Abstract
Purpose of review
Staphylococcus aureus is the most common invasive bacterial pathogen infecting children in the U.S. and many parts of the world. This major human pathogen continues to evolve, and recognition of recent trends in epidemiology, therapeutics and future horizons is of high importance.
Recent findings
Over the past decade, a relative rise of methicillin-susceptible S. aureus (MSSA) has occurred, such that methicillin-resistant S. aureus (MRSA) no longer dominates the landscape of invasive disease. Antimicrobial resistance continues to develop, however, and novel therapeutics or preventive modalities are urgently needed. Unfortunately, several recent vaccine attempts proved unsuccessful in humans.
Summary
Recent scientific breakthroughs highlight the opportunity for novel interventions against S. aureus by interfering with virulence rather than by traditional antimicrobial mechanisms. A S. aureus vaccine remains elusive; the reasons for this are multifactorial, and lessons learned from prior unsuccessful attempts may create a path toward an effective preventive. Finally, new diagnostic modalities have the potential to greatly enhance clinical care for invasive S. aureus disease in children.
Keywords: antimicrobial resistance, novel therapeutics, Staphylococcus aureus, Staphylococcus aureus vaccine
INTRODUCTION
Thanks in part to successful vaccine development against Streptococcus pneumoniae and Haemophilus influenzae, Staphylococcus aureus is now the leading invasive bacterial pathogen in children in the U.S. and many parts of the world [1■■,2,3]. In fact, current rates of invasive S. aureus disease in children (e.g. bloodstream infection, bone and joint infection) exceed peak rates of pneumococcal and H. influenzae disease at the time that vaccines were developed against those major pathogens [4]. S. aureus may be unrivalled as a pathogen both in terms of the number of failed vaccine attempts against the organism, as well as its protean clinical phenotypes and ability to invade nearly every tissue in the human body. The pathogen also continues to evolve and modulate its virulence, as it progressively gains antibiotic resistance mechanisms among circulating strains causing clinical disease. In this report, we will discuss current opinions and recent data regarding the epidemiology, treatment, prevention and future horizons of invasive S. aureus disease in children.
CHANGING EPIDEMIOLOGY OF PAEDIATRIC INVASIVE STAPHYLOCOCCUS AUREUS STRAINS
On a broad scale, the evolution of clinically relevant, actively circulating strains can be marked by a sequence of ‘eras’. For decades, methicillin-resistant S. aureus (MRSA) was almost exclusively a nosocomial pathogen, while methicillin-susceptible (MSSA) strains caused occasional disease in other-wise healthy hosts in the community [5]. This fundamentally shifted in the mid-1990s with the rise of the epidemic S. aureus clone, USA300, which drove the community-associated MRSA (CA-MRSA) epidemic in the early part of the twenty-first century. This era, while profoundly impactful both at the level of patient care and by its impact on new drug and attempted vaccine development, now appears to be shifting again.
Beginning around 2005, invasive MRSA infection rates began to gradually decline, and over the past decade, a relative rise in virulent MSSA infections has begun to define a new era of invasive S. aureus epidemiology in children [6]. USA300 CA-MRSA strains are no longer the predominant cause of invasive infections in children in the U.S. Rather, a more diverse group of currently circulating S. aureus strains populate the landscape, and the majority of infections now appear to be MSSA, with rates of paediatric bloodstream infection due to MSSA progressively rising since nearly 2009 [3,7■■]. This does not imply, however, that invasive infections due to methicillin-sensitive S. aureus are less severe. Although a variety of studies largely suggested that the USA300 CA-MRSA strain may have led to more severe disease phenotypes than contemporary MSSA comparators, this distinction no longer appears to hold. For example, a large series out of the University of Utah found that from 2009 to 2012, MSSA strains represented nearly 80% of invasive paediatric S. aureus infections, with six predominant strain types and no discernible severity differences comparing MRSA to MSSA [7■■]. This evolution of S. aureus, with a current predominance of MSSA strains, calls into question many current infection control practices, which focus exclusively on prevention of MRSA while ignoring the importance of invasive MSSA [8]. Typical hospital infection control policies mandate contact isolation when MRSA is present, though recent data would suggest an equal importance at preventing the spread of MSSA disease both in and out of the hospital setting [7■■,9■].
Critical future work in this area will involve the identification of virulence factors that are conserved across invasive/lethal strains of S. aureus, regardless of MRSA vs. MSSA resistance patterns. For example, the Panton Valentine leucocidin (PVL), thought to be a mediator of the clinical severity and invasion potential associated with USA300 CA-MRSA, is present in only nearly 10% of currently invasive S. aureus strains [7■■]. Identification of those factors that are necessary and sufficient for invasion across distantly related lineages of clinically relevant S. aureus will be central to identifying novel targets of intervention against this major paediatric pathogen.
ADVANCES IN TRADITIONAL ANTISTAPHYLOCOCCAL THERAPEUTICS
The dominance of CA-MRSA, along with multiply-drug-resistant strains of S. pneumoniae and Enterococcus species, as drivers of invasive paediatric infections throughout the early part of this century led to a dramatic response by the drug development industry to identify agents with activity against resistant Gram-positive pathogens, particularly MRSA. This has resulted in a relative surge of recently developed antimicrobials that maintain activity against MRSA and other resistant gram-positive pathogens (Table 1). Daptomycin, initially FDA approved for use in adults in 2003, is a bactericidal agent that provides an alternative to vancomycin for MRSA endocarditis and other invasive (nonpneumonic) infections. Daptomycin is now recommended by the Infectious Diseases Society of America (IDSA) expert guidelines as an equivalent option to vancomycin for the treatment of MRSA bacteraemia in adults [10], but similar recommendations from national guidelines are lagging in paediatric infections.
Table 1.
Recently developed antimicrobials with activity against methicillin-resistant Staphylococcus aureus
| Drug name | Initial US approval | Class/Mechanism of action | FDA indications | Paediatric labelling | Comments |
|---|---|---|---|---|---|
| Daptomycin | 2003 | Cyclic lipopeptide; rapid depolarization of cell membrane potential. | cSSSI; SAB including right-sided endocarditis | Dosing recommendations for ≥1 year of age. Not recommended <1 year of ago due to neuromuscular adverse effects in neonatal dogs. | Not indicated for pneumonia due to inactivation by surfactant. |
| Telavancin | 2009 | Lipoglycopeptide; inhibits cross-linking of cell wall peptidoglycan | cSSSI (daily infusion; half-life ~8h) | Safety and effectiveness in paediatric patients have not been studied. | Open-label PK study in patients aged 3 months–17 years is underway (NCT02013141) |
| Ceftaroline | 2010 | Cephalosporin; disruption of cell wall synthesis via binding to PBP (including PBP2a of MRSA) | ABSSSI and CABP | Safety and effectiveness in paediatric patients have not been established. | Significant PK and safety data are published for children down to 2 months of age (13) |
| Dalbavancin | 2014 | Lipoglycopeptide; inhibits cross-linking of cell wall peptidoglycan | ABSSSI (One or two-dose regimens; half-life ~8 days) | Safety and effectiveness in paediatric patients have not been established. | PK and safety appeared similar in adolescents in a small trial (15) |
| Oritavancin | 2014 | Lipoglycopeptide; inhibits cross-linking of cell wall peptidoglycan | ABSSSI (Single-dose therapy; half-life ~10 days) | Safety and effectiveness in paediatric patients have not been studied. | Limited PK data available for children aged 2–18 years (16) |
| Tedizolid | 2014 | Oxazolidinone; Inhibition of protein synthesis at the 50S ribosomal subunit. | ABSSSI | Safety and effectiveness in paediatric patients have not been established. | PK and safety appeared similar in adolescents in a small trial (17) |
| Delafloxacin | 2017 | Fluoroquinolone; inhibition of DNA gyrase and topoisomerase IV. | ABSSSI in adult patients | Use in patients <18 years of age is currently not recommended. | Box warning for tendon rupture and nervous system effects |
| Omadacycline | 2018 | Tetracycline; Inhibition of protein synthesis at the 30S ribosomal subunit | ABSSSI and CABP | Due to class effects on tooth development and bone growth, use in patients less <8 years of age is not recommended | Active-controlled safety study in children between 8 and 17 years old with CABP will be conducted |
ABSSSI, acute bacterial skin and skin structure infection; CABP, community-acquired bacterial pneumonia; cSSSI, complicated skin and skin structure infection; PBP, penicillin-binding protein; PK, pharmacokinetics.
In 2010, the drug ceftaroline was licensed for treatment of CAP and complicated SSTIs. Designated a fifth-generation cephalosporin, ceftaroline is highly notable as the first beta-lactam antimicrobial with activity against MRSA strains. There is increasing evidence of the utility of ceftaroline for the treatment of severe MRSA SSTI, bacteraemia, endocarditis and orthopaedic infections in adults [11,12]. Recent years have seen the accumulation of substantial experience in the paediatric use of ceftaroline, with significant pharmacokinetic and safety data down to infancy [13].
Newer glycopeptide antimicrobials such as dalbavancin, telavancin and oritavancin are also promising agents based on their much longer half-lives compared with vancomycin, potentially allowing for infusion-based weekly dosing rather than daily home antibiotic dosing via peripherally inserted central catheter (PICC) [14]. Like daptomycin, these agents are now commonly used to treat MRSA infections in adults, but widespread use in paediatrics is lagging significantly behind, in part related to a paucity of pharmacokinetic data, information which is of high importance given the prolonged half-life of these agents. Encouragingly, dalbavancin and oritavancin pharmacokinetics and safety in adolescents appear similar to adults [15,16], though data in younger children are unavailable at present. Finally, tedizolid may emerge as a better-tolerated analogue of linezolid in children, though again, paediatric data are limited beyond some encouraging pharmacokinetic and safety data in adolescents [17].
NOVEL APPROACHES TO ANTISTAPHYLOCOCCAL THERAPEUTICS
The widespread emergence of community-acquired MRSA in the 1990s and 2000s raised considerable concerns that S. aureus might continue to evolve resistance against all currently available antimicrobials. Fortunately, widespread resistance to important antibiotics for invasive staphylococcal infection (e.g. vancomycin and daptomycin) has yet to occur. Accordingly, unlike contemporary MDR Gram-negative pathogens, clinicians still typically have multiple antibiotic options for treating staphylococcal disease. However, treatment failure of invasive S. aureus infections commonly occurs [18■,19], even when the infection isolate has a susceptible MIC to the drug of choice. This phenomenon, which can be broadly referred to as antibiotic tolerance, reflects factors intrinsic to the pathogenesis of staphylococcal infection, as well as the limitations of the current clinical standards for antibiotic susceptibility testing [20].
The hallmark inflammatory lesion of invasive S. aureus infection is the abscess. Abscesses consist of a central bacterial microcolony, or ‘staphylococcal abscess community’, surrounded by viable and necrotic neutrophils. The neutrophilic infiltrate is further surrounded by a thin rim of macrophages, as well as a fibrous abscess capsule that eventually sequesters the lesion from surrounding healthy tissues [21]. Importantly, the staphylococcal abscess community is encased with a coagulase-dependent shell that impedes neutrophil access to the bacteria [22–24]. The staphylococcal coagulases leverage factors of the mammalian clotting system to build this barrier, and FDA-approved drugs that inhibit clotting have shown promise in preclinical models of staphylococcal infection [25]. It is likely that the abscess represents a critical barrier to the free diffusion of antibiotics, although experimental evidence is lacking due to the inherent difficulty in studying spatially heterogenous host-pathogen interactions in infected tissues. Outside of abscess formation, S. aureus exploits a number of host niches to escape immune effectors and antibiotic therapy. For example, staphylococcal invasion of resident macrophages in the liver is a critical determinant of antibiotic failure and subsequent disseminated disease in murine models [26,27]. Surewaard et al. [27] showed that this protective niche could be targeted by creating a liposomal formulation of vancomycin, which more efficiently eliminated intracellular bacteria. In the setting of osteomyelitis, which is one of the most common manifestations of invasive S. aureus infection, staphylococci exist in multiple niches that likely impact antibiotic efficacy. This includes bone marrow abscesses, invasion of necrotic fragments of bone known as sequestra, colonization of the osteocyte-canalicular network in the substructure of bone and growth within biofilms attached directly to the bone surface or to orthopaedic implants [20,28]. Moving forward, a better understanding of the heterogenous host tissue niches occupied by S. aureus during invasive infection will facilitate new developments in antibiotic and adjunctive therapeutic design. In addition, understanding how these host environments impose metabolic constraints on S. aureus that result in the emergence of tolerant subpopulations (e.g. small colony variants or persisters) will fuel new discoveries in antibiotic development [29,30].
A second factor that contributes to antibiotic failure for susceptible staphylococcal isolates is the inability of standard antimicrobial susceptibility testing (AST) methods using rich bacterial media to fully recapitulate the complex in-vivo host environment during infection. Invading bacteria encounter myriad physiologic alterations in vivo, such as tissue hypoxia, limited nutrient availability, the presence of antimicrobial peptides, pH differences, changes in buffering capacity and altered osmolarity, among others. Accordingly, some investigators have sought to perform AST in media that faithfully model the host environment [31,32,33■,34,35]. Such media compositions range from bodily fluids (e.g. urine or bronchoalveolar lavage fluid), to media with altered nutrient, pH or cation concentrations, to media containing antimicrobial peptides or immune cells. Modelling of the host environment during AST is a critical area for future research, and this effort will benefit from new developments for rapid AST profiling [36–41].
Another important emerging approach to develop new treatments for S. aureus infection is the repurposing of drugs already approved for other clinical indications. To this end, several recent studies have highlighted compounds with ‘antivirulence’ activity against S. aureus [42]. Antivirulence compounds are defined as those that target bacterial virulence factors or pathways that regulate virulence rather than directly inhibiting the growth of, or killing, bacteria. Potential advantages of such compounds include that they might be less prone to select for drug resistance, they can be combined with traditional antimicrobial agents to improve treatment outcomes, and they can mitigate tissue damage and dysfunction induced by bacteria. One example of antivirulence compounds for treatment of staphylococcal infection are so-called ‘quorum-quenching’ drugs, which target the accessory gene regulator (Agr) locus to inhibit multiple classes of virulence factors in S. aureus [42–45]. Examples of staphylococcal quorum quenching antivirulence compounds include the FDA-approved NSAID diflunisal [46], auto-inducing peptide analogues [44], savirin [47], Solonamide B [48], the fungal metabolite apicidin [49] and compounds produced by coagulase-negative staphylococci [50,51]. Other antivirulence compounds target bacterial stress responses that are necessary for survival in host tissues in the setting of inflammatory responses. For example, lipid-lowering squalene synthase inhibitors can also inhibit the production of the antioxidant staphyloxanthin, which is the golden carotenoid pigment for which S. aureus is named [52]. Finally, passive immunoprophylaxis with monoclonal antibodies that block staphylococcal toxins is a highly active area of research that is poised to deliver new antivirulence therapies. These strategies are discussed further in the following section.
PREVENTION OF STAPHYLOCOCCUS AUREUS DISEASE: VACCINE DEVELOPMENT
Few, if any, pathogens have resulted in as many unsuccessful vaccine attempts as S. aureus. The first known reported attempt at a staphylococcal vaccine construct was a whole-cell vaccine developed in 1902 [53]. Over the subsequent 119 years, many other putative vaccines were developed, with a surge in attempts in recent years. As of the time of this writing, however, S. aureus has eluded all passive and active immunization attempts to date. A clear need for a S. aureus vaccine exists, as current rates of invasive S. aureus infections in children exceed the rates of invasive H. influenzae type B and S. pneumoniae before vaccines were deployed against these other important invasive bacterial pathogens [4].
Current opinions in the field are that prior staphylococcal vaccine attempts have been unsuccessful in humans for a combination of reasons. First, many gaps still exist in our understanding of what defines functional immunity against S. aureus in the human host. Animal and in-vitro models are crucial for fundamental understanding of key aspects of S. aureus bacteriology and metabolism [54], but inferences regarding virulence factors and targets of the host response have shown to translate relatively poorly. Similarly, the field lacks a reliable serologic correlate of protection against S. aureus. Previous, successful bacterial vaccine development (e.g. Hib and pneumococcus) involved clear markers of functional immunity, such as opsonophagocytic assays that reliably translated to efficacy. Further, it remains unclear which of the many S. aureus virulence factors are actually produced in the setting of invasive human infection, are necessary for pathogenesis and intervenable by immunization. Traditional assessments have focused on immunodominant epitopes, while the most important antigens may be relatively less immunogenic. These gaps result in a poor understanding of which specific antigen(s) should be targeted in an ideal S. aureus vaccine.
Significant recent data suggest that the toxins of S. aureus represent a promising target for intervention against this organism [55], for several reasons. First, S. aureus expresses numerous host-evasion (and specifically antibody-evasion) mechanisms on the cell surface [56], such as staphylococcal protein A (SpA), which inhibits normal antibody function by binding the Fc antibody region with high affinity as well as facilitating B cell apoptosis [57,58]. Antibodies targeting toxins and acting extracellularly, may be relatively spared from S. aureus’ wealth of antiantibody mechanisms. Second, the pore-forming family of toxins [e.g. alpha-haemolysin (Hla), PVL, LukAB, LukED], a crucial component of staphylococcal evasion of innate host defenses, potently disrupt innate host immunity by a variety of mechanisms including the lysis of phagocytes [59]. Importantly, these toxins (particularly Hla and LukAB) are known to be produced in the setting of invasive human infection and targeted by the adaptive host response [60–62].
Recent findings suggest that the pore-forming toxins warrant continued investigation as potential targets of intervention, particularly for the amelioration or prevention of invasive disease and bloodstream infections [55]. This intervention could occur via active immunization against toxoid constructs, as recently demonstrated in animal models for LukAB [63■,64], PVL and Hla [65], or via passive immunization with neutralizing antibodies (i.e. monoclonal or oligoclonal preparations) in high-risk hosts [62,66,67].
Multivalent (i.e. simultaneously targeting a variety of important antigens) vaccine constructs are likely to be a necessary approach given the complexity of this organism. Moreover, a worthwhile goal is likely not the eradication of this organism nor prevention of colonization or even cutaneous infection, but rather amelioration of severe, invasive S. aureus disease and the associated morbidity and mortality. Numerous novel constructs and putative vaccines are in development (Table 2), many of which align with the priorities mentioned above, and ongoing work will determine whether the as-yet-insurmountable challenge of preventing S. aureus disease by vaccination will soon be overcome.
Table 2.
Staphylococcus aureus vaccine candidates currently in human trials, as of May 2021
| Specific Components | Name / Description | Manufacturer | Status of Development |
|---|---|---|---|
| Toxins/Leukocidins | |||
| Two pore-forming toxins: toxoid of α-toxin, and recombinant PVL subunit (rLukS-PV) | Two-component toxoid / subunit | Nabi Biopharmaceuticals | Phase 1 safety and immunogenicity complete |
| Human mAb targeting α-toxin | AR-301 (mAb) | Aridis Pharmaceuticals | Ongoing trial in ventilator-associated pneumonia (VAP), healthcare-associated bacterial pneumonia (HABP) and community-acquired pneumonia (CAP) |
| 5-valent mAB against α-toxin, PVL, HlgAB, HlbCD, LukED; and mAb against LukAB | ASN100 (mAb combination) | Arsanis | VAP; discontinued |
| Anti-Hla mAb | Suvratoxumab | AstraZeneca | SAATELLITE trial for VAP |
| Surface adhesins or capsular polysaccharides | |||
| rAls3p-N (Candidal adhesin) | NDV-3A | NovaDigm Therapeutics | Phase 2, prevention of S. aureus colonization– complete. (A separate Phase 2a trial in patients with Hyper IgE syndrome was discontinued due to adverse effects) |
| Capsular polysaccharides conjugated to tetanus toxin, α-toxoid, clumping factor A | 4C-Staph | Novartis/GlaxoSmithKline | Phase 1 Safety and immunogenicity–complete |
| Nutritional transporters | |||
| IsdB-N2, MntC, Hla, SEB, SpA | Five-component recombinant antigen | Chengdu Olymvax Biopharmaceuticals | Open-label phase 1b safety and immunogenicity; Ongoing |
| Not disclosed | |||
| Five recombinant antigens, bioconjugated and adjuvanted; components not publicly disclosed | Sa-5Ag | GlaxoSmithKline | Phase 1 safety and immunogenicity, patients with recent SSTI; Ongoing |
FUTURE HORIZONS AND OPPORTUNITIES
There are many exciting opportunities on the horizon to improve the diagnosis and treatment of staphylococcal infections. As noted above, continued innovation of AST methods will improve prediction of antibiotic success vs. failure in an increasingly medically complex population. Basic science investigations into the many niches that S. aureus can occupy in host tissues and how these niches impact antigen display, virulence factor production and bacterial metabolism, will improve efforts to predict and therapeutically target antibiotic failure.
Looking ahead, we suggest two areas of focus that could transform the care of patients suffering from S. aureus infection. First, as the electronic health record (EHR) becomes more sophisticated and integrated with patient metadata, there is an opportunity to discover factors that predict susceptibility to, or protection from infectious diseases. For example, multiple studies from the investigative team lead by Dr Vance Fowler have identified loci that predict enhanced susceptibility to staphylococcal disease in murine and human infection [68,69■■,70,71]. More recently, the MRSA Systems Immunobiology Group performed whole exome sequencing on a well matched cohort of prospectively enrolled patients with persistent or resolving MRSA bacteraemia. A single nucleotide polymorphism in the intronic region of DNMT3A was found to differentiate between persistent and resolving S. aureus bacteraemia. Mechanistically, DNMT3A variants were predicted to alter the host response to infection via methylation of key regulatory genes and reduced IL-10 production [69■■]. This study is paradigmatic of ‘bedside-to-bench’ research studies that leverage well constructed patient cohorts, EHR variables and genomic data to predict susceptibility to infectious diseases and resultant complications. As tools to mine the EHR and DNA databases become more sophisticated, it is possible to predict genetically encoded disease patterns using tools such as pheWAS (phenome-wide association studies) and phenotypic risk scoring [72,73]. Such tools and databases could also enable personalized antibiotic dosing regimens to maximize safety and efficacy.
A second area of focus for future research to transform management of S. aureus infection is the development of new diagnostic modalities that specifically detect viable bacteria or immune responses to S. aureus. Although rapid molecular diagnostics have improved the time to diagnosis for typical presentations of S. aureus disease, there remains a need for improved detection of invasive infection in those patients who have been pretreated with antibiotics, or in whom recrudescent infection is suspected following initial antibiotic therapy. In these cases, cultures are often negative, and diagnosis by traditional molecular diagnostics may require invasive procedures to obtain new specimens. One exciting development in both preclinical models and early human clinical trials is the development of new imaging modalities to specifically detect the presence of viable bacteria. For example, studies in animal models have shown the power of creating imaging probes to detect the activity of specific staphylococcal virulence factors such as staphylocoagulase or nuclease [74,75]. Similarly, labelling specific metabolites that are selectively imported by bacteria can enable the creation of pathogen-specific imaging probes [76–78]. Finally, chemically modifying antibiotics for use as PET probes has shown promise in the diagnosis of infection and monitoring of treatment responses [79–81]. These approaches might hold considerable value for patients with posttraumatic or postsurgical staphylococcal infection, wherein it can be difficult to distinguish sterile inflammation from ongoing infection.
Finally, the adaptive host response to invasive staphylococcal infections may also be leveraged as a diagnostic opportunity. Presence of specific antibodies targeting those factors strongly associated with invasive disease, rather than colonization or noninvasive infections, may assist with the diagnosis of invasive infections (e.g. osteomyelitis, septic arthritis or visceral abscesses) that may be culture-negative due to antimicrobial pretreatment or inadequate diagnostic specimens. Pilot data suggest that antibodies against the leukocidin LukAB, which is ubiquitous among clinical isolates and abundantly produced in the setting of invasive disease, may be promising in this regard [82]. Similarly, antiglucosaminidase antibodies have shown promise as a serologic diagnostic for S. aureus osteomyelitis [83]. Notably, the lack of antiglucosaminidase antibodies was associated with worsened clinical outcomes and higher mortality, suggesting a fundamental role of this aspect of the adaptive response in S. aureus infection [84].
CONCLUSION
Despite myriad scientific breakthroughs, discoveries and novel therapeutics, S. aureus remains a scourge and a major human pathogen causing a wide variety of clinical phenotypes. Notably, the organism continues to evolve, both in terms of its dominant circulating lineages (and the virulence factors contained therein), and regarding antimicrobial resistance. USA300 CA-MRSA is no longer the dominant clone driving invasive paediatric infections, and a relative rise of MSSA (every bit as severe as MRSA) is the hallmark in recent years. The identification and development of novel preventives and therapeutics will be paramount in attempting to keep pace with this highly successful organism.
KEY POINTS.
Currently, circulating invasive strains of S. aureus continue to evolve, and the USA300 CA-MRSA epidemic clone is no longer dominant; in fact, MSSA is more frequent in recent series, though equally as severe as invasive MRSA.
Numerous new antistaphylococcal antimicrobials have been developed in recent years, though paediatric data remain limited.
Recent scientific breakthroughs highlight the opportunity for novel interventions against S. aureus by interfering with virulence rather than by traditional antimicrobial mechanisms.
A S. aureus vaccine remains elusive; the reasons for this are multifactorial, and lessons learned from prior unsuccessful attempts may create a path towards an effective preventive.
Acknowledgements
No writing assistance was used in preparation of this manuscript.
Financial support and sponsorship
J.E.C. is supported by R01AI132560 (NIAID), R01AI145992 (NIAID), a Career Award for Medical Scientists from the Burroughs Wellcome Fund, and a Crohn’s and Colitis Foundation Senior Research Award. I.T. is supported by R01AI139172 (NIAID).
Footnotes
Conflicts of interest
I.T. has served as a consultant for Nashville Biosciences and Horizon Therapeutics.
REFERENCES AND RECOMMENDED READING
Papers of particular interest, published within the annual period of review, have been highlighted as:
■ of special interest
■■ of outstanding interest
- 1.■■.Kourtis AP, Hatfield K, Baggs J, et al. Vital signs: epidemiology and recent trends in methicillin-resistant and in methicillin-susceptible Staphylococcus aureus bloodstream infections: United States. MMWR Morb Mortal Wkly Rep 2019; 68:214–219. [DOI] [PMC free article] [PubMed] [Google Scholar]; This large study demonstrated that although MRSA rates have gradually declined (likely due in part to improved infection control measures), MSSA rates have not improved.
- 2.Klevens RM, Morrison MA, Nadle J, et al. Invasive methicillin-resistant Staphylococcus aureus infections in the United States. JAMA 2007; 298:1763–1771. [DOI] [PubMed] [Google Scholar]
- 3.Spaulding AB, Thurm C, Courter JD, et al. Epidemiology of Staphylococcus aureus infections in patients admitted to freestanding pediatric hospitals, 2009–2016. Infect Control Hosp Epidemiol 2018; 39:1487–1490. [DOI] [PubMed] [Google Scholar]
- 4.Proctor RA. Have we outlived the concept of commensalism for Staphylococcus aureus? Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2020. [Epub]. doi: 10.1093/cid/ciaa1431. [DOI] [PubMed] [Google Scholar]
- 5.Turner NA, Sharma-Kuinkel BK, Maskarinec SA, et al. Methicillin-resistant Staphylococcus aureus: an overview of basic and clinical research. Nat Rev Microbiol 2019; 17:203–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sutter DE, Milburn E, Chukwuma U, et al. Changing susceptibility of Staphylococcus aureus in a US pediatric population. Pediatrics 2016; 137:e20153099. [DOI] [PubMed] [Google Scholar]
- 7.■■.Crandall H, Kapusta A, Killpack J, et al. Clinical and molecular epidemiology of invasive Staphylococcus aureus infection in Utah children; continued dominance of MSSA over MRSA. PLoS One 2020; 15:e0238991. [DOI] [PMC free article] [PubMed] [Google Scholar]; A recent study of invasive paediatric S. aureus disease showing that currently circulating invasive isolates are diverse, and the majority were MSSA and lack PVL. Also, MSSA cases were as severe as MRSA.
- 8.Kavanagh KT. Control of MSSA and MRSA in the United States: protocols, policies, risk adjustment and excuses. Antimicrob Resist Infect Control 2019; 8:103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.■.Thomsen IP, Kadari P, Soper NR, et al. Molecular epidemiology of invasive Staphylococcus aureus infections and concordance with colonization isolates. J Pediatr 2019; 210:173–177. [DOI] [PMC free article] [PubMed] [Google Scholar]; Invasive MSSA infection was more common than MRSA infection in this cohort, and patients with MSSA infection were significantly more likely than those with MRSA infection to have concordant colonizing isolates across multiple anatomic.
- 10.Liu C, Bayer A, Cosgrove SE, et al. Clinical practice guidelines by the infectious diseases society of america for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children. Clin Infect Dis 2011; 52:e18–e55. [DOI] [PubMed] [Google Scholar]
- 11.Stevens DL, Bisno AL, Chambers HF, et al. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America. Clin Infect Dis 2014; 59:e10–e52. [DOI] [PubMed] [Google Scholar]
- 12.Zhanel GG, Sniezek G, Schweizer F, et al. Ceftaroline: a novel broad-spectrum cephalosporin with activity against meticillin-resistant Staphylococcus aureus. Drugs 2009; 69:809–831. [DOI] [PubMed] [Google Scholar]
- 13.Yim J, Molloy LM, Newland JG. Use of ceftaroline fosamil in children: review of current knowledge and its application. Infect Dis Ther 2017; 6:57–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhanel GG, Calic D, Schweizer F, et al. New lipoglycopeptides: a comparative review of dalbavancin, oritavancin and telavancin. Drugs 2010; 70:859–886. [DOI] [PubMed] [Google Scholar]
- 15.Bradley JS, Puttagunta S, Rubino CM, et al. Pharmacokinetics, safety and tolerability of single dose dalbavancin in children 12–17 years of age. Pediatr Infect Dis J 2015; 34:748–752. [DOI] [PubMed] [Google Scholar]
- 16.Bradley J, Arrieta A, Bokesch P, et al. 1719. Pharmacokinetics (PK) of oritavancin in children: the ORKIDS Trial. Open Forum Infect Dis 2018; 5(Suppl 1):S52. [Google Scholar]
- 17.Bradley JS, Flanagan SD, Arrieta AC, et al. Pharmacokinetics, safety and tolerability of single oral or intravenous administration of 200 mg tedizolid phosphate in adolescents. Pediatr Infect Dis J 2016; 35:628–633. [DOI] [PubMed] [Google Scholar]
- 18.■.Randolph AG, Xu R, Novak T, et al. Vancomycin monotherapy may be insufficient to treat methicillin-resistant Staphylococcus aureus coinfection in children with influenza-related critical illness. Clin Infect Dis 2019; 68:365–372. [DOI] [PMC free article] [PubMed] [Google Scholar]; Vancomycin monotherapy was associated with increased mortality in children with postinfluenza MRSA, highlighting a need for novel therapeutics and alternative regimens.
- 19.Thomsen IP. The concern for vancomycin failure in the treatment of pediatric Staphylococcus aureus disease. Clin Infect Dis 2019; 68:373–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gimza BD, Cassat JE. Mechanisms of antibiotic failure during Staphylococcus aureus osteomyelitis. Front Immunol 2021; 12:638085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Cheng AG, DeDent AC, Schneewind O, Missiakas D. A play in four acts: Staphylococcus aureus abscess formation. Trends Microbiol 2011; 19:225–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Cheng AG, McAdow M, Kim HK, et al. Contribution of coagulases towards Staphylococcus aureus disease and protective immunity. PLoS Pathog 2010; 6:e1001036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Guggenberger C, Wolz C, Morrissey JA, Heesemann J. Two distinct coagulase-dependent barriers protect Staphylococcus aureus from neutrophils in a three dimensional in vitro infection model. PLoS Pathog 2012; 8:e1002434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.McAdow M, Missiakas DM, Schneewind O. Staphylococcus aureus secretes coagulase and von Willebrand factor binding protein to modify the coagulation cascade and establish host infections. J Innate Immun 2012; 4:141−148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.McAdow M, Kim HK, Dedent AC, et al. Preventing Staphylococcus aureus sepsis through the inhibition of its agglutination in blood. PLoS Pathog 2011; 7:e1002307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jorch SK, Surewaard BG, Hossain M, et al. Peritoneal GATA6+ macrophages function as a portal for Staphylococcus aureus dissemination. J Clin Invest 2019; 129:4643–4656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Surewaard BG, Deniset JF, Zemp FJ, et al. Identification and treatment of the Staphylococcus aureus reservoir in vivo. J Exp Med 2016; 213:1141–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Urish KL, Cassat JE. Staphylococcus aureus osteomyelitis: bone, bugs, and surgery. Infect Immun 2020; 88:e00932–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bui LM, Conlon BP, Kidd SP. Antibiotic tolerance and the alternative lifestyles of Staphylococcus aureus. Essays Biochem 2017; 61:71−79. [DOI] [PubMed] [Google Scholar]
- 30.Rowe SE, Beam JE, Conlon BP. Recalcitrant Staphylococcus aureus infections: obstacles and solutions. Infect Immun 2021; 89:e00694–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ersoy SC, Heithoff DM, Barnes L, et al. Correcting a fundamental flaw in the paradigm for antimicrobial susceptibility testing. EBioMedicine 2017; 20:173–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lin L, Nonejuie P, Munguia J, et al. Azithromycin synergizes with cationic antimicrobial peptides to exert bactericidal and therapeutic activity against highly multidrug-resistant Gram-negative bacterial pathogens. EBioMedicine 2015; 2:690–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.■.Mercer DK, Torres MDT, Duay SS, et al. Antimicrobial susceptibility testing of antimicrobial peptides to better predict efficacy. Front Cell Infect Microbiol 2020; 10:326. [DOI] [PMC free article] [PubMed] [Google Scholar]; Antimicrobial peptides are promising as novel therapuetic agents against S. aureus, and this work outlines a pathway for efficient development of these molecules.
- 34.Mercier RC, Dietz RM, Mazzola JL, et al. Beneficial influence of platelets on antibiotic efficacy in an in vitro model of Staphylococcus aureus-induced endocarditis. Antimicrob Agents Chemother 2004; 48:2551−2557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sakoulas G, Okumura CY, Thienphrapa W, et al. Nafcillin enhances innate immune-mediated killing of methicillin-resistant Staphylococcus aureus. J Mol Med (Berl) 2014; 92:139–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Humphries RM. Update on susceptibility testing: genotypic and phenotypic methods. Clin Lab Med 2020; 40:433–446. [DOI] [PubMed] [Google Scholar]
- 37.Khan ZA, Siddiqui MF, Park S. Current and Emerging methods of antibiotic susceptibility testing. Diagnostics (Basel) 2019; 9:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Nonejuie P, Burkart M, Pogliano K, Pogliano J. Bacterial cytological profiling rapidly identifies the cellular pathways targeted by antibacterial molecules. Proc Natl Acad Sci U S A 2013; 110:16169–16174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Trotter AJ, Aydin A, Strinden MJ, O’Grady J. Recent and emerging technologies for the rapid diagnosis of infection and antimicrobial resistance. Curr Opin Microbiol 2019; 51:39–45. [DOI] [PubMed] [Google Scholar]
- 40.van Belkum A, Bachmann TT, Ludke G, et al. Developmental roadmap for antimicrobial susceptibility testing systems. Nat Rev Microbiol 2019; 17:51–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.van Belkum A, Dunne WM Jr. Next-generation antimicrobial susceptibility testing. J Clin Microbiol 2013; 51:2018–2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ford CA, Hurford IM, Cassat JE. Antivirulence strategies for the treatment of Staphylococcus aureus infections: a mini review. Front Microbiol 2020; 11:632706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Butrico CE, Cassat JE. Quorum sensing and toxin production in Staphylococcus aureus osteomyelitis: pathogenesis and paradox. Toxins (Basel) 2020; 12:516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Horswill AR, Gordon CP. Structure-activity relationship studies of small molecule modulators of the Staphylococcal accessory gene regulator. J Med Chem 2020; 63:2705–2730. [DOI] [PubMed] [Google Scholar]
- 45.Piewngam P, Chiou J, Chatterjee P, Otto M. Alternative approaches to treat bacterial infections: targeting quorum-sensing. Expert Rev Anti Infect Ther 2020; 18:499–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hendrix AS, Spoonmore TJ, Wilde AD, et al. Repurposing the nonsteroidal anti-inflammatory drug diflunisal as an osteoprotective, antivirulence therapy for Staphylococcus aureus osteomyelitis. Antimicrob Agents Chemother 2016; 60:5322–5330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Sully EK, Malachowa N, Elmore BO, et al. Selective chemical inhibition of agr quorum sensing in Staphylococcus aureus promotes host defense with minimal impact on resistance. PLoS Pathog 2014; 10:e1004174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Nielsen A, Mansson M, Bojer MS, et al. Solonamide B inhibits quorum sensing and reduces Staphylococcus aureus mediated killing of human neutrophils. PLoS One 2014; 9:e84992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Parlet CP, Kavanaugh JS, Crosby HA, et al. Apicidin attenuates MRSA virulence through quorum-sensing inhibition and enhanced host defense. Cell Rep 2019; 27:187–198. e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Brown MM, Kwiecinski JM, Cruz LM, et al. Novel peptide from commensal Staphylococcus simulans blocks methicillin-resistant Staphylococcus aureus quorum sensing and protects host skin from damage. Antimicrob Agents Chemother 2020; 64:e00172–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Williams MR, Costa SK, Zaramela LS, et al. Quorum sensing between bacterial species on the skin protects against epidermal injury in atopic dermatitis. Sci Transl Med 2019; 11:eaat8329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Liu CI, Liu GY, Song Y, et al. A cholesterol biosynthesis inhibitor blocks Staphylococcus aureus virulence. Science 2008; 319:1391−1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wright AE. Notes on the treatment of furunculosis, sycosis, and acne by the inoculation of a Staphylococcus aureus vaccine. The Lancet 1902; 159:874–884. [Google Scholar]
- 54.Thomsen IP, Liu GY. Targeting fundamental pathways to disrupt Staphylococcus aureus survival: clinical implications of recent discoveries. JCI Insight 2018; 3:e98216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Miller LS, Fowler VG, Shukla SK, et al. Development of a vaccine against Staphylococcus aureus invasive infections: evidence based on human immunity, genetics and bacterial evasion mechanisms. FEMS Microbiol Rev 2020; 44:123–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Foster TJ, Geoghegan JA, Ganesh VK, Hook M. Adhesion, invasion and evasion: the many functions of the surface proteins of Staphylococcus aureus. Nat Rev Microbiol 2014; 12:49–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kim HK, Falugi F, Thomer L, et al. Protein A suppresses immune responses during Staphylococcus aureus bloodstream infection in guinea pigs. mBio 2015; 6:e02369–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kim HK, Falugi F, Missiakas DM, Schneewind O. Peptidoglycan-linked protein A promotes T cell-dependent antibody expansion during Staphylococcus aureus infection. Proc Natl Acad Sci U S A 2016; 113:5718–5723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Spaan AN, van Strijp JAG, Torres VJ. Leukocidins: staphylococcal bi-component pore-forming toxins find their receptors. Nat Rev Microbiol 2017; 15:435–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Fritz SA, Tiemann KM, Hogan PG, et al. A serologic correlate of protective immunity against community-onset Staphylococcus aureus infection. Clin Infect Dis 2013; 56:1554–1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Thomsen IP, Dumont AL, James DB, et al. Children with invasive Staphylococcus aureusdisease exhibit a potently neutralizing antibody response to the cytotoxin LukAB. Infect Immun 2014; 82:1234–1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Thomsen IP, Sapparapu G, James DBA, et al. Monoclonal antibodies against the Staphylococcus aureus bicomponent leukotoxin AB isolated following invasive human infection reveal diverse binding and modes of action. J Infect Dis 2017; 215:1124–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.■.Fernandez J, Sanders H, Henn J, et al. Vaccination with detoxified leukocidin AB reduces bacterial load in a Staphylococcus aureus minipig deep surgical wound infection model. J Infect Dis 2021. jiab219 [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]; Preclinical data supporting the use of LukAB toxoids as a vaccine candidate.
- 64.Kailasan S, Kort T, Mukherjee I, et al. Rational design of toxoid vaccine candidates for Staphylococcus aureus leukocidin AB (LukAB). Toxins (Basel) 2019; 11:339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Tran VG, Venkatasubramaniam A, Adhikari RP, et al. Efficacy of active immunization with attenuated alpha-hemolysin and panton-valentine leukocidin in a rabbit model of Staphylococcus aureus necrotizing pneumonia. J Infect Dis 2020; 221:267–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hua L, Hilliard JJ, Shi Y, et al. Assessment of an antialpha-toxin monoclonal antibody for prevention and treatment of Staphylococcus aureus-induced pneumonia. Antimicrob Agents Chemother 2014; 58:1108–1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Verreault D, Ennis J, Whaley K, et al. Effective treatment of Staphylococcal enterotoxin B aerosol intoxication in Rhesus Macaques by using two parenterally administered high-affinity monoclonal antibodies. Antimicrob Agents Chemother 2019; 63:e02049–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ahn SH, Deshmukh H, Johnson N, et al. Two genes on A/J chromosome 18 are associated with susceptibility to Staphylococcus aureus infection by combined microarray and QTL analyses. PLoS Pathog 2010; 6:e1001088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.■■.Mba Medie F, Sharma-Kuinkel BK, Ruffin F, et al. Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients. Proc Natl Acad Sci U S A 2019; 116:20087–20096. [DOI] [PMC free article] [PubMed] [Google Scholar]; Example of genotype/phenotype correlations to identify host factors that may predispose patients to more severe S. aureus disease.
- 70.Scott WK, Medie FM, Ruffin F, et al. Human genetic variation in GLS2 is associated with development of complicated Staphylococcus aureus bacteremia. PLoS Genet 2018; 14:e1007667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yan Q, Sharma-Kuinkel BK, Deshmukh H, et al. Dusp3 and Psme3 are associated with murine susceptibility to Staphylococcus aureus infection and human sepsis. PLoS Pathog 2014; 10:e1004149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Bastarache L, Hughey JJ, Hebbring S, et al. Phenotype risk scores identify patients with unrecognized Mendelian disease patterns. Science 2018; 359:1233–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gamazon ER, Wheeler HE, Shah KP, et al. A gene-based association method for mapping traits using reference transcriptome data. Nat Genet 2015; 47:1091–1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Hernandez FJ, Huang L, Olson ME, et al. Noninvasive imaging of Staphylococcus aureus infections with a nuclease-activated probe. Nat Med 2014; 20:301–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Panizzi P, Nahrendorf M, Figueiredo JL, et al. In vivo detection of Staphylococcus aureus endocarditis by targeting pathogen-specific prothrombin activation. Nat Med 2011; 17:1142–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ordonez AA, Weinstein EA, Bambarger LE, et al. A systematic approach for developing bacteria-specific imaging tracers. J Nucl Med 2017; 58:144–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ordonez AA, Wintaco LM, Mota F, et al. Imaging Enterobacterales infections in patients using pathogen-specific positron emission tomography. Sci Transl Med 2021; 13:eabe9805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zhang Z, Ordonez AA, Wang H, et al. Positron emission tomography imaging with 2-[(18)F]F-p-aminobenzoic acid detects Staphylococcus aureus infections and monitors drug response. ACS Infect Dis 2018; 4:1635–1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ordonez AA, Wang H, Magombedze G, et al. Dynamic imaging in patients with tuberculosis reveals heterogeneous drug exposures in pulmonary lesions. Nat Med 2020; 26:529–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sellmyer MA, Lee I, Hou C, et al. Bacterial infection imaging with [(18)F]fluoropropyl-trimethoprim. Proc Natl Acad Sci U S A 2017;114:8372–8377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Tucker EW, Guglieri-Lopez B, Ordonez AA, et al. Noninvasive (11)C-rifampin positron emission tomography reveals drug biodistribution in tuberculous meningitis. Sci Transl Med 2018; 10:eaau0965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wood JB, Jones LS, Soper NR, et al. Serologic detection of antibodies targeting the leukocidin LukAB strongly predicts Staphylococcus aureus in children with invasive infection. J Pediatric Infect Dis Soc 2019; 8:128–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Nishitani K, Beck CA, Rosenberg AF, et al. A diagnostic serum antibody test for patients with Staphylococcus aureus osteomyelitis. Clin Orthop Relat Res 2015; 473:2735–2749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kates SL, Owen JR, Beck CA, et al. Lack of humoral immunity against glucosaminidase is associated with postoperative complications in Staphylococcus aureus osteomyelitis. J Bone Joint Surg Am 2020; 102: 1842–1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
