Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jun 1.
Published in final edited form as: Expert Opin Ther Targets. 2012 Apr 25;16(6):601–612. doi: 10.1517/14728222.2012.682573

The potential use of toxin antibodies as a strategy for controlling acute Staphylococcus aureus infections

Gordon Y C Cheung 1, Michael Otto 2,
PMCID: PMC3354029  NIHMSID: NIHMS371338  PMID: 22530584

Abstract

Introduction

The pandemic human pathogen, Staphylococcus aureus, displays high levels of antibiotic resistance and is a major cause of hospital- and community-associated infections. S. aureus disease manifestation is to a great extent due to the production of a large arsenal of virulence factors, which include a series of secreted toxins. Antibodies to S. aureus toxins are found in people who are infected or asymptomatically colonized with S. aureus. Immunotherapies consisting of neutralizing anti-toxin antibodies could provide immediate aid to patients with impaired immune systems or in advanced stages of disease.

Areas covered

Important S. aureus toxins, their roles in pathogenesis, rationales for selecting S. aureus toxins for immunization efforts, and caveats associated with monoclonal antibody-based passive immunization are discussed. This review will focus on hyper-virulent community-associated methicillin-resistant S. aureus (CA-MRSA) because of their recent surge and clinical importance.

Expert opinion

Antibodies against genome-encoded toxins may be more broadly applicable than those directed against toxins found only in a sub-population of S. aureus isolates. Furthermore, there is substantial functional redundancy among S. aureus toxins. Thus, an optimal anti-S. aureus formulation may consist of multiple antibodies directed against a series of key S. aureus genome-encoded toxins.

Keywords: Toxin, vaccine, immunotherapy, methicillin-resistant Staphylococcus aureus, monoclonal antibody, alpha-hemolysin, phenol-soluble modulins, superantigens, leukotoxins, Panton-Valentine leukocidin

1. The need for immunotherapies and vaccines against Staphylococcus aureus

The commensal microorganism, Staphylococcus aureus, is a primary colonizer of human nares (in ~ 25% of the population) and the etiological agent of many diseases [1, 2]. Since the 1960s, the use of antibiotics to treat S. aureus-associated infections has become increasingly problematic owing to the emergence of methicillin-resistant S. aureus (MRSA) and S. aureus strains that are resistant to a wide variety of different antibiotics [2, 3]. Originally a pathogen primarily of immune-compromised and otherwise predisposed patients in hospital settings (hospital associated-MRSA; HA-MRSA) [4], more aggressive methicillin-resistant strains of S. aureus have recently appeared in the community (community-associated-MRSA; CA-MRSA), which may successfully infect healthy human people [13]. The majority of CA-MRSA infections are soft skin and tissue infections (SSTIs) (~ 50% of total cases). However, invasive cases of CA-MRSA-related diseases, such as necrotizing pneumonia (~ 5% of total cases), are becoming increasingly more widespread. As a consequence, the treatment of MRSA-infections places great financial strains on public healthcare [5].

It is commonly believed that the epidemiological success of the pulsed-field type USA300 CA-MRSA isolate, which is now pandemic in the United States, is due to a combination of high virulence, antibiotic resistance, and colonization capacity, leading to sustainable spread in the community [6]. The high number of community-associated infections with USA300 has prompted considerable research efforts aimed to develop therapeutics to combat CA-MRSA. These primarily include strategies interfering with virulence, including most notably active and passive immunization efforts directed against CA-MRSA toxins and other virulence factors (reviewed extensively in [710]).

Here, an overview on a number of S. aureus toxin families, with a focus on S. aureus toxins encoded exclusively by, or showing increased expression in, CA-MRSA strains will be provided. The roles that those toxins have in S. aureus pathogenesis and disease will be discussed. Finally, the rationales and caveats regarding the use of anti-toxin monoclonal antibodies (mAbs) as therapeutics for the prevention and treatment of CA-MRSA-mediated disease will be explored.

2. Toxins involved in S. aureus pathogenesis

S. aureus produces and secretes many types of toxins with diverse roles in pathogenesis, particularly affecting immune evasion and activation of the immune response. These include alpha-hemolysin (α-hemolysin, Hla), beta-toxin (β-toxin), the superantigens (SAgs), the leukotoxins, and the phenol-soluble modulins (PSMs). The genes coding for β-toxin, the SAgs and leukotoxins are mostly encoded by mobile genetic elements (MGEs) [11]; therefore, toxin expression can differ tremendously between different strains of S. aureus. Only a few S. aureus toxins, such as α-hemolysin and the PSMs, are genome-encoded [12, 13] and expressed by both HA- and CA-MRSA strains. However, α-hemolysin and the PSMs are expressed more strongly in CA-MRSA compared to HA-MRSA strains. This suggests that differences in gene regulation may influence the epidemiological success of CA-MRSA strains such as USA300 [14, 15]. For example, it has been noted that USA300 shows increased expression of the global regulatory quorum sensing system, accessory gene regulator (agr) [16], which controls the expression of many virulence factors in a cell density-dependent manner. The agr locus consists of a divergent promoter, which controls the transcription of RNAII and RNAIII. The RNAII transcript contains 4 genes encoding a classical two-component quorum sensing system (agrAC), an auto-inducing peptide (AIP) precursor (agrD), and the AIP maturation and exporter protein (agrB). Upon increasing concentration of AIP during growth, the AgrAC two-component system becomes activated at a certain threshold concentration of AIP. Upon binding of AIP to the membrane protein AgrC, AgrA is activated, which in turn binds to the divergent promoter and induces transcription of RNAII and RNAIII. RNAIII is a regulatory RNA molecule that regulates the expression of a majority of toxins [16]. S. aureus mutants lacking a functional agr regulatory system are significantly less virulent than the corresponding wild-type strains, which has recently been demonstrated specifically for USA300 [14, 1719].

2.1 Leukotoxins

Neutrophils, or polymorphonuclear leukocytes (PMNs), are key components of the innate immune system and involved in controlling S. aureus infection [20, 21]. Possibly for that reason, S. aureus produces a large variety of virulence factors that inhibit neutrophil function [22], allowing S. aureus to circumvent elimination by innate host defense [23]. Particularly, pore-forming, bi-component leukotoxins with cytolytic affinity towards cells of myeloid lineage, such as monocytes, macrophages and neutrophils, represent key contributors to S. aureus immune evasion [24, 25]. Each leukotoxin requires one class S and one class F protein, which are individually non-toxic, to form a β-barreled pore-forming structure upon oligomerization [26]. Six class S subunits (LukS-PV, HlgA, HlgC, LukE, LukM, LukH) and five class F subunits (LukF-PV, HlgB, LukD, LukF′-PV, LukG) have been described [24, 25, 27, 28]. One exception to this monogamous pairing is the γ-hemolysin gene cluster, which comprises three genes ((hlgA [or hlg2], hlgC [or lukS] and hlgB [or lukF]), whose gene products allow the formation of two functional pairs of proteins: Hlg [HlgA + HlgB] and Luk [HlgB + HlgC]. Notably, all leukotoxins reportedly contribute to disease progression in at least some animal infection models, with the exception of LukM/LukF′-PV [2931].

While the hlg gene cluster occurs in 99% of S. aureus strains [3234], many other leukotoxin genes are not uniformly present among S. aureus isolates. The lukDE [35] and lukGH (lukAB) [24, 25] genes can be found in many sequenced strains or clinical isolates including USA300 [31]. However, for example, lukDE are not found in the HA-MRSA isolate, USA200 [36] and the phage-encoded lukF-PV and lukS-PV [33], which code for Panton-Valentine leukocidin (PVL), are mostly restricted to CA-MRSA strains [37], represented by only ~ 5% of all clinical isolates. Of note, the role of PVL in S. aureus pathogenesis remains controversial despite extensive research that has been performed on that leukotoxin [1, 15, 38, 39]. Owing to the initially observed epidemiological link of lukSF-PV with CA-MRSA, it has been suggested that PVL contributes to the pathogenesis of typical CA-MRSA disease manifestations, such as necrotizing pneumonia [15, 40] or the formation of skin abscesses [41, 42]. However, PVL-negative CA-MRSA strains have since been isolated, indicating that this epidemiological correlation is not absolute [1, 15]. Furthermore, animal infection studies investigating the role of PVL in S. aureus disease are not in agreement [4350]. These discrepancies may in part be due to differences of the animal models used, such as inoculum size and host species. Over the last decade, the PVL controversy may have overshadowed the importance of other, and perhaps more relevant, leukotoxins that are expressed by a majority of CA-MRSA strains, such as lukDE and lukGH. These leukotoxins are now being investigated more intensely. Studies using isogenic S. aureus deletion mutants show that LukDE and LukGH contribute to the virulence of S. aureus in murine sepsis [36] and renal abscess models [25], respectively. Furthermore, there have been some, albeit relatively few studies on the contribution of γ-hemolysin to S. aureus pathogenesis and disease. These studies indicate that γ-hemolysin may have roles in septic arthritis and weight loss in mice [51] and endophthalmitis in rabbits [52]. However, it is unknown whether LukDE, LukGH or γ-hemolysin contribute towards other facets of HA- and CA-MRSA disease, for example, severe cases of pneumonia, as has been suggested for PVL.

2.2. Staphylococcal superantigens

The superantigen (SAg) family consists of small, serologically distinct proteins including staphylococcal enterotoxins (SEs) A–E, G–J, staphylococcal enterotoxin-like toxins (SEl) K-R, U and X, and Toxic Shock Syndrome Toxin-1 (TSST-1), all of which are ~ 20 – 28 kDa in size [34, 53, 54]. Members of the SAg family are responsible for food poisoning, toxic shock syndrome and respiratory disease [55]. The SAgs share a common protein structure [53, 56] that enables each SAg to crosslink different alleles of major histocompatibility complex (MHC) class II molecules on the surfaces of antigen-presenting cells with different alleles of the variable region of T-cell receptors (TCRs) [5759]. These interactions allow the SAgs to bypass the conventional antigen processing pathway to non-specifically activate T-cells (~ 50%) and induce the irregular production of high levels of inflammatory cytokines [60]. This event induces multisystem disease that includes rash, hypotension, pyrexia, emesis and diarrhea [61]. The SAgs also cause severe life-threatening multiple organ failure [61].

Of the large number of SAgs that have been described, only the recently discovered SElX is genome-encoded [54]; all other SAgs are encoded by MGEs [6264]. Moreover, not all S. aureus isolates display superantigenic activity [65] because the SAgs are not distributed equally among S. aureus strains [66]. Furthermore, it is not unusual to see varied levels of SAg production among S. aureus strains that express SAgs [6772]. This is attributed to the involvement of at least three global regulators; agr [73], sarA [74], σB and saeRS [75]. Interestingly, TSST-1 and SEB have been reported to act as negative global regulators of toxin production [76].

The roles of SAgs in the pathogenesis of S. aureus disease have been investigated in animal models by administration of purified SAgs [77]. Other studies have compared the differences in the capacities of naturally occurring SAg+ and SAg strains [78] and isogenic SAg mutants [54, 7981] to cause disease in animals. The overall importance of the SAgs in CA-MRSA pathogenesis is debatable, because many SAgs that are classically associated with S. aureus disease are not expressed by CA-MRSA strains. For instance, USA300 does not typically produce TSST-1, SEB or SEC, SAgs that are most often associated with toxic shock [73, 82]. As one noticeable exception, SElX has been shown to contribute to USA300 necrotizing pneumonia in rabbits [54]. However, it is unknown if SElX contributes to other manifestations of CA-MRSA disease, such as SSTIs.

2.3. Alpha-hemolysin

The genome-encoded α-hemolysin (Hla), one of the first toxins described for S. aureus, has recently received considerable attention as a target for an anti-S. aureus vaccine and the production of neutralizing antibodies for passive immunization. α-hemolysin is produced by a majority of S. aureus strains and its expression is regulated by at least three global regulatory systems, which include agr [83]. It is well known for its pore-forming [13, 8489] and pro-inflammatory [90] properties. However, its cognate receptor, the zinc-dependent metalloprotease ADAM10 (A disintegrin and metalloproteinase 10), was only recently discovered [91, 92]. α-hemolysin contributes significantly to the pathogenesis of S. aureus USA300-induced skin infection and pneumonia [46, 93, 94]. Mice that do not express ADAM10 are resistant to lethal infection with S. aureus strain USA300 in a pneumonia model [91], but it is not known if ADAM10 is also required for α-hemolysin induced abscess formation by S. aureus. Another signal, which is found on immune cells, may also be required for α-hemolysin-induced pneumonia by S. aureus [95]. This second signal belongs to a class of cytosolic proteins called nucleotide-binding domain and leucine-rich repeat containing (NLR) proteins, which are important activators of the innate immune response. NLRs defend against pathogen infection and endogenous damage in response to a variety of immunogenic stimuli [96]. Upon activation, several members of this family assemble into large multimeric protein complexes called inflammasomes, which induce inflammatory responses. NLRP3 in particular is activated in response to bacterial toxins [97], including S. aureus β-toxin, γ-hemolysin [98] and α-hemolysin [95]. Thus, NLPR3 inflammasome activation and the presence of ADAM10 appear to contribute to the development of S. aureus pneumonia.

2.4 Phenol-soluble modulins

The phenol-soluble modulins (PSMs) are a family of small, amphipathic peptides with cytolytic and pro-inflammatory properties towards a range of cells types including erythrocytes and neutrophils [12]. The pro-inflammatory properties of PSMs (such as, induction of intracellular calcium flux, chemotaxis, and cytokine production in neutrophils) are dependent on binding to their cognate receptor, human formyl peptide receptor 2 (FPR2). Of note, PSM-dependent cell lysis occurs independently of FPR2 binding [99]. Two PSM sub-families can be distinguished: PSMα peptides, which include the well-known δ-toxin, are 20 – 26 amino acids, and PSMβ peptides 43 – 44 amino acids in length [12]. Generally, members of the PSMα class show more pronounced pro-inflammatory and especially lytic activities than members of the PSMβ class [12].

Similar to many other S. aureus virulence determinants, PSMs are under control of the agr global regulatory system [12, 100]. However, in contrast to most Agr targets, they are regulated directly by AgrA rather than via RNAIII [100, 101]. Most S. aureus strains contain the RNAIII-embedded hld gene encoding δ-toxin [102], and the operons coding for the psmα and the psmβ genes, which are located on two separate loci [12]. In contrast to the ubiquitous prevalence of the psmα and the psmβ genes, the psm-mec gene [103] is restricted to HA-MRSA strains carrying staphylococcal cassette chromosome mec (SCCmec) types II, III and VIII [101].

Importantly, CA-MRSA isolates secrete higher amounts of PSMs in average compared to HA-MRSA isolates [12], suggesting that the PSMs have a prominent role in CA-MRSA pathogenesis. Indeed, isogenic CA-MRSA psmα mutants are severely impaired in their ability to form abscesses and cause sepsis in mice [12]. Collectively, the α-class PSMs, including δ-toxin, and the β-class PSMs all play a role in the dissemination of CA-MRSA from biofilm-related catheter infections in mice [104]. Biofilms are sticky agglomerations consisting of a mixture of bacterial cells and extracellular polysaccharide matrices and are intrinsically resistant to antibiotics and mechanisms of host defense [105]. The detergent-like quality of PSMs [12, 106] influences the development of mature biofilms by breaking interactions between cell populations, introducing channel formation, followed by the detachment of cell clusters, leading to bacterial dissemination in vivo [104, 107]. Finally, PSMs act synergistically with β-toxin [108, 109] and PVL [110] in vitro. Since important CA-MRSA isolates do not produce β-toxin [111] and not all CA-MRSA isolates harbor the lukF-PV and lukS-PV genes, this synergism may infer a cooperative relationship between virulence factors produced by different strains of S. aureus that have different toxin expression profiles.

3. Anti-toxin antibody responses in humans

Soon after birth, 40 – 50% of the population carry S. aureus. By 14 months of age, there can be a ~ 40% reduction in carriage rates [112, 113] and S. aureus can remain a persistent colonizer in the nares of adults in ~ 25% of the population [114]. In healthy human individuals, antibody responses toward many S. aureus antigens can be detected, such as cell surface components, non-protein antigens, and toxins [54, 115118], indicating that S. aureus antigens are immunogenic.

Antibodies against most S. aureus toxins are not readily detected in healthy newborns although anti-TSST-1 antibodies were reported [119]. In healthy adults, carriers of S. aureus can produce greater levels of antibodies towards S. aureus antigens than non-carriers, but these titers are generally less pronounced at the age of 65 and higher [116]. Of the anti-toxin antibodies, only significantly higher levels of anti-TSST-1 and anti-SEA have been described in carriers compared to non-carriers; and it is thought that previous exposure(s) to different S. aureus strains may account for the diverse levels of antibody titers between individuals [116, 117].

Individuals infected with S. aureus generally produce greater levels of antibodies to PVL, α-hemolysin, and SAgs than healthy individuals [44, 115, 119, 120]. Furthermore, low levels of antibodies may be associated with more severe S. aureus infections [120]. However, presence of antibodies is not necessarily associated with protection, as recently described for the role of anti-PVL antibodies in PVL+ CA-MRSA infections in humans [121].

Sera from S. aureus-infected patients may demonstrate neutralizing activity against S. aureus toxins, such as SAgs [117, 122, 123] but whether there is neutralizing activity of these sera towards other S. aureus toxins remains to be determined. Altogether, although much information has been garnered from these serological studies, the roles that anti-S. aureus antibodies play during infection still remains largely unknown.

4. S. aureus toxins as targets for monoclonal antibody therapy

Unlike active immunization, which sometimes requires repeated boosters and long periods of time for maximum immune responses to be generated, passive immunization would provide immediate treatment for unvaccinated patients to help reduce the severity of acute S. aureus disease. Perhaps even more importantly, mAb therapy would benefit immune-compromised patients or infants with immature immune systems or even add to the existing pool of potentially protective anti-S. aureus antibodies. Clearly, it is important for such therapy to take effect that S. aureus is rapidly identified. Moreover, late stages of S. aureus infection may not be amenable to passive therapy, as advanced stages of toxin-mediated tissue destruction will have ensued. Even though there are caveats to passive immunization, much research has recently been performed on neutralizing mAbs against S. aureus toxins; and some of these successfully protected from S. aureus infection in animal models (Table 1). Polyclonal anti-toxin antibodies also are protective in animal models of S. aureus infection (Table 1). However, many of the investigated toxins, such as PVL and SEB, are not expressed by all S. aureus strains. Therefore, anti-PVL and anti-SEB antibodies may only be useful in treating acute infections caused by PVL and SEB-expressing S. aureus strains. Instead, core-genome encoded toxins, such as α-hemolysin, SElX, LukGH, and PSMs may better serve as targets for antibody generation, as antibodies against those toxins may be more broadly applicable for acute cases of S. aureus infections. The use of these toxins as targets for mAb generation will be discussed in more detail below.

Table 1.

Validated toxin targets

Toxin Challenge agent Antibody type Model Species Observed effect Reference
α-hemolysin S. aureus USA300 Rabbit polyclonal Pneumonia Mouse + [46]
S. aureus USA300 Mouse monoclonal Abscess Mouse + [146]
S. aureus USA300, CC30, CC5 Mouse humanized monoclonal Abscess Mouse + [144]
S. aureus USA300 Rabbit polyclonal Abscess Mouse + [145]
SEBa SEB Mouse monoclonal SEB-induced lethal shock Mouse + [136]
SEB Chicken polyclonal Aerosolized SEB Monkey + [137]
PVLb PVL Mouse humanized heavy chain Endophthlamitis Rabbit + [124]
S. aureus USA300 PVL+ Rabbit Polyclonal Pneumonia Mouse [46]
Several S. aureus USA300 PVL+ strains Rabbit Polyclonal Abscess Mouse [127]
S. epidermidis PSMβc S. epidermidis 1457 Mouse polyclonal Subcutaneous catheter-associated biofilm model Mouse + [107]

Abbreviations;

a

Staphylococcal enterotoxin B,

b

Panton-Valentine leukocidin,

c

Phenol-soluble modulin,

4.1 Antibody therapy against the leukotoxins

Even though there is controversy over whether PVL is a decisive virulence determinant in S. aureus, there is still strong interest in creating a vaccine against PVL and producing anti-PVL mAbs. There are several points supporting the use of anti-PVL antibodies for passive immunization. First, humans are capable of generating humoral immune responses against PVL [115, 117, 121]. Second, polyclonal and mAbs to PVL are neutralizing [121, 124, 125]. Third, injection of PVL together with a tetravalent anti-PVL mAb, which binds both LukS-PV and LukF-PV antigens simultaneously, reduced inflammation and tissue destruction in a non-lethal model of endophthalmitis in rabbits [124]. Finally, anti-PVL mAbs exhibit cross-reactivity with other leukotoxins, such as HlgC [124]. On the other hand, there are many important findings that negate the benefits of using anti-PVL antibodies. First, polyclonal anti-PVL antibodies failed to neutralize the cytotoxicity of a PVL-expressing USA300 strain toward human neutrophils in vitro [126]. Second, anti-PVL immune sera did not protect from S. aureus pneumonia in passive immunization experiments using mice [46]. Third, PVL-neutralizing antibodies did not protect children from primary or recurrent CA-MRSA-associated SSTIs [121]. Finally, the presence of anti-PVL antibodies may in fact exacerbate disease caused by PVL-expressing CA-MRSA during early stages of infection [127]. These antibodies may prevent the interactions of PVL with cells of the innate immune system, resulting in poor activation of the immune response and a decreased ability to mount an attack to the infection.

LukDE and LukGH (LukAB) also interact with innate immune cells [24, 25] and are lytic toward neutrophils [24, 25]. Furthermore, PVL and LukGH lyse other innate immune cells, such as human monocytes and macrophages [25, 128]. S. aureus strains that express one or more of these leukotoxins at sufficient levels thus may severely impair the ability of the host to mount an effective immune response. However, it remains to be investigated whether antibodies to leukotoxins other than PVL also may enhance the virulence of S. aureus in a way similar to anti-PVL antibodies. Finally, it has been reported that active immunization with PVL subunits relieves mice of severe skin and intranasal infection with PVL-expressing CA-MRSA [44]. Phase II clinical trials are currently underway using a recombinant LukS-PV toxin subunit. The results of this study have yet to be released [129].

Taken together, there are strong arguments against the use of anti-PVL antibodies to treat CA-MRSA disease. Furthermore, there is currently no compelling evidence for a necessary role of PVL in CA-MRSA pathogenesis, leaving the possibility that CA-MRSA strains without PVL may further expand or PVL-positive CA-MRSA strains may lose the PVL-encoding prophage without a substantial loss of virulence characteristics. Moreover, pursuing development of an anti-PVL mAb- or antigen-based vaccine is problematic from a marketing point of view, because only ~ 5% of S. aureus clinical isolates possess the genes coding for PVL, limiting its applicability. Finally, although active immunization with PVL subunits appears to be an encouraging alternative to passive immunization, active vaccination with a combination of LukGH or LukDE subunits may help protect against a larger number of S. aureus infections.

4.2 Antibody therapy against the superantigens

The SAgs are responsible for a range of symptoms and diseases in humans that range from food poisoning to toxic shock. Passive therapy with anti-SAg antibodies would likely help alleviate the symptoms associated with SAg-related disease by preventing the interactions of the SAgs with their ligands. Antibodies against TSST-1, SEA, SEB, SEC are readily detected in healthy individuals [117, 130132] and individuals with active S. aureus infections [119], indicating that these SAgs are immunogenic.

There is particular interest in developing neutralizing antibodies against the category B bioterrorism agent, SEB, which is resistant to denaturing and highly toxic to humans [133, 134]. Anti-SEB mAbs are neutralizing in vitro [135] and passive therapy with anti-SEB antibodies successfully prevents SEB-induced disease manifestations in mice [136] and monkeys [137] against lethal challenge with purified SEB. While there is certainly cross-reactivity of anti-SAg mAbs with several SAgs [138], it is unlikely that a monovalent SEB mAb will protect from infection by many SEB- expressing S. aureus strains of high clinical importance, such as the pandemic CA-MRSA strains. In contrast, passive immunization with several different anti-SAg mAbs would potentially be able to protect against a larger variety of S. aureus strains. Furthermore, the genome-encoded SElX may serve as an attractive alternative target for generating mAbs, because it is expressed by a majority of S. aureus isolates including the CA-MRSA strain USA300 [54]. Finally, passive immunization is not the only strategy for interfering with SAg-producing strains; other strategies include active immunization with detoxified SAgs [139141], the use of soluble forms of engineered TCR Vβ proteins [142], and hybrid TCR Vβ/MHCII proteins [143] that can block the binding activity of SEB toward its natural ligands found on T cells and antigen-presenting cells.

4.3 Antibody therapy against α-hemolysin

Healthy human individuals that are colonized with S. aureus produce high levels of antibodies directed toward α-hemolysin [116, 117]. As α-hemolysin is expressed by a majority of S. aureus strains, passive immunization with mAbs to α-hemolysin may be effective against many different S. aureus strains including strains of CA-MRSA. Several studies have shown the protective efficacy of anti-α-hemolysin antibodies in S. aureus abscess [144, 145] and pneumonia [46, 146] murine infection models. In addition, active immunization with a detoxified α-hemolysin derivative (H35L) was shown to reduce the severity of S. aureus-induced pneumonia [46] and S. aureus abscess [145] infections. Interestingly, recombinant α-hemolysin toxoid is being tested in the same phase II clinical trial as recombinant LukS-PV [129].

4.4 Antibody therapy against the phenol-soluble modulins

The genome-encoded PSMs show little variation in amino acid sequences between S. aureus strains [12]. Therefore, antibodies against PSMs could serve as an effective passive immunization strategy against a broad spectrum of S. aureus strains. Infants with invasive S. aureus infections do not appear to produce antibodies to the strongly cytolytic PSMα3 [147], but anti-PSM antibodies can be raised in animals injected with PSMs and adjuvant [107, 148] indicating that the PSMs have immunogenic potential. High levels of PSMs are produced by CA-MRSA strains in vitro [12, 14], but the levels of PSM in-vivo production are yet unknown. Therefore, the lack of anti-PSM antibody production, which is observed at least in infants, may either be due to a low production level of PSMs under those conditions, or if the infection is localized, such as in a SSTI, the host may not be able to detect enough PSMs in order to generate antibodies. Antibodies against PSMs may be more readily detected in adults or in other types of S. aureus diseases. However, this needs to be determined by including the relatively recently discovered PSMs as antigens in future human serological studies.

Although S. aureus catheter-related biofilm infections are not a focus of this review, this subject area warrants a brief discussion. PSMs play a crucial role in biofilm structuring and dissemination of biofilm-associated infection in these organisms [104, 107]. Notably, anti-PSMβ antibodies blocked the dissemination of S. epidermidis catheter infection in mice. Therefore, passive immunization with anti-PSM antibodies may be useful to inhibit the dissemination of bacteria from biofilm formed on catheters. However, more research is clearly needed to investigate antibody therapy against PSMs.

5. Expert opinion

There is an urgent need for research aimed to find a successful immunization strategy against S. aureus infections. This is warranted in particular because of the high levels of antibiotic resistance shown by pandemic strains of S. aureus. Antibodies against S. aureus toxins may have particular value, as they neutralize the binding affinity of the toxins, preventing pro-inflammatory responses and cytolytic activities. Furthermore, they may block toxin oligomerization even after insertion into cell membranes, thus preventing cytolytic activity of the toxin.

Choosing a specific S. aureus toxin as a singular vaccine target is complicated by the fact that S. aureus produces many, often functionally redundant toxins. Some toxins are immunogenic and play important roles in S. aureus pathogenesis, but many are not expressed by all S. aureus isolates. Therefore, targeting core-genome encoded toxins, such as α-hemolysin, LukGH, SElX or the PSMs, would serve as a better basis for vaccine development than toxins only produced by a sub-population of strains, such as PVL, TSST-1, SEA, SEB and SEC, especially when aiming for a vaccine preparation with broad applicability.

Monoclonal antibodies raised against only one S. aureus antigen are protective in animal models of S. aureus infection, but frequently, only disappointing results were achieved with many of those mAbs in human phase 1/2 clinical trials (reviewed in [149]). An alternative, and perhaps more successful, strategy would comprise a mAb preparation consisting of antibodies against different toxins and possibly other S. aureus virulence factors, such as surface proteins, to simultaneously target establishment and exacerbation of infection. In theory, such antibody therapy would thus be able to inhibit different aspects of S. aureus pathogenesis. Interestingly, it was recently shown that administration of two different mAbs against S. aureus SEB led to significantly more protection than one singular SEB-directed mAb [136]. This concept is not new - similar results have been achieved with mAbs against toxins produced by other bacterial species, such as Bacillus anthracis anthrax toxin [150], Clostridium botulinum neurotoxin A [151] and C. difficile toxins A and B [152]. However, it needs to be further explored whether such a vaccination strategy, i.e. using antibodies against different S. aureus antigens, would be broadly effective against S. aureus. Moreover, it needs to be cautiously evaluated whether antitoxin antibodies may have the opposite effect of exacerbating S. aureus disease, such as in the case of passive immunization with anti-PVL antibodies [127].

In summary, unsuccessful clinical trials with predominantly non-toxin-based S. aureus antigens warrant new approaches to protect against the new wave of pandemic CA-MRSA strains in addition to the continuously high disease burden due to HA-MRSA. There is strong evidence from laboratory experiments suggesting that passive immunization approaches using anti-toxin antibodies may be as successful as those directed toward other S. aureus antigens. However, human clinical trials will always be required as ultimate proof of efficacy against S. aureus infection [9, 136, 153].

Highlights.

  • Passive immunization with one antibody or active immunization with one antigen may not be sufficient to protect against different epidemic S. aureus strains.

  • Genome-encoded virulence factors, such as SElX, PSMs and α-hemolysin, as targets would allow universal treatment against different epidemic S. aureus strains.

  • Active immunization against leukotoxins may be advantageous. However, other leukotoxins, such as LukDE and LukGH, should be considered in addition to PVL.

  • Passive immunization with multiple mAbs against different S. aureus virulence factors may be more beneficial than passive immunization with one mAb.

Footnotes

Declaration of Interest:

This work was supported by the Intramural Research Program of the National Institute of Allergy and Infectious Diseases, NIH.

Contributor Information

Gordon Y. C. Cheung, Email: cheunggo@niaid.nih.gov, Laboratory of Human Bacterial Pathogenesis, NIAID, NIH, Bldg. 33, Room 1W10, 9000 Rockville Pike, Bethesda, MD 20892, USA, Tel. +1 301 443 5211

Michael Otto, Email: motto@niaid.nih.gov, Laboratory of Human Bacterial Pathogenesis, NIAID, NIH, Bldg. 33, Room 1W10, 9000 Rockville Pike, Bethesda, MD 20892, USA, Tel. +1 301 443 5209

References

Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.

  • 1••.Otto M. Basis of virulence in community-associated methicillin-resistant Staphylococcus aureus. Annu Rev Microbiol. 2010;64:143–62. doi: 10.1146/annurev.micro.112408.134309. A comprehensive overview on the basis of CA-MRSA pathogenesis and disease. [DOI] [PubMed] [Google Scholar]
  • 2.DeLeo FR, Otto M, Kreiswirth BN, Chambers HF. Community-associated meticillin-resistant Staphylococcus aureus. Lancet. 2010;375:1557–68. doi: 10.1016/S0140-6736(09)61999-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Daum RS. Clinical practice. Skin and soft-tissue infections caused by methicillin-resistant Staphylococcus aureus. N Engl J Med. 2007;357:380–90. doi: 10.1056/NEJMcp070747. [DOI] [PubMed] [Google Scholar]
  • 4.Lindsay JA, Holden MT. Staphylococcus aureus: superbug, super genome? Trends Microbiol. 2004;12:378–85. doi: 10.1016/j.tim.2004.06.004. [DOI] [PubMed] [Google Scholar]
  • 5.Klevens RM, Morrison MA, Nadle J, et al. Invasive methicillin-resistant Staphylococcus aureus infections in the United States. Jama. 2007;298:1763–71. doi: 10.1001/jama.298.15.1763. [DOI] [PubMed] [Google Scholar]
  • 6.Moran GJ, Krishnadasan A, Gorwitz RJ, et al. Methicillin-resistant S. aureus infections among patients in the emergency department. N Engl J Med. 2006;355:666–74. doi: 10.1056/NEJMoa055356. [DOI] [PubMed] [Google Scholar]
  • 7.Broughan J, Anderson R, Anderson AS. Strategies for and advances in the development of Staphylococcus aureus prophylactic vaccines. Expert Rev Vaccines. 2011;10:695–708. doi: 10.1586/erv.11.54. [DOI] [PubMed] [Google Scholar]
  • 8.Spellberg B, Daum R. Development of a vaccine against Staphylococcus aureus. Semin Immunopathol. 2011 doi: 10.1007/s00281-011-0293-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Schaffer AC, Lee JC. Vaccination and passive immunisation against Staphylococcus aureus. Int J Antimicrob Agents. 2008;32 (Suppl 1):S71–8. doi: 10.1016/j.ijantimicag.2008.06.009. [DOI] [PubMed] [Google Scholar]
  • 10.Dedent A, Kim HK, Missiakas D, Schneewind O. Exploring Staphylococcus aureus pathways to disease for vaccine development. Semin Immunopathol. 2011 doi: 10.1007/s00281-011-0299-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Li M, Cheung GY, Hu J, et al. Comparative analysis of virulence and toxin expression of global community-associated methicillin-resistant Staphylococcus aureus strains. J Infect Dis. 2010;202:1866–76. doi: 10.1086/657419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12•.Wang R, Braughton KR, Kretschmer D, et al. Identification of novel cytolytic peptides as key virulence determinants for community-associated MRSA. Nat Med. 2007;13:1510–4. doi: 10.1038/nm1656. First study showing the key contribution of PSMs in CA-MRSA disease. [DOI] [PubMed] [Google Scholar]
  • 13.Bhakdi S, Tranum-Jensen J. Alpha-toxin of Staphylococcus aureus. Microbiol Rev. 1991;55:733–51. doi: 10.1128/mr.55.4.733-751.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cheung GY, Wang R, Khan BA, et al. Role of the accessory gene regulator agr in community-associated methicillin-resistant Staphylococcus aureus pathogenesis. Infect Immun. 2011;79:1927–35. doi: 10.1128/IAI.00046-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Diep BA, Otto M. The role of virulence determinants in community-associated MRSA pathogenesis. Trends Microbiol. 2008;16:361–9. doi: 10.1016/j.tim.2008.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Novick RP. Autoinduction and signal transduction in the regulation of staphylococcal virulence. Mol Microbiol. 2003;48:1429–49. doi: 10.1046/j.1365-2958.2003.03526.x. [DOI] [PubMed] [Google Scholar]
  • 17.Gillaspy AF, Hickmon SG, Skinner RA, et al. Role of the accessory gene regulator (agr) in pathogenesis of staphylococcal osteomyelitis. Infect Immun. 1995;63:3373–80. doi: 10.1128/iai.63.9.3373-3380.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Cheung AL, Eberhardt KJ, Chung E, et al. Diminished virulence of a sar-/agr-mutant of Staphylococcus aureus in the rabbit model of endocarditis. J Clin Invest. 1994;94:1815–22. doi: 10.1172/JCI117530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Abdelnour A, Arvidson S, Bremell T, et al. The accessory gene regulator (agr) controls Staphylococcus aureus virulence in a murine arthritis model. Infect Immun. 1993;61:3879–85. doi: 10.1128/iai.61.9.3879-3885.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Miller LS, Cho JS. Immunity against Staphylococcus aureus cutaneous infections. Nat Rev Immunol. 2011;11:505–18. doi: 10.1038/nri3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Graves SF, Kobayashi SD, DeLeo FR. Community-associated methicillin-resistant Staphylococcus aureus immune evasion and virulence. J Mol Med. 2010;88:109–14. doi: 10.1007/s00109-009-0573-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22••.DeLeo FR, Diep BA, Otto M. Host defense and pathogenesis in Staphylococcus aureus infections. Infect Dis Clin North Am. 2009;23:17–34. doi: 10.1016/j.idc.2008.10.003. A comprehensive review on host immune responses to S. aureus and mechanisms of S. aureus immune evasion. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Voyich JM, Braughton KR, Sturdevant DE, et al. Insights into mechanisms used by Staphylococcus aureus to avoid destruction by human neutrophils. J Immunol. 2005;175:3907–19. doi: 10.4049/jimmunol.175.6.3907. [DOI] [PubMed] [Google Scholar]
  • 24.Ventura CL, Malachowa N, Hammer CH, et al. Identification of a novel Staphylococcus aureus two-component leukotoxin using cell surface proteomics. PLoS One. 2010;5:e11634. doi: 10.1371/journal.pone.0011634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Dumont AL, Nygaard TK, Watkins RL, et al. Characterization of a new cytotoxin that contributes to Staphylococcus aureus pathogenesis. Mol Microbiol. 2011;79:814–25. doi: 10.1111/j.1365-2958.2010.07490.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kaneko J, Kamio Y. Bacterial two-component and hetero-heptameric pore-forming cytolytic toxins: structures, pore-forming mechanism, and organization of the genes. Biosci Biotechnol Biochem. 2004;68:981–1003. doi: 10.1271/bbb.68.981. [DOI] [PubMed] [Google Scholar]
  • 27.Prevost G, Mourey L, Colin DA, Menestrina G. Staphylococcal pore-forming toxins. Curr Top Microbiol Immunol. 2001;257:53–83. doi: 10.1007/978-3-642-56508-3_4. [DOI] [PubMed] [Google Scholar]
  • 28.Morinaga N, Kaihou Y, Noda M. Purification, cloning and characterization of variant LukE-LukD with strong leukocidal activity of staphylococcal bi-component leukotoxin family. Microbiol Immunol. 2003;47:81–90. doi: 10.1111/j.1348-0421.2003.tb02789.x. [DOI] [PubMed] [Google Scholar]
  • 29.Kaneko J, Muramoto K, Kamio Y. Gene of LukF-PV-like component of Panton-Valentine leukocidin in Staphylococcus aureus P83 is linked with lukM. Biosci Biotechnol Biochem. 1997;61:541–4. doi: 10.1271/bbb.61.541. [DOI] [PubMed] [Google Scholar]
  • 30.Choorit W, Kaneko J, Muramoto K, Kamio Y. Existence of a new protein component with the same function as the LukF component of leukocidin or gamma-hemolysin and its gene in Staphylococcus aureus P83. FEBS Lett. 1995;357:260–4. doi: 10.1016/0014-5793(94)01372-8. [DOI] [PubMed] [Google Scholar]
  • 31.von Eiff C, Friedrich AW, Peters G, Becker K. Prevalence of genes encoding for members of the staphylococcal leukotoxin family among clinical isolates of Staphylococcus aureus. Diagn Microbiol Infect Dis. 2004;49:157–62. doi: 10.1016/j.diagmicrobio.2004.03.009. [DOI] [PubMed] [Google Scholar]
  • 32.Prevost G, Couppie P, Prevost P, et al. Epidemiological data on Staphylococcus aureus strains producing synergohymenotropic toxins. J Med Microbiol. 1995;42:237–45. doi: 10.1099/00222615-42-4-237. [DOI] [PubMed] [Google Scholar]
  • 33.Prevost G, Cribier B, Couppie P, et al. Panton-Valentine leucocidin and gamma-hemolysin from Staphylococcus aureus ATCC 49775 are encoded by distinct genetic loci and have different biological activities. Infect Immun. 1995;63:4121–9. doi: 10.1128/iai.63.10.4121-4129.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rahman A, Izaki K, Kamio Y. Gamma-hemolysin genes in the same family with lukF and lukS genes in methicillin resistant Staphylococcus aureus. Biosci Biotechnol Biochem. 1993;57:1234–6. doi: 10.1271/bbb.57.1234. [DOI] [PubMed] [Google Scholar]
  • 35.Gravet A, Colin DA, Keller D, et al. Characterization of a novel structural member, LukE-LukD, of the bi-component staphylococcal leucotoxins family. FEBS Lett. 1998;436:202–8. doi: 10.1016/s0014-5793(98)01130-2. [DOI] [PubMed] [Google Scholar]
  • 36.Alonzo F, 3rd, Benson MA, Chen J, et al. Staphylococcus aureus leukocidin ED contributes to systemic infection by targeting neutrophils and promoting bacterial growth in vivo. Mol Microbiol. 2011 doi: 10.1111/j.1365-2958.2011.07942.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Diep BA, Gill SR, Chang RF, et al. Complete genome sequence of USA300, an epidemic clone of community-acquired meticillin-resistant Staphylococcus aureus. Lancet. 2006;367:731–9. doi: 10.1016/S0140-6736(06)68231-7. [DOI] [PubMed] [Google Scholar]
  • 38•.Otto M. A MRSA-terious enemy among us: end of the PVL controversy? Nat Med. 2011;17:169–70. doi: 10.1038/nm0211-169. A concise and detailed summary of the PVL controversy. [DOI] [PubMed] [Google Scholar]
  • 39.Otto M. Panton-Valentine leukocidin antibodies for the treatment of MRSA skin infections? Expert Rev Anti Infect Ther. 2011;9:389–92. doi: 10.1586/eri.11.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Vandenesch F, Naimi T, Enright MC, et al. Community-acquired methicillin-resistant Staphylococcus aureus carrying Panton-Valentine leukocidin genes: worldwide emergence. Emerg Infect Dis. 2003;9:978–84. doi: 10.3201/eid0908.030089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.del Giudice P, Blanc V, de Rougemont A, et al. Primary skin abscesses are mainly caused by Panton-Valentine leukocidin-positive Staphylococcus aureus strains. Dermatology. 2009;219:299–302. doi: 10.1159/000232391. [DOI] [PubMed] [Google Scholar]
  • 42.Badiou C, Dumitrescu O, Croze M, et al. Panton-Valentine leukocidin is expressed at toxic levels in human skin abscesses. Clin Microbiol Infect. 2008;14:1180–3. doi: 10.1111/j.1469-0691.2008.02105.x. [DOI] [PubMed] [Google Scholar]
  • 43.Labandeira-Rey M, Couzon F, Boisset S, et al. Staphylococcus aureus Panton-Valentine leukocidin causes necrotizing pneumonia. Science. 2007;315:1130–3. doi: 10.1126/science.1137165. [DOI] [PubMed] [Google Scholar]
  • 44.Brown EL, Dumitrescu O, Thomas D, et al. The Panton-Valentine leukocidin vaccine protects mice against lung and skin infections caused by Staphylococcus aureus USA300. Clin Microbiol Infect. 2009;15:156–64. doi: 10.1111/j.1469-0691.2008.02648.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tseng CW, Kyme P, Low J, et al. Staphylococcus aureus Panton-Valentine leukocidin contributes to inflammation and muscle tissue injury. PLoS One. 2009;4:e6387. doi: 10.1371/journal.pone.0006387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Bubeck Wardenburg J, Schneewind O. Vaccine protection against Staphylococcus aureus pneumonia. J Exp Med. 2008;205:287–94. doi: 10.1084/jem.20072208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Villaruz AE, Bubeck Wardenburg J, Khan BA, et al. A point mutation in the agr locus rather than expression of the Panton-Valentine leukocidin caused previously reported phenotypes in Staphylococcus aureus pneumonia and gene regulation. J Infect Dis. 2009;200:724–34. doi: 10.1086/604728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Olsen RJ, Kobayashi SD, Ayeras AA, et al. Lack of a major role of Staphylococcus aureus Panton-Valentine leukocidin in lower respiratory tract infection in nonhuman primates. Am J Pathol. 2010;176:1346–54. doi: 10.2353/ajpath.2010.090960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Montgomery CP, Daum RS. Transcription of inflammatory genes in the lung after infection with community-associated methicillin-resistant Staphylococcus aureus: a role for Panton-Valentine leukocidin? Infect Immun. 2009;77:2159–67. doi: 10.1128/IAI.00021-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Voyich JM, Otto M, Mathema B, et al. Is Panton-Valentine leukocidin the major virulence determinant in community-associated methicillin-resistant Staphylococcus aureus disease? J Infect Dis. 2006;194:1761–70. doi: 10.1086/509506. [DOI] [PubMed] [Google Scholar]
  • 51.Nilsson IM, Hartford O, Foster T, Tarkowski A. Alpha-toxin and gamma-toxin jointly promote Staphylococcus aureus virulence in murine septic arthritis. Infect Immun. 1999;67:1045–9. doi: 10.1128/iai.67.3.1045-1049.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Supersac G, Piemont Y, Kubina M, et al. Assessment of the role of gamma-toxin in experimental endophthalmitis using a hlg-deficient mutant of Staphylococcus aureus. Microb Pathog. 1998;24:241–51. doi: 10.1006/mpat.1997.0192. [DOI] [PubMed] [Google Scholar]
  • 53.Thomas D, Chou S, Dauwalder O, Lina G. Diversity in Staphylococcus aureus enterotoxins. Chem Immunol Allergy. 2007;93:24–41. doi: 10.1159/000100856. [DOI] [PubMed] [Google Scholar]
  • 54••.Wilson GJ, Seo KS, Cartwright RA, et al. A novel core genome-encoded superantigen contributes to lethality of community-associated MRSA necrotizing pneumonia. PLoS Pathog. 2011;7:e1002271. doi: 10.1371/journal.ppat.1002271. The first description of a genome-encoded superantigen, SElX and its contribution to CA-MRSA disease. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Bachert C, Gevaert P, Zhang N, et al. Role of staphylococcal superantigens in airway disease. Chem Immunol Allergy. 2007;93:214–36. doi: 10.1159/000100897. [DOI] [PubMed] [Google Scholar]
  • 56.Dinges MM, Orwin PM, Schlievert PM. Exotoxins of Staphylococcus aureus. Clin Microbiol Rev. 2000;13:16–34. doi: 10.1128/cmr.13.1.16-34.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Dellabona P, Peccoud J, Kappler J, et al. Superantigens interact with MHC class II molecules outside of the antigen groove. Cell. 1990;62:1115–21. doi: 10.1016/0092-8674(90)90388-u. [DOI] [PubMed] [Google Scholar]
  • 58.Choi YW, Kotzin B, Herron L, et al. Interaction of Staphylococcus aureus toxin “superantigens” with human T cells. Proc Natl Acad Sci U S A. 1989;86:8941–5. doi: 10.1073/pnas.86.22.8941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fraser JD, Proft T. The bacterial superantigen and superantigen-like proteins. Immunol Rev. 2008;225:226–43. doi: 10.1111/j.1600-065X.2008.00681.x. [DOI] [PubMed] [Google Scholar]
  • 60.Langford MP, Stanton GJ, Johnson HM. Biological effects of staphylococcal enterotoxin A on human peripheral lymphocytes. Infect Immun. 1978;22:62–8. doi: 10.1128/iai.22.1.62-68.1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Drake CG, Kotzin BL. Superantigens: biology, immunology, and potential role in disease. J Clin Immunol. 1992;12:149–62. doi: 10.1007/BF00918083. [DOI] [PubMed] [Google Scholar]
  • 62.Ono HK, Omoe K, Imanishi K, et al. Identification and characterization of two novel staphylococcal enterotoxins, types S and T. Infect Immun. 2008;76:4999–5005. doi: 10.1128/IAI.00045-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Fitzgerald JR, Monday SR, Foster TJ, et al. Characterization of a putative pathogenicity island from bovine Staphylococcus aureus encoding multiple superantigens. J Bacteriol. 2001;183:63–70. doi: 10.1128/JB.183.1.63-70.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Johns MB, Jr, Khan SA. Staphylococcal enterotoxin B gene is associated with a discrete genetic element. J Bacteriol. 1988;170:4033–9. doi: 10.1128/jb.170.9.4033-4039.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Schmitz FJ, MacKenzie CR, Geisel R, et al. Enterotoxin and toxic shock syndrome toxin-1 production of methicillin resistant and methicillin sensitive Staphylococcus aureus strains. Eur J Epidemiol. 1997;13:699–708. doi: 10.1023/a:1007357206672. [DOI] [PubMed] [Google Scholar]
  • 66.Baba T, Takeuchi F, Kuroda M, et al. Genome and virulence determinants of high virulence community-acquired MRSA. Lancet. 2002;359:1819–27. doi: 10.1016/s0140-6736(02)08713-5. [DOI] [PubMed] [Google Scholar]
  • 67.Czop JK, Bergdoll MS. Staphylococcal enterotoxin synthesis during the exponential, transitional, and stationary growth phases. Infect Immun. 1974;9:229–35. doi: 10.1128/iai.9.2.229-235.1974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gaskill ME, Khan SA. Regulation of the enterotoxin B gene in Staphylococcus aureus. J Biol Chem. 1988;263:6276–80. [PubMed] [Google Scholar]
  • 69.Regassa LB, Couch JL, Betley MJ. Steady-state staphylococcal enterotoxin type C mRNA is affected by a product of the accessory gene regulator (agr) and by glucose. Infect Immun. 1991;59:955–62. doi: 10.1128/iai.59.3.955-962.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Munson SH, Tremaine MT, Betley MJ, Welch RA. Identification and characterization of staphylococcal enterotoxin types G and I from Staphylococcus aureus. Infect Immun. 1998;66:3337–48. doi: 10.1128/iai.66.7.3337-3348.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tremaine MT, Brockman DK, Betley MJ. Staphylococcal enterotoxin A gene (sea) expression is not affected by the accessory gene regulator (agr) Infect Immun. 1993;61:356–9. doi: 10.1128/iai.61.1.356-359.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Schlievert PM, Blomster DA. Production of staphylococcal pyrogenic exotoxin type C: influence of physical and chemical factors. J Infect Dis. 1983;147:236–42. doi: 10.1093/infdis/147.2.236. [DOI] [PubMed] [Google Scholar]
  • 73.Tseng CW, Zhang S, Stewart GC. Accessory gene regulator control of staphyloccoccal enterotoxin d gene expression. J Bacteriol. 2004;186:1793–801. doi: 10.1128/JB.186.6.1793-1801.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Andrey DO, Renzoni A, Monod A, et al. Control of the Staphylococcus aureus toxic shock tst promoter by the global regulator SarA. J Bacteriol. 2010;192:6077–85. doi: 10.1128/JB.00146-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kusch K, Hanke K, Holtfreter S, et al. The influence of SaeRS and sigma(B) on the expression of superantigens in different Staphylococcus aureus isolates. Int J Med Microbiol. 2011;301:488–99. doi: 10.1016/j.ijmm.2011.01.003. [DOI] [PubMed] [Google Scholar]
  • 76.Vojtov N, Ross HF, Novick RP. Global repression of exotoxin synthesis by staphylococcal superantigens. Proc Natl Acad Sci U S A. 2002;99:10102–7. doi: 10.1073/pnas.152152499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Neumann B, Engelhardt B, Wagner H, Holzmann B. Induction of acute inflammatory lung injury by staphylococcal enterotoxin B. J Immunol. 1997;158:1862–71. [PubMed] [Google Scholar]
  • 78.Strandberg KL, Rotschafer JH, Vetter SM, et al. Staphylococcal superantigens cause lethal pulmonary disease in rabbits. J Infect Dis. 2010;202:1690–7. doi: 10.1086/657156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.De Boer ML, Kum WW, Chow AW. Staphylococcus aureus isogenic mutant, deficient in toxic shock syndrome toxin-1 but not staphylococcal enterotoxin A production, exhibits attenuated virulence in a tampon-associated vaginal infection model of toxic shock syndrome. Can J Microbiol. 1999;45:250–6. [PubMed] [Google Scholar]
  • 80.Abdelnour A, Bremell T, Tarkowski A. Toxic shock syndrome toxin 1 contributes to the arthritogenicity of Staphylococcus aureus. J Infect Dis. 1994;170:94–9. doi: 10.1093/infdis/170.1.94. [DOI] [PubMed] [Google Scholar]
  • 81.de Azavedo JC, Foster TJ, Hartigan PJ, et al. Expression of the cloned toxic shock syndrome toxin 1 gene (tst) in vivo with a rabbit uterine model. Infect Immun. 1985;50:304–9. doi: 10.1128/iai.50.1.304-309.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zhang S, Stewart GC. Characterization of the promoter elements for the staphylococcal enterotoxin D gene. J Bacteriol. 2000;182:2321–5. doi: 10.1128/jb.182.8.2321-2325.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Xiong YQ, Willard J, Yeaman MR, et al. Regulation of Staphylococcus aureus alpha-toxin gene (hla) expression by agr, sarA, and sae in vitro and in experimental infective endocarditis. J Infect Dis. 2006;194:1267–75. doi: 10.1086/508210. [DOI] [PubMed] [Google Scholar]
  • 84.Liang X, Ji Y. Alpha-toxin interferes with integrin-mediated adhesion and internalization of Staphylococcus aureus by epithelial cells. Cell Microbiol. 2006;8:1656–68. doi: 10.1111/j.1462-5822.2006.00740.x. [DOI] [PubMed] [Google Scholar]
  • 85.Walev I, Martin E, Jonas D, et al. Staphylococcal alpha-toxin kills human keratinocytes by permeabilizing the plasma membrane for monovalent ions. Infect Immun. 1993;61:4972–9. doi: 10.1128/iai.61.12.4972-4979.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Thelestam M, Mollby R, Wadstrom T. Effects of staphylococcal alpha-, beta-, delta-, and gamma-hemolysins on human diploid fibroblasts and HeLa cells: evaluation of a new quantitative as say for measuring cell damage. Infect Immun. 1973;8:938–46. doi: 10.1128/iai.8.6.938-946.1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ezepchuk YV, Leung DY, Middleton MH, et al. Staphylococcal toxins and protein A differentially induce cytotoxicity and release of tumor necrosis factor-alpha from human keratinocytes. J Invest Dermatol. 1996;107:603–9. doi: 10.1111/1523-1747.ep12583377. [DOI] [PubMed] [Google Scholar]
  • 88.Valeva A, Walev I, Pinkernell M, et al. Transmembrane beta-barrel of staphylococcal alpha-toxin forms in sensitive but not in resistant cells. Proc Natl Acad Sci U S A. 1997;94:11607–11. doi: 10.1073/pnas.94.21.11607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hocke AC, Temmesfeld-Wollbrueck B, Schmeck B, et al. Perturbation of endothelial junction proteins by Staphylococcus aureus alpha-toxin: inhibition of endothelial gap formation by adrenomedullin. Histochem Cell Biol. 2006;126:305–16. doi: 10.1007/s00418-006-0174-5. [DOI] [PubMed] [Google Scholar]
  • 90.Bartlett AH, Foster TJ, Hayashida A, Park PW. Alpha-toxin facilitates the generation of CXC chemokine gradients and stimulates neutrophil homing in Staphylococcus aureus pneumonia. J Infect Dis. 2008;198:1529–35. doi: 10.1086/592758. [DOI] [PubMed] [Google Scholar]
  • 91.Inoshima I, Inoshima N, Wilke GA, et al. A Staphylococcus aureus pore-forming toxin subverts the activity of ADAM10 to cause lethal infection in mice. Nat Med. 2011;17:1310–4. doi: 10.1038/nm.2451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92•.Wilke GA, Bubeck Wardenburg J. Role of a disintegrin and metalloprotease 10 in Staphylococcus aureus alpha-hemolysin-mediated cellular injury. Proc Natl Acad Sci U S A. 2010;107:13473–8. doi: 10.1073/pnas.1001815107. A intricate study that demonstrates that ADAM10 is the receptor for α-hemolysin. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Bubeck Wardenburg J, Patel RJ, Schneewind O. Surface proteins and exotoxins are required for the pathogenesis of Staphylococcus aureus pneumonia. Infect Immun. 2007;75:1040–4. doi: 10.1128/IAI.01313-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Bubeck Wardenburg J, Bae T, Otto M, et al. Poring over pores: alpha-hemolysin and Panton-Valentine leukocidin in Staphylococcus aureus pneumonia. Nat Med. 2007;13:1405–6. doi: 10.1038/nm1207-1405. [DOI] [PubMed] [Google Scholar]
  • 95.Kebaier C, Chamberland RR, Allen IC, et al. Staphylococcus aureus alpha-hemolysin mediates virulence in a murine model of severe pneumonia through activation of the NLRP3 inflammasome. J Infect Dis. 2012 doi: 10.1093/infdis/jir846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Davis BK, Wen H, Ting JP. The inflammasome NLRs in immunity, inflammation, and associated diseases. Annu Rev Immunol. 2011;29:707–35. doi: 10.1146/annurev-immunol-031210-101405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Mariathasan S, Weiss DS, Newton K, et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440:228–32. doi: 10.1038/nature04515. [DOI] [PubMed] [Google Scholar]
  • 98.Munoz-Planillo R, Franchi L, Miller LS, Nunez G. A critical role for hemolysins and bacterial lipoproteins in Staphylococcus aureus-induced activation of the NLRP3 inflammasome. J Immunol. 2009;183:3942–8. doi: 10.4049/jimmunol.0900729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kretschmer D, Gleske AK, Rautenberg M, et al. Human formyl peptide receptor 2 senses highly pathogenic Staphylococcus aureus. Cell Host Microbe. 2010;7:463–73. doi: 10.1016/j.chom.2010.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Queck SY, Jameson-Lee M, Villaruz AE, et al. RNAIII-independent target gene control by the agr quorum-sensing system: insight into the evolution of virulence regulation in Staphylococcus aureus. Mol Cell. 2008;32:150–8. doi: 10.1016/j.molcel.2008.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Chatterjee SS, Chen L, Joo HS, et al. Distribution and regulation of the mobile genetic element-encoded phenol-soluble modulin psm-mec in methicillin-resistant Staphylococcus aureus. PLoS One. 2011;6:e28781. doi: 10.1371/journal.pone.0028781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Janzon L, Lofdahl S, Arvidson S. Identification and nucleotide sequence of the delta-lysin gene, hld, adjacent to the accessory gene regulator (agr) of Staphylococcus aureus. Mol Gen Genet. 1989;219:480–5. doi: 10.1007/BF00259623. [DOI] [PubMed] [Google Scholar]
  • 103.Queck SY, Khan BA, Wang R, et al. Mobile genetic element-encoded cytolysin connects virulence to methicillin resistance in MRSA. PLoS Pathog. 2009;5:e1000533. doi: 10.1371/journal.ppat.1000533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Periasamy S, Joo HS, Duong AC, et al. How Staphylococcus aureus biofilms develop their characteristic structure. Proc Natl Acad Sci U S A. 2012 doi: 10.1073/pnas.1115006109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common cause of persistent infections. Science. 1999;284:1318–22. doi: 10.1126/science.284.5418.1318. [DOI] [PubMed] [Google Scholar]
  • 106.Mellor IR, Thomas DH, Sansom MS. Properties of ion channels formed by Staphylococcus aureus delta-toxin. Biochim Biophys Acta. 1988;942:280–94. doi: 10.1016/0005-2736(88)90030-2. [DOI] [PubMed] [Google Scholar]
  • 107.Wang R, Khan BA, Cheung GY, et al. Staphylococcus epidermidis surfactant peptides promote biofilm maturation and dissemination of biofilm-associated infection in mice. J Clin Invest. 2011;121:238–48. doi: 10.1172/JCI42520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Cheung GY, Otto M. Direct and synergistic hemolysis caused by Staphylococcus phenol-soluble modulins: Implications for diagnosis and pathogenesis. Microbes Infect. 2011 doi: 10.1016/j.micinf.2011.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Giese B, Glowinski F, Paprotka K, et al. Expression of delta-toxin by Staphylococcus aureus mediates escape from phago-endosomes of human epithelial and endothelial cells in the presence of beta-toxin. Cell Microbiol. 2011;13:316–29. doi: 10.1111/j.1462-5822.2010.01538.x. [DOI] [PubMed] [Google Scholar]
  • 110.Hongo I, Baba T, Oishi K, et al. Phenol-soluble modulin alpha 3 enhances the human neutrophil lysis mediated by Panton-Valentine leukocidin. J Infect Dis. 2009;200:715–23. doi: 10.1086/605332. [DOI] [PubMed] [Google Scholar]
  • 111.Coleman DC, Arbuthnott JP, Pomeroy HM, Birkbeck TH. Cloning and expression in Escherichia coli and Staphylococcus aureus of the beta-lysin determinant from Staphylococcus aureus: evidence that bacteriophage conversion of beta-lysin activity is caused by insertional inactivation of the beta-lysin determinant. Microb Pathog. 1986;1:549–64. doi: 10.1016/0882-4010(86)90040-9. [DOI] [PubMed] [Google Scholar]
  • 112.Peacock SJ, Justice A, Griffiths D, et al. Determinants of acquisition and carriage of Staphylococcus aureus in infancy. J Clin Microbiol. 2003;41:5718–25. doi: 10.1128/JCM.41.12.5718-5725.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lebon A, Labout JA, Verbrugh HA, et al. Dynamics and determinants of Staphylococcus aureus carriage in infancy: the Generation R Study. J Clin Microbiol. 2008;46:3517–21. doi: 10.1128/JCM.00641-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Wertheim HF, Melles DC, Vos MC, et al. The role of nasal carriage in Staphylococcus aureus infections. 2005;5:751–62. doi: 10.1016/S1473-3099(05)70295-4. [DOI] [PubMed] [Google Scholar]
  • 115.Croze M, Dauwalder O, Dumitrescu O, et al. Serum antibodies against Panton-Valentine leukocidin in a normal population and during Staphylococcus aureus infection. Clin Microbiol Infect. 2009;15:144–8. doi: 10.1111/j.1469-0691.2008.02650.x. [DOI] [PubMed] [Google Scholar]
  • 116.Colque-Navarro P, Jacobsson G, Andersson R, et al. Levels of antibody against 11 Staphylococcus aureus antigens in a healthy population. Clin Vaccine Immunol. 2010;17:1117–23. doi: 10.1128/CVI.00506-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Verkaik NJ, de Vogel CP, Boelens HA, et al. Anti-staphylococcal humoral immune response in persistent nasal carriers and noncarriers of Staphylococcus aureus. J Infect Dis. 2009;199:625–32. doi: 10.1086/596743. [DOI] [PubMed] [Google Scholar]
  • 118.Holtfreter S, Kolata J, Broker BM. Towards the immune proteome of Staphylococcus aureus - The anti-S. aureus antibody response. Int J Med Microbiol. 2010;300:176–92. doi: 10.1016/j.ijmm.2009.10.002. [DOI] [PubMed] [Google Scholar]
  • 119.Verkaik NJ, Dauwalder O, Antri K, et al. Immunogenicity of toxins during Staphylococcus aureus infection. Clin Infect Dis. 2010;50:61–8. doi: 10.1086/648673. [DOI] [PubMed] [Google Scholar]
  • 120.Jacobsson G, Colque-Navarro P, Gustafsson E, et al. Antibody responses in patients with invasive Staphylococcus aureus infections. Eur J Clin Microbiol Infect Dis. 2010;29:715–25. doi: 10.1007/s10096-010-0919-x. [DOI] [PubMed] [Google Scholar]
  • 121.Hermos CR, Yoong P, Pier GB. High levels of antibody to panton-valentine leukocidin are not associated with resistance to Staphylococcus aureus-associated skin and soft-tissue infection. Clin Infect Dis. 2010;51:1138–46. doi: 10.1086/656742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Holtfreter S, Roschack K, Eichler P, et al. Staphylococcus aureus carriers neutralize superantigens by antibodies specific for their colonizing strain: a potential explanation for their improved prognosis in severe sepsis. J Infect Dis. 2006;193:1275–8. doi: 10.1086/503048. [DOI] [PubMed] [Google Scholar]
  • 123.Grumann D, Ruotsalainen E, Kolata J, et al. Characterization of infecting strains and superantigen-neutralizing antibodies in Staphylococcus aureus bacteremia. Clin Vaccine Immunol. 2011;18:487–93. doi: 10.1128/CVI.00329-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Laventie BJ, Rademaker HJ, Saleh M, et al. Heavy chain-only antibodies and tetravalent bispecific antibody neutralizing Staphylococcus aureus leukotoxins. Proc Natl Acad Sci U S A. 2011;108:16404–9. doi: 10.1073/pnas.1102265108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Gauduchon V, Cozon G, Vandenesch F, et al. Neutralization of Staphylococcus aureus Panton-Valentine leukocidin by intravenous immunoglobulin in vitro. J Infect Dis. 2004;189:346–53. doi: 10.1086/380909. [DOI] [PubMed] [Google Scholar]
  • 126.Loffler B, Hussain M, Grundmeier M, et al. Staphylococcus aureus Panton-Valentine leukocidin is a very potent cytotoxic factor for human neutrophils. PLoS Pathog. 2010;6:e1000715. doi: 10.1371/journal.ppat.1000715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127•.Yoong P, Pier GB. Antibody-mediated enhancement of community-acquired methicillin-resistant Staphylococcus aureus infection. Proc Natl Acad Sci U S A. 2010;107:2241–6. doi: 10.1073/pnas.0910344107. This study discusses the negative effects of early administration of anti-PVL antibodies during infection by PVL+ CA-MRSA strain. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Cribier B, Prevost G, Couppie P, et al. Staphylococcus aureus leukocidin: a new virulence factor in cutaneous infections? An epidemiological and experimental study. Dermatology. 1992;185:175–80. doi: 10.1159/000247443. [DOI] [PubMed] [Google Scholar]
  • 129.ClinicalTrials.gov; A service of the U.S. National Institutes of Health. Staphylococcus aureus Toxoids Phase. Vaccine Trial. :1–2. Available at: http://clinicaltrials.gov/ct2/show/NCT01011335.
  • 130.Takei S, Arora YK, Walker SM. Intravenous immunoglobulin contains specific antibodies inhibitory to activation of T cells by staphylococcal toxin superantigens. J Clin Invest. 1993;91:602–7. doi: 10.1172/JCI116240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Holtfreter S, Bauer K, Thomas D, et al. egc-Encoded superantigens from Staphylococcus aureus are neutralized by human sera much less efficiently than are classical staphylococcal enterotoxins or toxic shock syndrome toxin. Infect Immun. 2004;72:4061–71. doi: 10.1128/IAI.72.7.4061-4071.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Burian M, Grumann D, Holtfreter S, et al. Expression of staphylococcal superantigens during nasal colonization is not sufficient to induce a systemic neutralizing antibody response in humans. Eur J Clin Microbiol Infect Dis. 2011 doi: 10.1007/s10096-011-1302-2. [DOI] [PubMed] [Google Scholar]
  • 133.Rusnak JM, Kortepeter M, Ulrich R, et al. Laboratory exposures to staphylococcal enterotoxin B. Emerg Infect Dis. 2004;10:1544–9. doi: 10.3201/eid1009.040250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Gill DM. Bacterial toxins: a table of lethal amounts. Microbiol Rev. 1982;46:86–94. doi: 10.1128/mr.46.1.86-94.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Tilahun ME, Rajagopalan G, Shah-Mahoney N, et al. Potent neutralization of staphylococcal enterotoxin B by synergistic action of chimeric antibodies. Infect Immun. 2010;78:2801–11. doi: 10.1128/IAI.01121-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136•.Varshney AK, Wang X, Cook E, et al. Generation, characterization, and epitope mapping of neutralizing and protective monoclonal antibodies against staphylococcal enterotoxin B-induced lethal shock. J Biol Chem. 2011;286:9737–47. doi: 10.1074/jbc.M110.212407. A study that shows co-administration of multiple antibodies to SEB can lead to better protection in mice against SEB-toxin challenge. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.LeClaire RD, Hunt RE, Bavari S. Protection against bacterial superantigen staphylococcal enterotoxin B by passive vaccination. Infect Immun. 2002;70:2278–81. doi: 10.1128/IAI.70.5.2278-2281.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kum WW, Chow AW. Inhibition of staphylococcal enterotoxin A-induced superantigenic and lethal activities by a monoclonal antibody to toxic shock syndrome toxin-1. J Infect Dis. 2001;183:1739–48. doi: 10.1086/320732. [DOI] [PubMed] [Google Scholar]
  • 139.Boles JW, Pitt ML, LeClaire RD, et al. Generation of protective immunity by inactivated recombinant staphylococcal enterotoxin B vaccine in nonhuman primates and identification of correlates of immunity. Clin Immunol. 2003;108:51–9. doi: 10.1016/s1521-6616(03)00066-4. [DOI] [PubMed] [Google Scholar]
  • 140.Hu DL, Omoe K, Narita K, et al. Intranasal vaccination with a double mutant of staphylococcal enterotoxin C provides protection against Staphylococcus aureus infection. Microbes Infect. 2006;8:2841–8. doi: 10.1016/j.micinf.2006.09.001. [DOI] [PubMed] [Google Scholar]
  • 141.Hu DL, Omoe K, Sasaki S, et al. Vaccination with nontoxic mutant toxic shock syndrome toxin 1 protects against Staphylococcus aureus infection. J Infect Dis. 2003;188:743–52. doi: 10.1086/377308. [DOI] [PubMed] [Google Scholar]
  • 142.Buonpane RA, Churchill HR, Moza B, et al. Neutralization of staphylococcal enterotoxin B by soluble, high-affinity receptor antagonists. Nat Med. 2007;13:725–9. doi: 10.1038/nm1584. [DOI] [PubMed] [Google Scholar]
  • 143.Hong-Geller E, Mollhoff M, Shiflett PR, Gupta G. Design of chimeric receptor mimics with different TcRVbeta isoforms. Type-specific inhibition of superantigen pathogenesis. J Biol Chem. 2004;279:5676–84. doi: 10.1074/jbc.M309388200. [DOI] [PubMed] [Google Scholar]
  • 144.Tkaczyk C, Hua L, Varkey R, et al. Identification of anti-alpha toxin mAbs that reduce severity of Staphylococcus aureus dermonecrosis and exhibit a correlation between affinity and potency. Clin Vaccine Immunol. 2012;19:377–85. doi: 10.1128/CVI.05589-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Kennedy AD, Bubeck Wardenburg J, Gardner DJ, et al. Targeting of alpha-hemolysin by active or passive immunization decreases severity of USA300 skin infection in a mouse model. J Infect Dis. 2010;202:1050–8. doi: 10.1086/656043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Ragle BE, Bubeck Wardenburg J. Anti-alpha-hemolysin monoclonal antibodies mediate protection against Staphylococcus aureus pneumonia. Infect Immun. 2009;77:2712–8. doi: 10.1128/IAI.00115-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Brown EL, Bowden MG, Bryson RS, et al. Pediatric antibody response to community-acquired Staphylococcus aureus infection is directed to Panton-Valentine leukocidin. Clin Vaccine Immunol. 2009;16:139–41. doi: 10.1128/CVI.00360-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Nolte FS, Kapral FA. Immunogenicity of Staphylococcus aureus delta-toxin. Infect Immun. 1981;31:1251–60. doi: 10.1128/iai.31.3.1251-1260.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149•.Otto M. Novel targeted immunotherapy approaches for staphylococcal infection. Expert Opin Biol Ther. 2010;10:1049–59. doi: 10.1517/14712598.2010.495115. An overview of current therapeutic strategies against S. aureus infection. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Leysath CE, Chen KH, Moayeri M, et al. Mouse monoclonal antibodies to anthrax edema factor protect against infection. Infect Immun. 2011;79:4609–16. doi: 10.1128/IAI.05314-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Cheng LW, Stanker LH, Henderson TD, 2nd, et al. Antibody protection against botulinum neurotoxin intoxication in mice. Infect Immun. 2009;77:4305–13. doi: 10.1128/IAI.00405-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Babcock GJ, Broering TJ, Hernandez HJ, et al. Human monoclonal antibodies directed against toxins A and B prevent Clostridium difficile-induced mortality in hamsters. Infect Immun. 2006;74:6339–47. doi: 10.1128/IAI.00982-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Deresinski S, Herrera V. Immunotherapies for Staphylococcus aureus: current challenges and future prospects. Infect Control Hosp Epidemiol. 2010;31 (Suppl 1):S45–7. doi: 10.1086/655992. [DOI] [PubMed] [Google Scholar]

RESOURCES