the exceptionally broad-based potential of active and passive ......pnag has also been found to be...

14
Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=ierv20 Download by: [T&F Office Locations], [Florence Opara] Date: 14 April 2016, At: 06:21 Expert Review of Vaccines ISSN: 1476-0584 (Print) 1744-8395 (Online) Journal homepage: http://www.tandfonline.com/loi/ierv20 The exceptionally broad-based potential of active and passive vaccination targeting the conserved microbial surface polysaccharide PNAG David Skurnik, Colette Cywes-Bentley & Gerald B. Pier To cite this article: David Skurnik, Colette Cywes-Bentley & Gerald B. Pier (2016): The exceptionally broad-based potential of active and passive vaccination targeting the conserved microbial surface polysaccharide PNAG, Expert Review of Vaccines, DOI: 10.1586/14760584.2016.1159135 To link to this article: http://dx.doi.org/10.1586/14760584.2016.1159135 Accepted author version posted online: 26 Feb 2016. Published online: 16 Mar 2016. Submit your article to this journal Article views: 22 View related articles View Crossmark data Citing articles: 1 View citing articles

Upload: others

Post on 31-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

Full Terms & Conditions of access and use can be found athttp://www.tandfonline.com/action/journalInformation?journalCode=ierv20

Download by: [T&F Office Locations], [Florence Opara] Date: 14 April 2016, At: 06:21

Expert Review of Vaccines

ISSN: 1476-0584 (Print) 1744-8395 (Online) Journal homepage: http://www.tandfonline.com/loi/ierv20

The exceptionally broad-based potential of activeand passive vaccination targeting the conservedmicrobial surface polysaccharide PNAG

David Skurnik, Colette Cywes-Bentley & Gerald B. Pier

To cite this article: David Skurnik, Colette Cywes-Bentley & Gerald B. Pier (2016): Theexceptionally broad-based potential of active and passive vaccination targeting theconserved microbial surface polysaccharide PNAG, Expert Review of Vaccines, DOI:10.1586/14760584.2016.1159135

To link to this article: http://dx.doi.org/10.1586/14760584.2016.1159135

Accepted author version posted online: 26Feb 2016.Published online: 16 Mar 2016.

Submit your article to this journal

Article views: 22

View related articles

View Crossmark data

Citing articles: 1 View citing articles

Page 2: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

REVIEW

The exceptionally broad-based potential of active and passive vaccination targetingthe conserved microbial surface polysaccharide PNAGDavid Skurnik, Colette Cywes-Bentley and Gerald B. Pier

Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA

ABSTRACTA challenging component of vaccine development is the large serologic diversity of protective antigens.Remarkably, there is a conserved surface/capsular polysaccharide, one of the most effective vaccinetargets, expressed by a large number of bacterial, fungal and eukaryotic pathogens: poly-N-acetylglucosamine (PNAG). Natural antibodies to PNAG are poorly effective at mediating in vitro microbialkilling or in vivo protection. Removing most of the acetate substituents to produce a deacetylatedglycoform, or using synthetic oligosaccharides of poly-β-1–6-linked glucosamine conjugated to carrierproteins, results in vaccines that elicit high levels of broad-based immunity. A fully human monoclonalantibody is highly active in laboratory and preclinical studies and has been successfully tested in aphase-I setting. Both the synthetic oligosaccharide conjugate vaccine and MAb will be further tested inhumans starting in 2016; but, even if effective against only a fraction of the PNAG-producing patho-gens, a major advance in vaccine-preventable diseases will occur.

ARTICLE HISTORYReceived 4 January 2016Accepted 24 February 2016Published online16 March 2016

KEYWORDSPly-N-acetyl glucosamine;conjugate vaccine; capsule;antibiotic-resistance;monoclonal antibody

Vaccine potential of PNAG

Vaccines are the most exciting and effective therapeutics forhuman and animal diseases. They were called by the Centersfor Disease Control and Infection the most important medicaladvance of the twentieth century [1], as they are exceedinglysuccessful in preventing infections. Throughout the history ofvaccine development, the largest challenge has been theserologic diversity of many, if not most, of the protectiveantigens, requiring either a large compendium of vaccinecomponents for broad-based coverage (Streptococcus pneu-moniae) [2], finding a means to deal with a continually evol-ving target antigen (influenza virus and humanimmunodeficiency virus) [3,4], or trying to address high invivo antigenic shifts and drifts (Plasmodium spp.) [5]. Oftenvaccinologists must turn to finding conserved, but frequentlysuboptimal, antigens, or epitopes as vaccines. Thus, a majorparadigm underlying the success of many infectious agents isthe utilization of antigenic variation to avoid host immunityand thus retain their virulence. Overall, it would seem that aconserved antigen, readily accessible to major cellular andhumoral immune effectors, would be selected against by evo-lutionary forces that promote pathogen success. However,over the past 20 years, it has become apparent that such avaccine target may exist for many non-viral pathogens, andthis vaccine might be developed for use in a single formula-tion to induce humoral immunity to the most common micro-bial pathogens of humans and animals, encompassing mostGram-negative and Gram-positive bacteria, including tubercu-losis, as well as fungal pathogens such as Candida albicans andeven important protozoan pathogens such as those causingmalaria [6]. The target is poly-N-acetyl glucosamine (PNAG), a

polymer of β-(1–6)-linked N-acetylglucosamine residues with arelatively small amount of deacetylation occurring on 5–15%of the monosaccharide residues (Figure 1).

To thwart host acquired immunity from being effective atpreventing serious infection and disease against the collectionof PNAG-producing pathogens, natural antibodies to this poly-saccharide, found in most human and animal sera, are mostlyineffective at mediating microbial killing and protective immu-nity. Thus, these pathogens are unaffected by the native anti-body response to PNAG. Fortuitously, it was discovered that byremoving the acetates to produce a mostly polymeric β-1–6-linked N-glucosamine, or use of synthetic β-1–6-linked glucosa-mine (i.e. non-acetylated) oligosaccharides in conjugate vaccines[7–10]; antibodies could be induced that not only bound tonative, acetylated PNAG but also deposited complement ontomicrobial surfaces, mediated in vitro killing and showed protec-tive efficacy against a large variety of PNAG-producing patho-gens in preclinical studies [6,7,9]. We now stand on the brink ofdetermining in humans and economically important commercialand companion animals whether immunity to PNAGwill providea massive increase in vaccine-preventable diseases, be effica-cious against only some PNAG-producing microbes, or revealunexpected consequences that could make safety considera-tions and side effects outweigh the benefits of a PNAG vaccine.While animal studies to date, including some ongoing studies infarm animals, are highly encouraging that the benefits will behigh from a PNAG vaccine, these tests are still in the early stages.Importantly, the evaluation of the vaccine has been initiated infarm animals and will be tested in humans in 2016 along with afully human IgG1 monoclonal antibody (MAb) to PNAG that hasalready undergone phase I testing [11], with phase II testing also

CONTACT Gerald B. Pier [email protected]

EXPERT REVIEW OF VACCINES, 2016http://dx.doi.org/10.1586/14760584.2016.1159135

© 2016 Taylor & Francis

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 3: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

scheduled for commencement in 2016. Those of us working ondeveloping vaccines andMAbs to PNAG are cautiously optimisticabout our chances to have a major impact on human and animalhealth, but also fully cognizant of the challenges, roadblocks, andpotential of unanticipated consequences that confront the clin-ical development of all drugs.

Biology of PNAG

From PS/A to PIA to SAA to PNAG

Some of the properties of what eventually turned out to bePNAG were initially described as the capsular polysaccharide/adhesin (PS/A) of Staphylococcus epidermidis, although a defini-tive chemical composition and structure was not determined inthese studies [12–14]. Mack et al. [13] first described the chemicalproprieties of this surface polysaccharide and named it the poly-saccharide intercellular adhesin (PIA) and attributed to PIA theproperty of mediating intercellular adherence of coagulase-negative staphylococci. McKenney et al. [14] found that PS/Aand PIA were the same material and then reported in 1999 thatthe polymer was made by S. aureus [15], which was also reportedby Cramton et al. [16]. Another variant of the PNAG polymercontaining more than 70% glucosamine residues was describedas the slime-associated antigen (SAA) [17]. It is now clear that allfour of these entities (PNAG, PS/A, PIA, SAA) are chemicallyrelated and nearly identical [18–20].

Functional properties of PNAG

PNAG plays a major role in biofilm formation by different micro-bial species (see Ref. [21] for a review). In 2004, experimentsreported by Romeo and colleagues designed to identify compo-nents of the Escherichia coli biofilm led to the discovery that manystrains of this organism produced PNAG [22]. The locus in E. coliwas named pga for polyglucosamine and bioinformatic investiga-tions indicated a broad range of Gram-negative bacteria had thepga locus, implicating PNAG synthesis as a common factoramong these organisms. Further investigations confirmed PNAGwas a component of biofilms made by numerous Gram-negative

bacterial species. In Acinetobacter baumannii, deletion of the pgalocus led to loss of the biofilm phenotype, which was restored bycomplementation [23]. Similar findings were found withBurkholderia cepacia complex (BCC) [24], Klebsiella pneumoniae[25], and Bordetella pertussis [26]. Recently, PNAG’s role in biofilmformation was reported in Bacillus subtilis [27], the first Gram-positive bacillus found to produce PNAG.

PNAG has also been found to be an important virulencefactor for several pathogens. In three different strains of S.aureus, Mn8, Newman, and NCTC 10833 [28], loss of PNAGresulted in significantly reduced abilities to maintain bacteriallevels in blood following intravenous (IV) or intraperitoneal (IP)injection in mice, or to spread systemically to the kidneys orinduce a moribund/lethal state following IP infection. In E. coli,PNAG-deficient strains exhibited reduced fitness in the urinarybladder, kidneys, and spleen compared to wild-type strains inmurine models of ascending UTI and systemic infection,respectively [29]. In K. pneumoniae, PNAG-negative strains areless able to colonize the GI tract and to disseminate, and arelethal to significantly fewer mice after IP infection [25].

As a major component of the bacterial biofilm, PNAG has alsobeen reported to help bacteria evade the immune system, andparticularly opsonophagocytic killing (OPK) [30], presumably byeither preventing complement opsonins from binding to thebacterial cell surface or complement receptors on phagocytesfrom binding deposited opsonis, which are needed for efficientopsonic killing.

PNAG has also been reported to protect bacteria againstenvironmental stresses. For instance, S. aureus, E. coli, and K.pneumoniae producing PNAG were all better able to surviveexposure to ethanol than isogenic PNAG-deficient mutants, sug-gesting PNAG serves as a type of microbial armor regulatinginteractions of planktonic cells with the external environment [6].

Genetics and biosynthesis of PNAG

The ica and pga loci

The molecular basis for PNAG production was initially discov-ered in S. epidermidis [31]. In 1996, Heilmann et al. [31]

Figure 1. Structure of PNAG.

2 D. SKURNIK ET AL.

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 4: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

demonstrated that each one of three genes in a locus namedicaABC were necessary for the production of PNAG. An icaoperon mutant lost the ability to produce PNAG, which wasrestored by complementation with a plasmid carrying the icalocus. In addition, PNAG production was lost when any of thethree genes were individually inactivated. In 1998, a fourthgene (icaD), encoded between icaA and icaB was identified byGerke et al. [32] who also reported that the IcaA and IcaDproteins were located in the cytoplasmic membrane. Bothexhibited N-acetylglucosaminyltransferase activity, in whichIcaA represented a catalytic enzyme that required IcaD forfull activity.

In 2004, Wang et al. [22] used random transposon (Tn)mutagenesis to identify novel genes required for biofilmdevelopment in E. coli. They found 17 Tn-insertions clusteredin a small portion of the chromosome that resulted in E. colimutants that were producing significantly less biofilm (∼10-fold when compared to the parent strain). The locus, initiallyannotated as ycdSRQP, was renamed as pgaABCD. Each geneof the pga locus was necessary to produce a surface polysac-charide, whose chemical structure was determined by nuclearmagnetic resonance analyses to be PNAG. From this discovery,homologs of the pga locus were found in numerous Gram-negative bacterial species [6,23–26,33]. The presence of pgagenes is not enough, by itself, to firmly establish that thePNAG molecule is synthesized and expressed. Nonetheless, itis notable that, to date, essentially all the investigations find-ing pga genes in a microbe’s chromosome were able to

confirm the presence of PNAG at the bacterial surface [6,22–24,26,33,34]. Interestingly, the introduction of the pga locusinto a PNAG-negative bacteria can lead to the production ofPNAG [27]. A schematic depiction of the biosynthesis of PNAGbe proteins encoded by genes in the pga and ica loci is givenin Figure 2, taken from Ref. [35].

The hms locus of Yesinia pestis and eps locus of B.subtilis

PNAG has also been detected on the surface of bacterialspecies that have somewhat divergent genetic loci encodingPNAG biosynthetic enzymes and regulators. In Yesinia pestis,three operons, hmsHFRS, hmsT, and hmsP, are responsible forthe production of biofilms [33]. PNAG has been detected inbiofilms of Y. pestis, synthesized by proteins encoded withinthe hmsHFRS locus, and production is controlled by the digua-nylate cyclase HmsT and the EAL domain-containing phospho-diesterase, HmsP. These act as positive- and negativeregulators, respectively, of the transcription of the hmsHFRSlocus. Amino acid sequence analysis showed that HmsR andHmsF are homologues of the S. epidermidis β-1,6-GlcNAc gly-cosyltransferase IcaA and deacetylase IcaB, respectively, andall four HmsHFRS proteins have 50–83% similarity to productsencoded by pgaABCD.

PNAG was also detected in biofilms produced by B. subtilis[27]. In these bacteria, four genes encoding the proteins forPNAG synthesis, epsHIJK, were found in a cluster within the 15

Figure 2. Proposed pathways for enzymatic synthesis of PNAG. A. It has been proposed that IcaC may also be responsible for the O-succinlyation. B. Gram-negativebacteria use the PgaABCD proteins. C. Gram-positive Staphylococci use the IcaABCD proteins. From: Whitfield GB, Marmont LS, Howell PL. Enzymatic modifications ofexopolysaccharides enhance bacterial persistence. Front Microbiol. 15 May 2015;6:471. doi: 10.3389/fmicb.2015.00471. PMC4432689. Figure used from an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY) .

EXPERT REVIEW OF VACCINES 3

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 5: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

gene eps locus encompassing epsA-epsO. Cloning each of theB. subtilis epsH-K genes into E. coli with individual in-framedeletions in each of the four PNAG biosynthetic genes, pgaA-D, respectively, restored PNAG production in E. coli. Cloningthe entire B. subtilis epsHIJK locus into pga-deleted E. coli, K.pneumoniae, or alginate-negative Pseudomonas aeruginosaalso restored or conferred PNAG production [27]. In Y. pestisand B. subtilis, and about seven other bacterial species, whenPNAG was extracted and analyzed, it was found to be a β-1–6-linked poly-N-acetylglucosamine molecule, with only smallvariations in the levels of amino-group acetylation (95–100%)and variation in the amount of O-linked acetates and succi-nates [22,36,37].

Detection of PNAG expression by a broad range ofmicrobial pathogens

To detect the presence of PNAG onmicrobial cells, immunofluor-escence microscopy using an antigen-specific MAb to PNAG,designated F598, is routinely used [38]. The specificity of theMAb has been confirmed in studies wherein binding is stronglydetected using wild-type E. coli, S. aureus, and A. baumannii cells,but not to these bacteria deleted for the pga or ica loci [6,23,39].Binding is restored when an intact pga or ica locus is placed ontoa plasmid or back into the chromosome. Another test uses thePNAG-degrading enzyme dispersin B that specifically hydrolyzes

β-(1→6)-linked N-acetylglucosamine residues [40,41] to degradethe antigen and show concomitant loss of MAb F598 binding.Control treatment is with the β-1–4-linked N-acetylglucosaminehydrolyzing chitinase. Chitinase-resistant but dispersinB-sensitive binding of MAb F598 is highly specific to PNAG(Figure 3). This approach is particularly useful for identifyingPNAG expression when encoding genes have not been fullyidentified. At the moment, PNAG production has been identifiedin over 30 microbial organisms (Table 1) including fungal (C.albicans) and protozoan parasites (Plasmodium falciparum) [6],using chemical, genetic and/or immunologic analyses. For anumber of these, the protective efficacy of antibody to PNAG inmouse models of infection has confirmed the antigen isexpressed in vivo and a vaccine target [6]. Extensive work isongoing to determine the precise genetic and molecular basisfor PNAG production in all these species.

In vivo expression of PNAG

Previous studies have reported PNAG expression in the spu-tum and lung of CF patients infected with S. aureus [15], withPNAG detected by fluorescent microscopy at the surface of80–90% of the bacterial cells. PNAG was also found on thesurface of microbial cells in vivo in the lungs of patientsinfected by BCC [24] or Mycobacterium tuberculosis [6]. Inchildren, in vivo expression of PNAG was found in the middle

Figure 3. Detection of PNAG on bacterial surfaces. A. The PNAG-specific human IgG1 MAb F598, either directly conjugated to a fluorophore or detected with afluorescent anti human IgG is reacted with microbial cells (S. aureus in this example) treatted with chitinase, which has no effect on MAb F598 binding. (B) A controlhuman IgG1 MAb shows no immunofluorescence. C and D. Treatment with either dispersin B or 0.04 M sodium periodate destroys PNAG and renders bacterial cellsunreactive with Mab F598. Red-DNA stain. Bars = 10 µM.

4 D. SKURNIK ET AL.

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 6: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

ear effusions from S. pneumoniae or nontypable Hemophilusinfluenzae otitis media samples [6].

In animals, in vivo expression of PNAG was found in naso-pharyngeal fluid of chinchillas experimentally infected by S.pneumoniae serogroup 19A, in the gastrointestinal tract of amouse experimentally infected by C. rodentium, and in oculartissues from mice with C. albicans keratitis [6]. PNAG is alsoexpressed by major symbiont bacteria found in the humanmicrobiota: the GI tract (E. coli among others), the skin (S.epidermidis), and the oral flora (Aggregatibacter actinomyce-temcomitans). The extensive in vivo expression of PNAG raisedthe question of whether natural antibodies to PNAG werepresent in healthy individuals and whether these antibodieswere protective.

Naturally occurring antibodies to PNAG

Interestingly, in several studies, high levels of antibodies toPNAG were detected in healthy humans [8,38,42,43] byenzyme-linked immunosorbent assay (ELISA). Notably, theseantibodies do not appear to be functional or protective.Natural antibodies to PNAG are not able to mediate in vitrobactericidal or OPK [8] and they do not protect animals againstPNAG producing pathogenic bacteria such as S. aureus [42].The natural antibodies predominantly require that the acetatesubstituents on PNAG be present in order to bind to theantigen, but the result is an inability to mediate complement

activation and protective immunity [8,9,42]. Structural studiesof PNAG indicate that in its preferred glycoform conformationthe acetates are on the polymer’s surface, sticking out into thesurrounding medium, and this forms the basis for the immu-nodominance of the acetate-dependent epitopes [44].Apparently, then, the Fc regions of the antibodies bound tothe acetate-containing epitopes are not sufficiently closetogether to effectively bind C1q and initiate complementactivation [38,42].

Immunity to PNAG and capsular polysaccharides inS. aureus infections

In contrast to most normal human sera, in some infectedpatients functional antibodies to PNAG could be detected[8], especially opsonizing antibodies in patients with S. aureusbacteremia [43]. However, most patients with non-blood-stream, acute S. aureus infections, such as in the skin andlung, failed to make an opsonizing antibody response toPNAG. Thus, either a more intense, or longer term, infectionwas required to elicit opsonic antibodies to PNAG in thecontext of S. aureus infection. Notably, the fact that humanscould make such antibodies supported other data that alteredglycoforms of PNAG could induce opsonic, protective anti-body [9,39] indicating it would be feasible to develop aneffective passive or active immunotherapy targeting PNAG.

With the discovery of extensive production of PNAG byclinical isolates of S. aureus it became very attractive to con-sider a multivalent vaccine incorporating this antigen with theS. aureus capsular polysaccharides (CP), based on analogieswith successful capsular vaccines to other bacterial pathogens,including S. pneumoniae, Neisseria meningitidis, H. influenzaetype b, and Salmonella enterica serovar Typhi [2]. S. aureusmakes two CP antigens, types 5 and 8, expressed by 70–85%of clinical isolates and are considered to be excellent candi-date vaccine antigens [45]. Unfortunately, two clinical trials forpreventing bacteremia in hemodialysis patients using CP anti-gens conjugated to a carrier protein as a vaccine both failed tomeet their primary end points of reduced bacteremia [46,47].Notably, in the first trial [46], there was 57% protective efficacy(P = 0.02) after 40 weeks, as determined by a post hoc analysis;but at the predetermined 54-week endpoint efficacy was only26% (P = 0.23). Thus, a multivalent PNAG/CP5/CP8 vaccineseemed like a reasonable candidate to augment the limitedefficacy of the bivalent CP vaccine.

However, when testing of antisera containing antibodies tothese three components was started, instead of observing theexpected additive or synergist effects, what was actually foundwere potent and mutually inhibitory effects on opsonic killingusing in vitro assays and mouse models of bacteremia and skininfections [43]. Electron microscopy, isothermal calorimetry,and surface plasmon resonance experiments indicated thatantibodies to CP and PNAG bound together via an apparentcharge-dependent, idiotype–anti-idiotype interaction. In thesera of patients recovering from S. aureus, bacteremia anti-body titer increases to both PNAG and CP antigens werefound, but even in these sera there was a notable lack of invitro and in vivo protective efficacy [42,43]. Consistent with thelaboratory-based findings of potent inhibitory effects between

Table 1. Listing of organisms identified as making PNAG by chemical, genetic,or immunologic criteria.

A. Organisms verified to produce PNAG by chemical, genetic, andimmunologic criteria

Actinobacillus pleuropneumoniaeAcinetobacter baumanniiAggregatibacter actinomycetemcomitansBacillus subtilisEscherichia coliStaphylococcus aureusStaphylococcus epidermidisVibrio parahemolyticusYersinia pestis

B. Organisms verified to produce PNAG by genetic and immunologic criteriaBordetella pertussisBordetella parapertussisBordetella bronchisepticaBurkholderia cepacia complexEnterobacter cloacaeKlebsiella pneumoniaeShigella spp.Stenotrophomonas maltophiliaYersinia entercoliticaYersinia pseudotuberculosis

C. Organisms verified to produce PNAG by immunologic criteriaBacteroides fragilisCandida albicansHemophilus ducreyiHemophilus influenzaeListeria monocytogenesMycobacterium tuberculosisNeisseria gonorrhoeaeNeisseria meningitidesStreptococcus pyogenesStreptococcus agalactiaeStreptococcus pneumoniaePlasmodium spp.Trichmonas vaginalis

EXPERT REVIEW OF VACCINES 5

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 7: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

immunization-induced antibodies to PNAG and to S. aureus CPantigens, infection-induced antibodies to both PNAG and theS. aureus CP, which by themselves were able to mediateopsonic killing, did not do so when both antibodies were inthe same serum sample. When either antibodies to PNAG orcapsule were removed by absorption, the sera gained func-tional activity and this was again lost when the two antibodieswere recombined [43]. In a follow-up study, it was reportedthat natural, non-opsonic antibodies to PNAG that are presentin most NHS samples can also neutralize the functional activityof antibodies to S. aureus CP5 and CP8 antigens, but thesenatural antibodies to PNAG do not interfere with functionalantibodies to PNAG [42]. These findings raised the possibilitythat a potential explanation for the failure of the CP-basedvaccines in hemodialysis patients might have been loss ofefficacy when the titers induced by CP immunization declinedto the point where natural antibodies to PNAG neutralizedtheir protective efficacy. Altogether, these findings show thatthe presence of natural antibodies to PNAG in both healthyand infected subjects could impact newer attempts to use theCP antigens as vaccines but also highlight the complexity ofvaccine development using PNAG or CP antigens for S. aureus.

Discovery of the means to successfully inducefunctional immunity to PNAG

Before the full characterization of the chemical structure ofPNAG, immunization-induced antibodies raised to what wasmost likely semi-purified PNAG that had undergone extractionprocesses for purification that likely altered its compositionwere reported to be opsonic in vitro and active in vivo againstS. epidermidis [48,49]. Once the composition of native PNAGwas known, the basis for protective efficacy in comparison tothe lack of functionality of natural antibodies to PNAG couldbe explained.

Analysis of purified PNAG that induced opsonic killing andanimal protection showed a markedly reduced level ofN-acetylation in this material [9,19]. Based on this finding,two conjugate vaccines using native, highly acetylated(~95%) PNAG and the deacetylated glycoform, termeddPNAG (~15% acetylation), conjugated to a carrier protein ofmutant diphtheria toxoid were synthesized. Antibodies todPNAG-DT were found to be much more effective than anti-bodies to native PNAG-DT at inducing opsonic killing andreductions in bacterial levels in the blood of bacteremic miceand at protecting mice in a model of lethal S. aureus infec-tion [9].

An additional study examining the opsonic killing activityagainst S. aureus in serum samples from infected CF patientsfound that antibodies directed at the deacetylated (backbone)epitopes on PNAG (dPNAG) were superior at eliciting in vitroopsonic killing. More precisely, it was found that when opsonickilling activity was detected in the CF patient’s sera, antibodiesdirected to dPNAG were more functional in phagocyte-depen-dent killing than antibodies directed at the acetate-dependentepitope (PNAG). This result was obtained using both wholeserum and affinity-purified antibody populations directed atthe epitopes under examination [8]. Of note, the presence ofsuch antibodies correlated with a healthier overall clinical

condition in spite of the presence of S. aureus in sputumcultures. However, it is a hallmark of CF that patients with S.aureus are often healthier than those lacking this organism,although the most likely explanation is that lack of S. aureus isalso associated with the presence of P. aeruginosa, the patho-gen clearly most responsible for clinical deterioration in CF[50,51]. Nonetheless, the finding of functional antibodies toPNAG in S. aureus-infected CF patients could account for therelative lack of pathology associated with the presence of thispathogen in the lungs of these patients.

As both the animal-derived [9] and human-derived antibo-dies to dPNAG [8] were better able to mediate OPK activitythan antibodies to native PNAG, human monoclonal antibo-dies (MAbs) to PNAG and dPNAG were produced to furtherstudy this phenomena [38]. A MAb designated F598 thatbound well to both native PNAG (expressed on microbialsurfaces) and dPNAG (which binds the functional antibodies)had superior opsonophagocytic activity and protective activityin a mouse model of lethality induced by S. aureus than twoMAbs that bound optimally to PNAG and minimally to dPNAG(MAbs F628 and F630). The better activity of antibodies todPNAG was associated with deposition of osponically activefragments of the first and third components of complementonto PNAG [38] (Figure 4).

As the natural antibodies to PNAG are directed against itsnative, highly acetylated (>90% of N-acetyl groups) form,these results probably explain the lack of protection mediatedby natural antibodies: they do not bind to dPNAG or depositC3 opsonins onto bacterial surfaces [8,38]. In contrast, dPNAGelicits antibodies with excellent engagement of the comple-ment system leading to robust in vitro killing activity andprotection against infection in experimental animal models[8,38,42]. Thus, the dPNAG glycoform is the basis for thecurrent vaccine approaches being evaluated against PNAG-producing microbes.

Production of synthetic oligosaccharide-proteinconjugate vaccines

A further refinement to the development of a product foractive immunization was achieved by conjugating syntheticoligosaccharides composed of either non-acetylated β-(1→6)-D-glucosamine (GlcNH2) or β-(1→6)-N-acetylglucosamine(GlcNAc) to carrier proteins [7,10]. Oligosaccharides of eitherfive or nine monosaccharides in length, with linkers on thereducing termini that could be activated to produce sulfhydrylgroups for conjugation to bromoacetyl groups introducedonto carrier proteins, were synthesized [7]. Interestingly, syn-thetic 5-mer GlcNH2 (5GlcNH2) or 9GlcNH2 conjugated to teta-nus toxoid (TT) elicited mouse antibodies that mediatedopsonic killing, while the antibodies that were produced inresponse to the acetylated oligosaccharide conjugate vaccine,5GlcNAc- or 9GlcNAc-TT, did not mediate opsonic killing [7].Rabbit antibodies to 9GlcNH2-TT bound to PNAG and dPNAGantigens, mediated killing of S. aureus and E. coli, and pro-tected against S. aureus skin abscesses and lethal E. coli peri-tonitis [7]. Subsequent studies showed other carrier proteinscould induce protective antibodies against S. aureus and E. coliwhen conjugated to 5GlcNH2 or 9GlcNH2 [10,52]. Therefore,

6 D. SKURNIK ET AL.

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 8: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

the synthetic 5GlcNH2 oligosaccharide now forms the basis forthe clinical development of PNAG-targeting vaccines forhumans and animals.

In vitro correlates of PNAG-mediated immunity

Eliciting antibodies to PNAG with the proper properties tomediate microbial killing and clearance is essential for achiev-ing protective immunity, and predicting the level and func-tionality of antibodies that are indicative of protectiveimmunity is key for evaluating vaccine doses and constructsfor their potential to mediate protection. A number of in vitroassays have been developed as functional correlates ofimmunoprotection.

Antibody titer measured by ELISA

In order to distinguish the levels of naturally occurring,usually nonprotective, antibodies to PNAG from functionallyactive, vaccine-elicited antibodies to dPNAG, ELISA-basedassays using either purified PNAG- or dPNAG-coated platesare employed. The serum antibody titers are interpreted asthe amount of PNAG or dPNAG bound antibody determinedby optical density. Linear regression formulas are generated

to calculate titers. While antibodies to both PNAG anddPNAG will bind native PNAG, only dPNAG-elicited antibo-dies will bind dPNAG, in contrast to most natural antibodyto PNAG [7–9]. While a validated ELISA test or titer for eitherPNAG or dPNAG is not yet developed beyond those used inlaboratory investigations, it is clear that protective antibo-dies must bind to both of these antigens, indicating that ahigh titer to dPNAG is a good measure of likely protectiveefficacy.

Complement mediated opsonic and bactericidal killing

For Gram-positive bacteria, the OPK assay can be used todemonstrate both antibody specificity for PNAG recognition,and functional activity, by use of neutrophil- and complement-mediated killing of opsonized pathogens. Similarly, for Gram-negative bacteria, the bactericidal assay using antibody toPNAG/dPNAG and complement demonstrates antibody speci-ficity and functionality, requiring the complement membraneattack complex, consisting of complement components C5–C9, to be formed. These assays constitute core components ofthe in vitro evaluation of vaccine-induced antibodies to PNAG.Whether all strains of Gram-negative bacteria are killed in theabsence of phagocytes is not known.

Figure 4. Predicted binding of antibody (Ab) raised to highly acetylated PNAG or dPNAG to the native PNAG molecule and interaction with the first component ofcomplement, C1. A. Acetate molecules form an immunodominant epitope due to their projection out from the helix formed by PNAG. Binding of both natural andimmunization-induced antibody to native PNAG decreases markedly when the acetates are removed. B. In contrast, dPNAG presents more varied surface features,comprised of hydroxyl, hydroxymethyl and amino groups projecting out from the core helix. C. Acetate-dependent antibodies to native PNAG are predicted to bindto the PNAG molecule in a manner such that the Fc regions are not sufficiently close together to bind two arms of C1q. D. Antibodies raised to dPNAG bind to nativePNAG at acetate-independent epitopes bringing the Fc portion of IgG sufficiently close together to bind C1.

EXPERT REVIEW OF VACCINES 7

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 9: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

Complement deposition assays

Another means used to correlate the PNAG antibody titer withfunctional activity is use of a C3 deposition assay [38,42]. Inthe most recent iteration of this assay, ELISA plates are coatedwith purified PNAG and then varying concentrations of heat-inactivated (to remove endogenous complement activity) dilu-tions of pre- and postimmunization sera, and an active com-plement source, kept at a constant concentration, are added.Functionally active PNAG-specific antibodies bind to the pur-ified PNAG on the ELISA plate and activate complement,resulting in C3 deposition. A C3-specific antibody is used todetect the amount of C3 deposited in each well by the PNAG-specific antibody. Thus far, the amount of C3 deposited hascorrelated well with PNAG-specific antibody titers and protec-tion against experimental infection challenges, a findingapplicable to both polyclonal and MAbs [38].

More recently, it was found that protective antibodies toPNAG mediate their activity via activation of the classicalcomplement pathway (Figure 4). PNAG itself can activatecomplement via the lectin pathway, as evidenced by anti-body-independent C3 deposition occurring in C1-deficientserum [53]. However, this pathway is not optimal for mediat-ing killing of many Gram-positive bacterial species [54],whereas antibody-mediated activation of the classical path-way is much more effective. Therefore, we have developed aC1q-deposition assay similar to that for C3 deposition, exceptan antibody specific to C1q is used to detect complementactivation and binding of C1. Of note, C1q is highly conservedacross species and a single antiserum to purified human C1qcan be used to detect C1 deposition using sera obtained froma range of mammalian species as the complement source.

In vivo studies

Since the early 1990s, antibodies to PNAG have shown abroad-spectrum activity against numerous pathogens inlaboratory animal models. Published results are summarizedin Table 2 and Cywes-Bentley et al. [6] provide experimentalresults with Streptococcus pyogenes, Listeria monocytogenes, S.pneumoniae, and N. meningitidis serogroup B. In addition, thehuman MAb to PNAG protected T-bet/RAG2-deficient micefrom infectious colitis [6,55].

In vivo protection against staphylococci infections

The first report of protective activity of antibodies to PNAG(called PS/A at the time) was in a rabbit model of bacteremiausing catheters colonized by coagulase-negative staphylococciinserted into the right jugular vein and attached to a subcu-taneous osmotic pump [48]. In this model, antibodies to PNAGwere able to reduce the number of bacteremic days byapproximately 60%. This approach was then extended to arabbit model of endocarditis [49], and both passive and activeimmunization targeting PNAG protected against bacteremiaand endocarditis cause by S. epidermidis. The antibodies toPNAG were next tested against S. aureus in numerous studies.In the first report [15], active and passive immunization sig-nificantly reduced the colony forming units (CFU) recoveredfrom the kidneys of mice after IV infection with differentstrains of S. aureus. Similarly, passive immunization signifi-cantly protected mice in a lethality model. In the ensu-ing16 years, antibodies to PNAG were repetitively shown tobe protective against S. aureus infections, including mousemodels of bacteremia and skin abscesses as well as sheepmastitis [7–9,19,28,38,42,43,56,57].

In vivo protection against infections caused by E. coli

E. coli represents a major challenge for vaccine development,mostly due to the large serologic diversity of protective anti-gens such as LPS O side chains and capsules [58–60]. After thereport that the conserved PNAG was also present on the sur-face of E. coli [22], antibodies to this antigen were tested forprotection against this organism. In several studies, using amodel of systemic dissemination after IP infection [7,34,39],both polyclonal and MAbs to PNAG significantly protectedmice against various strains of E. coli. For infections causedby E. coli colonizing the GI tract, the 9GlcNH2 synthetic oligo-saccharide conjugated to Shiga toxin 1b subunit (9GlcNH2-Stx1b) was shown to induce rabbit antibodies that were pro-tective against infection caused by different strains of Shiga-toxin producing E. coli [52]. Thus, in multiple in vivo mousesettings, antibodies to PNAG were effective in reducing theconsequences of E. coli infections.

In vivo protection against infections caused by multi-drug resistant and pan-drug resistantGram-negativepathogens

Antibiotic resistance is a major concern impacting treatment ofillnesses caused by Gram-negative bacteria, and a vaccine orimmunotherapeutic alternative is highly desirous to participatein curing these infections. Among the main pathogenic Gram-negative bacteria that are naturally multi-drug resistant (MDR)as well as capable of acquiring additional resistances to becomepan-drug resistant (PDR) are A. baumannii and strains within theBCC. Studies with A. baumannii, a major cause of nosocomialinfections in immunocompromised patients, showed that rabbitantisera to 9GlcNH2-TT significantly reduced levels of A. bau-mannii in the lungs or blood of mice 2 or 24 h post-infection,respectively, compared to levels in control groups receivingnormal rabbit sera [36]. There are three major BCC species:

Table 2. Protective efficacy of antibody to the dPNAG glycoform against variousmicrobial pathogens in mouse settings of infection.

Staphylococcus aureus Lethal peritonitis, bacteremia, skin infection,pneumonia, keratitis

Escherichia coli Lethal peritonitis, GI infection by Stx+ strains, systemicinfection by multidrug resistant strains

Burkholderiacenocepacia

Lethal peritonitis

Acinetobacterbaumannii

Bacteremia and pneumonia

Neisseria meningitidisserogroup B

Neonatal mouse meningitis infection

Streptococcuspyogenes

Skin infection leading to lethal systemic infection

Listeria monocytogenes Systemic infectionCandida albicans KeratitisPlasmodium bergheiANKA

Mouse malaria

8 D. SKURNIK ET AL.

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 10: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

Burkholderia cenocepacia, B. multivorans, and B. dolosa and theycan all cause PDR infections, particularly in patients with cysticfibrosis. All three species were shown to produce PNAG both invitro and in vivo. Antibodies to PNAG were able kill thesebacteria in opsonophagocytic assays as well as protect micefrom lethal peritonitis caused by BCC strains injected eitheralone or in combination with MRSA, chosen as another exampleof a PNAG-producing pathogenic and antibiotic-resistant bac-terium [24].

Carbapenems are the main drug used to treat infectionscaused by MDR Gram-negative bacteria in general, and MDRenterobacteriacae in particular. Unfortunately, there are nowmore and more Carbapenem Resistant Enterobacteria (CRE),that have emerged mainly through the acquisition of genesencoding for production of carbapenemases such as the K.pneumoniae carbapenemase or the New Delhi metallo β–lac-tamase [61–63]. As with the other antibiotic resistant strains,antibodies to PNAG were able to kill CRE strains in vitro andprotect mice in vivo against three clinically relevant species ofenterobacteriacea (E. coli, Enterobacter cloacae and K. pneumo-niae), whether or not the strains produced a carbapenemase[34]. Finally, while P. aeruginosa is the only major drug-resis-tant pathogenic bacterium not producing PNAG naturally,antibodies to PNAG were shown to be able to protect miceinfected by a PNAG producing, genetically engineered P. aer-uginosa construct, suggesting that regardless of the Gram-negative bacterial species; PNAG expression is the sole deter-minant of the protective efficacy of antibodies to this antigen.The animal models used in these studies involved both IP andIV injections of the bacteria [34].

Killing and protection against pathogenic eukaryotes byantibodies to PNAG

PNAG has been detected at the surface of several eukaryotes,including human pathogens such as C. albicans and P. falci-parum [6]. Using a model of mouse keratitis, antibodies toPNAG significantly reduced CFU counts recovered from thecornea of C57BL/6 mice infected with C. albicans and treatedwith the human MAb F598 to PNAG either before or after thechallenge by C. albicans [6]. These antibodies also killed C.albicans in vitro using an OPK. When Plasmodium berghei wasused to infect mice, which is a relevant murine model for P.falciparum human infection, antibodies to PNAG significantlyreduced the mortality of C57BL/6 mice from systemic PNAG-positive P. berghei ANKA infection initiated by IP injection [6].Thus, the potential efficacy of PNAG as a vaccine extends to avery broad range of targets.

Route to a clinical testing

Although the first papers describing protective efficacy againstS. epidermidis using injections of what is now known to bePNAG-containing semi-purified antigens were published 25 toalmost 30 years ago [12,49], it has taken this long to bring thistechnology to the point of human testing. Two products arebeing evaluated. One is the fully human IgG1 moleculederived by cloning variable regions from a human B cellobtained from an individual recovering from a S. aureus

infection into the TCAE 6.2 vector encoding the humangamma1 heavy chain constant regions and lambda lightchain constant region [38]. The second is a synthetic pentamerof β-1–6-linked glucosamine conjugated to TT (5GlcNH2-TT)[7]. Notably, while designing phase II and III human trials ofvaccines is challenging even when the target antigen is on asingle species of pathogen, the plethora of pathogens makingPNAG provides multiple opportunities for testing, which can,at times, make decisions difficult. Nonetheless, both thehuman MAb and vaccine are currently being manufacturedunder GMP conditions for further testing commencing in2016.

Phase I clinical trial of MAb

The human IgG1 MAb was tested in 20 healthy humans in2010 in a dose-escalating trial involving four volunteers perdose, five doses given IV in a 2-h infusion starting at 0.85 mg/kg and ending at 17.2 mg/kg [11]. There were no significantadverse reactions, but two individuals given the highest dosehad recordable events, one involving a rash that developedafter infusion that was resolved by use of diphenhydramineand the other an infusion-reaction resolved by slowing therate of infusion. The half-life of the MAb was ~28 days andhigh titers of functional, opsonic antibody to S. aureus weremaintained throughout the 50-day observation and bloodsampling periods. No reports of distress that might be relatedto mucosal surface reactions or disruption to the normalmicrobiota were recorded. Overall, it appeared the MAb toPNAG is safe with an outstanding half-life and other proper-ties, paving the way for phase II trials.

Phase I clinical trial of vaccine

The synthetic pentameric 5GlcNH2-TT has undergone about2 years of development to scale up the synthesis of theoligosaccharide, which is produced as a dimer of two gluco-samine pentamers bound together by a disulfide bond at theend of a linker, as described [7]. Commercially available, GMP-grade TT is used as the carrier protein. Trials of this vaccine,containing from 20–39 oligosaccharides per TT monomer,have commenced in economically important farm animalswith some encouraging, albeit early, results indicating efficacy.As these trials are ongoing the full results are not yet available,but at least for some that are successful, it is anticipated thatfindings will be reported at appropriate meetings and poten-tially published in 2016.

Phase II clinical trials

Of note, with the broad range of pathogens producing PNAG,the challenge for phase II (and beyond) trials is finding asetting for testing both the MAb and vaccine where infectionrates will be high. One obvious setting is human challengemodels [64], and some that are currently available for PNAGproducing pathogens include Neisseria gonorrhoeae [65], P.falciparum [66], Hemophilus ducreyi [67], and several GI patho-gens [68,69]. Additional trials conducted in more traditionalsettings where infection rates with PNAG producing

EXPERT REVIEW OF VACCINES 9

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 11: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

pathogens is high might include efficacy in intensive careunits, GI tract surgery, mechanically-ventilated patients orsimilar high-risk locations within hospitals and health-caresettings. Interventional studies with both the MAb and vaccinein either newly diagnosed patients with long or difficultcourses of antibiotic treatments, such as drug-resistant tuber-culosis or MDR Gram-negative pathogens [24,34], or recurrentinfections, such as methicillin-resistant S. aureus skin and soft-tissue infections or E. coli urinary tract infections, are alsobeing considered. Overall the final decisions will depend onnumerous factors, taking into consideration concepts such asmedical need and potential impact on health, settings provid-ing a proof of principle, economic feasibility of the interven-tion and any other related risk/benefit considerations.

Expert commentary

The potential for a single vaccine component to provide pro-tective immunity to so many major pathogens is a clear para-digm shift in the field. In favor of realizing much of thispotential is the broad-based expression of PNAG [6], indicativeof an essential role in microbial biology inasmuch as so manydivergent genetic and enzymatic methods to produce PNAGhave evolved and survived in diverse microbial populations[22,23,27,31,33], detection of in vivo expression during humanand animal infection [6,15], and preclinical data mostly in micedemonstrating protection based on use of the deacetylatedglycoform of PNAG to induce immunity [7,9]. Furthermore, asa surface, essentially capsular, polysaccharide [6], PNAG is asavailable to immune effectors as are the more traditionalcapsules of S. pneumoniae, H. influenzae type B, and N. menin-gitides. Co-occurrence in the same physical space on themicrobial surface with classic capsules, among our most suc-cessful vaccines, places PNAG in an excellent position to be aneffective vaccine target. Many interesting questions remainrelated to the diversity of genes and proteins producingPNAG in so many different species, including their functions[32,70,71], how highly conserved the overall PNAG structure is,and how essential PNAG is to a given microbe’s fitness forsurvival. Nonetheless, the field is currently ready to see theresults of the animal and human testing to ascertain if insettings of infections that are relevant to specific but differentanimals hosts immunity to PNAG can be effective.

On the down-side, use of the MAb in humans and thevaccine in animals is still quite limited, so overall safety con-siderations are still predicated on a small sample size. Long-term effects are not known, nor is the duration of immunity,and whether protective antibody isotypes will be universallyinduced cannot be predicted from the small sample sizestested to date. Fortunately, there have been no indicationsthat the natural antibodies to PNAG that most everyone pos-sess, interfere with the vaccine-induced antibodies, but itwould not be surprising if some situations arise wherein theefficacy of vaccine-induced antibodies is compromised bynatural antibodies, including those to S. aureus CP antigens.Notably, several companies are pursuing S. aureus vaccinescontaining the CP5 and CP8 antigens [72,73] and if vaccinestargeting both these antigens and PNAG are found to beefficacious, this could present a difficult problem if these

vaccine-induced antibodies are cross-neutralizing, as hasbeen reported using rabbit and mouse antibodies [42,43].

Five-year view

At this point, the vast majority of vaccine research is centeredon finding individual vaccines for every important microbe.This is the basis for not only overall vaccine success to datebut is also consistent with our modern understanding ofimmunity and host defense in that serologically variant andorganism-specific antigens maximize their pathogenic poten-tial. In the next 5 years, extensive testing of the PNAG MAband vaccine will ensue. During this period, clear answers as tosafety will be obtained, as will a reasonable amount of efficacydata, particularly in economically important animals. Successof either reagent in human challenge models, or a clearlymedically important clinical setting, will likely propel the licen-sing of PNAG-directed immunotherapies. Given the exception-ally broad range of PNAG-targetable microbial species, itmight be anticipated there will be some failures for specificorganisms or in specific tissue settings. This could be due tolack of in vivo PNAG expression, difficulty in getting immuneeffectors (antibody, complement, phagocytes, Th17 cells, act)to sites of infection (i.e. skin, urogenital tract, GI tract) oremergence of PNAG-negative variants that retain pathogeni-city. Significant safety issues, mostly not anticipated at thistime, could be a major barrier to development. Concernsabout effects of antibody to PNAG on normal microbial florahas not been realized in any of the animal or short-termhuman studies conducted to date, but larger studies mightfind problems here. A difficult to predict consequence impact-ing the field might entail the success of immunity to PNAG inpreventing infections with some pathogens, but possiblyexacerbating infections by others, as antibody-mediatedenhancement of infection is not unknown in infectious dis-eases, particularly with viral pathogens [74]. Overall, significantclinically relevant results in humans with the MAb and the5GlcNH2-TT vaccine and in animals with the vaccine will beobtained in the next 5 years. These likely will determine thepotential success or failure of targeting PNAG with a broad-spectrum antibody and vaccine, not unlike broad-spectrumantibiotics. Optimistically, the immunotherapies will have littledownside and thus provide humanity with an important newtool to control infectious diseases.

Key issues

● PNAG is an exceptionally broadly expressed capsular/sur-face antigen on most major human and animal pathogens.

● Immunity to PNAG can be engendered by use of deacety-lated/non-acetylated glycoforms, particularly as compo-nents of conjugate vaccines.

● PNAG likely has an essential role in microbial biology basedon its broad expression and the diversity of genetic andenzymatic proteins involved in its synthesis among differ-ent microbial species.

● Natural immunity to PNAG is common but mostly ineffec-tive due to an inability of natural antibodies to activatecomplement.

10 D. SKURNIK ET AL.

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 12: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

● Preclinical data in mice show broad-protection against bac-terial, fungal, and protozoan pathogens.

● Phase I trials of a fully human IgG1 MAb to PNAG showed along half-life and overall excellent safety profile.

● Human clinical trials of the MAb and a conjugate vaccineconsisting of 5GlcNH2-TT will commence, barring unfore-seen difficulties, in 2016.

● Evaluation of the vaccine in economically important andcompanion animals has commenced with results likelyavailable in 2016.

● Challenges remain in regard to long-term safety, efficacyagainst a range of pathogens and unanticipatedconsequence.

Financial and competing interests disclosure

GB Pier is an inventor of intellectual properties [human monoclonal anti-body to PNAG and PNAG vaccines] that are licensed by Brigham andWomen’s Hospital to Alopexx Vaccine, LLC, and AlopexxPharmaceuticals, LLC, entities in which GB Pier also holds equity. As aninventor of intellectual properties, GB Pier also has the right to receive ashare of licensing-related income (royalties, fees) through Brigham andWomen’s Hospital from Alopexx Pharmaceuticals, LLC, and AlopexxVaccine, LLC. GB Pier’s interests were reviewed and are managed by theBrigham and Women’s Hospital and Partners Healthcare in accordancewith their conflict of interest policies. D Skurnik and C Cywes-Bentley areinventors of intellectual properties [use of human monoclonal antibody toPNAG and use of PNAG vaccines] that are licensed by Brigham andWomen’s Hospital to Alopexx Vaccine, LLC, and AlopexxPharmaceuticals, LLC. As inventors of intellectual properties, C Cywes-Bentley and D Skurnik also have the right to receive a share of licen-sing-related income (royalties, fees) through Brigham and Women’sHospital from Alopexx Pharmaceuticals, LLC, and Alopexx Vaccine, LLC.The authors have no other relevant affiliations or financial involvementwith any organization or entity with a financial interest in or financialconflict with the subject matter or materials discussed in the manuscriptapart from those disclosed.

ORCID

Gerald B. Pier http://orcid.org/0000-0002-9112-2331

References

Papers of special note have been highlighted as:• Of interest•• Of considerable interest

1. Achievements in Public Health. 1900-1999: control of infectiousdiseases. MMWR Morb Mort Wkly Rep. 1999;48(29):621–629.

2. Makela PH. Conjugate vaccines–a breakthrough in vaccine devel-opment. SE Asian J Trop Med Public Health. 2003;34(2):249–253.

3. Houser K, Subbarao K. Influenza vaccines: challenges and solutions.Cell Host Microbe. 2015;17(3):295–300.

4. Rubens M, Ramamoorthy V, Saxena A, et al.. HIV vaccine: Recentadvances, current roadblocks, and future directions. J Immunol Res.2015;2015:560347.

5. Sinha S, Medhi B, Sehgal R. Challenges of drug-resistant malaria.Parasite. 2014;21:61.

6. Cywes-Bentley C, Skurnik D, Zaidi T, et al. Antibody to a conservedantigenic target is protective against diverse prokaryotic andeukaryotic pathogens. Proc Natl Acad Sci USA. 2013;110(24):E2209–2218.

•• This paper describes the broad-based expression of PNAGamong a range of microbes and documents the protectiveefficacy of polyclonal and MAb to PNAG against these

pathogens. It also highlights the common in vivo expressionof PNAG in infected tissues.

7. Gening ML, Maira-Litran T, Kropec A, et al. Synthetic β-(1->6)-linkedN-acetylated and nonacetylated oligoglucosamines used to pro-duce conjugate vaccines for bacterial pathogens. Infect Immun.2012;78(2):764–772.

• This paper documents the utility of synthetic oligosaccharidesas a basis for a conjugate vaccine to PNAG. It also validates theneed for non-acetylated glucosamines as the PNAG glycoformneeded top-induced protective immunity.

8. Kelly-Quintos C, Kropec A, Briggs S, et al. The role of epitopespecificity in the human opsonic antibody response to the staphy-lococcal surface polysaccharide poly N-acetyl glucosamine. J InfectDis. 2012-2019;192(11):2005.

9. Maira-Litran T, Kropec A, Goldmann DA, et al. Comparative opsonicand protective activities of Staphylococcus aureus conjugate vac-cines containing native or deacetylated Staphylococcal Poly-N-acetyl-beta-(1-6)-glucosamine. Infect Immun. 2005;73(10):6752–6762.

10. Pozzi C, Wilk K, Lee JC, et al. Opsonic and protective properties ofantibodies raised to conjugate vaccines targeting sixStaphylococcus aureus antigens. Plos One. 2012;7(10):e46648.

11. Vlock D, Lee JC, Kropec-Huebner A, et al. Pre-clinical and initialphase i evaluations of a fully human monoclonal antibody directedagainst the PNAG surface polysaccharide on Staphylococcus aureus.Abstracts of the 50th ICAAC 2010; ASM Press; Abstract G1-1654/329;,2010.

12. Tojo M, Yamashita N, Goldmann DA, et al. Isolation and character-ization of a capsular polysaccharide adhesin from Staphylococcusepidermidis. J Infect Dis. 1988;157(4):713–722.

13. Mack D, Fischer W, Krokotsch A, et al. The intercellular adhesininvolved in biofilm accumulation of Staphylococcus epidermidis is alinear beta-1,6-linked glucosaminoglycan: purification and struc-tural analysis. J Bacteriol. 1996;178(1):175–183.

14. McKenney D, Hubner J, Muller E, et al. The ica locus ofStaphylococcus epidermidis encodes production of the capsularpolysaccharide/adhesin. Infect Immun. 1998;66(10):4711–4720.

15. McKenney D, Pouliot KL, Wang Y, et al. Broadly protective vaccinefor Staphylococcus aureus based on an in vivo-expressed antigen.Science. 1999;284(5419):1523–1527.

16. Cramton SE, Gerke C, Schnell NF, et al. The intercellular adhesion(ica) locus is present in Staphylococcus aureus and is required forbiofilm formation. Infect Immun. 1999;67(10):5427–5433.

17. Baldassarri L, Donnelli G, Gelosia A, et al. Purification and charac-terization of the staphylococcal slime-associated antigen and itsoccurrence among Staphylococcus epidermis clinical isolates. InfectImmun. 1996;64(8):3410–3415.

18. Muller E, Hübner J, Gutierrez N, et al. Isolation and characterizationof transposon mutants of Staphylococcus epidermidis deficient incapsular polysaccharide/adhesin and slime. Infect Immun. 1993;61(2):551–558.

19. Maira-Litran T, Kropec A, Abeygunawardana C, et al.Immunochemical properties of the staphylococcal poly-N-acetyl-glucosamine surface polysaccharide. Infect Immun. 2002;70(8):4433–4440.

20. Joyce JG, Abeygunawardana C, Xu Q, et al. Isolation, structuralcharacterization, and immunological evaluation of a high-molecu-lar-weight exopolysaccharide from Staphylococcus aureus.Carbohydr Res. 2003;338(9):903–922.

21. O’Gara JP. ica and beyond: biofilm mechanisms and regulation inStaphylococcus epidermidis and Staphylococcus aureus. FEMSMicrobiol Lett. 2007;270(2):179–188.

22. Wang X, Preston JF 3rd, Romeo T. The pgaABCD locus of Escherichiacoli promotes the synthesis of a polysaccharide adhesin requiredfor biofilm formation. J Bacteriol. 2004;186(9):2724–2734.

•• In 2004, Romeo and colleagues found the genes encoding thePNAG biosynthetic proteins in E. coli, leading to the identifica-tion of these genes in a large number of Gram-negativepathogens.

EXPERT REVIEW OF VACCINES 11

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 13: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

23. Choi AHK, Slamti L, Avci FY, et al. The pgaABCD locus ofAcinetobacter baumannii encodes the production of poly-beta-1-6-N-acetylglucosamine, which is critical for biofilm formation. JBacteriol. 2009;191(19):5953–5963.

24. Skurnik D, Davis MR Jr., Benedetti D, et al. Targeting pan-resistantbacteria with antibodies to a broadly conserved surface polysac-charide expressed during infection. J Infect Dis. 1709-1718;205(11):2012.

25. Chen K-M, Chiang M-K, Wang M, et al. The role of pgaC in Klebsiellapneumoniae virulence and biofilm formation. Microb Pathog.2014;77:89–99.

26. Parise G,MishraM, Itoh Y, et al. Role of a putative polysaccharide locusin Bordetella biofilm development. J Bacteriol. 2006;189(3):750–760.

27. Roux D, Cywes-Bentley C, Zhang YF, et al. Identification of poly-N-acetylglucosamine as a major polysaccharide component of theBacillus subtilis biofilm matrix. J Biol Chem. 2015;290(31):19261–19272.

28. Kropec A, Maira-Litran T, Jefferson KK, et al. Poly-N-acetylglucosa-mine production in Staphylococcus aureus is essential for virulencein murine models of systemic infection. Infect Immun. 2005;73(10):6868–6876.

29. Subashchandrabose S, Smith SN, Spurbeck RR, et al. Genome-widedetection of fitness genes in uropathogenic Escherichia coli duringsystemic infection. PLoS Pathog. 2013;9(12):e1003788.

30. Cerca N, Jefferson KK, Oliveira R, et al. Comparative antibody-mediated phagocytosis of Staphylococcus epidermidis cells grownin a biofilm or in the planktonic state. Infect Immun. 2006;74(8):4849–4855.

31. Heilmann C, Schweitzer O, Gerke C, et al. Molecular basis of inter-cellular adhesion in the biofilm-forming Staphylococcus epidermidis.Mol Microbiol. 1996;20(5):1083–1091.

• This publication provided the initial identification of thegenetic locus encoding proteins involved in PNAG synthesisand insight into the function of these proteins.

32. Gerke C, Kraft A, Süssmuth R, et al. Characterization of theN-acetylglucosaminyltransferase activity involved in the biosynth-esis of the Staphylococcus epidermidis polysaccharide intercellularadhesin. J Biol Chem. 1998;273(29):18586–18593.

33. Bobrov AG, Kirillina O, Forman S, et al. Insights into Yersinia pestisbiofilm development: topology and co-interaction of Hms innermembrane proteins involved in exopolysaccharide production.Environ Microbiol. 2008;10(6):1419–1432.

34. Skurnik D, Roux D, Pons S, et al. Extended spectrum antibodiesprotective against carbapenemase producing Enterobacteriaceae. JAntimicrob Chemother. 2016 Jan 7. pii: dkv448. [Epub ahead ofprint]

35. Whitfield GB, Marmont LS, Howell PL. Enzymatic modifications ofexopolysaccharides enhance bacterial persistence. Front Microbiol.2015;6:471.

• This review article presents an outstanding overview of thebiosynthesis of PNAG.

36. Bentancor LV, O’Malley JM, Bozkurt-Guzel C, et al. Poly-N-acetyl-beta-(1-6)-glucosamine is a target for protective immunity againstAcinetobacter baumannii infections. Infect Immun. 2012;80(2):651–656.

37. Yoong P, Cywes-Bentley C, Pier GB. Poly-N-acetylglucosamineexpression by wild-type Yersinia pestis is maximal at mammalian,not flea, temperatures. MBio. 2012;3(4):e00217–00212.

38. Kelly-Quintos C, Cavacini LA, Posner MR, et al. Characterization ofthe opsonic and protective activity against Staphylococcus aureusof fully human monoclonal antibodies specific for the bacterialsurface polysaccharide poly-N-acetylglucosamine. Infect Immun.2006;74(5):2742–2750.

•• This paper describes the production and characterization ofMAb F598 to PNAG, no in clinical trials in humans and alsoused as the reagent to detect PNAG among microbialpathogens.

39. Cerca N, Maira-Litrán T, Jefferson KK, et al. Protection againstEscherichia coli infection by antibody to the Staphylococcus aureus

poly-N-acetylglucosamine surface polysaccharide. Proc Natl AcadSci USA. 2007;104(18):7528–7533.

40. Kaplan JB, Velliyagounder K, Ragunath C, et al. Genes involved inthe synthesis and degradation of matrix polysaccharide inActinobacillus actinomycetemcomitans and Actinobacillus pleurop-neumoniae biofilms. J Bacteriol. 2004;186(24):8213–8220.

41. Ramasubbu N, Thomas LM, Ragunath C, et al. Structural analysis ofDispersin B, a biofilm-releasing glycoside hydrolase from the peri-odontopathogen Actinobacillus actinomycetemcomitans. J Mol Biol.2005;349(3):475–486.

•• The use of Dispersin B as a tool in identifying and studyingPNAG is exceptional. This paper reports on its properties andits specificity for the β-1-6-linkage between the glucosaminesin PNAG.

42. Skurnik D, Kropec A, Roux D, et al. Natural antibodies in normalhuman serum inhibit Staphylococcus aureus capsular polysacchar-ide vaccine efficacy. Clin Infect Dis. 2012;55(9):1188–1197.

43. Skurnik D, Merighi M, Grout M, et al. Animal and human antibodies todistinct Staphylococcus aureus antigens mutually neutralize opsonickilling and protection in mice. J Clin Invest. 2010;120(9):3220–3233.

44. Grachev AA, Gerbst AG, Gening ML, et al. NMR and conformationalstudies of linear and cyclic oligo-(1–>6)-beta-D-glucosamines.Carbohydr Res. 2011;346(15):2499–2510.

45. Verdier I, Durand G, Bes M, et al. Identification of the capsularpolysaccharides in Staphylococcus aureus clinical isolates by PCRand agglutination tests. J Clin Microbiol. 2007;45(3):725–729.

46. Shinefield H, Black S, Fattom A, et al. Use of a Staphylococcus aureusconjugate vaccine in patients receiving hemodialysis. N Engl J Med.2002;346(7):491–496.

47. Daum RS, Spellberg B. Progress toward a Staphylococcus aureusvaccine. Clin Infect Dis. 2012;54(4):560–567.

48. Kojima Y, Tojo M, Goldmann DA, et al. Antibody to the capsularpolysaccharide/adhesin protects rabbits against catheter-relatedbacteremia due to coagulase-negative staphylococci. J Infect Dis.1990;162(2):435–441.

49. Takeda S, Pier GB, Kojima Y, et al. Protection against endocarditisdue to Staphylococcus epidermidis by immunization with capsularpolysaccharide/adhesin. Circulation. 1991;84(6):2539–2546.

50. Farrell PM, Collins J, Broderick LS, et al. Association betweenmucoid Pseudomonas infection and bronchiectasis in childrenwith cystic fibrosis. Radiology. 2009;252(2):534–543.

51. Li Z, Kosorok MR, Farrell PM, et al. Longitudinal development ofmucoid Pseudomonas aeruginosa infection and lung disease pro-gression in children with cystic fibrosis. Jama. 2005;293(5):581–588.

52. Lu X, Skurnik D, Pozzi C, et al. A poly-N-acetylglucosamine-Shigatoxin broad-spectrum conjugate vaccine for Shiga toxin-producingEscherichia coli. Mbio. 2014;5(2):e00974-00914-e00974-00914.

53. Wallis R, Mitchell DA, Schmid R, et al. Paths reunited: Initiation ofthe classical and lectin pathways of complement activation.Immunobiol. 2010;215(1):1–11.

54. Brouwer N, Dolman KM, Van Houdt M, et al. Mannose-bindinglectin (MBL) facilitates opsonophagocytosis of yeasts but not ofbacteria despite MBL binding. J Immunol. 4124-4132;180(6):2008.

55. Garrett WS, Lord GM, Punit S, et al. Communicable ulcerative colitisinduced by T-bet deficiency in the innate immune system. Cell.2007;131(1):33–45.

56. Perez MM, Prenafeta A, Valle J, et al. Protection fromStaphylococcus aureus mastitis associated with poly-N-acetyl beta-1,6 glucosamine specific antibody production using biofilm-embedded bacteria. Vaccine. 2009;27(17):2379–2386.

57. Cerca N, Jefferson KK, Maira-Litran T, et al. Molecular basis forpreferential protective efficacy of antibodies directed to the poorlyacetylated form of staphylococcal poly-N-acetyl-beta-(1-6)-glucosa-mine. Infect Immun. 2007;75(7):3406–3413.

58. Lukacova M, Barak I, Kazar J. Role of structural variations of poly-saccharide antigens in the pathogenicity of Gram-negative bac-teria. Clin Microbiol Infect. 2008;14(3):200–206.

59. Samuel G, Reeves P. Biosynthesis of O-antigens: genes and path-ways involved in nucleotide sugar precursor synthesis andO-antigen assembly. Carbohydr Res. 2503-2519;338(23):2003.

12 D. SKURNIK ET AL.

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16

Page 14: The exceptionally broad-based potential of active and passive ......PNAG has also been found to be an important virulence factor for several pathogens. In three different strains of

60. Whitfield C. Biosynthesis and assembly of capsular polysaccharidesin Escherichia coli. Annu Rev Biochem. 2006;75:39–68.

61. Nordmann P, Cuzon G, Naas T. The real threat of Klebsiella pneu-moniae carbapenemase-producing bacteria. Lancet. 2009;9(4):228–236.

62. Poirel L, Pitout JD, Nordmann P. Carbapenemases: molecular diver-sity and clinical consequences. Future Microbiol. 2007;2(5):501–512.

63. Rasheed JK, Kitchel B, Zhu W, et al. New Delhi metallo-beta-lacta-mase-producing Enterobacteriaceae, United States. Emerg InfectDis. 2013;19(6):870–878.

64. Darton TC, Blohmke CJ, Moorthy VS, et al. Design, recruitment, andmicrobiological considerations in human challenge studies. Lancet.2015;15(7):840–851.

65. Hobbs MM, Sparling PF, Cohen MS, et al. Experimental gonococcalinfection in male volunteers: cumulative experience with Neisseriagonorrhoeae strains FA1090 and MS11mkC. Front Microbiol.2011;2:123.

66. Laurens MB, Billingsley P, Richman A, et al. Successful humaninfection with P. falciparum using three aseptic Anopheles stephensimosquitoes: A new model for controlled human malaria infection.Plos One. 2013;8(7):e68969.

67. Labandeira-Rey M, Dodd D, Fortney KR, et al. A Haemophilusducreyi CpxR deletion mutant is virulent in human volunteers. JInfect Dis. 2011;203(12):1859–1865.

68. Pollard AJ, Savulescu J, Oxford J, et al. Human microbial challenge:the ultimate animal model. Lancet. 2012;12(12):903–905.

69. Waddington CS, Darton TC, Jones C, et al. An outpatient, ambulant-design, controlled human infection model using escalating dosesof Salmonella typhi challenge delivered in sodium bicarbonatesolution. Clin Infect Dis. 2014;58(9):1230–1240.

70. Little DJ, Bamford NC, Pokrovskaya V, et al. Structural basis forthe de-N-acetylation of poly-beta-1,6-N-acetyl-D-glucosamine inGram-positive bacteria. J Biol Chem. 2014;289(52):35907–35917.

71. Little DJ, Li G, Ing C, et al. Modification and periplasmic trans-location of the biofilm exopolysaccharide poly-beta-1,6-N-acetyl-D-glucosamine. Proc Natl Acad Sci USA. 2014;111(30):11013–11018.

72. Nissen M, Marshall H, Richmond P, et al. A randomized phase Istudy of the safety and immunogenicity of three ascending doselevels of a 3-antigen Staphylococcus aureus vaccine (SA3Ag) inhealthy adults. Vaccine. 2015;33(15):1846–1854.

73. Lattar SM, Noto Llana M, Denoel P, et al. Protein antigens increase theprotective efficacy of a capsule-based vaccine against Staphylococcusaureus in a rat model of osteomyelitis. Infect Immun. 2014;82(1):83–91.

74. Taylor A, Foo SS, Bruzzone R, et al. Fc receptors in antibody-dependent enhancement of viral infections. Immunol Rev.2015;268(1):340–364.

EXPERT REVIEW OF VACCINES 13

Dow

nloa

ded

by [

T&

F O

ffic

e L

ocat

ions

], [

Flor

ence

Opa

ra]

at 0

6:21

14

Apr

il 20

16