WO2020130838A2 - Anticorps pour la prévention ou le traitement de la candidose - Google Patents

Anticorps pour la prévention ou le traitement de la candidose Download PDF

Info

Publication number
WO2020130838A2
WO2020130838A2 PCT/NL2019/050875 NL2019050875W WO2020130838A2 WO 2020130838 A2 WO2020130838 A2 WO 2020130838A2 NL 2019050875 W NL2019050875 W NL 2019050875W WO 2020130838 A2 WO2020130838 A2 WO 2020130838A2
Authority
WO
WIPO (PCT)
Prior art keywords
vhh
antibody
candidalysin
albicans
cells
Prior art date
Application number
PCT/NL2019/050875
Other languages
English (en)
Other versions
WO2020130838A3 (fr
Inventor
Cornelus Theodorus VERRIPS
Edward Dolk
Frank Martin SAUER
Bernhard HUBE
Selene MOGAVERO
Original Assignee
Qvq Holding B.V.
Leibniz Institute For Natural Product Research And Infection Biology, Hans Knoell Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Qvq Holding B.V., Leibniz Institute For Natural Product Research And Infection Biology, Hans Knoell Institute filed Critical Qvq Holding B.V.
Publication of WO2020130838A2 publication Critical patent/WO2020130838A2/fr
Publication of WO2020130838A3 publication Critical patent/WO2020130838A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/14Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from fungi, algea or lichens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • the invention relates to the field of microbial infections. More specifically, the invention relates to single heavy chain variable domain antibodies (VHH) which bind cell wall-associated proteins, or secreted proteins, such as Ecelp and peptides derived therefrom.
  • VHH single heavy chain variable domain antibodies
  • Candida spp. are fungal commensals which reside on epithelial surfaces of the skin and the whole digestive tract of mast humans. Colonisation by Candida spp. is usually harmless but in case of a person treated with antibiotics, a person with a compromised immune system, or a person who have undergone surgery, they can turn into invasive pathogens. In particular C. albicans present in the intestine are often translocated into the
  • Candida spp. frequently become invasive in immunocompromised people (estimated 400.000 cases per year), disseminate through the bloodstream and attack the whole body with mortality rates of 46 - 75% (Brown et al.,2012. Science Transl Med 4:165rvl3). Due to high prevalence and mortality rates Candida spp. are receiving increased scientific attention.
  • C. albicans is the most prevalent of the species causing invasive Candida infections worldwide (about 50% of all cases), and invasive C.
  • albicans infection is the third most common hospital-acquired infection. (Yapar, 2014. Ther Clin Risk Manag 10: 95-105).
  • Yapar, 2014. Ther Clin Risk Manag 10: 95-105 One of the main
  • C. albicans contributors to virulence of C. albicans is its ability to switch from a budding yeast form to invasive filamentous growth (Mayer et al., 2013. Virulence 4: 119-128) and the expression and secretion of Ecelp by hyphae.
  • Candidalysin is a 31 amino acid polypeptide that is released post- translational from the Ecelp precursor protein [Figure 1] by proteolytic cleavage during secretion via the Golgi apparatus (Richardson et al., 2018. mBio 9: e02178-17).
  • Candidalysin is structurally related to the main component of honeybee venom, the pore forming 26 amino acid peptide toxin melittin.
  • the 3D structures of candidalysin as well as melittin comprises two alpha-helices [Figure 2], connected by a proline at position 14, while the C- termini of candidalysin and melittin are basic (Raghuraman and
  • candidalysin was identified to be the main virulence factor during tissue infections and for translocation of C. albicans to the blood stream, it represents an attractive target for raising antibodies for diagnostic or therapeutic purposes.
  • Phage-display allows for targeted selection of antigen specific antibodies from antibody libraries and has been successfully applied to isolate detection and neutralisation antibodies for toxins off different origins (e. g. Clostridium botulinum or C. pulposus toxins and ricin (Kuhn et al., 2016. Proteomics Clin Appl 10: 922-948).
  • Candida albicans immune lama VHH libraries (the variable domains of the heavy-chains of heavy-chain only antibodies from camelids) were generated by immunisation of yeast and filamentous whole cells of C. albicans.
  • Ecelp a precursor protein from which candidalysin is produced and released after proteolytic cleavage in the Golgi apparatus, is highly expressed during filamentous growth (Birse et al., 1993. Inf Immunity 61: 3648-3655; Moyes et al., 2016. Nature 532: 64-68; Richardson et al., 2018. mBio 9: e02178-17).
  • the hyphal cells used for immunisation may comprise Ecepl, candidalysin and other Ecelp derived peptides.
  • VHH libraries were screened for VHH molecules that bind Ecelp and peptides derived from Ecelp, including candidalysin [see Figure 3]. As this approach worked quite well, VHH against other cell wall protein peptides and cell wall proteins were also selected in a similar set up.
  • the invention provides a single heavy chain variable domain antibody (VHH) that binds to a cell wall component and/or secreted protein, such as Ecelp, and/or a peptide derived therefrom, of Candida spp., preferably of C. albicans.
  • VHH single heavy chain variable domain antibody
  • a preferred VHH antibody according to the invention binds and neutralizes a cytolytic pore-forming peptide toxin, termed candidalysin.
  • VHH antibodies can be raised in llamas against Ecelp, candidalysin and other Ecelp derived peptides and these VHH antibodies can be selected using phage- display. Moreover, it is shown that these VHH antibodies can neutralize Ecelp derived peptides such as candidalysin and have great potency for
  • a preferred VHH antibody according to the invention comprises CDR1, CDR2 and CDR3 amino acid sequences as depicted in Figure 4A, preferably CDR1, CDR2 and CDR3 amino acid sequences from an individual VHH antibody.
  • a further preferred VHH antibody according to the invention comprises amino acid sequences as depicted in Figure 4B and/or Figure 4C.
  • a further preferred VHH antibody according to the invention binds to a non-candidalysin peptide and which antibody reduces or eliminates mammalian cell lysis that is induced by Ecelp derived peptides such as candidalysin.
  • a preferred VHH antibody that binds to a cell wall component of a Candida spp. binds a protein of the Als-, Hpw- and YflTHyr family of adhesin molecules of Candida spp. preferably C. albicans, most preferably to adhesin Als3.
  • a VHH antibody of the invention may be fused to an immunoglobulin Fc region or functional part thereof, preferably derived from IgGl, IgG2, IgG3, IgG4), more preferably derived from IgGl.
  • Said Fc region or functional part thereof preferably is human or a humanized lama Fc or functional part thereof.
  • the invention further provides a hi- or multi-specific antibody, such as a tri-specific antibody, comprising a VHH antibody according to the invention.
  • a hi- or multi-specific antibody such as a tri-specific antibody, comprising a VHH antibody according to the invention.
  • Said bi- or multi-specific antibody preferably comprises at least two VHH antibodies that are non-competing and non-interfering VHH antibodies.
  • Said bi- or multi-specific antibody preferably comprises at least a VHH that recognize a hyphae specific cell wall protein, and a VHH that recognizes ECE1 and an ECE1 derived peptide such as candidalysin.
  • a further preferred bi- or multi-specific antibody protects Galleria mellonella against death after infection with C. albicans.
  • the invention further provides a nucleic acid encoding an antibody according to the invention.
  • the invention further provides a method for producing an antibody, the method comprising expressing the nucleic acid of the invention in a relevant cell and recovering the thus produced antibody from the cell.
  • the invention further provides the antibody according to the invention for use in diagnostic applications.
  • the invention further provides the antibody according to the invention for use as a medicament.
  • the invention further provides an antibody according to the invention, for use in a method of prophylactic treatment of an immune compromised person that may become infected with C. albicans.
  • the invention further provides the antibody according to the invention for use in a method for treatment of an individual infected with a Candida spp.
  • the invention further provides a microbicide or apheresis device comprising an antibody according to the invention.
  • the invention further provides a pharmaceutical composition
  • the invention further provides a medical device, comprising a coating that prevents adhesion and biofilm formation of C. albicans hyphae, said coating preferably comprising one or more antibodies of the invention.
  • a host cell preferably a lactic acid bacterium, expressing a VHH antibody of the invention.
  • Said lactic acid bacterium can be used as a delivery system of these VHH to the gastro-intestinal tract to neutralize Ecelp, candidalysin and other factors causing translocation of C. albicans to the bloodstream, and/or a lactic acid bacterium that expresses VHH on its surface that may block adhesion of Candida spp., especially C. albicans, to the mucosal layer.
  • FIG. 1 A cartoon representation of Ecelp and the amino acid sequences of the peptides derived from Ecelp by the action of Kex2 and/or Kexl.
  • the peptides - fused to FLAG - used for the capturing of VHH during selection are indicated below the Ece lp-derived peptides.
  • FIG. 2 The 3D structure of Ecelp determined with the bio- informatics tool i-Tasser and the 3D model for candidalysin is superimposed on the 3D structure of Ecelp.
  • the Arg 81 and Arg 93 residues indicate the N- and C-termini of candidalysin, whereas the arrows indicate the a-helices of candidalysin.
  • FIG. 1 Amino acid sequences of FLAG-tagged candidalysin peptides used for the selection of anti- Ecelp and anti-candidalysin VHH.
  • a FLAG tag comprises the amino acid sequence DYKDDDDK.
  • FIG. 4 Amino acid sequences of VHH antibodies.
  • A Amino acid sequences of the CDR regions of the anti Ecelp and anti-candidalysin VHH of the FI subfamily and of the HI subfamily.
  • B Complete amino acid sequences of the FI and HI KQREL subfamilies of anti Ecelp and
  • candidalysin VHH Alternative amino acid residues are indicated in brackets after an amino acid residue.
  • C Amino acid sequences of anti-Als3 and anti- Als4 antibodies. Alternative amino acid residues are indicated in brackets after an amino acid residue.
  • C-terminal FLAG tag candidalysin with C-terminal (CLF) or N-terminal (FCL) FLAG tag and recombinant Ece lp (rEce lp) all of which had been coated to a polystyrene plate overnight.
  • Figure 7 SDS page and Western blots with recombinant Ecelp and VHH CALl-Fl and VHH CALl-Hl.
  • FIG. 1 Immunofluorescence of C. albicans during infection of epithelial cells; stained with anti-candidalysin CALl-Fl. Fluorescence is indicated by white arrows.
  • FIG. 10 Cell damage of human epithelial cells through C. albicans or candidalysin and the inhibition of this damage by VHH CALl-Fl and CALl- Hl. Shown is the damage in the presence of VHHs in relation to the damage detected in samples without VHHs. TR146 epithelial cells were exposed either to C. albicans cells (A) or to 32 mM synthetic candidalysin (B) in presence of VHHs. As a measure of cell damage the lactate-dehydrogenase release was determined. Each value represents the average and standard deviation of three technical replicates.
  • FIG. 12 Immunofluorescence microscopy of C. albicans deletion mutants as indicated in Table 1 with hyphae specific VHH CAW3A8.
  • the cell wall of C. albicans was stained with Calcofluor White Staining (Merck) and VHH CAW3A8 was detected by IgG-goat-anti-rabbit Alexa fluor 647 conjugate (far-red emission) (Thermoscientific) via IgG-rabbit-anti VHH.
  • FIG. 14 Protection of Galleria mellonella infection caused by C. albicans cells by [A] a single VHH; and [B] a mixture of VHHs.
  • LDH lactate dehydrogenase
  • Fig 16. Results of the damage inhibition assay induced with Candida albicans cells. Confluent TR146 cells were grown and yeast cells (WT cells or cells expressing Ecel) were added to damage the TR146 cells as measured by concentration of LDH (ng/ml). White bars indicate the wells where no antibody was added and dotted bars where 2,5 mM of Fl-Hl bihead was added and striped bars where 3,5 or 2,5 mM CalFl-CAW-3A8 bihead was added. Depicted is the mean of three technical replicates.
  • antibody' refers to an antigen binding protein comprising at least a heavy chain variable region (Vh) that binds to a target epitope.
  • Vh heavy chain variable region
  • the term antibody includes monoclonal antibodies comprising immunoglobulin heavy and hght chain molecules, single heavy chain variable domain antibodies, and variants and derivatives thereof, including chimeric variants of monoclonal and single heavy chain variable domain antibodies.
  • VHH refers to single heavy chain variable domain antibodies devoid of light chains.
  • a VHH is an antibody of the type that can be found in Camelidae which are naturally devoid of hght chains or a synthetic and non immunized VHH which can be constructed accordingly.
  • Lamalidae includes reference to Llamas such as, for example, Lamaglama, Lama vicugna ( Vicugna vicugna) and Lama pacos (Vicugna pacos), and to Camelus species including, for example, Camelus dromedarius and Camelus bactrianus ).
  • VHH family and sub-family
  • VHH are encoded by specific VHH V-genes, which are about 20 genes that can be divided into 4 groups by their VH genes (Deschacht et al., 2010. J Immunol 184: 5696-704) and the fusion of these V-genes to the various D- and J-genes. Whereas the D-genes can encode a very large number of sequences, there are only 7 J-genes in the genomes of lamas (Achour et al., 2008. J Immunol 181: 2001-9).
  • a family as a fusion product of one of the 3 V genes specific for VHH and the VH genes, and one of the 7 J genes.
  • a sub-family includes the variability of CDR3 mainly encoded by D-genes.
  • a family of VHH is thus defined as a particular combination of one of the 4 groups of V-genes and one of the 7 J-genes.
  • the amino acid sequence and structure of a heavy chain variable domain can be considered - without however being limited thereto - to be comprised of four framework regions or‘FR 1 , which are referred to in the art and herein as‘Framework region G or‘FR1’; as‘Framework region 2’ or’FR2’; as‘Framework region 3’ or‘FR3’; and as ‘Framework region 4’ or‘FR4’, respectively; which framework regions are interrupted by three complementary determining regions or‘CDR 1 s’, which are referred to in the art as‘Complementarity Determining Region G or ‘CDR1’; as‘Complementarity Determining Region 2' or‘CDR2’; and as ‘Complementarity Determining Region 3’ or * CDR3 ⁇ respectively.
  • CDR 1 framework regions
  • the amino acid residues of heavy chain variable regions are numbered according to the general numbering of Rabat (Rabat, et al. (1991) Sequences of Proteins of Immunological Interest, 5th edition. Public
  • amino acid residues 26-32 of VHH are defined as CDR1
  • amino acid residues 52-58 of VHH are defined as CDR2
  • amino acid residues 95-103 of VHH are defined as CDR3, with the amino acid residue numbering according to the Rabat numbering.
  • binding refers to the process of a non- covalent interaction between molecules.
  • said binding is specific.
  • the terms‘specific’ or‘specificity’ or grammatical variations thereof refer to the number of different types of antigens or their epitopes to which a particular antibody such as a VHH can bind.
  • the specificity of an antibody can be determined based on affinity.
  • a specific antibody preferably has a binding affinity Ed for its epitope of less than 10 -7 M, preferably less than 10 -8 M, most preferable less than 10 a M.
  • Ecelp refers to a 271 amino acid preproprotein which is expressed on an invasive form, termed hypha, of C. albicans.
  • the hyphae-specific preprotein is cleaved by proteases, especially Rex2 and Rexl, into 9 peptides termed SP, Ecel-I, Ecel-II, Ecel-III, Ecel-FV, Ecel-V, Ecel-VI, Ecel-VII, and Ecel-VIII, of which Ecel-III is also termed candidalysin. Only some of these non-candidalysin peptides can be detected in hyphal culture supernatants.
  • Ecelp derived peptide refers to the 9 peptides termed SP, Ecel-I, Ecel-II, Ecel-III, Ecel-FV, Ecel-V, Ecel-VI, Ecel-VII, and Ecel-VIII, that result from proteolytic cleavage of the precursor protein Ecelp.
  • Kex2/Kexl mediated hydrolysis of Ecelp is essential for the release of candidalysin and seven other peptides [see Fig 1].
  • candidalysin refers to a 31 amino acid polypeptide that is released post- translational from a Candida Ecelp precursor by proteolytic cleavage during secretion from the Golgi apparatus (Richardson et al., 2018. mBio 9: e02178- 17).
  • a primary amino acid sequence of candidalysin is provided in Figure 1.
  • epitope or antigenic determinant refers to a part of an antigen that is recognized by an antibody.
  • epitope includes linear epitopes and conformational epitopes.
  • a conformational epitope is based on 3-D surface features and shape and/or tertiary structure of the antigen.
  • neutralizing antibody refers to an antibody that, when bound to an epitope, interferes with the normal activity of the Ecelp-derived peptides.
  • a neutralizing antibody interferes with cytolytic activity of candidalysin and/or reduces the function of the non-candidalysin peptides.
  • affinity refers to the strength of a binding reaction between a binding domain of an antibody and an epitope. It is the sum of the attractive and repulsive forces operating between the binding domain and the epitope.
  • affinity refers to the dissociation constant, Kd.
  • microbicide refers to products that contain active components that block transfer of harmful living entities into the human body, in particular via the vagina and/or rectum.
  • Candida mucosal infection refers a severe mucosal infections that is associated with severe morbidity.
  • a defining feature of this pathogenesis is the transition from yeast to invasive filamentous hyphae which damage mucosal epithelia and induce activation of the activating protein- 1 (AP-1) transcription factor.
  • lactic acid bacteria refers to lactic acid bacteria that can survive for a considerable time in the gastro-intestinal tract or vagina and are able to produce VHH antibodies in that tract or the vagina that can neutralize Candida spp.
  • the lactic acid bacteria are preferably modified in such a way that, when excreted in the environment, they can’t survive.
  • products that protect implanted medical devices against Candidiases refers to sprays, gels and/or emulsions that contain VHH that block biofilm formation on these devices.
  • the invention relates to a specific class of antibodies, namely single heavy chain variable domain antibodies, or VHH antibodies, that are capable of binding to and neutralizing Ece lp-derived peptides including candidalysin and non-candidalysin peptides.
  • VHH antibodies Single heavy chain variable domain antibodies
  • These antibodies are termed“the Variable domains of the Heavy-chains of Heavy-chain only antibodies”, or abbreviated VHH.
  • the genes encoding heavy chain variable domain antibodies were isolated from lymphocytes of llamas that were immunized with hyphal Candida albicans cells that express high levels of Ecelp, the precursor protein of candidalysin and other peptides.
  • Ecelp is the precursor of candidalysin and other peptides we reasoned that in the rather complex immunogen that was used there was a reasonable probability that Ecelp, candidalysin and some of the other peptides were present. Consequently, we embarked on a strategy to select VHH antibodies from the immune libraries that recognize Ecelp,
  • Immune phage display libraries were generated from these animals at 43 days after the first injection. These libraries were used to select phages that bind to captured candidalysin. This resulted in the isolation of two families of VHH antibodies that showed binding and neutralization of candidalysin.
  • VHH antibodies that were isolated and showed binding to non- candidalysin Ecelp derived peptides, belong to 3 different families, notably the families containing V-genes characterized by the hall mark sequences KQREL, KEREG and KGLEW, as depicted in Figure 4. Only from the V- genes characterized by KEREF sequence no VHH antibodies were selected.
  • a VHH according to the invention preferably comprises CDR1, CDR2 and CDR3 amino acid sequences as depicted in Figure 4a, or a derivative, for example an exchange for an amino acid that has a related side group as described in Rajpal et al., 2005 [Rajpal et al., 2005. PNAS 102: 8466-8471].
  • a further preferred VHH antibody comprises any one of the variable domain amino acid sequences as depicted in Figure 4b and/or 4C, or a derivative, for example a conservative derivative, thereof.
  • a derivative preferably is more than 90% identical, more preferably more than 95% identical, more preferably more than 99% identical to the amino acid sequences depicted in Figure 4b and/or 4C.
  • Said derivative preferably has 10 or less conserved amino acid exchanges, such as 5 conserved amino acid exchanges, 4 conserved amino acid exchanges, 3 conserved amino acid exchanges, 2 conserved amino acid exchanges or 1 conserved amino acid exchange.
  • conserved amino acid exchange refers to an exchange for an amino acid that has a related side group as described in Rajpal et al., 2005.
  • the isolated anti-candidalysin VHH can be grouped into 2 families termed FI and HI, according to the germ line V- and J-genes of which the isolated VHH have been derived by selection and maturation (see Fig 4b).
  • the invention further provides an antibody that effectively competes with an anti-candidalysin antibody of families FI and HI for binding to its epitope on candidalysin.
  • the term effectively is used to indicate that the competing antibody binds with substantially the same affinity to the same or nearly the same epitope, when compared to the antibody of the invention.
  • the term substantially is used to indicate that the difference in affinity between an antibody of the invention and a competing antibody is preferably less than 10-fold, more preferred less than 5-fold, more preferred less than 2-fold, more preferred less than 1,5 fold.
  • a preferred competing antibody is capable of effectively competing with an antibody of the invention when the competing antibody lowers the affinity of the observed binding of an antibody of the invention to its epitope about 2-fold using the same molar amount of competing antibody.
  • Assays for measuring competition are known in the art and include, for example, competitive ELISA.
  • VHH that recognize proteins present in the cell membrane or the cell wall of C. albicans.
  • VHH that only bind to cell wall proteins that are only present in the hyphal form of C. albicans were selected.
  • One of the VHH selected in this way proofed to recognize the protein Als3, an adhesin.
  • a vaccine-like product using a modified form of Als3 give some protection against candidiasis [Schmidt et al., 2012. Vaccine 30: 7594-600; Edwards et al., 2018.
  • the HMA1 gene with a FLAG- tag-encoding sequence linked to it may be synthesized and cloned and expressed in E. coli using standard procedures.
  • FLAG- tagged Hmalp may subsequently be purified using an anti FLAG antibody column.
  • the Hmalp may then be captured on a microtiter plate coated with anti FLAG antibody as described above and phage selections may be carried out using the above indicated phage libraries as described in detail herein above.
  • a heavy chain variable domain antibody is small, does not or hardly aggregate and have a short half life when administered, for example, to humans. Therefore, VHH hardly induce an immune response after
  • de-immunization and/or humanization may be required for use of the antibodies of the invention in pharmaceutical compositions.
  • De-immunization is a preferred approach to reduce the immunogenicity of antibodies according to the invention. It involves the identification of linear T-cell epitopes in the antibody of interest, using bioinformatics, and their subsequent replacement by site-directed
  • mutagenesis to non-immunogenic sequences or, preferably human sequences.
  • Methods for de-immunization are known in the art, for example from
  • a further preferred approach to circumvent immunogenicity of antibodies according to the invention when applied to humans involves humanization.
  • Various recombinant DNA-based approaches have been estabhshed that are aimed at increasing the content of amino acid residues in antibodies that also occur at the same of similar position in human antibodies while retaining the specificity and affinity of the parental non-human antibody.
  • Most preferred are amino acid residues that occur in antibodies as they are encoded by genomic germ hne sequences.
  • Humanization may include the construction of VHH-human chimeric antibodies, in which the VHH binding regions are covalently attached, for example by amino acid bonds, to one or more human constant (C) regions.
  • Neutralization of candidalysin is measured by determining the loss of cell damage after incubation of cells with candidalysin or, preferably, with a C. albicans species, preferably with hyphae of a C. albicans species, in the presence and absence of an antibody according to the invention.
  • candidalysin and/or cells of C. albicans are mixed with the antibody under appropriate conditions and then provided to an indicator cell culture, preferably comprising epithelial cells, that is sensitive to candidalysin.
  • the loss of cell damage is brought about by interference by the bound antibody with any one of the steps leading to damage of the indicator cells, including membrane permeabilization, inward current concomitant with calcium influx, and activation of the activating protein- 1 (AP-1) transcription factor.
  • An antibody is preferably assayed for neutralization as a soluble antibody fragment.
  • Antibodies comprising an anti-Ecelp directed VHH antibody
  • the invention further provides a VHH antibody that binds and neutralizes Ecelp and Ecelp-derived peptides such as candidalysin.
  • a preferred antibody is a bispecific or multispecific antibody such as a trispecific antibody, comprising at least one neutralizing anti-candidalysin VHH antibody.
  • Said bispecific or multispecific antibody preferably comprises means for eliminating candidalysin and/or other Ecelp-derived peptides, via antibody-dependent cell-mediated cytotoxicity (ADCC) routes and/or complement dependent cytotoxicity (CDC) routes.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement dependent cytotoxicity
  • said antibody comprises an immunoglobulin Fc region or functional part thereof of an immunoglobulin heavy chain.
  • the Fc region or functional part thereof is preferably derived from IgGl, IgG2, IgG3, IgG4, IgM, IgD, IgA or IgE. It is further preferred that the Fc region or part thereof is a human Fc region or part thereof similar to VHH-Fc fusion antibodies as described for anti HIV VHH [McCoy et al., 2014. Retrovirology 11: 83].
  • Said camelid Fc region or part thereof preferably is humanized.
  • the single heavy chain variable domain is preferably connected to a Fc region or functional part thereof via a hinge region.
  • a preferred hinge region is the hinge region of a camelid or human immunoglobulin heavy chain molecules from IgGl, IgG2, IgG3, IgG4, IgM, IgD, IgA or IgE, most preferred from IgGl.
  • a hinge region of a camelid immunoglobulin heavy chain molecule preferably is humanized.
  • a preferred part of an Fc region is the region comprising the C2 domain, the C3 domain, or the C2 and C3 domains of IgGs, preferably IgGl or IgG3, most preferably C2 and C3 domains of human IgGl.
  • a further preferred antibody is a hi- or multivalent antibody comprising a neutralizing single heavy chain variable domain according to the invention.
  • Said hi- or multivalent antibody preferably is a bispecific or multispecific antibody comprising two or more different single heavy chain variable domains that recognize different epitopes on Ecelp or on an Ece lp-derived peptide such as candidalysin and/or another, non-candidalysin peptide derived from Ecelp.
  • a hi- or multispecific antibody preferably comprises two or more single heavy chain variable domains of which one binds candidalysin, and a second one binds another non-candidalysin peptide derived from Ecelp.
  • Non-limiting examples can be provided of combinations of different anti- Ecelp VHH antibodies, directed against candidalysin and/or against a non- candidalysin peptide derived from Ecelp. Any combination of these antibodies can be used in a hi- or multispecific antibody to generate functional bi-heads or even tri-heads that can be used in research, for imaging, or as therapeutics.
  • biheads formed from VHH that only recognize non-candidalysin peptides are included as they may have superior binding properties or biological functionalities.
  • biheads may be constructed that recognize both Ecelp, candidalysin or other peptides derived from Ecelp and VHH recognizing markers specific for hyphae of Candida spp.
  • a preferred bihead or multi-head such as trihead comprises an anti- candidalysin antibody and an antibody against a membrane-anchored target.
  • Said membrane-anchored target preferably is a protein such as adhesion molecule, preferably Als 3 and/or Als4.
  • said anti- membrane-anchored target antibody will localize the bihead or multi-head such as trihead on the Candida membrane, allowing it to effectively and specifically capture a candidalysin molecule after transport through the Candida membrane, before its diffusion and entering the extracellular matrix of the mammalian target cell, thereby lowering the concentration of the candidalysin molecule at the membrane of the mammalian target cell.
  • a preferred linker group is a linker polypeptide comprising from about 10 to about 50 amino acid residues, preferably from 10 to about 30 amino acid residues, most preferred about 15 amino acid residues.
  • gly-ser linkers for example of the type (Gly x Ser y ) z , such as, for example (Glyt Ser).3 ⁇ 4 or (Gly;iSer2>3 , as described in WO 99/42077, which is hereby incorporated by reference, and the GS30, GS15, GS9 and GS7 linkers described in, for example, WO 06/040153 and WO 06/122825, both of which are hereby incorporated by reference, as well as hinge-like regions, such as the hinge regions of naturally occurring heavy chain antibodies or similar sequences
  • a preferred bihead or multihead comprises at least two noncompeting and non-interfering monospecific anti- candidalysin VHH antibodies , or an anti- candidalysin VHH antibody and a second VHH antibody directed against a non-candidalysin peptide derived from Ecelp.
  • Another method to improve the binding of the selected VHH towards Ecelp, candidalysin or non-candidalysin peptides is the direct screening approach.
  • An immune library contains 10 4 to 10 5 genes encoding different VHH, low through-put selections only result in a small number of VHH.
  • High through put screening on functionality defined as reducing the lysis of mammalian cell by Ecelp, candidalysin or non-candidalysin Ecelp derived peptides, in combination with deep sequencing has been proven to be successful in the hands of the applicants [McCoy et al 2014, PLOS Path 10, e 1004552].
  • C. albicans can translocate from mucosal surfaces into the bloodstream.
  • translocation of the intestinal population of C. albicans results in candidases [Allert et al., 2018, mBio 9: e00915-18].
  • Methods to prevent translocation of C. albicans hyphae in the intestine by using specially selected - food grade - lactic acid bacteria that protect the epithelial layers of the intestine by interacting with surface proteins of epithelial cells and simultaneously secrete VHH that can neutralize the main factor causing translocation, ECE1 and its derivative candidalysin. In this way there is a more or less continuous delivery of neutralizing VHH at the place where they are mostly required, close to the epithelial cells of the intestine and vagina, will take place.
  • a preferred host cell for expression of anti- Candida VHH in general, preferably anti-C. albicans VHH, in particular anti candidalysin VHH, are lactic acid bacteria [Pant et al., 2006. J Infect Diseases 194: 1580-8; Hultberg et al., 2007. BMC Biotechn 7: 58; Marcotte et al., 2008. Future Virol 3: 327- 341; Anderson et al., 2016. Infection Immunity 84: 395-]. These bacteria can be cultivated at large scale and supplied as food ingredient to potential patients or immune compromised persons. The live lactic acid bacteria will produce the protective VHH in, for example, the intestine and/or vagina.
  • lactic acid bacteria that are common inhabitants of the vagina can be used to prevent candidiasis by exposing anti adhesin VHH on there surfaces.
  • Anti Ecelp, anti-candidalysin and/or anti Als3p VHH can also be taken up in a gel or emulsion that can be used to protect the epithelial cells of the vagina against necrosis caused by C. albicatis hyphae and products that are produced by the C. albicans hyphae.
  • said lactic acid bacteria preferably are food grade lactic acid bacteria that are frequently used in fermented food products. Said lactic acid bacteria preferably are generally recognised as safe (GRAS).
  • these transformed lactic acid bacteria are able to produce VHH that can neutrahze candidalysin, and/or VHH that block adhesion of Candida species such as C. albicans to the mucosal layer of the intestine or vagina.
  • lactic acid bacteria can be cultivated at large scale and supplied as food or food ingredient to potential patients or immune compromised persons. These still living lactic acid bacteria will produce the protective VHH in the intestine and vagina.
  • lactic acid bacteria that are common inhabitants of the vagina can be used to prevent candidiasis by exposing anti adhesin VHH on there surfaces.
  • Anti Ecelp, anti-candidalysin, and/or anti Als3p VHH can also be taken up in a gel or emulsion that can be used to protect the epithelial cells of the vagina against necrosis caused by C.
  • albicans hyphae and/or by products produced by the hyphae.
  • Said lactic acid bacteria preferably are provided as food, or food-like products or as a drink. Said lactic acid bacteria will bind to receptors of various mucosae of the gastro-intestinal tract and secrete the neutralizing VHH at the site where Candida spp. may translocate from mucosal surfaces of the gastro-intestinal tract into the bloodstream.
  • the invention provides products containing VHH that block the adhesion of C. albicans hyphae to implanted medical devices thereby preventing biofilm formation of these C. albicans hyphae and their production of candidalysin.
  • An antibody according to the invention for example a single heavy chain variable domain or an antibody comprising a single heavy chain variable domain, may be produced using antibody producing prokaryotic cells or eukaryotic cells, preferably mammalian cells such as CHO cells or HEK cells, or fungi, most preferably filamentous fungi such as Aspergillus awamori, or yeasts such as Saccharomyces cerevisiae or Pichia pastoris.
  • prokaryotic cells or eukaryotic cells preferably mammalian cells such as CHO cells or HEK cells, or fungi, most preferably filamentous fungi such as Aspergillus awamori, or yeasts such as Saccharomyces cerevisiae or Pichia pastoris.
  • An advantage of an eukaryotic production system is that folding of the protein results in proteins that are more suitable for treating a human individual.
  • eukaryotic cells often carry out desirable post translational modifications that resemble posttranslational modifications that occur in mamma
  • VHH production of VHH in filamentous fungi is preferably performed as described by Joosten et al. (2005). J Biotechnol 120:347-359, which is included herein by reference.
  • a preferred method for producing VHHs in Saccharomyces cerevisim is according to the method as described by v. d. Laar et al, (2007), Biotech Bioeng 96, 483-494; or Frenken et al. (2000). J Biotechnol 78:11-21, which are included herein by reference.
  • Another preferred method of VHH production is by expression in Pichia pastoris as described by Rahbarizadeh et al. (2006) J Mol Immunol 43:426—435, which is included herein by reference.
  • a further preferred method for production of therapeutic VHH comprises expression in mammalian cells such as a fibroblast cell, a Chinese hamster ovary cell, a mouse cell, a kidney cell, a retina cell, or a derivative of any of these cells.
  • a most preferred cell is a human cell such as, but not limited to, Hek293 and PER.C6.
  • a further preferred cell fine is a cell line in which alpha-(l,6)-fucosyltransferase has been inactivated, for example the AFUT8 CHO cell line, as described in Yamane-Ohnuki et al 2004, Biotechnol. Bioeng 87, 614-622. It was found that antibodies that are produced in AFUT8 cells enhance the ADCC route.
  • An antibody according to the invention is preferably produced by the provision of a nucleic acid encoding said antibody to a cell of interest.
  • the invention further provides a nucleic acid encoding an antibody according to the invention.
  • Said nucleic acid preferably DNA is preferably produced by recombinant technologies, including the use of polymerases, restriction enzymes, and ligases, from the single heavy chain variable domains that were isolated from the immunized animal, as is known to a skilled person.
  • said nucleic acid is provided by artificial gene synthesis, for example by synthesis of partially or completely overlapping oligonucleotides, or by a combination of organic chemistry and recombinant technologies, as is known to the skilled person.
  • Said nucleic acid is preferably codon-optimised to enhance expression of the antibody in the selected cell or cell line.
  • Further optimization preferably includes removal of cryptic splice sites, removal of cryptic polyA tails and/or removal of sequences that lead to unfavourable folding of the mRNA.
  • the presence of an intron flanked by splice sites may encourage export from the nucleus.
  • the nucleic acid preferably encodes a protein export signal for secretion of the antibody out of the cell into the periplasm of prokaryotes or into the growth medium, allowing efficient purification of the antibody.
  • the invention further provides a vector comprising a nucleic acid encoding an antibody according to the invention.
  • Said vector preferably additionally comprises means for high expression levels such as strong promoters, for example of viral origin (e.g., human cytomegalovirus) or promoters derived from genes that are highly expressed in a cell such as a mammalian cell (Running Deer and Allison (2004) Biotechnol Prog 20: 880- 889; US 5888809).
  • the vectors preferably comprise selection systems such as, for example, expression of glutamine synthetase or expression of
  • dihydrofolate reductase for amplification of the vector in a suitable recipient cell, as is known to the skilled person.
  • the invention further provides a method for producing an antibody, the method comprising expressing a nucleic acid according to the invention in a prokaryotic or eukaryotic cell and recovering the thus produced antibody from the cell cultures.
  • the nucleic acid preferably a vector comprising the nucleic acid, is preferably provided to a cell by transfection or electroporation.
  • the nucleic acid is either transiently, or, preferably, stably provided to the cell. Methods for transfection or electroporation of cells with nucleic acid are known to the skilled person.
  • a cell that expresses high amounts of the antibody may be selected. This cell is grown, for example in roller bottles, in fed-batch culture or continuous perfusion culture.
  • the invention additionally provides a host cell comprising a nucleic acid or vector according to the invention.
  • Said host cell may be grown or stored for future production of an antibody according to the invention.
  • the invention further relates to a product or composition containing or comprising at least one neutralizing anti-candidalysin antibody as described herein, and/or a VHH antibody against another, non-candidalysin peptide that is derived from Ecelp. Therefore, the invention provides an antibody according to the invention for use as a medicament.
  • the antibodies of the invention are preferably used for prophylactic administration or therapeutic administration in both humans and other animals that are infected with C. albicans.
  • the antibodies according to the invention are administered to high-risk individuals in order to lessen the likelihood of developing Candida infection related diseases such as candidiasis and invasive candidiasis, in these individuals or to lessen the severity of the disease or administered to subjects already evidencing active C. albicans infection.
  • an antibody of the invention is preferably provided in an effective amount.
  • An effective amount of an antibody of the invention is a dosage large enough to produce the desired effect in which the symptoms of the Candida infection are ameliorated or the likelihood of infection is decreased.
  • a therapeutically effective amount preferably does not cause adverse side effects, such as hyperviscosity syndrome, pulmonary edema, congestive heart failure, and the like.
  • a therapeutically effective amount varies with the subject's age, condition, and sex, as well as the extent of the disease in the subject and can be determined by one of skill in the art.
  • the dosage may be adjusted by the individual physician or veterinarian in the event of any complication.
  • a therapeutically effective amount may vary from about 0.01 mg/kg to about 500 mg/kg, preferably from about 0.1 mg/kg to about 200 mg/kg, mast preferably from about 0.2 mg/kg to about 20 mg/kg, in one or more dose administrations daily, for one or several days.
  • the antibodies of the invention can be administered by injection or by gradual infusion over time.
  • the administration of the antibodies preferably is parenteral such as, for example, intravenous, intraperitoneal, or
  • Preparations for parenteral administration include sterile aqueous or non- aqueous solutions suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like.
  • microbicide refers to a product that is used topically, preferably vaginally or rectally, to prevent infection.
  • a microbicide offers the potential for women to protect themselves and their sexual partners from Candida infections. For other anti-infective use, they may also be applied to the skin, mucous membranes, and orally.
  • Preferred microbicides are inexpensive, affordable, stable at ambient temperature, preferably at temperatures above 35 °C, more preferably at temperatures above 40 °C, compatible and active after mixture with cosmetically acceptable formulations, non-toxic and nondamaging to vulvar, vaginal, cervical, penile or other epithelium.
  • microbicide preferably further comprises a base or carrier, such as a foam, cream, wash, gel, suppository, ovule, lotion, ointment, film, tablet, foaming tablet, tampon, vaginal spray, aerosol, or other base or carrier as would be apparent to a skilled person.
  • Said microbicide can be coupled to, or included in, a support, for example a vaginal ring, for providing sustained protection against Candida species, as described, for example in Wahren et al. 2010 (Wahren et al. 2010. J Transl Med 8: 72).
  • Antimicrobial coatings must provide active resistance to microbe absorption and transference. In order to supply this microbial resistance, antimicrobial coatings often have release functions when exposed to bodily fluids in order to fight such infectious bacteria as E.coli and Staphylococcus, which they do by minimizing toxicity in organic tissue.
  • the invention further provides products that protect medical devices against Candidiases, prior to implantation. These products include sprays and gels emulsions that comprise VHH according to the invention that block biofilm formation on medical devices.
  • Such biofilm-blocking VHH preferably include Als3p, Hmalp and
  • the invention further provides a composition comprising an antibody according to the invention, preferably at least one antibody according to the invention.
  • Said composition preferably is a pharmaceutical composition.
  • a pharmaceutical composition preferably comprises a pharmaceutically acceptable carrier.
  • a carrier as used herein, means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredients.
  • physiologically acceptable refers to a non-toxic material that is compatible with a biological system such as a cell, cell culture, tissue, or organism. The characteristics of the carrier will depend on the route of administration. Physiologically and pharmaceutically acceptable carriers include diluents, fillers, salts buffers, stabilizers, solubilizers, and other materials which are well known in the art.
  • An anti- Ecelp or anti-Ece lp-derived peptide VHH antibody such as an anti-candidalysin VHH antibody of the invention may be labeled by a variety of means for use in diagnostic applications.
  • labels and methods of labeling known to those of ordinary skill in the art. Examples of the types of labels which can be used in the present invention include enzymes, radioisotopes, fluorescent compounds, colloidal metals,
  • radioisotopes may be bound to the VHH or the VHH-Fc fusion product either directly or indirectly by using an intermediate functional group.
  • Intermediate functional groups which are often used to bind radioisotopes which exist as metallic ions are the bifunctional chelating agents such as NOTA (1,4, 7-triazacyclononane- 1,4, 7- triacetic acid) and DOTA (1,4, 7, 10-tetraazacyclododecane- 1,4, 7, 10-tetraacetic acid) and derivatives thereof.
  • Typical examples of metallic ions which can be bound to the anti- Ecelp and/or anti-Als3p VHH antibodies, such as VHH antibodies directed against an Ecelp-derived peptide such as candidalysin, of the invention are .sup.lllln, .sup.97Ru, .sup.67Ga, .sup.GSGa. .sup.72As, .sup.89Zr and .sup.201Tl.
  • the antibodies of the invention can also be labeled with an infrared dye or with a paramagnetic isotope for purposes of in vivo diagnosis, as in, for example, magnetic resonance imaging (MRI), positron emission tomography (PET), single photon emission computed tomography (SPECT), computed tomography(CT) or photo-acoustic imaging.
  • MRI magnetic resonance imaging
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • CT computed tomography
  • photo-acoustic imaging acoustic imaging.
  • VHH used for imaging have an extended C-terminus containing at least the amino acid sequence A- C-A-A directly after the common terminus of VHH [VTVSS].
  • An extension at the C-terminus that contains the A-C-A-A amino acid sequence enables directed coupling of any imaging agent at the SH of the Cys residue.
  • the biggest advantage of labeling at the A-C-A-A amino acid sequence is that this
  • VHH retain full functionality.
  • the antibodies of the invention may further be used in vitro, for example, in immunoassays in which they can be utilized for detection of antigens in liquid phase or bound to a solid phase carrier.
  • the antibodies in these immunoassays can be detectably labeled in various ways. Examples of types of immunoassays which can utilize the antibodies of the invention are competitive and non-competitive immunoassays.
  • the assays either comprise a direct or an indirect format and include radioimmunoassay (RIA) and the sandwich assay.
  • RIA radioimmunoassay
  • a sample can be a liquid such as urine, saliva, cerebrospinal fluid, blood serum or the like; a solid or semisolid such as tissues, feces, or the like; or alternatively, a solid tissue such as those commonly used in histological diagnosis. 5. Examples
  • Candida albicans SC5314 C. a.
  • two llamas were immunised with both heat or formaldehyde inactivated C. albicans SC5314 yeast, germ and hyphae cells.
  • Filamentation of C. albicans was induced by incubation of yeast cells at 37 °C in RPMI-1640 (pH 7) supplemented with 165mM MOPS and 1.36 mM N- acetyl- glucosamine with shaking for 3 h (germ-tubes) or statically for 24 h (hyphae).
  • the fungal cells were inactivated with either 4% formaldehyde or by incubation for 2 h at 90 °C.
  • both llamas received full immunogen doses of formaldehyde and heat inactivated cells on day 0, 14, 28 and 35.
  • the immune response was verified by ELISA with llama sera from the day 0, 28 and 43 against coated Candida albicans SC5314 yeast, germ-tube and hyphae cells.
  • VHH libraries #7 and #11 have been created.
  • VHH coding sequences were cloned into phagemid pURSlOO and transformed to Escherichia coli TGI with an estimated diversity of 2.4* 10 7 clones for each library.
  • Ecelp in particularly the Ecelp-derived peptide candidalysin, a cytolytic peptide toxin secreted by the opportunistic pathogen Candida albicans which has been first described by Moyes et al., 2016 (Moyes et al., 2016. Nature 532: 64-68).
  • Candidalysin's C- terminus is hydrophobic, while its N-terminus is hydrophilic and basic.
  • the phage-display selections were conducted on immobilised candidalysin to keep the peptide in a solution and in a natural conformation, as described (Verheesen and Laeremans,
  • Candidalyins peptide was obtained from Peptide Protein Research Ltd.
  • the FLAG-tagged variants were obtained from Casio
  • E. coli amber suppressor Escherichia coli
  • TGI Amber suppressor Escherichia coli
  • K-12 F traD36 proAB lacIqZ DM15] supE thi-1
  • A(lac- proAB) A(mcrB-hsdSM)5(rK- mK-)
  • One vector was used for phages display and VHH production (pUR8100 derivative of pHENl with C- terminal c-myc and hexahistidine tag fusions to the gene insert; Hoogenboom et al., 1991. Nucleic Acids Res 19: 4133-4137).
  • Two other vectors were exclusively used for VHH production (pMEK219 and pMEK222, which are derivatives of pURSlOO without gene III, while in pMEK222 the c-myc- tag is substituted for a FLAG tag). All vectors contain the bacteriophage FI and the bacterial MB1 origin of replication as well as an ampicillin resistance gene coding for B-lactamase.
  • the inserted gene expression is under control of the lacZ promotor.
  • Gene inserts are N-terminally fused to a pelB leader sequence for translocation of the gene product to the periplasm for all three vectors.
  • a c-myc tag (EQKLISEEDL) followed by a hexahistidine tag (HHHHHH) are fused to the C-terminus of gene inserts in pURSlOO and pMEK219.
  • pMEK222 contains a FLAG tag (DYKDDDDK), followed by a hexahistidine tag.
  • TAA ochre stop codons
  • an amber stop codon TAG is present after the coding sequences, after which the M13 phage gene III has been inserted that codes for the M13 hull protein III.
  • the resulting supernatants were transferred to 250 pL aqueous solution containing 200 g/L PEG6000 (Sigma-Aldrich) and 2.5 M NaCl (Roth) for phage precipitation. After incubation for 30 min on ice the precipitated phages were sedimented by centrifugation (5 min at 17 000 x g). The supernatants were removed and the precipitated phages were resuspended in 1 mL PBS followed by a final centrifugation step (5 min at 17 000 x g) to remove any residual bacteria and storage of the resulting supernatants, which contain the purified phages, at 4 °C until further use in phage- display selections.
  • Phage productions from glycerol stocks of phage outputs from selections were carried out the same way with the following modifications until infection with helper phage: 5 mL 2x YT Medium (Roth) supplemented with 2% glucose (Roth) and 100 pg/mL ampicillin were inoculated with 75 pL of a selection output glycerol stock and incubated at 37°C and 200 rpm (Series 25 Incubator Shaker, New Brunswick Scientific) until they had reached an ODGOOnm of 0.5. Then 1 pL of helper phage (VCSM13, titer: 1.2*1013) was added to each sample. The remaining steps were as described for the phage production from libraries.
  • helper phage 5 mL 2x YT Medium (Roth) supplemented with 2% glucose (Roth) and 100 pg/mL ampicillin were inoculated with 75 pL of a selection output glycerol stock and in
  • an additional step has been performed. This step was necessary as selections on small - FLAG tagged peptides may result in false positive results because the selected phages recognize the capturing antibody, in this case the anti FLAG antibody, but not the peptides of interest.
  • the additional step was to substract from the original phage libraries all phages that have affinity to the capturing antibody, in particular phages that bind to the paratope of this capturing antibody. After removal of these phages according to methods known to a person skilled in the art, the subtracted phage libraries can be used to select phages that bind specifically to a non-candidalysin peptide.
  • the input phages which had been derived from the output phages of the lst-round of selection, were diluted 1:5 in 12,5% mouse serum (v/v) in 2,5% MPBS and pre-incubated for 30 min at room temperature on a rotating shaker at 20 rpm.
  • Tris/HCcl (NB) and TEA with Tris/HCl (2:1, ENB) were prepared of which 10 pL were transferred to 90 pL of the same E. cob TGI culture to ensure that the culture or the buffers had not been contaminated. After 30 min of incubation of the infected E. coli TGI samples at 37°C without shaking 5 pL of each dilution sample were spotted on LB/agar/glucose2%/ampicillinl00 pg/ml plates (Roth) and incubated overnight at 37°C.
  • each selection sample 75 pL of the neutralised phages from the elutions were used to infect 500 pL of the same E. coli TGI culture for storage of the phage outputs. After 30 min of incubation of the infected E. cob TGI samples at 37°C without shaking 5 pL of each dilution sample were spotted on LB/agar/glucose2%/ampicillinl00 pg/ml plates (Roth) and incubated overnight at 37°C.
  • glycerol stocks were made of the phage output samples by transfer of 1 ml culture to 0,5 ml 60% glycerol (Roth v/v) and stored at -80°C. The growth of the spotted dilution samples was documented by photographs and based on the number of colonies formed by the different dilutions of output phages, the number of eluted phages from each output and input was calculated.
  • Master plates were prepared by plating 100 pi of 10-5 dilutions of glycerol stocks from successful selection outputs onto
  • a control well with an E. cob TGI clone that carries a pURSlOO plasmid with an irrelevant VHH (well H5) and plasmid control with an E. cob TGI clone that carries an empty pUR8100 plasmid without any VHH (well Hll) were incorporated into the layout of the master plate.
  • Freshly prepared master plates were incubated overnight at 37°C then 40 pl/well of 60% glycerol were added. Master plates were stored at -80°C.
  • the pURSlOO phagemid's expression cassette contains a N-terminal pelB-leader sequence fused in frame within the open reading frame encoding VHH. Therefore, VHH expressed in E. coli TGI via pUR8100 is translocated to the periplasm.
  • Periplasmic extracts of induced E. coli TGI cultures were prepared from master plates for screening for antigen binding VHHs by ELISA. A copy plate of the master plate was prepared by transferring 1 gL of each well of the master plate to the corresponding wells of 96-well
  • polystyrene v-bottom microplates (# 651161, Greiner Bio-One) which had been filled with 80 pL 2xYT medium supplemented with 2% glucose and 100 pg/ml ampicillin (Roth). After incubation overnight at 37°C, 10 gl of each well of the copy plate were transferred to the respective wells of a polypropylene 96- well 2mL v-bottom assay block (Costar 3960, Coming Inc.) which had been filled with 900 gl /well 2xYT medium supplemented with 0.1% glucose and 100 gg/ml ampicillin (Roth) and pre- warmed to 37°C.
  • a polypropylene 96- well 2mL v-bottom assay block Costar 3960, Coming Inc.
  • the 96-well assay block was incubated for 3.5 h at 37°C and 200 rpm (Series 25 Incubator Shaker, New Brunswick Scientific), followed by addition of 100 gl/well of a 10 mM IPTG (Thermo Scientifc) solution in 2xYT-medium (Roth) for induction. The incubation was continued overnight at room temperature on vibrating platform shaker (Titramax 100, Heidolph Instruments) at 1350 rpm. The next morning the 96-well plate was centrifuged at 4 400 x g for 10 min resulting in sedimentation of the bacterial cells. The supernatants were discarded and the pellets were resuspended in 120 gl/well PBS.
  • PBS for peptide capturing or with 50 gL/well 5 pg/mL mouse IgG in PBS for detection of mouse IgG binders The plates were incubated overnight at 4°C. The next morning, after two washes with PBS, blocking was performed with 200 pL/well of 4% dried skimmed milk (Marvel, New York, NY) in PBS (4% MPBS) for 1 h at room temperature with shaking at 900 rpm on a vibrating platform shaker (Titramax 100, Heidolph Instruments GmbH & CO. KG, Schwab ach, Germany).
  • the blocking solution was discarded and the anti- FLAG M2 antibody coated wells were incubated with 50 pL/well of 5 mM peptide solution in 1% MPBs for 1 h at room temperature with shaking at 900 rpm on the platform shaker (mouse IgG coated wells were incubated solely with 1% MPBS). After two washes with PBS 50 pL/well of a 1:2 dilution of the periplasmic extracts in 1% MPBS were added to all coated wells (the layout of the master plate was conserved) followed by incubation for 1 h at room temperature and 900 rpm on the platform shaker.
  • Plasmid purification were performed with fresh overnight cultures of selected clones which had grown in 5 mL LB (Roth) medium supplemented with 2% glucose (m/v, Roth) and 100 mg/mL ampicillin (Roth) at 37°C and 200 rpm (Series 25 Incubator Shaker, New Brunswick Scientific).
  • NucleoSpin Plasmid Easy Pure Kit #740727.250; Macherey- Nagel, GmbH & Co. KG, Diiren, Germany
  • DNA sequencing of VHHs was conducted by the company Macrogen (Amsterdam. The Netherlands) and sequencing samples were prepared with purified plasmids and the M13rev primer according to Macrogen's recommendations.
  • the resulting supernatants (periplasmic extracts) were collected, mixed with 750 pL/sample of ddH20 and PBS washed iminodiacetic acid agarose/Co2+ beads (Roth, #1235.2) for Immobilized metal ion affinity chromatography (IMAC) purification and incubated on a rotating shaker at 20 rpm (Loopster basic, IKA-Works, Staufen im Breisgau, Germany) for 30 min at room temperature.
  • IMAC Immobilized metal ion affinity chromatography
  • the beads were collected by centrifugation (700 x g, 2 min) and washed one time with 50 mL PBS-Tween 20 (0,05%, v/v , Serva Electrophoresis) followed by two washes with 50 mL PBS (pH 7.4) and resuspension in 10 mL PBS.
  • the bead suspensions were transferred into chromatography columns ( Poly Prep, # 731-1550, BioRad, Hercules, CA) and rinsed with 10 mL PBS. Two pre-elutions were performed with 1 mL of 15 mM imidazole in PBS and 400 pL of 150 mM imidazole in PBS which both were discarded.
  • IMAC purified VHHs were analysed for their purity with the help of discontinuous sodium dodecylsulphate-polyacrylamide gelelectrophoresis (SDS-PAGE). Separation and stacking gel solutions had a concentration of 15% (pH 8.8) or 5% (pH 6.8) acrylamide, respectively. Discontinuous SDS- PAGE mini gels with a thickness of 0,75 mm were made by polymerization of separation and stacking gel solutions within the Mini-PROTEAN 3 Multi- Casting Chamber (BioRad).
  • the composition of the running gel was as follows: 13.5 mL ddH20, 15 mL 1.5 M TRIS (pH 8.8), 30 mL 30% acrylamide, 600 pL 10% SDS, 600 pL 10% ammonium persulfate (APS) and 60 pL tetramethylethyleendiamine (TEMED).
  • the composition of the stacking gel was as follows: 20.5 mL ddH20, 3.75 mL 1 M TRIS (pH 6.8), 5 mL 30% acrylamide, 300 pL 10% SDS, 300 pL 10% APS and 30 pL TEMED.
  • Digested VHH inserts were separated from the vector by DNA- gelelectrophoresis (1% agarose (Roth),TBE buffer (Roth), 80V constant) and extracted from the gel with the NucleoSpin Gel and PCR Clean-up kit (Macherey-N agel, # 740609.250) according to the manufacturer's instructions.
  • Digested VHH insert was ligated with pMEK219 vector hydrolysed with the same restriction enzymes by preparing samples with T4-DNA ligase (Thermo Scientific) according to the manufacturer's instruction. The ligation samples were transformed into chemically
  • Agarose gels were prepared by dissolving 1% agarose (w/v) in 0,5x TBE buffer (Roth) and adding 0.4 pg/mL ethidium bromide. Samples were mixed with 6x DNA loading dye (Thermo Scientific) before application. Separation was achieved with constant voltage of 5-8 V per cm distance between cathode and anode of the gel chamber. The running buffer was 0,5x TBE buffer and separated bands were visualized by exposition to UV radiation on a transilluminator.
  • PCR samples were composed of a colony sample resulting from transformation and 6.95 pL ddH20, 1 pL lOx Dream Taq green buffer (Thermo Scientific), 1 pL 2 mM dNTP mix, 0.5 pL 5 pM M13 forward primer (S'-GTAAAACGACGGCCAG-S 1 ), 0.5 pL 5 pM M13 reverse primer (5 1 - CAGGAAACAGCTATGAC- 3') , 0.05 pL Dream Taq DNA polymerase (5 U/pL, Thermo Scientific).
  • PCR conditions were 5 min 95°C/29 cycles of 30 s 95°C - 30 s 55°C - 45 s 72°C/ 10 min 72°C.
  • PCR products were analyzed by DNA agarose gelelectrophoresis, amplicon size of a positive clone was expected to be about 700 bp.
  • Colony samples were grown in parallel in 2xYT medium supplemented with 2% glucose and 100 pg/ml ampicillin at 37°C. Positive clones were prepared for plasmid purifications for DNA sequencing.
  • the cells were resuspended in 200 mL 100 mM CaC12 and incubated on ice for 25 min. Then cells were collected by centrifugation at 4°C, 2000 x g for 15 min. The supernatant was discarded and the pellet was resuspended in 50 mL of 85 mM CaCl2 with 15% glycerol followed by the final centrifugation step at 4°C, 2000 x g for 15 min. After that the supernatant was discarded and the pellet was resuspended in 2 mL of 85 mM CaC12 with 15% glycerol. Finally the suspension was divided into aliquots in 1,5 mL reaction tubes and stored at - 80°C until use.
  • N-terminally tagged Ecelp was expressed in E. coli. IMAC purification was performed with the help of a N-terminal hexa-histidine tag, which had been fused to the Ecel protein coding gene fragment.
  • the candidalysin bihead Cal-Fl-10 GS linker - Cal-Hl gene was ordered via Twist Bioscience (San Francisco, USA). The construct was ordered as a non-cloned gene with flanking cloning sites for restriction enzymes Sad and BstEII. The Fl-Hl VHH gene fragment was restricted with these enzymes and after cleaning of the restricted DNA the gene was ligated in the pYQVQll (Heukers et al., 2019. Antibodies 8: 26), which provides the VHH with a C-terminal C-Direct tag containing a free thiol (cysteine) and an EPEA (Glu, Pro, Glu, Ala) purification tag (C-tag, Thermo Fisher Scientific).
  • the pYQVQll plasmid containing the Fl-Hl gene was transformed through heat-shock into the Escherichia coli strain DH5a.
  • the colonies were allowed to grow overnight at 37 °C on LB agar plates supplemented with 2% glucose and 100 pg/ml ampicillin.
  • Six transformants were collected from the plate and grown again overnight at 37 °C with a rotation speed of 140 RPM in LB medium
  • the isolated plasmid DNA was transformed into Saccharomyces cerevisiae strain VWK18.
  • yeast cells Prior to transformation, yeast cells were prepared by growing the cells for 24 hours at 30 °C with a rotation speed of 140 RPM in Yeast Peptone medium supplemented with 2% glucose. The cells were collected by centrifugation at 5000 RCF for 10 minutes.
  • the DNA together with the transformation mix (50% (w fv) PEG3350, 1M LLAc, 5 mg/ml Fish sperm carrier DNA) and yeast cells transformed through the heat-shock method transforming the plasmid DNA into the yeast cell.
  • the VWK18 strain was grown for 2 - 4 days on a selective minimal medium plate (Yeast Nitrogen Base agar supplemented with 2% glucose) to form colonies.
  • Yeast Nitrogen Base medium supplemented with 2% glucose lacking leucine and grown for 24 hours at 30°C with a rotation speed of 140 RPM, thereby selecting for yeast cells with the pYQVQll which contains the gene for leucine which is essential for the yeast to grow.
  • the overnight culture is transferred to a rich medium where the culture was diluted 10 times in Yeast Peptone medium supplemented with 2% glucose and 1% galactose.
  • the culture was grown for 64 hours at 30°C with a rotation speed of 140 RPM.
  • the optical density at 600 nm of the culture was measured, where the desired optical density of > 20 was reached.
  • the supernatant was collect and filter sterilized after centrifugation at 5000 RCF for 15 minutes.
  • the produced VHH is excreted by the yeast cell and is present in the supernatant of the culture.
  • the supernatant was analysed on SDS-PAGE for the presence of VHH at 30kDa
  • DNA was isolated out of the two yeast transformant clones using the nucleospin plasmid easypure kit.
  • the two transformant clones are analysed on genetic scale by sequencing using yeast forward and reverse primers. The sequencing results were aligned against the initial Fl-Hl gene sequence and analysed.
  • VHH was purified using the tag containing the EPEA sequence and affinity chromatography, on the CaptureSelectTM C-tag column.
  • the purified VHH was buffer exchanged to PBS by desalting membrane.
  • the functionality of the bihead was tested using ELISA.
  • the candidalysin bihead F1-CAW3A8 gene was ordered via Twist Bioscience.
  • the construct was ordered as a non cloned gene with cloning sites for digestive enzymes Sad and BstEII.
  • the VHH gene was ligated in the desired plasmid pYQVQ 11.
  • the pYQVQ 11 plasmid containing the F1-CAW3A8 gene was transformed through heat-shock into the Escherichia coli strain DH5a.
  • the DH5a strain was grown overnight at 37 °C on LB agar plates supplemented with 2% glucose and 100 pg/ml ampicillin.
  • DNA was isolated out of two of the transformants using the nucleospin plasmid easypure kit. The isolated DNA was send for sequencing. The sequencing results were aligned against the initial F1-CAW3A8 gene sequence and analysed for correct insert.
  • the isolated plasmid DNA was transformed into the Saccharomyces cerevisiae strain VWK18 (Sagt et al., 1998. Appl Environ Microbiol 64: 316- 324).
  • yeast cells Prior to transformation, yeast cells were prepared by growing the cells for 24 hours at 30 °C with a rotation speed of 140 RPM in Yeast Peptone medium supplemented with 2% glucose. The cells were collected by centrifugation at 5000 RCF for 10 minutes and resuspended in a
  • transformation mix (50% (w/v) PEG3350, 1M LiAc, 5 mg/ml Fish sperm carrier DNA), together with the DNA.
  • Yeast cells were transformed through heat-shock method.
  • the VWK18 strain was grown for 2 - 4 days on a selective minimal medium plate to form colonies (Yeast Nitrogen Base agar supplemented with 2% glucose).
  • Two transformant clones were inoculated in Yeast Nitrogen Base medium supplemented with 2% glucose and grown for 24 hours at 30°C with a rotation speed of 140 RPM.
  • the overnight culture was transferred to a rich medium, in which the culture was diluted 10 times in Yeast Peptone medium supplemented with 2% glucose and 1% galactose.
  • the culture was grown for 64 hours at 30°C with a rotation speed of 140 RPM.
  • the optical density at 600 nm of the culture was measured, where a desired optical density of > 20 was reached.
  • the supernatant was collected and filter sterilized after
  • VHH was excreted by the yeast cell and was present in the supernatant of the culture. The supernatant was analysed on SDS-PAGE for the presence of VHH at 30kDa.
  • DNA was isolated out of the two yeast transformant clones using the nucleospin plasmid easypure kit.
  • the two transformant clones were sequenced and aligned against the initial F1-CAW3A8 gene sequence.
  • the VHH was purified using affinity chromatography on a
  • CaptureSelectTM C-tag column The purified VHH was desalted by the use of a desalting membrane. The desalted pure VHH was analysed on SDS-PAGE for the presence of VHH at 30 kDa and the absorbance at 280 nm wavelength was measured for the determination of VHH concentration. As the bihead Fl- CAW3A8 VHH also contains the VHH FI which recognizes Ecel-III-K cells, similar binding properties were expected in dose response ELISA using Ecel- III-K cells.
  • Recombinant Ecelp (rEcel) samples were separated by discontinuous SDS-PAGE (5% or 15% acrylamide for stacking or running gel, respectively). Separation was conducted at 60 V for 15 min followed by 80 V for 15 min and 100 V for 30 min. After equilibrating the membranes in blotting buffer (25 mM THIS, 192 mM glycine, 10% methanol, pH 8.5), transfer from the SDS- PAGE gel protein to PVDF membranes (Roche) was conducted at 2.5 mA/cm2 for 30 min with the Owl semi-dry electroblotter (Thermo Scientific) according to the manufacturer's instructions. All PVDF membrane incubations were performed on a rocking platform at room temperature.
  • PVDF membranes were blocked in PBS containing 4% dry milk powder (4% MPBS) for 1 h. Subsequent incubations were performed in 1% MPBS with 1 mM VHH raised against candidalysin (termed CAL1-F1 and CAL1-H1), polyclonal rabbit- anti- VHH purified antibody (QVQ, dilution 1:1000 ) and goat-anti-rabbit HRP conjugate IgG (#70748, dilution 1:3000, Cell Signaling Technologoy, Danvers, MA).
  • TR146 cells Human squamous cell carcinoma TR146 cells ( ECACC #10032305) were routinely grown in 20 mL DMEM/F-12 (Gibco, Thermo Scientific) with 10% FBS (Gibco, Thermo Scientific, heat inactivated) in 75 cm2 tissue culture flasks with vented caps (#83.3911.002, Sarstedt, Numbrecht, Germany) at 37°C, 5% CO2 until they reached confluency. Confluent TR146 cultures were washed with PBS and dry adherent cells were released by addition of 2 mL Accutase (#ACC-1B, Capricorn Scientific GmbH, Ebsdorferground, Germany) and incubation for 15 min at 37°C, 5% CO2.
  • Candida albicans strains and culture The Candida albicans strains used in this study are indicated in Table 2.
  • C. albicans was routinely grown on YPD agar at 30°C.
  • YPD broth was inoculated with a colony sample and incubated at 30°, 200 rpm on a platform shaker for 12-16 h.
  • TR146 cells 10 d oral epithelial cells (TR146 cells, ECACC #10032305) derived from a confluent culture were grown to confluence on round 12 mm coverslips (Roth, #P231.1) in 1 mL DMEM/F-12 with 10% FBS in 24-well flat clear bottom plates (Techno Plastic Products #92424) at 37°C, 5% CO2 for 2-3 days. After one wash with PBS, TR146 cells were infected with 2*10 4 yeast cells of an overnight culture of either C. albicans M2057 (ece 1D/D) or C. albicans M1477 (isogenic wildtype) in 1 mL of DMEM/F-12.
  • DMEM/F-12 containing candidalysin at sublytic concentrations (16-0.25 mM) were added to TR146 cells instead of yeast cell suspensions. After 6 hours of incubation at 37°C, 5% C02 samples were washed three times with PBS and fixed with HistoFix (4% buffered formaldehyde, Roth) overnight at 4°C. After three PBS washes samples were blocked with 1% BSA in PBS for 1 h at RT with gentles agitation. Subsequent incubation steps with antibodies were performed in 0.5% BSA.
  • TR146 cells 2*10 4 TR146 cells, derived from a confluent culture were grown to confluence in 200 pL DMEM/F-12) with 10% FBS in the inner wells of 96- well flat clear bottom plates (Techno Plastic Products #92696) at 37°C, 5% C02 for 1-2 days. The outer wells were filled with PBS. After one wash with PBS, the cells were infected with 2* 10 4 cells of an overnight culture of either C. albicans M2057 (ecel D/D) or C. albicans M1477 (isogenic wildtype) in 200 pL of DMEM/F-12. Additionally TR146 cells were exposed to 32 mM candidalysin in 200 pL of DMEM/F-12. Before application to TR146 all samples were preincubated for 1 h at 37°C, 5% C02.
  • the C. albicans suspensions or candialysin solutions were pre-incubated with serial dilutions of the VHHs CAL1-F1 or CAL1-H1. After 24 h of incubation at 37°C, 5% C02 supernatants of the TR146 samples were collected after centrifugation (10 min, 250 xg). As a measure of cell damage the lactate-dehydrogenase (LDH) release in relation to untreated controls was determined in the supernatants using the
  • VHHs that bind Ecelp and peptides derived from Ecelp such as candidalysin, were isolated from libraries created by immunisation of llama’s with various morphological forms of C. albicans cells, especially hyphae cells.
  • Two sequential rounds of phage display selections were conducted with Candida albicans SC5314 immune VHH libraries #7 and #11 on a candidalysin peptide that was either N-terminally (FCL) or C- terminally (CLF) fused to a FLAG protein tag.
  • the results of the first round of selection showed significant differences in phage outputs of selections on FCL, CLF or M2 antibody only for library 7 and 11. Outputs for library 7 were slightly higher than for library 11, PBS control outputs were lower than selection outputs. Enrichment of antigen binding phages was confirmed as phage outputs of the 2nd-round for library 7 of selections where roughly 100-1000 times higher than in the lst-round.
  • a master plate was generated and screened by periplasmic extract ELISA for binders to captured candidalysin.
  • M2 binder elimination for identification of relevant clones was performed because the outputs for 2nd round selections on M2 antibody were at best 10-times lower than captured candidalysin selection outputs, therefore a high proportion of M2 antibody binders was expected.
  • VHHs protect from candidalysin mediated cell damage.
  • damage inhibition assays with human TR146 epithelial cells were performed. TR146 cells were exposed for 24 hours to 10 5 C. albicans cells per mL or 32 mM synthesised
  • TR146 cell damage the lactate dehydrogenase release was determined in relation to epithelial cells exposed to the fungus or the toxin in absence of VHHs.
  • the results for C. albicans induced damage are shown in Figure 10A and indicate that both VHHs protect in a concentration dependent manner against C. albicans induced cell lysis. Both VHHs seem to abolish C. albicans induced cell damage at a concentration of 32 mM.
  • CALl-Hl is able to reduce cell damage caused by the fungus by approximately 80% at a concentration of 8 mM
  • CALl-Fl offers similar protection at a concentration of 4 mM and generally reduces damage better than CALl-Hl at equimolar concentrations.
  • VHH that recognize proteins involved in adhesin of C. albicans to cells or medical devices
  • Pathogenic fungi express a number of adhesins to bind to mammalian cells, in particular epithelial cells of the intestine or vagina. Subsequently these pathogenic fungi can secrete cytolytic peptides that destroy the cells to which they adhere. Blocking the adhesion of hyphae of C. albicans to these epithelial cells will contribute significantly to prevent translocation to the blood stream.
  • Adhesins can be grouped in three families: the Als, Hwp and Iff/Hyr families (de Groot et al., 2012. Eukaryotic Cell 12: 470—481). Although their sequences are quite different they have more or less the same molecular architecture. The method described below can be followed to select any adhesion.
  • Als3 is a multifunctional cell surface protein of C. albicans which plays a major role in adhesion and invasion of host cells.
  • Als3p is exclusively expressed at high levels during hyphal growth.
  • Als3p enables iron acquisition from host cell ferritin. (Liu and Filler, 2011. Eukaryotic Cell 10: 168-173).
  • SC5314 were prepared as described here for the C. albicans SC5314 immune library creation.
  • viable cells were either used immediately or fixed by overnight incubation in 4% formaldehyde at 4°C.
  • Three types of antigen preparations of these cells were used for phage display selections employing the C. albicans immune libraries 7 and 11: (1) Whole viable or formaldehyde inactivated cells, (2) purified cell walls derived from viable cells, or (3) supernatants of lyticase (Sigma) or zymolyase (Zymo Research) treated viable or formaldehyde inactivated cell suspensions.
  • Purified cells walls were prepared according to Kapteyn et al. 2000. Mol Microbiol 35: 601-611, with modifications. Cells were mechanically broken in lOmM Tris-HCl (pH 7.5 by using zirconia beads and cell homogenizer ( FastPrep-MP Biomedicals).
  • the cell debris containing cell walls, plasma membranes and cytoplasm were washed 5 times with 1 M NaCL, followed by three boiling steps at 100°C for 10 min in 500 mM Tris-HCl buffer [pH 7.5], 2 % [w/v] SDS, 0.3 M B-mercaptoethanol, and 1 mM EDTA and resuspended in 500 mM Tris-HCl buffer [pH 7.5] containing 2% [w/v] SDS, 0.3 M B- mercaptoethanol, and 1 mM EDTA). Finally the cell walls were lyophilised and stored at -80°C until further use.
  • Lyticase/zymolyase treatment of whole viable or formaldehyde inactivated cells was conducted by addition of 5-200 units of lyticase/zymolyase to a cell sample containing 2*10 ® - 2*10 ® cells in 10-70 mM sodium acetate buffer or potassium phosphate buffer (pH 5-8) and incubation at 30-37°C for 30-180 min.
  • Supernatants of treated cell samples were collected after centrifugation (4000-5000 x g, 5 min) and stored at 4°C until use in phage display selections
  • Phage display selections were performed on samples of these antigen preparations or recombinantly produced proteins in PBS which had been coated overnight at 4°C or for 2 h at RT to flat bottom hydrophilic polystyrene 96- well plates (Nunc Maxisorp #442404, Thermo Scientific). The phage display selections on the coated antigens were performed as described in Example 1. In addition to phage display selections on coated viable or formaldehyde inactivated C. albicans cells, these antigen preparations have been employed for selections on cells in suspension. Phage-display selections on cells in suspension were performed according to Romero-Martinez et. al., 2007. FEMS Immunol Medical Microbiol 50: 77-85, with modifications.
  • Immunofluorescence microscopy experiments with a set of deletion mutants demonstrated that the antigen is abundant in hyphal elongations and that no binding for an Als3 double deletion mutant could be detected.
  • Whole cell ELISAs were performed with VHH CAW3A8 on viable hyphae of the C. albicans mutant strains listed in Tables 3-5.
  • VHH CAW3A8 showed binding in ELISA to all of the hyphae forming mutants with exception of the Als3 double deletion mutant (1843).
  • Sequence analyses of the selected clones resulted in the amino acid sequences provided in the Table 3. In between brackets behind an amino acid residue are the amino acid residues that differ from the originally identified sequence. These substitutions are also able to bind to Als3.
  • Example 2 The method described in Example 2 to select VHH against cell wall protein Als3 was and can be followed for other cell wall proteins of C. albicans for which deletion mutants are available.
  • the results depicted in Figure 13 show that VHH were successfully isolated against a number of proteins from of C. albicans.
  • the fungal suspensions are pre-incubated with 10 mM or less endotoxin free VHH for 15-60 min at 37°C before injection.
  • Untreated or PBS- only, C. albicans cells-only, VHH-only and irrelevant- VHH control injected larvae are taken along. The injected larvae were kept in the dark at 37°C. Survival of larvae is documented and analyzed. Results of a single VHH and of a mixture of VHHs on the survival of G. mellonella are provided in Figures 14A and 14B.
  • Subfamily libraries contain VHH related to the parent VHH in terms of partial sequence identity with specificity towards Ecelp and Ecelp-derived peptides such as candidalysin.
  • the family member VHH may have higher affinity and/or neutralising activity towards Ecelp and Ecelp-derived peptides such as candidalysin, as well as improved physico-chemical stability or/and a higher yield during production.
  • Subfamily libraries for the parent VHH antibody CAL-F1 or HI is created by PCR amplification of VHH antibody DNA sequences as described herein.
  • amplicons containing related sequences to the parent VHH are created.
  • overlap extension PCR the two overlapping amplicons serve as template and amplification using the flanking primers of the constant plasmid or VHH framework regions yields a fusion product containing the complete VHH sequence due to hybridisation of the overlapping region.
  • the PCR or overlap extension PCR products are digested with restriction endonucleases, purified and ligated into a vector/phagemid (e. g pURSlOO). Highly efficient transformation of an appropriate bacterial or fungal strain, such as
  • Escherichia coli TGI by electroporation yields the subfamily library for application in VHH display selections, such as phage display. These selection will result in isolation of family specific VHH with improved properties over the parent VHH.
  • Primer pair examples for CALl-Fl subfamily library amplification from a library containing vector pURSlOO are:
  • primer concentrations are 0,05 -5 mM.
  • PCR and overlap extension PCR conditions depend on the primer pair and polymerase applied and can be e.g. 5 min 92-99°C/25-40 cycles of 30 s 92-99°C - 30 s 48-74°C - 30-90 s 74°C/ 1-10 min 68-74°C.
  • Example 5 Construction of vectors that can transform lactic acid bacteria Transformation of the described vectors results in either secretion of VHH by these lactic acid bacteria in the gastro intestinal tract or in expression of the VHH on their cell walls, thereby creating cells that bind to adhesin molecules on the surface of hyphae of C. albicans.
  • lactic acid bacteria have been used that bind to the mucosae of the intestine.
  • Other types of lactic acid bacteria have a long relative residence in the vagina as they bind to the mucosae of the vagina.
  • Lactic acid bacteria that secrete VHH carry one of the following genetic constructs:
  • VHHF1-E tag VHHH1-E tag and VHHAl E tag were codon-optimised for expression in lactic acid bacteria and synthesized (see Table 6).
  • These synthesized genes (QLF1 or QLH1 or QLA1) have on their 5’ end recognition sequences for the restriction enzyme Cfal and on their 3’ end, after the E-tag and a stop codon TAA, for the restriction enzyme Xhol.
  • These restriction sites enable integration of these constructs in pLP401 (Hultberg et al., 2007. BMC Biotechnol 7:58 10.1186/1472-6750-7-58), which plasmid was treated with Cfal and Xhol and the largest fragment was obtained by separation on molecular weight.
  • LAB were transformed with one of the plasmids pLPQOl or pLPQ02 or pLPQ03 and transformants were selected on MRS plates [Difco] with 3 pg/l erythromycin after cultivating anaerobically at 37 °C for 48 hours.
  • Table 6 Nucleotide sequences of Pamv funderlinedl - SSamv [italic! - fVHlTj - E- tag fnormall (A) and the amino acid sequences of SSamv [italic! and E- tae Tnormall (B)
  • Lactic acid bacteria that express on their surface VHH have one of the following genetic chemically synthesized constructs:
  • the anti-candidalysin VHH genes Cal-Fl- and Cal-Hl were ordered via Twist Bioscience.
  • the construct was ordered as a non cloned gene with flanking cloning sites for restriction enzymes Sad and BstEII.
  • the FI or HI VHH gene fragment was restricted with these enzymes and after cleaning of the restricted DNA the gene was ligated in pYQVQll.
  • the pYQVQll plasmid containing the FI or HI gene was transformed through heat-shock into the Escherichia coli strain DH5a. Colonies were allowed to grow overnight at 37 °C on LB agar plates supplemented with 2% glucose and 100 pg/ml ampicillin.
  • the isolated plasmid DNA was transformed into Saccharomyces cerevisiae strain VWK18 as described in Example 1.
  • DNA was isolated out of the two yeast transformant clones using the nucleospin plasmid easypure kit.
  • the two transformant clones were analysed by sequencing using yeast forward and reverse primers. The sequencing results were aligned against the initial FI or HI gene sequences and analysed.
  • VHH was purified using the tag containing the EPEA sequence and affinity chromatography, on the CaptureSelectTM C-tag column.
  • the purified VHH was buffer exchanged to PBS by desalting membrane.
  • the functionality of the individual clones was tested using ELISA similar as described in Example 1.
  • ECE1 or processed forms thereof use the same route, which means that ECE1 is mainly secreted at the tip of the hyphae. After secretion the ECE1 or processed forms thereof diffuse through the relative thin cell wall and enter in a relative thin water-like layer above the extra cellular matrix of epithelial cells. Subsequently, candidalysin, one of the processed forms of ECE1, passes this layer and enters the cell membrane of the epithelial cells and subsequently ensures lysis of these cells (see Figures 3 and 4).
  • VHH that bind to Als3, a protein characteristic for the tip of the hyphae and involved in the adhesion of the hyphae to epithelial cells.
  • VHH are coded VHH 3, VHH 14 [also described as clone 3A8 or VHH CAW3A8), VHH 15 and VHH 16 (see Figure 4B).
  • VHH 1 and 4 recognize biomolecules that are specific for hyphae, but the molecular nature of these antigens has not yet been determined.
  • Anti Als3, anti-Als4 and the 2 unknown hyphae specific biomolecules can serve as anchor for bi-specific VHH that capture Ecel or processed products thereof, like candidalysin, when passing the cell wall of the hyphae.
  • bihead should lower the number of candidalysin molecules that passes the ECM of epithelial cells. Even if not all candidalysin molecules are captured by these biheads, their effects can be very large, because only when the concentration of candidalysin at the surface of the target cell is high enough, multimeric forms of candidalysin will be formed and these multimeric forms can lyse the target cell.
  • the next step was testing the hypothesis that capturing of candidalysin by anti-candidalysin VHH was more effective when these VHH’s were present in a bi-specific bihead, comprising a VHH binding to Als3, notably VHH 14, 15 and 16 and a VHH binding the candidalysin.
  • Figures 15 and 16 show the protection provided by bihead, when compared to individual VHH’s.
  • the antibodies were tested against formaldehyde-inactivated cells and the absorbance measured at 490nm (subtracting the background 655nm) for yeast cells (a) and hyphae (b).
  • the grey scale for the absorbance was set on the background signal given by the control (no cells).
  • R indicates the signal intensity of the negative controls (0-15%), O (15-30%), Y (30-50%), C (50- 70%), L (70-85%), and G the highest signal in the plate (85-100%). The measurements were the average of two technical replicates.

Abstract

L'invention concerne un anticorps à domaine variable à chaîne lourde unique (VHH) qui se lie à Ece1p, et/ou un peptide dérivé de celui-ci, à partir de C. albicans. L'invention concerne en outre un VHH qui reconnaît les protéines de paroi cellulaire de C. albicans hyphae, telles que Als3p. L'invention concerne en outre un anticorps bi- ou multi-spécifique comprenant un tel VHH et un anticorps mono- ou bispécifique comprenant un tel VHH fusionné sur un niveau de gène à la partie Fc d'anticorps humains et des méthodes et des moyens de production de tels VHH. L'invention concerne en outre un anticorps de l'invention, destiné à être utilisé dans des applications diagnostiques et destiné à être utilisé en tant que médicament. L'invention concerne en outre des anticorps à domaine variable à chaîne lourde unique qui se lient aux adhésines de C. albicans et des champignons pathogènes associés. L'invention concerne en outre un anticorps de l'invention, destiné à être utilisé dans des applications diagnostiques ou destiné à être utilisé en tant que médicament. L'invention concerne en outre un microbicide comprenant un anticorps de l'invention, et une composition pharmaceutique comprenant un anticorps de l'invention. L'invention concerne enfin des bactéries lactiques de qualité alimentaire qui expriment un VHH contre Ece1p ou des adhésines.
PCT/NL2019/050875 2018-12-21 2019-12-23 Anticorps pour la prévention ou le traitement de la candidose WO2020130838A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NL2022281 2018-12-21
NL2022281 2018-12-21

Publications (2)

Publication Number Publication Date
WO2020130838A2 true WO2020130838A2 (fr) 2020-06-25
WO2020130838A3 WO2020130838A3 (fr) 2020-07-30

Family

ID=65576597

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2019/050875 WO2020130838A2 (fr) 2018-12-21 2019-12-23 Anticorps pour la prévention ou le traitement de la candidose

Country Status (1)

Country Link
WO (1) WO2020130838A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023039577A1 (fr) * 2021-09-10 2023-03-16 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Nanocorps à neutralisation large contre le coronavirus et leurs utilisations

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994004678A1 (fr) 1992-08-21 1994-03-03 Casterman Cecile Immunoglobulines exemptes de chaines legeres
WO1998052976A1 (fr) 1997-05-21 1998-11-26 Biovation Limited Procede de production de proteines non immunogenes
US5888809A (en) 1997-05-01 1999-03-30 Icos Corporation Hamster EF-1α transcriptional regulatory DNA
WO1999042077A2 (fr) 1998-02-19 1999-08-26 Xcyte Therapies, Inc. Compositions et procedes de regulation de l'activation des lymphocytes
WO2006040153A2 (fr) 2004-10-13 2006-04-20 Ablynx N.V. Nanocorps™ contre la proteine beta-amyloide et polypeptides les renfermant pour le traitement de maladies degeneratives neurales, telles que la maladie d'alzheimer
WO2006122825A2 (fr) 2005-05-20 2006-11-23 Ablynx Nv 'nanobodies™' (nanocorps) perfectionnes pour traiter des troubles medies par une agregation

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8092800B2 (en) * 2005-03-18 2012-01-10 Istituto Superiore Di Sanita Antibodies against Candida antigens
US20090186075A1 (en) * 2005-12-22 2009-07-23 Roitt Ivan M Composition
WO2014141094A1 (fr) * 2013-03-14 2014-09-18 Adamed Sp. Z O.O. Conjugué anticancéreux
GB201306588D0 (en) * 2013-04-11 2013-05-29 King S College London Peptides and Binding Partners Therefor

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994004678A1 (fr) 1992-08-21 1994-03-03 Casterman Cecile Immunoglobulines exemptes de chaines legeres
US5888809A (en) 1997-05-01 1999-03-30 Icos Corporation Hamster EF-1α transcriptional regulatory DNA
WO1998052976A1 (fr) 1997-05-21 1998-11-26 Biovation Limited Procede de production de proteines non immunogenes
WO1999042077A2 (fr) 1998-02-19 1999-08-26 Xcyte Therapies, Inc. Compositions et procedes de regulation de l'activation des lymphocytes
WO2006040153A2 (fr) 2004-10-13 2006-04-20 Ablynx N.V. Nanocorps™ contre la proteine beta-amyloide et polypeptides les renfermant pour le traitement de maladies degeneratives neurales, telles que la maladie d'alzheimer
WO2006122825A2 (fr) 2005-05-20 2006-11-23 Ablynx Nv 'nanobodies™' (nanocorps) perfectionnes pour traiter des troubles medies par une agregation

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
ACHOUR ET AL., J IMMUNOL, vol. 181, 2008, pages 2001 - 9
ANDERSON ET AL., INFECTION IMMUNITY, vol. 84, 2016, pages 395 - 341
BIRSE ET AL., INF IMMUNITY, vol. 61, 1993, pages 3648 - 3655
BROWN ET AL., SCIENCE TRANSL MED, vol. 4, 2012, pages 165rv13
DA SILVA DANTA ET AL., CURRENT OPINION MICROBIOL, vol. 34, 2016, pages 111 - 118
DE GROOT ET AL., EUKARYOTIC CELL, vol. 12, 2012, pages 470 - 481
DESCHACHT ET AL., J IMMUNOL, vol. 184, 2010, pages 5696 - 704
EDWARDS ET AL., CLIN INFECT DISEASES, vol. 66, 2018, pages 1928 - 1936
FRENKEN ET AL., J BIOTECHNOL, vol. 78, 2000, pages 11 - 21
FUCHS ET AL., VIRULENCE, vol. 1, 2010, pages 475 - 482
HEUKERS ET AL., ANTIBODIES, vol. 8, 2019, pages 26
HOOGENBOOM ET AL., NUCLEIC ACIDS RES, vol. 19, 1991, pages 4133 - 4137
HULTBERG ET AL., BMC BIOTECHN, vol. 7, 2007, pages 58
HULTBERG ET AL., BMC BIOTECHNOL, vol. 7, 2007, pages 58
JOOSTEN ET AL., J BIOTECHNOL, vol. 120, 2005, pages 347 - 359
KAPTEYN ET AL., MOL MICROBIOL, vol. 35, 2000, pages 601 - 611
KOH ET AL., J BIOL CHEM, vol. 285, 2010, pages 19116 - 19124
KUHN ET AL., PROTEOMICS CLIN APPL, vol. 10, 2016, pages 922 - 948
LAAR ET AL., BIOTECH BIOENG, vol. 96, 2007, pages 483 - 494
LIUFILLER, EUKARYOTIC CELL, vol. 10, 2011, pages 168 - 173
MARCOTTE ET AL., FUTURE VIROL, vol. 3, 2008, pages 327 - 341
MAYER ET AL., VIRULENCE, vol. 4, 2013, pages 119 - 128
MCCOY ET AL., PLOS PATH, vol. 10, 2014, pages e 1004552
MOYES ET AL., NATURE, vol. 532, 2016, pages 64 - 68
OOMEVAN DEN ACKERVEKEN, MPMI, vol. 27, 2014, pages 1081 - 1094
PANT ET AL., J INFECT DISEASES, vol. 194, 2006, pages 1580 - 8
PEREIRA ET AL., J FUNGI, vol. 4, 2018, pages 128
RAGHURAMANCHATTOPADHYAY, BIOSCI REP, vol. 27, 2007, pages 189 - 223
RAHBARIZADEH ET AL., J MOL IMMUNOL, vol. 43, 2006, pages 426 - 435
RAJPAL ET AL., PNAS, vol. 102, 2005, pages 84GG - 8471
RICHARDSON ET AL., MBIO, vol. 9, 2018, pages e02178 - 17
ROMERO-MARTINEZ, FEMS IMMUNOL MEDICAL MICROBIOL, vol. 50, 2007, pages 77 - 85
RUNNING DEERALLISON, BIOTECHNOL PROG, vol. 20, 2004, pages 880 - 889
SAGT ET AL., APPL ENVIRON MICROBIOL, vol. 64, 1998, pages 316 - 324
SCHMIDT ET AL., VACCINE, vol. 30, 2012, pages 7594 - 600
UPPULURI ET AL., FRONTIERS IMMUNOL, 2018, pages 9
VERHEESENLAEREMANS, METHODS MOL BIOL, vol. 911, 2012, pages 81 - 104
VERMA ET AL., SCIENCE IMMUNOLOGY, vol. 2, 2017, pages eaam8834
WAHREN ET AL., J TRANSL MED, vol. 8, 2010, pages 72
YAMANE-OHNUKI ET AL., BIOTECHNOL. BIOENG, vol. 87, 2004, pages 614 - 622
YAPAR, THER CLIN RISK MANAG, vol. 10, 2014, pages 95 - 105

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023039577A1 (fr) * 2021-09-10 2023-03-16 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Nanocorps à neutralisation large contre le coronavirus et leurs utilisations

Also Published As

Publication number Publication date
WO2020130838A3 (fr) 2020-07-30

Similar Documents

Publication Publication Date Title
JP7360262B2 (ja) 合成単一ドメイン抗体
DK2285408T3 (en) AMINO ACID SEQUENCES AGAINST COATING PROTEINS IN A VIRUS AND POLYPEPTIDES INCLUDING THESE FOR TREATMENT OF VIRUSAL DISEASES
WO2021183947A1 (fr) Molécules se liant au sars-cov-2
US20150158934A1 (en) Broadly neutralizing vhh against hiv-1
CN108610421B (zh) Cx3cr1结合多肽
EP2571898B1 (fr) Cd4 humain à domaine unique, soluble, complètement fonctionnel, à haute affinité, anticorps et protéines hybrides associées
CN111315784A (zh) 新的抗原结合嵌合蛋白及其方法和用途
JP2019503167A (ja) Cd38に対する抗原結合性ポリペプチド
JP2003284577A (ja) 癌マーカー用生物合成結合蛋白質
Mei et al. Research progress and applications of nanobody in human infectious diseases
JP2014525736A (ja) IgEに対する免疫グロブリン単一可変ドメイン
JP2023515505A (ja) Wntスーパーアゴニスト
JP2006523086A (ja) 発癌性形態のrasに対する細胞内発現抗体
JP2006521088A (ja) 抗活性化ras抗体
TWI489995B (zh) 具有抗綠膿桿菌作用之人化PcrV抗體
WO2020130838A2 (fr) Anticorps pour la prévention ou le traitement de la candidose
WO2022136685A1 (fr) Compositions d'anticorps pour traiter une infection par le virus corona
JP7433222B2 (ja) Alk7結合タンパク質およびその使用
US20220112276A1 (en) Biosynthetic materials and methods for multidirectional biotransportation
Lim et al. Single domain antibodies derived from ancient animals as broadly neutralizing agents for SARS-CoV-2 and other coronaviruses
US10882899B2 (en) Anti Francisella tularensis (FT) antibodies
US20180289833A1 (en) Antimicrobial compositions comprising single domain antibodies and pseudomonas exotoxin
CN113248610B (zh) 白细胞介素2结合分子、其衍生物、试剂盒及其生产方法和用途
US20230279077A1 (en) Anti-glycoprotein d antibodies, methods of preparation, and uses thereof
WO2024068744A1 (fr) Antiviraux dirigés contre le virus parainfluenza humain

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: 19835549

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19835549

Country of ref document: EP

Kind code of ref document: A2