Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

A Murine, Bispecific Monoclonal Antibody Simultaneously Recognizing β-Glucan and MP65 Determinants in Candida Species

  • Andrea Zito,

    Affiliation Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino and Transplant Immunology, Città della Salute e della Scienza, Torino, Italy

  • Carla Bromuro,

    Affiliation Istituto Superiore di Sanità, Department of Infectious, Parasitic and Immune-mediated Diseases, Rome, Italy

  • Giorgia Mandili,

    Affiliations Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino and Transplant Immunology, Città della Salute e della Scienza, Torino, Italy, Department of Molecular Biotechnology, University of Torino, Città della Salute e della Scienza, Torino, Italy

  • Paola Chiani,

    Affiliation Istituto Superiore di Sanità, Department of Infectious, Parasitic and Immune-mediated Diseases, Rome, Italy

  • Alberto L. Horenstein,

    Affiliation Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino and Transplant Immunology, Città della Salute e della Scienza, Torino, Italy

  • Fabio Malavasi,

    Affiliation Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino and Transplant Immunology, Città della Salute e della Scienza, Torino, Italy

  • Roberto Cauda,

    Affiliation Institute of Infectious Diseases, Catholic University of Rome, Rome, Italy

  • Antonio Cassone,

    Affiliation Center of Functional Genomics, Genetics and Biology, University of Perugia, Perugia, Italy

  • Antonella Torosantucci

    antonella.torosantucci@iss.it

    Affiliation Istituto Superiore di Sanità, Department of Infectious, Parasitic and Immune-mediated Diseases, Rome, Italy

Abstract

There is a real medical need of new diagnostic tools for the early recognition of invasive Candida infections. We exploited a rather simple and rapid redox methodology to construct a bispecific monoclonal antibody (bsmAb) that combines a monoclonal antibody (mAb) directed against 1,3-β-D-glucan, a well-known, pan-fungal diagnostic biomarker, with a mAb recognizing MP65, a major immunogenic mannoprotein secreted by C.albicans and other Candida species. The bsmAb (MP65/bglu mAb) was successfully produced and purified at high yields and proved to bind and reveal simultaneously, with high sensitivity, the β-glucan and MP65 antigens in both purified and native forms. The MP65/bglu mAb is the first bispecific antibody generated against a fungal microorganism and may prove useful for the concurrent detection of different and clinically significant Candida biomarkers in patient sera.

Introduction

Invasive fungal infections caused by C. albicans and other Candida species are a major cause of morbidity and mortality in hospitalized patients worldwide [1,2]. Despite the recent development and clinical availability of antifungal drugs of high efficacy and reduced toxicity, these infections remain difficult to control and their associated mortality rates are still unacceptably high, approaching 40–50% in some cases. This is further burdened by the high cost of treatment [1,2].

A main obstacle to an efficient clinical management of invasive candidiasis is the lack of diagnostic methodologies allowing the early detection and treatment of the disease [15]. Culture-based diagnostics typically are too insensitive and time-consuming. A number of noncultural assays, mostly based on the detection of fungal biomarkers in patient circulation, are making their way into the medical setting or are currently under investigation, but none have yet proved sufficiently sensitive and specific to detect the disease at a time when fungal burden is still relatively low and more responsive to antifungal therapy.

Several studies on the impact of treatment delays due to the time required for diagnosis show that patient mortality and hospitalization costs increase significantly for each day without anti-fungal therapy [35]. Clinicians and microbiologists agree that reliable tools for the early recognition of the fungal infection are a strong medical need [2], either as novel strategies for enhancing the sensitivity and specificity of current diagnostic methods or as new diagnostic assays.

Monoclonal antibodies (mAbs) are the gold standard for early detection of diagnostically relevant antigens. Highly specific and easily standardized, mAbs can reveal just picograms per milliliter of the specific target. The possibility of combining two pre-existing mAbs into one bispecific antibody (bsmAb)—a dual-Fab molecule recognizing both ligands of the mAbs of origin—has long been known [610]. To date, bsmAbs have been mostly investigated as candidate dual targeting therapeutics for treatment of cancer or inflammatory diseases; the potential of these reagents in the diagnosis of infectious diseases has remained almost unexplored [11,12].

The working hypothesis of this study is that the development of bsmAbs for the simultaneous detection of two different and significant Candida biomarkers would make it possible to design highly specific and sensitive assays for use in diagnosing invasive Candida infections. Our investigation exploited bsmAb technology to combine an anti-laminarin mAb, recognizing 1,3-β-D- and 1,6-β-D-glucan sequences, with a mAb directed against MP65, an immunodominant mannoprotein of C. albicans [1316].

Materials and Methods

MAb Production and Purification

MAb 2G8, an anti-laminarin mouse IgG2b [13,14], and 4C8, a murine IgG1 recognizing the protein moiety of the MP65 mannoprotein of C. albicans [15,16] were produced from culture supernatants of respective hybridoma cell lines. These were concentrated using a QuixStand benchtop system (GE Healthcare, Milano, Italy) equipped with a 30-kDa cutoff membrane and purified by high performance liquid chromatography (HPLC) on a rProtA MabSelect Sure column (GE Healthcare). To this aim, the concentrated supernatants were added with 112.6 g/l glycine, 175 g/l NaCl, and 3 g/l NaOH to achieve a pH of 8.9 and loaded onto the column at a 2 ml min−1 flow rate. Bound Igs were eluted with 3 column volumes of 0.1 M sodium citrate buffer, pH 5.8 (mAb 4C8) or pH 3.5 (mAb 2G8) and concentrated/diafiltrated against 10 mM sodium phosphate buffer, pH 6.7 using a Centricon Plus-80, 30-kDa MWCO, concentrator (Millipore, Milano, Italy) [17]. The samples were then processed by preparative chromatography on a Hydroxyapatite (HA) column (Bio-Rad, Milano, Italy) and eluted with a 110 min-linear gradient from 10 to 400 mM sodium phosphate buffer, pH 6.7, at a flow rate of 2 ml min−1. The pooled HA peak fractions containing the mAbs were concentrated and extensively dialyzed against phosphate-buffered saline (PBS), pH 7.2, using a C80 Centricon Plus (Millipore), and finally filtered through a 0.22-μm disposable hydrophilic Posidyne membrane syringe filter (Pall, Ann Arbor, MI, 25-mm diameter).

The IgG concentration was monitored and measured by absorbance at 280 nm throughout the entire process. Evaluation of retention times, peak heights and relative peak areas were made using the Gold Beckman software [18].

Redox Methodology

The 2G8/4C8 bsmAb was prepared by adapting the redox method described by Carling et al. [19]. To reduce the parental mAbs to monovalent antibody (mvAb), 2-mercaptoethanesulfonic acid sodium salt (Sigma-Aldrich, USA) was diluted in H2O and added to 1 mg of mAb 2G8 or to 1 mg of mAb 4C8, both diluted in PBS, pH 7.2, at the final concentration of 10 mM (mAb 2G8) or 30 mM (mAb 4C8). The samples were incubated at 37°C for 25 min. Re-association of mvAbs into bsmAbs was induced by mixing the reduced 2G8 and 4C8 mAbs (1:1 ratio), followed by oxidation through dialysis against 3 buffer exchanges of PBS, pH 7.2 (24 h at 4°C).

Purification and Characterization of the bsmAb

The bsmAb was purified by a two-step chromatography separation onto an HA column, after careful selection of appropriate fractionation gradient. Purification was performed at a flow-rate of 2 ml/min using 10 mM phosphate buffer, pH 6.7 as buffer A and 400 mM phosphate buffer, pH 6.7 as buffer B [18]. The eluted fractions from the first HA column were dialyzed against PBS and subsequently re-purified in a second HA column to isolate the peak containing the 2G8/4C8 bsmAb. The peak fractions corresponding to the active bsmAb were pooled, dialysed against sterile PBS and stored at -20°C.

SDS-PAGE analysis of purified bsmAb and parental mAbs was performed onto 10% polyacrylamide gels under reducing and non-reducing conditions, according to standard techniques [20]. Isotype class and subclass were analyzed by a standard immunodiffusion test [21], using goat anti-mouse IgG1, IgG2a, IgG2b, IgG3 and IgM antibodies, respectively (SouthernBiotech, Birmingham, AL).

2D-IEF analysis was performed using ready-made immobilized pH gradient (IPG) strips (7-cm IPG strips, pH 3–10). Each sample (250 μg of protein for preparative gels) was applied onto an IPG strip by in-gel rehydration for 20 h, with the addition of dithiothreitol (DTT) and Ampholine pH 3–10. IEF was carried out in a Protean IEF cell apparatus (Bio-Rad, Segrate, Italy). Focusing was performed at 18°C with a limit of 50 mA per strip. Subsequently, IPG strips were equilibrated for 15 min in 6 M urea, 2% SDS, 0.05 M Tris-HCl pH 8.8, 20% glycerol, 1% DTT buffer and for 12 min in 6 M urea, 2% SDS, 0.05 M Tris-HCl pH 8.8, 20% glycerol, 2.5% iodoacetamide buffer. For the second dimension, strips were run into 10% acrylamide gels in a Mini Protean system (Bio-Rad). Gels were stained with colloidal Coomassie blue G-250. Image analysis was performed using the PD-Quest software (version 7.2, Bio-Rad), according to the manufacturer’s instructions. Normalization of each individual spot was performed according to the total quantity of the valid spots in each gel, after subtraction of the background values. The spot volume was used as the analysis parameter to quantify protein expression.

Antigens

Laminarin was purchased from Sigma-Aldrich, USA. Recombinant MP65p, expressed in E.coli on the basis of the published sequence [22] and purified from bacterial culture supernatants, was produced by GeneScript, Piscataway, NJ. Concentrated and dyalized culture supernatants containing the antigenic material secreted by plastic-adherent, hyphal form-growing C.albicans (SAM) were obtained as described previously [13].

Biotin labelling of laminarin was performed essentially as described by Wang et al. [23]. Beta-1,6-linked, branch residues of the polysaccharide were oxidized by sodium metaperiodate (Sigma-Aldrich) and newly formed aldheyde groups were reacted with ethylenediamine (Sigma-Aldrich). Resulting Schiff base was stabilized by reduction with sodium borohydride (Sigma-Aldrich) and the aminated polysaccharide was finally reacted with the N-hydroxysuccinimide ester of biotin (BNHS, Sigma-Aldrich) and purified by precipitation with 95%, v/v ethanol in the cold and repeated washings. MP65p was biotinylated by direct reaction with BNHS in 1M carbonate buffer, pH 8.3 (ratio protein to BNHS, 10:1) and purified by diafiltration against PBS buffer in a 10 kDal MWCO ultrafiltration device (Millipore, Milano, Italy).

Analytical Determinations

Polysaccharide or protein content in the preparations of purified mAbs, bsmAb, biotinylated antigens or SAM were determined by the phenol-sulphuric acid method or by the MicroBCA Protein assay (Bio-Rad Laboratories), respectively, as previously reported [14].

Immunoenzymatic Assays

In indirect ELISA experiments, the various antigens were diluted in 0.05 M carbonate/bicarbonate buffer, pH 9.6 (laminarin, 50 μg/ml; MP65p, 5.0 μg/ml; SAM, 5.0 μg/ml protein, unless otherwise specified) and adsorbed onto polystyrene microtiter plates (MaxiSorp; NUNC). Plates were blocked with 3% bovine serum albumin (BSA, Fraction V, Sigma-Aldrich) in PBS and reacted with two-fold dilutions of the parental mAbs or bsmAb, followed by alkaline phosphatase-conjugated, anti-mouse IgG antibody (Sigma-Aldrich) and the enzyme substrate (p-nitrophenyl phosphate disodium, Sigma-Aldrich), as previously described [13]. Absorbance was read at 405 nm and readings from negative controls (wells non reacted with the mAbs or reacted with an irrelevant mAb) were subtracted from all absorbance values.

In capture ELISA, MaxiSorp plates were coated with an Fc-specific, goat anti-mouse IgG antibody (5 μg/ml in PBS, on, +4°C), washed and reacted with the bsmAb at the desired dilutions in PBS (1h, 37°C). After thorough washings and blocking with 3% BSA in PBS, the plate-bound bsmAb was reacted with different concentrations of biotinylated laminarin or MP65p for 1h at 37°C. Wells were washed again and the amount of bound antigen was evaluated by the addition of horse-radish peroxidase-conjugated streptavidin (Sigma-Aldrich) followed by 3,3′,5,5′-tetramethylbenzidine-H2O2 substrate solution (1-step Ultra-TMB ELISA, Thermo Scientific, USA). Wells were acidified by addition of 1M H2SO4 and absorbance read at 450 nm. Negative control wells (non reacted with the bsmAb or with the biotinylated antigens) were always run in parallel to evaluate background O.D. values.

Statistics

Linear regression analysis of bsmAb binding curves and Student’s t test were performed using the Prism 4 software (GraphPad, USA).

Results

Parental mAbs

To develop the MP65/bglu mAb we selected two mAbs that specifically recognize two different, disease-relevant, fungal antigens. The first (mAb 2G8) is a murine IgG2b specific for β-1,3-glucan, a cell wall component expressed and secreted by nearly all fungal pathogens [13,14]. The second mAb (4C8) is a mouse IgG1 that recognizes the protein moiety of MP65 (MP65p), a highly immunogenic mannoprotein involved in Candida cell wall maintenance and remodeling [15,16,24].

The parental mAbs were massively produced in cell cultures, and supernatants underwent all the steps for purification as described [17]. The purified mAbs maintained the purity grade and binding efficiency shown by reference mAb preparations, as assessed by SDS-PAGE analyses and ELISA testing (data not shown).

Production of the bsmAb

The construction of the MP65/bglu mAb relied upon a rapid method based on reduction-oxidation of the parental IgGs [19]. As illustrated in Fig 1, the method exploits the mild reducing agent 2-mercaptoethanesulfonic acid (MESNA) to break the inter-heavy chain bonds of the parental IgGs and to reduce them to monovalent mAbs (mvAbs). Subsequent dialysis under oxidizing conditions provided re-associations of the mvAbs to form bsmAb molecules.

thumbnail
Fig 1. Schematic diagram of MP65/bglu mAb generation by the redox method.

Modified from: Carlring et al., PLoS One. 2011;6: e22533.

https://doi.org/10.1371/journal.pone.0148714.g001

Preliminary experiments were performed to establish the optimal conditions of MESNA treatment for obtaining mvAbs from the parental 2G8 and 4C8 IgG. Different concentrations of MESNA and different incubation times were used to reduce the purified mAbs. The composition of the resulting products was then analyzed by non-reducing SDS-PAGE. The optimal reduction of the 2G8 mAb was obtained at a MESNA concentrations of 10 mM, as indicated by the presence of a sharp mvAb band at ~75 kDa in the electrophoretic profile of the final reaction mixture (Fig 2A). Instead, 30 mM MESNA yielded the highest amount of mvAb fragments, with a molecular weight of ~85 kDa, from the 4C8 mAb (Fig 2B). Both reduced mAb preparations showed only a faint band attributable to the intact mAb, confirming their high susceptibility to reduction at the conditions adopted (Fig 2A and 2B). MESNA reduction also led to the release of small amounts of free heavy and light mAb chains, which were almost totally removed by subsequent dialysis (Fig 2). These experiments defined the optimal incubation conditions for MESNA treatment were exposure of IgG at 37°C for 25 min (data not shown).

thumbnail
Fig 2. Tuning redox methodology for MP65/bglu mAb construction.

Panels A and B show the efficiency of different MESNA concentrations (0 to 50 mM, as indicated) in splitting mAb 2G8 (A) or mAb 4C8 (B) into mvAb fragments, as evaluated by their SDS-PAGE profiles under non-reducing conditions. The arrows indicate the presence of the whole mAb (1), mvAb (2), heavy chain (3), and light chain (4). Panel C shows a non-reducing SDS-PAGE analysis of the 2G8/4C8 mvAb mixture after oxidation by dialysis, demonstrating the re-association of mvAb fragments into whole Ig molecules. MW = molecular weight standards.

https://doi.org/10.1371/journal.pone.0148714.g002

Reassembling of mvAbs into bsmAbs was initiated by mixing equal amounts of reduced 2G8 and 4C8 mvAbs, followed by oxidation through dialysis against PBS [19]. SDS-PAGE analysis of the mvAbs mixture after the dialysis step showed an almost complete re-assemblage of the immunoglobulins, as shown by the presence of a major band at 150 kDa (Fig 2C). MvAbs re-associate randomly during the oxidizing reaction, hence the 150 kDa product represented a mixture of re-assembled original mAbs and newly formed bsmAb molecules (Fig 1).

MP65/bglu mAb Purification

The bsmAb was composed by two different and monovalent IgG moieties (mvIgG), each of which corresponded to one half of the parental IgGs (Fig 1). The resulting total net charge of this molecule (mvIgG1 x mvIgG2b) was thus different from that of the parental mAbs (mvIgG1 x mvIgG1 or mvIgG2b x mvIgG2b). HPLC on hydroxyapatite (HA) led to an efficient separation of the MP65/bglu mAb and the re-assembled 2G8 or 4C8 mAbs according to their individual charges [25]. Successful purification of the bsmAb, however, critically required the choice of gradients of appropriate ionic strength for the chromatographic separation, as to obtain significant differences in the retention times of the bsmAb and the parental mAbs. Gradients were selected by careful evaluation of the retention times of the parental mAbs in different conditions. As shown in Fig 3, HA chromatography allowed the purification of a definite peak attributable to the bsmAb, distinct from those originated by the parental 2G8 and 4C8 mAbs.

thumbnail
Fig 3. Purification of the MP65/bglu mAb by HA-ion-exchange chromatography.

Elution profiles of the 2G8 and 4C8 mAbs (Panel A), the 2G8 x 4C8 bsmAb (Panel B) and merge of all three profiles (Panel C); in C, the bsmAb peak is indicated by the dotted, red line. Chromatographic separation was performed on a 50 x 25 mm I.D. HA column at a flow rate of 2 ml/min. A 110 min linear gradient from 10 to 400 mM phosphate (dotted black line) was used. In B, dotted line inside of the bsmAb elution peak shows its separation into the two fractions 5/A and 5/B.

https://doi.org/10.1371/journal.pone.0148714.g003

To verify the identity of the MP65/bglu mAb peak, fractions corresponding to the ascending and descending part were collected separately and referred to as 5/A (ascending peak) and 5/B (descending peak) fraction (Fig 3C).

Classical Ouchterlony tests (Fig 4A) clearly showed that the two fractions were recognized by both anti-IgG1 and anti-IgG2b antibodies. Precipitin lines were of similar intensity, confirming that 5/A and 5/B were both a combined IgG1 x IgG2b complex.

thumbnail
Fig 4. Characterization of the 5/A and 5/B bsmAb fractions.

Panel A. Reactivity of 5/A and 5/B in the Ouchterlony double immunodiffusion assay. Central well 1 was loaded with fraction 5/A and central well 4 with fraction 5/B. Side well 2 contained a goat, anti-mouse IgG1 mAb and side well 3 a goat anti-mouse IgG2b mAb. Panel B. 2D-IEF analysis of the bsmAb fractions in comparison with the 2G8 and 4C8 parental mAbs. IEF separation was performed as described in Materials and Methods. Image analysis of the stained gels was performed using the PD-Quest software. Circle of the same color indicate corresponding protein spots in different mAb samples. Panel C. Binding of the bsmAb fractions to β-glucan and MP65p. Laminarin or MP65p adsorbed onto ELISA plates were reacted with fraction 5/A or 5/B or with the parental mAb 2G8 or 4C8, at the concentrations indicated. Binding of the Abs was revealed by an alkaline phosphatase-conjugated, anti-mouse IgG mAb followed by p-nitrophenyl phosphate disodium as the enzyme substrate. Binding is expressed as the mean, O.D. 405 nm readings from triplicate wells after subtraction of O.D. from the negative controls (wells with irrelevant Abs). The SD of all triplicate determinations was always <20% and is not shown in the graph.

https://doi.org/10.1371/journal.pone.0148714.g004

2D-IEF analysis showed that the fingerprints of the two fractions were almost identical in quantity and quality and substantially corresponded to the combination of the individual fingerprints exhibited by the parental mAbs (Fig 4B).

Finally, the fractions were assayed for reactivity with laminarin, a standard β-1,3 glucan polysaccharide, or with recombinant MP65p, used as solid-phase antigens in ELISA. The parental 2G8 and 4C8 mAbs were tested at the same concentrations as reactivity reference standards. The results demonstrated that both 5/A and 5/B were able to recognize plastic-adsorbed laminarin or MP65p, in a dose-dependent fashion (Fig 4C). Comparison of the binding efficiency of the two fractions, as exemplified by the final assay outputs produced (O.D. 405 nm), indicated an almost identical reaction patterns with MP65p and a slightly more efficiency in binding laminarin by 5/B (Fig 3C). However, the reactivity of the bsmAb fractions was comparable to that exhibited by the parental mAbs, at least at the highest doses.

Based on these results, subsequent experiments were performed using the entire bsmAb peak.

Validation of the bsmAb

Definitive validity of the construct was investigated by testing its ability to bind simultaneously and independently the epitopes recognized by the parental mAbs and by evaluating whether, and at what level of sensitivity, the MP65/bglu mAb was able to capture the free target antigens in solution. This was critical to envision the potential of the bsmAb as a single, specific reagent able to reveal as a whole both the β-glucan and MP65p diagnostic antigens in the sera of Candida infected patients.

First, the MP65/bglu mAb was assayed in indirect ELISA experiments against a combination of the antigens recognized by the parental mAbs as the plastic-adsorbed target (Fig 5). The combination was either a mixture of laminarin and recombinant MP65p or a concentrated culture supernatant of hyphal form-growing C.albicans (SAM), a preparation that is known to contain both the β-glucan and MP65 antigens as they are naturally expressed and secreted by the fungus [13,16,26]. Results from these experiments showed that reaction of the bsmAb with both the laminarin-MP65p mixture (Fig 5A) or the native fungal SAM (Fig 5B), at all doses tested, produced final O.D. readings approximately equivalent, if not slightly higher, to the sum of those produced by each parental mAb against its respective antigen. This clearly indicated that bsmAb could indeed bind the β-glucan and Mp65 antigens simultaneously and independently, as expected, with no loss of functional affinity with respect to the mAbs of origin and even in the form they are natively produced by fungal cells.

thumbnail
Fig 5. The bsmAb is able to bind contemporaneously and independently the β-glucan and MP65p antigens.

The ELISA plates were coated with a mixture of laminarin and MP65p (20 and 5.0 μg/ml, respectively) or with a concentrated culture supernatant of C.albicans hyphae (SAM, 5.0 μg/ml protein) containing naturally secreted β-glucan and MP65. The graph shows a comparison of mean O.D.405 nm values resulting upon reaction of the antigen mixtures with mAb 2G8, mAb 4C8 or bsmAb 2G8X4C8, at the indicated concentrations. SD between replicates was always <20% and is not reported.

https://doi.org/10.1371/journal.pone.0148714.g005

In all conditions tested, O.D. readings generated by the bsmAb were significantly higher (P<0.01, Student’s t test) than those generated by the mAb 2G8. Differences between readouts produced by the bsmAb and the mAb 4C8 were always statistically significant (P<0.05, Student t test) except when the two mAbs were compared at the concentration of 1μg/ml against the laminarin-MP65p mixture.

Following these experiments, we constructed ELISA assays in which the bsmAb was bound to the plate and evaluated for its ability to capture and reveal biotin-labelled laminarin or MP65p in solution. Checkerboard titration experiments showed that the bsmAb was efficient at capturing both antigens (Fig 6A and 6B). The outputs (final O.D. readings) of the assays were MP65/bglu.mAb- and antigen-dose dependent. Further, a good linearity was found for antigen dose-O.D. response curves obtained at all the different bsmAb concentrations assayed, with r2 values always approximating 1.0 (Fig 6A and 6B). Finally, the output of the experiments was readable at antigen concentrations as low as 50 pg/mL-1 (laminarin) or 150 pg/mL-1 (MP65p), demonstrating that the MP65/bglu mAb had a high avidity for both fungal biomarkers (Fig 6A and 6B). In other experiments (Fig 6C), the ELISA plate-bound MP65/bglu mAb was also reacted in parallel with different doses of biotinylated laminarin or MP65p, separate or mixed toghether. Similarly to what observed previously (see Fig 5), O.D. readings resulting upon capture of the antigen mixture roughly approximated the summed values generated by the capture of each single antigen, indicating that β-glucan and MP65p did not exert any mutual steric hindrance or generic interference in binding to the bsmAb (Fig 6C).

thumbnail
Fig 6. Antigen capture by the MP65/bglu mAb.

Panel A and B. Dose-response, β-glucan and MP65p capture curves. An Fc-specific, goat anti mouse IgG coating was used for the oriented binding of the bsmAb, at various concentrations, to the ELISA plates. Plate-bound bsmAb was then reacted with different doses of biotinylated laminarin (Panel A) or MP65p (Panel B). The amount of captured antigens was revealed by the addition of peroxidase-conjugated streptavidin followed by the enzyme substrate. O.D. 450 nm readings are the mean from duplicate wells after subtraction of O.D. values from the negative controls (wells with no antigen or no bsmAbs). R2 values of the resulting capture curves were calculated by linear regression analysis. Panel C. Dual antigen capture by the bsmAb. The ELISA plate-bound bsmAb (0.2 μgml) was reacted in comparison with different doses of biotinylated laminarin or biotinylated MP65p or with a mixture containing both biotynilated laminarin and MP65p. The ELISA assay was performed as described above for data in Panels A and B.

https://doi.org/10.1371/journal.pone.0148714.g006

Overall, these results suggested that the MP65/bglu mAb was very efficient and sensitive for the synchronous capture and revelation of the two fungal biomarkers.

Discussion

Despite the huge potentialities of bsmAbs for the detection and treatment of cancer and other human diseases, only few examples of bsmAb devised for anti-bacterial or anti-fungal therapy are reported in current literature [2732]. BsmAbs proposed for the diagnosis of human microbial infections are even fewer, and were all constructed to bind in combination a microbial antigen and various types of reporter molecule, in order to improve antigen recognition specificity in different immunoassays [3335].

We describe here the development and characterization of a novel, anti-Candida bsmAb (MP65/bglu mAb) designed for the dual-detection of two diverse, diagnostically relevant, fungal biomarkers in patient sera. The bsmAb was constructed from two previously described murine mAbs [13,14,16], that recognize two Candida antigens whose levels are known to increase in patient circulation in the course of the fungal infection. One of them is the pan-fungal polysaccharide β-glucan, a well-established disease biomarker that is currently assayed for the diagnosis of infections caused by different fungi, including Candida [36]. The companion antigen, MP65p, is a protein originally identified by us in C.albicans as the main fungal antigen targeted by the human cellular immune responses [15,16,37]. MP65p is secreted at very high levels, in vivo and in vitro, particularly by Candida hyphae which are the form of growth generally associated to infectious candidiasis [24,26]. Close homologues of this protein are also expressed and secreted by most of the pathogenic Candida species, such as C. guilliermondii, C. glabrata, C. parapsilosis and C. tropicalis [22]. Building upon our previous work, other authors have more recently proposed MP65p as a valid marker of Candida infection, although it is not yet used in clinical diagnostics [24].

Traditional methods of producing bsmAb rely on immunoglobulin G (IgG) heterocross-linking or the production of hybrid hybridomas [25]. Alternative methods, such as Fc-engineering [38], chemical redox [19,39] or both [4042], have been under development in the last few years. Hybrid hybridomas and engineering are highly sensitive as well as consistent, but they are time consuming and expensive. To construct the MP65/bglu mAb, we applied a manageable chemical redox methodology that has proven successful and time-saving for obtaining different types of bsmAbs, and that is particularly suitable for the production of bsmAb reagents for use in early pilot studies. Our results show that by coupling the redox strategy with HA purification, the production of bsmAbs from homospecific immunoglobulins of different isotypes is both simple and efficient. We were able to produce and purify the bsmAb with yields of up to 50%, which is twice that previously reported [19].

The bsmAb generated could simultaneously bind with high affinity the β-glucan and MP65p antigens. Indeed, both ELISA experiments with plastic-adsorbed antigen mixtures and ELISA dual-capture assays showed that the MP65/bglu mAb produced readout values that were essentially the sum of those individually produced by the two mAbs of origin. These results also indicate that, at least in our assay conditions, the overall avidity of the two bsmAb binding arms is preserved, if not increased, with respect to the two entire parental mAbs suggesting that assemblage of the 2G8 and 4C8 mAbs could enhance the affinity and/or avidity of one or both antigen binding sites in the resulting bispecific Ig. This might be caused by the structural diversity of the constant regions of the MP65/bglu mAb, that is a combined IgG1xIgG2 molecule, unlike the IgG1 and IgG2 mAbs of origin. In fact, it is well known that variations of the constant regions can significantly impact the affinity and even the fine specificity of Abs with identical antigen binding sites [43,44]. Moreover, an altered avidity of bsmAbs as compared to parental mAbs has been reported by several authors [4547].

ELISA capture curves revealed a high efficiency of the MP65/bglu mAb in detecting the fungal antigens. In particular, the sensitivity of the bsmAb effectively detected either β-glucan or MP65p at very low concentrations, such as those that are present and considered diagnostically significant in Candida-infected patients [24,36].

The standard β-glucan (laminarin) and the recombinant MP65p used in this investigation are mock antigens that express the epitopes recognized by the bmAb parental Igs but are structurally different from the native fungal antigens. Fungal β-glucan, for instance, is much more extensively branched than laminarin, while MP65p is released by fungal cells in a heavily mannosylated form. Therefore, the ability to recognize naturally released β-glucan and MP65p in hyphal secretion, a material that mimics the antigenic secretion produced by the infecting fungus in vivo, is another important and diagnostically relevant feature of our MP65/bglu mAb.

Early serological diagnosis of invasive candidiasis remains a real challenge. Specific approaches already attempted in this field have included the detection of fungal DNA using PCR-based methods or the analysis of circulating fungal biomarkers in patient sera [36]. PCR methodologies mostly proved difficult to standardize and are as yet experimental. Instead, commercial assays of 1,3-β-D-glucan or α-1,2-D-/α-1,6-D-mannan, two cell wall polysaccharides which increase in serum levels in the course of infection, are now commonly used in clinics [36]. These, however, have multiple drawbacks in terms of sensitivity and specificity and sometimes must be ran in combination to consolidate their diagnostic significance [36].

We consider that the MP65/bglu mAb produced in this work has the potential to contribute to the development of new, “two-in-one” biomarker assays for an earlier and more reliable diagnosis of candidiasis and to open the way to the use of bsmAbs as novel and potent reagents for the sensitive detection of infection biomarkers.

Author Contributions

Conceived and designed the experiments: AT AC RC FM AZ ALH. Performed the experiments: AZ CB GM PC. Analyzed the data: AT AC FM ALH AZ. Wrote the paper: AT AC FM RC AZ.

References

  1. 1. Delaloye J, Calandra T. Invasive candidiasis as a cause of sepsis in the critically ill patient. Virulence. 2014;5: 161–169. pmid:24157707
  2. 2. Clancy CJ, Nguyen MH. Finding the "missing 50%" of invasive candidiasis: how nonculture diagnostics will improve understanding of disease spectrum and transform patient care. Clin Infect Dis. 2013;56: 1284–1292. pmid:23315320
  3. 3. Moran C, Grussemeyer CA, Spalding JR, Benjamin DK Jr, Reed SD. Comparison of costs, length of stay, and mortality associated with Candida glabrata and Candida albicans bloodstream infections. Am J Infect Control. 2010;38: 78–80. pmid:19836856
  4. 4. Morrell M, Fraser VJ, Kollef MH. Delaying the empiric treatment of candida bloodstream infection until positive blood culture results are obtained: a potential risk factor for hospital mortality. Antimicrob Agents Chemother. 2005;49: 3640–3645. pmid:16127033
  5. 5. Garey KW, Rege M, Pai MP, Mingo DE, Suda KJ, Turpin RS, et al. Time to initiation of fluconazole therapy impacts mortality in patients with candidemia: a multi-institutional study. Clin Infect Dis. 2006;43: 25–31. pmid:16758414
  6. 6. De Monte L, Nistico P, Tecce R, Dellabona P, Momo M, Tarditi L, et al. Gene transfer by retrovirus-derived shuttle vectors in the generation of murine bispecific MAbs. Dev Biol Stand. 1990;71: 15–22. pmid:2401381
  7. 7. Malavasi F, De Monte LB, Funaro A, Magrini E, Bonino LD, Momo M, et al. Cell surface receptors and bispecific monoclonal antibodies: the link between basic science and medical oncology. Year Immunol. 1993;7: 74–80. pmid:8396823
  8. 8. Horenstein AL, Poiesi C, DeMonte L, Camagna M, Mariani M, Albertini A, et al. Real-time kinetic analysis applied to the production of bispecific monoclonal antibodies for radioimmunodetection of cancer. Int J Clin Lab Res. 1993;23: 199–205. pmid:8123875
  9. 9. Nistico P, Mortarini R, De Monte LB, Mazzocchi A, Mariani M, Malavasi F, et al. Cell retargeting by bispecific monoclonal antibodies. Evidence of bypass of intratumor susceptibility to cell lysis in human melanoma. J Clin Invest. 1992;90: 1093–1099. pmid:1387883
  10. 10. DeMonte LB, Nistico P, Tecce R, Dellabona P, Momo M, Anichini A, et al. Gene transfer by retrovirus-derived shuttle vectors in the generation of murine bispecific monoclonal antibodies. Proc Natl Acad Sci U S A. 1990;87: 2941–2945. pmid:2326256
  11. 11. Kontermann RE. Dual targeting strategies with bispecific antibodies. MAbs. 2012;4: 182–197. pmid:22453100
  12. 12. Byrne H, Conroy PJ, Whisstock JC, O'Kennedy RJ. A tale of two specificities: bispecific antibodies for therapeutic and diagnostic applications. Trends Biotechnol. 2013;31: 621–632. pmid:24094861
  13. 13. Torosantucci A, Chiani P, Bromuro C, De Bernardis F, Palma AS, Liu Y, et al. Protection by anti-beta-glucan antibodies is associated with restricted beta-1,3 glucan binding specificity and inhibition of fungal growth and adherence. PLoS One. 2009;4: e5392. pmid:19399183
  14. 14. 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. 2005;202: 597–606. pmid:16147975
  15. 15. Torosantucci A, Bromuro C, Gomez MJ, Ausiello CM, Urbani F, Cassone A. Identification of a 65-kDa mannoprotein as a main target of human cell-mediated immune response to Candida albicans. J Infect Dis. 1993;168: 427–435. pmid:8335981
  16. 16. Gomez MJ, Torosantucci A, Arancia S, Maras B, Parisi L, Cassone A. Purification and biochemical characterization of a 65-kilodalton mannoprotein (MP65), a main target of anti-Candida cell-mediated immune responses in humans. Infect Immun. 1996;64: 2577–2584. pmid:8698482
  17. 17. Horenstein AL, Durelli I, Malavasi F. Purification of clinical-grade monoclonal antibodies by chromatographic methods. Methods Mol Biol. 2005;308: 191–208. pmid:16082036
  18. 18. Horenstein AL, Crivellin F, Funaro A, Said M, Malavasi F. Design and scaleup of downstream processing of monoclonal antibodies for cancer therapy: from research to clinical proof of principle. J Immunol Methods. 2003;275: 99–112. pmid:12667674
  19. 19. Carlring J, De Leenheer E, Heath AW. A novel redox method for rapid production of functional bi-specific antibodies for use in early pilot studies. PLoS One. 2011;6: e22533. pmid:21811628
  20. 20. Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 1970;227: 680–685. pmid:5432063
  21. 21. Ouchterlony O. Antigen-antibody reactions in gels. IV. Types of reactions in coordinated systems of diffusion. Acta Pathol Microbiol Scand. 1953;32: 230–240. pmid:13079779
  22. 22. Candida Genome Database. Available: http://www.candidagenome.org/.
  23. 23. Wang G, Marquardt RR, Xiao H, Zhang Z. Development of a 96-well enzyme-linked solid-phase assay for beta-glucanase and xylanase. J Agric Food Chem. 1999;47: 1262–1267. pmid:10552447
  24. 24. Berzaghi R, Colombo AL, Machado AM, de Camargo ZP. New approach for diagnosis of candidemia based on detection of a 65-kilodalton antigen. Clin Vaccine Immunol. 2009;16: 1538–1545. pmid:19776195
  25. 25. Tarditi L, Camagna M, Parisi A, Vassarotto C, DeMonte LB, Letarte M, et al. Selective high-performance liquid chromatographic purification of bispecific monoclonal antibodies. J Chromatogr. 1992;599: 13–20. pmid:1618986
  26. 26. Bromuro C, Torosantucci A, Gomez MJ, Urbani F, Cassone A. Differential release of an immunodominant 65 kDa mannoprotein antigen from yeast and mycelial forms of Candida albicans. J Med Vet Mycol. 1994;32: 447–459. pmid:7738727
  27. 27. DiGiandomenico A, Keller AE, Gao C, Rainey GJ, Warrener P, Camara MM, et al. A multifunctional bispecific antibody protects against Pseudomonas aeruginosa. Sci Transl Med. 2014;6: 262ra155. pmid:25391481
  28. 28. Lindorfer MA, Nardin A, Foley PL, Solga MD, Bankovich AJ, Martin EN, et al. Targeting of Pseudomonas aeruginosa in the bloodstream with bispecific monoclonal antibodies. J Immunol. 2001;167: 2240–2249. pmid:11490011
  29. 29. De Bernardis F, Liu H, O'Mahony R, La Valle R, Bartollino S, Sandini S, et al. Human domain antibodies against virulence traits of Candida albicans inhibit fungus adherence to vaginal epithelium and protect against experimental vaginal candidiasis. J Infect Dis. 2007;195: 149–157. pmid:17152019
  30. 30. Yang Z, Schmidt D, Liu W, Li S, Shi L, Sheng J, et al. A novel multivalent, single-domain antibody targeting TcdA and TcdB prevents fulminant Clostridium difficile infection in mice. J Infect Dis. 2014;210: 964–972. pmid:24683195
  31. 31. Hellwig SM, van Spriel AB, Schellekens JF, Mooi FR, van de Winkel JG. Immunoglobulin A-mediated protection against Bordetella pertussis infection. Infect Immun. 2001;69: 4846–4850. pmid:11447159
  32. 32. Kulshreshtha P, Bhatnagar R. Inhibition of anthrax toxins with a bispecific monoclonal antibody that cross reacts with edema factor as well as lethal factor of Bacillus anthracis. Mol Immunol. 2011;48: 1958–1965. pmid:21704379
  33. 33. Wagstaffe SJ, Hill KE, Williams DW, Randle BJ, Thomas DW, Stephens P, et al. Bispecific antibody-mediated detection of the Staphylococcus aureus thermonuclease. Anal Chem. 2012;84: 5876–5884. pmid:22650421
  34. 34. Guttikonda S, Tang XL, Yang BM, Armstrong GD, Suresh MR. Monospecific and bispecific antibodies against E. coli O157 for diagnostics. J Immunol Methods. 2007;327: 1–9. pmid:17804009
  35. 35. Sarkar S, Tang XL, Das D, Spencer JS, Lowary TL, Suresh MR. A bispecific antibody based assay shows potential for detecting tuberculosis in resource constrained laboratory settings. PLoS One. 2012;7: e32340. pmid:22363820
  36. 36. Arvanitis M, Anagnostou T, Fuchs BB, Caliendo AM, Mylonakis E. Molecular and nonmolecular diagnostic methods for invasive fungal infections. Clin Microbiol Rev. 2014;27: 490–526. pmid:24982319
  37. 37. La Valle R, Sandini S, Gomez MJ, Mondello F, Romagnoli G, Nisini R, et al. Generation of a recombinant 65-kilodalton mannoprotein, a major antigen target of cell-mediated immune response to Candida albicans. Infect Immun. 2000;68: 6777–6784. pmid:11083795
  38. 38. Gramer MJ, van den Bremer ET, van Kampen MD, Kundu A, Kopfmann P, Etter E, et al. Production of stable bispecific IgG1 by controlled Fab-arm exchange: scalability from bench to large-scale manufacturing by application of standard approaches. MAbs. 2013;5: 962–973. pmid:23995617
  39. 39. Owais M, Kazmi S, Tufail S, Zubair S. An alternative chemical redox method for the production of bispecific antibodies: implication in rapid detection of food borne pathogens. PLoS One. 2014;9: e91255. pmid:24637674
  40. 40. Strop P, Ho WH, Boustany LM, Abdiche YN, Lindquist KC, Farias SE, et al. Generating bispecific human IgG1 and IgG2 antibodies from any antibody pair. J Mol Biol. 2012;420: 204–219. pmid:22543237
  41. 41. Spiess C, Merchant M, Huang A, Zheng Z, Yang NY, Peng J, et al. Bispecific antibodies with natural architecture produced by co-culture of bacteria expressing two distinct half-antibodies. Nat Biotechnol. 2013;31: 753–758. pmid:23831709
  42. 42. Labrijn AF, Meesters JI, de Goeij BE, van den Bremer ET, Neijssen J, van Kampen MD, et al. Efficient generation of stable bispecific IgG1 by controlled Fab-arm exchange. Proc Natl Acad Sci U S A. 2013;110: 5145–5150. pmid:23479652
  43. 43. Torres M, Casadevall A. The immunoglobulin constant region contributes to affinity and specificity. Trends Immunol. 2008;29: 91–97. pmid:18191616
  44. 44. Xia Y, Janda A, Eryilmaz E, Casadevall A, Putterman C. The constant region affects antigen binding of antibodies to DNA by altering secondary structure. Mol Immunol. 2013;56: 28–37. pmid:23665381
  45. 45. Yang CC, Chan HL. Preparation and characterization of beta 1-bungarotoxin bispecific monoclonal antibody. Biochem Mol Biol Int. 1999;47: 1039–1048. pmid:10410250
  46. 46. Dmitriev DA, Massino YS, Segal OL, Smirnova MB, Kolyaskina GI, Pavlova EV, et al. The comparison of the ability of monoclonal antibodies directed to different proteins (human IgG, human myoglobin and HRP) and bispecific antibodies derived thereof to bind antigens immobilized on a surface of a solid phase. Clin Chim Acta. 2001;309: 57–71. pmid:11408007
  47. 47. Harms BD, Kearns JD, Iadevaia S, Lugovskoy AA. Understanding the role of cross-arm binding efficiency in the activity of monoclonal and multispecific therapeutic antibodies. Methods. 2014;65: 95–104. pmid:23872324