WO2024240848A1 - Polypeptides dérivés du ldlr pour des utilisations antivirales - Google Patents
Polypeptides dérivés du ldlr pour des utilisations antivirales Download PDFInfo
- Publication number
- WO2024240848A1 WO2024240848A1 PCT/EP2024/064152 EP2024064152W WO2024240848A1 WO 2024240848 A1 WO2024240848 A1 WO 2024240848A1 EP 2024064152 W EP2024064152 W EP 2024064152W WO 2024240848 A1 WO2024240848 A1 WO 2024240848A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- egfa
- la6x
- la4x
- ldlr
- egfbx
- 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
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/177—Receptors; Cell surface antigens; Cell surface determinants
Definitions
- the present invention relates to an LDLR derived polypeptide that binds a Bunyavirales virus and is thus useful in the prophylaxis and/or treatment of a Bunyavirales virus infection in a subject, preferably a CCHV infection.
- LDLR derived polypeptides comprising an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain, which bind a Bunyavirales virus.
- LDLR derived polypeptides comprising a multimer of an LDLR ectodomain (SEQ ID NO 18), or of a functional fragment of said LDLR ectodomain, or of an amino acid variant of said LDLR ectodomain, or of an amino acid variant of said functional fragment of said LDLR ectodomain, wherein said LDLR derived polypeptide optionally comprises a peptide linker connecting said monomers.
- the LDLR polypeptides here described are particularly useful against Bunyavirales virus infections. Further, the LDLR polypeptides can be used for diagnostic detection of Bunyavirales virus infection, particularly CCHFV infection, and quality control testing of related vaccines. A method to identify new gene factors involved in an infection by a Bunyavirales virus is also provided herein.
- CHFV Crimean-Congo hemorrhagic fever virus
- CCHF Crimean-Congo hemorrhagic fever
- CCHF is transmitted by Hyalomma ticks and through direct contact with the blood and other bodily fluids of patients or infected animals.
- the CCHF incubation period is 2-14 days and correlates with the type of transmission and viral load.
- the clinical manifestations can be from subclinical illness to acute infection with haemorrhage such as pulmonary haemorrhage, intra-abdominal bleeding, or haematuria, and multiorgan failure.
- CCHF chronic myemorrhagic fever
- CCHFV CCHFV is the most widespread haemorrhagic fever, being endemic in certain regions of Africa and in Asia but also in Europe.
- CCHFV is a tick-borne pathogen that is also capable of interpersonal transmission.
- CCHFV CCHFV complex protein kinase
- CCHFV enters cells through clathrin-mediated endocytosis and uses the endosomal pathway to release viral RNA segments.
- CCHFV enters cells through clathrin-mediated endocytosis and uses the endosomal pathway to release viral RNA segments.
- many different cell types can be infected with CCHFV, suggesting the existence of either a widely distributed receptor or several redundant entry receptors.
- nucleoside analogue ribavirin is the only direct-acting anti-viral that has been widely used clinically in patients with CCHF.
- ribavirin is controversial, with continued debate about whether treatment improves outcome.
- antibody-based therapies have also been evaluated for treatment of CCHFV, however large- scale trials have not been performed so far.
- RNA viruses tend to generate vaccine-resistant escaper strains through mutations of antibody-binding sites.
- a host-related decoy receptor such as a LDLR-derived polypeptide as disclosed herein would require a gain-of-function mutation switching to another host cell receptor.
- Such a mutation is less likely to appear so that there is a decreased risk of generating resistant virus strains in response to treatment with a LDLR- derived polypeptide according to this invention compared to vaccine approaches.
- LDLR polypeptides comprising soluble LDL receptors, functional fragments thereof, or single amino acid variants thereof, which can be used for preventing and/or treating a Bunyavirales virus infection, preferably a CCHFV infection.
- a method to identify new factors involved in a CCHFV infection is also provided herein.
- an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain.
- an amino acid variant of an LDLR ectodomain refers to a single amino acid variant of an LDLR ectodomain. In some embodiments, an amino acid variant of an LDLR ectodomain refers to a multiple amino acid variant of an LDLR ectodomain.
- a preferred embodiment of present invention relates to LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection in a human, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain.
- said LDLR derived polypeptide for use comprises an LDLR ectodomain having amino acid sequence SEQ ID NO 18, or a functional fragment thereof, or an amino acid variant thereof.
- said LDLR derived polypeptide for use comprises a dimer, a trimer, or a multimer of an LDLR ectodomain monomer having amino acid sequence SEQ D NO 18, or of a functional fragment thereof, or of an amino acid variant thereof. More preferred said LDLR derived polypeptide comprises a dimer or trimer, still more preferred a dimer, still more preferred is a homodimer or a homotrimer, and still more preferred is a homodimer.
- said LDLR derived polypeptide for use comprises a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41 ), OLSD (SEQ ID NO 36), LA1 -LA2-LA3-LA4X-LA5-LA6X-LA7-EGFA-EGFBX-PROPX-EGFCX-OLSD, LA1 - LA2-LA3-LA4
- an amino acid variant of a functional fragment of an LDLR ectodomain refers to a single amino acid variant of a functional fragment of an LDLR ectodomain. In some embodiments, an amino acid variant of an LDLR ectodomain refers to a multiple amino acid variant of a functional fragment of an LDLR ectodomain.
- said LDLR derived polypeptide for use further comprises a peptide linker connecting said fragments or monomers.
- said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava-Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website
- CATV Longquan hantavirus
- LQUV Longquan hantavirus
- PUUV Puumala hantavirus
- SANGV Sangassou hantavirus
- SEOV Sin Nombre hantavirus
- SNV Sin Nombre hantavirus
- TAAIV Tula hantavirus
- NW New York hantavirus
- RVFV Rift Valley fever virus
- NSDV Sandfly Fever Naples virus
- said Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- said LDLR derived polypeptide for use comprises an LDLR ectodomain having amino acid sequence SEQ ID NO 18, and said Bunyavirales virus is CCHFV.
- a further embodiment of the invention is directed to a pharmaceutical composition
- a pharmaceutical composition comprising the LDLR derived polypeptide for use described above, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent.
- Another embodiment of the invention is directed to a recombinant nucleic acid molecule comprising a nucleic acid sequence encoding an LDLR derived polypeptide for use described above.
- a particular embodiment of the invention is directed to an LDLR derived polypeptide comprising a functional fragment of an LDLR ectodomain selected from the group comprising or consisting of:
- LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41 ), OLSD (SEQ ID NO 36), LA1 -LA2-LA3-LA4X-LA5-LA6X-LA7-EGFA-EGFBX-PROPX-EGFCX-OLSD, LA1 - LA2-LA3-LA4X-LA5-LA6X-LA7-EGFA-EGFBX-PROPX-EGFCX-OLSD, LA1 - LA2-
- LA2-LA3-LA4X-LA5-LA6X-LA7 LA2-LA3-LA4X-LA5-LA6, LA2-LA3-LA4X- LA5, LA2-LA3-LA4, LA2-LA3, LA3-LA4X-LA5-LA6X-LA7-EGFA-EGFBX-PROPX- EGFCX-OLSD, LA3-LA4X-LA5-LA6X-LA7-EGFA-EGFBX-PROPX-EGFC, LA3-LA4X-LA5-LA6X-LA7-EGFA-EGFBX-PROP, LA3-LA4X-LA5-LA6X-LA7- EGFA-EGFB, LA3-LA4X-LA5-LA6X-LA7- EGFA-EGFB, LA3-LA4X-LA5-LA6X-LA7-EGFA, LA3-LA4X-LA5-LA6X-LA7, LA3-LA4X-LA5-LA6X-LA7, LA3-LA
- a further particular embodiment of the invention is directed to LDLR derived polypeptide comprising a multimer of monomers, wherein each monomer is selected from the group comprising: an LDLR ectodomain (SEQ ID NO 18), a functional fragment of said LDLR ectodomain, an amino acid variant of said LDLR ectodomain, an amino acid variant of said functional fragment of said LDLR ectodomain, and wherein said multimer comprises between 2 and 8 monomers, preferably 2 or 3 monomers, and wherein said LDLR derived polypeptide optionally comprises a peptide linker connecting said monomers; and wherein said multimer is a homomultimer or a heteromultimer.
- a more particular embodiment of the invention is directed to an LDLR derived polypeptide comprising a functional fragment of an LDLR ectodomain as described above, or comprising a multimer of monomers as described above, wherein said LDLR derived polypeptide is for use in the prophylaxis and/or treatment of a virus infection, wherein said virus is preferably a Bunyavirales virus.
- a further particular embodiment of the invention is directed to a recombinant nucleic acid molecule comprising a nucleic acid sequence encoding an LDLR derived polypeptide comprising a functional fragment of an LDLR ectodomain or comprising a multimer of monomers as described above.
- a particular embodiment of the invention is directed to an in vitro screening method to identify a gene that modulates a Bunyavirales virus infection, the method comprising:
- step B) performing chemical random mutagenesis in said cells of step A) in order to obtain mutagenized cells
- step B) infecting the mutagenized cells of step B) with a Bunyavirales virus or with a Bunyavirales virus pseudotyped RNA viral particle;
- step D culturing the infected mutagenized cells of step C) to obtain resistant colonies; E) isolating the resistant colonies of step D);
- step E) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate a Bunyavirales virus infection
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof
- the pseudotyped RNA viral particle is a HIV particle or a MSV particle or a VSV particle, wherein the pseudotyped RNA viral particle express a Bunyavirales virus specific glycoprotein Gc and/or Gn
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hant
- whole exome sequencing refers to the sequencing of all exons in a specific cell or tissue type, wherein the exons are the protein coding regions of a gene. Therefore, in “whole exome sequencing” only the protein-coding DNA regions are sequenced as compared to whole genome sequencing.
- whole transcriptome sequencing refers to sequencing of expressed transcripts at the mRNA level to obtain sequence information on all mRNAs made in a cell or cell pool at a given time
- the pseudotyped RNA viral particle is preferably a VSV particle.
- performing chemical random mutagenesis of step B) comprises incubating the haploid cells with N-Ethyl-N-nitrosourea (ENU).
- the inventors were able to identify host factors important for CCHFV infections by using a novel method based on haploid murine stem cells mutagenized with N- Ethyl-N-nitrosourea (ENU), which induces single nucleotide variants into the genome, and subsequent infection with a CCHFV glycoprotein pseudotyped vesicular stomatitis virus (VSV-CCHF_G) ( Figure 1).
- ENU N- Ethyl-N-nitrosourea
- VSV-CCHF_G CCHFV glycoprotein pseudotyped vesicular stomatitis virus
- Haploid cells containing only a single genomic copy allow for direct translation of the introduced genetic changes such as gene knockouts and point mutations to a respective phenotype.
- LDLR Low Density Lipoprotein Receptor
- Table 2 Experiments using a soluble LDLR, ldlr knockout cells, and both a laboratory strain and a patient isolate of CCHFV, confirmed that LDLR mediates CCHFV infection ( Figure 2 and 4). Furthermore, experiments based on Bioluminescence Resonance Energy Transfer (BRET) technique confirmed that LDLR is a receptor for CCHFV, as the viral glycoprotein Gc directly attaches to LDLR ( Figures 3 and 8).
- BRET Bioluminescence Resonance Energy Transfer
- the low-density lipoprotein receptor gene family encodes for a class of structurally related cell surface receptors also named low density lipoprotein receptor-related proteins (LRP) that fulfil diverse biological functions in different organs, tissues, and cell types, including the regulation of lipid metabolism, protection against atherosclerosis, neurodevelopment, and transport of nutrients and vitamins.
- LRP low density lipoprotein receptor-related proteins
- LDLR low-density lipoprotein receptor
- VLDLR very low-density lipoprotein receptor
- ApoER2 also known as low density lipoprotein receptor-related protein- 8, LRP8;
- LRP4 low density lipoprotein receptor-related protein 4, LRP4, also known as multiple epidermal growth factor (EGF) repeat-containing protein, MEGF7;
- LRP1 LDLR-related protein 1
- LRP1 b LDLR-related protein 1b
- LDL is one of the major cholesterol-carrying lipoprotein of plasma, acting to regulate cholesterol homeostasis in mammalian cells.
- the LDL receptor binds and transports LDL particles into cells by acidic endocytosis. Once inside the cell, the LDLR separates from its ligand, which is degraded in the lysosomes, while the receptor returns to the cell surface.
- the internal dissociation of the LDLR with its ligand is mediated by proton pumps within the walls of the endosome that lower the pH.
- the LDLR is a multi-domain protein, containing the following five domains:
- LA repeats 40-amino acid LDLR class A cysteine-rich repeats, also named LA repeats, each of which contains a coordinated calcium ion and six cysteine residues involved in disulphide bond formation.
- LA1 40-amino acid LDLR class A cysteine-rich repeats
- LA7 a ligand-binding domain containing seven 40-amino acid LDLR class A cysteine-rich repeats, also named LA repeats, each of which contains a coordinated calcium ion and six cysteine residues involved in disulphide bond formation.
- LDLR class B repeats LB1 , LB2, LB2, LB3, LB4, LB5, and LB6
- EGFC EGF repeat
- the six LDLR class B repeats each contain a conserved YWTD motif and are predicted to form a [3-propeller structure, and are thus referred herein with the term "PROP”. This region is critical for ligand release and recycling of the receptor.
- LDLR ectodomain module or "LDLR module” or simply “module” is used herein to refer to the individual repeats and domains: LA1 , LA2, LA3, LA4, LA5, LA6, LA7, EGFA, EGFB, LB1 , LB2, LB2, LB3, LB4, LB5, LB6, PROP, EGFC, and OLSD.
- amino sequences of the "LDLR ectodomain modules" used in the present invention are:
- SEQ ID NO 36 OLSD Clustered O-linked oligosaccharides
- Soluble low-density lipoprotein receptor is the circulating LDLR ectodomain of transmembrane LDLR comprising the domains I) - III) described above, i.e. the LDLR derived polypeptide consisting of LA1-LA7 repeats, EGFA, EGFB, PROP, EGFC, and OLSD.
- LDLR ectodomain refers to the soluble form of the low-density lipoprotein receptor (sLDLR), comprising the "modules" LA1-LA7,
- LDLR ectodomain refers to the soluble form of the low-density lipoprotein receptor (sLDLR) that binds a Bunyavirales virus, and which comprises LA1-LA7, EGFA,
- the soluble "LDLR ectodomain” corresponds to the extracellular portion of 766 amino acid residues of the LDL receptor, which is purified to homogeneity from impurities. More in detail, the soluble "LDLR ectodomain” corresponds to the extracellular portion of mature LDL receptor, which has an amino acid sequence SEQ ID NO 18 corresponding to amino acid residue Ala22 to Arg788 of the mature LDL receptor as deposited at Uniprot ID P01130.
- LDLR derived polypeptide is used herein to refer to the following polypeptides (also referred to as “polypeptides of the disclosure”): an "LDLR ectodomain” having amino acid sequence SEQ ID NO 18; a “functional fragment” of said "LDLR ectodomain” having amino acid sequence SEQ ID NO 18; a “single or multiple amino acid variant” of said "LDLR ectodomain” having amino acid sequence SEQ ID NO 18; a single or multiple amino acid variant of said "functional fragment of said LDLR ectodomain” having amino acid sequence SEQ ID NO 18; a multimer of a "LDLR ectodomain” having amino acid sequence SEQ ID NO 18, wherein said multimer is a dimer, or a trimer or comprises more repetitions of said "LDLR ectodomain", and wherein said multimer is a homomultimer of said "LDLR ectodomain”; a multimer of a functional fragment of a "LDLR ectodomain”
- the multimer as disclosed herein is preferably a dimer or trimer, still more preferred a dimer, still more preferred is a homodimer or a homotrimer, and still more preferred is a homodimer.
- the present invention provides an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain.
- a preferred embodiment of present invention relates to LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection in a human, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain.
- An embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain having amino acid sequence SEQ ID NO 18, or a functional fragment thereof, or an amino acid variant thereof.
- amino acid variant refers to an analog of the natural soluble LDL receptor (LDLR ectodomain) or of a functional fragment thereof in which one or more amino acid residues of the natural soluble LDL receptor is replaced by a different amino acid residue or is deleted, so that the resulting LDLR derived polypeptide has an ability of binding to a Bunyavirales virus about equal to or greater than that of the natural LDLR ectodomain.
- LDLR ectodomain the natural soluble LDL receptor
- amino acid variants are prepared by known synthesis and/or by site-directed mutagenesis techniques, or any other known technique suitable therefor.
- amino acid variant comprises "single amino acid variant” and "multiple amino acid variant”.
- single amino acid variant refers to an analog of the natural soluble LDL receptor (LDLR ectodomain) or of a functional fragment thereof in which one amino acid residue of the natural soluble LDL receptor is replaced by a different amino acid residue, or is added, or is deleted, so that the resulting LDLR derived polypeptide has an ability of binding to a Bunyavirales virus about equal to or greater than that of the natural LDLR ectodomain.
- the term “multiple amino acid variant” refers to an analog of the natural soluble LDL receptor (LDLR ectodomain) or of a functional fragment thereof in which multiple amino acid residues, i.e. at least two amino acid residues of the natural soluble LDL receptor are replaced by different amino acid residues or deleted, or at least two amino acid residues are added compared to the natural soluble LDL receptor, so that the resulting LDLR derived polypeptide has an ability of binding to a Bunyavirales virus about equal to or greater than that of the natural LDLR ectodomain.
- a “multiple amino acid variant of an LDLR ectodomain or of a functional fragment thereof” comprises an amino acid sequence that shows high aminoacidic sequence identity to the parent LDLR ectodomain (SEQ ID NO 18) or a fragment thereof, such as it shows more than 60%, 70%, 75%, 80%, 85%, 90%, 95% or 98% sequence identity with the parent sequence and is preferably characterised by similar properties of the parent sequence, namely binding to a Bunyavirales virus.
- the percentage of "sequence identity” is determined by comparing two optimally aligned amino acid sequences over a “comparison window” on the full length of the reference sequence.
- a “comparison window” as used herein refers to the optimal alignment between the reference and variant sequence after that the two sequences are optimally aligned, wherein the variant amino acid sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment.
- Identity percentage is calculated by determining the number of positions at which the identical amino acids occur in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the full length in amino acid residues) and multiplying the results by 100 to yield the percentage of sequence identity.
- Two amino acid sequences are said to be "identical” if the sequence of amino acid residues in the two sequences is the same when optimally aligned as described above.
- the term "functional fragment of an LDLR ectodomain” refers to a fragment of the LDLR ectodomain comprising one or more "LDLR ectodomain modules" (i.e. LA1 to LA7, EGFA, EGFB, PROP, EGFC, and OLSD), provided that said fragment has an ability to bind to a Bunyavirales virus about equal to or greater than that of the natural LDLR ectodomain.
- LDLR ectodomain modules i.e. LA1 to LA7, EGFA, EGFB, PROP, EGFC, and OLSD
- the term “functional fragment” refers to a fragment of the LDLR ectodomain, wherein said “functional fragment” comprises one or more "LDLR ectodomain modules", wherein said "LDLR ectodomain modules” have the same order in the resulting "functional fragment” as in the natural LDLR ectodomain (SEQ ID NO 18), provided that said fragment has an ability to bind to a Bunyavirales virus about equal to or greater than that of the natural LDLR ectodomain.
- the term "functional fragment of an LDLR ectodomain” is selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41 ), OLSD (SEQ ID NO 36), LA1-LA2-LA3-LA4X-LA5-LA6X-LA7-EGFA- EGFBX-PROPX-EGFCX-OLSD, LA1 -LA2-LA3-LA4X-LA5-LA6X-OLSD, LA
- an embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41),
- said LDLR derived polypeptide for use is a homomultimer of functional fragment monomers, wherein each monomer is a functional fragment of an LDLR ectodomain, it is preferred that said homomultimers comprises between 2 and 10 monomers, 2 and 12 monomers, 2 and 14 monomers, 2 and 16 monomers, 2 and 18 monomers, 2 and 20 monomers, 2 and 8 monomers, 2 and 6 monomers, 2 and 4 monomers. More preferably, When said LDLR derived polypeptide for use is a homomultimer of monomers, wherein each monomer is a functional fragment, it is preferred that said homomultimers comprises between 2 and 8 monomers, preferably 2 or 3 monomers.
- a monomer in said LDLR derived polypeptide for use, can be a functional fragment of an LDLR ectodomain or an amino acid variant thereof.
- An exemplary LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises a homomultimer of a functional fragment of an LDLR ectodomain selected from the group reported above, has the following composition: (LA2-LA3-LA4) (LA2-LA3-LA4) (LA2-LA3-LA4) (LA2-LA3-LA4) (LA2-LA3-LA4) (LA2-LA3-LA4), or (LA3-LA4X-LA5-LA6X-LA7-EGFA-EGFBX-PROP) (LA3-LA4X-LA5-LA6X-LA7- EGFA-EGFBX-PROP), and similars. Multimers
- a further embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises a dimer, a trimer, a heteromultimer, or a homomultimer of an LDLR ectodomain monomer having amino acid sequence SEQ ID NO 18, or of a functional fragment thereof, or of an amino acid variant thereof. More preferred is a dimer or trimer, still more preferred a dimer, still more preferred is a homodimer or a homotrimer, and still more preferred is a homodimer.
- a monomer is thus selected from the group comprising: an LDLR ectodomain (SEQ ID NO 18), a functional fragment of said LDLR ectodomain, an amino acid variant of said LDLR ectodomain, an amino acid variant of said functional fragment of said LDLR ectodomain.
- said LDLR derived polypeptide for use is a homomultimer of the monomers defined above, it is preferred that said homomultimers comprises between 2 and 10 monomers, 2 and 12 monomers, 2 and 14 monomers, 2 and 16 monomers, 2 and 18 monomers, 2 and 20 monomers, 2 and 8 monomers, 2 and 6 monomers, 2 and 4 monomers. More preferably, said LDLR derived polypeptide for use comprises between 2 and 8 monomers as defined above, preferably 2 or 3 monomers.
- homomultimer of an LDLR ectodomain refers to a multimeric polypeptide consisting of two or more identical monomers, wherein each monomer is an LDLR ectodomain having amino acid sequence SEQ ID NO 18, or an amino acid variant thereof. Therefore, the term “homomultimer of an LDLR ectodomain” also refers to a multimeric polypeptide consisting of two or more identical monomers, wherein each monomer is an amino acid variant of LDLR ectodomain having amino acid sequence SEQ ID NO 18.
- the term "homomultimer of an LDLR ectodomain” refers to a dimer or trimer of an LDLR ectodomain monomer, wherein each LDLR ectodomain monomer has amino acid sequence SEQ ID NO 18, or an amino acid variant thereof. Therefore, the term “homomultimer of an LDLR ectodomain” also refers to a dimer or trimer of an LDLR ectodomain monomer, wherein each monomer is an amino acid variant of LDLR ectodomain having amino acid sequence SEQ ID NO 18.
- the term "homomultimer of an LDLR ectodomain functional fragment” refers to a multimeric polypeptide consisting of two or more identical monomers, wherein each monomer is an LDLR ectodomain functional fragment, and wherein each functional fragment is selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41 ), OLSD (SEQ ID NO 36), LA1 -LA2-LA3-LA4X-
- LA1-LA2-LA3-LA4X-LA5-LA6X-LA7 LA1 -LA2-LA3-LA4X-LA5-LA6, LA1- LA2-LA3-LA4X-LA5, LA1 -LA2-LA3-LA4, LA1-LA2-LA3, LA1-LA2, LA2-LA3-LA4X- LA5-LA6X-LA7-EGFA-EGFBX-PROPX-EGFCX-OLSD, LA2-LA3-LA4X-LA5-LA6X- LA7-EGFA-EGFBX-PROPX-EGFC, LA2-LA3-LA4X-LA5-LA6X-LA7-EGFA- EGFBX-PROPX-EGFC, LA2-LA3-LA4X-LA5-LA6X-LA7-EGFA- EGFBX-PROP, LA2-LA3-LA4X-LA5-LA6X-LA7-EGFA-EGFBX-PROP, LA2-LA3-LA4X-LA5-LA6X
- LA4X-LA5-LA6X-LA7-EGFA LA4X-LA5-LA6X-LA7
- LA4X-LA5-LA6X-LA7 LA4X-LA5-LA6, LA4X- LA5, LA5-LA6X-LA7-EGFA-EGFBX-PROPX-EGFCX-OLSD
- LA5-LA6X-LA7-EGFA-EGFBX-PROP LA5-LA6X- LA7-EGFA-EGFB
- LA5-LA6X-LA7-EGFA LA5-LA6X-LA7, LA5-LA6X-LA7, LA5-LA6X-LA7, LA5-LA6, LA6X-LA7- EGFA-EGFBX-PROPX-EGFCX-OLSD
- LA6X-LA7-EGFA-EGFBX-PROPX-EGFC LA6X-LA7-EGFA-EGFBX-PROPX-EGFC
- the disclosed dimer of an LDLR ectodomain monomer comprising two monomers, wherein each monomer is an LDLR ectodomain or an LDLR ectodomain functional fragment or an amino acid variant thereof, has a binding capacity for Bunyavirales virus that is about equal to or greater than the binding capacity of the single LDLR ectodomain monomers.
- the disclosed trimer of an LDLR ectodomain monomer comprising three monomers, wherein each monomer is an LDLR ectodomain or an LDLR ectodomain functional fragment or an amino acid variant thereof, has a binding capacity for Bunyavirales virus that is about equal to or greater than the binding capacity of the single LDLR ectodomain monomer.
- the disclosed homomultimer of an LDLR ectodomain monomer comprising a plurality of monomers, wherein each monomer is an LDLR ectodomain or an LDLR ectodomain functional fragment or an amino acid variant thereof, has a binding capacity for Bunyavirales virus, preferably CCHFV or RVFV and most preferably CCHFV, that is about equal to or greater than the binding capacity of an LDLR ectodomain monomer.
- each monomer or functional fragments or single amino acid variant thereof may be linked directly to the adjacent monomer or functional fragments or single amino acid variant thereof.
- the N-terminus of the second or successive monomer e.g. LDLR ectodomain of SEQ ID NO 18, or a functional fragment thereof or an amino acid variant thereof
- the C-terminus of the first or previous monomer of the disclosure e.g., LDLR ectodomain of SEQ ID NO 18, or a functional fragment thereof or an amino acid variant thereof.
- the LDLR ectodomain monomers, functional fragment thereof or single amino acid variant thereof can be associated in any suitable manner to form an "LDLR derived polypeptide" or an homomultimer forming an "LDLR derived polypeptide".
- Said various monomers or functional fragments may be covalently associated, e.g., by means of a peptide or disulfide bond.
- the polypeptide fragments or monomers can thus be directly fused.
- the resulting fusion protein representing the LDLR derived polypeptide of the invention can include any suitable number of modified bonds, e.g., isosteres, within or between the polypeptide fragments or monomers.
- the LDLR derived polypeptide can include a "peptide linker" or "peptide linker sequence” between any two polypeptide fragments or monomers that includes one or more amino acid sequences not forming part of the biologically active peptide portions.
- Any suitable peptide linker can be used.
- Such a linker can be of any suitable size.
- the linker predominantly may comprise or consist of neutral amino acid residues. If separation of peptide fragments or monomers is desirable a linker that facilitates separation can be used.
- Flexible linkers typically composed of combinations of glycine and/or serine residues, or combinations of glycine, proline, serine and alanine can be advantageous. Examples of such linkers are described in "Table 1".
- linker sequences are used in the "LDLR derived polypeptides" to connect different components of the "LDLR derived polypeptide".
- the amino terminal end of the linker sequence is joined by a peptide bond to a first (or previous) LDLR ectodomain monomer or fragment and the carboxy terminal end of the linker sequence is joined by a peptide bind to a second (or successive) LDLR ectodomain monomer or fragment.
- a particular embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain, and wherein said LDLR derived polypeptide further comprises a peptide linker connecting said fragments.
- a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1)
- a further embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises a dimer, a trimer, or a multimer of an LDLR ectodomain monomer having amino acid sequence SEQ ID NO 18, or of a functional fragment thereof, or of an amino acid variant thereof, and wherein said LDLR derived polypeptide further comprises a peptide linker connecting said fragments or monomers.
- a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1).
- said multimer comprises between 2 and 8 monomers, preferably 2 or 3 monomers.
- a further embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ
- said homomultimer comprises between
- This invention further concerns a recombinant nucleic acid molecule comprising a nucleic acid sequence encoding an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain or of a functional fragment thereof.
- nucleic acid molecule as used herein includes genomic DNA, cDNA, synthetic DNA, synthetic RNA, mRNA, ssDNA, dsDNA, self-amplifying RNA, dsRNA, and combinations thereof.
- nucleic acid molecule preferably includes synthetic DNA, synthetic RNA, and mRNA.
- the "nucleic acid molecule” is a “synthetic delivery RNA”.
- the “nucleic acid molecule” is a "therapeutic RNA” for delivery into a mammal cell.
- RNA or "RNA molecule” or “ribonucleic acid molecule” refers to a polymer of ribonucleotides (e.g., 2, 3, 4, 5, 10, 15, 20, 25, 30, or more ribonucleotides).
- DNA or "DNA molecule” or “deoxyribonucleic acid molecule” refers to a polymer of deoxyribonucleotides.
- DNA and RNA can be synthesized naturally e.g., by DNA replication or transcription of DNA, respectively. RNA can be post-transcriptionally modified. DNA and RNA can also be chemically synthesized. DNA and RNA can be single-stranded (i.e.
- mRNA or “messenger RNA” is a single-stranded RNA that specifies the amino acid sequence of one or more polypeptide chains.
- isolated RNA or isolated DNA refers to RNA or DNA molecules which are substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
- the polypeptides and recombinant nucleic acid molecules of interest are "purified” to essential homogeneity, i.e., contaminant species cannot be detected in the composition by conventional detection methods.
- Purity and homogeneity can be determined using a number of techniques well known in the art, such as agarose or polyacrylamide gel electrophoresis of a protein or nucleic acid sample, followed by visualization upon staining, or using a high-resolution technique, such as high performance liquid chromatography (HPLC).
- HPLC high performance liquid chromatography
- Synthetic delivery RNA as described herein can refer to a synthetic RNA for delivery into the nucleus of a cell.
- the invention also concerns replicable expression vectors containing said nucleic acid molecules, hosts transformed therewith and protein produced by expression of such transformed hosts.
- the production of the recombinant LDLR ectodomain or of the functional fragment thereof may be carried out by different techniques well known in the prior art.
- a DNA molecule, nucleic acid molecule or polypeptide is "recombinant" when it is artificial or engineered or derived from an artificial or engineered protein or nucleic acid.
- LDLR derived polypeptides of the invention Recombinant methods for producing and isolating LDLR derived polypeptides of the invention are those described in the prior art.
- the LDLR derived polypeptides may be produced by direct peptide synthesis using solid-phase techniques. Peptide synthesis may be performed using manual techniques or by automation. Automated synthesis may be achieved, for example, using Applied Biosystems 431 A Peptide Synthesizer (Perkin Elmer, Foster City, Calif.) in accordance with the instructions provided by the manufacturer. For example, sub-sequences may be chemically synthesized separately and combined using chemical methods to provide LDLR derived polypeptides of the invention or functional fragments thereof. Alternatively, such sequences may be ordered from a company or from a protein production facility that specialize in production of polypeptides.
- a recombinant method for producing and isolating polypeptides of the invention comprises introducing into a population of cells any "recombinant nucleic acid molecule" described herein, which is operatively linked to a regulatory sequence (e.g. a promoter) effective to produce the encoded polypeptide, culturing the cells in a culture medium to produce the polypeptide, and isolating the polypeptide from the cells or from the culture medium.
- a regulatory sequence e.g. a promoter
- An amount of nucleic acid sufficient to facilitate uptake by the cells (transfection) and/or expression of the polypeptide is utilized.
- Appropriate cell culture media are known to those of skill in the art.
- the nucleic acid is introduced into such cells by any delivery method known in the prior art, including, e.g., injection, needleless injection device, gene gun, transfection, transfection with lipofectamine, retroviral gene delivery, electroporation transdermal delivery, passive uptake, etc.
- the nucleic acid of the invention may be part of a vector, such as a recombinant expression vector, including a DNA plasmid vector, viral vector, or any suitable vector known in the prior art.
- the nucleic acid or vector comprising a nucleic acid of the invention may be prepared and formulated as known in the prior art. Such a nucleic acid or expression vector may be introduced into a population of in vitro cultured cells.
- a "vector” may be any agent that is able to deliver or maintain a nucleic acid in a host cell and includes, for example, but is not limited to, plasmids (e.g., DNA plasmids), naked nucleic acids, viral vectors, viruses, nucleic acids complexed with one or more polypeptide or other molecules, as well as nucleic acids immobilized onto solid phase particles.
- plasmids e.g., DNA plasmids
- naked nucleic acids e.g., viral vectors, viruses, nucleic acids complexed with one or more polypeptide or other molecules, as well as nucleic acids immobilized onto solid phase particles.
- a vector can be useful as an agent for delivering or maintaining an exogenous gene, nucleic acid, and/or protein in a host cell.
- a vector may be capable of transducing, transfecting, or transforming a cell, thereby causing the cell to replicate or express nucleic acids and/or proteins other than those native to the cell or in a manner not native to the cell.
- a vector may include materials to aid in achieving entry of a nucleic acid into the cell, such as a viral particle, liposome, protein coating, or the like. Any method of transferring a nucleic acid into the cell may be used; unless otherwise indicated, the term vector does not imply any particular method of delivering a nucleic acid into a cell or imply that any particular cell type is the subject of transduction.
- expression vector typically refers to a nucleic acid construct or sequence, generated recombinantly or synthetically, with a series of specific nucleic acid elements that permit transcription of a particular "nucleic acid molecule" in a host cell.
- the expression vector typically includes a nucleic acid to be transcribed operably linked to a promoter.
- expression includes any step involved in the production of the polypeptide (i.e. the LDLR ectodomain polypeptide, or a functional fragment thereof or an amino acid variant thereof) including, but not limited to, transcription, post-transcriptional modification, translation, post-translational modification, and/or secretion.
- the nucleic acids can also be modified to include one or more codons that provide for optimum expression in an expression system (e.g., mammalian cell or mammalian expression system), while, if desired, said one or more codons still encode the same amino acid(s).
- an expression system e.g., mammalian cell or mammalian expression system
- said one or more codons still encode the same amino acid(s).
- a DNA sequence encoding a desired linker sequence may be inserted between, and in the same reading frame as, for example, two adjacent DNA sequences encoding two monomers of the LDLR ectodomain using conventional techniques known in the art.
- a chemically synthesized oligonucleotide encoding the linker may be ligated between sequences encoding the first and second monomer, or the second and third monomer, or the third and fourth monomer, and fourth monomer and fifth monomer, etc.
- subject includes, but is not limited to, an organism or animal, including mammals and non-mammals.
- a mammal includes, e.g., but is not limited to, a human, non-human primate (e.g., baboon, orangutan, monkey, gorilla), mouse, dog, pig, cow, goat, cat, rabbit, rat, guinea pig, hamster, horse, sheep, or other non-human mammal.
- a non-mammal includes, e.g., but is not limited to, a non-mammalian invertebrate and non-mammalian vertebrate, such as a bird (e.g., a chicken or duck) or a fish.
- Bunyavirales order encompasses nine families of enveloped viruses containing a single-stranded negative-sense RNA genome divided into three segments.
- the small (S) and large (L) segments encode proteins participating in genome replication in the infected cell cytoplasm.
- Bunyavirales are vector-borne viruses transmitted mostly by bites of arthropods such as mosquitoes, ticks, flies, or livestock animals, with the exception of the viruses from the Hantaviridae family, which are transmitted by infectious excreta or bites of rodents and other small mammals. In certain cases, human-to-human transmission can occur due to close contact with infected blood or other bodily fluids.
- Bunyavirales are found throughout the world and are known to resist to adverse climate changes allowing for seasonal and persistent occurrence of the diseases. Bunyavirales are endemic in certain regions of the globe, such as Africa, the Middle East and Asia. In addition, outbreaks of Bunyavirales are often reported in both animals and humans.
- viruses of the order Bunyavirales are described as being human pathogens, and particularly the viruses of the genus Orthohantavirus (Hantaviridae family, also named Hantavirus) such as Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava-Belgrade hantavirus (DOBV), Haantan virus (HTNV), Vietnamese Negra hantavirus (LANV), Longquan hantavirus (LQIIV), Puumala hantavirus (PUIIV), Sangassou hantavirus (SANGV), Seoul hantavirus (SEOV), Sin Nombre hantavirus (SNV), Thailand hantavirus (THAIV), Tula hantavirus (TLILV), New York hantavirus (NYV), viruses of the genus Orthonairovirus such as Crimean-Congo hemorrhagic fever virus (CCHFV), Dugbe virus (DLIGV), Amsterdam sheep disease virus (CC
- Bunyavirales viruses cause a broad spectrum of clinical illnesses: self-limited febrile disease, respiratory and pulmonary syndromes, encephalitis, and life-threatening hemorrhagic fevers.
- CCHFV is a tripartite RNA genome virus with negative polarity of the genus Orthonairovirus and family Nairoviridae.
- the nairoviruses are predominantly tick- borne viruses.
- the genome of this virus is composed of the three different RNA segments small (S), medium (M) and large (L).
- S small segment ‘S’ encoding nucleoprotein (NP) is around 1.6kb long.
- the medium segment ‘M’ encoding glycoproteins is around 5.5kb long.
- the large segment ‘L’ encoding RNA - dependent RNA polymerase (RdRp or "L protein”) is around 12 kb long.
- the untranslated regions (UTRs) on 5' and 3' of the S, M and L segments are necessary for viral transcription, replication and packaging.
- the M segment generally encodes two structural glycoproteins, hereafter referred to as Gn and Gc, based on their location relative to the N and C termini of the (M segment encoded) polyprotein.
- the precursor of GP is cleaved and modified to generate the structural proteins Gn and Gc and the non-structural proteins GP38 and NSm.
- Gn and Gc structural glycoproteins
- NSm non-structural proteins
- the tri-segmented viral genome is coated with the viral NP and bound by the L protein.
- the viral proteins produce positive-sense viral RNA using the genomic negative-sense viral RNA as a template to initiate viral protein production and replication.
- the viral glycoproteins Gn and Gc are found on the virion surface and are responsible for receptor binding and viral entry.
- the present invention particularly relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain, and wherein said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Georgia Negra hantavirus (LANV), Longquan hantavirus (LQUV), Puumala hantavirus (PUUV), Sangassou
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Rift Valley fever virus (RVFV), Kenya sheep virus (NSDV), and Sandfly Fever Naples virus (SFNV), more preferably the Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- CCHFV Crimean-Congo Haemorrhagic Fever Virus
- RVFV Rift Valley fever virus
- NSDV Baltimore sheep virus
- SFNV Sandfly Fever Naples virus
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Rift Valley fever virus (RVFV), Kenya sheep virus (NSDV), and Sandfly Fever Naples virus (SFNV), more preferably the Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- CCHFV Crimean-Congo Haemorrhagic Fever Virus
- RVFV Rift Valley fever virus
- NSDV Baltimore sheep virus
- SFNV Sandfly Fever Naples virus
- the present invention more particularly relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an amino acid variant of an LDLR ectodomain, and wherein said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Madison Negra hantavirus (LANV), Longquan hantavirus (LQUV), Puumala hantavirus (PUUV), Sangassou hantavirus (SANGV), Seoul hantavirus (SEOV), Sin Nombre han
- An embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain having amino acid sequence SEQ ID NO 18, or a functional fragment thereof, or an amino acid variant thereof, and wherein said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Website), Georgia Negra hantavirus (LANV), Longquan hantavirus (LQUV), Puumala hantavirus (PUUV), Sangassou hanta
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Rift Valley fever virus (RVFV), Kenya sheep virus (NSDV), and Sandfly Fever Naples virus (SFNV), more preferably the Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- CCHFV Crimean-Congo Haemorrhagic Fever Virus
- RVFV Rift Valley fever virus
- NSDV Baltimore sheep virus
- SFNV Sandfly Fever Naples virus
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1)
- An alternative embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an amino acid variant of an LDLR ectodomain having amino acid sequence SEQ ID NO 18, or a functional fragment thereof, and wherein said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Website), Georgia Negra hantavirus (LANV), Longquan hantavirus (LQUV), Puumala hantavirus (PUUV), Sangassou hantavirus
- a further embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, wherein said LDLR derived polypeptide comprises a dimer, a trimer, or a multimer of an LDLR ectodomain monomer having amino acid sequence SEQ ID NO 18, or of a functional fragment thereof, or of an amino acid variant thereof, and wherein said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Georgia Negra hantavirus (LANV), Longquan hantavirus (LQUV), Puumal
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Rift Valley fever virus (RVFV), Kenya sheep virus (NSDV), and Sandfly Fever Naples virus (SFNV), more preferably the Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- CCHFV Crimean-Congo Haemorrhagic Fever Virus
- RVFV Rift Valley fever virus
- NSDV Baltimore sheep virus
- SFNV Sandfly Fever Naples virus
- a further alternative embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, wherein said LDLR derived polypeptide comprises a dimer, a trimer, or a multimer of an amino acid variant of an LDLR ectodomain monomer having amino acid sequence SEQ ID NO 18, or of a functional fragment thereof, and wherein said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Georgia Negra hantavirus (LANV), Longquan hantavirus (LQUV), Puumala
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1). More preferably, said multimer comprises between 2 and 8 monomers of an LDR ectodomain, or of a functional fragment thereof or of an amino acid variant thereof.
- a further embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41),
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Rift Valley fever virus (RVFV), Kenya sheep virus (NSDV), and Sandfly Fever Naples virus (SFNV), more preferably the Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- CCHFV Crimean-Congo Haemorrhagic Fever Virus
- RVFV Rift Valley fever virus
- NSDV Baltimore sheep virus
- SFNV Sandfly Fever Naples virus
- a further alternative embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an amino acid variant of a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1) Moreover, the present invention particularly relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain, and wherein said Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain having amino acid sequence SEQ ID NO 18, or a functional fragment thereof, or an amino acid variant thereof, and wherein said Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1)
- a further embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, wherein said LDLR derived polypeptide comprises a dimer, a trimer, a homodimer, a homotrimer, or a multimer of an LDLR ectodomain monomer having amino acid sequence SEQ ID NO 18, or of a functional fragment thereof, or of an amino acid variant thereof, and wherein said Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1). More preferably, said multimer comprises between 2 and 8 monomers, preferably 2 or 3 monomers of an LDLR ectodomain, or of a functional fragment thereof or of an amino acid variant thereof.
- a further embodiment of the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41),
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1). More preferably, said multimer comprises between 2 and 8 monomers of an LDR ectodomain, or of a functional fragment thereof or of an amino acid variant thereof.
- the present invention relates to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain having amino acid sequence SEQ ID NO 18, and wherein said Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- the present invention relates to a recombinant nucleic acid molecule comprising a nucleic acid sequence encoding an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain, and wherein said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Website, Website, Website, Georgia Negra hantavirus (LANV),
- the present invention relates to a recombinant nucleic acid molecule comprising a nucleic acid sequence encoding an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an amino acid variant of an LDLR ectodomain or a functional fragment thereof, and wherein said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Georgia Negra hantavirus (LANV), Longquan hantavirus (LQUV), Puumala hantavirus (CCHF
- An embodiment of the present invention is directed to a pharmaceutical composition
- a pharmaceutical composition comprising the LDLR derived polypeptide described herein, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent.
- composition refers to a composition suitable for pharmaceutical use in a subject, including an animal or human.
- a pharmaceutical composition typically comprises an effective amount of an active agent and a carrier, excipient, or diluent.
- the carrier, excipient, or diluent is typically a pharmaceutically acceptable carrier, excipient or diluent, respectively.
- compositions of the invention are prepared for administration by mixing the LDLR derived polypeptide, with physiologically acceptable carriers, stabilizers, and excipients, and prepared in dosage form, e.g. by lyophilization in dosage vials.
- a specific embodiment of the present invention is directed to a pharmaceutical composition
- a pharmaceutical composition comprising an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent.
- the present invention is also directed to a pharmaceutical composition
- a pharmaceutical composition comprising the LDLR derived polypeptide as disclosed herein together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent for use in the prophylaxis and/or treatment of an infection by a Bunyavirus.
- the present invention is directed to a recombinant nucleic acid molecule comprising a nucleic acid sequence encoding an LDLR derived polypeptide as disclosed herein for use in the prophylaxis and/or treatment of an infection by a Bunyavirus.
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1)
- Said Bunyavirales virus is preferably selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website
- LANV Longquan hantavirus
- LQUV Longquan hantavirus
- PUUV Puumala hantavirus
- SANGV Sangassou hantavirus
- SEOV Sin Nombre hantavirus
- SNV Sin Nombre hantavirus
- TAAIV Tula hantavirus
- NW New York hantavirus
- RVFV Rift Valley fever virus
- NSDV Sandfly Fever Naples virus
- SFTSV Severe fever with thrombocytopenia syndrome virus
- SFTSV Sandfly Fever Naples virus
- An alternative embodiment of the present invention is directed to a pharmaceutical composition
- a pharmaceutical composition comprising an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an amino acid variant of an LDLR ectodomain, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent, and wherein said Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Georgia Negra hantavirus (LANV), Longquan hantavirus (LQUV), Puumala
- a more specific embodiment of the present invention is directed to a pharmaceutical composition
- a pharmaceutical composition comprising an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain having amino acid sequence SEQ ID NO 18, or a functional fragment thereof, or an amino acid variant thereof.
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1)
- Said Bunyavirales virus is preferably selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website
- LANV Longquan hantavirus
- LQUV Longquan hantavirus
- PUUV Puumala hantavirus
- SANGV Sangassou hantavirus
- SEOV Sin Nombre hantavirus
- SNV Sin Nombre hantavirus
- TAAIV Tula hantavirus
- NW New York hantavirus
- RVFV Rift Valley fever virus
- NSDV Sandfly Fever Naples virus
- SFTSV Severe fever with thrombocytopenia syndrome virus
- SFTSV Sandfly Fever Naples virus
- a specific embodiment of the present invention is directed to a pharmaceutical composition
- a pharmaceutical composition comprising an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent, and wherein said LDLR derived polypeptide comprises a dimer, a trimer, a homodimer, a homotrimer, or a multimer of an LDLR ectodomain monomer having amino acid sequence SEQ ID NO 18, or of a functional fragment thereof, or of an amino acid variant thereof.
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1). More preferably, said multimer comprises between 2 and 8 monomers of an LDR ectodomain, or of a functional fragment thereof or of an amino acid variant thereof.
- Said Bunyavirales virus is preferably selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website
- LANV Longquan hantavirus
- LQUV Longquan hantavirus
- PUUV Puumala hantavirus
- SANGV Sangassou hantavirus
- SEOV Sin Nombre hantavirus
- SNV Sin Nombre hantavirus
- TAAIV Tula hantavirus
- NW New York hantavirus
- RVFV Rift Valley fever virus
- NSDV Sandfly Fever Naples virus
- SFTSV Severe fever with thrombocytopenia syndrome virus
- SFTSV Sandfly Fever Naples virus
- a specific embodiment of the present invention is directed to a pharmaceutical composition
- a pharmaceutical composition comprising an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent, and wherein said LDLR derived polypeptide comprises a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID
- said LDLR derived polypeptide further comprises a peptide linker connecting said fragments. More preferably, a peptide linker connecting said functional fragments is selected from the group comprising Linked to Linker76 (Table 1). More preferably, said multimer comprises between 2 and 8 monomers of an LDR ectodomain, or of a functional fragment thereof or of an amino acid variant thereof.
- Said Bunyavirales virus is preferably selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website
- LANV Longquan hantavirus
- LQUV Longquan hantavirus
- PUUV Puumala hantavirus
- SANGV Sangassou hantavirus
- SEOV Sin Nombre hantavirus
- SNV Sin Nombre hantavirus
- TAAIV Tula hantavirus
- NYV New York hantavirus
- RVFV Rift Valley fever virus
- NSDV Sandfly Fever Naples virus
- SFTSV Severe fever with thrombocytopenia syndrome virus
- SFNV Sandfly Fever Naples virus
- a specific embodiment of the present invention is directed to a pharmaceutical composition
- a pharmaceutical composition comprising an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain having amino acid sequence SEQ ID NO 18, and wherein said Bunyavirales virus is CCHFV or RVFV and most preferably CCHFV.
- the term "effective amount" refers to a dosage (or dose) or amount of a substance sufficient to produce a desired result.
- the desired result may comprise an objective or subjective improvement in the recipient of the dosage or amount.
- the desired result may comprise a measurable, or detectable inhibition of a virus infection in a subject to whom a dosage or amount of a particular LDLR ectodomain or a fragment thereof has been administered.
- a “prophylactic treatment” is a treatment administered to a subject who does not display signs or symptoms of, or displays only early signs or symptoms of, a disease, pathology, or disorder, such that treatment is administered for the purpose of preventing or decreasing the risk of developing the disease, pathology, or disorder.
- a prophylactic treatment functions as a preventative treatment against a disease, pathology, or disorder, or as a treatment that inhibits or reduces further development or enhancement of a disease, pathology or disorder.
- a “prophylactic activity” is an activity of an agent that, when administered to a subject who does not display signs or symptoms of, or who displays only early signs or symptoms of, a pathology, disease, or disorder, prevents or decreases the risk of the subject developing the pathology, disease, or disorder.
- a “prophylactically useful” agent refers to an agent that is useful in preventing development of a disease, pathology, or disorder, or useful in inhibiting or reducing further development or enhancement of a disease, pathology or disorder.
- a “therapeutic treatment” is a treatment administered to a subject who displays symptoms or signs of pathology, disease, or disorder, in which treatment is administered to the subject for the purpose of diminishing or eliminating those signs or symptoms.
- a “therapeutic activity” is an activity of an agent that eliminates or diminishes signs or symptoms of pathology, disease or disorder when administered to a subject suffering from such signs or symptoms.
- a “therapeutically useful” agent means the agent is useful in decreasing, treating, or eliminating signs or symptoms of a disease, pathology, or disorder.
- LDLR derived polypeptide of functional fragments or of a multimer thereof LDLR derived polypeptide of functional fragments or of a multimer thereof.
- a second aspect of the present invention is directed to an LDLR derived polypeptide comprising a functional fragment of an LDLR ectodomain selected from the group LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41), OLSD (SEQ ID NO 36), LA1-LA2-LA3-LA4X-LA5-LA6X-LA7-EGFA-EGFBX- PROPX-EGFCX-OLSD, LA1 -LA2-LA3-LA
- amino acid variant refers to a single amino acid variant or to a multiple amino acid variant as previously defined.
- An embodiment of the second aspect is directed to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a virus infection, wherein said virus is preferably a Bunyavirales virus, wherein said LDLR derived polypeptide comprises a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41 ), OLSD (SEQ ID NO 36), LA1-
- Said Bunyavirales virus is preferably selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website
- CATV Longquan hantavirus
- LQUV Longquan hantavirus
- PUUV Puumala hantavirus
- SANGV Sangassou hantavirus
- SEOV Sin Nombre hantavirus
- SNV Sin Nombre hantavirus
- TAAIV Tula hantavirus
- NW New York hantavirus
- RVFV Rift Valley fever virus
- NSDV Sandfly Fever Naples virus
- Said Bunyavirales virus is more preferably CCHFV or RVFV and most preferably CCHFV.
- a particular embodiment of the second aspect is directed to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a virus infection, wherein said virus is preferably a RVF virus (RVFV), wherein said LDLR derived polypeptide comprises an amino acid variant of a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID
- a further aspect of the present invention is directed to a LDLR derived polypeptide comprising a multimer of monomers, wherein each monomer is selected from the group comprising: an LDLR ectodomain (SEQ ID NO 18), a functional fragment of said LDLR ectodomain, an amino acid variant of said LDLR ectodomain, an amino acid variant of said functional fragment of said LDLR ectodomain, and wherein said multimer comprises between 2 and 8 monomers, and wherein said LDLR derived polypeptide optionally comprises a peptide linker connecting said monomers; and wherein said multimer is a homomultimer or a heteromultimer.
- amino acid variant refers to a single amino acid variant or to a multiple amino acid variant as previously defined.
- a particular further aspect is directed to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a virus infection, wherein said virus is preferably a Bunyavirales virus, wherein said LDLR derived polypeptide comprises a multimer of monomers, wherein each monomer is selected from the group comprising: an LDLR ectodomain (SEQ ID NO 18), a functional fragment of said LDLR ectodomain, an amino acid variant of said LDLR ectodomain, an amino acid variant of said functional fragment of said LDLR ectodomain, and wherein said multimer comprises between 2 and 8 monomers, and wherein said LDLR derived polypeptide optionally comprises a peptide linker connecting said monomers; and wherein said multimer is a homomultimer or a heteromultimer.
- Said Bunyavirales virus is preferably selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website
- HTNV Haantan virus
- LQLIV Longquan hantavirus
- PIIV Puumala hantavirus
- SEOV Seoul hantavirus
- Sin Nombre hantavirus SNV
- Thailand hantavirus TAAIV
- Tula hantavirus TILV
- NYV New York hantavirus
- RVFV Rift Valley fever virus
- NSDV Sandfly Fever Naples virus
- Said Bunyavirales virus is more preferably a CCHF virus or RVFV and most preferably CCHFV.
- a more particular further aspect is directed to an LDLR derived polypeptide for use in the prophylaxis and/or treatment of a virus infection, wherein said virus is preferably a RVF virus, wherein said LDLR derived polypeptide comprises a multimer of monomers, wherein each monomer is selected from the group comprising: an amino acid variant of an LDLR ectodomain (SEQ ID NO 18), an amino acid variant of a functional fragment of said LDLR ectodomain, and wherein said multimer comprises between 2 and 8 monomers, preferably 2 or 3 monomers, and wherein said LDLR derived polypeptide optionally comprises a peptide linker connecting said monomers; and wherein said multimer is a homomultimer or a heteromultimer.
- SEQ ID NO 18 amino acid variant of an LDLR ectodomain
- said multimer comprises between 2 and 8 monomers, preferably 2 or 3 monomers
- said LDLR derived polypeptide optionally comprises a peptide linker connecting said
- the present invention also discloses a pharmaceutical composition
- a pharmaceutical composition comprising said LDLR derived polypeptide, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent, wherein said LDLR derived polypeptide comprises a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41), OLSD (SEQ ID NO 36), LA1 -LA2-LA
- the present invention also discloses a pharmaceutical composition
- a pharmaceutical composition comprising said LDLR derived polypeptide, together with at least one pharmaceutically acceptable vehicle, excipient and/or diluent, wherein said LDLR derived polypeptide comprises a multimer of monomers, wherein each monomer is selected from the group comprising: an LDLR ectodomain (SEQ ID NO 18), a functional fragment of said LDLR ectodomain, an amino acid variant of said LDLR ectodomain, an amino acid variant of said functional fragment of said LDLR ectodomain, and wherein said multimer comprises between 2 and 8 monomers, preferably 2 or 3 monomers, and wherein said LDLR derived polypeptide optionally comprises a peptide linker connecting said monomers; and wherein said multimer is a homomultimer or a heteromultimer.
- the invention also contemplates a recombinant nucleic acid molecule comprising a nucleic acid sequence encoding an LDLR derived polypeptide, wherein said LDLR derived polypeptide comprises a functional fragment of an LDLR ectodomain selected from the group comprising LA1 (SEQ ID NO 19), LA2 (SEQ ID NO 20), LA3 (SEQ ID NO 21 ), LA4 (SEQ ID NO 22), LA4X (SEQ ID NO 37), LA5 (SEQ ID NO 23), LA6 (SEQ ID NO 24), LA6X (SEQ ID NO 38), LA7 (SEQ ID NO 25), EGFA (SEQ ID NO 26), EGFB (SEQ ID NO 27), EGFBX (SEQ ID NO 39), PROP (SEQ ID NO 34), PROPX (SEQ ID NO 40), EGFC (SEQ ID NO 35), EGFCX (SEQ ID NO 41), OLSD (SEQ ID NO 36), LA1 -LA2-LA3-LA4X-LA5
- the invention also contemplates a recombinant nucleic acid molecule comprising a nucleic acid sequence encoding an LDLR derived polypeptide, wherein said LDLR derived polypeptide comprises a multimer of monomers, wherein each monomer is selected from the group comprising: an LDLR ectodomain (SEQ ID NO 18), a functional fragment of said LDLR ectodomain, an amino acid variant of said LDLR ectodomain, an amino acid variant of said functional fragment of said LDLR ectodomain, and wherein said multimer comprises between 2 and 8 monomers, preferably 2 or 3 monomers, and wherein said LDLR derived polypeptide optionally comprises a peptide linker connecting said monomers; and wherein said multimer is a homomultimer or a heteromultimer.
- the inventors were able to identify LDLR as involved in CCHF virus infection by using a screening method based on infection of randomly mutagenized murine haploid cells with a CCHFV pseudotyped VSV, selection of mutant resistant clones and whole exome sequencing to individuate the gene mutations responsible for CCHFV resistance ( Figure 1a-c, Table 2).
- a diploid cell contains two copies of genomic DNA.
- a point mutation introduced by chemical mutagenesis always affects only one allele, while the second copy remains intact.
- One intact copy of a host cell factor is typically sufficient for a virus to hijack the cellular machinery and replicate.
- a haploid cell is a cell that only carries a single copy of the genome. Therefore, a point mutation introduced in a haploid cell alters 100% of the cellular protein. In case of a mutation interfering with viral entry or replication, this would completely block infection in the haploid cell and allow its identification in a screening setup. In sum, recessive mutations with regards to the viral infection can be identified.
- diploid is used herein to specifically refer to a cell or cell line including a genome wherein the cell is diploid for one or more specific genomic loci, or for the full genome, i.e. "fully diploid" cells.
- haploid as used herein shall specifically refer to a cell or cell line including a single copy of the genome. Haploidy may be determined or tested by known methods, e.g. spectral karyotyping, comparative genomic hybridization or comparative propidium iodide staining.
- near-haploid cell is a cell in which no more than 5 chromosomes are present in two or more copies. Near-haploid cells were found to maintain their status several months in culture.
- An exemplary near-haploid somatic human cell is of a KBM-7 cell line, which is haploid for most chromosomes with the exception of chromosome 8, and optionally a portion of chromosome 15, and is a non-adherent cell line.
- a further example of a near-haploid cell line is the HAP1 cell line, which is an adherent cell line obtained by engineering the KBM-7 cell line, which has lost the second copy of chromosome 8, and is hence “more haploid” than its KBM-7 parent, but still retains a portion of chromosome 15 and can therefore not be considered fully haploid.
- Further near-haploid cell lines may be derived from a cancer patient, or from a patient suffering from leukemia.
- HAP2 cell line which is obtained by engineering HAP1 cells through excision of the portion of chromosome 15 that retained its diploidy in the HAP1 cell line, thus, is considered truly or fully haploid. It turned out that the HAP2 cell line comprises the complete set of human chromosomes in the monosomic state.
- the eHAP1 cell line is a human fully haploid cell line deriving from HAP1 cells, which have been genetically engineered to delete the diploid portion of chromosome 15.
- eHAP1 is commercially available (e.g. Horizon Discovery (SKU: c669).
- mutagenesis as used in the context of the present invention shall refer to a method of providing mutants of a sequence, e.g. through insertion, deletion and/or substitution of one or more nucleotides or amino acids, so to obtain variants thereof. Mutagenesis may be through random, semi-random or site directed mutation.
- haploid murine stem cells refers to haploid murine stem cells that can be obtained and cultured as described in the prior art (Eiling, et al., Nat Protoc. 2019 Jul 1 ; 14(7): 1991-2014), or can be stabilized cell lines such as AN3-12, H129-2, H129-1. Therefore, in some embodiments, haploid murine stem cells are selected from the group comprising primary haploid murine stem cells, AN3-12, H129-2, H129-1 , and derivatives thereof.
- haploid human stem cells refers to haploid human stem cells that can be obtained and cultured as described in the prior art, e.g in Sagi I, et al. Identification and propagation of haploid human pluripotent stem cells. Nat Protoc. 2016 11(11 ): 2274-2286.
- haploid human stem cells refers to haploid human stem cells of different origins, such as for example, haploid human embryonic stem cells (hESCs), haploid human spermatogonial stem cells, haploid hematopoietic stem cell,
- PV pseudovirus
- pp pseudoparticles
- pseudotyped virus refers to a virus expressing heterologous viral envelope proteins.
- a pseudotyped VSV has the envelope protein of a heterologous virus assembled into the VSV membrane.
- VSV is a bullet-shaped, negative polarity enveloped RNA virus in the Rhabdoviridae family.
- VSV has a genome size of 11 kb that contains five main viral proteins: nucleoprotein (N), phosphoprotein (P), matrix protein (M), glycoprotein (G), and large polymerase protein (L).
- VSV-AG Recombinant virus VSV in which the glycoprotein (G) gene is deleted (rVSV-AG) (Whitt, J Virol Methods. 2010 Nov; 169(2): 365-374.) has been used to produce VSV pseudotypes containing the envelope glycoproteins of heterologous viruses, including viruses that require high-level biocontainment, such as Bunyavirales.
- rVSV-AG pseudotypes The advantage of rVSV-AG pseudotypes is that it performs only one single round of replication, so that the experiments can be performed using biosafety level 2 (BSL-2) containment.
- rVSV-AG expressing heterologous envelope glycoproteins can be used to analyse genetic factors involved in infection of viral pathogens without the need for specialized containment facilities.
- RNA viral particles pseudotyped with Bunyavirales glycoproteins can be obtained by using a method described in the prior art, such as for example in Cegolon et al 2021 .
- Preferred viruses are HIV, MSV, and VSV.
- Random mutagenesis can be performed by a method selected from chemical random mutagenesis, UV mediated random mutagenesis, transposon mutagenesis, or error prone PCR.
- random mutagenesis is performed by chemical random mutagenesis, and more preferably by incubating the haploid cells with Ethylnitrosurea (ENU) as described in the prior art.
- Ethylnitrosurea Ethylnitrosurea
- Bunyavirales glycoproteins Gn and Gc are derived from a precursor polyprotein by host cell proteases. Bunyavirales glycoproteins have been described in the prior art, for example in Hulswit, RJC et al, Viruses. 2021 Feb; 13(2): 353.
- whole exome sequencing refers to the sequencing of all exons in a specific cell or tissue type, wherein the exons are the protein coding regions of a gene. Therefore, in “whole exome sequencing” (WES) only the protein-coding DNA regions are sequenced as compared to whole genome sequencing (WGS).
- the term “whole transcriptome sequencing” refers to sequencing of expressed transcripts at the mRNA level to obtain sequence information on all mRNAs made in a cell or cell pool at a given time
- NGS next generation sequencing
- NGS refers to a group of platforms to perform sequencing of millions of small fragments of DNA in parallel. NGS can be used for whole genome sequencing, whole exome sequencing, or whole transcriptome sequencing.
- NGS can be used for whole genome sequencing, whole exome sequencing, or whole transcriptome sequencing.
- the terms “next generation sequencing”, “massively parallel sequencing", and “deep sequencing” are related terms and can be used interchangeably.
- a third aspect of the present invention is directed to an in vitro screening method to identify a gene that modulates a Bunyavirales virus infection, the method comprising:
- step B) performing chemical random mutagenesis in said cells of step A) in order to obtain mutagenized haploid or near-haploid cells;
- step B) infecting the mutagenized cells of step B) with a Bunyavirales virus or with a Bunyavirales virus pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized cells of step C) to obtain resistant colonies
- step E) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate said Bunyavirales virus infection
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof
- the pseudotyped RNA viral particle is a HIV particle or a MSV particle or a VSV particle
- the pseudotyped RNA viral particle express a Bunyavirales virus specific glycoprotein Gc and /or Gn
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade han
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid or near-haploid cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- the present invention is directed to an in vitro screening method to identify a gene that modulates a Bunyavirales virus infection, the method comprising:
- step B) performing chemical random mutagenesis in said cells of step A) in order to obtain mutagenized haploid or near-haploid cells;
- step B) infecting the mutagenized cells of step B) with a Bunyavirales virus or with a Bunyavirales virus pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized cells of step C) to obtain resistant colonies
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof, wherein the pseudotyped RNA viral particle is a HIV particle or a MSV particle or a VSV particle, and wherein the pseudotyped RNA viral particle express a Bunyavirales virus specific glycoprotein Gc and /or Gn, and wherein the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Bel
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid or near-haploid cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- An alternative embodiment of the present invention is directed to an in vitro screening method to identify a gene that modulates a Bunyavirales virus infection, the method comprising:
- step B) performing chemical random mutagenesis in said cells of step A) in order to obtain mutagenized haploid or near-haploid cells;
- step B) infecting the mutagenized cells of step B) with a Bunyavirales virus or with a Bunyavirales virus pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized cells of step C) to obtain resistant colonies
- step E) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate said Bunyavirales virus infection
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof
- the pseudotyped RNA viral particle is a HIV particle or a MSV particle or a VSV particle
- the pseudotyped RNA viral particle express a Bunyavirales virus specific glycoprotein Gc and /or Gn
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade han
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid or near-haploid cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- the present invention is directed to an in vitro screening method to identify a gene that modulates a Bunyavirales virus infection, the method comprising:
- step B) performing chemical random mutagenesis in said haploid or near- haploid cells of step A) in order to obtain mutagenized haploid or near- haploid cells;
- step B) infecting the mutagenized haploid or near-haploid cells of step B) with a Bunyavirales virus or with a Bunyavirales virus pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step E) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate said Bunyavirales virus infection
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof
- the pseudotyped RNA viral particle is a HIV particle or a MSV particle or a VSV particle
- the pseudotyped RNA viral particle express a Bunyavirales virus specific glycoprotein Gc and /or Gn
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Rift Valley fever virus (RVFV), Kenya sheep virus (NSDV), and Sandfly Fever Naples virus (SFNV), preferably
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid or near-haploid cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- the present invention is directed to in vitro screening method to identify a gene that modulates a Bunyavirales virus infection, the method comprising:
- step B) performing chemical random mutagenesis in said haploid or near- haploid cells of step A) in order to obtain mutagenized haploid or near- haploid cells;
- step C) infecting the mutagenized haploid or near-haploid cells of step B) with a Bunyavirales virus
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate said Bunyavirales virus infection, and wherein the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof, wherein the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Georgia Negra hantavirus (LANV), Longquan hantavirus (LQIIV), Puumala hantavirus (PUIIV), Sangas
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid cells with N-Ethyl- N-nitrosourea (ENU).
- ENU N-Ethyl- N-nitrosourea
- the present invention is directed to in vitro screening method to identify a gene that modulates a Bunyavirales virus infection, the method comprising:
- step B) performing chemical random mutagenesis in said haploid or near- haploid cells of step A) in order to obtain mutagenized haploid or near- haploid cells;
- step C) infecting the mutagenized haploid or near-haploid cells of step B) with a Bunyavirales virus pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step E) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate said Bunyavirales virus infection
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof
- the pseudotyped RNA viral particle is a HIV particle or a MSV particle or a VSV particle
- the pseudotyped RNA viral particle express a Bunyavirales virus specific glycoprotein Gc and /or Gn
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hant
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid cells with N-Ethyl- N-nitrosourea (ENU).
- ENU N-Ethyl- N-nitrosourea
- the present invention is directed to an in vitro screening method to identify a gene that modulates a Bunyavirales virus infection, the method comprising:
- step B) performing chemical random mutagenesis in said haploid or near- haploid cells of step A) in order to obtain mutagenized haploid or near- haploid cells;
- step C) infecting the mutagenized haploid or near-haploid cells of step B) with a Bunyavirales virus pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate said Bunyavirales virus infection, wherein the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof, wherein the pseudotyped RNA viral particle is a VSV particle, and wherein the pseudotyped RNA viral particle express a Bunyavirales virus specific glycoprotein Gc and /or Gn, wherein sequencing of step f) comprises whole genome sequencing or whole exome sequencing or whole transcriptome sequencing.
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid or near-haploid cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- the present invention is directed to an in vitro screening method to identify a gene that modulates a Bunyavirales virus infection, the method comprising:
- step B) performing chemical random mutagenesis in said haploid or near- haploid cells of step A) in order to obtain mutagenized haploid or near- haploid cells;
- step C) infecting the mutagenized haploid or near-haploid cells of step B) with a Bunyavirales virus pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step E) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate said Bunyavirales virus infection
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof
- the pseudotyped RNA viral particle is a VSV particle
- the pseudotyped RNA viral particle express a Bunyavirales virus specific glycoprotein Gc and /or Gn
- the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (CCHFV
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid or near-haploid cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- the third aspect of the invention is directed to an in vitro screening method to identify a gene that modulates a CCHFV infection, the method comprising:
- step B) performing chemical random mutagenesis in said haploid or near- haploid cells of step A) in order to obtain mutagenized haploid or near- haploid cells;
- step B) infecting the mutagenized haploid or near-haploid cells of step B) with a CCHFV or with a CCHFV glycoprotein pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate a CCHFV infection
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof, wherein the pseudotyped RNA viral particle is a HIV particle or a MSV particle or a VSV particle, and wherein said CCHFV glycoprotein is CCHFV glycoprotein Gc and/or Gn, wherein sequencing of step f) comprises whole genome sequencing or whole exome sequencing or whole transcriptome sequencing.
- performing chemical random mutagenesis in said haploid or near- haploid cells of step B) in order to obtain mutagenized haploid or near-haploid cells comprises incubating the haploid cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- a particular embodiment is directed to an in vitro screening method to identify a gene that modulates a CCHFV infection, the method comprising:
- step B) performing chemical random mutagenesis in said haploid or near- haploid cells of step A) in order to obtain mutagenized haploid or near- haploid cells;
- step B) infecting the mutagenized haploid or near-haploid cells of step B) with a CCHFV glycoprotein pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate a CCHFV infection, wherein the pseudotyped RNA viral particle is a VSV particle, and wherein said CCHFV glycoprotein is CCHFV glycoprotein Gc and/or Gn, wherein sequencing of step f) comprises whole genome sequencing or whole exome sequencing or whole transcriptome sequencing.
- a further embodiment is directed to an in vitro screening method to identify a gene that modulates a CCHFV infection, the method comprising:
- step B) performing chemical random mutagenesis in said haploid or near- haploid cells of step A) in order to obtain mutagenized haploid or near- haploid cells by incubating the haploid or near-haploid cells with N- Ethyl-N-nitrosourea (ENU);
- ENU N- Ethyl-N-nitrosourea
- step B) infecting the mutagenized haploid or near-haploid stem cells of step B) with a CCHFV glycoprotein pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate a CCHFV infection
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof, wherein the pseudotyped RNA viral particle is a HIV particle or a MSV particle or a VSV particle, and wherein said CCHFV glycoprotein is CCHFV glycoprotein Gc and/or Gn, wherein sequencing of step f) comprises whole genome sequencing or whole exome sequencing or whole transcriptome sequencing.
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid or near-haploid cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- a further particular embodiment is directed to an in vitro screening method to identify a gene that modulates a CCHFV infection, the method comprising:
- step B) performing chemical random mutagenesis in said cells of step A) in order to obtain mutagenized haploid or near-haploid cells by incubating said cells with N-Ethyl-N-nitrosourea (ENU);
- ENU N-Ethyl-N-nitrosourea
- step B) infecting the mutagenized haploid or near-haploid cells of step B) with a CCHFV glycoprotein pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate a CCHFV infection
- the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof, wherein the pseudotyped RNA viral particle is a VSV particle, and wherein said CCHFV glycoprotein is CCHFV glycoprotein Gc and/or Gn, wherein sequencing of step f) comprises whole genome sequencing or whole exome sequencing or whole transcriptome sequencing.
- a further more particular embodiment is directed to an in vitro screening method to identify a gene that modulates a CCHFV infection, the method comprising:
- step B) infecting the mutagenized haploid or near-haploid cells of step B) with CCHFV;
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate a CCHFV infection, wherein sequencing of step f) comprises whole genome sequencing or whole exome sequencing or whole transcriptome sequencing.
- step B) of performing chemical random mutagenesis in said haploid or near-haploid cells of step A) in order to obtain mutagenized haploid or near-haploid cells preferably comprises incubating the haploid or near-haploid cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- an embodiment is directed to an in vitro screening method to identify a gene that modulates a CCHFV infection, the method comprising:
- step B) performing chemical random mutagenesis in said haploid or near- haploid cells of step A) in order to obtain mutagenized haploid or near- halpoid cells by incubating the said cells with N-ethyl-N-nitrosourea (ENU);
- ENU N-ethyl-N-nitrosourea
- step B) infecting the mutagenized haploid or near-haploid cells of step B) with a CCHFV;
- step D) culturing the infected mutagenized haploid or near-haploid cells of step C) to obtain resistant colonies;
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate a CCHFV infection, wherein the haploid cells are selected from the group comprising human haploid stem cells, or murine haploid stem cells, or human eHAP1 , or human HAP2, or preferably murine haploid stem cells or wherein the near-haploid cells are selected from the group comprising KBM-7 cells, HAP1 cells, and derivatives thereof, wherein sequencing of step f) comprises whole genome sequencing or whole exome sequencing or whole transcriptome sequencing.
- a more specific embodiment is directed to an in vitro screening method to identify a gene that modulates a CCHFV infection, the method comprising:
- step B) performing chemical random mutagenesis in said cells of step A) in order to obtain mutagenized haploid murine stem cells
- step B) infecting the mutagenized cells of step B) with a Bunyavirales virus or with a Bunyavirales virus pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized cells of step C) to obtain resistant colonies
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate a Bunyavirales virus infection, wherein the pseudotyped RNA viral particle is a HIV particle or a MSV particle or a VSV particle, wherein the pseudotyped RNA viral particle express a Bunyavirales virus specific glycoprotein Gc and/or Gn, and wherein the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Website), Georgia Negra hantavirus (LANV), Longquan hantavirus (LQIIV), Puumala hantavirus (PUIIV), Sangassou hantavirus (SANGV), Seoul hantavirus (SEOV), Sin Nombre
- the pseudotyped RNA viral particle is preferably a VSV particle.
- step B) comprises incubating the haploid murine stem cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- step B) performing chemical random mutagenesis in said cells of step A) in order to obtain mutagenized haploid murine stem cells
- step B) infecting the mutagenized cells of step B) with a Bunyavirales virus or with a Bunyavirales virus pseudotyped RNA viral particle;
- step D) culturing the infected mutagenized cells of step C) to obtain resistant colonies
- step F) performing sequencing of the resistant colonies of step E) in order to identify the genes that modulate a Bunyavirales virus infection, wherein the pseudotyped RNA viral particle is a VSV particle, wherein the pseudotyped VSV particle express a Bunyavirales virus specific glycoprotein Gc and/or Gn, and wherein the Bunyavirales virus is selected from the group comprising or consisting of Crimean-Congo Haemorrhagic Fever Virus (CCHFV), Hantavirus, Andes hantavirus (ANDV), Black Creek Canal hantavirus virus (BCCV), Dobrava- Belgrade hantavirus (DOBV), Haantan virus (HTNV), Website Website, Longquan hantavirus (LANV), Longquan hantavirus (LQIIV), Puumala hantavirus (PUIIV), Sangassou hantavirus (SANGV), Seoul hantavirus (SEOV), Sin Nombre hantavirus (SNV), Thailand han
- step B) comprises incubating the haploid murine stem cells with N-Ethyl-N-nitrosourea (ENU).
- ENU N-Ethyl-N-nitrosourea
- the binding of the LDLR derived polypeptide can also be used for the detection of a Bunyavirales virus infection. Therefore, the present application is also directed to the use of an LDLR derived polypeptide in the diagnosis of a Bunyavirales virus infection, wherein said LDLR derived polypeptide binds to said Bunyavirales virus, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain as disclosed herein and wherein said binding diagnoses the infection by the Bunyavirales virus.
- the binding of the LDLR derived polypeptide can also be used for the quality control of vaccines produced to prevent or ameliorate Bunyavirales virus infection, and especially CCHFV infection. Therefore, the present application is also directed to the use of an LDLR derived polypeptide in quality control of a Bunyavirales virus vaccine, especially a CCHFV vaccine, wherein said LDLR derived polypeptide binds to vaccine ingredients, and wherein said LDLR derived polypeptide comprises an LDLR ectodomain or a functional fragment of an LDLR ectodomain or an amino acid variant of an LDLR ectodomain as disclosed herein and wherein said binding assesses the quality of that vaccine to prevent or ameliorate a Bunyavirales virus infection.
- Figure 1 shows generation of VSV-CCHF_G and haploid cells screening.
- a Schematic representation of the methods used to produce the VSV- CCHF_G pseudotyped virus in HEK293T cells.
- b Validation of resistant clones obtained in the primary haploid screen with VSV- CCHF_G. Each clone (1-13) was isolated, amplified and assessed for infection with the CCHFV lbAR10200 laboratory strain (MOI 0.1). The data show the level of infection for each clone compared to wild-type haploid cells (AN3-12) as determined by RT-PCR for CCHFV and RNase P RNA 24hpi.
- c Deep exome sequencing identified three distinct single point mutations in the Ldlr gene in three resistant clones (5, 8, 10) resulting in stop codons or a missense mutation.
- Figure 2 shows CCHFV infections in Ldlr knockout cells
- a Level of infections in control wild type AN3-12 haploid and sister knock out (KO) cells infected with VSV-CCHF_G, CCHFV lbAr10200 (MOI 0.1 , 48hpi). Level of infection was assessed by RT-qPCR for viral and RNase P RNA.
- b Levels of infection of lbAr10200 CCHFV in wild type (WT) and two different LDLR KO (clones C2 and C12) Vero cells and c, in three different clones of LDLR KO (clones C8, C10 and C11) A549 cells.
- Figure 3 shows that recombinant CCHFV Gc protein binds to and induces the internalization of LDLR.
- a Illustration depicting the BRET-based binding assay that was used to measure the interaction of BODIPY-FL labelled LDL and soluble BODIPY-FL labelled CCHFV Gc with the LDLR.
- Figure 4 shows inhibition of CCHFV infections by soluble LDLR.
- a Levels of VSV- CCHF_G infections in human SW13 cells, treated with the indicated range of soluble LDLR concentrations or left untreated (mock-treated) (MOI 0.01 , 6hpi)
- b Levels of lbAr10200 CCHFV infections in SW13 cells, treated with a range of soluble LDLR concentrations (MOI 0.01 , 24hpi).
- c Level of VSV infections in SW13 cells, treated with the indicated concentrations of soluble LDLR (MOI 0.01 , 6hpi).).
- Figure 5 shows CCHFV infections in human BVOs and Ldlr mutant mice, a, Scheme representing blood vessel organoids made from LDLR+ and LDLR- iPSC cells that were dissociated and seeded as 2D monolayer, b, Level of CCHFV (lbAr10200) infection of BVO-derived vascular cells generated from wild type (WT) and LDLR KO iPSCs. Levels of infections were determined by RT-PCR at 1day post-infection (1dpi) and 3 days post-infection (3dpi) (MOI 0.1 ). P values were calculating using unpaired student’s t-tests.
- c-e CCHFV (lbAr10200) infections of wild type or Ldlr KO mice.
- n 6 fenescence mice per group (400 PFU/mouse).
- c Weight loss was assessed the day of infection (day 0) and the day of euthanasia (day 4 post-infection),
- d Numbers of CCHFV RNA copies in serum, liver and spleen of wild type and Ldlr KO mice determined the day of euthanasia (day 4 post-infection). P values were calculating using unpaired student’s t-tests comparing two groups. All graphs are show as mean values ⁇ SD.
- Figure 6 shows validation of LDLR with a CCHFV patient isolate.
- CCHFV was isolated from the serum of a Turkish patient and this clinical isolate used for all subsequent experiments shown in Figure 6.
- a-c Levels of CCHFV infections (MOI 0.01 , 24hpi) of SW13 cells treated with the indicated concentrations of a, sLDLR, b, sLRP8, and c, sVLDLR.
- d Levels of infection with clinical CCHFV in wild type and LDLR KO (clones C2 and C12) Vero cells and in wild type and LDLR KO (clones C8, C10 and C11 ) A549 cells (MOI 0.1 , 24hpi).
- Graphs show mean value ⁇ SD.
- n 3 independent experiments. P values were calculating using unpaired student’s t-tests.
- hpi hour post-infection.
- Figure 7 shows generation and validation of Ldlr knockout in A549 and Vero cells, a, Schematic of CRISPR-Cas9 editing strategy. The extracellular region of LDLR was targeted, leading to putative N-terminally truncated proteins not displayed on the cell surface for entry, b, Schematic of editing and a-LDLR sorting procedure, c, PE intensity from a-LDLR- PE staining is shown. a-LDLR-PE staining was evaluated on single cells. Event densities were smoothened and are displayed as absolute counts or as counts normalization to the mode. Numbers indicate the percentage of single cells defined as a-LDLR-PE negative, d, Non- reactive and stained Tb1-Lu cells were used as negative control.
- PE intensity from a-LDLR-PE staining is shown. Event density was smoothened by normalization to the mode. e, Bulk sorting after transfection and transient Puromycin selection of a-LDLR-PE stained A549 or Vero cells, edited or unmodified (control) via CRISPR-Cas9. Event densities were smoothened and are displayed as counts normalization to the mode, f, Flow-cytometry result from A549-wild type and edited A549 clone 10 cells. PE intensity from a-LDLR-PE staining is shown. Event density was smoothened by normalization to the mode. Numbers indicate the percentage of single cells defined as a- LDLR-PE positive for A549 clone 10 and unmodified WT cells. Event densities were smoothened and are displayed as absolute counts.
- Figure 8 shows that ligand selectivity at LDLR drives receptor internalization.
- Figure 9 shows creation and validation of NC8 cells knocked out for LDLR.
- a Gating strategy and PE intensity from a-LDLR-PE staining are shown.
- a-LDLR-PE staining was evaluated on single cells
- b Sorting results from bulk NC8 iPSC after Cas9 LDLR editing.
- PE intensity from a-LDLR-PE staining is shown for cells targeted with an LDLR guide RNA or for unmodified control cells. Event densities were smoothened and are displayed as absolute counts or as counts normalization to the mode, c, Qualitative flow-cytometry result of selected clones stained with an a-LDLR-PE antibody.
- FIG. 11 Immunofluorescence assays a, Immunofluorescence staining of CCHFV in wild-type and LDLR KO cells. P values were calculating using two tailed student t-test. **P ⁇ 0.01. b, Immunofluorescence staining of CCHFV in SW13 cells infected with CCHFV mock or sLDLR treated.
- CCHFV produced on Hyalomma tick cells (a) and CCHFV from human patient serum (b) were tested for blocking with sLDLR or sLRP8 (MOI 0.01 , 24hpi).
- Graphs show mean value ⁇ SD.
- n 3 independent experiments. P values were calculating using two tailed student t-test. **P ⁇ 0.01 ; Non significant: p>0.05.
- FIG. 14 Protective effect of LDLR, LDLR-fragments, and multimers against CCHF-VLP infection CCHF-VLP infection assays in Huh-7 cells. Luminescence was normalized to infection control. VLPs were incubated with 10nM purified LDLR-fragments or commercially available rhLDLR (RnD System; Cat.- No. 2148-HP) or rhLDLR-mFc (SinoBiological; Cat.-No. 10231-H05H). Graph shows mean value + SEM; n > 3 independent experiments; p- values were calculated using two tailed student t-test: * p ⁇ 0.05, ** p ⁇ 0.01 , *** p ⁇ 0.001.
- sLDLR-sLRP8 expressed and purified from E.coli concentration ⁇ 5nM: sLDLR-sLRP8 (2.4nM), sLDLR-sVLDLR (1.7nM), sLRP8-sLDLR (4.4nM), sVLDLR-sLDLR (1.4nM).
- s soluble (receptor ectodomain without the transmembrane anchor)
- HEK293 ATCC®, CRL-1573
- HEK293T/17 HEK293T, ATCC® CRL-11268TM
- A549 ATCC® CCL-185
- Vero cells ATCC® CCL-81
- All cell lines were maintained in Dulbecco’s Modified Eagle’s Medium (DMEM, Life Technologies, Italy), supplemented with 10 % v/v of heat-inactivated Fetal Bovine Serum (FBS, Life Technologies) and incubated at 37° C with 95 % humidity and 5% CO2.
- DMEM Modified Eagle’s Medium
- FBS heat-inactivated Fetal Bovine Serum
- SW13 epidermal cells from adrenal gland, ATCC® CCL-105 cells were maintained in Leibovitz’s L15 medium (Thermofisher) at 37° C without CO2.
- Haploid mouse Stem-Cells mSCs, clone AN3-12 were maintained in standard embryonic stem-cell medium, supplemented with 10% (v/v) fetal bovine serum (Hyclone), recombinant mouse Leukemia Inhibitory Factor (LIF) and [3-mercaptoethanol at 37°C with 95 % humidity and 5% CO2. All cell lines were regularly tested for mycoplasma contamination.
- VSV-CCHF_G was produced as described below.
- CCHFV lbAr10200 strain was cultured on SW13 cells.
- CCHFV clinical strain was isolated on SW13 cells from a Turkish patient serum sampled as a part of another project. Ethical clearance has been obtained (Nr: 2017/1712-31/2) as well as fully informed patient consents.
- D-PBS, DMEM, Trypsin, PBS, penicillin/streptomycin, fetal bovine serum were from Gibco (ThermoFisher Scientific, Waltham, MA, USA).
- Polyethylenimine (PEI) was purchased from Alfa Aesar (ThermoFisher Scientific, Waltham, MA, USA).
- Unlabeled low-density lipoprotein (LDL) from human plasma and BOPIDY FL complexed LDL were purchased from ThermoFisher Scientific (Waltham, MA, USA).
- CCHFV Gc Human Fc-tagged CCHFV Gc, 6xHis-tagged CCHFV Gn and BODIPY FL complexed CCHFV Gc were purchased from Native Antigen (Kidlington, UK). Coelenterazine h was purchased from Nanolight Technologies (Pinetop, AZ, USA). NanoBRET Nano-Gio substrate was purchased from Promega (Madison, Wl, USA). Trizol was purchased from ThermoFisher (ThermoFisher Scientific, Waltham, MA, USA). Anti-IFN type I receptor antibody (MAR1 -5A3) was purchased from Leinco (MAR1 -5A3 [5A3]; Leinco Technologies, Inc.).
- Soluble LDLR, VLDLR and LRP8 were purchased from R&D systems.
- This soluble LDLR (Catalog #: 2148-LD R&D systems) corresponds to human LDLR protein (Uniprot P01130) Ala22-Arg788;
- soluble VLDLR (Catalog #: 8444-VL-025 R&D systems) corresponds to human VLDLR protein (UniProt: P98155.1) Thr25- Ser797;
- soluble LRP8 Catalog #: 3520-AR-050, R&D systems) corresponds to human LRP8 protein (UniProt: Q14114.4) Asp35-Lys818 (Ala262Val).
- the plasmid pC-G (Shtanko et al, 2014, Crimean-Congo hemorrhagic fever virus entry into host cells occurs through the multivesicular body and requires ESCRT regulators.
- VSV-G Vesicular Stomatitis Virus glycoprotein
- pVSV-G The plasmid expressing the Vesicular Stomatitis Virus (VSV) glycoprotein (pVSV-G) was previously described (Salata et al., 2009, vOX2 glycoprotein of human herpesvirus 8 modulates human primary macrophages activity. J Cell Physiol. 2009 Jun;219(3):698-706).
- VSVAG-GFP The recombinant VSV encoding the GFP in place of the VSV-G gene
- CCHFV-Gn/Gc pseudotyped VSVAG-GFP (CCHFV-pseudotyped virus) was generated as previously described.
- HEK293T cells were seeded in T75 flask and 24 h later transfected by calcium-phosphate protocol with 20 pg of pC-G plasmid and, 24 h later, infected with the recombinant VSVAG-GFP virus at the multiplicity of infection (MOI) of 4 fluorescent focus-forming units (FFU)/cell.
- MOI multiplicity of infection
- FFU fluorescent focus-forming units
- virus particles were pelleted by ultracentrifugation on a 20% p/v a sucrose cushion (27,000 rpm for 150 min at 4° C) in a Beckmann (Beckman Coulter Italia, Italy) SW 28 Ti swingingbucket rotor. Pellets were resuspended in 1 mL of ice-cold PBS1X/tube and mixed. Subsequently, the virus was aliquoted and stored at -80° C until use. Virus titer was determined by immunofluorescence on Vero cells seeded on 96-well plates. Viral stock was 10-fold serially diluted in DMEM and inoculated on confluent Vero cells for 1 h at 37° C.
- VSV-M [clone 23H12], Kerafast) antibody Alexa Fluor 488-conjugated goat anti-mouse IgG secondary antibodies (ThermoFisher). The fluorescent foci were counted and viral titer was expressed as FFU/mL.
- Chemical mutagenesis of haploid stem-cells Chemical mutagenesis using Ethylnitrosurea (ENU) was performed treating haploid AN3-12 cells for 2h with 0.1 mg/ml ENU in full medium while in suspension and under constant agitation. Cells were washed 5 times and transferred to a culture dish. Cells were left to recover for 48h, singled using Trypsin/EDTA and frozen in 10% DMSO, 40% FBS and 50% full medium. ENU libraries as well as untreated control libraries were used in screening experiments with VSV-CCHFV.
- ENU Ethylnitrosurea
- Haploid cells screens and analysis 50 million haploid mSCs were thawn and infected with VSV-CCHF_G at a MOI of 10 in 5 ml of ES medium without FBS. One hour after infection, the cells were supplemented with complete ES medium and incubated at 37°C with 5% CO2. After outgrowth of virus resistant cells, cell clones were picked separately and cultured before being validated by infection assay with CCHFV lbAr10200. Briefly, cells (AN3-12 wild-type and potentially resistant clones) were seeded at 5x10 4 cells per well in DMEM 5% FBS for 24h.
- A549 (ATCC CCL-185) and Vero (CCL-81) cells were grown in complete DMEM medium (DMEM High Glucose supplemented with 10% Fetal bovine serum (Gibco), 1x MEM-NEAA (Gibco), 1x Glutamax (Gibco), 1 mM Sodium Pyruvate (Gibco), 100 ll/rnl Penicillin-Streptomycin (Gibco)).
- the day before transfection 1 .05x10 5 cells were seeded per well of a 24- well plate in 0.5 complete DMEM medium.
- culture medium was replaced with fresh complete DMEM medium and transfected with a liposome:DNA mixture composed of 50pl Opti-MEM I (Gibco), 500ng of PX459 v2.0 plasmid coding (Addgene Plasmid #62988, Puro resistant), 1.5 pl Lipofectamine 3000 reagent and 1.0 pl P3000 reagent.
- Opti-MEM I Gibco
- PX459 v2.0 plasmid coding Additional Plasmid #62988, Puro resistant
- 1.5 pl Lipofectamine 3000 reagent 1.5 pl Lipofectamine 3000 reagent
- Several sgRNA were derived from CRISPick (https://portals.broadinstitute.org/gppx/crispick/public) using SpCas9 Cas9 knockout and the human LDLR gene as input.
- the final guide LDLR RNA sequence used for knockout studies was gATGAACAGGATCCACCACGA (SEQ ID NO 1 ), where lower latter g denotes preceding Guanosine to enhance transcription from the U6 Promoter.
- medium was replaced with complete DMEM supplemented with 1 pg/ml Puromycin for transient selection. 60 hours post transfection, each well containing selected A549, or Vero cells were expanded to one well of a 6-well plate in complete DMEM medium. Once cell reached 80% confluency, they were dissociated with 500 l with TrypLE Express enzyme solution (Gibco) for 5 minutes and collected in FACS Buffer (D-PBS containing 5% FBS).
- LDLR-PE Antibody R&D Systems FAB2148P
- 10 pl of a-LDLR-PE Antibody per 1.0 x 10 6 cells were added and stained for 1 h on ice in the dark. Unmodified cells were used as controls. After one hour of staining, cells were collected by centrifugation and washed twice in FACS Buffer. Finally, cells were resuspended in 1 ml of FACS Buffer and LDLR-negative cells were sorted into individual wells of a 96-well plate. LDLR-negative cells were defined as single cells displaying no PE fluorescence.
- Soluble LDLR, VLDLR and LRP8 assays SW13 were seeded at a density of 5.0 x 10 4 cells per well in a 48-well plate. 24h post-seeding, cells were counted to define the quantity of virus needed for an infection at a MOI of 0.01 . The virus was then mixed in 1.5 ml tubes (Sarstedt) with the appropriate quantity of sLDLR, sVLDR or sLRP8 in L15 medium containing 0.5% FBS. The tubes were the incubated for 30 min under shaking (75rpm) at 37°C.
- Plasmid DNA constructs for BRET assay To generate LDLR-Rlucll, codon- optimized LDLR was synthesized as a gBIock (Integrated DNA Technologies) and subcloned by Gibson assembly in pcDNA3.1/Hygro(+) GFP 10 -F?lucll db v.2 that had been linearized by PCR to exclude GFP 10 .
- To generate Nluc-LDLR codon- optimized LDLR from LDLR-Rlucll was amplified by PCR and subcloned by Gibson assembly in pcDNA3.1 NIuc-synFZDs that had been linearized by PCR to exclude FZDs.
- rGFP-FYVE (Namkung et al., 2016. Monitoring G protein-coupled receptor and (3-arrestin trafficking in live cells using enhanced bystander BRET. Nat Commun. 11 ;7:12178) has been described previously. All plasmid constructs were verified by Sanger sequencing.
- HEK293 cells were propagated in plastic flasks and grown at 37°C in 5% CO2 and 90% humidity. Cells (350,000 in 1 ml) were transfected in suspension with 1.0 pg of plasmid DNA complexed with linear polyethyleneimine (PEI; MW 25,000, 3:1 PEI:DNA ratio). All cell lines were regularly tested for mycoplasma contamination.
- PEI linear polyethyleneimine
- BRET assays Receptor trafficking-.
- HEK293 cells were transfected with LDLR-Rlucll and rGFP-FYVE and seeded in 6-well plates (7.0 x 10 5 cells/well). After a 48-hour incubation, cells were washed once with HBSS, detached and resuspended in HBSS containing 0.1 % BSA before distribution into white 96-well plates containing serial dilutions of LDL, CCHFV Gc or CCHFV Gn and returned to the incubator for 45 min at 37°C. Prior to BRET measurements, cells were incubated with coelenterazine h (10 min).
- NanoBRET binding assay To monitor the binding of fluorescent ligands to LDLR, HEK293 cells were transfected with Nluc-LDLR and seeded in white 96-well plates (3.5 x 10 4 cells/well). After a 48-hour incubation, cells were washed once with HBSS and maintained in the same buffer. Prior to BRET measurements, cells were incubated with NanoBRET Nano-Gio substrate (6 min) and then stimulated with either BODIPY FL LDL or BODIPY FL Gc for 90 min following a baseline measurement of 3 cycles.
- BRET measurements Plates were read on a Tecan Spark multimode microplate reader (Mannedorf, Switzerland) equipped with a double monochromator system to measure the emission of the Rlucll/rGFP donor-acceptor pair in receptor trafficking experiments [430-485 nm (donor) and 505-590 nm (acceptor)] or the Nluc/BODIPY FL donor-acceptor pair in the NanoBRET binding assay [445-470 nm (donor) and 520-575 nm (acceptor)].
- NC8 iPSC male, pericyte derived
- Matrigel hESC qualified, Coming
- Stemflex medium Gibco + 1 :100 Antibiotic-Antimycotic (Gibco) (Invivogen).
- Cells were passaged using 0.5mM EDTA at a ratio of 1 :6 every 3 to 4 days.
- iPSCs were dissociated into single cells using TrypLE select (Gibco) and seeded at 5.0 x 10 4 cells per 1-well of a rhLaminin521 (Gibco) coated 24-well plate in complete Stemflex medium supplemented with 1 :100 RevitaCell (Gibco).
- Blood-vessel organoids from NC8 clone 10 (LDLR+) and clone 4 (LDLR-) were produced as previously described (Wimmer et al, Generation of blood vessel organoids from human pluripotent stem cells. Nat Protoc. 2019; 14(11 ):3082-3100).
- the cells were collected through centrifugation (300xg, 5 min) and replated in PureCol (Advanced BioMatrix, 30pg/ml in PBS for 1 h at RT) coated T-25 flasks at 30840 cells/cm 2 in Sprouting media.
- PureCol Advanced BioMatrix, 30pg/ml in PBS for 1 h at RT
- mice In vivo experiments. In the current study were used six female C57BL/6J mice (Charles River, Germany) and six female B6.129S7Ldlrtm1 Her/J (LdLr KO) (stock#002207), (Jackson Laboratory, USA). All mice were 10 weeks old at the time of infection. The animals were housed according to Karolinska Institute ethical rules and observed daily.
- mice were challenged with 400 focus forming units (FFU) of CCHFV lbAr10200 in 100 pl via i.p injection.
- FFU focus forming units
- the mice were monitored daily with respect to clinical signs of disease, and their overall well-being.
- FFU focus forming units
- mice were euthanized independent on clinical signs.
- Blood was collected in microcontainer tubes for serum separation and serum was inactivated with Trizol for subsequent qRT-PCR analysis.
- liver, spleen and kidney were collected, a part in Trizol for qRT-PCR and a part were fixed in PFA 4% for histopathological analyzes.
- the experimenters were not blinded to the identity of the animals. However, the pathologist who analyzed livers as well as the scientist who runned the RT-qPCRs and the subsequent analysis were blinded.
- RNase P RNA was used as an endogenous control for normalization (Fwd: AGATTTGGACCTGCGAGCG (SEQ ID NO 12), Rev: GAGCGGCTGTCTCCACAAGT (SEQ ID NO 13), Probe: FAM- TTCTGACCTGAAGGCTCTGCGCG-MGB (SEQ ID NO 14)).
- CCHFV RNA Absolute quantification of CCHFV RNA for mice samples was performed by RT- qPCR.
- the standard synthetic RNA was solubilised in RNase-free water and the copy number calculated after quantification by nanodrop.
- SW13 were seeded at a density of 5.0 x 10 4 cells per well in a 48-well plate. 24h post-seeding, cells were counted to define the quantity of virus needed for an infection at a MOI of 0.01 .
- CCHFV was then mixed in 1.5 ml tubes (Sarstedt) with different concentration of LDL (Thermofisher, #L3486) or BSA (Saveen & Werner AB, #A1391 ) in L15 medium containing 0.5% FBS. Cells were rinsed once with PBS before being infected with virus only or with the mix virus/LDL or virus/BSA for 1 hour at 37°C.
- Hyalomma anatolicum embryo-derived cell lines HAE/CTVM9 were grown L- 15/M EM medium (equal volumes of L-15 and Minimal Essential Medium with Hank’s salts supplemented with 10% TPB), both supplemented with 2 mM l-glutamine, 20% FBS, and incubated in sealed flasks at 28°C, 0% CO2.
- CCHF L and N gene were seeded into each well of 24-well plates. Once cells attached, transfection of CCHF L and N gene (strain lbAr10200) was performed using GeneJammer Transfection Reagent (Agilent; Cat.-No. 204130). 16h post transfection indicator cells were infected with an 10Oul inoculum containing CCHF- VLPs (NanoLuc) after 1h pre-incubation with a protein-fragment of interest where applicable. 24hpi the wells were washed with PBS and analyzed using Nano-Gio® Luciferase Assay System (Promega; Cat.-No. N1110) according to manufacturer’s protocol. Luminescence was quantified with a plate reader (BMG Clario Star).
- Example 1 Haploid cells screening highlighted LDLR as an essential protein for VSV-CCHFGP/AG entry
- ENU mutagenized mES cells were infected with a viral RNA replication competent vesicular stomatitis virus pseudotyped with the glycoproteins of the Crimean-Congo hemorrhagic fever virus (VSV-CCHF_G) (see Figure 1A).
- VSV-CCHF_G Crimean-Congo hemorrhagic fever virus
- Table 2 Sequencing data of the three clones highlighting LDLR.
- Example 2 Validation of LDLR in CCHFV infections
- Ldlr mutant haploid mouse embryonic cells were assessed. These Lcf/r-knockout cells and wild-type sister cells were infected with VSV-CCHF_G and CCHFV. These murine Ldlr- knockout cells displayed more than a 90% decrease in infection rates compared to the wild type cells (Fig. 2a).
- Example 3 Gc binds to LDLR and induces endocytosis
- a bioluminescence resonance energy transfer (BRET) assay was developed to assess receptor-ligand interactions.
- BRET bioluminescence resonance energy transfer
- the inventors genetically engineered and expressed an LDLR that carries an N-terminal bioluminescent probe (NanoLuc or NIuc) in HEK293 cells. Addition of fluorescent ligands then allow to measure direct interaction through BRET (Fig. 3a).
- BODIPY-FL-labelled LDL was initially used to assess receptor binding, resulting in the expected concentration-dependent ligandbinding BRET signal in cells expressing Nluc-LDLR (Fig. 3b).
- sLDLR soluble LDLR
- VSV and VSV-CCHF_G infected cells have to be assessed at early time points post-infection to avoid a second round of infection.
- the cells were harvested and the levels of infection determined by qRT-PCR.
- sLDLR blocked the infection of rVSV-CCHF (Fig. 4a) and, importantly, CCHFV (Fig. 4b) in a dose-dependent manner.
- sLDLR was also able to inhibit VSV (Fig. 4c)
- sLRP8 is a one chain LRP8 Asp35-Lys818 (Ala262Val) fragment and sVLDLR a one chain VLDLR Thr25-Ser797 fragment.
- sLDLR neither sVLDLR nor sLRP8 decoys provided protection from CCHFV infections (Fig. 4d, e), while they were active against VSV infection (Fig. 4f, g). This data indicates that soluble LDLR can prevent CCHFV infections.
- Example 5 Infection of blood-vessels organoids
- Blood vessels are key target cells for viral tropism involved in hemorrhaging.
- Human blood vessel organoids were generated using the iPSC line NC8. Ldlr in the iPSCs was deleted using CRISPR/Cas9-mediated genome editing. Knockout iPSCs for Ldlr were validated by flow-cytometry (Fig. 9a - e). For infection were generated Ldlr mutant and wild type blood vessel organoids (BVOs), containing self-organizing bona fide capillaries formed by pericytes and endothelial cells.
- BVOs wild type blood vessel organoids
- the BVOs containing mature human capillaries were disaggregated, and the pericytes and endothelial cells were further cultured as monolayers in collagen-coated flasks (Fig. 5a). These cultures were subsequently infected with CCHFV. Knockout of Ldlr, using two different mutant iPSC clones, resulted in a significant reduction in CCHFV infections, detected one and three days post-infection (Fig. 5b). These data show that LDLRs are also involved in infections of human blood vessels.
- Example 6 LDLR KO protects mice against CCHF or LDLR mutant/KO mice are protected from CCHF
- C57BL/6J wild type and Ldlr-I- mice are naturally resistant to CCHF, but blockade or knockout of IFNa receptors render these mice susceptible to the infection, as previously reported. Therefore wild type and Ldlr-I- mice were treated with 2.5mg of anti-IFNa receptor antibodies at the time of CCHFV infection (400 PFU per mouse).
- All mice were euthanized, and their serum, liver, and spleen were analyzed for viral RNA and livers were assayed for pathologies using histology.
- Ldlr-I- mice did not show any weight loss or other macroscopic signs of disease (weakness, swollen eyes, marked incoordination, piloerection, light bleeding around the marking hole in the ear) (Fig. 5c). Ldlr-I- mice also exhibited significantly reduced level of viral RNA in the serum, with two mice even showing undetectable levels of circulating virus (Fig 5d). The mutant mice also exhibited reduced virus load in liver and spleen (Fig. 5d). Histopathological analysis of livers of CCHFV-infected wild type mice revealed midzonal necrosis (Fig. 5e upper left panel), periportal coagulative necrosis (Fig.
- Example 7 LDLR is also a receptor for CCHFV isolate
- LDLR mutant Vero and A549 cells challenged with the CCHFV patient isolate showed significantly reduced infection rates as compared to their respective LDLR expressing wild type control cells (Fig. 6d). This data confirms that LDLR also acts as a receptor for patient-derived CCHFV isolates.
- Example 8 Analysis of binding of LDLR fragments, and homo- and heteromultimers thereof to viruses.
- the LDLR fragments of the LDLR ectodomain as described above, and homo- and heteromultimers thereof covalently linked with or without a peptide linker, are expressed in bacteria, insect cells, or mammalian cells. Following purification, fragments are tested by: a) cell-based assays for their ability to block virus infections, preferably Bunyavirus infections, and more preferred, CCHFV infections. To this end wildtype virus strains, pseudotyped viruses, or viruses and pseudotyped viruses expressing marker proteins such as GFP can be used for infection of relevant cell lines.
- LDLR fragments Infection rates in the presence and absence of the LDLR fragments will be assessed by quantitative analysis of viral genomes or protein, including artificially expressed marker proteins such as GFP.
- biochemical binding assays for their ability to bind virus glycoproteins, preferred bunyavirus glycoproteins, more preferred Gc and Gn protein.
- Virus envelope gylcoproteins will be expressed and purified or purchased and then immobilized on suitable plates.
- LDLR fragments, and homo- and heteromultimers thereof will be extended by a peptide Tag such as a polyhistidine tag (6xH), a FLAG tag (DYKDDDDK), or a human influenza hemagglutinin (HA) tag (YPYDVPDYA), expressed and purified. Purified fragments and multimers thereof will be incubated with the immobilized glycoproteins. After several wash steps, high affinity binding peptides will be identified via antibody-based detection methods.
- VLPs transcription competent virus-like-particles
- the CCHFV L and N genes were co-transfected into the indicator cells 16h prior to infection.
- Pre-incubation of VLPs with full length recombinant human LDLR rhLDLR, RnD Systems; Cat.-No.
- LDLR purified soluble LDLR expressed from Hek293 cells
- CTR VLPs only
- fragments of LDLR containing only the ligand-binding domains LA1-7 or LA4-7 show protective effects against infection of Huh-7 cells with CCHF-VLPs. Multimerization of LDLR fragments containing a linker (I) between repetitive domains such as LDLR LA1-7I1-7 and LDLR LA2-3I2-3I2-3 also ameliorate infection, while a single LDLR LA2-3 fragment does not reach significance.
- Combinations of sLDLR fused to other LDLR family members (LRP8 and VLDLR) increases its potency to protect against CCHF-VLP infection (note that concentrations ⁇ 5nM were applied here).
- the closely related LDLR family member VLDLR alone shows significant protection against infection with CCHF-VLPs (Fig. 14).
- LDLR Nucleotide Sequence: LDLR (2-3) (SEQ ID NO 199) ATGGGACCATGGGGATGGAAGCTGCGCTGGACTGTGGCCCTTCTGCTTGCGGCGGCAGGAA CcgcaGTGGGTGACCGGGGCAGCGGCGTGAGCGGCTGGCGGCTGTTCAAGAAGATTAGCGG CGGAGTAACCTGCAAATCGGGAGACTTCAGCTGTGGTGGACGGGTTAACCGCTGTATACCTC AGTTTTGGCGGTGTGACGGACAAGTTGACTGTGATAATGGCTCAGATGAGCAGGGGTGCCCG CCAAAGACCTGCAGTCAGGACGAATTCCGATGCCACGATGGAAAATGTATTAGCCGTCAATTC GTTTGTGACTCGGACCGCGACTGTCTGGATGGTTCAGATGAAGCTTCGTGCCCAGTAgGAGA CGCTTCGgattacaaagacgatgacgataagCTGGAAGTTCTGTTCCAGGGGCCCGGTCATCACCACCA TCATCACCA
- LDLR (2-3) (SEQ ID NO 200) MGPWGWKLRWTVALLLAAAGTAVGDRGSGVSGWRLFKKISGGVTCKSGDFSCGGRVNRCIPQF
- sLDLR (SEQ ID NO 202) MGPWGWKLRWTVALLLAAAGTAVGDRGSGVSGWRLFKKISGGAVGDRCERNEFQCQDGKCISY KWVCDGSAECQDGSDESQETCLSVTCKSGDFSCGGRVNRCIPQFWRCDGQVDCDNGSDEQGC PPKTCSQDEFRCHDGKCISRQFVCDSDRDCLDGSDEASCPVLTCGPASFQCNSSTCIPQLWACD NDPDCEDGSDEWPQRCRGLYVFQGDSSPCSAFEFHCLSGECIHSSWRCDGGPDCKDKSDEEN CAVATCRPDEFQCSDGNCIHGSRQCDREYDCKDMSDEVGCVNVTLCEGPNKFKCHSGECITLDK VCNMARDCRDWSDEPIKECGTNECLDNNGGCSHVCNDLKIGYECLCPDGFQLVAQRRCEDIDEC QDPDTCSQLCVNLEGGYKCQCEEGFQLDPHTKACKAVGSIAYLF
- VLDLR (SEQ ID NO 209)
- VLDLR (SEQ ID NO 210)
- sLDLR-sVLDLR (SEQ ID NO 218) MGPWGWKLRWTVALLLAAAGTAVGDRGSGVSGWRLFKKISGGSGAVGDRCERNEFQCQDGKC ISYKWVCDGSAECQDGSDESQETCLSVTCKSGDFSCGGRVNRCIPQFWRCDGQVDCDNGSDE QGCPPKTCSQDEFRCHDGKCISRQFVCDSDRDCLDGSDEASCPVLTCGPASFQCNSSTCIPQLW ACDNDPDCEDGSDEWPQRCRGLYVFQGDSSPCSAFEFHCLSGECIHSSWRCDGGPDCKDKSD EENCAVATCRPDEFQCSDGNCIHGSRQCDREYDCKDMSDEVGCVNVTLCEGPNKFKCHSGECIT LDKVCNMARDCRDWSDEPIKECGTNECLDNNGGCSHVCNDLKIGYECLCPDGFQLVAQRRCEDI
- sLRP8-sLDLR (SEQ ID NO 220) MGPWGWKLRWTVALLLAAAGTAVGDRGSGVSGWRLFKKISGGSGHLAAAAADPLLGGQGPAKD CEKDQFQCRNERCIPSVWRCDEDDDCLDHSDEDDCPKKTCADSDFTCDNGHCIHERWKCDGEE ECPDGSDESEATCTKQVCPAEKLSCGPTSHKCVPASWRCDGEKDCEGGADEAGCATSLGTCRG DEFQCGDGTCVLAIKHCNQEQDCPDGSDEAGCLQVPPTFLGNRRRPRGLNECLHNNGGCSHICT DLKIGFECTCPAGFQLLDQKTCGDIDECKDPDACSQICVNYKGYFKCECYPGYEMDLLTKNCKAA AGKSPSLIFTNRHEVRRIDLVKRNYSRLIPMLKNVVALDVEVATNRIYWCDLSYRKIYSAYMDKASD PKEQEVLIDEQLHSPEGLAVDWVHKHI
- HiBiT-Tag VSGWRLFKKIS (SEQ ID NO 224) s: soluble (receptor ectodomain without the transmembrane anchor) *: stop codon encoded by the last triplet of the nucleotide sequence.
- Example 9 A phage-display approach to identify high affinity binding LDLR fragments and variants thereof to viral glycoproteins in a high-throughput format (Fig. 10).
- a viral glycoprotein of interest (preferred from Bunyaviridae, more preferred from CCHFV) is expressed, purified, and immobilized on a surface.
- a DNA pool encoding LDLR family fragments, and single and multiple amino acid variants thereof is cloned into a phagemid, i.e. an expression vector that expresses the cloned peptides as part of a bacteriophage envelope protein.
- the phagemids are transfected into bacteria cells to produces phages, wherein each phage displays a different peptide or set or peptides on its surface, wherein each peptide is a different LDLR fragment or an amino acid variant thereof.
- Display phages are incubated on the immobilized glycoprotein and will attach in dependence of the interaction strength between glycoprotein and LDLR fragment on phages.
- Example 10 Immunofluorescence stains confirm requirement of LDLR for CCHFV infections.
- Example 11 LDL outcompetes CCHFV for cell entry via the LDLR.
- CCHFV glycoproteins can bind to LDLR.
- a competition assay using CCHFV and LDL, the natural ligand for LDLR was conducted.
- LDL effectively competed with CCHFV infection in a dose-dependent manner for cell entry via LDLR.
- Bovine Serum Albumin (BSA) was used as a control and showed no impact on CCHFV infection.
- BSA Bovine Serum Albumin
- CCHFV lbAr10200 was initially cultured on a Hyalomma tick cell line, the natural vector, and the potential inhibitory effects of sLDLR and sLPR8 were evaluated.
- sLDLR demonstrated the ability to block the virus, while sLRP8 did not (Fig. 13a).
- a patient isolate that was cultivated on tick cells (see Example 7).
- the capacity of LDLR and LRP8 to block CCHFV was examined using virus present in patient serum without prior propagation on tick or human cells. As illustrated in Fig. 13b, both sLDLR and sLRP8 were effective in blocking the virus from this direct patient serum.
- LDLR LDLR family members like LRP8 can also interfere with virus spreading, potentially due to an altered protein decoration of the virus as compared to virus propagated in vitro.
- Example 13 Quality control of Bunyavirus vaccines.
- LDLR is used to assess Bunyavirus vaccine integrity or affinity of antigens for diagnostic purposes and especially to assess CCHFV vaccine integrity or affinity of antigens for diagnostic purposes.
- LDLR or LDLR fusion proteins or LDLR variants can be used to assess a bunyavirus vaccine, preferably a CCHFV vaccine, quality or the integrity of a diagnostic antigen, e.g. from patient samples.
- LDLR is a receptor that directly interacts with multiple bunyaviruses, such as CCHFV, it can be used to physically assess the quality and integrity of a bunyavirus vaccine, which is important to define lot-to-lot variation in commercial production. Further, LDLR can be used to detect diagnostic antigens, e.g. in a lateral flow immunoassay.
- Bunyavirus vaccine or diagnostic antigens can include, but are not limited to, live viruses from patient samples, live-attenuated viruses, virus-like particles, viral structural proteins, viral glycoproteins, or nucleic acids or vectors producing these viral proteins or particles.
- the format of the integrity assay can include immobilizing LDLR or LDLR fusion proteins (e.g. LDLR-Fc), or LDLR variants on a surface (e.g., microtiter well plate, chip, or slide) followed by incubation with the bunyavirus vaccine or diagnostic antigen and detection with a monoclonal or polyclonal anti-vaccine I anti- diagnostic antigen antibody.
- detection can be achieved via LDLR-Fc itself.
- the bunyavirus vaccine or diagnostic antigen can be captured on the surface (directly or indirectly) and then incubated with LDLR or an LDLR fusion protein or an LDLR variant for binding analysis.
- the assay could be performed by detection methods suitable to assess binding including, but not limited to ELISA, biolayer interferometry and surface plasmon resonance.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Cell Biology (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Medicinal Chemistry (AREA)
- Epidemiology (AREA)
- Biochemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Toxicology (AREA)
- Pharmacology & Pharmacy (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Molecular Biology (AREA)
- Engineering & Computer Science (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
La présente invention concerne un polypeptide dérivé de LDLR qui se lie à un virus Bunyavirales et est ainsi utile dans la prophylaxie et/ou le traitement d'une infection par un virus Bunyavirales chez un sujet, de préférence une infection par CCHV. L'invention concerne des polypeptides dérivés du LDLR comprenant un ectodomaine de LDLR ou un fragment fonctionnel d'un ectodomaine de LDLR ou un variant d'acide aminé d'un ectodomaine de LDLR, qui se lient à un virus Bunyavirales. L'invention concerne également des polypeptides dérivés du LDLR comprenant un multimère d'un ectodomaine de LDLR (SEQ ID NO 18), ou d'un fragment fonctionnel dudit ectodomaine de LDLR, ou d'un variant d'acide aminé dudit ectodomaine de LDLR, ou d'un variant d'acide aminé dudit fragment fonctionnel dudit ectodomaine de LDLR, ledit polypeptide dérivé du LDLR comprenant optionnellement un lieur peptidique reliant lesdits monomères. Les polypeptides de LDLR sont particulièrement utiles contre les infections par Bunyavirales. En outre, les polypeptides de LDLR peuvent être utilisés pour la détection diagnostique d'une infection par un virus Bunyavirales, en particulier une infection par le CCHFV, et le contrôle de la qualité de vaccins apparentés. L'invention divulgue également un procédé d'identification de nouveaux facteurs géniques impliqués dans une infection par le CCHFV.
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP23174811.2 | 2023-05-23 | ||
| EP23174811.2A EP4467563A1 (fr) | 2023-05-23 | 2023-05-23 | Polypeptides derives de ldlr pour utilisations antivirales |
| EP24163086 | 2024-03-12 | ||
| EP24163086.2 | 2024-03-12 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024240848A1 true WO2024240848A1 (fr) | 2024-11-28 |
Family
ID=91302640
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2024/064152 Pending WO2024240848A1 (fr) | 2023-05-23 | 2024-05-22 | Polypeptides dérivés du ldlr pour des utilisations antivirales |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024240848A1 (fr) |
Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP0553667A1 (fr) * | 1992-01-19 | 1993-08-04 | Yeda Research And Development Company Limited | Récepteur à LDL-soluble, sa production et son utilisation |
| WO2009055783A2 (fr) * | 2007-10-26 | 2009-04-30 | Schering Corporation | Anti-pcsk9 et méthodes de traitement de troubles du métabolisme lipidique et du cholestérol |
| WO2010077854A1 (fr) * | 2008-12-15 | 2010-07-08 | Regeneron Pharamaceuticals, Inc. | Anticorps humains à grande affinité contre pcsk9 |
| WO2019038332A1 (fr) * | 2017-08-22 | 2019-02-28 | Curevac Ag | Vaccin contre les bunyavirus |
| WO2021168270A1 (fr) * | 2020-02-19 | 2021-08-26 | Isolere Bio, Inc. | Matrice de purification à base de protéines et leurs méthodes d'utilisation |
-
2024
- 2024-05-22 WO PCT/EP2024/064152 patent/WO2024240848A1/fr active Pending
Patent Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP0553667A1 (fr) * | 1992-01-19 | 1993-08-04 | Yeda Research And Development Company Limited | Récepteur à LDL-soluble, sa production et son utilisation |
| WO2009055783A2 (fr) * | 2007-10-26 | 2009-04-30 | Schering Corporation | Anti-pcsk9 et méthodes de traitement de troubles du métabolisme lipidique et du cholestérol |
| WO2010077854A1 (fr) * | 2008-12-15 | 2010-07-08 | Regeneron Pharamaceuticals, Inc. | Anticorps humains à grande affinité contre pcsk9 |
| WO2019038332A1 (fr) * | 2017-08-22 | 2019-02-28 | Curevac Ag | Vaccin contre les bunyavirus |
| WO2021168270A1 (fr) * | 2020-02-19 | 2021-08-26 | Isolere Bio, Inc. | Matrice de purification à base de protéines et leurs méthodes d'utilisation |
Non-Patent Citations (9)
| Title |
|---|
| ELLING ET AL., NAT PROTOC., vol. 14, no. 7, 1 July 2019 (2019-07-01), pages 1991 - 2014 |
| HULSWIT, RJC ET AL., VIRUSES, vol. 13, no. 2, February 2021 (2021-02-01), pages 353 |
| NAMKUNG ET AL.: "Monitoring G protein-coupled receptor and β-arrestin trafficking in live cells using enhanced bystander BRET", NAT COMMUN., vol. 11, no. 7, 2016, pages 12178 |
| NAMKUNG, Y. ET AL.: "Monitoring G protein-coupled receptor and β-arrestin trafficking in live cells using enhanced bystander BRET", NAT COMMUN, vol. 7, 2016, pages 12178, XP055932276, DOI: 10.1038/ncomms12178 |
| SAGI I ET AL.: "Identification and propagation of haploid human pluripotent stem cells", NAT PROTOC., vol. 11, no. 11, 2016, pages 2274 - 2286, XP009507436, DOI: 10.1038/nprot.2016.145 |
| SALATA ET AL.: "2009, vOX2 glycoprotein of human herpesvirus 8 modulates human primary macrophages activity", J CELL PHYSIOL., vol. 219, no. 3, June 2009 (2009-06-01), pages 698 - 706 |
| SHTANKO ET AL.: "Crimean-Congo hemorrhagic fever virus entry into host cells occurs through the multivesicular body and requires ESCRT regulators", PLOS PATHOG, vol. 10, no. 9, 18 September 2014 (2014-09-18), pages e1004390 |
| WHITT, J VIROL METHODS, vol. 169, no. 2, November 2010 (2010-11-01), pages 365 - 374 |
| WIMMER ET AL.: "Generation of blood vessel organoids from human pluripotent stem cells", NAT PROTOC, vol. 14, no. 11, 2019, pages 3082 - 3100, XP036917363, DOI: 10.1038/s41596-019-0213-z |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP7754912B2 (ja) | インフルエンザウイルスにおける付加的遺伝子に遺伝的安定性を付与する突然変異 | |
| US12091434B2 (en) | Mutated glycoprotein of vesicular stomatitis virus | |
| Kuhn | Filoviruses: a compendium of 40 years of epidemiological, clinical, and laboratory studies | |
| ES3001108T3 (es) | Moléculas quiméricas y usos de las mismas | |
| CN109477074B (zh) | 用于疫苗开发的改进的乙型流感病毒复制 | |
| Zanker et al. | Influenza A virus infection induces viral and cellular defective ribosomal products encoded by alternative reading frames | |
| Yuan et al. | ADAM17 is an essential attachment factor for classical swine fever virus | |
| Monteil et al. | Crimean–Congo haemorrhagic fever virus uses LDLR to bind and enter host cells | |
| Jiang et al. | The V617I substitution in avian coronavirus IBV spike protein plays a crucial role in adaptation to primary chicken kidney cells | |
| Moreno et al. | A novel circulating tamiami mammarenavirus shows potential for zoonotic spillover | |
| EP2133358A1 (fr) | Particules de type virus de fièvre de la vallée du rift et leur utilisation pour l'immunisation et un système d'essai | |
| Ma et al. | The Single Amino Acid Change of R516K Enables Efficient Generation of Vesicular Stomatitis Virus‐Based Crimean–Congo Hemorrhagic Fever Reporter Virus | |
| Michaud et al. | The SARS-CoV-2 Spike mutation D614G increases entry fitness across a range of ACE2 levels, directly outcompetes the wild type, and is preferentially incorporated into trimers | |
| EP4467563A1 (fr) | Polypeptides derives de ldlr pour utilisations antivirales | |
| US20220088180A1 (en) | Immunogenic compositions and use thereof | |
| WO2024240848A1 (fr) | Polypeptides dérivés du ldlr pour des utilisations antivirales | |
| CN108601794A (zh) | 调节细胞因子产生 | |
| US20230357325A1 (en) | Composition and method to stabilize coronavirus spike glycoproteins in pre-fusion conformation | |
| JP2025524620A (ja) | 暗号化rna及びその使用方法 | |
| Han et al. | Engineering a streamlined virus-like particle for programmable tissue-specific gene delivery | |
| Cadena-López et al. | Assembly of Coronaviruses and CoV-Like-Particles | |
| Zheng et al. | CircMYO9A inhibits influenza A virus replication by dampening haemagglutinin cleavage via increasing SERPINE1/PAI-1 expression | |
| Huber | The Role of Bovine Complement Regulatory Protein 46 in Infection of Bovine Cells | |
| Emeterio | Insights on the adaptation of Sudan virus in guinea pigs and its implications in viral pathogenesis | |
| Su et al. | Resurrected ancestral influenza A viruses recapitulate the avian-to-mammalian adaptation of European avian-like swine influenza virus |
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: 24729203 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2024729203 Country of ref document: EP |
|
| ENP | Entry into the national phase |
Ref document number: 2024729203 Country of ref document: EP Effective date: 20251103 |