하는 고혈압 및 심혈관계 질환 치료용 약학조성물
EP2333074A1 (fr) * 2009-12-14 2011-06-15 Robert Steinfeld Substances et procédés pour le traitement de maladies liées au stockage lysosomal
US20150337030A1 (en) 2012-05-31 2015-11-26 The Trustees Of Columbia University In The City Of New York Methods to treat alzheimer's disease using apoe inhibitors
WO2015006504A1 (fr) * 2013-07-09 2015-01-15 Annexon, Inc. Anticorps anti-facteur du complément c1q et utilisations de ceux-ci
WO2017196874A1 (fr) * 2016-05-09 2017-11-16 Annexon, Inc. Compositions et méthodes pour traiter l'amyotrophie spinale progressive
WO2017196432A1 (fr) * 2016-05-12 2017-11-16 La Jolla Institute For Allergy And Immunology Compositions et méthodes de diagnostic et de traitement d'une maladie neurodégénérative

Non-Patent Citations (56)

* Cited by examiner, † Cited by third party
Title
ALMITAIRI JOM ET AL.: "Structure of the C1r-C1s interaction of the C1 complex of complement activation", PROC NATL ACAD SCI USA., vol. 115, no. 4, 2018, pages 768 - 773
ALMITAIRI JOM ET AL.: "Structure of the Clr-Cls interaction of the C1 complex of complement activation", PROC NATL ACAD SCI USA, vol. 115, no. 4, 2018, pages 768 - 773
ANDREI SA ET AL.: "Stabilization of protein-protein interactions in drug discovery", EXPERT OPIN DRUG DISCOV, vol. 12, no. 9, September 2017 (2017-09-01), pages 925 - 940
ARLAUD, G. J.; BIRO, A.; LING, W. L.: "Enzymatically Modified Low-Density Lipoprotein Is Recognized by Clq and Activates the Classical Complement Pathway", JOURNAL OF LIPIDS, vol. 2011, 2011, pages 376092
BELL, R. D. ET AL.: "Apolipoprotein E controls cerebrovascular integrity via cyclophilin A", NATURE, vol. 485, 2012, pages 512 - 516
BINDER, C. J.; PAPAC-MILICEVIC, N.; WITZTUM, J. L.: "Innate sensing of oxidation-specific epitopes in health and disease", NAT REV IMMUNOL, vol. 16, 2016, pages 485 - 497
BRAAK, H. ET AL.: "Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry", ACTA NEUROPATHOL, vol. 112, 2006, pages 389 - 404, XP019442875, DOI: doi:10.1007/s00401-006-0127-z
CRAMER, P. E. ET AL.: "ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models", SCIENCE, vol. 335, 2012, pages 1503 - 1506, XP055159821, DOI: doi:10.1126/science.1217697
DINARELLO, C. A.: "Anti-inflammatory Agents: Present and Future", CELL, vol. 140, 2010, pages 935 - 950
FLEMING S.D.: "Naturally occuring autoantibodies mediate ischemia/reperfusion-induced tissue injury", ADV. EXP. MED. BIOL., 2012, pages 750174 - 85
GABORIAUD C. ET AL.: "The crystal structure of the globular head of complement protein Clq provides a basis for its versatile recognition properties", J BIOL CHEM., vol. 278, no. 47, 2003, pages 46974 - 82, XP002597310, DOI: doi:10.1074/JBC.M307764200
GLASS, C. K. ET AL.: "Mechanisms Underlying Inflammation in Neurodegeneration", CELL, vol. 140, 2010, pages 918 - 934, XP055280584, DOI: doi:10.1016/j.cell.2010.02.016
GOLDMANN, T. ET AL.: "Origin, fate and dynamics of macrophages at central nervous system interfaces", NAT IMMUNOL, vol. 17, 2016, pages 797 - 805
GRONWALL C; SILVERMAN GJ: "Natural IgM: Beneficial autoantibodies for the control of inflammatory and autoimmune disease", J CLIN. IMMUNOL., vol. 34, no. 1512-21, 2014
HAJISHENGALLIS G ET AL.: "Novel mechanisms and functions of complement", NAT IMMUNOL, vol. 18, no. 12, 16 November 2017 (2017-11-16), pages 1288 - 1298
HANSSON, G. K.; HERMANSSON, A.: "The immune system in atherosclerosis", NAT IMMUNOL, vol. 12, 2011, pages 204 - 212
HASKARD DO ET AL.: "The role of complement in atherosclerosis", CURR OPIN LIPIDOL, vol. 19, no. 5, October 2008 (2008-10-01), pages 478 - 82
HESS, C.; KEMPER, C.: "Complement-Mediated Regulation of Metabolism and Basic Cellular Processes", IMMUNITY, vol. 45, 2016, pages 240 - 254, XP029687772, DOI: doi:10.1016/j.immuni.2016.08.003
HOFMAN, A. ET AL.: "Atherosclerosis, apolipoprotein E, and prevalence of dementia and Alzheimer's disease in the Rotterdam Study", LANCET, vol. 349, 1997, pages 151 - 154, XP005061658, DOI: doi:10.1016/S0140-6736(96)09328-2
HOLTZMAN, D. M.; HERZ, J.; BU, G.: "Apolipoprotein E and apolipoprotein E receptors: normal biology and roles in Alzheimer disease", COLD SPRING HARB PERSPECT MED, vol. 2, 2012, pages a006312
HOVLAND, A. ET AL.: "The complement system and toll-like receptors as integrated players in the pathophysiology of atherosclerosis", ATHEROSCLEROSIS, vol. 241, 2015, pages 480 - 494
KANEKIYO, T.; XU, H.; BU, G.: "ApoE and Ap in Alzheimer's Disease: Accidental Encounters or Partners?", NEURON, vol. 81, 2014, pages 740 - 754, XP028616093, DOI: doi:10.1016/j.neuron.2014.01.045
LIBBY, P.; RIDKER, P. M.; HANSSON, G. K.: "Progress and challenges in translating the biology of atherosclerosis", NATURE, vol. 473, 2011, pages 317 - 325
LIU ET AL.: "Targeted mouse complement inhibitor CR2-Crry protects against the development of atherosclerosis in mice", ATHEROSCLEROSIS, vol. 234, no. l, May 2014 (2014-05-01), pages 237 - 43
LUN, M. P.; MONUKI, E. S.; LEHTINEN, M. K.: "Development and functions of the choroid plexus-cerebrospinal fluid system", NAT REV NEUROSCI, vol. 16, 2015, pages 445 - 457
MAHLEY, R. W.; HUANG, Y.: "Apolipoprotein e sets the stage: response to injury triggers neuropathology", NEURON, vol. 76, 2012, pages 871 - 885, XP028960119, DOI: doi:10.1016/j.neuron.2012.11.020
MAJOR ET AL.: "Calcium-dependent conformational flexibility of a CUB domain controls activation of the complement serine protease Clr", J BIOL CHEM., vol. 285, no. 16, 2010, pages 11863 - 9
MATHERN, D. R.; P. S. HEEGER: "Molecules Great and Small: Complement System", CLIN JAM SOC NEPHROL, 2015
MILLER, CHOI S.H.; WIESNER P.; FANG L.; HARKEWICZ R; HARTVIGSEN K.; BOULLIER A.; GONEN A.; DIEHL C.J.; QUE X.; MONTANO E.: "Oxidation-Specific Epitopes are Danger Associated Molecular Patterns Recognized by Pattern Recognition Receptors of Innate", IMMUNITY CIRC RES, vol. 108, no. 2, 2011, pages 235 - 248
MOORE, G. R. ET AL.: "Complement and Humoral Adaptive Immunity in the Human Choroid Plexus: Roles for Stromal Concretions, Basement Membranes, and Epithelium", J NEUROPATHOL EXP NEUROL, vol. 75, 2016, pages 415 - 428
PANDA S; DING JL: "Natural antibodies bridge innate and adaptive immunity", J CLIN. IMMUNOL., vol. 194, 2015, pages 13 - 20, XP055456790, DOI: doi:10.4049/jimmunol.1400844
RANSOHOFF, R. M.; ENGELHARDT, B.: "The anatomical and cellular basis of immune surveillance in the central nervous system", NAT REV IMMUNOL, vol. 12, 2012, pages 623 - 635
RICKLIN D.; HAJISHENGALLIS G.; YANG K.; LAMBRIS J.D.: "The Complement - a key system for immune surveillance and homeostasis", NAT IMMUNOL, vol. 11, no. 9, 2010, pages 785 - 797, XP055306904, DOI: doi:10.1038/ni.1923
ROOS, A. ET AL.: "Specific Inhibition of the Classical Complement Pathway by Clq-Binding Peptides", J IMMUNOL, vol. 167, no. 12, 15 December 2001 (2001-12-15), pages 7052 - 7059, XP001056046
ROUMENINA L. ET AL.: "Complement Clq-target proteins recognition is inhibited by electric moment effectors", J MOL RECOGNIT, vol. 20, no. 5, September 2007 (2007-09-01), pages 405 - 15
SARMA, J. V.; P. A. WARD: "The complement system", CELL TISSUE RES, vol. 343, 2011, pages 227 - 235, XP019870471, DOI: doi:10.1007/s00441-010-1034-0
SAVIC N; SCHWANK G: "Advances in therapeutic CRISPR/Cas9 genome editing", TRANSL RES, vol. 168, February 2016 (2016-02-01), pages 15 - 21, XP029385959, DOI: doi:10.1016/j.trsl.2015.09.008
SCHWARTZ, M.; BARUCH, K.: "The resolution of neuroinflammation in neurodegeneration: leukocyte recruitment via the choroid plexus", EMBO J., vol. 33, 2014, pages 7 - 22
SHECHTER, R.; LONDON, A.; SCHWARTZ, M.: "Orchestrated leukocyte recruitment to immune-privileged sites: absolute barriers versus educational gates", NAT REV IMMUNOL, vol. 13, 2013, pages 206 - 218
SHUVAEV, V. V.; LAFFONT, I.; SIEST, G.: "Kinetics of apolipoprotein E isoforms-binding to the major glycosaminoglycans of the extracellular matrix", FEBS LETT, vol. 459, 1999, pages 353 - 357, XP004375859, DOI: doi:10.1016/S0014-5793(99)01285-5
SPEIDL WS ET AL.: "Complement in atherosclerosis: friend or foe?", J THROMB HAEMOST, vol. 9, no. 3, March 2011 (2011-03-01), pages 428 - 40
STARY, H. C.: "Natural history and histological classification of atherosclerotic lesions: an update", ARTERIOSCLER THROMB VASC BIOL, vol. 20, 2000, pages 1177 - 1178
TABAS, I.; GLASS, C. K.: "Anti-Inflammatory Therapy in Chronic Disease: Challenges and Opportunities", SCIENCE, vol. 339, 2013, pages 166 - 172
THAI, D. R.; RUB, U.; ORANTES, M.; BRAAK, H.: "Phases of Ap-deposition in the human brain and its relevance for the development of AD", NEUROLOGY, vol. 58, 2002, pages 1791 - 1800
THIELENS N.M. ET AL.: "Clq: A fresh look upon an old molecule", MOL IMMUNOL, vol. 89, September 2017 (2017-09-01), pages 73 - 83, XP085162865, DOI: doi:10.1016/j.molimm.2017.05.025
VENKATRAMAN GIRIJA, U. ET AL.: "Structural basis of the Clq/Cls interaction and its central role in assembly of the C1 complex of complement activation", PROC NATL ACAD SCI USA, vol. 110, 2013, pages 13916 - 13920
WEBER, C.; NOELS, H.: "Atherosclerosis: current pathogenesis and therapeutic options", NAT MED, vol. 17, 2011, pages 1410 - 1422
WEISMANN, D. ET AL.: "Complement factor H binds malondialdehyde epitopes and protects from oxidative stress", NATURE, vol. 478, 2011, pages 76 - 81, XP055044028, DOI: doi:10.1038/nature10449
WILES MV ET AL.: "CRISPR-Cas9-mediated genome editing and guide RNA design", MAMM GENOME, vol. 26, no. 9-10, October 2015 (2015-10-01), pages 501 - 10
WILSON, C. ET AL.: "Three-dimensional structure of the LDL receptor-binding domain of human apolipoprotein E", SCIENCE, vol. 252, 1991, pages 1817 - 1822
WITZTUM, J. L.; LICHTMAN, A. H.: "The influence of innate and adaptive immune responses on atherosclerosis", ANNU REV PATHOL, vol. 9, 2014, pages 73 - 102
YONG SHEN: "What does complement do in Alzheimer's disease? Old molecules with new insights", TRANSL NEURODEGENER, vol. 2, 2013, pages 21, XP021165528, DOI: doi:10.1186/2047-9158-2-21
ZHANG, B. ET AL.: "Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer's disease", CELL, vol. 153, 2013, pages 707 - 720, XP028589810, DOI: doi:10.1016/j.cell.2013.03.030
ZIPFEL, P. F.; C. SKERKA: "Complement regulators and inhibitory proteins", NAT REV IMMUNOL, vol. 9, 2009, pages 729 - 740
ZIPFEL, P. F.; SKERKA, C.: "Complement regulators and inhibitory proteins", NAT REV IMMUNOL, vol. 9, 2009, pages 729 - 740
ZLOKOVIC, B. V.: "Cerebrovascular effects of apolipoprotein E: implications for Alzheimer disease", JAMA NEUROL, vol. 70, 2013, pages 440 - 444

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022177724A1 (fr) * 2021-02-17 2022-08-25 University Of Rochester Procédés de traitement, de diagnostic et de prédiction du pronostic d'une sepsie

Also Published As

Publication number Publication date
EP3737472A1 (fr) 2020-11-18

Similar Documents

Publication Publication Date Title
Schartz et al. The good, the bad, and the opportunities of the complement system in neurodegenerative disease
AU2006222193B2 (en) Screening method, process for purifying of non-diffusible A-beta oligomers, selective antibodies against said non-diffusible A-beta oligomers and a process for manufacturing of said antibodies
El-Agnaf et al. A strategy for designing inhibitors of α‐synuclein aggregation and toxicity as a novel treatment for Parkinson's disease and related disorders
Thayanidhi et al. α-Synuclein delays endoplasmic reticulum (ER)-to-Golgi transport in mammalian cells by antagonizing ER/Golgi SNAREs
EP2052085B1 (fr) Procédés de modulation de set et utilisations associées
US20210395319A1 (en) Methods and compositions comprising tau oligomers
US11104710B2 (en) Methods and compositions comprising tau oligomers
Dunning et al. Direct high affinity interaction between Aβ42 and GSK3α stimulates hyperphosphorylation of tau. A new molecular link in Alzheimer’s disease?
US20200181250A1 (en) Anti-transthyretin human antibody
Yerbury et al. Extracellular chaperones modulate the effects of Alzheimer’s patient cerebrospinal fluid on Aβ1-42 toxicity and uptake
Qiu et al. The Sez6 family inhibits complement by facilitating factor I cleavage of C3b and accelerating the decay of C3 convertases
Haigh et al. Copper, endoproteolytic processing of the prion protein and cell signalling
CA2979617C (fr) Polypeptides derives de grp94 pour inhiber pcsk9 et procedes d'utilisation
US20110236455A1 (en) Novel Regulators of the Innate Immune System
WO2019137922A1 (fr) Modulateurs de c1q, en particulier de l'interaction de l'apoe avec c1q, et utilisations des modulateurs dans le traitement de maladies neuronales et de l'inflammation
Theunis et al. Novel Phospho-tau monoclonal antibody generated using a liposomal vaccine, with enhanced recognition of a conformational Tauopathy epitope
US20220034913A1 (en) Methods and compostions of detecting and treating neurodegenerative disorders
JP4937143B2 (ja) 非拡散性球状Aβ(X−38..43)オリゴマー構造エピトープに対して特異性を有する自己抗体を検出する方法
WO2020154363A1 (fr) Protéine de fusion nucléobindine 1-immunoglobuline génétiquement modifiée
RU2812207C2 (ru) Способы и фармацевтические композиции для лечения заболеваний, связанных с тубулин карбоксипептидазами (tcp)
RU2797200C1 (ru) Способы и фармацевтические композиции для лечения заболеваний, связанных с тубулин карбоксипептидазами (tcp)
Fathalla The generation of antibodies as tools to characterise triggering receptor expressed on Myeloid Cells 2 (TREM2) and explore its role in Alzheimer’s Disease
Cooley Dysregulated Membrane Trafficking and the Development of Pancreatitis
Wang et al. Cross Interactions between Apolipoprotein E and Amyloid Proteins in Neurodegenerative and other Diseases
US20180284137A1 (en) Diagnostic assay for alzheimer's disease

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19701776

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019701776

Country of ref document: EP

Effective date: 20200810