ANTIBODY MOLECULES AND USES THEREOF

Abstract
This invention relates to recombinant human antibody molecules. The antibodies bind fungal antigens, for example from Candida spp. Human antibody encoding genes targeting clinically relevant Candida epitopes have been isolated from single B cells from carefully selected donors and screened with specified types of protein or cell wall extract. The panel of purified, fully human recombinant IgG1 mAbs generated displayed a diverse range of specific binding profiles and demonstrated efficacy in a disease model. The fully human mAbs and derivatives thereof have utility in the generation of diagnostics, therapeutics and vaccines.
Description
TECHNICAL FIELD

This invention relates to recombinant human antibody molecules. The antibodies bind fungal antigens, for example from Candida spp. Such antibody molecules find use in the treatment, diagnosis and/or detection of fungal infections.


BACKGROUND ART

Fungi cause 3 million life-threatening infections each year, killing more people than tuberculosis and as many people as malaria (1). To make inroads into these high disease burdens and mortality figures, better diagnostics, treatments and fungal vaccines are urgently required.



Candida species collectively account for the majority of serious fungal infections and represent the fourth leading cause of healthcare-associated infections in the US (1, 2). Candida albicans is a common human commensal and the most prevalent fungal opportunistic pathogen (3). C. albicans is polymorphic, phenotypically variable and genetically diverse. Impairment of host immunity, due to mutation, pharmacological or surgical intervention, trauma or alteration in the natural microbiota, determines the frequency and severity of disease (4). Late diagnosis of invasive candidiasis using ‘gold standard’ blood culture methodologies and limitations in the versatility and accuracy if some diagnostic tests contribute to the overall poor prognosis and high mortality rates associated with septicaemia and invasive fungal disease (5-7).


Existing classes of antifungals are effective against infection but tend to have relatively narrow spectra of activity that means that informed therapy is predicated on accurate diagnosis (2, 8).


There are currently no vaccines for fungal infection in the clinic although experimental vaccines based on fungal cell wall targets are in pre-clinical development (20, 22-27). These include the investigational vaccine NDV-3 based on a recombinant fragment of the Als3 cell wall adhesin which has now entered phase II clinical trials for recurrent vulvovaginal candidiasis (RVVC) (26, 28) and a Candida-specific vaccine based on the recombinant N-terminal fragment of the Hyr1 protein expressed on C. albicans hyphae which has shown efficacy in a murine model of disseminated candidiasis (23, 29). These experimental vaccines exert their protective effects by eliciting neutralising and/or protective antibodies (23).


Protective monoclonal antibody (mAbs) for clinically relevant fungi have been reported (15-17). A number of protective mAbs targeting pan fungal and species-specific epitopes have been isolated, which are almost exclusively murine in origin, and generated via hybridoma technology (15, 18-22).


Increased mAb research in the field of mycotic disease has also led to progress in mAb-based diagnostics including the Aspergillus-specific mAb JF5 for the detection of invasive pulmonary aspergillosis (IPA), a C. albicans germ tube antibody (CAGTA) for deep-seated Candida infection and a new cryptococcal dipstick antigen test (30-33).


Nevertheless it can be seen that novel sources of diagnostic and therapeutic reagents targeting fungal pathogens would provide a contribution to the art.


DISCLOSURE OF THE INVENTION

The present invention seeks to provide novel diagnostics and therapeutics for fungal infections, through a mAb-based approach using C. albicans as the model organism.


The inventors have isolated human antibody encoding genes targeting clinically relevant Candida epitopes from single B cells that were derived from donors with a history of mucosal Candida infections screened with recombinant Candida albicans Hyr1 cell wall protein or whole fungal cell wall extracts. The panel of purified, fully human recombinant IgG1 mAbs generated displayed a diverse range of specific binding profiles to other pathogenic fungi and demonstrated efficacy in a murine model of disseminated candidiasis. The fully human mAbs have utility in the generation of diagnostics, therapeutics and vaccines.


In various aspects of the invention there are provided isolated recombinant human anti-Candida antibody molecule derived from single B cells, for example which specifically bind Candida cells or more specifically C. albicans hyphae.


Preferred antibody molecules have CDRs, FWs, VH and VL domains having sequences set out in Tables A-R, each Table being the sequence of one of the 18 antibodies of the Examples, or derivatives of those sequences having one or more amino acid substitutions, deletions or insertions.


Also provided are methods for producing an antibody antigen-binding domain for a fungal antigen, or for producing an antibody molecule that specifically binds to a fungal antigen, which methods comprise utilising or modifying one or more of the CDRs, FWs, VH and VL domains having sequences set out in Tables A-R.


Also provided are methods of identifying or labelling a Candida cell, or the hyphae of C. albicans, of opsonising, or increasing the rate of opsonisation of a Candida cell, of increasing the rate of engulfment of a Candida cell, or of increasing the rate of macrophage attraction to Candida cell, the methods utilising the antibody molecules of the invention.


The invention also provides therapeutic and diagnostic utilities for the antibody molecules of the invention, and diagnostic devices utilising them.


Some of these aspects and embodiments of the invention will now be described in more detail.


mAbs and Processes of Production


Pooled immunoglobulin from serum was one of the first widely available treatments for microbial infections and that hyperimmune human sera immunoglobulin is still used today to treat a number of infections including cytomegalovirus (CMV), hepatitis A and B virus (HAV, HBV) rabies and measles (12-14). Nevertheless, although in recent years humanised versions of mAbs have become some of the world's bestselling drugs, to date the majority of these mAbs have been licensed for the treatment of cancer and autoimmune diseases (9-11), there is currently only one mAb approved for the treatment of an infectious disease (13).


Methods for the production of mAbs for therapeutic and/or diagnostic use have diversified dramatically over the decades. Early mAbs were mainly of murine origin but tended to be immunogenic in the human host (34, 35). The majority of mAbs currently in the clinics are humanized or fully human IgG1 mAbs generated through hybridoma cell lines (14, 35). Combinatorial display technologies using phage or yeast have been valuable but require a period of in vitro affinity maturation and lose the natural antibody heavy and light chain pairings (14).


Recently, direct amplification of individual VH and VL chain domain genes from single human B cells to ensure retention of native antibody heavy and light chain pairings, has led to the generation of fully human mAbs with increased safety and relevance to human disease in areas where current treatments are suboptimal (14, 36-39).


Antibody Molecules

Anti-Candida recombinant human antibody molecules of the invention may include any polypeptide or protein comprising an antibody antigen-binding site described herein, including Fab, Fab2, Fab3, diabodies, triabodies, tetrabodies, minibodies and single-domain antibodies, as well as whole antibodies of any isotype or sub-class.


The anti-Candida recombinant human antibody molecules may also be a single-chain variable fragment (scFv) or single-chain antibody (scAb). An scFv fragment is a fusion of a variable heavy (VH) and variable light (VL) chain. A scAb has a constant light (CL) chain fused to the VL chain of an scFv fragment. The CL chain is optionally the human kappa light chain (HuCκ). A single chain Fv (scFv) may be comprised within a mini-immunoglobulin or small immunoprotein (SIP), e.g. as described in Li et al. (1997). An SIP may comprise an scFv molecule fused to the CH4 domain of the human IgE secretory isoform IgE-S2 (εS2-CH4; Batista, F. D., Anand, S., Presani, G., Efremov, D. G. and Burrone, O. R. (1996). The two membrane isoforms of human IgE assemble into functionally distinct B cell antigen receptors. J. Exp. Med. 184:2197-2205) forming an homo-dimeric mini-immunoglobulin antibody molecule.


Antibody molecules and methods for their construction and use are described, in for example, Holliger, P. and Hudson, P. J. (2005). Engineered antibody fragments and the rise of single domains. Nat. Biotechnol. 23:1126-1136.


Anti-Candida recombinant human antibody molecules as described herein may lack antibody constant regions.


However in some preferred embodiments, the anti-Candida recombinant human antibody molecule of the invention is a whole antibody. For example, the anti-Candida recombinant human antibody molecule may be an IgG, IgA, IgE or IgM or any of the isotype sub-classes, particularly IgG1.


Anti-Candida recombinant human antibody molecules as described will generally be provided in isolated form, in the sense of being free from contaminants, such as antibodies able to bind other polypeptides and/or serum components.


Anti-Candida recombinant human antibody molecules of the invention may be obtained in the light of the disclosure herein, for example using techniques described in reference (14).


Antibody molecules of the invention typically comprise an antigen binding domain comprising an immunoglobulin heavy chain variable domain (VH) and an immunoglobulin light chain variable domain (VL).


Each of the VH and VL domains typically comprise 3 complementarity determining regions (CDRs) responsible for antigen binding, interspersed by 4 framework (FW) regions.


In Tables A-R hereinafter, the sequences of each of the CDRs and FWs for each of the VH and VL domains is given for each of the preferred 18 antibodies of the invention i.e. Antibodies 120-124 (directed to the Hyr1 protein), and also 118-119, 126-127, 129-135, and 139-140 (directed to C. albicans ‘whole cell’):


Tables VH and VL give the entire VH and VL domains of these 18 antibodies.


In these tables, each antibody the sequences are numbered as follows:


















 1x
H FW1



 2x
H CDR1



 3x
H FW2



 4x
H CDR2



 5x
H FW3



 6x
H CDR3



 7x
H FW4



 8x
L FW1



 9x
L CDR1



10x
L FW2



11x
L CDR2



12x
L FW3



13x
L CDR3



14x
L FW4



15x
VH full sequence



16x
VL full sequence










Tables “VH-CDR3-mod” and “VL-CDR3-mod” show pairs of variants of the CDR3 sequences of some of the VH domains (i.e. SEQ ID No 6x) and VL domains (SEQ ID No 13x) respectively. These VH-CDR3 variants are numbered SEQ ID Nos 17x/18x and these VL-CDR3 variants are numbered SEQ ID No 19x/20x.


In each case ‘x’ represents any single letter of A-R, each letter representing one of the 18 antibodies 118-124, 126-127, 129-135, and 139-140, for example ‘A’ represents antibody


AB119 described in Table A and Tables VH and VL. It will be understood that the description in relation to sequence ‘x’ applies mutatis mutandis to any of the antibodies described in Tables A-R, as if that description was written individually for each antibody.


In some embodiments, Anti-Candida recombinant human antibody molecules of the invention may binding to the target wholly or substantially through a VHCDR3 sequence described herein.


Thus, for example, an anti-Candida recombinant human antibody molecule may comprise a VH domain comprising a HCDR3 having the amino acid sequence of SEQ ID NO: 6x or the sequence of SEQ ID NO: 6x with 1 or more, for example 2, or 3 or more amino acid substitutions, deletions or insertions (e.g. as shown in SEQ ID NO: 17x or 18x).


Substitutions as described herein may be conservative substitutions or may be present to remove Cys residues from the native sequence. In some embodiments, an antibody may comprise one or more substitutions, deletions or insertions which remove a glycosylation site.


The HCDR3 may be the only region of the antibody molecule that interacts with a target epitope or substantially the only region. The HCDR3 may therefore determine the specificity and/or affinity of the antibody molecule for the target.


The VH domain of an anti-Candida recombinant human antibody molecule may additionally comprise an HCDR2 having the amino acid sequence of SEQ ID NO: 4x or the sequence of SEQ ID NO: 4x with 1 or more, for example 2, or 3 or more amino acid substitutions, deletions or insertions.


The VH domain of an anti-Candida recombinant human antibody molecule may further comprise an HCDR1 having the amino acid sequence of SEQ ID NO: 2x or the sequence of SEQ ID NO: 2x with 1 or more, for example 2 or 3 or more amino acid substitutions, deletions or insertions.


In some embodiments, an antibody molecule may comprise a VH domain comprising a HCDR1, a HCDR2 and a HCDR3 having the sequences of SEQ ID NOs 2x, 4x and 6x respectively.


In some embodiments, an antibody molecule may comprise a VH domain comprising one or more or all of a FW1, a FW2, a FW3 and a FW4 having the sequences of SEQ ID NOs 1x, 3x, 5x and 7x respectively. Any of these FW regions may include 1 or more, for example 2 or 3 or more amino acid substitutions, deletions or insertions.


For example, an antibody molecule may comprise a VH domain having the sequence of SEQ ID NO: 15x or the sequence of SEQ ID NO: 15x with 1 or more, for example 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acid substitutions, deletions or insertions in SEQ ID NO: 15x.


The anti-Candida recombinant human antibody molecule will typically further comprise a VL domain, for example a VL domain comprising LCDR1, LCDR2 and LCDR3 having the sequences of SEQ ID NOs 9x, 11x and 13x respectively, or the sequences of SEQ ID NOs 9x, 11x and 13x respectively with, independently, 1 or more, for example 2 or 3 or more amino acid substitutions, deletions or insertions. Examples of variant LCDR3 sequences are shown in SEQ ID NOs: 19x and 20x.


In some embodiments, an antibody molecule may comprise a VL domain comprising one or more or all of a FW1, a FW2, a FW3 and a FW4 having the sequences of SEQ ID NOs 8x, 10x, 12x and 14x respectively. Any of these may include 1 or more, for example 2 or 3 or more amino acid substitutions, deletions or insertions.


For example, an antibody molecule may comprise a VL domain having the sequence of SEQ ID NO: 16x or the sequence of SEQ ID NO: 16x with 1 or more, for example 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acid substitutions, deletions or insertions in SEQ ID NO: 16x.


The anti-Candida recombinant human antibody molecule may for example comprise one or more amino acid substitutions, deletions or insertions which improve one or more properties of the antibody, for example affinity, functional half-life, on and off rates.


The techniques required in order to introduce substitutions, deletions or insertions within amino acid sequences of CDRs, antibody VH or VL domains and antibodies are generally available in the art. Variant sequences may be made, with substitutions, deletions or insertions that may or may not be predicted to have a minimal or beneficial effect on activity, and tested for ability to bind to C. albicans antigens and/or for any other desired property.


In some embodiments, an anti-Candida recombinant human antibody molecule may comprise a VH domain comprising a HCDR1, a HCDR2 and a HCDR3 having the sequences of SEQ ID NOs 2x, 4x, and 6x (or 17x or 18x), respectively, and a VL domain comprising a LCDR1, a LCDR2 and a LCDR3 having the sequences of SEQ ID NOs 9x, 11x and 13x (or 19x or 20x), respectively.


For example, the VH and VL domains may have the amino acid sequences of SEQ ID NO: 15x and SEQ ID NO: 16x respectively; or may have the amino acid sequences of SEQ ID NO: 15x and SEQ ID NO: 16x comprising, independently 1 or more, for example 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acid substitutions, deletions or insertions.


In some embodiments, an anti-Candida recombinant human antibody molecule VH domain may have at least about 60% sequence identity to SEQ ID NO: 15x, e.g. at least about 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to SEQ ID NO: 15x.


In some embodiments, an anti-Candida recombinant human antibody molecule VL domain may have at least about 60% sequence identity to SEQ ID NO: 16x, e.g. at least about 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to SEQ ID NO: 16x.


The anti-Candida recombinant human antibody molecule may be in any format, as described above, In some preferred embodiments, the anti-Candida recombinant human antibody molecule may be a whole antibody, for example an IgG, such as IgG1, IgA, IgE or IgM. In some preferred embodiments, than anti-Candida recombinant human antibody molecule is a scAb or scFv.


An anti-Candida recombinant human antibody molecule of the invention may be one which competes for binding to the target (e.g. Hyr1) with an antibody molecule described herein, for example an antibody molecule which


(i) binds Hyr1 and


(ii) comprises a VH domain of SEQ ID NO: 15x and/or VL domain of SEQ ID NO: 16x; an HCDR3 of SEQ ID NO: 6x; an HCDR1, HCDR2, LCDR1, LCDR2, or LCDR3 of SEQ ID NOS: 2x, 4x, 9x, 11x or 13x respectively; a VH domain comprising HCDR1, HCDR2 and HCDR3 sequences of SEQ ID NOS: 2x, 4x and 6x respectively; and/or a VH domain comprising HCDR1, HCDR2 and HCDR3 sequences of SEQ ID NOS: 2x, 4x and 6x and a VL domain comprising LCDR1, LDR2 and LCDR3 sequences of SEQ ID NOS: 9x, 11x and 13x respectively,

    • where x here is C, D, E, F or G.


An anti-Candida recombinant human antibody molecule of the invention may be one which competes for binding to the target (e.g. C. albicans whole cell wall extract) with an antibody molecule described herein, for example an antibody molecule which


(i) binds C. albicans whole cell wall extract, and


(ii) comprises a VH domain of SEQ ID NO: 15x and/or VL domain of SEQ ID NO: 16x; an HCDR3 of SEQ ID NO: 6x; an HCDR1, HCDR2, LCDR1, LCDR2, or LCDR3 of SEQ ID NOS: 2x, 4x, 9x, 11x or 13x respectively; a VH domain comprising HCDR1, HCDR2 and HCDR3 sequences of SEQ ID NOS: 2x, 4x and 6x respectively; and/or a VH domain comprising HCDR1, HCDR2 and HCDR3 sequences of SEQ ID NOS: 2x, 4x and 6x and a VL domain comprising LCDR1, LDR2 and LCDR3 sequences of SEQ ID NOS: 9x, 11x and 13x respectively,

    • where x here is A-B, or H-R.


Competition between antibody molecules may be assayed easily in vitro, for example using ELISA and/or by tagging a specific reporter molecule to one antibody molecule which can be detected in the presence of one or more other untagged antibody molecules, to enable identification of antibody molecules which bind the same epitope or an overlapping epitope. Such methods are readily known to one of ordinary skill in the art.


Thus, a further aspect of the present invention provides a binding member or antibody molecule comprising an antigen-binding site that competes with an antibody molecule, for example an antibody molecule comprising a VH and/or VL domain, CDR e.g. HCDR3 or set of CDRs of the parent antibody described above for binding to target antigen. A suitable antibody molecule may comprise an antibody antigen-binding site which competes with an antibody antigen-binding site for binding to target antigen wherein the antibody antigen-binding site is composed of a VH domain and a VL domain, and wherein the VH and VL domains comprise HCDR1, HCDR2 and HCDR3 sequences of SEQ ID NOS: 2x, 4x, and 6x (or 17x or 18x) and LCDR1, LDR2 and LCDR3 sequences of SEQ ID NOS: 9x, 11x, and 13x (or 19x or 20x) respectively, for example the VH and VL domains of SEQ ID NOS: 15x and 16x.


The VH and VL framework encoded by the genes encoded from the B cell antibody factories can be readily modified by molecular genetics to alter and refine the properties of the antibodies. Such modified sequences are termed “derived” from the B cells herein.


For example is may be desired to remove Cys residues in the sequence, to minimise potential incorrect Cys pairings.


Thus the invention also provides a method for producing an antibody antigen-binding domain for a fungal target as described herein, preferably C. albicans Hyr1 protein or whole cell wall extract, which comprises:

    • providing, by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VH domain comprising HCDR1, HCDR2 and


HCDR3, wherein the parent VH domain HCDR1, HCDR2 and HCDR3 have the amino acid sequences of SEQ ID NOS: 2x, 4x and 6x respectively, a VH domain which is an amino acid sequence variant of the parent VH domain, and;

    • optionally combining the VH domain thus provided with one or more VL domains to provide one or more VH/VL combinations; and
    • testing said VH domain which is an amino acid sequence variant of the parent VH domain or the VH/VL combination or combinations to identify an antibody antigen binding domain for target antigen.


A VH domain which is an amino acid sequence variant of the parent VH domain may have the HCDR3 sequence of SEQ ID NO: 6x or a variant with the addition, deletion, substitution or insertion of one, two, three or more amino acids e.g. 17x or 18x.


The VH domain which is an amino acid sequence variant of the parent VH domain may have the HCDR1 and HCDR2 sequences of SEQ ID NOS: 2x and 4x respectively, or variants of these sequences with the addition, deletion, substitution or insertion of one, two, three or more amino acids.


The invention also provides a method for producing an antibody antigen-binding domain for a fungal target as described herein, preferably C. albicans Hyr1 protein or whole cell wall extract, which comprises:

    • providing starting nucleic acid encoding a VH domain or a starting repertoire of nucleic acids each encoding a VH domain, wherein the VH domain or VH domains either comprise a HCDR1, HCDR2 and/or HCDR3 to be replaced or lack a HCDR1, HCDR2 and/or HCDR3 encoding region;
    • combining said starting nucleic acid or starting repertoire with donor nucleic acid or donor nucleic acids encoding or produced by mutation of the amino acid sequence of an HCDR1, HCDR2, and/or HCDR3 having the amino acid sequences of SEQ ID NOS: 2x, 4x and 6x respectively, such that said donor nucleic acid is or donor nucleic acids are inserted into the CDR1, CDR2 and/or CDR3 region in the starting nucleic acid or starting repertoire, so as to provide a product repertoire of nucleic acids encoding VH domains;
    • expressing the nucleic acids of said product repertoire to produce product VH domains;
    • optionally combining said product VH domains with one or more VL domains; selecting an antibody molecule that binds the fungal target, which antibody molecule comprises a product VH domain and optionally a VL domain; and


recovering said antibody molecule or nucleic acid encoding it.


Suitable techniques for the maturation and optimisation of antibody molecules are well-known in the art.


Anti-Candida recombinant human antibody molecules may be further modified by chemical modification, for example by PEGylation, or by incorporation in a liposome, to improve their pharmaceutical properties, for example by increasing in vivo half-life.


An anti-Candida recombinant human antibody molecule as described herein may conjugated to a toxic payload (e.g. ricin) that could kill the fungus and act as a therapeutic antibody.


An anti-Candida recombinant human antibody molecule as described herein may be one which binds Hyr1 with an EC50 values of 1 to 1500, e.g. 10 to 500, or 20 to 200 ng/ml.


An anti-Candida recombinant human antibody molecule as described herein may be one which binds C. albicans with an EC50 values of 1 to 1500, e.g. 1 to 500, or 1 to 40 ng/ml.


EC50 can be assessed as described hereinafter with ELISA e.g. as described in the Examples below for “Circulating IgG Enzyme-linked Immunosorbent assay (ELISA) to identify donors with B cells to take forward” and “B cell supernatant screen against target antigens via ELISA”.


Provided herein is a method of binding a fungal cell, for example C. albicans, the method comprising contacting the fungal cell with an anti-Candida recombinant human antibody molecule as described herein.


It is known that there are a large number of Candida species. Key Candida species which may be targeted by the antibodies described herein include Candida albicans, Candida glabrata, Candida tropicalis, Candida parapsilosis (a clonal complex of three species—C. parapsilosis, C. orthopsilosis and C. metapsilosis), and Candida krusei (synonym: Issatchenkia orientalis). Less-prominent species include Candida guiffiermondii, Candida lusitaniae, Candida kefyr, Candida famata (synonym: Debaryomyces hansenii), Candida inconspicua, Candida rugosa, Candida dubliniensis, Candida norvegensis, Candida auris, Candida haemulonii.


As described herein, the anti-Candida recombinant human antibody molecules of the invention can detect both morphology specific and morphology-independent epitopes with high specificity. The antibody molecules described herein may thus bind to C. albicans with high affinity relative to other fungal targets. For example, an antibody molecule of the invention may display a binding affinity for C. albicans which is at least 1000 fold or at least 2000 fold greater than a non-Candida pathogenic fungus such as Aspergillus fumigatus and Cryptococcus neoformans and Pneumocystis jirovecii.


Nevertheless an anti-Candida recombinant human antibody molecule as described herein may bind to the species closely related to C. albicans e.g. C. dubliniensis, C. tropicalis, C. parapsilosis (clonal complex), C. krusei, C. auris (clonal complex), C. glabrata and C. lusitaniae e.g. for example with an affinity within a 1000-fold o of the binding to C. albicans (assessed using EC50).


Provided herein is a method of opsonising, or increasing the rate of opsonisation of a fungal cell, for example C. albicans, the method comprising contacting or pre-incubating the fungal cell with an anti-Candida recombinant human antibody molecule as described herein.


Provided herein is a method of increasing the rate of engulfment of a fungal cell, for example C. albicans, by macrophages, the method comprising contacting the fungal cell with an anti-Candida recombinant human antibody molecule as described herein. The antibody molecule may optionally be one specific for the hyphal-specific protein Hyr1.


Provided herein is a method of increasing the rate of macrophage attraction to a fungal cell, for example C. albicans, the method comprising contacting or pre-incubating the fungal cell with an anti-Candida recombinant human antibody molecule as described herein. The antibody molecule may optionally be one raised to whole cell wall preparation of the fungal cell.


Treatment of Disease

An anti-Candida recombinant human antibody molecule as described herein may be used for clinical benefit in the treatment of a fungus-associated condition, and particularly infections caused by Candida species, i.e. candidiasis. Preferred antibody molecules are those specific for C. albicans cell wall preparations.



C. albicans is the most common serious fungal pathogen of humans, and the embodiments disclosed herein may be used in the prophylaxis or treatment of any condition related to infection caused by C. albicans.


The fungus is part of the normal gut flora of around 50% of the population and is normally harmless but can cause superficial mucosal infections such as oral and vaginal thrush and life-threatening systemic disseminated disease in immunocompromised individuals. Immunocompromised individuals may have a weakened immune system due to medical treatment (e.g. cancer treatment or organ transplant recipients), or due to a disease or disorder (e.g. HIV/AIDS, SCID, CVID). Other conditions that may be treated include lung infections in cystic fibrosis patients, mixed microbial infections, which include both bacteria (e.g. Pseudomonas spp.) and fungi, fungal infections on indwelling medical devices such as catheters, and skin and urinary tract infections.


The antibody molecules as described herein may be useful in the surgical and other medical procedures which may lead to immunosuppression, or medical procedures in patients who are already immunosuppressed.


Patients suitable for treatment as described herein include patients with conditions in which fungal infection is a symptom or a side-effect of treatment or which confer an increased risk of fungal infection or patients who are predisposed to or at increased risk of fungal infection, relative to the general population. For example, an anti-Candida recombinant human antibody molecule as described herein may also be useful in the treatment or prevention of fungal infection in cancer patients.


An anti-Candida recombinant human antibody molecule as described herein may be used in a method of treatment of the human or animal body, including prophylactic or preventative treatment (e.g. treatment before the onset of a condition in an individual to reduce the risk of the condition occurring in the individual; delay its onset; or reduce its severity after onset). The method of treatment may comprise administering an anti-Candida recombinant human antibody molecule to an individual in need thereof.


Aspects of the invention provide; an anti-Candida recombinant human antibody molecule as described herein for use in a method of treatment of the human or animal body; an anti-Candida recombinant human antibody molecule as described herein for use in a method of treatment of a fungal infection; the use of an anti-Candida recombinant human antibody molecule as described herein in the manufacture of a medicament for the treatment of a fungal infection; and a method of treatment of a fungal infection comprising administering an anti-Candida recombinant human antibody molecule as described herein to an individual in need thereof.


Pharmaceutical Compositions and Dosage Regimens

Anti-Candida recombinant human antibody molecules may be comprised in pharmaceutical compositions with a pharmaceutically acceptable excipient.


A pharmaceutically acceptable excipient may be a compound or a combination of compounds entering into a pharmaceutical composition which does not provoke secondary reactions and which allows, for example, facilitation of the administration of the anti-Candida recombinant human antibody molecule, an increase in its lifespan and/or in its efficacy in the body or an increase in its solubility in solution. These pharmaceutically acceptable vehicles are well known and will be adapted by the person skilled in the art as a function of the mode of administration of the anti-Candida recombinant human antibody molecule.


In some embodiments, anti-Candida recombinant human antibody molecules may be provided in a lyophilised form for reconstitution prior to administration. For example, lyophilised antibody molecules may be re-constituted in sterile water and mixed with saline prior to administration to an individual.


Anti-Candida recombinant human antibody molecules will usually be administered in the form of a pharmaceutical composition, which may comprise at least one component in addition to the antibody molecule. Thus pharmaceutical compositions may comprise, in addition to the anti-Candida recombinant human antibody molecule, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the anti-Candida recombinant human antibody molecule. The precise nature of the carrier or other material will depend on the route of administration, which may be by bolus, infusion, injection or any other suitable route, as discussed below.


For parenteral, for example sub-cutaneous or intra-venous administration, e.g. by injection, the pharmaceutical composition comprising the anti-Candida recombinant human antibody molecule may be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles, such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection. Preservatives, stabilisers, buffers, antioxidants and/or other additives may be employed as required including buffers such as phosphate, citrate and other organic acids; antioxidants, such as ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens, such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3′-pentanol; and m-cresol); low molecular weight polypeptides; proteins, such as serum albumin, gelatin or immunoglobulins; hydrophilic polymers, such as polyvinylpyrrolidone; amino acids, such as glycine, glutamine, asparagines, histidine, arginine, or lysine; monosaccharides, disaccharides and other carbohydrates including glucose, mannose or dextrins; chelating agents, such as EDTA; sugars, such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions, such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants, such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG).


Administration is normally in a “therapeutically effective amount”, this being sufficient to show benefit to a patient. Such benefit may be at least amelioration of at least one symptom. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the composition, the method of administration, the scheduling of administration and other factors known to medical practitioners. Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors and may depend on the severity of the symptoms and/or progression of a disease being treated. Appropriate doses of antibody molecules are well known in the art (Ledermann, J. A., Begent, R. H., Massof, C., Kelly, A. M., Adam, T. and Bagshawe, K. D. (1991). A phase-I study of repeated therapy with radiolabelled antibody to carcinoembryonic antigen using intermittent or continuous administration of cyclosporin A to suppress the immune response. Int. J. Cancer 47:659-664). Specific dosages may be indicated herein or in the Physician's Desk Reference (2003) as appropriate for the type of medicament being administered may be used. A therapeutically effective amount or suitable dose of an antibody molecule may be determined by comparing it's in vitro activity and in vivo activity in an animal model. Methods for extrapolation of effective dosages in mice and other test animals to humans are known. The precise dose will depend upon a number of factors, including whether the antibody is for prevention or for treatment, the size and location of the area to be treated, the precise nature of the antibody (e.g. whole antibody, fragment) and the nature of any detectable label or other molecule attached to the antibody.


A typical antibody dose will be in the range 100 μg to 1 g for systemic applications, and 1 μg to 1 mg for topical applications. An initial higher loading dose, followed by one or more lower doses, may be administered.


Typically, the antibody will be a whole antibody, e.g. the IgG1 isotype, and where a whole antibody is used, dosages at the lower end of the ranges described herein may be preferred. This is a dose for a single treatment of an adult patient, which may be proportionally adjusted for children and infants, and also adjusted for other antibody formats in proportion to molecular weight.


Preferably the antibody or fragment will be dosed at no more than 50 mg/kg or no more than 100 mg/kg in a human patient, for example between 1 and 50, e.g. 5 to 40, 10 to 30, 10 to 20 mg/kg.


Treatments may be repeated at daily, twice-weekly, weekly or monthly intervals, at the discretion of the physician. The treatment schedule for an individual may be dependent on the pharmocokinetic and pharmacodynamic properties of the antibody composition, the route of administration and the nature of the condition being treated.


Treatment may be periodic, and the period between administrations may be about two weeks or more, e.g. about three weeks or more, about four weeks or more, about once a month or more, about five weeks or more, or about six weeks or more. For example, treatment may be every two to four weeks or every four to eight weeks. Treatment may be given before, and/or after surgery, and/or may be administered or applied directly at the anatomical site of surgical treatment or invasive procedure. Suitable formulations and routes of administration are described above.


In some embodiments, anti-Candida recombinant human antibody molecules as described herein may be administered as sub-cutaneous injections. Sub-cutaneous injections may be administered using an auto-injector, for example for long term prophylaxis/treatment.


In some preferred embodiments, the therapeutic effect of the anti-Candida recombinant human antibody molecule may persist for several half-lives, depending on the dose. For example, the therapeutic effect of a single dose of anti-Candida recombinant human antibody molecule may persist in an individual for 1 month or more, 2 months or more, 3 months or more, 4 months or more, 5 months or more, or 6 months or more.


Combination Immunotherapy

It will be understood that the term “treatment” as used herein includes combination treatments and therapies, in which two or more treatments, therapies, or agents are combined, for example, sequentially or simultaneously.


The agents (i.e. the anti-Candida recombinant human antibody molecules described herein, plus one or more other agents) may be administered simultaneously or sequentially, and may be administered in individually varying dose schedules and via different routes. For example, when administered sequentially, the agents can be administered at closely spaced intervals (e.g., over a period of 5-10 minutes) or at longer intervals (e.g. 1, 2, 3, 4 or more hours apart, or even longer periods apart where required), the precise dosage regimen being commensurate with the properties of the therapeutic agent(s) as described herein, including their synergistic effect.


The agents (i.e. the anti-Candida recombinant human antibody molecules described here, plus one or more other agents) may be formulated together in a single dosage form, or alternatively, the individual agents may be formulated separately and presented together in the form of a kit, optionally with instructions for their use.


For example, the compounds described herein may in any aspect and embodiment also be used in combination therapies, e.g. in conjunction with other agents e.g. antifungal agents. The second antifungal agent may be selected from an azole (e.g. fluconazole), a polyene (e.g. amphotericin B), a echinocandin (e.g. caspofungin), an allylamine (e.g. terbinafine), and a flucytosine (also called 5-fluorocytosine). The skilled person will recognise that other antifungal agents may also be used. In some embodiments, the second antifungal agent is a second anti-fungal antibody or an antimicrobial peptide. In some embodiments, the anti-Candida recombinant human antibody molecule described herein is conjugated to the second antifungal agent.


Preparation of Other Therapeutic Moieties

The anti-Candida recombinant human antibody molecules described herein may be utilised to isolate and identify protective antigens for development as fungal vaccines, or prepare or identify other therapeutic moieties.


For example the antigens bound by the anti-whole cell mAbs described herein may be identified by methods known to those skilled in the art. For example they could be screened against protein and carbohydrate mutants to identify those mutants where binding is reduced. Alternatively antigens can be identified more directly by a proteomics-based approach, for example using 2D electrophoresis and immunoblotting, followed by analysis of spots by trypsinization and mass-spectroscopy (see e.g. Silva et al. Mol Biochem Parasitol. 2013 April;188(2):109-15.). Such antigens will have utility as potential vaccines.


Anti-idiotype anytibodies can be prepared to the antibodies described herein using methods well known to those in the art (see Polonelli, L et al. “Monoclonal Yeast Killer Toxin-like Candidacidal Anti-Idiotypic Antibodies.” Clinical and Diagnostic Laboratory Immunology 4.2 (1997): 142-146; also U.S. Pat. No. 5,233,024).


Detection and Diagnosis

Anti-Candida recombinant human antibody molecules as described herein may also be useful in in vitro testing, for example in the detection of fungus or a fungal infection, for example in a sample obtained from a patient.


Anti-Candida recombinant human antibody molecules as described herein may be useful for identifying C. albicans, and/or distinguishing C. albicans from other fungi.


The presence or absence of a fungus (e.g. C. albicans) may be detected by


(i) contacting a sample suspected of containing the fungus with an antibody molecule described herein, and


(ii) determining whether the antibody molecule binds to the sample, wherein binding of the antibody molecule to the sample indicates the presence of the fungus.


A fungal infection, e.g. C. albicans infection, in an individual may be diagnosed by


(i) obtaining a sample from the individual;


(ii) contacting the sample with an antibody molecule as described herein, and


(iii) determining whether the antibody molecule binds to the sample, wherein binding of the antibody molecule to the sample indicates the presence of the fungal infection.


Binding of antibodies to a sample may be determined using any of a variety of techniques known in the art, for example ELISA, immunocytochemistry, immunoprecipitation, affinity chromatography, and biochemical or cell-based assays. In some embodiments, the antibody is conjugated to a detectable label or a radioisotope.


Lateral Flow Devices

The invention also provides rapid and highly specific diagnostic tests for detecting fungal pathogens, for example multiple fungal pathogens, in a single test


Preferred tests detect not only C. albicans, but also one or more other major fungal pathogens e.g. Aspergillus fumigatus and Cryptococcus neoformans and Pneumocystis jirovecii. Other fungal pathogens which it may be desirable to detect include zygomycete fungi and skin dermatophytic (ringworm) fungi. Antibody molecules specific for these other pathogens may be provided in the light of the disclosure herein, for example.


Preferably the test is in the form of a lateral flow device (LFD). Such LFDs are particularly suitable for use as point-of-care fungal diagnostics.


A lateral flow assay device for the analysis of body fluid will comprise at its most basic:


(i) a housing, and


(ii) a flow path.


The devices, systems and methods described herein are for measuring analyte levels in body fluids of animals, particularly mammals including humans, or in environmental samples e.g. where it is believed fungal pathogens may exist.


As used anywhere herein, unless context demands otherwise, the term ‘body fluid’ may be taken to mean any fluid found in the body of which a sample can be taken for analysis. Examples of body fluids suitable for use in the present invention include, but are not limited to blood, urine, sweat and saliva. Preferably, the body fluid is blood. The fluid may be diluted by a pre-determined amount prior to assay, and any quantification indicator on the LFD may reflect that pre-determined dilution.


Some aspects of the LFD will now be discussed in more detail:


Flow Path of LFD

The flow path (e.g. a chromatographic strip) is preferably provided by a carrier, through which the test substance or body fluid can flow by capillary action. In one embodiment, the carrier is a porous carrier, for example a nitrocellulose or nylon membrane.


In a further embodiment, sections or all of the carrier may be non-porous. For example, the non-porous carrier may comprise areas of perpendicular projections (micropillars) around which lateral capillary flow is achieved, as described in for example WO2003/103835, WO2005/089082 and WO2006/137785, incorporated herein by reference.


The flow path will typically have an analyte-detection zone comprising a conjugate release zone and a detection zone where a visible signal reveals the presence (or absence) of the analyte of interest. The test substance can be introduced into the LFD and flows through to the detection zone.


Preferably the carrier material is in the form of a strip, sheet or similar to the material described in WO2006/137785 to which the reagents are applied in spatially distinct zones. The body fluid sample is allowed to permeate through the sheet, strip or other material from one side or end to another.


Analyte Detection Methods

Analyte detection may be based on competitive or sandwich (non-competitive) assays. Such assays may be used to detect analytes (antigens) from C. albicans, plus optionally one or more other major fungal pathogens e.g. from Aspergillus fumigatus, Cryptococcus neoformans and/or Pneumocystsis jirovecii. Other targets include zygomycete fungi and skin dermatophytic fungi.


The conjugate release zone may contain freely mobile antibodies to the analyte of interest. Alternatively, the conjugate release zone may comprise reagents for carrying out a particular assay to enable detection of the analyte, as described herein.


The binding partners may be attached to a mobile and visible label. A “label” is a composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Useful labels in the present invention include magnetic beads (e.g., Dynabeads™), fluorescent dyes, radiolabels, enzymes, and colorimetric labels such as colloidal gold, silver, selenium, or other metals, or coloured glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads. Preferred is a gold colloid or latex bead.


If the analyte is present in the sample, it will bind to the labelled binding partners. In preferred embodiments the intensity of the colour may be directly proportional to the amount of analyte. Here the detection zone comprises permanently immobilised unlabelled specific binding reagent for the same analyte. The relative positioning of the labelled binding partner and detection zone being such that a body fluid sample applied to the device can pick up labelled binding partner and thereafter permeate into the detection zone. The amount of bound label can be detected as a visible signal in the detection zone.


The label in the LFD will be quantifiable by conventional means or as described herein.


In one competitive format embodiment, the detection zone contains regions of immobile analyte-protein derivatives. These bind and immobilise any of the labelled binding partners not already bound by the analyte in the sample, producing a coloured line or stripe. In this case the amount of label bound in the detection zone (and hence the intensity of the coloured stripe) will be inversely proportional to the amount of analyte in the sample.


In another competitive format, a labelled analyte or analyte analogue may alternatively be provided and this is detected using immobilized specific binding partner (e. g. immobilized antibody specific for the analyte) in the detection zone.


In another competitive format, a labelled analyte or analyte analogue is provided along with a specific binding partner (e.g. an antibody specific for the analyte). The resulting mixture is conveyed to the detection zone presenting immobilized binding partner of the analyte or analyte analogue. The higher the amount of analyte in the sample, the higher the amount of free labelled analyte which leaves the conjugate release zone to be detected in the detection zone.


Control Zone

Preferably the LFD for use with the present invention contains a control zone, which may be located after the detection zone in the direction of sample flow, in which excess labelled binding partner binds to produce a visible signal showing that the test has been successfully run.


Alternatively or additionally, a control zone may be located before the detection zone in the direction of sample flow, indicating that enough sample has been collected to allow operation of the test.


In one embodiment, the control zone is used as a reference point for a reader (see below).


Multiplex Devices

In various aspects of the invention, the LFD may be capable of detecting two (or more) different analytes e.g. analytes (antigens) from C. albicans, plus optionally from one or more or all of Aspergillus fumigatus, Cryptococcus neoformans and/or Pneumocystsis jirovecii. Other targets include zygomycete fungi and skin dermatophytic fungi.


A number of multiplex formats are known in LFDs.


For example, the flow path may comprise two or more carriers. The carriers may be positioned along the flow path consecutively. In use, body fluid would flow along each carrier sequentially.


In a further embodiment, two or more carriers may be positioned in the flow path in parallel. In use, body fluid would flow along each carrier simultaneously.


In one embodiment, two analytes are analysed using two distinct flow path e.g. the housing of the LFD houses the two flow paths.


In one embodiment, the analyte-detecting means may comprise a first binding reagent that specifically binds the analyte and a second binding reagent that specifically binds the analyte, wherein the first binding reagent is labelled and is movable through a carrier under the influence of a liquid by capillary flow and the second binding reagent is immobilised at a detection site in the flow path. The analyte-detecting means comprises a labelled, mobile antibody, specific for the analyte and an immobilised unlabelled antibody, specific for the analyte.


In one embodiment, the analyte-detecting means for each analyte may be positioned together on the carrier, but the specific analyte-binding reagent for each different analyte may comprise a different label. The different labels will be capable of being distinguished as described herein or by conventional means.


Alternatively, the analyte-detecting means for each analyte may be spatially distinct. The flow path in the ‘multiplexed’ LFD may incorporate two or more discrete carriers of porous or non-porous solid phase material, e.g. each carrying mobile and immobilised reagents. These discrete bodies can be arranged in parallel, for example, such that a single application of body fluid sample to the device initiates sample flow in the discrete bodies simultaneously. The separate analytical results that can be determined in this way can be used as control results, or if different reagents are used on the different carriers, the simultaneous determination of a plurality of analytes in a single sample can be made. Alternatively, multiple samples can be applied individually to an array of carriers and analysed simultaneously.


Preferably, multiple analyte detection zones may be applied as lines spanning or substantially spanning the width of a test strip or sheet, preferably followed or preceded by one or more control zones in the direction of body fluid travel. However, multiple analyte detection zones may also, for example, be provided as spots, preferably as a series of discrete spots across the width of a test strip or sheet at the same height. In this case, a one or more control zones may again be provided after or before the analyte detection zones in the direction of body fluid travel.


Detection Systems

The presence or intensity of the signal in the detection zone may be determined by eye, optionally by comparison to a reference chart or card.


Where the intensity of the signal in the detection zone is to be converted to a quantitative reading of the concentration of analyte in the sample it will be preferred that the LFD can be used in conjunction with a screening device (‘reader’). The reader is preferably a handheld electronic device into which the LFD cartridge can be inserted after the sample has been applied.


The reader comprises a light source such as an LED, light from which illuminates the LFD membrane. The reflected image of the membrane may be detected and digitised, then analysed by a CPU and converted to a result which can be displayed on an LCD screen or other display technology (or output via a conventional interface to further storage or analytical means). A light-dependent resistor, phototransistor, photodiode, CCD or other photo sensor may be used to measure the amount of reflected light. The result may be displayed as positive or negative for a particular analyte of interest or, preferably, the concentration of the particular analyte may be displayed. More specifically the conventional reader comprises: illuminating means for illuminating an immunoassay test; photosensitive detector means for detecting the intensity of light from the illuminating means which is reflected from the immunoassay test; means, coupled to the output of the photosensitive detector means, for representing the intensity of the detected light by a data array; memory means for storing preset data; first data processing means, coupled to the memory means and to the output of the means for representing the intensity of the detected light by a data array, for segmenting the data array according to the preset data into control data, background data and test data; second data processing means, coupled to the first data processing means, for determining whether the test data exhibits a statistically significant result; and output means, coupled to the output of the second data processing means, for outputting the results from the second data processing means.


In embodiments of the present invention where multiple analytes are assessed, the reader may analyse the results to detect a plurality of spatially distinct detection or test zones pertaining to different analytes. The photosensitive detector means (e.g. light dependent resistor, phototransistor, photodiode, CCD or other light sensor) will therefore detect reflected light from all of these (optionally scanning them) and generate a discrete or segmented data stream for each zone. Respective control zonal data and background zonal data may also be gathered for the different analytes.


The colour of the LED or other source may vary dependent on the label or method of detecting the analyte.


For gold-labelled analytes, a white LED may be preferable, and therefore a reader may comprise both a red and white LED.


Unless stated otherwise, or clear from the context, antibody residues, where numbered herein, are numbered in accordance with the Kabat numbering scheme.


“and/or” where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. For example “A and/or B” is to be taken as specific disclosure of each of (i) A, (ii) B and (iii) A and B, just as if each is set out individually herein.


Any sub-titles herein are included for convenience only, and are not to be construed as limiting the disclosure in any way.


The invention will now be further described with reference to the following non-limiting Figures and Examples. Other embodiments of the invention will occur to those skilled in the art in the light of these.


The disclosure of all references cited herein, inasmuch as it may be used by those skilled in the art to carry out the invention, is hereby specifically incorporated herein by cross-reference.





FIGURES


FIG. 1—Workflow for the generation of human monoclonal antibodies from single B cells. Class-switched memory B cells were isolated from individuals and microcultured in activating media to promote IgG secretion for screening against target antigens. VH and VL genes from B cells positive for the target were amplified and cloned into a mammalian expression vector for expression and purification via fast protein liquid chromatography. Following QC, recombinant mAbs were assessed for functional activity in vitro and in vivo. Adapted from Huang et al. 2013 (38).



FIG. 2—Representative images from the process employed to generate fully human anti-Candida mAbs. FIG. 2A shows the ELISA screening of purified donor circulating IgG against the target antigens C. albicans ‘whole cell’ yeast and hyphae, and purified Hyr1 protein, to select the donors to take forward for B cell isolation. FIGS. 2B and 2C are representative agarose gel images following RT-PCR and nested PCR of VH and Vk-Ck genes respectively. FIGS. 2D and 2E are analytical mass spectrometry and analytical SEC traces of one of the purified recombinant IgG1 mAbs. Further quality control was carried out by SDS-PAGE gel analysis under non-reducing and reducing conditions as shown in FIGS. 2F and 2G.



FIG. 3—Concentration response curves showing anti-Candida mAbs binding to target antigens. FIG. 3A shows purified anti-Hyr1 mAbs binding to purified recombinant Hyr1 protein in a concentration-dependent manner via ELISA. Binding of purified anti-‘whole cell’ Candida mAbs to C. albicans yeast (FIGS. 3B and 3C) and hyphal cells (FIGS. 3D and 3E) via ELISA are also shown. Values represent mean ±SEM (n=2-4).



FIG. 4—Indirect immunofluorescence of AB120 binding to Hyr1 protein expressed on C. albicans hyphal cells. Indirect immunofluorescence with anti-Hyr1 mAb AB120 against WT CAl4-Clp10 (A), Hyr1 null mutant (B) and a Hyr1 re-integrant strain (C). A fluorescently conjugated secondary goat anti-human IgG antibody was used to detect anti-Hyr1 mAb binding. Scale bars represent 15 μm.



FIG. 5—Indirect immunofluorescence of anti-whole cell mAbs binding to WT CAl4-Clp10. Indirect immunofluorescence demonstrating the distinct binding patterns of the panel of anti-Candida mAbs. Shown are representative images of mAbs binding strongly to targets expressed on both CAl4-Clp10 yeast and hyphae (A), mAbs binding primarily to targets expressed on hyphae but with some binding to yeast (B), mAbs binding specifically to hyphae (C) and mAbs binding to yeast and the growing hyphal tip (D). A fluorescently conjugated secondary goat anti-human IgG antibody was used to detect anti-Candida mAb binding. Scale bars represent 19 μm.



FIG. 6—Heat-map of anti-Candida mAbs binding to Candida species and other pathogenic fungi. Immunofluorescence microscopy analysis of (a) anti-Hyr1 mAbs (AB120-AB123) and (b) cell wall mAbs (AB118-AB140) binding to C. albicans and other clinically relevant fungal species depicted in a heat map. Binding was graded from red (high) to yellow (none).



FIG. 7—Macrophage uptake of live C. albicans cells pre-incubated with saline, isotype control mAb or anti-Candida mAb. FIG. 7A shows the time at which an uptake event occurred over the first 90 min of the assay following C. albicans pre-incubation with saline, an IgG1 control antibody, an anti-whole cell mAb (AB118, AB119 and AB140) or an anti-Hyr1 mAb (AB120). FIG. 7B shows the morphology of C. albicans cells during each uptake event over the first 90 min of the assay following C. albicans pre-incubation with saline, an IgG1 control antibody, an anti-whole cell mAb (AB118, AB119 and AB140) or an anti-Hyr1 mAb (AB120). An uptake event was defined as the complete engulfment of a C. albicans cell by a macrophage. Bars represent percentage of uptake events±SEM (n=3). *p<0.05, **p<0.01, ****p<0.0001.



FIG. 8—Macrophage engulfment of live C. albicans cells pre-incubated with saline, isotype control mAb or anti-Candida mAb. FIGS. 8A-8C are snapshots taken from live cell video microscopy capturing the stages of C. albicans engulfment by J774.1 macrophages. FIG. 8A shows the macrophage (red, *) and C. albicans (green) prior to cell-cell contact, FIG. 8B shows the cells once cell-cell contact has been established and FIG. 8C shows the C. albicans within the phagocyte following ingestion. FIG. 8D shows the average time taken for a macrophage to engulf a live C. albicans cell following pre-incubation with saline, an IgG1 control antibody, an anti-whole cell mAb (AB118, AB119 and AB140) or an anti-Hyr1 mAb (AB120). FIG. 8E shows the time taken for a macrophage to ingest a filamentous C. albicans cell following pre-incubation of AB120 with hyphal C. albicans. Rate of engulfment was defined as the time taken from cell-cell contact to complete ingestion of the C. albicans cell inside the macrophage resulting in a loss of green fluorescence. Bars represent average time taken for a macrophage to ingest a C. albicans cell±SEM (n=3) *p<0.01, ***p<0.005.



FIG. 9—Macrophage engulfment of opsonized live C. albicans cells in the presence and absence of an FcγR blocker. The average time taken for a macrophage to ingest a live C. albicans cell following pre-incubation with saline or an anti-whole cell mAb (AB140) in the presence or absence of an FcγR block. Bars represent average time taken for a macrophage to ingest a C. albicans cell±SEM (n=3) *p<0.05.



FIG. 10—Macrophage migration towards C. albicans cells following pre-incubation with saline, an isotype control mAb or anti-Candida mAb. FIG. 10A shows mean velocity of macrophages as they migrate towards C. albicans cells following pre-incubation with saline, an IgG1 control mAb, or an anti-whole cell mAb (AB140). Bars represent macrophage mean track velocity±SEM (n=3). FIG. 10B shows average distance travelled by a macrophage to engulf a C. albicans cell following pre-incubation with saline, an IgG1 control mAb, or an anti-whole cell mAb (AB140). Bars represent average distance travelled±SEM (n=3). FIGS. 10C, 10D and 10E are tracking diagrams representing macrophage migration towards C. albicans cells pre-incubated with saline (blue), AB140 (pink) or IgG1 control mAb (green). Tracks represent the movement of individual macrophages relative to their starting position, up until the first uptake event. *p<0.05, **p<0.01, ***p<0.005.



FIG. 11—Assessment of anti-Candida mAbs in an in vivo model of disseminated candidiasis. C. albicans SC5314 was pre-incubated with saline, IgG1 control, anti-whole cell mAb (AB119) or anti-Hyr1 mAb (AB120) and then injected iv into the tail vein of BALB/c mice (n=6 per group). Kidney fungal burdens from each group were determined on day 3 post infection (FIG. 11A) and combined with the change in animal weight during the course of the infection to give an overall outcome score for disease progression (FIG. 11B). Dots represent individual animals; horizontal lines represent mean, *p<0.05, **p<0.01.



FIG. 12—Schematic of VH, Vκ-Cκ and Vλ-Cλ cloning into pTT5 expression vector. B cells positive for antigen binding in the initial ELISA screen were lysed. mRNA in B cell lysate was used as a template for VH, Vκ-Cκ and Vλ-Cλ gene amplification via RT-PCR. RT-PCR was carried out using forward primers specific to human V domain leader sequences and reverse primers specific for human IgCH1, Cκ or Cλ regions or light chain UTR. To increase the specificity of gene amplification, nested PCR was carried out using RT-PCR products as the template. Forward primers specific for human VH FW1 sequences and reverse primers specific for human VH FW4 sequences were used to amplify VH genes. To capture Vκ-Cκ and Vλ-Cλ genes, forward primers specific to human Vκ and human Vλ FW1 sequences were used in combination with reverse primers specific to the 3′ end of the human Cκ or human Cλ regions. Primers used in nested PCR reactions contained 15 bp extensions which were complementary to the pTT5 expression vector to facilitate downstream Infusion cloning. Amplification of VH, Vκ-Cκ and Vλ-Cλ genes were done in separate reactions. RT-PCR—reverse transcriptase polymerase chain reaction; UTR untranslated region; L—leader sequence; VH—heavy chain variable domain; Vκ—kappa chain variable domain; Vλ—lambda chain variable domain; CH—heavy chain constant domain; Cκ—kappa chain constant domain; Cλ—lambda chain constant domain.



FIG. 13—Concentration response curves of purified anti-Hyr1 mAbs screened for binding to unrelated proteins. (a, b) Purified anti-Hyr1 mAbs screened against HSA and HEK NA respectively via ELISA. Values represent mean (n=2-4).



FIG. 14—Concentration response curves showing anti-whole cell mAbs screened for binding to unrelated proteins. (a, b) Purified cell wall mAbs screened against HSA. (c, d) the same mAbs screened against HEK NA via ELISA. Values represent mean (n=2-4).



FIG. 15—Indirect immunofluorescence of mAbs binding to WT CAl4-Clp10 before and after enzymatic modification of the cell wall. Proteinase K treatment was used to reduce protein residues; Zymolyase 20T enzyme was used to digest β-1,3-glucans; Endoglycosidase H treatment reduced N-linked glycans on the CAl4-Clp10 cell wall. Decrease in indirect immunofluorescence after enzymatic treatments suggested the nature of the mAb epitopes. A fluorescently conjugated secondary goat anti-human IgG antibody was used to detect anti-Candida mAb binding. Scale bars represent 4 μm.



FIG. 16—Human monocyte-derived macrophage phagocytosis of live C. albicans cells pre-incubated with saline, isotype control mAb or anti-Candida mAb . (a) Time at which an uptake event occurred over the first 90 min of the assay following C. albicans pre-incubation with saline, an IgG1 control antibody, an anti-whole cell reactive mAb (AB119 and AB140) or an anti-Hyr1 mAb (AB120). Bars represent percentage of uptake events (n=2). (b) Percentage of these uptake events that occurred within the first 30 min of the assay. Dots represent average from individual experiments, line represents average (n=2) and (c) average time taken for a macrophage to engulf a live C. albicans cell following pre-incubation with saline, an IgG1 control antibody, an anti-whole cell mAb (AB119 and AB140) or an anti-Hyr1 mAb (AB120) at a MOI of 3 (n=2).



FIG. 17—Counterimmunoelectrophoresis of anti-Candida mAbs with C. albicans. Purified anti-Candida mAbs AB119, AB140 and AB118C101S reacted with yeast supernatant antigenic preparation and a crude yeast extract (a). AB119, AB140, AB118C101S and AB135 reacted with hyphal supernatant antigenic preparation and a crude hyphal extract (b).



FIG. 18—Immunogold localization of anti-Candida mAbs to the cell wall of C. albicans yeast (top panel) and hyphal (bottom panel) cell walls.





EXAMPLE 1
Generation of Fully Human Anti-Candida mAbs by Single B Cell Cloning

The generation of recombinant mAbs through direct amplification of VH and VL genes from single B cells produces fully human, affinity matured mAbs with the native antibody heavy and light chain pairing intact (14). We employed this technology to generate human recombinant anti-Candida mAbs to a defined C. albicans antigen—the morphogenesis-regulated protein 1 (Hyr1) protein expressed only in the hyphal cell wall (40), and to C. albicans whole cell wall preparations. Hyr1 protein was selected based on its role in proposed role in resisting phagocyte killing and pre-clinical data demonstrating that a recombinant N-terminal fragment of Hyr1 confers protection in a murine model of disseminated candidiasis (23, 29, 41). Furthermore, because Hyr1 is expressed solely on C. albicans hyphal cells so mAbs generated against this protein would serve as C. albicans-specific markers. In addition we used C. albicans whole cell wall extracts as a target to screen against allows for the isolation of mAbs that bind to an array of different antigens, anticipating that some of the resulting mAbs would be pan fungal and therefore possess a broad spectrum of therapeutic activity and pan-Candida diagnostic specificity.


To enhance the likelihood of isolating Candida-related antibodies, the class switched memory (CSM) B cells used in this study were isolated from the blood of individuals who had recovered from a superficial Candida infection within a year of sampling. Donors were selected from a panel of volunteers and the levels of target-specific circulating IgG in the donor plasma was assessed via ELISA. In this screen, donor 85 demonstrated the greatest IgG activity against C. albicans whole cell and donor 23 had the highest IgG titre against Hyr1 (FIG. 2A). These donors were selected to provide the source of B cells to use for the generation of Candida-specific recombinant antibodies. After the isolation of CSM B cells from a donor, approximately 80000-150000 cells were plated out at 5 cells/well and activated with a cocktail of cytokines and supplements to promote secretion of IgG into the supernatant. A high throughput screening platform was then employed to facilitate the detection of IgG in the B cell supernatant against target antigens by ELISA. Positive ELISA hits enabled identification of wells containing B cells secreting antigen-specific IgG into the supernatant. Typically, approximately 0.05% wells/screen were positive (OD>4xbackground). Non-specific hits were identified and eliminated by performing an ELISA screen against two unrelated proteins—human serum albumin (HSA) and human embryonic kidney nuclear antigen (HEK NA). CSM B cells from wells that were positive for the antigen screen and negative for the unrelated protein screen were then lysed and used as the source for VH, Vκ-Cκ and Vλ-Cλ gene amplification via RT-PCR and nested PCR (FIGS. 2B, C). VH, Vκ-Cκ and Vλ-Cλ genes were sub cloned into the pTT5 mammalian expression vector and the sequences analysed (data not shown). Corresponding heavy and light chains originating from the same hit well were co-transfected into Expi293F cells for small scale whole IgG1 expression. From these co-transfections, recombinant mAbs that demonstrated binding to the original target were selected for large scale recombinant expression. These were then purified via affinity-based FPLC using a protein A resin and quality control checked via analytical mass spectrometry, SDS-PAGE gel analysis and analytical SEC (FIGS. 2D-G).


In total, 18 purified recombinant IgG1 mAbs were generated using the single B cell technology described above. Five of these mAbs bound to purified Hyr1 protein and 13 bound to C. albicans whole cells (Table S3).


EXAMPLE 2
Purified Recombinant Anti-Candida mAbs Exhibit Specific Target Binding

Purified anti-Hyr1 mAbs were primarily assessed for functionality through binding to the purified recombinant N-terminus of Hyr1 protein via ELISA. Four of the five mAbs demonstrated strong binding to the purified antigen with EC50 values of 104 ng/ml, 76.5 ng/ml, 49.6 ng/ml and 53.3 ng/ml for AB120, AB121, AB122 and AB123 (FIG. 3A) respectively. AB124 bound to Hyr1 with a lower affinity with an EC50 value of 1050 ng/ml. To examine the specificity of these mAbs for the target protein, all five were tested against the unrelated antigens HSA and HEK nuclear antigen as negative controls and demonstrated no binding (FIG. 13).


The purified recombinant anti-whole cell mAbs were originally screened and isolated against C. albicans overnight culture. As such, the initial QC of these mAbs was to assess their binding to C. albicans whole cells via ELISA. The majority of purified anti-whole cell mAbs bound C. albicans yeast cells with high affinity with EC50 values ranging from 2.8 to 31.1 ng/ml (FIGS. 3B, C). AB134 and AB135, which have similar amino acid sequences, both demonstrated slightly lower affinity for the target with EC50 values of 1060 and 224 ng/ml respectively (FIG. 3C).


Purified anti-whole cell mAbs exhibited a variety of affinities when binding to C. albicans cells where both yeast and hyphal morphologies were present (FIGS. 3D, E). The majority bound these cells with high affinity with EC50 values ranging between 3 and 50 ng/ml. As observed with C. albicans yeast cell binding, AB134 and AB135 demonstrated slightly lower affinities with EC50 values of 684 and 69.4 ng/ml. EC50 values were used here as a simple comparison to demonstrate the variability in anti-whole cell mAbs binding to C. albicans cell surface antigens. Therefore this methodology generated a panel of mAbs which bound to a variety of specific cell targets. Specificity of the anti-whole cell mAbs for a target C. albicans antigen was assessed through binding to the two unrelated antigens HSA and HEK NA. All mAbs demonstrated no binding to these antigens confirming their specificity for the fungal cells (FIG. 14).


EXAMPLE 3
Purified Recombinant Anti-Candida mAbs Show Distinct Binding Patterns to C. Albicans and Other Fungal Species

The recombinant anti-Hyr1 mAbs generated by single B cell technology were initially isolated by screening against N-terminus of Hyr1 protein and, following purification, demonstrated binding to this recombinant antigen (above). We then visualized binding of these mAbs to Hyr1 protein expressed on the C. albicans cell surface by immunofluorescent staining using a fluorescently labelled secondary anti-human IgG mAb for detection. It was observed that the anti-Hyr1 mAbs bound to the predicted cellular location on the hyphae, and not the WT C. albicans yeast cells grown in different culture conditions (FIG. 4A). We verified that the anti-Hyr1 mAbs did not bind to hyphae of a Δhyr1 null mutant (FIG. 4B) and that binding was restored in a C. albicans strain containing a single reintegrated copy of the deleted HYR1 gene (FIG. 4C).


Next we visualised binding to WT C. albicans for the anti-whole cell mAbs via indirect immunofluorescent staining. The anti-whole cell mAbs demonstrated a range of binding profiles to WT C. albicans (FIG. 5). mAbs AB118, AB119, AB129, AB130, AB133, AB134, AB135, AB139, AB140 bound strongly to both C. albicans yeast and hyphae (FIG. 5A). AB132 bound to both yeast and hyphae but exhibited stronger binding to hyphae (FIG. 5B). AB126 and AB131 appeared to be hypha-specific (FIG. 5C) and AB127 stained the mother yeast cell and the tip of the growing hyphae (FIG. 5D). Therefore the panel of antibodies apparently detected both morphology specific and morphology-independent epitopes.



C. albicans cells were enzymatically treated with proteinase K, endoglycosidase H (endo-H) and zymolyase 20T and assessed for mAb binding. Proteinase K treatment reduced AB120 (anti-Hyr1) but not anti-whole cell mAbs binding to C. albicans confirming that anti-Hyr1 antibody recognised a protein epitope (FIG. 15a). Following zymolyase 20T and endo-H treatments, binding of other anti-whole cell mAbs decreased suggesting that the cognate epitopes might be β-glucan or N-mannan respectively (FIG. 15b, c). Some anti-whole cell mAbs demonstrated increased fluorescence after enzymatic treatment suggesting that their epitopes might be located deeper in the cell wall.


Commensurate with the C. albicans-specific nature of HYR1, anti-Hyr1 mAbs only bound to C. albicans and not to a range of other Candida species (FIG. 6a). In contrast, a range of binding patterns were observed for the binding of anti-whole cell mAbs to other pathogenic fungal species. The majority of mAbs bound strongly to the closely related species C. dubliniensis, C. tropicalis, C. parapsilosis and C. lusitaniae. There was little binding of mAbs to the more distantly related C. glabrata and C. krusei. Only the homologous AB131 and AB132 antibodies demonstrated some weak binding to C. krusei (FIG. 6b).


To assess for pan-fungal binding activity, all the anti-whole cell mAbs were tested against A. fumigatus. C. neoformans, C. gattii, P. carinii, M. circinelloides and M. dermatis but no binding was observed (FIG. 6b). Therefore the anti-Hyr1 mAbs are C. albicans-specific and the anti-whole cell mAbs demonstrate a variety of binding patterns to WT C. albicans and other pathogenic Candida species, indicating that they target a range of different antigens and the expression levels of these antigens varies from species to species.


In conclusion, all purified recombinant mAbs generated by this single B cell technology bound specifically to their target antigens with high affinity. As expected, the anti-whole cell mAbs demonstrated distinct binding patterns to WT C. albicans and other pathogenic fungi, indicating that they target a range of different antigens and the expression levels of these antigens varies from species to species.


EXAMPLE 4
Purified Recombinant Anti-Candida mAbs Opsonise C. Albicans for Phagocytosis by Macrophages

Phagocytic cells of the innate immune system are the first line of defence against fungal pathogens. Antibody binding enhances phagocytic clearance of pathogens. We utilised a live cell phagocytosis assay to examine whether the anti-Candida mAbs generated in this study opsonized C. albicans for phagocytosis by J774.1 macrophages and human monocyte-derived macrophages. The macrophages were challenged with live, C. albicans CAl4-Clp10 which had been pre-incubated with an anti-Candida mAb, an isotype control mAb or saline for 1 h. Live cell video microscopy using our standard phagocytosis assay (42, 43) was employed to determine the degree of opsonisation. No significant difference was observed between the saline control and anti-Candida mAb groups in terms of the overall number of C. albicans cells taken up during the 3 h by macrophages. However, there was a difference in the time by which the majority of uptake events had occurred (FIG. 7A). C. albicans cells that had been pre-incubated with either AB118, AB119 or AB140 (anti-whole cell mAbs) were taken more rapidly compared to the saline control-treated fungal cells, the IgG1 control pre-incubated fungal cells or AB120 pre-incubated fungal cells. The percentage of uptake events occurring by 20 min was 21±10, 54±9, 22±5 and 68±2, 44.3±0.6 and 7±2 (mean ±SD) for saline control, AB118, AB120, AB140, AB119 and isotype control respectively (FIG. 7A). A majority of C. albicans cells pre-incubated with AB118, AB119 or AB140 were taken up as yeast cells and the majority of cells taken up by the saline control group, AB120 group and isotype control group, were hyphal cells (FIG. 7B).


EXAMPLE 5
Macrophages Rapidly Engulf mAb-Bound C. Albicans Cells Through FcγR Binding

Next we used live cell video microscopy and image analysis to examine whether there was any difference in the rate of engulfment between C. albicans cells pre-incubated with saline compared to C. albicans cells pre-incubated with selected anti-Candida mAbs. As shown previously we defined the rate of engulfment as the time taken from establishment of cell-cell contact to the time at which a C. albicans cell had been completely engulfed by a macrophage as indicated by its loss of FITC green fluorescence (42, 43) (FIGS. 8A-C). When C. albicans yeast cells were pre-incubated with AB120 (anti-Hyr1 mAb) there was no difference in the rate of engulfment from the saline control or IgG1 control mAb however, in the presence of either AB118, AB119 or AB140 (anti-whole cell mAbs), fungal cells were engulfed at a significantly faster rate compared to the saline control and IgG1 control mAb, (FIG. 8D). The hypha-specific mAb AB120 stimulated faster macrophage engulfment of C. albicans hyphal cells by macrophages—taking an average of 5.8±0.3 min to engulf opsonised hyphae compared to 8.8±0.8 min for the control (FIG. 8E).


Similar observations were obtained using human monocyte-derived macrophages (FIG. 16).


Blocking FcγRs on the surface of the macrophage decreased the rate of engulfment of AB140-bound C. albicans compared to that of the saline control (FIG. 9) indicating that the increased rate of engulfment of mAb-bound Candida cells is, at least in part, due to uptake through the FcγRs.


EXAMPLE 6
Macrophages Migrate Further, Faster and More Direct Towards Anti-Candida mAb Bound C. Albicans Cells

We showed that antibody-bound C. albicans cells were cleared earlier by macrophages than control cells. To determine the effect of antibody binding on uptake dynamics, we used imaging analysis to digitise the migration of macrophages until their first uptake event, measuring the distance travelled, directionality and velocity of the macrophage towards control or antibody-bound fungal cells. Macrophages travelled further and at a greater velocity towards C. albicans yeast cells that had been pre-incubated with a whole-cell mAb (AB140) compared to control fungal cells or those pre-incubated with IgG1 control mAb (FIG. 10A,B). Furthermore we observed that macrophages moved in a more directional manner towards antibody-bound C. albicans cells compared to control cells or those pre-incubated with IgG1 control mAb (FIGS. 10 C, D and E).


EXAMPLE 7
Anti-Whole Cell mAb Reduces Fungal Burden in a Model of Disseminated Candidiasis

To determine whether the anti-Candida mAbs possessed therapeutic potential in vivo, their action was assessed in a murine model of systemic candidiasis (44). C. albicans SC5314 yeast cells were pre-incubated for 1 h with either saline, an IgG1 isotype control mAb, AB119 (anti-whole cell) or AB120 (anti-Hyr1) before iv injection into the mouse lateral tail vein. Disease progression was monitored by weight change and kidney fungal burdens at day 3 which together generated an overall outcome score for disease progression (44). When SC5314 was pre-incubated with AB120 there was no decrease in fungal burden compared to the saline control or the IgG1 control mAb (FIG. 11A). However, when AB119 was pre-incubated with SC5314, there was a significant decrease in kidney fungal burden compared to the saline control (FIG. 11A, p<0.01). This was also considerably less than the kidney fungal burden for the IgG1 isotype control. By weight change there was no significant difference in disease outcome score between AB120 and the saline control and isotype control (FIG. 11B). However, mice that had been injected with SC5314 pre-incubated with AB119 had a significantly lower disease outcome score than both the saline control group (p<0.01) and the isotype control group (p<0.05) indicating that when AB119 is present, the mice are able to clear infection more quickly and disease progression is limited (FIG. 11B). Therefore exposure to antibody improved the survival of mice in a systemic disease model.


EXAMPLE 8
Discussion of Examples 1-7

Monoclonal antibodies (mAbs) have the potential to be used in multiple fungal therapy and disease management situations. Here we describe and use for the first time a novel technology facilitating the isolation of fully human anti-Candida mAbs against whole cells and a specific cellular target. These mAbs were derived directly from single B cells from donors with a history of mucosal Candida infection and demonstrated distinct binding profiles to C. albicans and other pathogenic fungi, as well as the ability to opsonise fungal cells and to enhance phagocytosis and show partial protection in a murine model of disseminated candidiasis.


mAbs-based agents have been identified as an alternative strategy to complement the medical gaps associated with current antifungal treatments and diagnostics (13, 45, 46). In this study we generated 18 fully human recombinant anti-Candida mAbs through the direct amplification of mRNA isolated from VH and VL antibody genes produced naturally in vivo in response to a Candida infection. By employing this method, the purified, affinity matured recombinant mAbs generated were less likely to be immunogenic, had importantly retained their native antibody heavy and light chain pairings, and therefore are more likely to be of therapeutic benefit (35). IgG1 was selected as the antibody scaffold because this isotype makes up the majority of mAbs in the clinic and so is the best characterised in terms of drug development (47, 48). Thirteen of the mAbs generated bound to C. albicans whole cell and 5 bound to recombinant purified Hyr1 protein—a protein which is considered to be important for C. albicans resistance to phagocytosis and is currently in development as an experimental vaccine (29, 41) demonstrating that this novel technology can be utilised for screening against a wide range of specific antigens.


An antibody that recognises an antigen expressed across different fungal species could be highly beneficial as a pan-fungal therapeutic. At the same time, one of the major contributors to poor prognosis in the clinic is the lack of accurate and timely diagnostics with a knock on delay in appropriate treatment (6, 7, 49). In this case, it would be more beneficial to have a species-specific antibody which recognises an antigen only expressed on one species. As such, we assessed binding of our panel of mAbs to a number of emerging and resistant pathogenic fungi. We observed that anti-Hyr1 mAbs bound solely to C. albicans hyphae, correlating with findings that have reported that Hyr1 is only expressed on C. albicans hyphal cells (29, 40, 50). The binding pattern of anti-whole cell mAbs was more varied with the majority of mAbs binding strongly to the species that are closely related to C. albicans such as the emerging pathogens C. tropicalis and C. parapsilosis (51). As expected, little or no binding was observed to the more evolutionarily distinct species C. glabrata and C. krusei. Altogether this demonstrates that the novel technology employed here can be utilised to generate species-specific as well as pan fungal mAbs, which has great implications in terms of anti-fungal drug discovery and diagnostics. Furthermore, these mAbs could be utilised to isolate and identify protective antigens for development as fungal vaccines.


One of the many ways mAbs exert their protective effects is through opsonizing cells for phagocytosis (15). We have shown previously that by employing live cell imaging we can breakdown this process down into its component parts, thus allowing us to do a more in-depth analysis on the effect of mAbs on the individual stages of phagocytosis (42, 43). Here we observed that when yeast and hyphal cells were coated with an anti-whole cell mAb or a hyphal cell was coated with an anti-Hyr1 mAb, cells were engulfed at a significantly faster rate compared to unopsonized cells, and this was through engagement of the FcγR. Furthermore, macrophages migrated further, faster and in a more direct manner towards opsonized C. albicans cells and this contributed to earlier clearance of fungal cells.


A number of invasive infections occur in the immunocompetent patient population as a consequence of severe trauma, and in these situations opsonizing mAbs could be a viable treatment option. The majority of antibody therapeutics in the clinic are hIgG1 so this isotype has been routinely tested pre-clinically in murine models of disease (47). Furthermore, the literature shows that hIgG1 binds to all activating mFcγRs with a similar profile to the most potent IgG isotype in mice, mIgG2a, validating the use of mouse models to assess Fc-mediated effects of hIgG1 mAbs (47). As such, we utilised an established three-day murine model of disseminated candidiasis (44, 52) to assess the efficacy of anti-Candida mAbs in vivo and observed a significant decrease in kidney fungal burden and overall disease outcome score when C. albicans was pre-incubated with an anti-whole cell mAb.


We have generated fully human antibodies from single B-cells to create reagents that have high specificity for targets with utility in the antifungal diagnostic and therapeutic markets. The antibodies are of high affinity and are and can be synthesised in milligram quantities under defined conditions for heterologous protein expression.


The relative by which these antibodies can be produced means that they could be used singly or in multiplex formats to create novel polyvalent diagnostic tests, as vaccine Candidates or as therapeutic delivery systems to target toxic molecules to specific microbial or cellular targets.


EXAMPLE 9
CIE Analysis


FIG. 17 shows the results of counterimmunoelectrophoresis (CIE) analysis. This shows selected mAbs were able to detect C. albicans antigens in a format commonly used for the diagnosis of patients with a Candida infection.


EXAMPLE 10
TEM Analysis


FIG. 18 shows transmitting electron microscopy (TEM) images illustrating the binding of a select panel (one mAb from each CDR3 amino acid sequence cluster) of the anti-whole cell mAbs to C. albicans yeast and hyphal cell walls via immunogold labelling. The images show that the mAbs are very specific to the cell wall and that there are a variety of binding targets, for example AB126, AB127 and AB131 appear mainly to bind to hypha, whereas AB118C101S, AB119, AB140 and AB135 appear to bind to more abundantly expressed targets in both yeast and hyphal cells.


General Methods

Candida Strains and Growth Conditions


C. albicans serotype A strain CAl4+Clp10 (NGY152) was used as a control and its parent strain CAl4, used to construct the Δhyr1 null mutant C. albicans strain (40) and the hyr1 re-integrant strain (unpublished). The clinical isolates C. albicans SC5314, C. glabrata SC571182B, C. tropicalis AM2005/0546, C. parapsilosis ATCC22019, C. lusitaniae SC5211362H, C. krusei SC571987M, C. dubliniensis CD36 are shown in Table S1. All strains were obtained from glycerol stocks stored at −80° C. and plated onto YPD plates (2% (w/v) mycological peptone (Oxoid, Cambridge, UK), 1% (w/v) yeast extract (Oxoid), 2% (w/v) glucose (Fisher Scientific, Leicestershire, UK) and 2% (w/v) technical agar (Oxoid)). Candida strains tested were routinely grown in YPD (see above without the technical agar) except in the in vivo experiments where strains were grown in NGY medium (0.1% (w/v) Neopeptone (BD Biosciences), 0.4% (w/v) glucose (Fisher Scientific), 0.1% (w/v) yeast extract (Oxoid). Aspergillus fumigatus clinical isolate V05-27 was cultured on Potato Dextrose Agar slants for seven days before the spores were harvested by gentle shaking with sterile 0.1% Tween 20 in PBS. Harvested spores were purified, counted and re-suspended at a concentration of 1×108 spores/ml. Swollen spores were generated by incubation in RPMI media for 4 h at 37° C.



Malassezia dermatis CBS9169 was cultured on Modified Dixon agar (3.6% (w/v) Malt extract (Oxoid), 1% (w/v) Bacto peptone (BD Biosciences), 2% (w/v) Bile salts (Oxoid), 1% (w/v) Tween40 (Sigma), 0.2% (w/v) Glycerol (Acros Organics), 0.2% (w/v) Oleic acid (Fisher Scientific), 1.5% technical Agar (Oxoid)) supplemented with chloramphenicol (0.05% (w/v) Sigma) and cycloheximide (0.05% (w/v) Sigma)) . Overnight culture of M. dermatis was grown in Modified Dixon Medium. Mucor circinelloides CBS277.49 was grown on Potato Dextrose Agar for 7 days before spores were harvested in PBS and filtered through 40 μm Nylon Cell Strainer (BD Biosciences). Cryptococcus neoformans KN99α and Cryptococcus gattii R265 were grown in YPD overnight, washed in PBS and 1×107 cells were added to 6 ml RPMI+10% FCS in 6 well-plates. Plates were incubated at 37° C.+5% CO2 for 5 days to induce capsule formation. Harvested cells were washed in PBS. Rat lung tissue isolates of Pneumocystis carinii M167-6 were washed in PBS and immunostained.


Generation of Recombinant Hyr1 N-Protein

The recombinant N-terminus of the Hyr1 protein (amino acids 63 to 350—Table S2) incorporating an N-terminal 6xHis tag was expressed in HEK293F cells and purified by nickel-based affinity chromatography using a nickel NTA superflow column (QIAGEN, USA). Fractions containing the recombinant N-terminus of the Hyr1 protein were pooled and further purified via Analytical Superdex 200 gel filtration chromatography (GE Healthcare, USA) in PBS. QC of the recombinant protein via SDS-PAGE gel analysis, analytical size exclusion chromatography (SEC) and Western blot (using an anti-His antibody for detection) confirmed a protein of 32 kDa (data not shown).


Identification of Human Anti-Hyri and Anti-Whole Cell mAbs From Donor B Cells PBMC Isolation


In brief, peripheral venous blood from donors who had recovered from a Candida infection within the last year was collected in EDTA-coated vacutainers tubes and pooled. PBMCs and plasma were separated from the whole blood suspension via density gradient separation using Accuspin System-Histopaque-1077 kits (Sigma-Aldrich) according to manufacturer's instructions. Following separation, the plasma layer was aspirated and stored at 4° C. for later analysis of antibody titre and the PBMC layer was aspirated and washed in PBS and centrifugation at 250×g for 10 min three times before final resuspension at a concentration of 1×107 cells/ml in R10 media (RPMI 1640 (Gibco, Life Technologies), 10% FCS, 1 mM sodium pyruvate (Sigma), 10 mM HEPES (Gibco, Life Technologies), 4 mM L-glutamine (Sigma), 1× penicillin/streptomycin (Sigma)) containing additional 10% FCS and 10% DMSO. PBMCs were split into 1 ml aliquots and stored in liquid nitrogen until they were required.


Purification of Donor Plasma

IgG was purified from donor plasma using VivaPure MaxiPrepG Spin columns (Sartorius Stedman) according to manufacturer's instructions. In brief, plasma sample was applied to the spin column to facilitate IgG binding. The column was washed twice in PBS and then bound IgG was eluted in an amine buffer, pH 2.5 and neutralized with 1 M Tris buffer, pH8. Eluted IgG concentration was measured by absorbance at 280 nm using a NanoVue Plus Spectrophotometer (GE Healthcare).


Circulating IgG Enzyme-Linked Immunosorbent Assay (ELISA) To Identify Donors With B Cells To Take Forward

To identify the donor to use for subsequent class switched memory (CSM) B cell isolation and activation, ELISAs were carried out against the target antigens using IgG purified from donor plasma. NUNC maxisorp 384-well plates (Sigma) were coated with C. albicans overnight culture (whole cell) or 1 μg/ml purified, recombinant N-terminus hyr1 protein antigen in 1×PBS and incubated at 4° C. overnight. The next day, wells were washed three times with wash buffer (1×PBS+0.05% Tween) using a Zoom Microplate Washer (Titertek). Wells were then blocked with block buffer (1×PBS+0.05% Tween+0.5% BSA) for 1 h at room temperature with gentle shaking to inhibit non-specific binding. After three washes (as above), titrated purified IgG or IVIG in block buffer was added in duplicate, and the plates were incubated for 2 h at room temperature with gentle shaking. Wells were washed with wash buffer as above before addition of goat anti-human IgG, HRP conjugated (ThermoScientific) secondary antibody at 1:5000 dilution in blocking buffer. Plates were incubated for 45 min at room temperature with gentle shaking. To develop the ELISA, wells were washed three times with wash buffer (as above) before the addition of TMB (Thermo Scientific). Plates were incubated at room temperature for 5 min to allow the blue colour to develop and the reaction was quenched by the addition of 0.18 M sulphuric acid. The plates were then read at an OD of 450 nm on an Envision plate reader (PerkinElmer). Labstats software in Microsoft Excel was used to generate concentration-response curves for EC50 determination and donor selection for subsequent CSM B cell isolation and activation.


Isolation of Class Switched Memory B Cells

The PBMCs from donors who displayed a strong IgG response to the antigen of interest in the screening ELISA were taken forward for CSM B cell isolation and activation. The process of generating recombinant mAbs from a single donor's B cells to one particular antigen, beginning with the isolation of CSM B cells all the way through to expression and purification of recombinant mAbs, was termed an ‘Activation’. For each Activation, 5×107 PBMCs were removed from the liquid nitrogen store and thawed by adding pre-warmed R10 media drop wise to the cells. The diluted cell suspension was then transferred into a fresh polypropylene tube containing pre-warmed R10, resulting in a final cell dilution of approximately 1:10. Benzonase nuclease HC, purity>99% (Novagen) was added at a 1:10000 dilution (to ensure any lysed cells and their components didn't interfere with the live cells), and the cells were centrifuged at 300×g for 10 min at room temperature and the supernatant removed. PBMCs were then washed again in R10 before final resuspension in 1 ml R10 for PBMC cell number and viability determination.


Isolation of class switched memory B cells from PBMCs was carried out by magnetic bead separation using a Switched Memory B cell isolation kit with Pre-Separation Filters and LS columns (MACS Miltenyi Biotec) according to manufacturer's instructions. In brief, counted PBMCs were incubated with a cocktail of biotin-conjugated antibodies against CD2, CD14, CD16, CD36, CD43, CD235a (glycophorin A), IgM and IgD. Cells were then washed and incubated with anti-biotin microbeads. Following another wash step, the suspension was passed through a Pre-Separation Filter (to remove cell aggregates) before applying it to an LS column where the magnetically labelled cells were retained in the column and the unlabelled CSM B cells passed through and could be collected in the flow-through for determination of cell number and viability.


Activation of CSM B Cells

To activate CSM B cells and promote antibody secretion into the supernatant, a mixture of cytokines, mAb, TLR agonist and a supplement were added to the R10 media (see above) to make complete R10 media. CSM B cells were resuspended in complete R10 media at 56 cells/ml and then plated out at 90 μl/well (5 cells/well) in ThermoFisher Matrix 384 well plates using a Biomek FX (Beckman Coulter). Cells were incubated at 37° C., 5% CO2 for seven days. On day 7, 30 μl/well of supernatant was removed and replaced with 30 μl fresh complete R10. On day 13, all the supernatant was harvested from all plates and screened against the antigen of interest via ELISA. B cell activation and culturing was monitored by measuring IgG1 concentrations in B cell supernatants at day 7 and day 13.


B Cell Supernatant Screen Against Target Antigens via ELISA

For B cell supernatant screening against target antigens, NUNC maxisorp 384-well plates (Sigma) were coated with C. albicans overnight culture (whole cell) or 1 μg/ml purified, recombinant N-terminus hyr1 protein antigen in 1×PBS and incubated at 4° C. overnight. Wells were washed three times with wash buffer using a Zoom Microplate Washer (Titertek) as above before incubation with blocking buffer for 1 h at room temperature with gentle shaking. After another three washes (as above), B cell supernatant was added and the plates incubated for 2 h at room temperature with gentle shaking. Wells were washed with wash buffer as above before addition of goat anti-human IgG, HRP conjugated (ThermoScientific) secondary antibody at 1:5000 dilution in blocking buffer and incubation for 45 min at room temperature with gentle shaking. ELISAs were developed and plates read at an OD of 450nm on an Envision plate reader (PerkinElmer).


Positive hits were defined as wells with an OD450 reading >4xbackground. B cells in ‘positive hit’ wells were resuspended in lysis buffer (ml DEPC-treated H2O (Life Technologies), 10 μl 1 M Tris pH 8, 25 μl RNAsin Plus RNAse Inhibitor (Promega)) and stored at −80° C.


Generation of Recombinant Anti-Hyr1 and Anti-Whole Cell IgG1 mAbs: Amplification of VH, Vκ-Cκ and Vλ-Cλ Genes—cDNA Synthesis and PCR


A schematic of the cloning protocol is shown in FIG. 12. Primers used for the RT-PCR reaction were based on those used by Smith et. al., (36). To ensure all possible VH germline families were captured during the amplification, four forward primers specific to the leader sequences encompassing the different human VH germline families (VH1-7) were used in combination with two reverse primers; both placed in the human CgCH1 region. For the RT-PCR of human Vκ-Cκ genes, three forward primers specific to the leader sequences for the different human Vκ germline families (Vκ1-4) were used with a reverse primer specific to the human kappa constant region (Cκ) and two further reverse primers which were specific to the C- and N-terminal ends of the 3′ untranslated region (UTR). To capture the repertoire of human Vλ genes, 7 forward primers capturing the leader sequences for the different human Vλ germline families (Vλ1-8) were used in a mixture with two reverse primers which were complementary to the C- and N-terminal ends of the 3′ UTR and another reverse primer specific to the human lambda constant region (Cλ).


Prior to cDNA synthesis, B cell lysates were thawed and diluted 1:5, 1:15 and 1:25 in nuclease-free H2O (Life Technologies) before addition of oligodT20 (50 μM) (Invitrogen, Life Technologies) and incubation at 70° C. for 5 min. Reverse transcription and the first PCR reaction (RT-PCR) were done sequentially using the QIAGEN OneStep RT-PCR kit according to manufacturer's instructions. For this step and the subsequent nested PCR step, amplification of the variable domain of human Ig heavy chain genes (VH), the variable and constant domains of human Ig kappa light chain genes (Vκ-Cκ) and the variable and constant domains of human Ig lambda light chain genes (Vλ-Cλ), were done in separate reactions. In brief, a reaction mixture was prepared containing QIAGEN OneStep RT-PCR Buffer 5×, dNTPs (10 mM), gene-specific forward and reverse primer mixes (10 μM), QIAGEN OneStep RT-PCR Enzyme Mix and nuclease-free H2O. Reaction mixture was then added to wells of a 96-well PCR plate before addition of neat or diluted (1:5, 1:15, 1:25) B cell lysate as the template, resulting in a final reaction volume of 50 μl/well. The following cycling conditions were used for the RT-PCR reaction; 50° C. for 30 min, 95° C. for 15 min then 35-40 cycles of (94° C. for 1 min, 55° C. for 1 min and 72° C. for 1 min) with a final extension at 72° C. for 10 min.


Amplification of VH, Vκ-Cκ and Vλ-Cλ Genes—Nested PCR Reaction

Nested PCR reactions were carried out using the PCR products from the RT-PCR reaction as the template, nested gene-specific primers based on Smith et al. (36) and Platinum PCR SuperMix High-Fidelity (Invitrogen, Life Technologies). A total of 27 forward primers specific for the VH framework 1 (FW1) sequence were used together with two reverse primers specific for the framework 4 (FW4) region of the VH gene. For nested PCR of the Vκ-Cκ gene, a mixture of 18 forward primers specific for human Vκ FW1 sequence were used with a reverse primer specific to the human kappa constant region 3′ end. For amplification of the Vλ-Cλ gene, a mixture of 31 forward primers specific for human Vλ FW1 sequences were used together with a reverse primer that was placed at the 3′ end of the human lambda constant region. The primers used to generate the PCR fragments in these nested PCR reactions contained 15 bp extensions which were complementary to the target downstream pTT5 expression vector. Reaction mixtures containing Platinum PCR SuperMix High Fidelity, gene-specific forward primer mix (10 μM) and gene specific reverse primer mix (10 μM) was added to wells in a 96-well PCR plate before addition of cDNA template. Amplification of VH genes, Vκ-Cκ genes and Vλ-Cλ genes, were done in separate reactions. After the nested PCR reaction, samples were analysed via agarose gel electrophoresis and positive hits identified and taken forward for downstream InFusion cloning with pTT5 mammalian expression vector.


pTT5 Mammalian Expression Vector Preparation


The pTT5mammalian expression used for mAb expression (licensed from the National Research Council of Canada (NRCC)) (53). The pTT5 vector plasmid contained an IgG1 heavy chain gene in the multiple cloning site so digestion to generate the heavy chain (HC) backbone for downstream sub cloning of VH was done by double digestion using FastDigest Restriction enzymes (Thermo Scientific) with BssHII before the leader sequence of the VH region and SalI restriction after the FW4 of the VH domain. This yielded the heavy chain constant region in the vector backbone. For double digestion of the vector to generate the light chain (LC) backbone, the whole IgG1 heavy chain gene was with BssHII and BamHI astDigest Restriction enzymes (Thermo Scientific) to generate the vector ready for insertion of either κ-Cκ or Vλ-Cλ. Digestion reactions to generate HC and LC backbones were carried out separately. Following confirmation of digestion, samples were run on a 1% agarose gel and bands were excised from the gel and purified using the QIAquick Gel Extraction kit (QIAGEN). DNA was quantified on a NanoVue Plus Spectrophotometer (GE Healthcare). To prevent vector self-ligation, the 3′- and 5′-termini of the linearized plasmids were dephosphorylated using FastAP Thermosensitive Alkaline phosphatase (Thermo Scientific). Reaction mixtures were cleaned up using the MinElute Reaction Cleanup Kit (QIAGEN) and then run on a 1% agarose gel. Bands corresponding to dephosphorylated HC and LC backbones were excised from the gel and purified using the QIAQuick Gel Extraction kit (QIAGEN) as above. Dephosphorylated linearized vector DNA was quantified on a NanoVue Plus spectrophotometer (GE Healthcare).


In-Fusion Cloning

The In-Fusion HD Cloning Kit (Clontech, USA) was used to clone the IgG VH, Vκ-Cκ and Vλ-Cλ genes into a pTT5 mammalian expression vector. To avoid the need for nested PCR product purification before cloning, cloning enhancer (Clontech, USA) was added to each nested PCR product in a 96-well PCR plate and incubated at 37° C. for 15 min, then 80° C. for 15 min. The cloning enhancer-treated PCR product was then added to the In-Fusion Enzyme Premix and linearized vector DNA (˜5-10 ng). Reactions were made up to 10 μl with nuclease-free H2O and incubated for 15 min at 50° C. Samples were then either stored at −20° C. or placed on ice before transformation of Stellar Competent cells (Clontech). For transformation, 2 μl of each In-Fusion reaction mixture was added to cells in a 96-well plate format, and left on ice for 30 min before heat shock at 42° C. for 40 sec and then returning to ice for 2 min. Cells were then recovered in SOC medium (Clontech, USA) with gentle shaking at 37° C. for 45-60 min before plating out onto LB agar plates (1% (w/v) tryptone, 0.5% (w/v) yeast extract, 1% (w/v) NaCl, 1.5% (w/v) agar) containing 100 μg/ml ampicillin. Plates were incubated at 37° C. overnight and single colonies picked the next day.


Plasmid DNA Generation for Transfection

Following transformation, 8-16 single colonies per initial hit well for VH, Vκand Vλ were picked and used to inoculate 2×TY media containing 100 μg/ml ampicillin in a Greiner deep well, 96-well plate (Sigma). VH, Vκ and Vλ plates were set up separately with the same plate layout to facilitate visual screening. Cells were grown at 37° C., 200 rpm overnight, and glycerol stocks were made the following day and stored at −80° C. To ensure accurate tracking of DNA sequences for downstream sequencing and transfections, each well inoculated by a single colony was given a unique ID based on the colony's original hit well and its position in the deep well 96 well plate following transformations. To obtain plasmid DNA for gene sequencing and small scale mammalian transfections, DNA minipreps from the overnight cultures were carried out in a 96-well plate format using the EPmotion (Eppendorf), according to manufacturer's instructions. DNA not taken for gene sequencing was stored at −20° C. until required for small scale transfections. Sequence data was analysed for CDR diversity and comparisons to germline sequences and used to identify clones to take forward for small scale transfection.


Small Scale Expression of Recombinant mAbs


Following VH, Vκ and Vλ gene sequencing, a file was generated containing all possible VH and Vκ/Vλ combinations resulting from the original hit wells from the primary ELISA screen. Automated mixing of the native heavy and light chain DNA pairing combinations (1.5 μg of HC plasmid DNA and 1.5 μg of LC plasmid DNA) into a new 96-well plate was facilitated through a HAMILTON MICROLAB® Starline liquid handling platform (Life Science robotics, Hamilton Robotics). Subsequent mixed DNA was used for small scale transient transfection of 3 ml of suspension cultured Expi293F cells (Life Technologies, USA) at a density of 2.5×106 cells/ml in 24-well tissue culture plates using the Expifectamine 293 Transfection kit (Life Technologies, USA) in accordance with manufacturer's instructions. Expi293F cells were maintained in pre-warmed (37° C.) sterile Expi293 expression media (Invitrogen) without antibiotics at 37° C., 7% CO2, 120 rpm shaking. Supernatants were harvested on day 6 and recombinant mAb expression was quantified using anti-human IgG Fc sensors on an Octet QKe (ForteBio, Calif., USA) for identification of mAbs to upscale.


Large Scale Expression, Purification and QC of Recombinant mAbs


For downstream large scale mammalian transfections, where a greater amount of DNA was required, DNA was prepared using a QIAGEN Plasmid Maxi Kit (QIAGEN, USA) according to manufacturer's instructions with typical yields of 1.5 μg/μl.


For large scale mAb expression, 100 μg of total DNA (50 μg of HC plasmid DNA and 50 μg LC plasmid DNA) was used to transiently transfect 100 ml of suspension cultured Expi293F cells (Life Technologies, USA) at a density of 2.5×106 cells/ml using the Expifectamine 293 Transfection Kit (Life Technologies, USA) in accordance with the manufacturer's instructions. Supernatants were harvested on day 6 and recombinant mAb expression was quantified as above using an Octet QKe (ForteBio). Recombinant mAbs were purified via affinity based Fast Protein Liquid Chromatography using HiTrap Protein A HP columns on an ÄKTA (GE Healthcare) and eluted in 20 mM citric acid, 150 nM NaCl (pH2.5) before neutralisation with 1 M Tris buffer (pH8). Purified mAbs were dialysed in PBS overnight and IgG concentration was quantified on a NanoVue Spectrophotometer (GE Healthcare). All purified recombinant mAbs were quality control checked via SDS-PAGE gel analysis using 4-12% Bis-Tris SDS-PAGE gels under reducing and non-reducing conditions to confirm mass, analytical size exclusion chromatography (SEC) to check for protein aggregation/degradation and analytical mass spectrometry to confirm the amino acid sequence identity of each mAb. Purified recombinant mAbs were also tested for functionality by binding to target antigen/whole cell via ELISA.


ELISA with Purified Recombinant mAbs


For confirmation of binding to target as purified recombinant mAbs an ELISA was carried out using the protocol for B cell supernatant screen. The only change was that titrated purified recombinant mAb was added in place of B cell supernatant.


Immunofluorescence Imaging of Anti-Hyr1 and Anti-Whole Cell mAbs Binding to Fungal Cells


Indirect immunofluorescence was performed using purified recombinant mAbs. A single Candida colony was used to inoculate 10 ml YPD medium and incubated at 30° C., 200 rpm overnight. Overnight cultures were diluted 1:1333 in milliQ water and then added to a poly-L-lysine coated glass slide (Thermo Scientific, Menzel-Glaser) and incubated for 30 min at room temperature to allow for adherence of yeast cells to the slide. To induce filamentation, cells were incubated in pre-warmed RPMI+10% FCS at 37° C. for 90 min-2 h (this step was omitted for staining of yeast cells), after which they were washed in Dulbecco's Phosphate Buffered Saline (DPBS) and fixed with 4% paraformaldehyde. Cells were washed again and blocked with 1.5% normal goat serum (Life Technologies) before staining with an anti-Candida mAb at 1-10 μg/ml for 1 h at room temperature. After three PBS washes, cells were stained with Alexa Fluor® 488 goat anti-human IgG antibody (Life Technologies) at a 1:400 dilution and incubated at room temperature for 1 h in the dark. For additional staining of fungal cell wall chitin, Calcofluor White (CFW) was added at 25 μg/ml and cells were incubated for 10 min at room temperature in the dark and washed with DPBS. Slides were left to air dry before adding one drop of Vectashield mounting medium (Vector Labs) and applying a 20 mm×20 mm coverslip to the slide. Cells were imaged in 3D on an UltraVIEW® VoX spinning disk confocal microscope (Nikon, Surrey, UK).


Preparation of Human Monocyte-Derived Macrophages

Human monocyte-derived macrophages were isolated from the blood of healthy volunteers. In brief, the PBMC layer was isolated as described above and was then washed and re-suspended in DMEM medium (Lonza, Slough, UK) supplemented with 200 U/mI penicillin/streptomycin antibiotics (Invitrogen, Paisley, UK) and 2 mM L-glutamine (Invitrogen, Paisley, UK). Serum was separated from blood using standard methods and heat-inactivated at 56° C. for 20 min before use. Monocytes were isolated from PBMCs via positive selection using CD14 microbeads (MACS, Miltenyi Biotec) according to manufacturer's instructions. PBMCs were incubated with MicroBeads conjugated to monoclonal anti-human CD14 antibodies. Cells were then washed and run through an LS column in a magnetic field causing the CD14+ cells to be retained in the column and the unlabelled cells to run through. The CD14+ cells were then eluted and resuspended in supplemented DMEM containing 10% donor-specific serum, for determination of cell count and viability. Monocytes were then plated out at a density of 1.2×105 cells/well in an 8-well glass based imaging dish (Ibidi, Munich, Germany) and incubated at 37°, 5% CO2 for 7 days. Cells were used in imaging experiments on day 7. Immediately prior to phagocytosis experiments, supplemented DMEM was replaced with pre-warmed supplemented CO2-independent media (Gibco, Invitrogen, Paisley, UK) containing 1 μM LysoTracker Red DND-99 (Invitrogen, Paisley, UK). LysoTracker Red is a fluorescent dye that stains acidic compartments in live cells, enabling tracking of these cells during phagocytosis and phagolysosome maturation.


Preparation of J774.1 Mouse Macrophage Cell Line

J774.1 macrophages (ECACC, HPA, Salisbury, UK) were maintained in tissue culture flasks in DMEM medium (Lonza, Slough, UK) supplemented with 10% (v/v) FCS (Biosera, Ringmer, UK), 200 U/mI penicillin/streptomycin antibiotics (Invitrogen, Paisley, UK) and 2 mM L-glutamine (Invitrogen, Paisley, UK) and incubated at 37° C., 5% CO2. For phagocytosis assays, macrophages were seeded in 300 μl supplemented DMEM at a density of 1×105 cells/well in an 8-well glass based imaging dish (Ibidi, Munich, Germany) and incubated overnight at 37° C., 5% CO2. Immediately prior to phagocytosis experiments, supplemented DMEM was replaced with 300 μl pre-warmed supplemented CO2-independent media (Gibco, Invitrogen, Paisley, UK) containing 1 μM LysoTracker Red DND-99 (Invitrogen, Paisley, UK).


Preparation of Fluorescein Isothiocyanate (FITC)-Stained C. Albicans


C. albicans colonies were grown in YPD medium and incubated at 30° C., 200 rpm overnight. Live C. albicans cells were stained for 10 min at room temperature in the dark with 1 mg/ml FITC (Sigma, Dorset, UK) in 0.05 M carbonate-bicarbonate buffer (pH 9.6) (BDH Chemicals, VWR International, Leicestershire, UK). Following the 10 min incubation, in phagocytosis assays using C. albicans FITC-labelled yeast, the cells were washed three times in 1×PBS to remove any residual FITC and finally re-suspended in 1×PBS or 1×PBS containing purified anti-Candida mAb at 1-50 μg/ml. For assays where pre-germinated C. albicans was to be added to immune cells, cells were washed and re-suspended in supplemented CO2-independent media with or without anti-Candida mAb at 1-50 μg/ml and incubated at 37° C. with gentle shaking for 45 min.


Live Cell Video Microscopy Phagocytosis Assays

Phagocytosis assays were performed using our standard protocol with modifications (42, 43, 54). Following pre-incubation with/without anti-Candida mAb, live FITC-stained wild type C. albicans (CA14-Clp10) yeast or hyphal cells were added to LysoTracker Red DND-99-stained J774.1 murine macrophages or human monocyte-derived macrophages in an 8-well glass based imaging dish (Ibidi) at a multiplicity of infection (MOI) of 3. Video microscopy was performed using an UltraVIEW® VoX spinning disk confocal microscope (Nikon, Surrey, UK) in a 37° C. chamber and images were captured at 1 min intervals over a 3 h period. At least three independent experiments were performed for each antibody and at least 2 videos were analysed from each experiment using Volocity 6.3 imaging analysis software (Improvision, PerkinElmer, Coventry, UK). Twenty five macrophages were selected at random from each experiment and analysed individually at 1 min intervals over a 3 h period. Measurements taken included: C. albicans uptake—defined as the number of C. albicans cells taken up by an individual phagocyte over the 3 h period; C. albicans rate of engulfment—defined as the time point at which cell-cell contact was established until the time point at which C. albicans was fully engulfed (a fungal cell was considered to have been fully ingested when its FITC-fluorescent signal was lost, indicating that the fungal cell was now inside the phagocyte and not merely bound to the phagocyte cell surface) and finally Volocity 6.3 imaging analysis software was used to measure the distance travelled, directionality and velocity of macrophages at 1 min intervals during the first hour of the assay which provided a detailed overview of macrophage migration towards C. albicans cells.


Mean values and standard deviations were calculated. One- or two-way ANOVA followed by Bonferroni multiple comparison tests or unpaired, two-tailed t tests were used to determine statistical significance.


Systemic Candidiasis Infection Model

A well-established three-day model of disseminated candidiasis was employed to assess the efficacy of anti-Candida mAbs in vivo (44, 52). On day 0, ˜3.2×105 C. albicans SC5314 yeast cells were pre-incubated at RT with 7.5 mg/kg purified recombinant anti-Candida mAb for 60 min to allow binding of the antibody to the Candida cell surface before administration intravenously via the lateral tail vein. Assessment of disease progression was carried out by observation and weighing on successive days from day 0 up to and including day 3, at which point the animals were culled and the kidneys harvested for analysis of fungal burden. Fungal burdens were quantitated by homogenising the organ, and plating out serial dilutions on Sabouraud dextrose agar plates (1% mycological peptone (w/v), 4% glucose (w/v), 2% agar (w/v)) before incubation at 35° C. overnight. Colonies were counted the next day and fungal burden expressed as log CFU per gram of infected organ. An overall disease outcome score devised from the combination of 3-day weight loss and kidney burden data was also generated to assess disease progression.


Enzymatic Modification of Candida Albicans Cell Wall

For proteinase K treatment, single colonies of Candida were inoculated into 10 ml YPD medium and incubated at 30° C., 200 rpm overnight. Cultures were diluted in milliQ water and then adhered on poly-L-lysine coated glass slides. To induce filamentation, cells were incubated in pre-warmed RPMI+10% FCS at 37° C. for 90 min-2 h. Slides were washed with DPBS and cells were treated with 50 μg/ml proteinase K at 37° C. for 1 h. For Endo-H and zymolyase 20T treatments, C. albicans overnight yeast cells were washed and resuspended in DPBS. Filamentous cells were induced as above. Cells were washed in DPBS and resuspended in Glycobuffer and Endoglycosidase H (10 U/μl; NEB) or Buffer S and Zymolyase 20T (50 U/g wet cells; MPBIO) at 37° C. for 2 h. Cells were then washed in DPBS and fixed with 4% paraformaldehyde, washed and blocked with 1.5% normal goat serum (Life Technologies) before staining with an anti-Candida mAb at 1 μg/ml for 1 h at room temperature. After 3 washes with DPBS, cells were stained with Alexa Fluor® 488 goat anti-human IgG antibody (Life Technologies) at a 1:400 dilution and incubated at room temperature for 1 h prior to imaging in 3D on an UltraVIEW® VoX spinning disk confocal microscope (Nikon, Surrey, UK).


Preparation of Human Monocyte-Derived Macrophages

Human macrophages were derived from monocytes isolated from the blood of healthy volunteers. PBMCs were resuspended in Dulbecco's Modified Eagle's Medium (DMEM) (Lonza, Slough, UK) supplemented with 200 U/ml penicillin/streptomycin antibiotics (Invitrogen, Paisley, UK) and 2 mM L-glutamine (Invitrogen, Paisley, UK). Serum isolated from blood was heat inactivated for 20 min at 56° C. PBMCs were seeded at 6×105 in 300 μl/well supplemented DMEM medium containing 10% autologous human serum, onto an 8-well glass based imaging dish (Ibidi, Munich, Germany) and incubated at 37° C. with 5% CO2 for 1 h 45 min to facilitate monocyte adherence to the glass surface. Floating lymphocytes in the supernatant were aspirated and the same volume of fresh pre-warmed supplemented DMEM containing 10% autologous human serum added to the well. Cells were incubated at 37° C., 5% CO2 for 7 days with media changed on days 3 and 6. Cells were used in imaging experiments on day 7. Supplemented DMEM was replaced with pre-warmed supplemented CO2-independent media containing 1 μM LysoTracker Red DND-99 (Invitrogen) immediately prior to phagocytosis experiments.


Counterimmunoelectrophoresis

Agar gels were prepared (Veronal buffer+0.5% (w/v) purified agar+0.5% (w/v) LSA agarose+0.05% (w/v) sodium azide, pH 8.2) and wells were cut out using a cutter. Into one column of wells, 10 μl of neat anti-Candida mAb was added. The same volume of antigen (crude C. albicans yeast or hyphal preparation (following glass bead disruption of cells and 1 min centrifugation at 13000 rpm to generate disrupted cell wall/glass bead slurry and cell supernatant antigenic preparations)) was added to the second column of wells and gels were placed into an electrophoresis tank containing veronal buffer. Gels were oriented so that the antibody wells were lined up alongside the anode and the antigen wells alongside the cathode due to antibody migration towards the cathode via electroendosmosis and antigen migration towards the anode due to lower isoelectric points than the buffer pH. The gels were run at 100V for 90 min before removal and immersion in saline-trisodium citrate overnight. The following day the gels were rinsed with water and covered with moistened filter paper and left to dry in an oven for 2 h. Once dried, the filter paper was moistened and removed and the gels put back into the oven for a further 15 min to dry completely. Gels were then immersed in Buffalo black solution (0.05% (v/v) Buffalo black, 50% (v/v) distilled water, 40% (v/v) methylated spirit, 10% (v/v) acetic acid) for 10 min before destaining in destaining solution (45% (v/v) industrial methylated spirits, 10% (v/v) acetic acid, 45% (v/v) distilled water) for 10 min. Gels were then dried and examined for the formation of precipitin lines. The results are shown in FIG. 17.


High-Pressure Freezing (HPF) of Samples for Immunogold Labelling of C. Albicans Cells with Anti-Candida mAbs for Transmission Electron Microscopy (TEM).



C. albicans yeast and hyphal cell samples were prepared by high-pressure freezing using an EMPACT2 high-pressure freezer and rapid transport system (Leica Microsystems Ltd., Milton Keynes, United Kingdom). Using a Leica EMAFS2, cells were freeze-substituted in substitution reagent (1% (w/v) OsO4 in acetone) before embedding in Spurr resin and polymerizing at 60° C. for 48 h. A Diatome diamond knife on a Leica UC6 ultramicrotome was used to cut ultrathin sections which were then mounted onto nickel grids. Sections on nickel grids were blocked in blocking buffer (PBS+1% (w/v) BSA and 0.5% (v/v) Tween20) for 20 min before incubation in incubation buffer (PBS+0.1% (w/v) BSA) for 5 min×3. Sections were then incubated with anti-Candida mAb (5 μg/ml) for 90 min before incubation in incubation buffer for 5 min a total of 6 times. mAb binding was detected by incubation with Protein A gold 10 nm conjugate (Aurion) (diluted 1:40 in incubation buffer) for 60 min before another six 5 min washes in incubation buffer followed by three 5 min washes in PBS and three 5 min washes in water. Sections were then stained with uranyl acetate for 1 min before three 2 min washes in water and then left to dry. TEM images were taken using a JEM-1400 Plus using an AMT UltraVUE camera. The results are shown in FIG. 18.









TABLE S1







Clinical isolates and strains









Strain name
Genotype
Reference





CAl4 + Clp10 (NGY152)
ura3Δ::λimm434/ura3Δ::λimm434
Brand et al. 2004



RPS1/rps1::URA3



hyr1Δ
hyr1Δ::hisG/hyr1Δ:hisG-URA-3-hisG
Bailey et al. 1996


hyr1Δ + HYR1
hyr1::hisG/hyr1::hisG/RPS1/rps1::HYR1
Belmonte




(unpublished)


tup1Δ
tup1Δ::hisG/tup1Δ::hisG-URA3-hisG
Fonzi & Irwin 1993



C. albicans SC5314

Clinical isolate
Gillum et al. 1984



C. glabrata SCS71182B

Clinical isolate
Odds et al. 2007



C. tropicalis AM2005/0546

Clinical isolate
Clinical isolate from




Aberdeen



C. lusitaniae

Clinical isolate
Odds et al. 2007


SCS211362H





C. krusei SCS71987M

Clinical isolate
Odds et al. 2007



C. parapsilosis

Clinical isolate
Rudek 1978


ATCC22019





C. dubliniensis CD36

Clinical isolate
Moran et al. 1998



A. fumigatus V05-27

Clinical isolate
Netea et al. 2003



C. auris CBS 10913T

Clinical isolate
Satoh et al. 2009



C. haemulonii CBS 5149T

Clinical isolate
Khan et al. 2007



C. neoformans KN99α

H99 mating type α
Nielsen et al. 2003



C. gattii R265

Clinical isolate
Fyfe et al. 2002



P. carinii M167-6

Isolated from rat lung tissue




M. dermatis CBS 9169

CBS
Sugita et al. 2002



M. circinefioides CBS

CBS
Li et al. 2011


277.49


















TABLE S2







Recombinant Hyr1 protein amino acid sequence. The


leader sequence is underlined and the 6xHis tag is


in italics, and is followed by the linker ′G′.


Hyr1 protein amino acids 63-350 make up the


remainder of the sequence.








Recombinant protein
Amino acid sequence


antigen name
(amino acids 63-350)





Recombinant Hyr1

METDTLLLWVLLLWVPGSTGGSG
HHHHHH



N-terminus fragment
GEVEKGASLFIKSDNGPVLALNVALSTLV



RPVINNGVISLNSKSSTSFSNFDIGGSSF



TNNGEIYLASSGLVKSTAYLYAREWTNNG



LIVAYQNQKAAGNIAFGTAYQTITNNGQI



CLRHQDFVPATKIKGTGCVTADEDTWIKL



GNTILSVEPTHNFYLKDSKSSLIVHAVSS



NQTFTVHGFGNGNKLGLTLPLTGNRDHFR



FEYYPDTGILQLRAAALPQYFKIGKGYDS



KLFRIVNSRGLKNAVTYDGPVPNNEIPAV



CLIPCTNGPSAPESESDLNTPTTSSIGT
















TABLE S3







Purified recombinant human IgG1 mAbs


generated using the single B cell technology.









Antibody
Yield (mg)
Target












AB-120
12
Hyr1 protein


AB-121
28.5
Hyr1 protein


AB-122
67.9
Hyr1 protein


AB-123
67.3
Hyr1 protein


AB-124
38.9
Hyr1 protein


AB-118
7.5

C. albicans ‘whole cell’



AB-119
13.5

C. albicans ‘whole cell’



AB-126
60.9

C. albicans ‘whole cell’



AB-127
24.5

C. albicans ‘whole cell’



AB-129
2.3

C. albicans ‘whole cell’



AB-130
1.1

C. albicans ‘whole cell’



AB-131
24.1

C. albicans ‘whole cell’



AB-132
9.3

C. albicans ‘whole cell’



AB-133
19

C. albicans ‘whole cell’



AB-134
7.7

C. albicans ‘whole cell’



AB-135
16.5

C. albicans ‘whole cell’



AB-139
12.2

C. albicans ‘whole cell’



AB-140
19.5

C. albicans ‘whole cell’


























TABLE VH














SEQ


AB


VH





ID


name

VH FW1
CDR1
VH FW2
VH CDR2
VH FW3
VH CDR3
VH FW4
NO







06-
VH3
QVTLKESGGGLVQPG
RTY
WVRQDPG
RLDEVGRLT
RFTISRDNAKNILYLQMN
DLSGSADY
WGQGTLV
15A


AB-

GSLRLSCVASGFTF
WMH
KGLVWVS
SYADSVNG
SLRAEDTGVYYCAR

TVSS



119














06-
VH3
EVQLVESGGGLVQPG
SNY
WVRQVPG
RINEDGSVT
RFTISRDNAKNTLYLQM
DLCGERDD
WGQGTLV
15B


AB-

GSLRLSCSASQFIL
WVH
EGLVWVS
SYADSVKG
NSLRVDDTAVYYCVR

SVSS



118














06-
VH3
EVQLVQSGGGLVQPG
TSY
WVRQAPG
VITGNVGTS
RFTISRDNSKKTVSLQM
TRYDFSSGYY
WGQGTLV
15C


AB-

GSLGLSCAASGFIF
AMT
KGLEWVS
YYADSVKG
NSLRAEDTAIYYCVK
FDD
SVSS



120














06-
VH3
EVQLVESGGTLVQPG
SDY
WVRQAPG
NIKQDGSEK
RVTISRDNAQNSVFLQM
DGYTFGPATT
WGRGTLV
15D


AB-

GSLRLSCAASGFTF
WMN
KGLEWVA
YYVDSLRG
HSLSVEDTAVYYCAR
ELDH
SVSS



121














06-
VH3
EVQLVQSGGGLAQPG
DDF
WVRQPPG
GLIVVNGGSI
RFTISRDNAKNSLFLQM
GLSGGTMAPF
WGQGTMV
15E


AB-

RSLRLSCAASGFGF
AMH
KGLEWVS
DYAGSVRG
NSLRAEDTALYYCAK
DI
SVSS



122














06-
VH3
EVQLLESGGGVVQPG
SNY
WVRQAPG
VVWFDGSY
RFTISRDNSKSTLYLQM
PIMTSAFDI
WGPGTMV
15F


AB-

RSLRLSCAASGFTF
GMH
KGLEWVA
KYYTDSVKG
NSLRAEDTAVYYCVS

SVSS



123














06-
VH3
EVQLVESGGGVVQPG
SNY
WVRQAPG
VVWLDGSY
RFTISRDNSKSTLYLQM
PIMTSAFDI
WGPGTMV
15G


AB-

RSLRLSCAASGFTF
GMH
KGLEWVA
KYYTGSVKG
NSLRAEDTAAYYCVS

TVSS



124














06-
VH3
EVQLVESGGGLAQPG
AGN
WVRQAPG
AIGGSDDRT
RFTISRDKSKNTLSLQM
DIWRWAFDY
WGQGTLV
15H


AB-

GSLRLSCEASGFHL
AMA
KGLEWVA
DYADSVKG
NSLRVEDTAVYYCAK

SVSS



126














06-
VH3
EVQLVESGGGLVNPG
SNY
WVRQAPG
SISRSGDYIY
RSTISRDNAKNSLFLQM
DWGRLGYCSS
WGQGTRV
15I


AB-

GSLRLSCAASGFTF
AMN
KGLEWVS
YADSLKG
NSLRAEDSAVYYCAR
NNCPDAFDV
SVSS



127














06-
VH3
QVQLVESGGGLVQPG
SNY
WVRQVPG
RINEDGSVT
RFTISRDNAKNTLYLQM
DLCGERDD
WGQGTLV
15J


AB-

GSLRLSCSASQFIL
WVH
EGLVWVS
SYADSVKG
NSLRVDDTAVYYCVR

TVSS



129














06-
VH3
QLQLQESGGGLVQPG
SNY
WVRQVPG
RINEDGSVT
RFTISRDNAKNTLYLQM
DLCWERDD
WGQGTLV
15K


AB-

GSLRLSCSASQFIL
WVH
EGLVWVS
SYADSVKG
NSLRVDDTAVYYCVR

SVSS



130














06-
VH3
QVQLVQSGGGVVQPG
KISI
WVRQAPG
AMSYDGFSK
RLTISRDSSTNTLYLEMN
EAYTSGRAGC
WGQGVLV
15L


AB-

GSLRLSCAASPFTF
LH
KGLEWVS
YYADSVKG
SLRFEDTALYFCAR
FNP
SVSS



131














06-
VH3
QVLKESGGGVVQPGG
ETSI
WVRQAPG
AMSYDGFSK
RLTISRDSSTNTLYLEMN
EAYTSGRAGC
WGQGVLV
15M


AB-

SLRLSCAASPFTF
LH
KGLEWVS
YYADSVKG
SLRFEDTALYFCAR
FDP
SVSS



132














06-
VH3
EVQLVESGGGLVQPG
NTY
WVRQAPG
RINEDGTTIS
RFTISRDNAENTLYLQM
DFTGPFDS
WGQGTLV
15N


AB-

GSLRVSCAASGFTL
WMH
KGLVWVS
YADSVRG
HSLRAEDTGVYYCAR

SVSS



133














06-
VH3
QLQLQESGGGLVQPG
SSH
WVRQAPG
SISISGGDTF
RFTIFRDNSKNTVYLQM
ETSPNDY
WGQGTLV
15O


AB-

GSLRLSCVVSGFTF
AMS
KGLEWVS
YADSVRG
NSLRAEDTAVYYCAT

SVSS



134














06-
VH3
EVQLVETGGGLVQPG
SSH
WVRQAPG
SISISGGDTF
RFTIFRDNSKNTVYLQM
ETSPNDY
WGQGTLV
15P


AB-

GSLRLSCVVSGFTF
AMS
KGLEWVS
YADSVRG
NSLRAEDTAVYYCAT

TVSS



135














06-
VH3
EVQLVESGGGLVQPG
NTY
WVRQAPG
RINEDGTTIS
RFTISRDNAENTLYLQM
DFTGPFDS
WGQGTLV
15Q


AB-

GSLRVSCAASGFTL
WMH
KGLVWVS
YADSVRG
HSLRAEDTGVYYCAR

SVSS



139














06-
VH3
EVQLVESGGGLVQPG
NTY
WVRQAPG
RINEDGTTIS
RFTISRDNAENTLYLQM
DFTGPFDS
WGQGTLV
15R


AB-

GSLRVSCAASGFTL
WMH
KGLVWVS
YADSVRG
HSLRAEDTGVYYCAR

SVSS



140

























TABLE VL














SEQ


AB


VL

VL



ID


name

VL FW1
CDR1
VL FW2
CDR2
VL FW3
VL CDR3
VL FW4
NO







06-AB-
VK2
DVVLTQSPLFLPVT
RSSQSLLHS
WYLQKPGQS
SVFN
GVPDRFSGSGSGTDFTL
MQALEPPYT
FGQGTKLE
16A


119

PGEPASISC
RGHTSLH
PHLLIY
RAS
KISRVEAEDVGVYYC

IK






06-AB-
VK2
DIVMTQSPLSLPVT
RSSQSLLHR
WYLQKPGQS
LGSN
GVPDRFSGSGSGTDFTL
MQGLQTPY
FGQGTKLE
16B


118

PGEAASISC
NGKTFFA
PQILIY
RAS
KISRVEAEDVGIYYC
T
IK






06-AB-
VK2
DIVMTQSPSSVSAS
RASQGISRW
WYQQKPGEA
AASS
GVPSRFSGSGSGTDFTL
QQANSFPIT
FGQGTRL
16C


120

VGDKVTITC
LA
PELLIY
LOS
TISSLQPEDFATYYC

QIK






06-AB-
VL3
QLVLTQPPSVSVSP
SGDELRNKY
WYQQKSGQS
QDNN
GIPERFSGSQSGDTATL
QAWVSQTL
FGGGTKLT
16D


121

GQTASITC
TS
PVLVIY
RPS
TISGTQAVDEADYYC
V
VL






06-AB-
VL3
QAGLTQPPSVSVA
GGNNIGSKH
WYQQKPGQA
DDSD
GVPERFSGSNSGNTATL
QVWDRSSD
FGGGTRLT
16E


122

PGQTATIPC
VH
PVAVVY
RPS
TISSVEAGDEADYYC
HFWL
VL






06-AB-
VL2
QLVLTQPPSASGS
TGTSSDVGG
WYQHHPGKA
EVSQ
GVPDRFSGSKSGNTASL
SSYAGSVVL
FGGGTKLT
16F


123

PGQSVTISC
SNFVS
PKLMIY
RPS
TVSGLQADDEADYYC

VL






06-AB-
VL2
QLVLTQPPSASGS
TGTSSDVGG
WYQHHPGKA
EVSQ
GVPDRFSGSKSGNTASL
SSYAGSVVL
FGGGTKLT
16G


124

PGQSVTISC
SNFVS
PKLMIY
RPS
TVSGLQADDEADYYC

VL






06-AB-
VK3
DIVMTQSPATLSLS
WASQYINTY
WYQHKPGQA
DASK
GIPARFSGSGSGTDFTL
QQGSNWPL
FGQGTRL
16H


126

PGERATLSC
VN
PRLLIY
RAT
TISSLEPEDFAVYYC
T
EIK






06-AB-
VK1
EIVMTQSPSFVSAS
RASQDISNW
WYQQKPGKA
ASSN
GVPSRFSGSGSGTDFAL
QQENSFPY
FGQGTKLE
16I


127

VGDRVTITC
LV
PKLLIY
LOS
TIISLQPEDFATYYC
T
IK






06-AB-
VK2
VIWMTQSPLSLPVT
RSSQSLLHR
WYLQKPGQS
LGSN
GVPDRFSGSGSGTDFTL
MQGLQTPY
FGQGTKLE
16J


129

PGEAASISC
NGRTFFA
PQILIY
RAF
KISRVEAEDVGIYYC
T
IK






06-AB-
VK2
VIWMTQSPLSLPVT
RSSQSLLHR
WYLQKPGQS
LGSN
GVPDRFSGSGSGTDFTL
MQGLQTPY
FGQGTKLE
16K


130

PGEAASISC
NGRTFFA
PQILIY
RAF
KISRVEAEDVGIYYC
T
IK






06-AB-
VK1
DIVMTQTPSTQSAS
RASQSISIWL
WYQQKPGKA
DAST
GVPSRFSGSGSGTEFTL
QRYNDYPP
FGPGTKVE
16L


131

VGDRVTITC
A
PKLLIH
LES
TISSLQPDDSATYYC
T
IK






06-AB-
VK1
EIVMTQSPSTQSAS
RASQSISIWL
WYQQKPGKA
DAST
GVPSRFSGSGSGTEFTL
QRYNDYPP
FGPGTKVE
16M


132

VGDRVTITC
A
PKLLIH
LES
TISSLQPDDSATYYC
T
IK






06-AB-
VL1
QSVLTQPPSVSGT
SGSNSNAG
WYQQVPGTA
KNNQ
GVPDRFSGSKSGTSASL
IVWDGSLSG
FGTGTKVT
16N


133

PGQRVTISC
RDYVS
PKLLIY
RPS
AISGLRSEDDGDYYC
YV
VL






06-AB-
VL7
SYELTQPSSLTVSP
GLSSGAVTS
WFQQKPGQA
DTSR
WTPARFSGSLLGGKAAL
LLACNGACV
FGGGTKLT
16O


134

GGTVTLTC
GHYPY
PKTLIF
KHS
TLSGAQPEDDADYYC

VL






06-AB-
VL7
SYELTQPSSLTVSP
GLSSGAVTS
WFQQKPGQA
DTSR
WTPARFSGSLLGGKAAL
LLACNGACV
FGGGTKLT
16P


135

GGTVTLTC
GHYPY
PKTLIF
KHS
TLSGAQPEDDADYYC

VL






06-AB-
VL1
QSVLTQPPSVSGT
SGSNSNVG
WYQQVPGTA
KNNR
GVPDRFSGSKSGTSASL
IVWDGSLSG
FGTGTKVT
16Q


139

PGQRVTISC
RDYVS
PKLLIY
RPS
AISGLRSEDDGDYYC
YV
VL






06-AB-
VL1
QLVLTQPPSVSGT
SGSNSNVG
WYQQVPGTA
KNNQ
GVPDRFSGSKSGTSASL
IVWDGSLSG
FGTGTKVT
16R


140

PGQRVTISC
RDYVS
PKLLIY
RPS
AISGLRSEDDGDYYC
YV
VL









Antibody Sequences and Seq ID No.s









TABLE A







Antibody AB119









06-AB-

Seq.


119
Sequence
ID No.





VH FW1
QVTLKESGGGLVQPGGSLRLSCVASGFTF
 1A


VH CDR1
RTYWMH
 2A


VH FW2
WVRQDPGKGLVWVS
 3A


VH CDR2
RLDEVGRLTSYADSVNG
 4A


VH FW3
RFTISRDNAKNILYLQMNSLRAEDTGVYYCAR
 5A


VH CDR3
DLSGSADY
 6A


VH FW4
WGQGTLVTVSS
 7A





VL FW1
DVVLTQSPLFLPVTPGEPASISC
 8A


VL CDR1
RSSQSLLHSRGHTSLH
 9A


VL FW2
WYLQKPGQSPHLLIY
10A


VL CDR2
SVFNRAS
11A


VL FW3
GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
12A


VL CDR3
MQALEPPYT
13A


VL FW4
FGQGTKLEIK
14A
















TABLE B







Antibody AB118









06-AB-

Seq.


118
Sequence
ID No.





VH FW1
EVQLVESGGGLVQPGGSLRLSCSASQFIL
 1B


VH CDR1
SNYWVH
 2B


VH FW2
WVRQVPGEGLVWVS
 3B


VH CDR2
RINEDGSVTSYADSVKG
 4B


VH FW3
RFTISRDNAKNTLYLQMNSLRVDDTAVYYCVR
 5B


VH CDR3
DLCGERDD
 6B


VH FW4
WGQGTLVSVSS
 7B





VL FW1
DIVMTQSPLSLPVTPGEAASISC
 8B


VL CDR1
RSSQSLLHRNGKTFFA
 9B


VL FW2
WYLQKPGQSPQILIY
10B


VL CDR2
LGSNRAS
11B


VL FW3
GVPDRFSGSGSGTDFTLKISRVEAEDVGIYYC
12B


VL CDR3
MQGLQTPYT
13B


VL FW4
FGQGTKLEIK
14B
















TABLE C







Antibody AB120









06-AB-

Seq. 


120
Sequence
ID No.





VH FW1
EVQLVQSGGGLVQPGGSLGLSCAASGFIF
 1C


VH CDR1
TSYAMT
 2C


VH FW2
WVRQAPGKGLEWVS
 3C


VH CDR2
VITGNVGTSYYADSVKG
 4C


VH FW3
RFTISRDNSKKTVSLQMNSLRAEDTAIYYCVK
 5C


VH CDR3
TRYDFSSGYYFDD
 6C


VH FW4
WGQGTLVSVSS
 7C





VL FW1
DIVMTQSPSSVSASVGDKVTITC
 8C


VL CDR1
RASQGISRWLA
 9C


VL FW2
WYQQKPGEAPELLIY
10C


VL CDR2
AASSLQS
11C


VL FW3
GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC
12C


VL CDR3
QQANSFPIT
13C


VL FW4
FGQGTRLQIK
14C
















TABLE D







Antibody AB121









06-AB-

Seq.


121
Sequence
ID No.





VH FW1
EVQLVESGGTLVQPGGSLRLSCAASGFTF
 1D


VH CDR1
SDYWMN
 2D


VH FW2
WVRQAPGKGLEWVA
 3D


VH CDR2
NIKQDGSEKYYVDSLRG
 4D


VH FW3
RVTISRDNAQNSVFLQMHSLSVEDTAVYYCAR
 5D


VH CDR3
DGYTFGPATTELDH
 6D


VH FW4
WGRGTLVSVSS
 7D





VL FW1
QLVLTQPPSVSVSPGQTASITC
 8D


VL CDR1
SGDELRNKYTS
 9D


VL FW2
WYQQKSGQSPVLVIY
10D


VL CDR2
QDNNRPS
11D


VL FW3
GIPERFSGSQSGDTATLTISGTQAVDEADYYC
12D


VL CDR3
QAWVSQTLV
13D


VL FW4
FGGGTKLTVL
14D
















TABLE E







Antibody AB122









06-AB-

Seq.


122
Sequence
ID No.





VH FW1
EVQLVQSGGGLAQPGRSLRLSCAASGFGF
 1E


VH CDR1
DDFAMH
 2E


VH FW2
WVRQPPGKGLEWVS
 3E


VH CDR2
GLTWNGGSIDYAGSVRG
 4E


VH FW3
RFTISRDNAKNSLFLQMNSLRAEDTALYYCAK
 5E


VH CDR3
GLSGGTMAPFDI
 6E


VH FW4
WGQGTMVSVSS
 7E





VL FW1
QAGLTQPPSVSVAPGQTATIPC
 8E


VL CDR1
GGNNIGSKHVH
 9E


VL FW2
WYQQKPGQAPVAVVY
10E


VL CDR2
DDSDRPS
11E


VL FW3
GVPERFSGSNSGNTATLTISSVEAGDEADYYC
12E


VL CDR3
QVWDRSSDHFWL
13E


VL FW4
FGGGTRLTVL
14E
















TABLE F







Antibody AB123











Seq.


06-AB-123
Sequence
ID No.





VH FW1
EVQLLESGGGVVQPGRSLRLSCAASGFTF
 1F





VH CDR1
SNYGMH
 2F





VH FW2
WVRQAPGKGLEWVA
 3F





VH CDR2
VVWFDGSYKYYTDSVKG
 4F





VH FW3
RFTISRDNSKSTLYLQMNSLRAEDTAVYYCVS
 5F





VH CDR3
PIMTSAFDI
 6F





VH FW4
WGPGTMVSVSS
 7F





VL FW1
QLVLTQPPSASGSPGQSVTISC
 8F





VL CDR1
TGTSSDVGGSNFVS
 9F





VL FW2
WYQHHPGKAPKLMIY
10F





VL CDR2
EVSQRPS
11F





VL FW3
GVPDRFSGSKSGNTASLTVSGLQADDEADYYC
12F





VL CDR3
SSYAGSVVL
13F





VL FW4
FGGGTKLTVL
14F
















TABLE G







Antibody AB124











Seq.


06-AB-124
Sequence
ID No.





VH FW1
EVQLVESGGGVVQPGRSLRLSCAASGFTF
 1G





VH CDR1
SNYGMH
 2G





VH FW2
WVRQAPGKGLEWVA
 3G





VH CDR2
VVWLDGSYKYYTGSVKG
 4G





VH FW3
RFTISRDNSKSTLYLQMNSLRAEDTAAYYCVS
 5G





VH CDR3
PIMTSAFDI
 6G





VH FW4
WGPGTMVTVSS
 7G





VL FW1
QLVLTQPPSASGSPGQSVTISC
 8G





VL CDR1
TGTSSDVGGSNFVS
 9G





VL FW2
WYQHHPGKAPKLMIY
10G





VL CDR2
EVSQRPS
11G





VL FW3
GVPDRFSGSKSGNTASLTVSGLQADDEADYYC
12G





VL CDR3
SSYAGSVVL
13G





VL FW4
FGGGTKLTVL
14G
















TABLE H







Antibody AB126











Seq.


06-AB-126
Sequence
ID No.





VH FW1
EVQLVESGGGLAQPGGSLRLSCEASGFHL
 1H





VH CDR1
AGNAMA
 2H





VH FW2
WVRQAPGKGLEWVA
 3H





VH CDR2
AIGGSDDRTDYADSVKG
 4H





VH FW3
RFTISRDKSKNTLSLQMNSLRVEDTAVYYCAK
 5H





VH CDR3
DIWRWAFDY
 6H





VH FW4
WGQGTLVSVSS
 7H





VL FW1
DIVMTQSPATLSLSPGERATLSC
 8H





VL CDR1
WASQYINTYVN
 9H





VL FW2
WYQHKPGQAPRLLIY
10H





VL CDR2
DASKRAT
11H





VL FW3
GIPARFSGSGSGTDFTLTISSLEPEDFAVYYC
12H





VL CDR3
QQGSNWPLT
13H





VL FW4
FGQGTRLEIK
14H
















TABLE I







Antibody AB127











Seq.


06-AB-127
Sequence
ID No.





VH FW1
EVQLVESGGGLVNPGGSLRLSCAASGFTF
 1I





VH CDR1
SNYAMN
 2I





VH FW2
WVRQAPGKGLEWVS
 3I





VH CDR2
SISRSGDYIYYADSLKG
 4I





VH FW3
RSTISRDNAKNSLFLQMNSLRAEDSAVYYCAR
 5I





VH CDR3
DWGRLGYCSSNNCPDAFDV
 6I





VH FW4
WGQGTRVSVSS
 7I





VL FW1
EIVMTQSPSFVSASVGDRVTITC
 8I





VL CDR1
RASQDISNWLV
 9I





VL FW2
WYQQKPGKAPKLLIY
10I





VL CDR2
ASSNLQS
11I





VL FW3
GVPSRFSGSGSGTDFALTIISLQPEDFATYYC
12I





VL CDR3
QQENSFPYT
13I





VL FW4
FGQGTKLEIK
14I
















TABLE J







Antibody AB129











Seq.


06-AB-129
Sequence
ID No.





VH FW1
QVQLVESGGGLVQPGGSLRLSCSASQFIL
 1J





VH CDR1
SNYWVH
 2J





VH FW2
WVRQVPGEGLVWVS
 3J





VH CDR2
RINEDGSVTSYADSVKG
 4J





VH FW3
RFTISRDNAKNTLYLQMNSLRVDDTAVYYCVR
 5J





VH CDR3
DLCGERDD
 6J





VH FW4
WGQGTLVTVSS
 7J





VL FW1
VIWMTQSPLSLPVTPGEAASISC
 8J





VL CDR1
RSSQSLLHRNGRTFFA
 9J





VL FW2
WYLQKPGQSPQILIY
10J





VL CDR2
LGSNRAF
11J





VL FW3
GVPDRFSGSGSGTDFTLKISRVEAEDVGIYYC
12J





VL CDR3
MQGLQTPYT
13J





VL FW4
FGQGTKLEIK
14J
















TABLE K







Antibody AB130











Seq.


06-AB-130
Sequence
ID No.





VH FW1
QLQLQESGGGLVQPGGSLRLSCSASQFIL
 1K





VH CDR1
SNYWVH
 2K





VH FW2
WVRQVPGEGLVWVS
 3K





VH CDR2
RINEDGSVTSYADSVKG
 4K





VH FW3
RFTISRDNAKNTLYLQMNSLRVDDTAVYYCVR
 5K





VH CDR3
DLCWERDD
 6K





VH FW4
WGQGTLVSVSS
 7K





VL FW1
VIWMTQSPLSLPVTPGEAASISC
 8K





VL CDR1
RSSQSLLHRNGRTFFA
 9K





VL FW2
WYLQKPGQSPQILIY
10K





VL CDR2
LGSNRAF
11K





VL FW3
GVPDRFSGSGSGTDFTLKISRVEAEDVGIYYC
12K





VL CDR3
MQGLQTPYT
13K





VL FW4
FGQGTKLEIK
14K
















TABLE L







Antibody AB131











Seq.


06-AB-131
Sequence
ID No.





VH FW1
QVQLVQSGGGVVQPGGSLRLSCAASPFTF
 1L





VH CDR1
KTSILH
 2L





VH FW2
WVRQAPGKGLEWVS
 3L





VH CDR2
AMSYDGFSKYYADSVKG
 4L





VH FW3
RLTISRDSSTNTLYLEMNSLRFEDTALYFCAR
 5L





VH CDR3
EAYTSGRAGCFNP
 6L





VH FW4
WGQGVLVSVSS
 7L





VL FW1
DIVMTQTPSTQSASVGDRVTITC
 8L





VL CDR1
RASQSISIWLA
 9L





VL FW2
WYQQKPGKAPKLLIH
10L





VL CDR2
DASTLES
11L





VL FW3
GVPSRFSGSGSGTEFTLTISSLQPDDSATYYC
12L





VL CDR3
QRYNDYPPT
13L





VL FW4
FGPGTKVEIK
14L
















TABLE M







Antibody AB132











Seq.


06-AB-132
Sequence
ID No.





VH FW1
QVLKESGGGVVQPGGSLRLSCAASPFTF
 1M





VH CDR1
ETSILH
 2M





VH FW2
WVRQAPGKGLEWVS
 3M





VH CDR2
AMSYDGFSKYYADSVKG
 4M





VH FW3
RLTISRDSSTNTLYLEMNSLRFEDTALYFCAR
 5M





VH CDR3
EAYTSGRAGCFDP
 6M





VH FW4
WGQGVLVSVSS
 7M





VL FW1
EIVMTQSPSTQSASVGDRVTITC
 8M





VL CDR1
RASQSISIWLA
 9M





VL FW2
WYQQKPGKAPKLLIH
10M





VL CDR2
DASTLES
11M





VL FW3
GVPSRFSGSGSGTEFTLTISSLQPDDSATYYC
12M





VL CDR3
QRYNDYPPT
13M





VL FW4
FGPGTKVEIK
14M
















TABLE N







Antibody AB133











Seq.


06-AB-133
Sequence
ID No.





VH FW1
EVQLVESGGGLVQPGGSLRVSCAASGFTL
 1N





VH CDR1
NTYWMH
 2N





VH FW2
WVRQAPGKGLVWVS
 3N





VH CDR2
RINEDGTTISYADSVRG
 4N





VH FW3
RFTISRDNAENTLYLQMHSLRAEDTGVYYCAR
 5N





VH CDR3
DFTGPFDS
 6N





VH FW4
WGQGTLVSVSS
 7N





VL FW1
QSVLTQPPSVSGTPGQRVTISC
 8N





VL CDR1
SGSNSNAGRDYVS
 9N





VL FW2
WYQQVPGTAPKLLIY
10N





VL CDR2
KNNQRPS
11N





VL FW3
GVPDRFSGSKSGTSASLAISGLRSEDDGDYYC
12N





VL CDR3
IVWDGSLSGYV
13N





VL FW4
FGTGTKVTVL
14N
















TABLE O







Antibody AB134











Seq.


06-AB-134
Sequence
ID No.





VH FW1
QLQLQESGGGLVQPGGSLRLSCVVSGFTF
 1O





VH CDR1
SSHAMS
 2O





VH FW2
WVRQAPGKGLEWVS
 3O





VH CDR2
SISISGGDTFYADSVRG
 4O





VH FW3
RFTIFRDNSKNTVYLQMNSLRAEDTAVYYCAT
 5O





VH CDR3
ETSPNDY
 6O





VH FW4
WGQGTLVSVSS
 7O





VL FW1
SYELTQPSSLTVSPGGTVTLTC
 8O





VL CDR1
GLSSGAVTSGHYPY
 9O





VL FW2
WFQQKPGQAPKTLIF
10O





VL CDR2
DTSRKHS
11O





VL FW3
WTPARFSGSLLGGKAALTLSGAQPEDDADYYC
12O





VL CDR3
LLACNGACV
13O





VL FW4
FGGGTKLTVL
14O
















TABLE P







Antibody AB135











Seq.


06-AB-135
Sequence
ID No.





VH FW1
EVQLVETGGGLVQPGGSLRLSCVVSGFTF
 1P





VH CDR1
SSHAMS
 2P





VH FW2
WVRQAPGKGLEWVS
 3P





VH CDR2
SISISGGDTFYADSVRG
 4P





VH FW3
RFTIFRDNSKNTVYLQMNSLRAEDTAVYYCAT
 5P





VH CDR3
ETSPNDY
 6P





VH FW4
WGQGTLVTVSS
 7P





VL FW1
SYELTQPSSLTVSPGGTVTLTC
 8P





VL CDR1
GLSSGAVTSGHYPY
 9P





VL FW2
WFQQKPGQAPKTLIF
10p





VL CDR2
DTSRKHS
11P





VL FW3
WTPARFSGSLLGGKAALTLSGAQPEDDADYYC
12P





VL CDR3
LLACNGACV
13P





VL FW4
FGGGTKLTVL
14P
















TABLE Q







Antibody AB139











Seq.


06-AB-139
Sequence
ID No.





VH FW1
EVQLVESGGGLVQPGGSLRVSCAASGFTL
 1Q





VH CDR1
NTYWMH
 2Q





VH FW2
WVRQAPGKGLVWVS
 3Q





VH CDR2
RINEDGTTISYADSVRG
 4Q





VH FW3
RFTISRDNAENTLYLQMHSLRAEDTGVYYCAR
 5Q





VH CDR3
DFTGPFDS
 6Q





VH FW4
WGQGTLVSVSS
 7Q





VL FW1
QSVLTQPPSVSGTPGQRVTISC
 8Q





VL CDR1
SGSNSNVGRDYVS
 9Q





VL FW2
WYQQVPGTAPKLLIY
10Q





VL CDR2
KNNRRPS
11Q





VL FW3
GVPDRFSGSKSGTSASLAISGLRSEDDGDYYC
12Q





VL CDR3
IVWDGSLSGYV
13Q





VL FW4
FGTGTKVTVL
14Q
















TABLE R







Antibody AB140











Seq.


06-AB-140
Sequence
ID No.





VH FW1
EVQLVESGGGLVQPGGSLRVSCAASGFTL
 1R





VH CDR1
NTYWMH
 2R





VH FW2
WVRQAPGKGLVWVS
 3R





VH CDR2
RINEDGTTISYADSVRG
 4R





VH FW3
RFTISRDNAENTLYLQMHSLRAEDTGVYYCAR
 5R





VH CDR3
DFTGPFDS
 6R





VH FW4
WGQGTLVSVSS
 7R





VL FW1
QLVLTQPPSVSGTPGQRVTISC
 8R





VL CDR1
SGSNSNVGRDYVS
 9R





VL FW2
WYQQVPGTAPKLLIY
10R





VL CDR2
KNNQRPS
11R





VL FW3
GVPDRFSGSKSGTSASLAISGLRSEDDGDYYC
12R





VL CDR3
IVWDGSLSGYV
13R





VL FW4
FGTGTKVTVL
14R




















TABLE VH-CDR3-MOD








SEQ
(Variant




Light or
ID
of SEQ




Heavy CDR3
NO
ID NO)








06-AB-118.Heavy
DLAGERDD
17B
6B



C101A









06-AB-118.Heavy
DLSGERDD
18B
6B



C101S









06-AB-127.HeavyWY
DWGRLGYWSSNNY
17I
6I




PDAFDV








06-AB-127.HeavyAA
DWGRLGYASSNNA
18I
6I




PDAFDV








06-AB-131.HeavyW
EAYTSGRAGWFNP
17L
6L






06-AB-131.HeavyA
EAYTSGRAGAFNP
18L
6L






06-AB-132.HeavyW
EAYTSGRAGWFDP
17M
6M






06-AB-132.HeavyA
EAYTSGRAGAFDP
18M
6M






06-AB-129.HeavyW
DLWGERDD
17J
6J






06-AB-129.HeavyA
DLAGERDD
18J
6J




















TABLE VL-CDR3-MOD








06-AB-134.LightYW
LLAYNGAWV
19O
13O






06-AB-134.LightAA
LLAANGAAV
20O
13O






06-AB-135.LightYW
LLAYNGAWV
19P
13P






06-AB-135.LightAA
LLAANGAAV
20P
13P









REFERENCES

1. Brown G D. Innate antifungal immunity: The key role of phagocytes. Annu. Rev. Immuol. 29, 1-21 (2011).


2. Lockhart S R. Current epidemiology of Candida infection. Clin. Microbiol. Newsl. 36, 131-136 (2014).


3. Kim J, Sudbery P. Candida albicans, a major human fungal pathogen. J. Microbiol. 49, 171-177 (2011).


4. Gow N A R, Van De Veerdonk F L, Brown A J P, Netea M G. Candida albicans morphogenesis and host defence: Discriminating invasion from colonization. Nat. Rev. Microbiol. 10, 112-122 (2012).


5. Ellepola A N B, Morrison C J. Laboratory diagnosis of invasive candidiasis. J. Microbiol. 43, 65-84 (2005).


6. Ostrosky-Zeichner L. Invasive mycoses: Diagnostic challenges. Am. J. Med. 125, S14-24 (2012).


7. Perfect J R. Fungal diagnosis: How do we do it and can we do better? Curr. Med. Res. Opin. 29, 3-11 (2013).


8. Pfaller M A. Antifungal drug resistance: mechanisms, epidemiology, and consequences for treatment. Am. J. Med. 125(1 SUPPL.), S3-S13 (2012).


9. Rader C. Chemically programmed antibodies. Trends. Biotechnol. 32, 186-97 (2014).


10. Yoon S, Kim Y-, Shim H, Chung J. Current perspectives on therapeutic antibodies. Biotechnol. Bioprocess Eng. 15, 709-15 (2010).


11. Carter P J. Introduction to current and future protein therapeutics: a protein engineering perspective. Exp. Cell Res. 317, 1261-1269 (2011).


12. Berry J D, Gaudet R G. Antibodies in infectious diseases: polyclonals, monoclonals and niche biotechnology. New Biotech. 28, 489-501 (2011).


13. Saylor C, Dadachova E, Casadevall A. Monoclonal antibody-based therapies for microbial diseases. Vaccine. 27, G38-46 (2009).


14. Wilson P C, Andrews S F. Tools to therapeutically harness the human antibody response. Nat. Rev. Immunol. 12, 709-19 (2012).


15. Casadevall A, Pirofski L A. Immunoglobulins in defense, pathogenesis, and therapy of fungal diseases. Cell Host Microbe. 11, 447-56 (2012).


16. Dromer F, Salamero J, Contrepois A, Carbon C, Yeni P. Production, characterization, and antibody specificity of a mouse monoclonal antibody reactive with cryptococcus neoformans capsular polysaccharide. Infect. Immun. 55, 742-748 (1987).


17. Gigliotti F, Hughes W T. Passive immunoprophylaxis with specific monoclonal antibody confers partial protection against pneumocystis carinii pneumonitis in animal models. J. Clin. Invest. 81, 1666-1668 (1988).


18. Brena S, Cabezas-Olcoz J, Moragues M D, Fernández De Larrinoa I, Dominguez A, Quindós G, et al. Fungicidal monoclonal antibody C7 interferes with iron acquisition in Candida albicans. Antimicrob. Agents Chemother. 55, 3156-3163 (2011).


19. Moragues M D, Omaetxebarria M J, Elguezabal N, Sevilla M J, Conti S, Polonelli L, et al. A monoclonal antibody directed against a Candida albicans cell wall mannoprotein exerts three anti-C. albicans activities. Infect. Immun. 71, 5273-5279 (2003).


20. Torosantucci A, Bromuro C, Chiani P, De Bernardis F, Berti F, Galli C, et al. A novel glyco-conjugate vaccine against fungal pathogens. J. Exp. Med. 202, 597-606 (2005).


21. Torosantucci A, Chiani P, Bromuro C, De Bernardis F, Palma AS, Liu Y, et al. Protection by anti-β-glucan antibodies is associated with restricted β-1,3 glucan binding specificity and inhibition of fungal growth and adherence. PLoS ONE. 4, e5392 (2009).


22. Xin H, Cutler J E. Vaccine and monoclonal antibody that enhance mouse resistance to candidiasis. Clin. Vaccine Immunol. 18, 1656-1667 (2011).


23. Cassone A. Vulvovaginal Candida albicans infections: Pathogenesis, immunity and vaccine prospects. BJOG. doi: 10.1111/1471-0528.12994.(2014).


24. Cassone A. Development of vaccines for Candida albicans: fighting a skilled transformer. Nat. Rev. Microbiol. 11, 884-91 (2013).


25. Bromuro C, Romano M, Chiani P, Berti F, Tontini M, Proietti D, et al. Beta-glucan-CRM197 conjugates as Candidates antifungal vaccines. Vaccine. 28, 2615-23 (2010).


26. Schmidt C S, White C J, Ibrahim A S, Filler S G, Fu Y, Yeaman M R, et al. NDV-3, a recombinant alum-adjuvanted vaccine for Candida and Staphylococcus aureus, is safe and immunogenic in healthy adults. Vaccine. 30, 7594-7600 (2012).


27. Han Y, Morrison R P, Cutler J E. A vaccine and monoclonal antibodies that enhance mouse resistance to Candida albicans vaginal infection. Infect. Immun. 66, 5771-5776 (1998).


28. Ibrahim A S, Luo G, Gebremariam T, Lee H, Schmidt C S, Hennessey J P, et al. NDV-3 protects mice from vulvovaginal candidiasis through T- and B-cell immune response. Vaccine. 31, 5549-56 (2013).


29. Luo G, Ibrahim A S, French S W, Edwards Jr. J E, Fu Y. Active and passive immunization with rHyr1p-N protects mice against hematogenously disseminated candidiasis. PLoS ONE. 6, e25909. (2011).


30. Thornton C, Johnson G, Agrawal S. Detection of invasive pulmonary aspergillosis in haematological malignancy patients by using lateral-flow technology. J. Vis. Exp. 61, pii: 3721. doi: 10.3791/3721 (2012).


31. Thornton C R. Development of an immunochromatographic lateral-flow device for rapid serodiagnosis of invasive aspergillosis. Clin. Vaccine Immunol. 15, 1095-1105 (2008).


32. Martinez-Jiménez M C, Muñoz P, Guinea J, Valerio M, Alonso R, Escribano P, et al. Potential role of Candida albicans germ tube antibody in the diagnosis of deep-seated candidemia. Med. Mycol. 52, 270-275 (2014).


33. Jarvis J N, Percival A, Bauman S, Pelfrey J, Meintjes G, Williams G N, et al. Evaluation of a novel point-of-care cryptococcal antigen test on serum, plasma, and urine from patients with HIV-associated cryptococcal meningitis. Clin. Infect. Dis. 53, 1019-1023 (2011).


34. Carter P J. Potent antibody therapeutics by design. Nat. Rev. Immunol. 6, 343-357 (2006).


35. Tiller T. Single B cell antibody technologies. N. Biotechnol. 28, 453-457 (2011).


36. Smith K, Garman L, Wrammert J, Zheng N Y, Capra J D, Ahmed R, et al. Rapid generation of fully human monoclonal antibodies specific to a vaccinating antigen. Nat. Protoc. 4, 372-384 (2009).


37. Liao H X, Levesque M C, Nagel A, Dixon A, Zhang R, Walter E, et aL High-throughput isolation of immunoglobulin genes from single human B cells and expression as monoclonal antibodies. J. Virol. Methods. 158, 171-179 (2009).


38. Huang J, Doria-Rose N A, Longo N S, Laub L, Lin C-, Turk E, et al. Isolation of human monoclonal antibodies from peripheral blood B cells. Nat. Protoc. 8, 1907-1915 (2013).


39. Jiang X, Suzuki H, Hanai Y, Wada F, Hitomi K, Yamane T, et al. A novel strategy for generation of monoclonal antibodies from single B cells using RT-PCR technique and in vitro expression. Biotechnol. Prog. 22, 979-88 (2006).


40. Bailey D A, Feldmann P J F, Bovey M, Gow N A R, Brown A J P. The Candida albicans HYR1 gene, which is activated in response to hyphal development, belongs to a gene family encoding yeast cell wall proteins. J. Bacteriol. 178, 5353-5360 (1996).


41. Luo G, Ibrahim A S, Spellberg B, Nobile C J, Mitchell A P, Fu Y. Candida albicans Hyr1p confers resistance to neutrophil killing and is a potential vaccine target. J. Infect. Dis. 201, 1718-1728 (2010).


42. Lewis L E, Bain J M, Lowes C, Gillespie C, Rudkin F M, Gow N A R, et al. Stage specific assessment of Candida albicans phagocytosis by macrophages identifies cell wall composition and morphogenesis as key determinants. PLoS Pathog. 8, e1002578 (2012).


43. Rudkin F M, Bain J M, Walls C, Lewis L E, Gow N A R, Erwig L P. Altered dynamics of Candida albicans phagocytosis by macrophages and PMNs when both phagocyte subsets are present. mBio. 4, e00810-13. doi: 10.1128/mBio.00810-13 (2013).


44. MacCallum D M, Coste A, Ischer F, Jacobsen M D, Odds F C, Sanglard D. Genetic dissection of azole resistance mechanisms in Candida albicans and their validation in a mouse model of disseminated infection. Antimicrob. Agents Chemother. 54, 1476-1483 (2010).


45. Chames P, Van Regenmortel M, Weiss E, Baty D. Therapeutic antibodies:


successes, limitations and hopes for the future. Br. J. Pharmacol. 157, 220-233 (2009).


46. Beyda N D, Regen S, Lewis R E, Garey K W. Immunomodulatory agents as adjunctive therapy for the treatment of resistant Candida species. Curr. Fungal Infect. Rep. 7, 119-125 (2013).


47. Overdijk M B, Verploegen S, Buijsse A O, Vink T, Leusen J H W, Bleeker W K, et al. Crosstalk between human IgG isotypes and murine effector cells. J. Immunol. 189, 3430-3438 (2012).


48. Reichert J M. Marketed therapeutic antibodies compendium. mAbs. 4, 413-415 (2012).


49. Brown G D, Denning D W, Gow N A R, Levitz S M, Netea M G, White T C. Hidden killers: human fungal infections. Sci. Transl. Med. 4, 165rv13 (2012).


50. d'Enfert C, Goyard S, Rodriguez-Arnaveilhe S, Frangeul L, Jones L, Tekaia F, et al. CandidaDB: A genome database for Candida albicans pathogenomics. Nucleic Acids Res. 33(DATABASE ISS.), D353-357 (2005).


51. Yapar N. Epidemiology and risk factors for invasive candidiasis. Ther. Clin. Risk


Manag. 10, 95-105 (2014).


52. MacCallum D M. Hosting infection: experimental models to assay Candida virulence. Int. J. Microbiol. 2012, 363764 (2012).


53. Durocher Y, Perret S, Kamen A. High-level and high-throughput recombinant protein production by transient transfection of suspension-growing human 293-EBNA1 cells. Nucleic Acids Res. 30, e9 (2002).


54. McKenzie C G J, Koser U, Lewis L E, Bain J M, Mora-Montes H M, Barker R N, et al. Contribution of Candida albicans cell wall components to recognition by and escape from murine macrophages. Infect. Immun. 78, 1650-1658 (2010).


Additional references for Table S1:


Brand A, MacCallum D M, Brown A J, Gow N A, Odds F C. Ectopic expression of URA3 can influence the virulence phenotypes and proteome of Candida albicans but can be overcome by targeted reintegration of URA3 at the RPS10 locus. Eukaryot Cell 3: 900-909. doi: 10.1128/ec.3.4.900-909.2004 (2004).


Bailey D A, Feldmann P J F, Bovey M, Gow N A R, Brown A J P. The Candida albicans HYR1 gene, which is activated in response to hyphal development, belongs to a gene family encoding yeast cell wall proteins. J. Bacteriol. 178, 5353-5360 (1996).


Fonzi, W. A., Irwin M. Y. Isogenic strain construction and gene mapping in Candida albicans. Genetics 134, 717-728 (1993).


Gillum A M, Tsay E Y, Kirsch D R. Isolation of the Candida albicans gene for orotidine-5′-phosphate decarboxylase by complementation of S. cerevisiae ura3 and E. coli pyrF mutations. Mol Gen Genet. 198:179-8 (1984).


Odds F C, Hanson M F, Davidson A D, Jacobsen M D, Wright P, Whyte J A, Gow N A, Jones B L. One year prospective survey of Candida bloodstream infections in Scotland. J Med Microbiol. 56: 1066-1075 (2007).


Rudek W. Esterase activity in Candida species. J. Clin. Microbiol. 8: 756-759 (1978).


Moran G P, Sanglard D, Donnelly S M, Shanley D B, Sullivan D J, Coleman D C. Identification and expression of multidrug transporters responsible for fluconazole resistance in Candida dubliniensis. Antimicrob. Agents Chemother. 42:1819-1830 (1998).


Moran, G. P., Sanglard D., Donnelly S. M., Shanley D. B., Sullivan D. J., Coleman D. C. Identification and expression of multidrug transporters responsible for fluconazole resistance in Candida dubliniensis. Antimicrob. Agents Chemother. 42, 1819-1830 (1998).


Netea, M. G. et al. Aspergillus fumigatus evades immune recognition during germination through loss of toll-like receptor-4-mediated signal transduction. J. Infect. Dis. 188, 320-326 (2003).


Satoh, K., Makimura K., Hasumi Y., Nishiyama Y., Uchida K., Yamaguchi H. Candida auris sp. nov., a novel ascomycetous yeast isolated from the external ear canal of an inpatient in a Japanese hospital. Microbiol. Immunol . 53, 41-44 (2009).


Khan, Z. U. et al. Outbreak of fungemia among neonates caused by Candida haemulonii resistant to amphotericin B, itraconazole, and fluconazole. J. Clin. Microbiol. 45, 2025-2027 (2007).


Nielsen, K., Cox G. M., Wang P., Toffaletti D. L., Perfect J. R., Heitman J. Sexual cycle of Cryptococcus neoformans var. grubii and Virulence of congenic a and a isolates. Infect. Immun. 71, 4831-4841 (2003).


Fyfe, M., W. Black and M. Romney. Unprecedented outbreak of Cryptococcus neoformans var. gattii infections in British Columbia, Canada. Abstracts of the 5th International Conference on Cryptococcus and Cryptococcosis (2002).


Sugita, T. et al. New yeast species, Malassezia dermatis, isolated from patients with atopic dermatitis. J. Clin. Microbiol. 40, 1363-1367 (2002).


Li, C. H. et al. Sporangiospore size dimorphism is linked to virulence of Mucor circinelloides. PLoS Pathogens 7, e1002086 (2011).

Claims
  • 1.-3. (canceled)
  • 4. An anti-Candida recombinant human antibody molecule which comprises a VH domain comprising (i) an HCDR3 having the amino acid sequence of SEQ ID NO: 6x or the sequence of SEQ ID NO: 6x with 1, 2, or 3 amino acid substitutions, deletions or insertions; and optionally(ii) an HCDR2 having the amino acid sequence of SEQ ID NO: 4x or the sequence of SEQ ID NO: 4x with 1, 2, or 3 amino acid substitutions, deletions or insertions; and optionally(iii) an HCDR1 having the amino acid sequence of SEQ ID NO: 2x or the sequence of SEQ ID NO: 2x with 1, 2 or 3 amino acid substitutions, deletions or insertions, wherein ‘x’ is one letter from A to R, and said sequence is as shown in Table ‘x’ herein.
  • 5. (canceled)
  • 6. (canceled)
  • 7. An antibody molecule according to claim 4 wherein the antibody molecule comprises a VH domain comprising a HCDR1, a HCDR2 and a HCDR3 having the sequences of SEQ ID NOs 2x, 4x and 6x respectively.
  • 8. An antibody molecule according to claim 7 wherein the antibody molecule comprises a VH domain comprising one or more or all of a FW1, a FW2, a FW3 and a FW4 having the sequences of SEQ ID NOs 1x, 3x, 5x and 7x respectively.
  • 9. An antibody molecule according to claim 4 wherein the antibody molecule comprises a VH domain having an amino acid sequence at least about 80% identical to SEQ ID NO: 15x and\or having the amino acid sequence of SEQ ID NO: 15x and\or the sequence of SEQ ID NO: 15x with 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid substitutions, deletions or insertions in SEQ ID NO: 15x.
  • 10. An antibody molecule according to claim 4 wherein antibody molecule comprises a VL domain comprising LCDR1, LCDR2 and LCDR3 having the sequences of SEQ ID NOs 9x, 11x and 13x respectively, or the sequences of SEQ ID NOs 9x, 11x and 13x respectively with, independently, 1, 2 or 3 or more amino acid substitutions, deletions or insertions.
  • 11. An antibody molecule according to claim 10 wherein antibody molecule comprises a VL domain comprising LCDR1, LCDR2 and LCDR3 having the sequences of SEQ ID NOs 9x, 11x and 13x respectively.
  • 12. An antibody molecule according to claim 10 wherein the antibody molecule comprises a VL domain comprising one or more or all of a FW1, a FW2, a FW3 and a FW4 having the sequences of SEQ ID NOs 8x, 10x, 12x and 14x respectively.
  • 13. An antibody molecule according to claim 4 wherein the antibody molecule comprises a VL domain having an amino acid sequence at least about 80% identical to SEQ ID NO: 16x and\or having the sequence of SEQ ID NO: 16x and\or the sequence of SEQ ID NO: 16x with 1 or more, for example 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acid substitutions, deletions or insertions in SEQ ID NO: 16x.
  • 14. An antibody molecule according to claim 4 comprising a VH domain comprising a HCDR1, a HCDR2 and a HCDR3 having the sequences of SEQ ID NOs 2x, 4x, and 6x, 17x or 18x, respectively, and a VL domain comprising a LCDR1, a LCDR2 and a LCDR3 having the sequences of SEQ ID NOs 9x, 11x and 13x, 19x or 20x, respectively.
  • 15. An antibody molecule according to claim 4 comprising VH and VL domains having the amino acid sequences of SEQ ID NO: 15x and SEQ ID NO: 16x respectively.
  • 16. An antibody molecule according to claim 4 which binds C. albicans with an EC50 value of 1 to 1500 ng/ml, wherein ‘x’ is A-B, or H-R.
  • 17. (canceled)
  • 18. An antibody molecule according to claim 4 further comprising a payload which is cytotoxic.
  • 19. A method for producing an antibody antigen-binding domain for a fungal antigen, the method comprising; (i) providing, by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VH domain comprising HCDR1, HCDR2 and HCDR3, wherein the parent VH domain HCDR1, HCDR2 and HCDR3 have the amino acid sequences of SEQ ID NOS: 2x, 4x and 6x respectively, a VH domain which is an amino acid sequence variant of the parent VH domain,(ii) optionally combining the VH domain thus provided with one or more VL domains to provide one or more VH/VL combinations; and(iii) testing said VH domain which is an amino acid sequence variant of the parent VH domain or the VH/VL combination or combinations to identify an antibody antigen binding domain for the fungal antigen.
  • 20. A method for producing an antibody molecule that specifically binds to a fungal antigen, which method comprises: providing starting nucleic acid encoding a VH domain or a starting repertoire of nucleic acids each encoding a VH domain, wherein the VH domain or VH domains either comprise a HCDR1, HCDR2 and/or HCDR3 to be replaced or lack a HCDR1, HCDR2 and/or HCDR3 encoding region; combining said starting nucleic acid or starting repertoire with donor nucleic acid or donor nucleic acids encoding or produced by mutation of the amino acid sequence of an HCDR1, HCDR2, and/or HCDR3 having the amino acid sequences of SEQ ID NOS: 2x, 4x and 6x respectively, such that said donor nucleic acid is or donor nucleic acids are inserted into the CDR1, CDR2 and/or CDR3 region in the starting nucleic acid or starting repertoire, so as to provide a product repertoire of nucleic acids encoding VH domains;expressing the nucleic acids of said product repertoire to produce product VH domains;optionally combining said product VH domains with one or more VL domains; selecting an antibody molecule that binds the fungal antigen, which antibody molecule comprises a product VH domain and optionally a VL domain; andrecovering said antibody molecule or nucleic acid encoding it.
  • 21. An antibody molecule according to claim 4 which is a whole antibody or a scAb.
  • 22. A method comprising contacting a Candida cell with an antibody molecule as claimed in claim 4 for the purpose of: (i) identifying or labelling the Candida cell, or the hyphae of the cell; or(ii) opsonising, or increasing the rate of opsonisation of the Candida cell; or(iii) increasing the rate of macrophage engulfment of the Candida cell; or(iv) increasing the rate of macrophage attraction to the Candida cell.
  • 23.-25. (canceled)
  • 26. A pharmaceutical composition comprising an antibody molecule according to claim 4 and a pharmaceutically acceptable excipient optionally including a further antifungal agent.
  • 27.-30. (canceled)
  • 31. A method of treatment of a fungal infection comprising administering an antibody molecule according to claim 4 to an individual in need thereof.
  • 32. A method according to claim 31 wherein the fungal infection is a Candida infection, preferably a C. albicans, C. dubliniensis, C. tropicalis, C. parapsilosis or C. lusitaniae infection.
  • 33. A method according to claim 32 wherein the C. albicans infection is in a hyphal or yeast phase.
  • 34. A method according to claim 31, wherein the treatment comprises administering a second antifungal agent, wherein the second antifungal agent is optionally an azole, a polyene or an echinocandin.
  • 35. (canceled)
  • 36. A method for detecting the presence or absence of a fungus which is Candida spp, the method comprising (i) contacting a sample suspected of containing the fungus with an antibody molecule according to claim 4, and(ii) determining whether the antibody molecule binds to the sample, whereinbinding of the antibody molecule to the sample indicates the presence of the fungus.
  • 37. A method for diagnosing a fungal infection in an individual which is caused by Candida spp, the method comprising (i) contacting a biological sample obtained from the individual with an antibody molecule according to claim 4, and(ii) determining whether the antibody molecule binds to the biological sample, wherein binding of the antibody molecule to the biological sample indicates the presence of a fungal infection.
  • 38. A linear flow device (LFD) for detecting an analyte which is a fungal pathogen in a sample fluid, wherein said LFD comprises:
  • 39. (canceled)
  • 40. A device as claimed in claim 38 having a plurality of analyte-detecting means capable of distinguishing between multiple fungal pathogens, wherein one of the analyte-detecting means is an antibody molecule according to claim 4.
  • 41. A device as claimed in claim 40 wherein the multiple fungal pathogens comprise C. albicans, plus one or more or all of Aspergillus fumigatus, Cryptococcus neoformans, Pneumocystsis jirovecii, a zygomycete fungus, and a skin dermatophytic fungus.
  • 42. (canceled)
Priority Claims (1)
Number Date Country Kind
1503812.8 Mar 2015 GB national
PCT Information
Filing Document Filing Date Country Kind
PCT/GB2016/050577 3/4/2016 WO 00